Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Chin J Physiol ; 60(5): 259-266, 2017 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-28950689

RESUMEN

Basal ganglia hemorrhage accounts for approximately 50% of all hemorrhagic strokes. A good rat model that produces severe intrastriatal hemorrhage (ISH) mimicking human severe ISH is lacking. The present study compared the intra-striatal injection of 0.2 U with that of 0.6 U of collagenase in inducing severe ISH in rats. Three-Tesla (3T) magnetic resonance imaging (MRI) was used to evaluate brain injuries in terms of hematoma size (volume), midline shift (MLS), and brain edema. This evaluation was further substantiated by determination of behavior and neurologic functions and mortality over 56 h. The 0.2 U collagenase caused hematoma volume increases for 10.3 to 30.1 mm³, while the 0.6 U caused 36.4 to 114.8 mm³, at post-ISH 1 h to 56 h. The 0.6 U collagenase significantly increased MLS to 1.5-3.0 times greater than the 0.2 U did at all post-intracerebral hemorrhage (ICH) time points. The MLS increased dependently with hematoma expansion with high correlation coefficients, yet no mortality occurred. These two dosages, nevertheless, caused the same pattern and severity in relative apparent diffusion coefficient (rADC) changes for three regions of interest (ROIs). Both ISH models induced consistent behavior deficits. The larger dosage produced severe brain injuries as well as neurological deficits, more closely mimicking severe human ISH. Hematoma volume and MLS can be the most useful parameters for evaluating the ISH severity in the present experimental model. The larger dosage, therefore, would be useful for investigating the pathophysiology of the severer ISH in the striatum. This may be applied for evaluating potential therapeutic strategies and outcomes in the future.


Asunto(s)
Hemorragia Cerebral/etiología , Colagenasas/farmacología , Cuerpo Estriado , Modelos Animales de Enfermedad , Envejecimiento , Animales , Humanos , Imagen por Resonancia Magnética , Masculino , Ratas , Ratas Sprague-Dawley
2.
Toxicol Appl Pharmacol ; 305: 75-82, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27260673

RESUMEN

The abuse of ketamine and amphetamine analogs is associated with incidence of hypertension and strokes involving activation of sympathetic activities. Large cerebral arteries at the base of the brain from several species receive dense sympathetic innervation which upon activation causes parasympathetic-nitrergic vasodilation with increased regional blood flow via axo-axonal interaction mechanism, serving as a protective mechanism to meet O2 demand in an acutely stressful situation. The present study was designed to examine effects of ketamine and amphetamine analogs on axo-axonal interaction-mediated neurogenic nitrergic vasodilation in porcine basilar arteries using techniques of blood-vessel myography, patch clamp and two-electrode voltage clamp, and calcium imaging. In U46619-contracted basilar arterial rings, nicotine (100µM) and electrical depolarization of nitrergic nerves by transmural nerve stimulation (TNS, 8Hz) elicited neurogenic nitrergic vasodilations. Ketamine and amphetamine analogs concentration-dependently inhibited nicotine-induced parasympathetic-nitrergic vasodilation without affecting that induced by TNS, nitroprusside or isoproterenol. Ketamine and amphetamine analogs also concentration-dependently blocked nicotine-induced inward currents in Xenopus oocytes expressing α3ß2-nicotinic acetylcholine receptors (nAChRs), and nicotine-induced inward currents as well as calcium influxes in rat superior cervical ganglion neurons. The potency in inhibiting both inward-currents and calcium influxes is ketamine>methamphetamine>hydroxyamphetamine. These results indicate that ketamine and amphetamine analogs, by blocking nAChRs located on cerebral perivascular sympathetic nerves, reduce nicotine-induced, axo-axonal interaction mechanism-mediated neurogenic dilation of the basilar arteries. Chronic abuse of these drugs, therefore, may interfere with normal sympathetic-parasympathetic interaction mechanism resulting in diminished neurogenic vasodilation and, possibly, normal blood flow in the brainstem.


Asunto(s)
Anfetaminas/farmacología , Arteria Basilar/efectos de los fármacos , Ketamina/farmacología , Receptores Nicotínicos/fisiología , Vasoconstrictores/farmacología , Animales , Arteria Basilar/metabolismo , Arteria Basilar/fisiología , Calcio/metabolismo , Círculo Arterial Cerebral/efectos de los fármacos , Círculo Arterial Cerebral/fisiología , Técnicas In Vitro , Ketamina/análogos & derivados , Nicotina/farmacología , Oocitos , Ratas Sprague-Dawley , Receptores Nicotínicos/genética , Ganglio Cervical Superior/efectos de los fármacos , Ganglio Cervical Superior/metabolismo , Ganglio Cervical Superior/fisiología , Porcinos , Vasodilatación/efectos de los fármacos , Xenopus
3.
J Stroke Cerebrovasc Dis ; 25(1): 15-25, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26387045

RESUMEN

OBJECTIVE: Alcohol intoxication is associated with worse intracerebral hemorrhage (ICH) outcome, indicating the important role of alcohol in ICH pathogenesis. We intended to investigate the effects of ethanol pretreatment on the severity of ICH-induced brain injury in rats. METHODS: At 1 hour after intraperitoneal injection of ethanol (3 g/kg), 0.2 U bacterial collagenase was infused into the striatum of male Sprague-Dawley rats to induce ICH. Accumulative mortality rate, body weight changes, and motorsensory and neurological abnormalities were evaluated. The hemorrhagic volume, hematoma expansion, and water content were measured by Drabkin's method, morphometric assay, and dry/wet method, respectively. Blood-brain barrier disruption was assessed using Evans blue assay. Oxidative stress was evaluated by the enzymatic activity of glutathione peroxidase, oxidation of hydroethidine, and the production of malondialdehyde. Cerebral blood flow perfusion volume and hypo-/hyperperfusion neuroimaging were examined by magnetic resonance imaging. RESULTS: Ethanol pretreatment aggravates the hematoma hemolysis, hemorrhagic volume, hematoma expansion, brain edema, blood-brain barrier disruption, microglial activation, elevated oxidative stress, and neuroinflammation in the hemorrhagic striatum. The summation effect of these consequences is the major cause of marked neurological impairment and higher mortality rate (64%) in ethanol-pretreated rats with ICH. CONCLUSION: This is a novel model to evaluate the effects of high-dose alcohol administration on experimental ICH rats. IMPLICATIONS: The present study may provide clues for making novel strategies in the management of patients with ICH who overconsume alcoholic drinks before the attack.


Asunto(s)
Intoxicación Alcohólica/complicaciones , Daño Encefálico Crónico/etiología , Hemorragia Cerebral/complicaciones , Cuerpo Estriado/patología , Intoxicación Alcohólica/fisiopatología , Animales , Barrera Hematoencefálica , Daño Encefálico Crónico/patología , Edema Encefálico/etiología , Hemorragia Cerebral/fisiopatología , Circulación Cerebrovascular , Modelos Animales de Enfermedad , Etanol/administración & dosificación , Etanol/toxicidad , Hematoma/etiología , Inflamación , Inyecciones Intraperitoneales , Imagen por Resonancia Magnética , Masculino , Microglía/patología , Estrés Oxidativo , Imagen de Perfusión , Premedicación , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Prueba de Desempeño de Rotación con Aceleración Constante
4.
Cells ; 10(10)2021 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-34685754

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a progressive nervous system disease that causes motor neuron (MN) degeneration and results in patient death within a few years. To recapitulate the cytopathies of ALS patients' MNs, SOD1G85R mutant and corrected SOD1G85G isogenic-induced pluripotent stem cell (iPSC) lines were established. Two SOD1 mutant ALS (SOD1G85R and SOD1D90A), two SOD1 mutant corrected (SOD1G85G and SOD1D90D), and one sporadic ALS iPSC lines were directed toward MNs. After receiving ~90% purity for MNs, we first demonstrated that SOD1G85R mutant ALS MNs recapitulated ALS-specific nerve fiber aggregates, similar to SOD1D90A ALS MNs in a previous study. Moreover, we found that both SOD1 mutant MNs showed ALS-specific neurite degenerations and neurotransmitter-induced calcium hyperresponsiveness. In a small compound test using these MNs, we demonstrated that gastrodin, a major ingredient of Gastrodia elata, showed therapeutic effects that decreased nerve fiber cytopathies and reverse neurotransmitter-induced hyperresponsiveness. The therapeutic effects of gastrodin applied not only to SOD1 ALS MNs but also to sporadic ALS MNs and SOD1G93A ALS mice. Moreover, we found that coactivation of the GSK3ß and IGF-1 pathways was a mechanism involved in the therapeutic effects of gastrodin. Thus, the coordination of compounds that activate these two mechanisms could reduce nerve fiber cytopathies in SOD1 ALS MNs. Interestingly, the therapeutic role of GSK3ß activation on SOD1 ALS MNs in the present study was in contrast to the role previously reported in research using cell line- or transgenic animal-based models. In conclusion, we identified in vitro ALS-specific nerve fiber and neurofunctional markers in MNs, which will be useful for drug screening, and we used an iPSC-based model to reveal novel therapeutic mechanisms (including GSK3ß and IGF-1 activation) that may serve as potential targets for ALS therapy.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Neuronas Motoras/patología , Mutación/genética , Fibras Nerviosas/patología , Superóxido Dismutasa-1/genética , Animales , Axones/efectos de los fármacos , Axones/patología , Alcoholes Bencílicos/farmacología , Calcio/metabolismo , Diferenciación Celular , Glucósidos/farmacología , Ácido Glutámico/farmacología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Transgénicos , Degeneración Nerviosa , Neuritas/patología , Análisis de Supervivencia
5.
J Neurosci Res ; 86(6): 1277-89, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18092358

RESUMEN

We have characterized lipopolysaccharide (LPS) preconditioning-induced neuroprotective mechanisms against nitric oxide (NO) toxicity. Pretreatment of rat cortical cultures with LPS attenuated neurotoxicity of NO donors, including sodium nitroprusside (SNP) and diethylamine NONOate (NONOate). A transiently increased expression of endothelial nitric oxide synthase (eNOS) accompanied by an increase in NO production was observed during LPS preconditioning. Application of NOS inhibitors including L-N(5)-(1-iminoethyl)-ornithine (L-NIO) and L-nitroarginine methylester (L-NAME) abolished LPS-dependent protection against SNP toxicity. The LPS effect was also blocked by KT5823, an inhibitor of cGMP-dependent protein kinase (PKG). Consistently, application of 8-bromo-cyclic GMP (8-Br-cGMP), a slowly degradable cGMP analogue capable of PKG activation, was neuroprotective. LPS preconditioning resulted in a heightened neuronal expression of Bcl-2 protein that was abolished by L-NAME and KT5823, the respective inhibitors of NOS and PKG. Together, our results reveal the signaling cascade of "LPS --> eNOS --> NO --> cGMP/PKG --> Bcl-2" that might have contributed to the LPS protective effects in cortical neurons.


Asunto(s)
Precondicionamiento Isquémico/métodos , Lipopolisacáridos/farmacología , Degeneración Nerviosa/prevención & control , Neuronas/efectos de los fármacos , Óxido Nítrico/toxicidad , Transducción de Señal/fisiología , Animales , Western Blotting , Encéfalo/efectos de los fármacos , Células Cultivadas , GMP Cíclico/metabolismo , Inmunohistoquímica , Microscopía Confocal , Microscopía Fluorescente , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley
6.
J Neurosci ; 25(39): 8967-77, 2005 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-16192387

RESUMEN

Prion diseases are induced by pathologically misfolded prion protein (PrPSc), which recruit normal sialoglycoprotein PrPC by a template-directed process. In this study, we investigated the expression of PrPC in a rat model of cerebral ischemia to more fully understand its physiological role. Immunohistochemical analysis demonstrated that PrPC-immunoreactive cells increased significantly in the penumbra of ischemic rat brain compared with the untreated brain. Western blot analysis showed that PrPC protein expression increased in ischemic brain tissue in a time-dependent manner. In addition, PrPC protein expression was seen to colocalize with neuron, glial, and vascular endothelial cells in the penumbric region of the ischemic brain. Overexpression of PrPC by injection of rAd (replication-defective recombinant adenoviral)-PGK (phosphoglycerate kinase)-PrPC-Flag into ischemic rat brain improved neurological behavior and reduced the volume of cerebral infarction, which is supportive of a role for PrPC in the neuroprotective adaptive cellular response to ischemic lesions. Concomitant upregulation of PrPC and activated extracellular signal-regulated kinase (ERK1/2) under hypoxia-reoxygenation in primary cortical cultures was shown to be dependent on ERK1/2 phosphorylation. During hypoxia-reoxygenation, mouse neuroblastoma cell line N18 cells transfected with luciferase rat PrPC promoter reporter constructs, containing the heat shock element (HSE), expressed higher luciferase activities (3- to 10-fold) than those cells transfected with constructs not containing HSE. We propose that HSTF-1 (hypoxia-activated transcription factor), phosphorylated by ERK1/2, may in turn interact with HSE in the promoter of PrPC resulting in gene expression of the prion gene. In summary, we conclude that upregulation of PrPC expression after cerebral ischemia and hypoxia exerts a neuroprotective effect on injured neural tissue. This study suggests that PrPC has physiological relevance to cerebral ischemic injury and could be useful as a therapeutic target for the treatment of cerebral ischemia.


Asunto(s)
Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Marcación de Gen , Fármacos Neuroprotectores/metabolismo , Proteínas PrPC/metabolismo , Adenoviridae/genética , Animales , Apoptosis , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/complicaciones , Isquemia Encefálica/metabolismo , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Infarto Cerebral/patología , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Marcación de Gen/métodos , Vectores Genéticos , Humanos , Hipoxia/metabolismo , Masculino , Sistema Nervioso/fisiopatología , Oxígeno/farmacología , Proteínas PrPC/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Accidente Cerebrovascular/etiología , Distribución Tisular
7.
Circulation ; 110(13): 1847-54, 2004 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-15381647

RESUMEN

BACKGROUND: Stroke is a leading cause of death and disability worldwide; however, no effective treatment currently exists. METHODS AND RESULTS: Rats receiving subcutaneous granulocyte colony-stimulating factor (G-CSF) showed less cerebral infarction, as evaluated by MRI, and improved motor performance after right middle cerebral artery ligation than vehicle-treated control rats. Subcutaneous administration of G-CSF enhanced the availability of circulating hematopoietic stem cells to the brain and their capacity for neurogenesis and angiogenesis in rats with cerebral ischemia. CONCLUSIONS: G-CSF induced increases in bone marrow cell mobilization and targeting to the brain, reducing the volume of cerebral infarction and improving neural plasticity and vascularization.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/efectos de los fármacos , Infarto de la Arteria Cerebral Media/terapia , Animales , Biomarcadores , Médula Ósea/efectos de los fármacos , Química Encefálica , Diferenciación Celular , División Celular , Linaje de la Célula , Replicación del ADN , Evaluación Preclínica de Medicamentos , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Movimientos de la Cabeza , Células Madre Hematopoyéticas/citología , Infarto de la Arteria Cerebral Media/fisiopatología , Inyecciones Subcutáneas , Locomoción , Masculino , Neovascularización Fisiológica/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores CXCR4/biosíntesis , Receptores CXCR4/genética , Recuperación de la Función , Regulación hacia Arriba
8.
Neuropeptides ; 52: 89-96, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26055808

RESUMEN

Urocortin exerts neuroprotective effects in intracerebral hemorrhage (ICH) of rats. For pre-clinical trial, we intended to study the neuroprotective efficacy of human UCN (hUCN)-1, -2 and -3 in treating ICH rats. ICH was induced by infusing bacterial collagenase VII (0.23 U in sterile saline) to the striatum. The hUCN-1, -2, and -3 were administrated (2.5µg/kg, i.p.) at 1h after ICH insult, respectively. Neurological deficits were evaluated by modified Neurological Severity Scores. Brain edema and hematoma expansion was evaluated by coronal T2-WI and DWI magnetic resonance imaging on 1, 3, 6, 24, and 56h after ICH insult. Blood-brain barrier permeability was evaluated by Evans blue assay on day 3 after ICH. Brain lesion volume was evaluated by morphormetric measurement on day 7 after ICH. Our results demonstrated that the hUCN-1 significantly reduced hematoma, blood-brain barrier disruption and neurological deficits on day 3, and brain lesion volume on day 7 after ICH insult. The prediction of secondary structure of the hUCNs clarifies that the percentage of alpha-helix, random coil and extended strand between rat-UCN (rUCN)-1 and hUCN-1 are the same. The structure similarity between human- and rat-UCN-1 may be one of the reasons that both can exert similar therapeutic potential in ICH rats.


Asunto(s)
Hemorragia Cerebral/prevención & control , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/patología , Hormona Liberadora de Corticotropina/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Urocortinas/administración & dosificación , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Hemorragia Cerebral/inducido químicamente , Hormona Liberadora de Corticotropina/uso terapéutico , Humanos , Masculino , Colagenasa Microbiana , Fármacos Neuroprotectores/uso terapéutico , Ratas , Ratas Sprague-Dawley , Urocortinas/uso terapéutico
9.
Neurobiol Aging ; 25(7): 935-44, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15212847

RESUMEN

Neuregulin-1 (NRG-1) is expressed throughout the immature and adult central nervous system and it has been demonstrated to influence the migration of a variety of cell types in developing brain. Elevated levels of NRG-1 transcript are found in the adult brain after injury, leading to the suggestion that NRG-1 is involved in the physiological response to neuronal injury. Here, we report our findings that rats pre-treated with NRG-1 protein, undergoing cerebral ischemia 30 min later, had increased motor performance and less cerebral infarction than untreated rats. In the cortex of NRG-1 treated rats, ischemia induced a decrease in caspase-3 immunoreactivity and a reduction in the density of cells positive for terminal deoxynucleotidyl transferase-mediated dUTP-biotin in situ nick end-labeling. Improvement in behavioral assays was also found in animals treated with NRG-1. Pre-treatment with NRG-1 did not alter cerebral blood flow or other physiological parameters. NRG-1 reduced ischemia/reperfusion injury, indicating that it may act as an endogenous neuroprotective factor against stroke. Therefore, NRG-1 may represent a target for the development of new treatments for stroke.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Neurregulina-1/administración & dosificación , Neurregulina-1/metabolismo , Daño por Reperfusión/prevención & control , Animales , Apoptosis/fisiología , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Isquemia Encefálica/complicaciones , Isquemia Encefálica/tratamiento farmacológico , Caspasa 1/metabolismo , Supervivencia Celular , Infarto Cerebral/etiología , Infarto Cerebral/patología , Fragmentación del ADN/fisiología , ADN Nucleotidilexotransferasa/metabolismo , Inyecciones Intraventriculares , Masculino , Actividad Motora/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Ratas , Ratas Sprague-Dawley
10.
Neuropharmacology ; 62(4): 1651-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22155207

RESUMEN

Fluoxetine-induced relaxation of the smooth muscle of small cerebral arteries is thought beneficial in treating mental disorders. The present study was designed to examine effect of fluoxetine on neurogenic nitrergic vasodilation in large cerebral arteries, using in vitro tissue myography, techniques of electrophysiology, calcium imaging and biochemistry. In isolated porcine endothelium-denuded basilar arteries in the presence of U-46619-induced active muscle tone, fluoxetine in low concentration (<0.03 µM) significantly enhanced nicotine- and choline-induced relaxations. The vasorelaxation, however, was blocked by higher concentration of fluoxetine (>0.3 µM) with maximum inhibition at 3 µM. At this concentration, fluoxetine did not affect the basal tone or vasorelaxations induced by transmural nerve stimulation, sodium nitroprusside, or isoproterenol. Furthermore, fluoxetine exclusively blocked nicotine-induced inward currents and calcium influx in cultured neurons of rat superior cervical ganglion and Xenopus oocytes expressing human α7-, α3ß2-, or α4ß2-nicotinic acetylcholine receptors (nAChRs). In addition, fluoxetine at 0.03 µM and 3 µM significantly enhanced and blocked, respectively, nicotine-induced norepinephrine (NE) release from cerebral perivascular sympathetic nerves. These results indicate that fluoxetine via axo-axonal interaction mechanism exhibits bimodal effects on nAChR-mediated neurogenic nitrergic dilation of basilar arteries. Fluoxetine in high concentrations decreases while in low concentrations it increases neurogenic vasodilation. These results from in vitro experimentation suggest that optimal concentrations of fluoxetine which increase or minimally affect neurogenic vasodilation indicative of regional cerebral blood flow may be important consideration in treating mental disorders.


Asunto(s)
Arteria Basilar/efectos de los fármacos , Arterias Cerebrales/efectos de los fármacos , Fluoxetina/farmacología , Neuronas/efectos de los fármacos , Neuronas Nitrérgicas/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Vasodilatación/efectos de los fármacos , Animales , Arteria Basilar/inervación , Calcio/metabolismo , Células Cultivadas , Arterias Cerebrales/inervación , Neuronas/fisiología , Neuronas Nitrérgicas/fisiología , Nitroprusiato/farmacología , Ratas , Ganglio Cervical Superior/efectos de los fármacos , Ganglio Cervical Superior/fisiología , Porcinos , Vasodilatación/fisiología , Vasodilatadores/farmacología , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA