Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Physiol ; 602(13): 3151-3168, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38924526

RESUMEN

Aquaporin-4 (AQP4) is the main water channel in brain and is enriched in perivascular astrocyte processes abutting brain microvessels. There is a rich literature on the role of AQP4 in experimental stroke. While its role in oedema formation following middle cerebral artery occlusion (MCAO) has been studied extensively, its specific impact on infarct volume remains unclear. This study investigated the effects of total and partial AQP4 deletion on infarct volume in mice subjected to distal medial cerebral artery (dMCAO) occlusion. Compared to MCAO, this model induces smaller infarcts confined to neocortex, and less oedema. We show that AQP4 deletion significantly reduced infarct volume as assessed 1 week after dMCAO, suggesting that the role of AQP4 in stroke goes beyond its effect on oedema formation and dissolution. The reduction in infarct volume was associated with increased astrocyte reactivity in the peri-infarct areas. No significant differences were observed in the number of microglia among the genotypes. These findings provide new insights in the role of AQP4 in ischaemic injury indicating that AQP4 affects both infarct volume and astrocyte reactivity in the peri-infarct zone. KEY POINTS: Aquaporin-4 (AQP4) is the main water channel in brain and is enriched in perivascular astrocyte processes abutting microvessels. A rich literature exists on the role of AQP4 in oedema formation following middle cerebral artery occlusion (MCAO). We investigated the effects of total and partial AQP4 deletion on infarct volume in mice subjected to distal medial cerebral artery occlusion (dMCAO), a model inducing smaller infarcts confined to neocortex and less oedema compared to MCAO. AQP4 deletion significantly reduced infarct volume 1 week after dMCAO, suggesting a broader role for AQP4 in stroke beyond oedema formation. The reduction in infarct volume was associated with increased astrocyte reactivity in the peri-infarct areas, while no significant differences were observed in the number of microglia among the genotypes. These findings provide new insights into the role of AQP4 in stroke, indicating that AQP4 affects both infarct volume and astrocyte reactivity in the peri-infarct zone.


Asunto(s)
Acuaporina 4 , Astrocitos , Animales , Acuaporina 4/genética , Acuaporina 4/metabolismo , Astrocitos/metabolismo , Astrocitos/patología , Ratones , Masculino , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/genética , Ratones Noqueados , Edema Encefálico/patología , Edema Encefálico/metabolismo , Edema Encefálico/genética
2.
Glia ; 71(11): 2559-2572, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37439315

RESUMEN

Brain edema is a feared complication to disorders and insults affecting the brain. It can be fatal if the increase in intracranial pressure is sufficiently large to cause brain herniation. Moreover, accruing evidence suggests that even slight elevations of intracranial pressure have adverse effects, for instance on brain perfusion. The water channel aquaporin-4 (AQP4), densely expressed in perivascular astrocytic endfeet, plays a key role in brain edema formation. Using two-photon microscopy, we have studied AQP4-mediated swelling of astrocytes affects capillary blood flow and intracranial pressure (ICP) in unanesthetized mice using a mild brain edema model. We found improved regulation of capillary blood flow in mice devoid of AQP4, independently of the severity of ICP increase. Furthermore, we found brisk AQP4-dependent astrocytic Ca2+ signals in perivascular endfeet during edema that may play a role in the perturbed capillary blood flow dynamics. The study suggests that astrocytic endfoot swelling and pathological signaling disrupts microvascular flow regulation during brain edema formation.


Asunto(s)
Edema Encefálico , Animales , Ratones , Acuaporina 4/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Edema Encefálico/etiología , Edema Encefálico/patología , Edema
3.
Brain ; 143(8): 2576-2593, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32705145

RESUMEN

The glymphatic system, that is aquaporin 4 (AQP4) facilitated exchange of CSF with interstitial fluid (ISF), may provide a clearance pathway for protein species such as amyloid-ß and tau, which accumulate in the brain in Alzheimer's disease. Further, tau protein transference via the extracellular space, the compartment that is cleared by the glymphatic pathway, allows for its neuron-to-neuron propagation, and the regional progression of tauopathy in the disorder. The glymphatic system therefore represents an exciting new target for Alzheimer's disease. Here we aim to understand the involvement of glymphatic CSF-ISF exchange in tau pathology. First, we demonstrate impaired CSF-ISF exchange and AQP4 polarization in a mouse model of tauopathy, suggesting that this clearance pathway may have the potential to exacerbate or even induce pathogenic accumulation of tau. Subsequently, we establish the central role of AQP4 in the glymphatic clearance of tau from the brain; showing marked impaired glymphatic CSF-ISF exchange and tau protein clearance using the novel AQP4 inhibitor, TGN-020. As such, we show that this system presents as a novel druggable target for the treatment of Alzheimer's disease, and possibly other neurodegenerative diseases alike.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Acuaporina 4/metabolismo , Encéfalo/metabolismo , Sistema Glinfático/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Líquido Cefalorraquídeo/metabolismo , Modelos Animales de Enfermedad , Líquido Extracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
4.
Physiol Rev ; 93(4): 1543-62, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24137016

RESUMEN

Aquaporin-4 (AQP4) is one of the most abundant molecules in the brain and is particularly prevalent in astrocytic membranes at the blood-brain and brain-liquor interfaces. While AQP4 has been implicated in a number of pathophysiological processes, its role in brain physiology has remained elusive. Only recently has evidence accumulated to suggest that AQP4 is involved in such diverse functions as regulation of extracellular space volume, potassium buffering, cerebrospinal fluid circulation, interstitial fluid resorption, waste clearance, neuroinflammation, osmosensation, cell migration, and Ca(2+) signaling. AQP4 is also required for normal function of the retina, inner ear, and olfactory system. A review will be provided of the physiological roles of AQP4 in brain and of the growing list of data that emphasize the polarized nature of astrocytes.


Asunto(s)
Acuaporina 4/fisiología , Encéfalo/fisiología , Animales , Acuaporina 4/análisis , Acuaporina 4/química , Astrocitos/fisiología , Barrera Hematoencefálica/fisiología , Señalización del Calcio/fisiología , Homeostasis/fisiología , Humanos , Plasticidad Neuronal/fisiología
5.
Neuroimage ; 188: 515-523, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30557661

RESUMEN

There is currently a lack of non-invasive tools to assess water transport in healthy and pathological brain tissue. Aquaporin-4 (AQP4) water channels are central to many water transport mechanisms, and emerging evidence also suggests that AQP4 plays a key role in amyloid-ß (Aß) clearance, possibly via the glymphatic system. Here, we present the first non-invasive technique sensitive to AQP4 channels polarised at the blood-brain interface (BBI). We apply a multiple echo time (multi-TE) arterial spin labelling (ASL) MRI technique to the mouse brain to assess BBI water permeability via calculation of the exchange time (Texw), the time for magnetically labelled intravascular water to exchange across the BBI. We observed a 31% increase in exchange time in AQP4-deficient (Aqp4-/-) mice (452 ±â€¯90 ms) compared to their wild-type counterparts (343 ±â€¯91 ms) (p = 0.01), demonstrating the sensitivity of the technique to the lack of AQP4 water channels. More established, quantitative MRI parameters: arterial transit time (δa), cerebral blood flow (CBF) and apparent diffusion coefficient (ADC) detected no significant changes with the removal of AQP4. This clinically relevant tool may be crucial to better understand the role of AQP4 in water transport across the BBI, as well as clearance of proteins in neurodegenerative conditions such as Alzheimer's disease.


Asunto(s)
Acuaporina 4/fisiología , Transporte Biológico/fisiología , Barrera Hematoencefálica/fisiología , Agua Corporal , Sistema Glinfático/fisiología , Imagen por Resonancia Magnética/métodos , Neuroimagen/métodos , Animales , Barrera Hematoencefálica/diagnóstico por imagen , Femenino , Sistema Glinfático/diagnóstico por imagen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Marcadores de Spin
6.
Cereb Cortex ; 27(1): 24-33, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28365776

RESUMEN

Cortical spreading depression (CSD) is a phenomenon that challenges the homeostatic mechanisms on which normal brain function so critically depends. Analyzing the sequence of events in CSD holds the potential of providing new insight in the physiological processes underlying normal brain function as well as the pathophysiology of neurological conditions characterized by ionic dyshomeostasis. Here, we have studied the sequential progression of CSD in awake wild-type mice and in mice lacking aquaporin-4 (AQP4) or inositol 1,4,5-triphosphate type 2 receptor (IP3R2). By the use of a novel combination of genetically encoded sensors that a novel combination - an unprecedented temporal and spatial resolution, we show that CSD leads to brisk Ca2+ signals in astrocytes and that the duration of these Ca2+ signals is shortened in the absence of AQP4 but not in the absence of IP3R2. The decrease of the astrocytic, AQP4-dependent Ca2+ signals, coincides in time and space with a decrease in the duration of extracellular glutamate overflow but not with the initial peak of the glutamate release suggesting that in CSD, extracellular glutamate accumulation is extended through AQP4-dependent glutamate release from astrocytes. The present data point to a salient glial contribution to CSD and identify AQP4 as a new target for therapy.


Asunto(s)
Acuaporina 4/metabolismo , Astrocitos/fisiología , Depresión de Propagación Cortical/fisiología , Líquido Extracelular/metabolismo , Ácido Glutámico/metabolismo , Vigilia/fisiología , Animales , Acuaporina 4/genética , Señalización del Calcio/fisiología , Regulación hacia Abajo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
Crit Care Med ; 44(8): e702-10, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27035238

RESUMEN

OBJECTIVES: We tested the hypothesis that osmotherapy with hypertonic saline attenuates cerebral edema following experimental cardiac arrest and cardiopulmonary resuscitation by exerting its effect via the perivascular pool of aquaporin-4. We used mice with targeted disruption of the gene encoding α-syntrophin (α-Syn) that demonstrate diminished perivascular aquaporin-4 pool but retain the non-endfoot and ependymal pools. DESIGN: Laboratory animal study. SETTING: University animal research laboratory. INTERVENTIONS: Isoflurane-anesthetized adult male wild-type C57B/6 or α-Syn mice were subjected to cardiac arrest/cardiopulmonary resuscitation and treated with either a continuous IV infusion of 0.9% saline or various concentrations of hypertonic saline. Serum osmolality, regional brain water content, blood-brain barrier disruption, and aquaporin-4 protein expression were determined at 24 hours after cardiac arrest/cardiopulmonary resuscitation. MEASUREMENTS AND MAIN RESULTS: Hypertonic saline (7.5%) treatment significantly attenuated water content in the caudoputamen complex and cortex compared with 0.9% saline treatment in wild-type mice subjected to cardiac arrest/cardiopulmonary resuscitation. In contrast, in α-Syn mice subjected to cardiac arrest/cardiopulmonary resuscitation, 7.5% hypertonic saline treatment did not attenuate water content. Treatment with 7.5% hypertonic saline attenuated blood-brain barrier disruption at 24 hours following cardiac arrest/cardiopulmonary resuscitation in wild-type mice but not in α-Syn mice. Total aquaporin-4 protein expression was not different between 0.9% saline and hypertonic saline-treated wild-type mice. CONCLUSIONS: Following experimental cardiac arrest/cardiopulmonary resuscitation: 1) continuous hypertonic saline therapy maintained to achieve serum osmolality of ≈ 350 mOsm/L is beneficial for the treatment of cerebral edema; 2) perivascular pool of aquaporin-4 plays a critical role in water egress from brain; and 3) hypertonic saline attenuates blood-brain barrier disruption via perivascular aquaporin-4 pool.


Asunto(s)
Acuaporina 4/metabolismo , Edema Encefálico/etiología , Edema Encefálico/prevención & control , Paro Cardíaco/complicaciones , Solución Salina Hipertónica/farmacología , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/patología , Proteínas de Unión al Calcio/genética , Modelos Animales de Enfermedad , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/genética , Concentración Osmolar , Distribución Aleatoria , Solución Salina Hipertónica/administración & dosificación
8.
Glia ; 63(11): 2073-2091, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26119521

RESUMEN

The brain-blood interface holds the key to our understanding of how cerebral blood flow is regulated and how water and solutes are exchanged between blood and brain. The highly specialized astrocytic membranes that enwrap brain microvessels are salient constituents of the brain-blood interface. These endfoot membranes contain a distinct set of molecules that is anchored to the subendothelial basal lamina forming an endfoot-basal lamina junctional complex. Here we explore the mechanisms underpinning the formation of this complex. By use of a tailor made model system we show that endothelial cells promote AQP4 accumulation by exerting an inductive effect through extracellular matrix components such as agrin, as well as through a direct mechanical interaction with the endfoot processes. Through the compounds they secrete, the endothelial cells also increase AQP4 expression. The present data suggest that the highly specialized gliovascular interface is established through inductive processes that include both chemical and mechanical factors. GLIA 2015;63:2073-2091.

9.
Proc Natl Acad Sci U S A ; 108(6): 2563-8, 2011 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-21262839

RESUMEN

Regulatory volume decrease (RVD) is a key mechanism for volume control that serves to prevent detrimental swelling in response to hypo-osmotic stress. The molecular basis of RVD is not understood. Here we show that a complex containing aquaporin-4 (AQP4) and transient receptor potential vanilloid 4 (TRPV4) is essential for RVD in astrocytes. Astrocytes from AQP4-KO mice and astrocytes treated with TRPV4 siRNA fail to respond to hypotonic stress by increased intracellular Ca(2+) and RVD. Coimmunoprecipitation and immunohistochemistry analyses show that AQP4 and TRPV4 interact and colocalize. Functional analysis of an astrocyte-derived cell line expressing TRPV4 but not AQP4 shows that RVD and intracellular Ca(2+) response can be reconstituted by transfection with AQP4 but not with aquaporin-1. Our data indicate that astrocytes contain a TRPV4/AQP4 complex that constitutes a key element in the brain's volume homeostasis by acting as an osmosensor that couples osmotic stress to downstream signaling cascades.


Asunto(s)
Acuaporina 4/metabolismo , Astrocitos/metabolismo , Tamaño de la Célula , Canales Catiónicos TRPV/metabolismo , Animales , Acuaporina 1/genética , Acuaporina 1/metabolismo , Acuaporina 4/genética , Astrocitos/citología , Células COS , Calcio/metabolismo , Chlorocebus aethiops , Cricetinae , Humanos , Ratones , Ratones Noqueados , Presión Osmótica/fisiología , Transducción de Señal/fisiología , Canales Catiónicos TRPV/genética
10.
Proc Natl Acad Sci U S A ; 108(2): 846-51, 2011 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-21187412

RESUMEN

Aquaporin-4 (AQP4) is a primary influx route for water during brain edema formation. Here, we provide evidence that brain swelling triggers Ca(2+) signaling in astrocytes and that deletion of the Aqp4 gene markedly interferes with these events. Using in vivo two-photon imaging, we show that hypoosmotic stress (20% reduction in osmolarity) initiates astrocytic Ca(2+) spikes and that deletion of Aqp4 reduces these signals. The Ca(2+) signals are partly dependent on activation of P2 purinergic receptors, which was judged from the effects of appropriate antagonists applied to cortical slices. Supporting the involvement of purinergic signaling, osmotic stress was found to induce ATP release from cultured astrocytes in an AQP4-dependent manner. Our results suggest that AQP4 not only serves as an influx route for water but also is critical for initiating downstream signaling events that may affect and potentially exacerbate the pathological outcome in clinical conditions associated with brain edema.


Asunto(s)
Acuaporina 4/química , Acuaporina 4/genética , Astrocitos/metabolismo , Edema Encefálico/metabolismo , Calcio/metabolismo , Adenosina Trifosfato/química , Animales , Encéfalo/patología , Edema/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ósmosis , Fotones , Transducción de Señal , Agua/química
12.
Glia ; 60(6): 867-74, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22419561

RESUMEN

Little is known about the physiological roles of aquaporin-4 (AQP4) in the central nervous system. AQP4 water channels are concentrated in endfeet membranes of astrocytes but also localize to the fine astrocytic processes that abut central synapses. Based on its pattern of expression, we predicted that AQP4 could be involved in controlling water fluxes and changes in extracellular space (ECS) volume that are associated with activation of excitatory pathways. Here, we show that deletion of Aqp4 accentuated the shrinkage of the ECS that occurred in the mouse hippocampal CA1 region during activation of Schaffer collateral/commissural fibers. This effect was found in the stratum radiatum (where perisynaptic astrocytic processes abound) but not in the pyramidal cell layer (where astrocytic processes constitute but a minor volume fraction). For both genotypes the ECS shrinkage was most pronounced in the pyramidal cell layer. Our data attribute a physiological role to AQP4 and indicate that this water channel regulates extracellular volume dynamics in the mammalian brain.


Asunto(s)
Acuaporina 4/deficiencia , Astrocitos/fisiología , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/metabolismo , Potenciales Postsinápticos Excitadores/genética , Espacio Extracelular/genética , Animales , Astrocitos/ultraestructura , Fenómenos Biofísicos , Región CA1 Hipocampal/ultraestructura , Estimulación Eléctrica , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Estimulantes Ganglionares/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Técnicas de Placa-Clamp , Fosfopiruvato Hidratasa/metabolismo , Células Piramidales/fisiología , Compuestos de Amonio Cuaternario/farmacología , Sinapsis/genética , Sinapsis/ultraestructura
13.
Neurobiol Dis ; 47(3): 331-7, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22659305

RESUMEN

Increased extracellular brain glutamate has been implicated in the pathophysiology of human refractory temporal lobe epilepsy (TLE), but the cause of the excessive glutamate is unknown. Prior studies by us and others have shown that the glutamate degrading enzyme glutamine synthetase (GS) is deficient in astrocytes in the epileptogenic hippocampal formation in a subset of patients with TLE. We have postulated that the loss of GS in TLE leads to increased glutamate in astrocytes with elevated concentrations of extracellular glutamate and recurrent seizures as the ultimate end-points. Here we test the hypothesis that the deficiency in GS leads to increased glutamate in astrocytes. Rats were chronically infused with methionine sulfoximine (MSO, n=4) into the hippocampal formation to induce GS deficiency and recurrent seizures. A separate group of rats was infused with 0.9% NaCl (saline) as a control (n=6). At least 10days after the start of infusion, once recurrent seizures were established in the MSO-treated rats, the concentration of glutamate was assessed in CA1 of the hippocampal formation by immunogold electron microscopy. The concentration of glutamate was 47% higher in astrocytes in the MSO-treated vs. saline-treated rats (p=0.02), and the ratio of glutamate in astrocytes relative to axon terminals was increased by 74% in the MSO-treated rats (p=0.003). These data support our hypothesis that a deficiency in GS leads to increased glutamate in astrocytes. We additionally propose that the GS-deficient astrocytes in the hippocampal formation in TLE lead to elevated extracellular brain glutamate either through decreased clearance of extracellular glutamate or excessive release of glutamate into the extracellular space from these cells, or a combination of the two.


Asunto(s)
Astrocitos/metabolismo , Epilepsia del Lóbulo Temporal/patología , Ácido Glutámico/metabolismo , Animales , Astrocitos/ultraestructura , Ondas Encefálicas/efectos de los fármacos , Ondas Encefálicas/fisiología , Modelos Animales de Enfermedad , Estimulación Eléctrica/efectos adversos , Electroencefalografía , Epilepsia del Lóbulo Temporal/etiología , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/ultraestructura , Masculino , Metionina Sulfoximina/toxicidad , Microscopía Inmunoelectrónica , Ratas , Ratas Sprague-Dawley
14.
Front Neurol ; 12: 585189, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33841293

RESUMEN

Cognitive impairment is a common and disruptive outcome for stroke survivors, which is recognized to be notoriously difficult to treat. Previously, we have shown that low oxygen post-conditioning (LOPC) improves motor function and limits secondary neuronal loss in the thalamus after experimental stroke. There is also emerging evidence that LOPC may improve cognitive function post-stroke. In the current study we aimed to explore how exposure to LOPC may improve cognition post-stroke. Experimental stroke was induced using photothrombotic occlusion in adult, male C57BL/6 mice. At 72 h post-stroke animals were randomly assigned to either normal atmospheric air or to one of two low oxygen (11% O2) exposure groups (either 8 or 24 h/day for 14 days). Cognition was assessed during the treatment phase using a touchscreen based paired-associate learning assessment. At the end of treatment (17 days post-stroke) mice were euthanized and tissue was collected for subsequent histology and biochemical analysis. LOPC (both 8 and 24 h) enhanced learning and memory in the 2nd week post-stroke when compared with stroke animals exposed to atmospheric air. Additionally we observed LOPC was associated with lower levels of neuronal loss, the restoration of several vascular deficits, as well as a reduction in the severity of the amyloid-beta (Aß) burden. These findings provide further insight into the pro-cognitive benefits of LOPC.

15.
PLoS Comput Biol ; 5(1): e1000272, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19165313

RESUMEN

Neuronal stimulation causes approximately 30% shrinkage of the extracellular space (ECS) between neurons and surrounding astrocytes in grey and white matter under experimental conditions. Despite its possible implications for a proper understanding of basic aspects of potassium clearance and astrocyte function, the phenomenon remains unexplained. Here we present a dynamic model that accounts for current experimental data related to the shrinkage phenomenon in wild-type as well as in gene knockout individuals. We find that neuronal release of potassium and uptake of sodium during stimulation, astrocyte uptake of potassium, sodium, and chloride in passive channels, action of the Na/K/ATPase pump, and osmotically driven transport of water through the astrocyte membrane together seem sufficient for generating ECS shrinkage as such. However, when taking into account ECS and astrocyte ion concentrations observed in connection with neuronal stimulation, the actions of the Na(+)/K(+)/Cl(-) (NKCC1) and the Na(+)/HCO(3) (-) (NBC) cotransporters appear to be critical determinants for achieving observed quantitative levels of ECS shrinkage. Considering the current state of knowledge, the model framework appears sufficiently detailed and constrained to guide future key experiments and pave the way for more comprehensive astroglia-neuron interaction models for normal as well as pathophysiological situations.


Asunto(s)
Astrocitos/metabolismo , Espacio Extracelular/metabolismo , Transporte Iónico/fisiología , Modelos Biológicos , Neuronas/metabolismo , Animales , Bicarbonatos/metabolismo , Cloruros/metabolismo , Espacio Extracelular/química , Humanos , Potenciales de la Membrana/fisiología , Ósmosis/fisiología , Comunicación Paracrina/fisiología , Potasio/metabolismo , Sodio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Biología de Sistemas
16.
J Neurosci Res ; 87(6): 1310-22, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19115411

RESUMEN

AQP9 is an aquaglyceroporin that serves important functions in peripheral organs, including the liver. Reflecting the lack of AQP9 knockout mice, uncertainties still prevail regarding the localization and roles of AQP9 in the central nervous system. Here we present a comprehensive analysis of AQP9 gene expression in brain, based on a quantitative and multipronged approach that includes the use of animals with targeted deletion of the AQP9 gene. We show by real-time PCR that AQP9 mRNA concentration in rat and mouse brain is approximately 3% and approximately 0.5%, respectively, of that in rat and mouse liver, the organ with the highest level of AQP9. By blue native gel analysis it could be demonstrated that the brain contains tetrameric AQP9, corresponding to the functional form of AQP9. The band corresponding to the AQP9 tetramer was absent in AQP9 knockout brain and liver. Immunocytochemistry and in situ hybridization analyses with AQP9 knockout controls show that subpopulations of nigral neurons express AQP9 both at the mRNA and at the protein levels and that populations of cortical cells (including hilar neurons in the hippocampus) contain AQP9 mRNA but no detectable AQP9 immunosignal. The present data provide conclusive evidence for the presence of tetrameric AQP9 in brain and for the expression of AQP9 in neurons.


Asunto(s)
Acuaporinas/metabolismo , Encéfalo/metabolismo , Neuronas/metabolismo , Animales , Acuaporinas/genética , Encéfalo/ultraestructura , Expresión Génica , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estructura Cuaternaria de Proteína , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Tirosina 3-Monooxigenasa/metabolismo
17.
J Alzheimers Dis ; 16(2): 315-23, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19221421

RESUMEN

Little is known about how amyloid-beta (Abeta) is deposited in relation to the complex ultrastructure of the brain. Here we combined serial section immunoelectron microscopy with 3D reconstruction to elucidate the spatial relationship between Abeta deposits and ultrastructurally identified cellular compartments. The analysis was performed in a transgenic mouse model with mutant presenilin-1, and mutant amyloid-beta protein precursor (AbetaPP) and tau transgenes (3xTg-AD mice) and in aged dogs that develop Abeta plaques spontaneously. Reconstructions based on serial ultrathin sections of hippocampus (mice) or neocortex (dogs) that had been immunolabeled with Abeta (Abeta(1-42)) antibodies showed that the organization of extracellular Abeta deposits is more complex than anticipated from light microscopic analyses. In both species, deposits were tightly associated with plasma membranes of pyramidal cell bodies and major dendrites. The deposits typically consisted of thin sheets as well as slender tendrils that climbed along the large caliber dendritic stems of pyramidal neurons. No preferential association was observed between Abeta deposits and thin dendritic branches or spines, nor was there any evidence of preferential accumulation of Abeta around synaptic contacts or glial processes. Our data suggest that plaque formation is a precisely orchestrated process that involves specialized domains of dendrosomatic plasma membranes.


Asunto(s)
Envejecimiento/patología , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Encéfalo/patología , Membrana Celular/metabolismo , Dendritas/metabolismo , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/ultraestructura , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/ultraestructura , Membrana Celular/patología , Membrana Celular/ultraestructura , Dendritas/patología , Dendritas/ultraestructura , Modelos Animales de Enfermedad , Perros , Humanos , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión/métodos , Microscopía Inmunoelectrónica/métodos , Mutación , Fragmentos de Péptidos/ultraestructura , Presenilina-1/genética , Proteínas tau/genética
18.
PLoS One ; 13(3): e0194896, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29566083

RESUMEN

More than 90% of the cases of Parkinson's disease have unknown etiology. Gradual loss of dopaminergic neurons of substantia nigra is the main cause of morbidity in this disease. External factors such as environmental toxins are believed to play a role in the cell loss, although the cause of the selective vulnerability of dopaminergic neurons remains unknown. We have previously shown that aquaglyceroporin AQP9 is expressed in dopaminergic neurons and astrocytes of rodent brain. AQP9 is permeable to a broad spectrum of substrates including purines, pyrimidines, and lactate, in addition to water and glycerol. Here we test our hypothesis that AQP9 serves as an influx route for exogenous toxins and, hence, may contribute to the selective vulnerability of nigral dopaminergic (tyrosine hydroxylase-positive) neurons. Using Xenopus oocytes injected with Aqp9 cRNA, we show that AQP9 is permeable to the parkinsonogenic toxin 1-methyl-4-phenylpyridinium (MPP+). Stable expression of AQP9 in HEK cells increases their vulnerability to MPP+ and to arsenite-another parkinsonogenic toxin. Conversely, targeted deletion of Aqp9 in mice protects nigral dopaminergic neurons against MPP+ toxicity. A protective effect of Aqp9 deletion was demonstrated in organotypic slice cultures of mouse midbrain exposed to MPP+ in vitro and in mice subjected to intrastriatal injections of MPP+ in vivo. Seven days after intrastriatal MPP+ injections, the population of tyrosine hydroxylase-positive cells in substantia nigra is reduced by 48% in Aqp9 knockout mice compared with 67% in WT littermates. Our results show that AQP9 -selectively expressed in catecholaminergic neurons-is permeable to MPP+ and suggest that this aquaglyceroporin contributes to the selective vulnerability of nigral dopaminergic neurons by providing an entry route for parkinsonogenic toxins. To our knowledge this is the first evidence implicating a toxin permeable membrane channel in the pathophysiology of Parkinson's disease.


Asunto(s)
Acuaporinas/genética , Neuroprotección/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacocinética , Animales , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Femenino , Eliminación de Gen , Células HEK293 , Humanos , Intoxicación por MPTP/genética , Intoxicación por MPTP/metabolismo , Intoxicación por MPTP/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Fármacos Neuroprotectores/metabolismo , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/genética , Enfermedad de Parkinson Secundaria/metabolismo , Enfermedad de Parkinson Secundaria/patología , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Xenopus laevis
19.
Brain Struct Funct ; 222(9): 3959-3972, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28551776

RESUMEN

Aquaporin-4 (AQP4) is the predominant water channel in the brain and is expressed in high density in astrocytes. By fluxing water along osmotic gradients, AQP4 contributes to brain volume and ion homeostasis. Here we ask whether deletion of Aqp4 leads to upregulation of the gap junctional proteins connexin-43 (Cx43) and connexin-30 (Cx30). These molecules couple adjacent astrocytes to each other and allow water and ions to redistribute within the astrocyte syncytium. Immunogold analysis of parietal cortex and hippocampus showed that the number of gap junctions per capillary profile is increased in AQP4 knockout (AQP4 KO) mice. The most pronounced changes were observed for Cx43 in hippocampus where the number of connexin labeled gap junctions increased by 100% following AQP4 KO. Western blot analysis of whole tissue homogenates showed no change in the amount of Cx43 or Cx30 protein after AQP4 KO. However, AQP4 KO led to a significant increase in the amount of Cx43 in a Triton X-100 insoluble fraction. This fraction is associated with connexin assembly into gap junctional plaques in the plasma membrane. In line with our immunoblot data, RT-qPCR showed no significant increase in Cx43 and Cx30 mRNA levels after AQP4 KO. Our findings suggest that AQP4 KO leads to increased aggregation of Cx43 into gap junctions and provide a putative mechanistic basis for the enhanced tracer coupling in hippocampi of AQP4 KO mice. The increased number of gap junctions in AQP4 deficient mice may explain why Aqp4 deletion has rather modest effects on brain volume and K+ homeostasis.


Asunto(s)
Acuaporina 4/deficiencia , Astrocitos/metabolismo , Uniones Comunicantes/metabolismo , Regulación de la Expresión Génica/genética , Adenosina Trifosfatasas/metabolismo , Animales , Acuaporina 4/genética , Astrocitos/ultraestructura , Encéfalo/metabolismo , Encéfalo/ultraestructura , Conexina 30/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Conexinas/genética , Conexinas/metabolismo , Uniones Comunicantes/ultraestructura , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Microscopía Inmunoelectrónica
20.
FASEB J ; 19(11): 1459-67, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16126913

RESUMEN

Aquaporins are a family of water channels found in animals, plants, and microorganisms. A subfamily of aquaporins, the aquaglyceroporins, are permeable for water as well as certain solutes such as glycerol, lactate, and urea. Here we show that the brain contains two isoforms of AQP9--an aquaglyceroporin with a particularly broad substrate specificity--and that the more prevalent of these isoforms is expressed in brain mitochondria. The mitochondrial AQP9 isoform is detected as an approximately 25 kDa band in immunoblots. This isoform is likely to correspond to a new AQP9 mRNA that is obtained by alternative splicing and has a shorter ORF than the liver isoform. Subfractionation experiments and high-resolution immunogold analyses revealed that this novel AQP9 isoform is enriched in mitochondrial inner membranes. AQP9 immunopositive mitochondria occurred in astrocytes throughout the brain and in a subpopulation of neurons in the substantia nigra, ventral tegmental area, and arcuate nucleus. In the latter structures, the AQP9 immunopositive mitochondria were located in neurons that were also immunopositive for tyrosine hydroxylase, as demonstrated by double labeling immunogold electron microscopy. Our findings suggest that mitochondrial AQP9 is a hallmark of astrocytes and midbrain dopaminergic neurons. In physiological conditions, the flux of lactate and other metabolites through AQP9 may confer an advantage by allowing the mitochondria to adjust to the metabolic status of the extramitochondrial cytoplasm. We hypothesize that the complement of mitochondrial AQP9 in dopaminergic neurons may relate to the vulnerability of these neurons in Parkinson's disease.


Asunto(s)
Acuaporinas/análisis , Química Encefálica , Membranas Mitocondriales/química , Secuencia de Aminoácidos , Animales , Acuaporinas/genética , Acuaporinas/fisiología , Secuencia de Bases , Inmunohistoquímica , Masculino , Datos de Secuencia Molecular , Peso Molecular , Sistemas de Lectura Abierta , Isoformas de Proteínas , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA