Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Acta Neurochir Suppl ; 115: 259-66, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22890678

RESUMEN

INTRODUCTION: Statins influence immune system activities through mechanisms independent of their lipid-lowering properties. T cells can be subdivided based on cytokine secretion patterns into two subsets: T-helper cells type 1 (Th1) and type 2 (Th2). Independent laboratory studies have shown statins to be potent inducers of a Th2 switch in immune cell response and be neuroprotective in several models of central nervous system (CNS) disease. This study was the first to evaluate the immune modulating effects of statins in subarachnoid hemorrhage (SAH). METHODS: Simvastatin was administered to rats intraperitoneally in two dosages (1 and 20 mg/kg) 30 min after the induction of SAH using endovascular perforation. Neurological scores were assessed 24 h later. Animals were then sacrificed, and samples of cortex and brain stem were tested for expression of the T-regulatory cell cytokine transforming growth factor (TGF) ß1, as well as interleukin (IL) 1ß, a proinflammatory cytokine associated with Th1 immune responses. The presence of TGF-ß1 secreting T cells was evaluated with the use of brain slices. RESULTS: SAH significantly impaired neurological function in all SAH groups (treated and untreated) versus sham. Animals treated with high-dose simvastatin had less neurological impairment than both untreated and low-dose groups. Cortical and brain-stem levels of TGF-ß1 were significantly elevated following SAH in the high-dose group. IL-1ß was significantly elevated following the induction of SAH but was inhibited by high-dose simvastatin. Double-labeled fluorescent immunohistochemical data demonstrated the presence of lymphocytes in the subarachnoid and perivascular spaces following SAH. Expression of TGF-ß1 by lymphocytes was markedly increased following treatment with high-dose simvastatin. CONCLUSION: The present study elucidated the potential role of a Th2 immune switch in statin provided neuroprotection following SAH.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Hemorragia Subaracnoidea , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Análisis de Varianza , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Masculino , Examen Neurológico , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/tratamiento farmacológico , Hemorragia Subaracnoidea/patología , Hemorragia Subaracnoidea/fisiopatología , Linfocitos T Colaboradores-Inductores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
2.
Acta Neurochir Suppl ; 110(Pt 2): 9-11, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21125437

RESUMEN

3-Hydroxy-3-methylglutaryl coenzyme A (HMG CoA) reductase inhibitors, commonly known as statins, are widely used clinically for their lipid lowering properties. Recent experimental evidence shows that statins are also effective in ameliorating cerebral vasospasm, which occurs as sequelae of subarachnoid hemorrhage. This literature review focuses on the literature-based putative mechanisms involved in statin mediated attenuation of cerebral vasospasm, such as eNOS, vascular inflammation, apoptosis, especially the phosphatidylinositol 3-kinase/Akt (PI3K/Akt) pathway from our experimental study.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Simvastatina/uso terapéutico , Vasoespasmo Intracraneal/tratamiento farmacológico , Animales , Arterias Carótidas/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteína Oncogénica v-akt/metabolismo , Ratas , Literatura de Revisión como Asunto , Factores de Tiempo , Vasoespasmo Intracraneal/enzimología , Vasoespasmo Intracraneal/patología
3.
Acta Neurochir Suppl ; 111: 231-6, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21725761

RESUMEN

OBJECTS: We determined effects of recombinant OPN (r-OPN), a pleiotropic extracellular matrix protein, on blood-brain barrier (BBB) disruption and matrix metalloproteinase (MMP)-9 activation after subarachnoid hemorrhage (SAH) in rats. METHODS: The endovascular perforation model of SAH was used. SAH or sham-operated rats were treated with pre-SAH intracerebroventricular administration of two dosages of r-OPN, r-OPN+GRGDSP (an L-arginyl-glycyl-L-aspartate-dependent integrin receptor antagonist), albumin or vehicle. Neurological impairments, brain edema and BBB disruption were evaluated, and Western blot analyses were performed in the brain at 24 h after SAH. RESULTS: r-OPN significantly prevented brain edema and BBB disruption compared with the control rats, associated with the suppression of nuclear factor-κB and mitogen-activated protein kinase pathways, leading to MMP-9 inactivation. These effects were blocked by GRGDSP. CONCLUSIONS: L-arginyl-glycyl-L-aspartate-dependent integrin receptor-mediated multiple signaling pathways may be involved in the protective effects of r-OPN against BBB disruption after SAH.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiopatología , Osteopontina/uso terapéutico , Hemorragia Subaracnoidea/patología , Animales , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Distribución de Chi-Cuadrado , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Quinasa I-kappa B/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/etiología , Examen Neurológico , Oligopéptidos/uso terapéutico , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/uso terapéutico , Índice de Severidad de la Enfermedad , Hemorragia Subaracnoidea/tratamiento farmacológico
4.
Acta Neurochir Suppl ; 110(Pt 1): 75-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21116919

RESUMEN

BACKGROUND: Subarachnoid hemorrhage (SAH)-induced early brain injury (EBI) contributes to delayed ischemic neurological deficits, one of whose key pathologic manifestation is the blood-brain barrier (BBB) disruption. Although post-SAH BBB breakdown is a self-repairable phenomenon, the molecular pathways are unknown. We determined the role of osteopontin (OPN), a pleiotropic extracellular matrix glycoprotein, in the post-SAH BBB disruption in rats. METHOD: First, we produced the endovascular perforation model of SAH and studied if OPN is induced in the brain after SAH. Secondly, we examined the effects of blockage of endogenous OPN induction on neurological impairments and BBB disruption. Thirdly, we evaluated the effects of exogenous OPN on neurological impairments, brain edema and BBB disruption, and the related protein expression levels. FINDINGS: OPN was significantly induced and peaked at 72 h after SAH, in the recovery phase of EBI. OPN small interfering RNA significantly aggravated neurological impairment and BBB disruption 72 h after SAH. Exogenous OPN significantly prevented neurological impairment, brain edema and BBB disruption associated with the deactivation of nuclear factor-κB activity, the inhibition of matrix metalloproteinase (MMP)-9 induction and tissue inhibitor of MMP-1 reduction, and the consequent preservation of cerebral microvessel basal lamina protein laminin and tight junction protein zona occludens-1. CONCLUSIONS: These findings suggest the protective effects of OPN against BBB disruption after SAH, a finding which should provide a novel therapeutic approach for post-SAH EBI.


Asunto(s)
Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Osteopontina/metabolismo , Hemorragia Subaracnoidea/complicaciones , Análisis de Varianza , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiopatología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Lateralidad Funcional , Regulación de la Expresión Génica/efectos de los fármacos , Inyecciones Intraventriculares/métodos , Masculino , Modelos Biológicos , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Índice de Severidad de la Enfermedad , Factores de Tiempo
5.
Crit Care Med ; 38(2): 612-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19851092

RESUMEN

OBJECTIVE: Accumulated evidence suggests that the primary cause of poor outcome after subarachnoid hemorrhage is not only cerebral arterial narrowing but also early brain injury. Our objective was to determine the effect of recombinant osteopontin, a pleiotropic extracellular matrix glycoprotein, on early brain injury after subarachnoid hemorrhage in rats. DESIGN: Controlled in vivo laboratory study. SETTING: Animal research laboratory. SUBJECTS: One hundred seventy-seven male adult Sprague-Dawley rats weighing 300 to 370 g. INTERVENTIONS: The endovascular perforation model of subarachnoid hemorrhage was produced. Subarachnoid hemorrhage or sham-operated rats were treated with an equal volume (1 microL) of pre-subarachnoid hemorrhage intracerebroventricular administration of two dosages (0.02 and 0.1 microg) of recombinant osteopontin, albumin, or vehicle. Body weight, neurologic scores, brain edema, and blood-brain barrier disruption were evaluated, and Western blot analyses were performed to determine the effect of recombinant osteopontin on matrix metalloproteinase-9, substrates of matrix metalloproteinase-9 (zona occludens-1, laminin), tissue inhibitor of matrix metalloproteinase-1, inflammation (interleukin-1beta), and nuclear factor-kappaB signaling pathways. MEASUREMENTS AND MAIN RESULTS: Treatment with recombinant osteopontin prevented a significant loss in body weight, neurologic impairment, brain edema, and blood-brain barrier disruption after subarachnoid hemorrhage. These effects were associated with the deactivation of nuclear factor-kappaB activity, inhibition of matrix metalloproteinase-9 induction, the maintenance of tissue inhibitor of matrix metalloproteinase-1, the consequent preservation of the cerebral microvessel basal lamina protein laminin, and the tight junction protein zona occludens-1. CONCLUSIONS: These results demonstrate that recombinant osteopontin treatment is effective for early brain injury after subarachnoid hemorrhage.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Osteopontina/uso terapéutico , Hemorragia Subaracnoidea/tratamiento farmacológico , Animales , Barrera Hematoencefálica/metabolismo , Western Blotting , Lesiones Encefálicas/metabolismo , Modelos Animales de Enfermedad , Inyecciones Intraventriculares , Interleucina-1/biosíntesis , Masculino , Metaloproteinasa 1 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , FN-kappa B/biosíntesis , Fármacos Neuroprotectores/uso terapéutico , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/uso terapéutico , Hemorragia Subaracnoidea/metabolismo
6.
Turk Neurosurg ; 20(2): 159-66, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20401843

RESUMEN

Aneurysmal subarachnoid hemorrhage (SAH) is a devastating neurological disease that has a mortality rate as high as 67% in some series. Traditional research and treatment has focused on addressing the delayed events of cerebral vasospasm following SAH. However, the physiological and cellular events of early brain injury (EBI) make significant contributions to patient outcomes and may even be a more significant factor than delayed cerebral vasospasm. EBI is the result of physiological derangements such as increased intracranial pressure (ICP), decreased cerebral blood flow (CBF), and global cerebral ischemia, which results in blood brain barrier dysfunction, inflammation, and oxidative cascades that lead to neuronal cell death. The consequence of these events to the patient is often death or significant neurological disability. The link between EBI and outcome has come under intense focus with recent studies failing to show improved outcomes following significant inhibition of cerebral vasospasm, and research into the inhibition of EBI cascades is being perused as an effective means of treating SAH patients.


Asunto(s)
Lesiones Encefálicas/mortalidad , Lesiones Encefálicas/patología , Hemorragia Subaracnoidea/mortalidad , Hemorragia Subaracnoidea/patología , Apoptosis , Edema Encefálico/mortalidad , Edema Encefálico/patología , Infarto Cerebral/mortalidad , Infarto Cerebral/patología , Circulación Cerebrovascular , Encefalitis/mortalidad , Encefalitis/patología , Humanos , Hipertensión Intracraneal/mortalidad , Hipertensión Intracraneal/patología , Estrés Oxidativo , Vasoespasmo Intracraneal/mortalidad , Vasoespasmo Intracraneal/patología
7.
Stroke ; 40(4): 1530-2, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19228846

RESUMEN

BACKGROUND AND PURPOSE: We investigated the role of thrombin in early brain injury after subarachnoid hemorrhage (SAH). METHODS: The standard intravascular perforation model was used to produce experimental SAH in Sprague Dawley rats. Low-dose (0.3 mg/h) and high-dose (0.9 mg/h) argatroban, a direct thrombin inhibitor, were evaluated for effects on brain edema, blood-brain barrier (BBB) disruption, apoptotic cell death, inflammatory marker, and neurological outcomes after SAH. RESULTS: Both doses of argatroban attenuated BBB disruption; however, only high-dose was effective in lowering edema in all brain regions, reducing cell death, and inflammatory marker expression, and improving neurological outcomes. CONCLUSIONS: Thrombin inhibition by argatroban improves neurological outcomes and provides neuroprotection against acute events after SAH such as BBB disruption, brain edema, and cell death.


Asunto(s)
Anticoagulantes/farmacología , Edema Encefálico/tratamiento farmacológico , Ácidos Pipecólicos/farmacología , Recuperación de la Función/efectos de los fármacos , Hemorragia Subaracnoidea/tratamiento farmacológico , Trombina/antagonistas & inhibidores , Animales , Arginina/análogos & derivados , Barrera Hematoencefálica/efectos de los fármacos , Edema Encefálico/sangre , Edema Encefálico/patología , Muerte Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Actividad Motora/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/sangre , Hemorragia Subaracnoidea/patología , Sulfonamidas
8.
J Neurochem ; 111(3): 726-36, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19712057

RESUMEN

The present study was designed to investigate the role of matrix metalloproteinases (MMPs) in the immature brain and the long term effects of early MMPs inhibition after hypoxic-ischemic (HI) injury. HI was induced by unilateral ligation of the right carotid artery followed by hypoxia (8% O(2) for 2 h) in P7 rat pups. GM6001, a broad spectrum MMPs inhibitor, was injected (50 mg/kg or 100 mg/kg) intraperitoneally at 2 h and 24 h after HI injury. Blood-brain barrier (BBB) integrity, brain edema, MMP-2/-9 activity, TIMP-1/-2 and tight junction protein (TJP) level were evaluated using IgG staining, Evan's blue extravasation, brain water content, zymography and western blot. Doxycycline, another MMPs inhibitor, was injected (10 mg/kg or 30 mg/kg) intraperitoneally at 2 h after HI, then BBB integrity and brain edema were measured at 48 h post-HI using brain water content measurement and IgG staining. The long-term effects of early MMPs inhibition (GM6001, 100 mg/kg) were evaluated by neurobehavioral tests, body weight, and brain atrophy measurement. GM6001 attenuated brain edema and BBB disruption at the dosage of 100 mg/kg. MMP-2 activity increased at 24 h and peaked at 48 h after HI, whereas MMP-9 activity peaked at 24 h and tapered by 48 h after HI. MMP-9/-2 activities were significantly attenuated by GM6001 at 24 h and 48 h after HI. The degradation of TJPs (ZO-1 and occludin) at 48 h after HI was reversed by GM6001 treatment. Early MMPs inhibition had long-term effects that attenuated ipsilateral brain tissue loss, and improved neurobehavioral outcomes after HI. These results suggest that early MMPs inhibition with a broad-spectrum inhibitor provides both acute and long-term neuroprotection in the developing brain by reducing TJPs degradation, preserving BBB integrity, and ameliorating brain edema after neonatal HI injury.


Asunto(s)
Dipéptidos/uso terapéutico , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Metaloendopeptidasas/antagonistas & inhibidores , Fármacos Neuroprotectores/uso terapéutico , Análisis de Varianza , Animales , Animales Recién Nacidos , Atrofia/prevención & control , Barrera Hematoencefálica/efectos de los fármacos , Edema Encefálico/etiología , Edema Encefálico/prevención & control , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Hipoxia-Isquemia Encefálica/patología , Hipoxia-Isquemia Encefálica/fisiopatología , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Embarazo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/genética , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Proteína de la Zonula Occludens-1
9.
J Pineal Res ; 46(1): 43-8, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18573160

RESUMEN

The purpose of this study was to evaluate the effect of melatonin on oxidative stress occurring in the brain after routine lobectomy neurosurgery procedures. Different concentrations of melatonin (5, 15 and 150 mg/kg) were administered 1 hr before lobectomy in a rodent surgical brain injury (SBI) model. Neurological outcomes were assessed 24 hr before the killing of the rodents, for evaluation of brain water content (brain edema) and lipid peroxidation (oxidative stress). The results showed that lower doses (5 and 15 mg/kg) failed to reduce brain edema, but the 15 mg/kg dose did lower oxidative stress and improved several neurological parameters. High concentration of melatonin (150 mg/kg) significantly increased brain edema and elevated oxidative stress when compared with the vehicle-treated group. Furthermore, high-dose melatonin also worsened neurological outcomes compared with other groups. The study suggests that melatonin has dual effects: low-dose melatonin may provide neuroprotective effects against SBI but a high dose may aggravate some parameters after SBI.


Asunto(s)
Edema Encefálico/metabolismo , Lesiones Encefálicas/metabolismo , Encéfalo/efectos de los fármacos , Melatonina/farmacología , Estrés Oxidativo/efectos de los fármacos , Análisis de Varianza , Animales , Encéfalo/cirugía , Edema Encefálico/tratamiento farmacológico , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/etiología , Relación Dosis-Respuesta a Droga , Masculino , Destreza Motora/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Vibrisas
10.
Spine (Phila Pa 1976) ; 44(1): 17-24, 2019 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29894452

RESUMEN

STUDY DESIGN: Retrospective review. OBJECTIVE: Evaluate radiographic and clinical outcomes for infants and children, who underwent rigid occipito-cervical fixation for traumatic craniocervical dissociation (tCCD). SUMMARY OF BACKGROUND DATA: Traumatic craniocervical dissociation is devastating. Children are at high risk but make up a large number of survivors. Non-rigid fixation has traditionally been favored over screw and rod constructs due to inherent challenges involved with instrumenting the pediatric craniocervical junction. Therefore, outcomes for rigid occipito-cervical instrumentation in infants and young children with tCCD remain uncertain. METHODS: Retrospective review of children who survived tCCD between 2006 and 2016 and underwent rigid occipito-cervical fixation. RESULTS: Fifteen children, from 8 months to 8 years old (mean, 3.8 yr), were either a passenger (n = 11) or a pedestrian (n = 4) in a motor vehicle accident. Seven patients had weakness: five with quadriplegia, one with hemiparesis, and one with bilateral upper extremity paresis. Ten patients had concurrent C1-2 instability. At last follow-up, four patients had improved motor function: one with bilateral upper extremity paresis and one with hemiparesis regained full strength, one with quadriplegia regained function on one side while another regained function in bilateral upper extremities. All underwent rigid posterior occipito-cervical fixation, with two patients requiring additional anterior and posterior fixation at non-contiguous levels. Fourteen patients were stable on flexion-extension x-rays at a mean follow-up of 31 months (9-1 yr or longer, 7-2 yr or longer). There were no cases of deformity, growth disturbance, or subaxial instability. CONCLUSION: Children who survive tCCD may regain function after stabilization. Rigid internal rod and screw fixation in infants and young children safely provided long-term stability. We advocate using C2 translaminar screws to exploit the favorable anatomy of pediatric lamina to minimize the risks of occipitocervical (OC) instrumentation. LEVEL OF EVIDENCE: 4.


Asunto(s)
Vértebras Cervicales/cirugía , Fijación Interna de Fracturas , Fijadores Internos , Hueso Occipital/cirugía , Fusión Vertebral/métodos , Traumatismos del Sistema Nervioso/cirugía , Articulación Atlantoaxoidea/diagnóstico por imagen , Articulación Atlantoaxoidea/lesiones , Articulación Atlantoaxoidea/cirugía , Vértebras Cervicales/diagnóstico por imagen , Vértebras Cervicales/lesiones , Niño , Preescolar , Femenino , Estudios de Seguimiento , Fijación Interna de Fracturas/instrumentación , Fijación Interna de Fracturas/métodos , Humanos , Lactante , Masculino , Traumatismos del Cuello/diagnóstico por imagen , Traumatismos del Cuello/cirugía , Hueso Occipital/diagnóstico por imagen , Estudios Retrospectivos , Fusión Vertebral/instrumentación , Traumatismos del Sistema Nervioso/diagnóstico por imagen , Resultado del Tratamiento
11.
J Neurosci Res ; 86(16): 3635-43, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18683242

RESUMEN

The mechanisms involved in simvastatin-mediated attenuation of cerebral vasospasm after subarachnoid hemorrhage (SAH) are unclear. We investigated the role of the phosphatidylinositol 3-kinase/Akt (PI3K/Akt) pathway and endothelial nitric oxide synthase (eNOS) in the cerebral vasculature in statin-mediated attenuation of cerebral vasospasm using wortmannin, an irreversible pharmacological PI3K inhibitor, and a rat SAH endovascular perforation model. Simvastatin was administered intraperitoneally in two dosages (1 mg/kg and 20 mg/kg) at 0.5, 24, and 48 hr after SAH and histological parameters of ipsilateral intracranial carotid artery (ICA) were assessed at 24 and 72 hr. SAH significantly decreased ICA diameter and perimeter while increasing wall thickness at both 24 and 72 hr. High-dosage simvastatin prevented the reduction of ICA diameter and perimeter following SAH, whereas both high and low dosages reduced wall thickness significantly at 24 and 72 hr. The effects of simvastatin were significantly reversed by wortmannin. High-dosage simvastatin increased pAkt and peNOS (phosphorylated forms) levels without increasing Akt and eNOS expression compared with the SAH group and also improved neurological deficits at 24 and 72 hr. Simvastatin did not affect protein levels by itself compared with untreated sham group. The present study elucidates the critical role of the PI3K activation leading to phosphorylation of Akt and eNOS in simvastatin-mediated attenuation of cerebral vasospasm after SAH.


Asunto(s)
Óxido Nítrico Sintasa de Tipo III/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Simvastatina/farmacología , Hemorragia Subaracnoidea/complicaciones , Vasoespasmo Intracraneal/tratamiento farmacológico , Vasoespasmo Intracraneal/metabolismo , Androstadienos/farmacología , Animales , Estenosis Carotídea/tratamiento farmacológico , Estenosis Carotídea/etiología , Estenosis Carotídea/patología , Arterias Cerebrales/efectos de los fármacos , Arterias Cerebrales/metabolismo , Arterias Cerebrales/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Masculino , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Simvastatina/uso terapéutico , Hemorragia Subaracnoidea/fisiopatología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Vasoespasmo Intracraneal/fisiopatología , Wortmanina
12.
J Neurosci Methods ; 167(2): 327-34, 2008 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-17870179

RESUMEN

The endovascular perforation rodent model for experimental subarachnoid hemorrhage (SAH) studies is criticized for lack of control over bleeding. Presently, there is no practical grading system to categorize the severity of SAH depending on the amount of blood. We outline a simple and objective novel SAH grading system by examining the subarachnoid blood clots in the basal cisterns, and evaluate for correlation with neurological status and cerebral vasospasm. Effects of simvastatin, known to reduce vasospasm, were examined using this grading system. Seventy-seven adult male Sprague-Dawley rats were divided randomly into three groups: sham-operated (n=24), SAH (n=32), and SAH+simvastatin (n=25). High-resolution brain pictures were used to grade the severity of SAH and categorize animals into mild, moderate and severe groups. The SAH grades were compared with neurological scores and internal carotid artery parameters such as diameter, perimeter and wall thickness at 24h. Two investigators verified the grading system independently. The SAH grade showed linear correlation functionally with neurological status (r=0.42, p<0.01) and morphometrically with the degree of vasospasm (|r|>0.7, p<0.01), and also between two independent investigators (r=0.937, p<0.001). Simvastatin improved neurological score in moderate and severe (p<0.05) but not mild SAH groups (p=0.28). This grading system has the potential to be adopted for SAH experimental rodent models.


Asunto(s)
Hemorragia/tratamiento farmacológico , Hemorragia/etiología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Simvastatina/uso terapéutico , Hemorragia Subaracnoidea/complicaciones , Análisis de Varianza , Animales , Arteria Basilar/efectos de los fármacos , Arteria Basilar/fisiopatología , Modelos Animales de Enfermedad , Estudios de Evaluación como Asunto , Masculino , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Índice de Severidad de la Enfermedad , Estadística como Asunto , Hemorragia Subaracnoidea/patología , Vasoespasmo Intracraneal/tratamiento farmacológico , Vasoespasmo Intracraneal/etiología
13.
J Pineal Res ; 44(2): 197-204, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18289172

RESUMEN

Aneurysmal subarachnoid hemorrhage (SAH) is a devastating disease that is associated with significant morbidity and mortality. There is substantial evidence to suggest that oxidative stress is significant in the development of acute brain injury following SAH. Melatonin is a strong antioxidant that has low toxicity and easily passes through the blood-brain barrier. Previous studies have shown that melatonin provides neuroprotection in animal models of ischemic stroke. This study hypothesizes that melatonin will provide neuroprotection when administered 2 hr after SAH. The filament perforation model of SAH was performed in male Sprague-Dawley rats weighing between 300 and 380 g. Melatonin (15 or 150 mg/kg), or vehicle was given via intraperitoneal injection 2 hr after SAH. Mortality and neurologic deficits were assessed 24 hr after SAH. A significant reduction in 24-hr mortality was seen following treatment with high dose melatonin. There was no improvement in neurologic scores with treatment. Brain water content and lipid peroxidation were measured following the administration of high dose melatonin to identify a mechanism for the increased survival. High dose melatonin tended to reduce brain water content following SAH, but had no effect on the lipid peroxidation of brain samples. Large doses of melatonin significantly reduces mortality and brain water content in rats following SAH through a mechanism unrelated to oxidative stress.


Asunto(s)
Melatonina/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Hemorragia Subaracnoidea/mortalidad , Hemorragia Subaracnoidea/prevención & control , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Modelos Animales de Enfermedad , Masculino , Melatonina/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Índice de Severidad de la Enfermedad , Hemorragia Subaracnoidea/metabolismo
14.
Acta Neurochir Suppl ; 102: 327-30, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19388339

RESUMEN

BACKGROUND: Aneurysmal subarachnoid hemorrhage (SAH) is a devastating disease that is associated with significant morbidity and mortality. There is substantial evidence to suggest that oxidative stress is significant in the development of acute brain injury following SAH. Melatonin is a strong antioxidant that has low toxicity and easily passes through the BBB. Previous studies have shown that melatonin provides neuroprotection in other models of CNS injury. METHODS: This experiment evaluates melatonin as a neuroprotectant against early brain injury following SAH. The endovascular perforation model of SAH was performed in male Sprague Dawley rats followed by the administration of melatonin two hours after the insult. Mortality and brain water content were assessed 24 after SAH. FINDINGS: A significant reduction in 24 h mortality was seen following treatment with 150 mg/kg of melatonin. Brain water content was evaluated in the high dose treatment group to see if a reduction in brain edema was associated with reduced mortality. High dose melatonin tended to reduce brain water content following SAH. CONCLUSIONS: Large doses of melatonin significantly reduced mortality and brain water content in rats following SAH.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Melatonina/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Masculino , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/complicaciones , Factores de Tiempo
15.
Acta Neurochir Suppl ; 102: 391-4, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19388353

RESUMEN

BACKGROUND: Cerebral vasospasm is a common sequelae of subarachonoid hemorrhage (SAH), however, the mechanism of cerebral vasospasm is still unclear. Recently, statins have been shown to have efficacy in ameliorating cerebral vasospasm. The present study investigates whether simvastatin attenuates cerebral vasospasm after subarachnoid hemorrhage (SAH) via upregulation of the PI3K/Akt pathway. METHODS: 47 adult male Sprague-Dawley rats were divided into 6 groups: sham-operated, SAH treated with vehicle, SAH treated with low dose simvastatin (1 mg/kg), high dose simvastatin (20 mg/kg), SAH treated with simvastatin plus the PI3K inhibitor (wortmannin), and sham-operated plus wortmannin. Simvastatin was administered intraperitoneally 30 minutes after SAH created by the standard endovascular perforation model. Histological parameters of the ipsilateral internal carotid artery (ICA-diameter, perimeter, and wall thickness) and neurological score were assessed at 24 hours. FINDINGS: Mortality was reduced to zero in both the treated groups as compared to 20% in the vehicle-treated and 36% in the simvastatin plus wortmannin-treated groups. The decrease in ICA diameter and perimeter observed in vehicle-treated group (203.2 +/- 10.3 microm, 652.7 +/- 29.0 microm) as compared to sham (259.7 +/- 10.6, 865.4 +/- 39.5) were significantly attenuated by high-dose simvastatin (267.4 +/- 8.0, 882.4 +/- 30.0). The increase in wall thickness (vehicle 29.50 +/- 2.42 microm v/s sham 9.52 +/- 0.56 microm) was significantly attenuated by both high and low dose simvastatin (11.87 +/- 1.56, 19.75 +/- 1.40). These effects of simvastatin were blocked with the addition of wortmannin (162.7 +/- 20.6, 528.9 +/- 65.9, 29.19 +/- 1.97). High dose simvastatin improved the neurological deficits after SAH, but this was also blocked by wortmannin. CONCLUSIONS: The beneficial effects of high dose simvastatin in ameliorating cerebral vasospasm are likely mediated by upregulation of the PI3K/Akt pathway.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Simvastatina/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos , Vasoespasmo Intracraneal/tratamiento farmacológico , Análisis de Varianza , Animales , Arteria Carótida Interna/efectos de los fármacos , Arteria Carótida Interna/patología , Arteria Carótida Interna/ultraestructura , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/complicaciones , Vasoespasmo Intracraneal/etiología , Vasoespasmo Intracraneal/patología
16.
Acta Neurochir Suppl ; 102: 367-71, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19388347

RESUMEN

BACKGROUND: Surgical brain injury (SBI) to normal brain tissue can occur as inevitable sequelae of neurosurgical operations. SBI can contribute to post-operative complications such as brain edema following blood-brain barrier (BBB) disruption leading to neurological deficits. Melatonin is a commonly used drug with known antioxidant properties and neuroprotective effects in experimental animal studies (Chen et al., J Pineal Res 41:175-182, 2006; Chen et al., J Pineal Res 40(3):242-250, 2006; Cheung, J Pineal Res 34:153-160, 2003; Lee et al., J Pineal Res 42(3):297-309, 2007; Reiter et al., Exp Biol Med (Maywood) 230(2):104-117, 2005). METHODS: We tested different concentrations of melatonin (5 mg/kg, 15 mg/kg and 150 mg/kg) administered 1 hour before surgery for neuroprotection against SBI using a rodent model. Post-operative assessment included brain water content (brain edema), lipid peroxidation assays (oxidative stress), and neurological assessment. FINDINGS: The results showed a trend in decreasing brain edema with lower doses of melatonin (5 mg/kg and 15 mg/ kg), however, high concentration of melatonin (150 mg/kg) significantly increased brain edema compared to all other groups. This deleterious effect of high-dose melatonin was also observed in lipid-peroxidation assay wherein lower-dose melatonin (15 mg/kg) attenuated oxidative stress, but high-dose melatonin (150 mg/kg) increased oxidative stress as compared to vehicle-treated group. Furthermore, high-dose melatonin also worsened neurological outcomes compared to other groups whereas; the low-dose melatonin group (15 mg/kg) showed some improved neurological parameters. CONCLUSIONS: The study suggests that low-dose melatonin may provide neuroprotective effects against SBI. Further studies are needed to confirm this. More importantly, the findings of the study stress the need to carefully reassess safety issues with high doses of melatonin, which is considered to be a practically non-toxic drug.


Asunto(s)
Lesiones Encefálicas/prevención & control , Melatonina/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Análisis de Varianza , Animales , Edema Encefálico/etiología , Edema Encefálico/prevención & control , Lesiones Encefálicas/complicaciones , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Peroxidación de Lípido/efectos de los fármacos , Examen Neurológico/métodos , Ratas
17.
Cancer Res ; 65(6): 2125-9, 2005 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15781622

RESUMEN

p53 is a critical tumor suppressor which removes cells with DNA damage by regulating expression and activity of a select group of p53-induced genes (PIG) that subsequently induce apoptosis. PIG8 was also identified as a gene induced by etoposide and named etoposide-induced gene 24 (EI24). Later experiments established EI24/PIG8 as a proapoptotic factor and suggested that it may function as a tumor suppressor. Indeed, EI24/PIG8 is relatively highly mutated in aggressive breast cancers and is located in a region which expresses frequent loss of heterozygosity. However, despite these important observations, the activity and role of EI24/PIG8 remain largely unknown. We used (immmuno)fluorescence microscopy and subcellular fractionation techniques to show that EI24/PIG8 is localized in the endoplasmic reticulum (ER). Pull-down experiments showed that it specifically binds with Bcl-2, a death regulator known to reside in mitochondria, ER, and the nuclear envelope. EI24/PIG8-Bcl-2 binding was corroborated by coimmunoprecipitation and other in vitro and in vivo protein-protein binding assays. Further analysis showed that EI24/PIG8 uses its N-terminal region to bind the BH3 domain in Bcl-2. Finally, we used immunohistochemical techniques to analyze expression of EI24/PIG8 in breast cancer tissue progression arrays and showed that loss of EI24/PIG8 is associated with tumor invasiveness but not with the development of the primary tumor. These results suggest that EI24/PIG8 is a novel, ER-localized Bcl-2-binding protein which may contribute to apoptosis by modulating the activity and/or function of Bcl-2 in this organelle. EI24/PIG8 may serve to prevent tumor spreading, consistent with its suspected role as a tumor suppressor.


Asunto(s)
Neoplasias de la Mama/patología , Retículo Endoplásmico/metabolismo , Proteínas Nucleares/fisiología , Fosfoproteínas/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Neoplasias de la Mama/metabolismo , Humanos , Ratones , Microscopía Fluorescente , Células 3T3 NIH , Invasividad Neoplásica , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/metabolismo , Mapeo Peptídico , Fosfoproteínas/biosíntesis , Fosfoproteínas/metabolismo , Unión Proteica , Estructura Terciaria de Proteína
18.
Case Rep Surg ; 2017: 3056285, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29201484

RESUMEN

Myeloid sarcoma, a rare consequence of myeloproliferative disorders, is rarely seen in the central nervous system, most commonly in the pediatric population. Although there are a handful of case reports detailing initial presentation of CNS myeloid sarcoma in the adult population, we have been unable to find any reports of CNS myeloid sarcoma presenting as a large mass lesion in a herniating patient. Here, we present the case of a patient transferred to our facility for a very large subdural hematoma. Based on imaging characteristics, it was felt to be a spontaneous hematoma secondary to coagulopathy. No coagulopathy was found. Interestingly, he did have a history of acute myeloid leukemia (AML) diagnosed 2 months previously, and intraoperatively he was found to have a confluent white mass invading both the subdural and subarachnoid spaces. There was minimal associated hemorrhage and final pathology showed myeloid sarcoma. This is the first report we are aware of in which CNS myeloid sarcoma presented as a subdural metastasis and also the first report in which we are aware of this etiology causing a herniation syndrome secondary to mass effect.

19.
J Neurosurg ; 116(1): 246-53, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22017304

RESUMEN

OBJECT: Intracranial surgery causes cortical injury from incisions, hemorrhage, retraction, and electrocautery. The term "surgical brain injury" (SBI) has been developed to categorize this injury inherent to the procedure. Neuroinflammation plays a significant role in SBI. Traditional antiinflammatory therapies are often limited by their immunosuppressive side effects and poor CNS penetration. This study uses mucosal tolerance to develop an immune system that is tolerant to brain myelin basic protein (MBP) so that inflammation can be suppressed in a timely and site-specific manner following surgical disruption of the blood-brain barrier. METHODS: A standard SBI model using CD57 mice was used. Nasopharyngeal mucosa was exposed to vehicle, ovalbumin, or MBP to develop mucosal tolerance to these antigens. Immunological tolerance to MBP was confirmed in vivo through hypersensitivity testing. Neurological scores, cerebral edema, and interleukin (IL)-1ß and transforming growth factor (TGF)-ß1 cytokine levels were measured 48 hours postoperatively. RESULTS: Hypersensitivity testing confirmed the development of immune tolerance to MBP. Myelin basic protein-tolerant mice demonstrated reduced neurological injury, less cerebral edema, decreased levels of IL-1ß, and increased levels of TGFß1 following SBI. CONCLUSIONS: Developing preoperative immunological tolerance to brain antigens through mucosal tolerance provides neuroprotection, reduces brain edema, and modulates neuroinflammation following SBI.


Asunto(s)
Lesiones Encefálicas/inmunología , Encéfalo/inmunología , Encefalitis/inmunología , Tolerancia Inmunológica/inmunología , Animales , Encéfalo/metabolismo , Edema Encefálico/inmunología , Edema Encefálico/metabolismo , Lesiones Encefálicas/metabolismo , Citocinas/metabolismo , Encefalitis/metabolismo , Inmunidad Mucosa , Ratones , Proteína Básica de Mielina/metabolismo
20.
J Neurosurg Pediatr ; 8(3): 316-20, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21882925

RESUMEN

Encephalocraniocutaneous lipomatosis (ECCL) is a rare neurocutaneous syndrome whose hallmark lesions are benign lipomas of the brain and spinal cord. The authors present a case of a male infant with ECCL who had extensive brainstem and spinal cord lipomas. The management of this patient's hydrocephalus, cervicomedullary compression, tethered cord, and scoliosis over the course of his first 2 years of life is described. This case report and review of the literature is presented to provide a synopsis of the problems likely to be encountered by neurosurgeons who treat patients with this syndrome.


Asunto(s)
Oftalmopatías/patología , Oftalmopatías/cirugía , Lipomatosis/patología , Lipomatosis/cirugía , Síndromes Neurocutáneos/patología , Síndromes Neurocutáneos/cirugía , Procedimientos Neuroquirúrgicos , Oftalmopatías/complicaciones , Humanos , Hidrocefalia/etiología , Hidrocefalia/patología , Hidrocefalia/cirugía , Lactante , Lipomatosis/complicaciones , Masculino , Defectos del Tubo Neural/etiología , Defectos del Tubo Neural/patología , Defectos del Tubo Neural/cirugía , Síndromes Neurocutáneos/complicaciones , Escoliosis/etiología , Escoliosis/patología , Escoliosis/cirugía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA