1
|
Rivera R, Cruz JP, Merino-Osorio C, Rouchaud A, Mounayer C. Brain arteriovenous malformations: A scoping review of experimental models. INTERDISCIPLINARY NEUROSURGERY 2021. [DOI: 10.1016/j.inat.2021.101200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
2
|
Revuelta M, Zamarrón A, Fortes J, Rodríguez-Boto G, Gutiérrez-González R. Neuroprotective effect of indomethacin in normal perfusion pressure breakthrough phenomenon. Sci Rep 2020; 10:15466. [PMID: 32963342 PMCID: PMC7508825 DOI: 10.1038/s41598-020-72461-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 08/31/2020] [Indexed: 11/15/2022] Open
Abstract
Loss of cerebral autoregulation in normal perfusion pressure breakthrough (NPPB) phenomenon has been reported in other Central Nervous System diseases such as neonatal intraventricular haemorrhage. Several studies have demonstrated that low-dose indomethacin prevents this latter condition. A previous rat model was used to resemble NPPB phenomenon. Study animals were distributed in 4 groups that received 3 doses of indomethacin at different concentrations prior to fistula occlusion 60 days after its creation. Control animals received saline solution. Intracranial pressure (ICP) increased in all groups following fistula creation, whereas mean arterial pressure (MAP) and cerebral perfusion pressure (CPP) decreased as a manifestation of cerebral hypoperfusion and intracranial hypertension. The administration of indomethacin was associated with raised MAP and CPP, as well as decreased ICP. Sodium fluorescein extravasation was slight in study animals when comparing with control ones. Histological analysis evidenced diffuse ischaemic changes with signs of neuronal apoptosis in all brain layers in control animals. These findings were only focal and slight in study animals. The results suggest the usefulness of indomethacin to revert, at least partially, the haemodynamic effects of NPPB phenomenon in this experimental model, as well as to reduce BBB disruption and histological ischemia observed in absence of indomethacin.
Collapse
Affiliation(s)
- Manuel Revuelta
- Department of Neurosurgery, Puerta de Hierro University Hospital, Manuel de Falla 1, Majadahonda, 28222, Madrid, Spain
| | - Alvaro Zamarrón
- Department of Neurosurgery, La Paz University Hospital, Pº Castellana 261, 28046, Madrid, Spain
| | - Jose Fortes
- Health Research Institute-Fundación Jiménez Díaz (IIS-FJD), Avda Reyes Católicos S/N, 28040, Madrid, Spain
| | - Gregorio Rodríguez-Boto
- Department of Neurosurgery, Puerta de Hierro University Hospital, Manuel de Falla 1, Majadahonda, 28222, Madrid, Spain.,Department of Surgery, Faculty of Medicine, Autonomous University of Madrid, Arzobispo Morcillo 4, 28029, Madrid, Spain
| | - Raquel Gutiérrez-González
- Department of Neurosurgery, Puerta de Hierro University Hospital, Manuel de Falla 1, Majadahonda, 28222, Madrid, Spain. .,Health Research Institute-Fundación Jiménez Díaz (IIS-FJD), Avda Reyes Católicos S/N, 28040, Madrid, Spain.
| |
Collapse
|
3
|
Starke RM, McCarthy DJ, Chen CJ, Kano H, McShane BJ, Lee J, Patibandla MR, Mathieu D, Vasas LT, Kaufmann AM, Wang WG, Grills IS, Cifarelli CP, Paisan G, Vargo J, Chytka T, Janouskova L, Feliciano CE, Sujijantarat N, Matouk C, Chiang V, Hess J, Rodriguez-Mercado R, Tonetti DA, Lunsford LD, Sheehan JP. Hemorrhage risk of cerebral dural arteriovenous fistulas following Gamma Knife radiosurgery in a multicenter international consortium. J Neurosurg 2020; 132:1209-1217. [PMID: 30875690 PMCID: PMC6745287 DOI: 10.3171/2018.12.jns182208] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/12/2018] [Indexed: 11/12/2022]
Abstract
OBJECTIVE The authors performed a study to evaluate the hemorrhagic rates of cerebral dural arteriovenous fistulas (dAVFs) and the risk factors of hemorrhage following Gamma Knife radiosurgery (GKRS). METHODS Data from a cohort of patients undergoing GKRS for cerebral dAVFs were compiled from the International Radiosurgery Research Foundation. The annual posttreatment hemorrhage rate was calculated as the number of hemorrhages divided by the patient-years at risk. Risk factors for dAVF hemorrhage prior to GKRS and during the latency period after radiosurgery were evaluated in a multivariate analysis. RESULTS A total of 147 patients with dAVFs were treated with GKRS. Thirty-six patients (24.5%) presented with hemorrhage. dAVFs that had any cortical venous drainage (CVD) (OR = 3.8, p = 0.003) or convexity or torcula location (OR = 3.3, p = 0.017) were more likely to present with hemorrhage in multivariate analysis. Half of the patients had prior treatment (49.7%). Post-GRKS hemorrhage occurred in 4 patients, with an overall annual risk of 0.84% during the latency period. The annual risks of post-GKRS hemorrhage for Borden type 2-3 dAVFs and Borden type 2-3 hemorrhagic dAVFs were 1.45% and 0.93%, respectively. No hemorrhage occurred after radiological confirmation of obliteration. Independent predictors of hemorrhage following GKRS included nonhemorrhagic neural deficit presentation (HR = 21.6, p = 0.027) and increasing number of past endovascular treatments (HR = 1.81, p = 0.036). CONCLUSIONS Patients have similar rates of hemorrhage before and after radiosurgery until obliteration is achieved. dAVFs that have any CVD or are located in the convexity or torcula were more likely to present with hemorrhage. Patients presenting with nonhemorrhagic neural deficits and a history of endovascular treatments had higher risks of post-GKRS hemorrhage.
Collapse
Affiliation(s)
- Robert M Starke
- 1Department of Neurological Surgery, University of Miami, Florida
| | - David J McCarthy
- 1Department of Neurological Surgery, University of Miami, Florida
| | - Ching-Jen Chen
- 2Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia
| | - Hideyuki Kano
- 3Department of Neurological Surgery, University of Pittsburgh
| | - Brendan J McShane
- 4Department of Neurological Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John Lee
- 4Department of Neurological Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mohana Rao Patibandla
- 2Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia
| | - David Mathieu
- 5Department of Neurological Surgery, University of Sherbrooke, Quebec, Canada
| | - Lucas T Vasas
- 6Department of Neurological Surgery, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Anthony M Kaufmann
- 6Department of Neurological Surgery, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Wei Gang Wang
- 7Department of Radiation Oncology, Beaumont Health System, Royal Oak, Michigan
| | - Inga S Grills
- 7Department of Radiation Oncology, Beaumont Health System, Royal Oak, Michigan
| | | | - Gabriella Paisan
- 2Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia
| | - John Vargo
- 9Radiation Oncology, West Virginia University, Morgantown, West Virginia
| | - Tomas Chytka
- 10Department of Neurological Surgery, Na Homolce Hospital, Prague, Czech Republic
| | - Ladislava Janouskova
- 10Department of Neurological Surgery, Na Homolce Hospital, Prague, Czech Republic
| | - Caleb E Feliciano
- 11Department of Neurological Surgery, University of Puerto Rico, San Juan, Puerto Rico; and
| | - Nanthiya Sujijantarat
- 12Department of Neurological Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Charles Matouk
- 12Department of Neurological Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Veronica Chiang
- 12Department of Neurological Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Judith Hess
- 12Department of Neurological Surgery, Yale University School of Medicine, New Haven, Connecticut
| | | | | | - L Dade Lunsford
- 3Department of Neurological Surgery, University of Pittsburgh
| | - Jason P Sheehan
- 2Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
4
|
Revuelta JM, Zamarrón Á, Fortes J, Rodríguez-Boto G, Vaquero J, Gutiérrez-González R. Experimental rat model of chronic cerebral hypoperfusion-reperfusion mimicking normal perfusion pressure breakthrough phenomenon. Neurocirugia (Astur) 2020; 31:209-215. [PMID: 31948841 DOI: 10.1016/j.neucir.2019.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 11/13/2019] [Indexed: 11/25/2022]
Abstract
BACKGROUND Normal perfusion pressure breakthrough (NPPB) phenomenon is a major life-threatening complication that restricts the treatment of complex intracranial arteriovenous malformations. The aim of the study it to develop a rat model mimicking NPPB phenomenon that enables the evaluation of any therapy to prevent such complication. METHODS Twenty Wistar male rats were randomly assigned to either a study or a control group. Study animals underwent an end-to-side left external jugular vein-common carotid artery anastomosis and ligation of bilateral external carotid arteries. Control animals only underwent ligation of bilateral external carotid arteries. All animals were sacrificed sixty days after the procedure. Hemodynamic parameters [mean arterial pressure (MAP), intracranial pressure (ICP) and cerebral perfusion pressure (CPP)], blood-brain barrier (BBB) permeability (measured by fluorescein staining) and histological features were then compared between both groups. RESULTS A significant decrease in MAP and CPP was confirmed in the study group. An increase in ICP was also observed. A significant decrease in MAP and CPP was also present in the study group when comparing preoperative values with those recorded on days 0 (postoperative), 7 and 60. Fluorescein staining findings were consistent with signs of BBB disruption in study animals. Histological analysis demonstrated an increased number of pyknotic neurons in the ipsilateral hemisphere of rat brains included in the study group. CONCLUSION These results confirm that this model mimics a vascular steal state with chronic cerebral hypoperfusion comparable to patients with AVMs behavior and disruption of the BBB after fistula closure comparable to NPPB phenomenon disorders.
Collapse
Affiliation(s)
- Juan Manuel Revuelta
- Department of Neurosurgery, Puerta de Hierro University Hospital, Manuel de Falla 1, 28222 Majadahonda-Madrid, Spain
| | - Álvaro Zamarrón
- Department of Neurosurgery, La Paz University Hospital, P° Castellana 261, 28046 Madrid, Spain
| | - José Fortes
- Health Research Institute-Fundación Jiménez Díaz (IIS-FJD), Avda Reyes Católicos 2, 28040 Madrid, Spain
| | - Gregorio Rodríguez-Boto
- Department of Neurosurgery, Puerta de Hierro University Hospital, Manuel de Falla 1, 28222 Majadahonda-Madrid, Spain
| | - Jesús Vaquero
- Department of Neurosurgery, Puerta de Hierro University Hospital, Manuel de Falla 1, 28222 Majadahonda-Madrid, Spain
| | - Raquel Gutiérrez-González
- Department of Neurosurgery, Puerta de Hierro University Hospital, Manuel de Falla 1, 28222 Majadahonda-Madrid, Spain; Health Research Institute-Fundación Jiménez Díaz (IIS-FJD), Avda Reyes Católicos 2, 28040 Madrid, Spain.
| |
Collapse
|
5
|
Pike D, Shiu YT, Cho YF, Le H, Somarathna M, Isayeva T, Guo L, Symons JD, Kevil CG, Totenhagen J, Lee T. The effect of endothelial nitric oxide synthase on the hemodynamics and wall mechanics in murine arteriovenous fistulas. Sci Rep 2019; 9:4299. [PMID: 30862797 PMCID: PMC6414641 DOI: 10.1038/s41598-019-40683-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/19/2019] [Indexed: 11/12/2022] Open
Abstract
Creation of a hemodialysis arteriovenous fistula (AVF) causes aberrant vascular mechanics at and near the AVF anastomosis. When inadequately regulated, these aberrant mechanical factors may impede AVF lumen expansion to cause AVF maturation failure, a significant clinical problem with no effective treatments. The endothelial nitric oxide synthase (NOS3) system is crucial for vascular health and function, but its effect on AVF maturation has not been fully characterized. We hypothesize that NOS3 promotes AVF maturation by regulating local vascular mechanics following AVF creation. Here we report the first MRI-based fluid-structure interaction (FSI) study in a murine AVF model using three mouse strains: NOS3 overexpression (NOS3 OE) and knockout (NOS3-/-) on C57BL/6 background, with C57BL/6 as the wild-type control (NOS3+/+). When compared to NOS3+/+ and NOS3-/-, AVFs in the OE mice had larger lumen area. AVFs in the OE mice also had smoother blood flow streamlines, as well as lower blood shear stress at the wall, blood vorticity, inner wall circumferential stretch, and radial wall thinning at the anastomosis. Our results demonstrate that overexpression of NOS3 resulted in distinct hemodynamic and wall mechanical profiles associated with favorable AVF remodeling. Enhancing NOS3 expression may be a potential therapeutic approach for promoting AVF maturation.
Collapse
Affiliation(s)
- Daniel Pike
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Yan-Ting Shiu
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
- Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Yun-Fang Cho
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Ha Le
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Maheshika Somarathna
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tatyana Isayeva
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lingling Guo
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J David Symons
- Department of Nutrition and Integrative Physiology and Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Division of Endocrinology, Metabolism, and Diabetes, University of Utah, Salt Lake City, UT, USA
| | - Christopher G Kevil
- Departments of Pathology, Molecular and Cellular Physiology, and Cellular Biology and Anatomy, LSU Health Shreveport, Shreveport, LA, USA
| | - John Totenhagen
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Timmy Lee
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL, USA.
- Veterans Affairs Medical Center, Birmingham, AL, USA.
| |
Collapse
|
6
|
Herrera MI, Udovin LD, Toro-Urrego N, Kusnier CF, Luaces JP, Otero-Losada M, Capani F. Neuroprotection Targeting Protein Misfolding on Chronic Cerebral Hypoperfusion in the Context of Metabolic Syndrome. Front Neurosci 2018; 12:339. [PMID: 29904335 PMCID: PMC5990610 DOI: 10.3389/fnins.2018.00339] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 04/30/2018] [Indexed: 01/04/2023] Open
Abstract
Metabolic syndrome (MetS) is a cluster of risk factors that lead to microvascular dysfunction and chronic cerebral hypoperfusion (CCH). Long-standing reduction in oxygen and energy supply leads to brain hypoxia and protein misfolding, thereby linking CCH to Alzheimer's disease. Protein misfolding results in neurodegeneration as revealed by studying different experimental models of CCH. Regulating proteostasis network through pathways like the unfolded protein response (UPR), the ubiquitin-proteasome system (UPS), chaperone-mediated autophagy (CMA), and macroautophagy emerges as a novel target for neuroprotection. Lipoxin A4 methyl ester, baclofen, URB597, N-stearoyl-L-tyrosine, and melatonin may pose potential neuroprotective agents for rebalancing the proteostasis network under CCH. Autophagy is one of the most studied pathways of proteostatic cell response against the decrease in blood supply to the brain though the role of the UPR-specific chaperones and the UPS system in CCH deserves further research. Pharmacotherapy targeting misfolded proteins at different stages in the proteostatic pathway might be promising in treating cognitive impairment following CCH.
Collapse
Affiliation(s)
- María I Herrera
- Centro de Investigaciones en Psicología y Psicopedagogía, Facultad de Psicología y Psicopedagogía, Universidad Católica Argentina, Buenos Aires, Argentina.,Instituto de Investigaciones Cardiológicas (ININCA), Universidad de Buenos Aires (UBA-CONICET), Buenos Aires, Argentina
| | - Lucas D Udovin
- Instituto de Investigaciones Cardiológicas (ININCA), Universidad de Buenos Aires (UBA-CONICET), Buenos Aires, Argentina
| | - Nicolás Toro-Urrego
- Instituto de Investigaciones Cardiológicas (ININCA), Universidad de Buenos Aires (UBA-CONICET), Buenos Aires, Argentina
| | - Carlos F Kusnier
- Instituto de Investigaciones Cardiológicas (ININCA), Universidad de Buenos Aires (UBA-CONICET), Buenos Aires, Argentina
| | - Juan P Luaces
- Instituto de Investigaciones Cardiológicas (ININCA), Universidad de Buenos Aires (UBA-CONICET), Buenos Aires, Argentina
| | - Matilde Otero-Losada
- Instituto de Investigaciones Cardiológicas (ININCA), Universidad de Buenos Aires (UBA-CONICET), Buenos Aires, Argentina
| | - Francisco Capani
- Instituto de Investigaciones Cardiológicas (ININCA), Universidad de Buenos Aires (UBA-CONICET), Buenos Aires, Argentina.,Facultad de Medicina, Universidad Católica Argentina, Buenos Aires, Argentina.,Universidad Autónoma de Chile, Santiago de Chile, Chile
| |
Collapse
|
7
|
Zhou D, Meng R, Li S, Ya J, Ding J, Shang S, Ding Y, Ji X. Advances in chronic cerebral circulation insufficiency. CNS Neurosci Ther 2018; 24:5-17. [PMID: 29143463 PMCID: PMC6489997 DOI: 10.1111/cns.12780] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/25/2017] [Accepted: 10/26/2017] [Indexed: 12/30/2022] Open
Abstract
Chronic cerebral circulation insufficiency (CCCI) may not be an independent disease; rather, it is a pervasive state of long-term cerebral blood flow insufficiency caused by a variety of etiologies, and considered to be associated with either occurrence or recurrence of ischemic stroke, vascular cognitive impairment, and development of vascular dementia, resulting in disability and mortality worldwide. This review summarizes the features and recent progress of CCCI, mainly focusing on epidemiology, experimental research, pathophysiology, etiology, clinical manifestations, imaging presentation, diagnosis, and potential therapeutic regimens. Some research directions are briefly discussed as well.
Collapse
Affiliation(s)
- Da Zhou
- Departments of Neurology and NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
- Center of StrokeBeijing Institute for Brain DisordersBeijingChina
- Department of China‐America Institute of NeuroscienceXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Ran Meng
- Departments of Neurology and NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
- Center of StrokeBeijing Institute for Brain DisordersBeijingChina
- Department of China‐America Institute of NeuroscienceXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Si‐Jie Li
- Departments of Neurology and NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
- Center of StrokeBeijing Institute for Brain DisordersBeijingChina
| | - Jing‐Yuan Ya
- Departments of Neurology and NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
- Center of StrokeBeijing Institute for Brain DisordersBeijingChina
- Department of China‐America Institute of NeuroscienceXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Jia‐Yue Ding
- Departments of Neurology and NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
- Center of StrokeBeijing Institute for Brain DisordersBeijingChina
- Department of China‐America Institute of NeuroscienceXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Shu‐Ling Shang
- Departments of Neurology and NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Yu‐Chuan Ding
- Department of China‐America Institute of NeuroscienceXuanwu HospitalCapital Medical UniversityBeijingChina
- Department of NeurosurgeryWayne State University School of MedicineDetroitMIUSA
| | - Xun‐Ming Ji
- Departments of Neurology and NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
- Center of StrokeBeijing Institute for Brain DisordersBeijingChina
- Department of China‐America Institute of NeuroscienceXuanwu HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
8
|
Fennell VS, Martirosyan NL, Atwal GS, Kalani MYS, Ponce FA, Lemole GM, Dumont TM, Spetzler RF. Hemodynamics Associated With Intracerebral Arteriovenous Malformations: The Effects of Treatment Modalities. Neurosurgery 2017; 83:611-621. [DOI: 10.1093/neuros/nyx560] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 10/13/2017] [Indexed: 12/22/2022] Open
Abstract
Abstract
The understanding of the physiology of cerebral arteriovenous malformations (AVMs) continues to expand. Knowledge of the hemodynamics of blood flow associated with AVMs is also progressing as imaging and treatment modalities advance. The authors present a comprehensive literature review that reveals the physical hemodynamics of AVMs, and the effect that various treatment modalities have on AVM hemodynamics and the surrounding cortex and vasculature. The authors discuss feeding arteries, flow through the nidus, venous outflow, and the relative effects of radiosurgical monotherapy, endovascular embolization alone, and combined microsurgical treatments. The hemodynamics associated with intracranial AVMs is complex and likely changes over time with changes in the physical morphology and angioarchitecture of the lesions. Hemodynamic change may be even more of a factor as it pertains to the vast array of single and multimodal treatment options available. An understanding of AVM hemodynamics associated with differing treatment modalities can affect treatment strategies and should be considered for optimal clinical outcomes.
Collapse
Affiliation(s)
- Vernard S Fennell
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Nikolay L Martirosyan
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
- Division of Neurosurgery, University of Arizona College of Medicine, Tucson, Arizona
| | - Gursant S Atwal
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - M Yashar S Kalani
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Francisco A Ponce
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - G Michael Lemole
- Division of Neurosurgery, University of Arizona College of Medicine, Tucson, Arizona
| | - Travis M Dumont
- Division of Neurosurgery, University of Arizona College of Medicine, Tucson, Arizona
| | - Robert F Spetzler
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| |
Collapse
|
9
|
Du SQ, Wang XR, Xiao LY, Tu JF, Zhu W, He T, Liu CZ. Molecular Mechanisms of Vascular Dementia: What Can Be Learned from Animal Models of Chronic Cerebral Hypoperfusion? Mol Neurobiol 2016; 54:3670-3682. [PMID: 27206432 DOI: 10.1007/s12035-016-9915-1] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 05/03/2016] [Indexed: 01/06/2023]
Abstract
Vascular dementia (VD) is defined as a progressive neurodegenerative disease of cognitive decline, attributable to cerebrovascular factors. Numerous studies have demonstrated that chronic cerebral hypoperfusion (CCH) is associated with the initiation and progression of VD and Alzheimer's disease (AD). Suitable animal models were established to replicate such pathological condition in experimental research, which contributes largely to comprehending causal relationships between CCH and cognitive impairment. The most widely used experimental model of VD and CCH is permanent bilateral common carotid artery occlusion in rats. In CCH models, changes of learning and memory, cerebral blood flow (CBF), energy metabolism, and neuropathology initiated by ischemia were revealed. However, in order to achieve potential therapeutic targets, particular mechanisms in cognitive and neuropathological changes from CCH to dementia should be investigated. Recent studies have shown that hypoperfusion resulted in a chain of disruption of homeostatic interactions, including oxidative stress, neuroinflammation, neurotransmitter system dysfunction, mitochondrial dysfunction, disturbance of lipid metabolism, and alterations of growth factors. Evidence from experimental studies that elucidate the damaging effects of such imbalances suggests their critical roles in the pathogenesis of VD. The present review provides a summary of the achievements in mechanisms made with the CCH models, permits an understanding of the causative role played by CCH in VD, and highlights preventative and therapeutic prospects.
Collapse
Affiliation(s)
- Si-Qi Du
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Xue-Rui Wang
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Ling-Yong Xiao
- Beijing University of Chinese Medicine, 11 Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Jian-Feng Tu
- Beijing University of Chinese Medicine, 11 Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Wen Zhu
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Tian He
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Cun-Zhi Liu
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China.
| |
Collapse
|
10
|
Animal Models in Studying Cerebral Arteriovenous Malformation. BIOMED RESEARCH INTERNATIONAL 2015; 2015:178407. [PMID: 26649296 PMCID: PMC4663287 DOI: 10.1155/2015/178407] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/11/2015] [Accepted: 10/25/2015] [Indexed: 12/13/2022]
Abstract
Brain arteriovenous malformation (AVM) is an important cause of hemorrhagic stroke. The etiology is largely unknown and the therapeutics are controversial. A review of AVM-associated animal models may be helpful in order to understand the up-to-date knowledge and promote further research about the disease. We searched PubMed till December 31, 2014, with the term “arteriovenous malformation,” limiting results to animals and English language. Publications that described creations of AVM animal models or investigated AVM-related mechanisms and treatments using these models were reviewed. More than 100 articles fulfilling our inclusion criteria were identified, and from them eight different types of the original models were summarized. The backgrounds and procedures of these models, their applications, and research findings were demonstrated. Animal models are useful in studying the pathogenesis of AVM formation, growth, and rupture, as well as in developing and testing new treatments. Creations of preferable models are expected.
