1
|
Carlson AP, Mayer AR, Cole C, van der Horn HJ, Marquez J, Stevenson TC, Shuttleworth CW. Cerebral autoregulation, spreading depolarization, and implications for targeted therapy in brain injury and ischemia. Rev Neurosci 2024; 35:651-678. [PMID: 38581271 PMCID: PMC11297425 DOI: 10.1515/revneuro-2024-0028] [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: 02/22/2024] [Accepted: 03/25/2024] [Indexed: 04/08/2024]
Abstract
Cerebral autoregulation is an intrinsic myogenic response of cerebral vasculature that allows for preservation of stable cerebral blood flow levels in response to changing systemic blood pressure. It is effective across a broad range of blood pressure levels through precapillary vasoconstriction and dilation. Autoregulation is difficult to directly measure and methods to indirectly ascertain cerebral autoregulation status inherently require certain assumptions. Patients with impaired cerebral autoregulation may be at risk of brain ischemia. One of the central mechanisms of ischemia in patients with metabolically compromised states is likely the triggering of spreading depolarization (SD) events and ultimately, terminal (or anoxic) depolarization. Cerebral autoregulation and SD are therefore linked when considering the risk of ischemia. In this scoping review, we will discuss the range of methods to measure cerebral autoregulation, their theoretical strengths and weaknesses, and the available clinical evidence to support their utility. We will then discuss the emerging link between impaired cerebral autoregulation and the occurrence of SD events. Such an approach offers the opportunity to better understand an individual patient's physiology and provide targeted treatments.
Collapse
Affiliation(s)
- Andrew P. Carlson
- Department of Neurosurgery, University of New Mexico School of Medicine, MSC10 5615, 1 UNM, Albuquerque, NM, 87131, USA
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Albuquerque, NM, 87106, USA
| | - Andrew R. Mayer
- Mind Research Network, 1101 Yale, Blvd, NE, Albuquerque, NM, 87106, USA
| | - Chad Cole
- Department of Neurosurgery, University of New Mexico School of Medicine, MSC10 5615, 1 UNM, Albuquerque, NM, 87131, USA
| | | | - Joshua Marquez
- University of New Mexico School of Medicine, 915 Camino de Salud NE, Albuquerque, NM, 87106, USA
| | - Taylor C. Stevenson
- Department of Neurosurgery, University of New Mexico School of Medicine, MSC10 5615, 1 UNM, Albuquerque, NM, 87131, USA
| | - C. William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Albuquerque, NM, 87106, USA
| |
Collapse
|
2
|
van Hameren G, Aboghazleh R, Parker E, Dreier JP, Kaufer D, Friedman A. From spreading depolarization to blood-brain barrier dysfunction: navigating traumatic brain injury for novel diagnosis and therapy. Nat Rev Neurol 2024; 20:408-425. [PMID: 38886512 DOI: 10.1038/s41582-024-00973-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2024] [Indexed: 06/20/2024]
Abstract
Considerable strides in medical interventions during the acute phase of traumatic brain injury (TBI) have brought improved overall survival rates. However, following TBI, people often face ongoing, persistent and debilitating long-term complications. Here, we review the recent literature to propose possible mechanisms that lead from TBI to long-term complications, focusing particularly on the involvement of a compromised blood-brain barrier (BBB). We discuss evidence for the role of spreading depolarization as a key pathological mechanism associated with microvascular dysfunction and the transformation of astrocytes to an inflammatory phenotype. Finally, we summarize new predictive and diagnostic biomarkers and explore potential therapeutic targets for treating long-term complications of TBI.
Collapse
Affiliation(s)
- Gerben van Hameren
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Refat Aboghazleh
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Basic Medical Sciences, Faculty of Medicine, Al-Balqa Applied University, Al-Salt, Jordan
| | - Ellen Parker
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
- Division of Neurosurgery, Dalhousie University QEII Health Sciences Centre, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Jens P Dreier
- Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Daniela Kaufer
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Alon Friedman
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada.
- Department of Cell Biology, Cognitive and Brain Sciences, Zelman Inter-Disciplinary Center of Brain Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
3
|
Kowoll CM, Schumm L, Gieffers A, Lemale CL, Major S, Dohmen C, Fink GR, Brinker G, von Pidoll T, Dömer P, Dreier JP, Hecht N, Woitzik J. Duration of spreading depression is the electrophysiological correlate of infarct growth in malignant hemispheric stroke. J Cereb Blood Flow Metab 2024:271678X241262203. [PMID: 38902207 DOI: 10.1177/0271678x241262203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Spreading depolarizations (SD) contribute to lesion progression after experimental focal cerebral ischemia while such correlation has never been shown in stroke patients. In this prospective, diagnostic study, we investigate the association of SDs and secondary infarct progression after malignant hemispheric stroke. SDs were continuously monitored for 3-9 days with electrocorticography after decompressive hemicraniectomy for malignant hemispheric stroke. To ensure valid detection and analysis of SDs, a threshold based on the electrocorticographic baseline activity was calculated to identify valid electrocorticographic recordings. Subsequently SD characteristics were analyzed in association to infarct progression based on serial MRI. Overall, 62 patients with a mean stroke volume of 289.6 ± 68 cm3 were included. Valid electrocorticographic recordings were found in 44/62 patients with a mean recording duration of 139.6 ± 26.5 hours and 52.5 ± 39.5 SDs per patient. Infarct progression of more than 5% was found in 21/44 patients. While the number of SDs was similar between patients with and without infarct progression, the SD-induced depression duration per day was significantly longer in patients with infarct progression (593.8 vs. 314.1 minutes; *p = 0.046). Therefore, infarct progression is associated with a prolonged SD-induced depression duration. Real-time analysis of electrocorticographic recordings may identify secondary stroke progression and help implementing targeted management strategies.
Collapse
Affiliation(s)
- Christina M Kowoll
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Neurology, Märkische Kliniken Lüdenscheid, Lüdenscheid, Germany
| | - Leonie Schumm
- Department of Neurosurgery, Evangelisches Krankenhaus Oldenburg, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alexandra Gieffers
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- 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
| | - 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
| | - 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
| | - Christian Dohmen
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Neurology, LVR-Klinik Bonn, Bonn, Germany
| | - Gereon R Fink
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Jülich, Jülich, Germany
| | - Gerrit Brinker
- Department of Neurosurgery, University of Cologne, Cologne, Germany
| | - Tilmann von Pidoll
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Trauma Surgery, SANA-Dreifaltigkeitskrankenhaus Cologne, Cologne, Germany
| | - Patrick Dömer
- Department of Neurosurgery, Evangelisches Krankenhaus Oldenburg, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, 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 Centre for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Centre for Neurosciences Berlin, Berlin, Germany
| | - Nils Hecht
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- 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
| | - Johannes Woitzik
- Department of Neurosurgery, Evangelisches Krankenhaus Oldenburg, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
4
|
Schoknecht K, Maechler M, Wallach I, Dreier JP, Liotta A, Berndt N. Isoflurane lowers the cerebral metabolic rate of oxygen and prevents hypoxia during cortical spreading depolarization in vitro: An integrative experimental and modeling study. J Cereb Blood Flow Metab 2024; 44:1000-1012. [PMID: 38140913 PMCID: PMC11318408 DOI: 10.1177/0271678x231222306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 10/23/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023]
Abstract
Cortical spreading depolarization (SD) imposes a massive increase in energy demand and therefore evolves as a target for treatment following acute brain injuries. Anesthetics are empirically used to reduce energy metabolism in critical brain conditions, yet their effect on metabolism during SD remains largely unknown. We investigated oxidative metabolism during SD in brain slices from Wistar rats. Extracellular potassium ([K+]o), local field potential and partial tissue oxygen pressure (ptiO2) were measured simultaneously. The cerebral metabolic rate of oxygen (CMRO2) was calculated using a reaction-diffusion model. By that, we tested the effect of clinically relevant concentrations of isoflurane on CMRO2 during SD and modeled tissue oxygenation for different capillary pO2 values. During SD, CMRO2 increased 2.7-fold, resulting in transient hypoxia in the slice core. Isoflurane decreased CMRO2, reduced peak [K+]o, and prolonged [K+]o clearance, which indicates reduced synaptic transmission and sodium-potassium ATPase inhibition. Modeling tissue oxygenation during SD illustrates the need for increased capillary pO2 levels to prevent hypoxia. In the absence thereof, isoflurane could improve tissue oxygenation by lowering CMRO2. Therefore, isoflurane is a promising candidate for pre-clinical studies on neuronal survival in conditions involving SD.
Collapse
Affiliation(s)
- Karl Schoknecht
- Carl-Ludwig-Institute of Physiology, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Mathilde Maechler
- Department of Anesthesiology and Intensive Care, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Computer-Assisted Cardiovascular Medicine, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Iwona Wallach
- Institute of Computer-Assisted Cardiovascular Medicine, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jens P Dreier
- Centre for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Bernstein Centre for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Centre for Neurosciences Berlin, Berlin, Germany
| | - Agustin Liotta
- Department of Anesthesiology and Intensive Care, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Computer-Assisted Cardiovascular Medicine, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Health at Charité – Universitätsmedizin Berlin, Berlin
- Neuroscience Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nikolaus Berndt
- Institute of Computer-Assisted Cardiovascular Medicine, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Department of Molecular Toxicology, Nuthetal, Germany
| |
Collapse
|
5
|
Zdunczyk A, Schumm L, Helgers SOA, Nieminen-Kelhä M, Bai X, Major S, Dreier JP, Hecht N, Woitzik J. Ketamine-induced prevention of SD-associated late infarct progression in experimental ischemia. Sci Rep 2024; 14:10186. [PMID: 38702377 PMCID: PMC11068759 DOI: 10.1038/s41598-024-59835-5] [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: 01/04/2024] [Accepted: 04/16/2024] [Indexed: 05/06/2024] Open
Abstract
Spreading depolarizations (SDs) occur frequently in patients with malignant hemispheric stroke. In animal-based experiments, SDs have been shown to cause secondary neuronal damage and infarct expansion during the initial period of infarct progression. In contrast, the influence of SDs during the delayed period is not well characterized yet. Here, we analyzed the impact of SDs in the delayed phase after cerebral ischemia and the potential protective effect of ketamine. Focal ischemia was induced by distal occlusion of the left middle cerebral artery in C57BL6/J mice. 24 h after occlusion, SDs were measured using electrocorticography and laser-speckle imaging in three different study groups: control group without SD induction, SD induction with potassium chloride, and SD induction with potassium chloride and ketamine administration. Infarct progression was evaluated by sequential MRI scans. 24 h after occlusion, we observed spontaneous SDs with a rate of 0.33 SDs/hour which increased during potassium chloride application (3.37 SDs/hour). The analysis of the neurovascular coupling revealed prolonged hypoemic and hyperemic responses in this group. Stroke volume increased even 24 h after stroke onset in the SD-group. Ketamine treatment caused a lesser pronounced hypoemic response and prevented infarct growth in the delayed phase after experimental ischemia. Induction of SDs with potassium chloride was significantly associated with stroke progression even 24 h after stroke onset. Therefore, SD might be a significant contributor to delayed stroke progression. Ketamine might be a possible drug to prevent SD-induced delayed stroke progression.
Collapse
Affiliation(s)
- A Zdunczyk
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- 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
| | - L Schumm
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- 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
| | - S O A Helgers
- Department of Neurosurgery, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - M Nieminen-Kelhä
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- 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
| | - X Bai
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- 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
| | - S 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 Neurology, 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
| | - J 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 Neurology, 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
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - N Hecht
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- 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
| | - Johannes Woitzik
- Department of Neurosurgery, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.
- Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.
- University Clinic for Neurosurgery, Marienstr. 11, 26121, Oldenburg, Germany.
| |
Collapse
|
6
|
Joya A, Plaza-García S, Padro D, Aguado L, Iglesias L, Garbizu M, Gómez-Vallejo V, Laredo C, Cossío U, Torné R, Amaro S, Planas AM, Llop J, Ramos-Cabrer P, Justicia C, Martín A. Multimodal imaging of the role of hyperglycemia following experimental subarachnoid hemorrhage. J Cereb Blood Flow Metab 2024; 44:726-741. [PMID: 37728631 PMCID: PMC11197138 DOI: 10.1177/0271678x231197946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/27/2023] [Accepted: 08/06/2023] [Indexed: 09/21/2023]
Abstract
Hyperglycemia has been linked to worsening outcomes after subarachnoid hemorrhage (SAH). Nevertheless, the mechanisms involved in the pathogenesis of SAH have been scarcely evaluated so far. The role of hyperglycemia was assessed in an experimental model of SAH by T2 weighted, dynamic contrast-enhanced magnetic resonance imaging (T2W and DCE-MRI), [18F]BR-351 PET imaging and immunohistochemistry. Measures included the volume of bleeding, the extent of cerebral infarction and brain edema, blood brain barrier disruption (BBBd), neutrophil infiltration and matrix metalloprotease (MMP) activation. The neurofunctional outcome, neurodegeneration and myelinization were also investigated. The induction of hyperglycemia increased mortality, the size of the ischemic lesion, brain edema, neurodegeneration and worsened neurological outcome during the first 3 days after SAH in rats. In addition, these results show for the first time the exacerbating effect of hyperglycemia on in vivo MMP activation, Intercellular Adhesion Molecule 1 (ICAM-1) expression and neutrophil infiltration together with increased BBBd, bleeding volume and fibrinogen accumulation at days 1 and 3 after SAH. Notably, these data provide valuable insight into the detrimental effect of hyperglycemia on early BBB damage mediated by neutrophil infiltration and MMP activation that could explain the worse prognosis in SAH.
Collapse
Affiliation(s)
- Ana Joya
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
| | - Sandra Plaza-García
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
| | - Daniel Padro
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
| | - Laura Aguado
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
| | - Leyre Iglesias
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Neurovascular Group, Biocruces Health Research Institute, Barakaldo, Spain
| | - Maider Garbizu
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
| | | | - Carlos Laredo
- Institute of Neuroscience, Comprehensive Stroke Center, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Unai Cossío
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
| | - Ramon Torné
- Institute of Neuroscience, Neurosurgery Department, Hospital Clinic of Barcelona, Spain
| | - Sergio Amaro
- Institute of Neuroscience, Comprehensive Stroke Center, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Anna M Planas
- Area of Neurosciences. Institut d'Investigacions Biomèdiques Agustí Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Neuroscience and Experimental Therapeutics, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Jordi Llop
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
- Centro de Investigación Biomédica en Red - Enfermedades Respiratorias, CIBERES, Madrid, Spain
| | - Pedro Ramos-Cabrer
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, Spain
| | - Carles Justicia
- Area of Neurosciences. Institut d'Investigacions Biomèdiques Agustí Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Neuroscience and Experimental Therapeutics, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Abraham Martín
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
7
|
Menyhárt Á, Bálint AR, Kozák P, Bari F, Farkas E. Nimodipine accelerates the restoration of functional hyperemia during spreading oligemia. J Neurochem 2024; 168:888-898. [PMID: 36810711 DOI: 10.1111/jnc.15792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 02/24/2023]
Abstract
Spreading depolarization (SD) is assumed to be the pathophysiological correlate of migraine aura, leading to spreading depression of activity and a long-lasting vasoconstriction known as spreading oligemia. Furthermore, cerebrovascular reactivity is reversibly impaired after SD. Here, we explored the progressive restoration of impaired neurovascular coupling to somatosensory activation during spreading oligemia. Also, we evaluated whether nimodipine treatment accelerated the recovery of impaired neurovascular coupling after SD. Male, 4-9-month-old C57BL/6 mice (n = 11) were anesthetized with isoflurane (1%-1.5%), and SD was triggered with KCl through a burr hole made at the caudal parietal bone. EEG and cerebral blood flow (CBF) were recorded minimally invasively with a silver ball electrode and transcranial laser-Doppler flowmetry, rostral to SD elicitation. The L-type voltage-gated Ca2+ channel blocker nimodipine was administered i.p. (10 mg/kg). Whisker stimulation-related evoked potentials (EVPs) and functional hyperemia were assessed under isoflurane (0.1%)-medetomidine (0.1 mg/kg i.p.) anesthesia before, and repeatedly after SD, at 15-min intervals for 75 minutes. Nimodipine accelerated the recovery of CBF from spreading oligemia (time to full recovery, 52 ± 13 vs. 70 ± 8 min, nimodipine vs. control) and exhibited a tendency to shorten the duration of the SD-related EGG depression duration. The amplitudes of EVP and functional hyperemia were markedly reduced after SD, and progressively recovered over an hour post-SD. Nimodipine exerted no impact on EVP amplitude but consistently increased the absolute level of functional hyperemia from 20 min post-CSD (93 ± 11% vs. 66 ± 13%, nimodipine vs. control). A linear, positive correlation between EVP and functional hyperemia amplitude was skewed by nimodipine. In conclusion, nimodipine facilitated CBF restoration from spreading oligemia and the recovery of functional hyperemia post-SD, which were linked to a tendency of an accelerated return of spontaneous neural activity after SD. The use of nimodipine in migraine prophylaxis is suggested to be re-visited.
Collapse
Affiliation(s)
- Ákos Menyhárt
- Hungarian Centre of Excellence for Molecular Medicine-University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Armand Rafael Bálint
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Péter Kozák
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ferenc Bari
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Eszter Farkas
- Hungarian Centre of Excellence for Molecular Medicine-University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| |
Collapse
|
8
|
Dreier JP, Joerk A, Uchikawa H, Horst V, Lemale CL, Radbruch H, McBride DW, Vajkoczy P, Schneider UC, Xu R. All Three Supersystems-Nervous, Vascular, and Immune-Contribute to the Cortical Infarcts After Subarachnoid Hemorrhage. Transl Stroke Res 2024:10.1007/s12975-024-01242-z. [PMID: 38689162 DOI: 10.1007/s12975-024-01242-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 05/02/2024]
Abstract
The recently published DISCHARGE-1 trial supports the observations of earlier autopsy and neuroimaging studies that almost 70% of all focal brain damage after aneurysmal subarachnoid hemorrhage are anemic infarcts of the cortex, often also affecting the white matter immediately below. The infarcts are not limited by the usual vascular territories. About two-fifths of the ischemic damage occurs within ~ 48 h; the remaining three-fifths are delayed (within ~ 3 weeks). Using neuromonitoring technology in combination with longitudinal neuroimaging, the entire sequence of both early and delayed cortical infarct development after subarachnoid hemorrhage has recently been recorded in patients. Characteristically, cortical infarcts are caused by acute severe vasospastic events, so-called spreading ischemia, triggered by spontaneously occurring spreading depolarization. In locations where a spreading depolarization passes through, cerebral blood flow can drastically drop within a few seconds and remain suppressed for minutes or even hours, often followed by high-amplitude, sustained hyperemia. In spreading depolarization, neurons lead the event, and the other cells of the neurovascular unit (endothelium, vascular smooth muscle, pericytes, astrocytes, microglia, oligodendrocytes) follow. However, dysregulation in cells of all three supersystems-nervous, vascular, and immune-is very likely involved in the dysfunction of the neurovascular unit underlying spreading ischemia. It is assumed that subarachnoid blood, which lies directly on the cortex and enters the parenchyma via glymphatic channels, triggers these dysregulations. This review discusses the neuroglial, neurovascular, and neuroimmunological dysregulations in the context of spreading depolarization and spreading ischemia as critical elements in the pathogenesis of cortical infarcts after subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Jens P Dreier
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, 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.
| | - Alexander Joerk
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Hiroki Uchikawa
- Barrow Aneurysm & AVM Research Center, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Viktor Horst
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Neuropathology, 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, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, 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
| | - Helena Radbruch
- Institute of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Devin W McBride
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ulf C Schneider
- Department of Neurosurgery, Cantonal Hospital of Lucerne and University of Lucerne, Lucerne, Switzerland
| | - Ran Xu
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Berlin, Germany
| |
Collapse
|
9
|
Camassa A, Barbero-Castillo A, Bosch M, Dasilva M, Masvidal-Codina E, Villa R, Guimerà-Brunet A, Sanchez-Vives MV. Chronic full-band recordings with graphene microtransistors as neural interfaces for discrimination of brain states. NANOSCALE HORIZONS 2024; 9:589-597. [PMID: 38329118 DOI: 10.1039/d3nh00440f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Brain states such as sleep, anesthesia, wakefulness, or coma are characterized by specific patterns of cortical activity dynamics, from local circuits to full-brain emergent properties. We previously demonstrated that full-spectrum signals, including the infraslow component (DC, direct current-coupled), can be recorded acutely in multiple sites using flexible arrays of graphene solution-gated field-effect transistors (gSGFETs). Here, we performed chronic implantation of 16-channel gSGFET arrays over the rat cerebral cortex and recorded full-band neuronal activity with two objectives: (1) to test the long-term stability of implanted devices; and (2) to investigate full-band activity during the transition across different levels of anesthesia. First, we demonstrate it is possible to record full-band signals with stability, fidelity, and spatiotemporal resolution for up to 5.5 months using chronic epicortical gSGFET implants. Second, brain states generated by progressive variation of levels of anesthesia could be identified as traditionally using the high-pass filtered (AC, alternating current-coupled) spectrogram: from synchronous slow oscillations in deep anesthesia through to asynchronous activity in the awake state. However, the DC signal introduced a highly significant improvement for brain-state discrimination: the DC band provided an almost linear information prediction of the depth of anesthesia, with about 85% precision, using a trained algorithm. This prediction rose to about 95% precision when the full-band (AC + DC) spectrogram was taken into account. We conclude that recording infraslow activity using gSGFET interfaces is superior for the identification of brain states, and further supports the preclinical and clinical use of graphene neural interfaces for long-term recordings of cortical activity.
Collapse
Affiliation(s)
- A Camassa
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - A Barbero-Castillo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - M Bosch
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - M Dasilva
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - E Masvidal-Codina
- Instituto de Microelectrónica de Barcelona (IMB-CNM, CSIC), Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain
| | - R Villa
- Instituto de Microelectrónica de Barcelona (IMB-CNM, CSIC), Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain
| | - A Guimerà-Brunet
- Instituto de Microelectrónica de Barcelona (IMB-CNM, CSIC), Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain
| | - M V Sanchez-Vives
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- ICREA, Barcelona, Spain
| |
Collapse
|
10
|
Dreier JP, Lemale CL, Horst V, Major S, Kola V, Schoknecht K, Scheel M, Hartings JA, Vajkoczy P, Wolf S, Woitzik J, Hecht N. Similarities in the Electrographic Patterns of Delayed Cerebral Infarction and Brain Death After Aneurysmal and Traumatic Subarachnoid Hemorrhage. Transl Stroke Res 2024:10.1007/s12975-024-01237-w. [PMID: 38396252 DOI: 10.1007/s12975-024-01237-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024]
Abstract
While subarachnoid hemorrhage is the second most common hemorrhagic stroke in epidemiologic studies, the recent DISCHARGE-1 trial has shown that in reality, three-quarters of focal brain damage after subarachnoid hemorrhage is ischemic. Two-fifths of these ischemic infarctions occur early and three-fifths are delayed. The vast majority are cortical infarcts whose pathomorphology corresponds to anemic infarcts. Therefore, we propose in this review that subarachnoid hemorrhage as an ischemic-hemorrhagic stroke is rather a third, separate entity in addition to purely ischemic or hemorrhagic strokes. Cumulative focal brain damage, determined by neuroimaging after the first 2 weeks, is the strongest known predictor of patient outcome half a year after the initial hemorrhage. Because of the unique ability to implant neuromonitoring probes at the brain surface before stroke onset and to perform longitudinal MRI scans before and after stroke, delayed cerebral ischemia is currently the stroke variant in humans whose pathophysiological details are by far the best characterized. Optoelectrodes located directly over newly developing delayed infarcts have shown that, as mechanistic correlates of infarct development, spreading depolarizations trigger (1) spreading ischemia, (2) severe hypoxia, (3) persistent activity depression, and (4) transition from clustered spreading depolarizations to a negative ultraslow potential. Furthermore, traumatic brain injury and subarachnoid hemorrhage are the second and third most common etiologies of brain death during continued systemic circulation. Here, we use examples to illustrate that although the pathophysiological cascades associated with brain death are global, they closely resemble the local cascades associated with the development of delayed cerebral infarcts.
Collapse
Affiliation(s)
- Jens P Dreier
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, 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.
| | - Coline L Lemale
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, 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
| | - Viktor Horst
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Neuropathology, 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, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, 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
| | - Vasilis Kola
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Karl Schoknecht
- Medical Faculty, Carl Ludwig Institute for Physiology, University of Leipzig, Leipzig, Germany
| | - Michael Scheel
- Department of Neuroradiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jed A Hartings
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Stefan Wolf
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Johannes Woitzik
- Department of Neurosurgery, Evangelisches Krankenhaus Oldenburg, University of Oldenburg, Oldenburg, Germany
| | - Nils Hecht
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
11
|
Motawi TK, Sadik NAH, Shaker OG, Ghaleb MMH, Elbaz EM. Expression, Functional Polymorphism, and Diagnostic Values of MIAT rs2331291 and H19 rs217727 Long Non-Coding RNAs in Cerebral Ischemic Stroke Egyptian Patients. Int J Mol Sci 2024; 25:842. [PMID: 38255915 PMCID: PMC10815378 DOI: 10.3390/ijms25020842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/24/2023] [Accepted: 12/30/2023] [Indexed: 01/24/2024] Open
Abstract
Cerebral ischemic stroke (CIS) is a severe cerebral vascular event. This research aimed to evaluate the role of single-nucleotide polymorphisms (SNPs) of the lncRNAs MIAT rs2331291 and H19 rs217727 and epigenetic methylation in the expression patterns of serum lncRNA H19 in CIS Egyptian patients. It included 80 CIS cases and 40 healthy subjects. Serum MIAT expression levels decreased, whereas serum H19 expression levels increased among CIS compared to controls. For MIAT rs2331291, there were significant differences in the genotypic and allelic frequencies between the CIS and healthy subjects at p = 0.02 and p = 0.0001, respectively. Our findings illustrated a significantly increased MIAT T/T genotype frequency in hypertensive CIS compared to non-hypertensive CIS at p = 0.004. However, H19 rs217727 gene frequency C/C was not significantly higher in non-hypertensive CIS than in hypertensive CIS. The methylation of the H19 gene promoter was significantly higher in CIS patients compared to healthy subjects. The level of MIAT was positively correlated with serum H19 in CIS. Receiver operating characteristics (ROC) analysis revealed that serum MIAT and H19 have a high diagnostic potential for distinguishing CIS subjects from healthy ones. In conclusion, the MIAT-rs2331291 polymorphism might serve as a novel potential indicator of CIS.
Collapse
Affiliation(s)
- Tarek K. Motawi
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | | | - Olfat G. Shaker
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | | | - Eman M. Elbaz
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
12
|
Cancino-Fuentes N, Manasanch A, Covelo J, Suarez-Perez A, Fernandez E, Matsoukis S, Guger C, Illa X, Guimerà-Brunet A, Sanchez-Vives MV. Recording physiological and pathological cortical activity and exogenous electric fields using graphene microtransistor arrays in vitro. NANOSCALE 2024; 16:664-677. [PMID: 38100059 DOI: 10.1039/d3nr03842d] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Graphene-based solution-gated field-effect transistors (gSGFETs) allow the quantification of the brain's full-band signal. Extracellular alternating current (AC) signals include local field potentials (LFP, population activity within a reach of hundreds of micrometers), multiunit activity (MUA), and ultimately single units. Direct current (DC) potentials are slow brain signals with a frequency under 0.1 Hz, and commonly filtered out by conventional AC amplifiers. This component conveys information about what has been referred to as "infraslow" activity. We used gSGFET arrays to record full-band patterns from both physiological and pathological activity generated by the cerebral cortex. To this end, we used an in vitro preparation of cerebral cortex that generates spontaneous rhythmic activity, such as that occurring in slow wave sleep. This examination extended to experimentally induced pathological activities, including epileptiform discharges and cortical spreading depression. Validation of recordings obtained via gSGFETs, including both AC and DC components, was accomplished by cross-referencing with well-established technologies, thereby quantifying these components across different activity patterns. We then explored an additional gSGFET potential application, which is the measure of externally induced electric fields such as those used in therapeutic neuromodulation in humans. Finally, we tested the gSGFETs in human cortical slices obtained intrasurgically. In conclusion, this study offers a comprehensive characterization of gSGFETs for brain recordings, with a focus on potential clinical applications of this emerging technology.
Collapse
Affiliation(s)
| | - Arnau Manasanch
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Joana Covelo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Alex Suarez-Perez
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | | | - Stratis Matsoukis
- g.tec medical engineering, Schiedlberg, Austria
- Institute of Computational Perception, Johannes Kepler University, Linz, Austria
| | | | - Xavi Illa
- Instituto de Microelectrónica de Barcelona (IMB-CNM, CSIC), Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain
| | - Anton Guimerà-Brunet
- Instituto de Microelectrónica de Barcelona (IMB-CNM, CSIC), Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain
| | - Maria V Sanchez-Vives
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- ICREA, Barcelona, Spain
| |
Collapse
|
13
|
Brunner C, Denis NL, Gertz K, Grillet M, Montaldo G, Endres M, Urban A. Brain-wide continuous functional ultrasound imaging for real-time monitoring of hemodynamics during ischemic stroke. J Cereb Blood Flow Metab 2024; 44:6-18. [PMID: 37503862 PMCID: PMC10905631 DOI: 10.1177/0271678x231191600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023]
Abstract
Ischemic stroke occurs abruptly causing sudden neurologic deficits, and therefore, very little is known about hemodynamic perturbations in the brain immediately after stroke onset. Here, functional ultrasound imaging was used to monitor variations in relative cerebral blood volume (rCBV) compared to baseline. rCBV levels were analyzed brain-wide and continuously at high spatiotemporal resolution (100 μm, 2 Hz) until 70mins after stroke onset in rats. We compared two stroke models, with either a permanent occlusion of the middle cerebral artery (MCAo) or a tandem occlusion of both the common carotid and middle cerebral arteries (CCAo + MCAo). We observed a typical hemodynamic pattern, including a quick drop of the rCBV after MCAo, followed by spontaneous reperfusion of several brain regions located in the vicinity of the ischemic core. The severity and location of the ischemia were variable within groups. On average, the severity of the ischemia was in good agreement with the lesion volume (24 hrs after stroke) for MCAo group, while larger for the CCAo + MCAo model. For both groups, we observed that infarcts extended to initially non-ischemic regions located rostrally to the ischemic core. These regions strongly colocalize with the origin of transient hemodynamic events associated with spreading depolarizations.
Collapse
Affiliation(s)
- Clément Brunner
- Neuro-Electronics Research Flanders, Leuven, Belgium
- Vlaams Instituut voor Biotechnologie, Leuven, Belgium
- Interuniversity Microelectronics Centre, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Nielsen Lagumersindez Denis
- Department of Neurology and Center for Stroke Research Berlin, Charité-Universitätsmedizin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Karen Gertz
- Department of Neurology and Center for Stroke Research Berlin, Charité-Universitätsmedizin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Micheline Grillet
- Neuro-Electronics Research Flanders, Leuven, Belgium
- Vlaams Instituut voor Biotechnologie, Leuven, Belgium
- Interuniversity Microelectronics Centre, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Gabriel Montaldo
- Neuro-Electronics Research Flanders, Leuven, Belgium
- Vlaams Instituut voor Biotechnologie, Leuven, Belgium
- Interuniversity Microelectronics Centre, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Matthias Endres
- Department of Neurology and Center for Stroke Research Berlin, Charité-Universitätsmedizin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Alan Urban
- Neuro-Electronics Research Flanders, Leuven, Belgium
- Vlaams Instituut voor Biotechnologie, Leuven, Belgium
- Interuniversity Microelectronics Centre, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
14
|
van Hameren G, Muradov J, Minarik A, Aboghazleh R, Orr S, Cort S, Andrews K, McKenna C, Pham NT, MacLean MA, Friedman A. Mitochondrial dysfunction underlies impaired neurovascular coupling following traumatic brain injury. Neurobiol Dis 2023; 186:106269. [PMID: 37619791 DOI: 10.1016/j.nbd.2023.106269] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/26/2023] Open
Abstract
Traumatic brain injury (TBI) involves an acute injury (primary damage), which may evolve in the hours to days after impact (secondary damage). Seizures and cortical spreading depolarization (CSD) are metabolically demanding processes that may worsen secondary brain injury. Metabolic stress has been associated with mitochondrial dysfunction, including impaired calcium homeostasis, reduced ATP production, and elevated ROS production. However, the association between mitochondrial impairment and vascular function after TBI is poorly understood. Here, we explored this association using a rodent closed head injury model. CSD is associated with neurobehavioral decline after TBI. Craniotomy was performed to elicit CSD via electrical stimulation or to induce seizures via 4-aminopyridine application. We measured vascular dysfunction following CSDs and seizures in TBI animals using laser doppler flowmetry. We observed a more profound reduction in local cortical blood flow in TBI animals compared to healthy controls. CSD resulted in mitochondrial dysfunction and pathological signs of increased oxidative stress adjacent to the vasculature. We explored these findings further using electron microscopy and found that TBI and CSDs resulted in vascular morphological changes and mitochondrial cristae damage in astrocytes, pericytes and endothelial cells. Overall, we provide evidence that CSDs induce mitochondrial dysfunction, impaired cortical blood flow, and neurobehavioral deficits in the setting of TBI.
Collapse
Affiliation(s)
- Gerben van Hameren
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, NS B3H 4H7, Halifax, Canada.
| | - Jamil Muradov
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, NS B3H 4H7, Halifax, Canada
| | - Anna Minarik
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, NS B3H 4H7, Halifax, Canada
| | - Refat Aboghazleh
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, NS B3H 4H7, Halifax, Canada; Department of Basic Medical Sciences, Faculty of Medicine, Al-Balqa Applied University, Al-Salt, Jordan
| | - Sophie Orr
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, NS B3H 4H7, Halifax, Canada
| | - Shayna Cort
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, NS B3H 4H7, Halifax, Canada
| | - Keiran Andrews
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, NS B3H 4H7, Halifax, Canada
| | - Caitlin McKenna
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, NS B3H 4H7, Halifax, Canada
| | - Nga Thy Pham
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, NS B3H 4H7, Halifax, Canada
| | - Mark A MacLean
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, NS B3H 4H7, Halifax, Canada; Division of Neurosurgery, Department of Surgery, Dalhousie University, NS B3H 3A7, Halifax, Canada
| | - Alon Friedman
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, NS B3H 4H7, Halifax, Canada; Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
15
|
Sanchez-Porras R, Ramírez-Cuapio FL, Hecht N, Seule M, Díaz-Peregrino R, Unterberg A, Woitzik J, Dreier JP, Sakowitz OW, Santos E. Cerebrovascular Pressure Reactivity According to Long-Pressure Reactivity Index During Spreading Depolarizations in Aneurysmal Subarachnoid Hemorrhage. Neurocrit Care 2023; 39:135-144. [PMID: 36697998 PMCID: PMC10499750 DOI: 10.1007/s12028-022-01669-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/19/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Spreading depolarization (SD) has been linked to the impairment of neurovascular coupling. However, the association between SD occurrence and cerebrovascular pressure reactivity as a surrogate of cerebral autoregulation (CA) remains unclear. Therefore, we analyzed CA using the long-pressure reactivity index (L-PRx) during SDs in patients with aneurysmal subarachnoid hemorrhage (aSAH). METHODS A retrospective study of patients with aSAH who were recruited at two centers, Heidelberg (HD) and Berlin (BE), was performed. Continuous monitoring of mean arterial pressure (MAP) and intracranial pressure (ICP) was recorded. ICP was measured using an intraparenchymal probe in HD patients and was measure in BE patients through external ventricular drainage. Electrocorticographic (ECoG) activity was continuously recorded between 3 and 13 days after hemorrhage. Autoregulation according to L-PRx was calculated as a moving linear Pearson's correlation of 20-min averages of MAP and ICP. For every identified SD, 60-min intervals of L-PRx were averaged, plotted, and analyzed depending on SD occurrence. Random L-PRx recording periods without SDs served as the control. RESULTS A total of 19 patients (HD n = 14, BE n = 5, mean age 50.4 years, 9 female patients) were monitored for a mean duration of 230.4 h (range 96-360, STD ± 69.6 h), during which ECoG recordings revealed a total number of 277 SDs. Of these, 184 represented a single SD, and 93 SDs presented in clusters. In HD patients, mean L-PRx values were 0.12 (95% confidence interval [CI] 0.11-0.13) during SDs and 0.07 (95% CI 0.06-0.08) during control periods (p < 0.001). Similarly, in BE patients, a higher L-PRx value of 0.11 (95% CI 0.11-0.12) was detected during SDs than that during control periods (0.08, 95% CI 0.07-0.09; p < 0.001). In a more detailed analysis, CA changes registered through an intraparenchymal probe (HD patients) revealed that clustered SD periods were characterized by signs of more severely impaired CA (L-PRx during SD in clusters: 0.23 [95% CI 0.20-0.25]; single SD: 0.09 [95% CI 0.08-0.10]; control periods: 0.07 [95% CI 0.06-0.08]; p < 0.001). This group also showed significant increases in ICP during SDs in clusters compared with single SD and control periods. CONCLUSIONS Neuromonitoring for simultaneous assessment of cerebrovascular pressure reactivity using 20-min averages of MAP and ICP measured by L-PRx during SD events is feasible. SD occurrence was associated with significant increases in L-PRx values indicative of CA disturbances. An impaired CA was found during SD in clusters when using an intraparenchymal probe. This preliminary study validates the use of cerebrovascular reactivity indices to evaluate CA disturbances during SDs. Our results warrant further investigation in larger prospective patient cohorts.
Collapse
Affiliation(s)
- Renan Sanchez-Porras
- Department of Neurosurgery, Heidelberg University Hospital, Ruprecht Karls University of Heidelberg, Heidelberg, Germany
- Department of Neurosurgery, Evangelisches Krankenhaus Oldenburg, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| | - Francisco L Ramírez-Cuapio
- Department of Neurosurgery, Heidelberg University Hospital, Ruprecht Karls University of Heidelberg, Heidelberg, 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
- Center for Stroke Research Berlin, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Martin Seule
- Department of Neurosurgery, Heidelberg University Hospital, Ruprecht Karls University of Heidelberg, Heidelberg, Germany
- Department of Neurosurgery, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Roberto Díaz-Peregrino
- Department of Neurosurgery, Heidelberg University Hospital, Ruprecht Karls University of Heidelberg, Heidelberg, Germany
| | - Andreas Unterberg
- Department of Neurosurgery, Heidelberg University Hospital, Ruprecht Karls University of Heidelberg, Heidelberg, Germany
| | - Johannes Woitzik
- Department of Neurosurgery, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Neurosurgery, Evangelisches Krankenhaus Oldenburg, Carl von Ossietzky 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 Neurology, 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
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - Oliver W Sakowitz
- Department of Neurosurgery, Heidelberg University Hospital, Ruprecht Karls University of Heidelberg, Heidelberg, Germany
- Neurosurgery Center Ludwigsburg-Heilbronn, RKH Klinikum Ludwigsburg, Ludwigsburg, Germany
| | - Edgar Santos
- Department of Neurosurgery, Heidelberg University Hospital, Ruprecht Karls University of Heidelberg, Heidelberg, Germany.
- Department of Neurosurgery, Evangelisches Krankenhaus Oldenburg, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany.
| |
Collapse
|
16
|
Mingazov B, Vinokurova D, Zakharov A, Khazipov R. Comparative Study of Terminal Cortical Potentials Using Iridium and Ag/AgCl Electrodes. Int J Mol Sci 2023; 24:10769. [PMID: 37445945 DOI: 10.3390/ijms241310769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Brain ischemia induces slow voltage shifts in the cerebral cortex, including waves of spreading depolarization (SD) and negative ultraslow potentials (NUPs), which are considered as brain injury markers. However, different electrode materials and locations yield variable SD and NUP features. Here, we compared terminal cortical events during isoflurane or sevoflurane euthanasia using intracortical linear iridium electrode arrays and Ag/AgCl-based electrodes in the rat somatosensory cortex. Inhalation of anesthetics caused respiratory arrest, associated with hyperpolarization and followed by SD and NUP on both Ir and Ag electrodes. Ag-NUPs were bell shaped and waned within half an hour after death. Ir-NUPs were biphasic, with the early fast phase corresponding to Ag-NUP, and the late absent on Ag electrodes, phase of a progressive depolarizing voltage shift reaching -100 mV by two hours after death. In addition, late Ir-NUPs were more ample in the deep layers than at the cortical surface. Thus, intracortical Ag and Ir electrodes reliably assess early manifestations of terminal brain injury including hyperpolarization, SD and the early phase of NUP, while the late, giant amplitude phase of NUP, which is present only on Ir electrodes, is probably related to the sensitivity of Ir electrodes to a yet unidentified factor related to brain death.
Collapse
Affiliation(s)
- Bulat Mingazov
- Laboratory of Neurobiology, Kazan Federal University, Kazan 420008, Russia
| | - Daria Vinokurova
- Laboratory of Neurobiology, Kazan Federal University, Kazan 420008, Russia
| | - Andrei Zakharov
- Laboratory of Neurobiology, Kazan Federal University, Kazan 420008, Russia
- Department of Physiology, Kazan State Medical University, Kazan 420012, Russia
| | - Roustem Khazipov
- Laboratory of Neurobiology, Kazan Federal University, Kazan 420008, Russia
- Institut de Neurobiologie de la Méditerranée (Inserm U1249), Aix-Marseille Université, 13273 Marseille, France
| |
Collapse
|
17
|
Nash C, Powell K, Lynch DG, Hartings JA, Li C. Nonpharmacological modulation of cortical spreading depolarization. Life Sci 2023:121833. [PMID: 37302793 DOI: 10.1016/j.lfs.2023.121833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/01/2023] [Accepted: 06/03/2023] [Indexed: 06/13/2023]
Abstract
AIMS Cortical spreading depolarization (CSD) is a wave of pathologic neuronal dysfunction that spreads through cerebral gray matter, causing neurologic disturbance in migraine and promoting lesion development in acute brain injury. Pharmacologic interventions have been found to be effective in migraine with aura, but their efficacy in acutely injured brains may be limited. This necessitates the assessment of possible adjunctive treatments, such as nonpharmacologic methods. This review aims to summarize currently available nonpharmacological techniques for modulating CSDs, present their mechanisms of action, and provide insight and future directions for CSD treatment. MAIN METHODS A systematic literature review was performed, generating 22 articles across 3 decades. Relevant data is broken down according to method of treatment. KEY FINDINGS Both pharmacologic and nonpharmacologic interventions can mitigate the pathological impact of CSDs via shared molecular mechanisms, including modulating K+/Ca2+/Na+/Cl- ion channels and NMDA, GABAA, serotonin, and CGRP ligand-based receptors and decreasing microglial activation. Preclinical evidence suggests that nonpharmacologic interventions, including neuromodulation, physical exercise, therapeutic hypothermia, and lifestyle changes can also target unique mechanisms, such as increasing adrenergic tone and myelination and modulating membrane fluidity, which may lend broader modulatory effects. Collectively, these mechanisms increase the electrical initiation threshold, increase CSD latency, slow CSD velocity, and decrease CSD amplitude and duration. SIGNIFICANCE Given the harmful consequences of CSDs, limitations of current pharmacological interventions to inhibit CSDs in acutely injured brains, and translational potentials of nonpharmacologic interventions to modulate CSDs, further assessment of nonpharmacologic modalities and their mechanisms to mitigate CSD-related neurologic dysfunction is warranted.
Collapse
Affiliation(s)
- Christine Nash
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Barnard College, New York, NY, USA
| | - Keren Powell
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Daniel G Lynch
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Jed A Hartings
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH, USA
| | - Chunyan Li
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA; Department of Neurosurgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
18
|
Mehra A, Gomez F, Bischof H, Diedrich D, Laudanski K. Cortical Spreading Depolarization and Delayed Cerebral Ischemia; Rethinking Secondary Neurological Injury in Subarachnoid Hemorrhage. Int J Mol Sci 2023; 24:9883. [PMID: 37373029 DOI: 10.3390/ijms24129883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Poor outcomes in Subarachnoid Hemorrhage (SAH) are in part due to a unique form of secondary neurological injury known as Delayed Cerebral Ischemia (DCI). DCI is characterized by new neurological insults that continue to occur beyond 72 h after the onset of the hemorrhage. Historically, it was thought to be a consequence of hypoperfusion in the setting of vasospasm. However, DCI was found to occur even in the absence of radiographic evidence of vasospasm. More recent evidence indicates that catastrophic ionic disruptions known as Cortical Spreading Depolarizations (CSD) may be the culprits of DCI. CSDs occur in otherwise healthy brain tissue even without demonstrable vasospasm. Furthermore, CSDs often trigger a complex interplay of neuroinflammation, microthrombi formation, and vasoconstriction. CSDs may therefore represent measurable and modifiable prognostic factors in the prevention and treatment of DCI. Although Ketamine and Nimodipine have shown promise in the treatment and prevention of CSDs in SAH, further research is needed to determine the therapeutic potential of these as well as other agents.
Collapse
Affiliation(s)
- Ashir Mehra
- Department of Neurology, University of Missouri, Columbia, MO 65212, USA
| | - Francisco Gomez
- Department of Neurology, University of Missouri, Columbia, MO 65212, USA
| | - Holly Bischof
- Penn Presbyterian Medical Center, Philadelphia, PA 19104, USA
| | - Daniel Diedrich
- Department of Anesthesiology and Perioperative Care, Mayo Clinic, Rochester, MN 55905, USA
| | - Krzysztof Laudanski
- Department of Anesthesiology and Perioperative Care, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
19
|
Luo Y, Chen J, Huang HY, Lam ESY, Wong GKC. Narrative review of roles of astrocytes in subarachnoid hemorrhage. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:302. [PMID: 37181334 PMCID: PMC10170286 DOI: 10.21037/atm-22-5486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 03/16/2023] [Indexed: 03/28/2023]
Abstract
Background and Objective Astrocytes play an important role in healthy brain function, including the development and maintenance of blood-brain barrier (BBB), structural support, brain homeostasis, neurovascular coupling and secretion of neuroprotective factors. Reactive astrocytes participate in various pathophysiology after subarachnoid hemorrhage (SAH) including neuroinflammation, glutamate toxicity, brain edema, vasospasm, BBB disruption, cortical spreading depolarization (SD). Methods We searched PubMed up to 31 May, 2022 and evaluated the articles for screening and inclusion for subsequent systemic review. We found 198 articles with the searched terms. After exclusion based on the selection criteria, we selected 30 articles to start the systemic review. Key Content and Findings We summarized the response of astrocytes induced by SAH. Astrocytes are critical for brain edema formation, BBB reconstruction and neuroprotection in the acute stage of SAH. Astrocytes clear extracellular glutamate by increasing the uptake of glutamate and Na+/K+ ATPase activity after SAH. Neurotrophic factors released by astrocytes contribute to neurological recovery after SAH. Meanwhile, Astrocytes also form glial scars which hinder axon regeneration, produce proinflammatory cytokines, free radicals, and neurotoxic molecules. Conclusions Preclinical studies showed that therapeutic targeting the astrocytes response could have a beneficial effect in ameliorating neuronal injury and cognitive impairment after SAH. Clinical trials and preclinical animal studies are still urgently needed in order to determine where astrocytes stand in various pathway of brain damage and repair after SAH and, above all, to develop therapeutic approaches which benefit patient outcomes.
Collapse
Affiliation(s)
- Yujie Luo
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, the Chinese University of Hong Kong, Hong Kong, China
| | - Junfan Chen
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, the Chinese University of Hong Kong, Hong Kong, China
| | - Hiu Yin Huang
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, the Chinese University of Hong Kong, Hong Kong, China
| | - Erica Sin Yu Lam
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, the Chinese University of Hong Kong, Hong Kong, China
| | - George Kwok-Chu Wong
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, the Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
20
|
Kang EJ, Prager O, Lublinsky S, Oliveira-Ferreira AI, Reiffurth C, Major S, Müller DN, Friedman A, Dreier JP. Stroke-prone salt-sensitive spontaneously hypertensive rats show higher susceptibility to spreading depolarization (SD) and altered hemodynamic responses to SD. J Cereb Blood Flow Metab 2023; 43:210-230. [PMID: 36329390 PMCID: PMC9903222 DOI: 10.1177/0271678x221135085] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Spreading depolarization (SD) occurs in a plethora of clinical conditions including migraine aura, delayed ischemia after subarachnoid hemorrhage and malignant hemispheric stroke. It describes waves of near-breakdown of ion homeostasis, particularly Na+ homeostasis in brain gray matter. SD induces tone alterations in resistance vessels, causing either hyperperfusion in healthy tissue; or hypoperfusion (inverse hemodynamic response = spreading ischemia) in tissue at risk. Observations from mice with genetic dysfunction of the ATP1A2-encoded α2-isoform of Na+/K+-ATPase (α2NaKA) suggest a mechanistic link between (1) SD, (2) vascular dysfunction, and (3) salt-sensitive hypertension via α2NaKA. Thus, α2NaKA-dysfunctional mice are more susceptible to SD and show a shift toward more inverse hemodynamic responses. α2NaKA-dysfunctional patients suffer from familial hemiplegic migraine type 2, a Mendelian model disease of SD. α2NaKA-dysfunctional mice are also a genetic model of salt-sensitive hypertension. To determine whether SD thresholds and hemodynamic responses are also altered in other genetic models of salt-sensitive hypertension, we examined these variables in stroke-prone spontaneously hypertensive rats (SHRsp). Compared with Wistar Kyoto control rats, we found in SHRsp that electrical SD threshold was significantly reduced, propagation speed was increased, and inverse hemodynamic responses were prolonged. These results may have relevance to both migraine with aura and stroke.
Collapse
Affiliation(s)
- Eun-Jeung Kang
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ofer Prager
- Department of Physiology & Cell Biology, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Department of Cognitive & Brain Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Svetlana Lublinsky
- Department of Cognitive & Brain Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - 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
| | - Clemens Reiffurth
- 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
| | - Dominik N Müller
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Alon Friedman
- Department of Physiology & Cell Biology, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Department of Cognitive & Brain Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Department of Medical Neuroscience and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
| | - 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
|
21
|
Törteli A, Tóth R, Berger S, Samardzic S, Bari F, Menyhárt Á, Farkas E. Spreading depolarization causes reperfusion failure after cerebral ischemia. J Cereb Blood Flow Metab 2023; 43:655-664. [PMID: 36703609 PMCID: PMC10108181 DOI: 10.1177/0271678x231153745] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Despite successful recanalization, reperfusion failure associated with poor neurological outcomes develops in half of treated stroke patients. We explore here whether spreading depolarization (SD) is a predictor of reperfusion failure. Global forebrain ischemia/reperfusion was induced in male and female C57BL/6 mice (n = 57). SD and cerebral blood flow (CBF) changes were visualized with transcranial intrinsic optical signal and laser speckle contrast imaging. To block SD, MK801 was applied (n = 26). Neurological deficit, circle of Willis (CoW) anatomy and neuronal injury were evaluated 24 hours later. SD emerged after ischemia onset in one or both hemispheres under a perfusion threshold (CBF drop to 21.1 ± 4.6 vs. 33.6 ± 4.4%, SD vs. no SD). The failure of later reperfusion (44.4 ± 12.5%) was invariably linked to previous SD. In contrast, reperfusion was adequate (98.9 ± 7.4%) in hemispheres devoid of SD. Absence of the P1 segment of the posterior cerebral artery in the CoW favored SD occurrence and reperfusion failure. SD occurrence and reperfusion failure were associated with poor neurologic function, and neuronal necrosis 24 hours after ischemia. The inhibition of SD significantly improved reperfusion. SD occurrence during ischemia impairs later reperfusion, prognosticating poor neurological outcomes. The increased likelihood of SD occurrence is predicted by inadequate collaterals.
Collapse
Affiliation(s)
- Anna Törteli
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary.,Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Réka Tóth
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary.,Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Sarah Berger
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Sarah Samardzic
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ferenc Bari
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ákos Menyhárt
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary.,Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Eszter Farkas
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary.,Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| |
Collapse
|
22
|
Luckl J, Baker W, Boda K, Emri M, Yodh AG, Greenberg JH. Oxyhemoglobin and Cerebral Blood Flow Transients Detect Infarction in Rat Focal Brain Ischemia. Neuroscience 2023; 509:132-144. [PMID: 36460221 PMCID: PMC9852213 DOI: 10.1016/j.neuroscience.2022.11.028] [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: 08/21/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 11/30/2022]
Abstract
Spreading depolarizations (SD) refer to the near-complete depolarization of neurons that is associated with brain injuries such as ischemic stroke. The present gold standard for SD monitoring in humans is invasive electrocorticography (ECoG). A promising non-invasive alternative to ECoG is diffuse optical monitoring of SD-related flow and hemoglobin transients. To investigate the clinical utility of flow and hemoglobin transients, we analyzed their association with infarction in rat focal brain ischemia. Optical images of flow, oxy-hemoglobin, and deoxy-hemoglobin were continuously acquired with Laser Speckle and Optical Intrinsic Signal imaging for 2 h after photochemically induced distal middle cerebral artery occlusion in Sprague-Dawley rats (n = 10). Imaging was performed through a 6 × 6 mm window centered 3 mm posterior and 4 mm lateral to Bregma. Rats were sacrificed after 24 h, and the brain slices were stained for assessment of infarction. We mapped the infarcted area onto the imaging data and used nine circular regions of interest (ROI) to distinguish infarcted from non-infarcted tissue. Transients propagating through each ROI were characterized with six parameters (negative, positive, and total amplitude; negative and positive slope; duration). Transients were also classified into three morphology types (positive monophasic, biphasic, negative monophasic). Flow transient morphology, positive amplitude, positive slope, and total amplitude were all strongly associated with infarction (p < 0.001). Associations with infarction were also observed for oxy-hemoglobin morphology, oxy-hemoglobin positive amplitude and slope, and deoxy-hemoglobin positive slope and duration (all p < 0.01). These results suggest that flow and hemoglobin transients accompanying SD have value for detecting infarction.
Collapse
Affiliation(s)
- Janos Luckl
- Department of Neurology, University of Pennsylvania, Philadelphia, USA; Department of Neurology, University of Szeged, Szeged, Hungary; Department of Medical Physics and Informatics, Szeged, Hungary
| | - Wesley Baker
- Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, USA; Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, USA
| | - Krisztina Boda
- Department of Medical Physics and Informatics, Szeged, Hungary
| | - Miklos Emri
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Arjun G Yodh
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, USA
| | - Joel H Greenberg
- Department of Neurology, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
23
|
Meinert F, Lemâle CL, Major S, Helgers SOA, Dömer P, Mencke R, Bergold MN, Dreier JP, Hecht N, Woitzik J. Less-invasive subdural electrocorticography for investigation of spreading depolarizations in patients with subarachnoid hemorrhage. Front Neurol 2023; 13:1091987. [PMID: 36686541 PMCID: PMC9849676 DOI: 10.3389/fneur.2022.1091987] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction Wyler-strip electrodes for subdural electrocorticography (ECoG) are the gold standard for continuous bed-side monitoring of pathological cortical network events, such as spreading depolarizations (SD) and electrographic seizures. Recently, SD associated parameters were shown to be (1) a marker of early brain damage after aneurysmal subarachnoid hemorrhage (aSAH), (2) the strongest real-time predictor of delayed cerebral ischemia currently known, and (3) the second strongest predictor of patient outcome at 7 months. The strongest predictor of patient outcome at 7 months was focal brain damage segmented on neuroimaging 2 weeks after the initial hemorrhage, whereas the initial focal brain damage was inferior to the SD variables as a predictor for patient outcome. However, the implantation of Wyler-strip electrodes typically requires either a craniotomy or an enlarged burr hole. Neuromonitoring via an enlarged burr hole has been performed in only about 10% of the total patients monitored. Methods In the present pilot study, we investigated the feasibility of ECoG monitoring via a less invasive burrhole approach using a Spencer-type electrode array, which was implanted subdurally rather than in the depth of the parenchyma. Seven aSAH patients requiring extraventricular drainage (EVD) were included. For electrode placement, the burr hole over which the EVD was simultaneously placed, was used in all cases. After electrode implantation, continuous, direct current (DC)/alternating current (AC)-ECoG monitoring was performed at bedside in our Neurointensive Care unit. ECoGs were analyzed following the recommendations of the Co-Operative Studies on Brain Injury Depolarizations (COSBID). Results Subdural Spencer-type electrode arrays permitted high-quality ECoG recording. During a cumulative monitoring period of 1,194.5 hours and a median monitoring period of 201.3 (interquartile range: 126.1-209.4) hours per patient, 84 SDs were identified. Numbers of SDs, isoelectric SDs and clustered SDs per recording day, and peak total SD-induced depression duration of a recording day were not significantly different from the previously reported results of the prospective, observational, multicenter, cohort, diagnostic phase III trial, DISCHARGE-1. No adverse events related to electrode implantation were noted. Discussion In conclusion, our findings support the safety and feasibility of less-invasive subdural electrode implantation for reliable SD-monitoring.
Collapse
Affiliation(s)
- Franziska Meinert
- Department of Neurosurgery, Carl von Ossietzky University Oldenburg, Oldenburg, Germany,Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Coline L. Lemâle
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 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,Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 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, 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,Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Simeon O. A. Helgers
- Department of Neurosurgery, Carl von Ossietzky University Oldenburg, Oldenburg, Germany,Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Patrick Dömer
- Department of Neurosurgery, Carl von Ossietzky University Oldenburg, Oldenburg, Germany,Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Rik Mencke
- Department of Neurosurgery, Carl von Ossietzky University Oldenburg, Oldenburg, Germany,Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Martin N. Bergold
- Department of Anaesthesiology and Intensive Care Medicine, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Jens P. Dreier
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany,Department of Neurology, 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, Berlin Institute of Health, Berlin, Germany,Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany,Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - Nils Hecht
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany,Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Johannes Woitzik
- Department of Neurosurgery, Carl von Ossietzky University Oldenburg, Oldenburg, Germany,Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany,*Correspondence: Johannes Woitzik ✉
| |
Collapse
|
24
|
Álvarez-Merz I, Fomitcheva IV, Sword J, Hernández-Guijo JM, Solís JM, Kirov SA. Novel mechanism of hypoxic neuronal injury mediated by non-excitatory amino acids and astroglial swelling. Glia 2022; 70:2108-2130. [PMID: 35802030 PMCID: PMC9474671 DOI: 10.1002/glia.24241] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/14/2022] [Accepted: 06/24/2022] [Indexed: 11/08/2022]
Abstract
In ischemic stroke and post-traumatic brain injury (TBI), blood-brain barrier disruption leads to leaking plasma amino acids (AA) into cerebral parenchyma. Bleeding in hemorrhagic stroke and TBI also release plasma AA. Although excitotoxic AA were extensively studied, little is known about non-excitatory AA during hypoxic injury. Hypoxia-induced synaptic depression in hippocampal slices becomes irreversible with non-excitatory AA, alongside their intracellular accumulation and increased tissue electrical resistance. Four non-excitatory AA (l-alanine, glycine, l-glutamine, l-serine: AGQS) at plasmatic concentrations were applied to slices from mice expressing EGFP in pyramidal neurons or astrocytes during normoxia or hypoxia. Two-photon imaging, light transmittance (LT) changes, and electrophysiological field recordings followed by electron microscopy in hippocampal CA1 st. radiatum were used to monitor synaptic function concurrently with cellular swelling and injury. During normoxia, AGQS-induced increase in LT was due to astroglial but not neuronal swelling. LT raise during hypoxia and AGQS manifested astroglial and neuronal swelling accompanied by a permanent loss of synaptic transmission and irreversible dendritic beading, signifying acute damage. Neuronal injury was not triggered by spreading depolarization which did not occur in our experiments. Hypoxia without AGQS did not cause cell swelling, leaving dendrites intact. Inhibition of NMDA receptors prevented neuronal damage and irreversible loss of synaptic function. Deleterious effects of AGQS during hypoxia were prevented by alanine-serine-cysteine transporters (ASCT2) and volume-regulated anion channels (VRAC) blockers. Our findings suggest that astroglial swelling induced by accumulation of non-excitatory AA and release of excitotoxins through antiporters and VRAC may exacerbate the hypoxia-induced neuronal injury.
Collapse
Affiliation(s)
- Iris Álvarez-Merz
- Dept. de Farmacología y Terapéutica, ITH, Facultad de Medicina, Universidad Autónoma de Madrid, IRYCIS, 28029 Madrid, Spain
- Servicio de Neurobiología-Investigación, Hospital Universitario Ramón y Cajal, IRYCIS, 28034 Madrid, Spain
- Dept. of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, USA
| | - Ioulia V. Fomitcheva
- Dept. of Neurosurgery, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, USA
| | - Jeremy Sword
- Dept. of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, USA
| | - Jesús M. Hernández-Guijo
- Dept. de Farmacología y Terapéutica, ITH, Facultad de Medicina, Universidad Autónoma de Madrid, IRYCIS, 28029 Madrid, Spain
| | - José M. Solís
- Servicio de Neurobiología-Investigación, Hospital Universitario Ramón y Cajal, IRYCIS, 28034 Madrid, Spain
| | - Sergei A. Kirov
- Dept. of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, USA
| |
Collapse
|
25
|
Vinokurova D, Zakharov A, Chernova K, Burkhanova-Zakirova G, Horst V, Lemale CL, Dreier JP, Khazipov R. Depth-profile of impairments in endothelin-1 - induced focal cortical ischemia. J Cereb Blood Flow Metab 2022; 42:1944-1960. [PMID: 35702017 PMCID: PMC9536115 DOI: 10.1177/0271678x221107422] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The development of ischemic lesions has primarily been studied in horizontal cortical space. However, how ischemic lesions develop through the cortical depth remains largely unknown. We explored this question using direct current coupled recordings at different cortical depths using linear arrays of iridium electrodes in the focal epipial endothelin-1 (ET1) ischemia model in the rat barrel cortex. ET1-induced impairments were characterized by a vertical gradient with (i) rapid suppression of the spontaneous activity in the superficial cortical layers at the onset of ischemia, (ii) compartmentalization of spreading depolarizations (SDs) to the deep layers during progression of ischemia, and (iii) deeper suppression of activity and larger histological lesion size in superficial cortical layers. The level of impairments correlated strongly with the rate of spontaneous activity suppression, the rate of SD onset after ET1 application, and the amplitude of giant negative ultraslow potentials (∼-70 mV), which developed during ET1 application and were similar to the tent-shaped ultraslow potentials observed during focal ischemia in the human cortex. Thus, in the epipial ET1 ischemia model, ischemic lesions develop progressively from the surface to the cortical depth, and early changes in electrical activity at the onset of ET1-induced ischemia reliably predict the severity of ischemic damage.
Collapse
Affiliation(s)
- Daria Vinokurova
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia.,INMED, Aix-Marseille University, Marseille, France
| | - Andrey Zakharov
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia.,Department of Physiology, Kazan State Medical University, Kazan, Russia
| | - Kseniya Chernova
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| | | | - Viktor Horst
- Centre for Stroke Research Berlin, Department of Experimental Neurology and Department of Neurology, Charité Universitätsmedizin Berlin (Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Berlin, Germany
| | - Coline L Lemale
- Centre for Stroke Research Berlin, Department of Experimental Neurology and Department of Neurology, Charité Universitätsmedizin Berlin (Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Berlin, Germany
| | - Jens P Dreier
- Centre for Stroke Research Berlin, Department of Experimental Neurology and Department of Neurology, Charité Universitätsmedizin Berlin (Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Berlin, Germany.,Bernstein Centre for Computational Neuroscience Berlin, Berlin, Germany.,Einstein Centre for Neurosciences Berlin, Berlin, Germany
| | - Roustem Khazipov
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia.,INMED, Aix-Marseille University, Marseille, France
| |
Collapse
|
26
|
Meinert F, Dömer P, Helgers SOA, Schumm L, Hecht N, Dreier JP, Woitzik J. Subdural Placement of Electrocorticographic Electrode Array Through a Burr Hole Exposure: 2-Dimensional Operative Video. Oper Neurosurg (Hagerstown) 2022; 23:e169. [PMID: 35972097 DOI: 10.1227/ons.0000000000000299] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/24/2022] [Indexed: 02/04/2023] Open
Affiliation(s)
- Franziska Meinert
- Department of Neurosurgery, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Patrick Dömer
- Department of Neurosurgery, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | | | - Leonie Schumm
- Department of Neurosurgery, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Nils Hecht
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jens Peter 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
| | - Johannes Woitzik
- Department of Neurosurgery, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
27
|
Herreras O, Torres D, Martín-Vázquez G, Hernández-Recio S, López-Madrona VJ, Benito N, Makarov VA, Makarova J. Site-dependent shaping of field potential waveforms. Cereb Cortex 2022; 33:3636-3650. [PMID: 35972425 PMCID: PMC10068269 DOI: 10.1093/cercor/bhac297] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
The activity of neuron populations gives rise to field potentials (FPs) that extend beyond the sources. Their mixing in the volume dilutes the original temporal motifs in a site-dependent manner, a fact that has received little attention. And yet, it potentially rids of physiological significance the time-frequency parameters of individual waves (amplitude, phase, duration). This is most likely to happen when a single source or a local origin is erroneously assumed. Recent studies using spatial treatment of these signals and anatomically realistic modeling of neuron aggregates provide convincing evidence for the multisource origin and site-dependent blend of FPs. Thus, FPs generated in primary structures like the neocortex and hippocampus reach far and cross-contaminate each other but also, they add and even impose their temporal traits on distant regions. Furthermore, both structures house neurons that act as spatially distinct (but overlapped) FP sources whose activation is state, region, and time dependent, making the composition of so-called local FPs highly volatile and strongly site dependent. Since the spatial reach cannot be predicted without source geometry, it is important to assess whether waveforms and temporal motifs arise from a single source; otherwise, those from each of the co-active sources should be sought.
Collapse
Affiliation(s)
- Oscar Herreras
- Department of Translational Neuroscience, Cajal Institute, CSIC, Av. Doctor Arce 37, Madrid 28002, Spain
| | - Daniel Torres
- Department of Translational Neuroscience, Cajal Institute, CSIC, Av. Doctor Arce 37, Madrid 28002, Spain
| | - Gonzalo Martín-Vázquez
- Department of Translational Neuroscience, Cajal Institute, CSIC, Av. Doctor Arce 37, Madrid 28002, Spain
| | - Sara Hernández-Recio
- Department of Translational Neuroscience, Cajal Institute, CSIC, Av. Doctor Arce 37, Madrid 28002, Spain
| | - Víctor J López-Madrona
- Department of Translational Neuroscience, Cajal Institute, CSIC, Av. Doctor Arce 37, Madrid 28002, Spain
| | - Nuria Benito
- Department of Translational Neuroscience, Cajal Institute, CSIC, Av. Doctor Arce 37, Madrid 28002, Spain
| | - Valeri A Makarov
- Department of Applied Mathematics, Institute for Interdisciplinary Mathematics, Universidad Complutense of Madrid, Av. Paraninfo s/n, Madrid 28040, Spain
| | - Julia Makarova
- Department of Translational Neuroscience, Cajal Institute, CSIC, Av. Doctor Arce 37, Madrid 28002, Spain.,Department of Applied Mathematics, Institute for Interdisciplinary Mathematics, Universidad Complutense of Madrid, Av. Paraninfo s/n, Madrid 28040, Spain
| |
Collapse
|
28
|
Unekawa M, Tomita Y, Masamoto K, Kanno I, Nakahara J, Izawa Y. Close association between spreading depolarization and development of infarction under experimental ischemia in anesthetized male mice. Brain Res 2022; 1792:148023. [PMID: 35901965 DOI: 10.1016/j.brainres.2022.148023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 11/02/2022]
Abstract
Clinical and experimental evidence suggests that spreading depolarizations (SD) usually occur in patients with ischemic or hemorrhagic stroke when the gray matter of the brain is affected. In this study, we evaluated spatiotemporal changes of cerebral blood flow (CBF) during middle cerebral artery (MCA) occlusion and examined the relationship between SD occurrence and cerebral infarct development. In male isoflurane-anesthetized C57BL/6J mice, CBF changes over the ipsilateral parietal bone were recorded by laser speckle flowgraphy during and after transient (45 min, n = 22) or permanent occlusion (n = 22) of the distal MCA. Infarct volume was evaluated 24 hr after the operation. Upon MCA occlusion, CBF decreased by -55.6 ± 8.5 % in the lowest CBF and linearly recovered with increasing distance from the region. At 1-10 min after onset of occlusion, SD occurred and concentrically propagated from the core region, showing a decrease of CBF in the whole observed area along with a transient hyperemia and oligemia in the normal region. SD spontaneously re-occurred and propagated around the ischemic area in 37 % of mice, accompanied with a marked decrease of CBF in the core or a marked increase of CBF in the normal region. The CBF response to SDs gradually changed from the core to the normal area, depending upon the distance from the core region. Infarction was not observed in transiently (n = 2) or permanently (n = 4) occluded mice without SD. The infarct area tended to be larger with increasing number of SDs in transiently occluded mice. In conclusion, our findings suggest that the occurrence of SD during ischemia might elicit infarct formation and/or influence infarct development.
Collapse
Affiliation(s)
- Miyuki Unekawa
- Department of Neurology, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan.
| | - Yutaka Tomita
- Department of Neurology, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| | - Kazuto Masamoto
- Center for Neuroscience and Biomedical Engineering, University of Electro-Communications, Chofu, Tokyo 182-8585, Japan; Department of Functional Brain Imaging, National Institutes for Quantum Science and Technology, Inage, Chiba 263-8555, Japan
| | - Iwao Kanno
- Department of Functional Brain Imaging, National Institutes for Quantum Science and Technology, Inage, Chiba 263-8555, Japan
| | - Jin Nakahara
- Department of Neurology, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| | - Yoshikane Izawa
- Department of Neurology, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| |
Collapse
|
29
|
Eighteen-hour inhibitory effect of s-ketamine on potassium- and ischemia-induced spreading depolarizations in the gyrencephalic swine brain. Neuropharmacology 2022; 216:109176. [DOI: 10.1016/j.neuropharm.2022.109176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 05/15/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022]
|
30
|
Yan J, Li W, Zhou C, Wu N, Yang X, Pan Q, He T, Wu Y, Guo Z, Xia Y, Sun X, Cheng C. Dynamic Measurements of Cerebral Blood Flow Responses to Cortical Spreading Depolarization in the Murine Endovascular Perforation Subarachnoid Hemorrhage Model. Transl Stroke Res 2022:10.1007/s12975-022-01052-1. [PMID: 35749033 DOI: 10.1007/s12975-022-01052-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/30/2022] [Accepted: 06/10/2022] [Indexed: 12/13/2022]
Abstract
Delayed cerebral ischemia (DCI) is the most severe complication after subarachnoid hemorrhage (SAH), and cortical spreading depolarization (CSD) is believed to play a vital role in it. However, the dynamic changes in cerebral blood flow (CBF) in response to CSD in typical SAH models have not been well investigated. Here, SAH was established in mice with endovascular perforation. Subsequently, the spontaneous CBF dropped instantly and then returned to baseline rapidly. After KCl application to the cortex, subsequent hypoperfusion waves occurred across the groups, while a lower average perfusion level was found in the SAH groups (days 1-7). Moreover, in the SAH groups, the number of CSD decreased within day 7, and the duration and spreading velocity of the CSD increased within day 3 and day 14, respectively. Next, we continuously monitored the local field potential (LFP) in the prefrontal cortex. The results showed that the decrease in the percentage of gamma oscillations lasted throughout the whole process in the SAH group. In the chronic phase after SAH, we found that the mice still had cognitive deficits but experienced no obvious tissue damage. In summary, SAH negatively affects the CBF responses to CSD and the spontaneous LFP activity and causes long-term cognitive deficits in mice. Based on these findings, in the specific phase after SAH, DCI is induced or exacerbated more easily by potential causers of CSD in clinical practice (edema, erythrocytolysis, inflammation), which may lead to neurological deterioration.
Collapse
Affiliation(s)
- Jin Yan
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, People's Republic of China
| | - Wenlang Li
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chao Zhou
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, People's Republic of China
| | - Na Wu
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, People's Republic of China
| | - Xiaomin Yang
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, People's Republic of China
| | - Qiuling Pan
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, People's Republic of China
| | - Tao He
- Department of Orthopaedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Wu
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, People's Republic of China
| | - Zongduo Guo
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, People's Republic of China
| | - Yongzhi Xia
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, People's Republic of China
| | - Xiaochuan Sun
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, People's Republic of China.
| | - Chongjie Cheng
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
31
|
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: 18] [Impact Index Per Article: 9.0] [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
|
32
|
Hund SJ, Brown BR, Lemale CL, Menon PG, Easley KA, Dreier JP, Jones SC. Numerical Simulation of Concussive-Generated Cortical Spreading Depolarization to Optimize DC-EEG Electrode Spacing for Noninvasive Visual Detection. Neurocrit Care 2022; 37:67-82. [PMID: 35233716 PMCID: PMC9262830 DOI: 10.1007/s12028-021-01430-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 12/29/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Cortical spreading depolarization (SD) is a propagating depolarization wave of neurons and glial cells in the cerebral gray matter. SD occurs in all forms of severe acute brain injury, as documented by using invasive detection methods. Based on many experimental studies of mechanical brain deformation and concussion, the occurrence of SDs in human concussion has often been hypothesized. However, this hypothesis cannot be confirmed in humans, as SDs can only be detected with invasive detection methods that would require either a craniotomy or a burr hole to be performed on athletes. Typical electroencephalography electrodes, placed on the scalp, can help detect the possible presence of SD but have not been able to accurately and reliably identify SDs. METHODS To explore the possibility of a noninvasive method to resolve this hurdle, we developed a finite element numerical model that simulates scalp voltage changes that are induced by a brain surface SD. We then compared our simulation results with retrospectively evaluated data in patients with aneurysmal subarachnoid hemorrhage from Drenckhahn et al. (Brain 135:853, 2012). RESULTS The ratio of peak scalp to simulated peak cortical voltage, Vscalp/Vcortex, was 0.0735, whereas the ratio from the retrospectively evaluated data was 0.0316 (0.0221, 0.0527) (median [1st quartile, 3rd quartile], n = 161, p < 0.001, one sample Wilcoxon signed-rank test). These differing values provide validation because their differences can be attributed to differences in shape between concussive SDs and aneurysmal subarachnoid hemorrhage SDs, as well as the inherent limitations in human study voltage measurements. This simulated scalp surface potential was used to design a virtual scalp detection array. Error analysis and visual reconstruction showed that 1 cm is the optimal electrode spacing to visually identify the propagating scalp voltage from a cortical SD. Electrode spacings of 2 cm and above produce distorted images and high errors in the reconstructed image. CONCLUSIONS Our analysis suggests that concussive (and other) SDs can be detected from the scalp, which could confirm SD occurrence in human concussion, provide concussion diagnosis on the basis of an underlying physiological mechanism, and lead to noninvasive SD detection in the setting of severe acute brain injury.
Collapse
Affiliation(s)
- Samuel J Hund
- CerebroScope, SciencePlusPlease LLC, Pittsburgh, PA, USA
- SimulationSolutions, Pittsburgh, PA, USA
| | | | - Coline L Lemale
- Center for Stroke Research, 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
| | - Prahlad G Menon
- CerebroScope, SciencePlusPlease LLC, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kirk A Easley
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Jens P Dreier
- Center for Stroke Research, 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
|
33
|
Dreier JP, Winkler MKL, Major S, Horst V, Lublinsky S, Kola V, Lemale CL, Kang EJ, Maslarova A, Salur I, Lückl J, Platz J, Jorks D, Oliveira-Ferreira AI, Schoknecht K, Reiffurth C, Milakara D, Wiesenthal D, Hecht N, Dengler NF, Liotta A, Wolf S, Kowoll CM, Schulte AP, Santos E, Güresir E, Unterberg AW, Sarrafzadeh A, Sakowitz OW, Vatter H, Reiner M, Brinker G, Dohmen C, Shelef I, Bohner G, Scheel M, Vajkoczy P, Hartings JA, Friedman A, Martus P, Woitzik J. Spreading depolarizations in ischaemia after subarachnoid haemorrhage, a diagnostic phase III study. Brain 2022; 145:1264-1284. [PMID: 35411920 DOI: 10.1093/brain/awab457] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/18/2021] [Accepted: 11/21/2021] [Indexed: 02/06/2023] Open
Abstract
Focal brain damage after aneurysmal subarachnoid haemorrhage predominantly results from intracerebral haemorrhage, and early and delayed cerebral ischaemia. The prospective, observational, multicentre, cohort, diagnostic phase III trial, DISCHARGE-1, primarily investigated whether the peak total spreading depolarization-induced depression duration of a recording day during delayed neuromonitoring (delayed depression duration) indicates delayed ipsilateral infarction. Consecutive patients (n = 205) who required neurosurgery were enrolled in six university hospitals from September 2009 to April 2018. Subdural electrodes for electrocorticography were implanted. Participants were excluded on the basis of exclusion criteria, technical problems in data quality, missing neuroimages or patient withdrawal (n = 25). Evaluators were blinded to other measures. Longitudinal MRI, and CT studies if clinically indicated, revealed that 162/180 patients developed focal brain damage during the first 2 weeks. During 4.5 years of cumulative recording, 6777 spreading depolarizations occurred in 161/180 patients and 238 electrographic seizures in 14/180. Ten patients died early; 90/170 developed delayed infarction ipsilateral to the electrodes. Primary objective was to investigate whether a 60-min delayed depression duration cut-off in a 24-h window predicts delayed infarction with >0.60 sensitivity and >0.80 specificity, and to estimate a new cut-off. The 60-min cut-off was too short. Sensitivity was sufficient [= 0.76 (95% confidence interval: 0.65-0.84), P = 0.0014] but specificity was 0.59 (0.47-0.70), i.e. <0.80 (P < 0.0001). Nevertheless, the area under the receiver operating characteristic (AUROC) curve of delayed depression duration was 0.76 (0.69-0.83, P < 0.0001) for delayed infarction and 0.88 (0.81-0.94, P < 0.0001) for delayed ischaemia (reversible delayed neurological deficit or infarction). In secondary analysis, a new 180-min cut-off indicated delayed infarction with a targeted 0.62 sensitivity and 0.83 specificity. In awake patients, the AUROC curve of delayed depression duration was 0.84 (0.70-0.97, P = 0.001) and the prespecified 60-min cut-off showed 0.71 sensitivity and 0.82 specificity for reversible neurological deficits. In multivariate analysis, delayed depression duration (β = 0.474, P < 0.001), delayed median Glasgow Coma Score (β = -0.201, P = 0.005) and peak transcranial Doppler (β = 0.169, P = 0.016) explained 35% of variance in delayed infarction. Another key finding was that spreading depolarization-variables were included in every multiple regression model of early, delayed and total brain damage, patient outcome and death, strongly suggesting that they are an independent biomarker of progressive brain injury. While the 60-min cut-off of cumulative depression in a 24-h window indicated reversible delayed neurological deficit, only a 180-min cut-off indicated new infarction with >0.60 sensitivity and >0.80 specificity. Although spontaneous resolution of the neurological deficit is still possible, we recommend initiating rescue treatment at the 60-min rather than the 180-min cut-off if progression of injury to infarction is to be prevented.
Collapse
Affiliation(s)
- Jens P Dreier
- Centre 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 Centre for Computational Neuroscience Berlin, Berlin, Germany.,Einstein Centre for Neurosciences Berlin, Berlin, Germany
| | - Maren K L Winkler
- Centre for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Robert Koch-Institute, Berlin, Germany
| | - Sebastian Major
- Centre 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
| | - Viktor Horst
- Centre for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Svetlana Lublinsky
- Department of Brain & Cognitive Sciences, Zlotowski Centre for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Soroka University Medical Centre, Beer-Sheva, Israel.,Department of Physiology & Cell Biology, Zlotowski Centre for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Soroka University Medical Centre, Beer-Sheva, Israel
| | - Vasilis Kola
- Centre for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Coline L Lemale
- Centre for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Eun-Jeung Kang
- Centre 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
| | - Anna Maslarova
- Department of Neurosurgery, University Hospital and Friedrich-Wilhelms-University Bonn, Bonn, Germany.,Department of Neurosurgery, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Irmak Salur
- Department of Neurosurgery, University Hospital and Friedrich-Wilhelms-University Bonn, Bonn, Germany.,Department of Neurosurgery, KRH Klinikum Nordstadt, Hannover, Germany
| | - Janos Lückl
- Centre 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 Medical Physics and Informatics, University of Szeged, Szeged, Hungary.,Department of Neurology, University of Szeged, Szeged, Hungary
| | - Johannes Platz
- Department of Neurosurgery, Herz-Neuro-Zentrum Bodensee, Kreuzlingen, Switzerland
| | - Devi Jorks
- Centre for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Bernstein Centre for Computational Neuroscience Berlin, Berlin, Germany.,Clienia Schlössli AG, Privatklinik für Psychiatrie und Psychotherapie, Oetwil am See, Switzerland
| | - Ana I Oliveira-Ferreira
- Centre for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Neuro-Electronics Research Flanders, Leuven, Belgium.,VIB-KU, Leuven, Belgium.,Interuniversity Microelectronics Centre, Leuven, Belgium.,Laboratory of Neural Circuits, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Karl Schoknecht
- Centre for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Carl Ludwig Institute for Physiology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Clemens Reiffurth
- Centre 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
| | - Denny Milakara
- Centre for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Research Campus STIMULATE, Otto-von-Guericke-University, Magdeburg, Germany
| | - Dirk Wiesenthal
- Centre for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Univention GmbH, Bremen, Germany
| | - Nils Hecht
- Centre 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 Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nora F Dengler
- Centre 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 Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Agustin Liotta
- Centre 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 Anaesthesiology and Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Stefan Wolf
- Centre 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 Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Christina M Kowoll
- Department of Neurology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - André P Schulte
- Department of Spinal Surgery, Krankenhaus der Augustinerinnen, Cologne, Germany
| | - Edgar Santos
- Department of Neurosurgery, Heidelberg University Hospital, Ruprecht-Karls-University Heidelberg, Germany
| | - Erdem Güresir
- Department of Neurosurgery, University Hospital and Friedrich-Wilhelms-University Bonn, Bonn, Germany
| | - Andreas W Unterberg
- Department of Neurosurgery, Heidelberg University Hospital, Ruprecht-Karls-University Heidelberg, Germany
| | - Asita Sarrafzadeh
- Division of Neurosurgery, Department of Clinical Neurosciences, Faculty of Medicine, Geneva University Medical Centre, Geneva, Switzerland
| | - Oliver W Sakowitz
- Department of Neurosurgery, Heidelberg University Hospital, Ruprecht-Karls-University Heidelberg, Germany
| | - Hartmut Vatter
- Department of Neurosurgery, University Hospital and Friedrich-Wilhelms-University Bonn, Bonn, Germany
| | - Michael Reiner
- Medical Advisory Service of the Statutory Health Insurance of North Rhine, Germany
| | - Gerrit Brinker
- Department of Neurosurgery, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Christian Dohmen
- Department for Neurology and Neurological Intensive Care Medicine, LVR-Klinik Bonn, Bonn, Germany
| | - Ilan Shelef
- Department of Brain & Cognitive Sciences, Zlotowski Centre for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Soroka University Medical Centre, Beer-Sheva, Israel.,Department of Physiology & Cell Biology, Zlotowski Centre for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Soroka University Medical Centre, Beer-Sheva, Israel.,Institute of Radiology, Soroka University Medical Centre, Beer-Sheva, Israel
| | - Georg Bohner
- Department of Neuroradiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Michael Scheel
- Department of Neuroradiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Peter Vajkoczy
- Centre 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 Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jed A Hartings
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Alon Friedman
- Department of Brain & Cognitive Sciences, Zlotowski Centre for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Soroka University Medical Centre, Beer-Sheva, Israel.,Department of Physiology & Cell Biology, Zlotowski Centre for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Soroka University Medical Centre, Beer-Sheva, Israel.,Department of Medical Neuroscience and Brain Repair Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Peter Martus
- Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, Tübingen, Germany
| | - Johannes Woitzik
- Centre 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 Neurosurgery, Evangelisches Krankenhaus Oldenburg, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
34
|
Menyhárt Á, Frank R, Farkas AE, Süle Z, Varga VÉ, Nyúl-Tóth Á, Meiller A, Ivánkovits-Kiss O, Lemale CL, Szabó Í, Tóth R, Zölei-Szénási D, Woitzik J, Marinesco S, Krizbai IA, Bari F, Dreier JP, Farkas E. Malignant astrocyte swelling and impaired glutamate clearance drive the expansion of injurious spreading depolarization foci. J Cereb Blood Flow Metab 2022; 42:584-599. [PMID: 34427145 PMCID: PMC8943616 DOI: 10.1177/0271678x211040056] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Spreading depolarizations (SDs) indicate injury progression and predict worse clinical outcome in acute brain injury. We demonstrate in rodents that acute brain swelling upon cerebral ischemia impairs astroglial glutamate clearance and increases the tissue area invaded by SD. The cytotoxic extracellular glutamate accumulation (>15 µM) predisposes an extensive bulk of tissue (4-5 mm2) for a yet undescribed simultaneous depolarization (SiD). We confirm in rat brain slices exposed to osmotic stress that SiD is the pathological expansion of prior punctual SD foci (0.5-1 mm2), is associated with astrocyte swelling, and triggers oncotic neuron death. The blockade of astrocytic aquaporin-4 channels and Na+/K+/Cl- co-transporters, or volume-regulated anion channels mitigated slice edema, extracellular glutamate accumulation (<10 µM) and SiD occurrence. Reversal of slice swelling by hyperosmotic mannitol counteracted glutamate accumulation and prevented SiD. In contrast, inhibition of glial metabolism or inhibition of astrocyte glutamate transporters reproduced the SiD phenotype. Finally, we show in the rodent water intoxication model of cytotoxic edema that astrocyte swelling and altered astrocyte calcium waves are central in the evolution of SiD. We discuss our results in the light of evidence for SiD in the human cortex. Our results emphasize the need of preventive osmotherapy in acute brain injury.
Collapse
Affiliation(s)
- Ákos Menyhárt
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Rita Frank
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Attila E Farkas
- Neurovascular Unit Research Group, Molecular Neurobiology Research Unit, Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Zoltán Süle
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Viktória É Varga
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ádám Nyúl-Tóth
- Neurovascular Unit Research Group, Molecular Neurobiology Research Unit, Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anne Meiller
- Lyon Neuroscience Research Center, Inserm U1028, CNRS UMR 5292, University Claude Bernard Lyon I, Lyon, France
| | - Orsolya Ivánkovits-Kiss
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - 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
| | - Írisz Szabó
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Réka Tóth
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Dániel Zölei-Szénási
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - 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
| | - Stephane Marinesco
- Lyon Neuroscience Research Center, Inserm U1028, CNRS UMR 5292, University Claude Bernard Lyon I, Lyon, France
| | - István A Krizbai
- Neurovascular Unit Research Group, Molecular Neurobiology Research Unit, Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Institute of Life Sciences, Vasile Goldis Western University, Arad, Romania
| | - Ferenc Bari
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - 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
| | - Eszter Farkas
- HCEMM-USZ Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary.,Department of Cell Biology and Molecular Medicine, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged,Szeged, Hungary
| |
Collapse
|
35
|
Binder NF, Glück C, Middleham W, Alasoadura M, Pranculeviciute N, Wyss MT, Chuquet J, Weber B, Wegener S, El Amki M. Vascular Response to Spreading Depolarization Predicts Stroke Outcome. Stroke 2022; 53:1386-1395. [PMID: 35240860 PMCID: PMC10510800 DOI: 10.1161/strokeaha.121.038085] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 12/24/2021] [Accepted: 02/01/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cortical spreading depolarization (CSD) is a massive neuro-glial depolarization wave, which propagates across the cerebral cortex. In stroke, CSD is a necessary and ubiquitous mechanism for the development of neuronal lesions that initiates in the ischemic core and propagates through the penumbra extending the tissue injury. Although CSD propagation induces dramatic changes in cerebral blood flow, the vascular responses in different ischemic regions and their consequences on reperfusion and recovery remain to be defined. METHODS Ischemia was performed using the thrombin model of stroke and reperfusion was induced by r-tPA (recombinant tissue-type plasminogen activator) administration in mice. We used in vivo electrophysiology and laser speckle contrast imaging simultaneously to assess both electrophysiological and hemodynamic characteristics of CSD after ischemia onset. Neurological deficits were assessed on day 1, 3, and 7. Furthermore, infarct sizes were quantified using 2,3,5-triphenyltetrazolium chloride on day 7. RESULTS After ischemia, CSDs were evidenced by the characteristic propagating DC shift extending far beyond the ischemic area. On the vascular level, we observed 2 types of responses: some mice showed spreading hyperemia confined to the penumbra area (penumbral spreading hyperemia) while other showed spreading hyperemia propagating in the full hemisphere (full hemisphere spreading hyperemia). Penumbral spreading hyperemia was associated with severe stroke-induced damage, while full hemisphere spreading hyperemia indicated beneficial infarct outcome and potential viability of the infarct core. In all animals, thrombolysis with r-tPA modified the shape of the vascular response to CSD and reduced lesion volume. CONCLUSIONS Our results show that different types of spreading hyperemia occur spontaneously after the onset of ischemia. Depending on their shape and distribution, they predict severity of injury and outcome. Furthermore, our data show that modulating the hemodynamic response to CSD may be a promising therapeutic strategy to attenuate stroke outcome.
Collapse
Affiliation(s)
- Nadine Felizitas Binder
- Department of Neurology, University Hospital Zurich and University of Zurich (UZH), Switzerland (N.F.B., W.M., N.P., S.W., M.E.A.)
- Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Switzerland (N.F.B., C.G., W.M., N.P., B.W., S.W., M.E.A.)
| | - Chaim Glück
- Institute of Pharmacology and Toxicology, Experimental Imaging and Neuroenergetics, University of Zurich (UZH), Switzerland (C.G., M.T.W., B.W.)
- Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Switzerland (N.F.B., C.G., W.M., N.P., B.W., S.W., M.E.A.)
| | - William Middleham
- Department of Neurology, University Hospital Zurich and University of Zurich (UZH), Switzerland (N.F.B., W.M., N.P., S.W., M.E.A.)
- Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Switzerland (N.F.B., C.G., W.M., N.P., B.W., S.W., M.E.A.)
| | - Michael Alasoadura
- Normandie University, Unirouen, INSERM U1239, Rouen, France (M.A., J.C.)
| | - Nikolete Pranculeviciute
- Department of Neurology, University Hospital Zurich and University of Zurich (UZH), Switzerland (N.F.B., W.M., N.P., S.W., M.E.A.)
- Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Switzerland (N.F.B., C.G., W.M., N.P., B.W., S.W., M.E.A.)
| | - Matthias Tasso Wyss
- Institute of Pharmacology and Toxicology, Experimental Imaging and Neuroenergetics, University of Zurich (UZH), Switzerland (C.G., M.T.W., B.W.)
| | - Julien Chuquet
- Normandie University, Unirouen, INSERM U1239, Rouen, France (M.A., J.C.)
- Normandie University, Unirouen, IRIB, EA3830-GRHVN, Rouen, France (J.C.)
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, Experimental Imaging and Neuroenergetics, University of Zurich (UZH), Switzerland (C.G., M.T.W., B.W.)
- Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Switzerland (N.F.B., C.G., W.M., N.P., B.W., S.W., M.E.A.)
| | - Susanne Wegener
- Department of Neurology, University Hospital Zurich and University of Zurich (UZH), Switzerland (N.F.B., W.M., N.P., S.W., M.E.A.)
| | - Mohamad El Amki
- Department of Neurology, University Hospital Zurich and University of Zurich (UZH), Switzerland (N.F.B., W.M., N.P., S.W., M.E.A.)
- Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Switzerland (N.F.B., C.G., W.M., N.P., B.W., S.W., M.E.A.)
| |
Collapse
|
36
|
Zheng Z, Li Z, Lv J. Loss of Kir6.1 facilitates peri-infarct depolarizations in focal cerebral ischemia. Neurol Res 2022; 44:797-806. [PMID: 35271426 DOI: 10.1080/01616412.2022.2051132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Peri-infarct depolarizations (PIDs) are spontaneous waves that propagate slowly across the penumbra region following stroke, contributing to secondary infarct growth and negatively affecting stroke outcomes. KATP channels are generally spread in the brain. Under conditions of ischemia and/or hypoxia, KATP channels play a cytoprotective role in neurons. However, it is still unknown whether KATP channels are involved in the initiation and propagation of PIDs. METHODS The Kir6.1 knockout (Kir6.1-/-) mice, Kir6.2 knockout (Kir6.2-/-) mice, and wild-type C57Bl6 mice (n = 8) were used. The middle cerebral artery occlusion (MCAO) stroke model was made and PIDs were detected by an optical intrinsic signal (OIS) imaging system. RESULTS Much more PIDs appeared in Kir6.1-/-mice than that in Kir6.2-/- and WT mice in both the first hour and 4 hours following MCAO (3.9 ± 0.7 vs. 1.5 ± 0.3, p < 0,05; 3.9 ± 0.7 vs. 1.9 ± 0.3, p < 0.05; 20.0 ± 2.5 vs. 10.4 ± 2.4, p < 0.05; 20.0 ± 2.5 vs. 11.3 ± 1.4, p < 0.05). Furthermore, the first PID occurred much earlier in Kir6.1-/- mice than that in Kir6.2-/- mice and WT mice (21.3 ± 2.1 min vs. 34.1 ± 4.8 min, p < 0.05; 21.3 ± 2.1 min vs. 38.8 ± 3.4 min, p < 0.01). No significant differences in other characteristics of PIDs including originating sites, duration time, propagation patterns, and velocity were detected. Additionally, the migration of originating sites was observed. CONCLUSION This study shows that loss of Kir6.1, not Kir6.2 facilitates the induction of PIDs in focal cerebral ischemia, indicating that Kir6.1-forming channels in the brain may provide protection against PIDs.
Collapse
Affiliation(s)
- Zelong Zheng
- The Department of Neurosurgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Zhenyu Li
- The Department of Neurosurgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Jianping Lv
- The Department of Neurosurgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
37
|
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: 4.5] [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
|
38
|
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: 31] [Impact Index Per Article: 15.5] [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
|
39
|
Schumm L, Lemale CL, Major S, Hecht N, Nieminen-Kelhä M, Zdunczyk A, Kowoll CM, Martus P, Thiel CM, Dreier JP, Woitzik J. Physiological variables in association with spreading depolarizations in the late phase of ischemic stroke. J Cereb Blood Flow Metab 2022; 42:121-135. [PMID: 34427143 PMCID: PMC8721769 DOI: 10.1177/0271678x211039628] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Physiological effects of spreading depolarizations (SD) are only well studied in the first hours after experimental stroke. In patients with malignant hemispheric stroke (MHS), monitoring of SDs is restricted to the postoperative ICU stay, typically day 2-7 post-ictus. Therefore, we investigated the role of physiological variables (temperature, intracranial pressure, mean arterial pressure and cerebral perfusion pressure) in relationship to SD during the late phase after MHS in humans. Additionally, an experimental stroke model was used to investigate hemodynamic consequences of SD during this time window. In 60 patients with MHS, the occurrence of 1692 SDs was preceded by a decrease in mean arterial pressure (-1.04 mmHg; p = .02) and cerebral perfusion pressure (-1.04 mmHg; p = .03). Twenty-four hours after middle cerebral artery occlusion in 50 C57Bl6/J mice, hypothermia led to prolonged SD-induced hyperperfusion (+2.8 min; p < .05) whereas hypertension mitigated initial hypoperfusion (-1.4 min and +18.5%Δ rCBF; p < .01). MRI revealed that SDs elicited 24 hours after experimental stroke were associated with lesion progression (15.9 vs. 14.8 mm³; p < .01). These findings of small but significant effects of physiological variables on SDs in the late phase after ischemia support the hypothesis that the impact of SDs may be modified by adjusting physiological variables.
Collapse
Affiliation(s)
- Leonie Schumm
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurosurgery, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Coline L Lemale
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Major
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nils Hecht
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Melina Nieminen-Kelhä
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Zdunczyk
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Peter Martus
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute for Clinical Epidemiology and Applied Biostatistics, University of Tübingen, Tübingen, Germany
| | - Christiane M Thiel
- Biological Psychology, Department of Psychology, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Jens P Dreier
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Woitzik
- Department of Neurosurgery, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
40
|
Berhouma M, Eker OF, Dailler F, Rheims S, Balanca B. Cortical Spreading Depolarizations in Aneurysmal Subarachnoid Hemorrhage: An Overview of Current Knowledge and Future Perspectives. Adv Tech Stand Neurosurg 2022; 45:229-244. [PMID: 35976452 DOI: 10.1007/978-3-030-99166-1_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Despite significant advances in the management of aneurysmal subarachnoid hemorrhage (SAH), morbidity and mortality remain devastating particularly for high-grade SAH. Poor functional outcome usually results from delayed cerebral ischemia (DCI). The pathogenesis of DCI during aneurysmal SAH has historically been attributed to cerebral vasospasm, but spreading depolarizations (SDs) are now considered to play a central role in DCI. During SAH, SDs may produce an inverse hemodynamic response leading to spreading ischemia. Several animal models have contributed to a better understanding of the pathogenesis of SDs during aneurysmal SAH and provided new therapeutic approaches including N-methyl-D-aspartate receptor antagonists and phosphodiesterase inhibitors. Herein we review the current knowledge in the field of SDs' pathogenesis and we detail the key experimental and clinical studies that have opened interesting new therapeutic approaches to prevent DCI in aneurysmal SAH.
Collapse
Affiliation(s)
- Moncef Berhouma
- Department of Neurosurgical Oncology and Vascular Neurosurgery, Pierre Wertheimer Neurological and Neurosurgical Hospital, Hospices Civils de Lyon (Lyon University Hospital), Lyon, France.
- Creatis Lab, CNRS UMR 5220, INSERM U1206, Lyon 1 University, INSA Lyon, Lyon, France.
| | - Omer Faruk Eker
- Creatis Lab, CNRS UMR 5220, INSERM U1206, Lyon 1 University, INSA Lyon, Lyon, France
- Department of Interventional Neuroradiology, Pierre Wertheimer Neurological and Neurosurgical Hospital, Hospices Civils de Lyon (Lyon University Hospital), Lyon, France
| | - Frederic Dailler
- Department of Neuro-Anesthesia and Neuro-Critical Care, Pierre Wertheimer Neurological and Neurosurgical Hospital, Hospices Civils de Lyon (Lyon University Hospital), Lyon, France
| | - Sylvain Rheims
- Department of Functional Neurology and Epileptology, Pierre Wertheimer Neurological and Neurosurgical Hospital, Hospices Civils de Lyon (Lyon University Hospital), Lyon, France
- Lyon's Neurosciences Research Center, INSERM U1028/CNRS, UMR 5292, University of Lyon, Lyon, France
| | - Baptiste Balanca
- Department of Neuro-Anesthesia and Neuro-Critical Care, Pierre Wertheimer Neurological and Neurosurgical Hospital, Hospices Civils de Lyon (Lyon University Hospital), Lyon, France
- Lyon's Neurosciences Research Center, INSERM U1028/CNRS, UMR 5292, University of Lyon, Lyon, France
| |
Collapse
|
41
|
Andrew RD, Hartings JA, Ayata C, Brennan KC, Dawson-Scully KD, Farkas E, Herreras O, Kirov SA, Müller M, Ollen-Bittle N, Reiffurth C, Revah O, Robertson RM, Shuttleworth CW, Ullah G, Dreier JP. The Critical Role of Spreading Depolarizations in Early Brain Injury: Consensus and Contention. Neurocrit Care 2022; 37:83-101. [PMID: 35257321 PMCID: PMC9259543 DOI: 10.1007/s12028-021-01431-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 12/29/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND When a patient arrives in the emergency department following a stroke, a traumatic brain injury, or sudden cardiac arrest, there is no therapeutic drug available to help protect their jeopardized neurons. One crucial reason is that we have not identified the molecular mechanisms leading to electrical failure, neuronal swelling, and blood vessel constriction in newly injured gray matter. All three result from a process termed spreading depolarization (SD). Because we only partially understand SD, we lack molecular targets and biomarkers to help neurons survive after losing their blood flow and then undergoing recurrent SD. METHODS In this review, we introduce SD as a single or recurring event, generated in gray matter following lost blood flow, which compromises the Na+/K+ pump. Electrical recovery from each SD event requires so much energy that neurons often die over minutes and hours following initial injury, independent of extracellular glutamate. RESULTS We discuss how SD has been investigated with various pitfalls in numerous experimental preparations, how overtaxing the Na+/K+ ATPase elicits SD. Elevated K+ or glutamate are unlikely natural activators of SD. We then turn to the properties of SD itself, focusing on its initiation and propagation as well as on computer modeling. CONCLUSIONS Finally, we summarize points of consensus and contention among the authors as well as where SD research may be heading. In an accompanying review, we critique the role of the glutamate excitotoxicity theory, how it has shaped SD research, and its questionable importance to the study of early brain injury as compared with SD theory.
Collapse
Affiliation(s)
- R. David Andrew
- grid.410356.50000 0004 1936 8331Queen’s University, Kingston, ON Canada
| | - Jed A. Hartings
- grid.24827.3b0000 0001 2179 9593University of Cincinnati, Cincinnati, OH USA
| | - Cenk Ayata
- grid.38142.3c000000041936754XHarvard Medical School, Harvard University, Boston, MA USA
| | - K. C. Brennan
- grid.223827.e0000 0001 2193 0096The University of Utah, Salt Lake City, UT USA
| | | | - Eszter Farkas
- grid.9008.10000 0001 1016 96251HCEMM-USZ Cerebral Blood Flow and Metabolism Research Group, and the Department of Cell Biology and Molecular Medicine, Faculty of Science and Informatics & Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Oscar Herreras
- grid.419043.b0000 0001 2177 5516Instituto de Neurobiologia Ramon Y Cajal (Consejo Superior de Investigaciones Científicas), Madrid, Spain
| | - Sergei. A. Kirov
- grid.410427.40000 0001 2284 9329Medical College of Georgia, Augusta, GA USA
| | - Michael Müller
- grid.411984.10000 0001 0482 5331University of Göttingen, University Medical Center Göttingen, Göttingen, Germany
| | - Nikita Ollen-Bittle
- grid.39381.300000 0004 1936 8884University of Western Ontario, London, ON Canada
| | - Clemens Reiffurth
- grid.7468.d0000 0001 2248 7639Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; and the Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health., Berlin, Germany
| | - Omer Revah
- grid.168010.e0000000419368956School of Medicine, Stanford University, Stanford, CA USA
| | | | | | - Ghanim Ullah
- grid.170693.a0000 0001 2353 285XUniversity of South Florida, Tampa, FL USA
| | - Jens P. Dreier
- grid.7468.d0000 0001 2248 7639Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; and the Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health., Berlin, Germany
| |
Collapse
|
42
|
Risk of Re-Rupture, Vasospasm, or Re-Stroke after Clipping or Coiling of Ruptured Intracranial Aneurysms: Long-Term Follow-Up with a Propensity Score-Matched, Population-Based Cohort Study. J Pers Med 2021; 11:jpm11111209. [PMID: 34834561 PMCID: PMC8622401 DOI: 10.3390/jpm11111209] [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: 09/26/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 12/17/2022] Open
Abstract
Scarce evidence is available in Asia for estimating the long-term risk and prognostic factors of major complications such as re-rupture, vasospasm, or re-stroke for patients with aneurysmal subarachnoid hemorrhage (SAH) undergoing endovascular coil embolization or surgical clipping. This is the first head-to-head propensity score-matched study in an Asian population to demonstrate that endovascular coil embolization for aneurysmal SAH treatment is riskier than surgical clipping in terms of re-rupture, vasospasm, or re-stroke. In addition, the independent poor prognostic factors of vasospasm or re-stroke were endovascular coil embolization, male sex, older age (≥65 years; the risk of vasospasm increases with age), hypertension, congestive heart failure, diabetes, previous transient ischemic attack, or stroke in aneurysmal SAH treatment. Background: To estimate the long-term complications and prognostic factors of endovascular coil embolization or surgical clipping for patients with ruptured aneurysmal subarachnoid hemorrhage (SAH). Methods: We selected patients diagnosed with aneurysmal SAH between 1 January 2011 and 31 December 2017. Propensity score matching was performed, and Cox proportional hazards model curves were used to analyze the risk of re-rupture, vasospasm, and re-stroke in patients undergoing the different treatments. Findings: Multivariate Cox regression analysis revealed that the adjusted hazard ratio (aHR) of re-rupture for endovascular coil embolization compared with surgical clipping was 1.36 (95% confidence interval [CI]: 1.17–1.57; p < 0.0001). The aHRs of the secondary endpoints of vasospasm and re-stroke (delayed cerebral ischemia) for endovascular coil embolization compared with surgical clipping were 1.14 (1.02–1.27; p = 0.0214) and 2.04 (1.83–2.29; p < 0.0001), respectively. The independent poor prognostic factors for vasospasm and re-stroke were endovascular coil embolization, male sex, older age (≥65 years; risk increases with age), hypertension, congestive heart failure, diabetes, and previous transient ischemic attack or stroke. Interpretation: Endovascular coil embolization for aneurysmal SAH carries a higher risk than surgical clipping of both short- and long-term complications including re-rupture, vasospasm, and re-stroke.
Collapse
|
43
|
Brainstem and Cortical Spreading Depolarization in a Closed Head Injury Rat Model. Int J Mol Sci 2021; 22:ijms222111642. [PMID: 34769073 PMCID: PMC8584184 DOI: 10.3390/ijms222111642] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/14/2021] [Accepted: 10/25/2021] [Indexed: 11/17/2022] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of death in young individuals, and is a major health concern that often leads to long-lasting complications. However, the electrophysiological events that occur immediately after traumatic brain injury, and may underlie impact outcomes, have not been fully elucidated. To investigate the electrophysiological events that immediately follow traumatic brain injury, a weight-drop model of traumatic brain injury was used in rats pre-implanted with epidural and intracerebral electrodes. Electrophysiological (near-direct current) recordings and simultaneous alternating current recordings of brain activity were started within seconds following impact. Cortical spreading depolarization (SD) and SD-induced spreading depression occurred in approximately 50% of mild and severe impacts. SD was recorded within three minutes after injury in either one or both brain hemispheres. Electrographic seizures were rare. While both TBI- and electrically induced SDs resulted in elevated oxidative stress, TBI-exposed brains showed a reduced antioxidant defense. In severe TBI, brainstem SD could be recorded in addition to cortical SD, but this did not lead to the death of the animals. Severe impact, however, led to immediate death in 24% of animals, and was electrocorticographically characterized by non-spreading depression (NSD) of activity followed by terminal SD in both cortex and brainstem.
Collapse
|
44
|
Dreier JP. Vasospasm-Induced Spreading Depolarization and/or Spreading-Depolarization-Induced Vasospasm After Subarachnoid Hemorrhage. Neurocrit Care 2021; 37:5-7. [PMID: 34704217 PMCID: PMC9259518 DOI: 10.1007/s12028-021-01373-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 09/30/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Jens P Dreier
- Center for Stroke Research Berlin and Departments of Experimental Neurology and 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
|
45
|
Oxygen-Induced and pH-Induced Direct Current Artifacts on Invasive Platinum/Iridium Electrodes for Electrocorticography. Neurocrit Care 2021; 35:146-159. [PMID: 34622418 PMCID: PMC8496677 DOI: 10.1007/s12028-021-01358-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 09/15/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Spreading depolarization (SD) and the initial, still reversible phase of neuronal cytotoxic edema in the cerebral gray matter are two modalities of the same process. SD may thus serve as a real-time mechanistic biomarker for impending parenchyma damage in patients during neurocritical care. Using subdural platinum/iridium (Pt/Ir) electrodes, SD is observed as a large negative direct current (DC) shift. Besides SD, there are other causes of DC shifts that are not to be confused with SD. Here, we systematically analyzed DC artifacts in ventilated patients by observing changes in the fraction of inspired oxygen. For the same change in blood oxygenation, we found that negative and positive DC shifts can simultaneously occur at adjacent Pt/Ir electrodes. METHODS Nurses and intensivists typically increase blood oxygenation by increasing the fraction of inspired oxygen at the ventilator before performing manipulations on the patient. We retrospectively identified 20 such episodes in six patients via tissue partial pressure of oxygen (ptiO2) measurements with an intracortical O2 sensor and analyzed the associated DC shifts. In vitro, we compared Pt/Ir with silver/silver chloride (Ag/AgCl) to assess DC responses to changes in pO2, pH, or 5-min square voltage pulses and investigated the effect of electrode polarization on pO2-induced DC artifacts. RESULTS Hyperoxygenation episodes started from a ptiO2 of 37 (30-40) mmHg (median and interquartile range) reaching 71 (50-97) mmHg. During a total of 20 episodes on each of six subdural Pt/Ir electrodes in six patients, we observed 95 predominantly negative responses in six patients, 25 predominantly positive responses in four patients, and no brain activity changes. Adjacent electrodes could show positive and negative responses simultaneously. In vitro, Pt/Ir in contrast with Ag/AgCl responded to changes in either pO2 or pH with large DC shifts. In response to square voltage pulses, Pt/Ir falsely showed smaller DC shifts than Ag/AgCl, with the worst performance under anoxia. In response to pO2 increase, Pt/Ir showed DC positivity when positively polarized and DC negativity when negatively polarized. CONCLUSIONS The magnitude of pO2-induced subdural DC shifts by approximately 6 mV was similar to that of SDs, but they did not show a sequential onset at adjacent recording sites, could be either predominantly negative or positive in contrast with the always negative DC shifts of SD, and were not accompanied by brain activity depression. Opposing polarities of pO2-induced DC artifacts may result from differences in baseline electrode polarization or subdural ptiO2 inhomogeneities relative to subdermal ptiO2 at the quasi-reference.
Collapse
|
46
|
Robinson D, Hartings J, Foreman B. First Report of Spreading Depolarization Correlates on Scalp EEG Confirmed with a Depth Electrode. Neurocrit Care 2021; 35:100-104. [PMID: 34617254 DOI: 10.1007/s12028-021-01360-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 09/15/2021] [Indexed: 11/26/2022]
Affiliation(s)
- David Robinson
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, 234 Goodman Street, Cincinnati, OH, USA.
| | - Jed Hartings
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Collaborative for Research On Acute Neurological Injuries, Cincinnati, OH, USA
| | - Brandon Foreman
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, 234 Goodman Street, Cincinnati, OH, USA
- Collaborative for Research On Acute Neurological Injuries, Cincinnati, OH, USA
| |
Collapse
|
47
|
Zhao HT, Tuohy MC, Chow D, Kozberg MG, Kim SH, Shaik MA, Hillman EMC. Neurovascular dynamics of repeated cortical spreading depolarizations after acute brain injury. Cell Rep 2021; 37:109794. [PMID: 34610299 PMCID: PMC8590206 DOI: 10.1016/j.celrep.2021.109794] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/30/2021] [Accepted: 09/14/2021] [Indexed: 11/30/2022] Open
Abstract
Cortical spreading depolarizations (CSDs) are increasingly suspected to play an exacerbating role in a range of acute brain injuries, including stroke, possibly through their interactions with cortical blood flow. We use simultaneous wide-field imaging of neural activity and hemodynamics in Thy1-GCaMP6f mice to explore the neurovascular dynamics of CSDs during and following Rose Bengal-mediated photothrombosis. CSDs are observed in all mice as slow-moving waves of GCaMP fluorescence extending far beyond the photothrombotic area. Initial CSDs are accompanied by profound vasoconstriction and leave residual oligemia and ischemia in their wake. Later, CSDs evoke variable responses, from constriction to biphasic to vasodilation. However, CSD-evoked vasoconstriction is found to be more likely during rapid, high-amplitude CSDs in regions with stronger oligemia and ischemia, which, in turn, worsens after each repeated CSD. This feedback loop may explain the variable but potentially devastating effects of CSDs in the context of acute brain injury. Zhao et al. use wide-field optical mapping of neuronal and hemodynamic activity in mice, capturing CSDs immediately following photothrombosis. Initial CSDs are accompanied by strong vasoconstriction, leaving persistent oligemia and ischemia. Region-dependent neurovascular responses to subsequent CSDs demonstrate a potential vicious cycle of CSD-dependent damage in acute brain injury.
Collapse
Affiliation(s)
- Hanzhi T Zhao
- Laboratory for Functional Optical Imaging, Mortimer B. Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Mary Claire Tuohy
- Laboratory for Functional Optical Imaging, Mortimer B. Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Daniel Chow
- Laboratory for Functional Optical Imaging, Mortimer B. Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Mariel G Kozberg
- Laboratory for Functional Optical Imaging, Mortimer B. Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Sharon H Kim
- Laboratory for Functional Optical Imaging, Mortimer B. Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Mohammed A Shaik
- Laboratory for Functional Optical Imaging, Mortimer B. Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Elizabeth M C Hillman
- Laboratory for Functional Optical Imaging, Mortimer B. Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
48
|
Bazzigaluppi P, Mester J, Joo IL, Weisspapir I, Dorr A, Koletar MM, Beckett TL, Khosravani H, Carlen P, Stefanovic B. Frequency selective neuronal modulation triggers spreading depolarizations in the rat endothelin-1 model of stroke. J Cereb Blood Flow Metab 2021; 41:2756-2768. [PMID: 33969731 PMCID: PMC8504421 DOI: 10.1177/0271678x211013656] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Ischemia is one of the most common causes of acquired brain injury. Central to its noxious sequelae are spreading depolarizations (SDs), waves of persistent depolarizations which start at the location of the flow obstruction and expand outwards leading to excitotoxic damage. The majority of acute stage of stroke studies to date have focused on the phenomenology of SDs and their association with brain damage. In the current work, we investigated the role of peri-injection zone pyramidal neurons in triggering SDs by optogenetic stimulation in an endothelin-1 rat model of focal ischemia. Our concurrent two photon fluorescence microscopy data and local field potential recordings indicated that a ≥ 60% drop in cortical arteriolar red blood cell velocity was associated with SDs at the ET-1 injection site. SDs were also observed in the peri-injection zone, which subsequently exhibited elevated neuronal activity in the low-frequency bands. Critically, SDs were triggered by low- but not high-frequency optogenetic stimulation of peri-injection zone pyramidal neurons. Our findings depict a complex etiology of SDs post focal ischemia and reveal that effects of neuronal modulation exhibit spectral and spatial selectivity.
Collapse
Affiliation(s)
- Paolo Bazzigaluppi
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
- Paolo Bazzigaluppi, Sunnybrook Research Institute, 2075 Bayview Ave., S646, Toronto, ON M4N 3M5, Canada.
| | - James Mester
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Illsung L Joo
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
| | - Iliya Weisspapir
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
| | - Adrienne Dorr
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
| | | | - Tina L Beckett
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
| | - Houman Khosravani
- Division of Neurology and Interdepartmental Division of Critical Care, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
- Interdepartmental Division of Critical Care, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Peter Carlen
- Krembil Research Institute, University of Toronto, Toronto, ON, Canada
| | - Bojana Stefanovic
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
49
|
Emerging immune and cell death mechanisms in stroke: Saponins as therapeutic candidates. Brain Behav Immun Health 2021; 9:100152. [PMID: 34589895 PMCID: PMC8474497 DOI: 10.1016/j.bbih.2020.100152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
The complexity of the ischemic cascade is based on the integrated crosstalk of every cell type in the neurovascular unit. Depending on the features of the ischemic insult, several cell death mechanisms are triggered, such as apoptosis, necroptosis, ferroptosis/oxytosis, ETosis or pyroptosis, leading to reactive astrogliosis. However, emerging evidence demonstrates a dual role for the immune system in stroke pathophysiology, where it exerts both detrimental and also beneficial functions. In this review, we discuss the relevance of several cell death modalities and the dual role of the immune system in stroke pathophysiology. We also provide an overview of some emerging immunomodulatory therapeutic strategies, amongst which saponins, which are promising candidates that exert multiple pharmacological effects. Several cell death mechanisms coexist in stroke pathophysiology. Neurons are more vulnerable to necroptosis than glial cells. Inhibitors of receptor-interacting protein kinases and of ferroptosis induce neuroprotection. Saponins exert modulatory effects on inflammation and neuronal cell death in stroke.
Collapse
|
50
|
Full-Band EEG Recordings Using Hybrid AC/DC-Divider Filters. eNeuro 2021; 8:ENEURO.0246-21.2021. [PMID: 34380654 PMCID: PMC8387152 DOI: 10.1523/eneuro.0246-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/27/2021] [Accepted: 08/04/2021] [Indexed: 11/21/2022] Open
Abstract
Full-band DC recordings enable recording of slow electrical brain signals that are severely compromised during conventional AC recordings. However, full-band DC recordings may be limited by the amplifier's dynamic input range and the loss of small amplitude high-frequency signals. Recently, Neuralynx has proposed full-band recordings with inverse filtering for signal reconstruction based on hybrid AC/DC-divider RRC filters that enable only partial suppression of DC signals. However, the quality of signal reconstruction for biological signals has not yet been assessed. Here, we propose a novel digital inverse filter based on a mathematical model describing RRC filter properties, which provides high computational accuracy and versatility. Second, we propose procedures for the evaluation of the inverse filter coefficients, adapted for each recording channel to minimize the error caused by the deviation of the real values of the RRC filter elements from their nominal values. We demonstrate that this approach enables near 99% reconstruction quality of high-potassium-induced cortical spreading depolarizations (SDs), endothelin-induced ischemic negative ultraslow potentials (NUPs), and whole-cell recordings of membrane potential using RRC filters. The quality of the reconstruction was significantly higher than with the existing inverse filtering procedures. Thus, RRC filters with inverse filtering are optimal for full-band EEG recordings in various applications.
Collapse
|