Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Neurocrit Care ; 26(1): 48-57, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27430874

RESUMO

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.


Assuntos
Lesões Encefálicas , Inflamação/sangue , Avaliação de Resultados em Cuidados de Saúde , Ativação Plaquetária/fisiologia , Hemorragia Subaracnóidea , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores , Lesões Encefálicas/sangue , Lesões Encefálicas/etiologia , Lesões Encefálicas/imunologia , Lesões Encefálicas/fisiopatologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Hemorragia Subaracnóidea/sangue , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/imunologia , Hemorragia Subaracnóidea/fisiopatologia , Adulto Jovem
2.
J Neurol Neurosurg Psychiatry ; 86(1): 71-8, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24715224

RESUMO

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.


Assuntos
Lesões Encefálicas/patologia , Isquemia Encefálica/patologia , Hemorragia Subaracnoídea Traumática/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Lesões Encefálicas/complicações , Isquemia Encefálica/complicações , Imagem de Difusão por Ressonância Magnética , Feminino , Humanos , Aneurisma Intracraniano/complicações , Aneurisma Intracraniano/patologia , Masculino , Pessoa de Meia-Idade , Neuroimagem , Avaliação de Resultados em Cuidados de Saúde , Estudos Prospectivos , Fatores de Risco , Índice de Gravidade de Doença , Hemorragia Subaracnoídea Traumática/complicações , Hemorragia Subaracnoídea Traumática/mortalidade
3.
Acta Neurochir Suppl ; 120: 321-4, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25366645

RESUMO

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.


Assuntos
Circulação Cerebrovascular/fisiologia , Modelos Animais de Doenças , Procedimentos Endovasculares/métodos , Ratos , Hemorragia Subaracnóidea/fisiopatologia , Animais
4.
Acta Neurochir Suppl ; 120: 23-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25366594

RESUMO

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.


Assuntos
Química Encefálica/fisiologia , Isquemia Encefálica , Hiperemia , Hipertensão Intracraniana , Hemorragia Subaracnóidea , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Progressão da Doença , Humanos , Hiperemia/metabolismo , Hiperemia/patologia , Hiperemia/fisiopatologia , Hipertensão Intracraniana/metabolismo , Hipertensão Intracraniana/patologia , Hipertensão Intracraniana/fisiopatologia , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/patologia , Hemorragia Subaracnóidea/fisiopatologia
5.
Acta Neurochir Suppl ; 115: 199-205, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22890669

RESUMO

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.


Assuntos
Córtex Cerebral/patologia , Microvasos/fisiopatologia , Hemorragia Subaracnóidea/patologia , Animais , Modelos Animais de Doenças , Encefalite/etiologia , Encefalite/patologia , Humanos , Microvasos/ultraestrutura , Doenças do Sistema Nervoso/etiologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Estresse Oxidativo/fisiologia , Transdução de Sinais/fisiologia , Hemorragia Subaracnóidea/complicações
6.
J Neuroinflammation ; 8: 103, 2011 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-21854561

RESUMO

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.


Assuntos
Microcirculação , Ativação de Neutrófilo , Neutrófilos/fisiologia , Hemorragia Subaracnóidea/patologia , Animais , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Biomarcadores/metabolismo , Encéfalo/anatomia & histologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Permeabilidade Capilar , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Humanos , Masculino , Infiltração de Neutrófilos , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Prolina/análogos & derivados , Prolina/metabolismo , Prolina/farmacologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/fisiopatologia , Tiocarbamatos/metabolismo , Tiocarbamatos/farmacologia , Moduladores de Tubulina/farmacologia , Vimblastina/farmacologia
7.
Acta Neurochir Suppl ; 110(Pt 1): 49-55, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21116914

RESUMO

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.


Assuntos
Vasos Sanguíneos/fisiopatologia , Hemorragia Subaracnóidea/complicações , Vasoespasmo Intracraniano/etiologia , Vasoespasmo Intracraniano/patologia , Animais , Membrana Basal/fisiopatologia , Plaquetas/metabolismo , Endotélio/fisiopatologia , Humanos
8.
Acta Neurochir Suppl ; 110(Pt 1): 99-103, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21116923

RESUMO

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.


Assuntos
Lesões Encefálicas/etiologia , Lesões Encefálicas/metabolismo , Óxido Nítrico/metabolismo , Hemorragia Subaracnóidea/complicações , Animais , Encéfalo/metabolismo , Lesões Encefálicas/patologia , Humanos , Fatores de Tempo
9.
J Neurosurg ; 106(2): 321-9, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17410718

RESUMO

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.


Assuntos
Encéfalo/irrigação sanguínea , Doadores de Óxido Nítrico/uso terapêutico , S-Nitrosoglutationa/uso terapêutico , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/prevenção & controle , Animais , Encéfalo/patologia , Encéfalo/fisiopatologia , Permeabilidade Capilar/fisiologia , Masculino , Microcirculação/patologia , Microcirculação/fisiopatologia , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/patologia , Fatores de Tempo
10.
Neurol Res ; 28(4): 381-98, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16759442

RESUMO

Brain injury after subarachnoid hemorrhage (SAH) is a biphasic event with an acute ischemic insult at the time of the initial bleed and secondary events such as cerebral vasospasm 3 to 7 days later. Although much has been learned about the delayed effects of SAH, less is known about the mechanisms of acute SAH-induced injury. Distribution of blood in the subarachnoid space, elevation of intracranial pressure, reduced cerebral perfusion and cerebral blood flow (CBF) initiates the acute injury cascade. Together they lead to direct microvascular injury, plugging of vessels and release of vasoactive substances by platelet aggregates, alterations in the nitric oxide (NO)/nitric oxide synthase (NOS) pathways and lipid peroxidation. This review will summarize some of these mechanisms that contribute to acute cerebral injury after SAH.


Assuntos
Lesões Encefálicas/etiologia , Hemorragia Subaracnóidea/complicações , Animais , Circulação Cerebrovascular/fisiologia , Humanos , Pressão Intracraniana/fisiologia , Peroxidação de Lipídeos/fisiologia , Modelos Biológicos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Agregação Plaquetária/fisiologia , Vasoespasmo Intracraniano/etiologia , Vasoespasmo Intracraniano/fisiopatologia
11.
J Neurosurg ; 102(6): 1094-100, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16028769

RESUMO

OBJECT: The mechanisms underlying acute cerebral ischemia after subarachnoid hemorrhage (SAH) are not well established. Platelets aggregate within major cerebral vessels hours after SAH, but this has not been studied in the microvasculature. Platelet aggregates within the microvasculature could mechanically obstruct the lumen and initiate events that injure vessel structure. In the present study the authors examined the hypothesis that platelets aggregate within the cerebral microvasculature acutely after SAH. METHODS: Subarachnoid hemorrhage was induced in the rat by using the endovascular perforation model. The animals were killed between 10 minutes and 48 hours after SAH. Immunostaining for the platelet surface receptor glycoprotein (GP)IIb/IIIa, which mediates platelet aggregation, was used to detect platelet aggregation. Sham-operated animals were used as controls. The GPIIb/IIIa immunoreactive platelet aggregates were abundant in the microvasculature of the basal and frontal cortex, striatum, and hippocampus 10 minutes after SAH. These aggregates decreased in number from 1 to 6 hours post-SAH and then increased to a peak at 24 hours. No immunoreactive aggregates were observed 48 hours after SAH. CONCLUSIONS: The data indicate that widespread platelet aggregation occurs very rapidly in response to SAH followed by a decrease within 6 hours and a subsequent increase 24 hours after SAH. Microvascular platelet aggregates may contribute to decreased cerebral blood flow and ischemic injury after SAH via a number of mechanisms.


Assuntos
Isquemia Encefálica/fisiopatologia , Circulação Cerebrovascular , Agregação Plaquetária , Hemorragia Subaracnóidea/fisiopatologia , Doença Aguda , Animais , Plaquetas/metabolismo , Pressão Sanguínea , Isquemia Encefálica/sangue , Pressão Intracraniana , Masculino , Microcirculação , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/sangue
12.
Neurol Res ; 37(12): 1054-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26923576

RESUMO

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.


Assuntos
Regulação da Expressão Gênica/fisiologia , Caracteres Sexuais , Hemorragia Subaracnóidea , Animais , Arginina Vasopressina/genética , Arginina Vasopressina/metabolismo , Pressão Sanguínea/fisiologia , Circulação Cerebrovascular , Modelos Animais de Doenças , Feminino , Frequência Cardíaca/fisiologia , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Pressão Intracraniana , Masculino , Fosfopiruvato Hidratase/genética , Fosfopiruvato Hidratase/metabolismo , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/genética , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/fisiopatologia , Trombomodulina/genética , Trombomodulina/metabolismo , Fatores de Tempo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
PLoS One ; 10(8): e0136967, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26317208

RESUMO

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.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevenção & controle , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Proteína Smad1/deficiência , Animais , Astrócitos/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Células Cultivadas , Modelos Animais de Doenças , Regulação da Expressão Gênica , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Camundongos , Estresse Oxidativo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo
14.
Neurosurgery ; 55(3): 671-7; discussion 677-8, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15335435

RESUMO

OBJECTIVE: Subarachnoid hemorrhage (SAH) is associated with acute decreases and subsequent recovery of cerebral nitric oxide (NO) levels, but the mechanisms of these alterations are not known. In this study, we measured NO synthase (NOS) protein and kinetics to determine its involvement in the alterations of cerebral NO levels after SAH. METHODS: The endovascular rat model of SAH was used. The number of NOS-1 (neuronal) and NOS-2 (inducible)-positive cells (0-96 h) was determined by counting immunoreactive cells in 8-microm cryostat sections. The tissue content of active NOS and its kinetic parameters were studied with an enzymatic l-citrulline assay. RESULTS: The number of NOS-1-positive cells increased between 1 and 3 hours after SAH, decreased to and below control values at 6 and 72 hours after SAH, and increased to control values 96 hours after SAH. The number of NOS-2-positive cells increased 1 hour after SAH, decreased to control values at 24 hours, and increased above control values 96 hours after SAH. The Michaelis-Menten kinetic parameters (V(max), K(m), slope) of NOS remained unchanged at 10 and 90 minutes after SAH. CONCLUSION: NOS-1 and -2 proteins undergo a triphasic alteration after SAH, whereas the amount of active NOS and its kinetic parameters remain unchanged during the first 90 minutes after SAH. Depletion of NOS is not involved in the acute alterations of cerebral NO levels after SAH.


Assuntos
Encéfalo/enzimologia , Óxido Nítrico Sintase/fisiologia , Óxido Nítrico/metabolismo , Hemorragia Subaracnóidea/enzimologia , Animais , Encéfalo/patologia , Isquemia Encefálica/enzimologia , Isquemia Encefálica/patologia , Contagem de Células , Técnicas Imunoenzimáticas , Cinética , Masculino , Neurônios/enzimologia , Neurônios/patologia , Óxido Nítrico Sintase Tipo I , Óxido Nítrico Sintase Tipo II , Ratos , Ratos Sprague-Dawley , Valores de Referência , Hemorragia Subaracnóidea/patologia
15.
J Neurosurg ; 101(4): 633-40, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15481718

RESUMO

OBJECT: Aneurysmal subarachnoid hemorrhage (SAH) causes acute and delayed ischemic brain injuries. The mechanisms of acute ischemic injury following SAH are poorly understood, although an acute increase in microvascular permeability has been noted. The integrity of cerebral microvessels is maintained in part by components of basal lamina: collagen IV, elastin, lamina, and so forth. Destruction of basal lamina components by collagenases and matrix metalloproteinases (MMPs), especially MMP-9, has been known to occur in other ischemic models. The authors assessed the integrity of cerebral microvasculature after acute SAH by examining collagen IV and MMP-9 levels and collagenase activity in the microvessels. METHODS: Subarachnoid hemorrhage was induced in rats through endovascular perforation of the intracranial bifurcation of the internal carotid artery. Animals were killed 10 minutes to 48 hours after SAH or sham operation (time-matched controls). Levels of collagen IV and MMP-9 were studied in the microvasculature by performing immunoperoxidase and immunofluorescence staining, and collagenase activity was assessed by in situ zymography. Little change occurred in collagen IV and MMP-9 immunostaining or collagenase activity at 10 minutes or 1 hour after SAH. Starting 3 hours after SAH, collagen IV immunostaining was reduced or eliminated along segments of microvessels whereas MMP-9 staining was segmentally increased. These effects reached a maximum at 6 hours and returned toward those values in sham-operated controls at 48 hours. CONCLUSIONS: Results of this study demonstrated an acute loss of collagen IV from the cerebral microvasculature after SAH and indicated that MMP-9 contributes to this event. The loss of collagen IV might contribute to the known failure of the blood-brain barrier after SAH.


Assuntos
Membrana Basal/irrigação sanguínea , Membrana Basal/patologia , Isquemia Encefálica/etiologia , Aneurisma Intracraniano/complicações , Hemorragia Subaracnóidea/complicações , Doença Aguda , Animais , Colágeno Tipo IV/análise , Modelos Animais de Doenças , Masculino , Metaloproteinase 9 da Matriz/análise , Microcirculação , Ratos , Ratos Sprague-Dawley
16.
Transl Stroke Res ; 5(6): 660-8, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25213427

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Procedimentos Endovasculares , Hemorragia Subaracnóidea/etiologia , Animais , Artéria Carótida Interna/cirurgia , Feminino , Masculino , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Hemorragia Subaracnóidea/fisiopatologia
17.
Stroke Res Treat ; 2013: 615154, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23878760

RESUMO

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.

18.
PLoS One ; 8(11): e80101, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24250830

RESUMO

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.


Assuntos
Encéfalo/fisiopatologia , Inflamação/fisiopatologia , Caracteres Sexuais , Hemorragia Subaracnóidea/fisiopatologia , Animais , Apoptose , Plaquetas/patologia , Pressão Sanguínea , Encéfalo/irrigação sanguínea , Feminino , Humanos , Pressão Intracraniana , Masculino , Neurônios/patologia , Neutrófilos/patologia , Ratos , Fluxo Sanguíneo Regional
19.
Neurosci Lett ; 512(1): 6-11, 2012 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-22306092

RESUMO

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.


Assuntos
Apoptose , Hemorragia Subaracnóidea/patologia , Animais , Encéfalo/enzimologia , Encéfalo/patologia , Caspase 3/análise , Masculino , Necrose , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/enzimologia
20.
Prog Neurobiol ; 97(1): 14-37, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22414893

RESUMO

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.


Assuntos
Lesões Encefálicas/etiologia , Hemorragia Subaracnóidea/complicações , Fatores Etários , Humanos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA