1
|
Weisend JE, Carlson AP, Shuttleworth CW. Spreading Depolarization Induces a Transient Potentiation of Excitatory Synaptic Transmission. Neuroscience 2024; 551:323-332. [PMID: 38821241 PMCID: PMC11246225 DOI: 10.1016/j.neuroscience.2024.05.035] [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: 03/14/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
Spreading depolarization (SD) is a slowly propagating wave of prolonged activation followed by a period of synaptic suppression. Some prior reports have shown potentiation of synaptic transmission after recovery from synaptic suppression and noted similarities with the phenomenon of long-term potentiation (LTP). Since SD is increasingly recognized as participating in diverse neurological disorders, it is of interest to determine whether SD indeed leads to a generalized and sustained long-term strengthening of synaptic connections. We performed a characterization of SD-induced potentiation, and tested whether distinctive features of SD, including adenosine accumulation and swelling, contribute to reports of SD-induced plasticity. Field excitatory postsynaptic potentials (fEPSPs) were recorded in the hippocampal CA1 subregion of murine brain slices, and SD elicited using focal microinjection of KCl. A single SD was sufficient to induce a consistent potentiation of slope and amplitude of fEPSPs. Both AMPA- and NMDA-receptor mediated components were enhanced. Potentiation peaked ∼20 min after SD recovery and was sustained for ∼30 min. However, fEPSP amplitude and slope decayed over an extended 2-hour recording period and was estimated to reach baseline after ∼3 h. Potentiation was saturated after a single SD and adenosine A1 receptor activation did not mask additional potentiation. Induction of LTP with theta-burst stimulation was not altered by prior induction of SD and molecular mediators known to block LTP induction did not block SD-induced potentiation. Together, these results indicate an intermediate duration potentiation that is distinct from hippocampal LTP and may have implications for circuit function for 1-2 h following SD.
Collapse
Affiliation(s)
- Jordan E Weisend
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Andrew P Carlson
- Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - C William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA.
| |
Collapse
|
2
|
Gosalia H, Karsan N, Goadsby PJ. Genetic Mechanisms of Migraine: Insights from Monogenic Migraine Mutations. Int J Mol Sci 2023; 24:12697. [PMID: 37628876 PMCID: PMC10454024 DOI: 10.3390/ijms241612697] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/01/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
Migraine is a disabling neurological disorder burdening patients globally. Through the increasing development of preclinical and clinical experimental migraine models, advancing appreciation of the extended clinical phenotype, and functional neuroimaging studies, we can further our understanding of the neurobiological basis of this highly disabling condition. Despite increasing understanding of the molecular and chemical architecture of migraine mechanisms, many areas require further investigation. Research over the last three decades has suggested that migraine has a strong genetic basis, based on the positive family history in most patients, and this has steered exploration into possibly implicated genes. In recent times, human genome-wide association studies and rodent genetic migraine models have facilitated our understanding, but most migraine seems polygenic, with the monogenic migraine mutations being considerably rarer, so further large-scale studies are required to elucidate fully the genetic underpinnings of migraine and the translation of these to clinical practice. The monogenic migraine mutations cause severe aura phenotypes, amongst other symptoms, and offer valuable insights into the biology of aura and the relationship between migraine and other conditions, such as vascular disease and sleep disorders. This review will provide an outlook of what is known about some monogenic migraine mutations, including familial hemiplegic migraine, familial advanced sleep-phase syndrome, and cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy.
Collapse
Affiliation(s)
- Helin Gosalia
- Headache Group, The Wolfson Sensory, Pain and Rehabilitation Centre, NIHR King’s Clinical Research Facility, & SLaM Biomedical Research Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9PJ, UK; (H.G.); (N.K.)
| | - Nazia Karsan
- Headache Group, The Wolfson Sensory, Pain and Rehabilitation Centre, NIHR King’s Clinical Research Facility, & SLaM Biomedical Research Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9PJ, UK; (H.G.); (N.K.)
| | - Peter J. Goadsby
- Headache Group, The Wolfson Sensory, Pain and Rehabilitation Centre, NIHR King’s Clinical Research Facility, & SLaM Biomedical Research Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9PJ, UK; (H.G.); (N.K.)
- Department of Neurology, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Fomitcheva IV, Sword J, Shi Y, Kirov SA. Plasticity of perisynaptic astroglia during ischemia-induced spreading depolarization. Cereb Cortex 2023; 33:5469-5483. [PMID: 36368909 PMCID: PMC10152098 DOI: 10.1093/cercor/bhac434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 11/13/2022] Open
Abstract
High astroglial capacity for glutamate and potassium clearance aids in recovering spreading depolarization (SD)-evoked disturbance of ion homeostasis during stroke. Since perisynaptic astroglia cannot be imaged with diffraction-limited light microscopy, nothing is known about the impact of SD on the ultrastructure of a tripartite synapse. We used serial section electron microscopy to assess astroglial synaptic coverage in the sensorimotor cortex of urethane-anesthetized male and female mice during and after SD evoked by transient bilateral common carotid artery occlusion. At the subcellular level, astroglial mitochondria were remarkably resilient to SD compared to dendritic mitochondria that were fragmented by SD. Overall, 482 synapses in `Sham' during `SD' and `Recovery' groups were randomly selected and analyzed in 3D. Perisynaptic astroglia was present at the axon-spine interface (ASI) during SD and after recovery. Astrocytic processes were more likely found at large synapses on mushroom spines after recovery, while the length of the ASI perimeter surrounded by astroglia has also significantly increased at large synapses. These findings suggest that as larger synapses have a bigger capacity for neurotransmitter release during SD, they attract astroglial processes to their perimeter during recovery, limiting extrasynaptic glutamate escape and further enhancing the astrocytic ability to protect synapses in stroke.
Collapse
Affiliation(s)
- Ioulia V Fomitcheva
- Department of Neurosurgery, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
| | - Jeremy Sword
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
| | - Yang Shi
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
- Division of Biostatistics and Data Science, Department of Population Health Sciences, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
| | - Sergei A Kirov
- Department of Neurosurgery, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
| |
Collapse
|
4
|
Kalatharan V, Al-Karagholi MAM. Targeting Peripheral N-Methyl-D-Aspartate Receptor (NMDAR): A Novel Strategy for the Treatment of Migraine. J Clin Med 2023; 12:jcm12062156. [PMID: 36983158 PMCID: PMC10055974 DOI: 10.3390/jcm12062156] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/23/2023] [Accepted: 03/02/2023] [Indexed: 03/12/2023] Open
Abstract
Backgrounds: Several acute and preventive medications were developed for the treatment of migraine. Yet, a significant proportion of patients reports an inadequate response and a lack of tolerability, emphasizing the need for new options. Glutamate is the most important excitatory neurotransmitter in the brain, and glutamate receptors including N-Methyl-D-Aspartate Receptor (NMDAR) are expressed at several levels of the trigeminovascular system, which is the anatomical and physiological substrate of migraine pain. Objective: To review preclinical and clinical studies investigating the role of the NMDAR in migraine pathophysiology. Methods: No protocol was registered for this study. References for the present review were identified from a narrative search of the PubMed database. Search terms such as glutamate, migraine, N-Methyl-D-Aspartate Receptor, and NMDAR were used. No restrictions were made in terms of the language and date of publication. Results: In animal models, administration of monosodium glutamate (MSG) activated and sensitized trigeminovascular neurons. In healthy human participants, consumption of MSG caused headaches, craniofacial sensitivity, and nausea. In in vivo models and through immunolabeling, NMDAR subunits NR1, NR2A, and NR2B were expressed in trigeminal ganglion neurons. In humans, NMDAR antagonists such as ketamine and memantine caused a significant reduction in pain intensity and monthly headache frequency. Conclusions: Accumulative evidence indicates that NMDAR is a promising new target for the treatment of migraine. Selective NMDAR antagonists without central effects are needed to investigate their therapeutic benefit in the treatment of migraine.
Collapse
|
5
|
Curry RN, Aiba I, Meyer J, Lozzi B, Ko Y, McDonald MF, Rosenbaum A, Cervantes A, Huang-Hobbs E, Cocito C, Greenfield JP, Jalali A, Gavvala J, Mohila C, Serin Harmanci A, Noebels J, Rao G, Deneen B. Glioma epileptiform activity and progression are driven by IGSF3-mediated potassium dysregulation. Neuron 2023; 111:682-695.e9. [PMID: 36787748 PMCID: PMC9991983 DOI: 10.1016/j.neuron.2023.01.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/11/2022] [Accepted: 01/17/2023] [Indexed: 02/15/2023]
Abstract
Seizures are a frequent pathophysiological feature of malignant glioma. Recent studies implicate peritumoral synaptic dysregulation as a driver of brain hyperactivity and tumor progression; however, the molecular mechanisms that govern these phenomena remain elusive. Using scRNA-seq and intraoperative patient ECoG recordings, we show that tumors from seizure patients are enriched for gene signatures regulating synapse formation. Employing a human-to-mouse in vivo functionalization pipeline to screen these genes, we identify IGSF3 as a mediator of glioma progression and dysregulated neural circuitry that manifests as spreading depolarization (SD). Mechanistically, we discover that IGSF3 interacts with Kir4.1 to suppress potassium buffering and found that seizure patients exhibit reduced expression of potassium handlers in proliferating tumor cells. In vivo imaging reveals that dysregulated synaptic activity emanates from the tumor-neuron interface, which we confirm in patients. Our studies reveal that tumor progression and seizures are enabled by ion dyshomeostasis and identify SD as a driver of disease.
Collapse
Affiliation(s)
- Rachel Naomi Curry
- The Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Isamu Aiba
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jochen Meyer
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brittney Lozzi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Program in Genetics and Genomics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yeunjung Ko
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Program in Immunology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Malcolm Ford McDonald
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Program in Development, Disease, Models, and Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anna Rosenbaum
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexis Cervantes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Emmet Huang-Hobbs
- The Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Carolina Cocito
- Department of Pediatric Neurological Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Ali Jalali
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jay Gavvala
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Carrie Mohila
- Department of Pathology, Texas Children's Hospital, Houston, TX 77030, USA
| | - Akdes Serin Harmanci
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jeffrey Noebels
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ganesh Rao
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Benjamin Deneen
- The Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Program in Development, Disease, Models, and Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
6
|
Abstract
Headache disorders can produce recurrent, incapacitating pain. Migraine and cluster headache are notable for their ability to produce significant disability. The anatomy and physiology of headache disorders is fundamental to evolving treatment approaches and research priorities. Key concepts in headache mechanisms include activation and sensitization of trigeminovascular, brainstem, thalamic, and hypothalamic neurons; modulation of cortical brain regions; and activation of descending pain circuits. This review will examine the relevant anatomy of the trigeminal, brainstem, subcortical, and cortical brain regions and concepts related to the pathophysiology of migraine and cluster headache disorders.
Collapse
Affiliation(s)
- Andrea M Harriott
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yulia Orlova
- Department of Neurology, University of Florida, Gainesville, Florida
| |
Collapse
|
7
|
Extracellular Alterations in pH and K+ Modify the Murine Brain Endothelial Cell Total and Phospho-Proteome. Pharmaceutics 2022; 14:pharmaceutics14071469. [PMID: 35890365 PMCID: PMC9324801 DOI: 10.3390/pharmaceutics14071469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/31/2022] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
Pathologies of the blood-brain barrier (BBB) have been linked to a multitude of central nervous system (CNS) disorders whose pathology is poorly understood. Cortical spreading depression (CSD) has long been postulated to be involved in the underlying mechanisms of these disease states, yet a complete understanding remains elusive. This study seeks to utilize an in vitro model of the blood-brain barrier (BBB) with brain endothelial cell (b.End3) murine endothelioma cells to investigate the role of CSD in BBB pathology by characterizing effects of the release of major pronociceptive substances into the extracellular space of the CNS. The application of trans-endothelial electrical resistance (TEER) screening, transcellular uptake, and immunoreactive methods were used in concert with global proteome and phospho-proteomic approaches to assess the effect of modeled CSD events on the modeled BBB in vitro. The findings demonstrate relocalization and functional alteration to proteins associated with the actin cytoskeleton and endothelial tight junctions. Additionally, unique pathologic mechanisms induced by individual substances released during CSD were found to have unique phosphorylation signatures in phospho-proteome analysis, identifying Zona Occludins 1 (ZO-1) as a possible pathologic "checkpoint" of the BBB. By utilizing these phosphorylation signatures, possible novel diagnostic methods may be developed for CSD and warrants further investigation.
Collapse
|
8
|
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
|
9
|
Deep Brain Stimulation of the Medial Septal Area Can Modulate Gene Expression in the Hippocampus of Rats under Urethane Anesthesia. Int J Mol Sci 2022; 23:ijms23116034. [PMID: 35682713 PMCID: PMC9181580 DOI: 10.3390/ijms23116034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 11/17/2022] Open
Abstract
We studied the effects of stimulation of the medial septal area on the gene expression in the dorsal and ventral hippocampus. Rats under urethane anesthesia were implanted with a recording electrode in the right hippocampus and stimulating electrode in the dorsal medial septum (dMS) or medial septal nucleus (MSN). After one-hour-long deep brain stimulation, we collected ipsi- and contralateral dorsal and ventral hippocampi. Quantitative PCR showed that deep brain stimulation did not cause any changes in the intact contralateral dorsal and ventral hippocampi. A comparison of ipsi- and contralateral hippocampi in the control unstimulated animals showed that electrode implantation in the ipsilateral dorsal hippocampus led to a dramatic increase in the expression of immediate early genes (c-fos, arc, egr1, npas4), neurotrophins (ngf, bdnf) and inflammatory cytokines (il1b and tnf, but not il6) not only in the area close to implantation site but also in the ventral hippocampus. Moreover, the stimulation of MSN but not dMS further increased the expression of c-fos, egr1, npas4, bdnf, and tnf in the ipsilateral ventral but not dorsal hippocampus. Our data suggest that the activation of medial septal nucleus can change the gene expression in ventral hippocampal cells after their priming by other stimuli.
Collapse
|
10
|
Ellingsrud AJ, Dukefoss DB, Enger R, Halnes G, Pettersen K, Rognes ME. Validating a Computational Framework for Ionic Electrodiffusion with Cortical Spreading Depression as a Case Study. eNeuro 2022; 9:ENEURO.0408-21.2022. [PMID: 35365505 PMCID: PMC9045477 DOI: 10.1523/eneuro.0408-21.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/21/2022] [Accepted: 03/12/2022] [Indexed: 11/21/2022] Open
Abstract
Cortical spreading depression (CSD) is a wave of pronounced depolarization of brain tissue accompanied by substantial shifts in ionic concentrations and cellular swelling. Here, we validate a computational framework for modeling electrical potentials, ionic movement, and cellular swelling in brain tissue during CSD. We consider different model variations representing wild-type (WT) or knock-out/knock-down mice and systematically compare the numerical results with reports from a selection of experimental studies. We find that the data for several CSD hallmarks obtained computationally, including wave propagation speed, direct current shift duration, peak in extracellular K+ concentration as well as a pronounced shrinkage of extracellular space (ECS) are well in line with what has previously been observed experimentally. Further, we assess how key model parameters including cellular diffusivity, structural ratios, membrane water and/or K+ permeabilities affect the set of CSD characteristics.
Collapse
Affiliation(s)
- Ada J Ellingsrud
- Department for Numerical Analysis and Scientific Computing, Simula Research Laboratory, Oslo 0164, Norway
| | - Didrik B Dukefoss
- Letten Centre, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo 0317, Norway
| | - Rune Enger
- Letten Centre, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo 0317, Norway
| | - Geir Halnes
- CINPLA, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo 0316, Norway
- Institute of Physics, Faculty of Science and Technology, Norwegian University of Life Sciences, Ås 1432, Norway
| | - Klas Pettersen
- NORA, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo 0316, Norway
| | - Marie E Rognes
- Department for Numerical Analysis and Scientific Computing, Simula Research Laboratory, Oslo 0164, Norway
- Department of Mathematics, Faculty of Mathematics and Natural Sciences, University of Bergen, Bergen 5020, Norway
| |
Collapse
|
11
|
Foreman B, Lee H, Okonkwo DO, Strong AJ, Pahl C, Shutter LA, Dreier JP, Ngwenya LB, Hartings JA. The Relationship Between Seizures and Spreading Depolarizations in Patients with Severe Traumatic Brain Injury. Neurocrit Care 2022; 37:31-48. [PMID: 35174446 DOI: 10.1007/s12028-022-01441-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 01/04/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Both seizures and spreading depolarizations (SDs) are commonly detected using electrocorticography (ECoG) after severe traumatic brain injury (TBI). A close relationship between seizures and SDs has been described, but the implications of detecting either or both remain unclear. We sought to characterize the relationship between these two phenomena and their clinical significance. METHODS We performed a post hoc analysis of a prospective observational clinical study of patients with severe TBI requiring neurosurgery at five academic neurotrauma centers. A subdural electrode array was placed intraoperatively and ECoG was recorded during intensive care. SDs, seizures, and high-frequency background characteristics were quantified offline using published standards and terminology. The primary outcome was the Glasgow Outcome Scale-Extended score at 6 months post injury. RESULTS There were 138 patients with valid ECoG recordings; the mean age was 47 ± 19 years, and 104 (75%) were men. Overall, 2,219 ECoG-detected seizures occurred in 38 of 138 (28%) patients in a bimodal pattern, with peak incidences at 1.7-1.8 days and 3.8-4.0 days post injury. Seizures detected on scalp electroencephalography (EEG) were diagnosed by standard clinical care in only 18 of 138 (13%). Of 15 patients with ECoG-detected seizures and contemporaneous scalp EEG, seven (47%) had no definite scalp EEG correlate. ECoG-detected seizures were significantly associated with the severity and number of SDs, which occurred in 83 of 138 (60%) of patients. Temporal interactions were observed in 17 of 24 (70.8%) patients with both ECoG-detected seizures and SDs. After controlling for known prognostic covariates and the presence of SDs, seizures detected on either ECoG or scalp EEG did not have an independent association with 6-month functional outcome but portended worse outcome among those with clustered or isoelectric SDs. CONCLUSIONS In patients with severe TBI requiring neurosurgery, seizures were half as common as SDs. Seizures would have gone undetected without ECoG monitoring in 20% of patients. Although seizures alone did not influence 6-month functional outcomes in this cohort, they were independently associated with electrographic worsening and a lack of motor improvement following surgery. Temporal interactions between ECoG-detected seizures and SDs were common and held prognostic implications. Together, seizures and SDs may occur along a dynamic continuum of factors critical to the development of secondary brain injury. ECoG provides information integral to the clinical management of patients with TBI.
Collapse
Affiliation(s)
- Brandon Foreman
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, USA. .,Collaborative for Research on Acute Neurological Injuries, University of Cincinnati, Cincinnati, OH, USA.
| | - Hyunjo Lee
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, USA.,Collaborative for Research on Acute Neurological Injuries, University of Cincinnati, Cincinnati, OH, USA
| | - David O Okonkwo
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anthony J Strong
- Department of Basic and Clinical Neuroscience, King's College London, London, UK
| | - Clemens Pahl
- Department of Intensive Care Medicine, King's College Hospital, London, UK
| | - Lori A Shutter
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Critical Care Medicine and Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - 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
| | - Laura B Ngwenya
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, USA.,Collaborative for Research on Acute Neurological Injuries, University of Cincinnati, Cincinnati, OH, USA.,Department of Neurosurgery, University of Cincinnati, Cincinnati, OH, USA
| | - Jed A Hartings
- Collaborative for Research on Acute Neurological Injuries, University of Cincinnati, Cincinnati, OH, USA.,Department of Neurosurgery, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
12
|
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: 40] [Impact Index Per Article: 13.3] [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
|
13
|
Melo-Carrillo A, Strassman AM, Schain AJ, Broide RS, Cai BB, Rhéaume C, Brideau-Andersen AD, Ashina S, Flores-Montanez Y, Brin MF, Burstein R. OnabotulinumtoxinA affects cortical recovery period but not occurrence or propagation of cortical spreading depression in rats with compromised blood-brain barrier. Pain 2021; 162:2418-2427. [PMID: 34448754 PMCID: PMC8374711 DOI: 10.1097/j.pain.0000000000002230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/11/2021] [Accepted: 02/01/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT OnabotulinumtoxinA (BoNT-A) is an Food and Drug Administration-approved, peripherally acting preventive migraine drug capable of inhibiting meningeal nociceptors. Expanding our view of how else this neurotoxin attenuates the activation of the meningeal nociceptors, we reasoned that if the stimulus that triggers the activation of the nociceptor is lessened, the magnitude and/or duration of the nociceptors' activation could diminish as well. In the current study, we further examine this possibility using electrocorticogram recording techniques, immunohistochemistry, and 2-photon microscopy. We report (1) that scalp (head) but not lumbar (back) injections of BoNT-A shorten the period of profound depression of spontaneous cortical activity that follows a pinprick-induced cortical spreading depression (CSD); (2) that neither scalp nor lumbar injections prevent the induction, occurrence, propagation, or spreading velocity of a single wave of CSD; (3) that cleaved SNAP25-one of the most convincing tools to determine the anatomical targeting of BoNT-A treatment-could easily be detected in pericranial muscles at the injection sites and in nerve fibers of the intracranial dura, but not within any cortical area affected by the CSD; (4) that the absence of cleaved SNAP25 within the cortex and pia is unrelated to whether the blood-brain barrier is intact or compromised; and (5) that BoNT-A does not alter vascular responses to CSD. To the best of our knowledge, this is the first report of peripherally applied BoNT-A's ability to alter a neuronal function along a central nervous system pathway involved in the pathophysiology of migraine.
Collapse
Affiliation(s)
- Agustin Melo-Carrillo
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston MA, United States
- Department of Anesthesia, Harvard Medical School, Boston MA, United States
| | - Andrew M. Strassman
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston MA, United States
- Department of Anesthesia, Harvard Medical School, Boston MA, United States
| | - Aaron J. Schain
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston MA, United States
- Department of Anesthesia, Harvard Medical School, Boston MA, United States
| | - Ron S. Broide
- Allergan, an AbbVie Company, Irvine, CA, United States
| | - Brian B. Cai
- Allergan, an AbbVie Company, Irvine, CA, United States
| | | | | | - Sait Ashina
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston MA, United States
- Department of Anesthesia, Harvard Medical School, Boston MA, United States
| | - Yadira Flores-Montanez
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston MA, United States
- Department of Anesthesia, Harvard Medical School, Boston MA, United States
| | - Mitchell F. Brin
- Allergan, an AbbVie Company, Irvine, CA, United States
- University of California, Irvine, United States
| | - Rami Burstein
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston MA, United States
- Department of Anesthesia, Harvard Medical School, Boston MA, United States
| |
Collapse
|
14
|
Passlick S, Rose CR, Petzold GC, Henneberger C. Disruption of Glutamate Transport and Homeostasis by Acute Metabolic Stress. Front Cell Neurosci 2021; 15:637784. [PMID: 33603647 PMCID: PMC7884476 DOI: 10.3389/fncel.2021.637784] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/11/2021] [Indexed: 01/02/2023] Open
Abstract
High-affinity, Na+-dependent glutamate transporters are the primary means by which synaptically released glutamate is removed from the extracellular space. They restrict the spread of glutamate from the synaptic cleft into the perisynaptic space and reduce its spillover to neighboring synapses. Thereby, glutamate uptake increases the spatial precision of synaptic communication. Its dysfunction and the entailing rise of the extracellular glutamate concentration accompanied by an increased spread of glutamate result in a loss of precision and in enhanced excitation, which can eventually lead to neuronal death via excitotoxicity. Efficient glutamate uptake depends on a negative resting membrane potential as well as on the transmembrane gradients of the co-transported ions (Na+, K+, and H+) and thus on the proper functioning of the Na+/K+-ATPase. Consequently, numerous studies have documented the impact of an energy shortage, as occurring for instance during an ischemic stroke, on glutamate clearance and homeostasis. The observations range from rapid changes in the transport activity to altered expression of glutamate transporters. Notably, while astrocytes account for the majority of glutamate uptake under physiological conditions, they may also become a source of extracellular glutamate elevation during metabolic stress. However, the mechanisms of the latter phenomenon are still under debate. Here, we review the recent literature addressing changes of glutamate uptake and homeostasis triggered by acute metabolic stress, i.e., on a timescale of seconds to minutes.
Collapse
Affiliation(s)
- Stefan Passlick
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Division of Vascular Neurology, University Hospital Bonn, Bonn, Germany
| | - Christian Henneberger
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
15
|
MartInez-Coria H, Arrieta-Cruz I, Cruz ME, López-Valdés HE. Physiopathology of ischemic stroke and its modulation using memantine: evidence from preclinical stroke. Neural Regen Res 2021; 16:433-439. [PMID: 32985462 PMCID: PMC7996012 DOI: 10.4103/1673-5374.293129] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Ischemic stroke is the most common type of cerebrovascular disease and is caused by an interruption of blood flow in the brain. In this disease, two different damage areas are identifying: the lesion core, in which cells quickly die; and the penumbra (surrounding the lesion core), in which cells are functionally weakened but may recover and restore their functions. The currently approved treatments for ischemic stroke are the recombinant tissue plasminogen activator and endovascular thrombectomy, but they have a short therapeutic window (4.5 and 6 hours after stroke onset, respectively) and a low percentage of stroke patients actually receive these treatments. Memantine is an approved drug for the treatment of Alzheimer’s disease. Memantine is a noncompetitive, low affinity and use-dependent antagonist of N-methyl-D-aspartate glutamate receptor. Memantine has several advantages over developing a new drug to treat focal ischemic stroke, but the most important is that it has sufficient safe probes in preclinical models and humans, and if the preclinical studies provide more evidence about pharmacological actions in tissue protection and repair, this could help to increase the number of clinical trials. The present review summarizes the physiopathology of isquemic stroke and the pharmacological actions in neuroprotection and neuroplasticity of memantine in the post stroke stage of preclinical stroke models, to illustrate their potential to improve functional recovery in human patients.
Collapse
Affiliation(s)
- Hilda MartInez-Coria
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM); Laboratorio Experimental de Enfermedades Neurodegenerativas, Facultad de Medicina, UNAM/Instituto Nacional de Neurología y Neurocirugía, Ciudad de México, México
| | - Isabel Arrieta-Cruz
- Departamento de Investigación Básica, Instituto Nacional de Geriatría, Ciudad de México, México
| | - María-Esther Cruz
- Unidad de Investigación en Biología de la Reproducción, Laboratorio de Neuroendocrinología, Facultad de Estudios Superiores Zaragoza, UNAM, Ciudad de México, México
| | - Héctor E López-Valdés
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM); Unidad Periférica de Neurociencia, Facultad de Medicina, UNAM/Instituto Nacional de Neurología y Neurocirugía, Ciudad de México, México
| |
Collapse
|
16
|
Herreras O, Makarova J. Mechanisms of the negative potential associated with Leão's spreading depolarization: A history of brain electrogenesis. J Cereb Blood Flow Metab 2020; 40:1934-1952. [PMID: 32580670 PMCID: PMC7786845 DOI: 10.1177/0271678x20935998] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/04/2020] [Accepted: 05/18/2020] [Indexed: 12/16/2022]
Abstract
Spreading depolarization (SD) is a self-propagated wave that provokes transient disorder of numerous cell and tissue functions, and that may kill neurons in metabolically compromised tissue. We examined the mechanisms underlying the main hallmark of SD, a giant extracellular potential (ΔVo) for which multiple electromotive forces have been proposed. The end-point is that neurons and not glia, dendritic channels and not spatial currents, and increased sodium conductance rather than potassium gradients, appear to be the main actors in the generation of the negative ΔVo. Neuronal currents are established by two mechanisms, a voltage independent dendritic current, and the differential polarization along the neuron membranes. Notably, despite of a marked drop of ion gradients, these evolve significantly during SD, and yet the membrane potential remains clamped at zero no matter how much inward current is present. There may be substantial inward current or none in function of the evolving portion of the neuron dendrites with SD-activated channels. We propose that the ΔVo promotes swelling-induced dendritic damage. Understanding SD electrogenesis requires all elements relevant for membrane potential, action currents, field potentials and volume conduction to be jointly considered, and it has already encouraged the search for new targets to limit SD-related pathology.
Collapse
Affiliation(s)
- Oscar Herreras
- Department of Translational Neuroscience, Cajal Institute – CSIC, Madrid, Spain
| | - Julia Makarova
- Department of Translational Neuroscience, Cajal Institute – CSIC, Madrid, Spain
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| |
Collapse
|
17
|
Iftikhar W, Cheema FF, Khanal S, Khan QU. Migrainous Infarction and Cortical Spreading Depression. Discoveries (Craiova) 2020; 8:e112. [PMID: 33083518 PMCID: PMC7553730 DOI: 10.15190/d.2020.9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Migraine is a very common disorder of the nervous system. It shares similar physiological processes with stroke. Migrainous infarction is a rare complication of migraine with aura. The neuro-logical symptoms of migraine aura correspond to the cortical spreading depression and this depression can lead to a migrainous infarction. It is pertinent to state that the investigation and detection of the cortical depression might have a great clinical significance. Blood vessels in the cranium play an important role in the pathophysiology of migraine. In the case of injured states of brain, the cortical spreading depression causes extreme vasoconstriction rather than vasodilation. The endothelial damage caused by the cortical spreading depression can result in hypercoagulability, leading to an increased risk of stroke. There are many genetic disorders in which migraine and stroke are the major symptoms and an insight into these disorders can help us in the understanding of complex mechanisms of migrainous infarction. It is pertinent to state that some derangements in the vascular function accompany migraine which may also serve as targets for research and treatment. This article will describe the hemodynamic and genetic relationship between migraine induced stroke and how it relates to the cortical spreading depression.
Collapse
Affiliation(s)
- Waleed Iftikhar
- CMH Lahore Medical College and Institute of Dentistry (NUMS), Lahore, Pakistan
| | | | - Sneha Khanal
- Jahurul Islam Medical College and Hospital, Bajitpur, Kishoregonj, Bangladesh
| | - Qudsia Umaira Khan
- CMH Lahore Medical College and Institute of Dentistry (NUMS), Lahore, Pakistan
| |
Collapse
|
18
|
Melo-Carrillo A, Schain AJ, Stratton J, Strassman AM, Burstein R. Fremanezumab and its isotype slow propagation rate and shorten cortical recovery period but do not prevent occurrence of cortical spreading depression in rats with compromised blood-brain barrier. Pain 2020; 161:1037-1043. [PMID: 31895266 PMCID: PMC7166155 DOI: 10.1097/j.pain.0000000000001791] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Most centrally acting migraine preventive drugs suppress frequency and velocity of cortical spreading depression (CSD). The purpose of the current study was to determine how the new class of peripherally acting migraine preventive drug (ie, the anti-CGRP-mAbs) affect CSD-an established animal model of migraine aura, which affects about 1/3 of people with migraine-when allowed to cross the blood-brain barrier (BBB). Using standard electrocorticogram recording techniques and rats in which the BBB was intentionally compromised, we found that when the BBB was opened, the anti-CGRP-mAb fremanezumab did not prevent the induction, occurrence, or propagation of a single wave of CSD induced by a pinprick, but that both fremanezumab and its isotype were capable of slowing down the propagation velocity of CSD and shortening the period of profound depression of spontaneous cortical activity that followed the spreading depolarization. Fremanezumab's inability to completely block the occurrence of CSD in animals in which the BBB was compromised suggests that calcitonin gene-related peptide (CGRP) may not be involved in the initiation of CSD, at least not to the extent that it can prevent its occurrence. Similarly, we cannot conclude that CGRP is involved in the propagation velocity or the neuronal silencing period (also called cortical recovery period) that follows the CSD because similar effects were observed when the isotype was used. These finding call for caution with interpretations of studies that claim to show direct central nervous system effects of CGRP-mAbs.
Collapse
Affiliation(s)
- Agustin Melo-Carrillo
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston MA 02115
- Harvard Medical School, Boston, MA 02215, USA
| | - Aaron J. Schain
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston MA 02115
- Harvard Medical School, Boston, MA 02215, USA
| | | | - Andrew M. Strassman
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston MA 02115
- Harvard Medical School, Boston, MA 02215, USA
| | - Rami Burstein
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston MA 02115
- Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
19
|
Major S, Huo S, Lemale CL, Siebert E, Milakara D, Woitzik J, Gertz K, Dreier JP. Direct electrophysiological evidence that spreading depolarization-induced spreading depression is the pathophysiological correlate of the migraine aura and a review of the spreading depolarization continuum of acute neuronal mass injury. GeroScience 2020; 42:57-80. [PMID: 31820363 PMCID: PMC7031471 DOI: 10.1007/s11357-019-00142-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023] Open
Abstract
Spreading depolarization is observed as a large negative shift of the direct current potential, swelling of neuronal somas, and dendritic beading in the brain's gray matter and represents a state of a potentially reversible mass injury. Its hallmark is the abrupt, massive ion translocation between intraneuronal and extracellular compartment that causes water uptake (= cytotoxic edema) and massive glutamate release. Dependent on the tissue's energy status, spreading depolarization can co-occur with different depression or silencing patterns of spontaneous activity. In adequately supplied tissue, spreading depolarization induces spreading depression of activity. In severely ischemic tissue, nonspreading depression of activity precedes spreading depolarization. The depression pattern determines the neurological deficit which is either spreading such as in migraine aura or migraine stroke or nonspreading such as in transient ischemic attack or typical stroke. Although a clinical distinction between spreading and nonspreading focal neurological deficits is useful because they are associated with different probabilities of permanent damage, it is important to note that spreading depolarization, the neuronal injury potential, occurs in all of these conditions. Here, we first review the scientific basis of the continuum of spreading depolarizations. Second, we highlight the transition zone of the continuum from reversibility to irreversibility using clinical cases of aneurysmal subarachnoid hemorrhage and cerebral amyloid angiopathy. These illustrate how modern neuroimaging and neuromonitoring technologies increasingly bridge the gap between basic sciences and clinic. For example, we provide direct electrophysiological evidence for the first time that spreading depolarization-induced spreading depression is the pathophysiological correlate of the migraine aura.
Collapse
Affiliation(s)
- Sebastian Major
- Center for Stroke Research, Campus Charité Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Shufan Huo
- Center for Stroke Research, Campus Charité Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Coline L Lemale
- Center for Stroke Research, Campus Charité Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Eberhard Siebert
- Department of Neuroradiology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Denny Milakara
- Solution Centre for Image Guided Local Therapies (STIMULATE), Otto-von-Guericke-University, Magdeburg, Germany
| | - Johannes Woitzik
- Evangelisches Krankenhaus Oldenburg, University of Oldenburg, Oldenburg, Germany
| | - Karen Gertz
- Center for Stroke Research, Campus Charité Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jens P Dreier
- Center for Stroke Research, Campus Charité Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- Department of Neurology, Charité-Universitätsmedizin Berlin, 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
|
20
|
Taş YÇ, Solaroğlu İ, Gürsoy-Özdemir Y. Spreading Depolarization Waves in Neurological Diseases: A Short Review about its Pathophysiology and Clinical Relevance. Curr Neuropharmacol 2019; 17:151-164. [PMID: 28925885 PMCID: PMC6343201 DOI: 10.2174/1570159x15666170915160707] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/03/2017] [Accepted: 09/09/2017] [Indexed: 02/05/2023] Open
Abstract
Lesion growth following acutely injured brain tissue after stroke, subarachnoid hemorrhage and traumatic brain injury is an important issue and a new target area for promising therapeutic interventions. Spreading depolarization or peri-lesion depolarization waves were demonstrated as one of the significant contributors of continued lesion growth. In this short review, we discuss the pathophysiology for SD forming events and try to list findings detected in neurological disorders like migraine, stroke, subarachnoid hemorrhage and traumatic brain injury in both human as well as experimental studies. Pharmacological and non-pharmacological treatment strategies are highlighted and future directions and research limitations are discussed.
Collapse
Affiliation(s)
| | | | - Yasemin Gürsoy-Özdemir
- Address correspondence to these authors at the Department of Neurosurgery, School of Medicine, Koç University, İstanbul, Turkey; Tel: +90 850 250 8250; E-mails: ,
| |
Collapse
|
21
|
Hobbs CN, Johnson JA, Verber MD, Mark Wightman R. An implantable multimodal sensor for oxygen, neurotransmitters, and electrophysiology during spreading depolarization in the deep brain. Analyst 2018; 142:2912-2920. [PMID: 28715004 DOI: 10.1039/c7an00508c] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Brain tissue injury is often accompanied by spreading depolarization (SD) events, marked by widespread cellular depolarization and cessation of neuronal firing. SD recruits viable tissue into the lesion, making it a focus for intervention. During SD, drastic fluctuations occur in ion gradients, extracellular neurotransmitter concentrations, cellular metabolism, and cerebral blood flow. Measuring SD requires a multimodal approach to capture the array of changes. However, the use of multiple sensors can inflict tissue damage. Here, we use carbon-fiber microelectrodes to characterize several aspects of SD with a single, minimally invasive sensor in the deep brain region of the nucleus accumbens. Fast-scan cyclic voltammetry detects large changes in oxygen, which reflect the balance between cerebral blood flow and energy consumption, and also supraphysiological release of electroactive neurotransmitters (i.e., dopamine). We verify waves of SD with concurrent single-unit or DC potential electrophysiological recordings. The single-unit recordings reveal bursts of action potentials followed by inactivity. The DC potentials exhibit a slow negative voltage shift in the extracellular space indicative of wide-spread cellular depolarization. Here, we characterize the multiple modalities of our sensor and demonstrate its utility for improved SD recordings.
Collapse
Affiliation(s)
- Caddy N Hobbs
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | | | | | | |
Collapse
|
22
|
Lückl J, Lemale CL, Kola V, Horst V, Khojasteh U, Oliveira-Ferreira AI, Major S, Winkler MKL, Kang EJ, Schoknecht K, Martus P, Hartings JA, Woitzik J, Dreier JP. The negative ultraslow potential, electrophysiological correlate of infarction in the human cortex. Brain 2018; 141:1734-1752. [PMID: 29668855 PMCID: PMC5972557 DOI: 10.1093/brain/awy102] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 01/20/2018] [Accepted: 02/17/2018] [Indexed: 12/19/2022] Open
Abstract
Spreading depolarizations are characterized by abrupt, near-complete breakdown of the transmembrane ion gradients, neuronal oedema, mitochondrial depolarization, glutamate excitotoxicity and activity loss (depression). Spreading depolarization induces either transient hyperperfusion in normal tissue; or hypoperfusion (inverse coupling = spreading ischaemia) in tissue at risk for progressive injury. The concept of the spreading depolarization continuum is critical since many spreading depolarizations have intermediate characteristics, as opposed to the two extremes of spreading depolarization in either severely ischaemic or normal tissue. In animals, the spreading depolarization extreme in ischaemic tissue is characterized by prolonged depolarization durations, in addition to a slow baseline variation termed the negative ultraslow potential. The negative ultraslow potential is initiated by spreading depolarization and similar to the negative direct current (DC) shift of prolonged spreading depolarization, but specifically refers to a negative potential component during progressive recruitment of neurons into cell death in the wake of spreading depolarization. We here first quantified the spreading depolarization-initiated negative ultraslow potential in the electrocorticographic DC range and the activity depression in the alternate current range after middle cerebral artery occlusion in rats. Relevance of these variables to the injury was supported by significant correlations with the cortical infarct volume and neurological outcome after 72 h of survival. We then identified negative ultraslow potential-containing clusters of spreading depolarizations in 11 patients with aneurysmal subarachnoid haemorrhage. The human platinum/iridium-recorded negative ultraslow potential showed a tent-like shape. Its amplitude of 45.0 (39.0, 69.4) mV [median (first, third quartile)] was 6.6 times larger and its duration of 3.7 (3.3, 5.3) h was 34.9 times longer than the negative DC shift of spreading depolarizations in less compromised tissue. Using Generalized Estimating Equations applied to a logistic regression model, we found that negative ultraslow potential displaying electrodes were significantly more likely to overlie a developing ischaemic lesion (90.0%, 27/30) than those not displaying a negative ultraslow potential (0.0%, 0/20) (P = 0.004). Based on serial neuroimages, the lesions under the electrodes developed within a time window of 72 (56, 134) h. The negative ultraslow potential occurred in this time window in 9/10 patients. It was often preceded by a spreading depolarization cluster with increasingly persistent spreading depressions and progressively prolonged DC shifts and spreading ischaemias. During the negative ultraslow potential, spreading ischaemia lasted for 40.0 (28.0, 76.5) min, cerebral blood flow fell from 57 (53, 65) % to 26 (16, 42) % (n = 4) and tissue partial pressure of oxygen from 12.5 (9.2, 15.2) to 3.3 (2.4, 7.4) mmHg (n = 5). Our data suggest that the negative ultraslow potential is the electrophysiological correlate of infarction in human cerebral cortex and a neuromonitoring-detected medical emergency.awy102media15775596049001.
Collapse
Affiliation(s)
- Janos Lückl
- 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
| | - Coline L Lemale
- 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
| | - Vasilis Kola
- 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
| | - Viktor Horst
- 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
| | - Uldus Khojasteh
- 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
| | - 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
| | - Maren K L Winkler
- 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
| | - 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
| | - Karl Schoknecht
- 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
| | - Peter Martus
- Institute for Clinical Epidemiology and Applied Biostatistics, University of Tübingen, Tübingen, Germany
| | - Jed A Hartings
- UC Gardner Neuroscience Institute, University of Cincinnati (UC) College of Medicine, Cincinnati, OH, USA
- Department of Neurosurgery, University of Cincinnati (UC) College of Medicine, Cincinnati, OH, USA
| | - Johannes Woitzik
- Department of Neurosurgery, 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
|
23
|
Mayor D, Tymianski M. Neurotransmitters in the mediation of cerebral ischemic injury. Neuropharmacology 2017; 134:178-188. [PMID: 29203179 DOI: 10.1016/j.neuropharm.2017.11.050] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 01/09/2023]
Abstract
Under physiological conditions, neurotransmitters shape neuronal networks and control several cellular and synaptic functions. In the mammalian central nervous system (CNS), excitatory and inhibitory neurotransmission are mediated in large part by glutamate and gamma-aminobutyric acid (GABA), which are excitatory and inhibitory neurotransmitters, respectively. Glutamate and GABA also play crucial roles in neurological disorders such as cerebral ischemia. Glutamate in particular causes excitotoxicity, known as one of the hallmark mechanisms in the pathophysiology of cerebral ischemic injury for more than thirty years. Excitotoxicity occurs due to excessive glutamate release leading to overactivation of postsynaptic glutamate receptors, which evokes a downstream cascade that eventually leads to neuronal dysfunction and degeneration. Also, a reduction in GABA receptor response after ischemia impedes these inhibitory effectors from attenuating excitotoxicity and thereby further enabling the excitotoxic insult. This review focuses on the mechanisms by which glutamate and GABA mediate excitotoxicity and ischemic injury. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
Collapse
Affiliation(s)
- Diana Mayor
- Division of Fundamental Neurobiology, Krembil Institute, University Health Network, Toronto, Ontario, M5T 2S8, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Michael Tymianski
- Division of Fundamental Neurobiology, Krembil Institute, University Health Network, Toronto, Ontario, M5T 2S8, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada; Department of Neurosurgery, University of Toronto, Toronto, Ontario, M5G 1LG, Canada.
| |
Collapse
|
24
|
Hobbs CN, Holzberg G, Min AS, Wightman RM. Comparison of Spreading Depolarizations in the Motor Cortex and Nucleus Accumbens: Similar Patterns of Oxygen Responses and the Role of Dopamine. ACS Chem Neurosci 2017; 8:2512-2521. [PMID: 28820571 PMCID: PMC5691918 DOI: 10.1021/acschemneuro.7b00266] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Spreading depolarizations (SD) are pathophysiological phenomena that spontaneously arise in traumatized neural tissue and can promote cellular death. Most investigations of SD are performed in the cortex, a brain region that is susceptible to these depolarizing waves and accessible via a variety of monitoring techniques. Here, we describe SD responses in the cortex and the deep brain region of the nucleus accumbens (NAc) of the anesthetized rat with a minimally invasive, implantable sensor. With high temporal resolution, we characterize the time course of oxygen responses to SD in relation to the electrophysiological depolarization signal. The predominant oxygen pattern consists of four phases: (1) a small initial decrease, (2) a large increase during the SD, (3) a delayed increase, and (4) a persistent decrease from baseline after the SD. Oxygen decreases during SD were also recorded. The latter response occurred more often in the NAc than the cortex (56% vs 20% of locations, respectively), which correlates to denser cortical vascularization. We also find that SDs travel more quickly in the cortex than NAc, likely affected by regional differences in cell type populations. Finally, we investigate the previously uncharacterized effects of dopamine release during SD in the NAc with dopamine receptor blockade. Our results support an inhibitory role of the D2 receptor on SD. As such, the data presented here expands the current understanding of within- and between-region variance in responses to SD.
Collapse
Affiliation(s)
- Caddy N. Hobbs
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Gordon Holzberg
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Akira S. Min
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - R. Mark Wightman
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
25
|
Effects of experimental traumatic brain injury and impaired glutamate transport on cortical spreading depression. Exp Neurol 2017; 295:155-161. [DOI: 10.1016/j.expneurol.2017.05.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 04/14/2017] [Accepted: 05/03/2017] [Indexed: 12/21/2022]
|
26
|
Rakers C, Schmid M, Petzold GC. TRPV4 channels contribute to calcium transients in astrocytes and neurons during peri-infarct depolarizations in a stroke model. Glia 2017. [PMID: 28639721 DOI: 10.1002/glia.23183] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Stroke is one of the leading causes of death and long-term disability. In the penumbra, that is, the area surrounding the infarct core, peri-infarct depolarizations (PIDs) are accompanied by strong intracellular calcium elevations in astrocytes and neurons, thereby negatively affecting infarct size and clinical outcome. The dynamics of PIDs and the cellular pathways that are involved during PID formation and progression remain incompletely understood. We have previously shown that inositol triphosphate-gated calcium release from internal stores is a major component of PID-related astroglial calcium signals, but whether external calcium influx through membrane-localized channels also contributes to PIDs has remained unclear. In this study, we investigated the role of two astroglial membrane channels, transient receptor vanilloid 4 (TRPV4) channel and aquaporin-4 (AQP4). We combined in vivo multiphoton microscopy, electrophysiology as well as laser speckle contrast imaging with the middle cerebral artery occlusion stroke model. Using knockout mice and pharmacological inhibitors, we found that TRPV4 channels contribute to calcium influx into astrocytes and neurons and subsequent extracellular glutamate accumulation during PIDs. AQP4 neither influenced PID-related calcium signals nor PID-related edema of astrocyte somata. Both channels did not alter the dynamics, frequency and cerebrovascular response of PIDs in the penumbra. These data indicate that TRPV4 channels may represent a potential target to ameliorate the PID-induced calcium overload of astrocytes and neurons during acute stroke.
Collapse
Affiliation(s)
- Cordula Rakers
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Street 27, Bonn, 53127, Germany
| | - Matthias Schmid
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Street 27, Bonn, 53127, Germany.,Department of Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Sigmund-Freud-Street 25, Bonn, 53127, Germany
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Street 27, Bonn, 53127, Germany.,Department of Neurology, University Hospital Bonn, Sigmund-Freud-Street 25, Bonn, 53127, Germany
| |
Collapse
|
27
|
Urbach A, Baum E, Braun F, Witte OW. Cortical spreading depolarization increases adult neurogenesis, and alters behavior and hippocampus-dependent memory in mice. J Cereb Blood Flow Metab 2017; 37:1776-1790. [PMID: 27189903 PMCID: PMC5435280 DOI: 10.1177/0271678x16643736] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cortical spreading depolarizations are an epiphenomenon of human brain pathologies and associated with extensive but transient changes in ion homeostasis, metabolism, and blood flow. Previously, we have shown that cortical spreading depolarization have long-lasting consequences on the brains transcriptome and structure. In particular, we found that cortical spreading depolarization stimulate hippocampal cell proliferation resulting in a sustained increase in adult neurogenesis. Since the hippocampus is responsible for explicit memory and adult-born dentate granule neurons contribute to this function, cortical spreading depolarization might influence hippocampus-dependent cognition. To address this question, we induced cortical spreading depolarization in C57Bl/6 J mice by epidural application of 1.5 mol/L KCl and evaluated neurogenesis and behavior at two, four, or six weeks thereafter. Congruent with our previous findings in rats, we found that cortical spreading depolarization increases numbers of newborn dentate granule neurons. Moreover, exploratory behavior and object location memory were consistently enhanced. Reference memory in the water maze was virtually unaffected, whereas memory formation in the Barnes maze was impaired with a delay of two weeks and facilitated after four weeks. These data show that cortical spreading depolarization produces lasting changes in psychomotor behavior and complex, delay- and task-dependent changes in spatial memory, and suggest that cortical spreading depolarization-like events affect the emotional and cognitive outcomes of associated brain pathologies.
Collapse
Affiliation(s)
- Anja Urbach
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Eileen Baum
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Falko Braun
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Otto W Witte
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| |
Collapse
|
28
|
Balança B, Meiller A, Bezin L, Dreier JP, Marinesco S, Lieutaud T. Altered hypermetabolic response to cortical spreading depolarizations after traumatic brain injury in rats. J Cereb Blood Flow Metab 2017; 37:1670-1686. [PMID: 27356551 PMCID: PMC5435292 DOI: 10.1177/0271678x16657571] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 05/27/2016] [Accepted: 05/30/2016] [Indexed: 01/11/2023]
Abstract
Spreading depolarizations are waves of near-complete breakdown of neuronal transmembrane ion gradients, free energy starving, and mass depolarization. Spreading depolarizations in electrically inactive tissue are associated with poor outcome in patients with traumatic brain injury. Here, we studied changes in regional cerebral blood flow and brain oxygen (PbtO2), glucose ([Glc]b), and lactate ([Lac]b) concentrations in rats, using minimally invasive real-time sensors. Rats underwent either spreading depolarizations chemically triggered by KCl in naïve cortex in absence of traumatic brain injury or spontaneous spreading depolarizations in the traumatic penumbra after traumatic brain injury, or a cluster of spreading depolarizations triggered chemically by KCl in a remote window from which spreading depolarizations invaded penumbral tissue. Spreading depolarizations in noninjured cortex induced a hypermetabolic response characterized by a decline in [Glc]b and monophasic increases in regional cerebral blood flow, PbtO2, and [Lac]b, indicating transient hyperglycolysis. Following traumatic brain injury, spontaneous spreading depolarizations occurred, causing further decline in [Glc]b and reducing the increase in regional cerebral blood flow and biphasic responses of PbtO2 and [Lac]b, followed by prolonged decline. Recovery of PbtO2 and [Lac]b was significantly delayed in traumatized animals. Prespreading depolarization [Glc]b levels determined the metabolic response to clusters. The results suggest a compromised hypermetabolic response to spreading depolarizations and slower return to physiological conditions following traumatic brain injury-induced spreading depolarizations.
Collapse
Affiliation(s)
- Baptiste Balança
- Inserm U1028, CNRS UMR 5292, Lyon Neuroscience Research Center, Team TIGER, Lyon, France
- Centre hospitalier universitaire de Lyon, France
| | - Anne Meiller
- Université Claude Bernard Lyon I, Lyon Neuroscience Research Center, AniRA-Neurochem Technological platform, Lyon, France
| | - Laurent Bezin
- Inserm U1028, CNRS UMR 5292, Lyon Neuroscience Research Center, Team TIGER, Lyon, France
| | - Jens P. Dreier
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
- Department of Neurology and Department of Experimental Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Stéphane Marinesco
- Inserm U1028, CNRS UMR 5292, Lyon Neuroscience Research Center, Team TIGER, Lyon, France
- Université Claude Bernard Lyon I, Lyon Neuroscience Research Center, AniRA-Neurochem Technological platform, Lyon, France
| | - Thomas Lieutaud
- Inserm U1028, CNRS UMR 5292, Lyon Neuroscience Research Center, Team TIGER, Lyon, France
| |
Collapse
|
29
|
Pinczolits A, Zdunczyk A, Dengler NF, Hecht N, Kowoll CM, Dohmen C, Graf R, Winkler MK, Major S, Hartings JA, Dreier JP, Vajkoczy P, Woitzik J. Standard-sampling microdialysis and spreading depolarizations in patients with malignant hemispheric stroke. J Cereb Blood Flow Metab 2017; 37:1896-1905. [PMID: 28350195 PMCID: PMC5435299 DOI: 10.1177/0271678x17699629] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Spreading depolarizations (SD) occur in high frequency in patients with malignant hemispheric stroke (MHS). Experimentally, SDs cause marked increases in glutamate and lactate, whereas glucose decreases. Here, we studied extracellular brain glutamate, glucose, lactate, pyruvate and the lactate/pyruvate ratio in relationship to SDs after MHS. We inserted two microdialysis probes in peri-infarct tissue at 5 and 15 mm to the infarct in close proximity to a subdural electrode strip. During 2356.6 monitoring hours, electrocorticography (ECoG) revealed 697 SDs in 16 of 18 patients. Ninety-nine SDs in electrically active tissue (spreading depressions, SDd) were single (SDds) and 485 clustered (SDdc), whereas 10 SDs with at least one electrode in electrically inactive tissue (isoelectric SDs, SDi) were single (SDis) and 103 clustered (SDic). More SDs and a significant number of clustered SDs occurred during the first 36 h post-surgery when glutamate was significantly elevated (> 100 µM). In a grouped analysis, we observed minor glutamate elevations with more than two SDs per hour. Glucose slightly decreased during SDic at 5 mm from the infarct. Directions of SD-related metabolic changes correspond to the experimental setting but the long sampling time of standard microdialysis precludes a more adequate account of the dynamics revealed by ECoG.
Collapse
Affiliation(s)
- Alexandra Pinczolits
- 1 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Zdunczyk
- 1 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nora F Dengler
- 1 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nils Hecht
- 1 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christina M Kowoll
- 3 Department of Neurology, University of Cologne, Cologne, Germany.,4 Max Planck Institute for Neurological Research, Cologne, Germany
| | - Christian Dohmen
- 3 Department of Neurology, University of Cologne, Cologne, Germany.,4 Max Planck Institute for Neurological Research, Cologne, Germany
| | - Rudolf Graf
- 4 Max Planck Institute for Neurological Research, Cologne, Germany
| | - Maren Kl Winkler
- 2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,5 Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Major
- 2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,5 Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jed A Hartings
- 6 Department of Neurosurgery, University of Cincinnati College of Medicine, Mayfield Clinic, Cincinnati, OH, USA
| | - Jens P Dreier
- 2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,5 Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Vajkoczy
- 1 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Woitzik
- 1 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
30
|
Lauritzen M, Strong AJ. 'Spreading depression of Leão' and its emerging relevance to acute brain injury in humans. J Cereb Blood Flow Metab 2017; 37:1553-1570. [PMID: 27354095 PMCID: PMC5435290 DOI: 10.1177/0271678x16657092] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
A new research field in translational neuroscience has opened as a result of the recognition since 2002 that "spreading depression of Leão" can be detected in many patients with acute brain injury, whether vascular and spontaneous, or traumatic in origin, as well as in those many individuals experiencing the visual (or sensorimotor) aura of migraine. In this review, we trace from their first description in rabbits through to their detection and study in migraine and the injured human brain, and from our personal perspectives, the evolution of understanding of the importance of spread of mass depolarisations in cerebral grey matter. Detection of spontaneous depolarisations occurring and spreading in the periphery or penumbra of experimental focal cortical ischemic lesions and of their adverse effects on the cerebral cortical microcirculation and on the tissue glucose and oxygen pools has led to clearer concepts of how ischaemic and traumatic lesions evolve in the injured human brain, and of how to seek to improve clinical management and outcome. Recognition of the likely fundamental significance of spreading depolarisations for this wide range of serious acute encephalopathies in humans provides a powerful case for a fresh examination of neuroprotection strategies.
Collapse
Affiliation(s)
- Martin Lauritzen
- 1 Department of Neuroscience and Pharmacology and Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark.,2 Department of Clinical Neurophysiology, Rigshospitalet, Glostrup, Denmark
| | - Anthony J Strong
- 3 Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
31
|
Hartings JA, Shuttleworth CW, Kirov SA, Ayata C, Hinzman JM, Foreman B, Andrew RD, Boutelle MG, Brennan KC, Carlson AP, Dahlem MA, Drenckhahn C, Dohmen C, Fabricius M, Farkas E, Feuerstein D, Graf R, Helbok R, Lauritzen M, Major S, Oliveira-Ferreira AI, Richter F, Rosenthal ES, Sakowitz OW, Sánchez-Porras R, Santos E, Schöll M, Strong AJ, Urbach A, Westover MB, Winkler MK, Witte OW, Woitzik J, Dreier JP. The continuum of spreading depolarizations in acute cortical lesion development: Examining Leão's legacy. J Cereb Blood Flow Metab 2017; 37:1571-1594. [PMID: 27328690 PMCID: PMC5435288 DOI: 10.1177/0271678x16654495] [Citation(s) in RCA: 291] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
A modern understanding of how cerebral cortical lesions develop after acute brain injury is based on Aristides Leão's historic discoveries of spreading depression and asphyxial/anoxic depolarization. Treated as separate entities for decades, we now appreciate that these events define a continuum of spreading mass depolarizations, a concept that is central to understanding their pathologic effects. Within minutes of acute severe ischemia, the onset of persistent depolarization triggers the breakdown of ion homeostasis and development of cytotoxic edema. These persistent changes are diagnosed as diffusion restriction in magnetic resonance imaging and define the ischemic core. In delayed lesion growth, transient spreading depolarizations arise spontaneously in the ischemic penumbra and induce further persistent depolarization and excitotoxic damage, progressively expanding the ischemic core. The causal role of these waves in lesion development has been proven by real-time monitoring of electrophysiology, blood flow, and cytotoxic edema. The spreading depolarization continuum further applies to other models of acute cortical lesions, suggesting that it is a universal principle of cortical lesion development. These pathophysiologic concepts establish a working hypothesis for translation to human disease, where complex patterns of depolarizations are observed in acute brain injury and appear to mediate and signal ongoing secondary damage.
Collapse
Affiliation(s)
- Jed A Hartings
- 1 Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,2 Mayfield Clinic, Cincinnati, OH, USA
| | - C William Shuttleworth
- 3 Department of Neuroscience, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Sergei A Kirov
- 4 Department of Neurosurgery and Brain and Behavior Discovery Institute, Medical College of Georgia, Augusta, GA, USA
| | - Cenk Ayata
- 5 Neurovascular Research Unit, Department of Radiology, and Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jason M Hinzman
- 1 Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Brandon Foreman
- 6 Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - R David Andrew
- 7 Department of Biomedical & Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Martyn G Boutelle
- 8 Department of Bioengineering, Imperial College London, London, United Kingdom
| | - K C Brennan
- 9 Department of Neurology, University of Utah, Salt Lake City, UT, USA.,10 Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT, USA
| | - Andrew P Carlson
- 11 Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Markus A Dahlem
- 12 Department of Physics, Humboldt University of Berlin, Berlin, Germany
| | | | - Christian Dohmen
- 14 Department of Neurology, University of Cologne, Cologne, Germany
| | - Martin Fabricius
- 15 Department of Clinical Neurophysiology, Rigshospitalet, Glostrup, Denmark
| | - Eszter Farkas
- 16 Department of Medical Physics and Informatics, Faculty of Medicine, and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Delphine Feuerstein
- 17 Multimodal Imaging of Brain Metabolism, Max-Planck-Institute for Metabolism Research, Cologne, Germany
| | - Rudolf Graf
- 17 Multimodal Imaging of Brain Metabolism, Max-Planck-Institute for Metabolism Research, Cologne, Germany
| | - Raimund Helbok
- 18 Medical University of Innsbruck, Department of Neurology, Neurocritical Care Unit, Innsbruck, Austria
| | - Martin Lauritzen
- 15 Department of Clinical Neurophysiology, Rigshospitalet, Glostrup, Denmark.,19 Department of Neuroscience and Pharmacology and Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Sebastian Major
- 13 Department of Neurology, Charité University Medicine, Berlin, Germany.,20 Center for Stroke Research Berlin, Charité University Medicine, Berlin, Germany.,21 Department of Experimental Neurology, Charité University Medicine, Berlin, Germany
| | - Ana I Oliveira-Ferreira
- 20 Center for Stroke Research Berlin, Charité University Medicine, Berlin, Germany.,21 Department of Experimental Neurology, Charité University Medicine, Berlin, Germany
| | - Frank Richter
- 22 Institute of Physiology/Neurophysiology, Jena University Hospital, Jena, Germany
| | - Eric S Rosenthal
- 5 Neurovascular Research Unit, Department of Radiology, and Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Oliver W Sakowitz
- 23 Department of Neurosurgery, Klinikum Ludwigsburg, Ludwigsburg, Germany.,24 Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Renán Sánchez-Porras
- 24 Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Edgar Santos
- 24 Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Schöll
- 24 Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Anthony J Strong
- 25 Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London
| | - Anja Urbach
- 26 Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - M Brandon Westover
- 5 Neurovascular Research Unit, Department of Radiology, and Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maren Kl Winkler
- 20 Center for Stroke Research Berlin, Charité University Medicine, Berlin, Germany
| | - Otto W Witte
- 26 Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany.,27 Brain Imaging Center, Jena University Hospital, Jena, Germany
| | - Johannes Woitzik
- 20 Center for Stroke Research Berlin, Charité University Medicine, Berlin, Germany.,28 Department of Neurosurgery, Charité University Medicine, Berlin, Germany
| | - Jens P Dreier
- 13 Department of Neurology, Charité University Medicine, Berlin, Germany.,20 Center for Stroke Research Berlin, Charité University Medicine, Berlin, Germany.,21 Department of Experimental Neurology, Charité University Medicine, Berlin, Germany
| |
Collapse
|
32
|
van Lieshout JH, Dibué-Adjei M, Cornelius JF, Slotty PJ, Schneider T, Restin T, Boogaarts HD, Steiger HJ, Petridis AK, Kamp MA. An introduction to the pathophysiology of aneurysmal subarachnoid hemorrhage. Neurosurg Rev 2017; 41:917-930. [PMID: 28215029 DOI: 10.1007/s10143-017-0827-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/24/2017] [Accepted: 01/31/2017] [Indexed: 02/06/2023]
Abstract
Pathophysiological processes following subarachnoid hemorrhage (SAH) present survivors of the initial bleeding with a high risk of morbidity and mortality during the course of the disease. As angiographic vasospasm is strongly associated with delayed cerebral ischemia (DCI) and clinical outcome, clinical trials in the last few decades focused on prevention of these angiographic spasms. Despite all efforts, no new pharmacological agents have shown to improve patient outcome. As such, it has become clear that our understanding of the pathophysiology of SAH is incomplete and we need to reevaluate our concepts on the complex pathophysiological process following SAH. Angiographic vasospasm is probably important. However, a unifying theory for the pathophysiological changes following SAH has yet not been described. Some of these changes may be causally connected or present themselves as an epiphenomenon of an associated process. A causal connection between DCI and early brain injury (EBI) would mean that future therapies should address EBI more specifically. If the mechanisms following SAH display no causal pathophysiological connection but are rather evoked by the subarachnoid blood and its degradation production, multiple treatment strategies addressing the different pathophysiological mechanisms are required. The discrepancy between experimental and clinical SAH could be one reason for unsuccessful translational results.
Collapse
Affiliation(s)
- Jasper H van Lieshout
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany.
| | - Maxine Dibué-Adjei
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Jan F Cornelius
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Philipp J Slotty
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Toni Schneider
- Institute for Neurophysiology, Medical Faculty, University of Cologne, Robert-Koch-Str. 39, 50931, Köln, Germany
| | - Tanja Restin
- Zurich Centre for Integrative Human Physiology, Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.,Institute of Anesthesiology, Medical Faculty, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Hieronymus D Boogaarts
- Department of Neurosurgery, Medical Faculty, Radboud University Nijmegen, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
| | - Hans-Jakob Steiger
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Athanasios K Petridis
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Marcel A Kamp
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| |
Collapse
|
33
|
Rakers C, Petzold GC. Astrocytic calcium release mediates peri-infarct depolarizations in a rodent stroke model. J Clin Invest 2016; 127:511-516. [PMID: 27991861 DOI: 10.1172/jci89354] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 11/03/2016] [Indexed: 11/17/2022] Open
Abstract
Stroke is one of the most common diseases and a leading cause of death and disability. Cessation of cerebral blood flow (CBF) leads to cell death in the infarct core, but tissue surrounding the core has the potential to recover if local reductions in CBF are restored. In these areas, detrimental peri-infarct depolarizations (PIDs) contribute to secondary infarct growth and negatively affect stroke outcome. However, the cellular pathways underlying PIDs have remained unclear. Here, we have used in vivo multiphoton microscopy, laser speckle imaging of CBF, and electrophysiological recordings in a mouse model of focal ischemia to demonstrate that PIDs are associated with a strong increase of intracellular calcium in astrocytes and neurons. We found that astroglial calcium elevations during PIDs are mediated by inositol triphosphate receptor type 2-dependent (IP3R2-dependent) release from internal stores. Importantly, Ip3r2-deficient mice displayed a reduction of PID frequency and overall PID burden and showed increased neuronal survival after stroke. These effects were not related to local CBF changes in response to PIDs. However, we showed that the release and extracellular accumulation of glutamate during PIDs is strongly curtailed in Ip3r2-deficient mice, resulting in ameliorated calcium overload in neurons and astrocytes. Together, these data implicate astroglial calcium pathways as potential targets for stroke therapy.
Collapse
|
34
|
Karunasinghe RN, Grey AC, Telang R, Vlajkovic SM, Lipski J. Differential spread of anoxic depolarization contributes to the pattern of neuronal injury after oxygen and glucose deprivation (OGD) in the Substantia Nigra in rat brain slices. Neuroscience 2016; 340:359-372. [PMID: 27826106 DOI: 10.1016/j.neuroscience.2016.10.067] [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: 08/22/2016] [Revised: 10/14/2016] [Accepted: 10/29/2016] [Indexed: 12/21/2022]
Abstract
Anoxic depolarization (AD) is an acute event evoked by brain ischemia, involving a profound loss of cell membrane potential and swelling that spreads over susceptible parts of the gray matter. Its occurrence is a strong predictor of the severity of neuronal injury. Little is known about this event in the Substantia Nigra, a midbrain nucleus critical for motor control. We tested the effects of oxygen and glucose deprivation (OGD), an in vitro model of brain ischemia, in rat midbrain slices. AD developed within 4min from OGD onset and spread in the Substantia Nigra pars reticulata (SNr), but not through the Substantia Nigra pars compacta (SNc). This differential effect involved a contrasting pattern of changes in membrane potential between dopamine-producing SNc and non-dopaminergic SNr neurons. A fast depolarization in SNr neurons was not followed by repolarization after the end of OGD, and was associated with swollen somata and beaded dendrites. In contrast, slowly developing depolarization of SNc neurons led to repolarization after OGD ended, and no changes in neuronal morphology were observed. The AD-resistance of the SNc involved smaller dysregulations of K+ and Ca2+ ions, and a slower loss of energy metabolites. Our results show that acute ischemia profoundly impairs the function and morphology of SNr neurons but not adjacent SNc neurons, and that the surprising higher tolerance of SNc neurons correlates with the resistance of the SNc region to AD. This differential response may affect the pattern of early neuronal injury that develops in the brainstem after acute ischemic insults.
Collapse
Affiliation(s)
- Rashika N Karunasinghe
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Angus C Grey
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Ravindra Telang
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Srdjan M Vlajkovic
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Janusz Lipski
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand.
| |
Collapse
|
35
|
Hinzman JM, Wilson JA, Mazzeo AT, Bullock MR, Hartings JA. Excitotoxicity and Metabolic Crisis Are Associated with Spreading Depolarizations in Severe Traumatic Brain Injury Patients. J Neurotrauma 2016; 33:1775-1783. [PMID: 26586606 DOI: 10.1089/neu.2015.4226] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cerebral microdialysis has enabled the clinical characterization of excitotoxicity (glutamate >10 μM) and non-ischemic metabolic crisis (lactate/pyruvate ratio [LPR] >40) as important components of secondary damage in severe traumatic brain injury (TBI). Spreading depolarizations (SD) are pathological waves that occur in many patients in the days following TBI and, in animal models, cause elevations in extracellular glutamate, increased anaerobic metabolism, and energy substrate depletion. Here, we examined the association of SD with changes in cerebral neurochemistry by placing a microdialysis probe alongside a subdural electrode strip in peri-lesional cortex of 16 TBI patients requiring neurosurgery. In 107 h (median; range: 76-117 h) of monitoring, 135 SDs were recorded in six patients. Glutamate (50 μmol/L) and lactate (3.7 mmol/L) were significantly elevated on day 0 in patients with SD compared with subsequent days and with patients without SD, whereas pyruvate was decreased in the latter group on days 0 and 1 (two-way analysis of variance [ANOVA], p values <0.05). In patients with SD, both glutamate and LPR increased in a dose-dependent manner with the number of SDs in the microdialysis sampling period (0, 1, ≥2 SD) [glutamate: 2.1→7.0→52.3 μmol/L; LPR: 27.8→29.9→45.0, p values <0.05]. In these patients, there was a 10% probability of SD occurring when glutamate and LPR were in normal ranges, but a 60% probability when both variables were abnormal (>10 μmol/L and >40 μmol/L, respectively). Taken together with previous studies, these preliminary clinical results suggest SDs are a key pathophysiological process of secondary brain injury associated with non-ischemic glutamate excitotoxicity and severe metabolic crisis in severe TBI patients.
Collapse
Affiliation(s)
- Jason M Hinzman
- 1 Department of Neurosurgery, University of Cincinnati (UC) College of Medicine , Cincinnati, Ohio
| | - J Adam Wilson
- 1 Department of Neurosurgery, University of Cincinnati (UC) College of Medicine , Cincinnati, Ohio
| | - Anna Teresa Mazzeo
- 2 Division of Neurosurgery, Virginia Commonwealth University , Richmond, Virginia.,3 Department Anesthesia and Intensive Care, University of Torino , Torino, Italy
| | - M Ross Bullock
- 2 Division of Neurosurgery, Virginia Commonwealth University , Richmond, Virginia.,4 Department of Neurosurgery, University of Miami , Miami, Florida
| | - Jed A Hartings
- 1 Department of Neurosurgery, University of Cincinnati (UC) College of Medicine , Cincinnati, Ohio.,5 Neurotrauma Center, UC Neuroscience Institute , Cincinnati, Ohio.,6 Mayfield Clinic , Cincinnati, Ohio
| |
Collapse
|
36
|
Guan YZ, Ye JH. Glycine blocks long-term potentiation of GABAergic synapses in the ventral tegmental area. Neuroscience 2016; 318:134-42. [PMID: 26806277 DOI: 10.1016/j.neuroscience.2016.01.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 10/22/2022]
Abstract
The mesocorticolimbic dopamine system, originating in the ventral tegmental area (VTA) is normally constrained by GABA-mediated synaptic inhibition. Accumulating evidence indicates that long-term potentiation of GABAergic synapses (LTPGABA) in VTA dopamine neurons plays an important role in the actions of drugs of abuse, including ethanol. We previously showed that a single infusion of glycine into the VTA of rats strongly reduces ethanol intake for 24h. In the current study, we examined the effect of glycine on the electrophysiological activities of putative dopamine VTA neurons in midbrain slices from ethanol-naïve rats. We report here that a 15-min exposure to 10 μM glycine prevented trains of high-frequency stimulation (HFS) from producing LTPGABA, which was rescued by the glycine receptor (GlyR) antagonist strychnine. Glycine also concentration-dependently decreased the frequency of spontaneous excitatory postsynaptic currents (sEPSCs). By contrast, glycine pretreatment did not prevent potentiation of inhibitory postsynaptic currents (IPSCs) during a continuous exposure to the nitric oxide (NO) donor, SNAP (S-nitroso-N-acetylpenicillamine), or a brief exposure to 10 μM glycine and 10 μM NMDA (N-methyl-D-aspartate), an agonist of NMDA-type glutamate receptors. Thus, the blockade of LTPGABA by glycine is probably resulted from suppressing glutamate release by activating the GlyRs on the glutamatergic terminals. This effect of glycine may contribute to the reduction in ethanol intake induced by intra-VTA glycine observed in vivo.
Collapse
Affiliation(s)
- Y-Z Guan
- Department of Anesthesiology, Pharmacology and Physiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA; Department of Physiology, Mudanjiang Medical University, Mudanjiang, China.
| | - J-H Ye
- Department of Anesthesiology, Pharmacology and Physiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
37
|
Esteve C, Tolner EA, Shyti R, van den Maagdenberg AMJM, McDonnell LA. Mass spectrometry imaging of amino neurotransmitters: a comparison of derivatization methods and application in mouse brain tissue. Metabolomics 2016; 12:30. [PMID: 26793043 PMCID: PMC4705126 DOI: 10.1007/s11306-015-0926-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/19/2015] [Indexed: 12/12/2022]
Abstract
The detection of small polar compounds such as amino neurotransmitters by MALDI mass spectrometry imaging has been hindered by low-detection sensitivity and background interferences. Recently, several of on-tissue chemical derivatization strategies have been independently reported that enable their detection. Here, we present a comparison between these methods, and demonstrate the visualization of the distributions of up to 23 amino metabolites in tissue. We applied this methodology to detect alterations of these compounds after inducing an experimental cortical spreading depression in mouse brain, which causes profound transient alterations in key neurotransmitters in one hemisphere and is relevant for migraine and various other neurological disorders.
Collapse
Affiliation(s)
- Clara Esteve
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Else A. Tolner
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Reinald Shyti
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Arn M. J. M. van den Maagdenberg
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Liam A. McDonnell
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
- Fondazione Pisana per la Scienza ONLUS, Pisa, Italy
| |
Collapse
|
38
|
Enger R, Tang W, Vindedal GF, Jensen V, Johannes Helm P, Sprengel R, Looger LL, Nagelhus EA. Dynamics of Ionic Shifts in Cortical Spreading Depression. Cereb Cortex 2015; 25:4469-76. [PMID: 25840424 PMCID: PMC4816793 DOI: 10.1093/cercor/bhv054] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Cortical spreading depression is a slowly propagating wave of near-complete depolarization of brain cells followed by temporary suppression of neuronal activity. Accumulating evidence indicates that cortical spreading depression underlies the migraine aura and that similar waves promote tissue damage in stroke, trauma, and hemorrhage. Cortical spreading depression is characterized by neuronal swelling, profound elevation of extracellular potassium and glutamate, multiphasic blood flow changes, and drop in tissue oxygen tension. The slow speed of the cortical spreading depression wave implies that it is mediated by diffusion of a chemical substance, yet the identity of this substance and the pathway it follows are unknown. Intercellular spread between gap junction-coupled neurons or glial cells and interstitial diffusion of K(+) or glutamate have been proposed. Here we use extracellular direct current potential recordings, K(+)-sensitive microelectrodes, and 2-photon imaging with ultrasensitive Ca(2+) and glutamate fluorescent probes to elucidate the spatiotemporal dynamics of ionic shifts associated with the propagation of cortical spreading depression in the visual cortex of adult living mice. Our data argue against intercellular spread of Ca(2+) carrying the cortical spreading depression wavefront and are in favor of interstitial K(+) diffusion, rather than glutamate diffusion, as the leading event in cortical spreading depression.
Collapse
Affiliation(s)
- Rune Enger
- Department of Neurology, Oslo University Hospital, 0027 Oslo, Norway
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
- Department of Molecular Medicine, Letten Centre and GliaLab, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
| | - Wannan Tang
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
- Department of Molecular Medicine, Letten Centre and GliaLab, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
- Department of Molecular Neurobiology, Max Planck Institute for Medical Research, D69120 Heidelberg, Germany
| | - Gry Fluge Vindedal
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
- Department of Molecular Medicine, Letten Centre and GliaLab, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
| | - Vidar Jensen
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
- Department of Molecular Medicine, Letten Centre and GliaLab, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
| | - P. Johannes Helm
- Department of Molecular Medicine, Letten Centre and GliaLab, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
| | - Rolf Sprengel
- Department of Molecular Neurobiology, Max Planck Institute for Medical Research, D69120 Heidelberg, Germany
| | - Loren L. Looger
- Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Erlend A. Nagelhus
- Department of Neurology, Oslo University Hospital, 0027 Oslo, Norway
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
- Department of Molecular Medicine, Letten Centre and GliaLab, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
- Department of Molecular Neurobiology, Max Planck Institute for Medical Research, D69120 Heidelberg, Germany
| |
Collapse
|
39
|
Variation of repetitive cortical spreading depression waves is related with relative refractory period: a computational study. QUANTITATIVE BIOLOGY 2015. [DOI: 10.1007/s40484-015-0052-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
40
|
Ayata C, Lauritzen M. Spreading Depression, Spreading Depolarizations, and the Cerebral Vasculature. Physiol Rev 2015; 95:953-93. [PMID: 26133935 DOI: 10.1152/physrev.00027.2014] [Citation(s) in RCA: 386] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Spreading depression (SD) is a transient wave of near-complete neuronal and glial depolarization associated with massive transmembrane ionic and water shifts. It is evolutionarily conserved in the central nervous systems of a wide variety of species from locust to human. The depolarization spreads slowly at a rate of only millimeters per minute by way of grey matter contiguity, irrespective of functional or vascular divisions, and lasts up to a minute in otherwise normal tissue. As such, SD is a radically different breed of electrophysiological activity compared with everyday neural activity, such as action potentials and synaptic transmission. Seventy years after its discovery by Leão, the mechanisms of SD and its profound metabolic and hemodynamic effects are still debated. What we did learn of consequence, however, is that SD plays a central role in the pathophysiology of a number of diseases including migraine, ischemic stroke, intracranial hemorrhage, and traumatic brain injury. An intriguing overlap among them is that they are all neurovascular disorders. Therefore, the interplay between neurons and vascular elements is critical for our understanding of the impact of this homeostatic breakdown in patients. The challenges of translating experimental data into human pathophysiology notwithstanding, this review provides a detailed account of bidirectional interactions between brain parenchyma and the cerebral vasculature during SD and puts this in the context of neurovascular diseases.
Collapse
Affiliation(s)
- Cenk Ayata
- Neurovascular Research Laboratory, Department of Radiology, and Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Department of Neuroscience and Pharmacology and Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark; and Department of Clinical Neurophysiology, Glostrup Hospital, Glostrup, Denmark
| | - Martin Lauritzen
- Neurovascular Research Laboratory, Department of Radiology, and Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Department of Neuroscience and Pharmacology and Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark; and Department of Clinical Neurophysiology, Glostrup Hospital, Glostrup, Denmark
| |
Collapse
|
41
|
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
|
42
|
Carreira RJ, Shyti R, Balluff B, Abdelmoula WM, van Heiningen SH, van Zeijl RJ, Dijkstra J, Ferrari MD, Tolner EA, McDonnell LA, van den Maagdenberg AMJM. Large-scale mass spectrometry imaging investigation of consequences of cortical spreading depression in a transgenic mouse model of migraine. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2015; 26:853-61. [PMID: 25877011 PMCID: PMC4422864 DOI: 10.1007/s13361-015-1136-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/10/2015] [Accepted: 03/10/2015] [Indexed: 05/04/2023]
Abstract
Cortical spreading depression (CSD) is the electrophysiological correlate of migraine aura. Transgenic mice carrying the R192Q missense mutation in the Cacna1a gene, which in patients causes familial hemiplegic migraine type 1 (FHM1), exhibit increased propensity to CSD. Herein, mass spectrometry imaging (MSI) was applied for the first time to an animal cohort of transgenic and wild type mice to study the biomolecular changes following CSD in the brain. Ninety-six coronal brain sections from 32 mice were analyzed by MALDI-MSI. All MSI datasets were registered to the Allen Brain Atlas reference atlas of the mouse brain so that the molecular signatures of distinct brain regions could be compared. A number of metabolites and peptides showed substantial changes in the brain associated with CSD. Among those, different mass spectral features showed significant (t-test, P < 0.05) changes in the cortex, 146 and 377 Da, and in the thalamus, 1820 and 1834 Da, of the CSD-affected hemisphere of FHM1 R192Q mice. Our findings reveal CSD- and genotype-specific molecular changes in the brain of FHM1 transgenic mice that may further our understanding about the role of CSD in migraine pathophysiology. The results also demonstrate the utility of aligning MSI datasets to a common reference atlas for large-scale MSI investigations.
Collapse
Affiliation(s)
- Ricardo J. Carreira
- />Center for Proteomics and Metabolomics, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Reinald Shyti
- />Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Benjamin Balluff
- />Center for Proteomics and Metabolomics, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Walid M. Abdelmoula
- />Division of Image Processing, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Rene J. van Zeijl
- />Center for Proteomics and Metabolomics, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Jouke Dijkstra
- />Division of Image Processing, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michel D. Ferrari
- />Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Else A. Tolner
- />Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Liam A. McDonnell
- />Center for Proteomics and Metabolomics, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
- />Fondazione Pisana per la Scienza ONLUS, Pisa, Italy
| | - Arn M. J. M. van den Maagdenberg
- />Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
- />Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
43
|
Hinzman JM, DiNapoli VA, Mahoney EJ, Gerhardt GA, Hartings JA. Spreading depolarizations mediate excitotoxicity in the development of acute cortical lesions. Exp Neurol 2015; 267:243-53. [PMID: 25819105 DOI: 10.1016/j.expneurol.2015.03.014] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/09/2015] [Accepted: 03/17/2015] [Indexed: 11/19/2022]
Abstract
Spreading depolarizations (SD) are mass depolarizations of neurons and astrocytes that occur spontaneously in acute brain injury and mediate time-dependent lesion growth. Glutamate excitotoxicity has also been extensively studied as a mechanism of neuronal injury, although its relevance to in vivo pathology remains unclear. Here we hypothesized that excitotoxicity in acute lesion development occurs only as a consequence of SD. Using glutamate-sensitive microelectrodes, we found that SD induced by KCl in normal rat cortex elicits increases in extracellular glutamate (11.6±1.3μM) that are synchronous with the onset, sustainment, and resolution of the extracellular direct-current shift of SD. Inhibition of glutamate uptake with d,l-threo-β-benzyloxyaspartate (TBOA, 0.5 and 1mM) significantly prolonged the duration of the direct-current shift (148% and 426%, respectively) and the glutamate increase (167% and 374%, respectively) in a dose-dependent manner (P<0.05). These prolonged events produced significant cortical lesions as indicated by Fluoro-Jade staining (P<0.05), while no lesions were observed after SD in control conditions or after cortical injection of 1mM glutamate (extracellular increase: 243±50.8μM) or 0.5mM TBOA (glutamate increase: 8.5±1.6μM) without SD. We then used an embolic focal ischemia model to determine whether glutamate elevations occur independent of SD in the natural evolution of a cortical lesion. In both the ischemic core and penumbra, glutamate increased only in synchrony with anoxic terminal SD (6.1±1.1μM) and transient SDs (11.8±2.4μM), and not otherwise. Delayed terminal SDs were also observed in two animals at 98 and 150min after ischemic onset and induced similar glutamate elevations. Durations of SDs and glutamate increases were significantly correlated in both normal and ischemic animals (P<0.05). These data suggest that pathologically prolonged SDs are a required mechanism of acute cortical lesion development and that glutamate elevations and the mass electrochemical changes of SD and are merely different facets of the same pathophysiologic process.
Collapse
Affiliation(s)
- Jason M Hinzman
- Department of Neurosurgery, University of Cincinnati (UC) College of Medicine and Neurotrauma Center at UC Neuroscience Institute, Cincinnati, OH, USA.
| | - Vince A DiNapoli
- Department of Neurosurgery, University of Cincinnati (UC) College of Medicine and Neurotrauma Center at UC Neuroscience Institute, Cincinnati, OH, USA; Mayfield Clinic, Cincinnati, OH, USA
| | - Eric J Mahoney
- Department of Neurosurgery, University of Cincinnati (UC) College of Medicine and Neurotrauma Center at UC Neuroscience Institute, Cincinnati, OH, USA
| | - Greg A Gerhardt
- Department of Anatomy and Neurobiology, University of Kentucky Chandler Medical Center, Morris K. Udall Parkinson's Disease Research Center of Excellence, Center for Microelectrode Technology, Spinal Cord and Brain Injury Research Center, Lexington, KY, USA
| | - Jed A Hartings
- Department of Neurosurgery, University of Cincinnati (UC) College of Medicine and Neurotrauma Center at UC Neuroscience Institute, Cincinnati, OH, USA; Mayfield Clinic, Cincinnati, OH, USA
| |
Collapse
|
44
|
Aiba I, Shuttleworth CW. Characterization of inhibitory GABA-A receptor activation during spreading depolarization in brain slice. PLoS One 2014; 9:e110849. [PMID: 25338191 PMCID: PMC4206427 DOI: 10.1371/journal.pone.0110849] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 09/24/2014] [Indexed: 12/28/2022] Open
Abstract
Spreading depolarization (SD) is a slowly propagating wave of near complete depolarizations of neurons and glia. Previous studies have reported large GABA releases during SD, but there is limited understanding of how GABA release and receptor activation are regulated and influence the propagating SD wavefront, as well as an excitatory phase immediately following the passage of SD. The present study characterized GABA-A type receptor (GABAAR) currents during SD generated by KCl microinjection in acute hippocampal slices from adult mice. Spontaneous GABAAR-mediated currents (sIPSCs) were initially enhanced, and were followed by a large outward current at the wavefront. sIPSC were then transiently supressed during the late SD phase, resulting in a significant reduction of the sIPSC/sEPSC ratio. The large outward current generated during SD was eliminated by the GABAAR antagonist gabazine, but the channel potentiator/agonist propofol failed to potentiate the current, likely because of a ceiling effect. Extracellular Cl− decreases recorded during SD were reduced by the antagonist but were not increased by the potentiator. Together with effects of GABAAR modulators on SD propagation rate, these results demonstrate a significant inhibitory role of the initial GABAAR activation and suggest that intracellular Cl− loading is insufficient to generate excitatory GABAAR responses during SD propagation. These results provide a mechanistic explanation for facilitating effects of GABAAR antagonists, and the lack of inhibitory effect of GABAAR potentiators on SD propagation. In addition, selective suppression of GABA transmission in the late SD period and the lack of effect of GABAA modulators on the duration of SD suggests that GABA modulation may not be effective approach to protect neurons during the vulnerable phase of SD.
Collapse
Affiliation(s)
- Isamu Aiba
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
| | - C. William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
- * E-mail:
| |
Collapse
|
45
|
Autio JA, Shatillo A, Giniatullin R, Gröhn OH. Parenchymal spin-lock fMRI signals associated with cortical spreading depression. J Cereb Blood Flow Metab 2014; 34:768-75. [PMID: 24496172 PMCID: PMC4013757 DOI: 10.1038/jcbfm.2014.16] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/30/2013] [Accepted: 01/08/2014] [Indexed: 01/30/2023]
Abstract
We found novel types of parenchymal functional magnetic resonance imaging (fMRI) signals in the rat brain during large increases in metabolism. Cortical spreading depression (CSD), a self-propagating wave of cellular activation, is associated with several pathologic conditions such as migraine and stroke. It was used as a paradigm to evoke transient neuronal depolarization leading to enhanced energy consumption. Activation of CSD was investigated using spin-lock (SL), diffusion, blood oxygenation level-dependent and cerebral blood volume fMRI techniques. Our results show that the SL-fMRI signal is generated by endogenous parenchymal mechanisms during CSD propagation, and these mechanisms are not associated with hemodynamic changes or cellular swelling. Protein phantoms suggest that pH change alone does not explain the observed SL-fMRI signal changes. However, increased amounts of inorganic phosphates released from high-energy phosphates combined with pH changes may produce SL- power-dependent longitudinal relaxation in the rotating frame (R₁ρ) changes in protein phantoms that are similar to those observed during CSD, as seen before in acute ischemia under our experimental conditions. This links SL-fMRI changes intimately to energy metabolism and supports the use of the SL technique as a new, promising functional approach for noninvasive imaging of metabolic transitions in the active or pathologic brain.
Collapse
Affiliation(s)
- Joonas A Autio
- 1] Department of Neurobiology, A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland [2] Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Artem Shatillo
- Department of Neurobiology, A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Rashid Giniatullin
- Department of Neurobiology, A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Olli H Gröhn
- Department of Neurobiology, A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
46
|
Sánchez-Porras R, Robles-Cabrera A, Santos E. Despolarización cortical extendida: un nuevo mecanismo fisiopatológico en las enfermedades neurológicas. Med Clin (Barc) 2014; 142:457-62. [DOI: 10.1016/j.medcli.2013.05.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 05/15/2013] [Accepted: 05/16/2013] [Indexed: 01/11/2023]
|
47
|
Oláh G, Herédi J, Menyhárt A, Czinege Z, Nagy D, Fuzik J, Kocsis K, Knapp L, Krucsó E, Gellért L, Kis Z, Farkas T, Fülöp F, Párdutz A, Tajti J, Vécsei L, Toldi J. Unexpected effects of peripherally administered kynurenic acid on cortical spreading depression and related blood-brain barrier permeability. DRUG DESIGN DEVELOPMENT AND THERAPY 2013; 7:981-7. [PMID: 24068867 PMCID: PMC3782408 DOI: 10.2147/dddt.s44496] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Cortical spreading depression (CSD) involves a slowly-propagating depolarization wave in the cortex, which can appear in numerous pathophysiological conditions, such as migraine with aura, stroke, and traumatic brain injury. Neurons and glial cells are also depolarized transiently during the phenomena. CSD is followed by a massive increase in glutamate release and by changes in the brain microcirculation. The aim of this study was to investigate the effects of two N-methyl-D-aspartate receptor antagonists, endogenous kynurenic acid (KYNA) and dizocilpine, on CSD and the related blood-brain barrier (BBB) permeability in rats. In intact animals, KYNA hardly crosses the BBB but has some positive features as compared with its precursor L-Kynurenine, which is frequently used in animal studies (KYNA cannot be metabolized to excitotoxic agents such as 3-hydroxy-L-kynurenine and quinolinic acid). We therefore investigated the possible effects of peripherally administered KYNA. Repetitive CSD waves were elicited by the application of 1 M KCl solution to the cortex. Direct current-electrocorticograms were measured for 1 hour. Four parameters of the waves were compared. Evans blue dye and fluorescent microscopy were used to study the possible changes in the permeability of the BBB. The results demonstrated that N-methyl-D-aspartate receptor antagonists can reduce the number of CSD waves and decrease the permeability of the BBB during CSD. These results suggest that KYNA itself or its derivatives may offer a new approach in the therapy of migraines.
Collapse
Affiliation(s)
- Gáspár Oláh
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Közép fasor 52, Szeged, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Sánchez-Porras R, Zheng Z, Santos E, Schöll M, Unterberg AW, Sakowitz OW. The role of spreading depolarization in subarachnoid hemorrhage. Eur J Neurol 2013; 20:1121-7. [DOI: 10.1111/ene.12139] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 02/15/2013] [Indexed: 11/30/2022]
Affiliation(s)
- R. Sánchez-Porras
- Department of Neurosurgery; Heidelberg University Hospital; Heidelberg; Germany
| | - Z. Zheng
- Department of Neurosurgery; Heidelberg University Hospital; Heidelberg; Germany
| | - E. Santos
- Department of Neurosurgery; Heidelberg University Hospital; Heidelberg; Germany
| | - M. Schöll
- Department of Neurosurgery; Heidelberg University Hospital; Heidelberg; Germany
| | - A. W. Unterberg
- Department of Neurosurgery; Heidelberg University Hospital; Heidelberg; Germany
| | - O. W. Sakowitz
- Department of Neurosurgery; Heidelberg University Hospital; Heidelberg; Germany
| |
Collapse
|
49
|
Sakowitz OW, Santos E, Nagel A, Krajewski KL, Hertle DN, Vajkoczy P, Dreier JP, Unterberg AW, Sarrafzadeh AS. Clusters of spreading depolarizations are associated with disturbed cerebral metabolism in patients with aneurysmal subarachnoid hemorrhage. Stroke 2012; 44:220-3. [PMID: 23223504 DOI: 10.1161/strokeaha.112.672352] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE We studied the dynamics of extracellular brain tissue concentrations of glucose, lactate, pyruvate, and glutamate during the occurrence of spreading depolarizations (SDs) in patients with aneurysmal subarachnoid hemorrhage. METHODS In this prospective observational study, patients with aneurysmal subarachnoid hemorrhage received multimodal cerebral monitoring, including intracranial pressure, cerebral microdialysis, and subdural electrocorticography. RESULTS Seven of the 17 recruited patients had intracerebral hemorrhage, acute ischemia and severe brain oedema leading to acute ischemic neurological deficits associated with early disturbance of metabolism at the recording site. They displayed a total of 130 SDs. The remaining 10 patients without acute ischemic neurological deficits exhibited 138 single SDs and 68 SDs in clusters. In patients without acute ischemic neurological deficits, clustered SDs were associated with a significant transient decrease in glucose and increase in lactate compared with baseline during the first 140 minutes after SDs. Moreover, the number of clustered SDs correlated with the outcome (R=-0.659; P<0.01). CONCLUSIONS SDs can propagate in nonischemic human brain tissue. Clusters of SDs are related to metabolic changes suggestive of ongoing secondary damage in primarily nonischemic brain tissue.
Collapse
Affiliation(s)
- Oliver W Sakowitz
- University of Heidelberg, Department of Neurosurgery, Im Neuenheimer Feld 400, D-69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Imaging mass spectrometry to visualize biomolecule distributions in mouse brain tissue following hemispheric cortical spreading depression. J Proteomics 2012; 75:5027-5035. [PMID: 22776886 DOI: 10.1016/j.jprot.2012.06.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2012] [Accepted: 06/29/2012] [Indexed: 01/14/2023]
Abstract
MALDI mass spectrometry can simultaneously measure hundreds of biomolecules directly from tissue. Using essentially the same technique but different sample preparation strategies, metabolites, lipids, peptides and proteins can be analyzed. Spatially correlated analysis, imaging MS, enables the distributions of these biomolecular ions to be simultaneously measured in tissues. A key advantage of imaging MS is that it can annotate tissues based on their MS profiles and thereby distinguish biomolecularly distinct regions even if they were unexpected or are not distinct using established histological and histochemical methods e.g. neuropeptide and metabolite changes following transient electrophysiological events such as cortical spreading depression (CSD), which are spreading events of massive neuronal and glial depolarisations that occur in one hemisphere of the brain and do not pass to the other hemisphere , enabling the contralateral hemisphere to act as an internal control. A proof-of-principle imaging MS study, including 2D and 3D datasets, revealed substantial metabolite and neuropeptide changes immediately following CSD events which were absent in the protein imaging datasets. The large high dimensionality 3D datasets make even rudimentary contralateral comparisons difficult to visualize. Instead non-negative matrix factorization (NNMF), a multivariate factorization tool that is adept at highlighting latent features, such as MS signatures associated with CSD events, was applied to the 3D datasets. NNMF confirmed that the protein dataset did not contain substantial contralateral differences, while these were present in the neuropeptide dataset.
Collapse
|