Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Neurochem Res ; 42(2): 552-562, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27885575

RESUMEN

Neonatal hypoxia-ischemia (HI) is an etiologic component of several neurologic pathologies associated to cognitive impairment. The mechanisms involved in HI-induced tissue damage start immediately after HI and extend for days. Acetylcholine is an important neurotransmitter in the central nervous system and exerts a protector effect on tissue damage by modulating inflammation, and cholinesterase inhibitors have shown neuroprotective properties and their action are often attributed to inhibition of the immune response. The administration of Huperzia quadrifariata alkaloid extract (HqAE), with potent and selective cholinesterase inhibitor properties, will reduce the HI induced behavioral deficits and tissue damage. A total of 84 newborn Wistar rat pups at post natal day 7 (PND7) were subjected to right carotid occlusion followed by 1 h of hypoxia (8% of O2) and i.p. injections of saline, vehicle or HqAE (10 mg/kg). Morris Water Maze and inhibitory avoidance tests were used to assess the cognitive function. Flow cytometry was performed at PND11. Histological analysis was performed at PND45. HqAE treatment was able to prevent the HI induced cognitive deficits in both tests and, at PND45, histological analysis showed that HqAE treatment reduced hippocampus tissue damage. Flow cytometry of the injured hippocampus revealed that the treatment was able to reduce cellular death and the number of infiltrating T cells. Altogether, these results show the therapeutic potential of the Huperzia quadrifariata alkaloid extract to prevent cognitive deficits and histological damage caused by neonatal hypoxia-ischemia, probably by reducing cellular death and T cell mobilization.


Asunto(s)
Alcaloides/uso terapéutico , Inhibidores de la Colinesterasa/uso terapéutico , Huperzia , Hipoxia-Isquemia Encefálica/enzimología , Fármacos Neuroprotectores/uso terapéutico , Extractos Vegetales/uso terapéutico , Alcaloides/aislamiento & purificación , Alcaloides/farmacología , Animales , Animales Recién Nacidos , Inhibidores de la Colinesterasa/aislamiento & purificación , Inhibidores de la Colinesterasa/farmacología , Modelos Animales de Enfermedad , Femenino , Hipoxia-Isquemia Encefálica/prevención & control , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Fármacos Neuroprotectores/aislamiento & purificación , Fármacos Neuroprotectores/farmacología , Componentes Aéreos de las Plantas , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología , Ratas , Ratas Wistar , Resultado del Tratamiento
2.
Am J Physiol Cell Physiol ; 310(11): C903-10, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27009876

RESUMEN

Astrocytes, the most numerous cells in the human brain, play a central role in the metabolic homeostasis following hypoxic injury. Caveolin-1 (Cav-1), a transmembrane scaffolding protein, has been shown to converge prosurvival signaling in the central nerve system. The present study aimed to investigate the role of Cav-1 in the hypoxia-induced astrocyte injury. We also examined how Cav-1 alleviates apoptotic astrocyte death. To this end, primary astrocytes were exposed to oxygen-glucose deprivation (OGD) for 6 h and a subsequent 24-h reoxygenation to mimic hypoxic injury. OGD significantly reduced Cav-1 expression. Downregulation of Cav-1 using Cav-1 small interfering RNA dramatically worsened astrocyte cell damage and impaired cellular glutamate uptake after OGD, whereas overexpression of Cav-1 with Cav-1 scaffolding domain peptide attenuated OGD-induced cell apoptosis. Mechanistically, the expressions of Ras-GTP, phospho-Raf, and phospho-ERK were sequestered in Cav-1 small interfering RNA-treated astrocytes, yet were stimulated after supplementation with caveolin peptide. MEK/ERK inhibitor U0126 remarkably blocked the Cav-1-induced counteraction against apoptosis following hypoxia, indicating Ras/Raf/ERK pathway is required for the Cav-1's prosurvival role. Together, these findings support Cav-1 as a checkpoint for the in hypoxia-induced astrocyte apoptosis and warrant further studies targeting Cav-1 to treat hypoxic-ischemic brain injury.


Asunto(s)
Apoptosis , Astrocitos/enzimología , Encéfalo/enzimología , Caveolina 1/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipoxia-Isquemia Encefálica/enzimología , Quinasas raf/metabolismo , Proteínas ras/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/patología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Caveolina 1/genética , Hipoxia de la Célula , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Glucosa/deficiencia , Ácido Glutámico/metabolismo , Hipoxia-Isquemia Encefálica/genética , Hipoxia-Isquemia Encefálica/patología , Hipoxia-Isquemia Encefálica/prevención & control , Fosforilación , Cultivo Primario de Células , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Ratas Sprague-Dawley , Transducción de Señal , Transfección
3.
CNS Neurol Disord Drug Targets ; 15(1): 64-72, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26553162

RESUMEN

Recent findings have demonstrated a dual effect of the folic acid (FA) supplementation on the nervous system of rats. We found that FA treatment prevented memory impairment and Na(+), K(+)- ATPase inhibition in the striatum and cortex in adult rats that suffered neonatal hypoxia-ischemia (HI). However, spatial memory deficit has been associated with FA supplementation. In the present study we investigated the role of FA supplementation on spatial memory and Na(+), K(+)-ATPase activity in the hippocampus, as well as on morphologic alterations in adolescent rats submitted to neonatal HI. Wistar rats of both sexes at postnatal day (PND) 7 were submitted to Levine-Rice HI procedure. Intraperitoneal doses of FA were administered immediately before HI and repeated daily until the maximum PND 40. Hippocampal volume and striatum area were estimated and Na(+), K(+)-ATPase activity in the hippocampus was measured at PND 31. Also, the performance of the animals in the water maze was assessed and Na(+), K(+)-ATPase activity measured again at PND 52. Interestingly, HI and FA resulted in spatial memory deficits in the Morris water maze and the Na(+), K(+)-ATPase activity was impaired at PND 31 in HI rats which received FA. Additionally, Na(+), K(+)-ATPase activity in adulthood showed a decrease after HI and a recovery in supplemented animals. Hippocampal and striatal atrophy were partially reversed by FA. To conclude, the present results support the hypothesis that FA supplementation during development contributes to memory deficits caused by HI and Na(+), K(+)-ATPase failure in adolescent rats, although, in adulthood, FA has been effective in reversing enzymatic activity in the hippocampus.


Asunto(s)
Ácido Fólico/toxicidad , Hipocampo/enzimología , Hipoxia-Isquemia Encefálica/enzimología , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/enzimología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/patología , Hipoxia-Isquemia Encefálica/patología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/patología , Ratas , Ratas Wistar , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores
4.
PLoS One ; 9(12): e114470, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25474649

RESUMEN

BACKGROUND AND PURPOSE: Glucocorticoids pretreatment confers protection against neonatal hypoxic-ischemic (HI) brain injury. However, the molecular mechanism remains poorly elucidated. We tested the hypothesis that glucocorticoids protect against HI brain injury in neonatal rat by stimulation of lipocalin-type prostaglandin D synthase (L-PGDS)-induced prostaglandin D2 (PGD2)-DP1-pERK mediated signaling pathway. METHODS: Dexamethasone and inhibitors were administered via intracerebroventricular (i.c.v) injections into 10-day-old rat brains. Levels of L-PGD2, D prostanoid (DP1) receptor, pERK1/2 and PGD2 were determined by Western immunoblotting and ELISA, respectively. Brain injury was evaluated 48 hours after conduction of HI in 10-day-old rat pups. RESULTS: Dexamethasone pretreatment significantly upregulated L-PGDS expression and the biosynthesis of PGD2. Dexamethasone also selectively increased isoform pERK-44 level in the neonatal rat brains. Inhibitors of L-PGDS (SeCl4), DP1 (MK-0524) and MAPK (PD98059) abrogated dexamethasone-induced increases in pERK-44 level, respectively. Of importance, these inhibitors also blocked dexamethasone-mediated neuroprotective effects against HI brain injury in neonatal rat brains. CONCLUSION: Interaction of glucocorticoids-GR signaling and L-PGDS-PGD2-DP1-pERK mediated pathway underlies the neuroprotective effects of dexamethasone pretreatment in neonatal HI brain injury.


Asunto(s)
Dexametasona/uso terapéutico , Hipoxia-Isquemia Encefálica/prevención & control , Oxidorreductasas Intramoleculares/fisiología , Lipocalinas/fisiología , Sistema de Señalización de MAP Quinasas , Fármacos Neuroprotectores/uso terapéutico , Animales , Animales Recién Nacidos , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Dexametasona/farmacología , Evaluación Preclínica de Medicamentos , Inducción Enzimática/efectos de los fármacos , Femenino , Hipoxia-Isquemia Encefálica/enzimología , Masculino , Fármacos Neuroprotectores/farmacología , Prostaglandina D2/metabolismo , Ratas Sprague-Dawley , Factor de Transcripción DP1/metabolismo , Regulación hacia Arriba
5.
J Neurotrauma ; 25(6): 695-707, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18457515

RESUMEN

We previously reported that uridine blocked glucose deprivation-induced death of immunostimulated astrocytes by preserving ATP levels. Uridine phosphorylase (UPase), an enzyme catalyzing the reversible phosphorylation of uridine, was involved in this effect. Here, we tried to expand our previous findings by investigating the uridine effect on the brain and neurons using in vivo and in vitro ischemic injury models. Orally administrated uridine (50-200 mg/kg) reduced middle cerebral artery occlusion (1.5 h)/reperfusion (22 h)-induced infarct in mouse brain. Additionally, in the rat brain subjected to the same ischemic condition, UPase mRNA and protein levels were up-regulated. Next, we employed glucose deprivation-induced hypoglycemia in mixed cortical cultures of neurons and astrocytes as an in vitro model. Cells were deprived of glucose and, two hours later, supplemented with 20 mM glucose. Under this condition, a significant ATP loss followed by death was observed in neurons but not in astrocytes, which were blocked by treatment with uridine in a concentration-dependent manner. Inhibition of cellular uptake of uridine by S-(4-nitrobenzyl)-6-thioinosine blocked the uridine effect. Similar to our in vivo data, UPase expression was up-regulated by glucose deprivation in mRNA as well as protein levels. Additionally, 5-(phenylthio)acyclouridine, a specific inhibitor of UPase, prevented the uridine effect. Finally, the uridine effect was shown only in the presence of astrocytes. Taken together, the present study provides the first evidence that uridine protects neurons against ischemic insult-induced neuronal death, possibly through the action of UPase.


Asunto(s)
Hipoxia-Isquemia Encefálica/enzimología , Degeneración Nerviosa/enzimología , Neuronas/enzimología , Fármacos Neuroprotectores/farmacología , Uridina Fosforilasa/metabolismo , Uridina/farmacología , Adenosina Trifosfato/metabolismo , Administración Oral , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Infarto Encefálico/enzimología , Infarto Encefálico/fisiopatología , Infarto Encefálico/prevención & control , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/enzimología , Corteza Cerebral/fisiopatología , Técnicas de Cocultivo , Citoprotección/efectos de los fármacos , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Glucosa/deficiencia , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/fisiopatología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/enzimología , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Ratones , Ratones Endogámicos ICR , Degeneración Nerviosa/fisiopatología , Degeneración Nerviosa/prevención & control , Neuronas/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Uridina Fosforilasa/efectos de los fármacos , Uridina Fosforilasa/genética
6.
Life Sci ; 81(15): 1211-22, 2007 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-17904584

RESUMEN

Recently, it was demonstrated that TRPM7 is an essential mediator of anoxia-induced neuronal death. Meanwhile, nerve growth factor (NGF) is known to have survival and neuroprotective effects by interacting with the high affinity neurotrophin receptor, tropomyosin-related kinase A (trkA). In the present study, we found that electroacupuncture (EA) treatment could up-regulate trkA expression after focal cerebral ischemia in rats. At the same time, EA therapy obviously decreased the high expression of TRPM7 induced by ischemia. Using K252a to inhibit trkA, we found that the EA-mediated down-regulation of TRPM7 was significantly suppressed in rats subjected to cerebral ischemia. TrkA can utilize two distinct signaling pathways: the phosphatidylinositol 3-kinase (PI3K) pathway and the extracellular signal-related kinase (ERK) pathway. We found that the effect of EA on TRPM7 was also inhibited by a PI3K inhibitor, while an ERK inhibitor had no effect. Taken together, our findings suggest that EA can reverse the ischemia-induced increase of TRPM7 levels through the trkA-PI3K pathway.


Asunto(s)
Electroacupuntura , Hipoxia-Isquemia Encefálica/terapia , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor trkA/metabolismo , Daño por Reperfusión/terapia , Canales Catiónicos TRPM/biosíntesis , Animales , Western Blotting , Corteza Cerebral/enzimología , Corteza Cerebral/metabolismo , Regulación hacia Abajo , Hipocampo/enzimología , Hipocampo/metabolismo , Hipoxia-Isquemia Encefálica/complicaciones , Hipoxia-Isquemia Encefálica/enzimología , Hipoxia-Isquemia Encefálica/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/enzimología , Daño por Reperfusión/etiología , Daño por Reperfusión/metabolismo , Transducción de Señal , Canales Catiónicos TRPM/antagonistas & inhibidores
7.
Zhongguo Dang Dai Er Ke Za Zhi ; 8(3): 216-20, 2006 Jun.
Artículo en Chino | MEDLINE | ID: mdl-16787595

RESUMEN

OBJECTIVE: The application and therapeutic effect of hyperbaric oxygen (HBO) in hypoxic-ischemic brain damage (HIBD) remains controversial. Previous studies have focused on the early pathological and biochemical outcomes and there is a lack of long-term functional evaluation. This study was designed to evaluate the long-term pathological and behavioral changes of early HBO therapy on neonatal rats with HIBD. METHODS: Postnatal 7 days (PD7) rat pups were randomly assigned into Control (n=18), HIBD (n=17) and HBO treatment groups (n=17). HIBD was induced by ligating the left common carotid, followed by 2 hrs hypoxia exposure in the HIBD and HBO treatment groups. The Control group was sham-operated and was not subjected to hypoxia exposure. The HBO therapy with 2 atmosphere absolutes began 0.5-1 hr after HIBD in the HIBD treatment group, once daily for 2 days. The spatial learning and memory ability were evaluated by the Morris water maze test at PD37 to PD41. The morphological and histological changes of the brain, including brain weight, survival neurons, AchE positive unit and NOS positive neurons in hippocampal CA1 region, were detected at PD42. RESULTS: The rats in the HIBD group displayed significant morphological and histological deficits, as well as severe spatial learning and memory disability. In the Morris water maze test, the mean escape latency were longer (56.35 +/- 22.37 s vs 23.07 +/- 16.28 s; P < 0.05) and the probe time and probe length were shorter in the HIBD group (29.29 +/- 6.06 s vs 51.21 +/- 4.59 s and 548 +/- 92 cm vs 989 +/- 101 cm; both P < 0.05) compared with the Control group. The left brain weight in the HIBD group was lighter than that in the Control group (0.601 +/- 0.59 g vs 0.984 +/- 0.18 g; P < 0.05). The survival neurons in the hippocampal CA1 region were less (100 +/- 27/mm vs 183 +/- 8/mm; P < 0.05), as well as the AchE-positive unit and NOS-positive neurons (18.50 +/- 2.24% vs 27.50 +/- 2.18% and 19.25 +/- 4.33 vs 33.75 +/- 5.57 respectively; P < 0.05) after HIBD. Early HBO treatment improved the abilities of spatial learning and alleviated the morphological and histological damage. The mean escape latency (39.17 +/- 21.20 s) was shortened, the probe time (36.84 +/- 4.36 s) and the probe length (686 +/- 76 cm) were longer, and the brain weight (0.768 +/- 0.85 g), the survival neurons (133 +/- 25/mm) and the AchE-positive unit (21.94 +/- 2.73%) increased significantly compared with those of the HIBD group (P < 0.05). CONCLUSIONS: Early HBO treatment resulted in a protective effect against HIBD-induced long-term brain morphological and histological deficits and spatial learning and memory disability.


Asunto(s)
Oxigenoterapia Hiperbárica , Hipoxia-Isquemia Encefálica/terapia , Acetilcolinesterasa/análisis , Animales , Encéfalo/patología , Reacción de Fuga , Femenino , Hipocampo/enzimología , Hipocampo/patología , Hipoxia-Isquemia Encefálica/enzimología , Hipoxia-Isquemia Encefálica/patología , Masculino , Aprendizaje por Laberinto , Óxido Nítrico Sintasa/análisis , Ratas , Ratas Sprague-Dawley
8.
Pediatr Res ; 57(6): 858-64, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15774834

RESUMEN

Neonatal hypoxic-ischemic brain injury remains a significant cause of morbidity and mortality and lacks effective therapies for prevention and treatment. Recently, interest in the biology of polyphenol compounds has led to the discovery that dietary supplementation with foods rich in polyphenols (e.g. blueberries, green tea extract) provides neuroprotection in adult animal models of ischemia and Alzheimer's disease. We sought to determine whether protection of the neonatal brain against a hypoxic-ischemic insult could be attained through supplementation of the maternal diet with pomegranate juice, notable for its high polyphenol content. Mouse dams were provided ad libitum access to drinking water with pomegranate juice, at one of three doses, as well as plain water, sugar water, and vitamin C water controls during the last third of pregnancy and throughout the duration of litter suckling. At postnatal day 7, pups underwent unilateral carotid ligation followed by exposure to 8% oxygen for 45 min. Brain injury was assessed histologically after 1 wk (percentage of tissue area loss) and biochemically after 24 h (caspase-3 activity). Dietary supplementation with pomegranate juice resulted in markedly decreased brain tissue loss (>60%) in all three brain regions assessed, with the highest pomegranate juice dose having greatest significance (p < or = 0.0001). Pomegranate juice also diminished caspase-3 activation by 84% in the hippocampus and 64% in the cortex. Ellagic acid, a polyphenolic component in pomegranate juice, was detected in plasma from treated but not control pups. These results demonstrate that maternal dietary supplementation with pomegranate juice is neuroprotective for the neonatal brain.


Asunto(s)
Bebidas , Lesiones Encefálicas/prevención & control , Hipoxia-Isquemia Encefálica/prevención & control , Lythraceae , Animales , Animales Recién Nacidos , Lesiones Encefálicas/enzimología , Lesiones Encefálicas/patología , Caspasa 3 , Caspasas/metabolismo , Ácido Elágico/sangre , Femenino , Flavonoides/administración & dosificación , Hipoxia-Isquemia Encefálica/enzimología , Hipoxia-Isquemia Encefálica/patología , Intercambio Materno-Fetal , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/administración & dosificación , Fenoles/administración & dosificación , Fitoterapia , Polifenoles , Embarazo
9.
Zhongguo Zhong Yao Za Zhi ; 28(12): 1174-7, 2003 Dec.
Artículo en Chino | MEDLINE | ID: mdl-15617505

RESUMEN

OBJECTIVE: To study neuroprotective effects of astragulus membraneaceus on a neonatal rat hippocampus of hypoxia-ischemia brain damage (HIBD). METHOD: The neonatal hypoxia-ischemia model was established with 7-day-old rat pups. Brain injury was examined by neuron death rate in the hippocampal CA1 area. Caspase-3 (cysteinyl aspartate-specific proteinase) mRNA expression in ipsilateral hippocampal was measured by half-quantitative reverse transcription and polymerization chain reaction (RT-PCR). 90-day-old rats were used in tri-equal-arm maze to observe discrimination learning ability. Sham, model and astragulus-membraneaceus treated groups were set up. RESULT: In model group, caspase-3 mRNA showed an increase at 6h, with maximum arrivimg at 24 h - 48 h after HI. In astragulus-membraneaceus treated group, neurons death rate and caspase-3 mRNA were significantly reduced by astragulus membraneaceus, and discrimination learning ability of developed rats were improved obviously. CONCLUSION: Astragulus membraneaceus has a strong protective effect on neuronal damage in the immature rat hippocampus, which is ralated reducing caspase-3 expression.


Asunto(s)
Astragalus propinquus , Caspasas/biosíntesis , Medicamentos Herbarios Chinos/farmacología , Hipocampo/patología , Hipoxia-Isquemia Encefálica/patología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Astragalus propinquus/química , Caspasa 3 , Caspasas/genética , Muerte Celular/efectos de los fármacos , Aprendizaje Discriminativo/efectos de los fármacos , Medicamentos Herbarios Chinos/aislamiento & purificación , Femenino , Hipocampo/enzimología , Hipoxia-Isquemia Encefálica/enzimología , Masculino , Neuronas/patología , Fármacos Neuroprotectores/aislamiento & purificación , Fármacos Neuroprotectores/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley
10.
J Neurochem ; 79(6): 1196-206, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11752060

RESUMEN

The exact molecular mechanism of ischemic neuronal death still remains unclear from rodents to primates. A number of studies using lower species animals have suggested implication of apoptosis cascade, while using monkeys the authors recently claimed necrosis cascade by calpain-induced leakage of lysosomal cathepsins (calpain-cathepsin hypothesis). This paper is to study implications of apoptotic versus necrotic cascades for the development of hippocampal CA1 neuronal death in the primate brain undergoing complete global ischemia. Here, we focused on two terminal cell death effectors; caspase-activated DNase (CAD) and lysosomal enzyme DNase II, in the monkey CA1 sector undergoing 18 min ischemia. The expressions of their mRNA and proteins, and the subcellular localizations as well as ultrastructure and specific DNA gel electrophoresis were examined. Expression of CAD was much less in the normal brain, compared with the lymph node or heart tissues. On day 1 after ischemia, however, CAD mRNA and protein were significantly increased in the CA1 sector, and then CAD protein immunohistochemically showed a translocation from the perikarya into the nucleus. Activated DNase II protein was significantly increased on days 2 and 3 after ischemia, and also showed a similar translocation indicating lysosomal leakage. Although the post-ischemic CA1 neurons showed positive terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling (TUNEL) staining on days 3-5, they showed eosinophilic coagulation necrosis on light microscopy, and frank membrane disruption and mild chromatin condensation on electron microscopy. Furthermore, DNA smear pattern typical for necrosis was observed instead of DNA laddering. These data altogether suggest that the post-ischemic CA1 neuronal death of the monkey occurs not by apoptosis but by necrosis with participations of lysosomal enzymes DNase II and cathepsins as well as CAD. The interactions between apoptotic (caspase-3 and CAD) and necrotic (calpain, cathepsin and DNase II) cascades should be studied further.


Asunto(s)
Desoxirribonucleasas/fisiología , Endodesoxirribonucleasas/fisiología , Hipocampo/enzimología , Hipoxia-Isquemia Encefálica/enzimología , Lisosomas/enzimología , Proteínas del Tejido Nervioso/fisiología , Actinas/genética , Animales , Apoptosis/fisiología , Secuencia de Bases , Fragmentación del ADN , ADN Complementario/genética , Desoxirribonucleasas/biosíntesis , Desoxirribonucleasas/genética , Endodesoxirribonucleasas/biosíntesis , Endodesoxirribonucleasas/genética , Activación Enzimática , Inducción Enzimática , Precursores Enzimáticos/biosíntesis , Precursores Enzimáticos/genética , Perfilación de la Expresión Génica , Hipocampo/irrigación sanguínea , Hipocampo/patología , Humanos , Etiquetado Corte-Fin in Situ , Ataque Isquémico Transitorio/enzimología , Ganglios Linfáticos/enzimología , Macaca , Modelos Biológicos , Datos de Secuencia Molecular , Miocardio/enzimología , Necrosis , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Especificidad de Órganos , ARN Mensajero/biosíntesis , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie , Fracciones Subcelulares/enzimología
11.
Exp Neurol ; 168(2): 323-33, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11259120

RESUMEN

We examined the response of neuronal nitric oxide synthase (nNOS)-containing CNS neurons in rats exposed to a unilateral hypoxic-ischemic insult at 7 days of age. Animals were sacrificed at several time points after the injury, up to and including 7 days (Postnatal Day 14). Brain regions ipsilateral to the injury (including cerebral cortex, caudate-putamen, and thalamus) exhibited delayed, focal increases in nNOS immunoreactivity. The increase in nNOS immunoreactive fiber staining was prominent in areas adjacent to severe neuronal damage, especially in the cortex and the thalamus, regions that are also heavily and focally injured in term human neonates with hypoxic-ischemic encephalopathy. In cerebral cortex, these increases occurred despite modest declines in nNOS catalytic activity and protein levels. Proliferation of surviving nNOS immunoreactive fibers highlights regions of selective vulnerability to hypoxic-ischemic insult in the neonatal brain and may also contribute to plasticity of neuronal circuitry during recovery.


Asunto(s)
Corteza Cerebral/enzimología , Cuerpo Estriado/enzimología , Hipoxia-Isquemia Encefálica/enzimología , Neuronas/enzimología , Óxido Nítrico Sintasa/metabolismo , Tálamo/enzimología , Animales , Animales Recién Nacidos , Femenino , Óxido Nítrico Sintasa de Tipo I , Ratas
12.
J Neurosci ; 20(21): 7994-8004, 2000 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11050120

RESUMEN

Birth asphyxia can cause moderate to severe brain injury. It is unclear to what degree apoptotic or necrotic mechanisms of cell death account for damage after neonatal hypoxia-ischemia (HI). In a 7-d-old rat HI model, we determined the contributions of apoptosis and necrosis to neuronal injury in adjacent Nissl-stained, hematoxylin and eosin-stained, and terminal deoxynucleotidyl transferase-mediated UTP nick end-labeled sections. We found an apoptotic-necrotic continuum in the morphology of injured neurons in all regions examined. Eosinophilic necrotic neurons, typical in adult models, were rarely observed in neonatal HI. Electron microscopic analysis showed "classic" apoptotic and necrotic neurons and "hybrid" cells with intermediate characteristics. The time course of apoptotic injury varied regionally. In CA3, dentate gyrus, medial habenula, and laterodorsal thalamus, the density of apoptotic cells was highest at 24-72 hr after HI and then declined. In contrast, densities remained elevated from 12 hr to 7 d after HI in most cortical areas and in the basal ganglia. Temporal and regional patterns of neuronal death were compared with expression of caspase-3, a cysteine protease involved in the execution phase of apoptosis. Immunocytochemical and Western blot analyses showed increased caspase-3 expression in damaged hemispheres 24 hr to 7 d after HI. A p17 peptide fragment, which results from the proteolytic activation of the caspase-3 precursor, was detected in hippocampus, thalamus, and striatum but not in cerebral cortex. The continued expression of activated caspase-3 and the persistence of cells with an apoptotic morphology for days after HI suggests a prolonged role for apoptosis in neonatal hypoxic ischemic brain injury.


Asunto(s)
Apoptosis , Hipoxia-Isquemia Encefálica/patología , Enfermedades Neurodegenerativas/patología , Animales , Animales Recién Nacidos , Caspasa 3 , Caspasas/metabolismo , Corteza Cerebral/enzimología , Corteza Cerebral/patología , Cuerpo Estriado/enzimología , Cuerpo Estriado/patología , Giro Dentado/enzimología , Giro Dentado/patología , Modelos Animales de Enfermedad , Hipocampo/enzimología , Hipocampo/patología , Hipoxia-Isquemia Encefálica/complicaciones , Hipoxia-Isquemia Encefálica/enzimología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Necrosis , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/etiología , Neuronas/metabolismo , Neuronas/ultraestructura , Ratas , Tálamo/enzimología , Tálamo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA