1
|
Carretero VJ, Álvarez-Merz I, Hernández-Campano J, Kirov SA, Hernández-Guijo JM. Targeting harmful effects of non-excitatory amino acids as an alternative therapeutic strategy to reduce ischemic damage. Neural Regen Res 2025; 20:2454-2463. [PMID: 39314160 PMCID: PMC11801293 DOI: 10.4103/nrr.nrr-d-24-00536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/16/2024] [Accepted: 08/14/2024] [Indexed: 09/25/2024] Open
Abstract
The involvement of the excitatory amino acids glutamate and aspartate in cerebral ischemia and excitotoxicity is well-documented. Nevertheless, the role of non-excitatory amino acids in brain damage following a stroke or brain trauma remains largely understudied. The release of amino acids by necrotic cells in the ischemic core may contribute to the expansion of the penumbra. Our findings indicated that the reversible loss of field excitatory postsynaptic potentials caused by transient hypoxia became irreversible when exposed to a mixture of just four non-excitatory amino acids (L-alanine, glycine, L-glutamine, and L-serine) at their plasma concentrations. These amino acids induce swelling in the somas of neurons and astrocytes during hypoxia, along with permanent dendritic damage mediated by N-methyl-D-aspartate receptors. Blocking N-methyl-D-aspartate receptors prevented neuronal damage in the presence of these amino acids during hypoxia. It is likely that astroglial swelling caused by the accumulation of these amino acids via the alanine-serine-cysteine transporter 2 exchanger and system N transporters activates volume-regulated anion channels, leading to the release of excitotoxins and subsequent neuronal damage through N-methyl-D-aspartate receptor activation. Thus, previously unrecognized mechanisms involving non-excitatory amino acids may contribute to the progression and expansion of brain injury in neurological emergencies such as stroke and traumatic brain injury. Understanding these pathways could highlight new therapeutic targets to mitigate brain injury.
Collapse
Affiliation(s)
| | - Iris Álvarez-Merz
- Department of Pharmacology and Therapeutic, School of Medicine, Univ. Autónoma de Madrid, Madrid, Spain
- Ramón y Cajal Institute for Health Research (IRYCIS), Neurobiology-Research Service, Hospital Ramón y Cajal, Madrid, Spain
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jorge Hernández-Campano
- Department of Pharmacology and Therapeutic, School of Medicine, Univ. Autónoma de Madrid, Madrid, Spain
| | - Sergei A. Kirov
- Department of Neuroscience and Regenerative Medicine & Department of Neurosurgery, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Jesús M. Hernández-Guijo
- Department of Pharmacology and Therapeutic, School of Medicine, Univ. Autónoma de Madrid, Madrid, Spain
- Ramón y Cajal Institute for Health Research (IRYCIS), Neurobiology-Research Service, Hospital Ramón y Cajal, Madrid, Spain
| |
Collapse
|
2
|
Farkas E, Rose CR. A dangerous liaison: Spreading depolarization and tissue acidification in cerebral ischemia. J Cereb Blood Flow Metab 2025; 45:201-218. [PMID: 39535276 PMCID: PMC12000947 DOI: 10.1177/0271678x241289756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 11/16/2024]
Abstract
Brain pH is precisely regulated, and pH transients associated with activity are rapidly restored under physiological conditions. During ischemia, the brain's ability to buffer pH changes is rapidly depleted. Tissue oxygen deprivation causes a shift from aerobic to anaerobic metabolism and the accumulation of lactic acid and protons. Although the degree of tissue acidosis resulting from ischemia depends on the severity of the ischemia, spreading depolarization (SD) events emerge as central elements to determining ischemic tissue acidosis. A marked decrease in tissue pH during cerebral ischemia may exacerbate neuronal injury, which has become known as acidotoxicity, in analogy to excitotoxicity. The cellular pathways underlying acidotoxicity have recently been described in increasing detail. The molecular structure of acid or base carriers and acidosis-activated ion channels, the precise (dys)homeostatic conditions under which they are activated, and their possible role in severe ischemia have been addressed. The expanded understanding of acidotoxic mechanisms now provides an opportunity to reevaluate the contexts that lead to acidotoxic injury. Here, we review the specific cellular pathways of acidotoxicity and demonstrate that SD plays a central role in activating the molecular machinery leading to acid-induced damage. We propose that SD is a key contributor to acidotoxic injury in cerebral ischemia.
Collapse
Affiliation(s)
- Eszter Farkas
- Hungarian Centre of Excellence for Molecular Medicine – University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Christine R Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
3
|
Zoccal DB, Vieira BN, Mendes LR, Evangelista AB, Leirão IP. Hypoxia sensing in the body: An update on the peripheral and central mechanisms. Exp Physiol 2024; 109:461-469. [PMID: 38031809 PMCID: PMC10988761 DOI: 10.1113/ep091206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 11/15/2023] [Indexed: 12/01/2023]
Abstract
An adequate supply of O2 is essential for the maintenance of cellular activity. Systemic or local hypoxia can be experienced during decreased O2 availability or associated with diseases, or a combination of both. Exposure to hypoxia triggers adjustments in multiple physiological systems in the body to generate appropriate homeostatic responses. However, with significant reductions in the arterial partial pressure of O2, hypoxia can be life-threatening and cause maladaptive changes or cell damage and death. To mitigate the impact of limited O2 availability on cellular activity, O2 chemoreceptors rapidly detect and respond to reductions in the arterial partial pressure of O2, triggering orchestrated responses of increased ventilation and cardiac output, blood flow redistribution and metabolic adjustments. In mammals, the peripheral chemoreceptors of the carotid body are considered to be the main hypoxic sensors and the primary source of excitatory feedback driving respiratory, cardiovascular and autonomic responses. However, current evidence indicates that the CNS contains specialized brainstem and spinal cord regions that can also sense hypoxia and stimulate brain networks independently of the carotid body inputs. In this manuscript, we review the discoveries about the functioning of the O2 chemoreceptors and their contribution to the monitoring of O2 levels in the blood and brain parenchyma and mounting cardiorespiratory responses to maintain O2 homeostasis. We also discuss the implications of the chemoreflex-related mechanisms in paediatric and adult pathologies.
Collapse
Affiliation(s)
- Daniel B. Zoccal
- Department of Physiology and Pathology, School of Dentistry of AraraquaraSão Paulo State University (UNESP)AraraquaraSão PauloBrazil
| | - Beatriz N. Vieira
- Department of Physiology and Pathology, School of Dentistry of AraraquaraSão Paulo State University (UNESP)AraraquaraSão PauloBrazil
| | - Letícia R. Mendes
- Department of Physiology and Pathology, School of Dentistry of AraraquaraSão Paulo State University (UNESP)AraraquaraSão PauloBrazil
| | - Andressa B. Evangelista
- Department of Physiology and Pathology, School of Dentistry of AraraquaraSão Paulo State University (UNESP)AraraquaraSão PauloBrazil
| | - Isabela P. Leirão
- Department of Physiology and Pathology, School of Dentistry of AraraquaraSão Paulo State University (UNESP)AraraquaraSão PauloBrazil
| |
Collapse
|
4
|
Zhang X, Zhang Y, Su Q, Liu Y, Li Z, Yong VW, Xue M. Ion Channel Dysregulation Following Intracerebral Hemorrhage. Neurosci Bull 2024; 40:401-414. [PMID: 37755675 PMCID: PMC10912428 DOI: 10.1007/s12264-023-01118-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/14/2023] [Indexed: 09/28/2023] Open
Abstract
Injury to the brain after intracerebral hemorrhage (ICH) results from numerous complex cellular mechanisms. At present, effective therapy for ICH is limited and a better understanding of the mechanisms of brain injury is necessary to improve prognosis. There is increasing evidence that ion channel dysregulation occurs at multiple stages in primary and secondary brain injury following ICH. Ion channels such as TWIK-related K+ channel 1, sulfonylurea 1 transient receptor potential melastatin 4 and glutamate-gated channels affect ion homeostasis in ICH. They in turn participate in the formation of brain edema, disruption of the blood-brain barrier, and the generation of neurotoxicity. In this review, we summarize the interaction between ions and ion channels, the effects of ion channel dysregulation, and we discuss some therapeutics based on ion-channel modulation following ICH.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Yan Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Qiuyang Su
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Zhe Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
5
|
Hencz A, Magony A, Thomas C, Kovacs K, Szilagyi G, Pal J, Sik A. Mild hypoxia-induced structural and functional changes of the hippocampal network. Front Cell Neurosci 2023; 17:1277375. [PMID: 37841285 PMCID: PMC10576450 DOI: 10.3389/fncel.2023.1277375] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
Hypoxia causes structural and functional changes in several brain regions, including the oxygen-concentration-sensitive hippocampus. We investigated the consequences of mild short-term hypoxia on rat hippocampus in vivo. The hypoxic group was treated with 16% O2 for 1 h, and the control group with 21% O2. Using a combination of Gallyas silver impregnation histochemistry revealing damaged neurons and interneuron-specific immunohistochemistry, we found that somatostatin-expressing inhibitory neurons in the hilus were injured. We used 32-channel silicon probe arrays to record network oscillations and unit activity from the hippocampal layers under anaesthesia. There were no changes in the frequency power of slow, theta, beta, or gamma bands, but we found a significant increase in the frequency of slow oscillation (2.1-2.2 Hz) at 16% O2 compared to 21% O2. In the hilus region, the firing frequency of unidentified interneurons decreased. In the CA3 region, the firing frequency of some unidentified interneurons decreased while the activity of other interneurons increased. The activity of pyramidal cells increased both in the CA1 and CA3 regions. In addition, the regularity of CA1, CA3 pyramidal cells' and CA3 type II and hilar interneuron activity has significantly changed in hypoxic conditions. In summary, a low O2 environment caused profound changes in the state of hippocampal excitatory and inhibitory neurons and network activity, indicating potential changes in information processing caused by mild short-term hypoxia.
Collapse
Affiliation(s)
- Alexandra Hencz
- Institute of Physiology, Medical School, University of Pecs, Pecs, Hungary
| | - Andor Magony
- Institute of Physiology, Medical School, University of Pecs, Pecs, Hungary
- Institute of Transdisciplinary Discoveries, Medical School, University of Pecs, Pecs, Hungary
| | - Chloe Thomas
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Krisztina Kovacs
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Gabor Szilagyi
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pecs, Pecs, Hungary
| | - Jozsef Pal
- Institute of Physiology, Medical School, University of Pecs, Pecs, Hungary
| | - Attila Sik
- Institute of Physiology, Medical School, University of Pecs, Pecs, Hungary
- Institute of Transdisciplinary Discoveries, Medical School, University of Pecs, Pecs, Hungary
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
6
|
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
|
7
|
Zironi I, Aicardi G. Hypoxia Depresses Synaptic Transmission in the Primary Motor Cortex of the Infant Rat-Role of Adenosine A 1 Receptors and Nitric Oxide. Biomedicines 2022; 10:2875. [PMID: 36359395 PMCID: PMC9687150 DOI: 10.3390/biomedicines10112875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 09/08/2024] Open
Abstract
The acute and long-term consequences of perinatal asphyxia have been extensively investigated, but only a few studies have focused on postnatal asphyxia. In particular, electrophysiological changes induced in the motor cortex by postnatal asphyxia have not been examined so far, despite the critical involvement of this cortical area in epilepsy. In this study, we exposed primary motor cortex slices obtained from infant rats in an age window (16-18 day-old) characterized by high incidence of hypoxia-induced seizures associated with epileptiform motor behavior to 10 min of hypoxia. Extracellular field potentials evoked by horizontal pathway stimulation were recorded in layers II/III of the primary motor cortex before, during, and after the hypoxic event. The results show that hypoxia reversibly depressed glutamatergic synaptic transmission and neuronal excitability. Data obtained in the presence of specific blockers suggest that synaptic depression was mediated by adenosine acting on pre-synaptic A1 receptors to decrease glutamate release, and by a nitric oxide (NO)/cGMP postsynaptic pathway. These effects are neuroprotective because they limit energy failure. The present findings may be helpful in the preclinical search for therapeutic strategies aimed at preventing acute and long-term neurological consequences of postnatal asphyxia.
Collapse
Affiliation(s)
- Isabella Zironi
- Department of Physics and Astronomy, University of Bologna, 40127 Bologna, Italy
| | - Giorgio Aicardi
- Department for Life Quality Studies, University of Bologna, 40127 Bologna, Italy
| |
Collapse
|
8
|
Álvarez-Merz I, Fomitcheva IV, Sword J, Hernández-Guijo JM, Solís JM, Kirov SA. Novel mechanism of hypoxic neuronal injury mediated by non-excitatory amino acids and astroglial swelling. Glia 2022; 70:2108-2130. [PMID: 35802030 PMCID: PMC9474671 DOI: 10.1002/glia.24241] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/14/2022] [Accepted: 06/24/2022] [Indexed: 11/08/2022]
Abstract
In ischemic stroke and post-traumatic brain injury (TBI), blood-brain barrier disruption leads to leaking plasma amino acids (AA) into cerebral parenchyma. Bleeding in hemorrhagic stroke and TBI also release plasma AA. Although excitotoxic AA were extensively studied, little is known about non-excitatory AA during hypoxic injury. Hypoxia-induced synaptic depression in hippocampal slices becomes irreversible with non-excitatory AA, alongside their intracellular accumulation and increased tissue electrical resistance. Four non-excitatory AA (l-alanine, glycine, l-glutamine, l-serine: AGQS) at plasmatic concentrations were applied to slices from mice expressing EGFP in pyramidal neurons or astrocytes during normoxia or hypoxia. Two-photon imaging, light transmittance (LT) changes, and electrophysiological field recordings followed by electron microscopy in hippocampal CA1 st. radiatum were used to monitor synaptic function concurrently with cellular swelling and injury. During normoxia, AGQS-induced increase in LT was due to astroglial but not neuronal swelling. LT raise during hypoxia and AGQS manifested astroglial and neuronal swelling accompanied by a permanent loss of synaptic transmission and irreversible dendritic beading, signifying acute damage. Neuronal injury was not triggered by spreading depolarization which did not occur in our experiments. Hypoxia without AGQS did not cause cell swelling, leaving dendrites intact. Inhibition of NMDA receptors prevented neuronal damage and irreversible loss of synaptic function. Deleterious effects of AGQS during hypoxia were prevented by alanine-serine-cysteine transporters (ASCT2) and volume-regulated anion channels (VRAC) blockers. Our findings suggest that astroglial swelling induced by accumulation of non-excitatory AA and release of excitotoxins through antiporters and VRAC may exacerbate the hypoxia-induced neuronal injury.
Collapse
Affiliation(s)
- Iris Álvarez-Merz
- Dept. de Farmacología y Terapéutica, ITH, Facultad de Medicina, Universidad Autónoma de Madrid, IRYCIS, 28029 Madrid, Spain
- Servicio de Neurobiología-Investigación, Hospital Universitario Ramón y Cajal, IRYCIS, 28034 Madrid, Spain
- Dept. of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, USA
| | - Ioulia V. Fomitcheva
- Dept. of Neurosurgery, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, USA
| | - Jeremy Sword
- Dept. of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, USA
| | - Jesús M. Hernández-Guijo
- Dept. de Farmacología y Terapéutica, ITH, Facultad de Medicina, Universidad Autónoma de Madrid, IRYCIS, 28029 Madrid, Spain
| | - José M. Solís
- Servicio de Neurobiología-Investigación, Hospital Universitario Ramón y Cajal, IRYCIS, 28034 Madrid, Spain
| | - Sergei A. Kirov
- Dept. of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, USA
| |
Collapse
|
9
|
Andrew RD, Farkas E, Hartings JA, Brennan KC, Herreras O, Müller M, Kirov SA, Ayata C, Ollen-Bittle N, Reiffurth C, Revah O, Robertson RM, Dawson-Scully KD, Ullah G, Dreier JP. Questioning Glutamate Excitotoxicity in Acute Brain Damage: The Importance of Spreading Depolarization. Neurocrit Care 2022; 37:11-30. [PMID: 35194729 PMCID: PMC9259542 DOI: 10.1007/s12028-021-01429-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/20/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Within 2 min of severe ischemia, spreading depolarization (SD) propagates like a wave through compromised gray matter of the higher brain. More SDs arise over hours in adjacent tissue, expanding the neuronal damage. This period represents a therapeutic window to inhibit SD and so reduce impending tissue injury. Yet most neuroscientists assume that the course of early brain injury can be explained by glutamate excitotoxicity, the concept that immediate glutamate release promotes early and downstream brain injury. There are many problems with glutamate release being the unseen culprit, the most practical being that the concept has yielded zero therapeutics over the past 30 years. But the basic science is also flawed, arising from dubious foundational observations beginning in the 1950s METHODS: Literature pertaining to excitotoxicity and to SD over the past 60 years is critiqued. RESULTS Excitotoxicity theory centers on the immediate and excessive release of glutamate with resulting neuronal hyperexcitation. This instigates poststroke cascades with subsequent secondary neuronal injury. By contrast, SD theory argues that although SD evokes some brief glutamate release, acute neuronal damage and the subsequent cascade of injury to neurons are elicited by the metabolic stress of SD, not by excessive glutamate release. The challenge we present here is to find new clinical targets based on more informed basic science. This is motivated by the continuing failure by neuroscientists and by industry to develop drugs that can reduce brain injury following ischemic stroke, traumatic brain injury, or sudden cardiac arrest. One important step is to recognize that SD plays a central role in promoting early neuronal damage. We argue that uncovering the molecular biology of SD initiation and propagation is essential because ischemic neurons are usually not acutely injured unless SD propagates through them. The role of glutamate excitotoxicity theory and how it has shaped SD research is then addressed, followed by a critique of its fading relevance to the study of brain injury. CONCLUSIONS Spreading depolarizations better account for the acute neuronal injury arising from brain ischemia than does the early and excessive release of glutamate.
Collapse
Affiliation(s)
| | - Eszter Farkas
- Hungarian Centre of Excellence for Molecular Medicine-University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Department of Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| | | | | | | | | | | | - Cenk Ayata
- Harvard Medical School, Harvard University, Boston, MA USA
| | | | - Clemens Reiffurth
- Center for Stroke Research Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Omer Revah
- School of Medicine, Stanford University, Stanford, CA USA
| | | | | | | | - Jens P. Dreier
- Center for Stroke Research Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Corporate Member of Freie Universität Berlin, Berlin, Germany
- Department of Neurology, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Neurology, Berlin Institute of Health, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
| |
Collapse
|
10
|
Lemale CL, Lückl J, Horst V, Reiffurth C, Major S, Hecht N, Woitzik J, Dreier JP. Migraine Aura, Transient Ischemic Attacks, Stroke, and Dying of the Brain Share the Same Key Pathophysiological Process in Neurons Driven by Gibbs–Donnan Forces, Namely Spreading Depolarization. Front Cell Neurosci 2022; 16:837650. [PMID: 35237133 PMCID: PMC8884062 DOI: 10.3389/fncel.2022.837650] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/19/2022] [Indexed: 12/15/2022] Open
Abstract
Neuronal cytotoxic edema is the morphological correlate of the near-complete neuronal battery breakdown called spreading depolarization, or conversely, spreading depolarization is the electrophysiological correlate of the initial, still reversible phase of neuronal cytotoxic edema. Cytotoxic edema and spreading depolarization are thus different modalities of the same process, which represents a metastable universal reference state in the gray matter of the brain close to Gibbs–Donnan equilibrium. Different but merging sections of the spreading-depolarization continuum from short duration waves to intermediate duration waves to terminal waves occur in a plethora of clinical conditions, including migraine aura, ischemic stroke, traumatic brain injury, aneurysmal subarachnoid hemorrhage (aSAH) and delayed cerebral ischemia (DCI), spontaneous intracerebral hemorrhage, subdural hematoma, development of brain death, and the dying process during cardio circulatory arrest. Thus, spreading depolarization represents a prime and simultaneously the most neglected pathophysiological process in acute neurology. Aristides Leão postulated as early as the 1940s that the pathophysiological process in neurons underlying migraine aura is of the same nature as the pathophysiological process in neurons that occurs in response to cerebral circulatory arrest, because he assumed that spreading depolarization occurs in both conditions. With this in mind, it is not surprising that patients with migraine with aura have about a twofold increased risk of stroke, as some spreading depolarizations leading to the patient percept of migraine aura could be caused by cerebral ischemia. However, it is in the nature of spreading depolarization that it can have different etiologies and not all spreading depolarizations arise because of ischemia. Spreading depolarization is observed as a negative direct current (DC) shift and associated with different changes in spontaneous brain activity in the alternating current (AC) band of the electrocorticogram. These are non-spreading depression and spreading activity depression and epileptiform activity. The same spreading depolarization wave may be associated with different activity changes in adjacent brain regions. Here, we review the basal mechanism underlying spreading depolarization and the associated activity changes. Using original recordings in animals and patients, we illustrate that the associated changes in spontaneous activity are by no means trivial, but pose unsolved mechanistic puzzles and require proper scientific analysis.
Collapse
Affiliation(s)
- Coline L. Lemale
- Center for Stroke Research Berlin, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Experimental Neurology, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Janos Lückl
- Center for Stroke Research Berlin, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
- Department of Neurology, University of Szeged, Szeged, Hungary
| | - Viktor Horst
- Center for Stroke Research Berlin, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Clemens Reiffurth
- Center for Stroke Research Berlin, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Experimental Neurology, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sebastian Major
- Center for Stroke Research Berlin, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Experimental Neurology, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Neurology, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nils Hecht
- Department of Neurosurgery, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Johannes Woitzik
- Department of Neurosurgery, Evangelisches Krankenhaus Oldenburg, University of Oldenburg, Oldenburg, Germany
| | - Jens P. Dreier
- Center for Stroke Research Berlin, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Experimental Neurology, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Neurology, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
- *Correspondence: Jens P. Dreier,
| |
Collapse
|
11
|
Andrew RD, Hartings JA, Ayata C, Brennan KC, Dawson-Scully KD, Farkas E, Herreras O, Kirov SA, Müller M, Ollen-Bittle N, Reiffurth C, Revah O, Robertson RM, Shuttleworth CW, Ullah G, Dreier JP. The Critical Role of Spreading Depolarizations in Early Brain Injury: Consensus and Contention. Neurocrit Care 2022; 37:83-101. [PMID: 35257321 PMCID: PMC9259543 DOI: 10.1007/s12028-021-01431-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 12/29/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND When a patient arrives in the emergency department following a stroke, a traumatic brain injury, or sudden cardiac arrest, there is no therapeutic drug available to help protect their jeopardized neurons. One crucial reason is that we have not identified the molecular mechanisms leading to electrical failure, neuronal swelling, and blood vessel constriction in newly injured gray matter. All three result from a process termed spreading depolarization (SD). Because we only partially understand SD, we lack molecular targets and biomarkers to help neurons survive after losing their blood flow and then undergoing recurrent SD. METHODS In this review, we introduce SD as a single or recurring event, generated in gray matter following lost blood flow, which compromises the Na+/K+ pump. Electrical recovery from each SD event requires so much energy that neurons often die over minutes and hours following initial injury, independent of extracellular glutamate. RESULTS We discuss how SD has been investigated with various pitfalls in numerous experimental preparations, how overtaxing the Na+/K+ ATPase elicits SD. Elevated K+ or glutamate are unlikely natural activators of SD. We then turn to the properties of SD itself, focusing on its initiation and propagation as well as on computer modeling. CONCLUSIONS Finally, we summarize points of consensus and contention among the authors as well as where SD research may be heading. In an accompanying review, we critique the role of the glutamate excitotoxicity theory, how it has shaped SD research, and its questionable importance to the study of early brain injury as compared with SD theory.
Collapse
Affiliation(s)
- R. David Andrew
- grid.410356.50000 0004 1936 8331Queen’s University, Kingston, ON Canada
| | - Jed A. Hartings
- grid.24827.3b0000 0001 2179 9593University of Cincinnati, Cincinnati, OH USA
| | - Cenk Ayata
- grid.38142.3c000000041936754XHarvard Medical School, Harvard University, Boston, MA USA
| | - K. C. Brennan
- grid.223827.e0000 0001 2193 0096The University of Utah, Salt Lake City, UT USA
| | | | - Eszter Farkas
- grid.9008.10000 0001 1016 96251HCEMM-USZ Cerebral Blood Flow and Metabolism Research Group, and the Department of Cell Biology and Molecular Medicine, Faculty of Science and Informatics & Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Oscar Herreras
- grid.419043.b0000 0001 2177 5516Instituto de Neurobiologia Ramon Y Cajal (Consejo Superior de Investigaciones Científicas), Madrid, Spain
| | - Sergei. A. Kirov
- grid.410427.40000 0001 2284 9329Medical College of Georgia, Augusta, GA USA
| | - Michael Müller
- grid.411984.10000 0001 0482 5331University of Göttingen, University Medical Center Göttingen, Göttingen, Germany
| | - Nikita Ollen-Bittle
- grid.39381.300000 0004 1936 8884University of Western Ontario, London, ON Canada
| | - Clemens Reiffurth
- grid.7468.d0000 0001 2248 7639Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; and the Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health., Berlin, Germany
| | - Omer Revah
- grid.168010.e0000000419368956School of Medicine, Stanford University, Stanford, CA USA
| | | | | | - Ghanim Ullah
- grid.170693.a0000 0001 2353 285XUniversity of South Florida, Tampa, FL USA
| | - Jens P. Dreier
- grid.7468.d0000 0001 2248 7639Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; and the Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health., Berlin, Germany
| |
Collapse
|
12
|
Lu YL, Scharfman HE. New Insights and Methods for Recording and Imaging Spontaneous Spreading Depolarizations and Seizure-Like Events in Mouse Hippocampal Slices. Front Cell Neurosci 2021; 15:761423. [PMID: 34899190 PMCID: PMC8663723 DOI: 10.3389/fncel.2021.761423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/04/2021] [Indexed: 11/30/2022] Open
Abstract
Spreading depolarization (SD) is a sudden, large, and synchronous depolarization of principal cells which also involves interneurons and astrocytes. It is followed by depression of neuronal activity, and it slowly propagates across brain regions like cortex or hippocampus. SD is considered to be mechanistically relevant to migraine, epilepsy, and traumatic brain injury (TBI), but there are many questions about its basic neurophysiology and spread. Research into SD in hippocampus using slices is often used to gain insight and SD is usually triggered by a focal stimulus with or without an altered extracellular buffer. Here, we optimize an in vitro experimental model allowing us to record SD without focal stimulation, which we call spontaneous. This method uses only an altered extracellular buffer containing 0 mM Mg2+ and 5 mM K+ and makes it possible for simultaneous patch and extracellular recording in a submerged chamber plus intrinsic optical imaging in slices of either sex. We also add methods for quantification and show the quantified optical signal is much more complex than imaging alone would suggest. In brief, acute hippocampal slices were prepared with a chamber holding a submerged slice but with flow of artificial cerebrospinal fluid (aCSF) above and below, which we call interface-like. As soon as slices were placed in the chamber, aCSF with 0 Mg2+/5 K+ was used. Most mouse slices developed SD and did so in the first hour of 0 Mg2+/5 K+ aCSF exposure. In addition, prolonged bursts we call seizure-like events (SLEs) occurred, and the interactions between SD and SLEs suggest potentially important relationships. Differences between rats and mice in different chambers are described. Regarding optical imaging, SD originated in CA3 and the pattern of spread to CA1 and the dentate gyrus was similar in some ways to prior studies but also showed interesting differences. In summary, the methods are easy to use, provide new opportunities to study SD, new insights, and are inexpensive. They support previous suggestions that SD is diverse, and also suggest that participation by the dentate gyrus merits greater attention.
Collapse
Affiliation(s)
- Yi-Ling Lu
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - Helen E. Scharfman
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Department of Child and Adolescent Psychiatry, New York University Langone Health, New York, NY, United States
- Department of Neuroscience and Physiology, New York University Langone Health, New York, NY, United States
- Department of Psychiatry, New York University Langone Health, New York, NY, United States
| |
Collapse
|
13
|
Neocortical in vivo focal and spreading potassium responses and the influence of astrocytic gap junctional coupling. Neurobiol Dis 2020; 147:105160. [PMID: 33152505 DOI: 10.1016/j.nbd.2020.105160] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/29/2020] [Accepted: 10/31/2020] [Indexed: 11/21/2022] Open
Abstract
Raised extracellular potassium ion (K+) concentration is associated with several disorders including migraine, stroke, neurotrauma and epilepsy. K+ spatial buffering is a well-known mechanism for extracellular K+ regulation/distribution. Astrocytic gap junction-mediated buffering is a controversial candidate for K+ spatial buffering. To further investigate the existence of a K+ spatial buffering and to assess the involvement of astrocytic gap junctional coupling in K+ redistribution, we hypothesized that neocortical K+ and concomitant spreading depolarization (SD)-like responses are controlled by powerful local K+ buffering mechanisms and that K+ buffering/redistribution occurs partially through gap junctional coupling. Herein, we show, in vivo, that a threshold amount of focally applied KCl is required to trigger local and/or distal K+ responses, accompanied by a SD-like response. This observation indicates the presence of powerful local K+ buffering which mediates a rapid return of extracellular K+ to the baseline. Application of gap junctional blockers, carbenoxolone and Gap27, partially modulated the amplitude and shape of the K+ response and noticeably decreased the velocity of the spreading K+ and SD-like responses. Opening of gap junctions by trimethylamine, slightly decreased the amplitude of the K+ response and markedly increased the velocity of redistribution of K+ and SD-like events. We conclude that spreading K+ responses reflect powerful local K+ buffering mechanisms which are partially modulated by gap junctional communication. Gap junctional coupling mainly affected the velocity of the K+ and SD-like responses.
Collapse
|
14
|
Zhou T, Lin L, Hao C, Liao W. Environmental enrichment rescues cognitive impairment with suppression of TLR4-p38MAPK signaling pathway in vascular dementia rats. Neurosci Lett 2020; 737:135318. [PMID: 32846221 DOI: 10.1016/j.neulet.2020.135318] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 01/23/2023]
Abstract
Increasing evidence demonstrated the promising effects of environmental enrichment (EE) on brain recovery and cognitive performance in animal models of various diseases. However, the effect and molecular mechanisms of EE on vascular dementia (VD) remain to be studied. The aim of this study was to explore the effect of EE on cognitive decline and its mechanism. Sprague-Dawley rats underwent 2-vessel occlusion (2-VO) surgery or sham operation. Subsequently, rats were kept in EE for 4 weeks. In Morris water maze (MWM) test, we demonstrated that EE significantly improved cognitive function in rats with VD. HE staining exhibited morphological changes of neurons and quantitative analysis of TUNEL showed increased apoptotic neurons in hippocampal CA1 region following 2-VO. Results from RT-qPCR showed up-regulation of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) after 2-VO. Western blotting analysis revealed enhanced toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MYD88) and phosphorylated p38 mitogen-activated protein kinase (p-p38MAPK) in 2-VO rats. Whereas administration of EE reduced apoptotic neurons, down-regulated inflammatory factors. Moreover, EE suppressed protein expression of TLR4-p38MAPK pathway. Spearman correlation analysis showed that improved cognitive function was associated with decreased expression of TLR4 and p-p38MAPK proteins. Thus, our study proved that EE has a prominent effect on cognitive impairment and neuronal damage following 2-VO by attenuating inflammation and apoptosis, which may be realized via inhibiting the TLR4-P38MAPK signaling pathway.
Collapse
Affiliation(s)
- Tiantian Zhou
- Departmenta of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Lu Lin
- Departmenta of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chizi Hao
- Departmenta of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Weijing Liao
- Departmenta of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
15
|
Kondziella D. The Neurology of Death and the Dying Brain: A Pictorial Essay. Front Neurol 2020; 11:736. [PMID: 32793105 PMCID: PMC7385288 DOI: 10.3389/fneur.2020.00736] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/15/2020] [Indexed: 01/18/2023] Open
Abstract
As neurologists earn their living with the preservation and restoration of brain function, they are also well-positioned to address the science behind the transition from life to death. This essay in pictures highlights areas of neurological expertise needed for brain death determination; shows pitfalls to avoid during the clinical examination and interpretation of confirmatory laboratory tests in brain death protocols; illustrates the great variability of brain death legislations around the world; discusses arguments for the implementation of donation after circulatory death (DCD); points to unresolved questions related to DCD and the time between cardiac standstill and organ procurement (“hands-off period”); provides an overview of the epidemiology and semiology of near-death experiences, including their importance for religion, literature, and the visual arts; suggests biological mechanisms for near-death experiences such as dysfunction of temporoparietal cortex, N-methyl-D-aspartate receptor antagonism, migraine aura, and rapid eye movement sleep; hypothesizes that thanatosis (aka. death-feigning, a common behavioral trait in the animal kingdom) represents the evolutionary origin of near-death experiences; and speculates about the future implications of recent attempts of brain resuscitation in an animal model. The aim is to provide the reader with a thorough understanding that the boundaries within the neurology of death and the dying brain are being pushed just like everywhere else in the clinical neurosciences.
Collapse
Affiliation(s)
- Daniel Kondziella
- Department of Neurology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
Flavin Adenine Dinucleotide Fluorescence as an Early Marker of Mitochondrial Impairment During Brain Hypoxia. Int J Mol Sci 2020; 21:ijms21113977. [PMID: 32492921 PMCID: PMC7312830 DOI: 10.3390/ijms21113977] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/28/2020] [Accepted: 05/30/2020] [Indexed: 12/31/2022] Open
Abstract
Multimodal continuous bedside monitoring is increasingly recognized as a promising option for early treatment stratification in patients at risk for ischemia during neurocritical care. Modalities used at present are, for example, oxygen availability and subdural electrocorticography. The assessment of mitochondrial function could be an interesting complement to these modalities. For instance, flavin adenine dinucleotide (FAD) fluorescence permits direct insight into the mitochondrial redox state. Therefore, we explored the possibility of using FAD fluorometry to monitor consequences of hypoxia in brain tissue in vitro and in vivo. By combining experimental results with computational modeling, we identified the potential source responsible for the fluorescence signal and gained insight into the hypoxia-associated metabolic changes in neuronal energy metabolism. In vitro, hypoxia was characterized by a reductive shift of FAD, impairment of synaptic transmission and increasing interstitial potassium [K+]o. Computer simulations predicted FAD changes to originate from the citric acid cycle enzyme α-ketoglutarate dehydrogenase and pyruvate dehydrogenase. In vivo, the FAD signal during early hypoxia displayed a reductive shift followed by a short oxidation associated with terminal spreading depolarization. In silico, initial tissue hypoxia followed by a transient re-oxygenation phase due to glucose depletion might explain FAD dynamics in vivo. Our work suggests that FAD fluorescence could be readily used to monitor mitochondrial function during hypoxia and represents a potential diagnostic tool to differentiate underlying metabolic processes for complementation of multimodal brain monitoring.
Collapse
|
17
|
Oliveira-Ferreira AI, Major S, Przesdzing I, Kang EJ, Dreier JP. Spreading depolarizations in the rat endothelin-1 model of focal cerebellar ischemia. J Cereb Blood Flow Metab 2020; 40:1274-1289. [PMID: 31280632 PMCID: PMC7232780 DOI: 10.1177/0271678x19861604] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Focal brain ischemia is best studied in neocortex and striatum. Both show highly vulnerable neurons and high susceptibility to spreading depolarization (SD). Therefore, it has been hypothesized that these two variables generally correlate. However, this hypothesis is contradicted by findings in cerebellar cortex, which contains highly vulnerable neurons to ischemia, the Purkinje cells, but is said to be less susceptible to SD. Here, we found in the rat cerebellar cortex that elevated K+ induced a long-lasting depolarizing event superimposed with SDs. Cerebellar SDs resembled those in neocortex, but negative direct current (DC) shifts and regional blood flow responses were usually smaller. The K+ threshold for SD was higher in cerebellum than in previous studies in neocortex. We then topically applied endothelin-1 (ET-1) to the cerebellum, which is assumed to cause SD via vasoconstriction-induced focal ischemia. Although the blood flow decrease was similar to that in previous studies in neocortex, the ET-1 threshold for SD was higher. Quantitative cell counting found that the proportion of necrotic Purkinje cells was significantly higher in ET-1-treated rats than sham controls even if ET-1 had not caused SDs. Our results suggest that ischemic death of Purkinje cells does not require the occurrence of SD.
Collapse
Affiliation(s)
- Ana I Oliveira-Ferreira
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sebastian Major
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ingo Przesdzing
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Eun-Jeung Kang
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jens P Dreier
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany.,Einstein Center for Neurosciences Berlin, Berlin, Germany
| |
Collapse
|
18
|
Abstract
Cortical spreading depolarizations (SD) are strongly associated with worse tissue injury and clinical outcomes in the setting of aneurysmal subarachnoid hemorrhage (SAH). Animal studies have suggested a causal relationship, and new therapies to target SDs are starting to be tested in clinical studies. A recent set of single-center randomized trials assessed the effect of the phosphodiesterase inhibitor cilostazol in patients with SAH. Cilostazol led to improved functional outcomes and SD-related metrics in treated patients through a putative mechanism of improved cerebral blood flow. Another promising therapeutic approach includes attempts to block SDs with, for example, the NMDA receptor antagonist ketamine. SDs have emerged not only as a therapeutic target but also as a potentially useful biomarker for brain injury following SAH. Additional clinical and preclinical experimental work is greatly needed to assess the generalizability of existing therapeutic trials and to better delineate the relationship between SDs, SAH, and functional outcome.
Collapse
Affiliation(s)
- Kazutaka Sugimoto
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, 6403, Charlestown, MA, 02129, USA
- Department of Neurosurgery, Yamaguchi University School of Medicine, Ube, Japan
| | - David Y Chung
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, 6403, Charlestown, MA, 02129, USA.
- Division of Neurocritical Care, Department of Neurology, Boston Medical Center, Boston, MA, USA.
- Division of Neurocritical Care, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Reiffurth C, Alam M, Zahedi-Khorasani M, Major S, Dreier JP. Na +/K +-ATPase α isoform deficiency results in distinct spreading depolarization phenotypes. J Cereb Blood Flow Metab 2020; 40:622-638. [PMID: 30819023 PMCID: PMC7025397 DOI: 10.1177/0271678x19833757] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Compromised Na+/K+-ATPase function is associated with the occurrence of spreading depolarization (SD). Mutations in ATP1A2, the gene encoding the α2 isoform of the Na+/K+-ATPase, were identified in patients with familial hemiplegic migraine type 2 (FHM2), a Mendelian model disease for SD. This suggests a distinct role for the α2 isoform in modulating SD susceptibility and raises questions about underlying mechanisms including the roles of other Na+/K+-ATPase α isoforms. Here, we investigated the effects of genetic ablation and pharmacological inhibition of α1, α2, and α3 on SD using heterozygous knock-out mice. We found that only α2 heterozygous mice displayed higher SD susceptibility when challenged with prolonged extracellular high potassium concentration ([K+]o), a pronounced post SD oligemia and higher SD speed in-vivo. By contrast, under physiological [K+]o, α2 heterozygous mice showed similar SD susceptibility compared to wild-type littermates. Deficiency of α3 resulted in increased resistance against electrically induced SD in-vivo, whereas α1 deficiency did not affect SD. The results support important roles of the α2 isoform in SD. Moreover, they suggest that specific experimental conditions can be necessary to reveal an inherent SD phenotype by driving a (meta-) stable system into decompensation, reminiscent of the episodic nature of SDs in various diseases.
Collapse
Affiliation(s)
- Clemens Reiffurth
- Department of Experimental Neurology, Charité-University Medicine Berlin, Berlin, Germany.,Center for Stroke Research, Charité-University Medicine Berlin, Berlin, Germany
| | - Mesbah Alam
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - Mahdi Zahedi-Khorasani
- Research Center and Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Sebastian Major
- Department of Experimental Neurology, Charité-University Medicine Berlin, Berlin, Germany.,Center for Stroke Research, Charité-University Medicine Berlin, Berlin, Germany.,Department of Neurology, Charité-University Medicine Berlin, Berlin, Germany
| | - Jens P Dreier
- Department of Experimental Neurology, Charité-University Medicine Berlin, Berlin, Germany.,Center for Stroke Research, Charité-University Medicine Berlin, Berlin, Germany.,Department of Neurology, Charité-University Medicine Berlin, Berlin, Germany.,Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany.,Einstein Center for Neurosciences Berlin, Berlin, Germany
| |
Collapse
|
20
|
Kondziella D, Olsen MH, Lemale CL, Dreier JP. Migraine aura, a predictor of near-death experiences in a crowdsourced study. PeerJ 2019; 7:e8202. [PMID: 31824781 PMCID: PMC6898989 DOI: 10.7717/peerj.8202] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/13/2019] [Indexed: 12/20/2022] Open
Abstract
Background Near-death experiences (NDE) occur with imminent death and in situations of stress and danger but are poorly understood. Evidence suggests that NDE are associated with rapid eye movement (REM) sleep intrusion, a feature of narcolepsy. Previous studies further found REM abnormalities and an increased frequency of dream-enacting behavior in migraine patients, as well as an association between migraine with aura and narcolepsy. We therefore investigated if NDE are more common in people with migraine aura. Methods We recruited 1,037 laypeople from 35 countries and five continents, without any filters except for English language and age ≥18 years, via a crowdsourcing platform. Reports were validated using the Greyson NDE Scale. Results Eighty-one of 1,037 participants had NDE (7.8%; CI [6.3-9.7%]). There were no significant associations between NDE and age (p > 0.6, t-test independent samples) or gender (p > 0.9, Chi-square test). The only significant association was between NDE and migraine aura: 48 (6.1%) of 783 subjects without migraine aura and 33 (13.0%) of 254 subjects with migraine aura had NDE (p < 0.001, odds ratio (OR) = 2.29). In multiple logistic regression analysis, migraine aura remained significant after adjustment for age (p < 0.001, OR = 2.31), gender (p < 0.001, OR = 2.33), or both (p < 0.001, OR = 2.33). Conclusions In our sample, migraine aura was a predictor of NDE. This indirectly supports the association between NDE and REM intrusion and might have implications for the understanding of NDE, because a variant of spreading depolarization (SD), terminal SD, occurs in humans at the end of life, while a short-lasting variant of SD is considered the pathophysiological correlate of migraine aura.
Collapse
Affiliation(s)
- Daniel Kondziella
- Department of Neurology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Markus Harboe Olsen
- Department of Neuroanesthesiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Coline L Lemale
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Jens P Dreier
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany.,Einstein Center for Neurosciences Berlin, Berlin, Germany
| |
Collapse
|
21
|
Nordström T, Andersson LC, Åkerman KE. Regulation of intracellular pH by electrogenic Na+/HCO3– co-transporters in embryonic neural stem cell-derived radial glia-like cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:1037-1048. [DOI: 10.1016/j.bbamem.2019.03.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/11/2019] [Accepted: 03/14/2019] [Indexed: 01/19/2023]
|
22
|
Chrysin prevents cognitive and hippocampal long-term potentiation deficits and inflammation in rat with cerebral hypoperfusion and reperfusion injury. Life Sci 2019; 226:202-209. [DOI: 10.1016/j.lfs.2019.04.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/05/2019] [Accepted: 04/12/2019] [Indexed: 01/05/2023]
|
23
|
Memory deficits and hippocampal inflammation in cerebral hypoperfusion and reperfusion in male rats: Neuroprotective role of vanillic acid. Life Sci 2018; 211:126-132. [PMID: 30195619 DOI: 10.1016/j.lfs.2018.08.065] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 08/20/2018] [Accepted: 08/27/2018] [Indexed: 11/20/2022]
Abstract
Ischemic stroke is one of the leading causes of neurological deterioration and mortality worldwide. Neuroprotective strategies are being investigated to minimize cognitive deficits after ischemic events. Here we investigated the neuroprotective potential of vanillic acid (VA) in an animal model of transient bilateral common carotid artery occlusion and reperfusion (BCCAO/R). Adult male Wistar rats (250-300 g) were randomly divided in 4 groups and submitted to either cerebral hypoperfusion-reperfusion or a sham surgery after two-weeks of pretreatment with VA and/or normal saline. To induce the animal model of hypoperfusion, bilateral common carotid arteries were occluded (2VO model) for 30 min, followed by 72 h of reperfusion. Subsequently, their cognitive performance was evaluated in a Morris water maze (MWM) test, and also hippocampi were removed for ELISA assays and TUNEL staining test. The results showed that 2VO significantly reduced the spatial memory performance in MWM. As well as, BCCAO/R increased the level of IL-6, TNF-α and TUNEL positive cells, and also decreased the contents of IL-10 in the hippocampus of vehicle- pretreated groups as compared to the sham-operated groups. Furthermore, 14 consecutive days pretreatment with VA significantly restored the spatial memory, decreased the levels of IL-6, TNF-α and TUNEL positive cells and also increased the IL-10 levels in the hippocampi of the BCCAO/R rats. VA alone did not show any change neither in the status of various cytokines nor behavioral and TUNEL staining tests over sham values. Our data confirm that VA could potentially serve as a novel, promising, and accessible neuroprotective agent against cerebrovascular insufficiency states and vascular dementia.
Collapse
|
24
|
Khoshnam SE, Sarkaki A, Khorsandi L, Winlow W, Badavi M, Moghaddam HF, Farbood Y. Vanillic acid attenuates effects of transient bilateral common carotid occlusion and reperfusion in rats. Biomed Pharmacother 2017; 96:667-674. [DOI: 10.1016/j.biopha.2017.10.052] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 10/02/2017] [Accepted: 10/10/2017] [Indexed: 12/26/2022] Open
|
25
|
Dreier JP, Lemale CL, Kola V, Friedman A, Schoknecht K. Spreading depolarization is not an epiphenomenon but the principal mechanism of the cytotoxic edema in various gray matter structures of the brain during stroke. Neuropharmacology 2017; 134:189-207. [PMID: 28941738 DOI: 10.1016/j.neuropharm.2017.09.027] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 09/16/2017] [Accepted: 09/19/2017] [Indexed: 12/15/2022]
Abstract
Spreading depolarization (SD) is a phenomenon of various cerebral gray matter structures that only occurs under pathological conditions. In the present paper, we summarize the evidence from several decades of research that SD and cytotoxic edema in these structures are largely overlapping terms. SD/cytotoxic edema is a toxic state that - albeit initially reversible - leads eventually to cellular death when it is persistent. Both hemorrhagic and ischemic stroke are among the most prominent causes of SD/cytotoxic edema. SD/cytotoxic edema is the principal mechanism that mediates neuronal death in these conditions. This applies to gray matter structures in both the ischemic core and the penumbra. SD/cytotoxic edema is often a single terminal event in the core whereas, in the penumbra, a cluster of repetitive prolonged SDs is typical. SD/cytotoxic edema also propagates widely into healthy surrounding tissue as short-lasting, relatively harmless events so that regional electrocorticographic monitoring affords even remote detection of ischemic zones. Ischemia cannot only cause SD/cytotoxic edema but it can also be its consequence through inverse neurovascular coupling. Under this condition, ischemia does not start simultaneously in different regions but spreads in the tissue driven by SD/cytotoxic edema-induced microvascular constriction (= spreading ischemia). Spreading ischemia prolongs SD/cytotoxic edema. Thus, it increases the likelihood for the transition from SD/cytotoxic edema into cellular death. Vasogenic edema is the other major type of cerebral edema with relevance to ischemic stroke. It results from opening of the blood-brain barrier. SD/cytotoxic edema and vasogenic edema are distinct processes with important mutual interactions. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
Collapse
Affiliation(s)
- Jens P Dreier
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany; Departments of Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany; Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.
| | - Coline L Lemale
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Vasilis Kola
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Alon Friedman
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, Canada
| | - Karl Schoknecht
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany; Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
26
|
Lippmann K, Kamintsky L, Kim SY, Lublinsky S, Prager O, Nichtweiss JF, Salar S, Kaufer D, Heinemann U, Friedman A. Epileptiform activity and spreading depolarization in the blood-brain barrier-disrupted peri-infarct hippocampus are associated with impaired GABAergic inhibition and synaptic plasticity. J Cereb Blood Flow Metab 2017; 37:1803-1819. [PMID: 27252228 PMCID: PMC5435286 DOI: 10.1177/0271678x16652631] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Peri-infarct opening of the blood-brain barrier may be associated with spreading depolarizations, seizures, and epileptogenesis as well as cognitive dysfunction. We aimed to investigate the mechanisms underlying neural network pathophysiology in the blood-brain barrier-dysfunctional hippocampus. Photothrombotic stroke within the rat neocortex was associated with increased intracranial pressure, vasogenic edema, and peri-ischemic blood-brain barrier dysfunction that included the ipsilateral hippocampus. Intrahippocampal recordings revealed electrographic seizures within the first week in two-thirds of animals, accompanied by a reduction in gamma and increase in theta frequency bands. Synaptic interactions were studied in parasagittal hippocampal slices at 24 h and seven days post-stroke. Field potential recordings in CA1 and CA3 uncovered multiple population spikes, epileptiform episodes, and spreading depolarizations at 24 h. Input-output analysis revealed that fEPSP-spike coupling was significantly enhanced at seven days. In addition, CA1 feedback and feedforward inhibition were diminished. Slices generating epileptiform activity at seven days revealed impaired bidirectional long-term plasticity following high and low-frequency stimulation protocols. Microarray and PCR data confirmed changes in expression of astrocyte-related genes and suggested downregulation in expression of GABAA-receptor subunits. We conclude that blood-brain barrier dysfunction in the peri-infarct hippocampus is associated with early disinhibition, hyperexcitability, and abnormal synaptic plasticity.
Collapse
Affiliation(s)
- Kristina Lippmann
- 1 Institute of Neurophysiology, Charité - University Medicine Berlin, Berlin, Germany.,2 Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig, Germany
| | - Lyn Kamintsky
- 3 Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Soo Young Kim
- 4 Helen Wills Neuroscience Institute and the Department of Integrative Biology, University of California, Berkeley, CA, USA
| | - Svetlana Lublinsky
- 3 Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ofer Prager
- 3 Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Seda Salar
- 1 Institute of Neurophysiology, Charité - University Medicine Berlin, Berlin, Germany
| | - Daniela Kaufer
- 4 Helen Wills Neuroscience Institute and the Department of Integrative Biology, University of California, Berkeley, CA, USA
| | - Uwe Heinemann
- 5 Neuroscience Research Center, Berlin, Germany.,6 Excellence Cluster NeuroCure, Berlin, Germany
| | - Alon Friedman
- 3 Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,7 Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, Canada
| |
Collapse
|
27
|
Dergacheva O, Yamanaka A, Schwartz AR, Polotsky VY, Mendelowitz D. Hypoxia and hypercapnia inhibit hypothalamic orexin neurons in rats. J Neurophysiol 2016; 116:2250-2259. [PMID: 27559138 DOI: 10.1152/jn.00196.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 08/21/2016] [Indexed: 01/18/2023] Open
Abstract
Evidence of impaired function of orexin neurons has been found in individuals with cardiorespiratory disorders, such as obstructive sleep apnea (OSA) and sudden infant death syndrome (SIDS), but the mechanisms responsible are unknown. Individuals with OSA and SIDS experience repetitive breathing cessations and/or rebreathing of expired air, resulting in hypoxia/hypercapnia (H/H). In this study, we examined the responses of fluorescently identified rat orexin neurons in the lateral hypothalamus to acute H/H to test if and how these neurons alter their activity and function during this challenge. Experiments were conducted in an in vitro slice preparation using voltage-clamp and current-clamp configurations. H/H (10 min) induced hyperpolarization, accompanied by rapid depression, and finally, cessation of firing activity in orexin neurons. Hypoxia alone had similar but less potent effects. H/H did not alter the frequency of inhibitory glycinergic postsynaptic currents. The frequency of GABAergic currents was diminished but only at 8-10 min of H/H. In contrast, the frequency of excitatory glutamatergic postsynaptic events was diminished as early as 2-4 min of H/H. In the presence of glutamatergic receptor blockers, the inhibitory effects of H/H on the firing activity and membrane potential of orexin neurons persisted but to a lesser extent. In conclusion, both direct alteration of postsynaptic membrane properties and diminished glutamatergic neurotransmission likely contribute to the inhibition of orexin neurons by H/H. These mechanisms could be responsible for the decreased function of orexin in individuals at risk for OSA and SIDS.
Collapse
Affiliation(s)
- Olga Dergacheva
- Department of Pharmacology and Physiology, The George Washington University, Washington, DC;
| | - Akihiro Yamanaka
- Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan; and
| | - Alan R Schwartz
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - David Mendelowitz
- Department of Pharmacology and Physiology, The George Washington University, Washington, DC
| |
Collapse
|
28
|
Oikonomou KD, Singh MB, Rich MT, Short SM, Antic SD. Contribution of extrasynaptic N-methyl-D-aspartate and adenosine A1 receptors in the generation of dendritic glutamate-mediated plateau potentials. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0193. [PMID: 26009772 DOI: 10.1098/rstb.2014.0193] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Thin basal dendrites can strongly influence neuronal output via generation of dendritic spikes. It was recently postulated that glial processes actively support dendritic spikes by either ceasing glutamate uptake or by actively releasing glutamate and adenosine triphosphate (ATP). We used calcium imaging to study the role of NR2C/D-containing N-methyl-d-aspartate (NMDA) receptors and adenosine A1 receptors in the generation of dendritic NMDA spikes and plateau potentials in basal dendrites of layer 5 pyramidal neurons in the mouse prefrontal cortex. We found that NR2C/D glutamate receptor subunits contribute to the amplitude of synaptically evoked NMDA spikes. Dendritic calcium signals associated with glutamate-evoked dendritic plateau potentials were significantly shortened upon application of the NR2C/D receptor antagonist PPDA, suggesting that NR2C/D receptors prolong the duration of calcium influx during dendritic spiking. In contrast to NR2C/D receptors, adenosine A1 receptors act to abbreviate dendritic and somatic signals via the activation of dendritic K(+) current. This current is characterized as a slow-activating outward-rectifying voltage- and adenosine-gated current, insensitive to 4-aminopyridine but sensitive to TEA. Our data support the hypothesis that the release of glutamate and ATP from neurons or glia contribute to initiation, maintenance and termination of local dendritic glutamate-mediated regenerative potentials.
Collapse
Affiliation(s)
- Katerina D Oikonomou
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Mandakini B Singh
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Matthew T Rich
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Shaina M Short
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Srdjan D Antic
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
29
|
Bayat M, Sharifi MD, Haghani M, Shabani M. Enriched environment improves synaptic plasticity and cognitive deficiency in chronic cerebral hypoperfused rats. Brain Res Bull 2015; 119:34-40. [PMID: 26474515 DOI: 10.1016/j.brainresbull.2015.10.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 10/02/2015] [Accepted: 10/07/2015] [Indexed: 12/12/2022]
Abstract
Recent studies have indicated that environmental enrichment (EE) increases the sensorial and social stimulations and leads to strengthened plastic changes in the brain. In models of chronic cerebral hypoperfusion, the ability of an EE to restore the cognition depends on hippocampal synaptic plasticity. The mechanisms for this effect have not, however, been adequately studied. Thus, the aim of the present study was to evaluate the neuroprotective effects and underlying mechanism of environmental enrichment by assessment of spatial memory tasks as well as parameters of synaptic plasticity in rats subjected to occlusion of the bilateral common carotid arteries (2-VO) model. Male Sprague-Dawley rats were used in this study. The model group was established by occlusion of the bilateral common carotid arteries. The animals were tested for learning, memory performance and synaptic plasticity using Morris water maze (MWM), 8-arm Radial Maze (RM), and field potential recording, respectively. The rats subjected to 2-VO in EE exhibited a significantly lower number of working errors and reference errors in RM. Moreover, the enriched environment recovered the memory performance of hypoperfused rats and decreased the swimming time to reach the platform in MWM. In addition, conditions of the environment did not have any effect on baseline synaptic transmission and presynaptic plasticity, but housing the animals in EE rescued the impairment of LTP induction induced by 2-VO. These results suggest that EE ameliorates the LTP and spatial memory impairment induced by 2-VO. Our data indicated that the LTP recovery by EE in the rat models of 2-VO is probably mediated by post-synaptic mechanisms.
Collapse
Affiliation(s)
- Mahnaz Bayat
- Department of Physiology, International Branch, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad Davood Sharifi
- Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Masoud Haghani
- Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad Shabani
- Kerman Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
30
|
Abstract
The term spreading depolarization (SD) refers to waves of abrupt, sustained mass depolarization in gray matter of the CNS. SD, which spreads from neuron to neuron in affected tissue, is characterized by a rapid near-breakdown of the neuronal transmembrane ion gradients. SD can be induced by hypoxic conditions--such as from ischemia--and facilitates neuronal death in energy-compromised tissue. SD has also been implicated in migraine aura, where SD is assumed to ascend in well-nourished tissue and is typically benign. In addition to these two ends of the "SD continuum," an SD wave can propagate from an energy-depleted tissue into surrounding, well-nourished tissue, as is often the case in stroke and brain trauma. This review presents the neurobiology of SD--its triggers and propagation mechanisms--as well as clinical manifestations of SD, including overlaps and differences between migraine aura and stroke, and recent developments in neuromonitoring aimed at better diagnosis and more targeted treatments.
Collapse
Affiliation(s)
- Jens P Dreier
- Department of Neurology, Charité University Medicine Berlin, 10117 Berlin, Germany; Department of Experimental Neurology, Charité University Medicine Berlin, 10117 Berlin, Germany; Center for Stroke Research, Charité University Medicine Berlin, 10117 Berlin, Germany.
| | - Clemens Reiffurth
- Department of Experimental Neurology, Charité University Medicine Berlin, 10117 Berlin, Germany; Center for Stroke Research, Charité University Medicine Berlin, 10117 Berlin, Germany
| |
Collapse
|
31
|
Schoknecht K, David Y, Heinemann U. The blood-brain barrier-gatekeeper to neuronal homeostasis: clinical implications in the setting of stroke. Semin Cell Dev Biol 2014; 38:35-42. [PMID: 25444848 DOI: 10.1016/j.semcdb.2014.10.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 10/24/2014] [Accepted: 10/31/2014] [Indexed: 12/23/2022]
Abstract
The blood-brain barrier is part of the neurovascular unit and serves as a functional and anatomical barrier between the blood and the extracellular space. It controls the flow of solutes in and out of the brain thereby providing an optimal environment for neuronal functioning. Paracellular transport between endothelial cells is restricted by tight junctions and transendothelial transport is reduced and more selective compared to capillaries of other organs. Further, the blood-brain barrier is involved in controlling blood flow and it is the site for signaling damage of the nervous system to the peripheral immune system. As an important player in brain homeostasis, blood-brain barrier dysfunction has been implicated in the pathophysiology of many brain diseases including stroke, traumatic brain injury, brain tumors, epilepsy and neurodegenerative disorders. In this article - highlighting recent advances in basic science - we review the features of the blood-brain barrier and their significance for neuronal homeostasis to discuss clinical implications for neurological complications following cerebral ischemia.
Collapse
Affiliation(s)
- Karl Schoknecht
- Institute for Neurophysiology, Charité - University Medicine Berlin, Germany
| | - Yaron David
- Departments of Physiology & Cell Biology, Cognitive & Brain Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Uwe Heinemann
- Institute for Neurophysiology, Charité - University Medicine Berlin, Germany.
| |
Collapse
|
32
|
Geiseler SJ, Ludvigsen S, Folkow LP. KATP-channels play a minor role in the protective hypoxic shut-down of cerebellar activity in eider ducks (Somateria mollissima). Neuroscience 2014; 284:751-758. [PMID: 25451290 DOI: 10.1016/j.neuroscience.2014.10.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 10/02/2014] [Accepted: 10/15/2014] [Indexed: 10/24/2022]
Abstract
Eider duck (Somateria mollissima) cerebellar neurons are highly tolerant toward hypoxia in vitro, which in part is due to a hypoxia-induced depression of their spontaneous activity. We have studied whether this response involves ATP-sensitive potassium (KATP) channels, which are known to be involved in the hypoxic/ischemic defense of mammalian neural and muscular tissues, by causing hyperpolarization and reduced ATP demand. Extracellular recordings in the Purkinje layer of isolated normoxic eider duck cerebellar slices showed that their spontaneous neuronal activity decreased significantly compared to in control slices when the KATP channel opener diazoxide (600 μM) was added (F1,70=92.781, p<0.001). Adding the KATP channel blocker tolbutamide (400 μM) 5 min prior to diazoxide completely abolished its effect (F1,55=39.639, p<0.001), strongly suggesting that these drugs have a similar mode of action in this avian species as in mammals. The spontaneous activity of slices treated with tolbutamide in combined hypoxia/chemical anoxia (95% N2-5% CO2 and 2 mM NaCN) was not significantly different from that of control slices (F1,203=0.071, p=0.791). Recovery from hypoxia/anoxia was, however, slightly but significantly weaker in tolbutamide-treated slices than in control slices (F1,137=15.539, p<0.001). We conclude that KATP channels are present in eider duck cerebellar neurons and are activated in hypoxia/anoxia, but that they do not play a key role in the protective shut-down response to hypoxia/anoxia.
Collapse
Affiliation(s)
- S J Geiseler
- Department of Arctic and Marine Biology, University of Tromsø - the Arctic University of Norway, Breivika, NO-9037 Tromsø, Norway.
| | - S Ludvigsen
- Department of Arctic and Marine Biology, University of Tromsø - the Arctic University of Norway, Breivika, NO-9037 Tromsø, Norway
| | - L P Folkow
- Department of Arctic and Marine Biology, University of Tromsø - the Arctic University of Norway, Breivika, NO-9037 Tromsø, Norway
| |
Collapse
|
33
|
Erken HA, Erken G, Colak R, Genç O. Exercise and DHA prevent the negative effects of hypoxia on EEG and nerve conduction velocity. High Alt Med Biol 2014; 14:360-6. [PMID: 24377343 DOI: 10.1089/ham.2012.1125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
It is known that hypoxia has a negative effect on nervous system functions, but exercise and DHA (docosahexaenoic acid) have positive effect. In this study, it was investigated whether exercise and/or DHA can prevent the effects of hypoxia on EEG and nerve conduction velocity (NCV). 35 adult Wistar albino male rats were divided into five groups (n=7): control (C), hypoxia (H), hypoxia and exercise (HE), hypoxia and DHA (HD), and hypoxia and exercise and DHA (HED) groups. During the 28-day hypoxia exposure, the HE and HED groups of rats were exercised (0% incline, 30 m/min speed, 20 min/day, 5 days a week). In addition, DHA (36 mg/kg/day) was given by oral gavage to rats in the HD and HED groups. While EEG records were taken before and after the experimental period, NCV records were taken after the experimental period from anesthetized rats. Data were analyzed by paired t-test, one-way ANOVA, and post hoc Tukey test. In this study, it was shown that exposure to hypoxia decreased theta activity and NCV, but exercise and DHA reduced the delta activity, while theta, alpha, beta activities, and NCV were increased. These results have shown that the effects of hypoxia exposure on EEG and NCV can be prevented by exercise and/or DHA.
Collapse
Affiliation(s)
- Haydar Ali Erken
- 1 Department of Physiology, Faculty of Medicine, Balikesir University , Balikesir, Turkey
| | | | | | | |
Collapse
|
34
|
Antunes AP, Schiefecker AJ, Beer R, Pfausler B, Sohm F, Fischer M, Dietmann A, Lackner P, Hackl WO, Ndayisaba JP, Thomé C, Schmutzhard E, Helbok R. Higher brain extracellular potassium is associated with brain metabolic distress and poor outcome after aneurysmal subarachnoid hemorrhage. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2014; 18:R119. [PMID: 24920041 PMCID: PMC4229847 DOI: 10.1186/cc13916] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 05/21/2014] [Indexed: 12/11/2022]
Abstract
Introduction Elevated brain potassium levels ([K+]) are associated with neuronal damage in experimental models. The role of brain extracellular [K+] in patients with poor-grade aneurysmal subarachnoid hemorrhage (aSAH) and its association with hemorrhage load, metabolic dysfunction and outcome has not been studied so far. Methods Cerebral microdialysis (CMD) samples from 28 poor grade aSAH patients were analyzed for CMD [K+] for 12 consecutive days after ictus, and time-matched to brain metabolic and hemodynamic parameters as well as corresponding plasma [K+]. Statistical analysis was performed using a generalized estimating equation with an autoregressive function to handle repeated observations of an individual patient. Results CMD [K+] did not correlate with plasma [K+] (Spearman’s ρ = 0.114, P = 0.109). Higher CMD [K+] was associated with the presence of intracerebral hematoma on admission head computed tomography, CMD lactate/pyruvate ratio >40 and CMD lactate >4 mmol/L (P < 0.05). In vitro retrodialysis data suggest that high CMD [K+] was of brain cellular origin. Higher CMD [K+] was significantly associated with poor 3-month outcome, even after adjusting for age and disease severity (P < 0.01). Conclusions The results of this pilot study suggest that brain extracellular [K+] may serve as a biomarker for brain tissue injury in poor-grade aSAH patients. Further studies are needed to elucidate the relevance of brain interstitial K+ levels in the pathophysiology of secondary brain injury after aSAH.
Collapse
|
35
|
Attenuated effects of Neu2000 on hypoxia-induced synaptic activities in a rat hippocampus. Arch Pharm Res 2013; 37:232-8. [PMID: 23733585 DOI: 10.1007/s12272-013-0170-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 05/26/2013] [Accepted: 05/28/2013] [Indexed: 10/26/2022]
Abstract
Neu2000 (NEU; 2-hydroxy-5-(2,3,5,6-tetrafluoro-4-trifluoromethyl-benzylamino)-benzoic acid), a recently developed derivative of acetylsalicylic acid and sulfasalazine, potently protects against neuronal cell death following ischemic brain injury by antagonizing NMDA receptor-mediated neuronal toxicity and oxidative stress. However, it has yet to be determined whether NEU can attenuate hypoxia-induced impairment of neuronal electrical activity. In this study, we carried out extracellular recordings of hippocampal slices in order to investigate the effects of NEU on the electrical activity of neurons exposed to a hypoxic insult (oxygen and glucose deprivation). NEU prominently suppressed hypoxia-induced impairment of neuronal activity in a concentration-dependent manner. NEU, at a low dose (1 μM), competently depressed the hypoxia-induced convulsive activity in a manner similar to trolox. Furthermore, high concentrations of NEU (50 μM) markedly abolished all hypoxia-mediated impairment of neuronal activity and accelerated the slow recovery of neuronal activity more efficiently than ifenprodil and APV. These results suggest that NEU attenuates hypoxia-induced impairment of neuronal activity more potently than the antioxidant, trolox, and the NMDA receptor antagonists, ifenprodil and APV. We propose that NEU is a striking pharmacological candidate for neuroprotection against hypoxia because of its defensive action on hypoxia-mediated impairment of electrical neurotransmission as well as its neuroprotective action against neuronal cell death induced by exposure to pathological hypoxic conditions.
Collapse
|
36
|
Autonomic Nervous System In Vitro: Studying Tonically Active Neurons Controlling Vagal Outflow in Rodent Brainstem Slices. ISOLATED CENTRAL NERVOUS SYSTEM CIRCUITS 2012. [DOI: 10.1007/978-1-62703-020-5_1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
37
|
Maslarova A, Alam M, Reiffurth C, Lapilover E, Gorji A, Dreier JP. Chronically epileptic human and rat neocortex display a similar resistance against spreading depolarization in vitro. Stroke 2011; 42:2917-22. [PMID: 21836085 DOI: 10.1161/strokeaha.111.621581] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Experimental and clinical evidence suggests that prolonged spreading depolarizations (SDs) are a promising target for therapeutic intervention in stroke because they recruit tissue at risk into necrosis by protracted intracellular calcium surge and massive glutamate release. Unfortunately, unlike SDs in healthy tissue, they are resistant to drugs such as N-methyl-d-aspartate-receptor antagonists. This drug resistance of SD in low perfusion areas may be due to the gradual rise of extracellular potassium before SD onset. Brain slices from patients undergoing surgery for intractable epilepsy allow for screening of drugs, targeting pharmacoresistant SDs under elevated potassium in human tissue. However, network changes associated with epilepsy may interfere with tissue susceptibility to SD. This could distort the results of pharmacological tests. METHODS We investigated the threshold for SD, induced by a gradual rise of potassium, in neocortex slices of patients with intractable epilepsy and of chronically epileptic rats as well as age-matched and younger control rats using combined extracellular potassium/field recordings and intrinsic optical imaging. RESULTS Both age and epilepsy significantly increased the potassium threshold, which was similarly high in epileptic rat and human slices (23.6±2.4 mmol/L versus 22.3±2.8 mmol/L). CONCLUSIONS Our results suggest that chronic epilepsy confers resistance against SD. This should be considered when human tissue is used for screening of neuroprotective drugs. The finding of similar potassium thresholds for SD in epileptic human and rat neocortex challenges previous speculations that the resistance of the human brain against SD is markedly higher than that of the rodent brain.
Collapse
Affiliation(s)
- Anna Maslarova
- Institute of Neurophysiology, Center for Stroke Research Berlin, Department of Experimental Neurology, Charité University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | | | | | | | | | | |
Collapse
|
38
|
Shimoda LA, Polak J. Hypoxia. 4. Hypoxia and ion channel function. Am J Physiol Cell Physiol 2011; 300:C951-67. [PMID: 21178108 PMCID: PMC3093942 DOI: 10.1152/ajpcell.00512.2010] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 12/16/2010] [Indexed: 12/19/2022]
Abstract
The ability to sense and respond to oxygen deprivation is required for survival; thus, understanding the mechanisms by which changes in oxygen are linked to cell viability and function is of great importance. Ion channels play a critical role in regulating cell function in a wide variety of biological processes, including neuronal transmission, control of ventilation, cardiac contractility, and control of vasomotor tone. Since the 1988 discovery of oxygen-sensitive potassium channels in chemoreceptors, the effect of hypoxia on an assortment of ion channels has been studied in an array of cell types. In this review, we describe the effects of both acute and sustained hypoxia (continuous and intermittent) on mammalian ion channels in several tissues, the mode of action, and their contribution to diverse cellular processes.
Collapse
Affiliation(s)
- Larissa A Shimoda
- Div. of Pulmonary and Critical Care Medicine, Johns Hopkins University, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA.
| | | |
Collapse
|
39
|
The role of spreading depression, spreading depolarization and spreading ischemia in neurological disease. Nat Med 2011; 17:439-47. [PMID: 21475241 DOI: 10.1038/nm.2333] [Citation(s) in RCA: 822] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The term spreading depolarization describes a wave in the gray matter of the central nervous system characterized by swelling of neurons, distortion of dendritic spines, a large change of the slow electrical potential and silencing of brain electrical activity (spreading depression). In the clinic, unequivocal electrophysiological evidence now exists that spreading depolarizations occur abundantly in individuals with aneurismal subarachnoid hemorrhage, delayed ischemic stroke after subarachnoid hemorrhage, malignant hemispheric stroke, spontaneous intracerebral hemorrhage or traumatic brain injury. Spreading depolarization is induced experimentally by various noxious conditions including chemicals such as potassium, glutamate, inhibitors of the sodium pump, status epilepticus, hypoxia, hypoglycemia and ischemia, but it can can also invade healthy, naive tissue. Resistance vessels respond to it with tone alterations, causing either transient hyperperfusion (physiological hemodynamic response) in healthy tissue or severe hypoperfusion (inverse hemodynamic response, or spreading ischemia) in tissue at risk for progressive damage, which contributes to lesion progression. Therapies that target spreading depolarization or the inverse hemodynamic response may potentially treat these neurological conditions.
Collapse
|
40
|
Kron M, Müller M. Impaired hippocampal Ca2+ homeostasis and concomitant K+ channel dysfunction in a mouse model of Rett syndrome during anoxia. Neuroscience 2010; 171:300-15. [PMID: 20732392 DOI: 10.1016/j.neuroscience.2010.08.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2010] [Revised: 08/12/2010] [Accepted: 08/16/2010] [Indexed: 11/19/2022]
Abstract
Methyl-CpG-binding protein 2 (MeCP2) deficiency causes Rett syndrome (RTT), a neurodevelopmental disorder characterized by severe cognitive impairment, synaptic dysfunction, and hyperexcitability. Previously we reported that the hippocampus of MeCP2-deficient mice (Mecp2(-/y)), a mouse model for RTT, is more susceptible to hypoxia. To identify the underlying mechanisms we now focused on the anoxic responses of wildtype (WT) and Mecp2(-/y) CA1 neurons in acute hippocampal slices. Intracellular recordings revealed that Mecp2(-/y) neurons show only reduced or no hyperpolarizations early during cyanide-induced anoxia, suggesting potassium channel (K(+) channel) dysfunction. Blocking adenosine-5'-triphosphate-sensitive K(+) channels (K(ATP-)) and big-conductance Ca(2+)-activated K(+) channels (BK-channels) did not affect the early anoxic hyperpolarization in either genotype. However, blocking Ca(2+) release from the endoplasmic reticulum almost abolished the anoxic hyperpolarizations in Mecp2(-/y) neurons. Single-channel recordings confirmed that neither K(ATP)- nor BK-channels are the sole mediators of the early anoxic hyperpolarization. Instead, anoxia Ca(2+)-dependently activated various small/intermediate-conductance K(+) channels in WT neurons, which was less evident in Mecp2(-/y) neurons. Yet, pharmacologically increasing the Ca(2+) sensitivity of small/intermediate-conductance K(Ca) channels fully restored the anoxic hyperpolarization in Mecp2(-/y) neurons. Furthermore, Ca(2+) imaging unveiled lower intracellular Ca(2+) levels in resting Mecp2(-/y) neurons and reduced anoxic Ca(2+) transients with diminished Ca(2+) release from intracellular stores. In conclusion, the enhanced hypoxia susceptibility of Mecp2(-/y) hippocampus is primarily associated with disturbed Ca(2+) homeostasis and diminished Ca(2+) rises during anoxia. This secondarily attenuates the activation of K(Ca) channels and thereby increases the hypoxia susceptibility of Mecp2(-/y) neuronal networks. Since cytosolic Ca(2+) levels also determine neuronal excitability and synaptic plasticity, Ca(2+) homeostasis may constitute a promising target for pharmacotherapy in RTT.
Collapse
Affiliation(s)
- M Kron
- DFG Research Center Molecular Physiology of the Brain, Abteilung Neuro- und Sinnesphysiologie, Georg-August-Universität Göttingen, Humboldtallee 23, D-37073 Göttingen, Germany
| | | |
Collapse
|
41
|
Gee CE, Benquet P, Demont-Guignard S, Wendling F, Gerber U. Energy deprivation transiently enhances rhythmic inhibitory events in the CA3 hippocampal network in vitro. Neuroscience 2010; 168:605-12. [PMID: 20403414 DOI: 10.1016/j.neuroscience.2010.04.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Revised: 03/21/2010] [Accepted: 04/11/2010] [Indexed: 01/01/2023]
Abstract
Oxygen glucose deprivation (OGD) leads to rapid suppression of synaptic transmission. Here we describe an emergence of rhythmic activity at 8 to 20 Hz in the CA3 subfield of hippocampal slice cultures occurring for a few minutes prior to the OGD-induced cessation of evoked responses. These oscillations, dominated by inhibitory events, represent network activity, as they were abolished by tetrodotoxin. They were also completely blocked by the GABAergic antagonist picrotoxin, and strongly reduced by the glutamatergic antagonist NBQX. Applying CPP to block NMDA receptors had no effect and neither did UBP302, an antagonist of GluK1-containing kainate receptors. The gap junction blocker mefloquine disrupted rhythmicity. Simultaneous whole-cell voltage-clamp recordings from neighboring or distant CA3 pyramidal cells revealed strong cross-correlation of the incoming rhythmic activity. Interneurons in the CA3 area received similar correlated activity. Interestingly, oscillations were much less frequently observed in the CA1 area. These data, together with the observation that the recorded activity consists primarily of inhibitory events, suggest that CA3 interneurons are important for generating these oscillations. This transient increase in inhibitory network activity during OGD may represent a mechanism contributing to the lower vulnerability to ischemic insults of the CA3 area as compared to the CA1 area.
Collapse
Affiliation(s)
- C E Gee
- Brain Research Institute, University of Zurich, CH-8057 Zurich, Switzerland
| | | | | | | | | |
Collapse
|
42
|
Chao D, Xia Y. Ionic storm in hypoxic/ischemic stress: can opioid receptors subside it? Prog Neurobiol 2009; 90:439-70. [PMID: 20036308 DOI: 10.1016/j.pneurobio.2009.12.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2009] [Revised: 09/10/2009] [Accepted: 12/17/2009] [Indexed: 12/17/2022]
Abstract
Neurons in the mammalian central nervous system are extremely vulnerable to oxygen deprivation and blood supply insufficiency. Indeed, hypoxic/ischemic stress triggers multiple pathophysiological changes in the brain, forming the basis of hypoxic/ischemic encephalopathy. One of the initial and crucial events induced by hypoxia/ischemia is the disruption of ionic homeostasis characterized by enhanced K(+) efflux and Na(+)-, Ca(2+)- and Cl(-)-influx, which causes neuronal injury or even death. Recent data from our laboratory and those of others have shown that activation of opioid receptors, particularly delta-opioid receptors (DOR), is neuroprotective against hypoxic/ischemic insult. This protective mechanism may be one of the key factors that determine neuronal survival under hypoxic/ischemic condition. An important aspect of the DOR-mediated neuroprotection is its action against hypoxic/ischemic disruption of ionic homeostasis. Specially, DOR signal inhibits Na(+) influx through the membrane and reduces the increase in intracellular Ca(2+), thus decreasing the excessive leakage of intracellular K(+). Such protection is dependent on a PKC-dependent and PKA-independent signaling pathway. Furthermore, our novel exploration shows that DOR attenuates hypoxic/ischemic disruption of ionic homeostasis through the inhibitory regulation of Na(+) channels. In this review, we will first update current information regarding the process and features of hypoxic/ischemic disruption of ionic homeostasis and then discuss the opioid-mediated regulation of ionic homeostasis, especially in hypoxic/ischemic condition, and the underlying mechanisms.
Collapse
Affiliation(s)
- Dongman Chao
- Yale University School of Medicine, Department of Pediatrics, New Haven, CT 06520, USA
| | | |
Collapse
|
43
|
Park YK, Kim SJ. Impaired glial buffering hampers antidromic conduction of CA1 neurons during hypoxia. Brain Res 2009; 1280:90-7. [DOI: 10.1016/j.brainres.2009.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2009] [Revised: 04/29/2009] [Accepted: 05/02/2009] [Indexed: 11/29/2022]
|
44
|
Fischer M, Reuter J, Gerich FJ, Hildebrandt B, Hägele S, Katschinski D, Müller M. Enhanced hypoxia susceptibility in hippocampal slices from a mouse model of rett syndrome. J Neurophysiol 2008; 101:1016-32. [PMID: 19073793 DOI: 10.1152/jn.91124.2008] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Rett syndrome is a neurodevelopmental disorder caused by mutations in the X-chromosomal MECP2 gene encoding for the transcriptional regulator methyl CpG binding protein 2 (MeCP2). Rett patients suffer from episodic respiratory irregularities and reduced arterial oxygen levels. To elucidate whether such intermittent hypoxic episodes induce adaptation/preconditioning of the hypoxia-vulnerable hippocampal network, we analyzed its responses to severe hypoxia in adult Rett mice. The occurrence of hypoxia-induced spreading depression (HSD)--an experimental model for ischemic stroke--was hastened in Mecp2-/y males. The extracellular K+ rise during HSD was attenuated in Mecp2-/y males and the input resistance of CA1 pyramidal neurons decreased less before HSD onset. CA1 pyramidal neurons were smaller and more densely packed, but the cell swelling during HSD was unaffected. The intrinsic optical signal and the propagation of HSD were similar among the different genotypes. Basal synaptic function was intact, but Mecp2-/y males showed reduced paired-pulse facilitation and higher field potential/fiber volley ratios, but no increased seizure susceptibility. Synaptic failure during hypoxia was complete in all genotypes and the final degree of posthypoxic synaptic recovery indistinguishable. Cellular ATP content was normal in Mecp2-/y males, but their hematocrit was increased as was HIF-1alpha expression throughout the brain. This is the first study showing that in Rett syndrome, the susceptibility of telencephalic neuronal networks to hypoxia is increased; the underlying molecular mechanisms apparently involve disturbed K+ channel function. Such an increase in hypoxia susceptibility may potentially contribute to the vulnerability of male Rett patients who are either not viable or severely disabled.
Collapse
Affiliation(s)
- Marc Fischer
- Deutsche Forschungsgemeinschaft Research Center for Molecular Physiology of the Brain, Zentrum Physiologie und Pathophysiologie, Universität Göttingen, Humboldtallee 23, D-37073 Göttingen, Germany
| | | | | | | | | | | | | |
Collapse
|
45
|
Krnjević K. Electrophysiology of cerebral ischemia. Neuropharmacology 2008; 55:319-33. [DOI: 10.1016/j.neuropharm.2008.01.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Revised: 12/31/2007] [Accepted: 01/08/2008] [Indexed: 12/20/2022]
|
46
|
Chao D, Bazzy-Asaad A, Balboni G, Salvadori S, Xia Y. Activation of DOR attenuates anoxic K+ derangement via inhibition of Na+ entry in mouse cortex. Cereb Cortex 2008; 18:2217-27. [PMID: 18203692 DOI: 10.1093/cercor/bhm247] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We have recently found that in the mouse cortex, activation of delta-opioid receptor (DOR) attenuates the disruption of K(+) homeostasis induced by hypoxia or oxygen-glucose deprivation. This novel observation suggests that DOR may protect neurons from hypoxic/ischemic insults via the regulation of K(+) homeostasis because the disruption of K(+) homeostasis plays a critical role in neuronal injury under hypoxic/ischemic stress. The present study was performed to explore the ionic mechanism underlying the DOR-induced neuroprotection. Because anoxia causes Na(+) influx and thus stimulates K(+) leakage, we investigated whether DOR protects the cortex from anoxic K(+) derangement by targeting the Na(+)-based K(+) leakage. By using K(+)-sensitive microelectrodes in mouse cortical slices, we showed that 1) lowering Na(+) concentration and substituting with impermeable N-methyl-D-glucamine caused a concentration-dependent attenuation of anoxic K(+) derangement; 2) lowering Na(+) concentration by substituting with permeable Li(+) tended to potentiate the anoxic K(+) derangement; and 3) the DOR-induced protection against the anoxic K(+) responses was largely abolished by low-Na(+) perfusion irrespective of the substituted cation. We conclude that external Na(+) concentration greatly influences anoxic K(+) derangement and that DOR activation likely attenuates anoxic K(+) derangement induced by the Na(+)-activated mechanisms in the cortex.
Collapse
Affiliation(s)
- Dongman Chao
- Department of Pediatrics, Section of Respiratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | |
Collapse
|
47
|
Chao D, Bazzy-Asaad A, Balboni G, Xia Y. delta-, but not mu-, opioid receptor stabilizes K(+) homeostasis by reducing Ca(2+) influx in the cortex during acute hypoxia. J Cell Physiol 2007; 212:60-7. [PMID: 17373650 DOI: 10.1002/jcp.21000] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Past work has shown that delta-opioid receptor (DOR) activation by [D-Ala(2),D-Leu(5)]-enkephalin (DADLE) attenuated the disruption of K(+) homeostasis induced by hypoxia or oxygen-glucose deprivation (OGD) in the cortex, while naltrindole, a DOR antagonist blocked this effect, suggesting that DOR activity stabilizes K(+) homeostasis in the cortex during hypoxic/ischemic stress. However, several important issues remain unclear regarding this new observation, especially the difference between DOR and other opioid receptors in the stabilization of K(+) homeostasis and the underlying mechanism. In this study, we asked whether DOR is different from micro-opioid receptors (MOR) in stabilizing K(+) homeostasis and which membrane channel(s) is critically involved in the DOR effect. The main findings are that (1) similar to DADLE (10 microM), H-Dmt-Tic-NH-CH (CH(2)--COOH)-Bid (1-10 microM), a more specific and potent DOR agonist significantly attenuated anoxic K(+) derangement in cortical slice; (2) [D-Ala(2), N-Me-Phe(4), glycinol(5)]-enkephalin (DAGO; 10 microM), a MOR agonist, did not produce any appreciable change in anoxic disruption of K(+) homeostasis; (3) absence of Ca(2+) greatly attenuated anoxic K(+) derangement; (4) inhibition of Ca(2+)-activated K(+) (BK) channels with paxilline (10 microM) reduced anoxic K(+) derangement; (5) DADLE (10 microM) could not further reduce anoxic K(+) derangement in the Ca(2+)-free perfused slices or in the presence of paxilline; and (6) glybenclamide (20 microM), a K(ATP) channel blocker, decreased anoxia-induced K(+) derangement, but DADLE (10 microM) could further attenuate anoxic K(+) derangement in the glybenclamide-perfused slices. These data suggest that DOR, but not MOR, activation is protective against anoxic K(+) derangement in the cortex, at least partially via an inhibition of hypoxia-induced increase in Ca(2+) entry-BK channel activity.
Collapse
Affiliation(s)
- Dongman Chao
- Department of Pediatrics, Section of Respiratory Medicine, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | | | |
Collapse
|
48
|
Abstract
All mammals and birds must develop effective strategies to cope with reduced oxygen availability. These animals achieve tolerance to acute and chronic hypoxia by (a) reductions in metabolism, (b) the prevention of cellular injury, and (c) the maintenance of functional integrity. Failure to meet any one of these tasks is detrimental. Birds and mammals accomplish this triple task through a highly coordinated, systems-level reconfiguration involving the partial shutdown of some but not all organs. This reconfiguration is achieved through a similarly complex reconfiguration at the cellular and molecular levels. Reconfiguration at these various levels depends on numerous factors that include the environment, the degree of hypoxic stress, and developmental, behavioral, and ecological conditions. Although common molecular strategies exist, the cellular and molecular changes in any given cell are very diverse. Some cells remain metabolically active, whereas others shut down or rely on anaerobic metabolism. This cellular shutdown is temporarily regulated, and during hypoxic exposure, active cellular networks must continue to control vital functions. The challenge for future research is to explore the cellular mechanisms and conditions that transform an organ or a cellular network into a hypometabolic state, without loss of functional integrity. Much can be learned in this respect from nature: Diving, burrowing, and hibernating animals living in diverse environments are masters of adaptation and can teach us how to deal with hypoxia, an issue of great clinical significance.
Collapse
Affiliation(s)
- Jan-Marino Ramirez
- Department of Organismal Biology & Anatomy, University of Chicago, Chicago, Illinois 60637, USA.
| | | | | |
Collapse
|
49
|
Chao D, Donnelly DF, Feng Y, Bazzy-Asaad A, Xia Y. Cortical delta-opioid receptors potentiate K+ homeostasis during anoxia and oxygen-glucose deprivation. J Cereb Blood Flow Metab 2007; 27:356-68. [PMID: 16773140 DOI: 10.1038/sj.jcbfm.9600352] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Central neurons are extremely vulnerable to hypoxic/ischemic insult, which is a major cause of neurologic morbidity and mortality as a consequence of neuronal dysfunction and death. Our recent work has shown that delta-opioid receptor (DOR) is neuroprotective against hypoxic and excitotoxic stress, although the underlying mechanisms remain unclear. Because hypoxia/ischemia disrupts ionic homeostasis with an increase in extracellular K(+), which plays a role in neuronal death, we asked whether DOR activation preserves K(+) homeostasis during hypoxic/ischemic stress. To test this hypothesis, extracellular recordings with K(+)-sensitive microelectrodes were performed in mouse cortical slices under anoxia or oxygen-glucose deprivation (OGD). The main findings in this study are that (1) DOR activation with [D-Ala(2), D-Leu(5)]-enkephalinamide attenuated the anoxia- and OGD-induced increase in extracellular K(+) and decrease in DC potential in cortical slices; (2) DOR inhibition with naltrindole, a DOR antagonist, completely abolished the DOR-mediated prevention of increase in extracellular K(+) and decrease in DC potential; (3) inhibition of protein kinase A (PKA) with N-(2-[p-bromocinnamylamino]-ethyl)-5-isoquinolinesulfonamide dihydrochloride had no effect on the DOR protection; and (4) inhibition of protein kinase C (PKC) with chelerythrine chloride reduced the DOR protection, whereas the PKC activator (phorbol 12-myristate 13-acetate) mimicked the effect of DOR activation on K(+) homeostasis. These data suggest that activation of DOR protects the cortex against anoxia- or ODG-induced derangement of potassium homeostasis, and this protection occurs via a PKC-dependent and PKA-independent pathway. We conclude that an important aspect of DOR-mediated neuroprotection is its early action against derangement of K(+) homeostasis during anoxia or ischemia.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Cerebral Cortex/physiology
- Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Dose-Response Relationship, Drug
- Enkephalin, Leucine-2-Alanine/pharmacology
- Enzyme Inhibitors/pharmacology
- Extracellular Space/metabolism
- Glucose/deficiency
- Homeostasis/physiology
- Hypoxia, Brain/metabolism
- In Vitro Techniques
- Indicators and Reagents
- Male
- Mice
- Mice, Inbred C57BL
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Potassium/metabolism
- Protein Kinase C/antagonists & inhibitors
- Protein Kinase C/metabolism
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/physiology
- Signal Transduction/drug effects
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Dongman Chao
- Department of Pediatrics, Section of Respiratory Medicine, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | | | | | |
Collapse
|
50
|
Balfour RH, Trapp S. Ionic currents underlying the response of rat dorsal vagal neurones to hypoglycaemia and chemical anoxia. J Physiol 2007; 579:691-702. [PMID: 17218356 PMCID: PMC2151378 DOI: 10.1113/jphysiol.2006.126094] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
A proportion of dorsal vagal neurones (DVN) are glucosensors. These cells respond to brief hypoglycaemia either with a K(ATP) channel-mediated hyperpolarization or with depolarization owing to an as yet unknown mechanism. K(ATP) currents are observed not only during hypoglycaemia, but also in response to mitochondrial inhibition. Here we show that similarly to the observations for K(ATP) currents, both hypoglycaemia and inhibition of mitochondrial function elicited a small inward current that persisted in TTX in DVN of rat brainstem slices. Removal of glucose from the bath solution induced this inward current within 50 +/- 4 s in one subpopulation of DVN and in 279 +/- 36 s in another subpopulation. No such subpopulations were observed for the response to mitochondrial inhibition. Biophysical analysis revealed that mitochondrial inhibition or hypoglycaemia inhibited an openly rectifying K+ conductance in 25% of DVN. In the remaining cells, either an increase in conductance, with a reversal potential between -58 and +10 mV, or a parallel inward shift of the holding current was observed. This current most probably resulted from inhibition of the Na+-K+-ATPase and/or the opening of an ion channel. Recordings with electrodes containing 145 mm instead of 5 mm Cl- failed to shift the reversal potential of the inward current, indicating that a Cl- channel was not involved. In summary, glucosensing and non-glucosensing DVN appear to use common electrical pathways to respond to mitochondrial inhibition and to hypoglycaemia. We suggest that differences in glucose metabolism rather than differences in the complement of ion channels distinguish these two cell types.
Collapse
Affiliation(s)
- Robert H Balfour
- Department of Anaesthetics, Pain Medicine and Intensive Care, Chelsea & Westminster Hospital, Imperial College London, UK
| | | |
Collapse
|