Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Glia ; 71(12): 2770-2781, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37564028

ABSTRACT

Astrocytes are intricately involved in the activity of neural circuits; however, their basic physiology of interacting with nearby neurons is not well established. Using two-photon imaging of neurons and astrocytes during higher frequency stimulation of hippocampal CA3-CA1 Schaffer collateral (Scc) excitatory synapses, we could show that increasing levels of released glutamate accelerated local astrocytic Ca2+ elevation. However, blockage of glutamate transporters did not abolish this astrocytic Ca2+ response, suggesting that astrocytic Ca2+ elevation is indirectly associated with an uptake of extracellular glutamate. However, during the astrocytic glutamate uptake, the Na+ /Ca2+ exchanger (NCX) reverse mode was activated, and mediated extracellular Ca2+ entry, thereby triggering the internal release of Ca2+ . In addition, extracellular Ca2+ entry via membrane P2X receptors further facilitated astrocytic Ca2+ elevation via ATP binding. These findings suggest a novel mechanism of activity induced Ca2+ permeability increases of astrocytic membranes, which drives astrocytic responses during neuronal stimulation of CA3-CA1 Scc excitatory synapses.


Subject(s)
Astrocytes , Neurons , Astrocytes/metabolism , Neurons/metabolism , Hippocampus/metabolism , Synapses/metabolism , Glutamic Acid/metabolism , Permeability , Calcium/metabolism
2.
Glia ; 71(11): 2559-2572, 2023 11.
Article in English | MEDLINE | ID: mdl-37439315

ABSTRACT

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.


Subject(s)
Brain Edema , Animals , Mice , Aquaporin 4/metabolism , Astrocytes/metabolism , Brain/metabolism , