1
|
Marty-Lombardi S, Lu S, Ambroziak W, Schrenk-Siemens K, Wang J, DePaoli-Roach AA, Hagenston AM, Wende H, Tappe-Theodor A, Simonetti M, Bading H, Okun JG, Kuner R, Fleming T, Siemens J. Neuron-astrocyte metabolic coupling facilitates spinal plasticity and maintenance of inflammatory pain. Nat Metab 2024; 6:494-513. [PMID: 38443593 DOI: 10.1038/s42255-024-01001-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 01/31/2024] [Indexed: 03/07/2024]
Abstract
Long-lasting pain stimuli can trigger maladaptive changes in the spinal cord, reminiscent of plasticity associated with memory formation. Metabolic coupling between astrocytes and neurons has been implicated in neuronal plasticity and memory formation in the central nervous system, but neither its involvement in pathological pain nor in spinal plasticity has been tested. Here we report a form of neuroglia signalling involving spinal astrocytic glycogen dynamics triggered by persistent noxious stimulation via upregulation of the Protein Targeting to Glycogen (PTG) in spinal astrocytes. PTG drove glycogen build-up in astrocytes, and blunting glycogen accumulation and turnover by Ptg gene deletion reduced pain-related behaviours and promoted faster recovery by shortening pain maintenance in mice. Furthermore, mechanistic analyses revealed that glycogen dynamics is a critically required process for maintenance of pain by facilitating neuronal plasticity in spinal lamina 1 neurons. In summary, our study describes a previously unappreciated mechanism of astrocyte-neuron metabolic communication through glycogen breakdown in the spinal cord that fuels spinal neuron hyperexcitability.
Collapse
Affiliation(s)
| | - Shiying Lu
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Oliver Wyman GmbH, Munich, Germany
| | - Wojciech Ambroziak
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Department of Translational Disease Understanding, Grünenthal GmbH, Aachen, Germany
| | | | - Jialin Wang
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Anna A DePaoli-Roach
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anna M Hagenston
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, Heidelberg, Germany
| | - Hagen Wende
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Taconic Biosciences, Leverkusen, Germany
| | | | - Manuela Simonetti
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, Heidelberg, Germany
| | - Jürgen G Okun
- Dietmar-Hopp-Metabolic Center, Division of Neuropaediatrics and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| | - Rohini Kuner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Thomas Fleming
- Department of Endocrinology, Diabetology, Metabolism and Clinical Chemistry (Internal Medicine 1), Heidelberg University Hospital, Heidelberg, Germany
- German Center of Diabetes Research (DZD), Neuherberg, Germany
| | - Jan Siemens
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.
- Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
| |
Collapse
|
2
|
Su G, Farhat R, Laxman AK, Chapman-Natewa K, Nelson IE, Chan O. Astrocyte Glycogen Is a Major Source of Hypothalamic Lactate in Rats With Recurrent Hypoglycemia. Diabetes 2023; 72:1154-1160. [PMID: 37216640 PMCID: PMC10382650 DOI: 10.2337/db22-0902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 05/13/2023] [Indexed: 05/24/2023]
Abstract
Lactate is an important metabolic substrate for sustaining brain energy requirements when glucose supplies are limited. Recurring exposure to hypoglycemia (RH) raises lactate levels in the ventromedial hypothalamus (VMH), which contributes to counterregulatory failure. However, the source of this lactate remains unclear. The current study investigates whether astrocytic glycogen serves as the major source of lactate in the VMH of RH rats. By decreasing the expression of a key lactate transporter in VMH astrocytes of RH rats, we reduced extracellular lactate concentrations, suggesting excess lactate was locally produced from astrocytes. To determine whether astrocytic glycogen serves as the major source of lactate, we chronically delivered either artificial extracellular fluid or 1,4-dideoxy-1,4-imino-d-arabinitol to inhibit glycogen turnover in the VMH of RH animals. Inhibiting glycogen turnover in RH animals prevented the rise in VMH lactate and the development of counterregulatory failure. Lastly, we noted that RH led to an increase in glycogen shunt activity in response to hypoglycemia and elevated glycogen phosphorylase activity in the hours following a bout of hypoglycemia. Our data suggest that dysregulation of astrocytic glycogen metabolism following RH may be responsible, at least in part, for the rise in VMH lactate levels. ARTICLE HIGHLIGHTS Astrocytic glycogen serves as the major source of elevated lactate levels in the ventromedial hypothalamus (VMH) of animals exposed to recurring episodes of hypoglycemia. Antecedent hypoglycemia alters VMH glycogen turnover. Antecedent exposure to hypoglycemia enhances glycogen shunt activity in the VMH during subsequent bouts of hypoglycemia. In the immediate hours following a bout of hypoglycemia, sustained elevations in glycogen phosphorylase activity in the VMH of recurrently hypoglycemic animals contribute to sustained elevations in local lactate levels.
Collapse
Affiliation(s)
- Gong Su
- Cardiac Department, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Rawad Farhat
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Utah, Salt Lake City, UT
| | - Anil K. Laxman
- University of Utah Health Sciences Metabolic Phenotyping Core, University of Utah, Salt Lake City, UT
| | | | - Irvane E. Nelson
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Utah, Salt Lake City, UT
| | - Owen Chan
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Utah, Salt Lake City, UT
| |
Collapse
|
3
|
Chowdhury HH. Differences in cytosolic glucose dynamics in astrocytes and adipocytes measured by FRET-based nanosensors. Biophys Chem 2020; 261:106377. [PMID: 32302866 DOI: 10.1016/j.bpc.2020.106377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/08/2020] [Accepted: 04/08/2020] [Indexed: 11/17/2022]
Abstract
The cellular response to fluctuations in blood glucose levels consists of integrative regulation of cell glucose uptake and glucose utilization in the cytosol, resulting in altered levels of glucose in the cytosol. Cytosolic glucose is difficult to be measured in the intact tissue, however recently methods have become available that allow measurements of glucose in single living cells with fluorescence resonance energy transfer (FRET) based protein sensors. By studying the dynamics of cytosolic glucose levels in different experimental settings, we can gain insights into the properties of plasma membrane permeability to glucose and glucose utilization in the cytosol, and how these processes are modulated by different environmental conditions, agents and enzymes. In this review, we compare the cytosolic regulation of glucose in adipocytes and astrocytes - two important regulators of energy balance and glucose homeostasis in whole body and brain, respectively, with particular emphasis on the data obtained with FRET based protein sensors as well as other biochemical and molecular approaches.
Collapse
Affiliation(s)
- Helena H Chowdhury
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, 1000 Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia.
| |
Collapse
|
4
|
Matsui T, Liu YF, Soya M, Shima T, Soya H. Tyrosine as a Mechanistic-Based Biomarker for Brain Glycogen Decrease and Supercompensation With Endurance Exercise in Rats: A Metabolomics Study of Plasma. Front Neurosci 2019; 13:200. [PMID: 30941004 PMCID: PMC6433992 DOI: 10.3389/fnins.2019.00200] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 02/20/2019] [Indexed: 01/02/2023] Open
Abstract
Brain glycogen, localized in astrocytes, produces lactate as an energy source and/or a signal factor to serve neuronal functions involved in memory formation and exercise endurance. In rodents, 4 weeks of chronic moderate exercise-enhancing endurance and cognition increases brain glycogen in the hippocampus and cortex, which is an adaption of brain metabolism achieved through exercise. Although this brain adaptation is likely induced due to the accumulation of acute endurance exercise–induced brain glycogen supercompensation, its molecular mechanisms and biomarkers are unidentified. Since noradrenaline synthesized from blood-borne tyrosine activates not only glycogenolysis but also glycogenesis in astrocytes, we hypothesized that blood tyrosine is a mechanistic-based biomarker of acute exercise–induced brain glycogen supercompensation. To test this hypothesis, we used a rat model of endurance exercise, a microwave irradiation for accurate detection of glycogen in the brain (the cortex, hippocampus, and hypothalamus), and capillary electrophoresis mass spectrometry–based metabolomics to observe the comprehensive metabolic profile of the blood. Endurance exercise induced fatigue factors such as a decrease in blood glucose, an increase in blood lactate, and the depletion of muscle glycogen, but those parameters recovered to basal levels within 6 h after exercise. Brain glycogen decreased during endurance exercise and showed supercompensation within 6 h after exercise. Metabolomics detected 186 metabolites in the plasma, and 110 metabolites changed significantly during and following exhaustive exercise. Brain glycogen levels correlated negatively with plasma glycogenic amino acids (serine, proline, threonine, glutamate, methionine, tyrosine, and tryptophan) (r < −0.9). This is the first study to produce a broad picture of plasma metabolite changes due to endurance exercise–induced brain glycogen supercompensation. Our findings suggest that plasma glycogenic amino acids are sensitive indicators of brain glycogen levels in endurance exercise. In particular, plasma tyrosine as a precursor of brain noradrenaline might be a valuable mechanistic-based biomarker to predict brain glycogen dynamics in endurance exercise.
Collapse
Affiliation(s)
- Takashi Matsui
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, Tsukuba, Japan.,Sport Neuroscience Division, Advanced Research Initiative for Human High Performance (ARIHHP), University of Tsukuba, Tsukuba, Japan
| | - Yu-Fan Liu
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, Tsukuba, Japan
| | - Mariko Soya
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, Tsukuba, Japan.,Sport Neuroscience Division, Advanced Research Initiative for Human High Performance (ARIHHP), University of Tsukuba, Tsukuba, Japan
| | - Takeru Shima
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, Tsukuba, Japan
| | - Hideaki Soya
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, Tsukuba, Japan.,Sport Neuroscience Division, Advanced Research Initiative for Human High Performance (ARIHHP), University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
5
|
Morita M, Ikeshima-Kataoka H, Kreft M, Vardjan N, Zorec R, Noda M. Metabolic Plasticity of Astrocytes and Aging of the Brain. Int J Mol Sci 2019; 20:ijms20040941. [PMID: 30795555 PMCID: PMC6413111 DOI: 10.3390/ijms20040941] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 01/03/2023] Open
Abstract
As part of the blood-brain-barrier, astrocytes are ideally positioned between cerebral vasculature and neuronal synapses to mediate nutrient uptake from the systemic circulation. In addition, astrocytes have a robust enzymatic capacity of glycolysis, glycogenesis and lipid metabolism, managing nutrient support in the brain parenchyma for neuronal consumption. Here, we review the plasticity of astrocyte energy metabolism under physiologic and pathologic conditions, highlighting age-dependent brain dysfunctions. In astrocytes, glycolysis and glycogenesis are regulated by noradrenaline and insulin, respectively, while mitochondrial ATP production and fatty acid oxidation are influenced by the thyroid hormone. These regulations are essential for maintaining normal brain activities, and impairments of these processes may lead to neurodegeneration and cognitive decline. Metabolic plasticity is also associated with (re)activation of astrocytes, a process associated with pathologic events. It is likely that the recently described neurodegenerative and neuroprotective subpopulations of reactive astrocytes metabolize distinct energy substrates, and that this preference is supposed to explain some of their impacts on pathologic processes. Importantly, physiologic and pathologic properties of astrocytic metabolic plasticity bear translational potential in defining new potential diagnostic biomarkers and novel therapeutic targets to mitigate neurodegeneration and age-related brain dysfunctions.
Collapse
Affiliation(s)
- Mitsuhiro Morita
- Department of Biology, Graduate School of Sciences, Kobe University, 657-8501 Kobe, Japan.
| | - Hiroko Ikeshima-Kataoka
- Faculty of Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan.
| | - Marko Kreft
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia.
- Department of Biology, Biotechnical Faculty University of Ljubljana, 1000 Ljubljana, Slovenia.
| | - Nina Vardjan
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia.
| | - Robert Zorec
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia.
| | - Mami Noda
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| |
Collapse
|
6
|
Abstract
Glucose is the long-established, obligatory fuel for brain that fulfills many critical functions, including ATP production, oxidative stress management, and synthesis of neurotransmitters, neuromodulators, and structural components. Neuronal glucose oxidation exceeds that in astrocytes, but both rates increase in direct proportion to excitatory neurotransmission; signaling and metabolism are closely coupled at the local level. Exact details of neuron-astrocyte glutamate-glutamine cycling remain to be established, and the specific roles of glucose and lactate in the cellular energetics of these processes are debated. Glycolysis is preferentially upregulated during brain activation even though oxygen availability is sufficient (aerobic glycolysis). Three major pathways, glycolysis, pentose phosphate shunt, and glycogen turnover, contribute to utilization of glucose in excess of oxygen, and adrenergic regulation of aerobic glycolysis draws attention to astrocytic metabolism, particularly glycogen turnover, which has a high impact on the oxygen-carbohydrate mismatch. Aerobic glycolysis is proposed to be predominant in young children and specific brain regions, but re-evaluation of data is necessary. Shuttling of glucose- and glycogen-derived lactate from astrocytes to neurons during activation, neurotransmission, and memory consolidation are controversial topics for which alternative mechanisms are proposed. Nutritional therapy and vagus nerve stimulation are translational bridges from metabolism to clinical treatment of diverse brain disorders.
Collapse
Affiliation(s)
- Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences , Little Rock, Arkansas ; and Department of Cell Biology and Physiology, University of New Mexico , Albuquerque, New Mexico
| |
Collapse
|
7
|
Pederson BA. Structure and Regulation of Glycogen Synthase in the Brain. ADVANCES IN NEUROBIOLOGY 2019; 23:83-123. [PMID: 31667806 DOI: 10.1007/978-3-030-27480-1_3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Brain glycogen synthesis is a regulated, multi-step process that begins with glucose transport across the blood brain barrier and culminates with the actions of glycogen synthase and the glycogen branching enzyme to elongate glucose chains and introduce branch points in a growing glycogen molecule. This review focuses on the synthesis of glycogen in the brain, with an emphasis on glycogen synthase, but draws on salient studies in mammalian muscle and liver as well as baker's yeast, with the goal of providing a more comprehensive view of glycogen synthesis and highlighting potential areas for further study in the brain. In addition, deficiencies in the glycogen biosynthetic enzymes which lead to glycogen storage diseases in humans are discussed, highlighting effects on the brain and discussing findings in genetically modified animal models that recapitulate these diseases. Finally, implications of glycogen synthesis in neurodegenerative and other diseases that impact the brain are presented.
Collapse
|
8
|
Ratcliffe LE, Vázquez Villaseñor I, Jennings L, Heath PR, Mortiboys H, Schwartzentruber A, Karyka E, Simpson JE, Ince PG, Garwood CJ, Wharton SB. Loss of IGF1R in Human Astrocytes Alters Complex I Activity and Support for Neurons. Neuroscience 2018; 390:46-59. [PMID: 30056117 PMCID: PMC6372003 DOI: 10.1016/j.neuroscience.2018.07.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 11/15/2022]
Abstract
We have established a novel human astrocyte-neuron co-culture system. Astrocytes provided contact-mediated support for neurite outgrowth. IGF1R-impaired astrocytes are less able to protect neurons under stress conditions. Microarray analysis of these astrocytes identified changes in energy metabolism.
The insulin/insulin-like growth factor 1 (IGF1) signaling pathways are implicated in longevity and in progression of Alzheimer’s disease. Previously, we showed that insulin-like growth factor 1 receptor (IGF1R) and downstream signaling transcripts are reduced in astrocytes in human brain with progression of Alzheimer’s neuropathology and developed a model of IGF1 signaling impairment in human astrocytes using an IGF1R-specific monoclonal antibody, MAB391. Here, we have established a novel human astrocyte-neuron co-culture system to determine whether loss of astrocytic IGF1R affects their support for neurons. Astrocyte-neuron co-cultures were developed using human primary astrocytes and differentiated Lund Human Mesencephalic Cells (LUHMES). Neurite outgrowth assays, performed to measure astrocytic support for neurons, showed astrocytes provided contact-mediated support for neurite outgrowth. Loss of IGF1R did not affect neurite outgrowth under control conditions but when challenged with hydrogen peroxide IGF1R-impaired astrocytes were less able to protect LUHMES. To determine how loss of IGF1R affects neuronal support MAB391-treated astrocytes were FACS sorted from GFP-LUHMES and their transcriptomic profile was investigated using microarrays. Changes in transcripts involved in astrocyte energy metabolism were identified, particularly NDUFA2 and NDUFB6, which are related to complex I assembly. Loss of complex I activity in MAB391-treated astrocytes validated these findings. In conclusion, reduced IGF1 signaling in astrocytes impairs their support for neurons under conditions of stress and this is associated with defects in the mitochondrial respiratory chain in astrocytes.
Collapse
Affiliation(s)
- Laura E Ratcliffe
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Irina Vázquez Villaseñor
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Luke Jennings
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Aurelie Schwartzentruber
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Evangelia Karyka
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Paul G Ince
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Claire J Garwood
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK.
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| |
Collapse
|
9
|
Dean M, Rose J. Activation of the IGF1 receptor stimulates glycogen synthesis by mink uterine epithelial cells. Mol Reprod Dev 2018; 85:449-458. [DOI: 10.1002/mrd.22981] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 02/16/2018] [Accepted: 03/19/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Matthew Dean
- Department of Medicinal Chemistry and Pharmacognosy, Center for Biomolecular Sciences, College of Pharmacy; University of Illinois at Chicago; Chicago Illinois
- Department of Biological Sciences, College of Science and Engineering; Idaho State University; Idaho Pocatello
| | - Jack Rose
- Department of Biological Sciences, College of Science and Engineering; Idaho State University; Idaho Pocatello
| |
Collapse
|
10
|
Insulin-mediated synaptic plasticity in the CNS: Anatomical, functional and temporal contexts. Neuropharmacology 2017; 136:182-191. [PMID: 29217283 PMCID: PMC5988909 DOI: 10.1016/j.neuropharm.2017.12.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/01/2017] [Accepted: 12/03/2017] [Indexed: 12/17/2022]
Abstract
For decades the brain was erroneously considered an insulin insensitive organ. Although gaps in our knowledge base remain, conceptual frameworks are starting to emerge to provide insight into the mechanisms through which insulin facilitates critical brain functions like metabolism, cognition, and motivated behaviors. These diverse physiological and behavioral activities highlight the region-specific activities of insulin in the CNS; that is, there is an anatomical context to the activities of insulin in the CNS. Similarly, there is also a temporal context to the activities of insulin in the CNS. Indeed, brain insulin receptor activity can be conceptualized as a continuum in which insulin promotes neuroplasticity from development into adulthood where it is an integral part of healthy brain function. Unfortunately, brain insulin resistance likely contributes to neuroplasticity deficits in obesity and type 2 diabetes mellitus (T2DM). This neuroplasticity continuum can be conceptualized by the mechanisms through which insulin promotes cognitive function through its actions in brain regions like the hippocampus, as well as the ability of insulin to modulate motivated behaviors through actions in brain regions like the nucleus accumbens and the ventral tegmental area. Thus, the goals of this review are to highlight these anatomical, temporal, and functional contexts of insulin activity in these brain regions, and to identify potentially critical time points along this continuum where the transition from enhancement of neuroplasticity to impairment may take place.
Collapse
|
11
|
Fernandez AM, Hernandez E, Guerrero-Gomez D, Miranda-Vizuete A, Torres Aleman I. A network of insulin peptides regulate glucose uptake by astrocytes: Potential new druggable targets for brain hypometabolism. Neuropharmacology 2017; 136:216-222. [PMID: 28859884 DOI: 10.1016/j.neuropharm.2017.08.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/24/2017] [Accepted: 08/27/2017] [Indexed: 01/20/2023]
Abstract
Astrocytes are major players in brain glucose metabolism, supporting neuronal needs on demand through mechanisms that are not yet entirely clear. Understanding glucose metabolism in astrocytes is therefore of great consequence to unveil novel targets and develop new drugs to restore brain energy balance in pathology. Contrary to what has been held for many years, we now present evidence that insulin, in association with the related insulin-like growth factor I (IGF-I) modulates brain glucose metabolism through a concerted action on astrocytes. Cooperativity of insulin and IGF-I relies on the IGF-I receptor (IGF-IR), that acts as a scaffold of Glucose Transporter 1 (GluT1) regulating its activity by retaining it in the cytoplasm or, in response to a concerted action of insulin and IGF-I, translocating it to the cell membrane. Regulated translocation of GluT1 to the cell membrane by IGF-IR involves an intricate repertoire of protein-protein interactions amenable to drug modulation, particularly by interfering with IGF-IR/GluT1 interactions. We propose that this mechanism accounts for a substantial proportion of basal and regulated glucose uptake by astrocytes as GluT1 is the major glucose transporter in these brain cells. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- Ana M Fernandez
- Cajal Institute, Avda Dr Arce 37, 28002 Madrid, Spain; Ciberned, Madrid, Spain
| | - Edwin Hernandez
- Cajal Institute, Avda Dr Arce 37, 28002 Madrid, Spain; Ciberned, Madrid, Spain
| | - David Guerrero-Gomez
- Institute of Biomedicine (IBiS), Virgen del Rocío Hospital/CSIC/University of Seville, Seville, Spain
| | - Antonio Miranda-Vizuete
- Institute of Biomedicine (IBiS), Virgen del Rocío Hospital/CSIC/University of Seville, Seville, Spain
| | | |
Collapse
|
12
|
Astrocytic glycogen-derived lactate fuels the brain during exhaustive exercise to maintain endurance capacity. Proc Natl Acad Sci U S A 2017; 114:6358-6363. [PMID: 28515312 DOI: 10.1073/pnas.1702739114] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Brain glycogen stored in astrocytes provides lactate as an energy source to neurons through monocarboxylate transporters (MCTs) to maintain neuronal functions such as hippocampus-regulated memory formation. Although prolonged exhaustive exercise decreases brain glycogen, the role of this decrease and lactate transport in the exercising brain remains less clear. Because muscle glycogen fuels exercising muscles, we hypothesized that astrocytic glycogen plays an energetic role in the prolonged-exercising brain to maintain endurance capacity through lactate transport. To test this hypothesis, we used a rat model of exhaustive exercise and capillary electrophoresis-mass spectrometry-based metabolomics to observe comprehensive energetics of the brain (cortex and hippocampus) and muscle (plantaris). At exhaustion, muscle glycogen was depleted but brain glycogen was only decreased. The levels of MCT2, which takes up lactate in neurons, increased in the brain, as did muscle MCTs. Metabolomics revealed that brain, but not muscle, ATP was maintained with lactate and other glycogenolytic/glycolytic sources. Intracerebroventricular injection of the glycogen phosphorylase inhibitor 1,4-dideoxy-1,4-imino-d-arabinitol did not affect peripheral glycemic conditions but suppressed brain lactate production and decreased hippocampal ATP levels at exhaustion. An MCT2 inhibitor, α-cyano-4-hydroxy-cinnamate, triggered a similar response that resulted in lower endurance capacity. These findings provide direct evidence for the energetic role of astrocytic glycogen-derived lactate in the exhaustive-exercising brain, implicating the significance of brain glycogen level in endurance capacity. Glycogen-maintained ATP in the brain is a possible defense mechanism for neurons in the exhausted brain.
Collapse
|
13
|
Ruchti E, Roach P, DePaoli-Roach A, Magistretti P, Allaman I. Protein targeting to glycogen is a master regulator of glycogen synthesis in astrocytes. IBRO Rep 2016; 1:46-53. [PMID: 30135927 PMCID: PMC6084890 DOI: 10.1016/j.ibror.2016.10.002] [Citation(s) in RCA: 16] [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: 08/19/2016] [Revised: 10/03/2016] [Accepted: 10/03/2016] [Indexed: 01/21/2023] Open
Abstract
The storage and use of glycogen, the main energy reserve in the brain, is a metabolic feature of astrocytes. Glycogen synthesis is regulated by Protein Targeting to Glycogen (PTG), a member of specific glycogen-binding subunits of protein phosphatase-1 (PPP1). It positively regulates glycogen synthesis through de-phosphorylation of both glycogen synthase (activation) and glycogen phosphorylase (inactivation). In cultured astrocytes, PTG mRNA levels were previously shown to be enhanced by the neurotransmitter noradrenaline. To achieve further insight into the role of PTG in the regulation of astrocytic glycogen, its levels of expression were manipulated in primary cultures of mouse cortical astrocytes using adenovirus-mediated overexpression of tagged-PTG or siRNA to downregulate its expression. Infection of astrocytes with adenovirus led to a strong increase in PTG expression and was associated with massive glycogen accumulation (>100 fold), demonstrating that increased PTG expression is sufficient to induce glycogen synthesis and accumulation. In contrast, siRNA-mediated downregulation of PTG resulted in a 2-fold decrease in glycogen levels. Interestingly, PTG downregulation strongly impaired long-term astrocytic glycogen synthesis induced by insulin or noradrenaline. Finally, these effects of PTG downregulation on glycogen metabolism could also be observed in cultured astrocytes isolated from PTG-KO mice. Collectively, these observations point to a major role of PTG in the regulation of glycogen synthesis in astrocytes and indicate that conditions leading to changes in PTG expression will directly impact glycogen levels in this cell type.
Collapse
Affiliation(s)
- E. Ruchti
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Centre de Neurosciences Psychiatriques, CHUV, Département de Psychiatrie, Site de Cery, CH-1008 Prilly/Lausanne, Switzerland
| | - P.J. Roach
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - A.A. DePaoli-Roach
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - P.J. Magistretti
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Centre de Neurosciences Psychiatriques, CHUV, Département de Psychiatrie, Site de Cery, CH-1008 Prilly/Lausanne, Switzerland
- King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - I. Allaman
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
14
|
Hernandez-Garzón E, Fernandez AM, Perez-Alvarez A, Genis L, Bascuñana P, Fernandez de la Rosa R, Delgado M, Angel Pozo M, Moreno E, McCormick PJ, Santi A, Trueba-Saiz A, Garcia-Caceres C, Tschöp MH, Araque A, Martin ED, Torres Aleman I. The insulin-like growth factor I receptor regulates glucose transport by astrocytes. Glia 2016; 64:1962-71. [PMID: 27462832 DOI: 10.1002/glia.23035] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 07/05/2016] [Accepted: 07/07/2016] [Indexed: 12/20/2022]
Abstract
Previous findings indicate that reducing brain insulin-like growth factor I receptor (IGF-IR) activity promotes ample neuroprotection. We now examined a possible action of IGF-IR on brain glucose transport to explain its wide protective activity, as energy availability is crucial for healthy tissue function. Using (18) FGlucose PET we found that shRNA interference of IGF-IR in mouse somatosensory cortex significantly increased glucose uptake upon sensory stimulation. In vivo microscopy using astrocyte specific staining showed that after IGF-IR shRNA injection in somatosensory cortex, astrocytes displayed greater increases in glucose uptake as compared to astrocytes in the scramble-injected side. Further, mice with the IGF-IR knock down in astrocytes showed increased glucose uptake in somatosensory cortex upon sensory stimulation. Analysis of underlying mechanisms indicated that IGF-IR interacts with glucose transporter 1 (GLUT1), the main facilitative glucose transporter in astrocytes, through a mechanism involving interactions with the scaffolding protein GIPC and the multicargo transporter LRP1 to retain GLUT1 inside the cell. These findings identify IGF-IR as a key modulator of brain glucose metabolism through its inhibitory action on astrocytic GLUT1 activity. GLIA 2016;64:1962-1971.
Collapse
Affiliation(s)
| | | | - Alberto Perez-Alvarez
- Cajal Institute, CSIC, Madrid, Spain.,Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Laura Genis
- Cajal Institute, CSIC, Madrid, Spain.,Ciberned, Spain
| | - Pablo Bascuñana
- Pluridisciplinary Institute, Complutense University, Madrid, Spain
| | | | - Mercedes Delgado
- Pluridisciplinary Institute, Complutense University, Madrid, Spain
| | | | - Estefania Moreno
- Ciberned, Spain.,Dept Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain
| | - Peter J McCormick
- Ciberned, Spain.,Dept Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain.,School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - Andrea Santi
- Cajal Institute, CSIC, Madrid, Spain.,Ciberned, Spain
| | | | | | | | | | - Eduardo D Martin
- Science and Technology Park, Institute for Research in Neurological Disabilities, University of Castilla-La Mancha, Albacete, Spain
| | | |
Collapse
|
15
|
Astroglial glutamate transporters coordinate excitatory signaling and brain energetics. Neurochem Int 2016; 98:56-71. [PMID: 27013346 DOI: 10.1016/j.neuint.2016.03.014] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/15/2016] [Accepted: 03/17/2016] [Indexed: 12/22/2022]
Abstract
In the mammalian brain, a family of sodium-dependent transporters maintains low extracellular glutamate and shapes excitatory signaling. The bulk of this activity is mediated by the astroglial glutamate transporters GLT-1 and GLAST (also called EAAT2 and EAAT1). In this review, we will discuss evidence that these transporters co-localize with, form physical (co-immunoprecipitable) interactions with, and functionally couple to various 'energy-generating' systems, including the Na(+)/K(+)-ATPase, the Na(+)/Ca(2+) exchanger, glycogen metabolizing enzymes, glycolytic enzymes, and mitochondria/mitochondrial proteins. This functional coupling is bi-directional with many of these systems both being regulated by glutamate transport and providing the 'fuel' to support glutamate uptake. Given the importance of glutamate uptake to maintaining synaptic signaling and preventing excitotoxicity, it should not be surprising that some of these systems appear to 'redundantly' support the energetic costs of glutamate uptake. Although the glutamate-glutamine cycle contributes to recycling of neurotransmitter pools of glutamate, this is an over-simplification. The ramifications of co-compartmentalization of glutamate transporters with mitochondria for glutamate metabolism are discussed. Energy consumption in the brain accounts for ∼20% of the basal metabolic rate and relies almost exclusively on glucose for the production of ATP. However, the brain does not possess substantial reserves of glucose or other fuels. To ensure adequate energetic supply, increases in neuronal activity are matched by increases in cerebral blood flow via a process known as 'neurovascular coupling'. While the mechanisms for this coupling are not completely resolved, it is generally agreed that astrocytes, with processes that extend to synapses and endfeet that surround blood vessels, mediate at least some of the signal that causes vasodilation. Several studies have shown that either genetic deletion or pharmacologic inhibition of glutamate transport impairs neurovascular coupling. Together these studies strongly suggest that glutamate transport not only coordinates excitatory signaling, but also plays a pivotal role in regulating brain energetics.
Collapse
|
16
|
Abstract
Objective: To analyze the mechanism of neuroprotection of insulin and which blood glucose range was benefit for insulin exerting neuroprotective action. Data Sources: The study is based on the data from PubMed. Study Selection: Articles were selected with the search terms “insulin”, “blood glucose”, “neuroprotection”, “brain”, “glycogen”, “cerebral ischemia”, “neuronal necrosis”, “glutamate”, “γ-aminobutyric acid”. Results: Insulin has neuroprotection. The mechanisms include the regulation of neurotransmitter, promoting glycogen synthesis, and inhibition of neuronal necrosis and apoptosis. Insulin could play its role in neuroprotection by avoiding hypoglycemia and hyperglycemia. Conclusions: Intermittent and long-term infusion insulin may be a benefit for patients with ischemic brain damage at blood glucose 6–9 mmol/L.
Collapse
Affiliation(s)
| | - Yu Pei
- Department of Endocrinology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
17
|
Muhič M, Vardjan N, Chowdhury HH, Zorec R, Kreft M. Insulin and Insulin-like Growth Factor 1 (IGF-1) Modulate Cytoplasmic Glucose and Glycogen Levels but Not Glucose Transport across the Membrane in Astrocytes. J Biol Chem 2015; 290:11167-76. [PMID: 25792745 DOI: 10.1074/jbc.m114.629063] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Indexed: 01/21/2023] Open
Abstract
Astrocytes contain glycogen, an energy buffer, which can bridge local short term energy requirements in the brain. Glycogen levels reflect a dynamic equilibrium between glycogen synthesis and glycogenolysis. Many factors that include hormones and neuropeptides, such as insulin and insulin-like growth factor 1 (IGF-1) likely modulate glycogen stores in astrocytes, but detailed mechanisms at the cellular level are sparse. We used a glucose nanosensor based on Förster resonance energy transfer to monitor cytosolic glucose concentration with high temporal resolution and a cytochemical approach to determine glycogen stores in single cells. The results show that after glucose depletion, glycogen stores are replenished. Insulin and IGF-1 boost the process of glycogen formation. Although astrocytes appear to express glucose transporter GLUT4, glucose entry across the astrocyte plasma membrane is not affected by insulin. Stimulation of cells with insulin and IGF-1 decreased cytosolic glucose concentration, likely because of elevated glucose utilization for glycogen synthesis.
Collapse
Affiliation(s)
- Marko Muhič
- From the Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| | - Nina Vardjan
- From the Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia, Celica Biomedical, 1000 Ljubljana, Slovenia, and
| | - Helena H Chowdhury
- From the Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia, Celica Biomedical, 1000 Ljubljana, Slovenia, and
| | - Robert Zorec
- From the Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia, Celica Biomedical, 1000 Ljubljana, Slovenia, and
| | - Marko Kreft
- From the Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia, Celica Biomedical, 1000 Ljubljana, Slovenia, and the Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, 1000 Ljubljana, Slovenia
| |
Collapse
|
18
|
Zhang Q, Guo S, Zhang X, Tang S, Wang L, Han X, Shao W, Cong L, Du Y. Amyloid β oligomer-induced ERK1/2-dependent serine 636/639 phosphorylation of insulin receptor substrate-1 impairs insulin signaling and glycogen storage in human astrocytes. Gene 2015; 561:76-81. [PMID: 25667991 DOI: 10.1016/j.gene.2015.02.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 12/25/2014] [Accepted: 02/06/2015] [Indexed: 12/18/2022]
Abstract
AIMS This study is to investigate the effect of amyloid β1-42 oligomers on insulin signaling in astrocytes. METHODS Synthetic Aβ1-42 oligomers were prepared and the oligomeric form of Aβ1-42 was verified by an electron microscope. Normal human astrocytes were cultured in Dulbecco's Modified Eagle Medium. Western blotting was employed to measure the amount of lysate proteins. Immunofluorescence was performed to detect the distribution of phosphorylated insulin receptor substrate-1 and expression of P-GSK3β in astrocytes under confocal microscopy and fluorescent microscopy, respectively. Periodic Acid-Schiff staining was used to detect glycogen, the content of which was measured using glycogen assay. RESULTS Our data showed that Aβ1-42 oligomers inhibited insulin-induced serine phosphorylation of Akt at 473 and GSK3β at serine 9, as well as glycogen storage. However, the levels of phosphorylated GSK3β at tyrosine 216 were significantly increased in the presence of Aβ1-42 oligomers. In addition, the levels of phosphorylated ERK1/2 and insulin receptor substrate-1 at serine 636/639 were significantly increased in response to treatment with Aβ1-42 oligomers. Of note, the responses and inhibitory effects of Aβ1-42 oligomers on insulin signaling were partially reversed by ERK1/2 upstream inhibitor PD98059. CONCLUSIONS Our results demonstrated that Aβ1-42 oligomers impaired insulin signaling and suppressed insulin-induced glycogen storage in human astrocytes, probably due to ERK1/2-dependent serine phosphorylation of insulin receptor substrate-1 at 636/639 induced by Aβ1-42 oligomers.
Collapse
Affiliation(s)
- Qinghua Zhang
- Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan City, Shandong Province, PR China
| | - Shougang Guo
- Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan City, Shandong Province, PR China
| | - Xiao Zhang
- Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan City, Shandong Province, PR China
| | - Shi Tang
- Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan City, Shandong Province, PR China
| | - Lu Wang
- Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan City, Shandong Province, PR China
| | - Xiaojuan Han
- Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan City, Shandong Province, PR China
| | - Wen Shao
- Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan City, Shandong Province, PR China
| | - Lin Cong
- Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan City, Shandong Province, PR China
| | - Yifeng Du
- Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan City, Shandong Province, PR China.
| |
Collapse
|
19
|
Petit JM, Burlet-Godinot S, Magistretti PJ, Allaman I. Glycogen metabolism and the homeostatic regulation of sleep. Metab Brain Dis 2015; 30:263-79. [PMID: 25399336 PMCID: PMC4544655 DOI: 10.1007/s11011-014-9629-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/04/2014] [Indexed: 12/19/2022]
Abstract
In 1995 Benington and Heller formulated an energy hypothesis of sleep centered on a key role of glycogen. It was postulated that a major function of sleep is to replenish glycogen stores in the brain that have been depleted during wakefulness which is associated to an increased energy demand. Astrocytic glycogen depletion participates to an increase of extracellular adenosine release which influences sleep homeostasis. Here, we will review some evidence obtained by studies addressing the question of a key role played by glycogen metabolism in sleep regulation as proposed by this hypothesis or by an alternative hypothesis named "glycogenetic" hypothesis as well as the importance of the confounding effect of glucocorticoïds. Even though actual collected data argue in favor of a role of sleep in brain energy balance-homeostasis, they do not support a critical and direct involvement of glycogen metabolism on sleep regulation. For instance, glycogen levels during the sleep-wake cycle are driven by different physiological signals and therefore appear more as a marker-integrator of brain energy status than a direct regulator of sleep homeostasis. In support of this we provide evidence that blockade of glycogen mobilization does not induce more sleep episodes during the active period while locomotor activity is reduced. These observations do not invalidate the energy hypothesis of sleep but indicate that underlying cellular mechanisms are more complex than postulated by Benington and Heller.
Collapse
Affiliation(s)
- Jean-Marie Petit
- Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, 1015, Switzerland,
| | | | | | | |
Collapse
|
20
|
Jackson JG, O'Donnell JC, Krizman E, Robinson MB. Displacing hexokinase from mitochondrial voltage-dependent anion channel impairs GLT-1-mediated glutamate uptake but does not disrupt interactions between GLT-1 and mitochondrial proteins. J Neurosci Res 2014; 93:999-1008. [PMID: 25546576 DOI: 10.1002/jnr.23533] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 11/11/2014] [Accepted: 11/30/2014] [Indexed: 02/03/2023]
Abstract
The glutamate transporter GLT-1 is the major route for the clearance of extracellular glutamate in the forebrain, and most GLT-1 protein is found in astrocytes. This protein is coupled to the Na(+) electrochemical gradient, supporting the active intracellular accumulation of glutamate. We recently used a proteomic approach to identify proteins that may interact with GLT-1 in rat cortex, including the Na(+)/K(+) -ATPase, most glycolytic enzymes, and several mitochondrial proteins. We also showed that most GLT-1 puncta (∼ 70%) are overlapped by mitochondria in astroglial processes in organotypic slices. From this analysis, we proposed that the glycolytic enzyme hexokinase (HK)-1 might physically form a scaffold to link GLT-1 and mitochondria because HK1 is known to interact with the outer mitochondrial membrane protein voltage-dependent anion channel (VDAC). The current study validates the interactions among HK-1, VDAC, and GLT-1 by using forward and reverse immunoprecipitations and provides evidence that a subfraction of HK1 colocalizes with GLT-1 in vivo. A peptide known to disrupt the interaction between HK and VDAC did not disrupt interactions between GLT-1 and several mitochondrial proteins. In parallel experiments, displacement of HK from VDAC reduced GLT-1-mediated glutamate uptake. These results suggest that, although HK1 forms coimmunoprecipitatable complexes with both VDAC and GLT-1, it does not physically link GLT-1 to mitochondrial proteins. However, the interaction of HK1 with VDAC supports GLT-1-mediated transport activity.
Collapse
Affiliation(s)
- Joshua G Jackson
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania.,Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John C O'Donnell
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania.,Department of Pharmacology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth Krizman
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Michael B Robinson
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania.,Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Pharmacology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
21
|
Lanz B, Gruetter R, Duarte JMN. Metabolic Flux and Compartmentation Analysis in the Brain In vivo. Front Endocrinol (Lausanne) 2013; 4:156. [PMID: 24194729 PMCID: PMC3809570 DOI: 10.3389/fendo.2013.00156] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 10/07/2013] [Indexed: 12/16/2022] Open
Abstract
Through significant developments and progresses in the last two decades, in vivo localized nuclear magnetic resonance spectroscopy (MRS) became a method of choice to probe brain metabolic pathways in a non-invasive way. Beside the measurement of the total concentration of more than 20 metabolites, (1)H MRS can be used to quantify the dynamics of substrate transport across the blood-brain barrier by varying the plasma substrate level. On the other hand, (13)C MRS with the infusion of (13)C-enriched substrates enables the characterization of brain oxidative metabolism and neurotransmission by incorporation of (13)C in the different carbon positions of amino acid neurotransmitters. The quantitative determination of the biochemical reactions involved in these processes requires the use of appropriate metabolic models, whose level of details is strongly related to the amount of data accessible with in vivo MRS. In the present work, we present the different steps involved in the elaboration of a mathematical model of a given brain metabolic process and its application to the experimental data in order to extract quantitative brain metabolic rates. We review the recent advances in the localized measurement of brain glucose transport and compartmentalized brain energy metabolism, and how these reveal mechanistic details on glial support to glutamatergic and GABAergic neurons.
Collapse
Affiliation(s)
- Bernard Lanz
- Laboratory for Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Rolf Gruetter
- Laboratory for Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Department of Radiology, University of Lausanne, Lausanne, Switzerland
- Department of Radiology, University of Geneva, Geneva, Switzerland
| | - João M. N. Duarte
- Laboratory for Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Department of Radiology, University of Lausanne, Lausanne, Switzerland
- *Correspondence: João M. N. Duarte, Laboratory for Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne, Bâtiment CH, Station 6, CH-1015 Lausanne, Switzerland e-mail:
| |
Collapse
|
22
|
Pérezleón JA, Osorio-Paz I, Francois L, Salceda R. Immunohistochemical localization of glycogen synthase and GSK3β: control of glycogen content in retina. Neurochem Res 2013; 38:1063-9. [PMID: 23512644 DOI: 10.1007/s11064-013-1017-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 03/04/2013] [Accepted: 03/12/2013] [Indexed: 11/30/2022]
Abstract
Glycogen has an important role in energy handling in several brain regions. In the brain, glycogen is localized in astrocytes and its role in several normal and pathological processes has been described, whereas in the retina, glycogen metabolism has been scarcely investigated. The enzyme glycogen phosphorylase has been located in retinal Müller cells; however the cellular location of glycogen synthase (GS) and its regulatory partner, glycogen synthase kinase 3β (GSK3β), has not been investigated. Our aim was to localize these enzymes in the rat retina by immunofluorescence techniques. We found both GS and GSK3β in Müller cells in the synaptic layers, and within the inner segments of photoreceptor cells. The presence of these enzymes in Müller cells suggests that glycogen could be regulated within the retina as in other tissues. Indeed, we showed that glycogen content in the whole retina in vitro was increased by high glucose concentrations, glutamate, and insulin. In contrast, retina glycogen levels were not modified by norepinephrine nor by depolarization with high KCl concentrations. Insulin also induced an increase in glycogen content in cultured Müller cells. The effect of insulin in both, whole retina and cultured Müller cells was blocked by inhibitors of phosphatidyl-inositol 3-kinase, strongly suggesting that glycogen content in retina is modulated by the insulin signaling pathway. The expression of GS and GSK3β in the synaptic layers and photoreceptor cells suggests an important role of GSK3β regulating glycogen synthase in neurons, which opens multiple feasible roles of insulin within the retina.
Collapse
Affiliation(s)
- Jorge Alberto Pérezleón
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Anillo PRONAF y Estocolmo s/n, Colonia PRONAF, 31330 Ciudad Juárez, Chihuahua, Mexico.
| | | | | | | |
Collapse
|
23
|
Grondona JM, Granados-Durán P, Fernández-Llebrez P, López-Ávalos MD. A simple method to obtain pure cultures of multiciliated ependymal cells from adult rodents. Histochem Cell Biol 2012; 139:205-20. [PMID: 22878526 DOI: 10.1007/s00418-012-1008-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2012] [Indexed: 11/25/2022]
Abstract
Ependymal cells form an epithelium lining the ventricular cavities of the vertebrate brain. Numerous methods to obtain primary culture ependymal cells have been developed. Most of them use foetal or neonatal rat brain and the few that utilize adult brain hardly achieve purity. Here, we describe a simple and novel method to obtain a pure non-adherent ependymal cell culture from explants of the striatal and septal walls of the lateral ventricles. The combination of a low incubation temperature followed by a gentle enzymatic digestion allows the detachment of most of the ependymal cells from the ventricular wall in a period of 6 h. Along with ependymal cells, a low percentage (less than 6 %) of non-ependymal cells also detaches. However, they do not survive under two restrictive culture conditions: (1) a simple medium (alpha-MEM with glucose) without any supplement; and (2) a low density of 1 cell/µl. This purification method strategy does not require cell labelling with antibodies and cell sorting, which makes it a simpler and cheaper procedure than other methods previously described. After a period of 48 h, only ependymal cells survive such conditions, revealing the remarkable survival capacity of ependymal cells. Ependymal cells can be maintained in culture for up to 7-10 days, with the best survival rates obtained in Neurobasal supplemented with B27 among the tested media. After 7 days in culture, ependymal cells lose most of the cilia and therefore the mobility, while acquiring radial glial cell markers (GFAP, BLBP, GLAST). This interesting fact might indicate a reprogramming of the cell identity.
Collapse
Affiliation(s)
- J M Grondona
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Campus de Teatinos, Universidad de Málaga, 29071 Málaga, Spain.
| | | | | | | |
Collapse
|
24
|
Schmidt JM, Claassen J, Ko SB, Lantigua H, Presciutti M, Lee K, Connolly ES, Mayer SA, Seres DS, Badjatia N. Nutritional support and brain tissue glucose metabolism in poor-grade SAH: a retrospective observational study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2012; 16:R15. [PMID: 22277085 PMCID: PMC3396251 DOI: 10.1186/cc11160] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 11/01/2011] [Accepted: 01/25/2012] [Indexed: 01/04/2023]
Abstract
Introduction We sought to determine the effect of nutritional support and insulin infusion therapy on serum and brain glucose levels and cerebral metabolic crisis after aneurysmal subarachnoid hemorrhage (SAH). Methods We used a retrospective observational cohort study of 50 mechanically ventilated poor-grade (Hunt-Hess 4 or 5) aneurysmal SAH patients who underwent brain microdialysis monitoring for an average of 109 hours. Enteral nutrition was started within 72 hours of admission whenever feasible. Intensive insulin therapy was used to maintain serum glucose levels between 5.5 and 7.8 mmol/l. Serum glucose, insulin and caloric intake from enteral tube feeds, dextrose and propofol were recorded hourly. Cerebral metabolic distress was defined as a lactate to pyruvate ratio (LPR) > 40. Time-series data were analyzed using a general linear model extended by generalized estimation equations (GEE). Results Daily mean caloric intake received was 13.8 ± 6.9 cal/kg and mean serum glucose was 7.9 ± 1 mmol/l. A total of 32% of hourly recordings indicated a state of metabolic distress and < 1% indicated a state of critical brain hypoglycemia (< 0.2 mmol/l). Calories received from enteral tube feeds were associated with higher serum glucose concentrations (Wald = 6.07, P = 0.048), more insulin administered (Wald = 108, P < 0.001), higher body mass index (Wald = 213.47, P < 0.001), and lower body temperature (Wald = 4.1, P = 0.043). Enteral feeding (Wald = 1.743, P = 0.418) was not related to brain glucose concentrations after accounting for serum glucose concentrations (Wald = 67.41, P < 0.001). In the presence of metabolic distress, increased insulin administration was associated with a relative reduction of interstitial brain glucose concentrations (Wald = 8.26, P = 0.017), independent of serum glucose levels. Conclusions In the presence of metabolic distress, insulin administration is associated with reductions in brain glucose concentration that are independent of serum glucose levels. Further study is needed to understand how nutritional support and insulin administration can be optimized to minimize secondary injury after subarachnoid hemorrhage.
Collapse
Affiliation(s)
- J Michael Schmidt
- Columbia University, Department of Neurology, Milstein Hospital, 177 Fort Washington, Suite 8-300, New York, NY 10032, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ola MS, Hosoya KI, LaNoue KF. Influence of insulin on glutamine synthetase in the Müller glial cells of retina. Metab Brain Dis 2011; 26:195-202. [PMID: 21626103 DOI: 10.1007/s11011-011-9245-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 05/12/2011] [Indexed: 11/27/2022]
Abstract
Glutamine synthetase (GS), a Müller cell specific enzyme in the retina, is the key enzyme involve in glutamate metabolism. The goal of this study was to investigate the expression and regulation of GS by insulin in the cultured rat retinal Müller cells. Immunocytochemical and immunoblotting experiments showed that the cultured Müller cells express GS protein under normal cell culture conditions. Insulin treatments decreased the GS expression both in a time and dose dependent manner. Insulin also decreased the hydrocortisone induced GS expression. Furthermore, we investigated the expression and regulation of two other Müller cell specific enzymes known to be involved in glutamate metabolism, the mitochondrial branched chain aminotransferase (BCATm) and pyruvate carboxylase (PC). Immunoblotting experiments showed that Müller cells expressed both BCATm and PC. Treatments of cells with hydrocortisone or insulin did not influence the BCATm expression level. Hydrocortisone treatment of cells increased the PC expression but this induced expression was suppressed by insulin treatment. Müller cells expressed insulin receptor proteins (IRβ and IRS-1) and insulin activation induced the phosphotyrosine level of insulin receptor proteins. Moreover, hydrocortisone did not influence the expression or activation of these receptor proteins. The data suggests that insulin modulates the GS synthesis and may influence glutamate metabolism in the cultured retinal Müller cells but not by influencing the insulin signaling pathway.
Collapse
Affiliation(s)
- Mohammad Shamsul Ola
- Department of Ophthalmology, College of Medicine, King Saud University, Old Airport Road, KAUH, P.O. Box 245, Riyadh 11411, Kingdom of Saudi Arabia.
| | | | | |
Collapse
|
26
|
Tesfaye N, Seaquist ER, Oz G. Noninvasive measurement of brain glycogen by nuclear magnetic resonance spectroscopy and its application to the study of brain metabolism. J Neurosci Res 2011; 89:1905-12. [PMID: 21732401 DOI: 10.1002/jnr.22703] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Revised: 04/19/2011] [Accepted: 05/02/2011] [Indexed: 11/11/2022]
Abstract
Glycogen is the reservoir for glucose in the brain. Beyond the general agreement that glycogen serves as an energy source in the central nervous system, its exact role in brain energy metabolism has yet to be elucidated. Experiments performed in cell and tissue culture and animals have shown that glycogen content is affected by several factors, including glucose, insulin, neurotransmitters, and neuronal activation. The study of in vivo glycogen metabolism has been hindered by the inability to measure glycogen noninvasively, but, in the past several years, the development of a noninvasive localized (13) C nuclear magnetic resonance (NMR) spectroscopy method has allowed the study of glycogen metabolism in the conscious human. With this technique, (13) C-glucose is administered intravenously, and its incorporation into and washout from brain glycogen is tracked. One application of this method has been to the study of brain glycogen metabolism in humans during hypoglycemia: data have shown that mobilization of brain glycogen is augmented during hypoglycemia, and, after a single episode of hypoglycemia, glycogen synthesis rate is increased, suggesting that glycogen stores rebound to levels greater than baseline. Such studies suggest that glycogen may serve as a potential energy reservoir in hypoglycemia and may participate in the brain's adaptation to recurrent hypoglycemia and eventual development of hypoglycemia unawareness. Beyond this focused area of study, (13) C NMR spectroscopy has a broad potential for application in the study of brain glycogen metabolism and carries the promise of a better understanding of the role of brain glycogen in diabetes and other conditions.
Collapse
Affiliation(s)
- Nolawit Tesfaye
- Department of Medicine, Division of Endocrinology and Diabetes, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
27
|
Heni M, Hennige AM, Peter A, Siegel-Axel D, Ordelheide AM, Krebs N, Machicao F, Fritsche A, Häring HU, Staiger H. Insulin promotes glycogen storage and cell proliferation in primary human astrocytes. PLoS One 2011; 6:e21594. [PMID: 21738722 PMCID: PMC3124526 DOI: 10.1371/journal.pone.0021594] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 06/06/2011] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION In the human brain, there are at least as many astrocytes as neurons. Astrocytes are known to modulate neuronal function in several ways. Thus, they may also contribute to cerebral insulin actions. Therefore, we examined whether primary human astrocytes are insulin-responsive and whether their metabolic functions are affected by the hormone. METHODS Commercially available Normal Human Astrocytes were grown in the recommended medium. Major players in the insulin signaling pathway were detected by real-time RT-PCR and Western blotting. Phosphorylation events were detected by phospho-specific antibodies. Glucose uptake and glycogen synthesis were assessed using radio-labeled glucose. Glycogen content was assessed by histochemistry. Lactate levels were measured enzymatically. Cell proliferation was assessed by WST-1 assay. RESULTS We detected expression of key proteins for insulin signaling, such as insulin receptor β-subunit, insulin receptor substrat-1, Akt/protein kinase B and glycogen synthase kinase 3, in human astrocytes. Akt was phosphorylated and PI-3 kinase activity increased following insulin stimulation in a dose-dependent manner. Neither increased glucose uptake nor lactate secretion after insulin stimulation could be evidenced in this cell type. However, we found increased insulin-dependent glucose incorporation into glycogen. Furthermore, cell numbers increased dose-dependently upon insulin treatment. DISCUSSION This study demonstrated that human astrocytes are insulin-responsive at the molecular level. We identified glycogen synthesis and cell proliferation as biological responses of insulin signaling in these brain cells. Hence, this cell type may contribute to the effects of insulin in the human brain.
Collapse
Affiliation(s)
- Martin Heni
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, Member of the German Center for Diabetes Research (DZD), Eberhard Karls University Tübingen, Tübingen, Germany
| | - Anita M. Hennige
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, Member of the German Center for Diabetes Research (DZD), Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Peter
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, Member of the German Center for Diabetes Research (DZD), Eberhard Karls University Tübingen, Tübingen, Germany
| | - Dorothea Siegel-Axel
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, Member of the German Center for Diabetes Research (DZD), Eberhard Karls University Tübingen, Tübingen, Germany
| | - Anna-Maria Ordelheide
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, Member of the German Center for Diabetes Research (DZD), Eberhard Karls University Tübingen, Tübingen, Germany
| | - Norbert Krebs
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, Member of the German Center for Diabetes Research (DZD), Eberhard Karls University Tübingen, Tübingen, Germany
| | - Fausto Machicao
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, Member of the German Center for Diabetes Research (DZD), Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, Member of the German Center for Diabetes Research (DZD), Eberhard Karls University Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, Member of the German Center for Diabetes Research (DZD), Eberhard Karls University Tübingen, Tübingen, Germany
- * E-mail:
| | - Harald Staiger
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, Member of the German Center for Diabetes Research (DZD), Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
28
|
Hutchinson DS, Catus SL, Merlin J, Summers RJ, Gibbs ME. α₂-Adrenoceptors activate noradrenaline-mediated glycogen turnover in chick astrocytes. J Neurochem 2011; 117:915-26. [PMID: 21447002 DOI: 10.1111/j.1471-4159.2011.07261.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In the brain, glycogen is primarily stored in astrocytes where it is regulated by several hormones/neurotransmitters, including noradrenaline that controls glycogen breakdown (in the short term) and synthesis. Here, we have examined the adrenoceptor (AR) subtype that mediates the glycogenic effect of noradrenaline in chick primary astrocytes by the measurement of glycogen turnover (total (14) C incorporation of glucose into glycogen) following noradrenergic activation. Noradrenaline and insulin increased glycogen turnover in a concentration-dependent manner. The effect of noradrenaline was mimicked by stimulation of α(2) -ARs (and to a lesser degree by β(3) -ARs), but not by stimulation of α(1) -, β(1) -, or β(2) -ARs, and occurred only in astrocytes and not neurons. In chick astrocytes, studies using RT-PCR and radioligand binding showed that α(2A) - and α(2C) -AR mRNA and protein were present. α(2) -AR- or insulin-mediated glycogen turnover was inhibited by phosphatidylinositol-3 kinase inhibitors, and both insulin and clonidine caused phosphorylation of Akt and glycogen synthase kinase-3 in chick astrocytes. α(2) -AR but not insulin-mediated glycogen turnover was inhibited by pertussis toxin pre-treatment indicating involvement of Gi/o proteins. These results show that the increase in glycogen turnover caused by noradrenaline is because of activation of α(2) -ARs that increase glycogen turnover in astrocytes utilizing a Gi/o-PI3K pathway.
Collapse
Affiliation(s)
- Dana S Hutchinson
- Department of Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia.
| | | | | | | | | |
Collapse
|
29
|
Nielsen TT, Støttrup NB, Løfgren B, Bøtker HE. Metabolic fingerprint of ischaemic cardioprotection: importance of the malate-aspartate shuttle. Cardiovasc Res 2011; 91:382-91. [PMID: 21349875 DOI: 10.1093/cvr/cvr051] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The convergence of cardioprotective intracellular signalling pathways to modulate mitochondrial function as an end-target of cytoprotective stimuli is well described. However, our understanding of whether the complementary changes in mitochondrial energy metabolism are secondary responses or inherent mechanisms of ischaemic cardioprotection remains incomplete. In the heart, the malate-aspartate shuttle (MAS) constitutes the primary metabolic pathway for transfer of reducing equivalents from the cytosol into the mitochondria for oxidation. The flux of MAS is tightly linked to the flux of the tricarboxylic acid cycle and the electron transport chain, partly by the amino acid l-glutamate. In addition, emerging evidence suggests the MAS is an important regulator of cytosolic and mitochondrial calcium homeostasis. In the isolated rat heart, inhibition of MAS during ischaemia and early reperfusion by the aminotransferase inhibitor aminooxyacetate induces infarct limitation, improves haemodynamic responses, and modulates glucose metabolism, analogous to effects observed in classical ischaemic preconditioning. On the basis of these findings, the mechanisms through which MAS preserves mitochondrial function and cell survival are reviewed. We conclude that the available evidence is supportive of a down-regulation of mitochondrial respiration during lethal ischaemia with a gradual 'wake-up' during reperfusion as a pivotal feature of ischaemic cardioprotection. Finally, comments on modulating myocardial energy metabolism by the cardioprotective amino acids glutamate and glutamine are given.
Collapse
Affiliation(s)
- Torsten Toftegaard Nielsen
- Department of Cardiology, Skejby Hospital, Aarhus University Hospital, Brendstrupgaardsvej 100, Aarhus N, Denmark.
| | | | | | | |
Collapse
|
30
|
Glycogen synthesis in brain and astrocytes is inhibited by chronic lithium treatment. Neurosci Lett 2010; 482:128-32. [DOI: 10.1016/j.neulet.2010.07.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 07/06/2010] [Accepted: 07/08/2010] [Indexed: 02/06/2023]
|
31
|
Amyloid-beta aggregates cause alterations of astrocytic metabolic phenotype: impact on neuronal viability. J Neurosci 2010; 30:3326-38. [PMID: 20203192 DOI: 10.1523/jneurosci.5098-09.2010] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Amyloid-beta (Abeta) peptides play a key role in the pathogenesis of Alzheimer's disease and exert various toxic effects on neurons; however, relatively little is known about their influence on glial cells. Astrocytes play a pivotal role in brain homeostasis, contributing to the regulation of local energy metabolism and oxidative stress defense, two aspects of importance for neuronal viability and function. In the present study, we explored the effects of Abeta peptides on glucose metabolism in cultured astrocytes. Following Abeta(25-35) exposure, we observed an increase in glucose uptake and its various metabolic fates, i.e., glycolysis (coupled to lactate release), tricarboxylic acid cycle, pentose phosphate pathway, and incorporation into glycogen. Abeta increased hydrogen peroxide production as well as glutathione release into the extracellular space without affecting intracellular glutathione content. A causal link between the effects of Abeta on glucose metabolism and its aggregation and internalization into astrocytes through binding to members of the class A scavenger receptor family could be demonstrated. Using astrocyte-neuron cocultures, we observed that the overall modifications of astrocyte metabolism induced by Abeta impair neuronal viability. The effects of the Abeta(25-35) fragment were reproduced by Abeta(1-42) but not by Abeta(1-40). Finally, the phosphoinositide 3-kinase (PI3-kinase) pathway appears to be crucial in these events since both the changes in glucose utilization and the decrease in neuronal viability are prevented by LY294002, a PI3-kinase inhibitor. This set of observations indicates that Abeta aggregation and internalization into astrocytes profoundly alter their metabolic phenotype with deleterious consequences for neuronal viability.
Collapse
|
32
|
Abstract
OBJECTIVE We tested the hypotheses that human brain glycogen is mobilized during hypoglycemia and its content increases above normal levels ("supercompensates") after hypoglycemia. RESEARCH DESIGN AND METHODS We utilized in vivo (13)C nuclear magnetic resonance spectroscopy in conjunction with intravenous infusions of [(13)C]glucose in healthy volunteers to measure brain glycogen metabolism during and after euglycemic and hypoglycemic clamps. RESULTS After an overnight intravenous infusion of 99% enriched [1-(13)C]glucose to prelabel glycogen, the rate of label wash-out from [1-(13)C]glycogen was higher (0.12 +/- 0.05 vs. 0.03 +/- 0.06 micromol x g(-1) x h(-1), means +/- SD, P < 0.02, n = 5) during a 2-h hyperinsulinemic-hypoglycemic clamp (glucose concentration 57.2 +/- 9.7 mg/dl) than during a hyperinsulinemic-euglycemic clamp (95.3 +/- 3.3 mg/dl), indicating mobilization of glucose units from glycogen during moderate hypoglycemia. Five additional healthy volunteers received intravenous 25-50% enriched [1-(13)C]glucose over 22-54 h after undergoing hyperinsulinemic-euglycemic (glucose concentration 92.4 +/- 2.3 mg/dl) and hyperinsulinemic-hypoglycemic (52.9 +/- 4.8 mg/dl) clamps separated by at least 1 month. Levels of newly synthesized glycogen measured from 4 to 80 h were higher after hypoglycemia than after euglycemia (P <or= 0.01 for each subject), indicating increased brain glycogen synthesis after moderate hypoglycemia. CONCLUSIONS These data indicate that brain glycogen supports energy metabolism when glucose supply from the blood is inadequate and that its levels rebound to levels higher than normal after a single episode of moderate hypoglycemia in humans.
Collapse
Affiliation(s)
- Gülin Oz
- Center for MR Research, Department of Radiology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Poblete-Naredo I, Angulo C, Hernández-Kelly L, López-Bayghen E, Aguilera J, Ortega A. Insulin-dependent regulation of GLAST/EAAT1 in Bergmann glial cells. Neurosci Lett 2009; 451:134-8. [DOI: 10.1016/j.neulet.2008.12.049] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2008] [Revised: 12/01/2008] [Accepted: 12/24/2008] [Indexed: 11/25/2022]
|
34
|
Role of beta-adrenoceptors in memory consolidation: beta3-adrenoceptors act on glucose uptake and beta2-adrenoceptors on glycogenolysis. Neuropsychopharmacology 2008; 33:2384-97. [PMID: 18046311 DOI: 10.1038/sj.npp.1301629] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Noradrenaline, acting via beta(2)- and beta(3)-adrenoceptors (AR), enhances memory formation in single trial-discriminated avoidance learning in day-old chicks by mechanisms involving changes in metabolism of glucose and/or glycogen. Earlier studies of memory consolidation in chicks implicated beta(3)- rather than beta(2)-ARs in enhancement of memory consolidation by glucose, but did not elucidate whether stimulation of glucose uptake or of glycolysis was responsible. This study examines the role of glucose transport in memory formation using central injection of the nonselective facilitative glucose transporter (GLUT) inhibitor cytochalasin B, the endothelial/astrocytic GLUT-1 inhibitor phloretin and the Na(+)/energy-dependent endothelial glucose transporter (SGLT) inhibitor phlorizin. Cytochalasin B inhibited memory when injected into the mesopallium (avian cortex) either close to or between 25 and 45 min after training, whereas phloretin and phlorizin only inhibited memory at 30 min. This suggested that astrocytic/endothelial (GLUT-1) transport is critical at the time of consolidation, whereas a different transporter, probably the neuronal glucose transporter (GLUT-3), is important at the time of training. Inhibition of glucose transport by cytochalasin B, phloretin, or phlorizin also interfered with beta(3)-AR-mediated memory enhancement 20 min posttraining, whereas inhibition of glycogenolysis interfered with beta(2)-AR agonist enhancement of memory. We conclude that in astrocytes (1) activities of both GLUT-1 and SGLT are essential for memory consolidation 30 min posttraining; (2) neuronal GLUT-3 is essential at the time of training; and (3) beta(2)- and beta(3)-ARs consolidate memory by different mechanisms; beta(3)-ARs stimulate central glucose transport, whereas beta(2)-ARs stimulate central glycogenolysis.
Collapse
|
35
|
Hutchinson DS, Summers RJ, Gibbs ME. Energy metabolism and memory processing: role of glucose transport and glycogen in responses to adrenoceptor activation in the chicken. Brain Res Bull 2008; 76:224-34. [PMID: 18498935 DOI: 10.1016/j.brainresbull.2008.02.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2007] [Revised: 01/22/2008] [Accepted: 02/11/2008] [Indexed: 11/28/2022]
Abstract
From experiments using a discriminated bead task in young chicks, we have defined when and where adrenoceptors (ARs) are involved in memory modulation. All three ARs subtypes (alpha(1)-, alpha(2)- and beta-ARs) are found in the chick brain and in regions associated with memory. Glucose and glycogen are important in the role of memory consolidation in the chick since increasing glucose levels improves memory consolidation while inhibiting glucose transporters (GLUTs) or glycogen breakdown inhibits memory consolidation. The selective beta(3)-AR agonist CL316243 enhances memory consolidation by a glucose-dependent mechanism and the administration of the non-metabolized glucose analogue 2-deoxyglucose reduces the ability of CL316243 to enhance memory. Agents that reduce glucose uptake by GLUTs and its incorporation into the glycolytic pathway also reduce the effectiveness of CL316243, but do not alter the dose-response relationship to the beta(2)-AR agonist zinterol. However, beta(2)-ARs do have a role in memory related to glycogen breakdown and inhibition of glycogenolysis reduces the ability of zinterol to enhance memory. Both beta(2)- and beta(3)-ARs are found on astrocytes from chick forebrain, and the actions of beta(3)-ARs on glucose uptake, and beta(2)-ARs on the breakdown of glycogen is consistent with an effect on astrocytic metabolism at the time of memory consolidation 30 min after training. We have shown that both beta(2)- and beta(3)-ARs can increase glucose uptake in chick astrocytes but do so by different mechanisms. This review will focus on the role of ARs on memory consolidation and specifically the role of energy metabolism on AR modulation of memory.
Collapse
Affiliation(s)
- Dana S Hutchinson
- Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia.
| | | | | |
Collapse
|
36
|
Badaut J, Brunet J, Petit JM, Guérin C, Magistretti P, Regli L. Induction of brain aquaporin 9 (AQP9) in catecholaminergic neurons in diabetic rats. Brain Res 2008; 1188:17-24. [DOI: 10.1016/j.brainres.2007.10.087] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Revised: 10/18/2007] [Accepted: 10/24/2007] [Indexed: 11/28/2022]
|
37
|
Oz G, Seaquist ER, Kumar A, Criego AB, Benedict LE, Rao JP, Henry PG, Van De Moortele PF, Gruetter R. Human brain glycogen content and metabolism: implications on its role in brain energy metabolism. Am J Physiol Endocrinol Metab 2007; 292:E946-51. [PMID: 17132822 DOI: 10.1152/ajpendo.00424.2006] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The adult brain relies on glucose for its energy needs and stores it in the form of glycogen, primarily in astrocytes. Animal and culture studies indicate that brain glycogen may support neuronal function when the glucose supply from the blood is inadequate and/or during neuronal activation. However, the concentration of glycogen and rates of its metabolism in the human brain are unknown. We used in vivo localized 13C-NMR spectroscopy to measure glycogen content and turnover in the human brain. Nine healthy volunteers received intravenous infusions of [1-(13)C]glucose for durations ranging from 6 to 50 h, and brain glycogen labeling and washout were measured in the occipital lobe for up to 84 h. The labeling kinetics suggest that turnover is the main mechanism of label incorporation into brain glycogen. Upon fitting a model of glycogen metabolism to the time courses of newly synthesized glycogen, human brain glycogen content was estimated at approximately 3.5 micromol/g, i.e., three- to fourfold higher than free glucose at euglycemia. Turnover of bulk brain glycogen occurred at a rate of 0.16 micromol.g-1.h-1, implying that complete turnover requires 3-5 days. Twenty minutes of visual stimulation (n=5) did not result in detectable glycogen utilization in the visual cortex, as judged from similar [13C]glycogen levels before and after stimulation. We conclude that the brain stores a substantial amount of glycogen relative to free glucose and metabolizes this store very slowly under normal physiology.
Collapse
Affiliation(s)
- Gülin Oz
- Department of Radiology, Center for MR Research, University of Minnesota, 2021 6th St. SE, Minneapolis, MN 55455, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Pellerin L, Bouzier-Sore AK, Aubert A, Serres S, Merle M, Costalat R, Magistretti PJ. Activity-dependent regulation of energy metabolism by astrocytes: An update. Glia 2007; 55:1251-1262. [PMID: 17659524 DOI: 10.1002/glia.20528] [Citation(s) in RCA: 579] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Astrocytes play a critical role in the regulation of brain metabolic responses to activity. One detailed mechanism proposed to describe the role of astrocytes in some of these responses has come to be known as the astrocyte-neuron lactate shuttle hypothesis (ANLSH). Although controversial, the original concept of a coupling mechanism between neuronal activity and glucose utilization that involves an activation of aerobic glycolysis in astrocytes and lactate consumption by neurons provides a heuristically valid framework for experimental studies. In this context, it is necessary to provide a survey of recent developments and data pertaining to this model. Thus, here, we review very recent experimental evidence as well as theoretical arguments strongly supporting the original model and in some cases extending it. Aspects revisited include the existence of glutamate-induced glycolysis in astrocytes in vitro, ex vivo, and in vivo, lactate as a preferential oxidative substrate for neurons, and the notion of net lactate transfer between astrocytes and neurons in vivo. Inclusion of a role for glycogen in the ANLSH is discussed in the light of a possible extension of the astrocyte-neuron lactate shuttle (ANLS) concept rather than as a competing hypothesis. New perspectives offered by the application of this concept include a better understanding of the basis of signals used in functional brain imaging, a role for neuron-glia metabolic interactions in glucose sensing and diabetes, as well as novel strategies to develop therapies against neurodegenerative diseases based upon improving astrocyte-neuron coupled energetics.
Collapse
Affiliation(s)
- Luc Pellerin
- Département de Physiologie, Université de Lausanne, Switzerland
| | - Anne-Karine Bouzier-Sore
- Unité de Résonance Magnétique des Systèmes Biologiques, UMR5536 CNRS-Université Victor Segalen, Bordeaux, France
| | - Agnès Aubert
- Département de Physiologie, Université de Lausanne, Switzerland
| | - Sébastien Serres
- Unité de Résonance Magnétique des Systèmes Biologiques, UMR5536 CNRS-Université Victor Segalen, Bordeaux, France
| | - Michel Merle
- Unité de Résonance Magnétique des Systèmes Biologiques, UMR5536 CNRS-Université Victor Segalen, Bordeaux, France
| | - Robert Costalat
- INSERM U678, Université Pierre et Marie Curie-Paris 6, Paris, France
| | - Pierre J Magistretti
- Brain and Mind Institute, Ecole Polytechnique Fédérale de Lausanne and Centre de Neurosciences Psychiatriques, Hôpital de Cery, Prilly, Switzerland
| |
Collapse
|
39
|
Verleysdonk S, Kistner S, Pfeiffer-Guglielmi B, Wellard J, Lupescu A, Laske J, Lang F, Rapp M, Hamprecht B. Glycogen metabolism in rat ependymal primary cultures: regulation by serotonin. Brain Res 2005; 1060:89-99. [PMID: 16202983 DOI: 10.1016/j.brainres.2005.08.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Revised: 08/18/2005] [Accepted: 08/19/2005] [Indexed: 10/25/2022]
Abstract
Ependymal primary cultures are a model for studying ependymal energy metabolism. Intracellular glycogen is built up in the cultures dependent on culture age and the presence of glucose and glutamate. This energy store is mobilized upon glucose withdrawal, stimulation with isoproterenol, forskolin or serotonin and after uncoupling of oxidative phosphorylation from ATP production. Serotonin regulates ependymal glycogen metabolism predominantly via 5-HT receptor (5-HTR) 7, which elicits an increase in the level of ependymal cyclic AMP. Although the most abundant mRNAs for serotonin receptors are those of 5-HTR 2B and 5-HTR 3A, ependymal cells in primary culture do not respond to serotonin with an increase in their concentration of cytosolic calcium ions. The mRNAs of 5-HTRs 1A, 6, 1B, 5B, 7, 1/2C and 5A are also detectable in order of decreasing abundance. The mRNAs for 5-HTRs 1D, 1F, 3B and 4 are absent from the cultured cells. The ability of serotonin to mobilize ependymal glycogen depends on the culture age and the time allowed for glycogen buildup. During glycogen buildup time, glutamate is consumed by the cells. An increased ability of 5-HT to mobilize ependymal glycogen stores is noticed after the depletion of glutamate from the glycogen buildup medium. In ependymal primary cultures, cilia are colocalized with glycogen phosphorylase isozyme BB, while the MM isoform is not expressed. It is known from the literature that an increase in the concentration of cytosolic cAMP in ependymal cells leads to a decrease in ciliary beat frequency. Therefore, the present data point towards a function for ependymal glycogen other than supplying energy for the movement of cilia.
Collapse
Affiliation(s)
- Stephan Verleysdonk
- Interfaculty Institute for Biochemistry, University of Tübingen, Hoppe-Seyler-Str. 4, D-72076 Tübingen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hallschmid M, Schultes B, Marshall L, Mölle M, Kern W, Bredthauer J, Fehm HL, Born J. Transcortical direct current potential shift reflects immediate signaling of systemic insulin to the human brain. Diabetes 2004; 53:2202-8. [PMID: 15331528 DOI: 10.2337/diabetes.53.9.2202] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Circulating insulin is thought to provide a major feedback signal for the hypothalamic regulation of energy homeostasis and food intake, although this signaling appears to be slowed by a time-consuming blood-to-brain transport. Here we show, by recording direct current potentials, a rapid onset of the effects of circulating insulin on human brain activity. Recordings were obtained from 27 men who were intravenously injected with insulin (0.1 mU/kg body wt as bolus) and placebo. In a euglycemic condition, hypoglycemia was prevented, while in the hypoglycemic condition, plasma glucose reached a postinjection nadir of 43 mg/dl. Insulin injection induced a marked negative direct current (DC) potential shift starting within 7 min in all subjects. With euglycemic conditions, the DC potential at 10-60 min postinsulin injection averaged -621.3 microV (compared with preinjection baseline). Hypoglycemia reduced this potential to an average of -331.2 microV. While insulin per se did not affect oscillatory electroencephalographic activity, hypoglycemia peaking 25 min after insulin injection was accompanied by an immediate increase in theta activity. The rapid emergence of the DC potential shift, reflecting gross ionic changes in brain tissues, indicates that systemic insulin can serve as an immediate feedback signal in the control of hypothalamic and higher brain functions.
Collapse
Affiliation(s)
- Manfred Hallschmid
- University of Lübeck, Institute of Neuroendocrinology, Ratzeburger Allee 160, Haus 23a, 23538 Lübeck, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Verleysdonk S, Hirschner W, Wellard J, Rapp M, de los Angeles Garcia M, Nualart F, Hamprecht B. Regulation by insulin and insulin-like growth factor of 2-deoxyglucose uptake in primary ependymal cell cultures. Neurochem Res 2004; 29:127-34. [PMID: 14992271 DOI: 10.1023/b:nere.0000010441.08234.ca] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ependymal cells have been reported to express the facilitative glucose carriers GLUT1, GLUT2, and GLUT4, as well as glucokinase. They are therefore speculated to be part of the cerebral glucose sensing system and may also respond to insulin with alterations in their glucose uptake rate. A cell culture model was employed to study the functional status of ependymal insulin-regulated glucose uptake in vitro. Insulin increased the uptake of the model substrate 2-deoxyglucose (2-DG) dependent on the insulin concentration. This was due to a near doubling of the maximal 2-DG uptake rate. Insulin-like growth factor (IGF-1) was at least 10 times more potent than insulin in stimulating the rate of ependymal 2-DG uptake, suggesting that IGF-1, rather than insulin, is the physiological agonist regulating glucose transport in ependymal cells. The predominant glucose transporter in ependymal cell cultures was found to be GLUT1, which is apparently regulated by IGF-1 in ependymal cells.
Collapse
Affiliation(s)
- Stephan Verleysdonk
- Physiologisch-Chemisches Institut der Universität, Hoppe-Seyler-Str. 4, D-72076 Tübingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
42
|
Dienel GA, Cruz NF. Nutrition during brain activation: does cell-to-cell lactate shuttling contribute significantly to sweet and sour food for thought? Neurochem Int 2004; 45:321-51. [PMID: 15145548 DOI: 10.1016/j.neuint.2003.10.011] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2003] [Revised: 10/24/2003] [Accepted: 10/27/2003] [Indexed: 10/26/2022]
Abstract
Functional activation of astrocytic metabolism is believed, according to one hypothesis, to be closely linked to excitatory neurotransmission and to provide lactate as fuel for oxidative metabolism in neighboring neurons. However, review of emerging evidence suggests that the energetic demands of activated astrocytes are higher and more complex than recognized and much of the lactate presumably produced by astrocytes is not locally oxidized during activation. In vivo activation studies in normal subjects reveal that the rise in consumption of blood-borne glucose usually exceeds that of oxygen, especially in retina compared to brain. When the contribution of glycogen, the brain's major energy reserve located in astrocytes, is taken into account the magnitude of the carbohydrate-oxygen utilization mismatch increases further because the magnitude of glycogenolysis greatly exceeds the incremental increase in utilization of blood-borne glucose. Failure of local oxygen consumption to equal that of glucose plus glycogen in vivo is strong evidence against stoichiometric transfer of lactate from astrocytes to neighboring neurons for oxidation. Thus, astrocytes, not nearby neurons, use the glycogen for energy during physiological activation in normal brain. These findings plus apparent compartmentation of metabolism of glycogen and blood-borne glucose during activation lead to our working hypothesis that activated astrocytes have high energy demands in their fine perisynaptic processes (filopodia) that might be met by glycogenolysis and glycolysis coupled to rapid lactate clearance. Tissue culture studies do not consistently support the lactate shuttle hypothesis because key elements of the model, glutamate-induced increases in glucose utilization and lactate release, are not observed in many astrocyte preparations, suggesting differences in their oxidative capacities that have not been included in the model. In vivo nutritional interactions between working neurons and astrocytes are not as simple as implied by "sweet (glucose-glycogen) and sour (lactate) food for thought."
Collapse
Affiliation(s)
- Gerald A Dienel
- Department of Neurology, Slot 830, University of Arkansas for Medical Sciences, Room 715, Shorey Building, 4301 W. Markham Street, Little Rock, AR 72205, USA.
| | | |
Collapse
|
43
|
Abstract
The mammalian brain contains glycogen, which is located predominantly in astrocytes, but its function is unclear. A principal role for brain glycogen as an energy reserve, analogous to its role in the periphery, had been universally dismissed based on its relatively low concentration, an assumption apparently reinforced by the limited duration that the brain can function in the absence of glucose. However, during insulin-induced hypoglycaemia, where brain glucose availability is limited, glycogen content falls first in areas with the highest metabolic rate, suggesting that glycogen provides fuel to support brain function during pathological hypoglycaemia. General anaesthesia results in elevated brain glycogen suggesting quiescent neurones allow glycogen accumulation, and as long ago as the 1950s it was shown that brain glycogen accumulates during sleep, is mobilized upon waking, and that sleep deprivation results in region-specific decreases in brain glycogen, implying a supportive functional role for brain glycogen in the conscious, awake brain. Interest in brain glycogen has recently been re-awakened by the first continuous in vivo measurements using NMR spectroscopy, by the general acceptance of metabolic coupling between glia and neurones involving intercellular transfer of energy substrate, and by studies supporting a prominent physiological role for brain glycogen as a provider of supplemental energy substrate during periods of increased tissue energy demand, when ambient normoglycaemic glucose is unable to meet immediate energy requirements.
Collapse
Affiliation(s)
- Angus M Brown
- Department of Neurology, University of Washington School of Medicine, Seattle, Washington, USA.
| |
Collapse
|
44
|
Oz G, Henry PG, Seaquist ER, Gruetter R. Direct, noninvasive measurement of brain glycogen metabolism in humans. Neurochem Int 2003; 43:323-9. [PMID: 12742076 DOI: 10.1016/s0197-0186(03)00019-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The concentration and metabolism of the primary carbohydrate store in the brain, glycogen, is unknown in the conscious human brain. This study reports the first direct detection and measurement of glycogen metabolism in the human brain, which was achieved using localized 13C NMR spectroscopy. To enhance the NMR signal, the isotopic enrichment of the glucosyl moieties was increased by administration of 80 g of 99% enriched [1-13C]glucose in four subjects. 3 h after the start of the label administration, the 13C NMR signal of brain glycogen C1 was detected (0.36+/-0.07 micromol/g, mean+/-S.D., n=4). Based on the rate of 13C label incorporation into glycogen and the isotopic enrichment of plasma glucose, the flux through glycogen synthase was estimated at 0.17+/-0.05 micromol/(gh). This study establishes that brain glycogen can be measured in humans and indicates that its metabolism is very slow in the conscious human. The noninvasive detection of human brain glycogen opens the prospect of understanding the role and function of this important energy reserve under various physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Gülin Oz
- Department of Radiology, Center for MR Research, University of Minnesota, 2021 6th St. S.E., Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
45
|
Pfeiffer-Guglielmi B, Fleckenstein B, Jung G, Hamprecht B. Immunocytochemical localization of glycogen phosphorylase isozymes in rat nervous tissues by using isozyme-specific antibodies. J Neurochem 2003; 85:73-81. [PMID: 12641728 DOI: 10.1046/j.1471-4159.2003.01644.x] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Isozyme-specific antibodies were raised against peptides from the low-homology regions of the sequences of rat glycogen phosphorylase BB and MM isozymes by immunization of rabbits and guinea pigs. Immunocytochemical double-labelling experiments on frozen sections of rat nervous tissues were performed to investigate the isozyme localization pattern. Astrocytes throughout the brain and spinal cord expressed both isozymes in perfect co-localization. Ependymal cells only expressed the BB isozyme. Most neurones were not immunoreactive. The rare neurones that contained glycogen phosphorylase only expressed the BB isozyme. Nearly all of these neurones formed part of the afferent somatosensory system. These findings stress the general importance of glycogen in neural energy metabolism and indicate a special role for the glycogen phosphorylase BB isozyme in neurones in the somatosensory system.
Collapse
Affiliation(s)
- Brigitte Pfeiffer-Guglielmi
- Physiologisch-Chemisches Institut der Universität and Institut für Organische Chemie der Universität, Tübingen, Germany
| | | | | | | |
Collapse
|
46
|
Phillis JW, O'Regan MH. Energy utilization in the ischemic/reperfused brain. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2003; 51:377-414. [PMID: 12420365 DOI: 10.1016/s0074-7742(02)51011-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Affiliation(s)
- John W Phillis
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | |
Collapse
|
47
|
Bouzier-Sore AK, Merle M, Magistretti PJ, Pellerin L. Feeding active neurons: (re)emergence of a nursing role for astrocytes. JOURNAL OF PHYSIOLOGY, PARIS 2002; 96:273-82. [PMID: 12445906 DOI: 10.1016/s0928-4257(02)00016-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Despite unquestionable evidence that glucose is the major energy substrate for the brain, data collected over several decades with different approaches suggest that lactate may represent a supplementary metabolic substrate for neurons. Starting with the pioneering work of McIlwain in the early 1950s which showed that lactate can sustain the respiratory rate of small brain tissue pieces, this idea receives confirmation with more recent studies using nuclear magnetic resonance spectroscopy undoubtedly demonstrating that lactate is efficiently oxidized by neurons, both in vitro and in vivo. Not only is lactate able to maintain ATP levels and promote neuronal survival but it was also found to support neuronal activity, at least if low levels of glucose are present. Despite the early suggestion for a role of astrocytes in metabolic supply to neurons, it is only recently however that they have been considered as a potential source of lactate for neurons. Moreover, it has been proposed that astrocytes might provide lactate to neurons in response to enhanced synaptic activity by a well-characterized mechanism involving glutamate uptake. The description of specific transporters for lactate on both astrocytes and neurons further suggest that there exist a coordinated mechanism of lactate exchange between the two cell types. Thus it is proposed that astrocytes play a nursing role toward neurons by providing lactate as an additional energy substrate especially during periods of enhanced synaptic activity. The importance of this metabolic cooperation within the central nervous system, although not unique if compared to other organs, still remains to be explored.
Collapse
|
48
|
Qu H, Eloqayli H, Unsgård G, Sonnewald U. Glutamate decreases pyruvate carboxylase activity and spares glucose as energy substrate in cultured cerebellar astrocytes. J Neurosci Res 2001; 66:1127-32. [PMID: 11746445 DOI: 10.1002/jnr.10032] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The effects of glutamate on [U-(13)C]glucose metabolism were studied in cerebellar astrocytes using (13)C magnetic resonance spectroscopy. Labeled glutamate, glutamine, aspartate, lactate, and alanine were observed both in the cell extracts and in media, and, additionally, labeled glycogen was detected in the cell extracts. However, only labeled lactate and alanine were quantifiable in the medium in addition to [U-(13)C]glucose. In the presence of unlabeled glutamate, the amount of [U-(13)C]glucose removed from the medium was decreased, indicating that glutamate might spare glucose as an energy substrate and thus decrease the uptake of glucose. Labeled glycogen, [4,5-(13)C]glutamate, [3,4,5-(13)C]glutamate, [3,4-(13)C]aspartate, and [U-(13)C]alanine were increased in the presence of glutamate. However, the increase in the amount of [3,4,5-(13)C]glutamate from the second turn in the tricarboxylic acid (TCA) cycle was less pronounced than that of [4,5-(13)C]glutamate from the first turn in the TCA cycle. This indicates the dilution of label, probably resulting from the synthesis of unlabeled oxaloacetate from glutamate in the TCA cycle. Furthermore, exogenous glutamate had an inhibiting effect on pyruvate carboxylation, presumably by formation of oxaloacetate from 2-oxoglutarate derived from glutamate. It could be shown that glucose is a better substrate for energy production than glutamate; it is, however, less efficient in labeling amino acids than glutamate in cerebellar astrocytes.
Collapse
Affiliation(s)
- H Qu
- Department of Clinical Neuroscience, Faculty of Medicine, Norwegian University of Science and Technology, Olav Kyrresgt. 3, N-7489 Trondheim, Norway
| | | | | | | |
Collapse
|
49
|
Prothmann C, Wellard J, Berger J, Hamprecht B, Verleysdonk S. Primary cultures as a model for studying ependymal functions: glycogen metabolism in ependymal cells. Brain Res 2001; 920:74-83. [PMID: 11716813 DOI: 10.1016/s0006-8993(01)03021-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Ependymal cells form a single-layered, ciliated epithelium at the interface between the cerebrospinal fluid and the brain parenchyma. Although their morphology has been studied in detail, ependymal functions remain largely speculative. We have established and characterized a previously described cell culture model to investigate ependymal glycogen metabolism. During growth in minimal medium lacking many non-essential amino acids including L-glutamate, but containing glucose at physiological concentration, the cells contained negligible amounts of glycogen (7+/-3 nmol glucosyl residues/mg protein) despite the presence of insulin. However, during a period of 24 h, the cells accumulated glycogen to very high levels after transferal to a medium containing insulin, glucose at a 5-fold higher concentration, and all proteinogenic amino acids except L-asparagine and L-serine (990+/-112 nmol glucosyl residues/mg protein). Omission of insulin resulted in a 50% reduction in glycogen accumulation. Upon glucose deprivation, glycogen was degraded with a half-life of 21 min. The ependymal primary cultures contained 80+/-5 mU glycogen phosphorylase (Pho)/mg protein and stained positively with antibodies raised against this enzyme. Astroglial cultures built up less glycogen and had less Pho activity under identical conditions. Ependymal glycogen was mobilized by noradrenaline and serotonin. Our results indicate that ependymal cells maintain glycogen as a functional energy store, subject to rapid turnover dependent on the availability of energy substrates and the presence of appropriate signal molecules. Thus ependymocytes appear to be active players in the multitude of processes resulting in normal brain function, and ependymal primary cultures are suggested as a suitable model for studying the role of ependymal cells in these processes.
Collapse
Affiliation(s)
- C Prothmann
- Physiologisch-chemisches Institut der Universität, Hoppe-Seyler-Str. 4, D-72076, Tübingen, Germany
| | | | | | | | | |
Collapse
|
50
|
Béquet F, Gomez-Merino D, Berthelot M, Guezennec CY. Exercise-induced changes in brain glucose and serotonin revealed by microdialysis in rat hippocampus: effect of glucose supplementation. ACTA PHYSIOLOGICA SCANDINAVICA 2001; 173:223-30. [PMID: 11683680 DOI: 10.1046/j.1365-201x.2001.00859.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The aim of this study was to assess extracellular glucose changes in hippocampus in response to physical exercise and to determine the influence of glucose supplementation. In the same time, we have observed the changes in serotonin, in order to study the relationship between glucose and serotonin during exercise. Both glucose and serotonin were assessed using microdialysis. Exercise induced an increase in extracellular glucose levels over baseline during exercise to 121.1 +/- 3.0% (P < 0.001), then a decrease to baseline during recovery. The serotonin followed glucose changes during the first 90 min of exercise, but followed a different pattern during recovery, increasing to a maximum of 129.9 +/- 7.0% after 30 min of recovery (P < 0.001). When a 15% glucose solution was infused (10 microL x min(-1)) during exercise and recovery, blood glucose concentration was increased, but extracellular brain glucose decreased to reach a minimum of 73.3 +/- 4.6% after 90 min of recovery (P < 0.001). Serotonin was always the mirror-reflect of cerebral glucose, with a maximum increase of 142.0 +/- 6.9% after 90 min of recovery (P < 0.001). These results show that exercise induces changes in brain glucose and 5-hydroxytryptamine (5-HT) levels, which were dramatically modified by glucose infusion. Taking into account the implication of brain 5-HT in central fatigue, they suggest that if glucose supplementation, before and during exercise, undoubtedly increase performance because of its peripheral positive action, it would have a negative impact on the quality of recovery after the end of the exercise.
Collapse
Affiliation(s)
- F Béquet
- Department of Physiology, IMASSA, Brétigny-sur-Orge Cedex, France
| | | | | | | |
Collapse
|