1
|
Dreier JP, Joerk A, Uchikawa H, Horst V, Lemale CL, Radbruch H, McBride DW, Vajkoczy P, Schneider UC, Xu R. All Three Supersystems-Nervous, Vascular, and Immune-Contribute to the Cortical Infarcts After Subarachnoid Hemorrhage. Transl Stroke Res 2024:10.1007/s12975-024-01242-z. [PMID: 38689162 DOI: 10.1007/s12975-024-01242-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 05/02/2024]
Abstract
The recently published DISCHARGE-1 trial supports the observations of earlier autopsy and neuroimaging studies that almost 70% of all focal brain damage after aneurysmal subarachnoid hemorrhage are anemic infarcts of the cortex, often also affecting the white matter immediately below. The infarcts are not limited by the usual vascular territories. About two-fifths of the ischemic damage occurs within ~ 48 h; the remaining three-fifths are delayed (within ~ 3 weeks). Using neuromonitoring technology in combination with longitudinal neuroimaging, the entire sequence of both early and delayed cortical infarct development after subarachnoid hemorrhage has recently been recorded in patients. Characteristically, cortical infarcts are caused by acute severe vasospastic events, so-called spreading ischemia, triggered by spontaneously occurring spreading depolarization. In locations where a spreading depolarization passes through, cerebral blood flow can drastically drop within a few seconds and remain suppressed for minutes or even hours, often followed by high-amplitude, sustained hyperemia. In spreading depolarization, neurons lead the event, and the other cells of the neurovascular unit (endothelium, vascular smooth muscle, pericytes, astrocytes, microglia, oligodendrocytes) follow. However, dysregulation in cells of all three supersystems-nervous, vascular, and immune-is very likely involved in the dysfunction of the neurovascular unit underlying spreading ischemia. It is assumed that subarachnoid blood, which lies directly on the cortex and enters the parenchyma via glymphatic channels, triggers these dysregulations. This review discusses the neuroglial, neurovascular, and neuroimmunological dysregulations in the context of spreading depolarization and spreading ischemia as critical elements in the pathogenesis of cortical infarcts after subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Jens P Dreier
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- Department of Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany.
- Einstein Center for Neurosciences Berlin, Berlin, Germany.
| | - Alexander Joerk
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Hiroki Uchikawa
- Barrow Aneurysm & AVM Research Center, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Viktor Horst
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Coline L Lemale
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Helena Radbruch
- Institute of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Devin W McBride
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ulf C Schneider
- Department of Neurosurgery, Cantonal Hospital of Lucerne and University of Lucerne, Lucerne, Switzerland
| | - Ran Xu
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Berlin, Germany
| |
Collapse
|
2
|
Jussen D, Saeed S, Jablonski T, Krenzlin H, Lucia K, Kraemer T, Kempski O, Czabanka M, Ringel F, Alessandri B. Influence of Blood Components on Neuroinflammation, Blood-Brain Barrier Breakdown, and Functional Damage After Acute Subdural Hematoma in Rats. Neurotrauma Rep 2024; 5:215-225. [PMID: 38463418 PMCID: PMC10924060 DOI: 10.1089/neur.2023.0098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024] Open
Abstract
A central component of injury development after acute subdural hematoma (ASDH) is the increased intracranial pressure and consecutive mechanical reduction of cerebral blood flow (CBF). However, the role of different blood constituents in ASDH as additional lesioning factors remains unclear. This study examines the influence of blood components on neuroinflammation, blood-brain barrier (BBB) breakdown, and functional deficits in a rat model of ASDH. We infused corpuscular (whole blood, whole blood lysate, and red cell blood) and plasmatic (blood plasma, anticoagulated blood plasma, and aqueous isotonic solution) blood components into the subdural space while CBF was monitored. Rats then underwent behavioral testing. Lesion analysis and immunohistochemistry were performed 2 days after ASDH. Inflammatory reaction was assessed using staining for ionized calcium-binding adaptor molecule 1 and glial fibrillary acidic protein, interleukin-1ß, tumor necrosis factor-alpha, and membrane attack complex. Integrity of the BBB was evaluated with albumin and matrix metalloproteinase 9 (MMP9) staining. We observed a significant drop in CBF in the corpuscular group (75% ± 7.5% of baseline) with distinct post-operative deficits and larger lesion volume compared to the plasmatic group (13.6 ± 5.4 vs. 1.3 ± 0.4 mm3). Further, inflammation was significantly increased in the corpuscular group with stronger immunoreaction. After whole blood infusion, albumin and MMP9 immunoreaction were significantly increased, pointing toward a disrupted BBB. The interaction between corpuscular and plasmatic blood components seems to be a key factor in the detrimental impact of ASDH. This interaction results in neuroinflammation and BBB leakage. These findings underscore the importance of performing surgery as early as possible and also provide indications for potential pharmacological targets.
Collapse
Affiliation(s)
- Daniel Jussen
- Department of Neurosurgery, Goethe University, Frankfurt am Main, Germany
- Johannes Gutenberg University, Institute for Neurosurgical Pathophysiology, Mainz, Germany
| | - Syamend Saeed
- Johannes Gutenberg University, Institute for Neurosurgical Pathophysiology, Mainz, Germany
| | - Tatjana Jablonski
- Johannes Gutenberg University, Institute for Neurosurgical Pathophysiology, Mainz, Germany
| | - Harald Krenzlin
- Johannes Gutenberg University, Institute for Neurosurgical Pathophysiology, Mainz, Germany
- Department of Neurosurgery, University Medical Center Mainz, Mainz, Germany
| | - Kristin Lucia
- Department of Neurosurgery, Goethe University, Frankfurt am Main, Germany
| | - Tobias Kraemer
- Johannes Gutenberg University, Institute for Neurosurgical Pathophysiology, Mainz, Germany
| | - Oliver Kempski
- Johannes Gutenberg University, Institute for Neurosurgical Pathophysiology, Mainz, Germany
| | - Marcus Czabanka
- Department of Neurosurgery, Goethe University, Frankfurt am Main, Germany
| | - Florian Ringel
- Department of Neurosurgery, University Medical Center Mainz, Mainz, Germany
| | - Beat Alessandri
- Johannes Gutenberg University, Institute for Neurosurgical Pathophysiology, Mainz, Germany
- Department of Neurosurgery, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
3
|
Dreier JP, Lemale CL, Horst V, Major S, Kola V, Schoknecht K, Scheel M, Hartings JA, Vajkoczy P, Wolf S, Woitzik J, Hecht N. Similarities in the Electrographic Patterns of Delayed Cerebral Infarction and Brain Death After Aneurysmal and Traumatic Subarachnoid Hemorrhage. Transl Stroke Res 2024:10.1007/s12975-024-01237-w. [PMID: 38396252 DOI: 10.1007/s12975-024-01237-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024]
Abstract
While subarachnoid hemorrhage is the second most common hemorrhagic stroke in epidemiologic studies, the recent DISCHARGE-1 trial has shown that in reality, three-quarters of focal brain damage after subarachnoid hemorrhage is ischemic. Two-fifths of these ischemic infarctions occur early and three-fifths are delayed. The vast majority are cortical infarcts whose pathomorphology corresponds to anemic infarcts. Therefore, we propose in this review that subarachnoid hemorrhage as an ischemic-hemorrhagic stroke is rather a third, separate entity in addition to purely ischemic or hemorrhagic strokes. Cumulative focal brain damage, determined by neuroimaging after the first 2 weeks, is the strongest known predictor of patient outcome half a year after the initial hemorrhage. Because of the unique ability to implant neuromonitoring probes at the brain surface before stroke onset and to perform longitudinal MRI scans before and after stroke, delayed cerebral ischemia is currently the stroke variant in humans whose pathophysiological details are by far the best characterized. Optoelectrodes located directly over newly developing delayed infarcts have shown that, as mechanistic correlates of infarct development, spreading depolarizations trigger (1) spreading ischemia, (2) severe hypoxia, (3) persistent activity depression, and (4) transition from clustered spreading depolarizations to a negative ultraslow potential. Furthermore, traumatic brain injury and subarachnoid hemorrhage are the second and third most common etiologies of brain death during continued systemic circulation. Here, we use examples to illustrate that although the pathophysiological cascades associated with brain death are global, they closely resemble the local cascades associated with the development of delayed cerebral infarcts.
Collapse
Affiliation(s)
- Jens P Dreier
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany.
- Einstein Center for Neurosciences Berlin, Berlin, Germany.
| | - Coline L Lemale
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Viktor Horst
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sebastian Major
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Vasilis Kola
- Center for Stroke Research Berlin, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Karl Schoknecht
- Medical Faculty, Carl Ludwig Institute for Physiology, University of Leipzig, Leipzig, Germany
| | - Michael Scheel
- Department of Neuroradiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jed A Hartings
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Stefan Wolf
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Johannes Woitzik
- Department of Neurosurgery, Evangelisches Krankenhaus Oldenburg, University of Oldenburg, Oldenburg, Germany
| | - Nils Hecht
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
4
|
Lemale CL, Lückl J, Horst V, Reiffurth C, Major S, Hecht N, Woitzik J, Dreier JP. Migraine Aura, Transient Ischemic Attacks, Stroke, and Dying of the Brain Share the Same Key Pathophysiological Process in Neurons Driven by Gibbs–Donnan Forces, Namely Spreading Depolarization. Front Cell Neurosci 2022; 16:837650. [PMID: 35237133 PMCID: PMC8884062 DOI: 10.3389/fncel.2022.837650] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [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
|
5
|
Andrew RD, Hartings JA, Ayata C, Brennan KC, Dawson-Scully KD, Farkas E, Herreras O, Kirov SA, Müller M, Ollen-Bittle N, Reiffurth C, Revah O, Robertson RM, Shuttleworth CW, Ullah G, Dreier JP. The Critical Role of Spreading Depolarizations in Early Brain Injury: Consensus and Contention. Neurocrit Care 2022; 37:83-101. [PMID: 35257321 PMCID: PMC9259543 DOI: 10.1007/s12028-021-01431-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 12/29/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND When a patient arrives in the emergency department following a stroke, a traumatic brain injury, or sudden cardiac arrest, there is no therapeutic drug available to help protect their jeopardized neurons. One crucial reason is that we have not identified the molecular mechanisms leading to electrical failure, neuronal swelling, and blood vessel constriction in newly injured gray matter. All three result from a process termed spreading depolarization (SD). Because we only partially understand SD, we lack molecular targets and biomarkers to help neurons survive after losing their blood flow and then undergoing recurrent SD. METHODS In this review, we introduce SD as a single or recurring event, generated in gray matter following lost blood flow, which compromises the Na+/K+ pump. Electrical recovery from each SD event requires so much energy that neurons often die over minutes and hours following initial injury, independent of extracellular glutamate. RESULTS We discuss how SD has been investigated with various pitfalls in numerous experimental preparations, how overtaxing the Na+/K+ ATPase elicits SD. Elevated K+ or glutamate are unlikely natural activators of SD. We then turn to the properties of SD itself, focusing on its initiation and propagation as well as on computer modeling. CONCLUSIONS Finally, we summarize points of consensus and contention among the authors as well as where SD research may be heading. In an accompanying review, we critique the role of the glutamate excitotoxicity theory, how it has shaped SD research, and its questionable importance to the study of early brain injury as compared with SD theory.
Collapse
Affiliation(s)
- R. David Andrew
- grid.410356.50000 0004 1936 8331Queen’s University, Kingston, ON Canada
| | - Jed A. Hartings
- grid.24827.3b0000 0001 2179 9593University of Cincinnati, Cincinnati, OH USA
| | - Cenk Ayata
- grid.38142.3c000000041936754XHarvard Medical School, Harvard University, Boston, MA USA
| | - K. C. Brennan
- grid.223827.e0000 0001 2193 0096The University of Utah, Salt Lake City, UT USA
| | | | - Eszter Farkas
- grid.9008.10000 0001 1016 96251HCEMM-USZ Cerebral Blood Flow and Metabolism Research Group, and the Department of Cell Biology and Molecular Medicine, Faculty of Science and Informatics & Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Oscar Herreras
- grid.419043.b0000 0001 2177 5516Instituto de Neurobiologia Ramon Y Cajal (Consejo Superior de Investigaciones Científicas), Madrid, Spain
| | - Sergei. A. Kirov
- grid.410427.40000 0001 2284 9329Medical College of Georgia, Augusta, GA USA
| | - Michael Müller
- grid.411984.10000 0001 0482 5331University of Göttingen, University Medical Center Göttingen, Göttingen, Germany
| | - Nikita Ollen-Bittle
- grid.39381.300000 0004 1936 8884University of Western Ontario, London, ON Canada
| | - Clemens Reiffurth
- grid.7468.d0000 0001 2248 7639Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; and the Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health., Berlin, Germany
| | - Omer Revah
- grid.168010.e0000000419368956School of Medicine, Stanford University, Stanford, CA USA
| | | | | | - Ghanim Ullah
- grid.170693.a0000 0001 2353 285XUniversity of South Florida, Tampa, FL USA
| | - Jens P. Dreier
- grid.7468.d0000 0001 2248 7639Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; and the Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health., Berlin, Germany
| |
Collapse
|
6
|
Maruhashi T, Higashi Y. An overview of pharmacotherapy for cerebral vasospasm and delayed cerebral ischemia after subarachnoid hemorrhage. Expert Opin Pharmacother 2021; 22:1601-1614. [PMID: 33823726 DOI: 10.1080/14656566.2021.1912013] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction: Survival from aneurysmal subarachnoid hemorrhage has increased in the past few decades. However, functional outcome after subarachnoid hemorrhage is still suboptimal. Delayed cerebral ischemia (DCI) is one of the major causes of morbidity.Areas covered: Mechanisms underlying vasospasm and DCI after aneurysmal subarachnoid hemorrhage and pharmacological treatment are summarized in this review.Expert opinion: Oral nimodine, an L-type dihydropyridine calcium channel blocker, is the only FDA-approved drug for the prevention and treatment of neurological deficits after aneurysmal subarachnoid hemorrhage. Fasudil, a potent Rho-kinase inhibitor, has also been shown to improve the clinical outcome and has been approved in some countries for use in patients with aneurysmal subarachnoid hemorrhage. Although other drugs, including nicardipine, cilostazol, statins, clazosentan, magnesium and heparin, have been expected to have beneficial effects on DCI, there has been no convincing evidence supporting the routine use of those drugs in patients with aneurysmal subarachnoid hemorrhage in clinical practice. Further elucidation of the mechanisms underlying DCI and the development of effective therapeutic strategies for DCI, including combination therapy, are necessary to further improve the functional outcome and mortality after subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Tatsuya Maruhashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yukihito Higashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.,Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
7
|
Dannenberg F, Prager C, Schmidt F, Tietze A, Bittigau P, Kaindl AM. Intravenous Nimodipine Treatment for Severe Episode of ATP1A2 Hemiplegic Migraine. Pediatr Neurol 2020; 112:71-72. [PMID: 32920306 DOI: 10.1016/j.pediatrneurol.2020.07.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/05/2020] [Accepted: 07/18/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Fabian Dannenberg
- Department of Pediatric Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Center for Chronically Sick Children, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christine Prager
- Department of Pediatric Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Center for Chronically Sick Children, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Schmidt
- Pediatric Pulmology and Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Tietze
- Institute of Neuroradiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Bittigau
- Department of Pediatric Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Center for Chronically Sick Children, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Angela M Kaindl
- Department of Pediatric Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Center for Chronically Sick Children, Charité - Universitätsmedizin Berlin, Berlin, Germany; Institute for Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
8
|
Li R, Ng TS, Garlin MA, Weissleder R, Miller MA. Understanding the in vivo Fate of Advanced Materials by Imaging. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1910369. [PMID: 38545084 PMCID: PMC10972611 DOI: 10.1002/adfm.201910369] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/09/2020] [Indexed: 11/13/2024]
Abstract
Engineered materials are ubiquitous in biomedical applications ranging from systemic drug delivery systems to orthopedic implants, and their actions unfold across multiple time- and length-scales. The efficacy and safety of biologics, nanomaterials, and macroscopic implants are all dictated by the same general principles of pharmacology as apply to small molecule drugs, comprising how the body affects materials (pharmacokinetics, PK) and conversely how materials affect the body (pharmacodynamics, PD). Imaging technologies play an increasingly insightful role in monitoring both of these processes, often simultaneously: translational macroscopic imaging modalities such as MRI and PET/CT offer whole-body quantitation of biodistribution and structural or molecular response, while ex vivo approaches and optical imaging via in vivo (intravital) microscopy reveal behaviors at subcellular resolution. In this review, the authors survey developments in imaging the in situ behavior of systemically and locally administered materials, with a particular focus on using microscopy to understand transport, target engagement, and downstream host responses at a single-cell level. The themes of microenvironmental influence, controlled drug release, on-target molecular action, and immune response, especially as mediated by macrophages and other myeloid cells are examined. Finally, the future directions of how new imaging technologies may propel efficient clinical translation of next-generation therapeutics and medical devices are proposed.
Collapse
Affiliation(s)
- Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Thomas S.C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Michelle A. Garlin
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School
- Department of Systems Biology, Harvard Medical School
| | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School
| |
Collapse
|
9
|
Oliveira-Ferreira AI, Major S, Przesdzing I, Kang EJ, Dreier JP. Spreading depolarizations in the rat endothelin-1 model of focal cerebellar ischemia. J Cereb Blood Flow Metab 2020; 40:1274-1289. [PMID: 31280632 PMCID: PMC7232780 DOI: 10.1177/0271678x19861604] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Focal brain ischemia is best studied in neocortex and striatum. Both show highly vulnerable neurons and high susceptibility to spreading depolarization (SD). Therefore, it has been hypothesized that these two variables generally correlate. However, this hypothesis is contradicted by findings in cerebellar cortex, which contains highly vulnerable neurons to ischemia, the Purkinje cells, but is said to be less susceptible to SD. Here, we found in the rat cerebellar cortex that elevated K+ induced a long-lasting depolarizing event superimposed with SDs. Cerebellar SDs resembled those in neocortex, but negative direct current (DC) shifts and regional blood flow responses were usually smaller. The K+ threshold for SD was higher in cerebellum than in previous studies in neocortex. We then topically applied endothelin-1 (ET-1) to the cerebellum, which is assumed to cause SD via vasoconstriction-induced focal ischemia. Although the blood flow decrease was similar to that in previous studies in neocortex, the ET-1 threshold for SD was higher. Quantitative cell counting found that the proportion of necrotic Purkinje cells was significantly higher in ET-1-treated rats than sham controls even if ET-1 had not caused SDs. Our results suggest that ischemic death of Purkinje cells does not require the occurrence of SD.
Collapse
Affiliation(s)
- Ana I Oliveira-Ferreira
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sebastian Major
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ingo Przesdzing
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Eun-Jeung Kang
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jens P Dreier
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany.,Einstein Center for Neurosciences Berlin, Berlin, Germany
| |
Collapse
|
10
|
Major S, Huo S, Lemale CL, Siebert E, Milakara D, Woitzik J, Gertz K, Dreier JP. Direct electrophysiological evidence that spreading depolarization-induced spreading depression is the pathophysiological correlate of the migraine aura and a review of the spreading depolarization continuum of acute neuronal mass injury. GeroScience 2020; 42:57-80. [PMID: 31820363 PMCID: PMC7031471 DOI: 10.1007/s11357-019-00142-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023] Open
Abstract
Spreading depolarization is observed as a large negative shift of the direct current potential, swelling of neuronal somas, and dendritic beading in the brain's gray matter and represents a state of a potentially reversible mass injury. Its hallmark is the abrupt, massive ion translocation between intraneuronal and extracellular compartment that causes water uptake (= cytotoxic edema) and massive glutamate release. Dependent on the tissue's energy status, spreading depolarization can co-occur with different depression or silencing patterns of spontaneous activity. In adequately supplied tissue, spreading depolarization induces spreading depression of activity. In severely ischemic tissue, nonspreading depression of activity precedes spreading depolarization. The depression pattern determines the neurological deficit which is either spreading such as in migraine aura or migraine stroke or nonspreading such as in transient ischemic attack or typical stroke. Although a clinical distinction between spreading and nonspreading focal neurological deficits is useful because they are associated with different probabilities of permanent damage, it is important to note that spreading depolarization, the neuronal injury potential, occurs in all of these conditions. Here, we first review the scientific basis of the continuum of spreading depolarizations. Second, we highlight the transition zone of the continuum from reversibility to irreversibility using clinical cases of aneurysmal subarachnoid hemorrhage and cerebral amyloid angiopathy. These illustrate how modern neuroimaging and neuromonitoring technologies increasingly bridge the gap between basic sciences and clinic. For example, we provide direct electrophysiological evidence for the first time that spreading depolarization-induced spreading depression is the pathophysiological correlate of the migraine aura.
Collapse
Affiliation(s)
- Sebastian Major
- Center for Stroke Research, Campus Charité Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Shufan Huo
- Center for Stroke Research, Campus Charité Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Coline L Lemale
- Center for Stroke Research, Campus Charité Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Eberhard Siebert
- Department of Neuroradiology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Denny Milakara
- Solution Centre for Image Guided Local Therapies (STIMULATE), Otto-von-Guericke-University, Magdeburg, Germany
| | - Johannes Woitzik
- Evangelisches Krankenhaus Oldenburg, University of Oldenburg, Oldenburg, Germany
| | - Karen Gertz
- Center for Stroke Research, Campus Charité Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jens P Dreier
- Center for Stroke Research, Campus Charité Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- Department of Neurology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany.
- Einstein Center for Neurosciences Berlin, Berlin, Germany.
| |
Collapse
|
11
|
Carlson AP, Hänggi D, Macdonald RL, Shuttleworth CW. Nimodipine Reappraised: An Old Drug With a Future. Curr Neuropharmacol 2020; 18:65-82. [PMID: 31560289 PMCID: PMC7327937 DOI: 10.2174/1570159x17666190927113021] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/02/2019] [Accepted: 09/25/2019] [Indexed: 12/21/2022] Open
Abstract
Nimodipine is a dihydropyridine calcium channel antagonist that blocks the flux of extracellular calcium through L-type, voltage-gated calcium channels. While nimodipine is FDAapproved for the prevention and treatment of neurological deficits in patients with aneurysmal subarachnoid hemorrhage (aSAH), it affects myriad cell types throughout the body, and thus, likely has more complex mechanisms of action than simple inhibition of cerebral vasoconstriction. Newer understanding of the pathophysiology of delayed ischemic injury after a variety of acute neurologic injuries including aSAH, traumatic brain injury (TBI) and ischemic stroke, coupled with advances in the drug delivery method for nimodipine, have reignited interest in refining its potential therapeutic use. In this context, this review seeks to establish a firm understanding of current data on nimodipine's role in the mechanisms of delayed injury in aSAH, TBI, and ischemic stroke, and assess the extensive clinical data evaluating its use in these conditions. In addition, we will review pivotal trials using locally administered, sustained release nimodipine and discuss why such an approach has evaded demonstration of efficacy, while seemingly having the potential to significantly improve clinical care.
Collapse
Affiliation(s)
- Andrew P. Carlson
- Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Daniel Hänggi
- Department of Neurosurgery, University of Dusseldorf Hospital, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Robert L. Macdonald
- University of California San Francisco Fresno Department of Neurosurgery and University Neurosciences Institute and Division of Neurosurgery, Department of Surgery, University of Toronto, Canada
| | - Claude W. Shuttleworth
- Department of Neuroscience University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
12
|
Hartings JA, York J, Carroll CP, Hinzman JM, Mahoney E, Krueger B, Winkler MKL, Major S, Horst V, Jahnke P, Woitzik J, Kola V, Du Y, Hagen M, Jiang J, Dreier JP. Subarachnoid blood acutely induces spreading depolarizations and early cortical infarction. Brain 2019; 140:2673-2690. [PMID: 28969382 DOI: 10.1093/brain/awx214] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 07/10/2017] [Indexed: 01/05/2023] Open
Abstract
See Ghoshal and Claassen (doi:10.1093/brain/awx226) for a scientific commentary on this article.
Early cortical infarcts are common in poor-grade patients after aneurysmal subarachnoid haemorrhage. There are no animal models of these lesions and mechanisms are unknown, although mass cortical spreading depolarizations are hypothesized as a requisite mechanism and clinical marker of infarct development. Here we studied acute sequelae of subarachnoid haemorrhage in the gyrencephalic brain of propofol-anaesthetized juvenile swine using subdural electrode strips (electrocorticography) and intraparenchymal neuromonitoring probes. Subarachnoid infusion of 1–2 ml of fresh blood at 200 µl/min over cortical sulci caused clusters of spreading depolarizations (count range: 12–34) in 7/17 animals in the ipsilateral but not contralateral hemisphere in 6 h of monitoring, without meaningful changes in other variables. Spreading depolarization clusters were associated with formation of sulcal clots (P < 0.01), a high likelihood of adjacent cortical infarcts (5/7 versus 2/10, P < 0.06), and upregulation of cyclooxygenase-2 in ipsilateral cortex remote from clots/infarcts. In a second cohort, infusion of 1 ml of clotted blood into a sulcus caused spreading depolarizations in 5/6 animals (count range: 4–20 in 6 h) and persistent thick clots with patchy or extensive infarction of circumscribed cortex in all animals. Infarcts were significantly larger after blood clot infusion compared to mass effect controls using fibrin clots of equal volume. Haematoxylin and eosin staining of infarcts showed well demarcated zones of oedema and hypoxic-ischaemic neuronal injury, consistent with acute infarction. The association of spreading depolarizations with early brain injury was then investigated in 23 patients [14 female; age (median, quartiles): 57 years (47, 63)] after repair of ruptured anterior communicating artery aneurysms by clip ligation (n = 14) or coiling (n = 9). Frontal electrocorticography [duration: 54 h (34, 66)] from subdural electrode strips was analysed over Days 0–3 after initial haemorrhage and magnetic resonance imaging studies were performed at ∼ 24–48 h after aneurysm treatment. Patients with frontal infarcts only and those with frontal infarcts and/or intracerebral haemorrhage were both significantly more likely to have spreading depolarizations (6/7 and 10/12, respectively) than those without frontal brain lesions (1/11, P’s < 0.05). These results suggest that subarachnoid clots in sulci/fissures are sufficient to induce spreading depolarizations and acute infarction in adjacent cortex. We hypothesize that the cellular toxicity and vasoconstrictive effects of depolarizations act in synergy with direct ischaemic effects of haemorrhage as mechanisms of infarct development. Results further validate spreading depolarizations as a clinical marker of early brain injury and establish a clinically relevant model to investigate causal pathologic sequences and potential therapeutic interventions.
Collapse
Affiliation(s)
- Jed A Hartings
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,UC Gardner Neuroscience Institute and Mayfield Clinic, Cincinnati, OH, USA
| | - Jonathan York
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Christopher P Carroll
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jason M Hinzman
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Eric Mahoney
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Bryan Krueger
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Maren K L Winkler
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Germany
| | - Sebastian Major
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Germany.,Department of Neurology, Charité University Medicine Berlin, Germany.,Department of Experimental Neurology, Charité University Medicine Berlin, Germany
| | - Viktor Horst
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Germany
| | - Paul Jahnke
- Department of Radiology Charité University Medicine Berlin, Germany
| | - Johannes Woitzik
- Department of Neurosurgery, Charité University Medicine Berlin, Germany
| | - Vasilis Kola
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Germany
| | - Yifeng Du
- Division of Pharmaceutical Sciences, University of Cincinnati College of Pharmacy, Cincinnati, OH, USA
| | - Matthew Hagen
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jianxiong Jiang
- Division of Pharmaceutical Sciences, University of Cincinnati College of Pharmacy, Cincinnati, OH, USA
| | - Jens P Dreier
- Center for Stroke Research Berlin, Charité University Medicine Berlin, Germany.,Department of Neurology, Charité University Medicine Berlin, Germany.,Department of Experimental Neurology, Charité University Medicine Berlin, Germany
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW Spreading depolarizations are unique in being discrete pathologic entities that are well characterized experimentally and also occur commonly in patients with substantial acute brain injury. Here, we review essential concepts in depolarization monitoring, highlighting its clinical significance, interpretation, and future potential. RECENT FINDINGS Cortical lesion development in diverse animal models is mediated by tissue waves of mass spreading depolarization that cause the toxic loss of ion homeostasis and limit energy substrate supply through associated vasoconstriction. The signatures of such deterioration are observed in electrocorticographic recordings from perilesional cortex of patients with acute stroke or brain trauma. Experimental work suggests that depolarizations are triggered by energy supply-demand mismatch in focal hotspots of the injury penumbra, and depolarizations are usually observed clinically when other monitoring variables are within recommended ranges. These results suggest that depolarizations are a sensitive measure of relative ischemia and ongoing secondary injury, and may serve as a clinical guide for personalized, mechanistically targeted therapy. Both existing and future candidate therapies offer hope to limit depolarization recurrence. SUMMARY Electrocorticographic monitoring of spreading depolarizations in patients with acute brain injury provides a sensitive measure of relative energy shortage in focal, vulnerable brains regions and indicates ongoing secondary damage. Depolarization monitoring holds potential for targeted clinical trial design and implementation of precision medicine approaches to acute brain injury therapy.
Collapse
|
14
|
Dreier JP, Major S, Foreman B, Winkler MKL, Kang EJ, Milakara D, Lemale CL, DiNapoli V, Hinzman JM, Woitzik J, Andaluz N, Carlson A, Hartings JA. Terminal spreading depolarization and electrical silence in death of human cerebral cortex. Ann Neurol 2018; 83:295-310. [PMID: 29331091 PMCID: PMC5901399 DOI: 10.1002/ana.25147] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 11/10/2022]
Abstract
OBJECTIVE Restoring the circulation is the primary goal in emergency treatment of cerebral ischemia. However, better understanding of how the brain responds to energy depletion could help predict the time available for resuscitation until irreversible damage and advance development of interventions that prolong this span. Experimentally, injury to central neurons begins only with anoxic depolarization. This potentially reversible, spreading wave typically starts 2 to 5 minutes after the onset of severe ischemia, marking the onset of a toxic intraneuronal change that eventually results in irreversible injury. METHODS To investigate this in the human brain, we performed recordings with either subdural electrode strips (n = 4) or intraparenchymal electrode arrays (n = 5) in patients with devastating brain injury that resulted in activation of a Do Not Resuscitate-Comfort Care order followed by terminal extubation. RESULTS Withdrawal of life-sustaining therapies produced a decline in brain tissue partial pressure of oxygen (pti O2 ) and circulatory arrest. Silencing of spontaneous electrical activity developed simultaneously across regional electrode arrays in 8 patients. This silencing, termed "nonspreading depression," developed during the steep falling phase of pti O2 (intraparenchymal sensor, n = 6) at 11 (interquartile range [IQR] = 7-14) mmHg. Terminal spreading depolarizations started to propagate between electrodes 3.9 (IQR = 2.6-6.3) minutes after onset of the final drop in perfusion and 13 to 266 seconds after nonspreading depression. In 1 patient, terminal spreading depolarization induced the initial electrocerebral silence in a spreading depression pattern; circulatory arrest developed thereafter. INTERPRETATION These results provide fundamental insight into the neurobiology of dying and have important implications for survivable cerebral ischemic insults. Ann Neurol 2018;83:295-310.
Collapse
Affiliation(s)
- Jens P Dreier
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Departments of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany.,Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - Sebastian Major
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Departments of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Brandon Foreman
- UC Gardner Neuroscience Institute.,Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Maren K L Winkler
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Eun-Jeung Kang
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Denny Milakara
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Coline L Lemale
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Vince DiNapoli
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH.,Mayfield Clinic, Cincinnati, OH
| | - Jason M Hinzman
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Johannes Woitzik
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Norberto Andaluz
- UC Gardner Neuroscience Institute.,Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH.,Mayfield Clinic, Cincinnati, OH
| | - Andrew Carlson
- Department of Neurosurgery, University of New Mexico, Albuquerque, NM
| | - Jed A Hartings
- UC Gardner Neuroscience Institute.,Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
15
|
Dreier JP, Lemale CL, Kola V, Friedman A, Schoknecht K. Spreading depolarization is not an epiphenomenon but the principal mechanism of the cytotoxic edema in various gray matter structures of the brain during stroke. Neuropharmacology 2017; 134:189-207. [PMID: 28941738 DOI: 10.1016/j.neuropharm.2017.09.027] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 09/16/2017] [Accepted: 09/19/2017] [Indexed: 12/15/2022]
Abstract
Spreading depolarization (SD) is a phenomenon of various cerebral gray matter structures that only occurs under pathological conditions. In the present paper, we summarize the evidence from several decades of research that SD and cytotoxic edema in these structures are largely overlapping terms. SD/cytotoxic edema is a toxic state that - albeit initially reversible - leads eventually to cellular death when it is persistent. Both hemorrhagic and ischemic stroke are among the most prominent causes of SD/cytotoxic edema. SD/cytotoxic edema is the principal mechanism that mediates neuronal death in these conditions. This applies to gray matter structures in both the ischemic core and the penumbra. SD/cytotoxic edema is often a single terminal event in the core whereas, in the penumbra, a cluster of repetitive prolonged SDs is typical. SD/cytotoxic edema also propagates widely into healthy surrounding tissue as short-lasting, relatively harmless events so that regional electrocorticographic monitoring affords even remote detection of ischemic zones. Ischemia cannot only cause SD/cytotoxic edema but it can also be its consequence through inverse neurovascular coupling. Under this condition, ischemia does not start simultaneously in different regions but spreads in the tissue driven by SD/cytotoxic edema-induced microvascular constriction (= spreading ischemia). Spreading ischemia prolongs SD/cytotoxic edema. Thus, it increases the likelihood for the transition from SD/cytotoxic edema into cellular death. Vasogenic edema is the other major type of cerebral edema with relevance to ischemic stroke. It results from opening of the blood-brain barrier. SD/cytotoxic edema and vasogenic edema are distinct processes with important mutual interactions. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
Collapse
Affiliation(s)
- Jens P Dreier
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany; Departments of Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany; Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.
| | - Coline L Lemale
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Vasilis Kola
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Alon Friedman
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, Canada
| | - Karl Schoknecht
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany; Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
16
|
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
|
17
|
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: 243] [Impact Index Per Article: 30.4] [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
|
18
|
Richter F, Eitner A, Leuchtweis J, Bauer R, Lehmenkühler A, Schaible HG. Effects of interleukin-1ß on cortical spreading depolarization and cerebral vasculature. J Cereb Blood Flow Metab 2017; 37:1791-1802. [PMID: 27037093 PMCID: PMC5435277 DOI: 10.1177/0271678x16641127] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 01/26/2016] [Accepted: 02/22/2016] [Indexed: 11/16/2022]
Abstract
During brain damage and ischemia, the cytokine interleukin-1ß is rapidly upregulated due to activation of inflammasomes. We studied whether interleukin-1ß influences cortical spreading depolarization, and whether lipopolysaccharide, often used for microglial stimulation, influences cortical spreading depolarizations. In anaesthetized rats, cortical spreading depolarizations were elicited by microinjection of KCl. Interleukin-1ß, the IL-1 receptor 1 antagonist, the GABAA receptor blocker bicuculline, and lipopolysaccharide were administered either alone or combined (interleukin-1ß + IL-1 receptor 1 antagonist; interleukin-1ß + bicuculline; lipopolysaccharide + IL-1 receptor 1 antagonist) into a local cortical treatment area. Using microelectrodes, cortical spreading depolarizations were recorded in a non-treatment and in the treatment area. Plasma extravasation in cortical grey matter was assessed with Evans blue. Local application of interleukin-1ß reduced cortical spreading depolarization amplitudes in the treatment area, but not at a high dose. This reduction was prevented by IL-1 receptor 1 antagonist and by bicuculline. However, interleukin-1ß induced pronounced plasma extravasation independently on cortical spreading depolarizations. Application of lipopolysaccharide reduced cortical spreading depolarization amplitudes but prolonged their duration; EEG activity was still present. These effects were also blocked by IL-1 receptor 1 antagonist. Interleukin-1ß evokes changes of neuronal activity and of vascular functions. Thus, although the reduction of cortical spreading depolarization amplitudes at lower doses of interleukin-1ß may reduce deleterious effects of cortical spreading depolarizations, the sum of interleukin-1ß effects on excitability and on the vasculature rather promote brain damaging mechanisms.
Collapse
Affiliation(s)
- Frank Richter
- Institute of Physiology I/Neurophysiology, Jena University Hospital – Friedrich Schiller University Jena, Jena, Germany
| | - Annett Eitner
- Institute of Physiology I/Neurophysiology, Jena University Hospital – Friedrich Schiller University Jena, Jena, Germany
| | - Johannes Leuchtweis
- Institute of Physiology I/Neurophysiology, Jena University Hospital – Friedrich Schiller University Jena, Jena, Germany
| | - Reinhard Bauer
- Institute of Molecular Cell Biology, Jena University Hospital – Friedrich Schiller University Jena, Jena, Germany
| | | | - Hans-Georg Schaible
- Institute of Physiology I/Neurophysiology, Jena University Hospital – Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
19
|
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: 279] [Impact Index Per Article: 34.9] [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
|
20
|
[Intensive care treatment after aneurysmal subarachnoid hemorrhage]. Anaesthesist 2017; 65:951-970. [PMID: 27900416 DOI: 10.1007/s00101-016-0242-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Aneurysmal subarachnoid hemorrhage (SAH) is a devastating disease and nearly one third of patients die in the acute phase. Due to the bleeding event, a hyperactive sympathetic nervous system and an uncontrolled inflammatory response have a profound local and systemic impact on other organ functions. Neuroendocrinological disorders and cardiopulmonary morbidity are dominant. Despite a decrease in hospital mortality for high volume centers, a high proportion of survivors suffer from neurological deficits. Knowledge of the pathophysiology of vasospasms in the later stages of the disease has increased. Anti-inflammatory treatment does not improve the outcome. Nimodipine prophylaxis in the first 96 h after SAH seems to be the only intervention which has been proven to be advantageous in studies; however, nearly every second survivor of SAH suffers from some neurological deficits and more than one third of survivors report depressive episodes or symptoms of posttraumatic stress disorder.
Collapse
|
21
|
Santos E, León F, Silos H, Sanchez-Porras R, Shuttleworth CW, Unterberg A, Sakowitz OW. Incidence, hemodynamic, and electrical characteristics of spreading depolarization in a swine model are affected by local but not by intravenous application of magnesium. J Cereb Blood Flow Metab 2016; 36:2051-2057. [PMID: 27683450 PMCID: PMC5363671 DOI: 10.1177/0271678x16671317] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/26/2016] [Accepted: 08/28/2016] [Indexed: 11/17/2022]
Abstract
The aim was to characterize the effects of magnesium sulfate, using i.v. bolus and local administration, using intrinsic signal imaging, and on electrocorticographic activity during the induction and propagation of spreading depolarizations in the gyrencephalic porcine brain. Local application of magnesium sulfate led to a complete inhibition of spreading depolarizations. One hour after washing out the topical magnesium sulfate, re-incidence of the spreading depolarizations was observed in 50% of the hemispheres. Those spreading depolarizations showed attenuation in hemodynamic characteristics and speed in intrinsic optical signal imaging. The electrical amplitude decreased through electrocorticographic activity. Intravenous magnesium therapy showed no significant effects on spreading depolarization incidence and characteristics.
Collapse
Affiliation(s)
- Edgar Santos
- Department of Neurosurgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Fiorella León
- Department of Neurosurgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Humberto Silos
- Department of Neurosurgery, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | - Andreas Unterberg
- Department of Neurosurgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Oliver W Sakowitz
- Department of Neurosurgery, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
22
|
Abstract
OPINION STATEMENT New neuroprotective treatments aimed at preventing or minimizing "delayed brain injury" are attractive areas of investigation and hold the potential to have substantial beneficial effects on aneurysmal subarachnoid hemorrhage (aSAH) survivors. The underlying mechanisms for this "delayed brain injury" are multi-factorial and not fully understood. The most ideal treatment strategies would have the potential for a pleotropic effect positively modulating multiple implicated pathophysiological mechanisms at once. My personal management (RFJ) of patients with aneurysmal subarachnoid hemorrhage closely follows those treatment recommendations contained in modern published guidelines. However, over the last 5 years, I have also utilized a novel treatment strategy, originally developed at the University of Maryland, which consists of a 14-day continuous low-dose intravenous heparin infusion (LDIVH) beginning 12 h after securing the ruptured aneurysm. In addition to its well-known anti-coagulant properties, unfractionated heparin has potent anti-inflammatory effects and through multiple mechanisms may favorably modulate the neurotoxic and neuroinflammatory processes prominent in aneurysmal subarachnoid hemorrhage. In my personal series of patients treated with LDIVH, I have found significant preservation of neurocognitive function as measured by the Montreal Cognitive Assessment (MoCA) compared to a control cohort of my patients treated without LDIVH (RFJ unpublished data presented at the 2015 AHA/ASA International Stroke Conference symposium on neuroinflammation in aSAH and in abstract format at the 2015 AANS/CNS Joint Cerebrovascular Section Annual Meeting). It is important for academic physicians involved in the management of these complex patients to continue to explore new treatment options that may be protective against the potentially devastating "delayed brain injury" following cerebral aneurysm rupture. Several of the treatment options included in this review show promise and could be carefully adopted as the level of evidence for each improves. Other proposed neuroprotective treatments like statins and magnesium sulfate were previously thought to be very promising and to varying degrees were adopted at numerous institutions based on somewhat limited human evidence. Recent clinical trials and meta-analysis have shown no benefit for these treatments, and I currently no longer utilize either treatment as prophylaxis in my practice.
Collapse
|
23
|
Abstract
The term spreading depolarization (SD) refers to waves of abrupt, sustained mass depolarization in gray matter of the CNS. SD, which spreads from neuron to neuron in affected tissue, is characterized by a rapid near-breakdown of the neuronal transmembrane ion gradients. SD can be induced by hypoxic conditions--such as from ischemia--and facilitates neuronal death in energy-compromised tissue. SD has also been implicated in migraine aura, where SD is assumed to ascend in well-nourished tissue and is typically benign. In addition to these two ends of the "SD continuum," an SD wave can propagate from an energy-depleted tissue into surrounding, well-nourished tissue, as is often the case in stroke and brain trauma. This review presents the neurobiology of SD--its triggers and propagation mechanisms--as well as clinical manifestations of SD, including overlaps and differences between migraine aura and stroke, and recent developments in neuromonitoring aimed at better diagnosis and more targeted treatments.
Collapse
Affiliation(s)
- Jens P Dreier
- Department of Neurology, Charité University Medicine Berlin, 10117 Berlin, Germany; Department of Experimental Neurology, Charité University Medicine Berlin, 10117 Berlin, Germany; Center for Stroke Research, Charité University Medicine Berlin, 10117 Berlin, Germany.
| | - Clemens Reiffurth
- Department of Experimental Neurology, Charité University Medicine Berlin, 10117 Berlin, Germany; Center for Stroke Research, Charité University Medicine Berlin, 10117 Berlin, Germany
| |
Collapse
|
24
|
Beitzke M, Enzinger C, Wünsch G, Asslaber M, Gattringer T, Fazekas F. Contribution of convexal subarachnoid hemorrhage to disease progression in cerebral amyloid angiopathy. Stroke 2015; 46:1533-40. [PMID: 25953372 DOI: 10.1161/strokeaha.115.008778] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 04/01/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Cerebral amyloid angiopathy-related cortical superficial siderosis (cSS) seems to indicate an increased risk of subsequent intracerebral hemorrhage (ICH). We wanted to identify the mechanisms and sequence of hemorrhagic events which are responsible for this association. METHODS During a 9-year-period, we identified patients with spontaneous convexal subarachnoid hemorrhage (cSAH) and performed a careful longitudinal analysis of clinical and neuroimaging data. A close imaging-histopathologic correlation was performed in one patient. RESULTS Of 38 cSAH patients (mean age, 77±11 years), 29 (76%) had imaging features of cerebral amyloid angiopathy on baseline magnetic resonance imaging. Twenty-six (68%) had cSS. Sixteen subjects underwent postcontrast magnetic resonance imaging. Extravasation of gadolinium at the site of the acute cSAH was seen on all postcontrast scans. After a mean of 24±22 (range 1-78) months of follow-up, 15 (39%) had experienced recurrent cSAHs and 14 (37%) had suffered lobar ICHs. Of 22 new ICHs, 17 occurred at sites of previous cSAHs or cSS. Repeated neuroimaging showed expansion of cSAH into the brain parenchyma and evolution of a lobar ICH in 4 patients. Propagation of cSS was observed in 21 (55%) patients, with 14 of those having experienced recurrent cSAHs. In the autopsy case, leakage of meningeal vessels affected by cerebral amyloid angiopathy was noted. CONCLUSIONS In cerebral amyloid angiopathy, leakage of meningeal vessels seems to be a major cause for recurrent intrasulcal bleedings, which lead to the propagation of cSS and indicate sites with increased vulnerability for future ICH. Intracerebral bleedings may also develop directly from or in extension of a cSAH.
Collapse
Affiliation(s)
- Markus Beitzke
- From the Department of Neurology, Medical University of Graz, Graz, Austria (M.B., C.E., T.G., F.F.); Division of Neuroradiology, Department of Radiology, Medical University of Graz, Graz, Austria (C.E.); Department of Pathology, Medical University of Graz, Graz, Austria (M.A.); and Department for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria (G.W.).
| | - Christian Enzinger
- From the Department of Neurology, Medical University of Graz, Graz, Austria (M.B., C.E., T.G., F.F.); Division of Neuroradiology, Department of Radiology, Medical University of Graz, Graz, Austria (C.E.); Department of Pathology, Medical University of Graz, Graz, Austria (M.A.); and Department for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria (G.W.)
| | - Gerit Wünsch
- From the Department of Neurology, Medical University of Graz, Graz, Austria (M.B., C.E., T.G., F.F.); Division of Neuroradiology, Department of Radiology, Medical University of Graz, Graz, Austria (C.E.); Department of Pathology, Medical University of Graz, Graz, Austria (M.A.); and Department for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria (G.W.)
| | - Martin Asslaber
- From the Department of Neurology, Medical University of Graz, Graz, Austria (M.B., C.E., T.G., F.F.); Division of Neuroradiology, Department of Radiology, Medical University of Graz, Graz, Austria (C.E.); Department of Pathology, Medical University of Graz, Graz, Austria (M.A.); and Department for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria (G.W.)
| | - Thomas Gattringer
- From the Department of Neurology, Medical University of Graz, Graz, Austria (M.B., C.E., T.G., F.F.); Division of Neuroradiology, Department of Radiology, Medical University of Graz, Graz, Austria (C.E.); Department of Pathology, Medical University of Graz, Graz, Austria (M.A.); and Department for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria (G.W.)
| | - Franz Fazekas
- From the Department of Neurology, Medical University of Graz, Graz, Austria (M.B., C.E., T.G., F.F.); Division of Neuroradiology, Department of Radiology, Medical University of Graz, Graz, Austria (C.E.); Department of Pathology, Medical University of Graz, Graz, Austria (M.A.); and Department for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria (G.W.)
| |
Collapse
|
25
|
How spreading depolarization can be the pathophysiological correlate of both migraine aura and stroke. ACTA NEUROCHIRURGICA. SUPPLEMENT 2015; 120:137-40. [PMID: 25366613 DOI: 10.1007/978-3-319-04981-6_23] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
The term spreading depolarization describes a mechanism of abrupt, massive ion translocation between neurons and the interstitial space, which leads to a cytotoxic edema in the gray matter of the brain. In energy-compromised tissue, spreading depolarization is preceded by a nonspreading silencing (depression of spontaneous activity) because of a neuronal hyperpolarization. By contrast, in tissue that is not energy compromised, spreading depolarization is accompanied by a spreading silencing (spreading depression) of spontaneous activity caused by a depolarization block. It is assumed that the nonspreading silencing translates into the initial clinical symptoms of ischemic stroke and the spreading silencing (spreading depression) into the symptoms of migraine aura. In energy-compromised tissue, spreading depolarization facilitates neuronal death, whereas, in healthy tissue, it is relatively innocuous. Therapies targeting spreading depolarization in metabolically compromised tissue may potentially treat conditions of acute cerebral injury such as aneurysmal subarachnoid hemorrhage.
Collapse
|
26
|
Alieva M, Ritsma L, Giedt RJ, Weissleder R, van Rheenen J. Imaging windows for long-term intravital imaging: General overview and technical insights. INTRAVITAL 2014; 3:e29917. [PMID: 28243510 PMCID: PMC5312719 DOI: 10.4161/intv.29917] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 07/10/2014] [Accepted: 07/11/2014] [Indexed: 01/11/2023]
Abstract
Intravital microscopy is increasingly used to visualize and quantitate dynamic biological processes at the (sub)cellular level in live animals. By visualizing tissues through imaging windows, individual cells (e.g., cancer, host, or stem cells) can be tracked and studied over a time-span of days to months. Several imaging windows have been developed to access tissues including the brain, superficial fascia, mammary glands, liver, kidney, pancreas, and small intestine among others. Here, we review the development of imaging windows and compare the most commonly used long-term imaging windows for cancer biology: the cranial imaging window, the dorsal skin fold chamber, the mammary imaging window, and the abdominal imaging window. Moreover, we provide technical details, considerations, and trouble-shooting tips on the surgical procedures and microscopy setups for each imaging window and explain different strategies to assure imaging of the same area over multiple imaging sessions. This review aims to be a useful resource for establishing the long-term intravital imaging procedure.
Collapse
Affiliation(s)
- Maria Alieva
- Cancer Genomics Netherlands; Hubrecht Institute-KNAW and University Medical Centre Utrecht; CT Utrecht, The Netherlands
| | - Laila Ritsma
- Center for Cancer Research and Center for Regenerative Medicine; Massachusetts General Hospital; Richard B. Simches Research Center; Harvard Medical School; Boston, MA USA; Broad Institute of Harvard and Massachusetts Institute for Technology; Cambridge, MA USA
| | - Randy J Giedt
- Center for Systems Biology; Massachusetts General Hospital; Richard B. Simches Research Center; Harvard Medical School; Boston, MA USA
| | - Ralph Weissleder
- Center for Systems Biology; Massachusetts General Hospital; Richard B. Simches Research Center; Harvard Medical School; Boston, MA USA; Department of Systems Biology; Harvard Medical School; Boston, MA USA
| | - Jacco van Rheenen
- Cancer Genomics Netherlands; Hubrecht Institute-KNAW and University Medical Centre Utrecht; CT Utrecht, The Netherlands
| |
Collapse
|
27
|
Eikermann-Haerter K. Spreading depolarization may link migraine and stroke. Headache 2014; 54:1146-57. [PMID: 24913618 DOI: 10.1111/head.12386] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2014] [Indexed: 12/26/2022]
Abstract
Migraine increases the risk of stroke, particularly in young and otherwise healthy adults. Being the most frequent neurological condition, migraine prevalence is on a par with that of other common stroke risk factors, such as diabetes or hypertension. Several patterns of association have emerged: (1) migraine and stroke share a common association (eg, vasculopathies, patent foramen ovale, or pulmonary A-V malformations); (2) injury to the arterial wall such as acute arterial dissections can present as migraine aura attacks or stroke; (3) strokes rarely develop during a migraine attack, as described for "migrainous stroke." Increasing experimental evidence suggests that cerebral hyperexcitability and enhanced susceptibility to spreading depolarization, the electrophysiologic event underlying migraine, may serve as a mechanism underlying the migraine-stroke association. Mice carrying human vascular or neuronal migraine mutations exhibit an enhanced susceptibility to spreading depolarization while being particularly vulnerable to cerebral ischemia. The severe stroke phenotype in migraine mutant mice can be prevented by suppressing spreading depolarization. If confirmed in the clinical setting, inhibiting spreading depolarization might protect migraineurs at stroke risk as well as decrease attacks of migraine.
Collapse
Affiliation(s)
- Katharina Eikermann-Haerter
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
28
|
Richter F, Lütz W, Eitner A, Leuchtweis J, Lehmenkühler A, Schaible HG. Tumor necrosis factor reduces the amplitude of rat cortical spreading depression in vivo. Ann Neurol 2014; 76:43-53. [PMID: 24798682 DOI: 10.1002/ana.24176] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 05/02/2014] [Accepted: 05/02/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Brain damage and ischemia often trigger cortical spreading depression (CSD), which aggravates brain damage. The proinflammatory cytokine tumor necrosis factor (TNF) is significantly upregulated during brain damage, but it is unknown whether TNF influences spreading depression in cerebral cortex in vivo. This question is important because TNF not only furthers inflammatory reactions but might also be neuroprotective. Here we tested the hypothesis that TNF affects CSD, and we explored the direction in which CSD is modified by TNF. METHODS CSD, elicited by pressure microinjection of KCl, was recorded in anesthetized rats and mice. TNF was administered locally into a trough, providing local TNF treatment of a cortical area. For further analysis, antibodies to TNF receptor (TNFR) 1 or 2 were applied, or CSD was monitored in TNFR1 and TNFR2 knockout mice. γ-Aminobutyric acid (GABA)A receptors were blocked by bicuculline. Immunohistochemistry localized the cortical expression of TNFR1 and TNFR2. RESULTS Local application of TNF to the cortex reduced dose-dependently the amplitude of CSD. This effect was prevented by blockade or knockout of TNFR2 but not by blockade or knockout of TNFR1. TNFR2 was localized at cortical neurons including parvalbumin-positive inhibitory interneurons, and blockade of GABAA receptors by bicuculline prevented the reduction of CSD amplitudes by TNF. INTERPRETATION We identified a functional link between TNF and CSD. TNF activates TNFR2 in cortical inhibitory interneurons. The resulting release of GABA reduces CSD amplitudes. In this manner, TNF might be neuroprotective in pathological conditions.
Collapse
Affiliation(s)
- Frank Richter
- Institute of Physiology I/Neurophysiology, Jena University Hospital-Friedrich Schiller University Jena, Jena
| | | | | | | | | | | |
Collapse
|