1
|
Fanaee S, Austin W, Filiaggi M, Adibnia V. External Bleeding and Advanced Biomacromolecules for Hemostasis. Biomacromolecules 2024; 25:6936-6966. [PMID: 39463174 DOI: 10.1021/acs.biomac.4c00952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Hemorrhage is a significant medical problem that has been an active area of research over the past few decades. The human body has a complex response to bleeding that leads to blood clot formation and hemostasis. Many biomaterials based on various biomacromolecules have been developed to either accelerate or improve the body's natural response to bleeding. This review examines the mechanisms of hemostasis, types of bleeding, and the in vitro or in vivo models and techniques used to study bleeding and hemostatic materials. It provides a detailed overview of the diverse hemostatic materials, including those that are highly absorbent, wet adhesives, and those that accelerate the biochemical cascade of blood clotting. These materials are currently marketed, under preclinical testing, or being researched. In exploring the latest advancements in hemostatic technologies, this paper highlights the potential of these materials to significantly improve bleeding control in clinical and emergency situations.
Collapse
Affiliation(s)
- Sajjad Fanaee
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - William Austin
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Mark Filiaggi
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Vahid Adibnia
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Chemistry, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
2
|
Diwan D, Mehla J, Nelson JW, Quirk JD, Song SK, Cao S, Meron B, Mostofa A, Zipfel GJ. Development and Validation of a Prechiasmatic Mouse Model of Subarachnoid Hemorrhage to Measure Long-Term Cognitive Deficits. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2403977. [PMID: 39443821 DOI: 10.1002/advs.202403977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/17/2024] [Indexed: 10/25/2024]
Abstract
Controllable and reproducible animal models of aneurysmal subarachnoid hemorrhage (SAH) are crucial for the systematic study of the pathophysiology and treatment of this debilitating condition. However, current animal models have not been successful in replicating the pathology and disabilities seen in SAH patients, especially the long-term neurocognitive deficits that affect the survivor's quality of life. Therefore, there is an unmet need to develop experimental models that reliably replicate the long-term clinical ramifications of SAH - especially in mice where genetic manipulations are straightforward and readily available. To address this need, a standardized mouse SAH model is developed that reproducibly produced significant and trackable long-term cognitive deficits. SAH is induced by performing double blood injections into the prechiasmatic cistern - a simple modification to the well-characterized single prechiasmatic injection mouse model of SAH. Following SAH, mice recapitulated key characteristics of SAH patients, including cerebral edema measured by MRI - an indicator of early brain injury (EBI), neuroinflammation, apoptosis, and long-term cognitive impairment. This newly developed SAH mouse model is considered an ideal paradigm for investigating the complex SAH pathophysiology and identifying novel druggable therapeutic targets for treating SAH severity and SAH-associated long-term neurocognitive deficits in patients.
Collapse
Affiliation(s)
- Deepti Diwan
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jogender Mehla
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - James W Nelson
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - James D Quirk
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sheng-Kwei Song
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sarah Cao
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Benjamin Meron
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Aminah Mostofa
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Gregory J Zipfel
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
3
|
Diwan D, Mehla J, Nelson JW, Zipfel GJ. Development and validation of prechiasmatic mouse model of subarachnoid hemorrhage to measure long-term neurobehavioral impairment. RESEARCH SQUARE 2024:rs.3.rs-4176908. [PMID: 38645258 PMCID: PMC11030500 DOI: 10.21203/rs.3.rs-4176908/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Controllable and reproducible animal models of aneurysmal subarachnoid hemorrhage (SAH) are crucial for the systematic study of the pathophysiology and treatment of this debilitating condition. Despite the variety of animal models of SAH currently available, attempts to translate promising therapeutic strategies from preclinical studies to humans have largely failed. This failure is likely due, at least in part, to poor replication of pathology and disabilities in these preclinical models, especially the long-term neurocognitive deficits that drive poor quality of life / return to work in SAH survivors. Therefore, there is an unmet need to develop experimental models that reliably replicate the long-term clinical ramifications of SAH - especially in mice where genetic manipulations are straightforward and readily available. To address this need, we developed a standardized mouse model of SAH that reproducibly produced significant and trackable long-term neurobehavioral deficits. SAH was induced by performing double blood injections into the prechiasmatic cistern - a simple modification to the well-characterized single prechiasmatic injection mouse model of SAH. Following SAH, mice recapitulated key characteristics of SAH patients including long-term cognitive impairment as observed by a battery of behavioral testing and delayed pathophysiologic processes assayed by neuroinflammatory markers. We believe that this new SAH mouse model will be an ideal paradigm for investigating the complex pathophysiology of SAH and identifying novel druggable therapeutic targets for treating SAH-associated long-term neurocognitive deficits in patients.
Collapse
|
4
|
Tanner S, Zhou J, Bietar B, Lehmann C. Validation of a simplified model for subarachnoid hemorrhage in mice. Clin Hemorheol Microcirc 2024; 87:301-313. [PMID: 38701138 DOI: 10.3233/ch-231997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) represents a severe injury to the brain and is associated with a high mortality (40%). Several experimental SAH models are described in the literature requiring specialized equipment and a high degree of surgical expertise. Our goal was to validate a simplified, cost-effective model to permit future studies of SAH. METHODS SAH was induced by injection of homologous blood into the cisterna magna. Perfusion-fixation then perfusion of gelatinous India ink was performed. Brains and brainstems were collected and imaged for analysis of cerebral vasospasm. Triphenyl tetrazolium chloride (TTC) staining was used to analyze brain tissue cell death 24 hours following stroke. A composite neuroscore was utilized to assess SAH-related neurologic deficits. RESULTS Anterior cerebral artery and basilary artery diameters were significantly reduced at 24 hours post SAH induction. Middle cerebral artery diameter was also reduced; however, the results were not significant. TTC staining showed no infarcted tissue. Neuroscores were significantly lower in the SAH mice, indicating the presence of functional deficits. CONCLUSIONS This simplified model of SAH elicits pathological changes consistent with those described for more complex models in the literature. Therefore, it can be used in future preclinical studies examining the pathophysiology of SAH and novel treatment options.
Collapse
Affiliation(s)
- Sophie Tanner
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Juan Zhou
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Bashir Bietar
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Christian Lehmann
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| |
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 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
|
Kim HY, Back DB, Choi BR, Choi DH, Kwon KJ. Rodent Models of Post-Stroke Dementia. Int J Mol Sci 2022; 23:ijms231810750. [PMID: 36142661 PMCID: PMC9501431 DOI: 10.3390/ijms231810750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Post-stroke cognitive impairment is one of the most common complications in stroke survivors. Concomitant vascular risk factors, including aging, diabetes mellitus, hypertension, dyslipidemia, or underlying pathologic conditions, such as chronic cerebral hypoperfusion, white matter hyperintensities, or Alzheimer’s disease pathology, can predispose patients to develop post-stroke dementia (PSD). Given the various clinical conditions associated with PSD, a single animal model for PSD is not possible. Animal models of PSD that consider these diverse clinical situations have not been well-studied. In this literature review, diverse rodent models that simulate the various clinical conditions of PSD have been evaluated. Heterogeneous rodent models of PSD are classified into the following categories: surgical technique, special structure, and comorbid condition. The characteristics of individual models and their clinical significance are discussed in detail. Diverse rodent models mimicking the specific pathomechanisms of PSD could provide effective animal platforms for future studies investigating the characteristics and pathophysiology of PSD.
Collapse
Affiliation(s)
- Hahn Young Kim
- Department of Neurology, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Korea
- Correspondence: ; Tel.: +82-2-2030-7563; Fax: +82-2-2030-5169
| | - Dong Bin Back
- Department of Neurology, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Korea
| | - Bo-Ryoung Choi
- Department of Neurology, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Korea
| | - Dong-Hee Choi
- Department of Medicine, Konkuk University School of Medicine, Seoul 05030, Korea
| | - Kyoung Ja Kwon
- Department of Medicine, Konkuk University School of Medicine, Seoul 05030, Korea
| |
Collapse
|
7
|
Themistoklis KM, Papasilekas TI, Melanis KS, Boviatsis KA, Korfias SI, Vekrellis K, Sakas DE. The transient intraluminal filament Middle Cerebral Artery Occlusion stroke model in rats. A step by step guide and technical considerations. World Neurosurg 2022; 168:43-50. [PMID: 36115569 DOI: 10.1016/j.wneu.2022.09.043] [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: 06/21/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Stroke is a leading cause of disability and mortality worldwide. Related research, although already providing significant insights on the underlying pathophysiology and potential treatment strategies, is far from conclusive. In this respect, stroke models are proving of extreme significance for laboratories around the world. The scope of this article is to present in detail the most popular to date focal stroke model, the tifMCAO model in rats. This model mimics reliably stroke in humans and also approximates endovascular thrombectomy. METHODS The tifMCAO model was performed on Wistar rats with a weight of 300-400 gr. The surgical technique is described in a step-wise manner, while pictures and/or HD video accompany each step. Complete arteriotomy of the ECA stump is introduced during the procedure. RESULTS We performed the tifMCAO in 65 rats (male and female) involved in various experimental protocols. Although that initial mortality was 48%, practice reduced this number to10%. The mean procedural time was 53 min (range, 38 - 85 min). Stroke was confirmed in 87.5% of the cases. CONCLUSIONS The tifMCAO stroke model in rats is the most commonly utilized experimental model of focal ischemia because of its clinical relevance. We revisited the procedure and divided it, for instructional purposes, in 15 consecutive distinct steps.
Collapse
Affiliation(s)
- Konstantinos M Themistoklis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Department of Neurosurgery, "Korgialenio, Benakio, HRC" General Hospital of Athens, Athens, Greece.
| | - Themistoklis I Papasilekas
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Department of Neurosurgery, "Korgialenio, Benakio, HRC" General Hospital of Athens, Athens, Greece
| | - Konstantinos S Melanis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Department of Neurology, "Evaggelismos" General Hospital of Athens, Athens, Greece
| | | | - Stefanos I Korfias
- Department of Neurosurgery, University of Athens, "Evaggelismos" General Hospital of Athens, Athens, Greece
| | - Konstaninos Vekrellis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Damianos E Sakas
- Department of Neurosurgery, University of Athens, "Evaggelismos" General Hospital of Athens, Athens, Greece
| |
Collapse
|
8
|
Schwarzmaiera SM, Knarr MR, Hu S, Ertürk A, Hellal F, Plesnila N. Perfusion pressure determines vascular integrity and histomorphological quality following perfusion fixation of the brain. J Neurosci Methods 2022; 372:109493. [DOI: 10.1016/j.jneumeth.2022.109493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 01/25/2022] [Accepted: 02/03/2022] [Indexed: 10/19/2022]
|
9
|
In Vivo Vasospasm Induction by Ultrasound Application in the Chicken Chorioallantoic Membrane Model. Transl Stroke Res 2022; 13:616-624. [PMID: 35061211 PMCID: PMC9232457 DOI: 10.1007/s12975-021-00960-y] [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: 02/01/2021] [Revised: 10/01/2021] [Accepted: 10/26/2021] [Indexed: 11/29/2022]
Abstract
Cerebral vasospasm is a highly investigated phenomenon in neurovascular research. Experimental vasospasm models are irreplaceable for the evaluation of new antivasospastic drugs. In this study, we assessed the reliability of in vivo vasospasm induction by ultrasound application in the chicken chorioallantoic membrane (CAM) model. After incubation of fertilized chicken eggs for four days, a fenestration was performed to enable examination of the CAM vessels. On the thirteenth day, continuous-wave ultrasound (3 MHz, 1 W/cm2) was applied on the CAM vessels for 60 s. The ultrasound effect on the vessels was recorded by life imaging (5-MP HD-microscope camera, Leica®). The induced vessel diameter changes were evaluated in a defined time interval of 20 min using a Fiji macro. The vessel diameter before and after sonication was measured and the relative diameter reduction was determined. A first reduction of vessel diameter was observed after three minutes with an average vessel-diameter decrease to 77%. The maximum reduction in vessel diameter was reached eight minutes after sonication with an average vessel diameter decrease to 57% (mean relative diameter reduction of 43%, range 44–61%), ANOVA, p = 0.0002. The vasospasm persisted for all 20 recorded minutes post induction. Vasospasm can be reliably induced by short application of 3 MHz-ultrasound to the CAM vessels. This might be a suitable in vivo model for the evaluation of drug effects on vasospasm in an experimental setting as intermediary in the transition process from in vitro to in vivo assessment using animal models.
Collapse
|
10
|
Engler-Chiurazzi EB, Monaghan KL, Wan ECK, Ren X. Role of B cells and the aging brain in stroke recovery and treatment. GeroScience 2020; 42:1199-1216. [PMID: 32767220 PMCID: PMC7525651 DOI: 10.1007/s11357-020-00242-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 07/23/2020] [Indexed: 02/07/2023] Open
Abstract
As mitigation of brain aging continues to be a key public health priority, a wholistic and comprehensive consideration of the aging body has identified immunosenescence as a potential contributor to age-related brain injury and disease. Importantly, the nervous and immune systems engage in bidirectional communication and can exert profound influence on each other. Emerging evidence supports numerous impacts of innate, inflammatory immune responses and adaptive T cell-mediated immunity in neurological function and diseased or injured brain states, such as stroke. Indeed, a growing body of evidence supports key impacts of brain-resident immune cell activation and peripheral immune infiltration in both the post-stroke acute injury phase and the long-term recovery period. As such, modulation of the immune system is an attractive strategy for novel therapeutic interventions for a devastating age-related brain injury for which there are few readily available neuroprotective treatments or neurorestorative approaches. However, the role of B cells in the context of brain function, and specifically in response to stroke, has not been thoroughly elucidated and remains controversial, leaving our understanding of neuroimmune interactions incomplete. Importantly, emerging evidence suggests that B cells are not pathogenic contributors to stroke injury, and in fact may facilitate functional recovery, supporting their potential value as novel therapeutic targets. By summarizing the current knowledge of the role of B cells in stroke pathology and recovery and interpreting their role in the context of their interactions with other immune cells as well as the immunosenescence cascades that alter their function in aged populations, this review supports an increased understanding of the complex interplay between the nervous and immune systems in the context of brain aging, injury, and disease.
Collapse
Affiliation(s)
- E. B. Engler-Chiurazzi
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506 USA
- Center for Basic & Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506 USA
| | - K. L. Monaghan
- Center for Basic & Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506 USA
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26506 USA
| | - E. C. K. Wan
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506 USA
- Center for Basic & Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506 USA
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26506 USA
| | - X. Ren
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506 USA
- Center for Basic & Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506 USA
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26506 USA
| |
Collapse
|
11
|
Turan N, Heider RA, Nadeem M, Miller BA, Wali B, Yousuf S, Sayeed I, Stein DG, Pradilla G. Neurocognitive Outcomes in a Cisternal Blood Injection Murine Model of Subarachnoid Hemorrhage. J Stroke Cerebrovasc Dis 2020; 29:105249. [PMID: 33066928 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/13/2020] [Accepted: 08/10/2020] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) results in neurocognitive dysfunction and anxiety in humans and in animal models. Neurobehavioral tests such as the Morris Water Maze (MWM) and Elevated Plus Maze (EPM) tests are validated in several models of SAH but have not been tested in the murine cisternal blood injection SAH model. METHODS Adult C57BL/6 mice (n=16) were randomized into two groups. Group 1 (n=8) received sham surgery. Group 2 (n=8) underwent SAH with 60 µL of autologous blood injected into the cisterna magna. Mice were then tested using the Modified Garcia Score on post-operative day 2 (POD2), EPM on POD5 & POD16, and MWM on POD6-16.Brain tissues harvested on POD16 were stained with Fluoro-Jade C to identify neurodegeneration in the hippocampus and cortex and Iba-1 immunofluorescence staining for microglial activation in the dentate gyrus and CA1 region of the hippocampus. RESULTS SAH mice showed increased escape latency on POD10. Swim distance was significantly increased on POD9-10 and swim speed was significantly decreased on POD6&POD10 in SAH mice. SAH mice exhibited a trend for lowered proportion of covered arm entries in EPM on POD16. Modified Garcia Score was not significantly different between the groups on POD2. The area of microglial activation in the dentate gyrus and CA1 region of the hippocampus was mildly increased but not significantly different at day 16 after SAH. Similarly, no significant differences were noted in the number of Fluoro-Jade C (+) cells in cortex or hippocampus. CONCLUSIONS Cisternal single blood injection in mice produces mild neurocognitive deficits most pronounced in spatial learning and most evident 10 days after SAH.
Collapse
Affiliation(s)
- Nefize Turan
- Emory University School of Medicine, Department of Neurosurgery, Atlanta, GA, USA; Cerebrovascular Research Laboratory, Department of Neurosurgery, Atlanta, GA, USA
| | - Robert A Heider
- Emory University School of Medicine, Department of Neurosurgery, Atlanta, GA, USA
| | - Maheen Nadeem
- Emory University School of Medicine, Department of Neurosurgery, Atlanta, GA, USA
| | - Brandon A Miller
- Emory University School of Medicine, Department of Neurosurgery, Atlanta, GA, USA; Cerebrovascular Research Laboratory, Department of Neurosurgery, Atlanta, GA, USA
| | - Bushra Wali
- Emory University School of Medicine, Department of Emergency Medicine, Atlanta, GA, USA
| | - Seema Yousuf
- Emory University School of Medicine, Department of Emergency Medicine, Atlanta, GA, USA
| | - Iqbal Sayeed
- Emory University School of Medicine, Department of Emergency Medicine, Atlanta, GA, USA
| | - Donald G Stein
- Emory University School of Medicine, Department of Emergency Medicine, Atlanta, GA, USA
| | - Gustavo Pradilla
- Emory University School of Medicine, Department of Neurosurgery, Atlanta, GA, USA; Cerebrovascular Research Laboratory, Department of Neurosurgery, Atlanta, GA, USA.
| |
Collapse
|
12
|
Lan SH, Lai WT, Zheng SY, Yang L, Fang LC, Zhou L, Tang B, Duan J, Hong T. Upregulation of Connexin 40 Mediated by Nitric Oxide Attenuates Cerebral Vasospasm After Subarachnoid Hemorrhage via the Nitric Oxide-Cyclic Guanosine Monophosphate-Protein Kinase G Pathway. World Neurosurg 2020; 136:e476-e486. [PMID: 31953101 DOI: 10.1016/j.wneu.2020.01.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 01/04/2020] [Accepted: 01/06/2020] [Indexed: 01/07/2023]
Abstract
OBJECTIVE The present study was performed to elucidate the role of nitric oxide (NO) and connexin 40 (Cx40) in the induction of cerebral vasospasm after subarachnoid hemorrhage (SAH) in vivo. METHODS A SAH rat model was established using the double-bleed method. A total of 108 Sprague-Dawley rats weighing 250-300 g were randomly divided into 6 groups: SAH; SAH plus diethylenetriamine (DETA)/NO (exogenous NO donor); SAH plus 8-bromoadenosine (8-Br)-cyclic guanosine monophosphate (cGMP; protein kinase G [PKG] activator); SAH plus DETA/NO plus KT5823 (PKG inhibitor); SAH plus DETA/NO plus 40Gap27 (Cx40 inhibitor); and sham. The changes in the diameter of the branch microvessels in the middle cerebral artery were recorded. The neurological score was evaluated using the Garcia scoring system. Basilar artery (BA) tension was measured using the Danish Myo Technology myograph system. Cx40 protein expression was analyzed using immunofluorescence and Western blotting. Endothelial NO synthase, soluble guanylate cyclase, and PKG protein expression were measured by Western blotting. RESULTS A considerable narrowing of the cerebral vessels was detected in the SAH group compared with that in the sham group. Moreover, compared with the sham group, the SAH group showed a marked decrease in Cx40, endothelial NO synthase, soluble guanylate cyclase, and PKG expression. The expression of Cx40 and PKG were obviously higher in the SAH plus DETA/NO and SAH plus 8-Br-cGMP groups than in the SAH group. However, Cx40 was lower in the SAH plus DETA/NO plus KT5823 and SAH plus DETA/NO plus 40Gap27 groups than in the SAH plus ETA/NO group. The BAs showed significant vasodilation in the SAH plus DETA/NO and SAH plus 8-Br-cGMP groups. However, the vasodilation response of BAs was inhibited in the SAH plus DETA/NO plus KT5823 and SAH plus DETA-NO plus 40Gap27 groups. CONCLUSIONS The NO-cGMP-PKG pathway alleviated cerebral vasospasm via Cx40 upregulation.
Collapse
Affiliation(s)
- Shi Hai Lan
- Department of Neurosurgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wen Tao Lai
- Department of Neurosurgery, Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China
| | - Su Yue Zheng
- Department of Neurosurgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Le Yang
- Department of Neurosurgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lin Chun Fang
- Department of Neurosurgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lin Zhou
- Department of Neurosurgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bin Tang
- Department of Neurosurgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jian Duan
- Department of Neurosurgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tao Hong
- Department of Neurosurgery, First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
13
|
Wang Q, Luo Q, Zhao YH, Chen X. Toll-like receptor-4 pathway as a possible molecular mechanism for brain injuries after subarachnoid hemorrhage. Int J Neurosci 2020; 130:953-964. [PMID: 31903827 DOI: 10.1080/00207454.2019.1709845] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Subarachnoid hemorrhage (SAH) is known as an acute catastrophic neurological disease that continues to be a serious and significant health problem worldwide. The mechanisms contributing to brain injury after SAH remain unclear despite decades of study focusing on early brain injury (EBI) and delayed brain injury (DBI). Neuroinflammation is a well-recognized consequence of SAH and may be responsible for EBI, cerebral vasospasm, and DBI. Toll-like receptors (TLRs) play a crucial role in the inflammatory response by recognizing damage-associated molecular patterns derived from the SAH. TLR4 is the most studied Toll-like receptor and is widely expressed in the central nervous system (CNS). It can be activated by the extravasated blood components in myeloid differentiation primary response-88/Toll/interleukin-1 receptor-domain-containing adapter-inducing interferon-β (MyD88/TRIF)-dependent pathway after SAH. Transcription factors, such as nuclear factor-κB (NF-κB), mitogen-activated protein kinase (MAPK) and interferon regulatory factor (IRF), that regulate the expression of proinflammatory cytokine genes are initiated by the activation of TLR4, which cause the brain damage after SAH. TLR4 may therefore be a useful therapeutic target for overcoming EBI and DBI in post-SAH neuroinflammation, thereby improving SAH outcome. In the present review, we summarized recent findings from basic and clinical studies of SAH, with a primary focus on the biological characteristics and functions of TLR4 and discussed the mechanisms associated with TLR4 signaling pathway in EBI and DBI following SAH.
Collapse
Affiliation(s)
- Qunhui Wang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Qi Luo
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Yu-Hao Zhao
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Xuan Chen
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, P. R. China
| |
Collapse
|
14
|
Luo F, Wu L, Zhang Z, Zhu Z, Liu Z, Guo B, Li N, Ju J, Zhou Q, Li S, Yang X, Mak S, Han Y, Sun Y, Wang Y, Zhang G, Zhang Z. The dual-functional memantine nitrate MN-08 alleviates cerebral vasospasm and brain injury in experimental subarachnoid haemorrhage models. Br J Pharmacol 2019; 176:3318-3335. [PMID: 31180578 DOI: 10.1111/bph.14763] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 05/26/2019] [Accepted: 06/03/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Cerebral vasospasm and neuronal apoptosis after subarachnoid haemorrhage (SAH) is the major cause of morbidity and mortality in SAH patients. So far, single-target agents have not prevented its occurrence. Memantine, a non-competitive NMDA re3ceptor antagonist, is known to alleviate brain injury and vasospasm in experimental models of SAH. Impairment of NO availability also contributes to vasospasm. Recently, we designed and synthesized a memantine nitrate MN-08, which has potent dual functions: neuroprotection and vasodilation. Here, we have tested the therapeutic effects of MN-08 in animal models of SAH. EXPERIMENTAL APPROACH Binding to NMDA receptors (expressed in HEK293 cells), NO release and vasodilator effects of MN-08 were assessed in vitro. Therapeutic effects of MN-08 were investigated in vivo, using rat and rabbit SAH models. KEY RESULTS MN-08 bound to the NMDA receptor, slowly releasing NO in vitro and in vivo. Consequently, MN-08 relaxed the pre-contracted middle cerebral artery ex vivo and increased blood flow velocity in small vessels of the mouse cerebral cortex. It did not, however, lower systemic blood pressure. In an endovascular perforation rat model of SAH, MN-08 improved the neurological scores and ameliorated cerebral vasospasm. Moreover, MN-08 also alleviated cerebral vasospasm in a cisterna magna single-injection model in rabbits. MN-08 attenuated neural cell apoptosis in both rat and rabbit models of SAH. Importantly, the therapeutic benefit of MN-08 was greater than that of memantine. CONCLUSION AND IMPLICATIONS MN-08 has neuroprotective potential and can ameliorate vasospasm in experimental SAH models.
Collapse
Affiliation(s)
- Fangcheng Luo
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Liangmiao Wu
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Zhixiang Zhang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Zeyu Zhu
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Zheng Liu
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Baojian Guo
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Ning Li
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Jun Ju
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Qiang Zhou
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Shupeng Li
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Center for Disease Control and Prevention, Shenzhen, China
| | - Shinghung Mak
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.,Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Yifan Han
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.,Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Yewei Sun
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Yuqiang Wang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Gaoxiao Zhang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Zaijun Zhang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University College of Pharmacy, Guangzhou, China
| |
Collapse
|
15
|
Lei C, Ruan Y, Cai C, He B, Zhao D. Role of P38 mitogen-activated protein kinase on Cx43 phosphorylation in cerebral vasospasm after subarachnoid hemorrhage. Int J Neurosci 2018; 129:461-469. [PMID: 30369282 DOI: 10.1080/00207454.2018.1538992] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Chao Lei
- Department of Neurosurgery, First Affiliated Hospital of Medical College, Shihezi University, Xinjiang, China
| | - Yutian Ruan
- Department of Thoracic Surgery and Neurosurgery, Beitun Hospital of the Ten Division of the Xinjiang Production and Construction Corps, Xinjiang, China
| | - Changcheng Cai
- Department of Neurosurgery, First Affiliated Hospital of Medical College, Shihezi University, Xinjiang, China
| | - Bao He
- Department of Neurosurgery, First Affiliated Hospital of Medical College, Shihezi University, Xinjiang, China
| | - Dong Zhao
- Department of Neurosurgery, First Affiliated Hospital of Medical College, Shihezi University, Xinjiang, China
| |
Collapse
|
16
|
Wang KC, Tang SC, Lee JE, Tsai JC, Lai DM, Lin WC, Lin CP, Tu YK, Hsieh ST. Impaired microcirculation after subarachnoid hemorrhage in an in vivo animal model. Sci Rep 2018; 8:13315. [PMID: 30190518 PMCID: PMC6127197 DOI: 10.1038/s41598-018-31709-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 08/23/2018] [Indexed: 11/16/2022] Open
Abstract
The influence of aneurysmal subarachnoid hemorrhage (SAH) on brain microcirculation has not yet been systematically investigated. We established an animal model to examine (1) the brain surface microcirculation (2) the influences of cerebrospinal fluid (CSF) from aneurysmal SAH on the brain surface microcirculation. A rat SAH model was induced by injection of autologous arterial blood into the cisterna magnum, and the brain surface microcirculation was evaluated by a capillary videoscope with craniotomy at the fronto-parietal region. CSF from SAH rats and SAH patients was applied on the brain surface of naïve rats to assess the resulting microcirculatory changes. In the SAH rats, diffuse constriction of cortical arterioles within 24 hours of SAH was observed. Similar patterns of microcirculation impairment were induced on normal rat brain surfaces via application of CSF from SAH rats and SAH patients. Furthermore, the proportion of subjects with arteriolar vasoconstriction was significantly higher in the group of SAH patients with delayed ischemic neurological deficits (DIND) than in those without DIND (p < 0.001). This study demonstrated impaired microcirculation on brain surface arterioles in a rat model of SAH. CSF from SAH rats and patients was responsible for impairment of brain surface microcirculation.
Collapse
Affiliation(s)
- Kuo-Chuan Wang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Sung-Chun Tang
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Jing-Er Lee
- Department of Neurology, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan
| | - Jui-Chang Tsai
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Dar-Ming Lai
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Wei-Chou Lin
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Peng Lin
- Department of Anesthesiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yong-Kwang Tu
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Sung-Tsang Hsieh
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan. .,Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
17
|
modCHIMERA: a novel murine closed-head model of moderate traumatic brain injury. Sci Rep 2018; 8:7677. [PMID: 29769541 PMCID: PMC5955903 DOI: 10.1038/s41598-018-25737-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 04/26/2018] [Indexed: 01/18/2023] Open
Abstract
Traumatic brain injury is a major source of global disability and mortality. Preclinical TBI models are a crucial component of therapeutic investigation. We report a tunable, monitored model of murine non-surgical, diffuse closed-head injury—modCHIMERA—characterized by impact as well as linear and rotational acceleration. modCHIMERA is based on the Closed-Head Impact Model of Engineered Rotational Acceleration (CHIMERA) platform. We tested this model at 2 energy levels: 1.7 and 2.1 Joules—substantially higher than previously reported for this system. Kinematic analysis demonstrated linear acceleration exceeding injury thresholds in humans, although outcome metrics tracked impact energy more closely than kinematic parameters. Acute severity metrics were consistent with a complicated-mild or moderate TBI, a clinical population characterized by high morbidity but potentially reversible pathology. Axonal injury was multifocal and bilateral, neuronal death was detected in the hippocampus, and microglial neuroinflammation was prominent. Acute functional analysis revealed prolonged post-injury unconsciousness, and decreased spontaneous behavior and stimulated neurological scores. Neurobehavioral deficits were demonstrated in spatial learning/memory and socialization at 1-month. The overall injury profile of modCHIMERA corresponds with the range responsible for a substantial portion of TBI-related disability in humans. modCHIMERA should provide a reliable platform for efficient analysis of TBI pathophysiology and testing of treatment modalities.
Collapse
|
18
|
Mo Y, Huang L, Chen L, Veronesi M, Shi Y, Chen S, Peng L, Zhou L, Pu Y, Wang J. An experimental rabbit model of symptomatic cerebral vasospasm with in vivo neuroimaging assessment and ex vivo histological validation. Exp Ther Med 2018; 15:2411-2417. [PMID: 29456646 PMCID: PMC5795581 DOI: 10.3892/etm.2018.5690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 07/27/2017] [Indexed: 12/22/2022] Open
Abstract
Cerebral vasospasm (CVS) is a severe complication that occurs following aneurysmal subarachnoid hemorrhage (SAH). Magnetic resonance angiography (MRA) has been used to evaluate brain injury following SAH in humans. The present study was designed to assess a rabbit model of symptomatic CVS (SCVS) and the utility of MRA in evaluating SCVS in rabbits. Japanese white rabbits (n=24) were randomly divided into 2 equal groups: A sham group and a SAH group. Neurological scores were evaluated for 7 days following SAH. Basilar artery (BA) diameters were measured using MRA preoperatively and 7 days postoperatively. Rabbits were sacrificed 7 days following SAH and the BA diameter of each rabbit was determined using histological evaluation. Compared with the Sham group, neurological function was significantly reduced in the SAH group at all time points (P<0.05). Furthermore, the BA diameter was significantly smaller in the SAH group on day 7 compared with the baseline measurement (P<0.05). No significant difference was observed between histological and MRA findings in either group at day 7. Histological changes in the hippocampus consistent with ischemia were observed in the SAH group. Hippocampal ischemia was also identified in the SAH group via MRA and there was no difference in detection rates following the use of MRA and histochemistry. MRA appears to be an effective method for assessing vasospasms of the BA and ischemic changes to the hippocampus in a rabbit model of SCVS. Furthermore, the animal model used in the present study may be beneficial for the future study of SCVS.
Collapse
Affiliation(s)
- Yunchang Mo
- Department of Anesthesia, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Luping Huang
- Department of Anesthesia, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Linbi Chen
- Department of Anesthesia, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Michael Veronesi
- Department of Radiology, University of Chicago Hospital, Chicago, IL 60637, USA
| | - Yiyi Shi
- Department of Anesthesia, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Sijia Chen
- Department of Anesthesia, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Linli Peng
- Department of Anesthesia, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China.,Department of Anesthesia, Guangdong Provincial Maternity and Child Care Center, Guangzhou, Guangdong 510010, P.R. China
| | - Leping Zhou
- Department of Anesthesia, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China.,Department of Anesthesia, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Yonglin Pu
- Department of Radiology, University of Chicago Hospital, Chicago, IL 60637, USA
| | - Junlu Wang
- Department of Anesthesia, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
19
|
Fanizzi C, Sauerbeck AD, Gangolli M, Zipfel GJ, Brody DL, Kummer TT. Minimal Long-Term Neurobehavioral Impairments after Endovascular Perforation Subarachnoid Hemorrhage in Mice. Sci Rep 2017; 7:7569. [PMID: 28790425 PMCID: PMC5548778 DOI: 10.1038/s41598-017-07701-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/03/2017] [Indexed: 02/06/2023] Open
Abstract
Cognitive deficits are among the most severe and pervasive consequences of aneurysmal subarachnoid hemorrhage (SAH). A critical step in developing therapies targeting such outcomes is the characterization of experimentally-tractable pre-clinical models that exhibit multi-domain neurobehavioral deficits similar to those afflicting humans. We therefore searched for neurobehavioral abnormalities following endovascular perforation induction of SAH in mice, a heavily-utilized model. We instituted a functional screen to manage variability in injury severity, then assessed acute functional deficits, as well as activity, anxiety-related behavior, learning and memory, socialization, and depressive-like behavior at sub-acute and chronic time points (up to 1 month post-injury). Animals in which SAH was induced exhibited reduced acute functional capacity and reduced general activity to 1 month post-injury. Tests of anxiety-related behavior including central area time in the elevated plus maze and thigmotaxis in the open field test revealed increased anxiety-like behavior at subacute and chronic time-points, respectively. Effect sizes for subacute and chronic neurobehavioral endpoints in other domains, however, were small. In combination with persistent variability, this led to non-significant effects of injury on all remaining neurobehavioral outcomes. These results suggest that, with the exception of anxiety-related behavior, alternate mouse models are required to effectively analyze cognitive outcomes after SAH.
Collapse
Affiliation(s)
- Claudia Fanizzi
- Department of Neurology, Washington University School of Medicine in St. Louis, Missouri, USA
- Department of Neurosurgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrew D Sauerbeck
- Department of Neurology, Washington University School of Medicine in St. Louis, Missouri, USA
| | - Mihika Gangolli
- Department of Neurology, Washington University School of Medicine in St. Louis, Missouri, USA
| | - Gregory J Zipfel
- Department of Neurology, Washington University School of Medicine in St. Louis, Missouri, USA
- Department of Neurosurgery, Washington University School of Medicine in St. Louis, Missouri, USA
| | - David L Brody
- Department of Neurology, Washington University School of Medicine in St. Louis, Missouri, USA
| | - Terrance T Kummer
- Department of Neurology, Washington University School of Medicine in St. Louis, Missouri, USA.
| |
Collapse
|
20
|
Flynn LMC, Begg CJ, Macleod MR, Andrews PJD. Alpha Calcitonin Gene-Related Peptide Increases Cerebral Vessel Diameter in Animal Models of Subarachnoid Hemorrhage: A Systematic Review and Meta-analysis. Front Neurol 2017; 8:357. [PMID: 28790969 PMCID: PMC5524781 DOI: 10.3389/fneur.2017.00357] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/06/2017] [Indexed: 12/02/2022] Open
Abstract
Delayed cerebral ischemia (DCI) is a life-threatening complication after subarachnoid hemorrhage. There is a strong association between cerebral vessel narrowing and DCI. Alpha calcitonin gene-related peptide (αCGRP) is a potent vasodilator, which may be effective at reducing cerebral vessel narrowing after subarachnoid hemorrhage (SAH). Here, we report a meta-analysis of data from nine in vivo animal studies identified in a systematic review in which αCGRP was administered in SAH models. Our primary outcome was change in cerebral vessel diameter and the secondary outcome was change in neurobehavioral scores. There was a 40.8 ± 8.2% increase in cerebral vessel diameter in those animals treated with αCGRP compared with controls (p < 0.0005, 95% CI 23.7–57.9). Neurobehavioral scores were reported in four publications and showed a standardized mean difference of 1.31 in favor of αCGRP (CI −0.49 to 3.12). We conclude that αCGRP reduces cerebral vessel narrowing seen after SAH in animal studies but note that there is insufficient evidence to determine its effect on functional outcomes.
Collapse
Affiliation(s)
- Liam M C Flynn
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Caroline J Begg
- Emergency Department, Edinburgh Royal Infirmary, Edinburgh, United Kingdom
| | - Malcolm R Macleod
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter J D Andrews
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
21
|
Kawakita F, Fujimoto M, Liu L, Nakano F, Nakatsuka Y, Suzuki H. Effects of Toll-Like Receptor 4 Antagonists Against Cerebral Vasospasm After Experimental Subarachnoid Hemorrhage in Mice. Mol Neurobiol 2016; 54:6624-6633. [PMID: 27738873 DOI: 10.1007/s12035-016-0178-7] [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: 07/16/2016] [Accepted: 09/28/2016] [Indexed: 02/05/2023]
Abstract
Toll-like receptor 4 (TLR4) signaling may play a crucial role in the occurrence of cerebral vasospasm after subarachnoid hemorrhage (SAH). The main purpose of this study was to assess if selective blockage of TLR4 on cerebral arteries prevents cerebral vasospasm development and neurological impairments after SAH in mice. One hundred fourteen mice underwent endovascular perforation SAH or sham operation and were randomly divided into the following 6 groups: sham+vehicle, sham+LPS-RS ultrapure 8 μg, sham+LPS-RS ultrapure 40 μg, SAH+vehicle, SAH+LPS-RS ultrapure 8 μg, and SAH+LPS-RS ultrapure 40 μg. A selective TLR4 antagonist, LPS-RS ultrapure (8 or 40 μg), was administered intracerebroventricularly to mice at 30 min, and the effects were evaluated by neurobehavioral tests and India-ink angiography at 24-48 h, and Western blotting and immunohistochemistry on cerebral arteries at 24 h post-SAH. Higher but not lower dosages of LPS-RS ultrapure significantly prevented post-SAH neurological impairments and ameliorated cerebral vasospasm. SAH caused TLR4 activation and cyclooxygenase-1 (COX1) upregulation in the endothelial cells and smooth muscle cells of spastic cerebral arteries, both of which were significantly suppressed by LPS-RS ultrapure. Another selective TLR4 antagonist, IAXO-102, which has a different binding site from LPS-RS ultrapure, also showed similar protective effects to LPS-RS ultrapure. These findings suggest that TLR4 signaling is implicated in cerebral vasospasm development at least partly via COX1 upregulation, and that TLR4 antagonists have therapeutic potential as a new therapy against cerebral vasospasm.
Collapse
Affiliation(s)
- Fumihiro Kawakita
- Department of Neurosurgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Masashi Fujimoto
- Department of Neurosurgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Lei Liu
- Department of Neurosurgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Fumi Nakano
- Department of Neurosurgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Yoshinari Nakatsuka
- Department of Neurosurgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Hidenori Suzuki
- Department of Neurosurgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
| |
Collapse
|
22
|
Konczalla J, Brawanski N, Platz J, Senft C, Kashefiolasl S, Seifert V. Aneurysm Location as a Prognostic Outcome Factor After Subarachnoid Hemorrhage From Internal Carotid Artery Aneurysms and Potential Impact for Further Experimental Subarachnoid Hemorrhage Models. World Neurosurg 2016; 92:273-278. [DOI: 10.1016/j.wneu.2016.04.086] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 04/22/2016] [Accepted: 04/25/2016] [Indexed: 11/28/2022]
|
23
|
Backer-Grøndahl A, Lindal S, Lorentzen MA, Eldevik P, Vorren T, Kristiansen B, Vangberg T, Ytrebø LM. A new non-craniotomy model of subarachnoid hemorrhage in the pig: a pilot study. Lab Anim 2015; 50:379-89. [PMID: 26643281 DOI: 10.1177/0023677215619806] [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] [Indexed: 11/16/2022]
Abstract
Subarachnoid hemorrhage (SAH) from rupture of an intracranial arterial aneurysm is a devastating disease affecting young people, with serious lifelong disability or death as a frequent outcome. Large animal models that exhibit all the cardinal clinical features of human SAH are highly warranted. In this pilot study we aimed to develop a non-craniotomy model of SAH in pigs suitable for acute intervention studies. Six Norwegian Landrace pigs received advanced invasive hemodynamic and intracranial pressure (ICP) monitoring. The subarachnoid space, confirmed by a clear cerebrospinal fluid (CSF) tap, was reached by advancing a needle below the ocular bulb through the superior orbital fissure and into the interpeduncular cistern. SAH was induced by injecting 15 mL of autologous arterial blood into the subarachnoid space. Macro- and microanatomical investigations of the pig brain showed a typical blood distribution consistent with human aneurysmal SAH (aSAH) autopsy data. Immediately after SAH induction ICP sharply increased with a concomitant reduction in cerebral perfusion pressure (CPP). ICP returned to near normal values after 30 min, but increased subsequently during the experimental period. Signs of brain edema were confirmed by light microscopy post-mortem. None of the animals died during the experimental period. This new transorbital injection model of SAH in the pig mimics human aSAH and may be suitable for acute intervention studies. However, the model is technically challenging and needs further validation.
Collapse
Affiliation(s)
- Anders Backer-Grøndahl
- Department of Anesthesiology, University Hospital of North Norway, Tromsø, Norway Institute of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Sigurd Lindal
- Institute of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway Department of Pathology, University Hospital of North Norway, Tromsø, Norway
| | | | - Petter Eldevik
- Institute of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway Department of Radiology, University Hospital of North Norway, Tromsø, Norway
| | - Torgrim Vorren
- Department of Radiology, University Hospital of North Norway, Tromsø, Norway
| | - Bente Kristiansen
- Department of Radiology, University Hospital of North Norway, Tromsø, Norway
| | - Torgil Vangberg
- Institute of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway Department of Radiology, University Hospital of North Norway, Tromsø, Norway
| | - Lars Marius Ytrebø
- Department of Anesthesiology, University Hospital of North Norway, Tromsø, Norway Institute of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
24
|
Schneider UC, Davids AM, Brandenburg S, Müller A, Elke A, Magrini S, Atangana E, Turkowski K, Finger T, Gutenberg A, Gehlhaar C, Brück W, Heppner FL, Vajkoczy P. Microglia inflict delayed brain injury after subarachnoid hemorrhage. Acta Neuropathol 2015; 130:215-31. [PMID: 25956409 DOI: 10.1007/s00401-015-1440-1] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 04/17/2015] [Accepted: 05/01/2015] [Indexed: 01/08/2023]
Abstract
Inflammatory changes have been postulated to contribute to secondary brain injury after aneurysmal subarachnoid hemorrhage (SAH). In human specimens after SAH as well as in experimental SAH using mice, we show an intracerebral accumulation of inflammatory cells between days 4 and 28 after the bleeding. Using bone marrow chimeric mice allowing tracing of all peripherally derived immune cells, we confirm a truly CNS-intrinsic, microglial origin of these immune cells, exhibiting an inflammatory state, and rule out invasion of myeloid cells from the periphery into the brain. Furthermore, we detect secondary neuro-axonal injury throughout the time course of SAH. Since neuronal cell death and microglia accumulation follow a similar time course, we addressed whether the occurrence of activated microglia and neuro-axonal injury upon SAH are causally linked by depleting microglia in vivo. Given that the amount of neuronal cell death was significantly reduced after microglia depletion, we conclude that microglia accumulation inflicts secondary brain injury after SAH.
Collapse
Affiliation(s)
- Ulf C Schneider
- Department of Neurosurgery, Charité, Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Nikitina T, Zavaritskaya O, Semenyutin V, Persson PB, Patzak A, Sendeski M. Effects of iodinated contrast media in a novel model for cerebral vasospasm. ARQUIVOS DE NEURO-PSIQUIATRIA 2015; 73:125-31. [PMID: 25742582 DOI: 10.1590/0004-282x20140222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 11/10/2014] [Indexed: 11/22/2022]
Abstract
OBJECTIVE We developed an in vitro model for vasospasm post subarachnoid hemorrhage that was suitable for investigating brain vessel autoregulation. We further investigated the effects of iodinated contrast medium on the vascular tone and the myogenic response of spastic cerebral vessels. METHOD We isolated and perfused the superior cerebellar arteries of rats. The vessels were pressurized and studied under isobaric conditions. Coagulated blood was used to simulate subarachnoid hemorrhage. The contrast medium iodixanol was applied intraluminally. RESULTS Vessels exposed to blood developed significantly stronger myogenic tone (65.7 ± 2.0% vs 77.1 ± 1.2% of the maximum diameter, for the blood and the control group, respectively) and significantly decreased myogenic response, compared with the control groups. The contrast medium did not worsen the myogenic tone or the myogenic response in any group. CONCLUSION Our results show that deranged myogenic response may contribute to cerebral blood flow disturbances subsequent to subarachnoid hemorrhage. The contrast medium did not have any negative influence on vessel tone or myogenic response in this experimental setting.
Collapse
Affiliation(s)
- Tatiana Nikitina
- Charité-Universitaetsmedizin Berlin, Institut fuer Vegetative Physiologie, Berlin, Germany
| | - Olga Zavaritskaya
- Medical Faculty Mannheim, Research Division Cardiovascular Physiology, Mannheim, Germany
| | - Vladimir Semenyutin
- Russian Polenov Neurosurgical Institute, Laboratory of Brain Circulation Pathology, Saint-Petersburg, Russia
| | - Pontus B Persson
- Charité-Universitaetsmedizin Berlin, Institut fuer Vegetative Physiologie, Berlin, Germany
| | - Andreas Patzak
- Charité-Universitaetsmedizin Berlin, Institut fuer Vegetative Physiologie, Berlin, Germany
| | - Mauricio Sendeski
- Charité-Universitaetsmedizin Berlin, Institut fuer Vegetative Physiologie, Berlin, Germany
| |
Collapse
|
26
|
Shishido H, Egashira Y, Okubo S, Zhang H, Hua Y, Keep RF, Xi G. A magnetic resonance imaging grading system for subarachnoid hemorrhage severity in a rat model. J Neurosci Methods 2015; 243:115-9. [PMID: 25677406 DOI: 10.1016/j.jneumeth.2015.01.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/29/2015] [Accepted: 01/30/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND The endovascular perforation model of subarachnoid hemorrhage (SAH) has a large variation in outcomes. This may reflect differences in the SAH size. NEW METHOD Magnetic resonance imaging (MRI) was performed 24h after endovascular perforation in adult male (n=58) and female (n=58) rats. Rats were divided into five grades according to MRI characteristics: grade 0: no SAH or intraventricular hemorrhage (IVH); grade 1: minimal or thin SAH without IVH; grade 2: minimal or thin SAH with IVH; grade 3: thick SAH without IVH; grade 4: thick SAH with IVH. We investigated whether MRI grading scale reflected severity of SAH (determined post mortem) and neurological score. RESULTS There was a strong correlation between MRI grading scale and current SAH grading scale (P<0.01) and neurological score (P<0.01) in male rats. In female rats, there was also a strong correlation between MRI grading scale and SAH grading scale (P<0.01) but not with neurological score (P=0.24). COMPARISON WITH EXISTING METHODS The current grading system is based on the amount of SAH and needs animal euthanasia to evaluate SAH severity. There is no useful grading system to classify severity of SAH without decapitating animals. CONCLUSIONS We demonstrated a correlation between the MRI grading scale and the current SAH grading scale in an endovascular perforation rat model. The MRI grading scale allows evaluation of SAH severity without euthanizing animals.
Collapse
Affiliation(s)
- Hajime Shishido
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Yusuke Egashira
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Shuichi Okubo
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Haining Zhang
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
27
|
Milner E, Holtzman JC, Friess S, Hartman RE, Brody DL, Han BH, Zipfel GJ. Endovascular perforation subarachnoid hemorrhage fails to cause Morris water maze deficits in the mouse. J Cereb Blood Flow Metab 2014; 34:jcbfm2014108. [PMID: 24938403 PMCID: PMC4158664 DOI: 10.1038/jcbfm.2014.108] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 05/08/2014] [Accepted: 05/23/2014] [Indexed: 11/09/2022]
Abstract
Cognitive dysfunction is the primary driver of poor long-term outcome in aneurysmal subarachnoid hemorrhage (SAH) survivors; modeling such deficits preclinically is thus key for mechanistic and translational investigation. Although rat SAH causes long-term deficits in learning and memory, it remains unknown whether similar deficits are seen in the mouse, a species particularly amenable to powerful, targeted genetic manipulation. We thus subjected mice to endovascular perforation SAH and assessed long-term cognitive outcome via the Morris water maze (MWM), the most commonly used metric for rodent neurocognition. No significant differences in MWM performance (by either of two protocols) were seen in SAH versus sham mice. Moreover, SAH caused negligible hippocampal CA1 injury. These results undercut the potential of commonly used methods (of SAH induction and assessment of long-term neurocognitive outcome) for use in targeted molecular studies of SAH-induced cognitive deficits in the mouse.
Collapse
Affiliation(s)
- Eric Milner
- 1] Department of Neurological Surgery, Washington University School of Medicine, St Louis, Missouri, USA [2] Program in Neuroscience, Washington University School of Medicine, St Louis, Missouri, USA
| | - Jacob C Holtzman
- Department of Neurological Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| | - Stuart Friess
- Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, USA
| | - Richard E Hartman
- Department of Psychology, Loma Linda University, Loma Linda, California, USA
| | - David L Brody
- 1] Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA [2] Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, USA
| | - Byung H Han
- 1] Department of Neurological Surgery, Washington University School of Medicine, St Louis, Missouri, USA [2] Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, USA
| | - Gregory J Zipfel
- 1] Department of Neurological Surgery, Washington University School of Medicine, St Louis, Missouri, USA [2] Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA [3] Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
28
|
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: 3.6] [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
|
29
|
Mori K. Double cisterna magna blood injection model of experimental subarachnoid hemorrhage in dogs. Transl Stroke Res 2014; 5:647-52. [PMID: 24986149 DOI: 10.1007/s12975-014-0356-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 06/12/2014] [Accepted: 06/24/2014] [Indexed: 11/24/2022]
Abstract
Several animal subarachnoid hemorrhage (SAH) models have been proposed to study the etiology and treatment for cerebral vasospasm. We describe the experimental procedures of a canine double-hemorrhage model of SAH and discuss the pathophysiological parameters and occurrence of angiographic delayed cerebral vasospasm using magnetic resonance (MR) imaging and digital subtraction angiography. Autologous blood was injected twice on days 1 and 3 into the cerebellomedullary cistern of 36 female beagles. All animals showed delayed angiographic vasospasm in the vertebrobasilar arteries on day 7. The degree of vasospasm was 29-42 % of the arterial diameter. However, this model showed no symptomatic vasospasm or ischemic changes detected by MR imaging. This animal model can produce reproducible delayed vasospasm without detectable cerebral infarction on MR imaging. This model allows evaluation of the effect of treatment on delayed vasospasm in the same animals. The canine double-hemorrhage model of SAH is suitable for the quantitative and chronological study of delayed angiographic vasospasm, but not for investigating early brain injury and delayed cerebral ischemia.
Collapse
Affiliation(s)
- Kentaro Mori
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan,
| |
Collapse
|
30
|
Chen S, Feng H, Sherchan P, Klebe D, Zhao G, Sun X, Zhang J, Tang J, Zhang JH. Controversies and evolving new mechanisms in subarachnoid hemorrhage. Prog Neurobiol 2014; 115:64-91. [PMID: 24076160 PMCID: PMC3961493 DOI: 10.1016/j.pneurobio.2013.09.002] [Citation(s) in RCA: 270] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 09/07/2013] [Accepted: 09/12/2013] [Indexed: 12/13/2022]
Abstract
Despite decades of study, subarachnoid hemorrhage (SAH) continues to be a serious and significant health problem in the United States and worldwide. The mechanisms contributing to brain injury after SAH remain unclear. Traditionally, most in vivo research has heavily emphasized the basic mechanisms of SAH over the pathophysiological or morphological changes of delayed cerebral vasospasm after SAH. Unfortunately, the results of clinical trials based on this premise have mostly been disappointing, implicating some other pathophysiological factors, independent of vasospasm, as contributors to poor clinical outcomes. Delayed cerebral vasospasm is no longer the only culprit. In this review, we summarize recent data from both experimental and clinical studies of SAH and discuss the vast array of physiological dysfunctions following SAH that ultimately lead to cell death. Based on the progress in neurobiological understanding of SAH, the terms "early brain injury" and "delayed brain injury" are used according to the temporal progression of SAH-induced brain injury. Additionally, a new concept of the vasculo-neuronal-glia triad model for SAH study is highlighted and presents the challenges and opportunities of this model for future SAH applications.
Collapse
Affiliation(s)
- Sheng Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Prativa Sherchan
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Damon Klebe
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Gang Zhao
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi, China
| | - Xiaochuan Sun
- Department of Neurosurgery, First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiping Tang
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, USA; Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.
| |
Collapse
|
31
|
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: 9.3] [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]
|
32
|
Shiba M, Fujimoto M, Imanaka-Yoshida K, Yoshida T, Taki W, Suzuki H. Tenascin-C causes neuronal apoptosis after subarachnoid hemorrhage in rats. Transl Stroke Res 2014; 5:238-47. [PMID: 24481545 DOI: 10.1007/s12975-014-0333-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Revised: 01/15/2014] [Accepted: 01/16/2014] [Indexed: 10/25/2022]
Abstract
The role of tenascin-C (TNC), a matricellular protein, in brain injury is unknown. The aim of this study was to examine if TNC causes neuronal apoptosis after subarachnoid hemorrhage (SAH), a deadly cerebrovascular disorder, using imatinib mesylate (a selective inhibitor of platelet-derived growth factor receptor [PDGFR] that is reported to suppress TNC induction) and recombinant TNC. SAH by endovascular perforation caused caspase-dependent neuronal apoptosis in the cerebral cortex irrespective of cerebral vasospasm development at 24 and 72 h post-SAH, associated with PDGFR activation, mitogen-activated protein kinases (MAPKs) activation, and TNC induction in rats. PDGFR inactivation by an intraperitoneal injection of imatinib mesylate prevented neuronal apoptosis, as well as MAPKs activation and TNC induction in the cerebral cortex at 24 h. A cisternal injection of recombinant TNC reactivated MAPKs and abolished anti-apoptotic effects of imatinib mesylate. The TNC injection also induced TNC itself in SAH brain, which may internally augment neuronal apoptosis after SAH. These findings suggest that TNC upregulation by PDGFR activation causes neuronal apoptosis via MAPK activation, and that the positive feedback mechanisms may exist to augment neuronal apoptosis after SAH. TNC-induced neuronal apoptosis would be a new target to improve outcome after SAH.
Collapse
Affiliation(s)
- Masato Shiba
- Department of Neurosurgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | | | | | | | | | | |
Collapse
|
33
|
Majid A. Neuroprotection in stroke: past, present, and future. ISRN NEUROLOGY 2014; 2014:515716. [PMID: 24579051 PMCID: PMC3918861 DOI: 10.1155/2014/515716] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Accepted: 09/16/2013] [Indexed: 01/05/2023]
Abstract
Stroke is a devastating medical condition, killing millions of people each year and causing serious injury to many more. Despite advances in treatment, there is still little that can be done to prevent stroke-related brain damage. The concept of neuroprotection is a source of considerable interest in the search for novel therapies that have the potential to preserve brain tissue and improve overall outcome. Key points of intervention have been identified in many of the processes that are the source of damage to the brain after stroke, and numerous treatment strategies designed to exploit them have been developed. In this review, potential targets of neuroprotection in stroke are discussed, as well as the various treatments that have been targeted against them. In addition, a summary of recent progress in clinical trials of neuroprotective agents in stroke is provided.
Collapse
Affiliation(s)
- Arshad Majid
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
- Department of Neurology and Manchester Academic Health Sciences Centre, Salford Royal Hospital, Stott Lane, Salford M6 8HD, UK
| |
Collapse
|
34
|
Ostrowski RP, Zhang JH. Hyperbaric oxygen for cerebral vasospasm and brain injury following subarachnoid hemorrhage. Transl Stroke Res 2013; 2:316-27. [PMID: 23060945 DOI: 10.1007/s12975-011-0069-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The impact of acute brain injury and delayed neurological deficits due to cerebral vasospasm (CVS) are major determinants of outcomes after subarachnoid hemorrhage (SAH). Although hyperbaric oxygen (HBO) had been used to treat patients with SAH, the supporting evidence and underlying mechanisms have not been systematically reviewed. In the present paper, the overview of studies of HBO for cerebral vasospasm is followed by a discussion of HBO molecular mechanisms involved in the protection against SAH-induced brain injury and even, as hypothesized, in attenuating vascular spasm alone. Faced with the paucity of information as to what degree HBO is capable of antagonizing vasospasm after SAH, the authors postulate that the major beneficial effects of HBO in SAH include a reduction of acute brain injury and combating brain damage caused by CVS. Consequently, authors reviewed the effects of HBO on SAH-induced hypoxic signaling and other mechanisms of neurovascular injury. Moreover, authors hypothesize that HBO administered after SAH may "precondition" the brain against the detrimental sequelae of vasospasm. In conclusion, the existing evidence speaks in favor of administering HBO in both acute and delayed phase after SAH; however, further studies are needed to understand the underlying mechanisms and to establish the optimal regimen of treatment.
Collapse
Affiliation(s)
- Robert P Ostrowski
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus Street, Loma Linda, CA 92350, USA
| | | |
Collapse
|
35
|
Güresir E, Vasiliadis N, Konczalla J, Raab P, Hattingen E, Seifert V, Vatter H. Erythropoietin prevents delayed hemodynamic dysfunction after subarachnoid hemorrhage in a randomized controlled experimental setting. J Neurol Sci 2013; 332:128-35. [DOI: 10.1016/j.jns.2013.07.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 06/30/2013] [Accepted: 07/08/2013] [Indexed: 10/26/2022]
|
36
|
Agner C, Dujovny M. Cerebral arterial aneurysms: the early period of neuroendovascular therapy. Neurol Res 2013; 31:560-7. [DOI: 10.1179/174313209x455781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
37
|
Aneurysmal subarachnoid hemorrhage models: do they need a fix? Stroke Res Treat 2013; 2013:615154. [PMID: 23878760 PMCID: PMC3710594 DOI: 10.1155/2013/615154] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 05/31/2013] [Accepted: 06/04/2013] [Indexed: 11/17/2022] Open
Abstract
The discovery of tissue plasminogen activator to treat acute stroke is a success story of research on preventing brain injury following transient cerebral ischemia (TGI). That this discovery depended upon development of embolic animal model reiterates that proper stroke modeling is the key to develop new treatments. In contrast to TGI, despite extensive research, prevention or treatment of brain injury following aneurysmal subarachnoid hemorrhage (aSAH) has not been achieved. A lack of adequate aSAH disease model may have contributed to this failure. TGI is an important component of aSAH and shares mechanism of injury with it. We hypothesized that modifying aSAH model using experience acquired from TGI modeling may facilitate development of treatment for aSAH and its complications. This review focuses on similarities and dissimilarities between TGI and aSAH, discusses the existing TGI and aSAH animal models, and presents a modified aSAH model which effectively mimics the disease and has a potential of becoming a better resource for studying the brain injury mechanisms and developing a treatment.
Collapse
|
38
|
Pathophysiological Role of Global Cerebral Ischemia following Subarachnoid Hemorrhage: The Current Experimental Evidence. Stroke Res Treat 2013; 2013:651958. [PMID: 23844316 PMCID: PMC3694494 DOI: 10.1155/2013/651958] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 04/29/2013] [Indexed: 11/24/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is the subtype of stroke with one of the highest mortality rates and the least well-understood pathophysiologies. One of the very early events which may occur after SAH is a significant decrease of cerebral perfusion pressure (CPP) caused by the excessive increase of intracranial pressure during the initial bleeding. A severely decreased CPP results in global cerebral ischemia, an event also occurring after cardiac arrest. The aim of the current paper is to review the pathophysiological events occurring in experimental models of SAH and global cerebral ischemia and to evaluate the contribution and the importance of global cerebral ischemia for the pathophysiology of SAH.
Collapse
|
39
|
Acute microvascular changes after subarachnoid hemorrhage and transient global cerebral ischemia. Stroke Res Treat 2013; 2013:425281. [PMID: 23589781 PMCID: PMC3621372 DOI: 10.1155/2013/425281] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 02/26/2013] [Accepted: 02/28/2013] [Indexed: 01/27/2023] Open
Abstract
Subarachnoid hemorrhage and transient global cerebral ischemia result in similar pathophysiological changes in the cerebral microcirculation. These changes include microvascular constriction, increased leukocyte-endothelial interactions, blood brain barrier disruption, and microthrombus formation. This paper will look at various animal and preclinical studies that investigate these various microvascular changes, perhaps providing insight in how these microvessels can be a therapeutic target in both subarachnoid hemorrhage and transient global cerebral ischemia.
Collapse
|
40
|
Song JN, Chen H, Zhang M, Zhao YL, Ma XD. Dynamic change in cerebral microcirculation and focal cerebral metabolism in experimental subarachnoid hemorrhage in rabbits. Metab Brain Dis 2013; 28:33-43. [PMID: 23232626 DOI: 10.1007/s11011-012-9369-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Accepted: 11/26/2012] [Indexed: 10/27/2022]
Abstract
Regional cerebral blood flow (rCBF) in the cerebral metabolism and energy metabolism measurements can be used to assess blood flow of brain cells and to detect cell activity. Changes of rCBF in the cerebral microcirculation and energy metabolism were determined in an experimental model of subarachnoid hemorrhage (SAH) model in 56 large-eared Japanese rabbits about 12 to 16-month old. Laser Doppler flowmetry was used to detect the blood supply to brain cells. Internal carotid artery and vein blood samples were used for duplicate blood gas analysis to assess the energy metabolism of brain cells. Cerebral blood flow (CBF) was detected by single photon emission computed tomography (SPECT) perfusion imaging using Tc-99m ethyl cysteinate dimer (Tc-99m ECD) as an imaging reagent. The percentage of injected dose per gram of brain tissue was calculated and analyzed. There were positive correlations between the percentage of radionuclide injected per gram of brain tissue and rCBF supply and cerebral metabolic rate for oxygen (P < 0.05). However, there was a negative correlation between radioactivity counts per unit volume detected on the SPECT rheoencephalogram and lactic acid concentration in the homolateral internal carotid artery and vein. In summary, this study found abnormal CBF in metabolism and utilization of brain cells after SAH, and also found that deterioration of energy metabolism of brain cells played a significant role in the development of SAH. There are matched reductions in CBF and metabolism. Thus, SPECT imaging could be used as a noninvasive method to detect CBF.
Collapse
Affiliation(s)
- Jin-Ning Song
- First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | | | | | | | | |
Collapse
|
41
|
Relevance of animal models of subarachnoid hemorrhage for examining neurobehavioral changes. ACTA NEUROCHIRURGICA. SUPPLEMENT 2013; 115:225-32. [PMID: 22890673 DOI: 10.1007/978-3-7091-1192-5_41] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND PURPOSE For many years survival and neurological functionality of patients were the main outcome measures after treatment of intracranial aneurysms. But, the variable outcomes of patients operated on in a delayed fashion or before the aneurysm rupture indicate that more precise measures are needed for assessment of not only the neurological but also the neuropsychological outcome. However, development and testing of such new tools requires better understanding of pathomechanisms of neurobehavioral changes evoked by aneurysmal subarachnoid hemorrhage (aSAH), which can be achieved using animal models. METHODS We reviewed and selected (1) animal models developed to investigate delayed cerebral vasospasm that could be useful for examining effects of brain injury evoked by aSAH and (2) a battery of neurobehavioral animal testing that can be used for assessment of patients after aSAH. RESULTS For every species used as an aSAH model, a battery of neurobehavioral test exists. CONCLUSION Albeit some limitations must be recognized, research using animal models of SAH should continue to play a critical role in assessment of cognitive and behavioral functions after aSAH.
Collapse
|
42
|
Zoerle T, Ilodigwe D, Wan H, Lakovic K, Sabri M, Ai J, Macdonald RL. Pharmacologic reduction of angiographic vasospasm in experimental subarachnoid hemorrhage: systematic review. ACTA NEUROCHIRURGICA. SUPPLEMENT 2013; 115:247-51. [PMID: 22890676 DOI: 10.1007/978-3-7091-1192-5_44] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Animal models have been developed to simulate angiographic vasospasm secondary to subarachnoid hemorrhage (SAH) and to test pharmacologic treatments. Our aim was to evaluate the effect of pharmacologic treatments that have been tested in humans and in preclinical studies to determine if animal models inform results reported in humans. A systematic review and meta-analysis of SAH studies was performed. We investigated predictors of -translation from animals to humans with multivariate logistic regression. Pharmacologic reduction of vasospasm was effective in mice, rats, rabbits, dogs, nonhuman primates, and humans. Animal studies were generally of poor methodologic quality, and there was evidence of publication bias. Fresh blood injection to simulate SAH (vs. clot placement) and evaluation of vasospasm more than 3 days after SAH were independently associated with successful translation. We conclude that reduction of vasospasm is effective in animals and humans, and that injection of fresh blood and evaluation of vasospasm more than 3 days after SAH may be preferable for preclinical models.
Collapse
Affiliation(s)
- Tommaso Zoerle
- Department of Anesthesia and Critical Care Medicine, University of Milano, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
43
|
Shiba M, Suzuki H, Fujimoto M, Shimojo N, Imanaka-Yoshida K, Yoshida T, Kanamaru K, Matsushima S, Taki W. Role of platelet-derived growth factor in cerebral vasospasm after subarachnoid hemorrhage in rats. ACTA NEUROCHIRURGICA. SUPPLEMENT 2012; 115:219-23. [PMID: 22890672 DOI: 10.1007/978-3-7091-1192-5_40] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
BACKGROUND AND PURPOSE The role of platelet-derived growth factor (PDGF) remains unknown in cerebral vasospasm after subarachnoid hemorrhage (SAH). In this study, we examined the effects of PDGF receptor (PDGFR) inactivation on cerebral vasospasm in the endovascular perforation model of SAH in rats. METHODS Rats were assigned to sham, SAH plus vehicle, and SAH plus imatinib mesylate (imatinib) groups (n = 4 per group). Imatinib (50 mg/kg body weight), an inhibitor of the tyrosine kinases of PDGFR, or vehicle was administered intraperitoneally 30 min post-SAH. Vasospasm was evaluated in the left (perforation-sided) internal carotid artery by means of neurobehavioral tests, India ink angiography, and immunohistochemistry at 24 h after SAH. RESULTS Imatinib significantly inhibited post-SAH PDGFR activation in the left internal carotid artery, in which vasospasm was significantly prevented. Animal's neurobehavior also showed a tendency to improve by imatinib treatment. CONCLUSIONS PDGF may play an important role in the pathogenesis of vasospasm after SAH.
Collapse
Affiliation(s)
- Masato Shiba
- Department of Neurosurgery, Mie University Graduate School of Medicine, Mie, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Greenhalgh AD, Brough D, Robinson EM, Girard S, Rothwell NJ, Allan SM. Interleukin-1 receptor antagonist is beneficial after subarachnoid haemorrhage in rat by blocking haem-driven inflammatory pathology. Dis Model Mech 2012; 5:823-33. [PMID: 22679224 PMCID: PMC3484865 DOI: 10.1242/dmm.008557] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 04/23/2012] [Indexed: 12/19/2022] Open
Abstract
Subarachnoid haemorrhage (SAH) is a major contributor to the burden of stroke on society. Treatment options are limited and animal models of SAH do not always mimic key pathophysiological hallmarks of the disease, thus hindering development of new therapeutics. Inflammation is strongly associated with brain injury after SAH in animals and patients, and inhibition of the pro-inflammatory cytokine interleukin-1 (IL-1) represents a possible therapeutic target. Here we report that a rupture of the middle cerebral artery in the rat produces heterogeneous infarct patterns similar to those observed in human SAH. Administration of the IL-1 receptor antagonist (IL-1Ra) reduced blood-brain barrier breakdown, and the extent of breakdown correlated with brain injury. After SAH, haem oxygenase-1 (HO-1) was strongly expressed around the bleed site and in the cortex and striatum, indicating the presence of free haem, a breakdown product of haemoglobin. HO-1 expression was also found in the same regions as microglial/macrophage expression of IL-1α. The direct effect of haem on IL-1α expression was confirmed in vitro using organotypic slice culture (OSC). Haem-induced cell death was dependent on IL-1 signalling, with IL-1Ra completely blocking cellular injury. Furthermore, stimulation of mouse primary mixed glial cells with haem induced the release of IL-1α, but not IL-1β. Thus, we suggest that haem, released from lysed red blood cells (RBCs) in the subarachnoid space, acts as a danger-associated molecular pattern (DAMP) driving IL-1-dependent inflammation. These data provide new insights into inflammation after SAH-induced brain injury and suggest IL-1Ra as a candidate therapeutic for the disease.
Collapse
Affiliation(s)
- Andrew D Greenhalgh
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PL, UK.
| | | | | | | | | | | |
Collapse
|
45
|
Caner B, Hou J, Altay O, Fuj M, Zhang JH. Transition of research focus from vasospasm to early brain injury after subarachnoid hemorrhage. J Neurochem 2012; 123 Suppl 2:12-21. [DOI: 10.1111/j.1471-4159.2012.07939.x] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Basak Caner
- Department of Physiology; Loma Linda University, School of Medicine; Loma Linda; California; USA
| | - Jack Hou
- Department of Physiology; Loma Linda University, School of Medicine; Loma Linda; California; USA
| | - Orhan Altay
- Department of Physiology; Loma Linda University, School of Medicine; Loma Linda; California; USA
| | - Mutsumi Fuj
- Department of Physiology; Loma Linda University, School of Medicine; Loma Linda; California; USA
| | | |
Collapse
|
46
|
Krafft PR, Bailey EL, Lekic T, Rolland WB, Altay O, Tang J, Wardlaw JM, Zhang JH, Sudlow CLM. Etiology of stroke and choice of models. Int J Stroke 2012; 7:398-406. [PMID: 22712741 DOI: 10.1111/j.1747-4949.2012.00838.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Animal models of stroke contribute to the development of better stroke prevention and treatment through studies investigating the pathophysiology of different stroke subtypes and by testing promising treatments before trials in humans. There are two broad types of animal models: those in which stroke is induced through artificial means, modeling the consequences of a vascular insult but not the vascular pathology itself; and those in which strokes occur spontaneously. Most animal models of stroke are in rodents due to cost, ethical considerations, availability of standardized neurobehavioral assessments, and ease of physiological monitoring. While there are similarities in cerebrovascular anatomy and pathophysiology between rodents and humans, there are also important differences, including brain size, length and structure of perforating arteries, and gray to white matter ratio, which is substantially lower in humans. The wide range of rodent models of stroke includes models of global and focal ischemia, and of intracerebral and sub-arachnoid hemorrhage. The most widely studied model of spontaneous stroke is the spontaneously hypertensive stroke-prone rat, in which the predominant lesions are small subcortical infarcts resulting from a vascular pathology similar to human cerebral small vessel disease. Important limitations of animal models of stroke - they generally model only certain aspects of the disease and do not reflect the heterogeneity in severity, pathology and comorbidities of human stroke - and key methodological issues (especially the need for adequate sample size, randomization, and blinding in treatment trials) must be carefully considered for the successful translation of pathophysiological concepts and therapeutics from bench to bedside.
Collapse
Affiliation(s)
- Paul R Krafft
- Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Zoerle T, Ilodigwe DC, Wan H, Lakovic K, Sabri M, Ai J, Macdonald RL. Pharmacologic reduction of angiographic vasospasm in experimental subarachnoid hemorrhage: systematic review and meta-analysis. J Cereb Blood Flow Metab 2012; 32:1645-58. [PMID: 22534672 PMCID: PMC3437599 DOI: 10.1038/jcbfm.2012.57] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Animal models have been developed to simulate angiographic vasospasm secondary to subarachnoid hemorrhage (SAH) and to test pharmacologic treatments. Our aim was to evaluate the effect of pharmacologic treatments that have been tested in humans and in preclinical studies to determine if animal models inform results reported in humans. A systematic review and meta-analysis of SAH studies was performed. We investigated predictors of translation from animals to humans with multivariate logistic regression. Pharmacologic reduction of vasospasm was effective in mice, rats, rabbits, dogs, nonhuman primates (standard mean difference of -1.74; 95% confidence interval -2.04 to -1.44) and humans. Animal studies were generally of poor methodologic quality and there was evidence of publication bias. Subgroup analysis by drug and species showed that statins, tissue plasminogen activator, erythropoietin, endothelin receptor antagonists, calcium channel antagonists, fasudil, and tirilazad were effective whereas magnesium was not. Only evaluation of vasospasm >3 days after SAH was independently associated with successful translation. We conclude that reduction of vasospasm is effective in animals and humans and that evaluation of vasospasm >3 days after SAH may be preferable for preclinical models.
Collapse
Affiliation(s)
- Tommaso Zoerle
- Division of Neurosurgery, St Michael's Hospital, Labatt Family Centre of Excellence in Brain Injury and Trauma Research, Keenan Research Centre of the Li Ka Shing Knowledge Institute of St Michael's Hospital, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
48
|
Han BH, Vellimana AK, Zhou ML, Milner E, Zipfel GJ. Phosphodiesterase 5 inhibition attenuates cerebral vasospasm and improves functional recovery after experimental subarachnoid hemorrhage. Neurosurgery 2012; 70:178-86; discussion 186-7. [PMID: 21796010 DOI: 10.1227/neu.0b013e31822ec2b0] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Cerebral vasospasm is an independent predictor of poor outcome after subarachnoid hemorrhage (SAH). The nitric oxide-cyclic guanosine monophosphate (NO-cGMP) vasodilatory pathway is strongly implicated in its pathophysiology. Preliminary studies suggest that phosphodiesterase 5 (PDE5), an enzyme that degrades cGMP, may play a role because the PDE5 inhibitor sildenafil was found to reduce vasospasm after SAH. However, several questions that are critical when considering translational studies remain unanswered. OBJECTIVE To elucidate the mechanism of action of sildenafil against vasospasm and to assess whether sildenafil attenuates SAH-induced neuronal cell death, improves functional outcome after SAH, or causes significant physiological side effects when administered at therapeutically relevant doses. METHODS SAH was induced via endovascular perforation in male C57BL6 mice. Beginning 2 hours later, mice received sildenafil citrate (0.7, 2 or 5 mg/kg orally twice daily) or vehicle. Neurological outcome was assessed daily. Vasospasm was determined on post-SAH day 3. Brain PDE5 expression and activity, cGMP content, neuronal cell death, arterial blood pressure, and intracranial pressure were examined. RESULTS We found that PDE5 activity (but not expression) is increased after SAH, leading to decreased cGMP levels. Sildenafil attenuates this increase in PDE5 activity and restores cGMP levels after SAH. Post-SAH initiation of sildenafil was found to decrease vasospasm and neuronal cell death and markedly improve neurological outcome without causing significant physiological side effects. CONCLUSION Sildenafil, a US Food and Drug Administration-approved drug with a proven track record of safety in humans, is a promising new therapy for vasospasm and neurological deficits after SAH.
Collapse
Affiliation(s)
- Byung Hee Han
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|
49
|
A new rabbit model for the study of early brain injury after subarachnoid hemorrhage. J Neurosci Methods 2012; 208:138-45. [DOI: 10.1016/j.jneumeth.2012.05.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2012] [Revised: 05/07/2012] [Accepted: 05/08/2012] [Indexed: 11/24/2022]
|
50
|
A novel intravital method to evaluate cerebral vasospasm in rat models of subarachnoid hemorrhage: a study with synchrotron radiation angiography. PLoS One 2012; 7:e33366. [PMID: 22428033 PMCID: PMC3299776 DOI: 10.1371/journal.pone.0033366] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 02/13/2012] [Indexed: 02/06/2023] Open
Abstract
Precise in vivo evaluation of cerebral vasospasm caused by subarachnoid hemorrhage has remained a critical but unsolved issue in experimental small animal models. In this study, we used synchrotron radiation angiography to study the vasospasm of anterior circulation arteries in two subarachnoid hemorrhage models in rats. Synchrotron radiation angiography, laser Doppler flowmetry-cerebral blood flow measurement, [125I]N-isopropyl-p-iodoamphetamine cerebral blood flow measurement and terminal examinations were applied to evaluate the changes of anterior circulation arteries in two subarachnoid hemorrhage models made by blood injection into cisterna magna and prechiasmatic cistern. Using synchrotron radiation angiography technique, we detected cerebral vasospasm in subarachnoid hemorrhage rats compared to the controls (p<0.05). We also identified two interesting findings: 1) both middle cerebral artery and anterior cerebral artery shrunk the most at day 3 after subarachnoid hemorrhage; 2) the diameter of anterior cerebral artery in the prechiasmatic cistern injection group was smaller than that in the cisterna magna injection group (p<0.05), but not for middle cerebral artery. We concluded that synchrotron radiation angiography provided a novel technique, which could directly evaluate cerebral vasospasm in small animal experimental subarachnoid hemorrhage models. The courses of vasospasm in these two injection models are similar; however, the model produced by prechiasmatic cistern injection is more suitable for study of anterior circulation vasospasm.
Collapse
|