Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Neurosci ; 26(10): 1805-1819, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37735497

RESUMEN

The prefrontal cortex (PFC) enables mammals to respond to situations, including internal states, with appropriate actions. One such internal state could be 'tiredness'. Here, using activity tagging in the mouse PFC, we identified particularly excitable, fast-spiking, somatostatin-expressing, γ-aminobutyric acid (GABA) (PFCSst-GABA) cells that responded to sleep deprivation. These cells projected to the lateral preoptic (LPO) hypothalamus and the lateral hypothalamus (LH). Stimulating PFCSst-GABA terminals in the LPO hypothalamus caused sleep-preparatory behavior (nesting, elevated theta power and elevated temperature), and stimulating PFCSst-GABA terminals in the LH mimicked recovery sleep (non-rapid eye-movement sleep with higher delta power and lower body temperature). PFCSst-GABA terminals had enhanced activity during nesting and sleep, inducing inhibitory postsynaptic currents on diverse cells in the LPO hypothalamus and the LH. The PFC also might feature in deciding sleep location in the absence of excessive fatigue. These findings suggest that the PFC instructs the hypothalamus to ensure that optimal sleep takes place in a suitable place.


Asunto(s)
Área Hipotalámica Lateral , Neuronas , Ratones , Animales , Área Hipotalámica Lateral/metabolismo , Neuronas/fisiología , Somatostatina/metabolismo , Sueño/fisiología , Hipotálamo/fisiología , Ácido gamma-Aminobutírico , Corteza Prefrontal/fisiología , Mamíferos/metabolismo
2.
Neural Regen Res ; 18(9): 1961-1967, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36926720

RESUMEN

The inflammatory response plays an important role in neuroprotection and regeneration after ischemic insult. The use of non-steroidal anti-inflammatory drugs has been a matter of debate as to whether they have beneficial or detrimental effects. In this context, the effects of the anti-inflammatory agent meloxicam have been scarcely documented after stroke, but its ability to inhibit both cyclooxygenase isoforms (1 and 2) could be a promising strategy to modulate post-ischemic inflammation. This study analyzed the effect of meloxicam in a transient focal cerebral ischemia model in rats, measuring its neuroprotective effect after 48 hours and 7 days of reperfusion and the effects of the treatment on the glial scar and regenerative events such as the generation of new progenitors in the subventricular zone and axonal sprouting at the edge of the damaged area. We show that meloxicam's neuroprotective effects remained after 7 days of reperfusion even if its administration was restricted to the two first days after ischemia. Moreover, meloxicam treatment modulated glial scar reactivity, which matched with an increase in axonal sprouting. However, this treatment decreased the formation of neuronal progenitor cells. This study discusses the dual role of anti-inflammatory treatments after stroke and encourages the careful analysis of both the neuroprotective and the regenerative effects in preclinical studies.

3.
J Neurosci ; 42(27): 5389-5409, 2022 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-35649726

RESUMEN

The lateral preoptic (LPO) hypothalamus is a center for NREM and REM sleep induction and NREM sleep homeostasis. Although LPO is needed for NREM sleep, we found that calcium signals were, surprisingly, highest in REM sleep. Furthermore, and equally surprising, NMDA receptors in LPO were the main drivers of excitation. Deleting the NMDA receptor GluN1 subunit from LPO abolished calcium signals in all cells and produced insomnia. Mice of both sexes had highly fragmented NREM sleep-wake patterns and could not generate conventionally classified REM sleep. The sleep phenotype produced by deleting NMDA receptors depended on where in the hypothalamus the receptors were deleted. Deleting receptors from the anterior hypothalamic area (AHA) did not influence sleep-wake states. The sleep fragmentation originated from NMDA receptors on GABA neurons in LPO. Sleep fragmentation could be transiently overcome with sleeping medication (zolpidem) or sedatives (dexmedetomidine; Dex). By contrast, fragmentation persisted under high sleep pressure produced by sleep deprivation (SD), mice had a high propensity to sleep but woke up. By analyzing changes in δ power, sleep homeostasis (also referred to as "sleep drive") remained intact after NMDA receptor ablation. We suggest NMDA glutamate receptor activation stabilizes firing of sleep-on neurons and that mechanisms of sleep maintenance differ from that of the sleep drive itself.SIGNIFICANCE STATEMENT Insomnia is a common affliction. Most insomniacs feel that they do not get enough sleep, but in fact, often have good amounts of sleep. Their sleep, however, is fragmented, and sufferers wake up feeling unrefreshed. It is unknown how sleep is maintained once initiated. We find that in mice, NMDA-type glutamate receptors in the hypothalamus are the main drivers of excitation and are required for a range of sleep properties: they are, in fact, needed for both sustained NREM sleep periods, and REM sleep generation. When NMDA receptors are selectively reduced from inhibitory preoptic (PO) neurons, mice have normal total amounts of sleep but high sleep-wake fragmentation, providing a model for studying intractable insomnia.


Asunto(s)
Trastornos del Inicio y del Mantenimiento del Sueño , Sueño REM , Animales , Calcio , Electroencefalografía , Femenino , Hipotálamo , Masculino , Ratones , N-Metilaspartato , Receptores de N-Metil-D-Aspartato , Sueño/fisiología , Privación de Sueño , Sueño REM/fisiología , Vigilia/fisiología
4.
Mol Neurobiol ; 58(4): 1404-1417, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33184783

RESUMEN

Stroke is one of the main causes of death and disability worldwide. Ischemic stroke results in unfolded/misfolded protein accumulation in endoplasmic reticulum (ER), a condition known as ER stress. We hypothesized that previously reported neuroprotection of celecoxib, a selective inhibitor of cyclooxygenase-2, in transient middle cerebral artery occlusion (tMCAO) model, relies on the ER stress decrease. To probe this hypothesis, Sprague-Dawley rats were subjected to 1 h of tMCAO and treated with celecoxib or vehicle 1 and 24 h after ischemia. Protein and mRNA levels of the main hallmarks of ER stress, unfolded protein response (UPR) activation, UPR-induced cell death, and ubiquitin proteasome system (UPS) and autophagy, the main protein degradation pathways, were measured at 12 and 48 h of reperfusion. Celecoxib treatment decreased polyubiquitinated protein load and ER stress marker expression such as glucose-related protein 78 (GRP78), C/EBP (CCAAT/enhancer-binding protein) homologous protein (CHOP), and caspase 12 after 48 h of reperfusion. Regarding the UPR activation, celecoxib promoted inositol-requiring enzyme 1 (IRE1) pathway instead of double-stranded RNA-activated protein kinase-like ER kinase (PERK) pathway. Furthermore, celecoxib treatment increased proteasome catalytic subunits transcript levels and decreased p62 protein levels, while the microtubule-associated protein 1 light chain 3 (LC3B) II/I ratio remained unchanged. Thus, the ability of celecoxib treatment on reducing the ER stress correlates with the enhancement of IRE1-UPR pathway and UPS degradation. These data support the ability of anti-inflammatory therapy in modulating ER stress and reveal the IRE1 pathway as a promising therapeutic target in stroke therapy.Graphical abstract.


Asunto(s)
Celecoxib/farmacología , Infarto de la Arteria Cerebral Media/patología , Neuroprotección , Complejo de la Endopetidasa Proteasomal/metabolismo , Respuesta de Proteína Desplegada , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Biomarcadores/metabolismo , Isquemia Encefálica/complicaciones , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Endorribonucleasas/metabolismo , Proteínas de Choque Térmico/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Masculino , Complejos Multienzimáticos/metabolismo , Neuroprotección/efectos de los fármacos , Poliubiquitina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Subunidades de Proteína/metabolismo , Proteolisis/efectos de los fármacos , Ratas Sprague-Dawley , Respuesta de Proteína Desplegada/efectos de los fármacos , eIF-2 Quinasa/metabolismo
5.
J Neurochem ; 151(6): 777-794, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31165478

RESUMEN

Ischemic stroke is one of the most important causes of death and disability worldwide. Subroutines underlying cell death after stroke are largely unknown despite their importance in the design of novel therapies for this pathology. Necroptosis, a recently described form of regulated cell death, has been related with inflammation and, in some models, with endoplasmic reticulum (ER) stress. We hypothesize that alleviation of ER stress following a salubrinal treatment will reduce the ischemic-dependent necroptosis. To probe the hypothesis, we measured, at 48 and 72 h after transient global cerebral ischemia in rat, in cerebral cortex and cornu ammonis 1, the main hallmarks of necroptosis: mRNA levels and phosphorylation of mixed lineage kinase domain like pseudokinase as well as receptor interacting serine/threonine protein kinase 3, along the years 2017-2018. Selective neuronal loss after 7 days of the ischemic insult, and other markers related with the inflammatory response were also measured. This study shows that necroptosis in cerebral cortex can be detected after 72 h of the insult and seems to be elicited before 48 h of reperfusion. The type of necroptosis here observed seems to be tumor necrosis factor receptor 1 independent. Necroptotic response is less evident in the cornu ammonis 1 hippocampal area than in cerebral cortex. The treatment with salubrinal administered 1 and 24 h after the ischemia, decreased the necroptotic marker levels and reduced the areas of selective neuronal loss, supporting the presence of ischemic-dependent necroptosis, and the notion that ER stress is involved in the necroptotic response. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.


Asunto(s)
Isquemia Encefálica/patología , Isquemia Encefálica/prevención & control , Cinamatos/uso terapéutico , Modelos Animales de Enfermedad , Necroptosis/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Tiourea/análogos & derivados , Animales , Isquemia Encefálica/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Cinamatos/farmacología , Masculino , Necroptosis/fisiología , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Tiourea/farmacología , Tiourea/uso terapéutico
6.
J Cell Physiol ; 234(6): 9592-9604, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30317637

RESUMEN

Brain-derived neurotrophic factor (BDNF) is considered as a putative therapeutic agent against stroke. Since BDNF role on oxidative stress is uncertain, we have studied this role in a rat brain slice ischemia model, which allows BDNF reaching the neural parenchyma. Hippocampal and cerebral cortex slices were subjected to oxygen and glucose deprivation (OGD) and then returned to normoxic conditions (reperfusion-like, RL). OGD/RL increased a number of parameters mirroring oxidative stress in the hippocampus that were reduced by the BDNF presence. BDNF also reduced the OGD/RL-increased activity in a number of antioxidant enzymes in the hippocampus but no effects were observed in the cerebral cortex. In general, we conclude that alleviation of oxidative stress by BDNF in OGD/RL-exposed slices relies on decreasing cPLA2 activity, rather than modifying antioxidant enzyme activities. Moreover, a role for the oxidative stress in the differential ischemic vulnerability of cerebral cortex and hippocampus is also supported.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Encéfalo/patología , Glucosa/deficiencia , Modelos Biológicos , Estrés Oxidativo/efectos de los fármacos , Oxígeno/toxicidad , Animales , Antioxidantes/metabolismo , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citosol/enzimología , Peroxidación de Lípido/efectos de los fármacos , Masculino , NADPH Oxidasas/metabolismo , Fosfolipasas A2/genética , Fosfolipasas A2/metabolismo , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Reperfusión , Transcripción Genética/efectos de los fármacos
9.
J Pharmacol Exp Ther ; 367(3): 528-542, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30291174

RESUMEN

Areas of selective neuronal loss (SNL) represent the first morphologic signs of damage in the penumbra region and are considered putative targets for ischemic stroke therapy. We performed a novel assessment of measuring the effects of the anti-inflammatory agent celecoxib by analyzing simultaneously the different neural populations (neurons, astrocytes, and microglia cells) in SNL and non-SNL areas. Rats were subjected to 1 hour of middle cerebral artery occlusion (MCAO) and treated with celecoxib 1 and 24 hours after ischemia. Infarct volume measurements and triple immunostaining of neurons (neuronal nuclear antigen), microglia (ionized calcium-binding adaptor molecule 1), and astroglia were performed after 12 and 48 hours of reperfusion. Motor response was tested by standard behavioral assays at 3, 12, 24, and 48 hours. Confocal analysis revealed that the percentage of SNL areas, microglia densities, and glial activation increased at 48 hours of reperfusion. Celecoxib treatment improved the neurologic deficit, reduced the infarct volume by 50% after 48 hours of reperfusion, and resulted in a reduced percentage of SNL areas and microglia and astroglia reactivity after 48 hours of reperfusion. This study proves, for the first time, that celecoxib presents postischemic neuroprotective effects in a transient MCAO model, prevents or delays the presence of SNL areas, and reduces glial activation.


Asunto(s)
Celecoxib/farmacología , Infarto de la Arteria Cerebral Media/complicaciones , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/etiología , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Astrocitos/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/tratamiento farmacológico , Accidente Cerebrovascular/tratamiento farmacológico
10.
Neuroscience ; 386: 68-78, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-29949743

RESUMEN

We previously demonstrated that the non-steroidal anti-inflammatory agent meloxicam has neuroprotective effects in an oxygen and glucose deprivation model (OGD) of rat organotypic hippocampal slice cultures. We wondered if GABAergic transmission changed the neuroprotective effects of meloxicam and if meloxicam was able to modulate endoplasmic reticulum stress (ER stress) in this model. Mortality was measured using propidium iodide. Western blot assays were performed to measure levels of cleaved and non-cleaved caspase-3 to quantify apoptosis, while levels of GRP78, GRP94 and phosphorylated eIF2α were used to detect unfolded protein response (UPR). Transcript levels of GRP78, GRP94 and GABAergic receptor α, ß, and γ subunits were measured by real-time quantitative polymerase chain reaction (qPCR). In the present study, we show that the presence of meloxicam in a 30 min OGD assay, followed by 24 h of normoxic conditions, presented an antiapoptotic effect. The simultaneous presence of the GABAA receptor antagonist, bicuculline, in combination with meloxicam blocked the neuroprotective effect provided by the latter. However, in light of its effects on caspase 3 and PARP, bicuculline did not seem to promote the apoptotic pathway. Our results also showed that meloxicam modified the unfolded protein response (UPR), as well as the transcriptional response of different genes, including the GABAA receptor, alpha1, beta3 and gamma2 subunits. We concluded that meloxicam has a neuroprotective anti-apoptotic action, is able to enhance the UPR independently of the systemic anti-inflammatory response and its neuroprotective effect can be inhibited by blocking GABAA receptors.


Asunto(s)
Bicuculina/farmacología , Glucosa/deficiencia , Hipocampo/metabolismo , Meloxicam/farmacología , Fármacos Neuroprotectores/farmacología , Oxígeno , Animales , Antiinflamatorios no Esteroideos/farmacología , Muerte Celular/fisiología , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Relación Dosis-Respuesta a Droga , Antagonistas de Receptores de GABA-A/farmacología , Glucosa/metabolismo , Hipocampo/efectos de los fármacos , Técnicas de Cultivo de Órganos , Oxígeno/metabolismo , Ratas , Ratas Wistar , Resultado del Tratamiento
11.
Biochem Pharmacol ; 151: 26-37, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29499167

RESUMEN

BACKGROUND: Blood reperfusion of the ischemic tissue after stroke promotes increases in the inflammatory response as well as accumulation of unfolded/misfolded proteins in the cell, leading to endoplasmic reticulum (ER) stress. Both Inflammation and ER stress are critical processes in the delayed death of the cells damaged after ischemia. The aim of this study is to check the putative synergic neuroprotective effect by combining anti-inflammatory and anti-ER stress agents after ischemia. METHODS: The study was performed on a two-vessel occlusion global cerebral ischemia model. Animals were treated with salubrinal one hour after ischemia and with robenacoxib at 8 h and 32 h after ischemia. Parameters related to the integrity of the blood-brain barrier (BBB), such as matrix metalloproteinase 9 and different cell adhesion molecules (CAMs), were analyzed by qPCR at 24 h and 48 h after ischemia. Microglia and cell components of the neurovascular unit, including neurons, endothelial cells and astrocytes, were analyzed by immunofluorescence after 48 h and seven days of reperfusion. RESULTS: Pharmacologic control of ER stress by salubrinal treatment after ischemia, revealed a neuroprotective effect over neurons that reduces the transcription of molecules involved in the impairment of the BBB. Robenacoxib treatment stepped neuronal demise forward, revealing a detrimental effect of this anti-inflammatory agent. Combined treatment with robenacoxib and salubrinal after ischemia prevented neuronal loss and changes in components of the neurovascular unit and microglia observed when animals were treated only with robenacoxib. CONCLUSION: Combined treatment with anti-ER stress and anti-inflammatory agents is able to provide enhanced neuroprotective effects reducing glial activation, which opens new avenues in therapies against stroke.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Cinamatos/uso terapéutico , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Difenilamina/análogos & derivados , Estrés del Retículo Endoplásmico/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Fenilacetatos/uso terapéutico , Tiourea/análogos & derivados , Animales , Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/diagnóstico por imagen , Isquemia Encefálica/inmunología , Cinamatos/administración & dosificación , Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Difenilamina/administración & dosificación , Difenilamina/uso terapéutico , Esquema de Medicación , Quimioterapia Combinada , Inflamación , Masculino , Fármacos Neuroprotectores/administración & dosificación , Fenilacetatos/administración & dosificación , Ratas Sprague-Dawley , Tiourea/administración & dosificación , Tiourea/uso terapéutico
12.
J Neurochem ; 138(2): 295-306, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27123756

RESUMEN

This study describes the neuroprotective effect of treatment with salubrinal 1 and 24 h following 15 min of ischemia in a two-vessel occlusion model of global cerebral ischemia. The purpose of this study was to determine if salubrinal, an enhancer of the unfolded protein response, reduces the neural damage modulating the inflammatory response. The study was performed in CA1 and CA3 hippocampal areas as well as in the cerebral cortex whose different vulnerability to ischemic damage is widely described. Characterization of proteins was made by western blot, immunofluorescence, and ELISA, whereas mRNA levels were measured by Quantitative PCR. The salubrinal treatment decreased the cell demise in CA1 at 7 days as well as the levels of matrix metalloprotease 9 (MMP-9) in CA1 and cerebral cortex at 48 h and ICAM-1 and VCAM-1 cell adhesion molecules. However, increases in tumor necrosis factor α and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) inflammatory markers were observed at 24 h. Glial fibrillary acidic protein levels were not modified by salubrinal treatment in CA1 and cerebral cortex. We describe a neuroprotective effect of the post-ischemic treatment with salubrinal, measured as a decrease both in CA1 cell demise and in the blood-brain barrier impairment. We hypothesize that the ability of salubrinal to counteract the CA1 cell demise is because of a reduced ability of this structure to elicit unfolded protein response which would account for its greater ischemic vulnerability. Data of both treated and non-treated animals suggest that the neurovascular unit present a structure-dependent response to ischemia and a different course time for CA1/cerebral cortex compared with CA3. Finally, our study reveals a high responsiveness of endothelial cells to salubrinal in contrast to the limited responsiveness of astrocytes. The alleviation of ER stress by enhancing UPR with salubrinal treatment reduces the ischemic damage. This effect varies across the different neurovascular unit cell types. The salubrinal neuroprotective effect on CA1 supports differences in neurovascular unit for different brain regions and involves the inflammatory response and its time course. Thus, UPR modulation could be a therapeutic target in cerebral ischemia.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Fármacos Neuroprotectores/farmacología , Animales , Astrocitos/metabolismo , Isquemia Encefálica/patología , Infarto Cerebral/metabolismo , Infarto Cerebral/patología , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipocampo/metabolismo , Masculino , Ratas Sprague-Dawley
14.
Neuropathol Appl Neurobiol ; 41(4): e68-79, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24861158

RESUMEN

AIMS: To evaluate the neuroprotective role of autophagy in the cerebral cortex and hippocampus using an ex vivo animal model of stroke in brain slices. METHODS: Brain slices were maintained for 30 min in oxygen and glucose deprivation (OGD) followed by 3 h in normoxic conditions to simulate the reperfusion that follows ischaemia in vivo (RL, reperfusion-like). Phagophore formation (Beclin 1 and LC3B) as well as autophagy flux (p62/SQSTM1, Atg5, Atg7 and polyubiquitin) markers were quantified by Western blot and/or qPCR. The release of lactate dehydrogenase (LDH) and glutamate in the medium was used as a measure of the mortality in the absence and in the presence of the autophagy inhibitor 3-methyladenine. RESULTS: Striking differences in the autophagy markers were observed between the hippocampus and cerebral cortex in normoxic conditions. OGD/RL induced increases both in the phagophore formation and in the autophagy flux in the first three hours in the cerebral cortex that were not observed in the hippocampus. The blocking of autophagy increased the OGD/RL-induced mortality, increased the glutamate release in both the cerebral cortex and hippocampus and abolished the OGD-induced decrease in the polyubiquitinated proteins in the cerebral cortex. CONCLUSIONS: We conclude that OGD induces a rapid autophagic response in the cerebral cortex that plays a neuroprotective role. Polyubiquitination levels and control of the glutamate release appear to be involved in the neuroprotective role of autophagy.


Asunto(s)
Autofagia , Isquemia Encefálica/metabolismo , Corteza Cerebral/metabolismo , Hipocampo/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Hipoxia de la Célula , Glucosa/deficiencia , Ácido Glutámico/metabolismo , Técnicas In Vitro , Masculino , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo
15.
Age (Dordr) ; 36(5): 9703, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25182537

RESUMEN

Stroke is one of the leading causes of death and permanent disability in the elderly. However, most of the experimental studies on stroke are based on young animals, and we hypothesised that age can substantially affect the stroke response. The two-vessel occlusion model of global ischemia by occluding the common carotid arteries for 15 min at 40 mmHg of blood pressure was carried out in 3- and 18-month-old male Sprague-Dawley rats. The adhesion molecules E- and P-selectin, cell adhesion molecules (CAMs), both intercellular (ICAM-1) and vascular (VCAM-1), as well as glial fibrillary acidic protein (GFAP), and cleaved caspase-3 were measured at 48 h after ischemia in the cerebral cortex and hippocampus using Western blot, qPCR and immunofluorescence techniques. Diametric expression of GFAP and a different morphological pattern of caspase-3 labelling, although no changes in the cell number, were observed in the neurons of young and old animals. Expression of E-selectin and CAMs was also modified in an age- and ischemia/reperfusion-dependent manner. The hippocampus and cerebral cortex had similar response patterns for most of the markers studied. Our data suggest that old and young animals present different time-courses of neuroinflammation and apoptosis after ischemic damage. On the other hand, these results suggest that neuroinflammation is dependent on age rather than on the different vulnerability described for the hippocampus and cerebral cortex. These differences should be taken into account in searching for therapeutic targets.


Asunto(s)
Envejecimiento/metabolismo , Isquemia Encefálica/metabolismo , Selectina E/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas del Tejido Nervioso/genética , Reperfusión/métodos , Molécula 1 de Adhesión Celular Vascular/genética , Envejecimiento/genética , Envejecimiento/patología , Animales , Apoptosis , Western Blotting , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Selectina E/biosíntesis , Proteína Ácida Fibrilar de la Glía , Masculino , Proteínas del Tejido Nervioso/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Molécula 1 de Adhesión Celular Vascular/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...