Collapse
|
11
|
Bayat M, Sharifi MD, Haghani M, Shabani M. Enriched environment improves synaptic plasticity and cognitive deficiency in chronic cerebral hypoperfused rats. Brain Res Bull 2015; 119:34-40. [PMID: 26474515 DOI: 10.1016/j.brainresbull.2015.10.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 10/02/2015] [Accepted: 10/07/2015] [Indexed: 12/12/2022]
Abstract
Recent studies have indicated that environmental enrichment (EE) increases the sensorial and social stimulations and leads to strengthened plastic changes in the brain. In models of chronic cerebral hypoperfusion, the ability of an EE to restore the cognition depends on hippocampal synaptic plasticity. The mechanisms for this effect have not, however, been adequately studied. Thus, the aim of the present study was to evaluate the neuroprotective effects and underlying mechanism of environmental enrichment by assessment of spatial memory tasks as well as parameters of synaptic plasticity in rats subjected to occlusion of the bilateral common carotid arteries (2-VO) model. Male Sprague-Dawley rats were used in this study. The model group was established by occlusion of the bilateral common carotid arteries. The animals were tested for learning, memory performance and synaptic plasticity using Morris water maze (MWM), 8-arm Radial Maze (RM), and field potential recording, respectively. The rats subjected to 2-VO in EE exhibited a significantly lower number of working errors and reference errors in RM. Moreover, the enriched environment recovered the memory performance of hypoperfused rats and decreased the swimming time to reach the platform in MWM. In addition, conditions of the environment did not have any effect on baseline synaptic transmission and presynaptic plasticity, but housing the animals in EE rescued the impairment of LTP induction induced by 2-VO. These results suggest that EE ameliorates the LTP and spatial memory impairment induced by 2-VO. Our data indicated that the LTP recovery by EE in the rat models of 2-VO is probably mediated by post-synaptic mechanisms.
Collapse
Affiliation(s)
- Mahnaz Bayat
- Department of Physiology, International Branch, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad Davood Sharifi
- Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Masoud Haghani
- Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad Shabani
- Kerman Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
12
|
Lin JB, Phillips EH, Riggins TE, Sangha GS, Chakraborty S, Lee JY, Lycke RJ, Hernandez CL, Soepriatna AH, Thorne BRH, Yrineo AA, Goergen CJ. Imaging of small animal peripheral artery disease models: recent advancements and translational potential. Int J Mol Sci 2015; 16:11131-77. [PMID: 25993289 PMCID: PMC4463694 DOI: 10.3390/ijms160511131] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 03/10/2015] [Indexed: 12/11/2022] Open
Abstract
Peripheral artery disease (PAD) is a broad disorder encompassing multiple forms of arterial disease outside of the heart. As such, PAD development is a multifactorial process with a variety of manifestations. For example, aneurysms are pathological expansions of an artery that can lead to rupture, while ischemic atherosclerosis reduces blood flow, increasing the risk of claudication, poor wound healing, limb amputation, and stroke. Current PAD treatment is often ineffective or associated with serious risks, largely because these disorders are commonly undiagnosed or misdiagnosed. Active areas of research are focused on detecting and characterizing deleterious arterial changes at early stages using non-invasive imaging strategies, such as ultrasound, as well as emerging technologies like photoacoustic imaging. Earlier disease detection and characterization could improve interventional strategies, leading to better prognosis in PAD patients. While rodents are being used to investigate PAD pathophysiology, imaging of these animal models has been underutilized. This review focuses on structural and molecular information and disease progression revealed by recent imaging efforts of aortic, cerebral, and peripheral vascular disease models in mice, rats, and rabbits. Effective translation to humans involves better understanding of underlying PAD pathophysiology to develop novel therapeutics and apply non-invasive imaging techniques in the clinic.
Collapse
Affiliation(s)
- Jenny B Lin
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Evan H Phillips
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Ti'Air E Riggins
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Gurneet S Sangha
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Sreyashi Chakraborty
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Janice Y Lee
- Psychological Sciences, Purdue University, West Lafayette, IN 47907, USA.
| | - Roy J Lycke
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Clarissa L Hernandez
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Arvin H Soepriatna
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Bradford R H Thorne
- School of Sciences, Neuroscience, Purdue University, West Lafayette, IN 47907, USA.
| | - Alexa A Yrineo
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| |
Collapse
|
13
|
Gutiérrez-González R, Pérez-Zamarron A, Rodríguez-Boto G. Normal perfusion pressure breakthrough phenomenon: experimental models. Neurosurg Rev 2014; 37:559-67. [PMID: 24777643 DOI: 10.1007/s10143-014-0549-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 08/25/2013] [Accepted: 02/08/2014] [Indexed: 11/28/2022]
Abstract
One of the most life-threatening complications after the obliteration of intracranial arteriovenous malformations is the development of oedema and/or multifocal haemorrhage. Two main theories have been postulated so far in order to explain this situation. On one hand, "normal perfusion pressure breakthrough phenomenon" is based on the loss of cerebral vessel autoregulation due to the chronic vasodilation of perinidal microcirculation. On the other hand, the "occlusive hyperaemia" deals with thrombotic and venous obstruction phenomena that may also generate such manifestations. The aim of this study is to resume the main concepts of the "normal perfusion pressure breakthrough phenomenon" theory as well as the related animal models described up to date, their advantages and disadvantages, and the main conclusions obtained as a result of the experimental research.
Collapse
Affiliation(s)
- Raquel Gutiérrez-González
- Department of Neurosurgery, Fundación Jiménez Díaz (IIS-FJD), Avda Reyes Católicos 2, 28040, Madrid, Spain,
| | | | | |
Collapse
|
14
|
Yen CP, Lanzino G, Sheehan JP. Stereotactic Radiosurgery of Intracranial Dural Arteriovenous Fistulas. Neurosurg Clin N Am 2013; 24:591-6. [DOI: 10.1016/j.nec.2013.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
15
|
Jian H, Yi-Fang W, Qi L, Xiao-Song H, Gui-Yun Z. Cerebral blood flow and metabolic changes in hippocampal regions of a modified rat model with chronic cerebral hypoperfusion. Acta Neurol Belg 2013; 113:313-7. [PMID: 23111782 DOI: 10.1007/s13760-012-0154-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 10/16/2012] [Indexed: 10/27/2022]
Abstract
Chronic cerebral hypoperfusion (CCH) causes neurodegeneration which contributes to the cognitive impairment. This study utilized a modified rat model with CCH to investigate cerebral blood flow (CBF) and hippocampal metabolic changes. CBF was measured by laser Doppler flowmetry. Various metabolic ratios were evaluated from selective volumes of interest (VOI) in left hippocampal regions using in vivo proton magnetic resonance spectroscopy ((1)H-MRS). The ultrastructural changes with special respect to ribosomes in rat hippocampal CA1 neurons were studied by electron microscopy. CBF decreased immediately after CCH and remained reduced significantly at 1 day and 3 months postoperatively. (1)H-MRS revealed that CCH led to a significant decrease of N-acetyl aspartate/creatine (NAA/Cr) ratio in the hippocampal VOI in the model rats compared with the sham-operated control rats. However, no changes of myo-inositol/Cr, choline/Cr and glutamate and glutamine/Cr ratios between the model and control groups were observed. Under electron microscopy, most rosette-shaped polyribosomes were relatively evenly distributed in the hippocampal CA1 neuronal cytoplasms of the control rats. After CCH, most ribosomes were clumped into large abnormal aggregates in the model rats. Our data suggests that both permanent decrease of CBF and reduction of NAA/Cr ratio in the hippocampal regions may be related to the cognitive deficits in rats with CCH.
Collapse
|
16
|
Hai J, Lin Q, Deng DF, Pan QG, Ding MX. The pre-treatment effect on brain injury during restoration of normal perfusion pressure with hemodilution in a new rat model of chronic cerebral hypoperfusion. Neurol Res 2013; 29:583-7. [PMID: 17535558 DOI: 10.1179/016164107x166254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES To investigate the effect of hemodilution with high-concentration human serum albumin (HSA) on brain injury in a rat model of chronic cerebral hypoperfusion associated with arteriovenous malformations. METHODS The animal model was established by creating a fistula through an end-to-side anastomosis between the right distal external jugular vein and the ipsilateral common carotid artery, followed by ligation of the left vein draining the transverse sinus and bilateral external carotid arteries. The agent (20% HSA) or control solution (0.9% sodium chloride) was administered intravenously at a dosage of 1% body weight 24 hours before ligation of the fistula. Blood-brain barrier (BBB) disruption was judged by extravasation of Evans blue (EB) dye. EB, water content and the changes of myeloperoxidase (MPO) activity and superoxide dismutase (SOD) activity in rat brains 24 hours after ligation of the fistula were determined. RESULTS EB and water content in rat brains of the pre-treated group were significantly decreased compared with the control group accompanied by reduction of MPO activity and enhancement of SOD activity. DISCUSSION Hemodilution with high-concentration HSA has a certain pre-treatment effect on brain injury after ligation of the fistula in rat model of chronic cerebral hypoperfusion, which may be resulted from improved microcirculation, decrease in inflammatory cell infiltration and inactivation of oxygen free radicals.
Collapse
Affiliation(s)
- Jian Hai
- Department of Neurosurgery, Tongji Hospital, Tongji University, 389 XinCun Rd, Shanghai 200065, China.
| | | | | | | | | |
Collapse
|
17
|
Wu JS, Chen XC, Chen H, Shi YQ. A study on blood–brain barrier ultrastructural changes induced by cerebral hypoperfusion of different stages. Neurol Res 2013; 28:50-8. [PMID: 16464363 DOI: 10.1179/016164106x91870] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES To evaluate the ultrastructural changes in the blood-brain barrier (BBB) induced by cerebral hypoperfusion of different stages, which may predispose the brain to the formation of vasogenic edema and hemorrhage under cerebral luxury perfusion. METHODS Twenty cerebral steal models with left neck arteriovenous fistula (AVF) were surgically created in Wistar's rats, leading to a noninfarctional reduction in the cerebral blood flow (CBF) by between 25 and 50%, resulting in cerebral hypoperfusion in the AVF side for 3 days (acute stage), 3 weeks (subacute stage) and 3 months (chronic stage), respectively. Another six sham-operated models were made in age-matched rats as control. The BBB ultrastructural changes were assessed by transmission electron microscopy. Ridit analysis was conducted to compare the positive ratio of ultrastructural changes among multiple groups. RESULTS Electron microscopy demonstrated no ultrastructural change at the acute stage, however, at the subacute stage, slight vacuolar degeneration was found in the astrocytic foot process layer encircling the capillaries; furthermore, at the chronic stage, the astrocytic foot processes expressed marked vacuolization associated with the adjacent astrocytic degeneration. Meanwhile, in both capillary endothelium and basal lamina layers, no abnormal ultrastructures similar to those in the astrocytic foot processes layer were identified. After cerebral luxury perfusion took place, BBB was disrupted where astrocytic foot processes vacuolization was most distinguished. CONCLUSION Astrocytes generate ultrastructural abnormality as a result of chronic cerebral hypoperfusion. Astrocytic foot process vacuolization, which constitutes the major ultrastructural change in the BBB, is the extension of the degeneration of astrocyte body. It is inferred that BBB is prone to structure weakness and function instability, which forms the morphological basis of cerebral luxury perfusion.
Collapse
Affiliation(s)
- Jin-Song Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical School, Fudan University, 12 Wulumuqi Zhong Road, Shanghai 200040, China.
| | | | | | | |
Collapse
|
18
|
Dalyai RT, Ghobrial G, Chalouhi N, Dumont AS, Tjoumakaris S, Gonzalez LF, Rosenwasser R, Jabbour P. Radiosurgery for dural arterio-venous fistulas: A review. Clin Neurol Neurosurg 2013; 115:512-6. [PMID: 23481896 DOI: 10.1016/j.clineuro.2013.01.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 12/17/2012] [Accepted: 01/20/2013] [Indexed: 10/27/2022]
|
19
|
Hai J, Lin Q, Wu YF, Huang XS, Zhang GY, Wang F. Effects of N-stearoyl-L-tyrosine on the hippocampal ubiquitin-proteasome system in rats with chronic cerebral hypoperfusion. Neurol Res 2013; 35:734-43. [PMID: 23562289 DOI: 10.1179/1743132812y.0000000154] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Chronic cerebral hypoperfusion (CCH) leads to neurodegeneration and cognitive impairment. Ubiquitinated protein aggregates are commonly present in neurodegenerative disorders and are believed to cause neuronal degeneration. Here, we investigated the effects of N-stearoyl-L-tyrosine (NSTyr) on the hippocampal ubiquitin-proteasome system (UPS) in rats with CCH. METHODS After induction of CCH, NSTyr was intraperitoneally administered daily for 3 months. Protein aggregation was analyzed by ethanolic phosphotungstic acid (EPTA) electron microscopy (EM), immunogold EM, laser-scanning confocal microscopy, and Western blot. Proteasome peptidase activity was measured by peptidase activity assays. RESULTS By using EPTA EM, immunogold EM and high-resolution laser-scanning confocal microscopy, we found that CCH resulted in the accumulation of ubiquitinated protein aggregates in rat hippocampal CA1 neurons. Western blot revealed that the levels of free ubiquitin were significantly reduced and that the levels of ubiquitinated proteins were markedly increased in the hippocampus of CCH rats. Direct activity measurements demonstrated that proteasome peptidase activity in the hippocampal region of rats was decreased after CCH induction. In the hippocampal tissue of CCH rats treated with NSTyr, however, ubiquitinated protein aggregates decreased and proteasome peptidase activity increased. DISCUSSION These data indicate that NSTyr may exert protective effects on rat hippocampal UPS function via endogenous regulation.
Collapse
Affiliation(s)
- Jian Hai
- Tongji University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
20
|
Zinkel JL. Rat forebrain perfusion in vivo by 1 artery like the isolated kidney model: a robust recovery model permitting ischemia without anesthesia to compare multiple brain injury states. Neurosurgery 2013; 72:662-77; discussion 676-7. [PMID: 23277378 DOI: 10.1227/neu.0b013e3182846f4c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Rat brain perfusion models are critical to basic research, but they can be imprecise and/or not durable for extended outcome studies. OBJECTIVE To demonstrate a rat brain perfusion model that provides a simplified reliable brain perfusion circuit, reduces variables during experiment and recovery, and therefore permits more precise, reliable, and context-independent research data. METHODS Rat forebrain perfusion was reduced surgically to that by 1 internal carotid artery without injury to the animal. The next day, the fully awake rat was studied for brain ischemia painlessly yet in the absence of anesthesia or other interventions that might bias or alter the biochemistry of the event. This model was rigorously validated with isotope cerebral blood studies during ischemia and with histology studies at 72 hours after ischemia. The first application of this model was to compare ischemia injuries for global total, global penumbra, and global shock ischemia in a single experimental context. RESULTS This model is accurate, reliable, and remarkably durable. This model permits the severest brain ischemia by vessel occlusion ever demonstrated in a recovery model. It also confirms that, with conditions otherwise identical, penumbra ischemia is less injurious than total ischemia and that total ischemia is less injurious than shock ischemia. CONCLUSION Although meticulous in construction, this model creates ischemia more simply and more reliably than the Pulsinelli 4-vessel ischemia model that inspired it, with the inherent advantages of an isolated organ system, in which a known tissue volume is perfused at a predetermined volume and rate. This model permits robust long-term recovery.
Collapse
Affiliation(s)
- John L Zinkel
- Department of Neurological Surgery, Beaumont Hospital Grosse Pointe, St. Clair Shores, MI 48081, USA.
| |
Collapse
|
21
|
Gutierrez-Gonzalez R, Rodriguez-Boto G. Response to: Pathophysiology of normal perfusion pressure breakthrough: more than just abnormal arteries …. Br J Neurosurg 2012. [DOI: 10.3109/02688697.2012.717653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
22
|
Gutiérrez-González R, Gil A, Serna C, López-Ibor L, Boto GR. Normal perfusion pressure breakthrough phenomenon: what still remains unknown. Br J Neurosurg 2012; 26:403-5. [PMID: 22263544 DOI: 10.3109/02688697.2011.633637] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We report two cases of normal perfusion pressure breakthrough phenomenon after total brain arteriovenous malformation removal. Hereby, we demonstrate that not only autoregulation impairment in the ipsilateral hemisphere occurs but also contralateral remote vessels response does. Such findings may be observed at 2-4 weeks and may resolve after 1-3 months.
Collapse
Affiliation(s)
- Raquel Gutiérrez-González
- Department of Neurosurgery, Clínico San Carlos University Hospital, Prof. Martin Lagos, Madrid, Spain.
| | | | | | | | | |
Collapse
|
23
|
The changes of signal transduction pathways in hippocampal regions and postsynaptic densities after chronic cerebral hypoperfusion in rats. Brain Res 2012; 1429:9-17. [DOI: 10.1016/j.brainres.2011.10.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2011] [Revised: 10/11/2011] [Accepted: 10/12/2011] [Indexed: 11/15/2022]
|
24
|
Cifarelli CP, Kaptain G, Yen CP, Schlesinger D, Sheehan JP. Gamma knife radiosurgery for dural arteriovenous fistulas. Neurosurgery 2011; 67:1230-5; discussion 1235. [PMID: 20871448 DOI: 10.1227/neu.0b013e3181eff6f7] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The current management of intracranial dural arteriovenous fistulas (dAVFs) incorporates a multimodal approach involving microneurosurgery, endovascular embolization, and radiosurgery. OBJECTIVE To explore the role of Gamma Knife radiosurgery for dAVFs. METHODS The series includes patients with dAVFs who had Gamma Knife radiosurgery at the University of Virginia Medical Center between 1989 and 2005 with clinical follow-up through 2008. Medical records were reviewed to obtain clinical history, demographic data, and dosimetry. Radiographic records provided the location and anatomy of the dAVFs. Follow-up angiography was performed 2 to 3 years after treatment, with cure defined as complete obliteration of fistulous flow. Follow-up for clinical symptomology and quality of life was obtained from direct patient and primary physician questionnaires. RESULTS Fifty-five patients underwent Gamma Knife radiosurgery for dAVFs during the study period. Twenty patients (36%) presented with intracranial hemorrhage before radiosurgery. Gamma Knife radiosurgery was preceded by craniotomy for microneurosurgical ablation in 11 patients (20%) or endovascular embolization in 36 patients (65%). Follow-up angiography was performed on 46 patients (83%) with documented obliteration in 30 patients (65%). Patients lost to follow-up were classified as treatment failures, adjusting the range of efficacy from 65% to 54%. Three patients (5%) suffered a posttreatment hemorrhage during the follow-up period, but no new permanent neurological deficits resulted from these events. CONCLUSION Gamma Knife radiosurgery is an effective adjunct therapy for dAVFs with persistence of flow after open neurosurgical resection or endovascular treatment while still maintaining a role in nonaggressive dAVFs not amenable to either surgery or embolization.
Collapse
Affiliation(s)
| | - George Kaptain
- Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia
| | - Chun-Po Yen
- Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia
| | - David Schlesinger
- Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia
| | - Jason P Sheehan
- Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
25
|
Hai J, Lin Q, Su SH, Zhang L, Wan JF, Lu Y. Chronic cerebral hypoperfusion in rats causes proteasome dysfunction and aggregation of ubiquitinated proteins. Brain Res 2010; 1374:73-81. [PMID: 21167821 DOI: 10.1016/j.brainres.2010.12.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 11/30/2010] [Accepted: 12/10/2010] [Indexed: 11/18/2022]
Abstract
The deposition of abnormal protein aggregates is a feature of several neurodegenerative diseases. We have employed a rat model to investigate whether chronic cerebral hypoperfusion (CCH) induces proteasome dysfunction and the accumulation of ubiquitinated proteins and aggregates in the CNS. Protein aggregation was analyzed by ethanolic phosphotungstic acid (EPTA) electron microscopy (EM), immunogold EM, laser-scanning confocal microscopy, and Western blotting. Proteasome peptidase activity was studied by peptidase activity assays. EPTA EM and immunogold EM revealed that CCH led to the accumulation of protein aggregates in rat hippocampal CA1 neurons. High-resolution confocal microscopy demonstrated the presence of ubiquitin-positive protein aggregates surrounding nuclei and along dendrites. Western blotting revealed that levels of free ubiquitin were significantly reduced and that levels of ubiquitinated proteins were markedly increased in the hippocampus of CCH rats. Direct activity measurements revealed that proteasome peptidase activity in the hippocampal region of rats was decreased after CCH induction. These data suggest that reduced proteasome activity following CCH could impair the removal of abnormally folded proteins via the ubiquitin-proteasome pathway, leading to the accumulation of potentially toxic protein aggregates that could contribute to neurodegeneration.
Collapse
Affiliation(s)
- Jian Hai
- Department of Neurosurgery, Tongji Hospital, Tongji University, Shanghai 200065, China.
| | | | | | | | | | | |
Collapse
|
26
|
Lin Q, Hai J, Yao LY, Lu Y. Neuroprotective effects of NSTyr on cognitive function and neuronal plasticity in rats of chronic cerebral hypoperfusion. Brain Res 2010; 1325:183-90. [DOI: 10.1016/j.brainres.2010.02.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Revised: 02/07/2010] [Accepted: 02/08/2010] [Indexed: 02/01/2023]
|
27
|
Cognitive dysfunction induced by chronic cerebral hypoperfusion in a rat model associated with arteriovenous malformations. Brain Res 2009; 1301:80-8. [PMID: 19761762 DOI: 10.1016/j.brainres.2009.09.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 08/27/2009] [Accepted: 09/02/2009] [Indexed: 11/23/2022]
Abstract
The relationship between chronic cerebral hypoperfusion and cognitive function has not been completely delineated. In the present studies, we developed an experimental model associated with arteriovenous malformation to investigate the effects of chronic cerebral hypoperfusion on cognitive function and neuropathological changes. The rat model was established by creating a fistula through an end-to-side anastomosis between the right distal external jugular vein and the ipsilateral common carotid artery, followed by ligation of the left vein draining the transverse sinus and bilateral external carotid arteries. Age-matched rats comprised a control group. Three months after surgery, cognitive functions were evaluated by the Morris water maze and hippocampal long-term potentiation (LTP). Neuropathological changes were examined using light and electron microscopic techniques. We found that both learning capacity and spatial memory were significantly impaired in rats with chronic cerebral hypoperfusion concomitant with LTP inhibition and neurodegeneration in the hippocampal CA1 region of model rats compared with control rats. In addition, model rats showed a decrease at the protein level of cyclic AMP response element binding (CREB) phosphorylation in hippocampal tissues. Therefore, cognitive impairment caused by chronic cerebral hypoperfusion associated with arteriovenous malformations may be partially explained by the neurodegeneration and reduction of CREB phosphorylation in rat hippocampus.
Collapse
|
28
|
Gao P, Zhu Y, Ling F, Shen F, Lee B, Gabriel RA, Hao Q, Yang GY, Su H, Young WL. Nonischemic cerebral venous hypertension promotes a pro-angiogenic stage through HIF-1 downstream genes and leukocyte-derived MMP-9. J Cereb Blood Flow Metab 2009; 29:1482-90. [PMID: 19471278 PMCID: PMC2745831 DOI: 10.1038/jcbfm.2009.67] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cerebral venous hypertension (VH) and angiogenesis are implicated in the pathogenesis of brain arteriovenous malformation and dural arteriovenous fistulae. We studied the association of VH and angiogenesis using a mouse brain VH model. Sixty mice underwent external jugular vein and common carotid artery (CCA) anastomosis (VH model), CCA ligation, or sham dissection (n=20). Hypoxia-inducible factor-1alpha (HIF-1alpha), vascular endothelial growth factor (VEGF) and stromal-cell-derived factor-1alpha (SDF-1alpha) expression, and matrix metalloproteinase (MMP) activity were analyzed. We found VH animals had higher (P<0.05) sagittal sinus pressure (8+/-1 mm Hg) than control groups (1+/-1 mm Hg). Surface cerebral blood flow and mean arterial pressure did not change. Hypoxia-inducible factor-1alpha, VEGF, and SDF-1alpha expression increased (P<0.05). Neutrophils and MMP-9 activity increased 10-fold 1 day after surgery, gradually decreased afterward, and returned to baseline 2 weeks after surgery. Macrophages began to increase 3 days after surgery (P<0.05), which coincided with the changes in SDF-1alpha expression. Capillary density in the parasagittal cortex increased 17% compared with the controls. Our findings suggest that mild nonischemic VH results in a pro-angiogenic stage in the brain by upregulating HIF-1 and its downstream targets, VEGF and SDF-1alpha, increasing leukocyte infiltration and MMP-9 activity.
Collapse
Affiliation(s)
- Peng Gao
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California,San Francisco, CA
- Department of Neurosurgery, Xuanwu Hospital, Capital University of Medical Sciences, Beijing, China
- Department of Neurosurgery, Tongji Hospital, Tongji University, Shanghai, China
| | - Yiqian Zhu
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California,San Francisco, CA
| | - Feng Ling
- Department of Neurosurgery, Xuanwu Hospital, Capital University of Medical Sciences, Beijing, China
| | - Fanxia Shen
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California,San Francisco, CA
| | - Brian Lee
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California,San Francisco, CA
| | - Rodney Allanigue Gabriel
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California,San Francisco, CA
| | - Qi Hao
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California,San Francisco, CA
| | - Guo-Yuan Yang
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California,San Francisco, CA
- Med-X Research Institute, Shanghai JiaoTong University, Shanghai, China
| | - Hua Su
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California,San Francisco, CA
| | - William L. Young
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California,San Francisco, CA
- Department of Neurological Surgery, University of California, San Francisco, CA
- Department of Neurology, University of California, San Francisco, CA
| |
Collapse
|
29
|
Hossmann KA. Cerebral ischemia: Models, methods and outcomes. Neuropharmacology 2008; 55:257-70. [DOI: 10.1016/j.neuropharm.2007.12.004] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Revised: 12/06/2007] [Accepted: 12/10/2007] [Indexed: 01/31/2023]
|
30
|
Kojima T, Miyachi S, Sahara Y, Nakai K, Okamoto T, Hattori K, Kobayashi N, Hattori K, Negoro M, Yoshida J. The relationship between venous hypertension and expression of vascular endothelial growth factor: hemodynamic and immunohistochemical examinations in a rat venous hypertension model. ACTA ACUST UNITED AC 2007; 68:277-84; discussion 284. [PMID: 17719966 DOI: 10.1016/j.surneu.2006.10.075] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2006] [Accepted: 10/30/2006] [Indexed: 11/24/2022]
Abstract
BACKGROUND Venous hypertension is regarded as an important factor in the pathogenesis of dural arteriovenous fistula (DAVF). We investigate histologic reaction of dural sinus under the condition of venous hypertension using a rat venous hypertension model to present hemodynamic and immunohistochemical effect in the development of DAVF. METHODS Twenty-four Sprague-Dawley male rats were divided into venous hypertension and control groups. Venous hypertension was induced with a left common carotid artery-external jugular vein anastomosis and an occlusion of a right posterior facial vein. Measurements of systemic mean arterial pressure, draining vein pressure (DVP), and cerebral perfusion pressure (CPP) were conducted on the next day, at 7 days, and at 28 days after surgery, and the rats were killed for histologic examinations. RESULTS Postoperative DVP increased significantly in venous hypertension group compared to control group (35 +/- 5 vs 13 +/- 2 mm Hg, P < .05). Increased DVP remained above 30 mm Hg throughout the observation period. Postoperative CPP decreased significantly in venous hypertension group compared to control group (49 +/- 8 vs 86 +/- 9 mm Hg, P < .05). In venous hypertension group, there was a significant difference between days 1 and 28 (49 +/- 8 vs 64 +/- 8 mm Hg, P < .05). Histologic examination revealed thickening of connective tissues, proliferation of fibroblasts, and strong expression of vascular endothelial growth factor (VEGF) in endothelium under venous hypertension condition. Immunostained VEGF cells decreased significantly from day 7 to day 28 (100 +/- 16 vs 72 +/- 19 cells, P < .05). A positive correlation was observed between DVP and VEGF expression (Pearson correlation coefficient; r = 0.671, P = .0017). There was a negative correlation between CPP and immunostained VEGF cells (r = -0.702, P = .0089). CONCLUSIONS These results suggest that venous hypertension is associated with increased expression of VEGF, and a decreased CPP may have a potential effect in VEGF expression under venous hypertension condition. These factors are speculated to play an important role in progression of DAVF.
Collapse
Affiliation(s)
- Takao Kojima
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Brambilla P, Cerini R, Fabene PF, Andreone N, Rambaldelli G, Farace P, Versace A, Perlini C, Pelizza L, Gasparini A, Gatti R, Bellani M, Dusi N, Barbui C, Nosè M, Tournikioti K, Sbarbati A, Tansella M. Assessment of cerebral blood volume in schizophrenia: A magnetic resonance imaging study. J Psychiatr Res 2007; 41:502-10. [PMID: 16698038 DOI: 10.1016/j.jpsychires.2006.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2005] [Revised: 02/26/2006] [Accepted: 03/06/2006] [Indexed: 11/22/2022]
Abstract
Brain atrophy has consistently been observed in schizophrenia, representing a 'gross' evidence of anatomical abnormalities. Reduced cerebral blood volume (CBV) may accompany brain size decrement in schizophrenia, as suggested by prior small SPECT studies. In this study, we non-invasively investigated the hemisphere CBV in a large sample of patients suffering from schizophrenia with perfusion-weighted imaging (PWI). PWI images were obtained, following intravenous injection of paramagnetic contrast agent (Gadolinium-DTPA), for 54 DSM-IV patients with schizophrenia (mean age+/-SD=39.19+/-12.20 years; 34 males, 20 females) and 24 normal controls (mean age+/-SD=44.63+/-10.43 years; 9 males, 15 females) with a 1.5T Siemens magnet using an echo-planar sequence (TR=2160 ms, TE=47 ms, slice thickness=5mm). The contrast of enhancement (CE), a semi-quantitative parameter inversely estimating the CBV, were calculated pixel by pixel as the ratio of the maximum signal intensity drop during the passage of contrast agent (Sm) by the baseline pre-bolus signal intensity (So) (CE=Sm/Sox100) for right and left hemisphere on two axial images. Specifically, higher CE values correspond to lower CBV and viceversa Compared to normal controls, patients with schizophrenia had significantly higher bilateral hemisphere CE values (p=0.02) and inverse CE laterality index (p=0.02). This study showed abnormally reduced and inverse hemisphere CBV in a large population of patients with schizophrenia. Hypothetically, chronic low CBV may sustain neural hypoactivation and concomitant increase of free radicals, ultimately resulting in neuronal loss and cognitive impairments. Thus, altered intracranial hemodynamics may accompany brain atrophy and cognitive deficits, being a crucial factor in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Paolo Brambilla
- Department of Pathology and Experimental and Clinical Medicine, Section of Psychiatry, University of Udine, Udine, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
OBJECTIVE The aim of this paper is to analyse and summarize the main advances in experimental research on cerebral arteriovenous fistulas. METHODS A detailed analysis of the literature and my own research experience were employed to outline the methodology whereby experimental cerebral arteriovenous fistulas are created and further studied. RESULTS The analysis and quantification of the anatomical and functional variables in different experimental cerebral arteriovenous fistula models make it possible to develop more appropriate and individual ways of treatment in affected patients. CONCLUSION Experimental research on cerebral arteriovenous fistulas helps physicians to understand and predict more accurately the future evolution of arteriovenous malformations in humans.
Collapse
Affiliation(s)
- Mario N Carvi Y Nievas
- Department of Neurosurgery, Städtische Kliniken, Frankfurt am Main Höchst, Akademisches Lehrkrankenhaus der Johann-Wolfgang-Goethe-Universität, Germany.
| |
Collapse
|
33
|
Hai J, Lin Q, Li ST, Pan QG. Chronic cerebral hypoperfusion and reperfusion injury of restoration of normal perfusion pressure contributes to the neuropathological changes in rat brain. ACTA ACUST UNITED AC 2004; 126:137-45. [PMID: 15249137 DOI: 10.1016/j.molbrainres.2004.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2004] [Indexed: 10/26/2022]
Abstract
Restoration of normal perfusion pressure after resection of cerebral arteriovenous malformations (AVMs) is sometimes complicated by unexplained postoperative brain swelling and/or intracranial hemorrhage, which has been termed normal perfusion pressure breakthrough (NPPB). The precise mechanism of NPPB is still unclear. In this study, we investigated the time courses of blood-brain barrier (BBB) disruption, water content, neuronal apoptosis, myeloperoxidase (MPO) activity and superoxide dismutase (SOD) activity in the brain during restoration of normal perfusion pressure in a new rat model of chronic cerebral hypoperfusion associated with AVMs. Male Sprague-Dawley rats were randomly divided into either a sham-operated group, a control group, or a model group with reperfusion assessed at 1, 12, 24 and 72 h after restoration of normal perfusion pressure. BBB disruption was judged by extravasation of Evans blue (EB) dye. We observed that EB and water content in rat brains of the model group with reperfusion were significantly increased compared with the other groups. The most predominant increase occurred at 1 h after reperfusion, and the next at 24 h after reperfusion, representing biphasic changes which are similar to the pathological processes of acute cerebral ischemia/reperfusion injury. There was no difference of the percentage of apoptotic cells in rat brains between the sham-operated group and the control group using flow cytometry. No prominent apoptotic cells were found in the model group with reperfusion at 1 h. However, the percentage of apoptotic cells increased significantly in rat brains of the model group with reperfusion at 12 h, peaked at 24 h, and decreased at 72 h after reperfusion. Apoptotic cells were confirmed with electron microscopy and terminal deoxynuleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL). A significant enhancement of MPO activity in combination with reduction of SOD activity was seen at 12, 24 and 72 h in rat brains of the model group with reperfusion. Our data indicates that reperfusion after restoration of normal perfusion pressure with chronic cerebral hypoperfusion lead to secondary neuronal damage which may associate with cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Jian Hai
- Department of Neurosurgery, Tongji Hospital, Tongji University, 389 XinCun Rd., Shanghai 200065, China.
| | | | | | | |
Collapse
|
34
|
Lawton MT, Stewart CL, Wulfstat AA, Derugin N, Hashimoto T, Young WL. THE TRANSGENIC ARTERIOVENOUS FISTULA IN THE RAT: AN EXPERIMENTAL MODEL OF GENE THERAPY FOR BRAIN ARTERIOVENOUS MALFORMATIONS. Neurosurgery 2004; 54:1463-71; discussion 1471. [PMID: 15157304 DOI: 10.1227/01.neu.0000125004.89129.b9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2003] [Accepted: 02/11/2004] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To introduce the transgenic arteriovenous fistula model in the rat, constructed by interposing mouse aorta in a fistula between the common carotid artery and external jugular vein in a nude rat, and to describe the model's technical feasibility, long-term patency, and expression of reporter genes. METHODS Carotid-jugular fistulae were surgically created in 112 rats. In 25 immunodeficient nude rats, wild-type mouse thoracic aorta (TAo) was interposed in the fistula; in 10 immunocompetent rats, TAo was interposed; in 19 nude rats, transgenic TAo with reporter genes for beta-galactosidase or green fluorescent protein was interposed; in 18 nude rats, wild-type mouse ascending aorta was interposed; and in 40 rats, a simple fistula was constructed without an interpositional graft. Host tolerance and graft viability were analyzed by histopathology and immunohistochemistry for CD31 (mouse endothelial cell marker), endothelial nitric oxide synthase, smooth muscle actin, fibronectin, beta-galactosidase, and green fluorescent protein. RESULTS The transgenic arteriovenous fistula was technically feasible and immunologically tolerated in nude rats but not in immunocompetent rats. The overall angiographic patency rate was 41% with TAo grafts and 56% with ascending aorta grafts, both lower than the 98% patency rate in fistulae with a single anastomosis and no interpositional graft. Mouse endothelium survived on the graft for 3 months according to CD31 staining, but longer survival by transgenic smooth muscle cells resulted in continued expression of beta-galactosidase for 6 months and green fluorescent protein for 4 months. Endothelium and smooth muscle in the fistula were functional, with normal expression of endothelial nitric oxide synthase as well as smooth muscle actin and fibronectin, respectively. CONCLUSION The transgenic arteriovenous fistula model enhances other carotid-jugular fistula models by integrating transgenic tissue, thereby creating an experimental system for investigating the molecular biology of and gene therapies for arteriovenous malformations.
Collapse
Affiliation(s)
- Michael T Lawton
- Department of Neurological Surgery, Center for Cerebrovascular Research, University of California, San Francisco, 94143-0012, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Hai J, Li ST, Lin Q, Pan QG, Gao F, Ding MX. Vascular Endothelial Growth Factor Expression and Angiogenesis Induced by Chronic Cerebral Hypoperfusion in Rat Brain. Neurosurgery 2003; 53:963-70; discussion 970-2. [PMID: 14519228 DOI: 10.1227/01.neu.0000083594.10117.7a] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2003] [Accepted: 06/04/2003] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE In a rat model, we studied the time courses of vascular endothelial growth factor (VEGF) expression and angiogenesis induced by chronic cerebral hypoperfusion in the brain, and we investigated the histological basis of normal-perfusion pressure breakthrough. METHODS Twenty-one Sprague-Dawley rats were randomly divided into a control group (n = 3) and a model group assessed at various time points after the creation of a carotid artery-jugular vein fistula (12 h, n = 3; 24 h, n = 3; 72 h, n = 3; 7 d, n = 3; 21 d, n = 3; 90 d, n = 3). The time courses of the expression of VEGF messenger ribonucleic acid (mRNA) and protein in rat brain were analyzed with semiquantitative reverse transcriptase-polymerase chain reaction and Western blot assays, respectively. Immunohistochemical techniques were used to evaluate VEGF protein localization with rabbit polyclonal anti-rat VEGF, VEGF receptor (VEGFR) expression with rabbit polyclonal antibodies to VEGFR-1 and -2, microvascular density with mouse monoclonal anti-rat CD31, and astrocytic reactivity with polyclonal anti-glial fibrillary acidic protein, in cerebral cortical tissue of the right middle cerebral artery territory. RESULTS Three alternative splicing forms, i.e., VEGF(188), VEGF(164), and VEGF(120), were observed in cerebral cortical tissue of the right middle cerebral artery territory in semiquantitative reverse transcriptase-polymerase chain reaction analyses. VEGF(164) mRNA was the predominant isoform expressed in rat brain. VEGF(188) mRNA and VEGF(120) mRNA were also detected but at very low levels (not statistically significant). Low levels of VEGF(164) mRNA were observed in the control brains. However, VEGF(164) mRNA levels were significantly increased in the model brains at 24 hours postoperatively, peaked by 7 days, decreased by 21 days, and returned to basal levels by 90 days after fistula formation. VEGF protein expression, as measured in Western blot assays, was also increased in rat brains in the model group from 24 hours to 21 days postoperatively but returned to control levels by 90 days after fistula formation. VEGF immunohistochemical analyses indicated that this increased expression was mostly associated with endothelial cells. Consistent with the VEGF protein expression findings, up-regulation of VEGFR-1 but not VEGFR-2 expression on endothelial cells in the model brains was observed. Microvascular density in the rat brains began to increase significantly 7 days after fistula formation in the model group, as assessed immunohistochemically, and the increase was maintained for 90 days. Although no prominent astrocytic reactivity was observed in the rat brains throughout the experiments, there was an absence of astrocytic foot processes surrounding some cerebral capillaries 90 days after fistula formation in the model group. CONCLUSION These results demonstrated that chronic cerebral hypoperfusion could induce sustained up-regulation of VEGF mRNA and protein expression in rat brain, which was correlated with angiogenesis. An absence of corresponding astrocytic reactivity during angiogenesis may be an important factor accounting for structural deficits of the blood-brain barrier and the occurrence of normal-perfusion pressure breakthrough.
Collapse
Affiliation(s)
- Jian Hai
- Department of Neurosurgery, Tongji Hospital, Tongji University, Shanghai, People's Republic of China.
| | | | | | | | | | | |
Collapse
|