Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Neurocrit Care ; 26(1): 48-57, 2017 02.
Article in English | MEDLINE | ID: mdl-27430874

ABSTRACT

BACKGROUND: Early brain injury (EBI) following aneurysmal subarachnoid hemorrhage (SAH) is an important predictor of poor functional outcome, yet the underlying mechanism is not well understood. Animal studies suggest that platelet activation and inflammation with subsequent microthrombosis and ischemia may be a mechanism of EBI. METHODS: A prospective, hypothesis-driven study of spontaneous, SAH patients and controls was conducted. Platelet activation [thromboelastography maximum amplitude (MA)] and inflammation [C-reactive protein (CRP)] were measured serially over time during the first 72 h following SAH onset. Platelet activation and inflammatory markers were compared between controls and SAH patients with mild [Hunt-Hess (HH) 1-3] versus severe (HH 4-5) EBI. The association of these biomarkers with 3-month functional outcomes was evaluated. RESULTS: We enrolled 127 patients (106 SAH; 21 controls). Platelet activation and CRP increased incrementally with worse EBI/HH grade, and both increased over 72 h (all P < 0.01). Both were higher in severe versus mild EBI (MA 68.9 vs. 64.8 mm, P = 0.001; CRP 12.5 vs. 1.5 mg/L, P = 0.003) and compared to controls (both P < 0.003). Patients with delayed cerebral ischemia (DCI) had more platelet activation (66.6 vs. 64.9 in those without DCI, P = 0.02) within 72 h of ictus. At 3 months, death or severe disability was more likely with higher levels of platelet activation (mRS4-6 OR 1.18, 95 % CI 1.05-1.32, P = 0.007) and CRP (mRS4-6 OR 1.02, 95 % CI 1.00-1.03, P = 0.041). CONCLUSIONS: Platelet activation and inflammation occur acutely after SAH and are associated with worse EBI, DCI and poor 3-month functional outcomes. These markers may provide insight into the mechanism of EBI following SAH.


Subject(s)
Brain Injuries , Inflammation/blood , Outcome Assessment, Health Care , Platelet Activation/physiology , Subarachnoid Hemorrhage , Adult , Aged , Aged, 80 and over , Biomarkers , Brain Injuries/blood , Brain Injuries/etiology , Brain Injuries/immunology , Brain Injuries/physiopathology , Female , Humans , Male , Middle Aged , Severity of Illness Index , Subarachnoid Hemorrhage/blood , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/immunology , Subarachnoid Hemorrhage/physiopathology , Young Adult
2.
PLoS One ; 10(8): e0136967, 2015.
Article in English | MEDLINE | ID: mdl-26317208

ABSTRACT

Stroke results in brain tissue damage from ischemia and oxidative stress. Molecular regulators of the protective versus deleterious cellular responses after cerebral ischemia remain to be identified. Here, we show that deletion of Smad1, a conserved transcription factor that mediates canonical bone morphogenetic protein (BMP) signaling, results in neuroprotection in an ischemia-reperfusion (I/R) stroke model. Uninjured mice with conditional deletion of Smad1 in the CNS (Smad1 cKO) displayed upregulation of the reactive astrocyte marker GFAP and hypertrophic morphological changes in astrocytes compared to littermate controls. Additionally, cultured Smad1(-/-) astrocytes exhibited an enhanced antioxidant capacity. When subjected to I/R injury by transient middle cerebral artery occlusion (tMCAO), Smad1 cKO mice showed enhanced neuronal survival and improved neurological recovery at 7 days post-stroke. This neuroprotective phenotype is associated with attenuated reactive astrocytosis and neuroinflammation, along with reductions in oxidative stress, p53 induction, and apoptosis. Our data suggest that Smad1-mediated signaling pathway is involved in stroke pathophysiology and may present a new potential target for stroke therapy.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/prevention & control , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Smad1 Protein/deficiency , Animals , Astrocytes/metabolism , Brain Ischemia/genetics , Brain Ischemia/pathology , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Oxidative Stress , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction , Tumor Suppressor Protein p53/metabolism
3.
Neurol Res ; 37(12): 1054-9, 2015.
Article in English | MEDLINE | ID: mdl-26923576

ABSTRACT

BACKGROUND AND PURPOSE: Inflammation and compromise in structure and function of cerebral parenchymal microvasculature begins early after subarachnoid hemorrhage (SAH). We recently found greater inflammation and greater vascular compromise in male than in female rats following SAH. In this study, we investigated whether this cross-sexual difference in pathology is reflected in expression levels of genes related to vascular inflammation and structural compromise. METHOD: Age-matched male and female rats underwent sham surgery or SAH by endovascular perforation. Early physiology (intracranial pressure (ICP), blood pressure (BP), heart rate, and cerebral blood flow) was monitored. Cerebral RNA was extracted at sacrifice 3 h after surgery and assayed for expression of thrombomodulin (Thbd), endothelial nitric oxide synthase (eNos;Nos3), intracellular adhesion molecule-1 (Icam1), vascular endothelial growth factor (Vegf), interleukin-1beta (Il1ß) tumor necrosis factor-alpha (Tnf-α), and arginine vasopressin (Avp). RESULTS: Increases in ICP and BP at SAH appeared slightly greater in males but the difference did not reach statistical difference, indicating that SAH intensity did not differ significantly between the sexes. Of the seven genes studied two; Tnf-α and Vegf, did not change after injury, while the remainder showed significant responses to SAH. Response of Nos3 and Thbd was markedly different between the sexes, with expression greater in males. CONCLUSION: This study finds that sexual dimorphism is present in the response of some but not all genes to SAH. Since products of genes exhibiting sexual dimorphism have anti-inflammatory activities, our results indicate that previously found sex-based differences in vascular pathology are paralleled by sexually dimorphic changes in gene expression following SAH.


Subject(s)
Gene Expression Regulation/physiology , Sex Characteristics , Subarachnoid Hemorrhage , Animals , Arginine Vasopressin/genetics , Arginine Vasopressin/metabolism , Blood Pressure/physiology , Cerebrovascular Circulation , Disease Models, Animal , Female , Heart Rate/physiology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Intracranial Pressure , Male , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/genetics , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/physiopathology , Thrombomodulin/genetics , Thrombomodulin/metabolism , Time Factors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
4.
Acta Neurochir Suppl ; 120: 23-8, 2015.
Article in English | MEDLINE | ID: mdl-25366594

ABSTRACT

The first 72 h after aneurysmal subarachnoid hemorrhage (SAH) is a critical period for the patient. Most of the deaths in the SAH patient population occur during this time, and a number of key events activate and trigger mechanisms that not only contribute to early brain injury but evolve over time and participate in the delayed complications. This review highlights the contribution of key events to the early brain injury and to overall outcome after SAH.


Subject(s)
Brain Chemistry/physiology , Brain Ischemia , Hyperemia , Intracranial Hypertension , Subarachnoid Hemorrhage , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Disease Progression , Humans , Hyperemia/metabolism , Hyperemia/pathology , Hyperemia/physiopathology , Intracranial Hypertension/metabolism , Intracranial Hypertension/pathology , Intracranial Hypertension/physiopathology , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/pathology , Subarachnoid Hemorrhage/physiopathology
5.
Acta Neurochir Suppl ; 120: 321-4, 2015.
Article in English | MEDLINE | ID: mdl-25366645

ABSTRACT

The rat endovascular perforation model is considered the closest replica of human condition. Since its development, this model has been extensively used to study early brain injury after subarachnoid hemorrhage (SAH). However, like any other animal model, it has advantages and limitations. The following is a brief review of the rat endovascular perforation SAH model. One section is dedicated to technical considerations that can be used to overcome the model limitations.


Subject(s)
Cerebrovascular Circulation/physiology , Disease Models, Animal , Endovascular Procedures/methods , Rats , Subarachnoid Hemorrhage/physiopathology , Animals
6.
J Neurol Neurosurg Psychiatry ; 86(1): 71-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24715224

ABSTRACT

OBJECTIVE: To determine if ischaemia is a mechanism of early brain injury at the time of aneurysm rupture in subarachnoid haemorrhage (SAH) and if early MRI ischaemia correlates with admission clinical status and functional outcome. METHODS: In a prospective, hypothesis-driven study patients with SAH underwent MRI within 0-3 days of ictus (prior to vasospasm) and a repeat MRI (median 7 days). The volume and number of diffusion weighted imaging (DWI) positive/apparent diffusion coefficient (ADC) dark lesions on acute MRI were quantitatively assessed. The association of early ischaemia, admission clinical status, risk factors and 3-month outcome were analysed. RESULTS: In 61 patients with SAH, 131 MRI were performed. Early ischaemia occurred in 40 (66%) with a mean DWI/ADC volume 8.6 mL (0-198 mL) and lesion number 4.3 (0-25). The presence of any early DWI/ADC lesion and increasing lesion volume were associated with worse Hunt-Hess grade, Glasgow Coma Scale score and Acute Physiology and Chronic Health Evaluation II physiological subscores (all p<0.05). Early DWI/ADC lesions significantly predicted increased number and volume of infarcts on follow-up MRI (p<0.005). At 3 months, early DWI/ADC lesion volume was significantly associated with higher rates of death (21% vs. 3%, p=0.031), death/severe disability (modified Rankin Scale 4-6; 53% vs. 15%, p=0.003) and worse Barthel Index (70 vs. 100, p=0.004). After adjusting for age, Hunt-Hess grade and aneurysm size, early infarct volume correlated with death/severe disability (adjusted OR 1.7, 95% CI 1.0 to 3.2, p=0.066). CONCLUSIONS: Early ischaemia is related to poor acute neurological status after SAH and predicts future ischaemia and worse functional outcomes. Treatments addressing acute ischaemia should be evaluated for their effect on outcome.


Subject(s)
Brain Injuries/pathology , Brain Ischemia/pathology , Subarachnoid Hemorrhage, Traumatic/pathology , Adult , Aged , Aged, 80 and over , Brain Injuries/complications , Brain Ischemia/complications , Diffusion Magnetic Resonance Imaging , Female , Humans , Intracranial Aneurysm/complications , Intracranial Aneurysm/pathology , Male , Middle Aged , Neuroimaging , Outcome Assessment, Health Care , Prospective Studies , Risk Factors , Severity of Illness Index , Subarachnoid Hemorrhage, Traumatic/complications , Subarachnoid Hemorrhage, Traumatic/mortality
7.
Transl Stroke Res ; 5(6): 660-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25213427

ABSTRACT

Experimental animal models of aneurysmal subarachnoid hemorrhage (SAH) have provided a wealth of information on the mechanisms of brain injury. The rat endovascular perforation (EVP) model replicates the early pathophysiology of SAH and hence is frequently used to study early brain injury following SAH. This paper presents a brief review of historical development of the EVP model and details the technique used to create SAH and considerations necessary to overcome technical challenges.


Subject(s)
Disease Models, Animal , Endovascular Procedures , Subarachnoid Hemorrhage/etiology , Animals , Carotid Artery, Internal/surgery , Female , Male , Rats , Rats, Sprague-Dawley , Rats, Wistar , Subarachnoid Hemorrhage/physiopathology
9.
PLoS One ; 8(11): e80101, 2013.
Article in English | MEDLINE | ID: mdl-24250830

ABSTRACT

Aneurysmal subarachnoid hemorrhage (SAH) carries high early patient mortality. More women than men suffer from SAH and the average age of female SAH survivors is greater than that of male survivors; however, the overall mortality and neurological outcomes are not better in males despite their younger age. This pattern suggests the possibility of gender differences in the severity of initial impact and/or in subsequent pathophysiology. We explored gender differences in survival and pathophysiology following subarachnoid hemorrhage induced in age-matched male and female rats by endovascular puncture. Intracranial pressure (ICP), cerebral blood flow (CBF), blood pressure (BP) and cerebral perfusion pressure (CPP) were recorded at and after induction of SAH. Animals were sacrificed 3 hours after lesion and studied for subarachnoid hematoma size, vascular pathology (collagen and endothelium immunostaining), inflammation (platelet and neutrophil immunostaining), and cell death (TUNEL assay). In a second cohort, 24-hour survival was determined. Subarachnoid hematoma, post-hemorrhage ICP peak, BP elevation, reduction in CPP, intraluminal platelet aggregation and neutrophil accumulation, loss of vascular collagen, and neuronal and non-neuronal cell death were greater in male than in female rats. Hematoma size did not correlate with the number of apoptotic cells, platelet aggregates or neutrophil. The ICP peak correlated with hematoma size and with number of apoptotic cells but not with platelet aggregates and neutrophil number. This suggests that the intensity of ICP rise at SAH influences the severity of apoptosis but not of inflammation. Mortality was markedly greater in males than females. Our data demonstrate that in rats gender influences the initial impact of SAH causing greater bleed and early injury in males as compared to females.


Subject(s)
Brain/physiopathology , Inflammation/physiopathology , Sex Characteristics , Subarachnoid Hemorrhage/physiopathology , Animals , Apoptosis , Blood Platelets/pathology , Blood Pressure , Brain/blood supply , Female , Humans , Intracranial Pressure , Male , Neurons/pathology , Neutrophils/pathology , Rats , Regional Blood Flow
10.
Stroke Res Treat ; 2013: 615154, 2013.
Article in English | MEDLINE | ID: mdl-23878760

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.

11.
Acta Neurochir Suppl ; 115: 199-205, 2013.
Article in English | MEDLINE | ID: mdl-22890669

ABSTRACT

Most subarachnoid hemorrhage (SAH) patients exhibit clinical signs of cerebral ischemia at admission but no angiographic vasospasm. Consequently, the source of early cerebral ischemia is not understood. Parenchymal microvessels may contribute to early cerebral ischemia, but the low resolution of current imaging has prevented their analysis in SAH patients. Animal studies demonstrated that early after SAH structure and function of parenchymal vessels are compromised to the level that may very well contribute to early ischemia. We review these studies.


Subject(s)
Cerebral Cortex/pathology , Microvessels/physiopathology , Subarachnoid Hemorrhage/pathology , Animals , Disease Models, Animal , Encephalitis/etiology , Encephalitis/pathology , Humans , Microvessels/ultrastructure , Nervous System Diseases/etiology , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Oxidative Stress/physiology , Signal Transduction/physiology , Subarachnoid Hemorrhage/complications
12.
Prog Neurobiol ; 97(1): 14-37, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22414893

ABSTRACT

Aneurysmal subarachnoid hemorrhage (aSAH) is a medical emergency that accounts for 5% of all stroke cases. Individuals affected are typically in the prime of their lives (mean age 50 years). Approximately 12% of patients die before receiving medical attention, 33% within 48 h and 50% within 30 days of aSAH. Of the survivors 50% suffer from permanent disability with an estimated lifetime cost more than double that of an ischemic stroke. Traditionally, spasm that develops in large cerebral arteries 3-7 days after aneurysm rupture is considered the most important determinant of brain injury and outcome after aSAH. However, recent studies show that prevention of delayed vasospasm does not improve outcome in aSAH patients. This finding has finally brought in focus the influence of early brain injury on outcome of aSAH. A substantial amount of evidence indicates that brain injury begins at the aneurysm rupture, evolves with time and plays an important role in patients' outcome. In this manuscript we review early brain injury after aSAH. Due to the early nature, most of the information on this injury comes from animals and few only from autopsy of patients who died within days after aSAH. Consequently, we began with a review of animal models of early brain injury, next we review the mechanisms of brain injury according to the sequence of their temporal appearance and finally we discuss the failure of clinical translation of therapies successful in animal models of aSAH.


Subject(s)
Brain Injuries/etiology , Subarachnoid Hemorrhage/complications , Age Factors , Humans , Time Factors
13.
Neurosci Lett ; 512(1): 6-11, 2012 Mar 14.
Article in English | MEDLINE | ID: mdl-22306092

ABSTRACT

Brain injury begins early after aneurysmal subarachnoid hemorrhage (SAH). Although cell death via apoptosis and necrosis is known to be present in brain 24 h after SAH, it is not known how soon after SAH cell death begins. We have previously described structural changes in rat brain microvessels 10 min after induction of SAH by endovascular puncture. This study examined brain for evidence of cell death beginning 10 min after induction of SAH. Cleaved caspase-3 (cl-caspase-3) staining was evident in vascular and parenchymal cells at 10 min after SAH and was significantly greater than in time-matched, sham-operated controls. The number of cl-caspase-3 positive cells was increased further at 24 h after SAH. TUNEL assay revealed apoptotic cells present at 10 min, with substantially more at 24 h after SAH. Scattered Fluoro-Jade positive neurons appeared at 1h after SAH and their number increased with time. At 1 h Fluoro-Jade positive neurons were present in cortical and subcortical regions but not in hippocampus; at 24h they were also present in hippocampus and were significantly greater in the hemisphere ipsilateral to the vascular puncture. No Fluoro-Jade staining was present in shams. These data demonstrate an early activation of endothelial and parenchymal cells apoptosis and neuronal necrosis after SAH and identifies endpoints that can be targeted to reduce early brain injury after SAH.


Subject(s)
Apoptosis , Subarachnoid Hemorrhage/pathology , Animals , Brain/enzymology , Brain/pathology , Caspase 3/analysis , Male , Necrosis , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/enzymology
14.
J Neuroinflammation ; 8: 103, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21854561

ABSTRACT

BACKGROUND: Subarachnoid haemorrhage (SAH) elicits rapid pathological changes in the structure and function of parenchymal vessels (≤ 100 µm). The role of neutrophils in these changes has not been determined. This study investigates the role of neutrophils in early microvascular changes after SAH METHOD: Rats were either untreated, treated with vinblastine or anti-polymorphonuclear (PMN) serum, which depletes neutrophils, or treated with pyrrolidine dithiocarbamate (PDTC), which limits neutrophil activity. SAH was induced by endovascular perforation. Neutrophil infiltration and the integrity of vascular endothelium and basement membrane were assessed immunohistochemically. Vascular collagenase activity was assessed by in situ zymography. RESULTS: Vinblastine and anti-PMN serum reduced post-SAH accumulation of neutrophils in cerebral vessels and in brain parenchyma. PDTC increased the neutrophil accumulation in cerebral vessels and decreased accumulation in brain parenchyma. In addition, each of the three agents decreased vascular collagenase activity and post-SAH loss of vascular endothelial and basement membrane immunostaining. CONCLUSIONS: Our results implicate neutrophils in early microvascular injury after SAH and indicate that treatments which reduce neutrophil activity can be beneficial in limiting microvascular injury and increasing survival after SAH.


Subject(s)
Microcirculation , Neutrophil Activation , Neutrophils/physiology , Subarachnoid Hemorrhage/pathology , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Biomarkers/metabolism , Brain/anatomy & histology , Brain/drug effects , Brain/metabolism , Brain/pathology , Capillary Permeability , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Humans , Male , Neutrophil Infiltration , Neutrophils/cytology , Neutrophils/drug effects , Proline/analogs & derivatives , Proline/metabolism , Proline/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/physiopathology , Thiocarbamates/metabolism , Thiocarbamates/pharmacology , Tubulin Modulators/pharmacology , Vinblastine/pharmacology
15.
Mol Neurobiol ; 43(1): 27-40, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21161614

ABSTRACT

Delayed vasospasm that develops 3-7 days after aneurysmal subarachnoid hemorrhage (SAH) has traditionally been considered the most important determinant of delayed ischemic injury and poor outcome. Consequently, most therapies against delayed ischemic injury are directed towards reducing the incidence of vasospasm. The clinical trials based on this strategy, however, have so far claimed limited success; the incidence of vasospasm is reduced without reduction in delayed ischemic injury or improvement in the long-term outcome. This fact has shifted research interest to the early brain injury (first 72 h) evoked by SAH. In recent years, several pathological mechanisms that activate within minutes after the initial bleed and lead to early brain injury are identified. In addition, it is found that many of these mechanisms evolve with time and participate in the pathogenesis of delayed ischemic injury and poor outcome. Therefore, a therapy or therapies focused on these early mechanisms may not only prevent the early brain injury but may also help reduce the intensity of later developing neurological complications. This manuscript reviews the pathological mechanisms of early brain injury after SAH and summarizes the status of current therapies.


Subject(s)
Brain Injuries/etiology , Subarachnoid Hemorrhage/complications , Vasospasm, Intracranial/etiology , Animals , Brain Injuries/pathology , Brain Injuries/prevention & control , Brain Injuries/therapy , Cell Death , Cerebrovascular Circulation , Clinical Trials as Topic , Humans , Regional Blood Flow , Stroke/etiology , Stroke/pathology , Stroke/prevention & control , Stroke/therapy , Subarachnoid Hemorrhage/pathology , Subarachnoid Hemorrhage/therapy , Time Factors , Treatment Outcome , Vasospasm, Intracranial/pathology , Vasospasm, Intracranial/prevention & control , Vasospasm, Intracranial/therapy
16.
Acta Neurochir Suppl ; 110(Pt 1): 49-55, 2011.
Article in English | MEDLINE | ID: mdl-21116914

ABSTRACT

During the last decade much effort has been invested in understanding the events that occur early after SAH. It is now widely accepted that these early events not only participate in the early ischemic injury but also set the stage for the pathogenesis of delayed vasospasm. That early cerebral ischemia occurs after SAH is documented in both experimental SAH and in human autopsy studies; however, angiographic evidence for vasoconstriction early after SAH is lacking and the source of early ischemic injury is therefore unclear. Recently, the cerebral microvasculature has been identified as an early target of SAH. Changes in the anatomical structure of cerebral microvessels, sufficient to cause functional deficits, are found early after experimental SAH. These changes may explain cerebral ischemia in human in the absence of angiographic evidence of large vessel vasoconstriction. This paper summarizes known alterations in cerebral microvasculature during the first 48 h after SAH.


Subject(s)
Blood Vessels/physiopathology , Subarachnoid Hemorrhage/complications , Vasospasm, Intracranial/etiology , Vasospasm, Intracranial/pathology , Animals , Basement Membrane/physiopathology , Blood Platelets/metabolism , Endothelium/physiopathology , Humans
17.
Acta Neurochir Suppl ; 110(Pt 1): 99-103, 2011.
Article in English | MEDLINE | ID: mdl-21116923

ABSTRACT

Nitric Oxide (NO) is the major regulator of cerebral blood flow. In addition, it inhibits platelet adherence and aggregation, reduces adherence of leukocytes to the endothelium, and suppresses vessel injury. NO is produced on demand by nitric oxide synthase and has a very short half life. Hence maintenance of its cerebral level is crucial for normal vascular physiology. Time dependent alterations in cerebral NO level and the enzymes responsible for its synthesis are found after subarachnoid hemorrhage (SAH). Cerebral NO level decreases, recovers and increases within the first 24 h after SAH. Each change in cerebral NO level elicits a different pathological response form already compromised brain. These response range from constriction, platelet aggregation and vascular injury that occurs during the early hours and delayed occurring vasospasm, neuronal and axonal damage. This review summarizes the underlying mechanism and the consequence of alteration in cerebral NO level on brain during the first 72 h after SAH.


Subject(s)
Brain Injuries/etiology , Brain Injuries/metabolism , Nitric Oxide/metabolism , Subarachnoid Hemorrhage/complications , Animals , Brain/metabolism , Brain Injuries/pathology , Humans , Time Factors
18.
Brain Res ; 1354: 179-87, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20654597

ABSTRACT

The pathophysiology of early ischemic injury after aneurysmal subarachnoid hemorrhage (SAH) is not understood. This study examined the acute effect of endovascular puncture-induced SAH on parenchymal vessel function in rat, using intravascular fluorescent tracers to assess flow and vascular permeability and immunostaining to assess structural integrity and to visualize platelet aggregates. In sham-operated animals, vessels were well filled with tracer administered 10s before sacrifice, and parenchymal escape of tracer was rare. At ten minutes and three hours after hemorrhage, patches of poor vascular filling were distributed throughout the forebrain. Close examination of these regions revealed short segments of narrowed diameter along many profiles. Most vascular profiles with reduced perfusion contained platelet aggregates and in addition showed focal loss of collagen IV, a principal component of basal lamina. In contrast, vessels were well filled at 24h post-hemorrhage, indicating that vascular perfusion had recovered. Parenchymal escape of intravascular tracer was detected at 10 min post-hemorrhage and later as plumes of fluorescence emanating into parenchyma from restricted microvascular foci. These data demonstrate that parenchymal microvessels are compromised in function by 10 min after SAH and identify focal microvascular constriction and local accumulation of luminal platelet aggregates as potential initiators of that compromise.


Subject(s)
Blood Vessels/physiopathology , Brain/physiopathology , Capillary Permeability/physiology , Platelet Aggregation/physiology , Subarachnoid Hemorrhage/physiopathology , Analysis of Variance , Animals , Blood Platelets/metabolism , Blood Vessels/metabolism , Brain/blood supply , Brain/metabolism , Cerebrovascular Circulation/physiology , Collagen Type IV/metabolism , Immunohistochemistry , Platelet Count , Random Allocation , Rats , Rats, Sprague-Dawley
19.
J Neurosurg ; 113(4): 826-34, 2010 Oct.
Article in English | MEDLINE | ID: mdl-19895201

ABSTRACT

OBJECT: The role of adenosine A(2A) receptors in the early vascular response after subarachnoid hemorrhage (SAH) is unknown. In other forms of cerebral ischemia both activation and inhibition of A(2A) receptors is reported to be beneficial. However, these studies mainly used pharmacological receptor modulation, and most of the agents available exhibit low specificity. The authors used adenosine A(2A) receptor knockout mice to study the role of A(2A) receptors in the early vascular response to SAH. METHODS: Subarachnoid hemorrhage was induced in wild-type mice (C57BL/6) and A(2A) receptor knockout mice by endovascular puncture. Cerebral blood flow, intracranial pressure, and blood pressure were recorded, and cerebral perfusion pressure was deduced. Animals were sacrificed at 1, 3, or 6 hours after SAH or sham surgery. Coronal brain sections were immunostained for Type IV collagen, the major protein of the basal lamina. The internal diameter of major cerebral arteries and the area fraction of Type IV collagen-positive microvessels (< 100 µm) were determined. RESULTS: The initial increase in intracranial pressure and decrease in cerebral perfusion pressure at SAH induction was similar in both types of mice, but cerebral blood flow decline was significantly smaller in A(2A) receptor knockout mice as compared with wild-type cohorts. The internal diameter of major cerebral vessels decreased progressively after SAH. The extent of diameter reduction was significantly less in A(2A) receptor knockout mice than in wild-type mice. Type IV collagen immunostaining decreased progressively after SAH. This decrease was significantly less in A(2A) receptor knockout mice than in wild-type mice. CONCLUSIONS: These results demonstrate that global inactivation of A(2A) receptors decreases the intensity of the early vascular response to SAH. Early inhibition of A(2A) receptors after SAH might reduce cerebral injury.


Subject(s)
Brain Ischemia/metabolism , Receptor, Adenosine A2A/metabolism , Subarachnoid Hemorrhage/metabolism , Animals , Blood Gas Analysis , Blood Pressure/physiology , Brain/pathology , Brain Ischemia/pathology , Capillaries/metabolism , Capillaries/pathology , Cerebral Arteries/metabolism , Cerebral Arteries/pathology , Cerebrovascular Circulation/physiology , Collagen Type IV/metabolism , Fluorescent Antibody Technique , Intracranial Pressure/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Adenosine A2A/genetics , Subarachnoid Hemorrhage/pathology
20.
J Neurosurg ; 106(2): 321-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17410718

ABSTRACT

OBJECT: Structural changes in brain parenchymal vessels occur within minutes after subarachnoid hemorrhage (SAH). These changes include platelet aggregation, activation of vascular collagenases, and destruction of perivascular collagen IV. Because collagen IV is an important component of the basal lamina, the authors attempted to further define changes in vascular structure (length and luminal diameter) and their relationship to vascular permeability immediately after SAH. In addition, the authors explored whether such alterations were attenuated by administration of a nitric oxide (NO) donor. METHODS: Endovascular perforation was used to induce SAH in rats. Two sets of experiments were performed. The first established changes in vascular structure and permeability (collagen IV and endothelial barrier antigen [EBA] dual immunofluorescence) during the first 24 hours after SAH. In the second, the investigators examined the effects of an NO donor on vascular structure, permeability, and collagenase activity (in situ zymography). In this second study, animals received intravenous infusion of the NO donor S-nitrosoglutathione (GSNO, 1 microM/8 microl/min) 15 minutes after induction of SAH and were killed 3 hours after SAH onset. Controls were naive unoperated animals for the first study and saline-infused SAH animals for the second. The authors found a time-dependent decrease in area fraction, length, and luminal diameter of collagen IV- and EBA-immunofluorescent vessels after SAH. The greatest change occurred at 3 hours after onset of SAH. Administration of GSNO was associated with striking preservation of collagen IV and EBA immunofluorescence compared with saline treatment. Zymography indicated decreased collagenase activity in GSNO-treated SAH animals compared with saline-treated SAH animals. CONCLUSIONS: These results demonstrate changes in the structure and permeability of brain parenchymal microvessels after SAH and their reversal by treatment with an NO donor.


Subject(s)
Brain/blood supply , Nitric Oxide Donors/therapeutic use , S-Nitrosoglutathione/therapeutic use , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/prevention & control , Animals , Brain/pathology , Brain/physiopathology , Capillary Permeability/physiology , Male , Microcirculation/pathology , Microcirculation/physiopathology , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...