1
|
Dienel GA, Rothman DL. In vivo calibration of genetically encoded metabolite biosensors must account for metabolite metabolism during calibration and cellular volume. J Neurochem 2024; 168:506-532. [PMID: 36726217 DOI: 10.1111/jnc.15775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/21/2023] [Accepted: 01/28/2023] [Indexed: 02/03/2023]
Abstract
Isotopic assays of brain glucose utilization rates have been used for more than four decades to establish relationships between energetics, functional activity, and neurotransmitter cycling. Limitations of these methods include the relatively long time (1-60 min) for the determination of labeled metabolite levels and the lack of cellular resolution. Identification and quantification of fuels for neurons and astrocytes that support activation and higher brain functions are a major, unresolved issues. Glycolysis is preferentially up-regulated during activation even though oxygen level and supply are adequate, causing lactate concentrations to quickly rise during alerting, sensory processing, cognitive tasks, and memory consolidation. However, the fate of lactate (rapid release from brain or cell-cell shuttling coupled with local oxidation) is long disputed. Genetically encoded biosensors can determine intracellular metabolite concentrations and report real-time lactate level responses to sensory, behavioral, and biochemical challenges at the cellular level. Kinetics and time courses of cellular lactate concentration changes are informative, but accurate biosensor calibration is required for quantitative comparisons of lactate levels in astrocytes and neurons. An in vivo calibration procedure for the Laconic lactate biosensor involves intracellular lactate depletion by intravenous pyruvate-mediated trans-acceleration of lactate efflux followed by sensor saturation by intravenous infusion of high doses of lactate plus ammonium chloride. In the present paper, the validity of this procedure is questioned because rapid lactate-pyruvate interconversion in blood, preferential neuronal oxidation of both monocarboxylates, on-going glycolytic metabolism, and cellular volumes were not taken into account. Calibration pitfalls for the Laconic lactate biosensor also apply to other metabolite biosensors that are standardized in vivo by infusion of substrates that can be metabolized in peripheral tissues. We discuss how technical shortcomings negate the conclusion that Laconic sensor calibrations support the existence of an in vivo astrocyte-neuron lactate concentration gradient linked to lactate shuttling from astrocytes to neurons to fuel neuronal activity.
Collapse
Affiliation(s)
- Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Douglas L Rothman
- Magnetic Resonance Research Center and Departments of Radiology and Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
2
|
Dienel GA, Schousboe A, McKenna MC, Rothman DL. A tribute to Leif Hertz: The historical context of his pioneering studies of the roles of astrocytes in brain energy metabolism, neurotransmission, cognitive functions, and pharmacology identifies important, unresolved topics for future studies. J Neurochem 2024; 168:461-495. [PMID: 36928655 DOI: 10.1111/jnc.15812] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Leif Hertz, M.D., D.Sc. (honōris causā) (1930-2018), was one of the original and noteworthy participants in the International Conference on Brain Energy Metabolism (ICBEM) series since its inception in 1993. The biennial ICBEM conferences are organized by neuroscientists interested in energetics and metabolism underlying neural functions; they have had a high impact on conceptual and experimental advances in these fields and on promoting collaborative interactions among neuroscientists. Leif made major contributions to ICBEM discussions and understanding of metabolic and signaling characteristics of astrocytes and their roles in brain function. His studies ranged from uptake of K+ from extracellular fluid and its stimulation of astrocytic respiration, identification, and regulation of enzymes specifically or preferentially expressed in astrocytes in the glutamate-glutamine cycle of excitatory neurotransmission, a requirement for astrocytic glycogenolysis for fueling K+ uptake, involvement of glycogen in memory consolidation in the chick, and pharmacology of astrocytes. This tribute to Leif Hertz highlights his major discoveries, the high impact of his work on astrocyte-neuron interactions, and his unparalleled influence on understanding the cellular basis of brain energy metabolism. His work over six decades has helped integrate the roles of astrocytes into neurotransmission where oxidative and glycogenolytic metabolism during neurotransmitter glutamate turnover are key aspects of astrocytic energetics. Leif recognized that brain astrocytic metabolism is greatly underestimated unless the volume fraction of astrocytes is taken into account. Adjustment for pathway rates expressed per gram tissue for volume fraction indicates that astrocytes have much higher oxidative rates than neurons and astrocytic glycogen concentrations and glycogenolytic rates during sensory stimulation in vivo are similar to those in resting and exercising muscle, respectively. These novel insights are typical of Leif's astute contributions to the energy metabolism field, and his publications have identified unresolved topics that provide the neuroscience community with challenges and opportunities for future research.
Collapse
Affiliation(s)
- Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA
- Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico, 87131, USA
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Mary C McKenna
- Department of Pediatrics and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, 21201, USA
| | - Douglas L Rothman
- Department of Radiology, Magnetic Resonance Research Center (MRRC), Yale University, New Haven, Connecticut, 06520, USA
| |
Collapse
|
3
|
Matt RA, Martin RS, Evans AK, Gever JR, Vargas GA, Shamloo M, Ford AP. Locus Coeruleus and Noradrenergic Pharmacology in Neurodegenerative Disease. Handb Exp Pharmacol 2024; 285:555-616. [PMID: 37495851 DOI: 10.1007/164_2023_677] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Adrenoceptors (ARs) throughout the brain are stimulated by noradrenaline originating mostly from neurons of the locus coeruleus, a brainstem nucleus that is ostensibly the earliest to show detectable pathology in neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. The α1-AR, α2-AR, and β-AR subtypes expressed in target brain regions and on a range of cell populations define the physiological responses to noradrenaline, which includes activation of cognitive function in addition to modulation of neurometabolism, cerebral blood flow, and neuroinflammation. As these heterocellular functions are critical for maintaining brain homeostasis and neuronal health, combating the loss of noradrenergic tone from locus coeruleus degeneration may therefore be an effective treatment for both cognitive symptoms and disease modification in neurodegenerative indications. Two pharmacologic approaches are receiving attention in recent clinical studies: preserving noradrenaline levels (e.g., via reuptake inhibition) and direct activation of target adrenoceptors. Here, we review the expression and role of adrenoceptors in the brain, the preclinical studies which demonstrate that adrenergic stimulation can support cognitive function and cerebral health by reversing the effects of noradrenaline depletion, and the human data provided by pharmacoepidemiologic analyses and clinical trials which together identify adrenoceptors as promising targets for the treatment of neurodegenerative disease.
Collapse
Affiliation(s)
| | | | - Andrew K Evans
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | | | | | - Mehrdad Shamloo
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | | |
Collapse
|
4
|
Patani R, Hardingham GE, Liddelow SA. Functional roles of reactive astrocytes in neuroinflammation and neurodegeneration. Nat Rev Neurol 2023; 19:395-409. [PMID: 37308616 DOI: 10.1038/s41582-023-00822-1] [Citation(s) in RCA: 102] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/14/2023]
Abstract
Despite advances in uncovering the mechanisms that underlie neuroinflammation and neurodegenerative disease, therapies that prevent neuronal loss remain elusive. Targeting of disease-defining markers in conditions such as Alzheimer disease (amyloid-β and tau) or Parkinson disease (α-synuclein) has been met with limited success, suggesting that these proteins do not act in isolation but form part of a pathological network. This network could involve phenotypic alteration of multiple cell types in the CNS, including astrocytes, which have a major neurosupportive, homeostatic role in the healthy CNS but adopt reactive states under acute or chronic adverse conditions. Transcriptomic studies in human patients and disease models have revealed the co-existence of many putative reactive sub-states of astrocytes. Inter-disease and even intra-disease heterogeneity of reactive astrocytic sub-states are well established, but the extent to which specific sub-states are shared across different diseases is unclear. In this Review, we highlight how single-cell and single-nuclei RNA sequencing and other 'omics' technologies can enable the functional characterization of defined reactive astrocyte states in various pathological scenarios. We provide an integrated perspective, advocating cross-modal validation of key findings to define functionally important sub-states of astrocytes and their triggers as tractable therapeutic targets with cross-disease relevance.
Collapse
Affiliation(s)
- Rickie Patani
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, Human Stem Cells and Neurodegeneration Laboratory, London, UK
| | - Giles E Hardingham
- Euan MacDonald Centre for MND, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Neuroscience & Physiology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA.
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
5
|
Barros LF, Ruminot I, Sotelo-Hitschfeld T, Lerchundi R, Fernández-Moncada I. Metabolic Recruitment in Brain Tissue. Annu Rev Physiol 2023; 85:115-135. [PMID: 36270291 DOI: 10.1146/annurev-physiol-021422-091035] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Information processing imposes urgent metabolic demands on neurons, which have negligible energy stores and restricted access to fuel. Here, we discuss metabolic recruitment, the tissue-level phenomenon whereby active neurons harvest resources from their surroundings. The primary event is the neuronal release of K+ that mirrors workload. Astrocytes sense K+ in exquisite fashion thanks to their unique coexpression of NBCe1 and α2β2 Na+/K+ ATPase, and within seconds switch to Crabtree metabolism, involving GLUT1, aerobic glycolysis, transient suppression of mitochondrial respiration, and lactate export. The lactate surge serves as a secondary recruiter by inhibiting glucose consumption in distant cells. Additional recruiters are glutamate, nitric oxide, and ammonium, which signal over different spatiotemporal domains. The net outcome of these events is that more glucose, lactate, and oxygen are made available. Metabolic recruitment works alongside neurovascular coupling and various averaging strategies to support the inordinate dynamic range of individual neurons.
Collapse
Affiliation(s)
- L F Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile; .,Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile;
| | - I Ruminot
- Centro de Estudios Científicos (CECs), Valdivia, Chile; .,Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile;
| | - T Sotelo-Hitschfeld
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - R Lerchundi
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), MIRCen, Fontenay-aux-Roses, France
| | - I Fernández-Moncada
- NeuroCentre Magendie, INSERM U1215, University of Bordeaux, Bordeaux, France
| |
Collapse
|
6
|
Natsubori A, Hirai S, Kwon S, Ono D, Deng F, Wan J, Miyazawa M, Kojima T, Okado H, Karashima A, Li Y, Tanaka KF, Honda M. Serotonergic neurons control cortical neuronal intracellular energy dynamics by modulating astrocyte-neuron lactate shuttle. iScience 2023; 26:105830. [PMID: 36713262 PMCID: PMC9881222 DOI: 10.1016/j.isci.2022.105830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 08/15/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
The central serotonergic system has multiple roles in animal physiology and behavior, including sleep-wake control. However, its function in controlling brain energy metabolism according to the state of animals remains undetermined. Through in vivo monitoring of energy metabolites and signaling, we demonstrated that optogenetic activation of raphe serotonergic neurons increased cortical neuronal intracellular concentration of ATP, an indispensable cellular energy molecule, which was suppressed by inhibiting neuronal uptake of lactate derived from astrocytes. Raphe serotonergic neuronal activation induced cortical astrocytic Ca2+ and cAMP surges and increased extracellular lactate concentrations, suggesting the facilitation of lactate release from astrocytes. Furthermore, chemogenetic inhibition of raphe serotonergic neurons partly attenuated the increase in cortical neuronal intracellular ATP levels as arousal increased in mice. Serotonergic neuronal activation promoted an increase in cortical neuronal intracellular ATP levels, partly mediated by the facilitation of the astrocyte-neuron lactate shuttle, contributing to state-dependent optimization of neuronal intracellular energy levels.
Collapse
Affiliation(s)
- Akiyo Natsubori
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan,Corresponding author
| | - Shinobu Hirai
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Soojin Kwon
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Daisuke Ono
- Department of Neuroscience Ⅱ, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Fei Deng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China,PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Jinxia Wan
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China,PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Momoka Miyazawa
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan,Faculty of Science Division Ⅱ, Tokyo University of Science, Shinjuku-ku, Tokyo 162-8601, Japan
| | - Takashi Kojima
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Haruo Okado
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Akihiro Karashima
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, Sendai 982-8577, Japan
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China,PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Kenji F. Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Makoto Honda
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| |
Collapse
|
7
|
Affiliation(s)
- Gerald A. Dienel
- Department of Neurology University of Arkansas for Medical Sciences Little Rock Arkansas USA
- Department of Cell Biology and Physiology University of New Mexico School of Medicine Albuquerque New Mexico USA
| | - Lisa Gillinder
- Mater Hospital South Brisbane Queensland Australia
- Faculty of Medicine Mater Research Institute, University of Queensland St Lucia Queensland Australia
| | - Aileen McGonigal
- Mater Hospital South Brisbane Queensland Australia
- Faculty of Medicine Mater Research Institute, University of Queensland St Lucia Queensland Australia
| | - Karin Borges
- Faculty of Medicine School of Biomedical Sciences, University of Queensland St Lucia Queensland Australia
| |
Collapse
|
8
|
Dienel GA, Gillinder L, McGonigal A, Borges K. Potential new roles for glycogen in epilepsy. Epilepsia 2023; 64:29-53. [PMID: 36117414 PMCID: PMC10952408 DOI: 10.1111/epi.17412] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/15/2022] [Accepted: 09/15/2022] [Indexed: 01/21/2023]
Abstract
Seizures often originate in epileptogenic foci. Between seizures (interictally), these foci and some of the surrounding tissue often show low signals with 18 fluorodeoxyglucose (FDG) positron emission tomography (PET) in many epileptic patients, even when there are no radiologically detectable structural abnormalities. Low FDG-PET signals are thought to reflect glucose hypometabolism. Here, we review knowledge about metabolism of glucose and glycogen and oxidative stress in people with epilepsy and in acute and chronic rodent seizure models. Interictal brain glucose levels are normal and do not cause apparent glucose hypometabolism, which remains unexplained. During seizures, high amounts of fuel are needed to satisfy increased energy demands. Astrocytes consume glycogen as an additional emergency fuel to supplement glucose during high metabolic demand, such as during brain stimulation, stress, and seizures. In rodents, brain glycogen levels drop during induced seizures and increase to higher levels thereafter. Interictally, in people with epilepsy and in chronic epilepsy models, normal glucose but high glycogen levels have been found in the presumed brain areas involved in seizure generation. We present our new hypothesis that as an adaptive response to repeated episodes of high metabolic demand, high interictal glycogen levels in epileptogenic brain areas are used to support energy metabolism and potentially interictal neuronal activity. Glycogenolysis, which can be triggered by stress or oxidative stress, leads to decreased utilization of plasma glucose in epileptogenic brain areas, resulting in low FDG signals that are related to functional changes underlying seizure onset and propagation. This is (partially) reversible after successful surgery. Last, we propose that potential interictal glycogen depletion in epileptogenic and surrounding areas may cause energy shortages in astrocytes, which may impair potassium buffering and contribute to seizure generation. Based on these hypotheses, auxiliary fuels or treatments that support glycogen metabolism may be useful to treat epilepsy.
Collapse
Affiliation(s)
- Gerald A. Dienel
- Department of NeurologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
- Department of Cell Biology and PhysiologyUniversity of New Mexico School of MedicineAlbuquerqueNew MexicoUSA
| | - Lisa Gillinder
- Mater HospitalSouth BrisbaneQueenslandAustralia
- Faculty of MedicineMater Research Institute, University of QueenslandSt LuciaQueenslandAustralia
| | - Aileen McGonigal
- Mater HospitalSouth BrisbaneQueenslandAustralia
- Faculty of MedicineMater Research Institute, University of QueenslandSt LuciaQueenslandAustralia
| | - Karin Borges
- Faculty of MedicineSchool of Biomedical Sciences, University of QueenslandSt LuciaQueenslandAustralia
| |
Collapse
|
9
|
Alhamyani A, Napit PR, Bheemanapally K, Ibrahim MMH, Sylvester PW, Briski KP. Glycogen phosphorylase isoform regulation of glucose and energy sensor expression in male versus female rat hypothalamic astrocyte primary cultures. Mol Cell Endocrinol 2022; 553:111698. [PMID: 35718260 PMCID: PMC9332090 DOI: 10.1016/j.mce.2022.111698] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 11/21/2022]
Abstract
Astrocyte glycogen constitutes the primary energy fuel reserve in the brain. Current research investigated the novel premise that glycogen turnover governs astrocyte responsiveness to critical metabolic and neurotransmitter (norepinephrine) regulatory signals in a sex-dimorphic manner. Here, rat hypothalamic astrocyte glycogen phosphorylase (GP) gene expression was silenced by short-interfering RNA (siRNA) to investigate how glycogen metabolism controlled by GP-brain type (GPbb) or GP-muscle type (GPmm) activity affects glucose [glucose transporter-2 (GLUT2)] and energy [5'-AMP-activated protein kinase (AMPK)] sensor and adrenergic receptor (AR) proteins in each sex. Results show that in the presence of glucose, glycogen turnover is regulated by GPbb in the male or by GPmm in the female, yet in the absence of glucose, glycogen breakdown is controlled by GPbb in each sex. GLUT2 expression is governed by GPmm-mediated glycogen breakdown in glucose-supplied astrocytes of each sex, but glycogenolysis controls glucoprivic GLUT2 up-regulation in male only. GPbb-mediated glycogen disassembly causes divergent changes in total AMPK versus phosphoAMPK profiles in male. During glucoprivation, glycogenolysis up-regulates AMPK content in male astrocytes by GPbb- and GPmm-dependent mechanisms, whereas GPbb-mediated glycogen breakdown inhibits phosphoAMPK expression in female. GPbb and GPmm activity governs alpha2-AR and beta1-AR protein levels in male, but has no effect on these profiles in the female. Outcomes provide novel evidence for sex-specific glycogen regulation of glucose- and energy-sensory protein expression in hypothalamic astrocytes, and identify GP isoforms that mediate such control in each sex. Results also show that glycogen regulation of hypothalamic astrocyte receptivity to norepinephrine is male-specific. Further studies are needed to characterize the molecular mechanisms that underlie sex differences in glycogen control of astrocyte protein expression.
Collapse
Affiliation(s)
- Abdulrahman Alhamyani
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, 71201, USA
| | - Prabhat R Napit
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, 71201, USA
| | - Khaggeswar Bheemanapally
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, 71201, USA
| | - Mostafa M H Ibrahim
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, 71201, USA
| | - Paul W Sylvester
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, 71201, USA
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, 71201, USA.
| |
Collapse
|
10
|
Zhang M, Wang Y, Bai Y, Dai L, Guo H. Monocarboxylate Transporter 1 May Benefit Cerebral Ischemia via Facilitating Lactate Transport From Glial Cells to Neurons. Front Neurol 2022; 13:781063. [PMID: 35547368 PMCID: PMC9081727 DOI: 10.3389/fneur.2022.781063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
Monocarboxylate transporter 1 (MCT1) is expressed in glial cells and some populations of neurons. MCT1 facilitates astrocytes or oligodendrocytes (OLs) in the energy supplement of neurons, which is crucial for maintaining the neuronal activity and axonal function. It is suggested that MCT1 upregulation in cerebral ischemia is protective to ischemia/reperfusion (I/R) injury. Otherwise, its underlying mechanism has not been clearly discussed. In this review, it provides a novel insight that MCT1 may protect brain from I/R injury via facilitating lactate transport from glial cells (such as, astrocytes and OLs) to neurons. It extensively discusses (1) the structure and localization of MCT1; (2) the regulation of MCT1 in lactate transport among astrocytes, OLs, and neurons; and (3) the regulation of MCT1 in the cellular response of lactate accumulation under ischemic attack. At last, this review concludes that MCT1, in cerebral ischemia, may improve lactate transport from glial cells to neurons, which subsequently alleviates cellular damage induced by lactate accumulation (mostly in glial cells), and meets the energy metabolism of neurons.
Collapse
Affiliation(s)
- Mao Zhang
- Department of Medical Genetics, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Yanyan Wang
- Department of Medical Genetics, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Yun Bai
- Department of Medical Genetics, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Limeng Dai
- Department of Medical Genetics, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Hong Guo
- Department of Medical Genetics, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| |
Collapse
|
11
|
Napoli E, Panoutsopoulos AA, Kysar P, Satriya N, Sterling K, Shibata B, Imai D, Ruskin DN, Zarbalis KS, Giulivi C. Wdfy3 regulates glycophagy, mitophagy, and synaptic plasticity. J Cereb Blood Flow Metab 2021; 41:3213-3231. [PMID: 34187232 PMCID: PMC8669292 DOI: 10.1177/0271678x211027384] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Autophagy is essential to cell function, as it enables the recycling of intracellular constituents during starvation and in addition functions as a quality control mechanism by eliminating spent organelles and proteins that could cause cellular damage if not properly removed. Recently, we reported on Wdfy3's role in mitophagy, a clinically relevant macroautophagic scaffold protein that is linked to intellectual disability, neurodevelopmental delay, and autism spectrum disorder. In this study, we confirm our previous report that Wdfy3 haploinsufficiency in mice results in decreased mitophagy with accumulation of mitochondria with altered morphology, but expanding on that observation, we also note decreased mitochondrial localization at synaptic terminals and decreased synaptic density, which may contribute to altered synaptic plasticity. These changes are accompanied by defective elimination of glycogen particles and a shift to increased glycogen synthesis over glycogenolysis and glycophagy. This imbalance leads to an age-dependent higher incidence of brain glycogen deposits with cerebellar hypoplasia. Our results support and further extend Wdfy3's role in modulating both brain bioenergetics and synaptic plasticity by including glycogen as a target of macroautophagic degradation.
Collapse
Affiliation(s)
- Eleonora Napoli
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Alexios A Panoutsopoulos
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA
| | - Patricia Kysar
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, CA, USA
| | - Nathaniel Satriya
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Kira Sterling
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Bradley Shibata
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, CA, USA
| | - Denise Imai
- Anatomic Pathology Service, Veterinary Medical Teaching Hospital, University of California, Davis, CA, USA
| | - David N Ruskin
- Department of Psychology and Neuroscience Program, Trinity College, Hartford, CT, USA
| | - Konstantinos S Zarbalis
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA.,Medical Investigations of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, CA, USA
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA.,Medical Investigations of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, CA, USA
| |
Collapse
|
12
|
Horvat A, Zorec R, Vardjan N. Lactate as an Astroglial Signal Augmenting Aerobic Glycolysis and Lipid Metabolism. Front Physiol 2021; 12:735532. [PMID: 34658920 PMCID: PMC8514727 DOI: 10.3389/fphys.2021.735532] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/06/2021] [Indexed: 01/16/2023] Open
Abstract
Astrocytes, heterogeneous neuroglial cells, contribute to metabolic homeostasis in the brain by providing energy substrates to neurons. In contrast to predominantly oxidative neurons, astrocytes are considered primarily as glycolytic cells. They take up glucose from the circulation and in the process of aerobic glycolysis (despite the normal oxygen levels) produce L-lactate, which is then released into the extracellular space via lactate transporters and possibly channels. Astroglial L-lactate can enter neurons, where it is used as a metabolic substrate, or exit the brain via the circulation. Recently, L-lactate has also been considered to be a signaling molecule in the brain, but the mechanisms of L-lactate signaling and how it contributes to the brain function remain to be fully elucidated. Here, we provide an overview of L-lactate signaling mechanisms in the brain and present novel insights into the mechanisms of L-lactate signaling via G-protein coupled receptors (GPCRs) with the focus on astrocytes. We discuss how increased extracellular L-lactate upregulates cAMP production in astrocytes, most likely viaL-lactate-sensitive Gs-protein coupled GPCRs. This activates aerobic glycolysis, enhancing L-lactate production and accumulation of lipid droplets, suggesting that L-lactate augments its own production in astrocytes (i.e., metabolic excitability) to provide more L-lactate for neurons and that astrocytes in conditions of increased extracellular L-lactate switch to lipid metabolism.
Collapse
Affiliation(s)
- Anemari Horvat
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| | - Nina Vardjan
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| |
Collapse
|
13
|
Ibrahim MMH, Bheemanapally K, Sylvester PW, Briski KP. Norepinephrine Regulation of Adrenergic Receptor Expression, 5' AMP-Activated Protein Kinase Activity, and Glycogen Metabolism and Mass in Male Versus Female Hypothalamic Primary Astrocyte Cultures. ASN Neuro 2021; 12:1759091420974134. [PMID: 33176438 PMCID: PMC7672765 DOI: 10.1177/1759091420974134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Norepinephrine (NE) control of hypothalamic gluco-regulation involves astrocyte-derived energy fuel supply. In male rats, exogenous NE regulates astrocyte glycogen metabolic enzyme expression in vivo through 5’-AMP-activated protein kinase (AMPK)-dependent mechanisms. Current research utilized a rat hypothalamic astrocyte primary culture model to investigate the premise that NE imposes sex-specific direct control of AMPK activity and glycogen mass and metabolism in these glia. In male rats, NE down-regulation of pAMPK correlates with decreased CaMMKB and increased PP1 expression, whereas noradrenergic augmentation of female astrocyte pAMPK may not involve these upstream regulators. NE concentration is a critical determinant of control of hypothalamic astrocyte glycogen enzyme expression, but efficacy varies between sexes. Data show sex variations in glycogen synthase expression and glycogen phosphorylase-brain and –muscle type dose-responsiveness to NE. Narrow dose-dependent NE augmentation of astrocyte glycogen content during energy homeostasis infers that NE maintains, over a broad exposure range, constancy of glycogen content despite possible changes in turnover. In male rats, beta1- and beta2-adrenergic receptor (AR) profiles displayed bi-directional responses to increasing NE doses; female astrocytes exhibited diminished beta1-AR content at low dose exposure, but enhanced beta2-AR expression at high NE dosages. Thus, graded variations in noradrenergic stimulation may modulate astrocyte receptivity to NE in vivo. Sex dimorphic NE regulation of hypothalamic astrocyte AMPK activation and glycogen metabolism may be mediated, in part, by one or more ARs characterized here by divergent sensitivity to this transmitter.
Collapse
Affiliation(s)
- Mostafa M H Ibrahim
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, United States
| | - Khaggeswar Bheemanapally
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, United States
| | - Paul W Sylvester
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, United States
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, United States
| |
Collapse
|
14
|
Byman E, Martinsson I, Haukedal H, Gouras G, Freude KK, Wennström M. Neuronal α-amylase is important for neuronal activity and glycogenolysis and reduces in presence of amyloid beta pathology. Aging Cell 2021; 20:e13433. [PMID: 34261192 PMCID: PMC8373367 DOI: 10.1111/acel.13433] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/23/2021] [Accepted: 07/03/2021] [Indexed: 12/28/2022] Open
Abstract
Recent studies indicate a crucial role for neuronal glycogen storage and degradation in memory formation. We have previously identified alpha-amylase (α-amylase), a glycogen degradation enzyme, located within synaptic-like structures in CA1 pyramidal neurons and shown that individuals with a high copy number variation of α-amylase perform better on the episodic memory test. We reported that neuronal α-amylase was absent in patients with Alzheimer's disease (AD) and that this loss corresponded to increased AD pathology. In the current study, we verified these findings in a larger patient cohort and determined a similar reduction in α-amylase immunoreactivity in the molecular layer of hippocampus in AD patients. Next, we demonstrated reduced α-amylase concentrations in oligomer amyloid beta 42 (Aβ42 ) stimulated SH-SY5Y cells and neurons derived from human-induced pluripotent stem cells (hiPSC) with PSEN1 mutation. Reduction of α-amylase production and activity, induced by siRNA and α-amylase inhibitor Tendamistat, respectively, was further shown to enhance glycogen load in SH-SY5Y cells. Both oligomer Aβ42 stimulated SH-SY5Y cells and hiPSC neurons with PSEN1 mutation showed, however, reduced load of glycogen. Finally, we demonstrate the presence of α-amylase within synapses of isolated primary neurons and show that inhibition of α-amylase activity with Tendamistat alters neuronal activity measured by calcium imaging. In view of these findings, we hypothesize that α-amylase has a glycogen degrading function within synapses, potentially important in memory formation. Hence, a loss of α-amylase, which can be induced by Aβ pathology, may in part underlie the disrupted memory formation seen in AD patients.
Collapse
Affiliation(s)
- Elin Byman
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöLund UniversityMalmöSweden
| | - Isak Martinsson
- Experimental Dementia Research UnitDepartment of Experimental Medical ScienceBMC B11Lund UniversityLundSweden
| | - Henriette Haukedal
- Department of Veterinary and Animal SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenFrederiksbergDenmark
| | | | - Gunnar Gouras
- Experimental Dementia Research UnitDepartment of Experimental Medical ScienceBMC B11Lund UniversityLundSweden
| | - Kristine K. Freude
- Department of Veterinary and Animal SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenFrederiksbergDenmark
| | - Malin Wennström
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöLund UniversityMalmöSweden
| |
Collapse
|
15
|
van Putten MJ, Fahlke C, Kafitz KW, Hofmeijer J, Rose CR. Dysregulation of Astrocyte Ion Homeostasis and Its Relevance for Stroke-Induced Brain Damage. Int J Mol Sci 2021; 22:5679. [PMID: 34073593 PMCID: PMC8198632 DOI: 10.3390/ijms22115679] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke is a leading cause of mortality and chronic disability. Either recovery or progression towards irreversible failure of neurons and astrocytes occurs within minutes to days, depending on remaining perfusion levels. Initial damage arises from energy depletion resulting in a failure to maintain homeostasis and ion gradients between extra- and intracellular spaces. Astrocytes play a key role in these processes and are thus central players in the dynamics towards recovery or progression of stroke-induced brain damage. Here, we present a synopsis of the pivotal functions of astrocytes at the tripartite synapse, which form the basis of physiological brain functioning. We summarize the evidence of astrocytic failure and its consequences under ischemic conditions. Special emphasis is put on the homeostasis and stroke-induced dysregulation of the major monovalent ions, namely Na+, K+, H+, and Cl-, and their involvement in maintenance of cellular volume and generation of cerebral edema.
Collapse
Affiliation(s)
- Michel J.A.M. van Putten
- Department of Clinical Neurophysiology, University of Twente, 7522 NB Enschede, The Netherlands; (M.J.A.M.v.P.); (J.H.)
| | - Christoph Fahlke
- Institut für Biologische Informationsprozesse, Molekular-und Zellphysiologie (IBI-1), Forschungszentrum Jülich, 52425 Jülich, Germany;
| | - Karl W. Kafitz
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Jeannette Hofmeijer
- Department of Clinical Neurophysiology, University of Twente, 7522 NB Enschede, The Netherlands; (M.J.A.M.v.P.); (J.H.)
| | - Christine R. Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| |
Collapse
|
16
|
Horvat A, Muhič M, Smolič T, Begić E, Zorec R, Kreft M, Vardjan N. Ca 2+ as the prime trigger of aerobic glycolysis in astrocytes. Cell Calcium 2021; 95:102368. [PMID: 33621899 DOI: 10.1016/j.ceca.2021.102368] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/17/2022]
Abstract
Astroglial aerobic glycolysis, a process during which d-glucose is converted to l-lactate, a brain fuel and signal, is regulated by the plasmalemmal receptors, including adrenergic receptors (ARs) and purinergic receptors (PRs), modulating intracellular Ca2+ and cAMP signals. However, the extent to which the two signals regulate astroglial aerobic glycolysis is poorly understood. By using agonists to stimulate intracellular α1-/β-AR-mediated Ca2+/cAMP signals, β-AR-mediated cAMP and P2R-mediated Ca2+ signals and genetically encoded fluorescence resonance energy transfer-based glucose and lactate nanosensors in combination with real-time microscopy, we show that intracellular Ca2+, but not cAMP, initiates a robust increase in the concentration of intracellular free d-glucose ([glc]i) and l-lactate ([lac]i), both depending on extracellular d-glucose, suggesting Ca2+-triggered glucose uptake and aerobic glycolysis in astrocytes. When the glycogen shunt, a process of glycogen remodelling, was inhibited, the α1-/β-AR-mediated increases in [glc]i and [lac]i were reduced by ∼65 % and ∼30 %, respectively, indicating that at least ∼30 % of the utilization of d-glucose is linked to glycogen remodelling and aerobic glycolysis. Additional activation of β-AR/cAMP signals aided to α1-/β-AR-triggered [lac]i increase, whereas the [glc]i increase was unaltered. Taken together, an increase in intracellular Ca2+ is the prime mechanism of augmented aerobic glycolysis in astrocytes, while cAMP has only a moderate role. The results provide novel information on the signals regulating brain metabolism and open new avenues to explore whether astroglial Ca2+ signals are dysregulated and contribute to neuropathologies with impaired brain metabolism.
Collapse
Affiliation(s)
- Anemari Horvat
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| | - Marko Muhič
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tina Smolič
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Ena Begić
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| | - Marko Kreft
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia; Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Nina Vardjan
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia.
| |
Collapse
|
17
|
Astroglial Connexin43 as a Potential Target for a Mood Stabiliser. Int J Mol Sci 2020; 22:ijms22010339. [PMID: 33396966 PMCID: PMC7795839 DOI: 10.3390/ijms22010339] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/24/2020] [Accepted: 12/27/2020] [Indexed: 02/06/2023] Open
Abstract
Mood disorders remain a major public health concern worldwide. Monoaminergic hypotheses of pathophysiology of bipolar and major depressive disorders have led to the development of monoamine transporter-inhibiting antidepressants for the treatment of major depression and have contributed to the expanded indications of atypical antipsychotics for the treatment of bipolar disorders. In spite of psychopharmacological progress, current pharmacotherapy according to the monoaminergic hypothesis alone is insufficient to improve or prevent mood disorders. Recent approval of esketamine for treatment of treatment-resistant depression has attracted attention in psychopharmacology as a glutamatergic hypothesis of the pathophysiology of mood disorders. On the other hand, in the last decade, accumulated findings regarding the pathomechanisms of mood disorders emphasised that functional abnormalities of tripartite synaptic transmission play important roles in the pathophysiology of mood disorders. At first glance, the enhancement of astroglial connexin seems to contribute to antidepressant and mood-stabilising effects, but in reality, antidepressive and mood-stabilising actions are mediated by more complicated interactions associated with the astroglial gap junction and hemichannel. Indeed, several depressive mood-inducing stress stimulations suppress connexin43 expression and astroglial gap junction function, but enhance astroglial hemichannel activity. On the other hand, monoamine transporter-inhibiting antidepressants suppress astroglial hemichannel activity and enhance astroglial gap junction function, whereas several non-antidepressant mood stabilisers activate astroglial hemichannel activity. Based on preclinical findings, in this review, we summarise the effects of antidepressants, mood-stabilising antipsychotics, and anticonvulsants on astroglial connexin, and then, to establish a novel strategy for treatment of mood disorders, we reveal the current progress in psychopharmacology, changing the question from "what has been revealed?" to "what should be clarified?".
Collapse
|
18
|
Hagenston AM, Bading H, Bas-Orth C. Functional Consequences of Calcium-Dependent Synapse-to-Nucleus Communication: Focus on Transcription-Dependent Metabolic Plasticity. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035287. [PMID: 31570333 DOI: 10.1101/cshperspect.a035287] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the nervous system, calcium signals play a major role in the conversion of synaptic stimuli into transcriptional responses. Signal-regulated gene transcription is fundamental for a range of long-lasting adaptive brain functions that include learning and memory, structural plasticity of neurites and synapses, acquired neuroprotection, chronic pain, and addiction. In this review, we summarize the diverse mechanisms governing calcium-dependent transcriptional regulation associated with central nervous system plasticity. We focus on recent advances in the field of synapse-to-nucleus communication that include studies of the signal-regulated transcriptome in human neurons, identification of novel regulatory mechanisms such as activity-induced DNA double-strand breaks, and the identification of novel forms of activity- and transcription-dependent adaptations, in particular, metabolic plasticity. We summarize the reciprocal interactions between different kinds of neuroadaptations and highlight the emerging role of activity-regulated epigenetic modifiers in gating the inducibility of signal-regulated genes.
Collapse
Affiliation(s)
- Anna M Hagenston
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Carlos Bas-Orth
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
19
|
Heller JP, Odii T, Zheng K, Rusakov DA. Imaging tripartite synapses using super-resolution microscopy. Methods 2020; 174:81-90. [PMID: 31153907 PMCID: PMC7144327 DOI: 10.1016/j.ymeth.2019.05.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/03/2019] [Accepted: 05/28/2019] [Indexed: 01/02/2023] Open
Abstract
Astroglia are vital facilitators of brain development, homeostasis, and metabolic support. In addition, they are also essential to the formation and regulation of synaptic circuits. Due to the extraordinary complex, nanoscopic morphology of astrocytes, the underlying cellular mechanisms have been poorly understood. In particular, fine astrocytic processes that can be found in the vicinity of synapses have been difficult to study using traditional imaging techniques. Here, we describe a 3D three-colour super-resolution microscopy approach to unravel the nanostructure of tripartite synapses. The method is based on the SMLM technique direct stochastic optical reconstruction microscopy (dSTORM) which uses conventional fluorophore-labelled antibodies. This approach enables reconstructing the nanoscale localisation of individual astrocytic glutamate transporter (GLT-1) molecules surrounding presynaptic (bassoon) and postsynaptic (Homer1) protein localisations in fixed mouse brain sections. However, the technique is readily adaptable to other types of targets and tissues.
Collapse
Affiliation(s)
- Janosch Peter Heller
- UCL Queen Square Institute of Neurology, University College London, London, United Kingdom; FutureNeuro Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland.
| | - Tuamoru Odii
- UCL Queen Square Institute of Neurology, University College London, London, United Kingdom; Department of Physiology, Faculty of Basic Medical Sciences, Alex Ekwueme Federal University Ndufu-Alike Ikwo, PMB 1010 Abakaliki, Nigeria
| | - Kaiyu Zheng
- UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Dmitri A Rusakov
- UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.
| |
Collapse
|
20
|
Li Z, Lu Y, Liang S, Li S, Chen B, Zhang M, Xia M, Guan D, Verkhratsky A, Li B. Fluoxetine improves behavioural deficits induced by chronic alcohol treatment by alleviating RNA editing of 5-HT 2C receptors. Neurochem Int 2020; 134:104689. [PMID: 31968217 DOI: 10.1016/j.neuint.2020.104689] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/07/2020] [Accepted: 01/15/2020] [Indexed: 12/13/2022]
Abstract
The alcoholism and major depressive disorder are common comorbidity, with alcohol-induced depressive symptoms being eased by selective serotonin re-uptake inhibitors (SSRIs), although the mechanisms underlying pathology and therapy are poorly understood. Chronic alcohol consumption affects the activity of serotonin 2C receptors (5-HT2CR) by regulating adenosine deaminases acting on RNA (ADARs) in neurons. Astrogliopathic changes contribute to alcohol addiction, while decreased release of ATP from astrocytes can trigger depressive-like behaviours in mice. In this study, we discovered that chronic alcohol treatment increased editing of RNA of 5-HT2CR via up-regulating the expression of ADAR2, consequently reducing the release of ATP from astrocytes induced by 5-HT2CR agonist, MK212. Moreover, SSRI antidepressant fluoxetine decreased the expression of ADAR2 through the transactivation of EGFR/PI3K/AKT/cFos signalling pathway. The increased release of astroglial ATP by MK212 which was suppressed by chronic alcohol consumption, and reduction in ADAR2 activity eliminated the RNA editing of 5-HT2CR increased by alcohol in vitro and recovered the release of ATP from astrocytes induced by MK212. Meanwhile, fluoxetine improved the behavioural and motor symptoms induced by alcohol addiction and decreased the alcohol intake. Our study suggests that the astrocytic 5-HT2CR contribute to alcohol addiction; fluoxetine thus can be used to alleviate depression, treat alcohol addiction and improve motor coordination.
Collapse
Affiliation(s)
- Zexiong Li
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Yan Lu
- Key Laboratory of Health Ministry in Congenital Malformation, The Affiliated Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Shanshan Liang
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Shuai Li
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Beina Chen
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Manman Zhang
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Maosheng Xia
- Department of Orthopaedics, The First Hospital, China Medical University, Shenyang, PR China
| | - Dawei Guan
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, PR China.
| | - Alexei Verkhratsky
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, PR China; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
| | - Baoman Li
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, PR China.
| |
Collapse
|
21
|
Stovell MG, Mada MO, Carpenter TA, Yan JL, Guilfoyle MR, Jalloh I, Welsh KE, Helmy A, Howe DJ, Grice P, Mason A, Giorgi-Coll S, Gallagher CN, Murphy MP, Menon DK, Hutchinson PJ, Carpenter KL. Phosphorus spectroscopy in acute TBI demonstrates metabolic changes that relate to outcome in the presence of normal structural MRI. J Cereb Blood Flow Metab 2020; 40:67-84. [PMID: 30226401 PMCID: PMC6927074 DOI: 10.1177/0271678x18799176] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Metabolic dysfunction is a key pathophysiological process in the acute phase of traumatic brain injury (TBI). Although changes in brain glucose metabolism and extracellular lactate/pyruvate ratio are well known, it was hitherto unknown whether these translate to downstream changes in ATP metabolism and intracellular pH. We have performed the first clinical voxel-based in vivo phosphorus magnetic resonance spectroscopy (31P MRS) in 13 acute-phase major TBI patients versus 10 healthy controls (HCs), at 3T, focusing on eight central 2.5 × 2.5 × 2.5 cm3 voxels per subject. PCr/γATP ratio (a measure of energy status) in TBI patients was significantly higher (median = 1.09) than that of HCs (median = 0.93) (p < 0.0001), due to changes in both PCr and ATP. There was no significant difference in PCr/γATP between TBI patients with favourable and unfavourable outcome. Cerebral intracellular pH of TBI patients was significantly higher (median = 7.04) than that of HCs (median = 7.00) (p = 0.04). Alkalosis was limited to patients with unfavourable outcome (median = 7.07) (p < 0.0001). These changes persisted after excluding voxels with > 5% radiologically visible injury. This is the first clinical demonstration of brain alkalosis and elevated PCr/γATP ratio acutely after major TBI. 31P MRS has potential for non-invasively assessing brain injury in the absence of structural injury, predicting outcome and monitoring therapy response.
Collapse
Affiliation(s)
- Matthew G Stovell
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Marius O Mada
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - T Adrian Carpenter
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Jiun-Lin Yan
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Department of Neurosurgery, Keelung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Mathew R Guilfoyle
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Ibrahim Jalloh
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Karen E Welsh
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Adel Helmy
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Duncan J Howe
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Peter Grice
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Andrew Mason
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Susan Giorgi-Coll
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Clare N Gallagher
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Division of Neurosurgery, Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - David K Menon
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Peter J Hutchinson
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Keri Lh Carpenter
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
22
|
Rönnbäck C, Hansson E. The Importance and Control of Low-Grade Inflammation Due to Damage of Cellular Barrier Systems That May Lead to Systemic Inflammation. Front Neurol 2019; 10:533. [PMID: 31191433 PMCID: PMC6549124 DOI: 10.3389/fneur.2019.00533] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/03/2019] [Indexed: 01/04/2023] Open
Abstract
Systemic low-grade inflammation can be initiated in vivo after traumatic injury or in chronic diseases such as neurodegenerative, metabolic, and autoimmune diseases. Inducers of inflammation trigger production of inflammatory mediators, which alter the functionality of tissues and organs and leads to harmful induction of different barrier systems in the body, where the blood-brain barrier, the blood-retinal barrier, blood-nerve barrier, blood-lymph barrier and the blood-cerebrospinal fluid barrier play major roles. The different barriers are unique but structured in a similar way. They are equipped with sophisticated junctional complexes where different connexins, protein subunits of gap junction channels and hemichannels, constitute important partners. The cells involved in the various barriers are coupled in networks, are excitable but do not express action potentials and may be targets for inflammation leading to changes in several biochemical cellular parameters. During any type of inflammation barrier break-down is observed where any form of injury can start with low-grade inflammation and may lead to systemic inflammation.
Collapse
Affiliation(s)
- Cecilia Rönnbäck
- Department of Ophthalmology, Rigshospitalet, Glostrup, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Elisabeth Hansson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
23
|
Shah M, Addison A, Wang P, Zhu W, Chan O. Recurrent glucose deprivation leads to the preferential use of lactate by neurons in the ventromedial hypothalamus. Am J Physiol Endocrinol Metab 2019; 316:E948-E955. [PMID: 30888861 PMCID: PMC6580165 DOI: 10.1152/ajpendo.00468.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Increased GABAergic output in the ventromedial hypothalamus (VMH) contributes to counterregulatory failure in recurrently hypoglycemic (RH) rats, and lactate, an alternate fuel source in the brain, contributes to this phenomenon. The current study assessed whether recurring bouts of glucose deprivation enhanced neuronal lactate uptake and, if so, whether this influenced γ-aminobutyric acid (GABA) output and the counterregulatory responses. Glucose deprivation was induced using 5-thioglucose (5TG). Control rats received an infusion of artificial extracellular fluid. These groups were compared with RH animals. Subsequently, the rats underwent a hypoglycemic clamp with microdialysis. To test whether 5TG affected neuronal lactate utilization, a subgroup of 5TG-treated rats was microinjected with a lactate transporter inhibitor [cyano-4-hydroxycinnamate (4CIN)] just before the start of the clamp. Both RH and 5TG raised VMH GABA levels, and this was associated with impaired counterregulatory responses. 4CIN reduced VMH GABA levels and restored the hormone responses in the 5TG group. We then evaluated [14C]lactate uptake in hypothalamic neuronal cultures. Recurring exposure to low glucose increased monocarboxylate transporter-2 mRNA expression and augmented lactate uptake. Taken together, our data suggest that glucose deprivation, per se, enhances lactate utilization in hypothalamic neurons, and this may contribute to suppression of the counterregulatory responses to hypoglycemia.
Collapse
Affiliation(s)
- Maitreyee Shah
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine , New Haven, Connecticut
| | - Augustina Addison
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine , New Haven, Connecticut
| | - Peili Wang
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine , New Haven, Connecticut
| | - Wanling Zhu
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine , New Haven, Connecticut
| | - Owen Chan
- Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, University of Utah School of Medicine , Salt Lake City, Utah
| |
Collapse
|
24
|
Souza DG, Almeida RF, Souza DO, Zimmer ER. The astrocyte biochemistry. Semin Cell Dev Biol 2019; 95:142-150. [PMID: 30951895 DOI: 10.1016/j.semcdb.2019.04.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 03/19/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023]
Abstract
Astrocytes are a unique and dynamic subtype of glial cells in the central nervous system (CNS). Understanding their biochemical reactions and their influence in the surrounding cells is extremely important in the neuroscience field. They exert important influence in the neurotransmission, ionic homeostasis and also release neuroactive molecules termed gliotransmitters. Additionally, they metabolize, store and release metabolic substrates to meet high brain energy requirements. In this review, we highlight the main biochemical reactions regarding energy metabolism that take place in astrocytes. Special attention is given to synthesis, storage and catabolism of glucose, release of lactate, oxidation of fatty acids, production of ketone bodies, and metabolism of the main neurotransmitters, glutamate and GABA. The recent findings allow proposing these cells as key players controlling the energetic homeostasis in the CNS.
Collapse
Affiliation(s)
- Débora G Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Roberto F Almeida
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Exact and Biological Sciences Institute, Biological Sciences Department, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Diogo O Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Biochemistry, UFRGS, Porto Alegre, Brazil
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Pharmacology, UFRGS, Porto Alegre, Brazil; Graduate Program in Biological Sciences: Pharmacology and Therapeutics, UFRGS, Porto Alegre, Brazil; Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
25
|
Zhou Z, Ikegaya Y, Koyama R. The Astrocytic cAMP Pathway in Health and Disease. Int J Mol Sci 2019; 20:E779. [PMID: 30759771 PMCID: PMC6386894 DOI: 10.3390/ijms20030779] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 12/11/2022] Open
Abstract
Astrocytes are major glial cells that play critical roles in brain homeostasis. Abnormalities in astrocytic functions can lead to brain disorders. Astrocytes also respond to injury and disease through gliosis and immune activation, which can be both protective and detrimental. Thus, it is essential to elucidate the function of astrocytes in order to understand the physiology of the brain to develop therapeutic strategies against brain diseases. Cyclic adenosine monophosphate (cAMP) is a major second messenger that triggers various downstream cellular machinery in a wide variety of cells. The functions of astrocytes have also been suggested as being regulated by cAMP. Here, we summarize the possible roles of cAMP signaling in regulating the functions of astrocytes. Specifically, we introduce the ways in which cAMP pathways are involved in astrocyte functions, including (1) energy supply, (2) maintenance of the extracellular environment, (3) immune response, and (4) a potential role as a provider of trophic factors, and we discuss how these cAMP-regulated processes can affect brain functions in health and disease.
Collapse
Affiliation(s)
- Zhiwen Zhou
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-0033, Japan.
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-0033, Japan.
- Center for Information and Neural Networks, Suita City, Osaka 565-0871, Japan.
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
26
|
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
|
27
|
Dienel GA. Does shuttling of glycogen-derived lactate from astrocytes to neurons take place during neurotransmission and memory consolidation? J Neurosci Res 2019; 97:863-882. [PMID: 30667077 DOI: 10.1002/jnr.24387] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 12/24/2018] [Accepted: 01/07/2019] [Indexed: 12/17/2022]
Abstract
Glycogen levels in resting brain and its utilization rates during brain activation are high, but the functions fulfilled by glycogenolysis in living brain are poorly understood. Studies in cultured astrocytes have identified glycogen as the preferred fuel to provide ATP for Na+ ,K+ -ATPase for the uptake of extracellular K+ and for Ca2+ -ATPase to pump Ca2+ into the endoplasmic reticulum. Studies in astrocyte-neuron co-cultures led to the suggestion that glycogen-derived lactate is shuttled to neurons as oxidative fuel to support glutamatergic neurotransmission. Furthermore, both knockout of brain glycogen synthase and inhibition of glycogenolysis prior to a memory-evoking event impair memory consolidation, and shuttling of glycogen-derived lactate as neuronal fuel was postulated to be required for memory. However, lactate shuttling has not been measured in any of these studies, and procedures to inhibit glycogenolysis and neuronal lactate uptake are not specific. Testable alternative mechanisms to explain the observed findings are proposed: (i) disruption of K+ and Ca2+ homeostasis, (ii) release of gliotransmitters, (iii) imposition of an energy crisis on astrocytes and neurons by inhibition of mitochondrial pyruvate transport by compounds used to block neuronal monocarboxylic acid transporters, and (iv) inhibition of astrocytic filopodial movements that secondarily interfere with glutamate and K+ uptake from the synaptic cleft. Evidence that most pyruvate/lactate derived from glycogen is not oxidized and does not accumulate suggests predominant glycolytic metabolism of glycogen to support astrocytic energy demands. Sparing of blood-borne glucose for use by neurons is a reasonable explanation for the requirement for glycogenolysis in neurotransmission and memory processing.
Collapse
Affiliation(s)
- Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico
| |
Collapse
|
28
|
Juaristi I, Llorente-Folch I, Satrústegui J, Del Arco A. Extracellular ATP and glutamate drive pyruvate production and energy demand to regulate mitochondrial respiration in astrocytes. Glia 2019; 67:759-774. [PMID: 30623988 DOI: 10.1002/glia.23574] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/08/2018] [Accepted: 11/13/2018] [Indexed: 12/26/2022]
Abstract
Astrocytes respond to energetic demands by upregulating glycolysis, lactate production, and respiration. This study addresses the role of respiration and calcium regulation of respiration as part of the astrocyte response to the workloads caused by extracellular ATP and glutamate. Extracellular ATP (100 μM to 1 mM) causes a Ca2+ -dependent workload and fall of the cytosolic ATP/ADP ratio which acutely increases astrocytes respiration. Part of this increase is related to a Ca2+ -dependent upregulation of cytosolic pyruvate production. Conversely, glutamate (200 μM) causes a Na+ , but not Ca2+ , dependent workload even though glutamate-induced Ca2+ signals readily reach mitochondria. The glutamate workload triggers a rapid fall in the cytosolic ATP/ADP ratio and stimulation of respiration. These effects are mimicked by D-aspartate a nonmetabolized agonist of the glutamate transporter, but not by a metabotropic glutamate receptor agonist, indicating a major role of Na+ -dependent workload in stimulated respiration. Glutamate-induced increase in respiration is linked to a rapid increase in glycolytic pyruvate production, suggesting that both glutamate and extracellular ATP cause an increase in astrocyte respiration fueled by workload-induced increase in pyruvate production. However, glutamate-induced pyruvate production is partly resistant to glycolysis blockers (iodoacetate), indicating that oxidative consumption of glutamate also contributes to stimulated respiration. As stimulation of respiration by ATP and glutamate are similar and pyruvate production smaller in the first case, the results suggest that the response to extracellular ATP is a Ca2+ -dependent upregulation of respiration added to glycolysis upregulation. The global contribution of astrocyte respiratory responses to brain oxygen consumption is an open question.
Collapse
Affiliation(s)
- Inés Juaristi
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.,Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Irene Llorente-Folch
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Center for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jorgina Satrústegui
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.,Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Araceli Del Arco
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.,Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain.,Facultad de Ciencias Ambientales y Bioquímica, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla la Mancha, Toledo, Spain
| |
Collapse
|
29
|
Glycogenolysis in Cerebral Cortex During Sensory Stimulation, Acute Hypoglycemia, and Exercise: Impact on Astrocytic Energetics, Aerobic Glycolysis, and Astrocyte-Neuron Interactions. ADVANCES IN NEUROBIOLOGY 2019; 23:209-267. [DOI: 10.1007/978-3-030-27480-1_8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
30
|
Rothman DL, Dienel GA. Development of a Model to Test Whether Glycogenolysis Can Support Astrocytic Energy Demands of Na +, K +-ATPase and Glutamate-Glutamine Cycling, Sparing an Equivalent Amount of Glucose for Neurons. ADVANCES IN NEUROBIOLOGY 2019; 23:385-433. [PMID: 31667817 DOI: 10.1007/978-3-030-27480-1_14] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recent studies of glycogen in brain have suggested a much more important role in brain energy metabolism and function than previously recognized, including findings of much higher than previously recognized concentrations, consumption at substantial rates compared with utilization of blood-borne glucose, and involvement in ion pumping and in neurotransmission and memory. However, it remains unclear how glycogenolysis is coupled to neuronal activity and provides support for neuronal as well as astroglial function. At present, quantitative aspects of glycogenolysis in brain functions are very difficult to assess due to its metabolic lability, heterogeneous distributions within and among cells, and extreme sensitivity to physiological stimuli. To begin to address this problem, the present study develops a model based on pathway fluxes, mass balance, and literature relevant to functions and turnover of pathways that intersect with glycogen mobilization. A series of equations is developed to describe the stoichiometric relationships between net glycogen consumption that is predominantly in astrocytes with the rate of the glutamate-glutamine cycle, rates of astrocytic and neuronal glycolytic and oxidative metabolism, and the energetics of sodium/potassium pumping in astrocytes and neurons during brain activation. Literature supporting the assumptions of the model is discussed in detail. The overall conclusion is that astrocyte glycogen metabolism is primarily coupled to neuronal function via fueling glycolytically pumping of Na+ and K+ and sparing glucose for neuronal oxidation, as opposed to previous proposals of coupling neurotransmission via glutamate transport, lactate shuttling, and neuronal oxidation of lactate.
Collapse
Affiliation(s)
- Douglas L Rothman
- Magnetic Resonance Research Center and Department of Radiology, Yale University, New Haven, CT, USA.
| | - Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
31
|
Hansson E, Björklund U, Skiöldebrand E, Rönnbäck L. Anti-inflammatory effects induced by pharmaceutical substances on inflammatory active brain astrocytes-promising treatment of neuroinflammation. J Neuroinflammation 2018; 15:321. [PMID: 30447700 PMCID: PMC6240424 DOI: 10.1186/s12974-018-1361-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 11/07/2018] [Indexed: 02/06/2023] Open
Abstract
Background Pharmaceutical treatment with probable anti-inflammatory substances that attack cells in various ways including receptors, ion channels, or transporter systems may slow down the progression of inflammatory conditions. Astrocytes and microglia are the most prominent target cells for inflammation in the central nervous system. Their responses upon inflammatory stimuli work through the NO/cyclic GMP/protein kinase G systems that can downregulate the ATP-induced Ca2+ signaling, as well as G protein activities which alter Na+ transporters including Na+/K+-ATPase pump activity, Toll-like receptor 4 (TLR4), glutamate-induced Ca2+ signaling, and release of pro-inflammatory cytokines. The rationale for this project was to investigate a combination of pharmaceutical substances influencing the NO and the Gi/Gs activations of inflammatory reactive cells in order to make the cells return into a more physiological state. The ATP-evoked Ca2+ signaling is important maybe due to increased ATP release and subsequent activation of purinergic receptors. A balance between intercellular Ca2+ signaling through gap junctions and extracellular signaling mediated by extracellular ATP may be important for physiological function. Methods Astrocytes in primary cultures were incubated with lipopolysaccharide in a physiological glucose concentration for 24 h to induce inflammatory reactivity. The probable anti-inflammatory substances sildenafil and 1α,25-Dihydroxyvitamin D3 together with endomorphin-1, naloxone, and levetiracetam, were used in the presence of high glucose concentration in the medium to restore the cells. Glutamate-, 5-HT-, and ATP-evoked intracellular Ca2+ release, Na+/K+-ATPase expression, expression of inflammatory receptors, and release of tumor necrosis factor alpha were measured. Results Sildenafil in ultralow concentration together with 1α,25-Dihydroxyvitamin D3 showed most prominent effects on the ATP-evoked intracellular Ca2+ release. The μ-opioid agonist endomorphin-1, the μ-opioid antagonist naloxone in ultralow concentration, and the antiepileptic agent levetiracetam downregulated the glutamate-evoked intracellular Ca2+ release and TLR4. The combination of the pharmaceutical substances in high glucose concentration downregulated the glutamate- and ATP-evoked Ca2+ signaling and the TLR4 expression and upregulated the Na+/K+-ATPase pump. Conclusion Pharmaceutical treatment with the combination of substances that have potential anti-inflammatory effects, which attack different biochemical mechanisms in the cells may exert decisive effects to downregulate neuroinflammation in the nervous system.
Collapse
Affiliation(s)
- Elisabeth Hansson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, 3rd floor, SE 413 45, Gothenburg, Sweden.
| | - Ulrika Björklund
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, 3rd floor, SE 413 45, Gothenburg, Sweden
| | - Eva Skiöldebrand
- Section of Pathology, Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden.,Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital, Gothenburg University, Gothenburg, Sweden
| | - Lars Rönnbäck
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, 3rd floor, SE 413 45, Gothenburg, Sweden
| |
Collapse
|
32
|
Gandini Wheeler-Kingshott CAM, Riemer F, Palesi F, Ricciardi A, Castellazzi G, Golay X, Prados F, Solanky B, D'Angelo EU. Challenges and Perspectives of Quantitative Functional Sodium Imaging (fNaI). Front Neurosci 2018; 12:810. [PMID: 30473659 PMCID: PMC6237845 DOI: 10.3389/fnins.2018.00810] [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: 07/19/2018] [Accepted: 10/17/2018] [Indexed: 12/31/2022] Open
Abstract
Brain function has been investigated via the blood oxygenation level dependent (BOLD) effect using magnetic resonance imaging (MRI) for the past decades. Advances in sodium imaging offer the unique chance to access signal changes directly linked to sodium ions (23Na) flux across the cell membrane, which generates action potentials, hence signal transmission in the brain. During this process 23Na transiently accumulates in the intracellular space. Here we show that quantitative functional sodium imaging (fNaI) at 3T is potentially sensitive to 23Na concentration changes during finger tapping, which can be quantified in gray and white matter regions key to motor function. For the first time, we measured a 23Na concentration change of 0.54 mmol/l in the ipsilateral cerebellum, 0.46 mmol/l in the contralateral primary motor cortex (M1), 0.27 mmol/l in the corpus callosum and -11 mmol/l in the ipsilateral M1, suggesting that fNaI is sensitive to distributed functional alterations. Open issues persist on the role of the glymphatic system in maintaining 23Na homeostasis, the role of excitation and inhibition as well as volume distributions during neuronal activity. Haemodynamic and physiological signal recordings coupled to realistic models of tissue function will be critical to understand the mechanisms of such changes and contribute to meeting the overarching challenge of measuring neuronal activity in vivo.
Collapse
Affiliation(s)
- Claudia A M Gandini Wheeler-Kingshott
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom.,Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy.,Brain MRI 3T Mondino Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Frank Riemer
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom.,Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Fulvia Palesi
- Neuroradiology Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Antonio Ricciardi
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom.,Center for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Gloria Castellazzi
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom.,Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Xavier Golay
- NMR Research Unit, Queen Square MS Centre, Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - Ferran Prados
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom.,Center for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom.,Universitat Oberta de Catalunya, Barcelona, Spain
| | - Bhavana Solanky
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - Egidio U D'Angelo
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy.,Brain Connectivity Center, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
33
|
Coggan JS, Keller D, Calì C, Lehväslaiho H, Markram H, Schürmann F, Magistretti PJ. Norepinephrine stimulates glycogenolysis in astrocytes to fuel neurons with lactate. PLoS Comput Biol 2018; 14:e1006392. [PMID: 30161133 PMCID: PMC6160207 DOI: 10.1371/journal.pcbi.1006392] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 09/27/2018] [Accepted: 07/24/2018] [Indexed: 12/20/2022] Open
Abstract
The mechanism of rapid energy supply to the brain, especially to accommodate the heightened metabolic activity of excited states, is not well-understood. We explored the role of glycogen as a fuel source for neuromodulation using the noradrenergic stimulation of glia in a computational model of the neural-glial-vasculature ensemble (NGV). The detection of norepinephrine (NE) by the astrocyte and the coupled cAMP signal are rapid and largely insensitive to the distance of the locus coeruleus projection release sites from the glia, implying a diminished impact for volume transmission in high affinity receptor transduction systems. Glucosyl-conjugated units liberated from glial glycogen by NE-elicited cAMP second messenger transduction winds sequentially through the glycolytic cascade, generating robust increases in NADH and ATP before pyruvate is finally transformed into lactate. This astrocytic lactate is rapidly exported by monocarboxylate transporters to the associated neuron, demonstrating that the astrocyte-to-neuron lactate shuttle activated by glycogenolysis is a likely fuel source for neuromodulation and enhanced neural activity. Altogether, the energy supply for both astrocytes and neurons can be supplied rapidly by glycogenolysis upon neuromodulatory stimulus. Although efficient compared to computers, the human brain utilizes energy at 10-fold the rate of other organs by mass. How the brain is supplied with sufficient on-demand energy to support its activity in the absence of neuronal storage capacity remains unknown. Neurons are not capable of meeting their own energy requirements, instead energy supply in the brain is managed by an oligocellular cartel composed of neurons, glia and the local vasculature (NGV), wherein glia can provide the ergogenic metabolite lactate to the neuron in a process called the astrocyte-to-neuron shuttle (ANLS). The only means of energy storage in the brain is glycogen, a polymerized form of glucose that is localized largely to astrocytes, but its exact role and conditions of use are not clear. In this computational model we show that neuromodulatory stimulation by norepinephrine induces astrocytes to recover glucosyl subunits from glycogen for use in a glycolytic process that favors the production of lactate. The ATP and NADH produced support metabolism in the astrocyte while the lactate is exported to feed the neuron. Thus, rapid energy demands by both neurons and glia in a stimulated brain can be met by glycogen mobilization.
Collapse
Affiliation(s)
- Jay S. Coggan
- Blue Brain Project, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
- * E-mail: (JSC); (PJM)
| | - Daniel Keller
- Blue Brain Project, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Corrado Calì
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Heikki Lehväslaiho
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Henry Markram
- Blue Brain Project, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Felix Schürmann
- Blue Brain Project, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Pierre J. Magistretti
- Blue Brain Project, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
- * E-mail: (JSC); (PJM)
| |
Collapse
|
34
|
Choi SH, Kim HJ, Cho HJ, Park SD, Lee NE, Hwang SH, Cho IH, Hwang H, Rhim H, Kim HC, Nah SY. Gintonin, a Ginseng-Derived Exogenous Lysophosphatidic Acid Receptor Ligand, Protects Astrocytes from Hypoxic and Re-oxygenation Stresses Through Stimulation of Astrocytic Glycogenolysis. Mol Neurobiol 2018; 56:3280-3294. [PMID: 30117105 DOI: 10.1007/s12035-018-1308-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 08/08/2018] [Indexed: 11/25/2022]
Abstract
Astrocytes are a unique brain cell-storing glycogen and express lysophosphatidic acid (LPA) receptors. Gintonin is a ginseng-derived exogenous G protein-coupled LPA receptor ligand. Accumulating evidence shows that astrocytes serve as an energy supplier to neurons through astrocytic glycogenolysis under physiological and pathophysiological conditions. However, little is known about the relationships between LPA receptors and astrocytic glycogenolysis or about the roles of LPA receptors in hypoxia and re-oxygenation stresses. In the present study, we examined the functions of gintonin-mediated astrocytic glycogenolysis in adenosine triphosphate (ATP) production, glutamate uptake, and cell viability under normoxic, hypoxic, and re-oxygenation conditions. The application of gintonin or LPA to astrocytes induced glycogenolysis in concentration- and time-dependent manners. The stimulation of gintonin-mediated astrocytic glycogenolysis was achieved through the LPA receptor-Gαq/11 protein-phospholipase C-inositol 1,4,5-trisphosphate receptor-intracellular calcium ([Ca2+]i) transient pathway. Gintonin treatment to astrocytes increased the phosphorylation of brain phosphorylase kinase, with sensitive manner to K252a, an inhibitor of phosphorylase kinase. Gintonin-mediated astrocytic glycogenolysis was blocked by isofagomine, a glycogen phosphorylase inhibitor. Gintonin additionally increased astrocytic glycogenolysis under hypoxic and re-oxygenation conditions. Moreover, gintonin increased ATP production, glutamate uptake, and cell viability under the hypoxic and re-oxygenation conditions. Collectively, we found that the gintonin-mediated [Ca2+]i transients regulated by LPA receptors were coupled to astrocytic glycogenolysis and that stimulation of gintonin-mediated astrocytic glycogenolysis was coupled to ATP production and glutamate uptake under hypoxic and re-oxygenation conditions, ultimately protecting astrocytes. Hence, the gintonin-mediated astrocytic energy that is modulated via LPA receptors helps to protect astrocytes under hypoxia and re-oxygenation stresses.
Collapse
Affiliation(s)
- Sun-Hye Choi
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, 05029, South Korea
| | - Hyeon-Joong Kim
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, 05029, South Korea
| | - Hee-Jung Cho
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, 05029, South Korea
| | - Sang-Deuk Park
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, 05029, South Korea
| | - Na-Eun Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, 05029, South Korea
| | - Sung-Hee Hwang
- Department of Pharmaceutical Engineering, College of Health Sciences, Sangji University, Wonju, 26339, South Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hongik Hwang
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul, 02792, South Korea
| | - Hyewhon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul, 02792, South Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology program, College of Pharmacy, Kangwon National University, Chunchon, 24341, South Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, 05029, South Korea.
| |
Collapse
|
35
|
Jia S, Li B, Huang J, Verkhratsky A, Peng L. Regulation of Glycogen Content in Astrocytes via Cav-1/PTEN/AKT/GSK-3β Pathway by Three Anti-bipolar Drugs. Neurochem Res 2018; 43:1692-1701. [DOI: 10.1007/s11064-018-2585-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 06/15/2018] [Accepted: 06/20/2018] [Indexed: 12/27/2022]
|
36
|
Horvat A, Vardjan N. Astroglial cAMP signalling in space and time. Neurosci Lett 2018; 689:5-10. [PMID: 29908259 DOI: 10.1016/j.neulet.2018.06.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/11/2018] [Accepted: 06/13/2018] [Indexed: 01/14/2023]
Abstract
To maintain a high level of specificity and normal cell function, the cyclic adenosine monophosphate (cAMP) pathway is tightly regulated in space and time. Recent advances in cAMP reporter technology have provided insights into spatio-temporal characteristics of cAMP signalling in individual living cells, including astrocytes. Astrocytes are glial cells in the central nervous system with many homeostatic functions. In contrast to neurons, astrocytes are electrically silent, but, in response to extracellular stimuli through activation of surface receptors, they can increase intracellular levels of secondary messengers, e.g. Ca2+ and cAMP. This enables them to communicate with neighbouring cells, such as neurons and endothelial cells of blood vessels. The dynamics of receptor-mediated Ca2+ signalling in astrocytes has been extensively studied in the past in contrast to cAMP signalling. Here, we present the first insights into the temporal dynamics of cAMP signalling in living astrocytes, which revealed that cAMP signals in astrocytes exhibit tonic dynamics and are slower than Ca2+ signals with phasic dynamics. We debate on the heterogeneity of basal cAMP levels in astrocytes and how hypotonicity-induced astrocyte swelling affects temporal dynamics of cAMP signalling. Understanding the spatio-temporal characteristics of cAMP signalling in astrocytes is of extreme importance because cAMP governs many important cellular processes and any malfunctions may lead to pathology.
Collapse
Affiliation(s)
- Anemari Horvat
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Tehnološki Park 24, 1000 Ljubljana, Slovenia
| | - Nina Vardjan
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Tehnološki Park 24, 1000 Ljubljana, Slovenia.
| |
Collapse
|
37
|
Yapıcı-Eser H, Dönmez-Demir B, Kılıç K, Eren-Koçak E, Dalkara T. Stress modulates cortical excitability via α-2 adrenergic and glucocorticoid receptors: As assessed by spreading depression. Exp Neurol 2018; 307:45-51. [PMID: 29856967 DOI: 10.1016/j.expneurol.2018.05.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/15/2018] [Accepted: 05/27/2018] [Indexed: 12/29/2022]
Abstract
An increase in cortical excitability may be one of the factors mediating stress-induced vulnerability to neuropsychiatric disorders. Since stress increases extracellular glutamate and predisposes to migraine with aura attacks, we aimed to study the effect of stress on cortical spreading depression (CSD), the biological substrate of migraine aura and a measure of cortical excitability. CSD was induced by increasing concentrations of KCl applied over the dura with 5-minute intervals and recorded from parieto-occipital cortex to assess the CSD-induction threshold in acutely-stressed, chronically-stressed and naive mice. To study the mechanisms of acute stress-induced decrease in CSD threshold, we systemically administered clonidine, yohimbine, propranolol, CRH1 receptor antagonist NBI27914, mifepristone and spironolactone at doses shown to be effective on stress as well as a central noradrenergic neurotoxin (DSP-4) before acute stress. CSD threshold was significantly lowered by acute and chronic stress as well as central noradrenergic denervation. Clonidine and mifepristone further decreased the CSD threshold below the acute stress-induced levels, whereas yohimbine reversed the acute stress-induced decrease in CSD threshold compared to the saline-injected and stressed control groups. Propranolol, NBI27914 and spironolactone did not modify the effect of acute stress on CSD threshold. Stress increases cortical excitability as illustrated by a decrease in CSD-induction threshold. This action of acute stress is mediated by α2-adrenergic and glucocorticoid receptors. The decrease in CSD threshold may account for the stress-induced susceptibility to migraine. CSD may be used as a tool to study the link between stress-related disorders and cortical excitability.
Collapse
Affiliation(s)
- Hale Yapıcı-Eser
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey; School of Medicine, Department of Psychiatry, Koç University & Research Center for Translational Medicine (KUTTAM), Koç University, İstanbul, Turkey
| | - Buket Dönmez-Demir
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Kıvılcım Kılıç
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey; Neurophotonics Center, Boston University, Boston, MA, United States
| | - Emine Eren-Koçak
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
38
|
Pinto CB, Saleh Velez FG, Lopes F, de Toledo Piza PV, Dipietro L, Wang QM, Mazwi NL, Camargo EC, Black-Schaffer R, Fregni F. SSRI and Motor Recovery in Stroke: Reestablishment of Inhibitory Neural Network Tonus. Front Neurosci 2017; 11:637. [PMID: 29200995 PMCID: PMC5696576 DOI: 10.3389/fnins.2017.00637] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 11/02/2017] [Indexed: 12/23/2022] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are currently widely used in the field of the neuromodulation not only because of their anti-depressive effects but also due to their ability to promote plasticity and enhance motor recovery in patients with stroke. Recent studies showed that fluoxetine promotes motor recovery after stroke through its effects on the serotonergic system enhancing motor outputs and facilitating long term potentiation, key factors in motor neural plasticity. However, little is known in regards of the exact mechanisms underlying these effects and several aspects of it remain poorly understood. In this manuscript, we discuss evidence supporting the hypothesis that SSRIs, and in particular fluoxetine, modulate inhibitory pathways, and that this modulation enhances reorganization and reestablishment of excitatory-inhibitory control; these effects play a key role in learning induced plasticity in neural circuits involved in the promotion of motor recovery after stroke. This discussion aims to provide important insights and rationale for the development of novel strategies for stroke motor rehabilitation.
Collapse
Affiliation(s)
- Camila B. Pinto
- Laboratory of Neuromodulation and Center for Clinical Research Learning, Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, Harvard University, Boston, MA, United States
- Department of Neuroscience and Behavior, Psychology Institute, University of São Paulo, São Paulo, Brazil
| | - Faddi G. Saleh Velez
- Laboratory of Neuromodulation and Center for Clinical Research Learning, Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, Harvard University, Boston, MA, United States
| | - Fernanda Lopes
- Laboratory of Neuromodulation and Center for Clinical Research Learning, Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, Harvard University, Boston, MA, United States
| | - Polyana V. de Toledo Piza
- Laboratory of Neuromodulation and Center for Clinical Research Learning, Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, Harvard University, Boston, MA, United States
- Department of Severe Patients, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Qing M. Wang
- Stroke Biological Recovery Laboratory, Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, Harvard University, Boston, MA, United States
| | - Nicole L. Mazwi
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, Harvard University, Boston, MA, United States
| | - Erica C. Camargo
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Harvard University, Boston, MA, United States
| | - Randie Black-Schaffer
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, Harvard University, Boston, MA, United States
| | - Felipe Fregni
- Laboratory of Neuromodulation and Center for Clinical Research Learning, Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, Harvard University, Boston, MA, United States
| |
Collapse
|
39
|
Hertz L, Chen Y. Additional mechanisms for brain activation failure due to reduced glucose metabolism-a commentary on Zilberter and Zilberter: The vicious circle of hypometabolism in neurodegenerative diseases. J Neurosci Res 2017; 96:757-761. [PMID: 29095528 DOI: 10.1002/jnr.24192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/06/2017] [Accepted: 10/09/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Leif Hertz
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, Shenyang, People's Republic of China
| | - Ye Chen
- Henry M. Jackson Foundation, Bethesda, Maryland
| |
Collapse
|
40
|
Hertz L, Chen Y. Integration between Glycolysis and Glutamate-Glutamine Cycle Flux May Explain Preferential Glycolytic Increase during Brain Activation, Requiring Glutamate. Front Integr Neurosci 2017. [PMID: 28890689 DOI: 10.3389/fnint.2017.00018+10.3389/fnint.2017.00018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The 1988 observation by Fox et al. (1988) that brief intense brain activation increases glycolysis (pyruvate formation from glucose) much more than oxidative metabolism has been abundantly confirmed. Specifically glycolytic increase was unexpected because the amount of ATP it generates is much smaller than that formed by subsequent oxidative metabolism of pyruvate. The present article shows that preferential glycolysis can be explained by metabolic processes associated with activation of the glutamate-glutamine cycle. The flux in this cycle, which is essential for production of transmitter glutamate and GABA, equals 75% of brain glucose utilization and each turn is associated with utilization of ~1 glucose molecule. About one half of the association between cycle flux and glucose metabolism occurs during neuronal conversion of glutamine to glutamate in a process similar to the malate-aspartate shuttle (MAS) except that glutamate is supplied from glutamine, not formed from α-ketoglutarate (αKG) as during operation of conventional MAS. Regular MAS function is triggered by one oxidative process in the cytosol during glycolysis causing NAD+ reduction to NADH. Since NADH cannot cross the mitochondrial membrane (MEM) for oxidation NAD+ is re-generated by conversion of cytosolic oxaloacetate (OAA) to malate, which enters the mitochondria for oxidation and in a cyclic process regenerates cytosolic OAA. Therefore MAS as well as the "pseudo-MAS" necessary for neuronal glutamate formation can only operate together with cytosolic reduction of NAD+ to NADH. The major process causing NAD+ reduction is glycolysis which therefore also must occur during neuronal conversion of glutamine to glutamate and may energize vesicular glutamate uptake which preferentially uses glycolytically derived energy. Another major contributor to the association between glutamate-glutamine cycle and glucose utilization is the need for astrocytic pyruvate to generate glutamate. Although some oxidative metabolism occurs during glutamate formation it is only one half of that during normal tricarboxylic acid (TCA) cycle function. Glutamate's receptor stimulation leads to potassium ion (K+) release and astrocytic uptake, preferentially fueled by glycolysis and followed by release and neuronal re-accumulation. The activation-induced preferential glycolysis diminishes with continued activation and is followed by an increased ratio between oxidative metabolism and glycolysis, reflecting oxidation of generated glutamate and accumulated lactate.
Collapse
Affiliation(s)
- Leif Hertz
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical UniversityShenyang, China
| | - Ye Chen
- Henry M. Jackson FoundationBethesda, MD, United States
| |
Collapse
|
41
|
Hertz L, Chen Y. Integration between Glycolysis and Glutamate-Glutamine Cycle Flux May Explain Preferential Glycolytic Increase during Brain Activation, Requiring Glutamate. Front Integr Neurosci 2017; 11:18. [PMID: 28890689 PMCID: PMC5574930 DOI: 10.3389/fnint.2017.00018] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 08/11/2017] [Indexed: 11/13/2022] Open
Abstract
The 1988 observation by Fox et al. (1988) that brief intense brain activation increases glycolysis (pyruvate formation from glucose) much more than oxidative metabolism has been abundantly confirmed. Specifically glycolytic increase was unexpected because the amount of ATP it generates is much smaller than that formed by subsequent oxidative metabolism of pyruvate. The present article shows that preferential glycolysis can be explained by metabolic processes associated with activation of the glutamate-glutamine cycle. The flux in this cycle, which is essential for production of transmitter glutamate and GABA, equals 75% of brain glucose utilization and each turn is associated with utilization of ~1 glucose molecule. About one half of the association between cycle flux and glucose metabolism occurs during neuronal conversion of glutamine to glutamate in a process similar to the malate-aspartate shuttle (MAS) except that glutamate is supplied from glutamine, not formed from α-ketoglutarate (αKG) as during operation of conventional MAS. Regular MAS function is triggered by one oxidative process in the cytosol during glycolysis causing NAD+ reduction to NADH. Since NADH cannot cross the mitochondrial membrane (MEM) for oxidation NAD+ is re-generated by conversion of cytosolic oxaloacetate (OAA) to malate, which enters the mitochondria for oxidation and in a cyclic process regenerates cytosolic OAA. Therefore MAS as well as the "pseudo-MAS" necessary for neuronal glutamate formation can only operate together with cytosolic reduction of NAD+ to NADH. The major process causing NAD+ reduction is glycolysis which therefore also must occur during neuronal conversion of glutamine to glutamate and may energize vesicular glutamate uptake which preferentially uses glycolytically derived energy. Another major contributor to the association between glutamate-glutamine cycle and glucose utilization is the need for astrocytic pyruvate to generate glutamate. Although some oxidative metabolism occurs during glutamate formation it is only one half of that during normal tricarboxylic acid (TCA) cycle function. Glutamate's receptor stimulation leads to potassium ion (K+) release and astrocytic uptake, preferentially fueled by glycolysis and followed by release and neuronal re-accumulation. The activation-induced preferential glycolysis diminishes with continued activation and is followed by an increased ratio between oxidative metabolism and glycolysis, reflecting oxidation of generated glutamate and accumulated lactate.
Collapse
Affiliation(s)
- Leif Hertz
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical UniversityShenyang, China
| | - Ye Chen
- Henry M. Jackson FoundationBethesda, MD, United States
| |
Collapse
|
42
|
Hertz L, Chen Y. Integration between Glycolysis and Glutamate-Glutamine Cycle Flux May Explain Preferential Glycolytic Increase during Brain Activation, Requiring Glutamate. Front Integr Neurosci 2017. [PMID: 28890689 DOI: 10.3389/fnint.2017.00018 10.3389/fnint.2017.00018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The 1988 observation by Fox et al. (1988) that brief intense brain activation increases glycolysis (pyruvate formation from glucose) much more than oxidative metabolism has been abundantly confirmed. Specifically glycolytic increase was unexpected because the amount of ATP it generates is much smaller than that formed by subsequent oxidative metabolism of pyruvate. The present article shows that preferential glycolysis can be explained by metabolic processes associated with activation of the glutamate-glutamine cycle. The flux in this cycle, which is essential for production of transmitter glutamate and GABA, equals 75% of brain glucose utilization and each turn is associated with utilization of ~1 glucose molecule. About one half of the association between cycle flux and glucose metabolism occurs during neuronal conversion of glutamine to glutamate in a process similar to the malate-aspartate shuttle (MAS) except that glutamate is supplied from glutamine, not formed from α-ketoglutarate (αKG) as during operation of conventional MAS. Regular MAS function is triggered by one oxidative process in the cytosol during glycolysis causing NAD+ reduction to NADH. Since NADH cannot cross the mitochondrial membrane (MEM) for oxidation NAD+ is re-generated by conversion of cytosolic oxaloacetate (OAA) to malate, which enters the mitochondria for oxidation and in a cyclic process regenerates cytosolic OAA. Therefore MAS as well as the "pseudo-MAS" necessary for neuronal glutamate formation can only operate together with cytosolic reduction of NAD+ to NADH. The major process causing NAD+ reduction is glycolysis which therefore also must occur during neuronal conversion of glutamine to glutamate and may energize vesicular glutamate uptake which preferentially uses glycolytically derived energy. Another major contributor to the association between glutamate-glutamine cycle and glucose utilization is the need for astrocytic pyruvate to generate glutamate. Although some oxidative metabolism occurs during glutamate formation it is only one half of that during normal tricarboxylic acid (TCA) cycle function. Glutamate's receptor stimulation leads to potassium ion (K+) release and astrocytic uptake, preferentially fueled by glycolysis and followed by release and neuronal re-accumulation. The activation-induced preferential glycolysis diminishes with continued activation and is followed by an increased ratio between oxidative metabolism and glycolysis, reflecting oxidation of generated glutamate and accumulated lactate.
Collapse
Affiliation(s)
- Leif Hertz
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical UniversityShenyang, China
| | - Ye Chen
- Henry M. Jackson FoundationBethesda, MD, United States
| |
Collapse
|
43
|
Technical and experimental features of Magnetic Resonance Spectroscopy of brain glycogen metabolism. Anal Biochem 2017; 529:117-126. [DOI: 10.1016/j.ab.2016.12.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 08/31/2016] [Accepted: 12/23/2016] [Indexed: 01/20/2023]
|
44
|
Hertz L, Chen Y. Glycogenolysis, an Astrocyte-Specific Reaction, is Essential for Both Astrocytic and Neuronal Activities Involved in Learning. Neuroscience 2017; 370:27-36. [PMID: 28668486 DOI: 10.1016/j.neuroscience.2017.06.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/10/2017] [Accepted: 06/19/2017] [Indexed: 01/26/2023]
Abstract
In brain glycogen, formed from glucose, is degraded (glycogenolysis) in astrocytes but not in neurons. Although most of the degradation follows the same pathway as glucose, its breakdown product, l-lactate, is released from astrocytes in larger amounts than glucose when glycogenolysis is activated by noradrenaline. However, this is not the case when glycogenolysis is activated by high potassium ion (K+) concentrations - possibly because noradrenaline in contrast to high K+ stimulates glycogenolysis by an increase not only in free cytosolic Ca2+ concentration ([Ca2+]i) but also in cyclic AMP (c-AMP), which may increase the expression of the monocarboxylate transporter through which it is released. Several transmitters activate glycogenolysis in astrocytes and do so at different time points after training. This stimulation is essential for memory consolidation because glycogenolysis is necessary for uptake of K+ and stimulates formation of glutamate from glucose, and therefore is needed both for removal of increased extracellular K+ following neuronal excitation (which initially occurs into astrocytes) and for formation of transmitter glutamate and GABA. In addition the released l-lactate has effects on neurons which are essential for learning and for learning-related long-term potentiation (LTP), including induction of the neuronal gene Arc/Arg3.1 and activation of gene cascades mediated by CREB and cofilin. Inhibition of glycogenolysis blocks learning, LTP and all related molecular events, but all changes can be reversed by injection of l-lactate. The effect of extracellular l-lactate is due to both astrocyte-mediated signaling which activates noradrenergic activity on all brain cells and to a minor uptake, possibly into dendritic spines.
Collapse
Affiliation(s)
- Leif Hertz
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, Shenyang, PR China
| | - Ye Chen
- Henry M. Jackson Foundation, Bethesda, MD 20817, USA.
| |
Collapse
|
45
|
Chaban YHG, Chen Y, Hertz E, Hertz L. Severe Convulsions and Dysmyelination in Both Jimpy and Cx32/47 -/- Mice may Associate Astrocytic L-Channel Function with Myelination and Oligodendrocytic Connexins with Internodal K v Channels. Neurochem Res 2017; 42:1747-1766. [PMID: 28214987 DOI: 10.1007/s11064-017-2194-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 12/12/2022]
Abstract
The Jimpy mouse illustrates the importance of interactions between astrocytes and oligodendrocytes. It has a mutation in Plp coding for proteolipid protein and DM20. Its behavior is normal at birth but from the age of ~2 weeks it shows severe convulsions associated with oligodendrocyte/myelination deficits and early death. A normally occurring increase in oxygen consumption by highly elevated K+ concentrations is absent in Jimpy brain slices and cultured astrocytes, reflecting that Plp at early embryonic stages affects common precursors as also shown by the ability of conditioned medium from normal astrocytes to counteract histological abnormalities. This metabolic response is now known to reflect opening of L-channels for Ca2+. The resulting deficiency in Ca2+ entry has many consequences, including lack of K+-stimulated glycogenolysis and release of gliotransmitter ATP. Lack of purinergic stimulation compromises oligodendrocyte survival and myelination and affects connexins and K+ channels. Mice lacking the oligodendrocytic connexins Cx32 and 47 show similar neurological dysfunction as Jimpy. This possibly reflects that K+ released by intermodal axonal Kv channels is transported underneath a loosened myelin sheath instead of reaching the extracellular space via connexin-mediated transport to oligodendrocytes, followed by release and astrocytic Na+,K+-ATPase-driven uptake with subsequent Kir4.1-facilitated release and neuronal uptake.
Collapse
Affiliation(s)
| | - Ye Chen
- Henry M. Jackson Foundation, Bethesda, MD, 20817, USA
| | - Elna Hertz
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, Shenyang, People's Republic of China
| | - Leif Hertz
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, Shenyang, People's Republic of China.
| |
Collapse
|
46
|
Sonnay S, Gruetter R, Duarte JMN. How Energy Metabolism Supports Cerebral Function: Insights from 13C Magnetic Resonance Studies In vivo. Front Neurosci 2017; 11:288. [PMID: 28603480 PMCID: PMC5445183 DOI: 10.3389/fnins.2017.00288] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/04/2017] [Indexed: 12/25/2022] Open
Abstract
Cerebral function is associated with exceptionally high metabolic activity, and requires continuous supply of oxygen and nutrients from the blood stream. Since the mid-twentieth century the idea that brain energy metabolism is coupled to neuronal activity has emerged, and a number of studies supported this hypothesis. Moreover, brain energy metabolism was demonstrated to be compartmentalized in neurons and astrocytes, and astrocytic glycolysis was proposed to serve the energetic demands of glutamatergic activity. Shedding light on the role of astrocytes in brain metabolism, the earlier picture of astrocytes being restricted to a scaffold-associated function in the brain is now out of date. With the development and optimization of non-invasive techniques, such as nuclear magnetic resonance spectroscopy (MRS), several groups have worked on assessing cerebral metabolism in vivo. In this context, 1H MRS has allowed the measurements of energy metabolism-related compounds, whose concentrations can vary under different brain activation states. 1H-[13C] MRS, i.e., indirect detection of signals from 13C-coupled 1H, together with infusion of 13C-enriched glucose has provided insights into the coupling between neurotransmission and glucose oxidation. Although these techniques tackle the coupling between neuronal activity and metabolism, they lack chemical specificity and fail in providing information on neuronal and glial metabolic pathways underlying those processes. Currently, the improvement of detection modalities (i.e., direct detection of 13C isotopomers), the progress in building adequate mathematical models along with the increase in magnetic field strength now available render possible detailed compartmentalized metabolic flux characterization. In particular, direct 13C MRS offers more detailed dataset acquisitions and provides information on metabolic interactions between neurons and astrocytes, and their role in supporting neurotransmission. Here, we review state-of-the-art MR methods to study brain function and metabolism in vivo, and their contribution to the current understanding of how astrocytic energy metabolism supports glutamatergic activity and cerebral function. In this context, recent data suggests that astrocytic metabolism has been underestimated. Namely, the rate of oxidative metabolism in astrocytes is about half of that in neurons, and it can increase as much as the rate of neuronal metabolism in response to sensory stimulation.
Collapse
Affiliation(s)
- Sarah Sonnay
- Laboratory for Functional and Metabolic Imaging, École Polytechnique Fédérale de LausanneLausanne, Switzerland
| | - Rolf Gruetter
- Laboratory for Functional and Metabolic Imaging, École Polytechnique Fédérale de LausanneLausanne, Switzerland.,Department of Radiology, University of LausanneLausanne, Switzerland.,Department of Radiology, University of GenevaGeneva, Switzerland
| | - João M N Duarte
- Laboratory for Functional and Metabolic Imaging, École Polytechnique Fédérale de LausanneLausanne, Switzerland
| |
Collapse
|
47
|
Giorgi-Coll S, Amaral AI, Hutchinson PJA, Kotter MR, Carpenter KLH. Succinate supplementation improves metabolic performance of mixed glial cell cultures with mitochondrial dysfunction. Sci Rep 2017; 7:1003. [PMID: 28432362 PMCID: PMC5430749 DOI: 10.1038/s41598-017-01149-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 03/27/2017] [Indexed: 11/19/2022] Open
Abstract
Mitochondrial dysfunction, the inability to efficiently utilise metabolic fuels and oxygen, contributes to pathological changes following traumatic spinal cord or traumatic brain injury (TBI). In the present study, we tested the hypothesis that succinate supplementation can improve cellular energy state under metabolically stressed conditions in a robust, reductionist in vitro model of mitochondrial dysfunction in which primary mixed glial cultures (astrocytes, microglia and oligodendrocytes) were exposed to the mitochondrial complex I inhibitor rotenone. Cellular response was determined by measuring intracellular ATP, extracellular metabolites (glucose, lactate, pyruvate), and oxygen consumption rate (OCR). Rotenone produced no significant changes in glial ATP levels. However, it induced metabolic deficits as evidenced by lactate/pyruvate ratio (LPR) elevation (a clinically-established biomarker for poor outcome in TBI) and decrease in OCR. Succinate addition partially ameliorated these metabolic deficits. We conclude that succinate can improve glial oxidative metabolism, consistent our previous findings in TBI patients’ brains. The mixed glial cellular model may be useful in developing therapeutic strategies for conditions involving mitochondrial dysfunction, such as TBI.
Collapse
Affiliation(s)
- Susan Giorgi-Coll
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Box 167, Cambridge Biomedical Campus, CB2 0QQ, UK.
| | - Ana I Amaral
- Anne McLaren Laboratory, Wellcome Trust MRC Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, West Forvie Building, Robinson Way, Cambridge, CB2 0SZ, UK
| | - Peter J A Hutchinson
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Box 167, Cambridge Biomedical Campus, CB2 0QQ, UK
| | - Mark R Kotter
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Box 167, Cambridge Biomedical Campus, CB2 0QQ, UK. .,Anne McLaren Laboratory, Wellcome Trust MRC Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, West Forvie Building, Robinson Way, Cambridge, CB2 0SZ, UK.
| | - Keri L H Carpenter
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Box 167, Cambridge Biomedical Campus, CB2 0QQ, UK.
| |
Collapse
|
48
|
Bai Q, Song D, Gu L, Verkhratsky A, Peng L. Bi-phasic regulation of glycogen content in astrocytes via Cav-1/PTEN/PI3K/AKT/GSK-3β pathway by fluoxetine. Psychopharmacology (Berl) 2017; 234:1069-1077. [PMID: 28233032 DOI: 10.1007/s00213-017-4547-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/18/2017] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Here, we present the data indicating that chronic treatment with fluoxetine regulates Cav-1/PTEN/PI3K/AKT/GSK-3β signalling pathway and glycogen content in primary cultures of astrocytes with bi-phasic concentration dependence. RESULTS At lower concentrations, fluoxetine downregulates gene expression of Cav-1, decreases membrane content of PTEN, increases activity of PI3K/AKT, and elevates GSK-3β phosphorylation thus suppressing its activity. At higher concentrations, fluoxetine acts in an inverse fashion. As expected, fluoxetine at lower concentrations increased while at higher concentrations decreased glycogen content in astrocytes. CONCLUSIONS Our findings indicate that bi-phasic regulation of glycogen content via Cav-1/PTEN/PI3K/AKT/GSK-3β pathway by fluoxetine may be responsible for both therapeutic and side effects of the drug.
Collapse
Affiliation(s)
- Qiufang Bai
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, No. 77, Puhe Road, Shenbei District, Shenyang, People's Republic of China
| | - Dan Song
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, No. 77, Puhe Road, Shenbei District, Shenyang, People's Republic of China
| | - Li Gu
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, No. 77, Puhe Road, Shenbei District, Shenyang, People's Republic of China
| | - Alexei Verkhratsky
- Faculty of Life Science, The University of Manchester, Manchester, UK.,Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain
| | - Liang Peng
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, No. 77, Puhe Road, Shenbei District, Shenyang, People's Republic of China.
| |
Collapse
|
49
|
Wang Q, Jie W, Liu JH, Yang JM, Gao TM. An astroglial basis of major depressive disorder? An overview. Glia 2017; 65:1227-1250. [DOI: 10.1002/glia.23143] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 02/26/2017] [Accepted: 02/27/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Qian Wang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, Southern Medical University; Guangzhou 510515 China
| | - Wei Jie
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, Southern Medical University; Guangzhou 510515 China
| | - Ji-Hong Liu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, Southern Medical University; Guangzhou 510515 China
| | - Jian-Ming Yang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, Southern Medical University; Guangzhou 510515 China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Collaborative Innovation Center for Brain Science, Department of Neurobiology, Southern Medical University; Guangzhou 510515 China
| |
Collapse
|
50
|
Gerkau NJ, Rakers C, Petzold GC, Rose CR. Differential effects of energy deprivation on intracellular sodium homeostasis in neurons and astrocytes. J Neurosci Res 2017; 95:2275-2285. [PMID: 28150887 DOI: 10.1002/jnr.23995] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/10/2016] [Accepted: 11/10/2016] [Indexed: 12/11/2022]
Abstract
The maintenance of a low intracellular sodium concentration by the Na+ /K+ -ATPase (NKA) is critical for brain function. In both neurons and glial cells, NKA activity is required to counteract changes in the sodium gradient due to opening of voltage- and ligand-gated channels and/or activation of sodium-dependent secondary active transporters. Because NKA consumes about 50% of cellular ATP, sodium homeostasis is strictly dependent on an intact cellular energy metabolism. Despite the high energetic costs of electrical signaling, neurons do not contain significant energy stores themselves, but rely on a close metabolic interaction with surrounding astrocytes. A disruption of energy supply as observed during focal ischemia causes a rapid drop in ATP in both neurons and astrocytes. There is accumulating evidence that dysregulation of intracellular sodium is an inherent consequence of a reduction in cellular ATP, triggering secondary failure of extra- and intracellular homeostasis of other ions -in particular potassium, calcium, and protons- and thereby promoting excitotoxicity. The characteristics, cellular mechanisms and direct consequences of harmful sodium influx, however, differ between neurons and astrocytes. Moreover, recent work has shown that an intact astrocyte metabolism and sodium homeostasis are critical to maintain the sodium homeostasis of surrounding neurons as well as their capacity to recover from imposed sodium influx. Understanding the mechanisms of sodium increases upon metabolic failure and the differential responses of neurons and glial cells as well as their metabolic interactions will be critical to fully unravel the events causing cellular malfunction, failure and cell death following energy depletion. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Niklas J Gerkau
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, D-40225, Düsseldorf, Germany
| | - Cordula Rakers
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Strasse 27, 53127, Bonn, Germany
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Strasse 27, 53127, Bonn, Germany
| | - Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, D-40225, Düsseldorf, Germany
| |
Collapse
|