Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Dev Neurosci ; : 1-16, 2019 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-30861520

RESUMEN

The incidence of stroke in children is 2.4 per 100,000 person-years and results in long-term motor and cognitive disability. In ischemic stroke, white matter (WM) is frequently injured, but is relatively understudied compared to grey matter injury. Previous research suggests that the cellular response to WM ischemic injury is different at different ages. Little is known about whether WM repair mechanisms differ in children and adults. We utilized a model of focal ischemic WM injury to determine the oligodendrocyte (OL) response to focal WM ischemic injury in juvenile and adult mice. Methods: Juvenile (21-25 days of age) versus adult (2-3 months of age) mice underwent stereotaxic injection of the potent vasoconstrictor N5-(1-iminoethyhl)-L-ornithine (L-NIO) into the lateral corpus callosum (CC). Animals were sacrificed on postoperative day 3 (acute) or 21 (chronic). Cell birth-dating was performed acutely after WM stroke with 5-ethynyl-2-deoxyuridine (EdU) injected intraperitoneally. Immunohistochemistry was performed, as well as stereology, to measure injury volume. The acute oligodendrocyte progenitor cell (OPC) proliferation and the chronic OL cell fate were determined with immunohistochemistry. Compound action potentials were measured in the CC at acute and chronic time points. Results: Acutely WM injury volume was smaller in juveniles. There was significantly greater OPC proliferation in juvenile animals (acute) compared to adults, but newly born OLs did not survive and mature into myelinating cells at chronic time points. In addition, juveniles did not have improved histological or functional recovery when compared to adults. Protecting newly born OPCs is a potential therapeutic target in children with ischemic stroke.

2.
Stroke ; 48(8): 2274-2277, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28679853

RESUMEN

BACKGROUND AND PURPOSE: Childhood arterial ischemic stroke is frequently associated with an intracranial arteriopathy that often progresses in the first 3 to 6 months post stroke. We hypothesized that children with enhancing arteriopathies on vessel wall imaging (VWI) would have a higher risk of arteriopathy progression than those without enhancement. METHODS: Our institutional radiographic database was searched for cases of childhood stroke with VWI. Inclusion criteria consisted of age ranging from 1 month through 20 years, diagnosis of arterial ischemic stroke, available VWI, and follow-up magnetic resonance angiogram. Imaging was reviewed to systematically describe VWI findings, categorize arteriopathies, steroid therapy, and identify progressive arteriopathies using CACADE definitions. RESULTS: Sixteen cases of childhood stroke at Children's Hospital Colorado between January 1, 2010 and July 1, 2016 were reviewed. Strong vessel wall enhancement at presentation was associated with progressive arteriopathy in 83% of cases (10/12), when compared with 0% (0/4) without strong enhancement (P=0.008). CONCLUSIONS: Our case series demonstrates the potential benefit of VWI in children with stroke because it may identify patients who will have progressive arterial disease.


Asunto(s)
Arterias Cerebrales/diagnóstico por imagen , Progresión de la Enfermedad , Enfermedades Arteriales Intracraneales/diagnóstico por imagen , Angiografía por Resonancia Magnética/tendencias , Accidente Cerebrovascular/diagnóstico por imagen , Adolescente , Niño , Femenino , Estudios de Seguimiento , Humanos , Enfermedades Arteriales Intracraneales/complicaciones , Angiografía por Resonancia Magnética/métodos , Masculino , Valor Predictivo de las Pruebas , Estudios Retrospectivos , Accidente Cerebrovascular/complicaciones
3.
Glia ; 64(11): 1972-86, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27463063

RESUMEN

White matter injury following ischemic stroke is a major cause of functional disability. Injury to both myelinated axons and oligodendrocytes, the myelin producing cells in the central nervous system, occurs in experimental models of ischemic stroke. Age-related changes in white matter vulnerability to ischemia have been extensively studied and suggest that both the perinatal and the aged periods are times of increased white matter vulnerability. However, sensitivity of white matter following stroke in the juvenile brain has not been evaluated. Interestingly, the late pediatric period is an important developmental stage, as it is the time of maximal myelination. The current study demonstrates that neurons in late pediatric/juvenile striatum are vulnerable to ischemic damage, with neuronal injury being comparable in juvenile and adult mice following ischemia. By contrast, actively myelinating striatal oligodendrocytes in the juvenile brain are resistant to ischemia, whereas adult oligodendrocytes are quite sensitive. As a result, myelin sheaths are remarkably intact and axons survive well in the injured striatum of juvenile mice. In addition to relative resistance of juvenile white matter, other glial responses were very different in juvenile and adult mice following cerebral ischemia, including differences in astrogliosis, fibrosis, NG2-cell reactivity, and vascular integrity. Together, these responses lead to long-term preservation of brain parenchyma in juvenile mice, compared to severe tissue loss and scarring in adult mice. Overall, the current study suggests that equivalent ischemic insults may result in less functional deficit in children compared to adults and an environment more conducive to long-term recovery. GLIA 2016;64:1972-1986.


Asunto(s)
Cuerpo Estriado/patología , Infarto de la Arteria Cerebral Media/complicaciones , Leucoencefalopatías/etiología , Factores de Edad , Animales , Axones/patología , Vasos Sanguíneos/patología , Vasos Sanguíneos/ultraestructura , Infarto Encefálico/etiología , Modelos Animales de Enfermedad , Lateralidad Funcional , Transportador de Glucosa de Tipo 1/metabolismo , Glutatión Transferasa/metabolismo , Hemo-Oxigenasa 1/metabolismo , Leucoencefalopatías/patología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Proteínas de la Mielina/metabolismo , Fibras Nerviosas Mielínicas/patología , Fibras Nerviosas Mielínicas/ultraestructura , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/metabolismo , Oligodendroglía/ultraestructura , Factores de Tiempo
4.
J Stroke Cerebrovasc Dis ; 25(10): 2399-404, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27363622

RESUMEN

BACKGROUND AND OBJECTIVE: Infectious complications after ischemic stroke are frequent and lead to neurological deterioration, poor functional outcomes, and higher mortality. Local and systemic inflammatory responses to brain ischemia differ between males and females, but little is known about differences in poststroke susceptibility to infection by sex. The purpose of this study was to compare sex-related differences in the risk of hospital-acquired sepsis and pneumonia after acute ischemic stroke (AIS). MATERIALS AND METHODS: This is a retrospective, secondary analysis of the 2010-2011 California State Inpatient Database. Previously validated International Classification of Disease, Ninth Revision (ICD-9) codes were used to identify adult hospitalizations for AIS. The primary outcome was hospital-acquired sepsis or pneumonia, also identified using ICD-9 codes. Associations between sex and hospital-acquired sepsis or pneumonia were adjusted for baseline characteristics and comorbidities using multivariable logistic regression. RESULTS: There were 91,643 hospitalizations for AIS included in this analysis, of which 1027 had hospital-acquired sepsis and 1225 had hospital-acquired pneumonia. The in-hospital mortality without infection was 4.6%; the presence of hospital-acquired infections was associated with higher mortality for sepsis (32.7%) and pneumonia (21.9%). Female (versus male) sex was associated with lower adjusted odds of hospital-acquired sepsis (odds ratio [OR] .74, 95% confidence interval [CI] .65-.84) and pneumonia (OR .69, 95% CI .62-.78). This difference was similar across age strata. Among hospitalizations with either hospital-acquired sepsis or pneumonia, sex did not influence mortality. CONCLUSIONS: Female sex was associated with a lower risk of hospital-acquired sepsis and pneumonia after AIS. Further investigation is needed to determine the mechanisms underlying this clinical observation.


Asunto(s)
Isquemia Encefálica/epidemiología , Infección Hospitalaria/epidemiología , Neumonía/epidemiología , Sepsis/epidemiología , Accidente Cerebrovascular/epidemiología , Anciano , Anciano de 80 o más Años , Isquemia Encefálica/diagnóstico , Isquemia Encefálica/mortalidad , California/epidemiología , Comorbilidad , Infección Hospitalaria/diagnóstico , Infección Hospitalaria/mortalidad , Bases de Datos Factuales , Femenino , Mortalidad Hospitalaria , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Oportunidad Relativa , Neumonía/diagnóstico , Neumonía/mortalidad , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Sepsis/diagnóstico , Sepsis/mortalidad , Factores Sexuales , Accidente Cerebrovascular/diagnóstico , Accidente Cerebrovascular/mortalidad , Factores de Tiempo
5.
Stroke ; 44(3): 759-63, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23349190

RESUMEN

BACKGROUND AND PURPOSE: Pediatric stroke, birth to 18 years, is a significant cause of long-term disability in the United States; however, there is currently little experimental data on the pathophysiology of childhood stroke owing to lack of animal models. We developed a novel mouse model of experimental childhood-onset arterial ischemic stroke to characterize the sex-specific response of the adolescent brain to cerebral ischemia and assess the neuroprotective effect of estrogen at this developmental stage. METHODS: Postnatal day 20 to 25 mice were subjected to 90 minutes experimental stroke via the intraluminal filament middle cerebral artery occlusion model and ischemic damage assessed 22 hours after reperfusion. Real-time quantitative real-time polymerase chain reaction was performed 22 hours after middle cerebral artery occlusion to determine the effects of ischemia and estrogen treatment on the proapoptotic gene Bax. RESULTS: Ischemic injury did not differ between male and female juvenile (postnatal day 20-25) mice after middle cerebral artery occlusion. However, estrogen reduced ischemic injury in female mice, whereas having no effect in juvenile males. No differences in estrogen receptor expression were observed on postnatal day between 20 males and females. In contrast, estrogen minimized the ischemia-induced increase in the proapoptotic gene Bax in female mice, whereas having no effect on Bax induction in the male brain. CONCLUSIONS: Focal ischemia has fundamentally different effects in the juvenile brain compared with the adult, as evidenced by the lack of sex difference in ischemic injury in the murine postnatal day 20 to 25 middle cerebral artery occlusion model and the sexually dimorphic response to estrogen neuroprotection.


Asunto(s)
Envejecimiento/fisiología , Estrógenos/fisiología , Modelos Animales , Caracteres Sexuales , Transducción de Señal/fisiología , Accidente Cerebrovascular/fisiopatología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatología , Estrógenos/farmacología , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Infarto de la Arteria Cerebral Media/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/prevención & control , Proteína X Asociada a bcl-2/metabolismo
6.
Nat Med ; 9(8): 1085-90, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12872167

RESUMEN

In the past decade, it has become possible to use the nuclear (proton, 1H) signal of the hydrogen atoms in water for noninvasive assessment of functional and physiological parameters with magnetic resonance imaging (MRI). Here we show that it is possible to produce pH-sensitive MRI contrast by exploiting the exchange between the hydrogen atoms of water and the amide hydrogen atoms of endogenous mobile cellular proteins and peptides. Although amide proton concentrations are in the millimolar range, we achieved a detection sensitivity of several percent on the water signal (molar concentration). The pH dependence of the signal was calibrated in situ, using phosphorus spectroscopy to determine pH, and proton exchange spectroscopy to measure the amide proton transfer rate. To show the potential of amide proton transfer (APT) contrast for detecting acute stroke, pH effects were noninvasively imaged in ischemic rat brain. This observation opens the possibility of using intrinsic pH contrast, as well as protein- and/or peptide-content contrast, as diagnostic tools in clinical imaging.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Péptidos/química , Proteínas/química , Protones , Animales , Encéfalo/anatomía & histología , Concentración de Iones de Hidrógeno , Isquemia/metabolismo , Ratas , Ratas Sprague-Dawley , Agua/química
7.
Dev Cell ; 4(4): 575-85, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12689595

RESUMEN

BAK is a pro-apoptotic BCL-2 family protein that localizes to mitochondria. Here we evaluate the function of BAK in several mouse models of neuronal injury including neuronotropic Sindbis virus infection, Parkinson's disease, ischemia/stroke, and seizure. BAK promotes or inhibits neuronal death depending on the specific death stimulus, neuron subtype, and stage of postnatal development. BAK protects neurons from excitotoxicity and virus infection in the hippocampus. As mice mature, BAK is converted from anti- to pro-death function in virus-infected spinal cord neurons. In addition to regulating cell death, BAK also protects mice from kainate-induced seizures, suggesting a possible role in regulating synaptic activity. BAK can alter neurotransmitter release in a direction consistent with its protective effects on neurons and mice. These findings suggest that BAK inhibits cell death by modifying neuronal excitability.


Asunto(s)
Apoptosis/genética , Enfermedades del Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Transmisión Sináptica/genética , Factores de Edad , Animales , Animales Recién Nacidos , Sistema Nervioso Central/fisiopatología , Sistema Nervioso Central/virología , Enfermedades del Sistema Nervioso Central/genética , Enfermedades Virales del Sistema Nervioso Central/genética , Enfermedades Virales del Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Epilepsia/genética , Epilepsia/metabolismo , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Vectores Genéticos/genética , Hipocampo/metabolismo , Hipocampo/fisiopatología , Hipocampo/virología , Ácido Kaínico , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Neuronas/patología , Neuronas/virología , Neurotoxinas/genética , Neurotoxinas/metabolismo , Estructura Terciaria de Proteína/genética , Virus Sindbis/genética , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Transmisión Sináptica/efectos de los fármacos , Proteína Destructora del Antagonista Homólogo bcl-2
8.
Curr Opin Pharmacol ; 8(1): 104-10, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17962069

RESUMEN

While many pharmacological agents have been shown to protect the brain from cerebral ischemia in animal models, none have translated successfully to human patients. One potential clinical neuroprotective strategy in humans may involve increasing the brain's tolerance to ischemia by preischemic conditioning (preconditioning). There are many methods to induce tolerance via preconditioning such as ischemia itself, pharmacological, hypoxia, endotoxin, and others. Inhalational anesthetic agents have also been shown to result in brain preconditioning. Mechanisms responsible for brain preconditioning are many, complex, and unclear and may involve Akt activation, ATP-sensitive potassium channels, and nitric oxide, amongst many others. Anesthetics, however, may play an important and unique role as preconditioning agents, particularly during the perioperative period.


Asunto(s)
Anestésicos por Inhalación/farmacología , Isquemia Encefálica/prevención & control , Precondicionamiento Isquémico , Factores de Edad , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Canales KATP/fisiología , Masculino , Óxido Nítrico/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores Sexuales
9.
J Cereb Blood Flow Metab ; 39(6): 1005-1014, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-29283314

RESUMEN

The current study focuses on the ability to improve cognitive function after stroke with interventions administered at delayed/chronic time points. In light of recent studies demonstrating delayed GABA antagonists improve motor function, we utilized electrophysiology, biochemistry and neurobehavioral methods to investigate the role of α5 GABAA receptors on hippocampal plasticity and functional recovery following ischemic stroke. Male C57Bl/6 mice were exposed to 45 min transient middle cerebral artery occlusion and analysis of synaptic and functional deficits performed 7 or 30 days after recovery. Our findings indicate that hippocampal long-term potentiation (LTP) is impaired 7 days after stroke and remain impaired for at least 30 days. We demonstrate that ex vivo administration of L655,708 reversed ischemia-induced plasticity deficits and importantly, in vivo administration at delayed time-points reversed stroke-induced memory deficits. Western blot analysis of hippocampal tissue reveals proteins responsible for GABA synthesis are upregulated (GAD65/67 and MAOB), increasing GABA in hippocampal interneurons 30 days after stroke. Thus, our data indicate that both synaptic plasticity and memory impairments observed after stroke are caused by excessive tonic GABA activity, making inhibition of specific GABA activity at delayed timepoints a potential therapeutic approach to improve functional recovery and reverse cognitive impairments after stroke.


Asunto(s)
Isquemia Encefálica/fisiopatología , Cognición , Recuperación de la Función , Accidente Cerebrovascular/fisiopatología , Animales , Hipocampo/fisiopatología , Potenciación a Largo Plazo , Masculino , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal , Receptores de GABA-A/metabolismo , Factores de Tiempo , Ácido gamma-Aminobutírico/metabolismo
10.
J Neurochem ; 107(3): 668-78, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18699862

RESUMEN

Cerebellar Purkinje cells (PC) are particularly vulnerable to ischemic injury and excitotoxicity, although the molecular basis of this sensitivity remains unclear. We tested the hypothesis that ischemia causes rapid down-regulation of GABA(A) receptors in cerebellar PC, thereby increasing susceptibility to excitotoxicity. Oxygen-glucose deprivation (OGD) caused a decline in functional GABA(A) receptors, within the first hour of re-oxygenation. Decreased amplitude of miniature inhibitory post-synaptic potentials confirmed that OGD caused a significant decrease in functional synaptic GABA(A) receptors and quantitative Western blot analysis demonstrated the loss of GABA(A) receptor current was associated with a decline in total receptor protein. Interestingly, the potent neuroprotectant allopregnanolone (ALLO) prevented the decline in GABA(A) receptor current and protein. Consistent with our in vitro data, global ischemia in mice caused a significant decline in total cerebellar GABA(A) receptor protein and PC specific immunoreactivity. Moreover, ALLO provided strong protection of PC and prevented ischemia-induced decline in GABA(A) receptor protein. Our findings indicate that ischemia causes a rapid and sustained loss of GABA(A) receptors in PC, whereas ALLO prevents the decline in GABA(A) receptors and protects against ischemia-induced damage. Thus, interventions which prevent ischemia-induced decline in GABA(A) receptors may represent a novel neuroprotective strategy.


Asunto(s)
Isquemia Encefálica/metabolismo , Fármacos Neuroprotectores/farmacología , Pregnanolona/farmacología , Células de Purkinje/metabolismo , Receptores de GABA-A/metabolismo , Animales , Western Blotting , Inmunohistoquímica , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Ratones , Técnicas de Placa-Clamp , Células de Purkinje/efectos de los fármacos , Células de Purkinje/patología , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
J Cardiovasc Pharmacol ; 52(1): 82-9, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18645412

RESUMEN

Cocaine- and amphetamine-regulated transcript (CART) and its associated peptides have been implicated in a number of physiologic processes including modulation of the hypothalamo-pituitary-adrenal (HPA) axis and cardiovascular regulation. Recently, we reported that in isolated cerebral arterioles, CART peptide (CARTp) acts directly to produce endothelium-dependent constriction via the endothelin signaling pathway. We used the rat closed cranial window model to determine the in vivo effects of CARTp on pial arteriolar diameter. Intravenous administration of 30 microg/kg CARTp produced a significant pressor effect and constriction of pial arterioles. The pressor response to systemic CARTp was blocked by the beta-adrenergic receptor antagonist propranolol (2 mg/kg IV). Direct application of 0.1 nM-1 microM CARTp to pial arterioles produced a dose-dependent and long-lasting constriction to approximately 88% of baseline diameter. The constriction response to topically applied 100 nM CARTp was blocked by both the endothelin A (ETA) receptor antagonist BQ-123 (10 microM) and the inhibitor of endothelin-converting enzyme, phosphoramidon (100 nM). These results demonstrate for the first time that CARTp constricts cerebral vessels in vivo, an action mediated by its effects on the endothelin system, specifically via activation of ETA receptors. This supports the notion that CARTp plays a physiologic role in cerebrovascular regulation, particularly during times of HPA axis activation.


Asunto(s)
Circulación Cerebrovascular/fisiología , Proteínas del Tejido Nervioso/fisiología , Vasoconstricción/efectos de los fármacos , Animales , Arteriolas/efectos de los fármacos , Arteriolas/fisiología , Circulación Cerebrovascular/efectos de los fármacos , Inyecciones Intravenosas , Masculino , Proteínas del Tejido Nervioso/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Endotelina/fisiología
12.
Resuscitation ; 76(1): 89-94, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17728042

RESUMEN

The P450 eicosanoids epoxyeicosatrienoic acids (EETs) are produced by cytochrome P450 arachidonic acid epoxygenases and metabolized through multiple pathways, including soluble epoxide hydrolase (sEH). Pharmacological inhibition and gene deletion of sEH protect against ischemia/reperfusion injury in brain and heart, and against hypertension-related end-organ damage in kidney. We tested the hypothesis that sEH gene deletion improves survival, recovery of renal function and pathologic ischemic renal damage following transient whole-body ischemia induced by cardiac arrest (CA) and resuscitation. Mice with targeted deletion of sEH (sEH knockout, sEHKO) and C57Bl/6 wild-type control mice were subjected to 10-min CA, followed by cardiopulmonary resuscitation (CPR). Survival in wild-type mice was 93% and 80% at 10 min and 24 h after CA/CPR (n=15). Unexpectedly, survival in sEHKO mice was significantly lower than WT. Only 56% of sEHKO mice survived for 10 min (n=15, p=0.014 compared to WT) and no mice survived for 24 h after CA/CPR (p<0.0001 versus WT). We conclude that sEH plays an important role in cardiovascular regulation, and that reduced sEH levels or function reduces survival from cardiac arrest.


Asunto(s)
Reanimación Cardiopulmonar , Epóxido Hidrolasas/genética , Eliminación de Gen , Paro Cardíaco/enzimología , Paro Cardíaco/terapia , Animales , Electrocardiografía , Ratones , Ratones Endogámicos C57BL , Tasa de Supervivencia
13.
Comp Med ; 58(6): 588-96, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19149416

RESUMEN

Gender differences, sex steroid effects, and sex-specific candidate therapeutics in ischemic stroke have been studied in rodents but not in nonhuman primates. In this feasibility study (n = 3 per group), we developed a model of transient focal cerebral ischemia in adult male and female rhesus macaques that consistently includes white matter injury. The animals also were used to determine whether gender-linked differences in histopathologic outcomes could be evaluated in this model in future, larger preclinical trials. Histologic brain pathology was evaluated at 4 d after 90 min of reversible occlusion of the middle cerebral artery (MCA). MCA occlusion was accomplished by using a transorbital approach and temporary placement of an aneurysm clip. Male and female rhesus macaques 7 to 11 y of age were studied. Baseline and intraischemic blood glucose, systolic blood pressure, heart rate, oxygen saturation, end-tidal CO2, and rectal temperatures were not different among groups. The variability in injury volume was comparable to that observed in human focal cerebrovascular ischemia and in other nonhuman primate models using proximal MCA occlusion. In this small sample, the volume of injury was not different between male and female subjects, but observed variability was higher in female caudate nucleus, putamen, and hemisphere. This report is the first to compare cerebral ischemic outcomes in female and male rhesus macaques. The female rhesus macaque ischemic stroke model could be used after rodent studies to provide preclinical data for clinical trials in women.


Asunto(s)
Ataque Isquémico Transitorio/etiología , Animales , Encéfalo/patología , Dióxido de Carbono/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Ataque Isquémico Transitorio/patología , Ataque Isquémico Transitorio/fisiopatología , Macaca mulatta , Masculino , Caracteres Sexuales , Especificidad de la Especie
14.
J Mol Neurosci ; 64(3): 410-420, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29450697

RESUMEN

The role of biological sex in short-term and long-term outcome after traumatic brain injury (TBI) remains controversial. The observation that exogenous female sex steroids (progesterone and estrogen) reduce brain injury coupled with a small number of clinical studies showing smaller injury in women suggest that sex steroids may play a role in outcome from TBI. We used the controlled cortical impact (CCI) model of TBI in mice to test the hypothesis that after CCI, female mice would demonstrate less injury than male mice, related to the protective role of endogenous steroids. Indeed, adult females exhibit histological protection (3.7 ± 0.5 mm3) compared to adult male mice (6.8 ± 0.6 mm3), and females that lacked sex steroids (ovex) showed increased injury compared to intact females. Consistent with histology, sensorimotor deficits measured as reduced contralateral limb use were most pronounced in male mice (31.9 ± 6.9% reduced limb use) compared to a 12.7 ± 3.8% reduction in female mice. Ovex mice exhibited behavioral deficits similar to males (31.5 ± 3.9% reduced limb use). Ovex females demonstrated increased microglial activation relative to intact females in both the peri-injury cortex and the reticular thalamic nucleus. Ovex females also demonstrated increased astrogliosis in comparison to both females and males in the peri-injury cortex. These data indicate that female sex steroids reduce brain sensitivity to TBI and that reduced acute neuroinflammation may contribute to the relative protection observed in females.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Hormonas Esteroides Gonadales/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Femenino , Masculino , Ratones , Microglía/metabolismo , Microglía/patología , Factores Sexuales , Núcleos Talámicos/metabolismo , Núcleos Talámicos/patología
15.
Neuroscience ; 380: 1-13, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29649514

RESUMEN

Replacement of dead neurons following ischemia, either via enhanced endogenous neurogenesis or stem cell therapy, has long been sought. Unfortunately, while various therapies that enhance neurogenesis or stem cell therapies have proven beneficial in animal models, they have all uniformly failed to truly replace dead neurons in the ischemic core to facilitate long-term recovery. Remarkably, we observe robust repopulation of medium-spiny neurons within the ischemic core of juvenile mice following experimental stroke. Despite extensive neuronal cell death in the injured striatum of both juveniles and adults at acute time points after ischemia (24 h and 7 d), mature newborn neurons replaced lost striatal neurons at 30 d post-ischemia. This neuronal repopulation was found only in juveniles, not adults, and importantly, was accompanied by enhanced post-ischemic behavioral recovery at 30 d. Ablation of neurogenesis using irradiation prevented neuronal replacement and functional recovery in MCAo-injured juvenile mice. In contrast, findings in adults were consistent with previous reports, that newborn neurons failed to mature and died, offering little therapeutic potential. These data provide support for neuronal replacement and consequent functional recovery following ischemic stroke and new targets in the development of novel therapies to treat stroke.


Asunto(s)
Células Madre Adultas/citología , Isquemia Encefálica/patología , Regeneración Nerviosa/fisiología , Células-Madre Neurales/citología , Neurogénesis/fisiología , Neuronas/citología , Factores de Edad , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Recuperación de la Función
16.
J Cereb Blood Flow Metab ; 38(12): 2223-2235, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29611441

RESUMEN

Global ischemia in childhood often leads to poor neurologic outcomes, including learning and memory deficits. Using our novel model of childhood cardiac arrest/cardiopulmonary resuscitation (CA/CPR), we investigate the mechanism of ischemia-induced cognitive deficits and recovery. Memory is impaired seven days after juvenile CA/CPR and completely recovers by 30 days. Consistent with this remarkable recovery not observed in adults, hippocampal long-term potentiation (LTP) is impaired 7-14 days after CA/CPR, recovering by 30 days. This recovery is not due to the replacement of dead neurons (neurogenesis), but rather correlates with brain-derived neurotrophic factor (BDNF) expression, implicating BDNF as the molecular mechanism underlying impairment and recovery. Importantly, delayed activation of TrkB receptor signaling reverses CA/CPR-induced LTP deficits and memory impairments. These data provide two new insights (1) endogenous recovery of memory and LTP through development may contribute to improved neurological outcome in children compared to adults and (2) BDNF-enhancing drugs speed recovery from pediatric cardiac arrest during the critical school ages.


Asunto(s)
Isquemia Encefálica/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Recuperación de la Función/fisiología , Animales , Isquemia Encefálica/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal/fisiología , Transducción de Señal/fisiología
17.
J Cereb Blood Flow Metab ; 37(8): 3053-3064, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28168893

RESUMEN

Cardiac arrest and cardiopulmonary resuscitation (CA/CPR) produce brain ischemia that results in cognitive and motor coordination impairments subsequent to injury of vulnerable populations of neurons, including cerebellar Purkinje neurons. To determine the effects of CA/CPR on plasticity in the cerebellum, we used whole cell recordings from Purkinje neurons to examine long-term depression (LTD) at parallel fiber (PF) synapses. Acute slices were prepared from adult male mice subjected to 8 min cardiac arrest at 1, 7, and 30 days after resuscitation. Concurrent stimulation of PF and climbing fibers (CFs) resulted in robust LTD of PF-evoked excitatory postsynaptic currents (EPSCs) in controls. LTD was absent in recordings obtained from mice subjected to CA/CPR, with no change in EPSC amplitude from baseline at any time point tested. AMPA and mGluR-mediated responses at the PF were not altered by CA/CPR. In contrast, CF-evoked NMDA currents were reduced following CA/CPR, which could account for the loss of LTD observed. A loss of GluN1 protein was observed following CA/CPR that was surprisingly not associated with changes in mRNA expression. These data demonstrate sustained impairments in synaptic plasticity in Purkinje neurons that survive the initial injury and which likely contribute to motor coordination impairments observed after CA/CPR.


Asunto(s)
Reanimación Cardiopulmonar , Potenciales Postsinápticos Excitadores/fisiología , Paro Cardíaco/fisiopatología , Depresión Sináptica a Largo Plazo/fisiología , Células de Purkinje , Animales , Modelos Animales de Enfermedad , Paro Cardíaco/metabolismo , Paro Cardíaco/patología , Masculino , Ratones Endogámicos C57BL , Células de Purkinje/metabolismo , Células de Purkinje/fisiología , Receptores de Glutamato/metabolismo
18.
Cell Rep ; 18(5): 1109-1117, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28147268

RESUMEN

The Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a major mediator of physiological glutamate signaling, but its role in pathological glutamate signaling (excitotoxicity) remains less clear, with indications for both neuro-toxic and neuro-protective functions. Here, the role of CaMKII in ischemic injury is assessed utilizing our mouse model of cardiac arrest and cardiopulmonary resuscitation (CA/CPR). CaMKII inhibition (with tatCN21 or tatCN19o) at clinically relevant time points (30 min after resuscitation) greatly reduces neuronal injury. Importantly, CaMKII inhibition also works in combination with mild hypothermia, the current standard of care. The relevant drug target is specifically Ca2+-independent "autonomous" CaMKII activity generated by T286 autophosphorylation, as indicated by substantial reduction in injury in autonomy-incompetent T286A mutant mice. In addition to reducing cell death, tatCN19o also protects the surviving neurons from functional plasticity impairments and prevents behavioral learning deficits, even at extremely low doses (0.01 mg/kg), further highlighting the clinical potential of our findings.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Paro Cardíaco/metabolismo , Paro Cardíaco/fisiopatología , Neuroprotección/fisiología , Animales , Calcio/metabolismo , Muerte Celular/fisiología , Hipocampo/metabolismo , Hipocampo/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/fisiología , Fosforilación/fisiología
20.
Stroke ; 37(6): 1578-81, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16627792

RESUMEN

The National Heart, Lung, and Blood Institute (NHLBI) convened a conference call working group, consisting of experts in stroke and cerebrovascular biology on January 28, 2005. The purpose of this working group was to develop a prioritized set of recommendations for NHLBI to establish a focused and comprehensive set of research activities in cerebrovascular biology and disease. Three thematic areas of research emerged: (1) molecular and cellular neurobiology of cerebral blood vessels, focusing on genomics and proteomics, neurovascular signaling and cerebrovascular embryogenesis, development and plasticity; (2) resource development, involving the development of new methodological approaches for normal and altered function of the neurovascular unit, collaborative research, and training in cerebrovascular pathobiology; and (3) cerebrovascular diseases and translational approaches, addressing vascular mechanisms of disease, the role of risk factors, importance of biomarkers with the ultimate goal of developing new treatments.


Asunto(s)
Investigación Biomédica , Circulación Cerebrovascular , Trastornos Cerebrovasculares/fisiopatología , Animales , Vasos Sanguíneos/embriología , Vasos Sanguíneos/crecimiento & desarrollo , Circulación Cerebrovascular/fisiología , Modelos Animales de Enfermedad , Genómica , Humanos , Proteómica , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA