Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neurotherapeutics ; 20(1): 39-47, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36357662

RESUMEN

White matter injury is a progressive vascular disease that leads to neurological deficits and vascular dementia. It comprises up to 30% of all diagnosed strokes, though up to ten times as many events go undiagnosed in early stages. There are several pathologies that can lead to white matter injury. While some studies suggest that white matter injury starts as small infarcts in deep penetrating blood vessels in the brain, others point to the breakdown of endothelial function or the blood-brain barrier as the primary cause of the disease. Whether due to local endothelial or BBB dysfunction, or to local small infarcts (or a combination), white matter injury progresses, accumulates, and expands from preexisting lesions into adjacent white matter to produce motor and cognitive deficits that present as vascular dementia in the elderly. Vascular dementia is the second leading cause of dementia, and white matter injury-attributed vascular dementia represents 40% of all diagnosed dementias and aggravates Alzheimer's pathology. Despite the advances in the last 15 years, there are few animal models of progressive subcortical white matter injury or vascular dementia. This review will discuss recent progress in animal modeling of white matter injury and the emerging principles to enhance glial function as a means of promoting repair and recovery.


Asunto(s)
Lesiones Encefálicas , Demencia Vascular , Sustancia Blanca , Animales , Demencia Vascular/etiología , Demencia Vascular/patología , Encéfalo/patología , Lesiones Encefálicas/patología , Isquemia/complicaciones , Isquemia/patología , Infarto/complicaciones , Infarto/patología
2.
Stem Cell Res ; 55: 102458, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34274773

RESUMEN

White matter stroke (WMS) occurs as small infarcts in deep penetrating blood vessels in the brain and affects the regions of the brain that carry connections, termed the subcortical white matter. WMS progresses over years and has devastating clinical consequences. Unlike large grey matter strokes, WMS disrupts the axonal architecture of the brain and depletes astrocytes, oligodendrocyte lineage cells, axons and myelinating cells, resulting in abnormalities of gait and executive function. An astrocytic cell-based therapy is positioned as a strong therapeutic candidate after WMS. In this study we report, the reliable generation of a novel stem cell-based therapeutic product, glial enriched progenitors (GEPs) derived from human induced pluripotent stem cells (hiPSCs). By transient treatment of hiPSC derived neural progenitors (hiPSC-NPCs) with the small molecule deferoxamine, a prolyl hydroxylase inhibitor, for three days hiPSC-NPCs become permanently biased towards an astrocytic fate, producing hiPSC-GEPs. In preparation for clinical application, we have developed qualification assays to ensure identity, safety, purity, and viability of the cells prior to manufacture. Using tailored q-RT-PCR-based assays, we have demonstrated the lack of pluripotency in our final therapeutic candidate cells (hiPSC-GEPs) and we have identified the unique genetic profile of hiPSC-GEPs that is clearly distinct from the parent lines, hiPSCs and iPSC-NPCs. After completion of the viability assay, we have stablished the therapeutic window of use for hiPSC-GEPs in future clinical applications (7 h). Lastly, we were able to reliably and consistently produce a safe therapeutic final product negative for contamination by any human or murine viral pathogens, selected bacteria, common laboratory mycoplasmas, growth of any aerobes, anaerobes, yeast, or fungi and 100 times less endotoxin levels than the maximum acceptable value. This study demonstrates the reliable and safe generation of patient derived hiPSC-GEPs that are clinically ready as a cell-based therapeutic approach for WMS.


Asunto(s)
Células Madre Pluripotentes Inducidas , Animales , Astrocitos , Diferenciación Celular , Fibroblastos , Humanos , Ratones , Oligodendroglía
3.
Sci Transl Med ; 13(590)2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33883275

RESUMEN

Subcortical white matter stroke (WMS) accounts for up to 30% of all stroke events. WMS damages primarily astrocytes, axons, oligodendrocytes, and myelin. We hypothesized that a therapeutic intervention targeting astrocytes would be ideally suited for brain repair after WMS. We characterize the cellular properties and in vivo tissue repair activity of glial enriched progenitor (GEP) cells differentiated from human-induced pluripotent stem cells, termed hiPSC-derived GEPs (hiPSC-GEPs). hiPSC-GEPs are derived from hiPSC-neural progenitor cells via an experimental manipulation of hypoxia inducible factor activity by brief treatment with a prolyl hydroxylase inhibitor, deferoxamine. This treatment permanently biases these cells to further differentiate toward an astrocyte fate. hiPSC-GEPs transplanted into the brain in the subacute period after WMS in mice migrated widely, matured into astrocytes with a prorepair phenotype, induced endogenous oligodendrocyte precursor proliferation and remyelination, and promoted axonal sprouting. hiPSC-GEPs enhanced motor and cognitive recovery compared to other hiPSC-differentiated cell types. This approach establishes an hiPSC-derived product with easy scale-up capabilities that might be effective for treating WMS.


Asunto(s)
Demencia Vascular , Accidente Cerebrovascular , Sustancia Blanca , Animales , Diferenciación Celular , Humanos , Ratones , Vaina de Mielina , Oligodendroglía , Roedores , Accidente Cerebrovascular/terapia
5.
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
7.
Data Brief ; 10: 202-209, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27995155

RESUMEN

This article presents data related to the research article "Systematic optimization of an engineered hydrogel allows for selective control of human neural stem cell survival and differentiation after transplantation in the stroke brain" (P. Moshayedi, L.R. Nih, I.L. Llorente, A.R. Berg, J. Cinkornpumin, W.E. Lowry et al., 2016) [1] and focuses on the biocompatibility aspects of the hydrogel, including its stiffness and the inflammatory response of the transplanted organ. We have developed an injectable hyaluronic acid (HA)-based hydrogel for stem cell culture and transplantation, to promote brain tissue repair after stroke. This 3D biomaterial was engineered to bind bioactive signals such as adhesive motifs, as well as releasing growth factors while supporting cell growth and tissue infiltration. We used a Design of Experiment approach to create a complex matrix environment in vitro by keeping the hydrogel platform and cell type constant across conditions while systematically varying peptide motifs and growth factors. The optimized HA hydrogel promoted survival of encapsulated human induced pluripotent stem cell derived-neural progenitor cells (iPS-NPCs) after transplantation into the stroke cavity and differentially tuned transplanted cell fate through the promotion of glial, neuronal or immature/progenitor states. The highlights of this article include: (1) Data of cell and bioactive signals addition on the hydrogel mechanical properties and growth factor diffusion, (2) the use of a design of Experiment (DOE) approach (M.W. 2 Weible and T. Chan-Ling, 2007) [2] to select multi-factorial experimental conditions, and (3) Inflammatory response and cell survival after transplantation.

8.
Proc Natl Acad Sci U S A ; 113(52): E8453-E8462, 2016 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-27956620

RESUMEN

White matter stroke is a distinct stroke subtype, accounting for up to 25% of stroke and constituting the second leading cause of dementia. The biology of possible tissue repair after white matter stroke has not been determined. In a mouse stroke model, white matter ischemia causes focal damage and adjacent areas of axonal myelin disruption and gliosis. In these areas of only partial damage, local white matter progenitors respond to injury, as oligodendrocyte progenitors (OPCs) proliferate. However, OPCs fail to mature into oligodendrocytes (OLs) even in regions of demyelination with intact axons and instead divert into an astrocytic fate. Local axonal sprouting occurs, producing an increase in unmyelinated fibers in the corpus callosum. The OPC maturation block after white matter stroke is in part mediated via Nogo receptor 1 (NgR1) signaling. In both aged and young adult mice, stroke induces NgR1 ligands and down-regulates NgR1 inhibitors during the peak OPC maturation block. Nogo ligands are also induced adjacent to human white matter stroke in humans. A Nogo signaling blockade with an NgR1 antagonist administered after stroke reduces the OPC astrocytic transformation and improves poststroke oligodendrogenesis in mice. Notably, increased white matter repair in aged mice is translated into significant poststroke motor recovery, even when NgR1 blockade is provided during the chronic time points of injury. These data provide a perspective on the role of NgR1 ligand function in OPC fate in the context of a specific and common type of stroke and show that it is amenable to systemic intervention to promote recovery.


Asunto(s)
Envejecimiento , Vaina de Mielina/química , Receptor Nogo 1/metabolismo , Accidente Cerebrovascular/fisiopatología , Sustancia Blanca/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Axones/metabolismo , Encéfalo/patología , Diferenciación Celular , Enfermedades Desmielinizantes , Modelos Animales de Enfermedad , Humanos , Ligandos , Ratones , Ratones Transgénicos , Oligodendroglía/citología , Remielinización , Células Madre/citología , Rehabilitación de Accidente Cerebrovascular , Sustancia Blanca/patología
9.
Biomaterials ; 105: 145-155, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27521617

RESUMEN

Stem cell therapies have shown promise in promoting recovery in stroke but have been limited by poor cell survival and differentiation. We have developed a hyaluronic acid (HA)-based self-polymerizing hydrogel that serves as a platform for adhesion of structural motifs and a depot release for growth factors to promote transplant stem cell survival and differentiation. We took an iterative approach in optimizing the complex combination of mechanical, biochemical and biological properties of an HA cell scaffold. First, we optimized stiffness for a minimal reaction of adjacent brain to the transplant. Next hydrogel crosslinkers sensitive to matrix metalloproteinases (MMP) were incorporated as they promoted vascularization. Finally, candidate adhesion motifs and growth factors were systemically changed in vitro using a design of experiment approach to optimize stem cell survival or proliferation. The optimized HA hydrogel, tested in vivo, promoted survival of encapsulated human neural progenitor cells (iPS-NPCs) after transplantation into the stroke core and differentially tuned transplanted cell fate through the promotion of glial, neuronal or immature/progenitor states. This HA hydrogel can be tracked in vivo with MRI. A hydrogel can serve as a therapeutic adjunct in a stem cell therapy through selective control of stem cell survival and differentiation in vivo.


Asunto(s)
Encéfalo/patología , Hidrogeles/química , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/terapia , Andamios del Tejido , Animales , Encéfalo/cirugía , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Regeneración Tisular Dirigida/instrumentación , Humanos , Ácido Hialurónico/química , Masculino , Ensayo de Materiales , Ratones , Ratones Endogámicos C57BL , Trasplante de Células Madre/instrumentación , Trasplante de Células Madre/métodos , Propiedades de Superficie , Resistencia a la Tracción , Resultado del Tratamiento , Viscosidad
10.
J Vis Exp ; (109)2016 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-27023377

RESUMEN

Stroke affecting white matter accounts for up to 25% of clinical stroke presentations, occurs silently at rates that may be 5-10 fold greater, and contributes significantly to the development of vascular dementia. Few models of focal white matter stroke exist and this lack of appropriate models has hampered understanding of the neurobiologic mechanisms involved in injury response and repair after this type of stroke. The main limitation of other subcortical stroke models is that they do not focally restrict the infarct to the white matter or have primarily been validated in non-murine species. This limits the ability to apply the wide variety of murine research tools to study the neurobiology of white matter stroke. Here we present a methodology for the reliable production of a focal stroke in murine white matter using a local injection of an irreversible eNOS inhibitor. We also present several variations on the general protocol including two unique stereotactic variations, retrograde neuronal tracing, as well as fresh tissue labeling and dissection that greatly expand the potential applications of this technique. These variations allow for multiple approaches to analyze the neurobiologic effects of this common and understudied form of stroke.


Asunto(s)
Axones/patología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/toxicidad , Degeneración Nerviosa/patología , Ornitina/análogos & derivados , Accidente Cerebrovascular/patología , Sustancia Blanca/efectos de los fármacos , Animales , Axones/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Degeneración Nerviosa/inducido químicamente , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Ornitina/toxicidad , Accidente Cerebrovascular/inducido químicamente , Sustancia Blanca/patología
11.
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
12.
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
13.
Brain Res ; 1533: 141-51, 2013 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-23969196

RESUMEN

Assays on "ex vivo" sections of rat hippocampus and rat cerebral cortex, subjected to oxygen and glucose deprivation (OGD) and a three-hour reperfusion-like (RL) recovery, were performed in the presence of either GABA or the GABA(A) receptor binding site antagonist, bicuculline. Lactate dehydrogenase (LDH) and propidium iodide were used to quantify cell mortality. We also measured, using real-time quantitative polymerase chain reaction (qPCR), the early transcriptional response of a number of genes of the glutamatergic and GABAergic systems. Specifically, glial pre- and post-synaptic glutamatergic transporters (namely GLAST1a, EAAC-1, GLT-1 and VGLUT1), three GABAA receptor subunits (α1, ß2 and γ2), and the GABAergic presynaptic marker, glutamic acid decarboxylase (GAD65), were studied. Mortality assays revealed that GABAA receptor chloride channels play an important role in the neuroprotective effect of GABA in the cerebral cortex, but have a much smaller effect in the hippocampus. We also found that GABA reverses the OGD-dependent decrease in GABA(A) receptor transcript levels, as well as mRNA levels of the membrane and vesicular glutamate transporter genes. Based on the markers used, we conclude that OGD results in differential responses in the GABAergic presynaptic and postsynaptic systems.


Asunto(s)
Corteza Cerebral/metabolismo , Hipocampo/metabolismo , Fármacos Neuroprotectores/farmacología , Receptores de GABA-A/fisiología , Ácido gamma-Aminobutírico/farmacología , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Animales , Hipoxia de la Célula , Supervivencia Celular , Corteza Cerebral/patología , Glucosa/fisiología , Hipocampo/patología , Técnicas In Vitro , Masculino , Ratas , Ratas Sprague-Dawley , Reperfusión
14.
J Neurochem ; 127(5): 701-10, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23763503

RESUMEN

The unfolded protein response (UPR) in the hippocampal regions Cornu Ammonis 1 hippocampal region, Cornu Ammonis 3 hippocampal region, and dentate gyrus, as well as in the cerebral cortex of 3-month-old and 18-month-old rats were studied in a model of 15 min of global cerebral ischemia followed by 48 h of reperfusion. UPR was measured by quantifying the protein disulfide isomerase (PDI), C/EBP-homologous protein (CHOP), GRP78 and GRP94 transcripts using qPCR and the amounts of PDI and GRP78 by western blot. The study shows how the mRNA levels of these genes were similar in 3-month-old and 18-month-old sham-operated animals, but the ischemic insult elicited a noticeable increase in the expression of these genes in young animals that was scarcely appreciable in older animals. The striking increase in the mRNA levels of these genes in 3-month-old animals was abolished or even reverted by treatment with meloxicam, an anti-inflammatory agent. Western blot assays showed that the UPR was still detectable 48 h after ischemia in some of the studied areas, and provided evidence that the UPR is different between young and older animals. Western blot assays carried out in young animals also showed that meloxicam elicited different effects on the levels of PDI and GRP78 in the cerebral cortex and the hippocampus. We conclude that the UPR response to ischemic/reperfusion insult is age- and probably inflammation-dependent and could play an important role in ischemic vulnerability. The UPR appears to be strongly decreased in aged animals, suggesting a reduced ability for cell survival. In this study, we conclude that the unfolded protein response (UPR) to ischemic/reperfusion insult is age- and probably inflammation-dependent and could play an important role in ischemic vulnerability. The UPR strongly decreased in aged rats, suggesting a reduced ability for cell survival. The increase in the mRNA levels of UPR gene transcripts in 3-month-old animals was abolished or even reverted by treatment with meloxicam, an anti-inflammatory agent.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Tiazinas/farmacología , Tiazoles/farmacología , Respuesta de Proteína Desplegada/fisiología , Factores de Edad , Animales , Isquemia Encefálica/patología , Inhibidores de la Ciclooxigenasa/farmacología , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Masculino , Meloxicam , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/patología , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos
15.
Brain Res Bull ; 94: 90-7, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23458738

RESUMEN

AIMS: This study analyzes how age and inflammation modify the response of the vesicular glutamate transporters (VGLUTs), VGLUT1-3 to global brain ischemia/reperfusion (I/R) in brain areas with different I/R vulnerabilities. RESULTS: Global ischemia was induced in 3- and 18-month-old male Sprague-Dawley rats and CA1 and CA3 hippocampal areas, dentate gyrus and cerebral cortex of sham-operated and I/R animals were removed 48 h after insult. Real-time PCR analysis revealed that I/R challenge resulted in a significant decrease of the VGLUT mRNA levels in young animals. Western blot assays showed a lessened age-dependent response to the ischemic damage in VGLUT1 and VGLUT3, while VGLUT2 presented an age and structure-dependent response to challenge. The use of the anti-inflammatory agent meloxicam following challenge showed that COX2 inhibition promotes the expression of VGLUTs in both sham and injured animals, which results in a lessened response to I/R injury. CONCLUSIONS: VGLUT1 and VGLUT3 presented an age-dependent response to ischemic damage, while this VGLUT response was age both and structure-dependent. In addition, COX-2 inhibition resulted in an increase of VGLUT1 and VGLUT2 protein amounts both in sham and injured animals together with a lessening of the transporters' response to ischemia.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Isquemia Encefálica/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Tiazinas/farmacología , Tiazoles/farmacología , Proteínas de Transporte Vesicular de Glutamato/biosíntesis , Factores de Edad , Animales , Western Blotting , Modelos Animales de Enfermedad , Masculino , Meloxicam , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa
16.
Brain Res ; 1358: 11-9, 2010 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-20709031

RESUMEN

This study reports the mRNA levels of some excitatory amino acid transporters (EAATs) in response to ischemia-reperfusion (I/R) in rat hippocampus and cerebral cortex. The study was performed in 3-month-old and 18-month-old animals to analyze the possible role of age in the I/R response of these transporters. The I/R resulted in a reduced transcription of both the neuronal EAAC1 (excitatory amino acid carrier-1) and the neuronal and glial GLT-1 (glial glutamate transporter 1), while the glial GLAST1a (l-glutamate/l-aspartate transporter 1a) transcription increased following I/R. The changes observed were more striking in 3-month-old animals than in 18-month-old animals. We hypothesize that increases in the GLAST1a mRNA levels following I/R insult can be explained by increases in glial cells, while the GLT-1 response to I/R mirrors neuronal changes. GLAST1a transcription increases in 3-month-old animals support the hypothesis that this transporter would be the main mechanism for extracellular glutamate clearance after I/R. Decreases in EAAC1 and GLT-1 mRNA levels would represent either neuronal changes due to the delayed neuronal death or a putative protective down-regulation of these transporters to decrease the amount of glutamate inside the neurons, which would decrease their glutamate release. This study also reports how the treatment with the anti-inflammatory agent meloxicam attenuates the transcriptional response to I/R in 3-month-old rats and decreases the survival of the I/R-injured animals.


Asunto(s)
Envejecimiento , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 3 de Aminoácidos Excitadores/genética , Isquemia/metabolismo , ARN Mensajero/metabolismo , Reperfusión , Análisis de Varianza , Animales , Antiinflamatorios/uso terapéutico , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Encefalitis/tratamiento farmacológico , Encefalitis/etiología , Transportador 2 de Aminoácidos Excitadores/metabolismo , Transportador 3 de Aminoácidos Excitadores/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Isquemia/complicaciones , Isquemia/patología , Masculino , Meloxicam , Fosfopiruvato Hidratasa/genética , Fosfopiruvato Hidratasa/metabolismo , Ratas , Ratas Sprague-Dawley , Tiazinas/uso terapéutico , Tiazoles/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...