1
|
Barbour MA, Whitehead B, Gumbo C, Karelina K, Weil ZM. Traumatic brain injury persistently increases the incidence of both ischemic and hemorrhagic strokes: Potential mechanisms. Prog Neurobiol 2025; 248:102749. [PMID: 40113130 PMCID: PMC12021558 DOI: 10.1016/j.pneurobio.2025.102749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
Traumatic brain injuries (TBI) significantly increase the risk of both ischemic and hemorrhagic strokes, with effects persisting for years after the initial injury. The mechanisms underlying this increased stroke risk are complex, multifactorial, and incompletely understood but likely include chronic cerebrovascular dysfunction, blood-brain barrier disruption, and inflammatory responses. Epidemiological studies consistently show that TBI is an independent risk factor for stroke, with more severe injuries associated with greater risk, especially for hemorrhagic strokes. Traditional risk factors for stroke, such as hypertension, poor diet, and sedentary lifestyle, further elevate the risk in TBI survivors. Modifiable lifestyle factors, such as improving sleep, increasing physical activity, and adopting heart-healthy diets, offer potential intervention points to mitigate stroke risk. Pharmacological considerations, including the use of antidepressants, anticoagulants, and statins, also influence stroke risk, particularly with regard to hemorrhagic complications. This review explores the pathophysiological mechanisms linking TBI and stroke, emphasizing the need for future research to identify specific biomarkers and imaging techniques to predict stroke vulnerability in TBI patients. Addressing the gaps in understanding, particularly regarding small vessel pathology, will be essential to developing targeted therapies for reducing stroke incidence in TBI survivors.
Collapse
Affiliation(s)
- Mikaela A Barbour
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Road, 313 BMRC, Morgantown, WV 26506, USA.
| | - Bailey Whitehead
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Road, 313 BMRC, Morgantown, WV 26506, USA
| | - Claymore Gumbo
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Road, 313 BMRC, Morgantown, WV 26506, USA
| | - Kate Karelina
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Road, 313 BMRC, Morgantown, WV 26506, USA
| | - Zachary M Weil
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Road, 313 BMRC, Morgantown, WV 26506, USA
| |
Collapse
|
2
|
Kapapa T, Pfnür A, Halbgebauer R, Broer P, Halbgebauer S, Tumani H, Friedrichs AK, Huber-Lang M, Dörfer L. Biomarkers in Aneurysmatic and Spontaneous Subarachnoid Haemorrhage: A Clinical Prospective Multicentre Biomarker Panel Study of S100B, Claudin-5, Interleukin-10, TREM-1, TREM-2 and Neurofilament Light Chain As Well As Immunoglobulin G and M. Mol Neurobiol 2025:10.1007/s12035-025-04889-3. [PMID: 40295361 DOI: 10.1007/s12035-025-04889-3] [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: 10/21/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025]
Abstract
Following aneurysmatic subarachnoid haemorrhage (SAH), complex pathophysiological processes take place which result in ischaemia, dysfunction of the blood-brain barrier and the clinical development of vasospasms and delayed cerebral ischaemia (DCI). The aim of this study was to present a biomarker panel that can be used for temporal assignment in the pathophysiological process after haemorrhage, a prediction of vasospasm, DCI or outcome. In a prospective multicentre approach, complex laboratory chemistry tests were used to determine the value of the biomarkers S100B, Claudin-5, Interleukin (IL) -10, Triggering receptor expresses on myeloid cells (TREM)-1 and TREM-2, and neurofilament light chain (NfL) as well as IgG and IgM in plasma and Cerebro-spinal-fluid (CSF) in SAH patients. The predictive power of mentioned biomarkers with regard to the occurrence of vasospasms, DCI and the outcome (Glasgow Outcome Scale) were defined by using sophisticated statistical methods with the level of significance at p ≤ 0.05. Mean age of the 12 patients included was 56 (SD:14) years with 67% female patients and that of the 11 control subjects was 74 (SD:3) years with 55% female subjects. S100B showed higher concentrations compared to the control patients on the first four days (p ≤ 0.0141). For IL-10, the CSF concentrations showed a continuous increase: day 2 (p = 0.0074), day 4 (p = 0.0012), and day 5 (p < 0.0001). Regarding the TREM1 and TREM2 balance, CSF concentrations of TREM1 increased until day eight (p ≤ 0.0055). TREM-2 plasma concentrations decreased below the levels of control patients and appeared unchanged for the further course. The greatest difference in the CSF concentration of NfL between the patients and the control group was seen on day 8 (p = 0.0104). The differentiation between patients with and without DCI showed different concentration curves of the TREM1 CSF-plasma index with increasing concentrations for patients with DCI. The TREM 2 CSF-plasma index showed higher concentrations for patients with DCI. Patients without DCI showed a decreasing concentration of the NfL CSF-plasma index compared to an increase when vasospasm was detected. NfL, TREM-1 and TREM-2 have the potential to be relevant biomarkers for SAH in the intermediate and delayed injury phase.
Collapse
Affiliation(s)
- Thomas Kapapa
- Department of Neurosurgery, University Hospital Ulm, Albert-Einstein-Allee 23, Ulm, 89081, Germany.
| | - Andreas Pfnür
- Department of Neurosurgery, University Hospital Ulm, Albert-Einstein-Allee 23, Ulm, 89081, Germany
| | - Rebecca Halbgebauer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Helmholtzstraße 8/1, Ulm, 89081, Germany
| | - Patrik Broer
- Department of Intensive Care Medicine, Hospital Winterthur, Brauerstrasse 15, Winterthur, 8401, Austria
| | - Steffen Halbgebauer
- Department of Neurology, University Hospital Ulm, Oberer Eselsberg 45, Ulm, 89081, Germany
| | - Hayrettin Tumani
- Department of Neurology, University Hospital Ulm, Oberer Eselsberg 45, Ulm, 89081, Germany
| | - Ann-Kathrin Friedrichs
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Helmholtzstraße 8/1, Ulm, 89081, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Helmholtzstraße 8/1, Ulm, 89081, Germany
| | - Lena Dörfer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Helmholtzstraße 8/1, Ulm, 89081, Germany
| |
Collapse
|
3
|
Pfnür A, Mayer B, Dörfer L, Tumani H, Spitzer D, Huber-Lang M, Kapapa T. Regulatory T Cell- and Natural Killer Cell-Mediated Inflammation, Cerebral Vasospasm, and Delayed Cerebral Ischemia in Aneurysmal Subarachnoid Hemorrhage-A Systematic Review and Meta-Analysis Approach. Int J Mol Sci 2025; 26:1276. [PMID: 39941044 PMCID: PMC11818301 DOI: 10.3390/ijms26031276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Aneurysmal subarachnoid hemorrhage (SAH) involves a significant influx of blood into the cerebrospinal fluid, representing a severe form of stroke. Despite advancements in aneurysm closure and neuro-intensive care, outcomes remain impaired due to cerebral vasospasm and delayed cerebral ischemia (DCI). Previous pharmacological therapies have not successfully reduced DCI while improving overall outcomes. As a result, significant efforts are underway to better understand the cellular and molecular mechanisms involved. This review focuses on the activation and effects of immune cells after SAH and their interactions with neurotoxic and vasoactive substances as well as inflammatory mediators. Particular attention is given to clinical studies highlighting the roles of natural killer (NK) cells and regulatory T cells (Treg) cells. Alongside microglia, astrocytes, and oligodendrocytes, NK cells and Treg cells are key contributors to the inflammatory cascade following SAH. Their involvement in modulating the neuro-inflammatory response, vasospasm, and DCI underscores their potential as therapeutic targets and prognostic markers in the post-SAH recovery process. We conducted a systematic review on T cell- and natural killer cell-mediated inflammation and their roles in cerebral vasospasm and delayed cerebral ischemia. We conducted a meta-analysis to evaluate outcomes and mortality in studies focused on NK cell- and T cell-mediated mechanisms.
Collapse
Affiliation(s)
- Andreas Pfnür
- Department of Neurosurgery, University Hospital Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, University of Ulm, Helmholtzstr. 22, 89081 Ulm, Germany
| | - Lena Dörfer
- Institute for Clinical and Experimental Trauma Immunology, University Hospital Ulm, Helmholtzstr. 8/, 89081 Ulm, Germany
| | - Hayrettin Tumani
- Department of Neurology, University Hospital Ulm, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Daniel Spitzer
- Department of Neurology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma Immunology, University Hospital Ulm, Helmholtzstr. 8/, 89081 Ulm, Germany
| | - Thomas Kapapa
- Department of Neurosurgery, University Hospital Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| |
Collapse
|
4
|
Ryu JY, Zhang J, Tirado SR, Dagen S, Frerichs KU, Patel NJ, Aziz-Sultan MA, Brown A, Rogers-Grazado M, Amr SS, Weiss ST, Du R. MiRNA expression profiling reveals a potential role of microRNA-148b-3p in cerebral vasospasm in subarachnoid hemorrhage. Sci Rep 2024; 14:22539. [PMID: 39341923 PMCID: PMC11438990 DOI: 10.1038/s41598-024-73579-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Cerebral vasospasm (CVS) is an important contributor to delayed cerebral ischemia following aneurysmal subarachnoid hemorrhage (aSAH), leading to high morbidity and long-term disability. While several microRNAs (miRNAs) have been implicated in vasospasm, the underlying mechanisms for CVS remain poorly understood. Our study aims to identify miRNAs that may contribute to the development of CVS. Whole-blood samples were obtained during or outside of vasospasm from aSAH patients whose maximal vasospasm was moderate or severe. MiRNAs were isolated from serial whole-blood samples, and miRNA sequencing was performed. Differentially expressed miRNAs were identified and the expression levels in patients' samples were verified using real-time qPCR. The biological functions of identified miRNA were evaluated in human brain endothelial cells (HBECs). MiRNA profiling revealed significant upregulation of miR-148b-3p in patients during CVS. We demonstrated that miR-148b-3p directly targeted and decreased the expression of ROCK1, affecting cell proliferation, migration, and invasion of HBECs through the ROCK-LIMK-Cofilin pathway. We propose that the upregulation of miRNA-148b-3p plays a role in the development of CVS by regulating actin cytoskeletal dynamics in HBECs, which is crucial for vascular function. Our study highlights miR-148b-3p as a potential diagnostic marker as well as therapeutic target for CVS following aSAH.
Collapse
Affiliation(s)
- Jee-Yeon Ryu
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Jianing Zhang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Selena-Rae Tirado
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Sarajune Dagen
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Kai U Frerichs
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Nirav J Patel
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - M Ali Aziz-Sultan
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Alison Brown
- Mass General Brigham Personalized Medicine, Mass General Brigham, Cambridge, MA, USA
| | | | - Sami S Amr
- Mass General Brigham Personalized Medicine, Mass General Brigham, Cambridge, MA, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rose Du
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|
5
|
Becker K. Animal Welfare Aspects in Planning and Conducting Experiments on Rodent Models of Subarachnoid Hemorrhage. Cell Mol Neurobiol 2023; 43:3965-3981. [PMID: 37861870 PMCID: PMC11407738 DOI: 10.1007/s10571-023-01418-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023]
Abstract
Subarachnoid hemorrhage is an acute life-threatening cerebrovascular disease with high socio-economic impact. The most frequent cause, the rupture of an intracerebral aneurysm, is accompanied by abrupt changes in intracerebral pressure, cerebral perfusion pressure and, consequently, cerebral blood flow. As aneurysms rupture spontaneously, monitoring of these parameters in patients is only possible with a time delay, upon hospitalization. To study alterations in cerebral perfusion immediately upon ictus, animal models are mandatory. This article addresses the points necessarily to be included in an animal project proposal according to EU directive 2010/63/EU for the protection of animals used for scientific purposes and herewith offers an insight into animal welfare aspects of using rodent models for the investigation of cerebral perfusion after subarachnoid hemorrhage. It compares surgeries, model characteristics, advantages, and drawbacks of the most-frequently used rodent models-the endovascular perforation model and the prechiasmatic and single or double cisterna magna injection model. The topics of discussing anesthesia, advice on peri- and postanesthetic handling of animals, assessing the severity of suffering the animals undergo during the procedure according to EU directive 2010/63/EU and weighing the use of these in vivo models for experimental research ethically are also presented. In conclusion, rodent models of subarachnoid hemorrhage display pathophysiological characteristics, including changes of cerebral perfusion similar to the clinical situation, rendering the models suited to study the sequelae of the bleeding. A current problem is low standardization of the models, wherefore reporting according to the ARRIVE guidelines is highly recommended. Animal welfare aspects of rodent models of subarachnoid hemorrhage. Rodent models for investigation of cerebral perfusion after subarachnoid hemorrhage are compared regarding surgeries and model characteristics, and 3R measures are suggested. Anesthesia is discussed, and advice given on peri- and postanesthetic handling. Severity of suffering according to 2010/63/EU is assessed and use of these in vivo models weighed ethically.
Collapse
Affiliation(s)
- Katrin Becker
- Institute for Translational Neurosurgery, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.
- Institute for Cardiovascular Sciences, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
6
|
Sanicola HW, Stewart CE, Luther P, Yabut K, Guthikonda B, Jordan JD, Alexander JS. Pathophysiology, Management, and Therapeutics in Subarachnoid Hemorrhage and Delayed Cerebral Ischemia: An Overview. PATHOPHYSIOLOGY 2023; 30:420-442. [PMID: 37755398 PMCID: PMC10536590 DOI: 10.3390/pathophysiology30030032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/21/2023] [Accepted: 09/07/2023] [Indexed: 09/28/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) is a type of hemorrhagic stroke resulting from the rupture of an arterial vessel within the brain. Unlike other stroke types, SAH affects both young adults (mid-40s) and the geriatric population. Patients with SAH often experience significant neurological deficits, leading to a substantial societal burden in terms of lost potential years of life. This review provides a comprehensive overview of SAH, examining its development across different stages (early, intermediate, and late) and highlighting the pathophysiological and pathohistological processes specific to each phase. The clinical management of SAH is also explored, focusing on tailored treatments and interventions to address the unique pathological changes that occur during each stage. Additionally, the paper reviews current treatment modalities and pharmacological interventions based on the evolving guidelines provided by the American Heart Association (AHA). Recent advances in our understanding of SAH will facilitate clinicians' improved management of SAH to reduce the incidence of delayed cerebral ischemia in patients.
Collapse
Affiliation(s)
- Henry W. Sanicola
- Department of Neurology, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA;
| | - Caleb E. Stewart
- Department of Neurosurgery, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA;
| | - Patrick Luther
- School of Medicine, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA; (P.L.); (K.Y.)
| | - Kevin Yabut
- School of Medicine, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA; (P.L.); (K.Y.)
| | - Bharat Guthikonda
- Department of Neurosurgery, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA;
| | - J. Dedrick Jordan
- Department of Neurology, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA;
| | - J. Steven Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA
| |
Collapse
|
7
|
Tartara F, Montalbetti A, Crobeddu E, Armocida D, Tavazzi E, Cardia A, Cenzato M, Boeris D, Garbossa D, Cofano F. Compartmental Cerebrospinal Fluid Events Occurring after Subarachnoid Hemorrhage: An "Heparin Oriented" Systematic Review. Int J Mol Sci 2023; 24:7832. [PMID: 37175544 PMCID: PMC10178276 DOI: 10.3390/ijms24097832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) represents a severe acute event with high morbidity and mortality due to the development of early brain injury (EBI), secondary delayed cerebral ischemia (DCI), and shunt-related hydrocephalus. Secondary events (SSE) such as neuroinflammation, vasospasm, excitotoxicity, blood-brain barrier disruption, oxidative cascade, and neuronal apoptosis are related to DCI. Despite improvement in management strategies and therapeutic protocols, surviving patients frequently present neurological deficits with neurocognitive impairment. The aim of this paper is to offer to clinicians a practical review of the actually documented pathophysiological events following subarachnoid hemorrhage. To reach our goal we performed a literature review analyzing reported studies regarding the mediators involved in the pathophysiological events following SAH occurring in the cerebrospinal fluid (CSF) (hemoglobin degradation products, platelets, complement, cytokines, chemokines, leucocytes, endothelin-1, NO-synthase, osteopontin, matricellular proteins, blood-brain barrier disruption, microglia polarization). The cascade of pathophysiological events secondary to SAH is very complex and involves several interconnected, but also distinct pathways. The identification of single therapeutical targets or specific pharmacological agents may be a limited strategy able to block only selective pathophysiological paths, but not the global evolution of SAH-related events. We report furthermore on the role of heparin in SAH management and discuss the rationale for use of intrathecal heparin as a pleiotropic therapeutical agent. The combination of the anticoagulant effect and the ability to interfere with SSE theoretically make heparin a very interesting molecule for SAH management.
Collapse
Affiliation(s)
- Fulvio Tartara
- IRCCS Fondazione Istituto Neurologico Nazionale C. Mondino, 27100 Pavia, Italy
| | - Andrea Montalbetti
- A.O.U. Maggiore della Carità University Hospital, Department of Neurosurgery, 28100 Novara, Italy
| | - Emanuela Crobeddu
- A.O.U. Maggiore della Carità University Hospital, Department of Neurosurgery, 28100 Novara, Italy
| | - Daniele Armocida
- A.U.O. Policlinico Umberto I, Neurosurgery Division, Human Neurosciences Department, Sapienza University, 00185 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Eleonora Tavazzi
- IRCCS Fondazione Istituto Neurologico Nazionale C. Mondino, 27100 Pavia, Italy
| | - Andrea Cardia
- Department of Neurosurgery, Neurocenter of Southern Switzerland, EOC, 6900 Lugano, Switzerland
| | - Marco Cenzato
- Ospedale Niguarda Ca’ Granda, Department of Neurosurgery, 20162 Milan, Italy
| | - Davide Boeris
- Ospedale Niguarda Ca’ Granda, Department of Neurosurgery, 20162 Milan, Italy
| | - Diego Garbossa
- Department of Neuroscience Rita Levi Montalcini, Neurosurgery Unit, University of Turin, 10095 Turin, Italy
| | - Fabio Cofano
- Department of Neuroscience Rita Levi Montalcini, Neurosurgery Unit, University of Turin, 10095 Turin, Italy
| |
Collapse
|
8
|
Weng W, Cheng F, Zhang J. Specific signature biomarkers highlight the potential mechanisms of circulating neutrophils in aneurysmal subarachnoid hemorrhage. Front Pharmacol 2022; 13:1022564. [PMID: 36438795 PMCID: PMC9685413 DOI: 10.3389/fphar.2022.1022564] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022] Open
Abstract
Background: Aneurysmal subarachnoid hemorrhage (aSAH) is a devastating hemorrhagic stroke with high disability and mortality. Neuroinflammation and the immunological response after aSAH are complex pathophysiological processes that have not yet been fully elucidated. Therefore, attention should be paid to exploring the inflammation-related genes involved in the systemic response to the rupture of intracranial aneurysms. Methods: The datasets of gene transcriptomes were downloaded from the Gene Expression Omnibus database. We constructed a gene co-expression network to identify cluster genes associated with aSAH and screened out differentially expressed genes (DEGs). The common gene was subsequently applied to identify hub genes by protein-protein interaction analysis and screen signature genes by machine learning algorithms. CMap analysis was implemented to identify potential small-molecule compounds. Meanwhile, Cibersort and ssGSEA were used to evaluate the immune cell composition, and GSEA reveals signal biological pathways. Results: We identified 602 DEGs from the GSE36791. The neutrophil-related module associated with aSAH was screened by weighted gene co-expression network analysis (WGCNA) and functional enrichment analysis. Several small molecular compounds were predicted based on neutrophil-related genes. MAPK14, ITGAM, TLR4, and FCGR1A have been identified as crucial genes involved in the peripheral immune activation related to neutrophils. Six significant genes (CST7, HSP90AB1, PADI4, PLBD1, RAB32, and SLAMF6) were identified as signature biomarkers by performing the LASSO analysis and SVM algorithms. The constructed machine learning model appears to be robust by receiver-operating characteristic curve analysis. The immune feature analysis demonstrated that neutrophils were upregulated post-aSAH and PADI4 was positively correlated with neutrophils. The NETs pathway was significantly upregulated in aSAH. Conclusion: We identified core regulatory genes influencing the transcription profiles of circulating neutrophils after the rupture of intracranial aneurysms using bioinformatics analysis and machine learning algorithms. This study provides new insight into the mechanism of peripheral immune response and inflammation after aSAH.
Collapse
|
9
|
Xu J, Zhang Z, Jin B, Geng Y, Lin L, Zhang S. Delayed filling of the superficial middle cerebral vein in acute large artery occlusion. Front Neurol 2022; 13:955804. [PMID: 36303557 PMCID: PMC9592715 DOI: 10.3389/fneur.2022.955804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 08/02/2022] [Indexed: 11/25/2022] Open
Abstract
Objective This study aimed to determine whether baseline delayed filling of the superficial middle cerebral vein (SMCV) was an independent cause of stroke prognosis in patients with acute anterior large vessel occlusion (LVO). Methods Consecutive patients with acute LVO [middle cerebral artery M1 ± intracranial internal carotid artery (ICA)] between March 2019 and May 2020 were included. Delayed SMCV was defined as delayed filling of SMCV in the affected side compared with the normal side throughout the venous phase on four-dimensional computed tomographic angiography (4D-CTA) reconstructed from CT perfusion imaging. The modified Rankin scale (mRS) was used to evaluate the prognosis of these patients 3 months after stroke. Results Of 54 patients in total, 47 (87.0%) patients presented with baseline delayed SMCV, and 36 (76.6%) patients achieved SMCV reversal (ipsilateral delayed SMCV reversed to bilateral symmetrical SMCV) after reperfusion therapy. Successful reperfusion was independently associated with SMCV reversal [odds ratio (OR) = 69.328, 95% confidence interval (CI) = 2.818–175.342]. A significant association between baseline SMCV delay and a 3-month poor outcome (OR = 19.623, 95% CI = 1.567–245.727, p = 0.021) was observed using a multivariable regression analysis. Compared with patients with persistent delayed SMCV, patients with reversed SMCV did not show a significant difference in the risk of a 3-month poor outcome (OR = 1.177, 95% CI = 0.147–9.448). Conclusions In patients with acute LVO, baseline delayed SMCV was an independent cause of poor stroke prognosis, and SMCV reversal cannot reverse the 3-month stroke prognosis. Therefore, the evaluation of baseline SMCV filling status should be strengthened in clinical practice.
Collapse
Affiliation(s)
- Jingsi Xu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zheyu Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Neurology, People's Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Bo Jin
- Department of Neurology, People's Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Yu Geng
- Department of Neurology, People's Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Longting Lin
- Department of Neurology, John Hunter Hospital, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Sheng Zhang
- Department of Neurology, People's Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, China
- *Correspondence: Sheng Zhang
| |
Collapse
|
10
|
Motwani K, Dodd WS, Laurent D, Lucke-Wold B, Chalouhi N. Delayed cerebral ischemia: A look at the role of endothelial dysfunction, emerging endovascular management, and glymphatic clearance. Clin Neurol Neurosurg 2022; 218:107273. [PMID: 35537284 DOI: 10.1016/j.clineuro.2022.107273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/08/2022] [Accepted: 04/27/2022] [Indexed: 12/12/2022]
Abstract
Delayed cerebral ischemia (DCI) contributes to extensive morbidity and mortality for patients with aneurysmal subarachnoid hemorrhage (SAH). Recent contributions to the basic and translational investigation of DCI have shed light on emerging concepts that may aid in the development of novel therapeutics. A clear association between cerebral vasospasm (CV) and DCI exists, but it is also known that DCI can affect brain parenchyma remote from sites of vasospasm. In this review, we highlight the most recent contributions to the understanding of the underlying pathophysiology of DCI including the emerging role of the glymphatic system. Furthermore, we discuss treatments for DCI, including both pharmacologic therapies and endovascular treatment of vasospasm. There continues to be a disconnect between interventions and targeted treatment against pathophysiology. This review is intended to serve as a catalyst for further research and discovery that can aid in improved treatment options for DCI.
Collapse
Affiliation(s)
- Kartik Motwani
- Department of Neurosurgery, University of Florida, Gainesville, USA
| | - William S Dodd
- Department of Neurosurgery, University of Florida, Gainesville, USA
| | - Dimitri Laurent
- Department of Neurosurgery, University of Florida, Gainesville, USA
| | | | - Nohra Chalouhi
- Department of Neurosurgery, University of Florida, Gainesville, USA
| |
Collapse
|
11
|
Monsour M, Croci DM, Agazzi S. Microclots in subarachnoid hemorrhage: an underestimated factor in delayed cerebral ischemia? Clin Neurol Neurosurg 2022; 219:107330. [PMID: 35700661 DOI: 10.1016/j.clineuro.2022.107330] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 12/01/2022]
Abstract
Subarachnoid hemorrhage has a poor prognosis due to the wide array of associated complications such as vasospasm, early brain injury, cortical spreading depression, oxidative stress, inflammation, and apoptosis. Each of these complications increases the risk of delayed cerebral ischemia (DCI), but recent research has suggested microclots play a substantial role in DCI incidence. This review will focus on the underlying inflammatory and coagulative mechanisms of microthrombosis while also outlining the current literature relating microclot burden to DCI. With a better understanding DCI pathophysiology as it relates to microthrombosis, more effective therapies can be developed in the future to improve clinical outcomes of SAH.
Collapse
Affiliation(s)
- Molly Monsour
- University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Davide Marco Croci
- Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| | - Siviero Agazzi
- Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
12
|
Minocycline Attenuates Microglia/Macrophage Phagocytic Activity and Inhibits SAH-Induced Neuronal Cell Death and Inflammation. Neurocrit Care 2022; 37:410-423. [PMID: 35585424 PMCID: PMC9519684 DOI: 10.1007/s12028-022-01511-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 04/05/2022] [Indexed: 01/28/2023]
Abstract
Background Neuroprotective treatment strategies aiming at interfering with either inflammation or cell death indicate the importance of these mechanisms in the development of brain injury after subarachnoid hemorrhage (SAH). This study was undertaken to evaluate the influence of minocycline on microglia/macrophage cell activity and its neuroprotective and anti-inflammatory impact 14 days after aneurismal SAH in mice. Methods Endovascular filament perforation was used to induce SAH in mice. SAH + vehicle-operated mice were used as controls for SAH vehicle-treated mice and SAH + minocycline-treated mice. The drug administration started 4 h after SAH induction and was daily repeated until day 7 post SAH and continued until day 14 every second day. Brain cryosections were immunolabeled for Iba1 to detect microglia/macrophages and NeuN to visualize neurons. Phagocytosis assay was performed to determine the microglia/macrophage activity status. Apoptotic cells were stained using terminal deoxyuridine triphosphate nick end labeling. Real-time quantitative polymerase chain reaction was used to estimate cytokine gene expression. Results We observed a significantly reduced phagocytic activity of microglia/macrophages accompanied by a lowered spatial interaction with neurons and reduced neuronal apoptosis achieved by minocycline administration after SAH. Moreover, the SAH-induced overexpression of pro-inflammatory cytokines and neuronal cell death was markedly attenuated by the compound. Conclusions Minocycline treatment may be implicated as a therapeutic approach with long-term benefits in the management of secondary brain injury after SAH in a clinically relevant time window. Supplementary Information The online version contains supplementary material available at 10.1007/s12028-022-01511-5.
Collapse
|
13
|
Dienel A, Kumar T P, Blackburn SL, McBride DW. Role of platelets in the pathogenesis of delayed injury after subarachnoid hemorrhage. J Cereb Blood Flow Metab 2021; 41:2820-2830. [PMID: 34112003 PMCID: PMC8756481 DOI: 10.1177/0271678x211020865] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) patients develop delayed cerebral ischemia and delayed deficits (DCI) within 2 weeks of aneurysm rupture at a rate of approximately 30%. DCI is a major contributor to morbidity and mortality after SAH. The cause of DCI is multi-factorial with contributions from microthrombi, blood vessel constriction, inflammation, and cortical spreading depolarizations. Platelets play central roles in hemostasis, inflammation, and vascular function. Within this review, we examine the potential roles of platelets in microthrombi formation, large artery vasospasm, microvessel constriction, inflammation, and cortical spreading depolarization. Evidence from experimental and clinical studies is provided to support the role(s) of platelets in each pathophysiology which contributes to DCI. The review concludes with a suggestion for future therapeutic targets to prevent DCI after aSAH.
Collapse
Affiliation(s)
- Ari Dienel
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Peeyush Kumar T
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Spiros L Blackburn
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - 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
| |
Collapse
|
14
|
Schenck H, Netti E, Teernstra O, De Ridder I, Dings J, Niemelä M, Temel Y, Hoogland G, Haeren R. The Role of the Glycocalyx in the Pathophysiology of Subarachnoid Hemorrhage-Induced Delayed Cerebral Ischemia. Front Cell Dev Biol 2021; 9:731641. [PMID: 34540844 PMCID: PMC8446455 DOI: 10.3389/fcell.2021.731641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 08/06/2021] [Indexed: 12/02/2022] Open
Abstract
The glycocalyx is an important constituent of blood vessels located between the bloodstream and the endothelium. It plays a pivotal role in intercellular interactions in neuroinflammation, reduction of vascular oxidative stress, and provides a barrier regulating vascular permeability. In the brain, the glycocalyx is closely related to functions of the blood-brain barrier and neurovascular unit, both responsible for adequate neurovascular responses to potential threats to cerebral homeostasis. An aneurysmal subarachnoid hemorrhage (aSAH) occurs following rupture of an intracranial aneurysm and leads to immediate brain damage (early brain injury). In some cases, this can result in secondary brain damage, also known as delayed cerebral ischemia (DCI). DCI is a life-threatening condition that affects up to 30% of all aSAH patients. As such, it is associated with substantial societal and healthcare-related costs. Causes of DCI are multifactorial and thought to involve neuroinflammation, oxidative stress, neuroinflammation, thrombosis, and neurovascular uncoupling. To date, prediction of DCI is limited, and preventive and effective treatment strategies of DCI are scarce. There is increasing evidence that the glycocalyx is disrupted following an aSAH, and that glycocalyx disruption could precipitate or aggravate DCI. This review explores the potential role of the glycocalyx in the pathophysiological mechanisms contributing to DCI following aSAH. Understanding the role of the glycocalyx in DCI could advance the development of improved methods to predict DCI or identify patients at risk for DCI. This knowledge may also alter the methods and timing of preventive and treatment strategies of DCI. To this end, we review the potential and limitations of methods currently used to evaluate the glycocalyx, and strategies to restore or prevent glycocalyx shedding.
Collapse
Affiliation(s)
- Hanna Schenck
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Eliisa Netti
- Department of Neurosurgery, Helsinki University Hospital, Helsinki, Finland
| | - Onno Teernstra
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Inger De Ridder
- Department of Neurology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, Netherlands
| | - Jim Dings
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Mika Niemelä
- Department of Neurosurgery, Helsinki University Hospital, Helsinki, Finland
| | - Yasin Temel
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Govert Hoogland
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Roel Haeren
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, Netherlands.,Department of Neurosurgery, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
15
|
Ikram A, Javaid MA, Ortega-Gutierrez S, Selim M, Kelangi S, Anwar SMH, Torbey MT, Divani AA. Delayed Cerebral Ischemia after Subarachnoid Hemorrhage. J Stroke Cerebrovasc Dis 2021; 30:106064. [PMID: 34464924 DOI: 10.1016/j.jstrokecerebrovasdis.2021.106064] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/25/2021] [Accepted: 08/15/2021] [Indexed: 12/23/2022] Open
Abstract
Delayed cerebral ischemia (DCI) is the most feared complication of aneurysmal subarachnoid hemorrhage (aSAH). It increases the mortality and morbidity associated with aSAH. Previously, large cerebral artery vasospasm was thought to be the sole major contributing factor associated with increased risk of DCI. Recent literature has challenged this concept. We conducted a literature search using PUBMED as the prime source of articles discussing various other factors which may contribute to the development of DCI both in the presence or absence of large cerebral artery vasospasm. These factors include microvascular spasm, micro-thrombosis, cerebrovascular dysregulation, and cortical spreading depolarization. These factors collectively result in inflammation of brain parenchyma, which is thought to precipitate early brain injury and DCI. We conclude that diagnostic modalities need to be refined in order to diagnose DCI more efficiently in its early phase, and newer interventions need to be developed to prevent and treat this condition. These newer interventions are currently being studied in experimental models. However, their effectiveness on patients with aSAH is yet to be determined.
Collapse
Affiliation(s)
- Asad Ikram
- Department of Neurology, University of New Mexico, MSC10-5620, 1, Albuquerque, NM 87131, USA
| | - Muhammad Ali Javaid
- Department of Neurology, University of New Mexico, MSC10-5620, 1, Albuquerque, NM 87131, USA
| | | | - Magdy Selim
- Stroke Division, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sarah Kelangi
- Department of Neurology, University of New Mexico, MSC10-5620, 1, Albuquerque, NM 87131, USA
| | | | - Michel T Torbey
- Department of Neurology, University of New Mexico, MSC10-5620, 1, Albuquerque, NM 87131, USA
| | - Afshin A Divani
- Department of Neurology, University of New Mexico, MSC10-5620, 1, Albuquerque, NM 87131, USA.
| |
Collapse
|
16
|
Schwarting J, Nehrkorn K, Liu H, Plesnila N, Terpolilli NA. Role of Pial Microvasospasms and Leukocyte Plugging for Parenchymal Perfusion after Subarachnoid Hemorrhage Assessed by In Vivo Multi-Photon Microscopy. Int J Mol Sci 2021; 22:8444. [PMID: 34445151 PMCID: PMC8395146 DOI: 10.3390/ijms22168444] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 12/05/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is associated with acute and delayed cerebral ischemia. We suggested spasms of pial arterioles as a possible mechanism; however, it remained unclear whether and how pial microvasospasms (MVSs) induce cerebral ischemia. Therefore, we used in vivo deep tissue imaging by two-photon microscopy to investigate MVSs together with the intraparenchymal microcirculation in a clinically relevant murine SAH model. Male C57BL/6 mice received a cranial window. Cerebral vessels and leukocytes were labelled with fluorescent dyes and imaged by in vivo two-photon microscopy before and three hours after SAH induced by filament perforation. After SAH, a large clot formed around the perforation site at the skull base, and blood distributed along the perivascular space of the middle cerebral artery up to the cerebral cortex. Comparing the cerebral microvasculature before and after SAH, we identified three different patterns of constrictions: pearl string, global, and bottleneck. At the same time, the volume of perfused intraparenchymal vessels and blood flow velocity in individual arterioles were significantly reduced by more than 60%. Plugging of capillaries by leukocytes was observed but infrequent. The current study demonstrates that perivascular blood is associated with spasms of pial arterioles and that these spasms result in a significant reduction in cortical perfusion after SAH. Thus, the pial microvasospasm seems to be an important mechanism by which blood in the subarachnoid space triggers cerebral ischemia after SAH. Identifying the mechanisms of pial vasospasm may therefore result in novel therapeutic options for SAH patients.
Collapse
Affiliation(s)
- Julian Schwarting
- Institute for Stroke and Dementia Research, Munich University Hospital, Graduate School of Systemic Neurosciences, Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians-University, 81377 Munich, Germany; (J.S.); (K.N.); (H.L.); (N.A.T.)
- Department of Neurosurgery, Munich University Hospital, 81377 Munich, Germany
| | - Kathrin Nehrkorn
- Institute for Stroke and Dementia Research, Munich University Hospital, Graduate School of Systemic Neurosciences, Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians-University, 81377 Munich, Germany; (J.S.); (K.N.); (H.L.); (N.A.T.)
| | - Hanhan Liu
- Institute for Stroke and Dementia Research, Munich University Hospital, Graduate School of Systemic Neurosciences, Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians-University, 81377 Munich, Germany; (J.S.); (K.N.); (H.L.); (N.A.T.)
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, Munich University Hospital, Graduate School of Systemic Neurosciences, Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians-University, 81377 Munich, Germany; (J.S.); (K.N.); (H.L.); (N.A.T.)
| | - Nicole Angela Terpolilli
- Institute for Stroke and Dementia Research, Munich University Hospital, Graduate School of Systemic Neurosciences, Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians-University, 81377 Munich, Germany; (J.S.); (K.N.); (H.L.); (N.A.T.)
- Department of Neurosurgery, Munich University Hospital, 81377 Munich, Germany
| |
Collapse
|
17
|
Dodd WS, Laurent D, Dumont AS, Hasan DM, Jabbour PM, Starke RM, Hosaka K, Polifka AJ, Hoh BL, Chalouhi N. Pathophysiology of Delayed Cerebral Ischemia After Subarachnoid Hemorrhage: A Review. J Am Heart Assoc 2021; 10:e021845. [PMID: 34325514 PMCID: PMC8475656 DOI: 10.1161/jaha.121.021845] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/09/2021] [Indexed: 01/23/2023]
Abstract
Delayed cerebral ischemia is a major predictor of poor outcomes in patients who suffer subarachnoid hemorrhage. Treatment options are limited and often ineffective despite many years of investigation and clinical trials. Modern advances in basic science have produced a much more complex, multifactorial framework in which delayed cerebral ischemia is better understood and novel treatments can be developed. Leveraging this knowledge to improve outcomes, however, depends on a holistic understanding of the disease process. We conducted a review of the literature to analyze the current state of investigation into delayed cerebral ischemia with emphasis on the major themes that have emerged over the past decades. Specifically, we discuss microcirculatory dysfunction, glymphatic impairment, inflammation, and neuroelectric disruption as pathological factors in addition to the canonical focus on cerebral vasospasm. This review intends to give clinicians and researchers a summary of the foundations of delayed cerebral ischemia pathophysiology while also underscoring the interactions and interdependencies between pathological factors. Through this overview, we also highlight the advances in translational studies and potential future therapeutic opportunities.
Collapse
Affiliation(s)
- William S. Dodd
- Department of NeurosurgeryCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Dimitri Laurent
- Department of NeurosurgeryCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Aaron S. Dumont
- Department of Neurological SurgerySchool of MedicineTulane UniversityNew OrleansLA
| | - David M. Hasan
- Department of NeurosurgeryCarver College of MedicineUniversity of IowaIowa CityIA
| | - Pascal M. Jabbour
- Department of Neurological SurgerySidney Kimmel Medical CollegeThomas Jefferson UniversityPhiladelphiaPA
| | - Robert M. Starke
- Department of Neurological SurgeryMiller School of MedicineUniversity of MiamiFL
| | - Koji Hosaka
- Department of NeurosurgeryCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Adam J. Polifka
- Department of NeurosurgeryCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Brian L. Hoh
- Department of NeurosurgeryCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Nohra Chalouhi
- Department of NeurosurgeryCollege of MedicineUniversity of FloridaGainesvilleFL
| |
Collapse
|
18
|
Lin J, Cheng Z, Shi Y, Cai X, Huang L. Evaluating the Velocity and Extent of Cortical Venous Filling in Patients With Severe Middle Cerebral Artery Stenosis or Occlusion. Front Neurol 2021; 12:610658. [PMID: 33897584 PMCID: PMC8060485 DOI: 10.3389/fneur.2021.610658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 03/16/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: To investigate the velocity and extent of cortical venous filling (CVF) and its association with clinical manifestations in patients with severe stenosis or occlusion of the middle cerebral artery (MCA) using dynamic computed tomography angiography (CTA). Methods: Fifty-eight patients (36 symptomatic and 22 asymptomatic) with severe unilateral stenosis (≥70%) or occlusion of the MCA M1 segment who underwent dynamic CTA were included. Collateral status, antegrade flow, and CVF of each patient were observed using dynamic CTA. Three types of cortical veins were selected to observe the extent of CVF, and the absence of CVF (CVF-) was recorded. Based on the appearance of CVF in the superior sagittal sinus, instances of CVF, including early (CVF1), peak (CVF2), and late (CVF3) venous phases, were recorded. The differences in CVF times between the affected and contralateral hemispheres were represented as rCVFs, and CVF velocity was defined compared to the median time of each rCVF. Results: All CVF times in the affected hemisphere were longer than those in the contralateral hemisphere (p < 0.05). Patients with symptomatic MCA stenosis had more ipsilateral CVF- (p = 0.02) and more delayed CVF at rCVF2 and rCVF21 (rCVF2-rCVF1) (p = 0.03 and 0.001, respectively) compared to those with asymptomatic MCA stenosis. For symptomatic patients, fast CVF at rCVF21 was associated with poor collateral status (odds ratio [OR] 6.42, 95% confidence interval [CI] 1.37-30.05, p = 0.02), and ipsilateral CVF- in two cortical veins was associated with poor 3-month outcomes (adjusted OR 0.025, 95% CI 0.002-0.33, p = 0.005). Conclusions: Complete and fast CVF is essential for patients with symptomatic MCA stenosis or occlusion. The clinical value of additional CVF assessment should be explored in future studies to identify patients with severe MCA stenosis or occlusion at a higher risk of stroke occurrence and poor recovery.
Collapse
Affiliation(s)
- Jia'Xing Lin
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Zhong'Yuan Cheng
- Medical Imaging Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ying'Ying Shi
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xiang'Ran Cai
- Medical Imaging Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Li'An Huang
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
19
|
Di Bella D, Ferreira JPS, Silva RDNO, Echem C, Milan A, Akamine EH, Carvalho MH, Rodrigues SF. Gold nanoparticles reduce inflammation in cerebral microvessels of mice with sepsis. J Nanobiotechnology 2021; 19:52. [PMID: 33608025 PMCID: PMC7893894 DOI: 10.1186/s12951-021-00796-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
Background Sepsis is an emergency medical condition that can lead to death and it is defined as a life-threatening organ dysfunction caused by immune dysregulation in response to an infection. It is considered the main killer in intensive care units. Sepsis associated-encephalopathy (SAE) is mostly caused by a sepsis-induced systemic inflammatory response. Studies report SAE in 14–63% of septic patients. Main SAE symptoms are not specific and usually include acute impairment of consciousness, delirium and/or coma, along with electroencephalogram (EEG) changes. For those who recover from sepsis and SAE, impaired cognitive function, mobility and quality of life are often observed months to years after hospital discharge, and there is no treatment available today to prevent that. Inflammation and oxidative stress are key players for the SAE pathophysiology. Gold nanoparticles have been demonstrated to own important anti-inflammatory properties. It was also reported 20 nm citrate-covered gold nanoparticles (cit-AuNP) reduce oxidative stress. In this context, we tested whether 20 nm cit-AuNP could alleviate the acute changes caused by sepsis in brain of mice, with focus on inflammation. Sepsis was induced in female C57BL/6 mice by cecal ligation and puncture (CLP), 20 nm cit-AuNP or saline were intravenously (IV) injected 2 h after induction of sepsis and experiments performed 6 h after induction. Intravital microscopy was used for leukocyte and platelet adhesion study in brain, blood brain barrier (BBB) permeability carried out by Evans blue assay, cytokines measured by ELISA and real time PCR, cell adhesion molecules (CAMs) by flow cytometry and immunohistochemistry, and transcription factors, by western blotting. Results 20 nm cit-AuNP treatment reduced leukocyte and platelet adhesion to cerebral blood vessels, prevented BBB failure, reduced TNF- concentration in brain, and ICAM-1 expression both in circulating polymorphonuclear (PMN) leukocytes and cerebral blood vessels of mice with sepsis. Furthermore, 20 nm cit-AuNP did not interfere with the antibiotic effect on the survival rate of mice with sepsis. Conclusions Cit-AuNP showed important anti-inflammatory properties in the brain of mice with sepsis, being a potential candidate to be used as adjuvant drug along with antibiotics in the treatment of sepsis to avoid SAE ![]()
Collapse
Affiliation(s)
- Davide Di Bella
- Laboratory of Hypertension, Diabetes and Vascular Biology, Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1524, ICB I, sala 205, 2º andar, Butanta, 05508-900, Sao Paulo, Brazil
| | - João P S Ferreira
- Laboratory of Hypertension, Diabetes and Vascular Biology, Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1524, ICB I, sala 205, 2º andar, Butanta, 05508-900, Sao Paulo, Brazil
| | - Renee de Nazare O Silva
- Laboratory of Hypertension, Diabetes and Vascular Biology, Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1524, ICB I, sala 205, 2º andar, Butanta, 05508-900, Sao Paulo, Brazil
| | - Cinthya Echem
- Laboratory of Hypertension, Diabetes and Vascular Biology, Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1524, ICB I, sala 205, 2º andar, Butanta, 05508-900, Sao Paulo, Brazil
| | - Aline Milan
- Laboratory of Hypertension, Diabetes and Vascular Biology, Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1524, ICB I, sala 205, 2º andar, Butanta, 05508-900, Sao Paulo, Brazil
| | - Eliana H Akamine
- Laboratory of Hypertension, Diabetes and Vascular Biology, Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1524, ICB I, sala 205, 2º andar, Butanta, 05508-900, Sao Paulo, Brazil
| | - Maria H Carvalho
- Laboratory of Hypertension, Diabetes and Vascular Biology, Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1524, ICB I, sala 205, 2º andar, Butanta, 05508-900, Sao Paulo, Brazil
| | - Stephen F Rodrigues
- Laboratory of Hypertension, Diabetes and Vascular Biology, Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1524, ICB I, sala 205, 2º andar, Butanta, 05508-900, Sao Paulo, Brazil. .,Laboratory of Vascular Nanopharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1524, ICB I, sala 319, 3º andar, Butanta, 05508-900, Sao Paulo, Brazil.
| |
Collapse
|
20
|
Wang Z, Chen J, Toyota Y, Keep RF, Xi G, Hua Y. Ultra-Early Cerebral Thrombosis Formation After Experimental Subarachnoid Hemorrhage Detected on T2* Magnetic Resonance Imaging. Stroke 2021; 52:1033-1042. [PMID: 33535782 DOI: 10.1161/strokeaha.120.032397] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND AND PURPOSE The mechanisms of brain damage during ultra-early subarachnoid hemorrhage (SAH) have not been well studied. The current study examined the SAH-induced hyperacute brain damage at 4 hours using magnetic resonance imaging and brain histology in a mouse model. METHODS SAH was induced by endovascular perforation in adult mice. First, adult male wild-type mice underwent magnetic resonance imaging T2 and T2* 4 hours after an endovascular perforation or a sham operation and were euthanized to assess brain histology. Second, male and female adult lipocalin-2 knockout mice had SAH. All animals underwent magnetic resonance imaging at 4 hours, and the brains were harvested for brain histology. RESULTS T2* hypointensity vessels were observed in the brain 4 hours after SAH in male wild-type mice. The numbers of T2*-positive vessels were significantly higher in SAH brains than in sham-operated mice. Brain histology showed thrombosis and erythrocyte plugs in the T2*-positive cerebral vessels which may be venules. The number of T2*-positive vessels correlated with SAH grade and the presence of T2 lesions. Brain thrombosis was also accompanied by albumin leakage and neuronal injury. LCN2 deficient male mice had lower numbers of T2*-positive vessels after SAH compared with wild-type male mice. CONCLUSIONS SAH causes ultra-early brain vessel thrombosis that can be detected by T2* gradient-echo sequence at 4 hours after SAH. LCN2 deficiency decreased the number of T2*-positive vessels.
Collapse
Affiliation(s)
- Zhepei Wang
- Department of Neurosurgery, University of Michigan, Ann Arbor (Z.W., J.C., Y.T., R.F.K., G.X., Y.H.).,Department of Neurosurgery, The First Hospital of Ningbo, Zhejiang, China (Z.W.)
| | - Jingyin Chen
- Department of Neurosurgery, University of Michigan, Ann Arbor (Z.W., J.C., Y.T., R.F.K., G.X., Y.H.)
| | - Yasunori Toyota
- Department of Neurosurgery, University of Michigan, Ann Arbor (Z.W., J.C., Y.T., R.F.K., G.X., Y.H.)
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor (Z.W., J.C., Y.T., R.F.K., G.X., Y.H.)
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor (Z.W., J.C., Y.T., R.F.K., G.X., Y.H.)
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor (Z.W., J.C., Y.T., R.F.K., G.X., Y.H.)
| |
Collapse
|
21
|
Suzuki H, Kanamaru H, Kawakita F, Asada R, Fujimoto M, Shiba M. Cerebrovascular pathophysiology of delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. Histol Histopathol 2020; 36:143-158. [PMID: 32996580 DOI: 10.14670/hh-18-253] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Aneurysmal subarachnoid hemorrhage (SAH) remains a serious cerebrovascular disease. Even if SAH patients survive the initial insults, delayed cerebral ischemia (DCI) may occur at 4 days or later post-SAH. DCI is characteristics of SAH, and is considered to develop by blood breakdown products and inflammatory reactions, or secondary to early brain injury, acute pathophysiological events that occur in the brain within the first 72 hours of aneurysmal SAH. The pathology underlying DCI may involve large artery vasospasm and/or microcirculatory disturbances by microvasospasm, microthrombosis, dysfunction of venous outflow and compression of microvasculature by vasogenic or cytotoxic tissue edema. Recent clinical evidence has shown that large artery vasospasm is not the only cause of DCI, and that both large artery vasospasm-dependent and -independent cerebral infarction causes poor outcome. Animal studies suggest that mechanisms of vasospasm may differ between large artery and arterioles or capillaries, and that many kinds of cells in the vascular wall and brain parenchyma may be involved in the pathogenesis of microcirculatory disturbances. The impairment of the paravascular and glymphatic systems also may play important roles in the development of DCI. As pathological mediators for DCI, glutamate and several matricellular proteins have been investigated in addition to inflammatory molecules. Glutamate is involved in excitotoxicity contributing to cortical spreading ischemia and epileptic activity-related events. Microvascular dysfunction is an attractive mechanism to explain the cause of poor outcomes independently of large cerebral artery vasospasm, but needs more studies to clarify the pathophysiologies or mechanisms and to develop a novel therapeutic strategy.
Collapse
Affiliation(s)
- Hidenori Suzuki
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan.
| | - Hideki Kanamaru
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Fumihiro Kawakita
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Reona Asada
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Masashi Fujimoto
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Masato Shiba
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
22
|
Clarke JV, Suggs JM, Diwan D, Lee JV, Lipsey K, Vellimana AK, Zipfel GJ. Microvascular platelet aggregation and thrombosis after subarachnoid hemorrhage: A review and synthesis. J Cereb Blood Flow Metab 2020; 40:1565-1575. [PMID: 32345104 PMCID: PMC7370365 DOI: 10.1177/0271678x20921974] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Delayed cerebral ischemia (DCI) after aneurysmal subarachnoid hemorrhage (SAH) has been associated with numerous pathophysiological sequelae, including large artery vasospasm and microvascular thrombosis. The focus of this review is to provide an overview of experimental animal model studies and human autopsy studies that explore the temporal-spatial characterization and mechanism of microvascular platelet aggregation and thrombosis following SAH, as well as to critically assess experimental studies and clinical trials highlighting preventative therapeutic options against this highly morbid pathophysiological process. Upon review of the literature, we discovered that microvascular platelet aggregation and thrombosis occur after experimental SAH across multiple species and SAH induction techniques in a similar time frame to other components of DCI, occurring in the cerebral cortex and hippocampus across both hemispheres. We discuss the relationship of these findings to human autopsy studies. In the final section of this review, we highlight the important therapeutic options for targeting microvascular platelet aggregation and thrombosis, and emphasize why therapeutic targeting of this neurovascular pathology may improve patient care. We encourage ongoing research into the pathophysiology of SAH and DCI, especially in regard to microvascular platelet aggregation and thrombosis and the translation to randomized clinical trials.
Collapse
Affiliation(s)
- Julian V Clarke
- Washington University School of Medicine, Saint Louis, MO, USA
| | - Julia M Suggs
- Washington University School of Medicine, Saint Louis, MO, USA
| | - Deepti Diwan
- Washington University School of Medicine, Saint Louis, MO, USA
| | - Jin V Lee
- Washington University School of Medicine, Saint Louis, MO, USA
| | - Kim Lipsey
- Washington University School of Medicine, Saint Louis, MO, USA
| | - Ananth K Vellimana
- Neurological Surgery, Washington University School of Medicine, , Saint Louis, MO, USA
| | - Gregory J Zipfel
- Neurological Surgery, Washington University School of Medicine, , Saint Louis, MO, USA
| |
Collapse
|
23
|
Balbi M, Vega MJ, Lourbopoulos A, Terpolilli NA, Plesnila N. Long-term impairment of neurovascular coupling following experimental subarachnoid hemorrhage. J Cereb Blood Flow Metab 2020; 40:1193-1202. [PMID: 31296132 PMCID: PMC7238370 DOI: 10.1177/0271678x19863021] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
CO2-reactivity and neurovascular coupling are sequentially lost within the first 24 h after subarachnoid hemorrhage (SAH). Whether and when these impairments recover is not known. Therefore, we investigated the reactivity of pial and intraparenchymal vessels by in vivo two-photon microscopy one month after experimental SAH. C57BL/6 mice were subjected to either sham surgery or SAH by filament perforation. One month later, cerebral blood flow following CO2-challenge and forepaw stimulation was assessed by laser Doppler fluxmetry. Diameters of pial and intraparenchymal arterioles were quantified by in vivo two-photon microscopy. One month after SAH, pial and parenchymal vessels dilated in response to CO2. Neurovascular coupling was almost completely absent after SAH: vessel diameter did not change upon forepaw stimulation compared to a 20% increase in sham-operated mice. The current results demonstrate that neurovascular function differentially recovers after SAH: while CO2-reactivity normalizes within one month after SAH, neurovascular coupling is still absent. These findings show an acute and persistent loss of neurovascular coupling after SAH that may serve as a link between early brain injury and delayed cerebral ischemia, two distinct pathophysiological phenomena after SAH that were so far believed not to be directly related.
Collapse
Affiliation(s)
- Matilde Balbi
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Munich, Germany.,Graduate School of Systemic Neurosciences (GSN), Munich University Hospital, Munich, Germany.,Munich Cluster of Systems Neurology (Synergy), Munich, Germany
| | - Max Jativa Vega
- Graduate School of Systemic Neurosciences (GSN), Munich University Hospital, Munich, Germany
| | - Athanasios Lourbopoulos
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Munich, Germany.,Munich Cluster of Systems Neurology (Synergy), Munich, Germany
| | - Nicole A Terpolilli
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Munich, Germany.,Munich Cluster of Systems Neurology (Synergy), Munich, Germany.,Department of Neurosurgery, Munich University Hospital, Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Munich, Germany.,Graduate School of Systemic Neurosciences (GSN), Munich University Hospital, Munich, Germany.,Munich Cluster of Systems Neurology (Synergy), Munich, Germany
| |
Collapse
|
24
|
Increased Mean Platelet Volume is Associated with Poor Outcome in Patients with Aneurysmal Subarachnoid Hemorrhage. World Neurosurg 2020; 137:e118-e125. [DOI: 10.1016/j.wneu.2020.01.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/09/2020] [Accepted: 01/09/2020] [Indexed: 12/26/2022]
|
25
|
James RF, Khattar NK, Aljuboori ZS, Page PS, Shao EY, Carter LM, Meyer KS, Daniels MW, Craycroft J, Gaughen JR, Chaudry MI, Rai SN, Everhart DE, Simard JM. Continuous infusion of low-dose unfractionated heparin after aneurysmal subarachnoid hemorrhage: a preliminary study of cognitive outcomes. J Neurosurg 2019; 130:1460-1467. [PMID: 29749915 DOI: 10.3171/2017.11.jns17894] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 11/29/2017] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Cognitive dysfunction occurs in up to 70% of aneurysmal subarachnoid hemorrhage (aSAH) survivors. Low-dose intravenous heparin (LDIVH) infusion using the Maryland protocol was recently shown to reduce clinical vasospasm and vasospasm-related infarction. In this study, the Montreal Cognitive Assessment (MoCA) was used to evaluate cognitive changes in aSAH patients treated with the Maryland LDIVH protocol compared with controls. METHODS A retrospective analysis of all patients treated for aSAH between July 2009 and April 2014 was conducted. Beginning in 2012, aSAH patients were treated with LDIVH in the postprocedural period. The MoCA was administered to all aSAH survivors prospectively during routine follow-up visits, at least 3 months after aSAH, by trained staff blinded to treatment status. Mean MoCA scores were compared between groups, and regression analyses were performed for relevant factors. RESULTS No significant differences in baseline characteristics were observed between groups. The mean MoCA score for the LDIVH group (n = 25) was 26.4 compared with 22.7 in controls (n = 22) (p = 0.013). Serious cognitive impairment (MoCA ≤ 20) was observed in 32% of controls compared with 0% in the LDIVH group (p = 0.008). Linear regression analysis demonstrated that only LDIVH was associated with a positive influence on MoCA scores (β = 3.68, p =0.019), whereas anterior communicating artery aneurysms and fevers were negatively associated with MoCA scores. Multivariable linear regression analysis resulted in all 3 factors maintaining significance. There were no treatment complications. CONCLUSIONS This preliminary study suggests that the Maryland LDIVH protocol may improve cognitive outcomes in aSAH patients. A randomized controlled trial is needed to determine the safety and potential benefit of unfractionated heparin in aSAH patients.
Collapse
Affiliation(s)
- Robert F James
- 1Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
- 2Department of Surgery, Brody School of Medicine at East Carolina University, Greenville, North Carolina
| | - Nicolas K Khattar
- 1Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Zaid S Aljuboori
- 1Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Paul S Page
- 1Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Elaine Y Shao
- 2Department of Surgery, Brody School of Medicine at East Carolina University, Greenville, North Carolina
| | - Lacey M Carter
- 2Department of Surgery, Brody School of Medicine at East Carolina University, Greenville, North Carolina
| | - Kimberly S Meyer
- 1Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Michael W Daniels
- 3Department of Bioinformatics and Biostatistics, University of Louisville School of Public Health, Louisville, Kentucky
| | - John Craycroft
- 3Department of Bioinformatics and Biostatistics, University of Louisville School of Public Health, Louisville, Kentucky
| | - John R Gaughen
- 4Sentara Martha Jefferson Hospital, Charlottesville, Virginia
| | - M Imran Chaudry
- 5Comprehensive Stroke and Cerebrovascular Center, Medical University of South Carolina, Charleston, South Carolina
| | - Shesh N Rai
- 3Department of Bioinformatics and Biostatistics, University of Louisville School of Public Health, Louisville, Kentucky
| | - D Erik Everhart
- Departments of6Psychology and
- 7Internal Medicine, East Carolina University, Greenville, North Carolina; and
| | - J Marc Simard
- Departments of8Neurosurgery
- 9Pathology, and
- 10Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
26
|
Yang L, Lai WT, Wu YS, Zhang JA, Zhou XH, Yan J, Fang C, Zeng EM, Tang B, Peng CL, Zhao Y, Hong T. Simple and efficient rat model for studying delayed cerebral ischemia after subarachnoid hemorrhage. J Neurosci Methods 2018; 304:146-153. [DOI: 10.1016/j.jneumeth.2018.04.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 12/22/2022]
|
27
|
Possible Role of Inflammation and Galectin-3 in Brain Injury after Subarachnoid Hemorrhage. Brain Sci 2018; 8:brainsci8020030. [PMID: 29414883 PMCID: PMC5836049 DOI: 10.3390/brainsci8020030] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 02/05/2018] [Accepted: 02/05/2018] [Indexed: 12/14/2022] Open
Abstract
Aneurysmal subarachnoid hemorrhage (SAH) is known as one of the most devastating diseases in the central nervous system. In the past few decades, research on SAH has focused on cerebral vasospasm to prevent post-SAH delayed cerebral ischemia (DCI) and to improve outcomes. However, increasing evidence has suggested that early brain injury (EBI) is an important mechanism contributing to DCI, cerebral vasospasm as well as poor outcomes. Though the mechanism of EBI is very complex, inflammation is thought to play a pivotal role in EBI. Galectin-3 is a unique chimera type in the galectin family characterized by its β-galactoside-binding lectin, which mediates various pathologies, such as fibrosis, cell adhesion, and inflammation. Recently, two clinical studies revealed galectin-3 to be a possible prognostic biomarker in SAH patients. In addition, our recent report suggested that higher acute-stage plasma galectin-3 levels correlated with subsequent development of delayed cerebral infarction that was not associated with vasospasm in SAH patients. We review the possible role and molecular mechanisms of inflammation as well as galectin-3 in brain injuries, especially focusing on EBI after SAH, and discuss galectin-3 as a potential new therapeutic or research target in post-SAH brain injuries.
Collapse
|
28
|
Yang XM, Chen XH, Lu JF, Zhou CM, Han JY, Chen CH. In vivo observation of cerebral microcirculation after experimental subarachnoid hemorrhage in mice. Neural Regen Res 2018; 13:456-462. [PMID: 29623930 PMCID: PMC5900508 DOI: 10.4103/1673-5374.228728] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Acute brain injury caused by subarachnoid hemorrhage is the major cause of poor prognosis. The pathology of subarachnoid hemorrhage likely involves major morphological changes in the microcirculation. However, previous studies primarily used fixed tissue or delayed injury models. Therefore, in the present study, we used in vivo imaging to observe the dynamic changes in cerebral microcirculation after subarachnoid hemorrhage. Subarachnoid hemorrhage was induced by perforation of the bifurcation of the middle cerebral and anterior cerebral arteries in male C57/BL6 mice. The diameter of pial arterioles and venules was measured by in vivo fluorescence microscopy at different time points within 180 minutes after subarachnoid hemorrhage. Cerebral blood flow was examined and leukocyte adhesion/albumin extravasation was determined at different time points before and after subarachnoid hemorrhage. Cerebral pial microcirculation was abnormal and cerebral blood flow was reduced after subarachnoid hemorrhage. Acute vasoconstriction occurred predominantly in the arterioles instead of the venules. A progressive increase in the number of adherent leukocytes in venules and substantial albumin extravasation were observed between 10 and 180 minutes after subarachnoid hemorrhage. These results show that major changes in microcirculation occur in the early stage of subarachnoid hemorrhage. Our findings may promote the development of novel therapeutic strategies for the early treatment of subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Xiao-Mei Yang
- Department of Human Anatomy and Embryology, Peking University Health Science Center, Beijing, China
| | - Xu-Hao Chen
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jian-Fei Lu
- Department of Human Anatomy and Embryology, Peking University Health Science Center, Beijing, China
| | - Chang-Man Zhou
- Department of Human Anatomy and Embryology, Peking University Health Science Center, Beijing, China
| | - Jing-Yan Han
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
| | - Chun-Hua Chen
- Department of Human Anatomy and Embryology, Peking University Health Science Center, Beijing, China
| |
Collapse
|
29
|
Ray B, Pandav VM, Mathews EA, Thompson DM, Ford L, Yearout LK, Bohnstedt BN, Chaudhary S, Dale GL, Prodan CI. Coated-Platelet Trends Predict Short-Term Clinical OutcomeAfter Subarachnoid Hemorrhage. Transl Stroke Res 2017; 9:459-470. [DOI: 10.1007/s12975-017-0594-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 12/21/2022]
|
30
|
Balbi M, Koide M, Wellman GC, Plesnila N. Inversion of neurovascular coupling after subarachnoid hemorrhage in vivo. J Cereb Blood Flow Metab 2017; 37:3625-3634. [PMID: 28112024 PMCID: PMC5669344 DOI: 10.1177/0271678x16686595] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Subarachnoid hemorrhage (SAH) induces acute changes in the cerebral microcirculation. Recent findings ex vivo suggest neurovascular coupling (NVC), the process that increases cerebral blood flow upon neuronal activity, is also impaired after SAH. The aim of the current study was to investigate whether this occurs also in vivo. C57BL/6 mice were subjected to either sham surgery or SAH by filament perforation. Twenty-four hours later NVC was tested by forepaw stimulation and CO2 reactivity by inhalation of 10% CO2. Vessel diameter was assessed in vivo by two-photon microscopy. NVC was also investigated ex vivo using brain slices. Cerebral arterioles of sham-operated mice dilated to 130% of baseline upon CO2 inhalation or forepaw stimulation and cerebral blood flow (CBF) increased. Following SAH, however, CO2 reactivity was completely lost and the majority of cerebral arterioles showed paradoxical constriction in vivo and ex vivo resulting in a reduced CBF response. As previous results showed intact NVC 3 h after SAH, the current findings indicate that impairment of NVC after cerebral hemorrhage occurs secondarily and is progressive. Since neuronal activity-induced vasoconstriction (inverse NVC) is likely to further aggravate SAH-induced cerebral ischemia and subsequent brain damage, inverse NVC may represent a novel therapeutic target after SAH.
Collapse
Affiliation(s)
- Matilde Balbi
- 1 Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Ludwig-Maximilians University (LMU), Munich, Germany.,2 Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilians University (LMU), Munich, Germany
| | - Masayo Koide
- 3 Department of Pharmacology, University of Vermont, Burlington, VT, USA
| | - George C Wellman
- 3 Department of Pharmacology, University of Vermont, Burlington, VT, USA
| | - Nikolaus Plesnila
- 1 Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Ludwig-Maximilians University (LMU), Munich, Germany.,2 Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilians University (LMU), Munich, Germany.,4 Munich Cluster for Systems Neurology, Munich, Germany
| |
Collapse
|
31
|
Frontera JA, Provencio JJ, Sehba FA, McIntyre TM, Nowacki AS, Gordon E, Weimer JM, Aledort L. The Role of Platelet Activation and Inflammation in Early Brain Injury Following Subarachnoid Hemorrhage. Neurocrit Care 2017; 26:48-57. [PMID: 27430874 DOI: 10.1007/s12028-016-0292-4] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND Early brain injury (EBI) following aneurysmal subarachnoid hemorrhage (SAH) is an important predictor of poor functional outcome, yet the underlying mechanism is not well understood. Animal studies suggest that platelet activation and inflammation with subsequent microthrombosis and ischemia may be a mechanism of EBI. METHODS A prospective, hypothesis-driven study of spontaneous, SAH patients and controls was conducted. Platelet activation [thromboelastography maximum amplitude (MA)] and inflammation [C-reactive protein (CRP)] were measured serially over time during the first 72 h following SAH onset. Platelet activation and inflammatory markers were compared between controls and SAH patients with mild [Hunt-Hess (HH) 1-3] versus severe (HH 4-5) EBI. The association of these biomarkers with 3-month functional outcomes was evaluated. RESULTS We enrolled 127 patients (106 SAH; 21 controls). Platelet activation and CRP increased incrementally with worse EBI/HH grade, and both increased over 72 h (all P < 0.01). Both were higher in severe versus mild EBI (MA 68.9 vs. 64.8 mm, P = 0.001; CRP 12.5 vs. 1.5 mg/L, P = 0.003) and compared to controls (both P < 0.003). Patients with delayed cerebral ischemia (DCI) had more platelet activation (66.6 vs. 64.9 in those without DCI, P = 0.02) within 72 h of ictus. At 3 months, death or severe disability was more likely with higher levels of platelet activation (mRS4-6 OR 1.18, 95 % CI 1.05-1.32, P = 0.007) and CRP (mRS4-6 OR 1.02, 95 % CI 1.00-1.03, P = 0.041). CONCLUSIONS Platelet activation and inflammation occur acutely after SAH and are associated with worse EBI, DCI and poor 3-month functional outcomes. These markers may provide insight into the mechanism of EBI following SAH.
Collapse
Affiliation(s)
- Jennifer A Frontera
- Cerebrovascular Center of the Neurological Institute, Cleveland Clinic, 9500 Euclid Ave. S80, Cleveland, OH, 44195, USA. .,Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - J Javier Provencio
- Department of Neurology and Neuroscience, Brain Immunology and Glia Center, University of Virginia, Charlottesville, VA, USA
| | - Fatima A Sehba
- Department of Neurosurgery, Mount Sinai School of Medicine, New York, NY, USA
| | - Thomas M McIntyre
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Amy S Nowacki
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Errol Gordon
- Department of Critical Care, Mount Sinai School of Medicine, New York, NY, USA
| | - Jonathan M Weimer
- Cerebrovascular Center of the Neurological Institute, Cleveland Clinic, 9500 Euclid Ave. S80, Cleveland, OH, 44195, USA
| | - Louis Aledort
- Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| |
Collapse
|
32
|
Neural Vascular Mechanism for the Cerebral Blood Flow Autoregulation after Hemorrhagic Stroke. Neural Plast 2017; 2017:5819514. [PMID: 29104807 PMCID: PMC5634612 DOI: 10.1155/2017/5819514] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/11/2017] [Indexed: 12/21/2022] Open
Abstract
During the initial stages of hemorrhagic stroke, including intracerebral hemorrhage and subarachnoid hemorrhage, the reflex mechanisms are activated to protect cerebral perfusion, but secondary dysfunction of cerebral flow autoregulation will eventually reduce global cerebral blood flow and the delivery of metabolic substrates, leading to generalized cerebral ischemia, hypoxia, and ultimately, neuronal cell death. Cerebral blood flow is controlled by various regulatory mechanisms, including prevailing arterial pressure, intracranial pressure, arterial blood gases, neural activity, and metabolic demand. Evoked by the concept of vascular neural network, the unveiled neural vascular mechanism gains more and more attentions. Astrocyte, neuron, pericyte, endothelium, and so forth are formed as a communicate network to regulate with each other as well as the cerebral blood flow. However, the signaling molecules responsible for this communication between these new players and blood vessels are yet to be definitively confirmed. Recent evidence suggested the pivotal role of transcriptional mechanism, including but not limited to miRNA, lncRNA, exosome, and so forth, for the cerebral blood flow autoregulation. In the present review, we sought to summarize the hemodynamic changes and underline neural vascular mechanism for cerebral blood flow autoregulation in stroke-prone state and after hemorrhagic stroke and hopefully provide more systematic and innovative research interests for the pathophysiology and therapeutic strategies of hemorrhagic stroke.
Collapse
|
33
|
Zhang S, Lai Y, Ding X, Parsons M, Zhang JH, Lou M. Absent Filling of Ipsilateral Superficial Middle Cerebral Vein Is Associated With Poor Outcome After Reperfusion Therapy. Stroke 2017; 48:907-914. [PMID: 28265013 DOI: 10.1161/strokeaha.116.016174] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 01/11/2017] [Accepted: 01/20/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND PURPOSE Our aim was to study the effect of drainage of cortical veins, including the superficial middle cerebral vein (SMCV), vein of Trolard, and vein of Labbé on neurological outcomes after reperfusion therapy. METHODS Consecutive ischemic stroke patients who underwent pretreatment computed tomographic perfusion and 24-hour computed tomographic perfusion or magnetic resonance perfusion after intravenous thrombolysis were included. We defined "absent filling of ipsilateral cortical vein" (eg, SMCV-) as no contrast filling of the vein across the whole venous phase on 4-dimensional computed tomographic angiography in the ischemic hemisphere. RESULTS Of 228 patients, SMCV-, vein of Trolard- and vein of Labbé- were observed in 50 (21.9%), 27 (11.8%), and 32 (14.0%) patients, respectively. Only SMCV- independently predicted poor outcome (3-month modified Rankin Scale score of >2; odds ratio, 2.710; P=0.040). No difference was found in reperfusion rate after treatment between patients with and without SMCV- (P>0.05). In patients achieving major reperfusion (≥80%), there was no difference in 24-hour infarct volume, or rate of poor outcome between patients with and without SMCV- (P>0.05). However, in those without major reperfusion, patients with SMCV- had larger 24-hour infarct volume (P=0.011), higher rate of poor outcome (P=0.012), and death (P=0.032) compared with those with SMCV filling. SMCV- was significantly associated with brain edema at 24 hours (P=0.037), which, in turn, was associated with poor 3-month outcome (P=0.002). CONCLUSIONS Lack of SMCV filling contributed to poor outcome after thrombolysis, especially when reperfusion was not achieved. The main deleterious effect of poor venous filling appears related to the development of brain edema.
Collapse
Affiliation(s)
- Sheng Zhang
- From the Departments of Neurology (S.Z., Y.L., M.L.) and Radiology (X.D.), The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China; Department of Neurology, John Hunter Hospital, The University of Newcastle, NSW, Australia (M.P.); and Department of Physiology and Pharmacology, Loma Linda University, CA (J.H.Z.)
| | - Yangxiao Lai
- From the Departments of Neurology (S.Z., Y.L., M.L.) and Radiology (X.D.), The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China; Department of Neurology, John Hunter Hospital, The University of Newcastle, NSW, Australia (M.P.); and Department of Physiology and Pharmacology, Loma Linda University, CA (J.H.Z.)
| | - Xinfa Ding
- From the Departments of Neurology (S.Z., Y.L., M.L.) and Radiology (X.D.), The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China; Department of Neurology, John Hunter Hospital, The University of Newcastle, NSW, Australia (M.P.); and Department of Physiology and Pharmacology, Loma Linda University, CA (J.H.Z.)
| | - Mark Parsons
- From the Departments of Neurology (S.Z., Y.L., M.L.) and Radiology (X.D.), The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China; Department of Neurology, John Hunter Hospital, The University of Newcastle, NSW, Australia (M.P.); and Department of Physiology and Pharmacology, Loma Linda University, CA (J.H.Z.)
| | - John H Zhang
- From the Departments of Neurology (S.Z., Y.L., M.L.) and Radiology (X.D.), The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China; Department of Neurology, John Hunter Hospital, The University of Newcastle, NSW, Australia (M.P.); and Department of Physiology and Pharmacology, Loma Linda University, CA (J.H.Z.)
| | - Min Lou
- From the Departments of Neurology (S.Z., Y.L., M.L.) and Radiology (X.D.), The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China; Department of Neurology, John Hunter Hospital, The University of Newcastle, NSW, Australia (M.P.); and Department of Physiology and Pharmacology, Loma Linda University, CA (J.H.Z.).
| |
Collapse
|
34
|
Kamp MA, Lieshout JHV, Dibué-Adjei M, Weber JK, Schneider T, Restin T, Fischer I, Steiger HJ. A Systematic and Meta-Analysis of Mortality in Experimental Mouse Models Analyzing Delayed Cerebral Ischemia After Subarachnoid Hemorrhage. Transl Stroke Res 2017; 8:206-219. [DOI: 10.1007/s12975-016-0513-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 11/23/2016] [Accepted: 11/27/2016] [Indexed: 01/18/2023]
|
35
|
Balbi M, Koide M, Schwarzmaier SM, Wellman GC, Plesnila N. Acute changes in neurovascular reactivity after subarachnoid hemorrhage in vivo. J Cereb Blood Flow Metab 2017; 37:178-187. [PMID: 26676226 PMCID: PMC5363735 DOI: 10.1177/0271678x15621253] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/15/2015] [Accepted: 06/29/2015] [Indexed: 12/21/2022]
Abstract
Subarachnoid hemorrhage causes acute and long-lasting constrictions of pial arterioles. Whether these vessels dilate normally to neuronal activity is of great interest since a mismatch between delivery and consumption of glucose and oxygen may cause additional neuronal damage. Therefore, we investigated neurovascular reactivity of pial and parenchymal arterioles after experimental subarachnoid hemorrhage. C57BL/6 mice were subjected to subarachnoid hemorrhage by filament perforation or sham surgery. Neurovascular reactivity was assessed 3 h later by forepaw stimulation or inhalation of 10% CO2 Diameters of cerebral arterioles were assessed using two-photon microscopy. Neurovascular coupling and astrocytic endfoot Ca2+ were measured in brain slices using two-photon and infrared-differential interference contrast microscopy. Vessels of sham-operated mice dilated normally to CO2 and forepaw stimulation. Three hours after subarachnoid hemorrhage, CO2 reactivity was completely lost in both pial and parenchymal arterioles, while neurovascular coupling was not affected. Brain slices studies also showed normal neurovascular coupling and a normal increase in astrocytic endfoot Ca2+ acutely after subarachnoid hemorrhage. These findings suggest that communication between neurons, astrocytes, and parenchymal arterioles is not affected in the first few hours after subarachnoid hemorrhage, while CO2 reactivity, which is dependent on NO signaling, is completely lost.
Collapse
Affiliation(s)
- Matilde Balbi
- Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilians University (LMU), Munich, Germany
| | - Masayo Koide
- Department of Pharmacology, University of Vermont, Burlington, VT, USA
| | - Susanne M Schwarzmaier
- Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Munich, Germany
| | - George C Wellman
- Department of Pharmacology, University of Vermont, Burlington, VT, USA
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilians University (LMU), Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| |
Collapse
|
36
|
Terpolilli NA, Feiler S, Dienel A, Müller F, Heumos N, Friedrich B, Stover J, Thal S, Schöller K, Plesnila N. Nitric oxide inhalation reduces brain damage, prevents mortality, and improves neurological outcome after subarachnoid hemorrhage by resolving early pial microvasospasms. J Cereb Blood Flow Metab 2016; 36:2096-2107. [PMID: 26661144 PMCID: PMC5363657 DOI: 10.1177/0271678x15605848] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/29/2015] [Accepted: 07/31/2015] [Indexed: 01/15/2023]
Abstract
Subarachnoid hemorrhage is a stroke subtype with particularly bad outcome. Recent findings suggest that constrictions of pial arterioles occurring early after hemorrhage may be responsible for cerebral ischemia and - subsequently - unfavorable outcome after subarachnoid hemorrhage. Since we recently hypothesized that the lack of nitric oxide may cause post-hemorrhagic microvasospasms, our aim was to investigate whether inhaled nitric oxide, a treatment paradigm selectively delivering nitric oxide to ischemic microvessels, is able to dilate post-hemorrhagic microvasospasms; thereby improving outcome after experimental subarachnoid hemorrhage. C57BL/6 mice were subjected to experimental SAH. Three hours after subarachnoid hemorrhage pial artery spasms were quantified by intravital microscopy, then mice received inhaled nitric oxide or vehicle. For induction of large artery spasms mice received an intracisternal injection of autologous blood. Inhaled nitric oxide significantly reduced number and severity of subarachnoid hemorrhage-induced post-hemorrhage microvasospasms while only having limited effect on large artery spasms. This resulted in less brain-edema-formation, less hippocampal neuronal loss, lack of mortality, and significantly improved neurological outcome after subarachnoid hemorrhage. This suggests that spasms of pial arterioles play a major role for the outcome after subarachnoid hemorrhage and that lack of nitric oxide is an important mechanism of post-hemorrhagic microvascular dysfunction. Reversing microvascular dysfunction by inhaled nitric oxide might be a promising treatment strategy for subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Nicole A Terpolilli
- Department of Neurosurgery, University of Munich Medical Center, Munich, Germany.,Institute for Surgical Research, University of Munich Medical Center, Munich, Germany.,Institute for Stroke and Dementia Research, University of Munich Medical Center, Munich, Germany
| | - Sergej Feiler
- Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - Ari Dienel
- Department of Neurosurgery, University of Munich Medical Center, Munich, Germany.,Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - Frank Müller
- Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - Nicole Heumos
- Department of Neurosurgery, University of Munich Medical Center, Munich, Germany.,Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - Benjamin Friedrich
- Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - John Stover
- Department of Surgery, University of Zurich, Switzerland
| | - Serge Thal
- Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - Karsten Schöller
- Department of Neurosurgery, University of Munich Medical Center, Munich, Germany.,Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - Nikolaus Plesnila
- Institute for Surgical Research, University of Munich Medical Center, Munich, Germany .,Institute for Stroke and Dementia Research, University of Munich Medical Center, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
37
|
The effect of subarachnoid erythrocyte lysate on brain injury: a preliminary study. Biosci Rep 2016; 36:BSR20160100. [PMID: 27279653 PMCID: PMC4945991 DOI: 10.1042/bsr20160100] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 06/08/2016] [Indexed: 02/01/2023] Open
Abstract
We found that more severe brain injury was caused by subarachnoid erythrocyte lysate, and inflammation associated with Prx2 might be involved in mechanism of brain injury. Abundant erythrocytes remain and lyse partially in the subarachnoid space after severe subarachnoid haemorrhage (SAH). But the effect of subarachnoid erythrocyte lysate on brain injury is still not completely clear. In this study, autologous erythrocytes (the non-lysate group) and their lysate (the lysate group) were injected separately into the cistern magna of rabbits to induce a model of experimental SAH, although the control group received isotonic sodium chloride solution instead of erythrocyte solution. Results showed that vasospasm of the basilar artery was observed at 72 h after experimental SAH, but there was no significant difference between the non-lysate group and the lysate group. Brain injury was more severe in the lysate group than in the non-lysate group. Meanwhile, the levels of peroxiredoxin 2 (Prx2), IL-6 and TNF-α in brain cortex and in CSF were significantly higher in the lysate group than those in the non-lysate group. These results demonstrated that brain injury was more likely to be caused by erythrocyte lysate than by intact erythrocytes in subarachnoid space, and inflammation response positively correlated with Prx2 expression might be involved in mechanism of brain injury after SAH.
Collapse
|
38
|
Cortical microcirculatory disturbance in the super acute phase of subarachnoid hemorrhage - In vivo analysis using two-photon laser scanning microscopy. J Neurol Sci 2016; 368:326-33. [PMID: 27538658 DOI: 10.1016/j.jns.2016.06.067] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 05/28/2016] [Accepted: 06/29/2016] [Indexed: 11/21/2022]
Abstract
OBJECTIVE Subarachnoid hemorrhage (SAH) causes cerebral ischemia and drastically worsens the clinical status at onset. However, the arterial flow is surprisingly well maintained on the cerebral surface. We investigated cortical microcirculatory changes in the super acute phase of SAH using two-photon laser scanning microscopy (TPLSM). METHODS SAH was induced at the skull base in 10 mice using a prone endovascular perforation model. Before SAH, and 1, 2, 5, 10, 20, 30 and 60min after SAH, the cortical microcirculation was observed with TPLSM through a cranial window. Diameters of penetrating and precapillary arterioles were measured and red blood cell (RBC) velocities in precapillary arterioles were analyzed using a line-scan method after administration of Q-dot 655 nanocrystals. RESULTS One minute after SAH, RBC velocity and flow in precapillary arterioles drastically decreased to <20% of the pre-SAH values, while penetrating and precapillary arterioles dilated significantly. Subsequently, the arterioles either dilated or constricted inconsistently for 60min with continual decreases in RBC velocity and flow in the arterioles, suggesting neurovascular dysfunction. CONCLUSION SAH caused sudden worsening of the cortical arteriolar velocity and flow at onset. The neurovascular unit cannot function sufficiently to maintain cortical microcirculatory flow in the super acute phase of SAH.
Collapse
|
39
|
Leukocyte plugging and cortical capillary flow after subarachnoid hemorrhage. Acta Neurochir (Wien) 2016; 158:1057-67. [PMID: 27040552 DOI: 10.1007/s00701-016-2792-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/22/2016] [Indexed: 10/22/2022]
Abstract
BACKGROUND It is believed that increased intracranial pressure immediately after subarachnoid hemorrhage (SAH) causes extensive brain ischemia and results in worsening clinical status. Arterial flow to the cerebral surfaces is clinically well maintained during clipping surgery regardless of the severity of the World Federation of Neurological Societies grade after SAH. To explore what kinds of changes occur in the cortical microcirculation, not at the cerebral surface, we examined cortical microcirculation after SAH using two-photon laser scanning microscopy (TPLSM). METHODS SAH was induced in mice with an endovascular perforation model. Following continuous injection of rhodamine 6G, velocities of labeled platelets and leukocytes and unlabeled red blood cells (RBCs) were measured in the cortical capillaries 60 min after SAH with a line-scan method using TPLSM, and the data were compared to a sham group and P-selectin monoclonal antibody-treated group. RESULTS Velocities of leukocytes, platelets, and RBCs in capillaries decreased significantly 60 min after SAH. Rolling and adherent leukocytes suddenly prevented other blood cells from flowing in the capillaries. Flowing blood cells also decreased significantly in each capillary after SAH. This no-reflow phenomenon induced by plugging leukocytes was often observed in the SAH group but not in the sham group. The decreased velocities of blood cells were reversed by pretreatment with the monoclonal antibody of P-selection, an adhesion molecule expressed on the surfaces of both endothelial cells and platelets. CONCLUSIONS SAH caused sudden worsening of cortical microcirculation at the onset. Leukocyte plugging in capillaries is one of the reasons why cortical microcirculation is aggravated after SAH.
Collapse
|
40
|
Szklener S, Melges A, Korchut A, Zaluska W, Trojanowski T, Rejdak R, Rejdak K. Predictive model for patients with poor-grade subarachnoid haemorrhage in 30-day observation: a 9-year cohort study. BMJ Open 2015; 5:e007795. [PMID: 26070797 PMCID: PMC4466620 DOI: 10.1136/bmjopen-2015-007795] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE The purpose of this study was to identify prognostic factors and build the predictive model based on poor-grade subarachnoid haemorrhage (SAH) population received only supportive symptomatic treatment. DESIGN Prospective observational cohort study. SETTING Intensive care unit at the Clinical Department of Neurology. PARTICIPANTS A total of 101 patients with spontaneous SAH disqualified from neurosurgical operative treatment due to poor clinical condition. Data were collected over a 9-year period. OUTCOME MEASURES Unfavourable outcome was defined as a modified Rankin Score ≥ 5 at 30 days of observation. RESULTS Multivariable logistic regression analysis indicated the World Federation of Neurosurgical Societies Scale score, increasing age, Fisher grade and admission leucocytosis as independent predictive factors. The proposed scale subdivides the study population into four prognostic groups with significantly different outcomes: grade I: probability of favourable outcome 89.9%; grade II: 47.5%; grade III: 4.2%; grade IV: 0%. The receiver operating characteristic (ROC) curve for the prediction of outcome performed by the new scale had an area under the curve (AUC)=0.910 (excellent accuracy). CONCLUSIONS Unfavourable outcome in non-operated patients with poor-grade SAH is strongly predicted by traditional unmodifiable factors such as age, amount of bleeding in CT, level of consciousness as well as leucocytosis. A new predictive scale based on the above parameters seems to reliably predict the outcome and may contribute to more effective planning of therapeutic management in patients with poor-grade SAH.
Collapse
Affiliation(s)
| | - Anna Melges
- Department of Neurology, Medical University of Lublin, Poland
| | | | | | | | - Robert Rejdak
- Department of General Ophthalmology, Medical University of Lublin, Poland
| | - Konrad Rejdak
- Department of Neurology, Medical University of Lublin, Poland
| |
Collapse
|
41
|
Muroi C, Fujioka M, Marbacher S, Fandino J, Keller E, Iwasaki K, Mishima K. Mouse model of subarachnoid hemorrhage: technical note on the filament perforation model. ACTA NEUROCHIRURGICA. SUPPLEMENT 2015; 120:315-20. [PMID: 25366644 DOI: 10.1007/978-3-319-04981-6_54] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Experiments using genetically engineered mice are regarded as indispensable to gaining a better understanding of the molecular pathophysiology in neuronal injury after subarachnoid hemorrhage (SAH). Therefore, mouse SAH models are becoming increasingly important. The circle of Willis perforation (cWp) model is the most frequently used mouse SAH model. We report and discuss the technical surgical approach, results, and difficulties associated with the cWp model, with reference to the existing literature. Our results largely confirmed previously published results. This model may be the first choice at present, because important pathologies can be reproduced in this model and most findings in the literature are based on it.
Collapse
Affiliation(s)
- Carl Muroi
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan,
| | | | | | | | | | | | | |
Collapse
|
42
|
Chen S, Chen Y, Xu L, Matei N, Tang J, Feng H, Zhang J. Venous system in acute brain injury: Mechanisms of pathophysiological change and function. Exp Neurol 2015; 272:4-10. [PMID: 25783658 DOI: 10.1016/j.expneurol.2015.03.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 03/09/2015] [Indexed: 01/31/2023]
Abstract
Cerebral vascular injury is a major component of acute brain injury. Currently, neuroprotective strategies primarily focus on the recanalization of cerebral arteries and capillaries, and the protection of insulted neurons. Hitherto, the role of vein drainage in the pathophysiology of acute brain injury has been overlooked, due to an under appreciation of the magnitude of the impact of veins in circulation. In this review, we summarize the changes in the vein morphology and functions that are known, or likely to occur related to acute brain injury, and aim to advance the therapeutic management of acute brain injury by shifting the focus from reperfusion to another term: recirculation. Recent progress in the neurobiological understanding of the vascular neural network has demonstrated that cerebral venous systems are able to respond to acute brain injury by regulating the blood flow disharmony following brain edema, blood brain barrier disruption, ischemia, and hemorrhage. With the evidence presented in this review, future clinical management of acutely brain injured patients will expand to include the recirculation concept, establishing a harmony between arterial and venous systems, in addition to the established recanalization and reperfusion strategies.
Collapse
Affiliation(s)
- Sheng Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Liang Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Nathanael Matei
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - JohnH Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA
| |
Collapse
|
43
|
Schwarzmaier SM, Plesnila N. Contributions of the immune system to the pathophysiology of traumatic brain injury - evidence by intravital microscopy. Front Cell Neurosci 2014; 8:358. [PMID: 25408636 PMCID: PMC4219391 DOI: 10.3389/fncel.2014.00358] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/13/2014] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) results in immediate brain damage that is caused by the mechanical impact and is non-reversible. This initiates a cascade of delayed processes which cause additional—secondary—brain damage. Among these secondary mechanisms, the inflammatory response is believed to play an important role, mediating actions that can have both protective and detrimental effects on the progression of secondary brain damage. Histological data generated extensive information; however, this is only a snapshot of processes that are, in fact, very dynamic. In contrast, in vivo microscopy provides detailed insight into the temporal and spatial patterns of cellular dynamics. In this review, we aim to summarize data which was generated by in vivo microscopy, specifically investigating the immune response following brain trauma, and its potential effects on secondary brain damage.
Collapse
Affiliation(s)
- Susanne M Schwarzmaier
- Department of Anesthesiology, University of Munich Medical Center Munich, Germany ; Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center Munich, Germany ; Munich Cluster of Systems Neurology Munich, Germany
| |
Collapse
|
44
|
Rat endovascular perforation model. Transl Stroke Res 2014; 5:660-8. [PMID: 25213427 DOI: 10.1007/s12975-014-0368-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 08/18/2014] [Accepted: 08/21/2014] [Indexed: 12/31/2022]
Abstract
Experimental animal models of aneurysmal subarachnoid hemorrhage (SAH) have provided a wealth of information on the mechanisms of brain injury. The rat endovascular perforation (EVP) model replicates the early pathophysiology of SAH and hence is frequently used to study early brain injury following SAH. This paper presents a brief review of historical development of the EVP model and details the technique used to create SAH and considerations necessary to overcome technical challenges.
Collapse
|
45
|
Bühler D, Schüller K, Plesnila N. Protocol for the induction of subarachnoid hemorrhage in mice by perforation of the Circle of Willis with an endovascular filament. Transl Stroke Res 2014; 5:653-9. [PMID: 25123204 PMCID: PMC4213389 DOI: 10.1007/s12975-014-0366-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 07/25/2014] [Accepted: 08/05/2014] [Indexed: 12/05/2022]
Abstract
Genetically engineered mice are a valuable tool to investigate the molecular and cellular mechanisms leading to brain damage following subarachnoid hemorrhage (SAH). Therefore, several murine SAH models were developed during the last 15 years. Among those models, the perforation of the Circle of Willis by an endovascular filament or “filament model” turned out to become the most popular one, since it is believed to reproduce some of the most prominent pathophysiological features observed after human SAH. Despite the importance of the endovascular filament model for SAH research, relatively few studies were published using this technique during the past years and a number of laboratories reported problems establishing the technique. This triggered discussions about the standardization, reproducibility, and the reliability of the model. In order to improve this situation, the current paper aims to provide a comprehensive hands-on protocol of the murine endovascular filament model. The protocol proved to result in induction of SAH in mice with high intrapersonal and interpersonal reproducibility and is based on our experience with this technique for more than 10 years. By sharing our experience with this valuable model, we aim to initiate a constantly ongoing discussion process on the improvement of standards and techniques in the field of experimental SAH research.
Collapse
Affiliation(s)
- Dominik Bühler
- Institute for Stroke and Dementia Research, University of Munich Medical Center, Max-Lebsche Platz 30, 81377, Munich, Germany
| | | | | |
Collapse
|
46
|
Muroi C, Fujioka M, Okuchi K, Fandino J, Keller E, Sakamoto Y, Mishima K, Iwasaki K, Fujiwara M. Filament perforation model for mouse subarachnoid hemorrhage: Surgical-technical considerations. Br J Neurosurg 2014; 28:722-32. [DOI: 10.3109/02688697.2014.918579] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
47
|
Fukuoka T, Hayashi T, Hirayama M, Maruyama H, Tanahashi N. Cilostazol Inhibits Platelet–Endothelial Cell Interaction in Murine Microvessels after Transient Bilateral Common Carotid Artery Occlusion. J Stroke Cerebrovasc Dis 2014; 23:1056-61. [DOI: 10.1016/j.jstrokecerebrovasdis.2013.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 08/30/2013] [Accepted: 09/04/2013] [Indexed: 11/26/2022] Open
|
48
|
Subarachnoid Hemorrhage: a Review of Experimental Studies on the Microcirculation and the Neurovascular Unit. Transl Stroke Res 2014; 5:174-89. [DOI: 10.1007/s12975-014-0323-4] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/20/2013] [Accepted: 01/03/2014] [Indexed: 11/29/2022]
|
49
|
Cilostazol inhibits leukocyte-endothelial cell interactions in murine microvessels after transient bilateral common carotid artery occlusion. Brain Res 2014; 1543:173-8. [PMID: 24309140 DOI: 10.1016/j.brainres.2013.11.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 11/09/2013] [Accepted: 11/15/2013] [Indexed: 11/20/2022]
Abstract
Leukocyte behavior in the cerebral microvasculature following vessel occlusion has not been fully elucidated. The purpose of this study was to investigate the effects of cilostazol on leukocyte behavior (rolling and adhesion) in murine cerebral microvessels following transient bilateral carotid artery occlusion using intravital fluorescence microscopy. Four groups of mice were assigned: a sham group (n=16); an ischemia (induced by 15-min occlusion of bilateral common carotid arteries) and reperfusion (I/R) group (n=13); I/R+cilostazol (I/R+CZ3 mg/kg) group (I/R after oral administration of cilostazol at 3 mg/kg) (n=8); and I/R+cilostazol (I/R+CZ30 mg/kg) group (I/R after oral administration of cilostazol at 30 mg/kg) (n=12). Leukocytes labeled with 0.05% acridine orange were administered intravenously and their behavior was investigated at 3 and 6 h after reperfusion. Numbers of rolling or adherent leukocytes were expressed as the count per square millimeter per 30s. Numbers of rolling and adherent leukocytes at 3 and 6h after reperfusion were significantly higher in the I/R group than in the sham or I/R+CZ30 mg/kg groups in both pial veins (P<0.05) and pial arteries (P<0.05). Cilostazol (30 mg/kg) inhibited leukocyte-endothelial interactions following cerebral ischemia and reperfusion.
Collapse
|
50
|
Brathwaite S, Macdonald RL. Current management of delayed cerebral ischemia: update from results of recent clinical trials. Transl Stroke Res 2013; 5:207-26. [PMID: 24338266 DOI: 10.1007/s12975-013-0316-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 11/23/2013] [Accepted: 11/29/2013] [Indexed: 01/19/2023]
Abstract
Subarachnoid hemorrhage (SAH) accounts for 5-7% of all strokes worldwide and is associated with high mortality and morbidity. Even after surgical intervention, approximately 30% of patients develop long-term cognitive and neurological deficits that significantly affect their capacity to return to work or daily life unassisted. Much of this stems from a secondary ischemic phenomenon referred to as delayed cerebral ischemia (DCI). While DCI has been historically attributed to the narrowing of the large basal cerebral arteries, it is now recognized that numerous pathways contribute to its pathogenesis, including microcirculatory dysfunction, microthrombosis, cortical spreading depression, and early brain injury. This paper seeks to summarize some of the key pathophysiological events that are associated with poor outcome after SAH, provide a general overview of current methods of treating SAH patients, and review the results of recent clinical trials directed at improving outcome after SAH. The scientific basis of these studies will be discussed, in addition to the available results and recommendations for effective patient management. Therapeutic methods under current clinical investigation will also be addressed. In particular, the mechanisms by which they are expected to elicit improved outcome will be investigated, as well as the specific study designs and anticipated time lines for completion.
Collapse
Affiliation(s)
- Shakira Brathwaite
- Division of Neurosurgery, Department of Surgery, St. Michael's Hospital, 30 Bond Street, Toronto, Ontario, Canada, M5B 1W8
| | | |
Collapse
|