1
|
Álvarez-Merz I, Eitelmann S. An Empirical-Theoretical Approach to Determine Astroglial Potassium Upon Ischemic Stress. Methods Mol Biol 2025; 2896:33-49. [PMID: 40111595 DOI: 10.1007/978-1-0716-4366-2_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Dynamic regulation of intracellular potassium is a key feature of astrocytes, as it will determine their capability to regulate extracellular potassium homeostasis during physiology and pathology. However, the study of intracellular potassium dynamics is hindered by the lack of adequate techniques. This chapter outlines a method for calculating astrocytic potassium concentration using the Goldman-Hodgkin-Katz equation. The method integrates experimental measurements of astroglial sodium concentration and membrane potential, along with extracellular potassium and sodium concentrations. We will show how this empirical-theoretical method reveals an initial loss of intracellular potassium and a subsequent transient potassium overshoot upon a protocol that mimics metabolic ischemic stress in acute brain slices.
Collapse
Affiliation(s)
- Iris Álvarez-Merz
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sara Eitelmann
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
2
|
Pushie MJ, Sylvain NJ, Hou H, Pendleton N, Wang R, Zimmermann L, Pally M, Cayabyab FS, Peeling L, Kelly ME. X-ray fluorescence mapping of brain tissue reveals the profound extent of trace element dysregulation in stroke pathophysiology. Metallomics 2024; 16:mfae054. [PMID: 39547935 PMCID: PMC11631071 DOI: 10.1093/mtomcs/mfae054] [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: 06/18/2024] [Accepted: 11/13/2024] [Indexed: 11/17/2024]
Abstract
The brain is a privileged organ with regard to its trace element composition and maintains a robust barrier system to sequester this specialized environment from the rest of the body and the vascular system. Stroke is caused by loss of adequate blood flow to a region of the brain. Without adequate blood flow ischaemic changes begin almost immediately, triggering an ischaemic cascade, characterized by ion dysregulation, loss of function, oxidative damage, cellular degradation, and breakdown of the barrier that helps maintain this environment. Ion dysregulation is a hallmark of stroke pathophysiology and we observe that most elements in the brain are dysregulated after stroke. X-ray fluorescence-based detection of physiological changes in the neurometallome after stroke reveals profound ion dysregulation within the lesion and surrounding tissue. Not only are most elements significantly dysregulated after stroke, but the level of dysregulation cannot be predicted from a cell-level description of dysregulation. X-ray fluorescence imaging reveals that the stroke lesion retains <25% of essential K+ after stroke, but this element is not concomitantly elevated elsewhere in the organ. Moreover, elements like Na+, Ca2+, and Cl- are vastly elevated above levels available in normal brain tissue (>400%, >200%, and >150%, respectively). We hypothesize that weakening of the blood-brain barrier after stroke allows elements to freely diffuse down their concentration gradient so that the stroke lesion is in equilibrium with blood (and the compartments containing brain interstitial fluid and cerebrospinal fluid). The change observed for the neurometallome likely has consequences for the potential to rescue infarcted tissue, but also presents specific targets for treatment.
Collapse
Affiliation(s)
- M Jake Pushie
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Nicole J Sylvain
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Huishu Hou
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Nicole Pendleton
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Richard Wang
- College of Medicine, Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Liam Zimmermann
- College of Medicine, Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Maxwell Pally
- College of Arts & Science, Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Francisco S Cayabyab
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Lissa Peeling
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Michael E Kelly
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
3
|
Obeagu EI. Potassium dynamics in sickle cell anemia: clinical implications and pathophysiological insights. Ann Med Surg (Lond) 2024; 86:6037-6045. [PMID: 39359761 PMCID: PMC11444568 DOI: 10.1097/ms9.0000000000002551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/27/2024] [Indexed: 10/04/2024] Open
Abstract
Potassium dynamics are critical in the pathophysiology of sickle cell anemia (SCA), a genetic disorder characterized by the presence of abnormally shaped red blood cells that lead to various complications such as vaso-occlusive crises and hemolytic anemia. This review focuses on the clinical implications and pathophysiological insights of potassium regulation in SCA, highlighting its impact on disease progression and potential therapeutic strategies. The dysregulation of potassium transport in SCA leads to significant K+ efflux and cellular dehydration, exacerbating the sickling process. Dehydrated sickle cells, due to potassium loss, become more rigid and prone to causing blockages in small blood vessels, leading to painful vaso-occlusive crises and ischemia. Furthermore, chronic hemolysis in SCA, aggravated by potassium imbalance, contributes to severe anemia and systemic complications. These insights underscore the importance of maintaining potassium homeostasis to mitigate disease severity and improve patient outcomes. Therapeutic strategies targeting potassium regulation show promise in managing SCA. Inhibitors of the Gardos channel, such as senicapoc, have demonstrated potential in reducing sickling and hemolysis. Additionally, hydration therapy plays a crucial role in maintaining electrolyte balance and preventing RBC dehydration. A comprehensive approach that includes monitoring and correcting electrolyte imbalances, along with standard treatments like hydroxyurea and blood transfusions, is essential for effective disease management.
Collapse
|
4
|
Hermanova Z, Valihrach L, Kriska J, Maheta M, Tureckova J, Kubista M, Anderova M. The deletion of AQP4 and TRPV4 affects astrocyte swelling/volume recovery in response to ischemia-mimicking pathologies. Front Cell Neurosci 2024; 18:1393751. [PMID: 38818517 PMCID: PMC11138210 DOI: 10.3389/fncel.2024.1393751] [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: 02/29/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024] Open
Abstract
Introduction Astrocytic Transient receptor potential vanilloid 4 (TRPV4) channels, together with Aquaporin 4 (AQP4), are suspected to be the key players in cellular volume regulation, and therefore may affect the development and severity of cerebral edema during ischemia. In this study, we examined astrocytic swelling/volume recovery in mice with TRPV4 and/or AQP4 deletion in response to in vitro ischemic conditions, to determine how the deletion of these channels can affect the development of cerebral edema. Methods We used three models of ischemia-related pathological conditions: hypoosmotic stress, hyperkalemia, and oxygenglucose deprivation (OGD), and observed their effect on astrocyte volume changes in acute brain slices of Aqp4-/-, Trpv4-/- and double knockouts. In addition, we employed single-cell RT-qPCR to assess the effect of TRPV4 and AQP4 deletion on the expression of other ion channels and transporters involved in the homeostatic functioning of astrocytes. Results Quantification of astrocyte volume changes during OGD revealed that the deletion of AQP4 reduces astrocyte swelling, while simultaneous deletion of both AQP4 and TRPV4 leads to a disruption of astrocyte volume recovery during the subsequent washout. Of note, astrocyte exposure to hypoosmotic stress or hyperkalemia revealed no differences in astrocyte swelling in the absence of AQP4, TRPV4, or both channels. Moreover, under ischemia-mimicking conditions, we identified two distinct subpopulations of astrocytes with low and high volumetric responses (LRA and HRA), and their analyses revealed that mainly HRA are affected by the deletion of AQP4, TRPV4, or both channels. Furthermore, gene expression analysis revealed reduced expression of the ion transporters KCC1 and ClC2 as well as the receptors GABAB and NMDA in Trpv4-/- mice. The deletion of AQP4 instead caused reduced expression of the serine/cysteine peptidase inhibitor Serpina3n. Discussion Thus, we showed that in AQP4 or TRPV4 knockouts, not only the specific function of these channels is affected, but also the expression of other proteins, which may modulate the ischemic cascade and thus influence the final impact of ischemia.
Collapse
Affiliation(s)
- Zuzana Hermanova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
- Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Lukas Valihrach
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Vestec, Czechia
| | - Jan Kriska
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
| | - Mansi Maheta
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Vestec, Czechia
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
| | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Vestec, Czechia
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
| |
Collapse
|
5
|
Ding F, Sun Q, Long C, Rasmussen RN, Peng S, Xu Q, Kang N, Song W, Weikop P, Goldman SA, Nedergaard M. Dysregulation of extracellular potassium distinguishes healthy ageing from neurodegeneration. Brain 2024; 147:1726-1739. [PMID: 38462589 PMCID: PMC11068329 DOI: 10.1093/brain/awae075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 03/12/2024] Open
Abstract
Progressive neuronal loss is a hallmark feature distinguishing neurodegenerative diseases from normal ageing. However, the underlying mechanisms remain unknown. Extracellular K+ homeostasis is a potential mediator of neuronal injury as K+ elevations increase excitatory activity. The dysregulation of extracellular K+ and potassium channel expressions during neurodegeneration could contribute to this distinction. Here we measured the cortical extracellular K+ concentration ([K+]e) in awake wild-type mice as well as murine models of neurodegeneration using K+-sensitive microelectrodes. Unexpectedly, aged wild-type mice exhibited significantly lower cortical [K+]e than young mice. In contrast, cortical [K+]e was consistently elevated in Alzheimer's disease (APP/PS1), amyotrophic lateral sclerosis (ALS) (SOD1G93A) and Huntington's disease (R6/2) models. Cortical resting [K+]e correlated inversely with neuronal density and the [K+]e buffering rate but correlated positively with the predicted neuronal firing rate. Screening of astrocyte-selective genomic datasets revealed a number of potassium channel genes that were downregulated in these disease models but not in normal ageing. In particular, the inwardly rectifying potassium channel Kcnj10 was downregulated in ALS and Huntington's disease models but not in normal ageing, while Fxyd1 and Slc1a3, each of which acts as a negative regulator of potassium uptake, were each upregulated by astrocytes in both Alzheimer's disease and ALS models. Chronic elevation of [K+]e in response to changes in gene expression and the attendant neuronal hyperexcitability may drive the neuronal loss characteristic of these neurodegenerative diseases. These observations suggest that the dysregulation of extracellular K+ homeostasis in a number of neurodegenerative diseases could be due to aberrant astrocytic K+ buffering and as such, highlight a fundamental role for glial dysfunction in neurodegeneration.
Collapse
Affiliation(s)
- Fengfei Ding
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Pharmacology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qian Sun
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Pharmacology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Carter Long
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Rune Nguyen Rasmussen
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, Neurology Department, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sisi Peng
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Qiwu Xu
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Ning Kang
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Wei Song
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Pia Weikop
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, Neurology Department, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, Neurology Department, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, Neurology Department, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
6
|
Harea GT, Thrailkill M, Garcia I, Beely BM, Wendorff DS, Roberts TR, Golobish TD, Gruda M, Kovacs T, Guliashvili T, Chan PP, Stewart IJ, Chung KK, Guda T, Batchinsky AI. K +ontrol rapidly and efficiently reduces potassium in donor blood during ex vivo circulation. Perfusion 2024; 39:134-141. [PMID: 36196521 DOI: 10.1177/02676591221130175] [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] [Indexed: 11/16/2022]
Abstract
BACKGROUND Patients with kidney failure are at risk for lethal complications from hyperkalemia. Resuscitation, medications, and hemodialysis are used to mitigate increased potassium (K+) levels in circulating blood; however, these approaches may not always be readily available or effective, especially in a resource limited environment. We tested a sorbent cartridge (KC, K+ontrol CytoSorbents Medical Inc., Monmouth Junction, New Jersey) which contains a resin adsorber for K+. The objective of this study was to test the utility of KC in an ex vivo circulation system. We hypothesized that KC reduces K+ levels in extracorporeal circulation of donor swine whole blood infused with KCl. METHODS A six-hour circulation study was carried out using KC, a NxStage (NxStage Medical, Inc., Lawrence, MA) membrane, blood bag containing heparinized whole blood with KCl infusion, 3/16-inch ID tubing, a peristaltic pump, and flow sensors. The NxStage permeate line was connected back to the main circuit in the Control group (n = 6), creating a recirculation loop. For KC group (n = 6), KC was added to the recirculation loop, and a continuous infusion of KCl at 10 mEq/hour was administered for two hours. Blood samples were acquired at baseline and every hour for 6 h. RESULTS In the control group, K+ levels remained at ∼9 mmol/L; 9.1 ± 0.4 mmol/L at 6 h. In the KC group, significant decreases in K+ at hour 1 (4.3 ± 0.3 mmol/L) and were sustained for the experiment duration equilibrating at 4.6 ± 0.4 mmol/L after 6 h (p = 0.042). Main loop blood flow was maintained under 400 mL/min; recirculation loop flow varied between 60 and 70 mL/min in the control group and 45-55 mL/min in the KC group. Decreases in recirculation loop flow in KC group required 7% increase of pump RPM. CONCLUSIONS During ex-vivo extracorporeal circulation using donor swine blood, KC removed approximately 50% of K+, normalizing circulating levels.
Collapse
Affiliation(s)
- George T Harea
- Autonomous Reanimation and Evacuation Research Program, San Antonio, TX, USA
| | - Marianne Thrailkill
- Autonomous Reanimation and Evacuation Research Program, San Antonio, TX, USA
| | - Isabella Garcia
- Autonomous Reanimation and Evacuation Research Program, San Antonio, TX, USA
| | - Brendan M Beely
- Autonomous Reanimation and Evacuation Research Program, San Antonio, TX, USA
- Department of Translational Medicine, School of Osteopathic Medicine, University of the Incarnate Word, San Antonio, TX, USA
| | - Daniel S Wendorff
- Autonomous Reanimation and Evacuation Research Program, San Antonio, TX, USA
- Department of Translational Medicine, School of Osteopathic Medicine, University of the Incarnate Word, San Antonio, TX, USA
| | - Teryn R Roberts
- Autonomous Reanimation and Evacuation Research Program, San Antonio, TX, USA
- Department of Translational Medicine, School of Osteopathic Medicine, University of the Incarnate Word, San Antonio, TX, USA
| | | | | | - Tim Kovacs
- Cytosorbents Inc., Monmouth Junction, NJ, USA
| | | | | | - Ian J Stewart
- Department of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Kevin K Chung
- Department of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Teja Guda
- University of Texas at San Antonio, San Antonio, TX, USA
| | - Andriy I Batchinsky
- Autonomous Reanimation and Evacuation Research Program, San Antonio, TX, USA
- Department of Translational Medicine, School of Osteopathic Medicine, University of the Incarnate Word, San Antonio, TX, USA
| |
Collapse
|
7
|
Dutta S, Iyer KK, Vanhatalo S, Breakspear M, Roberts JA. Mechanisms underlying pathological cortical bursts during metabolic depletion. Nat Commun 2023; 14:4792. [PMID: 37553358 PMCID: PMC10409751 DOI: 10.1038/s41467-023-40437-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/27/2023] [Indexed: 08/10/2023] Open
Abstract
Cortical activity depends upon a continuous supply of oxygen and other metabolic resources. Perinatal disruption of oxygen availability is a common clinical scenario in neonatal intensive care units, and a leading cause of lifelong disability. Pathological patterns of brain activity including burst suppression and seizures are a hallmark of the recovery period, yet the mechanisms by which these patterns arise remain poorly understood. Here, we use computational modeling of coupled metabolic-neuronal activity to explore the mechanisms by which oxygen depletion generates pathological brain activity. We find that restricting oxygen supply drives transitions from normal activity to several pathological activity patterns (isoelectric, burst suppression, and seizures), depending on the potassium supply. Trajectories through parameter space track key features of clinical electrophysiology recordings and reveal how infants with good recovery outcomes track toward normal parameter values, whereas the parameter values for infants with poor outcomes dwell around the pathological values. These findings open avenues for studying and monitoring the metabolically challenged infant brain, and deepen our understanding of the link between neuronal and metabolic activity.
Collapse
Affiliation(s)
- Shrey Dutta
- Brain Modelling Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
- School of Psychological Sciences, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW, Australia.
| | - Kartik K Iyer
- Brain Modelling Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Sampsa Vanhatalo
- Pediatric Research Center, Department of Physiology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Michael Breakspear
- School of Psychological Sciences, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW, Australia
- School of Medicine and Public Health, College of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - James A Roberts
- Brain Modelling Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
8
|
Lu X, Han X, Meirovitch Y, Sjöstedt E, Schalek RL, Lichtman JW. Preserving extracellular space for high-quality optical and ultrastructural studies of whole mammalian brains. CELL REPORTS METHODS 2023; 3:100520. [PMID: 37533653 PMCID: PMC10391564 DOI: 10.1016/j.crmeth.2023.100520] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/08/2023] [Accepted: 06/07/2023] [Indexed: 08/04/2023]
Abstract
Analysis of brain structure, connectivity, and molecular diversity relies on effective tissue fixation. Conventional tissue fixation causes extracellular space (ECS) loss, complicating the segmentation of cellular objects from electron microscopy datasets. Previous techniques for preserving ECS in mammalian brains utilizing high-pressure perfusion can give inconsistent results owing to variations in the hydrostatic pressure within the vasculature. A more reliable fixation protocol that uniformly preserves the ECS throughout whole brains would greatly benefit a wide range of neuroscience studies. Here, we report a straightforward transcardial perfusion strategy that preserves ECS throughout the whole rodent brain. No special setup is needed besides sequential solution changes, and the protocol offers excellent reproducibility. In addition to better capturing tissue ultrastructure, preservation of ECS has many downstream advantages such as accelerating heavy-metal staining for electron microscopy, improving detergent-free immunohistochemistry for correlated light and electron microscopy, and facilitating lipid removal for tissue clearing.
Collapse
Affiliation(s)
- Xiaotang Lu
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Xiaomeng Han
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Yaron Meirovitch
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Evelina Sjöstedt
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Richard L. Schalek
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Jeff W. Lichtman
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| |
Collapse
|
9
|
Robbins EM, Jaquins-Gerstl AS, Okonkwo DO, Boutelle MG, Michael AC. Dexamethasone-Enhanced Continuous Online Microdialysis for Neuromonitoring of O 2 after Brain Injury. ACS Chem Neurosci 2023. [PMID: 37369003 PMCID: PMC10360069 DOI: 10.1021/acschemneuro.2c00703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a major public health crisis in many regions of the world. Severe TBI may cause a primary brain lesion with a surrounding penumbra of tissue that is vulnerable to secondary injury. Secondary injury presents as progressive expansion of the lesion, possibly leading to severe disability, a persistent vegetive state, or death. Real time neuromonitoring to detect and monitor secondary injury is urgently needed. Dexamethasone-enhanced continuous online microdialysis (Dex-enhanced coMD) is an emerging paradigm for chronic neuromonitoring after brain injury. The present study employed Dex-enhanced coMD to monitor brain K+ and O2 during manually induced spreading depolarization in the cortex of anesthetized rats and after controlled cortical impact, a widely used rodent model of TBI, in behaving rats. Consistent with prior reports on glucose, O2 exhibited a variety of responses to spreading depolarization and a prolonged, essentially permanent decline in the days after controlled cortical impact. These findings confirm that Dex-enhanced coMD delivers valuable information regarding the impact of spreading depolarization and controlled cortical impact on O2 levels in the rat cortex.
Collapse
Affiliation(s)
- Elaine M Robbins
- Department of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Andrea S Jaquins-Gerstl
- Department of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - David O Okonkwo
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, United States
| | - Martyn G Boutelle
- Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Adrian C Michael
- Department of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
10
|
Aryal SP, Xia M, Ortinski PI, Richards CI. Study of Calcium Signaling in Astrocytes with a Novel Endoplasmic Reticulum-Targeted GCaMP Sensor. Curr Protoc 2022; 2:e491. [PMID: 35938843 PMCID: PMC9373517 DOI: 10.1002/cpz1.491] [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] [Indexed: 06/15/2023]
Abstract
The endoplasmic reticulum (ER), the major organelle for the storage of Ca2+ , maintains a concentration of Ca2+ much higher than in the cytosol or other subcellular organelles, such as the mitochondria. A variety of tools have been developed for measuring Ca2+ activity in neuronal and glial cells, but most of these sensors target either the plasma membrane (PM) or the cytosol. Though these sensors are important for measuring Ca2+ transients, they lack the capability to measure activity in the periphery of the ER or to measure low-amplitude events resulting from Ca2+ exchange between the ER and other organelles, such as the mitochondria. We recently developed an ER-targeted GCaMP6f anchored to the cytosolic side of the ER that can measure minute calcium exchange occurring in this region. In this article, we discuss detailed methods to characterize the ER-GCaMP6f sensor, utilize it for calcium imaging in cultured astrocytes, and assess its expression and calcium imaging in astrocytes in rodent brains. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Expression and characterization of ER-GCaMP6f Support Protocol 1: ER-GCaMP6f-expressing stable cell line generation Basic Protocol 2: In vitro calcium imaging with ER-GCaMP6f Support Protocol 2: Imaging of drug-induced calcium activity Alternate Protocol 1: Transduction of astrocytes with ER-GCaMP6f AAV Alternate Protocol 2: Calcium imaging of astrocytes with Fluo-4 AM Basic Protocol 3: In vivo ER-GCaMP6f expression and slice calcium imaging Support Protocol 3: Pharmacological studies with 2-APB in brain slices.
Collapse
Affiliation(s)
- Surya P Aryal
- Department of Chemistry, University of Kentucky, Lexington, Kentucky
| | - Mengfan Xia
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
| | - Pavel I Ortinski
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
| | | |
Collapse
|
11
|
Sanchez-Conde FG, Jimenez-Vazquez EN, Auerbach DS, Jones DK. The ERG1 K+ Channel and Its Role in Neuronal Health and Disease. Front Mol Neurosci 2022; 15:890368. [PMID: 35600076 PMCID: PMC9113952 DOI: 10.3389/fnmol.2022.890368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022] Open
Abstract
The ERG1 potassium channel, encoded by KCNH2, has long been associated with cardiac electrical excitability. Yet, a growing body of work suggests that ERG1 mediates physiology throughout the human body, including the brain. ERG1 is a regulator of neuronal excitability, ERG1 variants are associated with neuronal diseases (e.g., epilepsy and schizophrenia), and ERG1 serves as a potential therapeutic target for neuronal pathophysiology. This review summarizes the current state-of-the-field regarding the ERG1 channel structure and function, ERG1’s relationship to the mammalian brain and highlights key questions that have yet to be answered.
Collapse
Affiliation(s)
| | - Eric N. Jimenez-Vazquez
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - David S. Auerbach
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY, United States
- *Correspondence: David S. Auerbach,
| | - David K. Jones
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
- David K. Jones,
| |
Collapse
|
12
|
Eitelmann S, Stephan J, Everaerts K, Durry S, Pape N, Gerkau NJ, Rose CR. Changes in Astroglial K + upon Brief Periods of Energy Deprivation in the Mouse Neocortex. Int J Mol Sci 2022; 23:ijms23094836. [PMID: 35563238 PMCID: PMC9102782 DOI: 10.3390/ijms23094836] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/21/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022] Open
Abstract
Malfunction of astrocytic K+ regulation contributes to the breakdown of extracellular K+ homeostasis during ischemia and spreading depolarization events. Studying astroglial K+ changes is, however, hampered by a lack of suitable techniques. Here, we combined results from fluorescence imaging, ion-selective microelectrodes, and patch-clamp recordings in murine neocortical slices with the calculation of astrocytic [K+]. Brief chemical ischemia caused a reversible ATP reduction and a transient depolarization of astrocytes. Moreover, astrocytic [Na+] increased by 24 mM and extracellular [Na+] decreased. Extracellular [K+] increased, followed by an undershoot during recovery. Feeding these data into the Goldman-Hodgkin-Katz equation revealed a baseline astroglial [K+] of 146 mM, an initial K+ loss by 43 mM upon chemical ischemia, and a transient K+ overshoot of 16 mM during recovery. It also disclosed a biphasic mismatch in astrocytic Na+/K+ balance, which was initially ameliorated, but later aggravated by accompanying changes in pH and bicarbonate, respectively. Altogether, our study predicts a loss of K+ from astrocytes upon chemical ischemia followed by a net gain. The overshooting K+ uptake will promote low extracellular K+ during recovery, likely exerting a neuroprotective effect. The resulting late cation/anion imbalance requires additional efflux of cations and/or influx of anions, the latter eventually driving delayed astrocyte swelling.
Collapse
|
13
|
Du Y, Brennan FH, Popovich PG, Zhou M. Microglia maintain the normal structure and function of the hippocampal astrocyte network. Glia 2022; 70:1359-1379. [PMID: 35394085 PMCID: PMC9324808 DOI: 10.1002/glia.24179] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 03/09/2022] [Accepted: 03/28/2022] [Indexed: 12/14/2022]
Abstract
Microglial control of activity‐dependent plasticity and synaptic remodeling in neuronal networks has been the subject of intense research in the past several years. Although microglia–neuron interactions have been extensively studied, less is known about how microglia influence astrocyte‐dependent control over neuronal structure and function. Here, we explored a role for microglia in regulating the structure and function of the astrocyte syncytium in mouse hippocampus. After depleting microglia using a CSF1R antagonist (PLX5622, Plexxikon), we observed severe disruption of astrocyte syncytial isopotentiality and dye coupling. A decrease in astrocyte‐specific gap junction connexin (Cx) 30 and 43 expression, at least partially accounts for these microglia‐dependent changes in astrocytes. Because neuronal function requires intact astrocyte coupling, we also evaluated the effects of microglia depletion on synaptic transmission in the hippocampus. Without microglia, the strength of synaptic transmission was reduced at baseline and after long‐term potentiation (LTP). Conversely, priming microglia with systemic injections of lipopolysaccharide enhanced CA3‐CA1 synaptic transmission. This microglia‐induced scaling of synaptic transmission was associated with increased expression of post‐synaptic scaffold proteins (Homer1) in CA1. However, astrocyte network function was not affected by microglia priming, indicating that microglia‐dependent effects on astrocytes and neurons vary across functional states. Through manipulation of microglia in the brain, our results reveal the importance of microglia in homeostatic regulation of the astrocyte syncytium and scaling of synaptic transmission. These novel mechanisms uncover a new direction for future studies interrogating microglia function in various physiological and pathological contexts.
Collapse
Affiliation(s)
- Yixing Du
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Faith H Brennan
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.,Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Phillip G Popovich
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.,Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Min Zhou
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
14
|
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: 26] [Impact Index Per Article: 6.5] [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
|
15
|
Piccialli I, Tedeschi V, Boscia F, Ciccone R, Casamassa A, de Rosa V, Grieco P, Secondo A, Pannaccione A. The Anemonia sulcata Toxin BDS-I Protects Astrocytes Exposed to Aβ 1-42 Oligomers by Restoring [Ca 2+] i Transients and ER Ca 2+ Signaling. Toxins (Basel) 2020; 13:20. [PMID: 33396295 PMCID: PMC7823622 DOI: 10.3390/toxins13010020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 12/28/2022] Open
Abstract
Intracellular calcium concentration ([Ca2+]i) transients in astrocytes represent a highly plastic signaling pathway underlying the communication between neurons and glial cells. However, how this important phenomenon may be compromised in Alzheimer's disease (AD) remains unexplored. Moreover, the involvement of several K+ channels, including KV3.4 underlying the fast-inactivating currents, has been demonstrated in several AD models. Here, the effect of KV3.4 modulation by the marine toxin blood depressing substance-I (BDS-I) extracted from Anemonia sulcata has been studied on [Ca2+]i transients in rat primary cortical astrocytes exposed to Aβ1-42 oligomers. We showed that: (1) primary cortical astrocytes expressing KV3.4 channels displayed [Ca2+]i transients depending on the occurrence of membrane potential spikes, (2) BDS-I restored, in a dose-dependent way, [Ca2+]i transients in astrocytes exposed to Aβ1-42 oligomers (5 µM/48 h) by inhibiting hyperfunctional KV3.4 channels, (3) BDS-I counteracted Ca2+ overload into the endoplasmic reticulum (ER) induced by Aβ1-42 oligomers, (4) BDS-I prevented the expression of the ER stress markers including active caspase 12 and GRP78/BiP in astrocytes treated with Aβ1-42 oligomers, and (5) BDS-I prevented Aβ1-42-induced reactive oxygen species (ROS) production and cell suffering measured as mitochondrial activity and lactate dehydrogenase (LDH) release. Collectively, we proposed that the marine toxin BDS-I, by inhibiting the hyperfunctional KV3.4 channels and restoring [Ca2+]i oscillation frequency, prevented Aβ1-42-induced ER stress and cell suffering in astrocytes.
Collapse
Affiliation(s)
- Ilaria Piccialli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, 80131 Napoli, Italy; (I.P.); (V.T.); (F.B.); (R.C.); (A.C.); (V.d.R.)
| | - Valentina Tedeschi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, 80131 Napoli, Italy; (I.P.); (V.T.); (F.B.); (R.C.); (A.C.); (V.d.R.)
| | - Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, 80131 Napoli, Italy; (I.P.); (V.T.); (F.B.); (R.C.); (A.C.); (V.d.R.)
| | - Roselia Ciccone
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, 80131 Napoli, Italy; (I.P.); (V.T.); (F.B.); (R.C.); (A.C.); (V.d.R.)
| | - Antonella Casamassa
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, 80131 Napoli, Italy; (I.P.); (V.T.); (F.B.); (R.C.); (A.C.); (V.d.R.)
| | - Valeria de Rosa
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, 80131 Napoli, Italy; (I.P.); (V.T.); (F.B.); (R.C.); (A.C.); (V.d.R.)
| | - Paolo Grieco
- Department of Pharmacy, School of Medicine, Federico II Universityof Naples, 80131 Napoli, Italy;
| | - Agnese Secondo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, 80131 Napoli, Italy; (I.P.); (V.T.); (F.B.); (R.C.); (A.C.); (V.d.R.)
| | - Anna Pannaccione
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, 80131 Napoli, Italy; (I.P.); (V.T.); (F.B.); (R.C.); (A.C.); (V.d.R.)
| |
Collapse
|
16
|
Koumangoye R, Bastarache L, Delpire E. NKCC1: Newly Found as a Human Disease-Causing Ion Transporter. FUNCTION 2020; 2:zqaa028. [PMID: 33345190 PMCID: PMC7727275 DOI: 10.1093/function/zqaa028] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 01/06/2023] Open
Abstract
Among the electroneutral Na+-dependent chloride transporters, NKCC1 had until now evaded identification as a protein causing human diseases. The closely related SLC12A transporters, NKCC2 and NCC have been identified some 25 years ago as responsible for Bartter and Gitelman syndromes: two renal-dependent salt wasting disorders. Absence of disease was most surprising since the NKCC1 knockout mouse was shown in 1999 to be viable, albeit with a wide range of deleterious phenotypes. Here we summarize the work of the past 5 years that introduced us to clinical cases involving NKCC1. The most striking cases are of 3 children with inherited mutations, who have complete absence of NKCC1 expression. These cases establish that lack of NKCC1 causes deafness; CFTR-like secretory defects with mucus accumulation in lung and intestine; severe xerostomia, hypotonia, dysmorphic facial features, and severe neurodevelopmental disorder. Another intriguing case is of a patient with a dominant deleterious SLC12A2 allele. This de novo mutation introduced a premature stop codon leading to a truncated protein. This mutant transporter seems to exert dominant-negative effect on wild-type transporter only in epithelial cells. The patient who suffers from lung, bladder, intestine, pancreas, and multiple endocrine abnormalities has, however, normal hearing and cognition. Finally, new reports substantiate the haploinsufficiency prediction of the SLC12A2 gene. Cases with single allele mutations in SLC12A2 have been linked to hearing loss and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Rainelli Koumangoye
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA,Corresponding author. E-mail:
| |
Collapse
|
17
|
Abstract
Cerebral edema is a pathological hallmark of various central nervous system (CNS) insults, including traumatic brain injury (TBI) and excitotoxic injury such as stroke. Due to the rigidity of the skull, edema-induced increase of intracranial fluid significantly complicates severe CNS injuries by raising intracranial pressure and compromising perfusion. Mortality due to cerebral edema is high. With mortality rates up to 80% in severe cases of stroke, it is the leading cause of death within the first week. Similarly, cerebral edema is devastating for patients of TBI, accounting for up to 50% mortality. Currently, the available treatments for cerebral edema include hypothermia, osmotherapy, and surgery. However, these treatments only address the symptoms and often elicit adverse side effects, potentially in part due to non-specificity. There is an urgent need to identify effective pharmacological treatments for cerebral edema. Currently, ion channels represent the third-largest target class for drug development, but their roles in cerebral edema remain ill-defined. The present review aims to provide an overview of the proposed roles of ion channels and transporters (including aquaporins, SUR1-TRPM4, chloride channels, glucose transporters, and proton-sensitive channels) in mediating cerebral edema in acute ischemic stroke and TBI. We also focus on the pharmacological inhibitors for each target and potential therapeutic strategies that may be further pursued for the treatment of cerebral edema.
Collapse
|
18
|
Jiang M, Li R, Lyu J, Li X, Wang W, Wang Z, Sheng H, Zhang W, Karhausen J, Yang W. MCC950, a selective NLPR3 inflammasome inhibitor, improves neurologic function and survival after cardiac arrest and resuscitation. J Neuroinflammation 2020; 17:256. [PMID: 32867797 PMCID: PMC7457538 DOI: 10.1186/s12974-020-01933-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022] Open
Abstract
Background Cardiac arrest (CA) is associated with high morbidity and mortality, even after spontaneous circulation is re-established. This dire situation is partly due to post-CA syndrome for which no specific and effective intervention is available. One key component of post-CA syndrome is sterile inflammation, which affects various organs including the brain. A major effector of sterile inflammation is activated NLRP3 inflammasome, which leads to increased release of interleukin (IL)-1β. However, how NLRP3 inflammasome impacts neuroinflammation and neurologic outcome after CA is largely undefined. Methods Mice were subjected to a potassium-based murine CA and cardiopulmonary resuscitation (CPR) model. MCC950 was used to suppress activation of NLRP3 inflammasome after CA/CPR. Levels of protein and mRNA were examined by Western blotting and quantitative PCR, respectively. Immunologic changes were assessed by measuring cytokine expression and immune cell compositions. CA outcomes, including neurologic deficits, bacterial load in the lung, and survival rate, were evaluated. Results Using our CA/CPR model, we found that NLRP3 inflammasome was activated in the post-CA brain, and that pro-inflammatory cytokine levels, including IL-1β, were increased. After treatment with MCC950, a potent and selective NLRP3 inflammasome inhibitor, mice exhibited improved functional recovery and survival rate during the 14-day observational period after CA/CPR. In line with these findings, IL-1β mRNA levels in the post-CA brain were significantly suppressed after MCC950 treatment. Interestingly, we also found that in MCC950- vs. vehicle-treated CA mice, immune homeostasis in the spleen was better preserved and bacterial load in the lung was significantly reduced. Conclusions Our data demonstrate that activation of NLRP3 inflammasome could be a key event shaping the post-CA immuno- and neuro-pathology, and identify this pathway as a unique and promising therapeutic target to improve outcomes after CA/CPR.
Collapse
Affiliation(s)
- Maorong Jiang
- Department of Anesthesiology, Center for Perioperative Organ Protection, Duke University Medical Center, Durham, NC, USA.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ran Li
- Department of Anesthesiology, Center for Perioperative Organ Protection, Duke University Medical Center, Durham, NC, USA
| | - Jingjun Lyu
- Department of Anesthesiology, Center for Perioperative Organ Protection, Duke University Medical Center, Durham, NC, USA.,Department of Emergency Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xuan Li
- Department of Anesthesiology, Center for Perioperative Organ Protection, Duke University Medical Center, Durham, NC, USA
| | - Wei Wang
- Department of Anesthesiology, Center for Perioperative Organ Protection, Duke University Medical Center, Durham, NC, USA
| | - Zhuoran Wang
- Department of Anesthesiology, Center for Perioperative Organ Protection, Duke University Medical Center, Durham, NC, USA
| | - Huaxin Sheng
- Department of Anesthesiology, Center for Perioperative Organ Protection, Duke University Medical Center, Durham, NC, USA
| | - Weiguo Zhang
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Jörn Karhausen
- Department of Anesthesiology, Center for Perioperative Organ Protection, Duke University Medical Center, Durham, NC, USA
| | - Wei Yang
- Department of Anesthesiology, Center for Perioperative Organ Protection, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
19
|
Gerkau NJ, Rakers C, Durry S, Petzold GC, Rose CR. Reverse NCX Attenuates Cellular Sodium Loading in Metabolically Compromised Cortex. Cereb Cortex 2019; 28:4264-4280. [PMID: 29136153 DOI: 10.1093/cercor/bhx280] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 10/04/2017] [Indexed: 01/05/2023] Open
Abstract
In core regions of ischemic stroke, disruption of blood flow causes breakdown of ionic gradients and, ultimately, calcium overload and cell death. In the surrounding penumbra, cells may recover upon reperfusion, but recovery is hampered by additional metabolic demands imposed by peri-infarct depolarizations (PIDs). There is evidence that sodium influx drives PIDs, but no data exist on PID-related sodium accumulations in vivo. Here, we found that PIDs in mouse neocortex are associated with propagating sodium elevations in neurons and astrocytes. Similar transient sodium elevations were induced in acute tissue slices by brief chemical ischemia. Blocking NMDA-receptors dampened sodium and accompanying calcium loads of neurons in tissue slices, while inhibiting glutamate transport diminished sodium influx into astrocytes, but amplified neuronal sodium loads. In both cell types, inhibition of sodium/calcium exchange (NCX) increased sodium transients. Blocking NCX also significantly reduced calcium transients, a result confirmed in vivo. Our study provides the first quantitative data on sodium elevations in peri-infarct regions in vivo. They suggest that sodium influx drives reversal of NCX, triggering a massive secondary calcium elevation while promoting export of sodium. Reported neuroprotective effects of NCX activity in stroke models might thus be related to its dampening of ischemia-induced sodium loading.
Collapse
Affiliation(s)
- Niklas J Gerkau
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Universitaetsstrasse 1, Duesseldorf, Germany
| | - Cordula Rakers
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, Bonn, Germany
| | - Simone Durry
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Universitaetsstrasse 1, Duesseldorf, Germany
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, Bonn, Germany.,Department of Neurology, University Hospital Bonn, Sigmund-Freud-Str. 25, Bonn, Germany
| | - Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Universitaetsstrasse 1, Duesseldorf, Germany
| |
Collapse
|
20
|
Weiss S, Melom JE, Ormerod KG, Zhang YV, Littleton JT. Glial Ca 2+signaling links endocytosis to K + buffering around neuronal somas to regulate excitability. eLife 2019; 8:44186. [PMID: 31025939 PMCID: PMC6510531 DOI: 10.7554/elife.44186] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/25/2019] [Indexed: 12/30/2022] Open
Abstract
Glial-neuronal signaling at synapses is widely studied, but how glia interact with neuronal somas to regulate their activity is unclear. Drosophila cortex glia are restricted to brain regions devoid of synapses, providing an opportunity to characterize interactions with neuronal somas. Mutations in the cortex glial NCKXzydeco elevate basal Ca2+, predisposing animals to seizure-like behavior. To determine how cortex glial Ca2+ signaling controls neuronal excitability, we performed an in vivo modifier screen of the NCKXzydeco seizure phenotype. We show that elevation of glial Ca2+ causes hyperactivation of calcineurin-dependent endocytosis and accumulation of early endosomes. Knockdown of sandman, a K2P channel, recapitulates NCKXzydeco seizures. Indeed, sandman expression on cortex glial membranes is substantially reduced in NCKXzydeco mutants, indicating enhanced internalization of sandman predisposes animals to seizures. These data provide an unexpected link between glial Ca2+ signaling and the well-known role of glia in K+ buffering as a key mechanism for regulating neuronal excitability.
Collapse
Affiliation(s)
- Shirley Weiss
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Jan E Melom
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Kiel G Ormerod
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Yao V Zhang
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - J Troy Littleton
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
21
|
Chmelova M, Sucha P, Bochin M, Vorisek I, Pivonkova H, Hermanova Z, Anderova M, Vargova L. The role of aquaporin-4 and transient receptor potential vaniloid isoform 4 channels in the development of cytotoxic edema and associated extracellular diffusion parameter changes. Eur J Neurosci 2019; 50:1685-1699. [PMID: 30633415 DOI: 10.1111/ejn.14338] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 12/27/2018] [Accepted: 01/04/2019] [Indexed: 11/30/2022]
Abstract
The proper function of the nervous system is dependent on the balance of ions and water between the intracellular and extracellular space (ECS). It has been suggested that the interaction of aquaporin-4 (AQP4) and the transient receptor potential vaniloid isoform 4 (TRPV4) channels play a role in water balance and cell volume regulation, and indirectly, of the ECS volume. Using the real-time iontophoretic method, we studied the changes of the ECS diffusion parameters: ECS volume fraction α (α = ECS volume fraction/total tissue volume) and tortuosity λ (λ2 = free/apparent diffusion coefficient) in mice with a genetic deficiency of AQP4 or TRPV4 channels, and in control animals. The used models of cytotoxic edema included: mild and severe hypotonic stress or oxygen-glucose deprivation (OGD) in situ and terminal ischemia/anoxia in vivo. This study shows that an AQP4 or TRPV4 deficit slows down the ECS volume shrinkage during severe ischemia in vivo. We further demonstrate that a TRPV4 deficit slows down the velocity and attenuates an extent of the ECS volume decrease during OGD treatment in situ. However, in any of the cytotoxic edema models in situ (OGD, mild or severe hypotonic stress), we did not detect any alterations in the cell swelling or volume regulation caused by AQP4 deficiency. Overall, our results indicate that the AQP4 and TRPV4 channels may play a crucial role in severe pathological states associated with their overexpression and enhanced cell swelling. However, detailed interplay between AQP4 and TRPV4 channels requires further studies and additional research.
Collapse
Affiliation(s)
- Martina Chmelova
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic.,Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - Petra Sucha
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic.,Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - Marcel Bochin
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic.,Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - Ivan Vorisek
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - Helena Pivonkova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - Zuzana Hermanova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - Miroslava Anderova
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic.,Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - Lydia Vargova
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic.,Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| |
Collapse
|
22
|
Susceptibility of the cerebral cortex to spreading depolarization in neurological disease states: The impact of aging. Neurochem Int 2018; 127:125-136. [PMID: 30336178 DOI: 10.1016/j.neuint.2018.10.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/10/2018] [Accepted: 10/13/2018] [Indexed: 12/17/2022]
Abstract
Secondary injury following acute brain insults significantly contributes to poorer neurological outcome. The spontaneous, recurrent occurrence of spreading depolarization events (SD) has been recognized as a potent secondary injury mechanism in subarachnoid hemorrhage, malignant ischemic stroke and traumatic brain injury. In addition, SD is the underlying mechanism of the aura symptoms of migraineurs. The susceptibility of the nervous tissue to SD is subject to the metabolic status of the tissue, the ionic composition of the extracellular space, and the functional status of ion pumps, voltage-gated and other cation channels, glutamate receptors and excitatory amino acid transporters. All these mechanisms tune the excitability of the nervous tissue. Aging has also been found to alter SD susceptibility, which appears to be highest at young adulthood, and decline over the aging process. The lower susceptibility of the cerebral gray matter to SD in the old brain may be caused by the age-related impairment of mechanisms implicated in ion translocations between the intra- and extracellular compartments, glutamate signaling and surplus potassium and glutamate clearance. Even though the aging nervous tissue is thus less able to sustain SD, the consequences of SD recurrence in the old brain have proven to be graver, possibly leading to accelerated lesion maturation. Taken that recurrent SDs may pose an increased burden in the aging injured brain, the benefit of therapeutic approaches to restrict SD generation and propagation may be particularly relevant for elderly patients.
Collapse
|
23
|
Sadeghian H, Lacoste B, Qin T, Toussay X, Rosa R, Oka F, Chung DY, Takizawa T, Gu C, Ayata C. Spreading depolarizations trigger caveolin-1-dependent endothelial transcytosis. Ann Neurol 2018; 84:409-423. [PMID: 30014540 PMCID: PMC6153037 DOI: 10.1002/ana.25298] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 07/08/2018] [Accepted: 07/11/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Cortical spreading depolarizations (CSDs) are intense and ubiquitous depolarization waves relevant for the pathophysiology of migraine and brain injury. CSDs disrupt the blood-brain barrier (BBB), but the mechanisms are unknown. METHODS A total of six CSDs were evoked over 1 hour by topical application of 300 mM of KCl or optogenetically with 470 nm (blue) LED over the right hemisphere in anesthetized mice (C57BL/6 J wild type, Thy1-ChR2-YFP line 18, and cav-1-/- ). BBB disruption was assessed by Evans blue (2% EB, 3 ml/kg, intra-arterial) or dextran (200 mg/kg, fluorescein, 70,000 MW, intra-arterial) extravasation in parietotemporal cortex at 3 to 24 hours after CSD. Endothelial cell ultrastructure was examined using transmission electron microscopy 0 to 24 hours after the same CSD protocol in order to assess vesicular trafficking, endothelial tight junctions, and pericyte integrity. Mice were treated with vehicle, isoform nonselective rho-associated kinase (ROCK) inhibitor fasudil (10 mg/kg, intraperitoneally 30 minutes before CSD), or ROCK-2 selective inhibitor KD025 (200 mg/kg, per oral twice-daily for 5 doses before CSD). RESULTS We show that CSD-induced BBB opening to water and large molecules is mediated by increased endothelial transcytosis starting between 3 and 6 hours and lasting approximately 24 hours. Endothelial tight junctions, pericytes, and basement membrane remain preserved after CSDs. Moreover, we show that CSD-induced BBB disruption is exclusively caveolin-1-dependent and requires rho-kinase 2 activity. Importantly, hyperoxia failed to prevent CSD-induced BBB breakdown, suggesting that the latter is independent of tissue hypoxia. INTERPRETATION Our data elucidate the mechanisms by which CSDs lead to transient BBB disruption, with diagnostic and therapeutic implications for migraine and brain injury.
Collapse
Affiliation(s)
- Homa Sadeghian
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Baptiste Lacoste
- The Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, ON, Canada
- The University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Tao Qin
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Xavier Toussay
- The Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, ON, Canada
| | - Roberto Rosa
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Fumiaki Oka
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - David Y Chung
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Tsubasa Takizawa
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Chenghua Gu
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Cenk Ayata
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
24
|
Abstract
Brain waves are rhythmic voltage oscillations emerging from the synchronization of individual neurons into a neuronal network. These oscillations range from slow to fast fluctuations, and are classified by power and frequency band, with different frequency bands being associated with specific behaviours. It has been postulated that at least ten distinct mechanisms are required to cover the frequency range of neural oscillations, however the mechanisms that gear the transition between distinct oscillatory frequencies are unknown. In this study, we have used electrophysiological recordings to explore the involvement of astrocytic K+ clearance processes in modulating neural oscillations at both network and cellular levels. Our results indicate that impairment of astrocytic K+ clearance capabilities, either through blockade of K+ uptake or astrocytic connectivity, enhance network excitability and form high power network oscillations over a wide range of frequencies. At the cellular level, local increases in extracellular K+ results in modulation of the oscillatory behaviour of individual neurons, which underlies the network behaviour. Since astrocytes are central for maintaining K+ homeostasis, our study suggests that modulation of their inherent capabilities to clear K+ from the extracellular milieu is a potential mechanism to optimise neural resonance behaviour and thus tune neural oscillations.
Collapse
|
25
|
Bylicky MA, Mueller GP, Day RM. Mechanisms of Endogenous Neuroprotective Effects of Astrocytes in Brain Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6501031. [PMID: 29805731 PMCID: PMC5901819 DOI: 10.1155/2018/6501031] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/19/2018] [Indexed: 12/11/2022]
Abstract
Astrocytes, once believed to serve only as "glue" for the structural support of neurons, have been demonstrated to serve critical functions for the maintenance and protection of neurons, especially under conditions of acute or chronic injury. There are at least seven distinct mechanisms by which astrocytes protect neurons from damage; these are (1) protection against glutamate toxicity, (2) protection against redox stress, (3) mediation of mitochondrial repair mechanisms, (4) protection against glucose-induced metabolic stress, (5) protection against iron toxicity, (6) modulation of the immune response in the brain, and (7) maintenance of tissue homeostasis in the presence of DNA damage. Astrocytes support these critical functions through specialized responses to stress or toxic conditions. The detoxifying activities of astrocytes are essential for maintenance of the microenvironment surrounding neurons and in whole tissue homeostasis. Improved understanding of the mechanisms by which astrocytes protect the brain could lead to the development of novel targets for the development of neuroprotective strategies.
Collapse
Affiliation(s)
- Michelle A. Bylicky
- Department of Anatomy, Physiology, and Genetics, The Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Gregory P. Mueller
- Department of Anatomy, Physiology, and Genetics, The Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Regina M. Day
- Department of Pharmacology and Molecular Therapeutics, The Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| |
Collapse
|
26
|
Du Y, Wang W, Lutton AD, Kiyoshi CM, Ma B, Taylor AT, Olesik JW, McTigue DM, Askwith CC, Zhou M. Dissipation of transmembrane potassium gradient is the main cause of cerebral ischemia-induced depolarization in astrocytes and neurons. Exp Neurol 2018; 303:1-11. [PMID: 29407729 DOI: 10.1016/j.expneurol.2018.01.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/02/2018] [Accepted: 01/25/2018] [Indexed: 01/16/2023]
Abstract
Membrane potential (VM) depolarization occurs immediately following cerebral ischemia and is devastating for the astrocyte homeostasis and neuronal signaling. Previously, an excessive release of extracellular K+ and glutamate has been shown to underlie an ischemia-induced VM depolarization. Ischemic insults should impair membrane ion channels and disrupt the physiological ion gradients. However, their respective contribution to ischemia-induced neuronal and glial depolarization and loss of neuronal excitability are unanswered questions. A short-term oxygen-glucose deprivation (OGD) was used for the purpose of examining the acute effect of ischemic conditions on ion channel activity and physiological K+ gradient in neurons and glial cells. We show that a 30 min OGD treatment exerted no measurable damage to the function of membrane ion channels in neurons, astrocytes, and NG2 glia. As a result of the resilience of membrane ion channels, neuronal spikes last twice as long as our previously reported 15 min time window. In the electrophysiological analysis, a 30 min OGD-induced dissipation of transmembrane K+ gradient contributed differently in brain cell depolarization: severe in astrocytes and neurons, and undetectable in NG2 glia. The discrete cellular responses to OGD corresponded to a total loss of 69% of the intracellular K+ contents in hippocampal slices as measured by Inductively Coupled Plasma Mass Spectrometry (ICP-MS). A major brain cell depolarization mechanism identified here is important for our understanding of cerebral ischemia pathology. Additionally, further understanding of the resilient response of NG2 glia to ischemia-induced intracellular K+ loss and depolarization should facilitate the development of future stroke therapy.
Collapse
Affiliation(s)
- Yixing Du
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Wei Wang
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Physiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Anthony D Lutton
- Trace Element Research Laboratory, The Ohio State University, Columbus, OH 43210, USA
| | - Conrad M Kiyoshi
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Baofeng Ma
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Anne T Taylor
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - John W Olesik
- Trace Element Research Laboratory, The Ohio State University, Columbus, OH 43210, USA
| | - Dana M McTigue
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Candice C Askwith
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Min Zhou
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
27
|
Abstract
Oncotic cell death or oncosis represents a major mechanism of cell death in ischaemic stroke, occurring in many central nervous system (CNS) cell types including neurons, glia and vascular endothelial cells. In stroke, energy depletion causes ionic pump failure and disrupts ionic homeostasis. Imbalance between the influx of Na+ and Cl- ions and the efflux of K+ ions through various channel proteins and transporters creates a transmembrane osmotic gradient, with ensuing movement of water into the cells, resulting in cell swelling and oncosis. Oncosis is a key mediator of cerebral oedema in ischaemic stroke, contributing directly through cytotoxic oedema, and indirectly through vasogenic oedema by causing vascular endothelial cell death and disruption of the blood-brain barrier (BBB). Hence, inhibition of uncontrolled ionic flux represents a novel and powerful strategy in achieving neuroprotection in stroke. In this review, we provide an overview of oncotic cell death in the pathology of stroke. Importantly, we summarised the therapeutically significant pathways of water, Na+, Cl- and K+ movement across cell membranes in the CNS and their respective roles in the pathobiology of stroke.
Collapse
|
28
|
Helleringer R, Chever O, Daniel H, Galante M. Oxygen and Glucose Deprivation Induces Bergmann Glia Membrane Depolarization and Ca 2+ Rises Mainly Mediated by K + and ATP Increases in the Extracellular Space. Front Cell Neurosci 2017; 11:349. [PMID: 29163059 PMCID: PMC5675856 DOI: 10.3389/fncel.2017.00349] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 10/20/2017] [Indexed: 01/24/2023] Open
Abstract
During brain ischemia, intense energy deficiency induces a complex succession of events including pump failure, acidosis and exacerbated glutamate release. In the cerebellum, glutamate is the principal mediator of Purkinje neuron anoxic depolarization during episodes of oxygen and glucose deprivation (OGD). Here, the impact of OGD is studied in Bergmann glia, specialized astrocytes closely associated to Purkinje neurons. Patch clamp experiments reveal that during OGD Bergmann glial cells develop a large depolarizing current that is not mediated by glutamate and purinergic receptors but is mainly due to the accumulation of K+ in the extracellular space. Furthermore, we also found that increases in the intracellular Ca2+ concentration appear in Bergmann glia processes several minutes following OGD. These elevations require, in an early phase, Ca2+ mobilization from internal stores via P2Y receptor activation, and, over longer periods, Ca2+ entry through store-operated calcium channels. Our results suggest that increases of K+ and ATP concentrations in the extracellular space are primordial mediators of the OGD effects on Bergmann glia. In the cerebellum, glial responses to energy deprivation-triggering events are therefore highly likely to follow largely distinct rules from those of their neuronal counterparts.
Collapse
Affiliation(s)
- Romain Helleringer
- Pharmacology and Biochemistry of the Synapse, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Université Paris-Sud, CNRS, UMR 9197, Orsay, France
| | - Oana Chever
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, UMR 7241, INSERM U1050, Labex Memolife, PSL Research University Paris, Paris, France
| | - Hervé Daniel
- Pharmacology and Biochemistry of the Synapse, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Université Paris-Sud, CNRS, UMR 9197, Orsay, France
| | - Micaela Galante
- Pharmacology and Biochemistry of the Synapse, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Université Paris-Sud, CNRS, UMR 9197, Orsay, France
| |
Collapse
|
29
|
Roque C, Baltazar G. Impact of Astrocytes on the Injury Induced by In Vitro Ischemia. Cell Mol Neurobiol 2017; 37:1521-1528. [PMID: 28315110 PMCID: PMC11482236 DOI: 10.1007/s10571-017-0483-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/12/2017] [Indexed: 12/29/2022]
Abstract
Cell cultures are characterized by their simplicity, controllability, and ability to provide detailed basic information on how a particular cell population responds to specific stimuli or insult. These characteristics led to their extensive application in the study of molecular interactions and represent a valuable tool in the study of different pathologies. However, due to the lack of interactions between the different components that form an in vivo system, the results obtained in pure cell cultures not always translate what occurs in vivo. In this context, the use of co-cultures has the advantage of allowing the study of interactions between different types of cells present in a tissue, which in many situations are determinant for the effects obtained. The present study aimed to characterize cortical neuron-glia and neuron-enriched primary cultures and evaluate their response to an ischemic insult. Cell viability was assessed by the MTT assay and cell number/phenotype was analyzed by immunocytochemistry in control cultures and in cells subjected to 4 h of oxygen and glucose deprivation. The results obtained demonstrate that astrocytes have a substantial impact on the injury induced by an ischemic insult, thus suggesting that the crosstalk between glia and neurons is crucial to the neuronal protection in conditions of ischemia.
Collapse
Affiliation(s)
- Cláudio Roque
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal
- Instituto Politécnico de Castelo Branco, Escola Superior de Saúde Dr. Lopes Dias, IPCB-ESALD, Castelo Branco, Portugal
| | - Graça Baltazar
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal.
- Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
30
|
Ellman DG, Isaksen TJ, Lund MC, Dursun S, Wirenfeldt M, Jørgensen LH, Lykke-Hartmann K, Lambertsen KL. The loss-of-function disease-mutation G301R in the Na +/K +-ATPase α 2 isoform decreases lesion volume and improves functional outcome after acute spinal cord injury in mice. BMC Neurosci 2017; 18:66. [PMID: 28886701 PMCID: PMC5590116 DOI: 10.1186/s12868-017-0385-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 09/01/2017] [Indexed: 12/12/2022] Open
Abstract
Background The Na+/K+-ATPases are transmembrane ion pumps important for maintenance of ion gradients across the plasma membrane that serve to support multiple cellular functions, such as membrane potentials, regulation of cellular volume and pH, and co-transport of signaling transmitters in all animal cells. The α2Na+/K+-ATPase subunit isoform is predominantly expressed in astrocytes, which us the sharp Na+-gradient maintained by the sodium pump necessary for astroglial metabolism. Prolonged ischemia induces an elevation of [Na+]i, decreased ATP levels and intracellular pH owing to anaerobic metabolism and lactate accumulation. During ischemia, Na+/K+-ATPase-related functions will naturally increase the energy demand of the Na+/K+-ATPase ion pump. However, the role of the α2Na+/K+-ATPase in contusion injury to the spinal cord remains unknown. We used mice heterozygous mice for the loss-of-function disease-mutation G301R in the Atp1a2 gene (α2+/G301R) to study the effect of reduced α2Na+/K+-ATPase expression in a moderate contusion spinal cord injury (SCI) model. Results We found that α2+/G301R mice display significantly improved functional recovery and decreased lesion volume compared to littermate controls (α2+/+) 7 days after SCI. The protein level of the α1 isoform was significantly increased, in contrast to the α3 isoform that significantly decreased 3 days after SCI in both α2+/G301R and α2+/+ mice. The level of the α2 isoform was significantly decreased in α2+/G301R mice both under naïve conditions and 3 days after SCI compared to α2+/+ mice. We found no differences in astroglial aquaporin 4 levels and no changes in the expression of chemokines (CCL2, CCL5 and CXCL1) and cytokines (TNF, IL-6, IL-1β, IL-10 and IL-5) between genotypes, just as no apparent differences were observed in location and activation of CD45 and F4/80 positive microglia and infiltrating leukocytes. Conclusion Our proof of concept study demonstrates that reduced expression of the α2 isoform in the spinal cord is protective following SCI. Importantly, the BMS and lesion volume were assessed at 7 days after SCI, and longer time points after SCI were not evaluated. However, the α2 isoform is a potential possible target of therapeutic strategies for the treatment of SCI.
Collapse
Affiliation(s)
- Ditte Gry Ellman
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense C, Denmark
| | - Toke Jost Isaksen
- Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark.,Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Danish National Research Foundation, Aarhus University, 8000, Aarhus C, Denmark
| | - Minna Christiansen Lund
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense C, Denmark
| | - Safinaz Dursun
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense C, Denmark
| | - Martin Wirenfeldt
- Department of Pathology, University of Southern Denmark/Odense University Hospital, Odense, 5000, Odense C, Denmark.,Department of Clinical Research, University of Southern Denmark/Odense University Hospital, Odense, 5000, Odense C, Denmark
| | - Louise Helskov Jørgensen
- Department of Pathology, University of Southern Denmark/Odense University Hospital, Odense, 5000, Odense C, Denmark.,Department of Clinical Research, University of Southern Denmark/Odense University Hospital, Odense, 5000, Odense C, Denmark
| | - Karin Lykke-Hartmann
- Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark. .,Department of Clinical Medicine, Aarhus University, 8000, Aarhus C, Denmark. .,Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Danish National Research Foundation, Aarhus University, 8000, Aarhus C, Denmark. .,Department of Clinical Genetics, Aarhus University Hospital, 8000, Aarhus C, Denmark.
| | - Kate Lykke Lambertsen
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense C, Denmark. .,Department of Neurology, Odense University Hospital, 5000, Odense C, Denmark. .,BRIDGE, Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, 5000, Odense C, Denmark.
| |
Collapse
|
31
|
Astrocytic modulation of neuronal excitability through K + spatial buffering. Neurosci Biobehav Rev 2017; 77:87-97. [PMID: 28279812 DOI: 10.1016/j.neubiorev.2017.03.002] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 03/05/2017] [Accepted: 03/05/2017] [Indexed: 11/22/2022]
Abstract
The human brain contains two major cell populations, neurons and glia. While neurons are electrically excitable and capable of discharging short voltage pulses known as action potentials, glial cells are not. However, astrocytes, the prevailing subtype of glia in the cortex, are highly connected and can modulate the excitability of neurons by changing the concentration of potassium ions in the extracellular environment, a process called K+ clearance. During the past decade, astrocytes have been the focus of much research, mainly due to their close association with synapses and their modulatory impact on neuronal activity. It has been shown that astrocytes play an essential role in normal brain function including: nitrosative regulation of synaptic release in the neocortex, synaptogenesis, synaptic transmission and plasticity. Here, we discuss the role of astrocytes in network modulation through their K+ clearance capabilities, a theory that was first raised 50 years ago by Orkand and Kuffler. We will discuss the functional alterations in astrocytic activity that leads to aberrant modulation of network oscillations and synchronous activity.
Collapse
|
32
|
Seifert G, Henneberger C, Steinhäuser C. Diversity of astrocyte potassium channels: An update. Brain Res Bull 2016; 136:26-36. [PMID: 27965079 DOI: 10.1016/j.brainresbull.2016.12.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 12/08/2016] [Accepted: 12/09/2016] [Indexed: 01/08/2023]
Abstract
Astrocyte K+ channels and the K+ currents they mediate dwarf all other transmembrane conductances in these cells. This defining feature of astrocytes and its functional implications have been investigated intensely over the past decades. Nonetheless, many aspects of astrocyte K+ handling and signaling remain incompletely understood. In this review, we provide an update on the diversity of K+ channels expressed by astrocytes and new functional implications. We focus on inwardly-rectifying K+ channels (particularly Kir4.1), two-pore K+ channels and voltage and Ca2+-dependent K+ channels. We further discuss new insights into the involvement of these K+ channels in K+ buffering, control of synaptic transmission, regulation of the vasculature and in diseases of the central nervous system.
Collapse
Affiliation(s)
- Gerald Seifert
- Institute of Cellular Neurosciences, University of Bonn Medical School, Bonn, Germany.
| | - Christian Henneberger
- Institute of Cellular Neurosciences, University of Bonn Medical School, Bonn, Germany; German Center of Neurodegenerative Diseases (DZNE), Bonn, Germany; Institute of Neurology, University College London, London, United Kingdom
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, University of Bonn Medical School, Bonn, Germany.
| |
Collapse
|
33
|
Spong KE, Andrew RD, Robertson RM. Mechanisms of spreading depolarization in vertebrate and insect central nervous systems. J Neurophysiol 2016; 116:1117-27. [PMID: 27334953 PMCID: PMC5013167 DOI: 10.1152/jn.00352.2016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 06/15/2016] [Indexed: 11/22/2022] Open
Abstract
Spreading depolarization (SD) is generated in the central nervous systems of both vertebrates and invertebrates. SD manifests as a propagating wave of electrical depression caused by a massive redistribution of ions. Mammalian SD underlies a continuum of human pathologies from migraine to stroke damage, whereas insect SD is associated with environmental stress-induced neural shutdown. The general cellular mechanisms underlying SD seem to be evolutionarily conserved throughout the animal kingdom. In particular, SD in the central nervous system of Locusta migratoria and Drosophila melanogaster has all the hallmarks of mammalian SD. Locust SD is easily induced and monitored within the metathoracic ganglion (MTG) and can be modulated both pharmacologically and by preconditioning treatments. The finding that the fly brain supports repetitive waves of SD is relatively recent but noteworthy, since it provides a genetically tractable model system. Due to the human suffering caused by SD manifestations, elucidating control mechanisms that could ultimately attenuate brain susceptibility is essential. Here we review mechanisms of SD focusing on the similarities between mammalian and insect systems. Additionally we discuss advantages of using invertebrate model systems and propose insect SD as a valuable model for providing new insights to mammalian SD.
Collapse
Affiliation(s)
- Kristin E Spong
- Department of Biology, Queen's University, Kingston, Ontario, Canada
| | - R David Andrew
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada; and Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - R Meldrum Robertson
- Department of Biology, Queen's University, Kingston, Ontario, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada; and Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
34
|
Boscia F, Begum G, Pignataro G, Sirabella R, Cuomo O, Casamassa A, Sun D, Annunziato L. Glial Na(+) -dependent ion transporters in pathophysiological conditions. Glia 2016; 64:1677-97. [PMID: 27458821 DOI: 10.1002/glia.23030] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/22/2016] [Accepted: 06/29/2016] [Indexed: 12/12/2022]
Abstract
Sodium dynamics are essential for regulating functional processes in glial cells. Indeed, glial Na(+) signaling influences and regulates important glial activities, and plays a role in neuron-glia interaction under physiological conditions or in response to injury of the central nervous system (CNS). Emerging studies indicate that Na(+) pumps and Na(+) -dependent ion transporters in astrocytes, microglia, and oligodendrocytes regulate Na(+) homeostasis and play a fundamental role in modulating glial activities in neurological diseases. In this review, we first briefly introduced the emerging roles of each glial cell type in the pathophysiology of cerebral ischemia, Alzheimer's disease, epilepsy, Parkinson's disease, Amyotrophic Lateral Sclerosis, and myelin diseases. Then, we discussed the current knowledge on the main roles played by the different glial Na(+) -dependent ion transporters, including Na(+) /K(+) ATPase, Na(+) /Ca(2+) exchangers, Na(+) /H(+) exchangers, Na(+) -K(+) -Cl(-) cotransporters, and Na(+) - HCO3- cotransporter in the pathophysiology of the diverse CNS diseases. We highlighted their contributions in cell survival, synaptic pathology, gliotransmission, pH homeostasis, and their role in glial activation, migration, gliosis, inflammation, and tissue repair processes. Therefore, this review summarizes the foundation work for targeting Na(+) -dependent ion transporters in glia as a novel strategy to control important glial activities associated with Na(+) dynamics in different neurological disorders. GLIA 2016;64:1677-1697.
Collapse
Affiliation(s)
- Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Gulnaz Begum
- Department of Neurology, University of Pittsburgh Medical School
| | - Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Rossana Sirabella
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Ornella Cuomo
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Antonella Casamassa
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh Medical School.,Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational and Clinical Center, Pittsburgh, Pennsylvania, 15213
| | - Lucio Annunziato
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| |
Collapse
|
35
|
Logica T, Riviere S, Holubiec MI, Castilla R, Barreto GE, Capani F. Metabolic Changes Following Perinatal Asphyxia: Role of Astrocytes and Their Interaction with Neurons. Front Aging Neurosci 2016; 8:116. [PMID: 27445788 PMCID: PMC4921470 DOI: 10.3389/fnagi.2016.00116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 05/03/2016] [Indexed: 11/13/2022] Open
Abstract
Perinatal Asphyxia (PA) represents an important cause of severe neurological deficits including delayed mental and motor development, epilepsy, major cognitive deficits and blindness. The interaction between neurons, astrocytes and endothelial cells plays a central role coupling energy supply with changes in neuronal activity. Traditionally, experimental research focused on neurons, whereas astrocytes have been more related to the damage mechanisms of PA. Astrocytes carry out a number of functions that are critical to normal nervous system function, including uptake of neurotransmitters, regulation of pH and ion concentrations, and metabolic support for neurons. In this work, we aim to review metabolic neuron-astrocyte interactions with the purpose of encourage further research in this area in the context of PA, which is highly complex and its mechanisms and pathways have not been fully elucidated to this day.
Collapse
Affiliation(s)
- Tamara Logica
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Facultad de Medicina, Instituto de Investigaciones Cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, CABA Buenos Aires, Argentina
| | - Stephanie Riviere
- Laboratorio de Biología Molecular, Facultad de Medicina, Instituto de Investigaciones cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, CABA Buenos Aires, Argentina
| | - Mariana I Holubiec
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Facultad de Medicina, Instituto de Investigaciones Cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, CABA Buenos Aires, Argentina
| | - Rocío Castilla
- Laboratorio de Biología Molecular, Facultad de Medicina, Instituto de Investigaciones cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, CABA Buenos Aires, Argentina
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá Bogotá, Colombia
| | - Francisco Capani
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Facultad de Medicina, Instituto de Investigaciones Cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, CABABuenos Aires, Argentina; Departamento de Biología, Universidad Argentina JF KennedyBuenos Aires, Argentina; Investigador Asociado, Universidad Autónoma de ChileSantiago, Chile
| |
Collapse
|
36
|
Murakami S, Kurachi Y. Mechanisms of astrocytic K(+) clearance and swelling under high extracellular K(+) concentrations. J Physiol Sci 2016; 66:127-42. [PMID: 26507417 PMCID: PMC10717000 DOI: 10.1007/s12576-015-0404-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/16/2015] [Indexed: 12/24/2022]
Abstract
In response to the elevation of extracellular K(+) concentration ([K(+)]out), astrocytes clear excessive K(+) to maintain conditions necessary for neural activity. K(+) clearance in astrocytes occurs via two processes: K(+) uptake and K(+) spatial buffering. High [K(+)]out also induces swelling in astrocytes, leading to edema and cell death in the brain. Despite the importance of astrocytic K(+) clearance and swelling, the underlying mechanisms remain unclear. Here, we report results from a simulation analysis of astrocytic K(+) clearance and swelling. Astrocyte models were constructed by incorporating various mechanisms such as intra/extracellular ion concentrations of Na(+), K(+), and Cl(-), cell volume, and models of Na,K-ATPase, Na-K-Cl cotransporter (NKCC), K-Cl cotransporter, inwardly-rectifying K(+) (KIR) channel, passive Cl(-) current, and aquaporin channel. The simulated response of astrocyte models under the uniform distribution of high [K(+)]out revealed significant contributions of NKCC and Na,K-ATPase to increases of intracellular K(+) and Cl(-) concentrations, and swelling. Moreover, we found that, under the non-uniform distribution of high [K(+)]out, KIR channels localized at synaptic clefts absorbed excess K(+) by depolarizing the equivalent potential of K(+) (E K) above membrane potential, while K(+) released through perivascular KIR channels was enhanced by hyperpolarizing E K and depolarizing membrane potential. Further analysis of simulated drug effects revealed that astrocyte swelling was modulated by blocking each of the ion channels and transporters. Our simulation analysis revealed controversial mechanisms of astrocytic K(+) clearance and swelling resulting from complex interactions among ion channels and transporters.
Collapse
Affiliation(s)
- Shingo Murakami
- Division of Molecular and Cellular Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
- The Global Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| | - Yoshihisa Kurachi
- Division of Molecular and Cellular Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
- The Global Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
37
|
Cheng H, Qiu X, Zhao X, Meng W, Huo D, Wei H. Functional Nucleic Acid Probe for Parallel Monitoring K+ and Protoporphyrin IX in Living Organisms. Anal Chem 2016; 88:2937-43. [DOI: 10.1021/acs.analchem.5b04936] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Hanjun Cheng
- Department
of Biomedical Engineering, College of Engineering and Applied Sciences,
Collaborative Innovation Center of Chemistry for Life Sciences, Nanjing National Laboratory of Microstructures, Nanjing, Jiangsu 210093, China
- State
Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210093, China
| | - Xuefeng Qiu
- Department
of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Xiaozhi Zhao
- Department
of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Wei Meng
- School
of Physics, Collaborative Innovation Center of Advanced Microstructures,
Nanjing National Laboratory of Microstructures, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Da Huo
- Department
of Biomedical Engineering, College of Engineering and Applied Sciences,
Collaborative Innovation Center of Chemistry for Life Sciences, Nanjing National Laboratory of Microstructures, Nanjing, Jiangsu 210093, China
| | - Hui Wei
- Department
of Biomedical Engineering, College of Engineering and Applied Sciences,
Collaborative Innovation Center of Chemistry for Life Sciences, Nanjing National Laboratory of Microstructures, Nanjing, Jiangsu 210093, China
| |
Collapse
|
38
|
Seidel JL, Faideau M, Aiba I, Pannasch U, Escartin C, Rouach N, Bonvento G, Shuttleworth CW. Ciliary neurotrophic factor (CNTF) activation of astrocytes decreases spreading depolarization susceptibility and increases potassium clearance. Glia 2015; 63:91-103. [PMID: 25092804 PMCID: PMC5141616 DOI: 10.1002/glia.22735] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 07/17/2014] [Indexed: 11/08/2022]
Abstract
Waves of spreading depolarization (SD) have been implicated in the progressive expansion of acute brain injuries. SD can persist over several days, coincident with the time course of astrocyte activation, but little is known about how astrocyte activation may influence SD susceptibility. We examined whether activation of astrocytes modified SD threshold in hippocampal slices. Injection of a lentiviral vector encoding Ciliary neurotrophic factor (CNTF) into the hippocampus in vivo, led to sustained astrocyte activation, verified by up-regulation of glial fibrillary acidic protein (GFAP) at the mRNA and protein levels, as compared to controls injected with vector encoding LacZ. In acute brain slices from LacZ controls, localized 1M KCl microinjections invariably generated SD in CA1 hippocampus, but SD was never induced with this stimulus in CNTF tissues. No significant change in intrinsic excitability was observed in CA1 neurons, but excitatory synaptic transmission was significantly reduced in CNTF samples. mRNA levels of the predominantly astrocytic Na(+) /K(+) -ATPase pump α2 subunit were higher in CNTF samples, and the kinetics of extracellular K(+) transients during matched synaptic activation were consistent with increased K(+) uptake in CNTF tissues. Supporting a role for the Na(+) /K(+) -ATPase pump in increased SD threshold, ouabain, an inhibitor of the pump, was able to generate SD in CNTF tissues. These data support the hypothesis that activated astrocytes can limit SD onset via increased K(+) clearance and suggest that therapeutic strategies targeting these glial cells could improve the outcome following acute brain injuries associated with SD.
Collapse
Affiliation(s)
- Jessica L Seidel
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
A novel optical intracellular imaging approach for potassium dynamics in astrocytes. PLoS One 2014; 9:e109243. [PMID: 25275375 PMCID: PMC4183569 DOI: 10.1371/journal.pone.0109243] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 09/09/2014] [Indexed: 11/19/2022] Open
Abstract
Astrocytes fulfill a central role in regulating K+ and glutamate, both released by neurons into the extracellular space during activity. Glial glutamate uptake is a secondary active process that involves the influx of three Na+ ions and one proton and the efflux of one K+ ion. Thus, intracellular K+ concentration ([K+]i) is potentially influenced both by extracellular K+ concentration ([K+]o) fluctuations and glutamate transport in astrocytes. We evaluated the impact of these K+ ion movements on [K+]i in primary mouse astrocytes by microspectrofluorimetry. We established a new noninvasive and reliable approach to monitor and quantify [K+]i using the recently developed K+ sensitive fluorescent indicator Asante Potassium Green-1 (APG-1). An in situ calibration procedure enabled us to estimate the resting [K+]i at 133±1 mM. We first investigated the dependency of [K+]i levels on [K+]o. We found that [K+]i followed [K+]o changes nearly proportionally in the range 3–10 mM, which is consistent with previously reported microelectrode measurements of intracellular K+ concentration changes in astrocytes. We then found that glutamate superfusion caused a reversible drop of [K+]i that depended on the glutamate concentration with an apparent EC50 of 11.1±1.4 µM, corresponding to the affinity of astrocyte glutamate transporters. The amplitude of the [K+]i drop was found to be 2.3±0.1 mM for 200 µM glutamate applications. Overall, this study shows that the fluorescent K+ indicator APG-1 is a powerful new tool for addressing important questions regarding fine [K+]i regulation with excellent spatial resolution.
Collapse
|
40
|
Sayre NL, Chen Y, Sifuentes M, Stoveken B, Lechleiter JD. Purinergic receptor stimulation decreases ischemic brain damage by energizing astrocyte mitochondria. ADVANCES IN NEUROBIOLOGY 2014; 11:121-50. [PMID: 25236727 DOI: 10.1007/978-3-319-08894-5_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
As a leading cause of death in the world, cerebral ischemic stroke has limited treatment options. The lack of glucose and oxygen after stroke is particularly harmful in the brain because neuronal metabolism accounts for significantly more energy consumption per gram of body weight compared to other organs. Our laboratory has identified mitochondrial metabolism of astrocytes to be a key target for pharmacologic intervention, not only because astrocytes play a central role in regulating brain metabolism, but also because they are essential for neuronal health and support. Here we review current literature pertaining to the pathobiology of stroke, along with the role of astrocytes and metabolism in stroke. We also discuss our research, which has revealed that pharmacologic stimulation of metabotropic P2Y1 receptor signaling in astrocytes can increase mitochondrial energy production and also reduce damage after stroke.
Collapse
Affiliation(s)
- Naomi L Sayre
- Department of Cellular and Structural Biology, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | | | | | | | | |
Collapse
|
41
|
Huang X, Li Y, Li J, Feng Y, Xu X. Tanshinone IIA dampens the cell proliferation induced by ischemic insult in rat astrocytes via blocking the activation of HIF-1α/SDF-1 signaling. Life Sci 2014; 112:59-67. [DOI: 10.1016/j.lfs.2014.07.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/24/2014] [Accepted: 07/12/2014] [Indexed: 12/13/2022]
|
42
|
Farkas E, Bari F. Spreading depolarization in the ischemic brain: does aging have an impact? J Gerontol A Biol Sci Med Sci 2014; 69:1363-70. [PMID: 24809351 DOI: 10.1093/gerona/glu066] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Recurrent waves of spreading depolarization (SD) spontaneously occur minutes after the onset of focal ischemia in the brain and keep generating for a number of days to follow. It has become widely accepted that ischemia-related SDs are part of the pathophysiology of cerebrovascular diseases and predict worse outcome. SDs may exacerbate ischemic injury via related atypical hemodynamic responses. The incidence of ischemic stroke is known to increase markedly with age; yet, very few studies investigated whether age alters SD evolution and whether a potential age-specific pattern of SD would contribute to the age-related intensification of infarct development. Experimental data demonstrate that aging has a marked impact on SD evolution and corresponding changes in cerebral blood flow. We hypothesize that an age-specific pattern of the SD-associated hemodynamic response must be involved in augmenting the expansion of ischemic brain damage in the elderly patients and that structural and functional (mal)adaptation of the cerebrovascular system with aging serves as a potential basis for compromised vascular reactivity and subsequent tissue damage. The concept put forward is expected to stimulate further investigation to achieve a comprehensive overview of the implication of SD in injury progression in the aged brain.
Collapse
Affiliation(s)
- Eszter Farkas
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary.
| | - Ferenc Bari
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| |
Collapse
|
43
|
Sato S, Kawauchi S, Okuda W, Nishidate I, Nawashiro H, Tsumatori G. Real-time optical diagnosis of the rat brain exposed to a laser-induced shock wave: observation of spreading depolarization, vasoconstriction and hypoxemia-oligemia. PLoS One 2014; 9:e82891. [PMID: 24416150 PMCID: PMC3885400 DOI: 10.1371/journal.pone.0082891] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 10/28/2013] [Indexed: 11/26/2022] Open
Abstract
Despite many efforts, the pathophysiology and mechanism of blast-induced traumatic brain injury (bTBI) have not yet been elucidated, partially due to the difficulty of real-time diagnosis and extremely complex factors determining the outcome. In this study, we topically applied a laser-induced shock wave (LISW) to the rat brain through the skull, for which real-time measurements of optical diffuse reflectance and electroencephalogram (EEG) were performed. Even under conditions showing no clear changes in systemic physiological parameters, the brain showed a drastic light scattering change accompanied by EEG suppression, which indicated the occurrence of spreading depression, long-lasting hypoxemia and signal change indicating mitochondrial energy impairment. Under the standard LISW conditions examined, hemorrhage and contusion were not apparent in the cortex. To investigate events associated with spreading depression, measurement of direct current (DC) potential, light scattering imaging and stereomicroscopic observation of blood vessels were also conducted for the brain. After LISW application, we observed a distinct negative shift in the DC potential, which temporally coincided with the transit of a light scattering wave, showing the occurrence of spreading depolarization and concomitant change in light scattering. Blood vessels in the brain surface initially showed vasodilatation for 3-4 min, which was followed by long-lasting vasoconstriction, corresponding to hypoxemia. Computer simulation based on the inverse Monte Carlo method showed that hemoglobin oxygen saturation declined to as low as ∼35% in the long-term hypoxemic phase. Overall, we found that topical application of a shock wave to the brain caused spreading depolarization/depression and prolonged severe hypoxemia-oligemia, which might lead to pathological conditions in the brain. Although further study is needed, our findings suggest that spreading depolarization/depression is one of the key events determining the outcome in bTBI. Furthermore, a rat exposed to an LISW(s) can be a reliable laboratory animal model for blast injury research.
Collapse
Affiliation(s)
- Shunichi Sato
- Division of Biomedical Information Sciences, National Defense Medical College Research Institute, Tokorozawa, Saitama, Japan
| | - Satoko Kawauchi
- Division of Biomedical Information Sciences, National Defense Medical College Research Institute, Tokorozawa, Saitama, Japan
| | - Wataru Okuda
- Graduate School of Bio-Application and Systems Engineering, Tokyo University of Agriculture and Technology, Koganei, Tokyo, Japan
| | - Izumi Nishidate
- Graduate School of Bio-Application and Systems Engineering, Tokyo University of Agriculture and Technology, Koganei, Tokyo, Japan
| | - Hiroshi Nawashiro
- Division of Neurosurgery, Tokorozawa Central Hospital, Tokorozawa, Saitama, Japan
| | - Gentaro Tsumatori
- Department of Defense Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| |
Collapse
|
44
|
Spong KE, Robertson RM. Pharmacological blockade of gap junctions induces repetitive surging of extracellular potassium within the locust CNS. JOURNAL OF INSECT PHYSIOLOGY 2013; 59:1031-1040. [PMID: 23916994 DOI: 10.1016/j.jinsphys.2013.07.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 07/24/2013] [Accepted: 07/25/2013] [Indexed: 06/02/2023]
Abstract
The maintenance of cellular ion homeostasis is crucial for optimal neural function and thus it is of great importance to understand its regulation. Glial cells are extensively coupled by gap junctions forming a network that is suggested to serve as a spatial buffer for potassium (K(+)) ions. We have investigated the role of glial spatial buffering in the regulation of extracellular K(+) concentration ([K(+)]o) within the locust metathoracic ganglion by pharmacologically inhibiting gap junctions. Using K(+)-sensitive microelectrodes, we measured [K(+)]o near the ventilatory neuropile while simultaneously recording the ventilatory rhythm as a model of neural circuit function. We found that blockade of gap junctions with either carbenoxolone (CBX), 18β-glycyrrhetinic acid (18β-GA) or meclofenamic acid (MFA) reliably induced repetitive [K(+)]o surges and caused a progressive impairment in the ability to maintain baseline [K(+)]o levels throughout the treatment period. We also show that a low dose of CBX that did not induce surging activity increased the vulnerability of locust neural tissue to spreading depression (SD) induced by Na(+)/K(+)-ATPase inhibition with ouabain. CBX pre-treatment increased the number of SD events induced by ouabain and hindered the recovery of [K(+)]o back to baseline levels between events. Our results suggest that glial spatial buffering through gap junctions plays an essential role in the regulation of [K(+)]o under normal conditions and also contributes to a component of [K(+)]o clearance following physiologically elevated levels of [K(+)]o.
Collapse
Affiliation(s)
- Kristin E Spong
- Department of Biology, Queen's University, Kingston, Ontario K7L 3N6, Canada.
| | | |
Collapse
|
45
|
Ionic transporter activity in astrocytes, microglia, and oligodendrocytes during brain ischemia. J Cereb Blood Flow Metab 2013; 33:969-82. [PMID: 23549380 PMCID: PMC3705429 DOI: 10.1038/jcbfm.2013.44] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 02/27/2013] [Accepted: 02/28/2013] [Indexed: 01/09/2023]
Abstract
Glial cells constitute a large percentage of cells in the nervous system. During recent years, a large number of studies have critically attributed to glia a new role which no longer reflects the long-held view that glia constitute solely a silent and passive supportive scaffolding for brain cells. Indeed, it has been hypothesized that glia, partnering neurons, have a much more actively participating role in brain function. Alteration of intraglial ionic homeostasis in response to ischemic injury has a crucial role in inducing and maintaining glial responses in the ischemic brain. Therefore, glial transporters as potential candidates in stroke intervention are becoming promising targets to enhance an effective and additional therapy for brain ischemia. In this review, we will describe in detail the role played by ionic transporters in influencing astrocyte, microglia, and oligodendrocyte activity and the implications that these transporters have in the progression of ischemic lesion.
Collapse
|
46
|
Monzack EL, Cunningham LL. Lead roles for supporting actors: critical functions of inner ear supporting cells. Hear Res 2013; 303:20-9. [PMID: 23347917 DOI: 10.1016/j.heares.2013.01.008] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 12/20/2012] [Accepted: 01/10/2013] [Indexed: 12/27/2022]
Abstract
Many studies that aim to investigate the underlying mechanisms of hearing loss or balance disorders focus on the hair cells and spiral ganglion neurons of the inner ear. Fewer studies have examined the supporting cells that contact both of these cell types in the cochlea and vestibular end organs. While the roles of supporting cells are still being elucidated, emerging evidence indicates that they serve many functions vital to maintaining healthy populations of hair cells and spiral ganglion neurons. Here we review recent studies that highlight the critical roles supporting cells play in the development, function, survival, death, phagocytosis, and regeneration of other cell types within the inner ear. Many of these roles have also been described for glial cells in other parts of the nervous system, and lessons from these other systems continue to inform our understanding of supporting cell functions. This article is part of a Special Issue entitled "Annual Reviews 2013".
Collapse
Affiliation(s)
- Elyssa L Monzack
- National Institute on Deafness and Other Communication Disorders, 5 Research Court, Rockville, MD 20850, USA.
| | | |
Collapse
|
47
|
Sutton AC, Yu W, Calos ME, Mueller LE, Berk M, Shim J, Molho ES, Brotchie JM, Carlen PL, Shin DS. Elevated potassium provides an ionic mechanism for deep brain stimulation in the hemiparkinsonian rat. Eur J Neurosci 2012; 37:231-41. [PMID: 23121286 DOI: 10.1111/ejn.12040] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 09/02/2012] [Accepted: 09/05/2012] [Indexed: 11/30/2022]
Abstract
The mechanism of high-frequency stimulation used in deep brain stimulation (DBS) for Parkinson's disease (PD) has not been completely elucidated. Previously, high-frequency stimulation of the rat entopeduncular nucleus, a basal ganglia output nucleus, elicited an increase in [K(+)](e) to 18 mm, in vitro. In this study, we assessed whether elevated K(+) can elicit DBS-like therapeutic effects in hemiparkinsonian rats by employing the limb-use asymmetry test and the self-adjusting stepping test. We then identified how these effects were meditated with in-vivo and in-vitro electrophysiology. Forelimb akinesia improved in hemiparkinsonian rats undergoing both tests after 20 mm KCl injection into the substantia nigra pars reticulata (SNr) or the subthalamic nucleus. In the SNr, neuronal spiking activity decreased from 38.2 ± 1.2 to 14.6 ± 1.6 Hz and attenuated SNr beta-frequency (12-30 Hz) oscillations after K(+) treatment. These oscillations are commonly associated with akinesia/bradykinesia in patients with PD and animal models of PD. Pressure ejection of 20 mm KCl onto SNr neurons in vitro caused a depolarisation block and sustained quiescence of SNr activity. In conclusion, our data showed that elevated K(+) injection into the hemiparkinsonian rat SNr improved forelimb akinesia, which coincided with a decrease in SNr neuronal spiking activity and desynchronised activity in SNr beta frequency, and subsequently an overall increase in ventral medial thalamic neuronal activity. Moreover, these findings also suggest that elevated K(+) may provide an ionic mechanism that can contribute to the therapeutic effects of DBS for the motor treatment of advanced PD.
Collapse
Affiliation(s)
- Alexander C Sutton
- Center for Neuropharmacology and Neuroscience, Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Risher WC, Croom D, Kirov SA. Persistent astroglial swelling accompanies rapid reversible dendritic injury during stroke-induced spreading depolarizations. Glia 2012; 60:1709-20. [PMID: 22821441 PMCID: PMC3435464 DOI: 10.1002/glia.22390] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 06/20/2012] [Indexed: 11/10/2022]
Abstract
Spreading depolarizations are a key event in the pathophysiology of stroke, resulting in rapid dendritic beading, which represents acute damage to synaptic circuitry. The impact of spreading depolarizations on the real-time injury of astrocytes during ischemia is less clear. We used simultaneous in vivo 2-photon imaging and electrophysiological recordings in adult mouse somatosensory cortex to examine spreading depolarization-induced astroglial structural changes concurrently with signs of neuronal injury in the early periods of focal and global ischemia. Astrocytes in the metabolically compromised ischemic penumbra-like area showed a long lasting swelling response to spontaneous spreading depolarizations despite rapid dendritic recovery in a photothrombotic occlusion model of focal stroke. Astroglial swelling was often facilitated by recurrent depolarizations and the magnitude of swelling strongly correlated with the total duration of depolarization. In contrast, spreading depolarization-induced astroglial swelling was transient in normoxic healthy tissue. In a model of transient global ischemia, the occurrence of a single spreading depolarization elicited by a bilateral common carotid artery occlusion coincided with astroglial swelling alongside dendritic beading. With immediate reperfusion, dendritic beading subsides. Astroglial swelling was either transient during short ischemic periods distinguished by a short-lasting spreading depolarization, or persistent during severe ischemia characterized by a long-lasting depolarization with the ultraslow negative voltage component. We propose that persistent astroglial swelling is initiated and exacerbated during spreading depolarization in brain tissue with moderate to severe energy deficits, disrupting astroglial maintenance of normal homeostatic function thus contributing to the negative outcome of ischemic stroke as astrocytes fail to provide neuronal support.
Collapse
Affiliation(s)
- W. Christopher Risher
- Graduate Program in Neuroscience, Georgia Health Sciences University, Augusta, Georgia 30912
- Brain and Behavior Discovery Institute, Georgia Health Sciences University, Augusta, Georgia 30912
| | - Deborah Croom
- Department of Neurosurgery, Georgia Health Sciences University, Augusta, Georgia 30912
- Brain and Behavior Discovery Institute, Georgia Health Sciences University, Augusta, Georgia 30912
| | - Sergei A. Kirov
- Department of Neurosurgery, Georgia Health Sciences University, Augusta, Georgia 30912
- Brain and Behavior Discovery Institute, Georgia Health Sciences University, Augusta, Georgia 30912
| |
Collapse
|
49
|
Carlen PL. Curious and contradictory roles of glial connexins and pannexins in epilepsy. Brain Res 2012; 1487:54-60. [PMID: 22796594 DOI: 10.1016/j.brainres.2012.06.059] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 06/19/2012] [Accepted: 06/20/2012] [Indexed: 10/28/2022]
Abstract
Glia play an under-recognized role in epilepsy. This review examines the involvement of glial connexins (Cxs) and pannexins (Panxs), proteins which form gap junctions and membrane hemichannels (connexins) and hemichannels (pannexins), in epilepsy. These proteins, particularly glial Cx43, have been shown to be upregulated in epileptic brain tissue. In a cobalt model of in vitro seizures, seizures increased Panxs1 and 2 and Cx43 expression, and remarkably reorganized the interrelationships between their mRNA levels (transcriptome) which then became statistically significant. Gap junctions are highly implicated in synchronous seizure activity. Blocking gap junctional communication (GJC) is often anticonvulsant, and assumed to be due to blocking gap junctionally-medicated electrotonic coupling between neurons. However, in organotypic hippocampal slice cultures, connexin43 specific peptides, which attenuate GJC possibly by blocking connexon docking, diminished spontaneous seizures. Glia have many functions including extracellular potassium redistribution, in part via gap junctions, which if blocked, can be seizuregenic. Glial gap junctions are critical for the delivery of nutrients to neurons, which if interrupted, can depress seizure activity. Other functions of glia possibly related to epileptogenesis are mentioned including anatomic reorganization in chronic seizure models greatly increasing the overlapping domains of glial processes, changes in neurotransmitter re-uptake, and possible glial generation of currents and fields during seizure activity. Finally there is recent evidence for Cx43 hemichannels and Panx1 channels in glial membranes which could play a role in brain damage and seizure activity. Although glial Cxs and Panxs are increasingly recognized as contributing to fundamental mechanisms of epilepsy, the data are often contradictory and controversial, requiring much more research. This article is part of a Special Issue entitled Electrical Synapses.
Collapse
Affiliation(s)
- Peter L Carlen
- Toronto Western Research Institute, Epilepsy Program, University Health Network, Toronto, Ontario, Canada M5T2S8.
| |
Collapse
|
50
|
Pasantes-Morales H, Vázquez-Juárez E. Transporters and channels in cytotoxic astrocyte swelling. Neurochem Res 2012; 37:2379-87. [PMID: 22544664 DOI: 10.1007/s11064-012-0777-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 03/30/2012] [Accepted: 04/03/2012] [Indexed: 12/29/2022]
Abstract
Brain edema is a severe clinical complication in a number of pathologies and is a major cause of increased morbidity and death. The swelling of astrocytes caused by a disruption of water and ion homeostasis, is the primary event contributing to the cytotoxic form of brain edema. Astrocyte cytotoxic swelling ultimately leads to transcapillary fluxes of ions and water into the brain parenchyma. This review focuses on the implication of transporters and channels in cytotoxic astrocyte swelling in hyponatremia, ischemia, trauma and hepatic encephalopathy. Emphasis is put on some salient features of the astrocyte physiology, all related to cell swelling, i.e. predominance of aquaporins, control of K(+) homeostasis and ammonia accumulation during the brain ammonia-detoxifying process.
Collapse
Affiliation(s)
- Herminia Pasantes-Morales
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| | | |
Collapse
|