Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Childs Nerv Syst ; 37(4): 1363-1368, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32740674

RESUMEN

Thoracolumbar fractures in children are relatively uncommon and should be regarded as a separate entity from those in adults. While percutaneous pedicle fixation has emerged as an effective alternative to open fixation in adults with unstable thoracolumbar fractures, this technique is rarely applied in children. We report a 6-year-old girl with an L3 chance fracture, which was treated via short-segment percutaneous pedicle fixation. We also discussed the technical challenges and caveats of this surgical technique in young children. While potentially more challenging, percutaneous pedicle fixation is feasible in young children with thoracolumbar fractures. Specific differences between the developing and mature spine in regard to anatomical and biomechanical characteristics, including ligamentous laxity and intrinsic elasticity, should be taken into consideration. Future studies are needed to compare outcomes of minimally invasive spinal techniques to open surgery in children.


Asunto(s)
Tornillos Pediculares , Fracturas de la Columna Vertebral , Adulto , Niño , Preescolar , Femenino , Fijación Interna de Fracturas , Humanos , Vértebras Lumbares/diagnóstico por imagen , Vértebras Lumbares/cirugía , Fracturas de la Columna Vertebral/diagnóstico por imagen , Fracturas de la Columna Vertebral/cirugía , Vértebras Torácicas/diagnóstico por imagen , Vértebras Torácicas/cirugía , Resultado del Tratamiento
2.
Pediatr Emerg Care ; 37(10): e602-e608, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-30624426

RESUMEN

AIMS: The aims of this study were to document the injury pattern in pediatric traumatic craniocervical dissociation (CCD) and identify features of survivors. METHODS: Pediatric traumatic CCDs, diagnosed between January 2004 and July 2016, were reviewed. Survivors and nonsurvivors were compared. Categorical and continuous variables were analyzed with Fisher exact and t tests, respectively. RESULTS: Twenty-seven children were identified; 10 died (37%). The median age was 60 months (ranges, 6-109 months [survivors], 2-98 months [nonsurvivors]). For survivors, the median follow-up was 13.4 months (range, 1-109 months). The median time to mortality was 1.5 days (range, 1-7 days). The injury modality was motor vehicle collision in 18 (67%), pedestrian struck in 8 (30%), and 1 shaken infant (3%). For nonsurvivors, CCD was equally diagnosed by plain radiograph and head/cervical spine computed tomography scan. For survivors, CCD was diagnosed by computed tomography in 7 (41%), magnetic resonance imaging in 10 (59%), and none by radiograph. Seven diagnosed by magnetic resonance imaging (41%) had nondiagnostic initial imaging but persistent neck pain. Magnetic resonance imaging was obtained and was diagnostic of CCD in all 7 (P < 0.01). Survivors required significantly less cardiopulmonary resuscitation (P < 0.01), had lower Injury Severity Scores (P < 0.01), higher Glasgow Coma Scale scores (P < 0.01), and shorter transport times (P < 0.01). Significantly more involved in motor vehicle collisions survived (P = 0.04). Nine (53%) had no disability at follow-up evaluation. CONCLUSIONS: In pediatric CCD, high-velocity mechanism, cardiac arrest, high Injury Severity Score, and low Glasgow Coma Scale score are associated with mortality. If CCD is correctly managed in the absence of cardiac arrest or traumatic brain or spinal cord injury, children may survive intact.


Asunto(s)
Luxaciones Articulares , Vértebras Cervicales/diagnóstico por imagen , Niño , Preescolar , Escala de Coma de Glasgow , Humanos , Lactante , Puntaje de Gravedad del Traumatismo , Luxaciones Articulares/diagnóstico , Luxaciones Articulares/terapia , Estudios Retrospectivos
3.
J Neurosci Res ; 98(1): 105-120, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-30793349

RESUMEN

In addition to being the leading cause of morbidity and mortality in premature infants, germinal matrix hemorrhage (GMH) is also the leading cause of acquired infantile hydrocephalus. The pathophysiology of posthemorrhagic hydrocephalus (PHH) development after GMH is complex and vaguely understood, although evidence suggests fibrosis and gliosis in the periventricular and subarachnoid spaces disrupts normal cerebrospinal fluid (CSF) dynamics. Theories explaining general hydrocephalus etiology have substantially evolved from the original bulk flow theory developed by Dr. Dandy over a century ago. Current clinical and experimental evidence supports a new hydrodynamic theory for hydrocephalus development involving redistribution of vascular pulsations and disruption of Starling forces in the brain microcirculation. In this review, we discuss CSF flow dynamics, history and development of theoretical hydrocephalus pathophysiology, and GMH epidemiology and etiology as it relates to PHH development. We highlight known mechanisms and propose new avenues that will further elucidate GMH pathophysiology, specifically related to hydrocephalus.


Asunto(s)
Plexo Coroideo/metabolismo , Hidrocefalia/metabolismo , Hemorragias Intracraneales/metabolismo , Transducción de Señal/fisiología , Plexo Coroideo/patología , Humanos , Hidrocefalia/etiología , Hidrocefalia/patología , Lactante , Recién Nacido , Recien Nacido Prematuro , Hemorragias Intracraneales/complicaciones , Hemorragias Intracraneales/patología
4.
Stroke ; 49(12): 3020-3029, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30571407

RESUMEN

Background and Purpose- Heme and iron are considered to be key factors responsible for secondary insults after intracerebral hemorrhage (ICH). Our previous study showed that LRP1 (low-density lipoprotein receptor-related protein-1)-Hx (hemopexin) facilitates removal of heme. The TLR7 (Toll-like receptor 7)-BTK (Bruton tyrosine kinase)-CRT (calreticulin) pathway regulates the expression of LRP1-Hx. This study is designed to clarify whether TLR7 activation facilitates heme scavenging and to establish the potential role of the BTK-CRT-LRP1-Hx signaling pathway in the pathophysiology of ICH. Methods- ICH was induced by stereotactic, intrastriatal injection of type VII collagenase. Mice received TLR7 agonist (imiquimod) via intraperitoneal injection after ICH induction. TLR7 inhibitor (ODN2088), BTK inhibitor (LFM-A13), and CRT agonist (thapsigargin) were given in different groups to further evaluate the underlying pathway. Mice were randomly divided into sham, ICH+vehicle (normal saline), ICH+Imiquimod (2.5, 5, and 10 µg/g), ICH+ODN2088, ICH+LFM-A13, ICH+thapsigargin, and ICH+ODN2088+thapsigargin. Imiquimod was administered twice daily starting at 6 hours after ICH; ODN2088 was administered by intracerebroventricular injection at 30 minutes, and LFM-A13 or thapsigargin was administered by intraperitoneal injection at 3 hours after ICH induction. Neurological scores, cognitive abilities, as well as brain edema, blood-brain barrier permeability, hemoglobin level, brain expression of TLR7/BTK/CRT/LRP1/Hx were analyzed. Results- Low dosage imiquimod significantly attenuated hematoma volume, brain edema, BBB permeability, and neurological deficits after ICH. Imiquimod also increased protein expressions of TLR7, BTK, CRT, LRP1, and Hx; ODN2088 reduced TLR7, BTK, CRT, LRP1, and Hx expressions. Conclusions- TLR7 plays an important role in heme scavenging after ICH by modulating the BTK-CRT-LRP1-Hx pathway. TLR7 may offer protective effects by promoting heme resolution and reduction of brain edema after ICH.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/metabolismo , Encéfalo/metabolismo , Calreticulina/metabolismo , Hemorragia Cerebral/metabolismo , Hemo/metabolismo , Hemopexina/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de LDL/metabolismo , Receptor Toll-Like 7/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Agammaglobulinemia Tirosina Quinasa/efectos de los fármacos , Amidas/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Edema Encefálico/metabolismo , Calreticulina/agonistas , Calreticulina/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Hemopexina/efectos de los fármacos , Imiquimod/farmacología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/efectos de los fármacos , Ratones , Nitrilos/farmacología , Oligodesoxirribonucleótidos/farmacología , Receptores de LDL/efectos de los fármacos , Transducción de Señal , Tapsigargina/farmacología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/antagonistas & inhibidores , Receptor Toll-Like 7/efectos de los fármacos , Proteínas Supresoras de Tumor/efectos de los fármacos
5.
J Neurochem ; 140(5): 776-786, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28054340

RESUMEN

Fingolimod, a sphingosine-1-phosphate receptor (S1PR) agonist, is clinically available to treat multiple sclerosis and is showing promise in treating stroke. We investigated if fingolimod provides long-term protection from experimental neonatal germinal matrix hemorrhage (GMH), aiming to support a potential mechanism of acute fingolimod-induced protection. GMH was induced in P7 rats by infusion of collagenase (0.3 U) into the right ganglionic eminence. Animals killed at 4 weeks post-GMH received low- or high-dose fingolimod (0.25 or 1.0 mg/kg) or vehicle, and underwent neurocognitive testing before histopathological evaluation. Subsequently, a cohort of animals killed at 72 h post-GMH received 1.0 mg/kg fingolimod; the specific S1PR1 agonist, SEW2871; or fingolimod co-administered with the S1PR1/3/4 inhibitor, VPC23019, or the Rac1 inhibitor, EHT1864. All drugs were injected intraperitoneally 1, 24, and 48 h post-surgery. At 72 h post-GMH, brain water content, extravasated Evans blue dye, and hemoglobin were measured as well as the expression levels of phospho-Akt, Akt, GTP-Rac1, Total-Rac1, ZO1, occludin, and claudin-3 determined. Fingolimod significantly improved long-term neurocognitive performance and ameliorated brain tissue loss. At 72 h post-GMH, fingolimod reduced brain water content and Evans blue dye extravasation as well as reversed GMH-induced loss of tight junctional proteins. S1PR1 agonism showed similar protection, whereas S1PR or Rac1 inhibition abolished the protective effect of fingolimod. Fingolimod treatment improved functional and morphological outcomes after GMH, in part, by tempering acute post-hemorrhagic blood-brain barrier disruption via the activation of the S1PR1/Akt/Rac1 pathway.


Asunto(s)
Clorhidrato de Fingolimod/farmacología , Hemorragias Intracraneales/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Proteína de Unión al GTP rac1/metabolismo , Animales , Agua Corporal/metabolismo , Encéfalo/patología , Química Encefálica/efectos de los fármacos , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Cognición/efectos de los fármacos , Femenino , Hemorragias Intracraneales/metabolismo , Hemorragias Intracraneales/psicología , Recuento de Leucocitos , Masculino , Oxadiazoles/farmacología , Fosfoserina/análogos & derivados , Fosfoserina/farmacología , Embarazo , Pironas/farmacología , Quinolinas/farmacología , Ratas , Ratas Sprague-Dawley , Tiofenos/farmacología , Proteínas de Uniones Estrechas/metabolismo , Proteína de Unión al GTP rac1/antagonistas & inhibidores
6.
Neurobiol Dis ; 87: 124-33, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26739391

RESUMEN

Germinal matrix hemorrhage remains the leading cause of morbidity and mortality in preterm infants in the United States with little progress made in its clinical management. Survivors are often afflicted with long-term neurological sequelae, including cerebral palsy, mental retardation, hydrocephalus, and psychiatric disorders. Blood clots disrupting normal cerebrospinal fluid circulation and absorption after germinal matrix hemorrhage are thought to be important contributors towards post-hemorrhagic hydrocephalus development. We evaluated if upregulating CD36 scavenger receptor expression in microglia and macrophages through PPARγ stimulation, which was effective in experimental adult cerebral hemorrhage models and is being evaluated clinically, will enhance hematoma resolution and ameliorate long-term brain sequelae using a neonatal rat germinal matrix hemorrhage model. PPARγ stimulation (15d-PGJ2) increased short-term PPARγ and CD36 expression levels as well as enhanced hematoma resolution, which was reversed by a PPARγ antagonist (GW9662) and CD36 siRNA. PPARγ stimulation (15d-PGJ2) also reduced long-term white matter loss and post-hemorrhagic ventricular dilation as well as improved neurofunctional outcomes, which were reversed by a PPARγ antagonist (GW9662). PPARγ-induced upregulation of CD36 in macrophages and microglia is, therefore, critical for enhancing hematoma resolution and ameliorating long-term brain sequelae.


Asunto(s)
Antígenos CD36/metabolismo , Hematoma/fisiopatología , Hemorragias Intracraneales/fisiopatología , PPAR gamma/metabolismo , Anilidas/farmacología , Animales , Animales Recién Nacidos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/fisiopatología , Antígenos CD36/genética , Fármacos del Sistema Nervioso Central/farmacología , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Hematoma/tratamiento farmacológico , Hematoma/patología , Hemorragias Intracraneales/tratamiento farmacológico , Hemorragias Intracraneales/patología , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/fisiología , Microglía/efectos de los fármacos , Microglía/fisiología , Fármacos Neuroprotectores/farmacología , PPAR gamma/antagonistas & inhibidores , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacología , ARN Interferente Pequeño/administración & dosificación , Distribución Aleatoria , Ratas Sprague-Dawley , Regulación hacia Arriba
7.
Acta Neurochir Suppl ; 121: 203-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463949

RESUMEN

Germinal matrix hemorrhage (GMH) is a major cause of brain damage in prematurity and has long-lasting neurological implications. The development of brain inflammation contributes to brain injury, leading to a lifetime of neurologic deficits. PAR-1 and 4 receptors are involved with inflammatory pathways after brain hemorrhage in adult models of stroke, of which cyclooxygenase-2 (COX-2) is a potential mediator. We therefore hypothesized a role for PAR-1, 4/ COX-2 signaling following GMH. Postnatal day 7 Sprague-Dawley rats were subjected to GMH induction, which entailed stereotactic collagenase infusion into the ganglionic eminence. Animals were euthanized at two time points: 72 h (short-term) or 4 weeks (long-term). Short-term COX-2 expression was evaluated in the context of PAR-1 (SCH-79797) and PAR-4 (P4pal10) inhibition. Pups in the long-term group were administered the selective COX-2 inhibitor (NS-398); and the neurobehavioral and pathological examinations were performed 4 weeks later. Pharmacological PAR-1, 4 antagonism normalized COX-2 expression following GMH and reduced hydrocephalus. Early inhibition of COX-2 by NS-398 improved long-term neurobehavioral outcomes. COX-2 signaling plays an important role in brain injury following neonatal GMH, possibly through upstream PAR-1, 4 receptor mechanisms.


Asunto(s)
Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Ciclooxigenasa 2/efectos de los fármacos , Hemorragias Intracraneales/metabolismo , Oligopéptidos/farmacología , Pirroles/farmacología , Quinazolinas/farmacología , Animales , Animales Recién Nacidos , Western Blotting , Encéfalo/patología , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Hemorragias Intracraneales/patología , Hemorragias Intracraneales/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptor PAR-1/antagonistas & inhibidores , Receptores de Trombina/antagonistas & inhibidores
8.
Acta Neurochir Suppl ; 121: 213-6, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463951

RESUMEN

Germinal matrix hemorrhage (GMH) is the most common cause of neurological complications of prematurity and has lasting implications. PAR-1 and PAR-4 receptors are involved with upstream signaling pathways following brain hemorrhage in adult models of stroke, of which the mammalian target of rapamycin (mTOR) is a potential downstream mediator. Therefore, we hypothesized a role for PAR-1, -4/ mTOR signaling following GMH brain injury. Postnatal day 7 Sprague-Dawley rats were subjected to GMH through stereotactic infusion of collagenase into the right ganglionic eminence. Rodents were euthanized at 72 h (short term), or 4 weeks (long term). Short-term mTOR expression was evaluated by Western blot in the context of PAR-1 (SCH-79797) and PAR-4 (P4pal10) inhibition. Pups in the long-term group were administered the selective mTOR inhibitor (rapamycin) with neurobehavioral and brain pathological examinations performed at 4 weeks. Pharmacological PAR-1, -4 antagonism normalized the increased mTOR expression following GMH. Early inhibition of mTOR by rapamycin improved long-term outcomes in rats. Mammalian-TOR signaling plays an important role in brain injury following neonatal GMH, possibly involving upstream PAR-1, -4 mechanisms.


Asunto(s)
Encéfalo/efectos de los fármacos , Hemorragias Intracraneales/metabolismo , Oligopéptidos/farmacología , Pirroles/farmacología , Quinazolinas/farmacología , Receptor PAR-1/antagonistas & inhibidores , Receptores de Trombina/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/efectos de los fármacos , Trombina/metabolismo , Animales , Animales Recién Nacidos , Conducta Animal/efectos de los fármacos , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Inmunosupresores/farmacología , Hemorragias Intracraneales/patología , Hemorragias Intracraneales/fisiopatología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
9.
Acta Neurochir Suppl ; 121: 237-41, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463955

RESUMEN

Hemorrhagic transformation occurs in as many as 48 % of stroke patients and is a major contributor to post-insult morbidity and mortality. Experimental models of hemorrhagic transformation are utilized for understanding the mechanisms behind its development, as well as for investigating potential therapeutics for prevention and reduction of bleeding. Thoroughly studying animal models of hemorrhagic transformation is critically important for testing novel treatments. Thus far, no study has examined the progression of brain swelling and hemorrhagic transformation after transient middle cerebral artery occlusion (MCAO). Herein, we investigate the development of infarction, brain swelling, and hemorrhagic transformation following MCAO in hyperglycemic rats. Twenty-five Sprague-Dawley rats were subjected to either 1.5 h of MCAO or sham surgery 15 min after induction of hyperglycemia. Animals were sacrificed at 0.25, 1, 3, or 24 h after reperfusion for measurement of infarct volume, brain swelling, and hemoglobin volume. Within 15 min of reperfusion, the infarct volume was significantly larger than in sham animals and did not increase in size over the 24 h. However, both brain swelling and hemorrhagic transformation, which began immediately after reperfusion, increase over 24 h after reperfusion.


Asunto(s)
Glucemia/metabolismo , Edema Encefálico/metabolismo , Hemorragia Cerebral/metabolismo , Hiperglucemia/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Animales , Edema Encefálico/etiología , Hemorragia Cerebral/etiología , Modelos Animales de Enfermedad , Glucosa/farmacología , Hiperglucemia/inducido químicamente , Infarto de la Arteria Cerebral Media/complicaciones , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Riesgo , Edulcorantes/farmacología , Factores de Tiempo
10.
Neurobiol Dis ; 82: 349-358, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26176793

RESUMEN

BACKGROUND AND PURPOSE: Edema formation, inflammation and increased blood-brain barrier permeability contribute to poor outcomes after intracerebral hemorrhage (ICH). This study examined the therapeutic effect of dimethyl fumarate (DMF), a fumaric acid ester that activates nuclear factor erythroid-2 related factor 2 (Nrf2) and Nrf2 heterodimerization effector protein musculo-aponeurotic fibrosarcoma-G (MAFG) in a murine ICH model. METHODS: Male CD-1 mice (n=176) were subjected to intrastriatal infusion of bacterial collagenase (n=126), autologous blood (n=18) or sham surgery (n=32). Four (4) animals not subjected to ICH (naive) were also included in the study. After ICH, animals either received vehicle, dimethyl fumarate (10 mg or 100 mg/kg) or casein kinase 2 inhibitor (E)-3-(2,3,4,5-tetrabromophenyl)acrylic acid (TBCA). Thirty-two mice also received scrambled siRNA or MAFG siRNA 24h before ICH. Brain water content and neurological function were evaluated. RESULTS: Dimethyl fumarate reduced Evans blue dye extravasation, decreased brain water content, and improved neurological deficits at 24 and 72 h after ICH. Casein kinase 2 inhibitor TBCA and MAFG siRNA prevented the effect of dimethyl fumarate on brain edema and neurological function. After ICH, ICAM-1 levels increased and casein kinase 2 levels decreased. Dimethyl fumarate reduced ICAM-1 but enhanced casein kinase 2 levels. Again, casein kinase 2 inhibitor TBCA and MAFG siRNA abolished the effect of dimethyl fumarate on ICAM-1 and casein kinase 2. Dimethyl fumarate preserved pNrf2 and MAFG expression in the nuclear lysate after ICH and the effect of dimethyl fumarate was abolished by casein kinase 2 inhibitor TBCA and MAFG siRNA. Dimethyl fumarate reduced microglia activation in peri-hematoma areas after ICH. The protective effect of dimethyl fumarate on brain edema and neurological function was also observed in a blood injection mouse model. CONCLUSION: Dimethyl fumarate ameliorated inflammation, reduced blood-brain barrier permeability, and improved neurological outcomes by casein kinase 2 and Nrf2 signaling pathways after experimental ICH in mice.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Hemorragia Cerebral/tratamiento farmacológico , Dimetilfumarato/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Fármacos Neuroprotectores/farmacología , Acrilatos/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/enzimología , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/enzimología , Quinasa de la Caseína II/antagonistas & inhibidores , Hemorragia Cerebral/enzimología , Colagenasas , Modelos Animales de Enfermedad , Molécula 1 de Adhesión Intercelular/metabolismo , Factor de Transcripción MafG/genética , Factor de Transcripción MafG/metabolismo , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/enzimología , Neuroinmunomodulación/efectos de los fármacos , Neuroinmunomodulación/fisiología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
11.
J Neurosci Res ; 93(1): 94-103, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25196554

RESUMEN

Blood-brain barrier (BBB) disruption and consequent edema formation contribute to the development of early brain injury following subarachnoid hemorrhage (SAH). Various cerebrovascular insults result in increased platelet-derived growth factor receptor (PDGFR)-α stimulation, which has been linked to BBB breakdown and edema formation. This study examines whether imatinib, a PDGFR inhibitor, can preserve BBB integrity in a rat endovascular perforation SAH model. Imatinib (40 or 120 mg/kg) or a vehicle was administered intraperitoneally at 1 hr after SAH induction. BBB leakage, brain edema, and neurological deficits were evaluated. Total and phosphorylated protein expressions of PDGFR-α, c-Src, c-Jun N-terminal kinase (JNK), and c-Jun were measured, and enzymatic activities of matrix metalloproteinase (MMP)-2 and MMP-9 were determined in the injured brain. Imatinib treatment significantly ameliorated BBB leakage and edema formation 24 hr after SAH, which was paralleled by improved neurological functions. Decreased brain expressions of phosphorylated PDGFR-α, c-Src, JNK, and c-Jun as well as reduced MMP-9 activities were found in treated animals. PDGFR-α inhibition preserved BBB integrity following experimental SAH; however, the protective mechanisms remain to be elucidated. Targeting PDGFR-α signaling might be advantageous to ameliorate early brain injury following SAH.


Asunto(s)
Benzamidas/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Piperazinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Hemorragia Subaracnoidea/patología , Animales , Barrera Hematoencefálica/fisiopatología , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Permeabilidad Capilar/fisiología , Modelos Animales de Enfermedad , Mesilato de Imatinib , Inmunoprecipitación , MAP Quinasa Quinasa 4/metabolismo , Masculino , Examen Neurológico , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
12.
Cell Mol Neurobiol ; 35(1): 85-99, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25257832

RESUMEN

Autophagy involves degradation of dysfunctional cellular components through the actions of lysosomes. Apoptosis is the process of programmed cell death involving a series of characteristic cell changes. Autophagy and apoptosis, as self-destructive processes, play an important role in the pathogenesis of neurological diseases; and a crosstalk between "self-eating" (autophagy) and "self-killing" (apoptosis) plays an important role in pathological cellular adaptation. Expert knowledge of autophagy and apoptosis has increased in recent years, particularly in regards to cellular and molecular mechanisms. The crosstalk between autophagy and apoptosis was partially uncovered and several key molecules, including Bcl-2 family members, Beclin 1, and p53 were identified. However, the precise mechanisms of such a crosstalk remain to be elucidated. This current review article aims to summarize key mediators of the autophagy-apoptosis crosstalk in pathological conditions, and to highlight recent advances in the field, as well as to discuss further investigations and therapeutic potentials of manipulating those mechanisms in central nervous system diseases.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/fisiología , Autofagia/fisiología , Enfermedades del Sistema Nervioso Central/metabolismo , Genes p53/fisiología , Proteínas de la Membrana/metabolismo , Animales , Beclina-1 , Enfermedades del Sistema Nervioso Central/patología , Humanos
13.
Stroke ; 45(8): 2475-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24947291

RESUMEN

BACKGROUND AND PURPOSE: This study investigated if acute and delayed deferoxamine treatment attenuates long-term sequelae after germinal matrix hemorrhage (GMH). METHODS: Bacterial collagenase (0.3 U) was infused intraparenchymally into the right hemispheric ganglionic eminence in P7 rat pups to induce GMH. GMH animals received either deferoxamine or vehicle twice a day for 7 consecutive days. Deferoxamine administration was initiated at either 1 hour or 72 hours post-GMH. Long-term neurocognitive deficits and motor coordination were assessed using Morris water maze, rotarod, and foot fault tests between day 21 to 28 post-GMH. At 28 days post-GMH, brain morphology was assessed and extracellular matrix protein (fibronectin and vitronectin) expression was determined. RESULTS: Acute and delayed deferoxamine treatment improved long-term motor and cognitive function at 21 to 28 days post-GMH. Attenuated neurofunction was paralleled with improved overall brain morphology at 28 days post-GMH, reducing white matter loss, basal ganglia loss, posthemorrhagic ventricular dilation, and cortical loss. GMH resulted in significantly increased expression of fibronectin and vitronectin, which was reversed by acute and delayed deferoxamine treatment. CONCLUSIONS: Acute and delayed deferoxamine administration ameliorated long-term sequelae after GMH.


Asunto(s)
Encéfalo/efectos de los fármacos , Deferoxamina/uso terapéutico , Hemorragias Intracraneales/tratamiento farmacológico , Aprendizaje por Laberinto/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Animales , Animales Recién Nacidos , Deferoxamina/administración & dosificación , Modelos Animales de Enfermedad , Hemorragias Intracraneales/fisiopatología , Aprendizaje por Laberinto/fisiología , Actividad Motora/fisiología , Ratas , Factores de Tiempo
14.
Neurocirugia (Astur : Engl Ed) ; 35(1): 51-56, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-36934973

RESUMEN

Neurosurgical management of basilar invagination (BI) has traditionally been aimed at direct cervicomedullary decompression through transoral dens resection or suboccipital decompression with supplemental instrumented fixation. Dr. Goel introduced chronic atlantoaxial dislocation (AAD) as the etiology in most cases of BI and described a technique for distracting the C1-C2 joint with interfacet spacers to achieve reduction and anatomic realignment. We present our modification to Goel's surgical technique, in which we utilize anterior cervical discectomy (ACD) cages as C1-C2 interfacet implants. A young adult male presented to our institution with BI, cervicomedullary compression, occipitalization of C1, and Chiari 1 malformation. There was AAD of C1 over the C2 lateral masses. This reduced some with preoperative traction. He underwent successful C1-C2 interfacet joint reduction and arthrodesis with anterior cervical discectomy (ACD) cages and concomittant occiput to C2 instrumented fusion. BI can be effectively treated through reduction of AAD and by utilizing ACD cages as interfacet spacers.


Asunto(s)
Articulación Atlantoaxoidea , Luxaciones Articulares , Adulto Joven , Masculino , Humanos , Articulación Atlantoaxoidea/diagnóstico por imagen , Articulación Atlantoaxoidea/cirugía , Luxaciones Articulares/diagnóstico por imagen , Luxaciones Articulares/cirugía , Descompresión Quirúrgica/métodos
15.
Neuroscience ; 549: 138-144, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38734302

RESUMEN

Postoperative stroke is a challenging and potentially devastating complication after elective carotid endarterectomy (CEA). We previously demonstrated that transmembrane protein 166 (TMEM166) levels were directly related to neuronal damage after cerebral ischemia-reperfusion injury in rats. In this subsequent clinical study, we aimed to evaluate the prognostic value of TMEM166 in patients suffering from post-CEA strokes. Thirty-five patients undergoing uncomplicated elective CEA and 8 patients who suffered ischemic strokes after CEA were recruited. We evaluated the protein level and expression of TMEM166 in patients diagnosed with postoperative strokes and compared it to those in patients who underwent uncomplicated elective CEA. Blood samples and carotid artery plaques were collected and analyzed. High expressions of TMEM166 were detected by immunofluorescence staining and Western Blot in carotid artery plaques of all patients who underwent CEA. Furthermore, circulating TMEM166 concentrations were statistically higher in post-CEA stroke patients than in patients allocated to the control group. Mean plasma concentrations of inflammatory markers, including interleukin 6 (IL-6) and C-reactive protein (CRP), were also elevated in patients with postoperative strokes. Therefore, based on these findings, we hypothesize that elevated TMEM166 levels, accompanied by a strong inflammatory response, serve as a useful biomarker for risk assessment of postoperative stroke following CEA.


Asunto(s)
Endarterectomía Carotidea , Proteínas de la Membrana , Complicaciones Posoperatorias , Accidente Cerebrovascular , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Biomarcadores/sangre , Proteína C-Reactiva/metabolismo , Estenosis Carotídea/cirugía , Endarterectomía Carotidea/efectos adversos , Interleucina-6/sangre , Interleucina-6/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso , Complicaciones Posoperatorias/metabolismo , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/sangre
16.
Stroke ; 44(6): 1743-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23613493

RESUMEN

BACKGROUND AND PURPOSE: Blood-brain barrier disruption and consequent vasogenic edema formation codetermine the clinical course of intracerebral hemorrhage (ICH). This study examined the effect of PHA-543613, a novel α7 nicotinic acetylcholine receptor agonist, on blood-brain barrier preservation after ICH. METHODS: Male CD-1 mice, subjected to intrastriatal blood infusion, received PHA-543613 alone or in combination with α7 nicotinic acetylcholine receptor antagonist methyllycaconitine or phosphatidylinositol 3-kinase inhibitor wortmannin. RESULTS: PHA-543613 alone, but not in combination with methyllycaconitine or wortmannin, inhibited glycogen synthase kinase-3ß, thus, stabilizing ß-catenin and tight junction proteins, which was paralleled by improved blood-brain barrier stability and ameliorated neurofunctional deficits in ICH animals. CONCLUSIONS: PHA-543613 preserved blood-brain barrier integrity after ICH, possibly through phosphatidylinositol 3-kinase-Akt-induced inhibition of glycogen synthase kinase-3ß and ß-catenin stabilization.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Hemorragia Cerebral/fisiopatología , Quinuclidinas/farmacología , Receptores Nicotínicos/fisiología , Transducción de Señal/efectos de los fármacos , Aconitina/análogos & derivados , Aconitina/farmacología , Androstadienos/farmacología , Animales , Barrera Hematoencefálica/fisiología , Hemorragia Cerebral/metabolismo , Claudina-3/metabolismo , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta , Masculino , Ratones , Ratones Endogámicos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Wortmanina , beta Catenina/metabolismo
17.
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
18.
Stroke ; 44(12): 3587-90, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24149004

RESUMEN

BACKGROUND AND PURPOSE: This study investigated whether isoflurane ameliorates neurological sequelae after germinal matrix hemorrhage (GMH) through activation of the cytoprotective sphingosine kinase/sphingosine-1-phosphate receptor/Akt pathway. METHODS: GMH was induced in P7 rat pups by intraparenchymal infusion of bacterial collagenase (0.3 U) into the right hemispheric germinal matrix. GMH animals received 2% isoflurane either once 1 hour after surgery or every 12 hours for 3 days. Isoflurane treatment was then combined with sphingosine-1-phosphate receptor-1/2 antagonist VPC23019 or sphingosine kinase 1/2 antagonist N,N-dimethylsphingosine. RESULTS: Brain protein expression of sphingosine kinase-1 and phosphorylated Akt were significantly increased after isoflurane post-treatment, and cleaved caspase-3 was decreased at 24 hours after surgery, which was reversed by the antagonists. Isoflurane significantly reduced posthemorrhagic ventricular dilation and improved motor, but not cognitive, functions in GMH animals 3 weeks after surgery; no improvements were observed after VPC23019 administration. CONCLUSIONS: Isoflurane post-treatment improved the neurological sequelae after GMH possibly by activation of the sphingosine kinase/Akt pathway.


Asunto(s)
Encéfalo/efectos de los fármacos , Hemorragias Intracraneales/tratamiento farmacológico , Isoflurano/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Hemorragias Intracraneales/inducido químicamente , Hemorragias Intracraneales/metabolismo , Isoflurano/farmacología , Fármacos Neuroprotectores/farmacología , Fosforilación/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Receptores de Lisoesfingolípidos/metabolismo , Recuperación de la Función/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacología , Esfingosina/uso terapéutico
19.
Neurobiol Dis ; 51: 133-43, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23146993

RESUMEN

Treatments that could extend the therapeutic window of opportunity for stroke patients are urgently needed. Early administration of hyperbaric oxygen therapy (HBOT) has been proven neuroprotective in the middle cerebral artery occlusion (MCAo) in rodents. Our aim was to determine: 1) whether delayed HBOT after permanent MCAo (pMCAo) can still convey neuroprotection and restorative cell proliferation, and 2) whether these beneficial effects rely on HBO-induced activation of protein phosphatase-1γ (PP1-γ) leading to a decreased phosphorylation and ubiquitination of CREB and hence its stabilization. The experiments were performed in one hundred thirty-two male Sprague-Dawley rats with the body weight ranging from 240 to 270 g. Permanent MCAo was induced with the intraluminal filament occluding the right middle cerebral artery (MCA). In the first experiment, HBOT (2.5 ATA, 1h daily for 10 days) was started 48 h after pMCAo. Neurobehavioral deficits and infarct size as well as cyclic AMP response element-binding protein (CREB) expression and BrdU-DAB staining in the hippocampus and the peri-infarct region were evaluated on day 14 and day 28 post-MCAo. In the second experiment, HBOT (2.5 ATA, 1h) was started 3h after pMCAo. The effects of CREB siRNA or PP1-γ siRNA on HBO-induced infarct size alterations and target protein expression were studied. HBOT started with 48 h delay reduced infarct size, ameliorated neurobehavioral deficits and increased protein expression of CREB, resulting in increased cell proliferations in the hippocampus and peri-infarct region, on day 14 and day 28 post-MCAo. In the acute experiment pMCAo resulted in cerebral infarction and functional deterioration and reduced brain expression of PP1-γ, which led to increased phosphorylation and ubiquitination of CREB 24h after MCAo. However HBOT administered 3h after ischemia reversed these molecular events and resulted in CREB stabilization, infarct size reduction and neurobehavioral improvement. Gene silencing with CREB siRNA or PP1-γ siRNA reduced acute beneficial effects of HBO. In conclusion, delayed daily HBOT presented as potent neuroprotectant in pMCAo rats, increased CREB expression and signaling activity, and bolstered regenerative type cell proliferation in the injured brain. As shown in the acute experiment these effects of HBO were likely to be mediated by reducing ubiquitin-dependent CREB degradation owing to HBO-induced activation of PP1γ.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Oxigenoterapia Hiperbárica/métodos , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/terapia , Animales , Western Blotting , Proliferación Celular , Modelos Animales de Enfermedad , Inmunohistoquímica , Inmunoprecipitación , Masculino , Ratas , Ratas Sprague-Dawley
20.
Neurobiol Dis ; 58: 296-307, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23816751

RESUMEN

Neuroinflammation contributes to the pathogenesis of early brain injury (EBI) after subarachnoid hemorrhage (SAH). Cytotoxic events following SAH, such as extracellular accumulation of adenosine triphosphate (ATP), may activate the P2X purinoceptor 7 (P2X7R)/cryopyrin inflammasome axis, thus inducing the proinflammatory cytokine IL-1ß/IL-18 secretion. We therefore hypothesized that inhibition of P2X7R/cryopyrin inflammasome axis would ameliorate neuroinflammation after SAH. In the present study, SAH was induced by the endovascular perforation in rats. Small interfering RNAs (siRNAs) of P2X7R or cryopyrin were administered intracerebroventricularly 24h before SAH. Brilliant blue G (BBG), a non-competitive antagonist of P2X7R, was administered intraperitoneally 30min following SAH. Post-assessments including SAH severity score, neurobehavioral test, brain water content, Western blot and immunofluorescence, were performed. Administration of P2X7R and cryopyrin siRNA as well as pharmacologic blockade of P2X7R by BBG ameliorated neurological deficits and brain edema at 24h following SAH. Inhibition of P2X7R/cryopyrin inflammasome axis suppressed caspase-1 activation, which subsequently decreased maturation of IL-1ß/IL-18. To investigate the link between P2X7R and cryopyrin inflammasome in vivo, Benzoylbenzoyl-ATP (BzATP), a P2X7R agonist, was given to lipopolysaccharide (LPS) primed naive rats with scramble or cryopyrin siRNAs. In LPS-primed naive rats, BzATP induced caspase-1 activation and mature IL-1ß release were neutralized by cryopyrin siRNA. Thus, the P2X7R/cryopyrin inflammasome axis may contribute to neuroinflammation via activation of caspase-1 and thereafter mature IL-1ß/IL-18 production following SAH. Therapeutic interventions targeting P2X7R/cryopyrin pathway may be a novel approach to ameliorate EBI following SAH.


Asunto(s)
Encefalitis/tratamiento farmacológico , Encefalitis/etiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Transducción de Señal/fisiología , Hemorragia Subaracnoidea/complicaciones , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Animales , Edema Encefálico/etiología , Proteínas Portadoras , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inyecciones Intraventriculares , Lipopolisacáridos , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR , Inhibidores de Agregación Plaquetaria/farmacología , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/genética , Receptores Purinérgicos P2X7/genética , Colorantes de Rosanilina/farmacología , Transducción de Señal/efectos de los fármacos , Hemorragia Subaracnoidea/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA