Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Neurol Neurosurg ; 228: 107704, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37003100

RESUMEN

OBJECTIVE: The course of the vertebral artery and its relationship to the C0-1-2 structures render it particularly vulnerable to mechanical trauma. In the present study, we investigated the course of vertebral arteries along the craniovertebral junction (CJ) to cast light on the biomechanical aspects of aneurysm formation, focusing mainly on the relation of the vertebral artery injuries to the CJ bony landmarks. Herein, we report our experience with fourteen cases of craniovertebral junction vertebral artery (CJVA) aneurysms and their presentations, management, and outcomes. MATERIALS AND METHODS: We extracted from 83 vertebral artery aneurysms only those 14 cases whose aneurysms were located at the C0-1-2. We reviewed all medical records, including operative reports and radiologic images. We divided the CJVA into 5 segments and then carefully reviewed the cases, largely focusing on the CJVA segments involved in the aneurysm. Angiographic outcomes were determined by angiography, which was scheduled at 3-6 months, 1, 2.5, and 5 years postoperatively. RESULTS: A total of 14 patients with CJVA aneurysms were included in the present study. 35.7 % had cerebrovascular risk factors, while 23.5 % had other predisposing factors such as an AVM, an AVF, or a foramen magnum tumor. Predisposing factors in the form of neck trauma, both direct and indirect, were identified in 50 % of cases. The segmental distribution of aneurysms was as follows: three (21.4 %) at CJV 1, one (7.1 %) at CJV 2, four (28.6 %) at CJV 3, two (14.3 %) at CJV 4, and four (28.6 %) isolated to the CJV 5 segment. Of the 6 indirect traumatic aneurysms, 1 (16.7 %) was located at CJV 1, 4 (66.7 %) were located at CJV 3 and 1 (16.7 %) was located at CJV 5. The 1/1 direct traumatic aneurysm (100 %) from the penetrating injury was located at CJV 1. 100 % of cases with cerebrovascular risk factors, the affected vessels were on the dominant side. 42.9 % of cases presented symptoms of a vertebrobasilar stroke. All 14 aneurysms were managed only endovascularly. 85.8 % of patients we implemented flow diverters only. 57.1 % of follow-up cases were completely occluded angiographically, and 42.9 % of cases were near-completely or incompletely occluded at 1, 2.5, and 5-year follow-ups. CONCLUSIONS: The current article is the first report of a series of vertebral artery aneurysms located in CJ. Herein, the association of vertebral artery aneurysm, hemodynamics, and trauma is well established. We clarified all segments of the CJVA and showed that the segmental distribution of CJVA aneurysms significantly differs between traumatic and spontaneous cases. We showed that treatment with flow diverters should be the mainstay of CJVA aneurysm treatment.


Asunto(s)
Aneurisma Intracraneal , Accidente Cerebrovascular , Humanos , Aneurisma Intracraneal/diagnóstico por imagen , Aneurisma Intracraneal/cirugía , Aneurisma Intracraneal/complicaciones , Angiografía Cerebral/efectos adversos , Arteria Vertebral/diagnóstico por imagen , Accidente Cerebrovascular/etiología , Resultado del Tratamiento
2.
Oxid Med Cell Longev ; 2021: 9739977, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34804372

RESUMEN

Blood-brain barrier (BBB) disruption is a common and critical pathology following subarachnoid hemorrhage (SAH). We investigated the BBB disruption property of secreted protein acidic and rich in cysteine (SPARC) after SAH. A total of 197 rats underwent endovascular perforation to induce SAH or sham operation. Small interfering ribonucleic acid (siRNA) for SPARC or scrambled siRNA was administered intracerebroventricularly to rats 48 h before SAH. Anti-SPARC monoclonal antibody (mAb) 236 for functional blocking or normal mouse immunoglobulin G (IgG) was administered intracerebroventricularly 1 h after SAH. Selective integrin αVß3 inhibitor cyclo(-RGDfK) or phosphate-buffered saline was administered intranasally 1 h before SAH, along with recombinant SPARC treatment. Neurobehavior, SAH severity, brain edema, immunohistochemical staining, and Western blot were evaluated. The expression of SPARC and integrin αVß3 was upregulated after SAH in the endothelial cells. SPARC siRNA and anti-SPARC mAb 236 prevented neuroimpairments and brain edema through protection of BBB as measured by IgG extravasation 24 and 72 h after SAH. Recombinant SPARC aggravated neuroimpairments and cyclo(-RGDfK) suppressed the harmful neurological effects via inhibition of activated c-Jun N-terminal kinase, p38, and matrix metalloproteinase-9 followed by retention of endothelial junction proteins. SPARC may induce post-SAH BBB disruption via integrin αVß3 signaling pathway.


Asunto(s)
Barrera Hematoencefálica/patología , Regulación de la Expresión Génica , Integrina alfaVbeta3/metabolismo , Sistema de Señalización de MAP Quinasas , Metaloproteinasa 9 de la Matriz/metabolismo , Osteonectina/metabolismo , Hemorragia Subaracnoidea/fisiopatología , Animales , Barrera Hematoencefálica/metabolismo , Integrina alfaVbeta3/genética , Masculino , Metaloproteinasa 9 de la Matriz/genética , Osteonectina/genética , Ratas , Ratas Sprague-Dawley
3.
Neurosci Lett ; 733: 135142, 2020 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-32522601

RESUMEN

The first step to treat aneurysmal subarachnoid hemorrhage (SAH) is aneurysmal obliteration under general anesthesia but not treat the SAH itself and the secondary effects. However, the identification of anesthetics with properties that help to attenuate post-SAH brain injury can be useful for improving outcomes of SAH patients. We examined whether 2% isoflurane and 3% sevoflurane posttreatment are protective against early brain injury (EBI) after SAH. This study used 87 8-week-old male CD-1 mice. We induced SAH by endovascular perforation in mice. Animals were randomly divided into 4 groups: sham-operated (n = 16), SAH + vehicle-medical air (n = 26), SAH + 2% isoflurane (n = 22), and SAH + 3% sevoflurane (n = 23). Neurobehavioral function, brain water content and Western blotting were evaluated at 24 h. The expression of sphingosine kinase (SphK), cleaved caspase-3 and cyclooxygenase-2 (COX2) was determined by Western blotting. Cell death was examined by terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end-labeling staining. Both 2% isoflurane and 3% sevoflurane significantly improved neurobehavioral function, and brain edema at 24 h after SAH and attenuated cell death, associated with an increase in SphK1, a decrease in cleaved caspase-3 and COX2. The neuroprotective effects were similar between 2% isoflurane and 3% sevoflurane. These findings suggest that both 2% isoflurane and 3% sevoflurane significantly inhibited EBI by suppressing post-SAH apoptosis and brain inflammation possibly via the SphK1-related pathway.


Asunto(s)
Isoflurano/farmacología , Fármacos Neuroprotectores/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Sevoflurano/farmacología , Hemorragia Subaracnoidea/patología , Animales , Apoptosis/efectos de los fármacos , Edema Encefálico/etiología , Edema Encefálico/prevención & control , Modelos Animales de Enfermedad , Masculino , Ratones , Recuperación de la Función/efectos de los fármacos , Hemorragia Subaracnoidea/complicaciones
4.
Neurosci Lett ; 728: 134979, 2020 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-32302701

RESUMEN

BACKGROUND: Sphingosine kinase (SphK) 1 has been reported as an important signaling node in anti-apoptotic signaling. Heparin is a pleiotropic drug that antagonizes many pathophysiological mechanisms. In this study, we evaluated if heparin prevents early brain injury (EBI) after subarachnoid hemorrhage (SAH) by anti-apoptotic mechanisms including SphK1. METHODS: SAH was induced by endovascular perforation in mice, which were randomly assigned to sham-operated (n = 23), SAH + vehicle (n = 36), SAH + 10U heparin pretreatment (n = 13), SAH + 30U heparin pretreatment (n = 15), SAH + 10U heparin posttreatment (n = 31), and SAH + 30U heparin posttreatment (n = 23). At 24 hours post-SAH, neurological scores, brain water content and Evans blue extravasation were evaluated. Also, the expression of SphK, phosphorylated Akt, and cleaved caspase-3 was determined by Western blotting, and cell death was examined by terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end-labeling staining. RESULTS: Low-dose heparin posttreatment improved neurobehavioral function, brain edema, blood-brain barrier disruption and cell death in the cortex, associated with an increase in SphK1 and phosphorylated Akt, and a decrease in cleaved caspase-3. High-dose heparin had a tendency for increased SAH severity, which obscured the neuroprotective effects by heparin. CONCLUSIONS: Low-dose heparin posttreatment may decrease the development of post-SAH EBI through anti-apoptotic mechanisms including sphingosine-related pathway activation.


Asunto(s)
Edema Encefálico/tratamiento farmacológico , Lesiones Encefálicas/tratamiento farmacológico , Heparina/farmacología , Esfingosina/farmacología , Hemorragia Subaracnoidea/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Lesiones Encefálicas/fisiopatología , Masculino , Ratones , Fármacos Neuroprotectores/farmacología , Transducción de Señal/efectos de los fármacos
5.
Turk Neurosurg ; 30(3): 354-360, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-30901072

RESUMEN

AIM: To examine whether post-treatment sevoflurane is protective against early brain injury (EBI) after subarachnoid hemorrhage (SAH) and how this neuroprotective effect occurs at different concentrations and durations of administration in mice. Furthermore, we tested whether the neuroprotective effect of post-treatment sevoflurane is associated with inhibition of apoptosis. MATERIAL AND METHODS: SAH was induced in mice by endovascular perforation. Animals were randomly assigned to five groups in each study. Study 1, sham-operated; SAH+vehicle-air; and SAH+1.5% sevoflurane for 30, 60, and 90 min. Study 2, SAH+3% sevoflurane for 30, 60, and 90 min. Study 3, SAH+4.5% sevoflurane for 30, 60, and 90 min. Neurobehavioral function and brain edema (brain water content) were evaluated 24 h after SAH. Neuroglial cell death was examined by terminal deoxynucleotidyl transferase-mediated uridine 5′-triphosphate-biotin nick end-labeling (TUNEL) staining. RESULTS: Administration of 1.5% sevoflurane for 60 min and 3% sevoflurane for 30 and 60 min significantly improved neurobehavioral function, brain edema, and attenuated neuronal cell death in the basal cortex at 24 h after SAH. CONCLUSION: Administration of 1.5% sevoflurane for 60 min and 3% for 30 and 60 min sevoflurane application attenuated the development of EBI after SAH, implying its use for anesthesia during acute aneurysm surgery or intervention.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/uso terapéutico , Sevoflurano/uso terapéutico , Hemorragia Subaracnoidea/tratamiento farmacológico , Animales , Apoptosis/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/metabolismo , Edema Encefálico/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Inhibidores de Agregación Plaquetaria/farmacología , Distribución Aleatoria , Sevoflurano/farmacología , Hemorragia Subaracnoidea/metabolismo , Hemorragia Subaracnoidea/patología
6.
Acta Neurochir Suppl ; 121: 127-30, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463935

RESUMEN

Heparin is a pleiotropic drug that antagonizes many pathophysiological mechanisms. In this study, we evaluated whether heparin prevents early brain injury (EBI) after subarachnoid hemorrhage (SAH) in mice. SAH was induced by endovascular perforation in mice randomly assigned to sham-operated (n = 8), SAH + vehicle (n = 12), SAH + 10 U heparin pretreatment (n = 11), and SAH + 30 U heparin pretreatment (n = 14) groups. At 24 h post-SAH, severity of SAH, neurological scores, and brain water content were evaluated. Low-dose heparin pretreatment improved neurobehavioral function, and decreased brain edema in the ipsilateral cerebral hemisphere to the perforation side. High-dose heparin had a tendency for increased SAH, which obscured the neuroprotective effects by heparin. Low-dose heparin pretreatment may decrease the development of post-SAH EBI.


Asunto(s)
Apoptosis/efectos de los fármacos , Edema Encefálico/fisiopatología , Lesiones Encefálicas/fisiopatología , Encéfalo/efectos de los fármacos , Fibrinolíticos/farmacología , Heparina/farmacología , Hemorragia Subaracnoidea/fisiopatología , Animales , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Procedimientos Endovasculares , Fibrinolíticos/administración & dosificación , Heparina/administración & dosificación , Masculino , Ratones , Mortalidad , Fármacos Neuroprotectores/farmacología , Punciones , Distribución Aleatoria , Índice de Severidad de la Enfermedad
7.
Stroke ; 46(6): 1710-3, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25931468

RESUMEN

BACKGROUND AND PURPOSE: This study examines the role of thrombin's protease-activated receptor (PAR)-1, PAR-4 in mediating cyclooxygenase-2 and mammalian target of rapamycin after germinal matrix hemorrhage. METHODS: Germinal matrix hemorrhage was induced by intraparenchymal infusion of bacterial collagenase into the right ganglionic eminence of P7 rat pups. Animals were treated with PAR-1, PAR-4, cyclooxygenase-2, or mammalian target of rapamycin inhibitors by 1 hour, and ≤5 days. RESULTS: We found increased thrombin activity 6 to 24 hours after germinal matrix hemorrhage, and PAR-1, PAR-4, inhibition normalized cyclooxygenase-2, and mammalian target of rapamycin by 72 hours. Early treatment with NS398 or rapamycin substantially improved long-term outcomes in juvenile animals. CONCLUSIONS: Suppressing early PAR signal transduction, and postnatal NS398 or rapamycin treatment, may help reduce germinal matrix hemorrhage severity in susceptible preterm infants.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Lesiones Encefálicas/tratamiento farmacológico , Hemorragia Cerebral/tratamiento farmacológico , Inmunosupresores/farmacología , Nitrobencenos/farmacología , Receptor PAR-1/antagonistas & inhibidores , Receptores de Trombina/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Sulfonamidas/farmacología , Animales , Animales Recién Nacidos , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Ciclooxigenasa 2/metabolismo , Ratas
8.
Neurobiol Dis ; 62: 365-71, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24084689

RESUMEN

Brain inflammation may play an important role in the pathophysiology of early brain injury after subarachnoid hemorrhage (SAH). Our aim was to demonstrate brain inflammation development and to determine whether isoflurane, a clinically available volatile anesthetic agent, prevents brain inflammation after SAH. This study used 162 8-week-old male CD-1 mice. We induced SAH with endovascular perforation in mice and randomly assigned animals to sham-operated (n=21), SAH+vehicle-air (n=35) and SAH+2% isoflurane (n=31). In addition to the evaluation of brain injury (neurological scores, brain edema and Evans blue dye extravasation), brain inflammation was evaluated by means of expression changes in markers of inflammatory cells (ionized calcium binding adaptor molecule-1, myeloperoxidase), cytokines (tumor necrosis factor [TNF]-α, interleukin-1ß), adhesion molecules (intercellular adhesion molecule [ICAM]-1, P-selectin), inducers of inflammation (cyclooxygenase-2, phosphorylated c-Jun N-terminal kinase [p-JNK]) and endothelial cell activation (von Willebrand factor) at 24h post-SAH. Sphingosine kinase inhibitor (N, N-dimethylsphingosine [DMS]) and sphingosine-1-phosphate receptor-1/3 antagonist (VPC23019) were used to block isoflurane's effects (n=22, each). SAH caused early brain injury, which was associated with inflammation so that all evaluated markers of inflammation were increased. Isoflurane significantly inhibited both brain injury (P<0.001, respectively) and inflammation (myeloperoxidase, P=0.022; interleukin-1ß, P=0.002; TNF-α, P=0.015; P-selectin, P=0.010; ICAM-1, P=0.016; p-JNK, P<0.001; cyclooxygenase-2, P=0.003, respectively). This beneficial effect of isoflurane was abolished with DMS and VPC23019. Isoflurane may suppress post-SAH brain inflammation possibly via the sphingosine-related pathway.


Asunto(s)
Encefalitis/tratamiento farmacológico , Isoflurano/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Hemorragia Subaracnoidea/tratamiento farmacológico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encefalitis/etiología , Encefalitis/metabolismo , Encefalitis/patología , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos , Distribución Aleatoria , Hemorragia Subaracnoidea/complicaciones
9.
Transl Stroke Res ; 4(5): 524-32, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24187597

RESUMEN

Although there is evidence that sphingosine-1-phosphate receptor-1 (S1P1) activation occurs following experimental brain injury, there is little information about its metabolic pathway in cerebral ischemia. The purpose of this study was to evaluate the role of the sphingosine metabolic pathway including S1P1, sphingosine kinases 1 (SphK1), and 2 (SphK2) in transient middle cerebral artery occlusion (MCAO). Fifty-eight male Sprague-Dawley rats were used to asses temporal profiles of S1P1, SphK1 and 2 on neurons in infarct and periinfarct cortices at pre-infarct state, 6, and 24 hours after MCAO. The animals were then treated with vehicle and 0.25 mg/kg FTY720, which is an agonist of S1P receptors, and evaluated regarding neurological function, infarct volume, and S1P1 expression on neurons at 24 hours after MCAO. The expressions of S1P1, SphK1, and SphK2 were significantly decreased after MCAO. Labeling of all markers were reduced in the infarct cortex but remained present in the periinfarct cortex, and some were found to be on neurons. Significant improvements of neurological function and brain injury were observed in the FTY720 group compared with the vehicle and untreated groups, although S1P1 expression on neurons was reduced in the FTY720 group compared with the vehicle group. We demonstrated that S1P1, SphK1, and SphK2 were downregulated in the infarct cortex, whereas they were preserved in the periinfarct cortex where FTY720 reduced neuronal injury possibly via S1P1 activation. Our findings suggest that activation of the sphingosine metabolic pathway may be neuroprotective in cerebral ischemia.


Asunto(s)
Isquemia Encefálica/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Neuronas/metabolismo , Esfingosina/metabolismo , Animales , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod , Inmunosupresores/farmacología , Masculino , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Glicoles de Propileno/farmacología , Ratas , Ratas Sprague-Dawley , Esfingosina/análogos & derivados , Esfingosina/farmacología
10.
Stroke ; 44(11): 3189-94, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24008574

RESUMEN

BACKGROUND AND PURPOSE: Neuronal apoptosis is a key pathological process in subarachnoid hemorrhage (SAH)-induced early brain injury. Given that recombinant osteopontin (rOPN), a promising neuroprotectant, cannot pass through the blood-brain barrier, we aimed to examine whether nasal administration of rOPN prevents neuronal apoptosis after experimental SAH. METHODS: Male Sprague-Dawley rats (n=144) were subjected to the endovascular perforation SAH model. rOPN was administered via the nasal route and neurological scores as well as brain water content were evaluated at 24 and 72 hours after SAH induction. The expressions of cleaved caspase-3, phosphorylated focal adhesion kinase (FAK), and phosphorylated Akt were examined using Western blot analysis. Neuronal cell death was demonstrated with terminal deoxynucleotid transferase-deoxyuridine triphosphate (dUTP) nick end labeling. We also administered FAK inhibitor 14 and phosphatidylinositol 3-kinase inhibitor, Wortmannin, prior to rOPN to establish its neuroprotective mechanism. ELISA was used to measure rOPN delivery into the cerebrospinal fluid. RESULTS: Cerebrospinal fluid level of rOPN increased after its nasal administration. This was associated with improved neurological scores and reduced brain edema at 24 hours after SAH. rOPN increased phosphorylated FAK and phosphorylated Akt expressions and decreased caspase-3 cleavage, resulting in attenuation of neuronal cell death within the cerebral cortex. These effects were abolished by FAK inhibitor 14 and Wortmannin. CONCLUSIONS: Nasal administration of rOPN decreased neuronal cell death and brain edema and improved the neurological status in SAH rats, possibly through FAK-phosphatidylinositol 3-kinase-Akt-induced inhibition of capase-3 cleavage.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Osteopontina/uso terapéutico , Proteínas Recombinantes/uso terapéutico , Hemorragia Subaracnoidea/tratamiento farmacológico , Administración Intranasal , Animales , Apoptosis , Encéfalo/patología , Lesiones Encefálicas/complicaciones , Caspasa 3/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Masculino , Neuronas/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/complicaciones , Factores de Tiempo
11.
Exp Neurol ; 241: 45-55, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23261767

RESUMEN

T-lymphocytes promote cerebral inflammation, thus aggravating neuronal injury after stroke. Fingolimod, a sphingosine 1-phosphate receptor analog, prevents the egress of lymphocytes from primary and secondary lymphoid organs. Based on these findings, we hypothesized fingolimod treatment would reduce the number of T-lymphocytes migrating into the brain, thereby ameliorating cerebral inflammation following experimental intracerebral hemorrhage (ICH). We investigated the effects of fingolimod in two well-established murine models of ICH, implementing intrastriatal infusions of either bacterial collagenase (cICH) or autologous blood (bICH). Furthermore, we tested the long term neurological improvements by Fingolimod in a collagenase-induced rat model of ICH. Fingolimod, in contrast to vehicle administration alone, improved neurological functions and reduced brain edema at 24 and 72 h following experimental ICH in CD-1 mice (n=103; p<0.05). Significantly fewer lymphocytes were found in blood and brain samples of treated animals when compared to the vehicle group (p<0.05). Moreover, fingolimod treatment significantly reduced the expression of intercellular adhesion molecule-1 (ICAM-1), interferon-γ (INF-γ), and interleukin-17 (IL-17) in the mouse brain at 72 h post-cICH (p<0.05 compared to vehicle). Long-term neurocognitive performance and histopathological analysis were evaluated in Sprague-Dawley rats between 8 and 10 weeks post-cICH (n=28). Treated rats showed reduced spatial and motor learning deficits, along with significantly reduced brain atrophy and neuronal cell loss within the basal ganglia (p<0.05 compared to vehicle). We conclude that fingolimod treatment ameliorated cerebral inflammation, at least to some extent, by reducing the availability and subsequent brain infiltration of T-lymphocytes, which improved the short and long-term sequelae after experimental ICH in rodents.


Asunto(s)
Corteza Cerebral/patología , Hemorragia Cerebral/tratamiento farmacológico , Hemorragia Cerebral/patología , Inmunosupresores/uso terapéutico , Linfocitos/efectos de los fármacos , Glicoles de Propileno/uso terapéutico , Esfingosina/análogos & derivados , Análisis de Varianza , Animales , Ganglios Basales/patología , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Transfusión de Sangre Autóloga/efectos adversos , Edema Encefálico/etiología , Edema Encefálico/prevención & control , Complejo CD3/metabolismo , Recuento de Células , Hemorragia Cerebral/inducido químicamente , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/etiología , Hemorragia Cerebral/inmunología , Colagenasas/toxicidad , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod , Miembro Anterior/fisiopatología , Lateralidad Funcional/efectos de los fármacos , Molécula 1 de Adhesión Intercelular/metabolismo , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Leucocitos/efectos de los fármacos , Linfocitos/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/prevención & control , Glicoles de Propileno/farmacología , Trastornos Psicomotores/tratamiento farmacológico , Trastornos Psicomotores/etiología , Ratas , Ratas Sprague-Dawley , Percepción Espacial/efectos de los fármacos , Esfingosina/farmacología , Esfingosina/uso terapéutico , Factores de Tiempo
12.
J Neurochem ; 123 Suppl 2: 12-21, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23050638

RESUMEN

Subarachnoid hemorrhage is a devastating disease that can be difficult to manage. Not only is the initial bleeding and rebleeding associated with high mortality, but a large fraction of patients also develop a delayed neurological deficit even when the aneurysm was successfully secured with clipping or coiling. Past research effort has traditionally been focused on vasospasm, which was conceived to be the sole factor for delayed neurological deficit. The failure of anti-vasospastic drugs to improve outcome in clinical trials has brought into focus the significance of early brain injury. The immediate events associated with subarachnoid hemorrhage, including increased intracranial pressure, decreased cerebral blood flow and global ischemia initiate a cascade of pathological changes that occur before the onset of delayed vasospasm. These pathological changes in the very early stage of the hemorrhage propagate and cause blood-brain barrier disruption, inflammation, oxidative stress and cell death. Focusing only on the treatment of vasospasm with complete disregard for early brain injury is insufficient for the management of subarachnoid hemorrhage. Instead, a therapeutic intervention has to aim at stopping the molecular cascades of early brain injury that may lead to long-term deficits in addition to vasospasm. We review the pathological mechanisms of early brain injury, which may reveal new therapeutic avenues that can be exploited to serve as combination therapy with anti-vasospasm medications in the future.


Asunto(s)
Lesiones Encefálicas/etiología , Ensayos Clínicos como Asunto/métodos , Hemorragia Subaracnoidea/complicaciones , Vasoespasmo Intracraneal/etiología , Barrera Hematoencefálica/fisiopatología , Lesiones Encefálicas/terapia , Muerte Celular , Circulación Cerebrovascular/fisiología , Humanos , Inflamación/etiología , Presión Intracraneal , Estrés Oxidativo , Vasoespasmo Intracraneal/terapia
13.
Stroke ; 43(9): 2513-6, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22773559

RESUMEN

BACKGROUND AND PURPOSE: We examined effects of isoflurane, volatile anesthetics, on blood-brain barrier disruption in the endovascular perforation model of subarachnoid hemorrhage (SAH) in mice. METHODS: Animals were assigned to sham-operated, SAH+vehicle-air, SAH+1%, or 2% isoflurane groups. Neurobehavioral function, brain water content, Evans blue dye extravasation, and Western blotting for sphingosine kinases, occludin, claudin-5, junctional adhesion molecule, and vascular endothelial cadherin were evaluated at 24 hours post-SAH. Effects of sphingosine kinase (N,N-dimethylsphingosine) or sphingosine-1-phosphate receptor-1/3 (S1P1/3) inhibitors (VPC23019) on isoflurane's action were also examined. RESULTS: SAH aggravated neurological scores, brain edema, and blood-brain barrier permeability, which were prevented by 2% but not 1% isoflurane posttreatment. Two percent isoflurane increased sphingosine kinase-1 expression and prevented a post-SAH decrease in expressions of the blood-brain barrier-related proteins. Both N,N-dimethylsphingosine and VPC23019 abolished the beneficial effects of isoflurane. CONCLUSIONS: Two percent isoflurane can suppress post-SAH blood-brain barrier disruption, which may be mediated by sphingosine kinase 1 expression and sphingosine-1-phosphate receptor-1/3 activation.


Asunto(s)
Anestésicos por Inhalación/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Isoflurano/uso terapéutico , Hemorragia Subaracnoidea/tratamiento farmacológico , Hemorragia Subaracnoidea/fisiopatología , Animales , Biomarcadores/análisis , Western Blotting , Edema Encefálico/patología , Colorantes , Azul de Evans , Lateralidad Funcional/fisiología , Masculino , Ratones , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/antagonistas & inhibidores
14.
Int J Stroke ; 7(5): 398-406, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22712741

RESUMEN

Animal models of stroke contribute to the development of better stroke prevention and treatment through studies investigating the pathophysiology of different stroke subtypes and by testing promising treatments before trials in humans. There are two broad types of animal models: those in which stroke is induced through artificial means, modeling the consequences of a vascular insult but not the vascular pathology itself; and those in which strokes occur spontaneously. Most animal models of stroke are in rodents due to cost, ethical considerations, availability of standardized neurobehavioral assessments, and ease of physiological monitoring. While there are similarities in cerebrovascular anatomy and pathophysiology between rodents and humans, there are also important differences, including brain size, length and structure of perforating arteries, and gray to white matter ratio, which is substantially lower in humans. The wide range of rodent models of stroke includes models of global and focal ischemia, and of intracerebral and sub-arachnoid hemorrhage. The most widely studied model of spontaneous stroke is the spontaneously hypertensive stroke-prone rat, in which the predominant lesions are small subcortical infarcts resulting from a vascular pathology similar to human cerebral small vessel disease. Important limitations of animal models of stroke - they generally model only certain aspects of the disease and do not reflect the heterogeneity in severity, pathology and comorbidities of human stroke - and key methodological issues (especially the need for adequate sample size, randomization, and blinding in treatment trials) must be carefully considered for the successful translation of pathophysiological concepts and therapeutics from bench to bedside.


Asunto(s)
Modelos Animales de Enfermedad , Accidente Cerebrovascular/etiología , Animales , Animales Endogámicos , Isquemia Encefálica/etiología , Conducta de Elección , Humanos , Infarto de la Arteria Cerebral Media/etiología , Ratones , Ratas , Proyectos de Investigación , Accidente Cerebrovascular/prevención & control , Hemorragia Subaracnoidea/etiología
15.
Exp Neurol ; 236(1): 69-78, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22524990

RESUMEN

Germinal matrix hemorrhage (GMH) is the most common neurological disease of premature newborns. GMH causes neurological sequelae such as cerebral palsy, post-hemorrhagic hydrocephalus, and mental retardation. Despite this, there is no standardized animal model of spontaneous GMH using newborn rats to depict the condition. We asked whether stereotactic injection of collagenase type VII (0.3 U) into the ganglionic eminence of neonatal rats would reproduce the acute brain injury, gliosis, hydrocephalus, periventricular leukomalacia, and attendant neurological consequences found in humans. To test this hypothesis, we used our neonatal rat model of collagenase-induced GMH in P7 pups, and found that the levels of free-radical adducts (nitrotyrosine and 4-hyroxynonenal), proliferation (mammalian target of rapamycin), inflammation (COX-2), blood components (hemoglobin and thrombin), and gliosis (vitronectin and GFAP) were higher in the forebrain of GMH pups, than in controls. Neurobehavioral testing showed that pups with GMH had developmental delay, and the juvenile animals had significant cognitive and motor disability, suggesting clinical relevance of the model. There was also evidence of white-matter reduction, ventricular dilation, and brain atrophy in the GMH animals. This study highlights an instructive animal model of the neurological consequences after germinal matrix hemorrhage, with evidence of brain injuries that can be used to evaluate strategies in the prevention and treatment of post-hemorrhagic complications.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Hemorragia Cerebral/fisiopatología , Hidrocefalia/fisiopatología , Recien Nacido Prematuro/fisiología , Animales , Animales Recién Nacidos , Lesiones Encefálicas/diagnóstico , Lesiones Encefálicas/patología , Hemorragia Cerebral/diagnóstico , Hemorragia Cerebral/patología , Colágeno Tipo VII/toxicidad , Diagnóstico Diferencial , Modelos Animales de Enfermedad , Femenino , Humanos , Hidrocefalia/diagnóstico , Hidrocefalia/patología , Recién Nacido , Embarazo , Ratas , Ratas Sprague-Dawley , Telencéfalo/efectos de los fármacos , Telencéfalo/embriología , Telencéfalo/patología
16.
Crit Care Med ; 40(6): 1908-13, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22488000

RESUMEN

OBJECTIVE: Isoflurane, a volatile anesthetic agent, has been recognized for its potential neuroprotective properties and has antiapoptotic effects. We examined whether isoflurane posttreatment is protective against early brain injury after subarachnoid hemorrhage and determined whether this effect needs sphingosine-related pathway activation. DESIGN: Controlled in vivo laboratory study. SETTING: Animal research laboratory. SUBJECTS: One hundred seventy-nine 8-wk-old male CD-1 mice weighing 30-38 g. INTERVENTIONS: Subarachnoid hemorrhage was induced in mice by endovascular perforation. Animals were randomly assigned to sham-operated, subarachnoid hemorrhage-vehicle, and subarachnoid hemorrhage+2% isoflurane. Neurobehavioral function and brain edema were evaluated at 24 and 72 hrs. The expression of sphingosine kinase, phosphorylated Akt, and cleaved caspase-3 was determined by Western blotting and immunofluorescence. Neuronal cell death was examined by terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end-labeling staining. Effects of a sphingosine kinase inhibitor N, N-dimethylsphingosine or a sphingosine 1 phosphate receptor inhibitor VPC23019 on isoflurane's protective action against postsubarachnoid hemorrhage early brain injury were also examined. MEASUREMENTS AND MAIN RESULTS: Isoflurane significantly improved neurobehavioral function and brain edema at 24 hrs but not 72 hrs after subarachnoid hemorrhage. At 24 hrs, isoflurane attenuated neuronal cell death in the cortex, associated with an increase in sphingosine kinase 1 and phosphorylated Akt, and a decrease in cleaved caspase-3. The beneficial effects of isoflurane were abolished by N, N-dimethylsphingosine and VPC23019. CONCLUSIONS: Isoflurane posttreatment delays the development of postsubarachnoid hemorrhage early brain injury through antiapoptotic mechanisms including sphingosine-related pathway activation, implying its use for anesthesia during acute aneurysm surgery or intervention.


Asunto(s)
Apoptosis/efectos de los fármacos , Lesiones Encefálicas/prevención & control , Isoflurano/farmacología , Fármacos Neuroprotectores/farmacología , Hemorragia Subaracnoidea/tratamiento farmacológico , Animales , Edema Encefálico/tratamiento farmacológico , Lesiones Encefálicas/etiología , Lisofosfolípidos/metabolismo , Masculino , Ratones , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Distribución Aleatoria , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Hemorragia Subaracnoidea/complicaciones
17.
Neurochem Int ; 60(3): 327-33, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22226843

RESUMEN

The blood-brain barrier (BBB) disruption and brain edema are important pathophysiologies of early brain injury after subarachnoid hemorrhage (SAH). This study is to evaluate whether Rho kinase (Rock) enhances BBB permeability via disruption of tight junction proteins during early brain injury. Adult male rats were assigned to five groups; Sham-operated, SAH treated with saline, a Rock inhibitor hydroxyfasudil (HF) (10 mg/kg) treatment at 0.5 h after SAH, HF treatment at 0.5 and 6 h (10 mg/kg, each) after SAH, and another Rock inhibitor Y27632 (10 mg/kg) treatment at 0.5 h after SAH. The perforation model of SAH was performed and neurological score and brain water content were evaluated 24 and 72 h after surgery. Evans blue extravasation, Rock activity assay, and western blotting analyses were evaluated 24 h after surgery. Treatment of HF significantly improved neurological scores 24 h after SAH. Single treatment with HF and Y27632, and two treatments with HF reduced brain water content in the ipsilateral hemisphere. HF reduced Evans blue extravasation in the ipsilateral hemisphere after SAH. Rock activity increased 24 h after SAH, and HF reversed the activity. SAH significantly decreased the levels of tight junction proteins, occludin and zonula occludens-1 (ZO-1), and HF preserved the levels of occluding and ZO-1 in ipsilateral hemisphere. In conclusion, HF attenuated BBB permeability after SAH, possibly by protection of tight junction proteins.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Hemorragia Subaracnoidea/complicaciones , Quinasas Asociadas a rho/antagonistas & inhibidores , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Amidas/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Western Blotting , Agua Corporal/metabolismo , Química Encefálica/fisiología , Edema Encefálico/mortalidad , Inhibidores Enzimáticos/farmacología , Lateralidad Funcional/fisiología , Masculino , Proteínas de la Membrana/metabolismo , Procedimientos Neuroquirúrgicos , Ocludina , Fosfoproteínas/metabolismo , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/mortalidad , Uniones Estrechas/efectos de los fármacos , Resultado del Tratamiento , Proteína de la Zonula Occludens-1 , Quinasas Asociadas a rho/metabolismo
18.
J Neurosci Res ; 90(3): 691-7, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22183833

RESUMEN

Attenuation of blood-brain barrier (BBB) disruption is one of the therapeutic candidates for treatment of subarachnoid hemorrhage (SAH). In this study, the protective effect of sodium orthovanadate (SOV) on BBB disruption was investigated in SAH using the endovascular perforation model. Fifty-five rats were randomly assigned to sham-operated, SAH treated with saline (as a vehicle), or 10 mg/kg SOV groups and were evaluated for neurofunction and Evans blue dye extravasation. The phosphorylation of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and mitogen-activated protein kinase (MAPK) and the expression of matrix metalloproteinase-9 (MMP-9), occludin, and collagen-IV were examined by Western blot analyses. Cell death among endothelial cells was revealed by immunofluorescence and terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end-labeling staining. SOV significantly improved neurofunction and reduced Evans blue dye extravasation in brains after SAH. SOV phosphorylated PTEN, decreased phospho-JNK and MMP-9, and preserved occludin expression. SOV also attenuated SAH-induced capillary endothelial cell death. The current study showed that SOV was protective against BBB disruption after SAH, possibly via PTEN phosphorylation.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Fosfohidrolasa PTEN/metabolismo , Hemorragia Subaracnoidea/tratamiento farmacológico , Vanadatos/farmacología , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiopatología , Muerte Celular/efectos de los fármacos , Colágeno Tipo IV/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Ocludina , Permeabilidad/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/metabolismo , Hemorragia Subaracnoidea/fisiopatología , Vanadatos/uso terapéutico
19.
Stroke ; 43(3): 844-50, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22207510

RESUMEN

BACKGROUND AND PURPOSE: Perihematomal edema formation and consequent cell death contribute to the delayed brain injury evoked by intracerebral hemorrhage (ICH). We aimed to evaluate the effect of α7 nicotinic acetylcholine receptor (α7nAChR) stimulation on behavior, brain edema, and neuronal apoptosis. Furthermore, we aimed to determine the role of the proapoptotic glycogen synthase kinase-3ß (GSK-3ß) after experimental ICH. METHODS: Male CD-1 mice (n=109) were subjected to intracerebral infusion of autologous blood (n=88) or sham surgery (n=21). ICH animals received vehicle administration, 4 or 12 mg/kg of α7nAChR agonist PHA-543613, 12 mg/kg of α7nAChR agonist PNU-282987, 6 mg/kg of α7nAChR antagonist methyllycaconitine (MLA), 15 µg/kg of phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin, or PHA-543613 combined with MLA or wortmannin. Behavioral deficits and brain water content were evaluated at 24 and 72 hours after surgery. Western blotting and immunofluorescence staining were used for the quantification and localization of activated Akt (p-Akt), GSK-3ß (p-GSK-3ß), and cleaved caspase-3 (CC3). Neuronal cell death was quantified through terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL). RESULTS: α7nAChR stimulation improved neurological outcome and reduced brain edema at 24 and 72 hours after surgery (P<0.05 compared with vehicle). Furthermore, PHA-543613 treatment increased p-Akt and decreased p-GSK-3ß and CC3 expressions in the ipsilateral hemisphere (P<0.05, respectively), which was reversed by MLA and wortmannin. P-Akt, p-GSK-3ß, and CC3 were generally localized in neurons. PHA-543613 reduced neuronal cell death in the perihematomal area (P<0.05). CONCLUSIONS: α7nAChR stimulation improved functional and morphological outcomes after experimental ICH in mice. PHA-543613 reduced the expression of proapoptotic GSK-3ß through the PI3K-Akt signaling pathway.


Asunto(s)
Benzamidas/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Compuestos Bicíclicos con Puentes/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Hemorragias Intracraneales/tratamiento farmacológico , Fármacos Neuroprotectores , Agonistas Nicotínicos/farmacología , Quinuclidinas/farmacología , Receptores Nicotínicos/fisiología , Animales , Apoptosis/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Western Blotting , Agua Corporal/metabolismo , Química Encefálica/efectos de los fármacos , Edema Encefálico/etiología , Edema Encefálico/patología , Caspasa 3/fisiología , Inhibidores de Caspasas , Técnica del Anticuerpo Fluorescente , Glucógeno Sintasa Quinasa 3 beta , Hemorragias Intracraneales/psicología , Masculino , Ratones , Neuronas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/fisiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Receptores Nicotínicos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/psicología , Resultado del Tratamiento , Receptor Nicotínico de Acetilcolina alfa 7
20.
Stroke ; 42(2): 477-83, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21193742

RESUMEN

BACKGROUND AND PURPOSE: Recent studies reported that apoptosis was involved in the pathogenesis of early brain injury after subarachnoid hemorrhage (SAH). The aim of this study was to examine whether sodium orthovanadate (SOV) prevents post-SAH apoptosis by modulating growth factors and its downstream receptor tyrosine kinases. Method- Rats were operated on with the endovascular perforation model. SAH animals were treated with vehicle, 3 mg/kg and 10 mg/kg SOV, and evaluated regarding neurofunction and brain edema. The expression of growth factors such as mature brain-derived neurotrophic factor, insulin-like growth factor-1, and vascular endothelial growth factor and phosphorylation of tropomyosin-related kinase B, which is a receptor tyrosine kinase for brain-derived neurotrophic factor and the downstream pathway in antiapoptosis, was examined by Western blot analysis. Neuronal cell death was measured with terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end-labeling staining. We also administered K252a, a tropomyosin-related kinase B antagonist, to examine the mechanisms for neuroprotective effects by SOV. RESULTS: SOV significantly improved neurofunction and reduced brain edema after SAH. SOV increased mature brain-derived neurotrophic factor and prevented post-SAH tropomyosin-related kinase B inactivation and caspase-3 activation, resulting in attenuation of neuronal cell death in the cortex and hippocampal CA1 region. Preinjection of K252a abolished the beneficial effects of SOV. CONCLUSIONS: The current study showed that brain-derived neurotrophic factor-induced tropomyosin-related kinase B activation by SOV was necessary for protection against early brain injury after SAH.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/fisiología , Receptor trkB/fisiología , Transducción de Señal/fisiología , Hemorragia Subaracnoidea/tratamiento farmacológico , Hemorragia Subaracnoidea/enzimología , Tropomiosina/fisiología , Vanadatos/uso terapéutico , Animales , Lesiones Encefálicas/enzimología , Lesiones Encefálicas/etiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Masculino , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Hemorragia Subaracnoidea/complicaciones , Factores de Tiempo , Vanadatos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...