1
|
Thorens B. Neuronal glucose sensing mechanisms and circuits in the control of insulin and glucagon secretion. Physiol Rev 2024; 104:1461-1486. [PMID: 38661565 DOI: 10.1152/physrev.00038.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 04/16/2024] [Accepted: 04/20/2024] [Indexed: 04/26/2024] Open
Abstract
Glucose homeostasis is mainly under the control of the pancreatic islet hormones insulin and glucagon, which, respectively, stimulate glucose uptake and utilization by liver, fat, and muscle and glucose production by the liver. The balance between the secretions of these hormones is under the control of blood glucose concentrations. Indeed, pancreatic islet β-cells and α-cells can sense variations in glycemia and respond by an appropriate secretory response. However, the secretory activity of these cells is also under multiple additional metabolic, hormonal, and neuronal signals that combine to ensure the perfect control of glycemia over a lifetime. The central nervous system (CNS), which has an almost absolute requirement for glucose as a source of metabolic energy and thus a vital interest in ensuring that glycemic levels never fall below ∼5 mM, is equipped with populations of neurons responsive to changes in glucose concentrations. These neurons control pancreatic islet cell secretion activity in multiple ways: through both branches of the autonomic nervous system, through the hypothalamic-pituitary-adrenal axis, and by secreting vasopressin (AVP) in the blood at the level of the posterior pituitary. Here, we present the autonomic innervation of the pancreatic islets; the mechanisms of neuron activation by a rise or a fall in glucose concentration; how current viral tracing, chemogenetic, and optogenetic techniques allow integration of specific glucose sensing neurons in defined neuronal circuits that control endocrine pancreas function; and, finally, how genetic screens in mice can untangle the diversity of the hypothalamic mechanisms controlling the response to hypoglycemia.
Collapse
Affiliation(s)
- Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
2
|
Le J, Chen Y, Yang W, Chen L, Ye J. Metabolic basis of solute carrier transporters in treatment of type 2 diabetes mellitus. Acta Pharm Sin B 2024; 14:437-454. [PMID: 38322335 PMCID: PMC10840401 DOI: 10.1016/j.apsb.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/10/2023] [Accepted: 08/09/2023] [Indexed: 02/08/2024] Open
Abstract
Solute carriers (SLCs) constitute the largest superfamily of membrane transporter proteins. These transporters, present in various SLC families, play a vital role in energy metabolism by facilitating the transport of diverse substances, including glucose, fatty acids, amino acids, nucleotides, and ions. They actively participate in the regulation of glucose metabolism at various steps, such as glucose uptake (e.g., SLC2A4/GLUT4), glucose reabsorption (e.g., SLC5A2/SGLT2), thermogenesis (e.g., SLC25A7/UCP-1), and ATP production (e.g., SLC25A4/ANT1 and SLC25A5/ANT2). The activities of these transporters contribute to the pathogenesis of type 2 diabetes mellitus (T2DM). Notably, SLC5A2 has emerged as a valid drug target for T2DM due to its role in renal glucose reabsorption, leading to groundbreaking advancements in diabetes drug discovery. Alongside SLC5A2, multiple families of SLC transporters involved in the regulation of glucose homeostasis hold potential applications for T2DM therapy. SLCs also impact drug metabolism of diabetic medicines through gene polymorphisms, such as rosiglitazone (SLCO1B1/OATP1B1) and metformin (SLC22A1-3/OCT1-3 and SLC47A1, 2/MATE1, 2). By consolidating insights into the biological activities and clinical relevance of SLC transporters in T2DM, this review offers a comprehensive update on their roles in controlling glucose metabolism as potential drug targets.
Collapse
Affiliation(s)
- Jiamei Le
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yilong Chen
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Wei Yang
- Metabolic Disease Research Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Ligong Chen
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Jianping Ye
- Metabolic Disease Research Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
- Research Center for Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
3
|
Sun B, Chen H, Xue J, Li P, Fu X. The role of GLUT2 in glucose metabolism in multiple organs and tissues. Mol Biol Rep 2023; 50:6963-6974. [PMID: 37358764 PMCID: PMC10374759 DOI: 10.1007/s11033-023-08535-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 05/17/2023] [Indexed: 06/27/2023]
Abstract
The glucose transporter family has an important role in the initial stage of glucose metabolism; Glucose transporters 2 (GLUTs, encoded by the solute carrier family 2, SLC2A genes) is the major glucose transporter in β-cells of pancreatic islets and hepatocytes but is also expressed in the small intestine, kidneys, and central nervous system; GLUT2 has a relatively low affinity to glucose. Under physiological conditions, GLUT2 transports glucose into cells and allows the glucose concentration to reach balance on the bilateral sides of the cellular membrane; Variation of GLUT2 is associated with various endocrine and metabolic disorders; In this study, we discussed the role of GLUT2 in participating in glucose metabolism and regulation in multiple organs and tissues and its effects on maintaining glucose homeostasis.
Collapse
Affiliation(s)
- Bo Sun
- Endorcrine and Metabolism Department, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Department of Infantile Endocrine Genetic Metabolism, Gansu Maternal and child Health Care Hospital, Lanzhou, 730000, China
| | - Hui Chen
- Endorcrine and Metabolism Department, Lanzhou University Second Hospital, Lanzhou, 730000, China.
| | - Jisu Xue
- EndEnorcrine and Metabolism Department, Shenzhen Bao 'an People's Hospital (Group), Shenzhen, 518100, China
| | - Peiwu Li
- Key Laboratory of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Xu Fu
- Key Laboratory of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, 730000, China
| |
Collapse
|
4
|
Su Q, Yuan F, Li X, Wang X, Yang K, Shao L, Li W. Wfs1 loss-of-function disrupts the composition of mouse pancreatic endocrine cells from birth and impairs Glut2 localization to cytomembrane in pancreatic β cells. Biochem Biophys Res Commun 2023; 658:80-87. [PMID: 37027908 DOI: 10.1016/j.bbrc.2023.03.074] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
Wfs1 is an endoplasmic reticulum (ER) membrane located protein highly expressed in pancreatic β cells and brain. Wfs1 deficiency causes adult pancreatic β cells dysfunction following β cells apoptosis. Previous studies mainly focus on the Wfs1 function in adult mouse pancreatic β cells. However, whether Wfs1 loss-of-function impairs mouse pancreatic β cell from its early development is unknown. In our study, Wfs1 deficiency disrupts the composition of mouse pancreatic endocrine cells from early postnatal day 0 (P0) to 8 weeks old, with decreased percentage of β cells and increased percentage of α and δ cells. Meanwhile, Wfs1 loss-of-function leads to reduced intracellular insulin content. Notably, Wfs1 deficiency impairs Glut2 localization and causes the accumulation of Glut2 in mouse pancreatic β cell cytoplasm. In Wfs1-deficient mice, glucose homeostasis is disturbed from early 3 weeks old to 8 weeks old. This work reveals that Wfs1 is significantly required for the composition of pancreatic endocrine cells and is essential for Glut2 localization in mouse pancreatic β cells.
Collapse
Affiliation(s)
- Qiang Su
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Fei Yuan
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xiaobo Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xuan Wang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Kaijiang Yang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Li Shao
- Department of VIP Clinic, Shanghai East Hospital, Tongji University School of Medicine, No. 1800 Yuntai Road, Pudong District, Shanghai, 200092, China.
| | - Weida Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China; Reg-Verse Therapeutics (Shanghai) Co. Ltd., Shanghai, 200120, China.
| |
Collapse
|
5
|
Barahona MJ, Langlet F, Labouèbe G, Croizier S, Picard A, Thorens B, García-Robles MA. GLUT2 expression by glial fibrillary acidic protein-positive tanycytes is required for promoting feeding-response to fasting. Sci Rep 2022; 12:17717. [PMID: 36271117 PMCID: PMC9587252 DOI: 10.1038/s41598-022-22489-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/14/2022] [Indexed: 01/18/2023] Open
Abstract
Feeding behavior is a complex process that depends on the ability of the brain to integrate hormonal and nutritional signals, such as glucose. One glucosensing mechanism relies on the glucose transporter 2 (GLUT2) in the hypothalamus, especially in radial glia-like cells called tanycytes. Here, we analyzed whether a GLUT2-dependent glucosensing mechanism is required for the normal regulation of feeding behavior in GFAP-positive tanycytes. Genetic inactivation of Glut2 in GFAP-expressing tanycytes was performed using Cre/Lox technology. The efficiency of GFAP-tanycyte targeting was analyzed in the anteroposterior and dorsoventral axes by evaluating GFP fluorescence. Feeding behavior, hormonal levels, neuronal activity using c-Fos, and neuropeptide expression were also analyzed in the fasting-to-refeeding transition. In basal conditions, Glut2-inactivated mice had normal food intake and meal patterns. Implementation of a preceeding fasting period led to decreased total food intake and a delay in meal initiation during refeeding. Additionally, Glut2 inactivation increased the number of c-Fos-positive cells in the ventromedial nucleus in response to fasting and a deregulation of Pomc expression in the fasting-to-refeeding transition. Thus, a GLUT2-dependent glucose-sensing mechanism in GFAP-tanycytes is required to control food consumption and promote meal initiation after a fasting period.
Collapse
Affiliation(s)
- M. J. Barahona
- grid.5380.e0000 0001 2298 9663Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile ,grid.5380.e0000 0001 2298 9663Present Address: Laboratorio de Neurobiología y células madres (NeuroCellT), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - F. Langlet
- grid.9851.50000 0001 2165 4204Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland ,grid.9851.50000 0001 2165 4204Present Address: Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - G. Labouèbe
- grid.9851.50000 0001 2165 4204Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - S. Croizier
- grid.9851.50000 0001 2165 4204Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - A. Picard
- grid.9851.50000 0001 2165 4204Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Bernard Thorens
- grid.9851.50000 0001 2165 4204Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - María A. García-Robles
- grid.5380.e0000 0001 2298 9663Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile ,grid.412185.b0000 0000 8912 4050Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaiso, Chile
| |
Collapse
|
6
|
Lipid biosynthesis enzyme Agpat5 in AgRP-neurons is required for insulin-induced hypoglycemia sensing and glucagon secretion. Nat Commun 2022; 13:5761. [PMID: 36180454 PMCID: PMC9525695 DOI: 10.1038/s41467-022-33484-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
The counterregulatory response to hypoglycemia that restores normal blood glucose levels is an essential physiological function. It is initiated, in large part, by incompletely characterized brain hypoglycemia sensing neurons that trigger the secretion of counterregulatory hormones, in particular glucagon, to stimulate hepatic glucose production. In a genetic screen of recombinant inbred BXD mice we previously identified Agpat5 as a candidate regulator of hypoglycemia-induced glucagon secretion. Here, using genetic mouse models, we demonstrate that Agpat5 expressed in agouti-related peptide neurons is required for their activation by hypoglycemia, for hypoglycemia-induced vagal nerve activity, and glucagon secretion. We find that inactivation of Agpat5 leads to increased fatty acid oxidation and ATP production and that suppressing Cpt1a-dependent fatty acid import into mitochondria restores hypoglycemia sensing. Collectively, our data show that AgRP neurons are involved in the control of glucagon secretion and that Agpat5, by partitioning fatty acyl-CoAs away from mitochondrial fatty acid oxidation and ATP generation, ensures that the fall in intracellular ATP, which triggers neuronal firing, faithfully reflects changes in glycemia. During hypoglycemia, glucagon secretion is part of the mechanism needed to restore normal blood glucose levels. Here, Strembitska et al. report that sensing of hypoglycemia by AgRP neurons requires Agpat5, an enzyme which prevents fatty acids from entering the mitochondria for ATP production, ensuring correct neuronal activation and glucagon secretion.
Collapse
|
7
|
Targeting Ceramides and Adiponectin Receptors in the Islet of Langerhans for Treating Diabetes. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27186117. [PMID: 36144859 PMCID: PMC9502927 DOI: 10.3390/molecules27186117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022]
Abstract
Ceramides belong to the sphingolipid family and represent the central hub of the sphingolipid network. In obesity, oversupply of saturated fatty acids including palmitate raises ceramide levels which can be detrimental to cells. Elevated ceramides can cause insulin resistance, endoplasmic reticulum stress, and mitochondrial dysfunction. Studies over the last few decades have highlighted the role played by ceramides in pancreatic islet β-cell apoptosis, especially under glucolipotoxic and inflammatory conditions. This review focuses on ceramides and adiponectin receptor signaling, summarizing recent advancements in our understanding of their roles in islet β-cells and the discovery of zinc-dependent lipid hydrolase (ceramidase) activity of adiponectin receptors. The therapeutic potential of targeting these events to prevent islet β-cell loss for treating diabetes is discussed.
Collapse
|
8
|
MicroRNA-21 promotes pancreatic β cell function through modulating glucose uptake. Nat Commun 2022; 13:3545. [PMID: 35729232 PMCID: PMC9213410 DOI: 10.1038/s41467-022-31317-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/08/2022] [Indexed: 02/07/2023] Open
Abstract
Pancreatic β cell dysfunction contributes to the pathogenesis of type 2 diabetes. MiR-21 has been shown to be induced in the islets of glucose intolerant patients and type 2 diabetic mice. However, the role of miR-21 in the regulation of pancreatic β cell function remains largely elusive. In the current study, we identify the pathway by which miR-21 regulates glucose-stimulated insulin secretion utilizing mice lacking miR-21 in their β cells (miR-21βKO). We find that miR-21βKO mice develop glucose intolerance due to impaired glucose-stimulated insulin secretion. Mechanistic studies reveal that miR-21 enhances glucose uptake and subsequently promotes insulin secretion by up-regulating Glut2 expression in a miR-21-Pdcd4-AP-1 dependent pathway. Over-expression of Glut2 in knockout islets results in rescue of the impaired glucose-stimulated insulin secretion. Furthermore, we demonstrate that delivery of miR-21 into the pancreas of type 2 diabetic db/db male mice is able to promote Glut2 expression and reduce blood glucose level. Taking together, our results reveal that miR-21 in islet β cell promotes insulin secretion and support a role for miR-21 in the regulation of pancreatic β cell function in type 2 diabetes.
Collapse
|
9
|
Yang YHC, Briant LJB, Raab CA, Mullapudi ST, Maischein HM, Kawakami K, Stainier DYR. Innervation modulates the functional connectivity between pancreatic endocrine cells. eLife 2022; 11:64526. [PMID: 35373736 PMCID: PMC9007585 DOI: 10.7554/elife.64526] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 04/03/2022] [Indexed: 11/20/2022] Open
Abstract
The importance of pancreatic endocrine cell activity modulation by autonomic innervation has been debated. To investigate this question, we established an in vivo imaging model that also allows chronic and acute neuromodulation with genetic and optogenetic tools. Using the GCaMP6s biosensor together with endocrine cell fluorescent reporters, we imaged calcium dynamics simultaneously in multiple pancreatic islet cell types in live animals in control states and upon changes in innervation. We find that by 4 days post fertilization in zebrafish, a stage when islet architecture is reminiscent of that in adult rodents, prominent activity coupling between beta cells is present in basal glucose conditions. Furthermore, we show that both chronic and acute loss of nerve activity result in diminished beta–beta and alpha–beta activity coupling. Pancreatic nerves are in contact with all islet cell types, but predominantly with beta and delta cells. Surprisingly, a subset of delta cells with detectable peri-islet neural activity coupling had significantly higher homotypic coupling with other delta cells suggesting that some delta cells receive innervation that coordinates their output. Overall, these data show that innervation plays a vital role in the maintenance of homotypic and heterotypic cellular connectivity in pancreatic islets, a process critical for islet function.
Collapse
Affiliation(s)
- Yu Hsuan Carol Yang
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Christopher A Raab
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Sri Teja Mullapudi
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Hans-Martin Maischein
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Japan
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
10
|
Moullé VS. Autonomic control of pancreatic beta cells: What is known on the regulation of insulin secretion and beta-cell proliferation in rodents and humans. Peptides 2022; 148:170709. [PMID: 34896576 DOI: 10.1016/j.peptides.2021.170709] [Citation(s) in RCA: 4] [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: 09/05/2021] [Revised: 11/17/2021] [Accepted: 12/07/2021] [Indexed: 11/21/2022]
Abstract
Insulin secretion and pancreatic beta-cell proliferation are tightly regulated by several signals such as hormones, nutrients, and neurotransmitters. However, the autonomic control of beta cells is not fully understood. In this review, we describe mechanisms involved in insulin secretion as well as metabolic and mitogenic actions on its target tissues. Since pancreatic islets are physically connected to the brain by nerves, parasympathetic and sympathetic neurotransmitters can directly potentiate or repress insulin secretion and beta-cell proliferation. Finally, we highlight the role of the autonomic nervous system in metabolic diseases such as diabetes and obesity.
Collapse
|
11
|
Thorens B. Neuronal regulation of glucagon secretion and gluconeogenesis. J Diabetes Investig 2022; 13:599-607. [PMID: 34989155 PMCID: PMC9017634 DOI: 10.1111/jdi.13745] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/02/2022] [Indexed: 11/29/2022] Open
Abstract
Hypoglycemia almost never develops in healthy individuals because multiple hypoglycemia sensing systems, located in the periphery and in the central nervous system trigger a coordinated counterregulatory hormonal response to restore normoglycemia. This involves not only the secretion of glucagon but also of epinephrine, norepinephrine, cortisol and growth hormone. Increased hepatic glucose production is also stimulated by direct autonomous nervous connections to the liver that stimulate glycogenolysis and gluconeogenesis. This counterregulatory response, however, becomes deregulated in a significant fraction of diabetic patients that receive insulin therapy. This leads to risk of developing hypoglycemic episodes, of increasing severity, which negatively impact the quality of life of the patients. How hypoglycemia is detected by the central nervous system is being actively investigated. Recent studies using novel molecular biological, optogenetic and chemogenetic techniques, allow the characterization of glucose sensing neurons, the mechanisms of hypoglycemia detection, the neuronal circuits in which they are integrated and the physiological responses they control. This review will discuss recent studies aimed at identifying central hypoglycemia sensing neuronal circuits, how neurons are activated by hypoglycemia, and how they restore normoglycemia.
Collapse
Affiliation(s)
- Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, 1015, Lausanne, Switzerland
| |
Collapse
|
12
|
Mirzadeh Z, Faber CL, Schwartz MW. Central Nervous System Control of Glucose Homeostasis: A Therapeutic Target for Type 2 Diabetes? Annu Rev Pharmacol Toxicol 2022; 62:55-84. [PMID: 34990204 PMCID: PMC8900291 DOI: 10.1146/annurev-pharmtox-052220-010446] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Historically, pancreatic islet beta cells have been viewed as principal regulators of glycemia, with type 2 diabetes (T2D) resulting when insulin secretion fails to compensate for peripheral tissue insulin resistance. However, glycemia is also regulated by insulin-independent mechanisms that are dysregulated in T2D. Based on evidence supporting its role both in adaptive coupling of insulin secretion to changes in insulin sensitivity and in the regulation of insulin-independent glucose disposal, the central nervous system (CNS) has emerged as a fundamental player in glucose homeostasis. Here, we review and expand upon an integrative model wherein the CNS, together with the islet, establishes and maintains the defended level of glycemia. We discuss the implications of this model for understanding both normal glucose homeostasis and T2D pathogenesis and highlight centrally targeted therapeutic approaches with the potential to restore normoglycemia to patients with T2D.
Collapse
Affiliation(s)
- Zaman Mirzadeh
- Ivy Brain Tumor Center, Department of Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona 85013, USA;
| | - Chelsea L Faber
- Ivy Brain Tumor Center, Department of Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona 85013, USA;
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, Washington 98109, USA;
| | - Michael W Schwartz
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, Washington 98109, USA;
| |
Collapse
|
13
|
Yang M, Chen Y, Vagionitis S, Körtvely E, Ueffing M, Schmachtenberg O, Hu Z, Jiao K, Paquet-Durand F. Expression of glucose transporter-2 in murine retina: Evidence for glucose transport from horizontal cells to photoreceptor synapses. J Neurochem 2021; 160:283-296. [PMID: 34726780 DOI: 10.1111/jnc.15533] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 01/30/2023]
Abstract
The retina has the highest relative energy consumption of any tissue, depending on a steady supply of glucose from the bloodstream. Glucose uptake is mediated by specific transporters whose regulation and expression are critical for the pathogenesis of many diseases, including diabetes and diabetic retinopathy. Here, we used immunofluorescence to show that glucose transporter-2 (GLUT2) is expressed in horizontal cells of the mouse neuroretina in proximity to inner retinal capillaries. To study the function of GLUT2 in the murine retina, we used organotypic retinal explants, cultivated under entirely controlled, serum-free conditions and exposed them to streptozotocin, a cytotoxic drug transported exclusively by GLUT2. Contrary to our expectations, streptozotocin did not measurably affect horizontal cell viability, while it ablated rod and cone photoreceptors in a concentration-dependent manner. Staining for poly-ADP-ribose (PAR) indicated that the detrimental effect of streptozotocin on photoreceptors may be associated with DNA damage. The negative effect of streptozotocin on the viability of rod photoreceptors was counteracted by co-administration of either the inhibitor of connexin-formed hemi-channels meclofenamic acid or the blocker of clathrin-mediated endocytosis dynasore. Remarkably, cone photoreceptors were not protected from streptozotocin-induced degeneration by neither of the two drugs. Overall, these data suggest the existence of a GLUT2-dependent glucose transport shuttle, from horizontal cells into photoreceptor synapses. Moreover, our study points at different glucose uptake mechanisms in rod and cone photoreceptors.
Collapse
Affiliation(s)
- Ming Yang
- Affiliated Hospital of Yunnan University & 2nd People's Hospital of Yunnan Province, Kunming, China.,Yunnan Eye Institute & Key Laboratory of Yunnan Province, Kunming, China.,1st Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yiyi Chen
- Institute for Ophthalmic Research, Eberhard-Karls-Universität, Tübingen, Germany
| | - Stavros Vagionitis
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Elöd Körtvely
- Roche Pharma Research and Early Development, Immunology, Infectious Diseases and Ophthalmology (I2O), Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Marius Ueffing
- Institute for Ophthalmic Research, Eberhard-Karls-Universität, Tübingen, Germany
| | - Oliver Schmachtenberg
- CINV, Instituto de Biología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Zhulin Hu
- Affiliated Hospital of Yunnan University & 2nd People's Hospital of Yunnan Province, Kunming, China.,Yunnan Eye Institute & Key Laboratory of Yunnan Province, Kunming, China
| | - Kangwei Jiao
- Affiliated Hospital of Yunnan University & 2nd People's Hospital of Yunnan Province, Kunming, China.,Yunnan Eye Institute & Key Laboratory of Yunnan Province, Kunming, China
| | | |
Collapse
|
14
|
Reed J, Bain S, Kanamarlapudi V. A Review of Current Trends with Type 2 Diabetes Epidemiology, Aetiology, Pathogenesis, Treatments and Future Perspectives. Diabetes Metab Syndr Obes 2021; 14:3567-3602. [PMID: 34413662 PMCID: PMC8369920 DOI: 10.2147/dmso.s319895] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/09/2021] [Indexed: 12/13/2022] Open
Abstract
Type 2 diabetes (T2D), which has currently become a global pandemic, is a metabolic disease largely characterised by impaired insulin secretion and action. Significant progress has been made in understanding T2D aetiology and pathogenesis, which is discussed in this review. Extrapancreatic pathology is also summarised, which demonstrates the highly multifactorial nature of T2D. Glucagon-like peptide (GLP)-1 is an incretin hormone responsible for augmenting insulin secretion from pancreatic beta-cells during the postprandial period. Given that native GLP-1 has a very short half-life, GLP-1 mimetics with a much longer half-life have been developed, which are currently an effective treatment option for T2D by enhancing insulin secretion in patients. Interestingly, there is continual emerging evidence that these therapies alleviate some of the post-diagnosis complications of T2D. Additionally, these therapies have been shown to induce weight loss in patients, suggesting they could be an alternative to bariatric surgery, a procedure associated with numerous complications. Current GLP-1-based therapies all act as orthosteric agonists for the GLP-1 receptor (GLP-1R). Interestingly, it has emerged that GLP-1R also has allosteric binding sites and agonists have been developed for these sites to test their therapeutic potential. Recent studies have also demonstrated the potential of bi- and tri-agonists, which target multiple hormonal receptors including GLP-1R, to more effectively treat T2D. Improved understanding of T2D aetiology/pathogenesis, coupled with the further elucidation of both GLP-1 activity/targets and GLP-1R mechanisms of activation via different agonists, will likely provide better insight into the therapeutic potential of GLP-1-based therapies to treat T2D.
Collapse
Affiliation(s)
- Josh Reed
- Institute of Life Science 1, Medical School, Swansea University, Swansea, SA2 8PP, UK
| | - Stephen Bain
- Institute of Life Science 1, Medical School, Swansea University, Swansea, SA2 8PP, UK
| | | |
Collapse
|
15
|
Picard A, Metref S, Tarussio D, Dolci W, Berney X, Croizier S, Labouebe G, Thorens B. Fgf15 Neurons of the Dorsomedial Hypothalamus Control Glucagon Secretion and Hepatic Gluconeogenesis. Diabetes 2021; 70:1443-1457. [PMID: 33883213 PMCID: PMC8336003 DOI: 10.2337/db20-1121] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 04/03/2021] [Indexed: 11/18/2022]
Abstract
The counterregulatory response to hypoglycemia is an essential survival function. It is controlled by an integrated network of glucose-responsive neurons, which trigger endogenous glucose production to restore normoglycemia. The complexity of this glucoregulatory network is, however, only partly characterized. In a genetic screen of a panel of recombinant inbred mice we previously identified Fgf15, expressed in neurons of the dorsomedial hypothalamus (DMH), as a negative regulator of glucagon secretion. Here, we report on the generation of Fgf15CretdTomato mice and their use to further characterize these neurons. We show that they were glutamatergic and comprised glucose-inhibited and glucose-excited neurons. When activated by chemogenetics, Fgf15 neurons prevented the increase in vagal nerve firing and the secretion of glucagon normally triggered by insulin-induced hypoglycemia. On the other hand, they increased the activity of the sympathetic nerve in the basal state and prevented its silencing by glucose overload. Higher sympathetic tone increased hepatic Creb1 phosphorylation, Pck1 mRNA expression, and hepatic glucose production leading to glucose intolerance. Thus, Fgf15 neurons of the DMH participate in the counterregulatory response to hypoglycemia by a direct adrenergic stimulation of hepatic glucose production while suppressing vagally induced glucagon secretion. This study provides new insights into the complex neuronal network that prevents the development of hypoglycemia.
Collapse
Affiliation(s)
- Alexandre Picard
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Salima Metref
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - David Tarussio
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Wanda Dolci
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Xavier Berney
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Sophie Croizier
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Gwenaël Labouebe
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
16
|
Gervais M, Labouèbe G, Picard A, Thorens B, Croizier S. EphrinB1 modulates glutamatergic inputs into POMC-expressing progenitors and controls glucose homeostasis. PLoS Biol 2020; 18:e3000680. [PMID: 33253166 PMCID: PMC7728393 DOI: 10.1371/journal.pbio.3000680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 12/10/2020] [Accepted: 11/05/2020] [Indexed: 12/25/2022] Open
Abstract
Proopiomelanocortin (POMC) neurons are major regulators of energy balance and glucose homeostasis. In addition to being regulated by hormones and nutrients, POMC neurons are controlled by glutamatergic input originating from multiple brain regions. However, the factors involved in the formation of glutamatergic inputs and how they contribute to bodily functions remain largely unknown. Here, we show that during the development of glutamatergic inputs, POMC neurons exhibit enriched expression of the Efnb1 (EphrinB1) and Efnb2 (EphrinB2) genes, which are known to control excitatory synapse formation. In vivo loss of Efnb1 in POMC-expressing progenitors decreases the amount of glutamatergic inputs, associated with a reduced number of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-D-aspartate (NMDA) receptor subunits and excitability of these cells. We found that mice lacking Efnb1 in POMC-expressing progenitors display impaired glucose tolerance due to blunted vagus nerve activity and decreased insulin secretion. However, despite reduced excitatory inputs, mice lacking Efnb2 in POMC-expressing progenitors showed no deregulation of insulin secretion and only mild alterations in feeding behavior and gluconeogenesis. Collectively, our data demonstrate the role of ephrins in controlling excitatory input amount into POMC-expressing progenitors and show an isotype-specific role of ephrins on the regulation of glucose homeostasis and feeding.
Collapse
Affiliation(s)
- Manon Gervais
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Gwenaël Labouèbe
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Alexandre Picard
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Sophie Croizier
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
17
|
Quenneville S, Labouèbe G, Basco D, Metref S, Viollet B, Foretz M, Thorens B. Hypoglycemia-Sensing Neurons of the Ventromedial Hypothalamus Require AMPK-Induced Txn2 Expression but Are Dispensable for Physiological Counterregulation. Diabetes 2020; 69:2253-2266. [PMID: 32839348 PMCID: PMC7576557 DOI: 10.2337/db20-0577] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/18/2020] [Indexed: 12/23/2022]
Abstract
The ventromedial nucleus of the hypothalamus (VMN) is involved in the counterregulatory response to hypoglycemia. VMN neurons activated by hypoglycemia (glucose-inhibited [GI] neurons) have been assumed to play a critical although untested role in this response. Here, we show that expression of a dominant negative form of AMPK or inactivation of AMPK α1 and α2 subunit genes in Sf1 neurons of the VMN selectively suppressed GI neuron activity. We found that Txn2, encoding a mitochondrial redox enzyme, was strongly downregulated in the absence of AMPK activity and that reexpression of Txn2 in Sf1 neurons restored GI neuron activity. In cell lines, Txn2 was required to limit glucopenia-induced reactive oxygen species production. In physiological studies, absence of GI neuron activity after AMPK suppression in the VMN had no impact on the counterregulatory hormone response to hypoglycemia or on feeding. Thus, AMPK is required for GI neuron activity by controlling the expression of the antioxidant enzyme Txn2. However, the glucose-sensing capacity of VMN GI neurons is not required for the normal counterregulatory response to hypoglycemia. Instead, it may represent a fail-safe system in case of impaired hypoglycemia sensing by peripherally located glucose detection systems that are connected to the VMN.
Collapse
Affiliation(s)
- Simon Quenneville
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Gwenaël Labouèbe
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Davide Basco
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Salima Metref
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Benoit Viollet
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | - Marc Foretz
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
18
|
Faber CL, Deem JD, Campos CA, Taborsky GJ, Morton GJ. CNS control of the endocrine pancreas. Diabetologia 2020; 63:2086-2094. [PMID: 32894319 PMCID: PMC7983553 DOI: 10.1007/s00125-020-05204-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/07/2020] [Indexed: 12/30/2022]
Abstract
Increasing evidence suggests that, although pancreatic islets can function autonomously to detect and respond to changes in the circulating glucose level, the brain cooperates with the islet to maintain glycaemic control. Here, we review the role of the central and autonomic nervous systems in the control of the endocrine pancreas, including mechanisms whereby the brain senses circulating blood glucose levels. We also examine whether dysfunction in these systems might contribute to complications of type 1 diabetes and the pathogenesis of type 2 diabetes. Graphical abstract.
Collapse
Affiliation(s)
- Chelsea L Faber
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, 750 Republican St, Box 358062, Seattle, WA, 98109, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer D Deem
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, 750 Republican St, Box 358062, Seattle, WA, 98109, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Carlos A Campos
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, 750 Republican St, Box 358062, Seattle, WA, 98109, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Gerald J Taborsky
- Department of Medicine, University of Washington, Seattle, WA, USA
- Veterans Affairs Puget Sound Health Care System, Department of Veterans Affairs Medical Center, Seattle, WA, USA
| | - Gregory J Morton
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, 750 Republican St, Box 358062, Seattle, WA, 98109, USA.
- Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
19
|
Sharari S, Abou-Alloul M, Hussain K, Ahmad Khan F. Fanconi-Bickel Syndrome: A Review of the Mechanisms That Lead to Dysglycaemia. Int J Mol Sci 2020; 21:E6286. [PMID: 32877990 PMCID: PMC7504390 DOI: 10.3390/ijms21176286] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/30/2020] [Accepted: 08/02/2020] [Indexed: 12/13/2022] Open
Abstract
Accumulation of glycogen in the kidney and liver is the main feature of Fanconi-Bickel Syndrome (FBS), a rare disorder of carbohydrate metabolism inherited in an autosomal recessive manner due to SLC2A2 gene mutations. Missense, nonsense, frame-shift (fs), in-frame indels, splice site, and compound heterozygous variants have all been identified in SLC2A2 gene of FBS cases. Approximately 144 FBS cases with 70 different SLC2A2 gene variants have been reported so far. SLC2A2 encodes for glucose transporter 2 (GLUT2) a low affinity facilitative transporter of glucose mainly expressed in tissues playing important roles in glucose homeostasis, such as renal tubular cells, enterocytes, pancreatic β-cells, hepatocytes and discrete regions of the brain. Dysfunctional mutations and decreased GLUT2 expression leads to dysglycaemia (fasting hypoglycemia, postprandial hyperglycemia, glucose intolerance, and rarely diabetes mellitus), hepatomegaly, galactose intolerance, rickets, and poor growth. The molecular mechanisms of dysglycaemia in FBS are still not clearly understood. In this review, we discuss the physiological roles of GLUT2 and the pathophysiology of mutants, highlight all of the previously reported SLC2A2 mutations associated with dysglycaemia, and review the potential molecular mechanisms leading to dysglycaemia and diabetes mellitus in FBS patients.
Collapse
Affiliation(s)
- Sanaa Sharari
- Division of Biological and Biomedical Sciences, College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Education City, Doha, Qatar;
- Department of Pediatric Medicine, Division of Endocrinology, Sidra Medicine, Doha, Qatar;
| | - Mohamad Abou-Alloul
- Department of Pediatric Medicine, Saida Governmental University Hospital, Beirut Arab University, Beirut 115020, Lebanon;
| | - Khalid Hussain
- Department of Pediatric Medicine, Division of Endocrinology, Sidra Medicine, Doha, Qatar;
| | - Faiyaz Ahmad Khan
- Department of Pediatric Medicine, Division of Endocrinology, Sidra Medicine, Doha, Qatar;
| |
Collapse
|
20
|
Rebelos E, Mari A, Bucci M, Honka M, Hannukainen JC, Virtanen KA, Hirvonen J, Nummenmaa L, Heni M, Iozzo P, Ferrannini E, Nuutila P. Brain substrate metabolism and ß-cell function in humans: A positron emission tomography study. Endocrinol Diabetes Metab 2020; 3:e00136. [PMID: 32704559 PMCID: PMC7375082 DOI: 10.1002/edm2.136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 03/06/2020] [Accepted: 03/28/2020] [Indexed: 12/16/2022] Open
Abstract
AIMS Recent clinical studies have shown enhanced brain glucose uptake during clamp and brain fatty acid uptake in insulin-resistant individuals. Preclinical studies suggest that the brain may be involved in the control of insulin secretion. The aim of this study was to investigate whether brain metabolism assessed as brain glucose and fatty acid uptake is associated with the parameters of β-cell function in humans. MATERIALS AND METHODS We analysed cross-sectional data of 120 subjects across a wide range of BMI and insulin sensitivity. Brain glucose uptake (BGU) was measured during euglycaemic-hyperinsulinaemic clamp (n = 67) and/or during fasting (n = 45) using [18F]-fluorodeoxyglucose (FDG) positron emission tomography (PET). In another group of subjects (n = 34), brain fatty acid uptake was measured using [18F]-fluoro-6-thia-heptadecanoic acid (FTHA) PET during fasting. The parameters of β-cell function were derived from OGTT modelling. Statistical analysis was performed with whole-brain voxel-based statistical parametric mapping. RESULTS In non-diabetics, BGU during euglycaemic hyperinsulinaemic clamp correlated positively with basal insulin secretion rate (r = 0.51, P = .0008) and total insulin output (r = 0.51, P = .0008), whereas no correlation was found in type 2 diabetics. BGU during clamp correlated positively with potentiation in non-diabetics (r = 0.33, P = .02) and negatively in type 2 diabetics (r = -0.61, P = .02). The associations in non-diabetics were not explained with whole-body insulin sensitivity or BMI. No correlations were found between baseline (fasting) BGU and basal insulin secretion rate, whereas baseline brain fatty acid uptake correlated directly with basal insulin secretion rate (r = 0.39, P = .02) and inversely with potentiation (r = -0.36, P = .04). CONCLUSIONS Our study provides coherent, though correlative, evidence that, in humans, the brain may be involved in the control of insulin secretion independently of insulin sensitivity.
Collapse
Affiliation(s)
| | - Andrea Mari
- Institute of NeuroscienceNational Research CouncilPaduaItaly
| | - Marco Bucci
- Turku PET CentreUniversity of TurkuTurkuFinland
| | | | | | - Kirsi A. Virtanen
- Turku PET CentreUniversity of TurkuTurkuFinland
- Clinical NutritionInstitute of Public Health and Clinical NutritionUniversity of Eastern Finland (UEF)KuopioFinland
| | - Jussi Hirvonen
- Department of RadiologyTurku University Hospital and University of TurkuTurkuFinland
| | - Lauri Nummenmaa
- Turku PET CentreUniversity of TurkuTurkuFinland
- Department of PsychologyUniversity of TurkuTurkuFinland
| | - Martin Heni
- Department of Internal MedicineDivision of EndocrinologyDiabetology, Angiology, Nephrology and Clinical ChemistryEberhard Karls University TuebingenTuebingenGermany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz CenterMunich at the University of TuebingenTuebingenGermany
- German Center for Diabetes Research (DZD e.V.)NeuherbergGermany
| | - Patricia Iozzo
- Turku PET CentreUniversity of TurkuTurkuFinland
- Institute of Clinical PhysiologyNational Research Council (CNR)PisaItaly
| | - Ele Ferrannini
- Institute of Clinical PhysiologyNational Research Council (CNR)PisaItaly
| | - Pirjo Nuutila
- Turku PET CentreUniversity of TurkuTurkuFinland
- Department of EndocrinologyTurku University HospitalTurkuFinland
| |
Collapse
|
21
|
Dumayne C, Tarussio D, Sanchez-Archidona AR, Picard A, Basco D, Berney XP, Ibberson M, Thorens B. Klf6 protects β-cells against insulin resistance-induced dedifferentiation. Mol Metab 2020; 35:100958. [PMID: 32244185 PMCID: PMC7093812 DOI: 10.1016/j.molmet.2020.02.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/30/2020] [Accepted: 02/02/2020] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES In the pathogenesis of type 2 diabetes, development of insulin resistance triggers an increase in pancreatic β-cell insulin secretion capacity and β-cell number. Failure of this compensatory mechanism is caused by a dedifferentiation of β-cells, which leads to insufficient insulin secretion and diabetic hyperglycemia. The β-cell factors that normally protect against dedifferentiation remain poorly defined. Here, through a systems biology approach, we identify the transcription factor Klf6 as a regulator of β-cell adaptation to metabolic stress. METHODS We used a β-cell specific Klf6 knockout mouse model to investigate whether Klf6 may be a potential regulator of β-cell adaptation to a metabolic stress. RESULTS We show that inactivation of Klf6 in β-cells blunts their proliferation induced by the insulin resistance of pregnancy, high-fat high-sucrose feeding, and insulin receptor antagonism. Transcriptomic analysis showed that Klf6 controls the expression of β-cell proliferation genes and, in the presence of insulin resistance, it prevents the down-expression of genes controlling mature β-cell identity and the induction of disallowed genes that impair insulin secretion. Its expression also limits the transdifferentiation of β-cells into α-cells. CONCLUSION Our study identifies a new transcription factor that protects β-cells against dedifferentiation, and which may be targeted to prevent diabetes development.
Collapse
Affiliation(s)
- Christopher Dumayne
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - David Tarussio
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Ana Rodriguez Sanchez-Archidona
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; Vital-IT, Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland.
| | - Alexandre Picard
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Davide Basco
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Xavier Pascal Berney
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Mark Ibberson
- Vital-IT, Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland.
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
22
|
Iozzo P, Guzzardi MA. Imaging of brain glucose uptake by PET in obesity and cognitive dysfunction: life-course perspective. Endocr Connect 2019; 8:R169-R183. [PMID: 31590145 PMCID: PMC6865363 DOI: 10.1530/ec-19-0348] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 10/07/2019] [Indexed: 12/17/2022]
Abstract
The prevalence of obesity has reached epidemic proportions and keeps growing. Obesity seems implicated in the pathogenesis of cognitive dysfunction, Alzheimer's disease and dementia, and vice versa. Growing scientific efforts are being devoted to the identification of central mechanisms underlying the frequent association between obesity and cognitive dysfunction. Glucose brain handling undergoes dynamic changes during the life-course, suggesting that its alterations might precede and contribute to degenerative changes or signaling abnormalities. Imaging of the glucose analog 18F-labeled fluorodeoxyglucose (18FDG) by positron emission tomography (PET) is the gold-standard for the assessment of cerebral glucose metabolism in vivo. This review summarizes the current literature addressing brain glucose uptake measured by PET imaging, and the effect of insulin on brain metabolism, trying to embrace a life-course vision in the identification of patterns that may explain (and contribute to) the frequent association between obesity and cognitive dysfunction. The current evidence supports that brain hypermetabolism and brain insulin resistance occur in selected high-risk conditions as a transient phenomenon, eventually evolving toward normal or low values during life or disease progression. Associative studies suggest that brain hypermetabolism predicts low BDNF levels, hepatic and whole body insulin resistance, food desire and an unfavorable balance between anticipated reward from food and cognitive inhibitory control. Emerging mechanistic links involve the microbiota and the metabolome, which correlate with brain metabolism and cognition, deserving attention as potential future prevention targets.
Collapse
Affiliation(s)
- Patricia Iozzo
- Institute of Clinical Physiology, National Research Council (CNR), Pisa, Italy
- Correspondence should be addressed to P Iozzo:
| | | |
Collapse
|
23
|
Lei H, Preitner F, Labouèbe G, Gruetter R, Thorens B. Glucose transporter 2 mediates the hypoglycemia-induced increase in cerebral blood flow. J Cereb Blood Flow Metab 2019; 39:1725-1736. [PMID: 29561214 PMCID: PMC6727137 DOI: 10.1177/0271678x18766743] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Glucose transporter 2 (Glut2)-positive cells are sparsely distributed in brain and play an important role in the stimulation of glucagon secretion in response to hypoglycemia. We aimed to determine if Glut2-positive cells can influence another response to hypoglycemia, i.e. increased cerebral blood flow (CBF). CBF of adult male mice devoid of Glut2, either globally (ripglut1:glut2-/-) or in the nervous system only (NG2KO), and their respective controls were studied under basal glycemia and insulin-induced hypoglycemia using quantitative perfusion magnetic resonance imaging at 9.4 T. The effect on CBF of optogenetic activation of hypoglycemia responsive Glut2-positive neurons of the paraventricular thalamic area was measured in mice expressing channelrhodopsin2 under the control of the Glut2 promoter. We found that in both ripglut1:glut2-/- mice and NG2KO mice, CBF in basal conditions was higher than in their respective controls and not further activated by hypoglycemia, as measured in the hippocampus, hypothalamus and whole brain. Conversely, optogenetic activation of Glut2-positive cells in the paraventricular thalamic nucleus induced a local increase in CBF similar to that induced by hypoglycemia. Thus, Glut2 expression in the nervous system is required for the control of CBF in response to changes in blood glucose concentrations.
Collapse
Affiliation(s)
- Hongxia Lei
- 1 AIT, Center for Biomedical Imaging (CIBM-AIT), Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,2 Department of Radiology, University of Geneva, Geneva, Switzerland
| | - Frédéric Preitner
- 3 Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.,4 Mouse Metabolic Evaluation Facility, Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Gwenaël Labouèbe
- 3 Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Rolf Gruetter
- 1 AIT, Center for Biomedical Imaging (CIBM-AIT), Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,2 Department of Radiology, University of Geneva, Geneva, Switzerland.,5 Department of Radiology, University of Lausanne, Lausanne, Switzerland
| | - Bernard Thorens
- 3 Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
24
|
Moullé VS, Tremblay C, Castell AL, Vivot K, Ethier M, Fergusson G, Alquier T, Ghislain J, Poitout V. The autonomic nervous system regulates pancreatic β-cell proliferation in adult male rats. Am J Physiol Endocrinol Metab 2019; 317:E234-E243. [PMID: 31013146 PMCID: PMC6732465 DOI: 10.1152/ajpendo.00385.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The pancreatic β-cell responds to changes in the nutrient environment to maintain glucose homeostasis by adapting its function and mass. Nutrients can act directly on the β-cell and also indirectly through the brain via autonomic nerves innervating islets. Despite the importance of the brain-islet axis in insulin secretion, relatively little is known regarding its involvement in β-cell proliferation. We previously demonstrated that prolonged infusions of nutrients in rats provoke a dramatic increase in β-cell proliferation in part because of the direct action of nutrients. Here, we addressed the contribution of the autonomic nervous system. In isolated islets, muscarinic stimulation increased, whereas adrenergic stimulation decreased, glucose-induced β-cell proliferation. Blocking α-adrenergic receptors reversed the effect of epinephrine on glucose + nonesterified fatty acids (NEFA)-induced β-cell proliferation, whereas activation of β-adrenergic receptors was without effect. Infusion of glucose + NEFA toward the brain stimulated β-cell proliferation, and this effect was abrogated following celiac vagotomy. The increase in β-cell proliferation following peripheral infusions of glucose + NEFA was not inhibited by vagotomy or atropine treatment but was blocked by coinfusion of epinephrine. We conclude that β-cell proliferation is stimulated by parasympathetic and inhibited by sympathetic signals. Whereas glucose + NEFA in the brain stimulates β-cell proliferation through the vagus nerve, β-cell proliferation in response to systemic nutrient excess does not involve parasympathetic signals but may be associated with decreased sympathetic tone.
Collapse
Affiliation(s)
- Valentine S Moullé
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
| | - Caroline Tremblay
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
| | - Anne-Laure Castell
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
| | - Kevin Vivot
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
| | - Mélanie Ethier
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
| | - Grace Fergusson
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
| | - Thierry Alquier
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
- Department of Medicine, University of Montreal , Quebec , Canada
| | - Julien Ghislain
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
- Department of Medicine, University of Montreal , Quebec , Canada
| |
Collapse
|
25
|
Abstract
Glucose-sensitive neurons have long been implicated in glucose homeostasis, but how glucose-sensing information is used by the brain in this process remains uncertain. Here, we propose a model in which (1) information relevant to the circulating glucose level is essential to the proper function of this regulatory system, (2) this input is provided by neurons located outside the blood-brain barrier (BBB) (since neurons situated behind the BBB are exposed to glucose in brain interstitial fluid, rather than that in the circulation), and (3) while the efferent limb of this system is comprised of neurons situated behind the BBB, many of these neurons are also glucose sensitive. Precedent for such an organizational scheme is found in the thermoregulatory system, which we draw upon in this framework for understanding the role played by brain glucose sensing in glucose homeostasis.
Collapse
Affiliation(s)
- Marie Aare Bentsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Blegdamsvej 3B, Building 7 (Maersk Tower), Copenhagen N 2200, Denmark; University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington at South Lake Union, 750 Republican St, F704, Box 358062, Seattle, WA 98109, USA
| | - Zaman Mirzadeh
- Department of Neurological Surgery, Barrow Neurological Institute, 350 West Thomas Road, Phoenix, AZ 85013, USA
| | - Michael W Schwartz
- University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington at South Lake Union, 750 Republican St, F704, Box 358062, Seattle, WA 98109, USA.
| |
Collapse
|
26
|
Landsman L. Pancreatic Pericytes in Glucose Homeostasis and Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1122:27-40. [DOI: 10.1007/978-3-030-11093-2_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
27
|
Idelevich A, Baron R. Brain to bone: What is the contribution of the brain to skeletal homeostasis? Bone 2018; 115:31-42. [PMID: 29777919 PMCID: PMC6110971 DOI: 10.1016/j.bone.2018.05.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022]
Abstract
The brain, which governs most, if not all, physiological functions in the body, from the complexities of cognition, learning and memory, to the regulation of basal body temperature, heart rate and breathing, has long been known to affect skeletal health. In particular, the hypothalamus - located at the base of the brain in close proximity to the medial eminence, where the blood-brain-barrier is not as tight as in other regions of the brain but rather "leaky", due to fenestrated capillaries - is exposed to a variety of circulating body cues, such as nutrients (glucose, fatty acids, amino acids), and hormones (insulin, glucagon, leptin, adiponectin) [1-3].Information collected from the body via these peripheral cues is integrated by hypothalamic sensing neurons and glial cells [4-7], which express receptors for these nutrients and hormones, transforming these cues into physiological outputs. Interestingly, many of the same molecules, including leptin, adiponectin and insulin, regulate both energy and skeletal homeostasis. Moreover, they act on a common set of hypothalamic nuclei and their residing neurons, activating endocrine and neuronal systems, which ultimately fine-tune the body to new physiological states. This review will focus exclusively on the brain-to-bone pathway, highlighting the most important anatomical sites within the brain, which are known to affect bone, but not covering the input pathways and molecules informing the brain of the energy and bone metabolic status, covered elsewhere [8-10]. The discussion in each section will present side by side the metabolic and bone-related functions of hypothalamic nuclei, in an attempt to answer some of the long-standing questions of whether energy is affected by bone remodeling and homeostasis and vice versa.
Collapse
Affiliation(s)
- Anna Idelevich
- Department of Medicine, Harvard Medical School and Endocrine Unit MGH, Division of Bone and Mineral Metabolism, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Roland Baron
- Department of Medicine, Harvard Medical School and Endocrine Unit MGH, Division of Bone and Mineral Metabolism, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA.
| |
Collapse
|
28
|
Ma Y, Ratnasabapathy R, Izzi-Engbeaya C, Nguyen-Tu MS, Richardson E, Hussain S, De Backer I, Holton C, Norton M, Carrat G, Schwappach B, Rutter GA, Dhillo WS, Gardiner J. Hypothalamic arcuate nucleus glucokinase regulates insulin secretion and glucose homeostasis. Diabetes Obes Metab 2018; 20:2246-2254. [PMID: 29748994 PMCID: PMC6099255 DOI: 10.1111/dom.13359] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 04/30/2018] [Accepted: 05/09/2018] [Indexed: 01/08/2023]
Abstract
AIMS To investigate the role of arcuate glucokinase (GK) in the regulation of glucose homeostasis. MATERIALS AND METHODS A recombinant adeno-associated virus expressing either GK or an antisense GK construct was used to alter GK activity specifically in the hypothalamic arcuate nucleus (arc). GK activity in this nucleus was also increased by stereotactic injection of the GK activator, compound A. The effect of altered arc GK activity on glucose homeostasis was subsequently investigated using glucose and insulin tolerance tests. RESULTS Increased GK activity specifically within the arc increased insulin secretion and improved glucose tolerance in rats during oral glucose tolerance tests. Decreased GK activity in this nucleus reduced insulin secretion and increased glucose levels during the same tests. Insulin sensitivity was not affected in either case. The effect of arc GK was maintained in a model of type 2 diabetes. CONCLUSIONS These results demonstrate a role for arc GK in systemic glucose homeostasis.
Collapse
Affiliation(s)
- Yue Ma
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Risheka Ratnasabapathy
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Chioma Izzi-Engbeaya
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Marie-Sophie Nguyen-Tu
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Errol Richardson
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Sufyan Hussain
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Ivan De Backer
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Christopher Holton
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Mariana Norton
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Gaelle Carrat
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Blanche Schwappach
- Department of Molecular Biology, Centre for Biochemistry and Molecular Cell Biology, Heart Research Centre Göttingen, University Medicine Göttingen, Göttingen, Germany
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - James Gardiner
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| |
Collapse
|
29
|
López-Gambero AJ, Martínez F, Salazar K, Cifuentes M, Nualart F. Brain Glucose-Sensing Mechanism and Energy Homeostasis. Mol Neurobiol 2018; 56:769-796. [PMID: 29796992 DOI: 10.1007/s12035-018-1099-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/25/2018] [Indexed: 01/02/2023]
Abstract
The metabolic and energy state of the organism depends largely on the availability of substrates, such as glucose for ATP production, necessary for maintaining physiological functions. Deregulation in glucose levels leads to the appearance of pathological signs that result in failures in the cardiovascular system and various diseases, such as diabetes, obesity, nephropathy, and neuropathy. Particularly, the brain relies on glucose as fuel for the normal development of neuronal activity. Regions adjacent to the cerebral ventricles, such as the hypothalamus and brainstem, exercise central control in energy homeostasis. These centers house nuclei of neurons whose excitatory activity is sensitive to changes in glucose levels. Determining the different detection mechanisms, the phenotype of neurosecretion, and neural connections involving glucose-sensitive neurons is essential to understanding the response to hypoglycemia through modulation of food intake, thermogenesis, and activation of sympathetic and parasympathetic branches, inducing glucagon and epinephrine secretion and other hypothalamic-pituitary axis-dependent counterregulatory hormones, such as glucocorticoids and growth hormone. The aim of this review focuses on integrating the current understanding of various glucose-sensing mechanisms described in the brain, thereby establishing a relationship between neuroanatomy and control of physiological processes involved in both metabolic and energy balance. This will advance the understanding of increasingly prevalent diseases in the modern world, especially diabetes, and emphasize patterns that regulate and stimulate intake, thermogenesis, and the overall synergistic effect of the neuroendocrine system.
Collapse
Affiliation(s)
- A J López-Gambero
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile.,Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, Málaga, Spain
| | - F Martínez
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - K Salazar
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - M Cifuentes
- Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, Málaga, Spain.
| | - F Nualart
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile. .,Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepción, Chile.
| |
Collapse
|
30
|
Barahona MJ, Llanos P, Recabal A, Escobar-Acuña K, Elizondo-Vega R, Salgado M, Ordenes P, Uribe E, Sepúlveda FJ, Araneda RC, García-Robles MA. Glial hypothalamic inhibition of GLUT2 expression alters satiety, impacting eating behavior. Glia 2017; 66:592-605. [PMID: 29178321 PMCID: PMC5814884 DOI: 10.1002/glia.23267] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 10/31/2017] [Accepted: 11/07/2017] [Indexed: 12/22/2022]
Abstract
Glucose is a key modulator of feeding behavior. By acting in peripheral tissues and in the central nervous system, it directly controls the secretion of hormones and neuropeptides and modulates the activity of the autonomic nervous system. GLUT2 is required for several glucoregulatory responses in the brain, including feeding behavior, and is localized in the hypothalamus and brainstem, which are the main centers that control this behavior. In the hypothalamus, GLUT2 has been detected in glial cells, known as tanycytes, which line the basal walls of the third ventricle (3V). This study aimed to clarify the role of GLUT2 expression in tanycytes in feeding behavior using 3V injections of an adenovirus encoding a shRNA against GLUT2 and the reporter EGFP (Ad‐shGLUT2). Efficient in vivo GLUT2 knockdown in rat hypothalamic tissue was demonstrated by qPCR and Western blot analyses. Specificity of cell transduction in the hypothalamus and brainstem was evaluated by EGFP‐fluorescence and immunohistochemistry, which showed EGFP expression specifically in ependymal cells, including tanycytes. The altered mRNA levels of both orexigenic and anorexigenic neuropeptides suggested a loss of response to increased glucose in the 3V. Feeding behavior analysis in the fasting‐feeding transition revealed that GLUT2‐knockdown rats had increased food intake and body weight, suggesting an inhibitory effect on satiety. Taken together, suppression of GLUT2 expression in tanycytes disrupted the hypothalamic glucosensing mechanism, which altered the feeding behavior.
Collapse
Affiliation(s)
- María J Barahona
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Paula Llanos
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Antonia Recabal
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Kathleen Escobar-Acuña
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Roberto Elizondo-Vega
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.,Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Magdiel Salgado
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Patricio Ordenes
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Elena Uribe
- Departamento de Bioquímica y Biología Molecular, Universidad de Concepción, Chile
| | - Fernando J Sepúlveda
- Departamento de Bioquímica y Biología Molecular, Universidad de Concepción, Chile.,Departamento de Ciencias Biológica Universidad Andrés Bello, Concepción, Chile
| | - Ricardo C Araneda
- Department of Biology, University of Maryland, College Park, Maryland
| | - María A García-Robles
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
31
|
Hypothalamic ΔFosB prevents age-related metabolic decline and functions via SNS. Aging (Albany NY) 2017; 9:353-369. [PMID: 28121620 PMCID: PMC5361668 DOI: 10.18632/aging.101157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 01/15/2017] [Indexed: 12/31/2022]
Abstract
The ventral hypothalamus (VHT) integrates several physiological cues to maintain glucose homeostasis and energy balance. Aging is associated with increased glucose intolerance but the underlying mechanisms responsible for age-related metabolic decline, including neuronal signaling in the VHT, remain elusive. We have shown that mice with VHT-targeted overexpression of ∆FosB, a splice variant of the AP1 transcription factor FosB, exhibit increased energy expenditure, leading to decreased adiposity. Here, we show that VHT-targeted overexpression of ∆FosB also improves glucose tolerance, increases insulin sensitivity in target organs and thereby suppresses insulin secretion. These effects are also observed by the overexpression of dominant negative JunD, demonstrating that they occur via AP1 antagonism within the VHT. Furthermore, the improved glucose tolerance and insulin sensitivity persisted in aged animals overexpressing ∆FosB in the VHT. These beneficial effects on glucose metabolism were abolished by peripheral sympathectomy and α-adrenergic, but not β-adrenergic, blockade. Taken together, our results show that antagonizing AP1 transcription activity in the VHT leads to a marked improvement in whole body glucose homeostasis via activation of the SNS, conferring protection against age-related impairment in glucose metabolism. These findings may open novel avenues for therapeutic intervention in diabetes and age-related glucose intolerance.
Collapse
|
32
|
Inhibition of central de novo ceramide synthesis restores insulin signaling in hypothalamus and enhances β-cell function of obese Zucker rats. Mol Metab 2017; 8:23-36. [PMID: 29233519 PMCID: PMC5985020 DOI: 10.1016/j.molmet.2017.10.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/17/2017] [Accepted: 10/27/2017] [Indexed: 12/29/2022] Open
Abstract
Objectives Hypothalamic lipotoxicity has been shown to induce central insulin resistance and dysregulation of glucose homeostasis; nevertheless, elucidation of the regulatory mechanisms remains incomplete. Here, we aimed to determine the role of de novo ceramide synthesis in hypothalamus on the onset of central insulin resistance and the dysregulation of glucose homeostasis induced by obesity. Methods Hypothalamic GT1-7 neuronal cells were treated with palmitate. De novo ceramide synthesis was inhibited either by pharmacological (myriocin) or molecular (si-Serine Palmitoyl Transferase 2, siSPT2) approaches. Obese Zucker rats (OZR) were intracerebroventricularly infused with myriocin to inhibit de novo ceramide synthesis. Insulin resistance was determined by quantification of Akt phosphorylation. Ceramide levels were quantified either by a radioactive kinase assay or by mass spectrometry analysis. Glucose homeostasis were evaluated in myriocin-treated OZR. Basal and glucose-stimulated parasympathetic tonus was recorded in OZR. Insulin secretion from islets and β-cell mass was also determined. Results We show that palmitate impaired insulin signaling and increased ceramide levels in hypothalamic neuronal GT1-7 cells. In addition, the use of deuterated palmitic acid demonstrated that palmitate activated several enzymes of the de novo ceramide synthesis pathway in hypothalamic cells. Importantly, myriocin and siSPT2 treatment restored insulin signaling in palmitate-treated GT1-7 cells. Protein kinase C (PKC) inhibitor or a dominant-negative PKCζ also counteracted palmitate-induced insulin resistance. Interestingly, attenuating the increase in levels of hypothalamic ceramides with intracerebroventricular infusion of myriocin in OZR improved their hypothalamic insulin-sensitivity. Importantly, central myriocin treatment partially restored glucose tolerance in OZR. This latter effect is related to the restoration of glucose-stimulated insulin secretion and an increase in β-cell mass of OZR. Electrophysiological recordings also showed an improvement of glucose-stimulated parasympathetic nerve activity in OZR centrally treated with myriocin. Conclusion Our results highlight a key role of hypothalamic de novo ceramide synthesis in central insulin resistance installation and glucose homeostasis dysregulation associated with obesity. de novo ceramide synthesis induces hypothalamic insulin resistance through PKCζ. Hypothalamic ceramides induce glucose homeostasis dysregulation seen with obesity. Hypothalamic ceramides mediate inhibition of insulin secretion induced by obesity. Hypothalamic ceramides decreases β cell mass in obese rats. Hypothalamic ceramides decreases parasympathetic tonus.
Collapse
|
33
|
Epshtein A, Rachi E, Sakhneny L, Mizrachi S, Baer D, Landsman L. Neonatal pancreatic pericytes support β-cell proliferation. Mol Metab 2017; 6:1330-1338. [PMID: 29031732 PMCID: PMC5641631 DOI: 10.1016/j.molmet.2017.07.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE The maintenance and expansion of β-cell mass rely on their proliferation, which reaches its peak in the neonatal stage. β-cell proliferation was found to rely on cells of the islet microenvironment. We hypothesized that pericytes, which are components of the islet vasculature, support neonatal β-cell proliferation. METHODS To test our hypothesis, we combined in vivo and in vitro approaches. Briefly, we used a Diphtheria toxin-based transgenic mouse system to specifically deplete neonatal pancreatic pericytes in vivo. We further cultured neonatal pericytes isolated from the neonatal pancreas and combined the use of a β-cell line and primary cultured mouse β-cells. RESULTS Our findings indicate that neonatal pancreatic pericytes are required and sufficient for β-cell proliferation. We observed impaired proliferation of neonatal β-cells upon in vivo depletion of pancreatic pericytes. Furthermore, exposure to pericyte-conditioned medium stimulated proliferation in cultured β-cells. CONCLUSIONS This study introduces pancreatic pericytes as regulators of neonatal β-cell proliferation. In addition to advancing current understanding of the physiological β-cell replication process, these findings could facilitate the development of protocols aimed at expending these cells as a potential cure for diabetes.
Collapse
Affiliation(s)
- Alona Epshtein
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eleonor Rachi
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lina Sakhneny
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shani Mizrachi
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Daria Baer
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Limor Landsman
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
34
|
Deem JD, Muta K, Scarlett JM, Morton GJ, Schwartz MW. How Should We Think About the Role of the Brain in Glucose Homeostasis and Diabetes? Diabetes 2017; 66:1758-1765. [PMID: 28603139 PMCID: PMC5482090 DOI: 10.2337/dbi16-0067] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 02/25/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Jennifer D Deem
- Department of Medicine, University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Kenjiro Muta
- Department of Medicine, University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Jarrad M Scarlett
- Department of Medicine, University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Gregory J Morton
- Department of Medicine, University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Michael W Schwartz
- Department of Medicine, University of Washington Diabetes Institute, University of Washington, Seattle, WA
| |
Collapse
|
35
|
Ramírez S, Gómez-Valadés AG, Schneeberger M, Varela L, Haddad-Tóvolli R, Altirriba J, Noguera E, Drougard A, Flores-Martínez Á, Imbernón M, Chivite I, Pozo M, Vidal-Itriago A, Garcia A, Cervantes S, Gasa R, Nogueiras R, Gama-Pérez P, Garcia-Roves PM, Cano DA, Knauf C, Servitja JM, Horvath TL, Gomis R, Zorzano A, Claret M. Mitochondrial Dynamics Mediated by Mitofusin 1 Is Required for POMC Neuron Glucose-Sensing and Insulin Release Control. Cell Metab 2017; 25:1390-1399.e6. [PMID: 28591639 DOI: 10.1016/j.cmet.2017.05.010] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 04/03/2017] [Accepted: 05/23/2017] [Indexed: 11/29/2022]
Abstract
Proopiomelanocortin (POMC) neurons are critical sensors of nutrient availability implicated in energy balance and glucose metabolism control. However, the precise mechanisms underlying nutrient sensing in POMC neurons remain incompletely understood. We show that mitochondrial dynamics mediated by Mitofusin 1 (MFN1) in POMC neurons couple nutrient sensing with systemic glucose metabolism. Mice lacking MFN1 in POMC neurons exhibited defective mitochondrial architecture remodeling and attenuated hypothalamic gene expression programs during the fast-to-fed transition. This loss of mitochondrial flexibility in POMC neurons bidirectionally altered glucose sensing, causing abnormal glucose homeostasis due to defective insulin secretion by pancreatic β cells. Fed mice lacking MFN1 in POMC neurons displayed enhanced hypothalamic mitochondrial oxygen flux and reactive oxygen species generation. Central delivery of antioxidants was able to normalize the phenotype. Collectively, our data posit MFN1-mediated mitochondrial dynamics in POMC neurons as an intrinsic nutrient-sensing mechanism and unveil an unrecognized link between this subset of neurons and insulin release.
Collapse
Affiliation(s)
- Sara Ramírez
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Alicia G Gómez-Valadés
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Marc Schneeberger
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Luis Varela
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Jordi Altirriba
- Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Eduard Noguera
- Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain
| | - Anne Drougard
- Institute of Research in Digestive Health (IRSD) - INSERM U1220, European Associated Laboratory "NeuroMicrobiota", University Paul Sabatier, 31024 Toulouse, France
| | - Álvaro Flores-Martínez
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Mónica Imbernón
- Instituto de Investigaciones Sanitarias (IDIS), CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Iñigo Chivite
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Macarena Pozo
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Andrés Vidal-Itriago
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Ainhoa Garcia
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain; Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Sara Cervantes
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Rosa Gasa
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain; Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Ruben Nogueiras
- Instituto de Investigaciones Sanitarias (IDIS), CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Pau Gama-Pérez
- Department of Physiological Sciences, University of Barcelona, 08907 Barcelona, Spain
| | - Pablo M Garcia-Roves
- Department of Physiological Sciences, University of Barcelona, 08907 Barcelona, Spain; Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - David A Cano
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Claude Knauf
- Institute of Research in Digestive Health (IRSD) - INSERM U1220, European Associated Laboratory "NeuroMicrobiota", University Paul Sabatier, 31024 Toulouse, France
| | - Joan-Marc Servitja
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain; Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Tamas L Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Anatomy and Hystology, University of Veterinary Medicine, Budapest 1078, Hungary
| | - Ramon Gomis
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain; Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Department of Endocrinology and Nutrition, Hospital Clínic. School of Medicine, University of Barcelona, 08036 Barcelona, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain.
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain.
| |
Collapse
|
36
|
Shirakawa J, De Jesus DF, Kulkarni RN. Exploring inter-organ crosstalk to uncover mechanisms that regulate β-cell function and mass. Eur J Clin Nutr 2017; 71:896-903. [PMID: 28294170 DOI: 10.1038/ejcn.2017.13] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 01/24/2017] [Indexed: 02/07/2023]
Abstract
Impaired β-cell function and insufficient β-cell mass compensation are twin pathogenic features that underlie type 2 diabetes (T2D). Current therapeutic strategies continue to evolve to improve treatment outcomes in different ethnic populations and include approaches to counter insulin resistance and improve β-cell function. Although the effects of insulin secretion on metabolic organs such as liver, skeletal muscle and adipose is directly relevant for improving glucose uptake and reduce hyperglycemia, the ability of pancreatic β-cells to crosstalk with multiple non-metabolic tissues is providing novel insights into potential opportunities for improving β-cell function and/or mass that could have beneficial effects in patients with diabetes. For example, the role of the gastrointestinal system in the regulation of β-cell biology is well recognized and has been exploited clinically to develop incretin-related antidiabetic agents. The microbiome and the immune system are emerging as important players in regulating β-cell function and mass. The rich innervation of islet cells indicates it is a prime organ for regulation by the nervous system. In this review, we discuss the potential implications of signals from these organ systems as well as those from bone, placenta, kidney, thyroid, endothelial cells, reproductive organs and adrenal and pituitary glands that can directly impact β-cell biology. An added layer of complexity is the limited data regarding the relative relevance of one or more of these systems in different ethnic populations. It is evident that better understanding of this paradigm would provide clues to enhance β-cell function and/or mass in vivo in the long-term goal of treating or curing patients with diabetes.
Collapse
Affiliation(s)
- J Shirakawa
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - D F De Jesus
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA.,Graduate Program in Areas of Basic and Applied Biology (GABBA), Abel Salazar Biomedical Sciences Institute, University of Porto, Porto, Portugal
| | - R N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
37
|
Intestinal PPARγ signalling is required for sympathetic nervous system activation in response to caloric restriction. Sci Rep 2016; 6:36937. [PMID: 27853235 PMCID: PMC5113069 DOI: 10.1038/srep36937] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 10/05/2016] [Indexed: 02/08/2023] Open
Abstract
Nuclear receptor PPARγ has been proven to affect metabolism in multiple tissues, and has received considerable attention for its involvement in colon cancer and inflammatory disease. However, its role in intestinal metabolism has been largely ignored. To investigate this potential aspect of PPARγ function, we submitted intestinal epithelium-specific PPARγ knockout mice (iePPARγKO) to a two-week period of 25% caloric restriction (CR), following which iePPARγKO mice retained more fat than their wild type littermates. In attempting to explain this discrepancy, we analysed the liver, skeletal muscle, intestinal lipid trafficking, and the microbiome, none of which appeared to contribute to the adiposity phenotype. Interestingly, under conditions of CR, iePPARγKO mice failed to activate their sympathetic nervous system (SNS) and increase CR-specific locomotor activity. These KO mice also manifested a defective control of their body temperature, which was overly reduced. Furthermore, the white adipose tissue of iePPARγKO CR mice showed lower levels of both hormone-sensitive lipase, and its phosphorylated form. This would result from impaired SNS signalling and possibly cause reduced lipolysis. We conclude that intestinal epithelium PPARγ plays an essential role in increasing SNS activity under CR conditions, thereby contributing to energy mobilization during metabolically stressful episodes.
Collapse
|
38
|
Ferrario CR, Labouèbe G, Liu S, Nieh EH, Routh VH, Xu S, O'Connor EC. Homeostasis Meets Motivation in the Battle to Control Food Intake. J Neurosci 2016; 36:11469-11481. [PMID: 27911750 PMCID: PMC5125214 DOI: 10.1523/jneurosci.2338-16.2016] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 08/30/2016] [Accepted: 09/05/2016] [Indexed: 01/09/2023] Open
Abstract
Signals of energy homeostasis interact closely with neural circuits of motivation to control food intake. An emerging hypothesis is that the transition to maladaptive feeding behavior seen in eating disorders or obesity may arise from dysregulation of these interactions. Focusing on key brain regions involved in the control of food intake (ventral tegmental area, striatum, hypothalamus, and thalamus), we describe how activity of specific cell types embedded within these regions can influence distinct components of motivated feeding behavior. We review how signals of energy homeostasis interact with these regions to influence motivated behavioral output and present evidence that experience-dependent neural adaptations in key feeding circuits may represent cellular correlates of impaired food intake control. Future research into mechanisms that restore the balance of control between signals of homeostasis and motivated feeding behavior may inspire new treatment options for eating disorders and obesity.
Collapse
Affiliation(s)
- Carrie R Ferrario
- University of Michigan Medical School, Department of Pharmacology, Ann Arbor, Michigan 48109-5632
| | - Gwenaël Labouèbe
- University of Lausanne, Center for Integrative Genomics, Lausanne, CH1015, Switzerland
| | - Shuai Liu
- University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Edward H Nieh
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | | | - Shengjin Xu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia 20147, and
| | - Eoin C O'Connor
- University of Geneva, Department of Basic Neuroscience, Geneva, CH1211, Switzerland
| |
Collapse
|
39
|
Picard A, Soyer J, Berney X, Tarussio D, Quenneville S, Jan M, Grouzmann E, Burdet F, Ibberson M, Thorens B. A Genetic Screen Identifies Hypothalamic Fgf15 as a Regulator of Glucagon Secretion. Cell Rep 2016; 17:1795-1806. [PMID: 27829151 PMCID: PMC5120348 DOI: 10.1016/j.celrep.2016.10.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 10/04/2016] [Accepted: 10/13/2016] [Indexed: 12/26/2022] Open
Abstract
The counterregulatory response to hypoglycemia, which restores normal blood glucose levels to ensure sufficient provision of glucose to the brain, is critical for survival. To discover underlying brain regulatory systems, we performed a genetic screen in recombinant inbred mice for quantitative trait loci (QTL) controlling glucagon secretion in response to neuroglucopenia. We identified a QTL on the distal part of chromosome 7 and combined this genetic information with transcriptomic analysis of hypothalami. This revealed Fgf15 as the strongest candidate to control the glucagon response. Fgf15 was expressed by neurons of the dorsomedial hypothalamus and the perifornical area. Intracerebroventricular injection of FGF19, the human ortholog of Fgf15, reduced activation by neuroglucopenia of dorsal vagal complex neurons, of the parasympathetic nerve, and lowered glucagon secretion. In contrast, silencing Fgf15 in the dorsomedial hypothalamus increased neuroglucopenia-induced glucagon secretion. These data identify hypothalamic Fgf15 as a regulator of glucagon secretion.
Collapse
Affiliation(s)
- Alexandre Picard
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Josselin Soyer
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Xavier Berney
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - David Tarussio
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Simon Quenneville
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Maxime Jan
- Vital-IT, Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Eric Grouzmann
- Service de Biomédicine, Laboratoire des Catécholamines et Peptides, Centre Hospitalier Universitaire Vaudois CHUV, 1011 Lausanne, Switzerland
| | - Frédéric Burdet
- Vital-IT, Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Mark Ibberson
- Vital-IT, Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
40
|
Steinbusch LKM, Picard A, Bonnet MS, Basco D, Labouèbe G, Thorens B. Sex-Specific Control of Fat Mass and Counterregulation by Hypothalamic Glucokinase. Diabetes 2016; 65:2920-31. [PMID: 27422385 DOI: 10.2337/db15-1514] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 07/07/2016] [Indexed: 11/13/2022]
Abstract
Glucokinase (Gck) is a critical regulator of glucose-induced insulin secretion by pancreatic β-cells. It has been suggested to also play an important role in glucose signaling in neurons of the ventromedial hypothalamic nucleus (VMN), a brain nucleus involved in the control of glucose homeostasis and feeding. To test the role of Gck in VMN glucose sensing and physiological regulation, we studied mice with genetic inactivation of the Gck gene in Sf1 neurons of the VMN (Sf1Gck(-/-) mice). Compared with control littermates, Sf1Gck(-/-) mice displayed increased white fat mass and adipocyte size, reduced lean mass, impaired hypoglycemia-induced glucagon secretion, and a lack of parasympathetic and sympathetic nerve activation by neuroglucopenia. However, these phenotypes were observed only in female mice. To determine whether Gck was required for glucose sensing by Sf1 neurons, we performed whole-cell patch clamp analysis of brain slices from control and Sf1Gck(-/-) mice. Absence of Gck expression did not prevent the glucose responsiveness of glucose-excited or glucose-inhibited Sf1 neurons in either sex. Thus Gck in the VMN plays a sex-specific role in the glucose-dependent control of autonomic nervous activity; this is, however, unrelated to the control of the firing activity of classical glucose-responsive neurons.
Collapse
Affiliation(s)
| | - Alexandre Picard
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Marion S Bonnet
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Davide Basco
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Gwenaël Labouèbe
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
41
|
Semplici F, Mondragon A, Macintyre B, Madeyski-Bengston K, Persson-Kry A, Barr S, Ramne A, Marley A, McGinty J, French P, Soedling H, Yokosuka R, Gaitan J, Lang J, Migrenne-Li S, Philippe E, Herrera PL, Magnan C, da Silva Xavier G, Rutter GA. Cell type-specific deletion in mice reveals roles for PAS kinase in insulin and glucagon production. Diabetologia 2016; 59:1938-47. [PMID: 27338626 PMCID: PMC4969360 DOI: 10.1007/s00125-016-4025-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/01/2016] [Indexed: 12/12/2022]
Abstract
AIMS/HYPOTHESIS Per-Arnt-Sim kinase (PASK) is a nutrient-regulated domain-containing protein kinase previously implicated in the control of insulin gene expression and glucagon secretion. Here, we explore the roles of PASK in the control of islet hormone release, by generating mice with selective deletion of the Pask gene in pancreatic beta or alpha cells. METHODS Floxed alleles of Pask were produced by homologous recombination and animals bred with mice bearing beta (Ins1 (Cre); PaskBKO) or alpha (Ppg (Cre) [also known as Gcg]; PaskAKO) cell-selective Cre recombinase alleles. Glucose homeostasis and hormone secretion in vivo and in vitro, gene expression and islet cell mass were measured using standard techniques. RESULTS Ins1 (Cre)-based recombination led to efficient beta cell-targeted deletion of Pask. Beta cell mass was reduced by 36.5% (p < 0.05) compared with controls in PaskBKO mice, as well as in global Pask-null mice (38%, p < 0.05). PaskBKO mice displayed normal body weight and fasting glycaemia, but slightly impaired glucose tolerance, and beta cell proliferation, after maintenance on a high-fat diet. Whilst glucose tolerance was unaffected in PaskAKO mice, glucose infusion rates were increased, and glucagon secretion tended to be lower, during hypoglycaemic clamps. Although alpha cell mass was increased (21.9%, p < 0.05), glucagon release at low glucose was impaired (p < 0.05) in PaskAKO islets. CONCLUSIONS/INTERPRETATION The findings demonstrate cell-autonomous roles for PASK in the control of pancreatic endocrine hormone secretion. Differences between the glycaemic phenotype of global vs cell type-specific null mice suggest important roles for tissue interactions in the control of glycaemia by PASK.
Collapse
Affiliation(s)
- Francesca Semplici
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, Imperial Centre for Translational and Experimental Medicine, Hammersmith Hospital, du Cane Road, London, W12 0NN, UK
| | - Angeles Mondragon
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, Imperial Centre for Translational and Experimental Medicine, Hammersmith Hospital, du Cane Road, London, W12 0NN, UK
| | - Benedict Macintyre
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, Imperial Centre for Translational and Experimental Medicine, Hammersmith Hospital, du Cane Road, London, W12 0NN, UK
| | - Katja Madeyski-Bengston
- AstraZeneca R&D, DECS, AstraZeneca R&D, Mölndal, Sweden
- AstraZeneca R&D, HC3020, AstraZeneca R&D, Mölndal, Sweden
| | - Anette Persson-Kry
- AstraZeneca R&D, DECS, AstraZeneca R&D, Mölndal, Sweden
- AstraZeneca R&D, HC3020, AstraZeneca R&D, Mölndal, Sweden
| | - Sara Barr
- AstraZeneca R&D, DECS, AstraZeneca R&D, Mölndal, Sweden
- AstraZeneca R&D, HC3020, AstraZeneca R&D, Mölndal, Sweden
| | - Anna Ramne
- AstraZeneca R&D, DECS, AstraZeneca R&D, Mölndal, Sweden
- AstraZeneca R&D, HC3020, AstraZeneca R&D, Mölndal, Sweden
| | | | - James McGinty
- Photonics Group, Department of Physics, Imperial College London, London, UK
| | - Paul French
- Photonics Group, Department of Physics, Imperial College London, London, UK
| | - Helen Soedling
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, Imperial Centre for Translational and Experimental Medicine, Hammersmith Hospital, du Cane Road, London, W12 0NN, UK
| | - Ryohsuke Yokosuka
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, Imperial Centre for Translational and Experimental Medicine, Hammersmith Hospital, du Cane Road, London, W12 0NN, UK
| | - Julien Gaitan
- Université de Bordeaux, Institut de Chimie et Biologie des Membranes et des Nano-objets, CNRS UMR 5248, Pessac, France
| | - Jochen Lang
- Université de Bordeaux, Institut de Chimie et Biologie des Membranes et des Nano-objets, CNRS UMR 5248, Pessac, France
| | - Stephanie Migrenne-Li
- Paris Diderot University, Unit of Functional and Adaptive Biology (BFA), CNRS UMR 8251, Paris, France
| | - Erwann Philippe
- Paris Diderot University, Unit of Functional and Adaptive Biology (BFA), CNRS UMR 8251, Paris, France
| | - Pedro L Herrera
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Christophe Magnan
- Paris Diderot University, Unit of Functional and Adaptive Biology (BFA), CNRS UMR 8251, Paris, France
| | - Gabriela da Silva Xavier
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, Imperial Centre for Translational and Experimental Medicine, Hammersmith Hospital, du Cane Road, London, W12 0NN, UK.
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, Imperial Centre for Translational and Experimental Medicine, Hammersmith Hospital, du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
42
|
Labouèbe G, Boutrel B, Tarussio D, Thorens B. Glucose-responsive neurons of the paraventricular thalamus control sucrose-seeking behavior. Nat Neurosci 2016; 19:999-1002. [PMID: 27322418 PMCID: PMC4964931 DOI: 10.1038/nn.4331] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 05/24/2016] [Indexed: 11/30/2022]
Abstract
Feeding behavior is governed by homeostatic needs and motivational drive to obtain palatable foods. Here, we identify a population of glutamatergic neurons in the paraventricular thalamus, which express the glucose transporter Glut2 (Scl2a2) and project to the nucleus accumbens. These neurons are activated by hypoglycemia and, in freely moving mice, their activation by optogenetics or Slc2a2 inactivation increases motivated sucrose but not saccharin-seeking behavior. These neurons may control sugar overconsumption in obesity and diabetes.
Collapse
Affiliation(s)
- Gwenaël Labouèbe
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Benjamin Boutrel
- Center for Psychiatric Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | - David Tarussio
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
43
|
Abstract
Glucose and hormone responsiveness of pancreatic β cells is acquired during postnatal maturation and is critical for appropriate insulin secretion. In a recent issue of Cell Metabolism, Yoshihara et al. (2016) report that estrogen-related receptor γ (ERRγ) promotes functional maturation of both mouse neonatal β cells and human iPSC-derived β-like cells.
Collapse
Affiliation(s)
- Jun Shirakawa
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA; Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
44
|
Triterpene derivative: A potential signaling pathway for the fern-9(11)-ene-2α,3β-diol on insulin secretion in pancreatic islet. Life Sci 2016; 154:58-65. [PMID: 27108785 DOI: 10.1016/j.lfs.2016.04.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 04/13/2016] [Accepted: 04/19/2016] [Indexed: 11/22/2022]
Abstract
AIM Triterpenes and their derivatives influence on carbohydrate metabolism. In vivo and in vitro treatment investigated the effect of the natural triterpene fern-9(11)-ene-2α,3β-diol (1), isolated from Croton heterodoxus, and a derivative triterpene (2) on glucose homeostasis. MAIN METHODS The antidiabetic effect of the crude extract from C. heterodoxus leaves, the natural triterpene (1) as well as the derivative triterpene (2) were assayed on glucose tolerance. The effect and the mechanism of action on in vivo treatment with triterpene 2 on glycaemia and insulin secretion were studied. In addition, in vitro studies investigated the mechanism of triterpene 2 on glucose uptake and calcium influx on insulin secretion in pancreatic islets. KEY FINDINGS The results show the extract slightly reduced the glycaemia when compared with hyperglycemic group. However, the presence of the substituent electron-withdrawing 4-nitrobenzoyl group in the A-ring of triterpene 2 powered the serum glucose lowering compared to triterpene 1. In addition, in vivo treatment with triterpene 2 significantly increased the insulin secretion induced by glucose and stimulated the glucose uptake and calcium influx in pancreatic islet. The effect of triterpene on calcium influx was completely inhibited by diazoxide, nifedipine and stearoylcarnitine treatment. SIGNIFICANCE The stimulatory effect of triterpene 2 on glucose uptake, calcium influx, regulation of potassium (K(+)-ATP) and calcium (L-VDCCs) channels activity as well as the pathway of PKC highlights the mechanism of action of the compound in pancreatic islets on insulin secretion and glucose homeostasis. In addition, this compound did not induce toxicity in this experimental condition.
Collapse
|
45
|
Modi H, Jacovetti C, Tarussio D, Metref S, Madsen OD, Zhang FP, Rantakari P, Poutanen M, Nef S, Gorman T, Regazzi R, Thorens B. Autocrine Action of IGF2 Regulates Adult β-Cell Mass and Function. Diabetes 2015; 64:4148-57. [PMID: 26384384 DOI: 10.2337/db14-1735] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 08/30/2015] [Indexed: 11/13/2022]
Abstract
Insulin-like growth factor 2 (IGF2), produced and secreted by adult β-cells, functions as an autocrine activator of the β-cell insulin-like growth factor 1 receptor signaling pathway. Whether this autocrine activity of IGF2 plays a physiological role in β-cell and whole-body physiology is not known. Here, we studied mice with β-cell-specific inactivation of Igf2 (βIGF2KO mice) and assessed β-cell mass and function in aging, pregnancy, and acute induction of insulin resistance. We showed that glucose-stimulated insulin secretion (GSIS) was markedly reduced in old female βIGF2KO mice; glucose tolerance was, however, normal because of increased insulin sensitivity. While on a high-fat diet, both male and female βIGF2KO mice displayed lower GSIS compared with control mice, but reduced β-cell mass was observed only in female βIGF2KO mice. During pregnancy, there was no increase in β-cell proliferation and mass in βIGF2KO mice. Finally, β-cell mass expansion in response to acute induction of insulin resistance was lower in βIGF2KO mice than in control mice. Thus, the autocrine action of IGF2 regulates adult β-cell mass and function to preserve in vivo GSIS in aging and to adapt β-cell mass in response to metabolic stress, pregnancy hormones, and acute induction of insulin resistance.
Collapse
Affiliation(s)
- Honey Modi
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Cecile Jacovetti
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - David Tarussio
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Salima Metref
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Ole D Madsen
- Hagedorn Research, Diabetes Biology, Novo Nordisk A/S, Måløv, Denmark
| | - Fu-Ping Zhang
- Department of Physiology and Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Pia Rantakari
- Department of Physiology and Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Matti Poutanen
- Department of Physiology and Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Tracy Gorman
- AstraZeneca, High-Content Biology, Discovery Sciences, Alderley Park, Macclesfield, Cheshire, U.K
| | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
46
|
Postnatal β-cell maturation is associated with islet-specific microRNA changes induced by nutrient shifts at weaning. Nat Commun 2015; 6:8084. [PMID: 26330140 PMCID: PMC4569696 DOI: 10.1038/ncomms9084] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 07/16/2015] [Indexed: 12/21/2022] Open
Abstract
Glucose-induced insulin secretion is an essential function of pancreatic β-cells that is partially lost in individuals affected by Type 2 diabetes. This unique property of β-cells is acquired through a poorly understood postnatal maturation process involving major modifications in gene expression programs. Here we show that β-cell maturation is associated with changes in microRNA expression induced by the nutritional transition that occurs at weaning. When mimicked in newborn islet cells, modifications in the level of specific microRNAs result in a switch in the expression of metabolic enzymes and cause the acquisition of glucose-induced insulin release. Our data suggest microRNAs have a central role in postnatal β-cell maturation and in the determination of adult functional β-cell mass. A better understanding of the events governing β-cell maturation may help understand why some individuals are predisposed to developing diabetes and could lead to new strategies for the treatment of this common metabolic disease. Pancreatic β-cells are less responsive to changes in glucose concentration in newborn than in adult rats. Here, the authors show that functional β-cell maturation is associated with changes in miRNA expression induced by nutritional shifts at the suckling-to-weaning transition.
Collapse
|
47
|
Steinbusch L, Labouèbe G, Thorens B. Brain glucose sensing in homeostatic and hedonic regulation. Trends Endocrinol Metab 2015; 26:455-66. [PMID: 26163755 DOI: 10.1016/j.tem.2015.06.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/15/2015] [Accepted: 06/16/2015] [Indexed: 11/21/2022]
Abstract
Glucose homeostasis as well as homeostatic and hedonic control of feeding is regulated by hormonal, neuronal, and nutrient-related cues. Glucose, besides its role as a source of metabolic energy, is an important signal controlling hormone secretion and neuronal activity, hence contributing to whole-body metabolic integration in coordination with feeding control. Brain glucose sensing plays a key, but insufficiently explored, role in these metabolic and behavioral controls, which when deregulated may contribute to the development of obesity and diabetes. The recent introduction of innovative transgenic, pharmacogenetic, and optogenetic techniques allows unprecedented analysis of the complexity of central glucose sensing at the molecular, cellular, and neuronal circuit levels, which will lead to a new understanding of the pathogenesis of metabolic diseases.
Collapse
Affiliation(s)
- Laura Steinbusch
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Gwenaël Labouèbe
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Nutrient-specific sensor systems in enteroendocrine cells detect intestinal contents and cause gut hormone release upon activation. Among these peptide hormones, the incretins glucose-dependent insulinotropic polypeptide and glucagon-like peptide 1 are of particular interest by their role in glucose homeostasis, metabolic control and for proper ß-cell function. This review focuses on intestinal nutrient-sensing processes and their role in health and disease. RECENT FINDINGS All macronutrients, respectively, their digestion products can cause incretin release by targeting specific sensors. Luminal glucose is the strongest stimulant for incretin release with the Na-dependent glucose transporter as the prime sensor. For peptides, the H-dependent peptide transporter together with calcium-sensing-receptor act as a sensing system. That transporters can function as nutrient-sensing 'transceptors' is conceptually new as G-protein coupled receptors so far were thought to be the sensing entities. This still holds true for GPR40 and GPR120 as sensors for medium/long-chain fatty acids and GPR41 and GPR43 for microbiota-derived short-chain fatty acids. Synthetic agonists for these receptors show impressive effects on glucagon-like peptide 1 output and glycemic control. Moreover, the remarkable and immediate antidiabetic effects of bariatric surgery/gastric bypass put intestinal nutrient sensing into focus of new strategies for metabolic control. SUMMARY Targeting the intestinal nutrient-sensing machinery by dietary and/or pharmacological means holds promises in particular for treatment of type 2 diabetes. This interest may help to better understand the nutrient-sensing processes and the involvement of the intestine in overall endocrine, neuronal and metabolic control.
Collapse
Affiliation(s)
- Tamara Zietek
- ZIEL - Institute for Food & Health, Technische Universität München, Freising, Germany
| | | |
Collapse
|
49
|
Thorens B, Tarussio D, Maestro MA, Rovira M, Heikkilä E, Ferrer J. Ins1(Cre) knock-in mice for beta cell-specific gene recombination. Diabetologia 2015; 58:558-65. [PMID: 25500700 PMCID: PMC4320308 DOI: 10.1007/s00125-014-3468-5] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/14/2014] [Indexed: 01/01/2023]
Abstract
AIMS/HYPOTHESIS Pancreatic beta cells play a central role in the control of glucose homeostasis by secreting insulin to stimulate glucose uptake by peripheral tissues. Understanding the molecular mechanisms that control beta cell function and plasticity has critical implications for the pathophysiology and therapy of major forms of diabetes. Selective gene inactivation in pancreatic beta cells, using the Cre-lox system, is a powerful approach to assess the role of particular genes in beta cells and their impact on whole body glucose homeostasis. Several Cre recombinase (Cre) deleter mice have been established to allow inactivation of genes in beta cells, but many show non-specific recombination in other cell types, often in the brain. METHODS We describe the generation of Ins1(Cre) and Ins1(CreERT2) mice in which the Cre or Cre-oestrogen receptor fusion protein (CreERT2) recombinases have been introduced at the initiation codon of the Ins1 gene. RESULTS We show that Ins1(Cre) mice induce efficient and selective recombination of floxed genes in beta cells from the time of birth, with no recombination in the central nervous system. These mice have normal body weight and glucose homeostasis. Furthermore, we show that tamoxifen treatment of adult Ins1(CreERT2) mice crossed with Rosa26-tdTomato mice induces efficient recombination in beta cells. CONCLUSIONS/INTERPRETATION These two strains of deleter mice are useful new resources to investigate the molecular physiology of pancreatic beta cells.
Collapse
Affiliation(s)
- Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Genopode Building, CH-1015 Lausanne, Switzerland
| | - David Tarussio
- Center for Integrative Genomics, University of Lausanne, Genopode Building, CH-1015 Lausanne, Switzerland
| | - Miguel Angel Maestro
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain, http://www.ciberdem.org/
| | - Meritxell Rovira
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain, http://www.ciberdem.org/
| | - Eija Heikkilä
- Center for Integrative Genomics, University of Lausanne, Genopode Building, CH-1015 Lausanne, Switzerland
| | - Jorge Ferrer
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain, http://www.ciberdem.org/
- Department of Medicine, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, W12 0NN UK
| |
Collapse
|
50
|
Taylor BL, Benthuysen J, Sander M. Postnatal β-cell proliferation and mass expansion is dependent on the transcription factor Nkx6.1. Diabetes 2015; 64:897-903. [PMID: 25277396 PMCID: PMC4338594 DOI: 10.2337/db14-0684] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
All forms of diabetes are characterized by a loss of functional β-cell mass, and strategies for expanding β-cell mass could have significant therapeutic benefit. We have recently identified the transcription factor Nkx6.1 as an essential maintenance factor of the functional β-cell state. In addition, Nkx6.1 has been proposed to control β-cell proliferation, but a role for Nkx6.1 in regulating β-cell mass has not been demonstrated. Here, we show that Nkx6.1 is required for postnatal β-cell mass expansion. Genetic inactivation of Nkx6.1 in newly formed β-cells caused a drastic decrease in early postnatal β-cell proliferation, leading to reduced β-cell mass and glucose intolerance. Interestingly, Nkx6.1 was dispensable for prenatal β-cell proliferation. We found that Nkx6.1 regulates the expression of several β-cell maturation markers as well as expression of the nutrient sensors Glut2 and Glp1r. Manifestation of the β-cell mass defect at the transition to postnatal feeding suggests that Nkx6.1 could regulate β-cell growth by enabling β-cells to respond to nutrient-dependent proliferation signals, such as glucose and Glp1. Identification of β-cell-intrinsic regulators that connect nutrient-sensing and proliferation suggests new therapeutic targets for expanding functional β-cell mass.
Collapse
Affiliation(s)
- Brandon L Taylor
- Departments of Pediatrics and Cellular and Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA
| | - Jacqueline Benthuysen
- Departments of Pediatrics and Cellular and Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA
| | - Maike Sander
- Departments of Pediatrics and Cellular and Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA
| |
Collapse
|