1
|
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; 44:1550-1560. [PMID: 38902207 PMCID: PMC11572034 DOI: 10.1177/0271678x241262203] [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: 11/30/2023] [Revised: 05/20/2024] [Accepted: 05/26/2024] [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
|
2
|
Frank R, Szarvas PA, Pesti I, Zsigmond A, Berkecz R, Menyhárt Á, Bari F, Farkas E. Nimodipine inhibits spreading depolarization, ischemic injury, and neuroinflammation in mouse live brain slice preparations. Eur J Pharmacol 2024; 977:176718. [PMID: 38849040 DOI: 10.1016/j.ejphar.2024.176718] [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: 04/11/2024] [Revised: 05/28/2024] [Accepted: 06/05/2024] [Indexed: 06/09/2024]
Abstract
Nimodipine is used to prevent delayed ischemic deficit in patients with aneurysmal subarachnoid hemorrhage (aSAH). Spreading depolarization (SD) is recognized as a factor in the pathomechanism of aSAH and other acute brain injuries. Although nimodipine is primarily known as a cerebral vasodilator, it may have a more complex mechanism of action due to the expression of its target, the L-type voltage-gated calcium channels (LVGCCs) in various cells in neural tissue. This study was designed to investigate the direct effect of nimodipine on SD, ischemic tissue injury, and neuroinflammation. SD in control or nimodipine-treated live mouse brain slices was induced under physiological conditions using electrical stimulation, or by subjecting the slices to hypo-osmotic stress or mild oxygen-glucose deprivation (mOGD). SD was recorded applying local field potential recording or intrinsic optical signal imaging. Histological analysis was used to estimate tissue injury, the number of reactive astrocytes, and the degree of microglia activation. Nimodipine did not prevent SD occurrence in mOGD, but it did reduce the rate of SD propagation and the cortical area affected by SD. In contrast, nimodipine blocked SD occurrence in hypo-osmotic stress, but had no effect on SD propagation. Furthermore, nimodipine prevented ischemic injury associated with SD in mOGD. Nimodipine also exhibited anti-inflammatory effects in mOGD by reducing reactive astrogliosis and microglial activation. The results demonstrate that nimodipine directly inhibits SD, independent of nimodipine's vascular effects. Therefore, the use of nimodipine may be extended to treat acute brain injuries where SD plays a central role in injury progression.
Collapse
Affiliation(s)
- Rita Frank
- 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.
| | - Péter Archibald Szarvas
- 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
| | - István Pesti
- 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
| | - Anna Zsigmond
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Róbert Berkecz
- Institute of Pharmaceutical Analysis, Faculty of Pharmacy, University of Szeged, Szeged, Hungary; Department of Forensic Medicine, Albert Szent-Györgyi Health Centre, Kossuth Lajos Sgt. 40, 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
| | - 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
|
3
|
Sword J, Fomitcheva IV, Kirov SA. Spreading depolarization causes reversible neuronal mitochondria fragmentation and swelling in healthy, normally perfused neocortex. J Cereb Blood Flow Metab 2024:271678X241257887. [PMID: 39053498 PMCID: PMC11574936 DOI: 10.1177/0271678x241257887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/28/2024] [Accepted: 05/12/2024] [Indexed: 07/27/2024]
Abstract
Mitochondrial function is tightly linked to morphology, and fragmentation of dendritic mitochondria during noxious conditions suggests loss of function. In the normoxic cortex, spreading depolarization (SD) is a phenomenon underlying migraine aura. It is unknown whether mitochondria structure is affected by normoxic SD. In vivo two-photon imaging followed by quantitative serial section electron microscopy (ssEM) was used to monitor dendritic mitochondria in the normoxic cortex of urethane-anesthetized mature male and female mice during and after SD initiated by focal KCl microinjection. Structural dynamics of dendrites and their mitochondria were visualized by transfecting excitatory, glutamatergic neurons of the somatosensory cortex with bicistronic AAV, which induced tdTomoto labeling in neuronal cytoplasm and mitochondria labeling with roGFP. Normoxic SD triggered rapidly reversible fragmentation of dendritic mitochondria alongside dendritic beading; however, mitochondria took significantly longer to recover. Several rounds of SD resulted in transient mitochondrial fragmentation and dendritic beading without accumulating injury, as both recovered. SsEM corroborated normoxic SD-elicited dendritic and mitochondrial swelling and transformation of the filamentous mitochondrial network into shorter, swollen tubular, and globular structures. Our results revealed normoxic SD-induced disruption of the dendritic mitochondrial structure that might impact mitochondrial bioenergetics during migraine with aura.
Collapse
Affiliation(s)
- Jeremy Sword
- Dept. of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Ioulia V Fomitcheva
- Dept. of Neurosurgery, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Sergei A Kirov
- Dept. of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Dept. of Neurosurgery, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
4
|
Hladky SB, Barrand MA. Alterations in brain fluid physiology during the early stages of development of ischaemic oedema. Fluids Barriers CNS 2024; 21:51. [PMID: 38858667 PMCID: PMC11163777 DOI: 10.1186/s12987-024-00534-8] [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: 02/01/2024] [Accepted: 03/22/2024] [Indexed: 06/12/2024] Open
Abstract
Oedema occurs when higher than normal amounts of solutes and water accumulate in tissues. In brain parenchymal tissue, vasogenic oedema arises from changes in blood-brain barrier permeability, e.g. in peritumoral oedema. Cytotoxic oedema arises from excess accumulation of solutes within cells, e.g. ischaemic oedema following stroke. This type of oedema is initiated when blood flow in the affected core region falls sufficiently to deprive brain cells of the ATP needed to maintain ion gradients. As a consequence, there is: depolarization of neurons; neural uptake of Na+ and Cl- and loss of K+; neuronal swelling; astrocytic uptake of Na+, K+ and anions; swelling of astrocytes; and reduction in ISF volume by fluid uptake into neurons and astrocytes. There is increased parenchymal solute content due to metabolic osmolyte production and solute influx from CSF and blood. The greatly increased [K+]isf triggers spreading depolarizations into the surrounding penumbra increasing metabolic load leading to increased size of the ischaemic core. Water enters the parenchyma primarily from blood, some passing into astrocyte endfeet via AQP4. In the medium term, e.g. after three hours, NaCl permeability and swelling rate increase with partial opening of tight junctions between blood-brain barrier endothelial cells and opening of SUR1-TPRM4 channels. Swelling is then driven by a Donnan-like effect. Longer term, there is gross failure of the blood-brain barrier. Oedema resolution is slower than its formation. Fluids without colloid, e.g. infused mock CSF, can be reabsorbed across the blood-brain barrier by a Starling-like mechanism whereas infused serum with its colloids must be removed by even slower extravascular means. Large scale oedema can increase intracranial pressure (ICP) sufficiently to cause fatal brain herniation. The potentially lethal increase in ICP can be avoided by craniectomy or by aspiration of the osmotically active infarcted region. However, the only satisfactory treatment resulting in retention of function is restoration of blood flow, providing this can be achieved relatively quickly. One important objective of current research is to find treatments that increase the time during which reperfusion is successful. Questions still to be resolved are discussed.
Collapse
Affiliation(s)
- Stephen B Hladky
- Department of Pharmacology, Tennis Court Rd., Cambridge, CB2 1PD, UK.
| | - Margery A Barrand
- Department of Pharmacology, Tennis Court Rd., Cambridge, CB2 1PD, UK
| |
Collapse
|
5
|
Planas AM. Role of microglia in stroke. Glia 2024; 72:1016-1053. [PMID: 38173414 DOI: 10.1002/glia.24501] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024]
Abstract
Microglia play key roles in the post-ischemic inflammatory response and damaged tissue removal reacting rapidly to the disturbances caused by ischemia and working to restore the lost homeostasis. However, the modified environment, encompassing ionic imbalances, disruption of crucial neuron-microglia interactions, spreading depolarization, and generation of danger signals from necrotic neurons, induce morphological and phenotypic shifts in microglia. This leads them to adopt a proinflammatory profile and heighten their phagocytic activity. From day three post-ischemia, macrophages infiltrate the necrotic core while microglia amass at the periphery. Further, inflammation prompts a metabolic shift favoring glycolysis, the pentose-phosphate shunt, and lipid synthesis. These shifts, combined with phagocytic lipid intake, drive lipid droplet biogenesis, fuel anabolism, and enable microglia proliferation. Proliferating microglia release trophic factors contributing to protection and repair. However, some microglia accumulate lipids persistently and transform into dysfunctional and potentially harmful foam cells. Studies also showed microglia that either display impaired apoptotic cell clearance, or eliminate synapses, viable neurons, or endothelial cells. Yet, it will be essential to elucidate the viability of engulfed cells, the features of the local environment, the extent of tissue damage, and the temporal sequence. Ischemia provides a rich variety of region- and injury-dependent stimuli for microglia, evolving with time and generating distinct microglia phenotypes including those exhibiting proinflammatory or dysfunctional traits and others showing pro-repair features. Accurate profiling of microglia phenotypes, alongside with a more precise understanding of the associated post-ischemic tissue conditions, is a necessary step to serve as the potential foundation for focused interventions in human stroke.
Collapse
Affiliation(s)
- Anna M Planas
- Cerebrovascular Research Laboratory, Department of Neuroscience and Experimental Therapeutics, Instituto de Investigaciones Biomédicas de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
- Cerebrovascular Diseases, Area of Clinical and Experimental Neuroscience, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clínic, Barcelona, Spain
| |
Collapse
|
6
|
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
|
7
|
Palopoli-Trojani K, Trumpis M, Chiang CH, Wang C, Williams AJ, Evans CL, Turner DA, Viventi J, Hoffmann U. High-density cortical µECoG arrays concurrently track spreading depolarizations and long-term evolution of stroke in awake rats. Commun Biol 2024; 7:263. [PMID: 38438529 PMCID: PMC10912118 DOI: 10.1038/s42003-024-05932-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 02/18/2024] [Indexed: 03/06/2024] Open
Abstract
Spreading depolarizations (SDs) are widely recognized as a major contributor to the progression of tissue damage from ischemic stroke even if blood flow can be restored. They are characterized by negative intracortical waveforms of up to -20 mV, propagation velocities of 3 - 6 mm/min, and massive disturbance of membrane ion homeostasis. High-density, micro-electrocorticographic (μECoG) epidural electrodes and custom, DC-coupled, multiplexed amplifiers, were used to continuously characterize and monitor SD and µECoG cortical signal evolution in awake, moving rats over days. This highly innovative approach can define these events over a large brain surface area (~ 3.4 × 3.4 mm), extending across the boundaries of the stroke, and offers sufficient electrode density (60 contacts total per array for a density of 5.7 electrodes / mm2) to measure and determine the origin of SDs in relation to the infarct boundaries. In addition, spontaneous ECoG activity can simultaneously be detected to further define cortical infarct regions. This technology allows us to understand dynamic stroke evolution and provides immediate cortical functional activity over days. Further translational development of this approach may facilitate improved treatment options for acute stroke patients.
Collapse
Affiliation(s)
| | | | | | - Charles Wang
- Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Cody L Evans
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, USA
| | - Dennis A Turner
- Biomedical Engineering, Duke University, Durham, NC, USA
- Neurosurgery, Neurobiology, Duke University, Durham, USA
- Research and Surgery Services, Durham VAMC, Durham, USA
| | | | - Ulrike Hoffmann
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, USA.
| |
Collapse
|
8
|
Sword J, Fomitcheva IV, Kirov SA. Spreading depolarization causes reversible neuronal mitochondria fragmentation and swelling in healthy, normally perfused neocortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576364. [PMID: 38328069 PMCID: PMC10849532 DOI: 10.1101/2024.01.22.576364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Mitochondrial function is tightly linked to their morphology, and fragmentation of dendritic mitochondria during noxious conditions suggests loss of function. In the normoxic cortex, spreading depolarization (SD) is a phenomenon underlying migraine aura. It is unknown whether mitochondria structure is affected by normoxic SD. In vivo two-photon imaging followed by quantitative serial section electron microscopy (ssEM) was used to monitor dendritic mitochondria in the normoxic cortex of urethane-anesthetized mature male and female mice during and after SD initiated by focal KCl microinjection. Structural dynamics of dendrites and their mitochondria were visualized by transfecting excitatory, glutamatergic neurons of the somatosensory cortex with bicistronic AAV, which induced tdTomoto labeling in neuronal cytoplasm and mitochondria labeling with roGFP. Normoxic SD triggered a rapid fragmentation of dendritic mitochondria alongside dendritic beading, both reversible; however, mitochondria took significantly longer to recover. Several rounds of SD resulted in transient mitochondrial fragmentation and dendritic beading without accumulating injury, as both recovered. SsEM corroborated normoxic SD-elicited dendritic and mitochondrial swelling and transformation of the filamentous mitochondrial network into shorter, swollen tubular and globular structures. Our results revealed normoxic SD-induced disruption of the dendritic mitochondrial structure that might impact mitochondrial bioenergetics during migraine with aura.
Collapse
|
9
|
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: 3] [Impact Index Per Article: 3.0] [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
|
10
|
Walther J, Kirsch EM, Hellwig L, Schmerbeck SS, Holloway PM, Buchan AM, Mergenthaler P. Reinventing the Penumbra - the Emerging Clockwork of a Multi-modal Mechanistic Paradigm. Transl Stroke Res 2023; 14:643-666. [PMID: 36219377 PMCID: PMC10444697 DOI: 10.1007/s12975-022-01090-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/25/2022]
Abstract
The concept of the ischemic penumbra was originally defined as the area around a necrotic stroke core and seen as the tissue at imminent risk of further damage. Today, the penumbra is generally considered as time-sensitive hypoperfused brain tissue with decreased oxygen and glucose availability, salvageable tissue as treated by intervention, and the potential target for neuroprotection in focal stroke. The original concept entailed electrical failure and potassium release but one short of neuronal cell death and was based on experimental stroke models, later confirmed in clinical imaging studies. However, even though the basic mechanisms have translated well, conferring brain protection, and improving neurological outcome after stroke based on the pathophysiological mechanisms in the penumbra has yet to be achieved. Recent findings shape the modern understanding of the penumbra revealing a plethora of molecular and cellular pathophysiological mechanisms. We now propose a new model of the penumbra, one which we hope will lay the foundation for future translational success. We focus on the availability of glucose, the brain's central source of energy, and bioenergetic failure as core pathophysiological concepts. We discuss the relation of mitochondrial function in different cell types to bioenergetics and apoptotic cell death mechanisms, autophagy, and neuroinflammation, to glucose metabolism in what is a dynamic ischemic penumbra.
Collapse
Affiliation(s)
- Jakob Walther
- Charité - Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Elena Marie Kirsch
- Charité - Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Lina Hellwig
- Charité - Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Sarah S Schmerbeck
- Charité - Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Paul M Holloway
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK
| | - Alastair M Buchan
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK.
| | - Philipp Mergenthaler
- Charité - Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, NeuroCure Clinical Research Center, Charitéplatz 1, 10117, Berlin, Germany.
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK.
| |
Collapse
|
11
|
Sugimoto K, Yang J, Fischer P, Takizawa T, Mulder I, Qin T, Erdogan TD, Yaseen MA, Sakadžić S, Chung DY, Ayata C. Optogenetic Spreading Depolarizations Do Not Worsen Acute Ischemic Stroke Outcome. Stroke 2023; 54:1110-1119. [PMID: 36876481 PMCID: PMC10050120 DOI: 10.1161/strokeaha.122.041351] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 02/01/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND Spreading depolarizations (SDs) are believed to contribute to injury progression and worsen outcomes in focal cerebral ischemia because exogenously induced SDs have been associated with enlarged infarct volumes. However, previous studies used highly invasive methods to trigger SDs that can directly cause tissue injury (eg, topical KCl) and confound the interpretation. Here, we tested whether SDs indeed enlarge infarcts when induced via a novel, noninjurious method using optogenetics. METHODS Using transgenic mice expressing channelrhodopsin-2 in neurons (Thy1-ChR2-YFP), we induced 8 optogenetic SDs to trigger SDs noninvasively at a remote cortical location in a noninjurious manner during 1-hour distal microvascular clip or proximal an endovascular filament occlusion of the middle cerebral artery. Laser speckle imaging was used to monitor cerebral blood flow. Infarct volumes were then quantified at 24 or 48 hours. RESULTS Infarct volumes in the optogenetic SD arm did not differ from the control arm in either distal or proximal middle cerebral artery occlusion, despite a 6-fold and 4-fold higher number of SDs, respectively. Identical optogenetic illumination in wild-type mice did not affect the infarct volume. Full-field laser speckle imaging showed that optogenetic stimulation did not affect the perfusion in the peri-infarct cortex. CONCLUSIONS Altogether, these data show that SDs induced noninvasively using optogenetics do not worsen tissue outcomes. Our findings compel a careful reexamination of the notion that SDs are causally linked to infarct expansion.
Collapse
Affiliation(s)
- Kazutaka Sugimoto
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
- Department of Neurosurgery, Yamaguchi University School of Medicine, Ube, Yamaguchi 7558505, Japan
| | - Joanna Yang
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| | - Paul Fischer
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| | - Tsubasa Takizawa
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| | - Inge Mulder
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| | - Tao Qin
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| | - Taylan D. Erdogan
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| | - Mohammad A. Yaseen
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129
| | - Sava Sakadžić
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129
| | - David Y. Chung
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Cenk Ayata
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| |
Collapse
|
12
|
Alkhachroum A, Appavu B, Egawa S, Foreman B, Gaspard N, Gilmore EJ, Hirsch LJ, Kurtz P, Lambrecq V, Kromm J, Vespa P, Zafar SF, Rohaut B, Claassen J. Electroencephalogram in the intensive care unit: a focused look at acute brain injury. Intensive Care Med 2022; 48:1443-1462. [PMID: 35997792 PMCID: PMC10008537 DOI: 10.1007/s00134-022-06854-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/31/2022] [Indexed: 02/04/2023]
Abstract
Over the past decades, electroencephalography (EEG) has become a widely applied and highly sophisticated brain monitoring tool in a variety of intensive care unit (ICU) settings. The most common indication for EEG monitoring currently is the management of refractory status epilepticus. In addition, a number of studies have associated frequent seizures, including nonconvulsive status epilepticus (NCSE), with worsening secondary brain injury and with worse outcomes. With the widespread utilization of EEG (spot and continuous EEG), rhythmic and periodic patterns that do not fulfill strict seizure criteria have been identified, epidemiologically quantified, and linked to pathophysiological events across a wide spectrum of critical and acute illnesses, including acute brain injury. Increasingly, EEG is not just qualitatively described, but also quantitatively analyzed together with other modalities to generate innovative measurements with possible clinical relevance. In this review, we discuss the current knowledge and emerging applications of EEG in the ICU, including seizure detection, ischemia monitoring, detection of cortical spreading depolarizations, assessment of consciousness and prognostication. We also review some technical aspects and challenges of using EEG in the ICU including the logistics of setting up ICU EEG monitoring in resource-limited settings.
Collapse
Affiliation(s)
- Ayham Alkhachroum
- Department of Neurology, University of Miami, Miami, FL, USA
- Department of Neurology, Jackson Memorial Hospital, Miami, FL, USA
| | - Brian Appavu
- Department of Child Health and Neurology, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
- Department of Neurosciences, Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Satoshi Egawa
- Neurointensive Care Unit, Department of Neurosurgery, and Stroke and Epilepsy Center, TMG Asaka Medical Center, Saitama, Japan
| | - Brandon Foreman
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, USA
| | - Nicolas Gaspard
- Department of Neurology, Erasme Hospital, Free University of Brussels, Brussels, Belgium
| | - Emily J Gilmore
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Neurocritical Care and Emergency Neurology, Department of Neurology, Ale University School of Medicine, New Haven, CT, USA
| | - Lawrence J Hirsch
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Pedro Kurtz
- Department of Intensive Care Medicine, D'or Institute for Research and Education, Rio de Janeiro, Brazil
- Neurointensive Care, Paulo Niemeyer State Brain Institute, Rio de Janeiro, Brazil
| | - Virginie Lambrecq
- Department of Clinical Neurophysiology and Epilepsy Unit, AP-HP, Pitié Salpêtrière Hospital, Reference Center for Rare Epilepsies, 75013, Paris, France
| | - Julie Kromm
- Departments of Critical Care Medicine and Clinical Neurosciences, Cumming School of Medicine, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, Calgary, AB, Canada
| | - Paul Vespa
- Brain Injury Research Center, Department of Neurosurgery, University of California, Los Angeles, USA
| | - Sahar F Zafar
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Benjamin Rohaut
- Department of Neurology, Sorbonne Université, Pitié-Salpêtrière-AP-HP and Paris Brain Institute, ICM, Inserm, CNRS, Paris, France
| | - Jan Claassen
- Department of Neurology, Neurological Institute, Columbia University, New York Presbyterian Hospital, 177 Fort Washington Avenue, MHB 8 Center, Room 300, New York, NY, 10032, USA.
| |
Collapse
|
13
|
Fernández-Serra R, Martínez-Alonso E, Alcázar A, Chioua M, Marco-Contelles J, Martínez-Murillo R, Ramos M, Guinea GV, González-Nieto D. Postischemic Neuroprotection of Aminoethoxydiphenyl Borate Associates Shortening of Peri-Infarct Depolarizations. Int J Mol Sci 2022; 23:ijms23137449. [PMID: 35806455 PMCID: PMC9266990 DOI: 10.3390/ijms23137449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/29/2022] [Accepted: 07/03/2022] [Indexed: 11/28/2022] Open
Abstract
Brain stroke is a highly prevalent pathology and a main cause of disability among older adults. If not promptly treated with recanalization therapies, primary and secondary mechanisms of injury contribute to an increase in the lesion, enhancing neurological deficits. Targeting excitotoxicity and oxidative stress are very promising approaches, but only a few compounds have reached the clinic with relatively good positive outcomes. The exploration of novel targets might overcome the lack of clinical translation of previous efficient preclinical neuroprotective treatments. In this study, we examined the neuroprotective properties of 2-aminoethoxydiphenyl borate (2-APB), a molecule that interferes with intracellular calcium dynamics by the antagonization of several channels and receptors. In a permanent model of cerebral ischemia, we showed that 2-APB reduces the extent of the damage and preserves the functionality of the cortical territory, as evaluated by somatosensory evoked potentials (SSEPs). While in this permanent ischemia model, the neuroprotective effect exerted by the antioxidant scavenger cholesteronitrone F2 was associated with a reduction in reactive oxygen species (ROS) and better neuronal survival in the penumbra, 2-APB did not modify the inflammatory response or decrease the content of ROS and was mostly associated with a shortening of peri-infarct depolarizations, which translated into better cerebral blood perfusion in the penumbra. Our study highlights the potential of 2-APB to target spreading depolarization events and their associated inverse hemodynamic changes, which mainly contribute to extension of the area of lesion in cerebrovascular pathologies.
Collapse
Affiliation(s)
- Rocío Fernández-Serra
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Madrid, Spain; (R.F.-S.); (M.R.); (G.V.G.)
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Silk Biomed SL, 28260 Madrid, Spain
| | - Emma Martínez-Alonso
- Department of Research, Hospital Universitario Ramón y Cajal, IRYCIS, 28034 Madrid, Spain; (E.M.-A.); (A.A.)
| | - Alberto Alcázar
- Department of Research, Hospital Universitario Ramón y Cajal, IRYCIS, 28034 Madrid, Spain; (E.M.-A.); (A.A.)
| | - Mourad Chioua
- Laboratory of Medicinal Chemistry, Institute of General Organic Chemistry (CSIC), 28006 Madrid, Spain; (M.C.); (J.M.-C.)
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry, Institute of General Organic Chemistry (CSIC), 28006 Madrid, Spain; (M.C.); (J.M.-C.)
| | | | - Milagros Ramos
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Madrid, Spain; (R.F.-S.); (M.R.); (G.V.G.)
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Gustavo V. Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Madrid, Spain; (R.F.-S.); (M.R.); (G.V.G.)
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Silk Biomed SL, 28260 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Biomaterials and Regenerative Medicine Group, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Madrid, Spain; (R.F.-S.); (M.R.); (G.V.G.)
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Silk Biomed SL, 28260 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-910679280
| |
Collapse
|
14
|
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]
|
15
|
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: 14] [Impact Index Per Article: 4.7] [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
|
16
|
Microglia Modulate Cortical Spreading Depolarizations After Ischemic Stroke: A Narrative Review. Neurocrit Care 2022; 37:133-138. [PMID: 35288861 PMCID: PMC9259539 DOI: 10.1007/s12028-022-01469-4] [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: 09/14/2021] [Accepted: 02/08/2022] [Indexed: 01/06/2023]
Abstract
Cortical spreading depolarizations (CSDs) are characterized by waves of diminished electroencephalography activity that propagate across the cortex with subsequent loss of ionic homeostasis. CSDs have been found in many pathological conditions, including migraine, traumatic brain injury, and ischemic stroke. Because of CSD-associated ionic and metabolic disturbances at the peri-infarct area after ischemic stroke, it is thought that CSDs exacerbate tissue infarction and worsen clinical outcomes. Microglia, the main innate immune cells in the brain, are among the first responders to brain tissue damage. Recent studies demonstrated that microglia play a critical role in CSD initiation and propagation. In this article, we discuss the significance of CSD in the setting of ischemic stroke and how microglia may modulate peri-infarct CSDs, also known as iso-electric depolarizations. Finally, we discuss the significance of microglial Ca2+ and how it might be used as a potential therapeutic target for patients with ischemic stroke.
Collapse
|
17
|
Lemale CL, Lückl J, Horst V, Reiffurth C, Major S, Hecht N, Woitzik J, Dreier JP. Migraine Aura, Transient Ischemic Attacks, Stroke, and Dying of the Brain Share the Same Key Pathophysiological Process in Neurons Driven by Gibbs–Donnan Forces, Namely Spreading Depolarization. Front Cell Neurosci 2022; 16:837650. [PMID: 35237133 PMCID: PMC8884062 DOI: 10.3389/fncel.2022.837650] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/19/2022] [Indexed: 12/15/2022] Open
Abstract
Neuronal cytotoxic edema is the morphological correlate of the near-complete neuronal battery breakdown called spreading depolarization, or conversely, spreading depolarization is the electrophysiological correlate of the initial, still reversible phase of neuronal cytotoxic edema. Cytotoxic edema and spreading depolarization are thus different modalities of the same process, which represents a metastable universal reference state in the gray matter of the brain close to Gibbs–Donnan equilibrium. Different but merging sections of the spreading-depolarization continuum from short duration waves to intermediate duration waves to terminal waves occur in a plethora of clinical conditions, including migraine aura, ischemic stroke, traumatic brain injury, aneurysmal subarachnoid hemorrhage (aSAH) and delayed cerebral ischemia (DCI), spontaneous intracerebral hemorrhage, subdural hematoma, development of brain death, and the dying process during cardio circulatory arrest. Thus, spreading depolarization represents a prime and simultaneously the most neglected pathophysiological process in acute neurology. Aristides Leão postulated as early as the 1940s that the pathophysiological process in neurons underlying migraine aura is of the same nature as the pathophysiological process in neurons that occurs in response to cerebral circulatory arrest, because he assumed that spreading depolarization occurs in both conditions. With this in mind, it is not surprising that patients with migraine with aura have about a twofold increased risk of stroke, as some spreading depolarizations leading to the patient percept of migraine aura could be caused by cerebral ischemia. However, it is in the nature of spreading depolarization that it can have different etiologies and not all spreading depolarizations arise because of ischemia. Spreading depolarization is observed as a negative direct current (DC) shift and associated with different changes in spontaneous brain activity in the alternating current (AC) band of the electrocorticogram. These are non-spreading depression and spreading activity depression and epileptiform activity. The same spreading depolarization wave may be associated with different activity changes in adjacent brain regions. Here, we review the basal mechanism underlying spreading depolarization and the associated activity changes. Using original recordings in animals and patients, we illustrate that the associated changes in spontaneous activity are by no means trivial, but pose unsolved mechanistic puzzles and require proper scientific analysis.
Collapse
Affiliation(s)
- Coline L. Lemale
- Center for Stroke Research Berlin, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Experimental Neurology, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Janos Lückl
- Center for Stroke Research Berlin, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
- Department of Neurology, University of Szeged, Szeged, Hungary
| | - Viktor Horst
- Center for Stroke Research Berlin, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Clemens Reiffurth
- Center for Stroke Research Berlin, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Experimental Neurology, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sebastian Major
- Center for Stroke Research Berlin, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Experimental Neurology, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Neurology, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nils Hecht
- Department of Neurosurgery, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Johannes Woitzik
- Department of Neurosurgery, Evangelisches Krankenhaus Oldenburg, University of Oldenburg, Oldenburg, Germany
| | - Jens P. Dreier
- Center for Stroke Research Berlin, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Experimental Neurology, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Neurology, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
- *Correspondence: Jens P. Dreier,
| |
Collapse
|
18
|
del Zoppo GJ, Moskowitz MA, Nedergaard M. The Neurovascular Unit and Responses to Ischemia. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00007-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
19
|
Bioactive Flavonoids Icaritin and Icariin Protect against Cerebral Ischemia-Reperfusion-Associated Apoptosis and Extracellular Matrix Accumulation in an Ischemic Stroke Mouse Model. Biomedicines 2021; 9:biomedicines9111719. [PMID: 34829948 PMCID: PMC8615444 DOI: 10.3390/biomedicines9111719] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/10/2021] [Accepted: 11/18/2021] [Indexed: 01/01/2023] Open
Abstract
Stroke, which is the second leading cause of mortality in the world, is urgently needed to explore the medical strategies for ischemic stroke treatment. Both icariin (ICA) and icaritin (ICT) are the major active flavonoids extracted from Herba epimedii that have been regarded as the neuroprotective agents in disease models. In this study, we aimed to investigate and compare the neuroprotective effects of ICA and ICT in a middle cerebral artery occlusion (MCAO) mouse model. Male ICR mice were pretreated with both ICA and ICT, which ameliorated body weight loss, neurological injury, infarct volume, and pathological change in acute ischemic stroke mice. Furthermore, administration of both ICA and ICT could also protect against neuronal cell apoptotic death, oxidative and nitrosative stress, lipid peroxidation, and extracellular matrix (ECM) accumulation in the brains. The neuroprotective effects of ICT are slightly better than that of ICA in acute cerebral ischemic stroke mice. These results suggest that pretreatment with both ICA and ICT improves the neuronal cell apoptosis and responses of oxidative/nitrosative stress and counteracts the ECM accumulation in the brains of acute cerebral ischemic stroke mice. Both ICA and ICT treatment may serve as a useful therapeutic strategy for acute ischemic stroke.
Collapse
|
20
|
Faillot M, Chaillet A, Palfi S, Senova S. Rodent models used in preclinical studies of deep brain stimulation to rescue memory deficits. Neurosci Biobehav Rev 2021; 130:410-432. [PMID: 34437937 DOI: 10.1016/j.neubiorev.2021.08.012] [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: 02/08/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 11/28/2022]
Abstract
Deep brain stimulation paradigms might be used to treat memory disorders in patients with stroke or traumatic brain injury. However, proof of concept studies in animal models are needed before clinical translation. We propose here a comprehensive review of rodent models for Traumatic Brain Injury and Stroke. We systematically review the histological, behavioral and electrophysiological features of each model and identify those that are the most relevant for translational research.
Collapse
Affiliation(s)
- Matthieu Faillot
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Antoine Chaillet
- Laboratoire des Signaux et Systèmes (L2S-UMR8506) - CentraleSupélec, Université Paris Saclay, Institut Universitaire de France, France
| | - Stéphane Palfi
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Suhan Senova
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France.
| |
Collapse
|
21
|
Petzold GC, Dreier JP. Spreading depolarization evoked by endothelin-1 is inhibited by octanol but not by carbenoxolone. BRAIN HEMORRHAGES 2021. [DOI: 10.1016/j.hest.2020.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
22
|
Telles JPM, Welling LC, Coelho ACSDS, Rabelo NN, Teixeira MJ, Figueiredo EG. Cortical spreading depolarization and ketamine: a short systematic review. Neurophysiol Clin 2021; 51:145-151. [PMID: 33610431 DOI: 10.1016/j.neucli.2021.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Cortical spreading depolarization (SD) describes pathological waves characterized by an almost complete sustained depolarization of neurons and astrocytes that spreads throughout the cortex. In this study, we carried out a qualitative review of all available evidence, clinical and preclinical, on the use of ketamine in SD. METHODS We performed a systematic review of Medline, with no restrictions regarding publishing date or language, in search of articles reporting the use of ketamine in SD. The search string was composed of "ketamine," "spreading," "depolarization," and "depression" in both (AND) and (OR) combinations. RESULTS Twenty studies were included in the final synthesis. Many studies showed that ketamine effectively blocks SD in rats, swine, and humans. The first prospective randomized trial was published in 2018. Ten patients with severe traumatic brain injury or subarachnoid hemorrhage were enrolled, and ketamine showed a significant, dose-dependent effect on the reduction of SD. CONCLUSION The available evidence from preclinical studies is helping to translate the role of ketamine in blocking spreading depolarizations to clinical practice, in the settings of migraine with aura, traumatic brain injury, subarachnoid hemorrhage, and hemorrhagic and ischemic stroke. More randomized controlled trials are needed to determine whether interrupting the ketamine-blockable SDs effectively leads to an improvement in outcome and to assess the real occurrence of adverse effects.
Collapse
Affiliation(s)
- João Paulo Mota Telles
- Division of Neurosurgery, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HC-FMUSP), Brazil
| | | | | | - Nícollas Nunes Rabelo
- Division of Neurosurgery, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HC-FMUSP), Brazil
| | - Manoel Jacobsen Teixeira
- Division of Neurosurgery, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HC-FMUSP), Brazil
| | - Eberval Gadelha Figueiredo
- Division of Neurosurgery, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HC-FMUSP), Brazil.
| |
Collapse
|
23
|
Gamma frequency activation of inhibitory neurons in the acute phase after stroke attenuates vascular and behavioral dysfunction. Cell Rep 2021; 34:108696. [PMID: 33535035 DOI: 10.1016/j.celrep.2021.108696] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/06/2020] [Accepted: 01/06/2021] [Indexed: 11/24/2022] Open
Abstract
Alterations in gamma oscillations occur in several neurological disorders, and the entrainment of gamma oscillations has been recently proposed as a treatment for neurodegenerative disease. Optogenetic stimulation enhances recovery in models of stroke when applied weeks after injury; however, the benefits of acute brain stimulation have not been investigated. Here, we report beneficial effects of gamma-frequency modulation in the acute phase, within 1 h, after stroke. Transgenic VGAT-ChR2 mice are subject to awake photothrombotic stroke in an area encompassing the forelimb sensory and motor cortex. Optogenetic stimulation at 40 Hz in the peri-infarct zone recovers neuronal activity 24 h after stroke in motor and parietal association areas, as well as blood flow over the first week after stroke. Stimulation significantly reduces lesion volume and improves motor function. Our results suggest that acute-phase modulation of cortical oscillatory dynamics may serve as a target for neuroprotection against stroke.
Collapse
|
24
|
MartInez-Coria H, Arrieta-Cruz I, Cruz ME, López-Valdés HE. Physiopathology of ischemic stroke and its modulation using memantine: evidence from preclinical stroke. Neural Regen Res 2021; 16:433-439. [PMID: 32985462 PMCID: PMC7996012 DOI: 10.4103/1673-5374.293129] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Ischemic stroke is the most common type of cerebrovascular disease and is caused by an interruption of blood flow in the brain. In this disease, two different damage areas are identifying: the lesion core, in which cells quickly die; and the penumbra (surrounding the lesion core), in which cells are functionally weakened but may recover and restore their functions. The currently approved treatments for ischemic stroke are the recombinant tissue plasminogen activator and endovascular thrombectomy, but they have a short therapeutic window (4.5 and 6 hours after stroke onset, respectively) and a low percentage of stroke patients actually receive these treatments. Memantine is an approved drug for the treatment of Alzheimer’s disease. Memantine is a noncompetitive, low affinity and use-dependent antagonist of N-methyl-D-aspartate glutamate receptor. Memantine has several advantages over developing a new drug to treat focal ischemic stroke, but the most important is that it has sufficient safe probes in preclinical models and humans, and if the preclinical studies provide more evidence about pharmacological actions in tissue protection and repair, this could help to increase the number of clinical trials. The present review summarizes the physiopathology of isquemic stroke and the pharmacological actions in neuroprotection and neuroplasticity of memantine in the post stroke stage of preclinical stroke models, to illustrate their potential to improve functional recovery in human patients.
Collapse
Affiliation(s)
- Hilda MartInez-Coria
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM); Laboratorio Experimental de Enfermedades Neurodegenerativas, Facultad de Medicina, UNAM/Instituto Nacional de Neurología y Neurocirugía, Ciudad de México, México
| | - Isabel Arrieta-Cruz
- Departamento de Investigación Básica, Instituto Nacional de Geriatría, Ciudad de México, México
| | - María-Esther Cruz
- Unidad de Investigación en Biología de la Reproducción, Laboratorio de Neuroendocrinología, Facultad de Estudios Superiores Zaragoza, UNAM, Ciudad de México, México
| | - Héctor E López-Valdés
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM); Unidad Periférica de Neurociencia, Facultad de Medicina, UNAM/Instituto Nacional de Neurología y Neurocirugía, Ciudad de México, México
| |
Collapse
|
25
|
Sueiras M, Thonon V, Santamarina E, Sánchez-Guerrero Á, Riveiro M, Poca MA, Quintana M, Gándara D, Sahuquillo J. Is Spreading Depolarization a Risk Factor for Late Epilepsy? A Prospective Study in Patients with Traumatic Brain Injury and Malignant Ischemic Stroke Undergoing Decompressive Craniectomy. Neurocrit Care 2020; 34:876-888. [PMID: 33000378 DOI: 10.1007/s12028-020-01107-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 09/05/2020] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Spreading depolarizations (SDs) have been described in patients with ischemic and haemorrhagic stroke, traumatic brain injury, and migraine with aura, among other conditions. The exact pathophysiological mechanism of SDs is not yet fully established. Our aim in this study was to evaluate the relationship between the electrocorticography (ECoG) findings of SDs and/or epileptiform activity and subsequent epilepsy and electroclinical outcome. METHODS This was a prospective observational study of 39 adults, 17 with malignant middle cerebral artery infarction (MMCAI) and 22 with traumatic brain injury, who underwent decompressive craniectomy and multimodal neuromonitoring including ECoG in penumbral tissue. Serial electroencephalography (EEG) recordings were obtained for all surviving patients. Functional disability at 6 and 12 months after injury were assessed using the Barthel, modified Rankin (mRS), and Extended Glasgow Outcome (GOS-E) scales. RESULTS SDs were recorded in 58.9% of patients, being more common-particularly those of isoelectric type-in patients with MMCAI (p < 0.04). At follow-up, 74.7% of patients had epileptiform abnormalities on EEG and/or seizures. A significant correlation was observed between the degree of preserved brain activity on EEG and disability severity (R [mRS]: + 0.7, R [GOS-E, Barthel]: - 0.6, p < 0.001), and between the presence of multifocal epileptiform abnormalities on EEG and more severe disability on the GOS-E at 6 months (R: - 0.3, p = 0.03) and 12 months (R: - 0.3, p = 0.05). Patients with more SDs and higher depression ratios scored worse on the GOS-E (R: - 0.4 at 6 and 12 months) and Barthel (R: - 0.4 at 6 and 12 months) disability scales (p < 0.05). The number of SDs (p = 0.064) and the depression ratio (p = 0.1) on ECoG did not show a statistically significant correlation with late epilepsy. CONCLUSIONS SDs are common in the cortex of ischemic or traumatic penumbra. Our study suggests an association between the presence of SDs in the acute phase and worse long-term outcome, although no association with subsequent epilepsy was found. More comprehensive studies, involving ECoG and EEG could help determine their association with epileptogenesis.
Collapse
Affiliation(s)
- Maria Sueiras
- Department of Clinical Neurophysiology, Vall d'Hebron University Hospital, Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain. .,Neurotrauma and Neurosurgery Research Unit (UNINN), Vall d'Hebron Research Institute (VHIR), Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain. .,Universitat Autònoma de Barcelona (UAB), Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain.
| | - Vanessa Thonon
- Department of Clinical Neurophysiology, Vall d'Hebron University Hospital, Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Estevo Santamarina
- Epilepsy Unit, Department of Neurology, Vall d'Hebron University Hospital, Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Ángela Sánchez-Guerrero
- Neurotrauma and Neurosurgery Research Unit (UNINN), Vall d'Hebron Research Institute (VHIR), Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Marilyn Riveiro
- Neurotrauma Intensive Care Unit, Vall d'Hebron University Hospital, Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Maria-Antonia Poca
- Neurotrauma and Neurosurgery Research Unit (UNINN), Vall d'Hebron Research Institute (VHIR), Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain.,Universitat Autònoma de Barcelona (UAB), Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain.,Department of Neurosurgery, Vall d'Hebron University Hospital, Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Manuel Quintana
- Epilepsy Unit, Department of Neurology, Vall d'Hebron University Hospital, Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Dario Gándara
- Neurotrauma and Neurosurgery Research Unit (UNINN), Vall d'Hebron Research Institute (VHIR), Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain.,Department of Neurosurgery, Vall d'Hebron University Hospital, Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Juan Sahuquillo
- Neurotrauma and Neurosurgery Research Unit (UNINN), Vall d'Hebron Research Institute (VHIR), Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain.,Universitat Autònoma de Barcelona (UAB), Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain.,Department of Neurosurgery, Vall d'Hebron University Hospital, Paseo Vall d'Hebron 119-129, 08035, Barcelona, Spain
| |
Collapse
|
26
|
Ashayeri Ahmadabad R, Khaleghi Ghadiri M, Gorji A. The role of Toll-like receptor signaling pathways in cerebrovascular disorders: the impact of spreading depolarization. J Neuroinflammation 2020; 17:108. [PMID: 32264928 PMCID: PMC7140571 DOI: 10.1186/s12974-020-01785-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/24/2020] [Indexed: 02/08/2023] Open
Abstract
Cerebral vascular diseases (CVDs) are a group of disorders that affect the blood supply to the brain and lead to the reduction of oxygen and glucose supply to the neurons and the supporting cells. Spreading depolarization (SD), a propagating wave of neuroglial depolarization, occurs in different CVDs. A growing amount of evidence suggests that the inflammatory responses following hypoxic-ischemic insults and after SD plays a double-edged role in brain tissue injury and clinical outcome; a beneficial effect in the acute phase and a destructive role in the late phase. Toll-like receptors (TLRs) play a crucial role in the activation of inflammatory cascades and subsequent neuroprotective or harmful effects after CVDs and SD. Here, we review current data regarding the pathophysiological role of TLR signaling pathways in different CVDs and discuss the role of SD in the potentiation of the inflammatory cascade in CVDs through the modulation of TLRs.
Collapse
Affiliation(s)
- Rezan Ashayeri Ahmadabad
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
- Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | | | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany.
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster, Germany.
- Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany.
- Neuroscience research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
27
|
Shibata K, Hashimoto T, Miyazaki T, Miyazaki A, Nobe K. Thrombolytic Therapy for Acute Ischemic Stroke: Past and Future. Curr Pharm Des 2020; 25:242-250. [PMID: 30892155 DOI: 10.2174/1381612825666190319115018] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/13/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Thromboembolic ischemic stroke, which is mainly caused by hypertension, as well as plasma dyslipidemia, arterial fibrillation and diabetes, is a leading cause of death in the US and other countries. Numerous clinical trials for thrombolytic drugs, which aimed to pharmacologically dissolve thrombi, were conducted in the 1950s, when the first thrombolytic therapy was performed. METHODS In this study, we summarize the pathophysiologic features of ischemic stroke, and the history of thrombolytic therapy, and discuss the recent progress that has been made in the ongoing development of thrombolytic drugs. CONCLUSION Thrombolytic therapy is sometimes accompanied by harmful hemorrhagic insults; accordingly, a window of time wherein therapy can safely be performed has been established for this approach. Several basic and clinical studies are ongoing to develop next-generation thrombolytic drugs to expand the time window.
Collapse
Affiliation(s)
- Keita Shibata
- Division of Pharmacology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Terumasa Hashimoto
- Division of Pharmacology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Takuro Miyazaki
- Department of Biochemistry, Showa University School of Medicine, Shinagawaku, Tokyo 142-8555, Japan
| | - Akira Miyazaki
- Department of Biochemistry, Showa University School of Medicine, Shinagawaku, Tokyo 142-8555, Japan
| | - Koji Nobe
- Division of Pharmacology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| |
Collapse
|
28
|
Baron JC. Protecting the ischaemic penumbra as an adjunct to thrombectomy for acute stroke. Nat Rev Neurol 2019; 14:325-337. [PMID: 29674752 DOI: 10.1038/s41582-018-0002-2] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
After ischaemic stroke, brain damage can be curtailed by rescuing the 'ischaemic penumbra' - that is, the severely hypoperfused, at-risk but not yet infarcted tissue. Current evidence-based treatments involve restoration of blood flow so as to salvage the penumbra before it evolves into irreversibly damaged tissue, termed the 'core'. Intravenous thrombolysis (IVT) can salvage the penumbra if given within 4.5 h after stroke onset; however, the early recanalization rate is only ~30%. Direct removal of the occluding clot by mechanical thrombectomy considerably improves outcomes over IVT alone, but despite early recanalization in > 80% of cases, ~50% of patients who receive this treatment do not enjoy functional independence, usually because the core is already too large at the time of recanalization. Novel therapies aiming to 'freeze' the penumbra - that is, prevent core growth until recanalization is complete - hold potential as adjuncts to mechanical thrombectomy. This Review focuses on nonpharmacological approaches that aim to restore the physiological balance between oxygen delivery to and oxygen demand of the penumbra. Particular emphasis is placed on normobaric oxygen therapy, hypothermia and sensory stimulation. Preclinical evidence and early pilot clinical trials are critically reviewed, and future directions, including clinical translation and trial design issues, are discussed.
Collapse
Affiliation(s)
- Jean-Claude Baron
- Department of Neurology, Hôpital Sainte-Anne, Université Paris 5, INSERM U894, Paris, France.
| |
Collapse
|
29
|
Sudhakar SK, Choi TJ, Ahmed OJ. Biophysical Modeling Suggests Optimal Drug Combinations for Improving the Efficacy of GABA Agonists after Traumatic Brain Injuries. J Neurotrauma 2019; 36:1632-1645. [PMID: 30484362 PMCID: PMC6531909 DOI: 10.1089/neu.2018.6065] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Traumatic brain injuries (TBI) lead to dramatic changes in the surviving brain tissue. Altered ion concentrations, coupled with changes in the expression of membrane-spanning proteins, create a post-TBI brain state that can lead to further neuronal loss caused by secondary excitotoxicity. Several GABA receptor agonists have been tested in the search for neuroprotection immediately after an injury, with paradoxical results. These drugs not only fail to offer neuroprotection, but can also slow down functional recovery after TBI. Here, using computational modeling, we provide a biophysical hypothesis to explain these observations. We show that the accumulation of intracellular chloride ions caused by a transient upregulation of Na+-K+-2Cl- (NKCC1) co-transporters as observed following TBI, causes GABA receptor agonists to lead to excitation and depolarization block, rather than the expected hyperpolarization. The likelihood of prolonged, excitotoxic depolarization block is further exacerbated by the extremely high levels of extracellular potassium seen after TBI. Our modeling results predict that the neuroprotective efficacy of GABA receptor agonists can be substantially enhanced when they are combined with NKCC1 co-transporter inhibitors. This suggests a rational, biophysically principled method for identifying drug combinations for neuroprotection after TBI.
Collapse
Affiliation(s)
| | - Thomas J. Choi
- Department of Psychology, University of Michigan, Ann Arbor, Michigan
| | - Omar J. Ahmed
- Department of Psychology, University of Michigan, Ann Arbor, Michigan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
- Department of Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan
- Department of Kresge Hearing Research Institute, University of Michigan, Ann Arbor, Michigan
- Department of Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
30
|
Taş YÇ, Solaroğlu İ, Gürsoy-Özdemir Y. Spreading Depolarization Waves in Neurological Diseases: A Short Review about its Pathophysiology and Clinical Relevance. Curr Neuropharmacol 2019; 17:151-164. [PMID: 28925885 PMCID: PMC6343201 DOI: 10.2174/1570159x15666170915160707] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/03/2017] [Accepted: 09/09/2017] [Indexed: 02/05/2023] Open
Abstract
Lesion growth following acutely injured brain tissue after stroke, subarachnoid hemorrhage and traumatic brain injury is an important issue and a new target area for promising therapeutic interventions. Spreading depolarization or peri-lesion depolarization waves were demonstrated as one of the significant contributors of continued lesion growth. In this short review, we discuss the pathophysiology for SD forming events and try to list findings detected in neurological disorders like migraine, stroke, subarachnoid hemorrhage and traumatic brain injury in both human as well as experimental studies. Pharmacological and non-pharmacological treatment strategies are highlighted and future directions and research limitations are discussed.
Collapse
Affiliation(s)
| | | | - Yasemin Gürsoy-Özdemir
- Address correspondence to these authors at the Department of Neurosurgery, School of Medicine, Koç University, İstanbul, Turkey; Tel: +90 850 250 8250; E-mails: ,
| |
Collapse
|
31
|
Hobbs CN, Johnson JA, Verber MD, Mark Wightman R. An implantable multimodal sensor for oxygen, neurotransmitters, and electrophysiology during spreading depolarization in the deep brain. Analyst 2018; 142:2912-2920. [PMID: 28715004 DOI: 10.1039/c7an00508c] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Brain tissue injury is often accompanied by spreading depolarization (SD) events, marked by widespread cellular depolarization and cessation of neuronal firing. SD recruits viable tissue into the lesion, making it a focus for intervention. During SD, drastic fluctuations occur in ion gradients, extracellular neurotransmitter concentrations, cellular metabolism, and cerebral blood flow. Measuring SD requires a multimodal approach to capture the array of changes. However, the use of multiple sensors can inflict tissue damage. Here, we use carbon-fiber microelectrodes to characterize several aspects of SD with a single, minimally invasive sensor in the deep brain region of the nucleus accumbens. Fast-scan cyclic voltammetry detects large changes in oxygen, which reflect the balance between cerebral blood flow and energy consumption, and also supraphysiological release of electroactive neurotransmitters (i.e., dopamine). We verify waves of SD with concurrent single-unit or DC potential electrophysiological recordings. The single-unit recordings reveal bursts of action potentials followed by inactivity. The DC potentials exhibit a slow negative voltage shift in the extracellular space indicative of wide-spread cellular depolarization. Here, we characterize the multiple modalities of our sensor and demonstrate its utility for improved SD recordings.
Collapse
Affiliation(s)
- Caddy N Hobbs
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | | | | | | |
Collapse
|
32
|
Pruvost-Robieux E, Calvet D, Ben Hassen W, Turc G, Marchi A, Mélé N, Seners P, Oppenheim C, Baron JC, Mas JL, Gavaret M. Design and Methodology of a Pilot Randomized Controlled Trial of Transcranial Direct Current Stimulation in Acute Middle Cerebral Artery Stroke (STICA). Front Neurol 2018; 9:816. [PMID: 30356889 PMCID: PMC6190876 DOI: 10.3389/fneur.2018.00816] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 09/10/2018] [Indexed: 01/01/2023] Open
Abstract
Background: Stroke is a major cause of death and disability worldwide. The related burden is expected to further increase due to aging populations, calling for more efficient treatment. Ischemic stroke results from a focal reduction in cerebral blood flow due to the sudden occlusion of a brain artery. Ischemic brain injury results from a sequence of pathophysiological events that evolve over time and space. This cascade includes excitotoxicity and peri-infarct depolarizations (PIDs). Focal impairment of cerebral blood flow restricts the delivery of energetics substrates and impairs ionic gradients. Membrane potential is eventually lost, and neurons depolarize. Although recanalization therapies target the ischemic penumbra, they can only rescue the penumbra still present at the time of reperfusion. A promising novel approach is to "freeze" the penumbra until reperfusion occurs. Transcranial direct current stimulation (tDCS) is a non-invasive method of neuromodulation. Based on preclinical evidence, we propose to test the penumbra freezing concept in a clinical phase IIa trial assessing whether cathodal tDCS-shown in rodents to reduce infarction volume-prevents early infarct growth in human acute Middle Cerebral Artery (MCA) stroke, in adjunction to conventional revascularization methods. Methods: This is a monocentric randomized, double-blind, and placebo-controlled trial performed in patients with acute MCA stroke eligible to revascularization procedures. Primary outcome is infarct volume growth on diffusion weighted imaging (DWI) at day 1 relative to baseline. Secondary outcomes include safety and clinical efficacy. Significance: Results from this clinical trial are expected to provide rationale for a phase III study. Clinical trial registration-EUDRACT: 2016-A00160-51.
Collapse
Affiliation(s)
- Estelle Pruvost-Robieux
- Department of Neurophysiology, Sainte-Anne Hospital, Paris, France.,Faculty of Medicine, Paris Descartes University, Paris, France
| | - David Calvet
- Faculty of Medicine, Paris Descartes University, Paris, France.,INSERM UMR S894, Paris, France.,Department of Neurology, Sainte-Anne Hospital, Paris, France
| | - Wagih Ben Hassen
- Faculty of Medicine, Paris Descartes University, Paris, France.,INSERM UMR S894, Paris, France.,Department of Neuroradiology, Sainte-Anne Hospital, Paris, France
| | - Guillaume Turc
- Faculty of Medicine, Paris Descartes University, Paris, France.,INSERM UMR S894, Paris, France.,Department of Neurology, Sainte-Anne Hospital, Paris, France
| | - Angela Marchi
- Department of Neurophysiology, Sainte-Anne Hospital, Paris, France
| | - Nicolas Mélé
- Department of Neurology, Sainte-Anne Hospital, Paris, France
| | - Pierre Seners
- Faculty of Medicine, Paris Descartes University, Paris, France.,INSERM UMR S894, Paris, France.,Department of Neurology, Sainte-Anne Hospital, Paris, France
| | - Catherine Oppenheim
- Faculty of Medicine, Paris Descartes University, Paris, France.,INSERM UMR S894, Paris, France.,Department of Neuroradiology, Sainte-Anne Hospital, Paris, France
| | - Jean-Claude Baron
- Faculty of Medicine, Paris Descartes University, Paris, France.,INSERM UMR S894, Paris, France.,Department of Neurology, Sainte-Anne Hospital, Paris, France
| | - Jean-Louis Mas
- Faculty of Medicine, Paris Descartes University, Paris, France.,INSERM UMR S894, Paris, France.,Department of Neurology, Sainte-Anne Hospital, Paris, France
| | - Martine Gavaret
- Department of Neurophysiology, Sainte-Anne Hospital, Paris, France.,Faculty of Medicine, Paris Descartes University, Paris, France.,INSERM UMR S894, Paris, France
| |
Collapse
|
33
|
Pál B. Involvement of extrasynaptic glutamate in physiological and pathophysiological changes of neuronal excitability. Cell Mol Life Sci 2018; 75:2917-2949. [PMID: 29766217 PMCID: PMC11105518 DOI: 10.1007/s00018-018-2837-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/27/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
Glutamate is the most abundant neurotransmitter of the central nervous system, as the majority of neurons use glutamate as neurotransmitter. It is also well known that this neurotransmitter is not restricted to synaptic clefts, but found in the extrasynaptic regions as ambient glutamate. Extrasynaptic glutamate originates from spillover of synaptic release, as well as from astrocytes and microglia. Its concentration is magnitudes lower than in the synaptic cleft, but receptors responding to it have higher affinity for it. Extrasynaptic glutamate receptors can be found in neuronal somatodendritic location, on astroglia, oligodendrocytes or microglia. Activation of them leads to changes of neuronal excitability with different amplitude and kinetics. Extrasynaptic glutamate is taken up by neurons and astrocytes mostly via EAAT transporters, and astrocytes, in turn metabolize it to glutamine. Extrasynaptic glutamate is involved in several physiological phenomena of the central nervous system. It regulates neuronal excitability and synaptic strength by involving astroglia; contributing to learning and memory formation, neurosecretory and neuromodulatory mechanisms, as well as sleep homeostasis.The extrasynaptic glutamatergic system is affected in several brain pathologies related to excitotoxicity, neurodegeneration or neuroinflammation. Being present in dementias, neurodegenerative and neuropsychiatric diseases or tumor invasion in a seemingly uniform way, the system possibly provides a common component of their pathogenesis. Although parts of the system are extensively discussed by several recent reviews, in this review I attempt to summarize physiological actions of the extrasynaptic glutamate on neuronal excitability and provide a brief insight to its pathology for basic understanding of the topic.
Collapse
Affiliation(s)
- Balázs Pál
- Department of Physiology, Faculty of Medicine, University of Debrecen, Nagyerdei krt 98, Debrecen, 4012, Hungary.
| |
Collapse
|
34
|
Lückl J, Lemale CL, Kola V, Horst V, Khojasteh U, Oliveira-Ferreira AI, Major S, Winkler MKL, Kang EJ, Schoknecht K, Martus P, Hartings JA, Woitzik J, Dreier JP. The negative ultraslow potential, electrophysiological correlate of infarction in the human cortex. Brain 2018; 141:1734-1752. [PMID: 29668855 PMCID: PMC5972557 DOI: 10.1093/brain/awy102] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 01/20/2018] [Accepted: 02/17/2018] [Indexed: 12/19/2022] Open
Abstract
Spreading depolarizations are characterized by abrupt, near-complete breakdown of the transmembrane ion gradients, neuronal oedema, mitochondrial depolarization, glutamate excitotoxicity and activity loss (depression). Spreading depolarization induces either transient hyperperfusion in normal tissue; or hypoperfusion (inverse coupling = spreading ischaemia) in tissue at risk for progressive injury. The concept of the spreading depolarization continuum is critical since many spreading depolarizations have intermediate characteristics, as opposed to the two extremes of spreading depolarization in either severely ischaemic or normal tissue. In animals, the spreading depolarization extreme in ischaemic tissue is characterized by prolonged depolarization durations, in addition to a slow baseline variation termed the negative ultraslow potential. The negative ultraslow potential is initiated by spreading depolarization and similar to the negative direct current (DC) shift of prolonged spreading depolarization, but specifically refers to a negative potential component during progressive recruitment of neurons into cell death in the wake of spreading depolarization. We here first quantified the spreading depolarization-initiated negative ultraslow potential in the electrocorticographic DC range and the activity depression in the alternate current range after middle cerebral artery occlusion in rats. Relevance of these variables to the injury was supported by significant correlations with the cortical infarct volume and neurological outcome after 72 h of survival. We then identified negative ultraslow potential-containing clusters of spreading depolarizations in 11 patients with aneurysmal subarachnoid haemorrhage. The human platinum/iridium-recorded negative ultraslow potential showed a tent-like shape. Its amplitude of 45.0 (39.0, 69.4) mV [median (first, third quartile)] was 6.6 times larger and its duration of 3.7 (3.3, 5.3) h was 34.9 times longer than the negative DC shift of spreading depolarizations in less compromised tissue. Using Generalized Estimating Equations applied to a logistic regression model, we found that negative ultraslow potential displaying electrodes were significantly more likely to overlie a developing ischaemic lesion (90.0%, 27/30) than those not displaying a negative ultraslow potential (0.0%, 0/20) (P = 0.004). Based on serial neuroimages, the lesions under the electrodes developed within a time window of 72 (56, 134) h. The negative ultraslow potential occurred in this time window in 9/10 patients. It was often preceded by a spreading depolarization cluster with increasingly persistent spreading depressions and progressively prolonged DC shifts and spreading ischaemias. During the negative ultraslow potential, spreading ischaemia lasted for 40.0 (28.0, 76.5) min, cerebral blood flow fell from 57 (53, 65) % to 26 (16, 42) % (n = 4) and tissue partial pressure of oxygen from 12.5 (9.2, 15.2) to 3.3 (2.4, 7.4) mmHg (n = 5). Our data suggest that the negative ultraslow potential is the electrophysiological correlate of infarction in human cerebral cortex and a neuromonitoring-detected medical emergency.awy102media15775596049001.
Collapse
Affiliation(s)
- Janos Lückl
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Coline L Lemale
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Vasilis Kola
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Viktor Horst
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Uldus Khojasteh
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ana I Oliveira-Ferreira
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sebastian Major
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Neurology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Maren K L Winkler
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Eun-Jeung Kang
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Karl Schoknecht
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Peter Martus
- Institute for Clinical Epidemiology and Applied Biostatistics, University of Tübingen, Tübingen, Germany
| | - Jed A Hartings
- UC Gardner Neuroscience Institute, University of Cincinnati (UC) College of Medicine, Cincinnati, OH, USA
- Department of Neurosurgery, University of Cincinnati (UC) College of Medicine, Cincinnati, OH, USA
| | - Johannes Woitzik
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jens P Dreier
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Neurology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
| |
Collapse
|
35
|
Uncensored EEG: The role of DC potentials in neurobiology of the brain. Prog Neurobiol 2018; 165-167:51-65. [PMID: 29428834 DOI: 10.1016/j.pneurobio.2018.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/24/2017] [Accepted: 02/03/2018] [Indexed: 12/11/2022]
Abstract
Brain direct current (DC) potentials denote sustained shifts and slow deflections of cerebral potentials superimposed with conventional electroencephalography (EEG) waves and reflect alterations in the excitation level of the cerebral cortex and subcortical structures. Using galvanometers, such sustained displacement of the EEG baseline was recorded in the early days of EEG recordings. To stabilize the EEG baseline and eliminate artefacts, EEG was performed later by voltage amplifiers with high-pass filters that dismiss slow DC potentials. This left slow DC potential recordings as a neglected diagnostic source in the routine clinical setting over the last few decades. Brain DC waves may arise from physiological processes or pathological phenomena. Recordings of DC potentials are fundamental electro-clinical signatures of some neurological and psychological disorders and may serve as diagnostic, prognostic, and treatment monitoring tools. We here review the utility of both physiological and pathological brain DC potentials in different aspects of neurological and psychological disorders. This may enhance our understanding of the role of brain DC potentials and improve our fundamental clinical and research strategies for brain disorders.
Collapse
|
36
|
Ludwig PE, Thankam FG, Patil AA, Chamczuk AJ, Agrawal DK. Brain injury and neural stem cells. Neural Regen Res 2018; 13:7-18. [PMID: 29451199 PMCID: PMC5840995 DOI: 10.4103/1673-5374.224361] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2017] [Indexed: 12/26/2022] Open
Abstract
Many therapies with potential for treatment of brain injury have been investigated. Few types of cells have spurred as much interest and excitement as stem cells over the past few decades. The multipotentiality and self-renewing characteristics of stem cells confer upon them the capability to regenerate lost tissue in ischemic or degenerative conditions as well as trauma. While stem cells have not yet proven to be clinically effective in many such conditions as was once hoped, they have demonstrated some effects that could be manipulated for clinical benefit. The various types of stem cells have similar characteristics, and largely differ in terms of origin; those that have differentiated to some extent may exhibit limited capability in differentiation potential. Stem cells can aid in decreasing lesion size and improving function following brain injury.
Collapse
Affiliation(s)
- Parker E. Ludwig
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA
| | - Finosh G. Thankam
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA
| | - Arun A. Patil
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA
- Department of Neurosurgery, Creighton University School of Medicine, Omaha, NE, USA
| | - Andrea J. Chamczuk
- Department of Neurosurgery, Creighton University School of Medicine, Omaha, NE, USA
| | - Devendra K. Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA
| |
Collapse
|
37
|
The Role of Circular RNAs in Cerebral Ischemic Diseases: Ischemic Stroke and Cerebral Ischemia/Reperfusion Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1087:309-325. [PMID: 30259377 DOI: 10.1007/978-981-13-1426-1_25] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cerebral ischemic diseases including ischemic stroke and cerebral ischemia reperfusion injury can result in serious dysfunction of the brain, which leads to extremely high mortality and disability. There are no effective therapeutics for cerebral ischemic diseases to date. Circular RNAs are a kind of newly investigated noncoding RNAs. It is reported that circular RNAs are enriched in multiple organs, especially abundant in the brain, which indicates that circular RNAs may be involved in cerebral physiological and pathological processes. In this chapter, we will firstly review the pathophysiology, underlying mechanisms, and current treatments of cerebral ischemic diseases including ischemic stroke and cerebral ischemia/reperfusion injury. Secondly, the characteristics and function of circular RNAs will be outlined, and then we are going to introduce the roles circular RNAs play in human diseases. Finally, we will summarize the function of circular RNAs in cerebral ischemic diseases.
Collapse
|
38
|
Mayor D, Tymianski M. Neurotransmitters in the mediation of cerebral ischemic injury. Neuropharmacology 2017; 134:178-188. [PMID: 29203179 DOI: 10.1016/j.neuropharm.2017.11.050] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 01/09/2023]
Abstract
Under physiological conditions, neurotransmitters shape neuronal networks and control several cellular and synaptic functions. In the mammalian central nervous system (CNS), excitatory and inhibitory neurotransmission are mediated in large part by glutamate and gamma-aminobutyric acid (GABA), which are excitatory and inhibitory neurotransmitters, respectively. Glutamate and GABA also play crucial roles in neurological disorders such as cerebral ischemia. Glutamate in particular causes excitotoxicity, known as one of the hallmark mechanisms in the pathophysiology of cerebral ischemic injury for more than thirty years. Excitotoxicity occurs due to excessive glutamate release leading to overactivation of postsynaptic glutamate receptors, which evokes a downstream cascade that eventually leads to neuronal dysfunction and degeneration. Also, a reduction in GABA receptor response after ischemia impedes these inhibitory effectors from attenuating excitotoxicity and thereby further enabling the excitotoxic insult. This review focuses on the mechanisms by which glutamate and GABA mediate excitotoxicity and ischemic injury. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
Collapse
Affiliation(s)
- Diana Mayor
- Division of Fundamental Neurobiology, Krembil Institute, University Health Network, Toronto, Ontario, M5T 2S8, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Michael Tymianski
- Division of Fundamental Neurobiology, Krembil Institute, University Health Network, Toronto, Ontario, M5T 2S8, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada; Department of Neurosurgery, University of Toronto, Toronto, Ontario, M5G 1LG, Canada.
| |
Collapse
|
39
|
Kawauchi S, Nishidate I, Nawashiro H, Sato S. Near-infrared diffuse reflectance signals for monitoring spreading depolarizations and progression of the lesion in a male rat focal cerebral ischemia model. J Neurosci Res 2017; 96:875-888. [PMID: 29150867 DOI: 10.1002/jnr.24201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/24/2017] [Accepted: 11/08/2017] [Indexed: 11/07/2022]
Abstract
In ischemic stroke research, a better understanding of the pathophysiology and development of neuroprotection methods are crucial, for which in vivo imaging to monitor spreading depolarizations (SDs) and evolution of tissue damage is desired. Since these events are accompanied by cellular morphological changes, light-scattering signals, which are sensitive to cellular and subcellular morphology, can be used for monitoring them. In this study, we performed transcranial imaging of near-infrared (NIR) diffuse reflectance at ∼800 nm, which sensitively reflects light-scattering change, and examined how NIR reflectance is correlated with simultaneously measured cerebral blood flow (CBF) for a rat middle cerebral artery occlusion (MCAO) model. After MCAO, wavelike NIR reflectance changes indicating occurrence of SDs were generated and propagated around the ischemic core for ∼90 min, during which time NIR reflectance increased not only within the ischemic core but also in the peripheral region. The area with increased reflectance expanded with increase in the number of SD occurrences, the correlation coefficient being 0.7686 (n = 5). The area with increased reflectance had become infarcted at 24 hr after MCAO. The infarct region was found to be associated with hypoperfusion or no-flow response to SD, but hyperemia or hypoperfusion followed by hyperemia response to SD was also observed, and the regional heterogeneity seemed to be connected with the rat cerebrovasculature and hence existence/absence of collateral flow. The results suggest that NIR reflectance signals depicted early evolution of tissue damage, which was not seen by CBF changes, and enabled lesion progression monitoring in the present stroke model.
Collapse
Affiliation(s)
- Satoko Kawauchi
- Division of Bioinformation and Therapeutic Systems, National Defense Medical College Research Institute, Tokorozawa, Saitama, Japan
| | - Izumi Nishidate
- Graduate School of Bio-Applications & Systems Engineering, Tokyo University of Agriculture and Technology, Koganei, Tokyo, Japan
| | - Hiroshi Nawashiro
- Division of Neurosurgery, Tokorozawa Central Hospital, Tokorozawa, Saitama, Japan
| | - Shunichi Sato
- Division of Bioinformation and Therapeutic Systems, National Defense Medical College Research Institute, Tokorozawa, Saitama, Japan
| |
Collapse
|
40
|
Hobbs CN, Holzberg G, Min AS, Wightman RM. Comparison of Spreading Depolarizations in the Motor Cortex and Nucleus Accumbens: Similar Patterns of Oxygen Responses and the Role of Dopamine. ACS Chem Neurosci 2017; 8:2512-2521. [PMID: 28820571 PMCID: PMC5691918 DOI: 10.1021/acschemneuro.7b00266] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Spreading depolarizations (SD) are pathophysiological phenomena that spontaneously arise in traumatized neural tissue and can promote cellular death. Most investigations of SD are performed in the cortex, a brain region that is susceptible to these depolarizing waves and accessible via a variety of monitoring techniques. Here, we describe SD responses in the cortex and the deep brain region of the nucleus accumbens (NAc) of the anesthetized rat with a minimally invasive, implantable sensor. With high temporal resolution, we characterize the time course of oxygen responses to SD in relation to the electrophysiological depolarization signal. The predominant oxygen pattern consists of four phases: (1) a small initial decrease, (2) a large increase during the SD, (3) a delayed increase, and (4) a persistent decrease from baseline after the SD. Oxygen decreases during SD were also recorded. The latter response occurred more often in the NAc than the cortex (56% vs 20% of locations, respectively), which correlates to denser cortical vascularization. We also find that SDs travel more quickly in the cortex than NAc, likely affected by regional differences in cell type populations. Finally, we investigate the previously uncharacterized effects of dopamine release during SD in the NAc with dopamine receptor blockade. Our results support an inhibitory role of the D2 receptor on SD. As such, the data presented here expands the current understanding of within- and between-region variance in responses to SD.
Collapse
Affiliation(s)
- Caddy N. Hobbs
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Gordon Holzberg
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Akira S. Min
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - R. Mark Wightman
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
41
|
Tamaki R, Orie SI, Alessandri B, Kempski O, Heimann A. Spreading depression and focal venous cerebral ischemia enhance cortical neurogenesis. Neural Regen Res 2017; 12:1278-1286. [PMID: 28966642 PMCID: PMC5607822 DOI: 10.4103/1673-5374.213547] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2017] [Indexed: 11/29/2022] Open
Abstract
Endogenous neurogenesis can arise from a variety of physiological stimuli including exercise, learning, or "enriched environment" as well as pathological conditions such as ischemia, epilepsy or cortical spreading depression. Whether all these conditions use a common trigger to set off endogenous neurogenesis is yet unclear. We hypothesized that cortical spreading depression (CSD) induces neurogenesis in the cerebral cortex and dentate gyrus after cerebral venous ischemia. Forty-two Wistar rats alternatively underwent sham operation (Sham), induction of ten CSDs or venous ischemia provoked via occlusion of two adjacent superficial cortical vein followed by ten induced CSDs (CSD + 2-VO). As an additional control, 15 naïve rats received no intervention except 5-bromo-2'-deoxyuridine (BrdU) treatment for 7 days. Sagittal brain slices (40 μm thick) were co-stained for BrdU and doublecortin (DCX; new immature neuronal cells) on day 9 or NeuN (new mature neuronal cells) on day 28. On day 9 after sham operation, cell proliferation and neurogenesis occurred in the cortex in rats. The sole induction of CSD had no effect. But on days 9 and 28, more proliferating cells and newly formed neurons in the ipsilateral cortex were observed in rats subjected to CSD + 2VO than in rats subjected to sham operation. On days 9 and 28, cell proliferation and neurogenesis in the ipsilateral dentate gyrus was increased in sham-operated rats than in naïve rats. Our data supports the hypothesis that induced cortical neurogenesis after CSD + 2-VO is a direct effect of ischemia, rather than of CSD alone.
Collapse
Affiliation(s)
- Ryo Tamaki
- Department of Neurosurgery, Nara Medical University, Nara, Japan
| | - Samuel Ige Orie
- University Medical Center of the Johannes Gutenberg-University of Mainz, Institute for Neurosurgical Pathophysiology, Mainz, Germany
| | - Beat Alessandri
- University Medical Center of the Johannes Gutenberg-University of Mainz, Institute for Neurosurgical Pathophysiology, Mainz, Germany
| | - Oliver Kempski
- University Medical Center of the Johannes Gutenberg-University of Mainz, Institute for Neurosurgical Pathophysiology, Mainz, Germany
| | - Axel Heimann
- University Medical Center of the Johannes Gutenberg-University of Mainz, Institute for Neurosurgical Pathophysiology, Mainz, Germany
| |
Collapse
|
42
|
Pan HC, Liao LD, Lo YC, Chen JW, Wang HL, Yang L, Liang YW, Huang PY, Yang MH, Chen YY. Neurovascular function recovery after focal ischemic stroke by enhancing cerebral collateral circulation via peripheral stimulation-mediated interarterial anastomosis. NEUROPHOTONICS 2017; 4:035003. [PMID: 28983488 PMCID: PMC5621356 DOI: 10.1117/1.nph.4.3.035003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 09/12/2017] [Indexed: 05/22/2023]
Abstract
Current treatments for ischemic stroke have focused on the administration of a tissue plasminogen activator, although the associated side effects and subsequent reperfusion injury remain challenging. Peripheral electrical stimulation has shed light on therapeutic interventions for ischemia by increasing cerebral blood flow (CBF) to the target region through collateral circulation, although the mechanism remains elusive. Here, a focal photothrombotic ischemic (PTI) stroke was induced in the right hemispheric primary somatosensory forelimb cortex (S1FL) of rat brains, and the therapeutic effects of forelimb and hindlimb stimulation were characterized at the contralesional S1FL. We observed that PTI stroke rats that received forelimb stimulation exhibited significantly restored CBF of the ischemic penumbra ([Formula: see text] for the S1FL and [Formula: see text] for the primary somatosensory hindlimb cortex, respectively), electrocorticography (ECoG) delta band coherence of the intercortical S1FL ([Formula: see text]) at the 75th min poststroke and an ischemic infarct ([Formula: see text]) via collateral circulation recruitment. Importantly, anterior cerebral artery/middle cerebral artery (ACA-MCA) interarterial anastomotic regulation occurred upon forelimb stimulation and played roles in the recovery of neurovascular functions. These results indicated that receptive field-specific stimulation further restores CBF, neuronal activities, and tissue viability through the enhancement of ACA-MCA interarterial anastomosis-mediated collateral circulation and provides a feasible therapeutic intervention for stroke recovery.
Collapse
Affiliation(s)
- Han-Chi Pan
- National Health Research Institutes, Institute of Biomedical Engineering and Nanomedicine, Zhunan Town, Miaoli County, Taiwan
| | - Lun-De Liao
- National Health Research Institutes, Institute of Biomedical Engineering and Nanomedicine, Zhunan Town, Miaoli County, Taiwan
| | - Yu-Chun Lo
- Taipei Medical University, The PhD Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei, Taiwan
| | - Jia-Wei Chen
- National Yang Ming University, Department of Biomedical Engineering, Taipei, Taiwan
| | - Han-Lin Wang
- National Yang Ming University, Department of Biomedical Engineering, Taipei, Taiwan
| | - Li Yang
- National Yang Ming University, Department of Biomedical Engineering, Taipei, Taiwan
| | - Yao-Wen Liang
- National Yang Ming University, Department of Life Sciences and Institute of Genome Sciences, Taipei, Taiwan
| | - Po-Yu Huang
- National Yang Ming University, Department of Medicine, Taipei, Taiwan
| | - Ming-Hsun Yang
- Cheng Hsin General Hospital, Division of General Surgery, Department of Surgery, Taipei, Taiwan
- Address all correspondence to: Ming-Hsun Yang, E-mail: ; You-Yin Chen, E-mail:
| | - You-Yin Chen
- National Yang Ming University, Department of Biomedical Engineering, Taipei, Taiwan
- Address all correspondence to: Ming-Hsun Yang, E-mail: ; You-Yin Chen, E-mail:
| |
Collapse
|
43
|
Rakers C, Schmid M, Petzold GC. TRPV4 channels contribute to calcium transients in astrocytes and neurons during peri-infarct depolarizations in a stroke model. Glia 2017. [PMID: 28639721 DOI: 10.1002/glia.23183] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Stroke is one of the leading causes of death and long-term disability. In the penumbra, that is, the area surrounding the infarct core, peri-infarct depolarizations (PIDs) are accompanied by strong intracellular calcium elevations in astrocytes and neurons, thereby negatively affecting infarct size and clinical outcome. The dynamics of PIDs and the cellular pathways that are involved during PID formation and progression remain incompletely understood. We have previously shown that inositol triphosphate-gated calcium release from internal stores is a major component of PID-related astroglial calcium signals, but whether external calcium influx through membrane-localized channels also contributes to PIDs has remained unclear. In this study, we investigated the role of two astroglial membrane channels, transient receptor vanilloid 4 (TRPV4) channel and aquaporin-4 (AQP4). We combined in vivo multiphoton microscopy, electrophysiology as well as laser speckle contrast imaging with the middle cerebral artery occlusion stroke model. Using knockout mice and pharmacological inhibitors, we found that TRPV4 channels contribute to calcium influx into astrocytes and neurons and subsequent extracellular glutamate accumulation during PIDs. AQP4 neither influenced PID-related calcium signals nor PID-related edema of astrocyte somata. Both channels did not alter the dynamics, frequency and cerebrovascular response of PIDs in the penumbra. These data indicate that TRPV4 channels may represent a potential target to ameliorate the PID-induced calcium overload of astrocytes and neurons during acute stroke.
Collapse
Affiliation(s)
- Cordula Rakers
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Street 27, Bonn, 53127, Germany
| | - Matthias Schmid
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Street 27, Bonn, 53127, Germany.,Department of Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Sigmund-Freud-Street 25, Bonn, 53127, Germany
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Street 27, Bonn, 53127, Germany.,Department of Neurology, University Hospital Bonn, Sigmund-Freud-Street 25, Bonn, 53127, Germany
| |
Collapse
|
44
|
Pinczolits A, Zdunczyk A, Dengler NF, Hecht N, Kowoll CM, Dohmen C, Graf R, Winkler MK, Major S, Hartings JA, Dreier JP, Vajkoczy P, Woitzik J. Standard-sampling microdialysis and spreading depolarizations in patients with malignant hemispheric stroke. J Cereb Blood Flow Metab 2017; 37:1896-1905. [PMID: 28350195 PMCID: PMC5435299 DOI: 10.1177/0271678x17699629] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Spreading depolarizations (SD) occur in high frequency in patients with malignant hemispheric stroke (MHS). Experimentally, SDs cause marked increases in glutamate and lactate, whereas glucose decreases. Here, we studied extracellular brain glutamate, glucose, lactate, pyruvate and the lactate/pyruvate ratio in relationship to SDs after MHS. We inserted two microdialysis probes in peri-infarct tissue at 5 and 15 mm to the infarct in close proximity to a subdural electrode strip. During 2356.6 monitoring hours, electrocorticography (ECoG) revealed 697 SDs in 16 of 18 patients. Ninety-nine SDs in electrically active tissue (spreading depressions, SDd) were single (SDds) and 485 clustered (SDdc), whereas 10 SDs with at least one electrode in electrically inactive tissue (isoelectric SDs, SDi) were single (SDis) and 103 clustered (SDic). More SDs and a significant number of clustered SDs occurred during the first 36 h post-surgery when glutamate was significantly elevated (> 100 µM). In a grouped analysis, we observed minor glutamate elevations with more than two SDs per hour. Glucose slightly decreased during SDic at 5 mm from the infarct. Directions of SD-related metabolic changes correspond to the experimental setting but the long sampling time of standard microdialysis precludes a more adequate account of the dynamics revealed by ECoG.
Collapse
Affiliation(s)
- Alexandra Pinczolits
- 1 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Zdunczyk
- 1 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nora F Dengler
- 1 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nils Hecht
- 1 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christina M Kowoll
- 3 Department of Neurology, University of Cologne, Cologne, Germany.,4 Max Planck Institute for Neurological Research, Cologne, Germany
| | - Christian Dohmen
- 3 Department of Neurology, University of Cologne, Cologne, Germany.,4 Max Planck Institute for Neurological Research, Cologne, Germany
| | - Rudolf Graf
- 4 Max Planck Institute for Neurological Research, Cologne, Germany
| | - Maren Kl Winkler
- 2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,5 Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Major
- 2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,5 Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jed A Hartings
- 6 Department of Neurosurgery, University of Cincinnati College of Medicine, Mayfield Clinic, Cincinnati, OH, USA
| | - Jens P Dreier
- 2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,5 Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Vajkoczy
- 1 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Woitzik
- 1 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,2 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
45
|
Dreier JP, Fabricius M, Ayata C, Sakowitz OW, William Shuttleworth C, Dohmen C, Graf R, Vajkoczy P, Helbok R, Suzuki M, Schiefecker AJ, Major S, Winkler MKL, Kang EJ, Milakara D, Oliveira-Ferreira AI, Reiffurth C, Revankar GS, Sugimoto K, Dengler NF, Hecht N, Foreman B, Feyen B, Kondziella D, Friberg CK, Piilgaard H, Rosenthal ES, Westover MB, Maslarova A, Santos E, Hertle D, Sánchez-Porras R, Jewell SL, Balança B, Platz J, Hinzman JM, Lückl J, Schoknecht K, Schöll M, Drenckhahn C, Feuerstein D, Eriksen N, Horst V, Bretz JS, Jahnke P, Scheel M, Bohner G, Rostrup E, Pakkenberg B, Heinemann U, Claassen J, Carlson AP, Kowoll CM, Lublinsky S, Chassidim Y, Shelef I, Friedman A, Brinker G, Reiner M, Kirov SA, Andrew RD, Farkas E, Güresir E, Vatter H, Chung LS, Brennan KC, Lieutaud T, Marinesco S, Maas AIR, Sahuquillo J, Dahlem MA, Richter F, Herreras O, Boutelle MG, Okonkwo DO, Bullock MR, Witte OW, Martus P, van den Maagdenberg AMJM, Ferrari MD, Dijkhuizen RM, Shutter LA, Andaluz N, Schulte AP, MacVicar B, Watanabe T, Woitzik J, Lauritzen M, Strong AJ, Hartings JA. Recording, analysis, and interpretation of spreading depolarizations in neurointensive care: Review and recommendations of the COSBID research group. J Cereb Blood Flow Metab 2017; 37:1595-1625. [PMID: 27317657 PMCID: PMC5435289 DOI: 10.1177/0271678x16654496] [Citation(s) in RCA: 255] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 05/04/2016] [Accepted: 05/06/2016] [Indexed: 01/18/2023]
Abstract
Spreading depolarizations (SD) are waves of abrupt, near-complete breakdown of neuronal transmembrane ion gradients, are the largest possible pathophysiologic disruption of viable cerebral gray matter, and are a crucial mechanism of lesion development. Spreading depolarizations are increasingly recorded during multimodal neuromonitoring in neurocritical care as a causal biomarker providing a diagnostic summary measure of metabolic failure and excitotoxic injury. Focal ischemia causes spreading depolarization within minutes. Further spreading depolarizations arise for hours to days due to energy supply-demand mismatch in viable tissue. Spreading depolarizations exacerbate neuronal injury through prolonged ionic breakdown and spreading depolarization-related hypoperfusion (spreading ischemia). Local duration of the depolarization indicates local tissue energy status and risk of injury. Regional electrocorticographic monitoring affords even remote detection of injury because spreading depolarizations propagate widely from ischemic or metabolically stressed zones; characteristic patterns, including temporal clusters of spreading depolarizations and persistent depression of spontaneous cortical activity, can be recognized and quantified. Here, we describe the experimental basis for interpreting these patterns and illustrate their translation to human disease. We further provide consensus recommendations for electrocorticographic methods to record, classify, and score spreading depolarizations and associated spreading depressions. These methods offer distinct advantages over other neuromonitoring modalities and allow for future refinement through less invasive and more automated approaches.
Collapse
Affiliation(s)
- Jens P Dreier
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
- Department of Neurology, Charité University Medicine Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Martin Fabricius
- Department of Clinical Neurophysiology, Rigshospitalet, Copenhagen, Denmark
| | - Cenk Ayata
- Neurovascular Research Laboratory, Department of Radiology, and Stroke Service and Neuroscience Intensive Care Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Oliver W Sakowitz
- Department of Neurosurgery, Klinikum Ludwigsburg, Ludwigsburg, Germany
- Department of Neurosurgery, University Hospital, Heidelberg, Germany
| | - C William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Christian Dohmen
- Department of Neurology, University of Cologne, Cologne, Germany
- Multimodal Imaging of Brain Metabolism, Max-Planck-Institute for Metabolism Research, Cologne, Germany
| | - Rudolf Graf
- Multimodal Imaging of Brain Metabolism, Max-Planck-Institute for Metabolism Research, Cologne, Germany
| | - Peter Vajkoczy
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
- Department of Neurosurgery, Charité University Medicine Berlin, Berlin, Germany
| | - Raimund Helbok
- Department of Neurology, Neurocritical Care Unit, Medical University Innsbruck, Innsbruck, Austria
| | - Michiyasu Suzuki
- Department of Neurosurgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Alois J Schiefecker
- Department of Neurology, Neurocritical Care Unit, Medical University Innsbruck, Innsbruck, Austria
| | - Sebastian Major
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
- Department of Neurology, Charité University Medicine Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Maren KL Winkler
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
| | - Eun-Jeung Kang
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Denny Milakara
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
| | - Ana I Oliveira-Ferreira
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Clemens Reiffurth
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Gajanan S Revankar
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
| | - Kazutaka Sugimoto
- Department of Neurosurgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Nora F Dengler
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
- Department of Neurosurgery, Charité University Medicine Berlin, Berlin, Germany
| | - Nils Hecht
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
- Department of Neurosurgery, Charité University Medicine Berlin, Berlin, Germany
| | - Brandon Foreman
- Department of Neurology and Rehabilitation Medicine, Neurocritical Care Division, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Bart Feyen
- Department of Neurosurgery, Antwerp University Hospital and University of Antwerp, Edegem, Belgium
| | | | | | - Henning Piilgaard
- Department of Clinical Neurophysiology, Rigshospitalet, Copenhagen, Denmark
| | - Eric S Rosenthal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - M Brandon Westover
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anna Maslarova
- Department of Neurosurgery, University Hospital and University of Bonn, Bonn, Germany
| | - Edgar Santos
- Department of Neurosurgery, University Hospital, Heidelberg, Germany
| | - Daniel Hertle
- Department of Neurosurgery, University Hospital, Heidelberg, Germany
| | | | - Sharon L Jewell
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Baptiste Balança
- Inserm U10128, CNRS UMR5292, Lyon Neuroscience Research Center, Team TIGER, Lyon, France
- Université Claude Bernard, Lyon, France
| | - Johannes Platz
- Department of Neurosurgery, Goethe-University, Frankfurt, Germany
| | - Jason M Hinzman
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Janos Lückl
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
| | - Karl Schoknecht
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité University Medicine Berlin, Berlin, Germany
- Neuroscience Research Center, Charité University Medicine Berlin, Berlin, Germany
| | - Michael Schöll
- Department of Neurosurgery, University Hospital, Heidelberg, Germany
- Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Christoph Drenckhahn
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
- Neurological Center, Segeberger Kliniken, Bad Segeberg, Germany
| | - Delphine Feuerstein
- Multimodal Imaging of Brain Metabolism, Max-Planck-Institute for Metabolism Research, Cologne, Germany
| | - Nina Eriksen
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen, Denmark
- Research Laboratory for Stereology and Neuroscience, Bispebjerg-Frederiksberg Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Viktor Horst
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
- Department of Neuroradiology, Charité University Medicine Berlin, Berlin, Germany
| | - Julia S Bretz
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
- Department of Neuroradiology, Charité University Medicine Berlin, Berlin, Germany
| | - Paul Jahnke
- Department of Neuroradiology, Charité University Medicine Berlin, Berlin, Germany
| | - Michael Scheel
- Department of Neuroradiology, Charité University Medicine Berlin, Berlin, Germany
| | - Georg Bohner
- Department of Neuroradiology, Charité University Medicine Berlin, Berlin, Germany
| | - Egill Rostrup
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen, Denmark
| | - Bente Pakkenberg
- Research Laboratory for Stereology and Neuroscience, Bispebjerg-Frederiksberg Hospital, Rigshospitalet, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Uwe Heinemann
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
- Neuroscience Research Center, Charité University Medicine Berlin, Berlin, Germany
| | - Jan Claassen
- Neurocritical Care, Columbia University College of Physicians & Surgeons, New York, NY, USA
| | - Andrew P Carlson
- Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Christina M Kowoll
- Department of Neurology, University of Cologne, Cologne, Germany
- Multimodal Imaging of Brain Metabolism, Max-Planck-Institute for Metabolism Research, Cologne, Germany
| | - Svetlana Lublinsky
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Beer-Sheva, Israel
- Department of Neuroradiology, Soroka University Medical Center and Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yoash Chassidim
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Beer-Sheva, Israel
- Department of Neuroradiology, Soroka University Medical Center and Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ilan Shelef
- Department of Neuroradiology, Soroka University Medical Center and Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alon Friedman
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Beer-Sheva, Israel
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, Canada
| | - Gerrit Brinker
- Department of Neurosurgery, University of Cologne, Cologne, Germany
| | - Michael Reiner
- Department of Neurosurgery, University of Cologne, Cologne, Germany
| | - Sergei A Kirov
- Department of Neurosurgery and Brain and Behavior Discovery Institute, Medical College of Georgia, Augusta, GA, USA
| | - R David Andrew
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, Canada
| | - Eszter Farkas
- Department of Medical Physics and Informatics, Faculty of Medicine, and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Erdem Güresir
- Department of Neurosurgery, University Hospital and University of Bonn, Bonn, Germany
| | - Hartmut Vatter
- Department of Neurosurgery, University Hospital and University of Bonn, Bonn, Germany
| | - Lee S Chung
- Department of Neurology, University of Utah, Salt Lake City, UT, USA
| | - KC Brennan
- Department of Neurology, University of Utah, Salt Lake City, UT, USA
| | - Thomas Lieutaud
- Inserm U10128, CNRS UMR5292, Lyon Neuroscience Research Center, Team TIGER, Lyon, France
- Université Claude Bernard, Lyon, France
| | - Stephane Marinesco
- Inserm U10128, CNRS UMR5292, Lyon Neuroscience Research Center, Team TIGER, Lyon, France
- AniRA-Neurochem Technological Platform, Lyon, France
| | - Andrew IR Maas
- Department of Neurosurgery, Antwerp University Hospital and University of Antwerp, Edegem, Belgium
| | - Juan Sahuquillo
- Department of Neurosurgery, Neurotraumatology and Neurosurgery Research Unit (UNINN), Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Frank Richter
- Institute of Physiology I/Neurophysiology, Friedrich Schiller University Jena, Jena, Germany
| | - Oscar Herreras
- Department of Systems Neuroscience, Cajal Institute-CSIC, Madrid, Spain
| | | | - David O Okonkwo
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - M Ross Bullock
- Department of Neurological Surgery, University of Miami, Miami, FL, USA
| | - Otto W Witte
- Hans Berger Department of Neurology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Peter Martus
- Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, Tübingen, Germany
| | - Arn MJM van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Michel D Ferrari
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Rick M Dijkhuizen
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Lori A Shutter
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Department of Critical Care Medicine and Neurology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Norberto Andaluz
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Mayfield Clinic, Cincinnati, OH, USA
| | - André P Schulte
- Department of Spinal Surgery, St. Franziskus Hospital Cologne, Cologne, Germany
| | - Brian MacVicar
- Department of Psychiatry, University of British Columbia, Vancouver, Canada
| | | | - Johannes Woitzik
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin, Germany
- Department of Neurosurgery, Charité University Medicine Berlin, Berlin, Germany
| | - Martin Lauritzen
- Department of Clinical Neurophysiology, Rigshospitalet, Copenhagen, Denmark
- Department of Neuroscience and Pharmacology, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Anthony J Strong
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Jed A Hartings
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Mayfield Clinic, Cincinnati, OH, USA
| |
Collapse
|
46
|
Lindquist BE, Shuttleworth CW. Evidence that adenosine contributes to Leao's spreading depression in vivo. J Cereb Blood Flow Metab 2017; 37:1656-1669. [PMID: 27217381 PMCID: PMC5435284 DOI: 10.1177/0271678x16650696] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Leao's spreading depression of cortical activity is a propagating silencing of neuronal activity resulting from spreading depolarization (SD). We evaluated the contributions of action potential (AP) failure and adenosine A1 receptor (A1R) activation to the depression of evoked and spontaneous electrocorticographic (ECoG) activity after SD in vivo, in anesthetized mice. We compared depression with SD-induced effects on AP-dependent transmission, and synaptic potentials in the transcallosal and thalamocortical pathways. After SD, APs recovered rapidly, within 1-2 min, as demonstrated by evoked activity in distant projection targets. Evoked corticocortical postsynaptic potentials recovered next, within ∼5 min. Spontaneous ECoG and evoked thalamocortical postsynaptic potentials recovered together, after ∼10-15 min. The duration of ECoG depression was shortened 20% by systemic (10 mg/kg) or focal (30 µM) administration of A1R competitive antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX). ECoG depression was also shortened by focal application of exogenous adenosine deaminase (ADA; 100 U/mL), and conversely, was prolonged 50% by the non-competitive ADA inhibitor deoxycoformycin (DCF; 100 µM). We concluded that while initial depolarization block is brief, adenosine A1R activation, in part, contributes to the persistent secondary phase of Leao's cortical spreading depression.
Collapse
Affiliation(s)
- Britta E Lindquist
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - C William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
47
|
Hartings JA, Shuttleworth CW, Kirov SA, Ayata C, Hinzman JM, Foreman B, Andrew RD, Boutelle MG, Brennan KC, Carlson AP, Dahlem MA, Drenckhahn C, Dohmen C, Fabricius M, Farkas E, Feuerstein D, Graf R, Helbok R, Lauritzen M, Major S, Oliveira-Ferreira AI, Richter F, Rosenthal ES, Sakowitz OW, Sánchez-Porras R, Santos E, Schöll M, Strong AJ, Urbach A, Westover MB, Winkler MK, Witte OW, Woitzik J, Dreier JP. The continuum of spreading depolarizations in acute cortical lesion development: Examining Leão's legacy. J Cereb Blood Flow Metab 2017; 37:1571-1594. [PMID: 27328690 PMCID: PMC5435288 DOI: 10.1177/0271678x16654495] [Citation(s) in RCA: 291] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
A modern understanding of how cerebral cortical lesions develop after acute brain injury is based on Aristides Leão's historic discoveries of spreading depression and asphyxial/anoxic depolarization. Treated as separate entities for decades, we now appreciate that these events define a continuum of spreading mass depolarizations, a concept that is central to understanding their pathologic effects. Within minutes of acute severe ischemia, the onset of persistent depolarization triggers the breakdown of ion homeostasis and development of cytotoxic edema. These persistent changes are diagnosed as diffusion restriction in magnetic resonance imaging and define the ischemic core. In delayed lesion growth, transient spreading depolarizations arise spontaneously in the ischemic penumbra and induce further persistent depolarization and excitotoxic damage, progressively expanding the ischemic core. The causal role of these waves in lesion development has been proven by real-time monitoring of electrophysiology, blood flow, and cytotoxic edema. The spreading depolarization continuum further applies to other models of acute cortical lesions, suggesting that it is a universal principle of cortical lesion development. These pathophysiologic concepts establish a working hypothesis for translation to human disease, where complex patterns of depolarizations are observed in acute brain injury and appear to mediate and signal ongoing secondary damage.
Collapse
Affiliation(s)
- Jed A Hartings
- 1 Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,2 Mayfield Clinic, Cincinnati, OH, USA
| | - C William Shuttleworth
- 3 Department of Neuroscience, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Sergei A Kirov
- 4 Department of Neurosurgery and Brain and Behavior Discovery Institute, Medical College of Georgia, Augusta, GA, USA
| | - Cenk Ayata
- 5 Neurovascular Research Unit, Department of Radiology, and Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jason M Hinzman
- 1 Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Brandon Foreman
- 6 Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - R David Andrew
- 7 Department of Biomedical & Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Martyn G Boutelle
- 8 Department of Bioengineering, Imperial College London, London, United Kingdom
| | - K C Brennan
- 9 Department of Neurology, University of Utah, Salt Lake City, UT, USA.,10 Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT, USA
| | - Andrew P Carlson
- 11 Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Markus A Dahlem
- 12 Department of Physics, Humboldt University of Berlin, Berlin, Germany
| | | | - Christian Dohmen
- 14 Department of Neurology, University of Cologne, Cologne, Germany
| | - Martin Fabricius
- 15 Department of Clinical Neurophysiology, Rigshospitalet, Glostrup, Denmark
| | - Eszter Farkas
- 16 Department of Medical Physics and Informatics, Faculty of Medicine, and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Delphine Feuerstein
- 17 Multimodal Imaging of Brain Metabolism, Max-Planck-Institute for Metabolism Research, Cologne, Germany
| | - Rudolf Graf
- 17 Multimodal Imaging of Brain Metabolism, Max-Planck-Institute for Metabolism Research, Cologne, Germany
| | - Raimund Helbok
- 18 Medical University of Innsbruck, Department of Neurology, Neurocritical Care Unit, Innsbruck, Austria
| | - Martin Lauritzen
- 15 Department of Clinical Neurophysiology, Rigshospitalet, Glostrup, Denmark.,19 Department of Neuroscience and Pharmacology and Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Sebastian Major
- 13 Department of Neurology, Charité University Medicine, Berlin, Germany.,20 Center for Stroke Research Berlin, Charité University Medicine, Berlin, Germany.,21 Department of Experimental Neurology, Charité University Medicine, Berlin, Germany
| | - Ana I Oliveira-Ferreira
- 20 Center for Stroke Research Berlin, Charité University Medicine, Berlin, Germany.,21 Department of Experimental Neurology, Charité University Medicine, Berlin, Germany
| | - Frank Richter
- 22 Institute of Physiology/Neurophysiology, Jena University Hospital, Jena, Germany
| | - Eric S Rosenthal
- 5 Neurovascular Research Unit, Department of Radiology, and Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Oliver W Sakowitz
- 23 Department of Neurosurgery, Klinikum Ludwigsburg, Ludwigsburg, Germany.,24 Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Renán Sánchez-Porras
- 24 Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Edgar Santos
- 24 Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Schöll
- 24 Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Anthony J Strong
- 25 Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London
| | - Anja Urbach
- 26 Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - M Brandon Westover
- 5 Neurovascular Research Unit, Department of Radiology, and Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maren Kl Winkler
- 20 Center for Stroke Research Berlin, Charité University Medicine, Berlin, Germany
| | - Otto W Witte
- 26 Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany.,27 Brain Imaging Center, Jena University Hospital, Jena, Germany
| | - Johannes Woitzik
- 20 Center for Stroke Research Berlin, Charité University Medicine, Berlin, Germany.,28 Department of Neurosurgery, Charité University Medicine, Berlin, Germany
| | - Jens P Dreier
- 13 Department of Neurology, Charité University Medicine, Berlin, Germany.,20 Center for Stroke Research Berlin, Charité University Medicine, Berlin, Germany.,21 Department of Experimental Neurology, Charité University Medicine, Berlin, Germany
| |
Collapse
|
48
|
Shimizu T, Hishikawa T, Nishihiro S, Shinji Y, Takasugi Y, Haruma J, Hiramatsu M, Kawase H, Sato S, Mizoue R, Takeda Y, Sugiu K, Morimatsu H, Date I. NADH fluorescence imaging and the histological impact of cortical spreading depolarization during the acute phase of subarachnoid hemorrhage in rats. J Neurosurg 2017; 128:137-143. [PMID: 28128691 DOI: 10.3171/2016.9.jns161385] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Although cortical spreading depolarization (CSD) has been observed during the early phase of subarachnoid hemorrhage (SAH) in clinical settings, the pathogenicity of CSD is unclear. The aim of this study is to elucidate the effects of loss of membrane potential on neuronal damage during the acute phase of SAH. METHODS Twenty-four rats were subjected to SAH by the perforation method. The propagation of depolarization in the brain cortex was examined by using electrodes to monitor 2 direct-current (DC) potentials and obtaining NADH (reduced nicotinamide adenine dinucleotide) fluorescence images while exposing the parietal-temporal cortex to ultraviolet light. Cerebral blood flow (CBF) was monitored in the vicinity of the lateral electrode. Twenty-four hours after onset of SAH, histological damage was evaluated at the DC potential recording sites. RESULTS Changes in DC potentials (n = 48 in total) were sorted into 3 types according to the appearance of ischemic depolarization in the entire hemisphere following induction of SAH. In Type 1 changes (n = 21), ischemic depolarization was not observed during a 1-hour observation period. In Type 2 changes (n = 13), the DC potential demonstrated ischemic depolarization on initiation of SAH and recovered 80% from the maximal DC deflection during a 1-hour observation period (33.3 ± 15.8 minutes). In Type 3 changes (n = 14), the DC potential displayed ischemic depolarization and did not recover during a 1-hour observation period. Histological evaluations at DC potential recording sites showed intact tissue at all sites in the Type 1 group, whereas in the Type 2 and Type 3 groups neuronal damage of varying severity was observed depending on the duration of ischemic depolarization. The duration of depolarization that causes injury to 50% of neurons (P50) was estimated to be 22.4 minutes (95% confidence intervals 17.0-30.3 minutes). CSD was observed in 3 rats at 6 sites in the Type 1 group 5.1 ± 2.2 minutes after initiation of SAH. On NADH fluorescence images CSD was initially observed in the anterior cortex; it propagated through the entire hemisphere in the direction of the occipital cortex at a rate of 3 mm/minute, with repolarization in 2.3 ± 1.2 minutes. DC potential recording sites that had undergone CSD were found to have intact tissue 24 hours later. Compared with depolarization that caused 50% neuronal damage, the duration of CSD was too short to cause histological damage. CONCLUSIONS CSD was successfully visualized using NADH fluorescence. It propagated from the anterior to the posterior cortex along with an increase in CBF. The duration of depolarization in CSD (2.3 ± 1.2 minutes) was far shorter than that causing 50% neuronal damage (22.4 minutes) and was not associated with histological damage in the current experimental setting.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hirokazu Kawase
- 2Department of Anesthesiology and Resuscitology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Sachiko Sato
- 2Department of Anesthesiology and Resuscitology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ryoichi Mizoue
- 2Department of Anesthesiology and Resuscitology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshimasa Takeda
- 2Department of Anesthesiology and Resuscitology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | - Hiroshi Morimatsu
- 2Department of Anesthesiology and Resuscitology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Isao Date
- 1Department of Neurological Surgery and
| |
Collapse
|
49
|
Kramer DR, Fujii T, Ohiorhenuan I, Liu CY. Interplay between Cortical Spreading Depolarization and Seizures. Stereotact Funct Neurosurg 2017; 95:1-5. [PMID: 28088802 DOI: 10.1159/000452841] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 10/18/2016] [Indexed: 11/19/2022]
Abstract
Cortical spreading depolarization (CSD) is an electrophysiologic phenomenon found mostly in the setting of neurologic injury resulting in the disturbance of ion homeostasis and leading to changes in the local vascular response. The bioelectric etiology of CSD shares similarities to those in epileptic disorders, yet the relationship between seizures and CSD is unclear, with several studies observing cortical depression before, during, and after seizure activity, thus obscuring our understanding of whether CSD activity potentiates or limits seizures and vice versa. Cortical sampling has exhibited how the redistribution of ion concentrations in the intra- and extracellular environments interplay between the excitation of seizures and the electrical depression of CSD. Modeling of both environments has suggested that CSD synchronizes the affected tissue, creating a favorable environment for seizure activity; however, other studies have demonstrated the opposite: epileptiform activity initiating waves of CSD. Further studies have underscored the role of the vascular response and subsequent ischemia in CSD that contributes to epileptogenesis. Investigations in migraine, traumatic brain injury, and other neurologic injuries suggest that several drugs may target CSD. Manipulations in the occurrence and nature of CSD can potentially alter the threshold for seizure activity, and perhaps minimize immediate and long-term sequelae associated with epilepsy.
Collapse
Affiliation(s)
- Daniel R Kramer
- Department of Neurosurgery, University of Southern California, Los Angeles, CA, USA
| | | | | | | |
Collapse
|
50
|
Muir KW, Macrae IM. Neuroimaging as a Selection Tool and Endpoint in Clinical and Pre-clinical Trials. Transl Stroke Res 2016; 7:368-77. [PMID: 27543177 PMCID: PMC5014902 DOI: 10.1007/s12975-016-0487-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/29/2016] [Accepted: 07/19/2016] [Indexed: 12/03/2022]
Abstract
Standard imaging in acute stroke enables the exclusion of non-stroke structural CNS lesions and cerebral haemorrhage from clinical and pre-clinical ischaemic stroke trials. In this review, the potential benefit of imaging (e.g., angiography and penumbral imaging) as a translational tool for trial recruitment and the use of imaging endpoints are discussed for both clinical and pre-clinical stroke research. The addition of advanced imaging to identify a “responder” population leads to reduced sample size for any given effect size in phase 2 trials and is a potentially cost-efficient means of testing interventions. In pre-clinical studies, technical failures (failed or incomplete vessel occlusion, cerebral haemorrhage) can be excluded early and continuous multimodal imaging of the animal from stroke onset is feasible. Pre- and post-intervention repeat scans provide real time assessment of the intervention over the first 4–6 h. Negative aspects of advanced imaging in animal studies include increased time under general anaesthesia, and, as in clinical studies, a delay in starting the intervention. In clinical phase 3 trial designs, the negative aspects of advanced imaging in patient selection include higher exclusion rates, slower recruitment, overestimated effect size and longer acquisition times. Imaging may identify biological effects with smaller sample size and at earlier time points, compared to standard clinical assessments, and can be adjusted for baseline parameters. Mechanistic insights can be obtained. Pre-clinically, multimodal imaging can non-invasively generate data on a range of parameters, allowing the animal to be recovered for subsequent behavioural testing and/or the brain taken for further molecular or histological analysis.
Collapse
Affiliation(s)
- Keith W Muir
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - I Mhairi Macrae
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK.
| |
Collapse
|