Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neurotox Res ; 41(6): 526-545, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37378827

RESUMEN

Neonatal hypoxia-ischemia (HI) is one of the main causes of tissue damage, cell death, and imbalance between neuronal excitation and inhibition and synaptic loss in newborns. GABA, the major inhibitory neurotransmitter of the central nervous system (CNS) in adults, is excitatory at the onset of neurodevelopment and its action depends on the chloride (Cl-) cotransporters NKCC1 (imports Cl-) and KCC2 (exports Cl-) expression. Under basal conditions, the NKCC1/KCC2 ratio decreases over neurodevelopment. Thus, changes in this ratio caused by HI may be related to neurological disorders. The present study evaluated the effects of bumetanide (NKCC cotransporters inhibitor) on HI impairments in two neurodevelopmental periods. Male Wistar rat pups, 3 (PND3) and 11 (PND11) days old, were submitted to the Rice-Vannucci model. Animals were divided into 3 groups: SHAM, HI-SAL, and HI-BUM, considering each age. Bumetanide was administered intraperitoneally at 1, 24, 48, and 72 h after HI. NKCC1, KCC2, PSD-95, and synaptophysin proteins were analyzed after the last injection by western blot. Negative geotaxis, righting reflex, open field, object recognition test, and Morris water maze task were performed to assess neurological reflexes, locomotion, and memory function. Tissue atrophy and cell death were evaluated by histology. Bumetanide prevented neurodevelopmental delay, hyperactivity, and declarative and spatial memory deficits. Furthermore, bumetanide reversed HI-induced brain tissue damage, reduced neuronal death and controlled GABAergic tone, maintained the NKCC1/KCC2 ratio, and synaptogenesis close to normality. Thereby, bumetanide appears to play an important therapeutic role in the CNS, protecting the animals against HI damage and improving functional performance.


Asunto(s)
Bumetanida , Hipoxia-Isquemia Encefálica , Ratas , Animales , Masculino , Bumetanida/farmacología , Bumetanida/uso terapéutico , Ratas Wistar , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Isquemia/tratamiento farmacológico , Hipoxia/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/complicaciones , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Encéfalo/metabolismo , Cognición , Animales Recién Nacidos
2.
Behav Brain Res ; 430: 113935, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35605797

RESUMEN

Chronic cerebral hypoperfusion leads to neuronal loss in the hippocampus and spatial memory impairments. Physical exercise is known to prevent cognitive deficits in animal models; and there is evidence of sex differences in behavioral neuroprotective approaches. The aim of present study was to investigate the effects of acrobatic training in male and female rats submitted to chronic cerebral hypoperfusion. Males and females rats underwent 2VO (two-vessel occlusion) surgery and were randomly allocated into 4 groups of males and 4 groups of females, as follows: 2VO acrobatic, 2VO sedentary, Sham acrobatic and Sham sedentary. The acrobatic training started 45 days after surgery and lasted 4 weeks; animals were then submitted to object recognition and water maze testing. Brain samples were collected for histological and morphological assessment and flow cytometry. 2VO causes cognitive impairments and acrobatic training prevented spatial memory deficits assessed in the water maze, mainly for females. Morphological analysis showed that 2VO animals had less NeuN labeling and acrobatic training prevented it. Increased number of GFAP positive cells was observerd in females; moreover, males had more branched astrocytes and acrobatic training prevented the branching after 2VO. Flow cytometry showed higher mitochondrial potential in trained animals and more reactive oxygen species production in males. Acrobatic training promoted neuronal survival and improved mitochondrial function in both sexes, and influenced the glial scar in a sex-dependent manner, associated to greater cognitive benefit to females after chronic cerebral hypoperfusion.


Asunto(s)
Isquemia Encefálica , Memoria Espacial , Animales , Femenino , Masculino , Ratas , Astrocitos/patología , Isquemia Encefálica/patología , Cicatriz/patología , Modelos Animales de Enfermedad , Hipocampo , Aprendizaje por Laberinto , Memoria Espacial/fisiología
3.
Mol Neurobiol ; 58(5): 2297-2308, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33417220

RESUMEN

Neonatal hypoxia-ischemia (HI) is among the main causes of mortality and morbidity in newborns. Experimental studies show that the immature rat brain is less susceptible to HI injury, suggesting that changes that occur during the first days of life drastically alter its susceptibility. Among the main developmental changes observed is the mitochondrial function, namely, the tricarboxylic acid (TCA) cycle and respiratory complex (RC) activities. Therefore, in the present study, we investigated the influence of neonatal HI on mitochondrial functions, redox homeostasis, and cell damage at different postnatal ages in the hippocampus of neonate rats. For this purpose, animals were divided into four groups: sham postnatal day 3 (ShP3), HIP3, ShP11, and HIP11. We initially observed increased apoptosis in the HIP11 group only, indicating a higher susceptibility of these animals to brain injury. Mitochondrial damage, as determined by flow cytometry showing mitochondrial swelling and loss of mitochondrial membrane potential, was also demonstrated only in the HIP11 group. This was consistent with the decreased mitochondrial oxygen consumption, reduced TCA cycle enzymes, and RC activities and induction of oxidative stress in this group of animals. Considering that HIP3 and the sham animals showed no alteration of mitochondrial functions, redox homeostasis, and showed no apoptosis, our data suggest an age-dependent vulnerability of the hippocampus to hypoxia-ischemia. The present results highlight age-dependent metabolic differences in the brain of neonate rats submitted to HI indicating that different treatments might be needed for HI newborns with different gestational ages.


Asunto(s)
Apoptosis/fisiología , Hipocampo/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Factores de Edad , Animales , Modelos Animales de Enfermedad , Femenino , Homeostasis/fisiología , Oxidación-Reducción , Consumo de Oxígeno/fisiología , Ratas , Ratas Wistar
4.
J Neurochem ; 157(6): 1911-1929, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33098090

RESUMEN

Prenatal and early postnatal periods are important for brain development and neural function. Neonatal insults such as hypoxia-ischemia (HI) causes prolonged neural and metabolic dysregulation, affecting central nervous system maturation. There is evidence that brain hypometabolism could increase the risk of adult-onset neurodegenerative diseases. However, the impact of non-pharmacologic strategies to attenuate HI-induced brain glucose dysfunction is still underexplored. This study investigated the long-term effects of early environmental enrichment in metabolic, cell, and functional responses after neonatal HI. Thereby, male Wistar rats were divided according to surgical procedure, sham, and HI (performed at postnatal day 3), and the allocation to standard (SC) or enriched condition (EC) during gestation and lactation periods. In-vivo cerebral metabolism was assessed by means of [18 F]-FDG micro-positron emission tomography, and cognitive, biochemical, and histological analyses were performed in adulthood. Our findings reveal that HI causes a reduction in glucose metabolism and glucose transporter levels as well as hyposynchronicity in metabolic brain networks. However, EC during prenatal or early postnatal period attenuated these metabolic disturbances. A positive correlation was observed between [18 F]-FDG values and volume ratios in adulthood, indicating that preserved tissue by EC is metabolically active. EC promotes better cognitive scores, as well as down-regulation of amyloid precursor protein in the parietal cortex and hippocampus of HI animals. Furthermore, growth-associated protein 43 was up-regulated in the cortex of EC animals. Altogether, results presented support that EC during gestation and lactation period can reduce HI-induced impairments that may contribute to functional decline and progressive late neurodegeneration.


Asunto(s)
Encéfalo/metabolismo , Ambiente , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/prevención & control , Plasticidad Neuronal/fisiología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Animales , Animales Recién Nacidos , Femenino , Hipoxia-Isquemia Encefálica/psicología , Lactancia/metabolismo , Lactancia/psicología , Masculino , Aprendizaje por Laberinto/fisiología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/prevención & control , Enfermedades Neurodegenerativas/psicología , Tomografía de Emisión de Positrones/métodos , Embarazo , Efectos Tardíos de la Exposición Prenatal/psicología , Ratas , Ratas Wistar
5.
Neurobiol Learn Mem ; 171: 107207, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32147586

RESUMEN

BACKGROUND AND PURPOSE: Hypoxia and cerebral ischemia (HI) events are capable of triggering important changes in brain metabolism, including glucose metabolism abnormalities, which may be related to the severity of the insult. Using positron emission microtomography (microPET) with [18F]fluorodeoxyglucose (18F-FDG), this study proposes to assess abnormalities of brain glucose metabolism in adult rats previously submitted to the neonatal HI model. We hypothesize that cerebral metabolic outcomes will be associated with cognitive deficits and magnitude of brain injury. METHODS: Seven-day-old rats were subjected to an HI model, induced by permanent occlusion of the right common carotid artery and systemic hypoxia. 18F-FDG-microPET was used to assess regional and whole brain glucose metabolism in rats at 60 postnatal days (PND 60). An interregional cross-correlation matrix was utilized to construct metabolic brain networks (MBN). Rats were also subjected to the Morris Water Maze (MWM) to evaluate spatial memory and their brains were processed for volumetric evaluation. RESULTS: Brain glucose metabolism changes were observed in adult rats after neonatal HI insult, limited to the right brain hemisphere. However, not all HI animals exhibited significant cerebral hypometabolism. Hippocampal glucose metabolism was used to stratify HI animals into HI hypometabolic (HI-h) and HI non-hypometabolic (HI non-h) groups. The HI-h group had drastic MBN disturbance, cognitive deficit, and brain tissue loss, concomitantly. Conversely, HI non-h rats had normal brain glucose metabolism and brain tissue preserved, but also presented MBN changes and spatial memory impairment. Furthermore, data showed that brain glucose metabolism correlated with cognitive deficits and brain volume outcomes. CONCLUSIONS: Our findings demonstrated that long-term changes in MBN drive memory impairments in adult rats subjected to neonatal hypoxic ischemia, using in vivo imaging microPET-FDG. The MBN analyses identified glucose metabolism abnormalities in HI non-h animals, which were not detected by conventional 18F-FDG standardized uptake value (SUVr) measurements. These animals exhibited a metabolic brain signature that may explain the cognitive deficit even with no identifiable brain damage.


Asunto(s)
Encéfalo/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Trastornos de la Memoria/metabolismo , Red Nerviosa/metabolismo , Animales , Encéfalo/diagnóstico por imagen , Modelos Animales de Enfermedad , Glucosa/metabolismo , Hipoxia-Isquemia Encefálica/complicaciones , Hipoxia-Isquemia Encefálica/diagnóstico por imagen , Masculino , Trastornos de la Memoria/diagnóstico por imagen , Trastornos de la Memoria/etiología , Red Nerviosa/diagnóstico por imagen , Tomografía de Emisión de Positrones , Ratas , Ratas Wistar
6.
Neurochem Res ; 43(12): 2268-2276, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30255215

RESUMEN

Neonatal hypoxia-ischemia (HI) is associated to cognitive and motor impairments and until the moment there is no proven treatment. The underlying neuroprotective mechanisms of stem cells are partially understood and include decrease in excitotoxicity, apoptosis and inflammation suppression. This study was conducted in order to test the effects of intracardiac transplantation of human dental pulp stem cells (hDPSCs) for treating HI damage. Seven-day-old Wistar rats were divided into four groups: sham-saline, sham-hDPSCs, HI-saline, and HI-hDPSCs. Motor and cognitive tasks were performed from postnatal day 30. HI-induced cognitive deficits in the novel-object recognition test and in spatial reference memory impairment which were prevented by hDPSCs. No motor impairments were observed in HI animals. Immunofluorescence analysis showed human-positive nuclei in hDPSC-treated animals closely associated with anti-GFAP staining in the lesion scar tissue, suggesting that these cells were able to migrate to the injury site and could be providing support to CNS cells. Our study evidence novel evidence that hDPSC can contribute to the recovery following hypoxia-ischemia and highlight the need of further investigation in order to better understand the exact mechanisms underlying its neuroprotective effects.


Asunto(s)
Disfunción Cognitiva/prevención & control , Pulpa Dental/trasplante , Hipoxia-Isquemia Encefálica/terapia , Trasplante de Células Madre/métodos , Animales , Animales Recién Nacidos , Células Cultivadas , Disfunción Cognitiva/etiología , Disfunción Cognitiva/patología , Pulpa Dental/citología , Pulpa Dental/fisiología , Femenino , Ventrículos Cardíacos , Humanos , Hipoxia-Isquemia Encefálica/complicaciones , Hipoxia-Isquemia Encefálica/patología , Inyecciones , Masculino , Aprendizaje por Laberinto/fisiología , Embarazo , Distribución Aleatoria , Ratas , Ratas Wistar , Células Madre/fisiología
7.
J Perinat Med ; 46(4): 433-439, 2018 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-28841577

RESUMEN

Neonatal hypoxia ischemia (HI) is the main cause of mortality and morbidity in newborns. The mechanisms involved in its progression start immediately and persist for several days. Oxidative stress and inflammation are determinant factors of the severity of the final lesion. The spleen plays a major part in the inflammatory response to HI. This study assessed the temporal progression of HI-induced alterations in oxidative stress parameters in the hippocampus, the most affected brain structure, and in the spleen. HI was induced in Wistar rat pups in post-natal day 7. Production of reactive oxygen species (ROS), and the activity of the anti oxidant enzyme superoxide dismutase and catalase were assessed 24 h, 96 h and 38 days post-HI. Interestingly, both structures showed a similar pattern, with few alterations in the production of ROS species up to 96 h often combined with an increased activity of the anti oxidant enzymes. However, 38 days after the injury, ROS were at the highest in both structures, coupled with a decrease in the activity of the enzymes. Altogether, present results suggest that HI causes long lasting alterations in the hippocampus as well as in the spleen, suggesting a possible target for delayed treatments for HI.


Asunto(s)
Hipocampo/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Estrés Oxidativo , Bazo/metabolismo , Animales , Animales Recién Nacidos , Catalasa/metabolismo , Modelos Animales de Enfermedad , Femenino , Hipocampo/patología , Hipoxia-Isquemia Encefálica/patología , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Bazo/patología , Superóxido Dismutasa/metabolismo
8.
Neurochem Res ; 42(5): 1422-1429, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28210957

RESUMEN

Regular physical activity has shown to improve the quality of life and to prevent age-related memory deficits. Memory processing requires proper regulation of several enzymes such as sodium-potassium adenosine triphosphatase (Na+, K+-ATPase) and acetylcholinesterase (AChE), which have a pivotal role in neuronal transmission. The present study investigated the effects of a treadmill running protocol in young (3 months), mature (6 months) and aged (22 months) Wistar rats, on: (a) cognitive function, as assessed in the Water maze spatial tasks; (b) Na+, K+-ATPase and AChE activities in the hippocampus following cognitive training alone or treadmill running combined with cognitive training. Animals of all ages were assigned to naïve (with no behavioral or exercise training), sedentary (non-exercised, with cognitive training) and exercised (20 min of daily running sessions, 3 times per week for 4 weeks and with cognitive training) groups. Cognition was assessed by reference and working memory tasks run in the Morris Water maze; 24 h after last session of behavioral testing, hippocampi were collected for biochemical analysis. Results demonstrated that: (a) a moderate treadmill running exercise prevented spatial learning and memory deficits in aged rats; (b) training in the Water maze increased both Na+, K+-ATPase and AChE activities in the hippocampus of mature and aged rats; (c) aged exercised rats displayed an even further increase of Na+, K+-ATPase activity in the hippocampus, (d) enzyme activity correlated with memory performance in aged rats. It is suggested that exercise prevents spatial memory deficits in aged rats probably through the activation of Na+, K+-ATPase in the hippocampus.


Asunto(s)
Envejecimiento/metabolismo , Hipocampo/enzimología , Trastornos de la Memoria/enzimología , Condicionamiento Físico Animal/fisiología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Memoria Espacial/fisiología , Animales , Activación Enzimática/fisiología , Prueba de Esfuerzo/métodos , Prueba de Esfuerzo/psicología , Masculino , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/prevención & control , Condicionamiento Físico Animal/métodos , Condicionamiento Físico Animal/psicología , Distribución Aleatoria , Ratas , Ratas Wistar
9.
J Neurosci Res ; 95(1-2): 409-421, 2017 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-27870406

RESUMEN

Neonatal hypoxia-ischemia (HI) is an important cause of neurological deficits in humans, and the Levine-Rice model of experimental HI in the rat mimics the human brain lesion and the following sensory motor deficits and cognitive disabilities. With the growing evidence that sex influences all levels of brain functions, this Mini-Review highlights studies in which sex was a controlled variable and that provided evidence of sexual dimorphism in behavioral outcome, extension of brain damage, mechanisms of lesion, and treatment efficacy in the rat neonatal HI model. It was shown that 1) females have greater memory deficits; 2) cell death is dependent mainly on caspase activation in females; 3) males are more susceptible to oxidative stress; and 4) treatments acting on distinct cell death pathways afford sex-dependent neuroprotection. These tentative conclusions, along with growing evidence from other fields of neurobiology, support the need for scientists to design their experiments considering sex as an important variable; otherwise, important knowledge will continue to be missed. It is conceivable that sex can influence the development of efficacious therapeutic tools to treat neonates suffering from brain HI. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Encéfalo/patología , Discapacidades del Desarrollo/etiología , Hipoxia-Isquemia Encefálica/complicaciones , Caracteres Sexuales , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Femenino , Hipoxia-Isquemia Encefálica/patología , Hipoxia-Isquemia Encefálica/fisiopatología , Masculino , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...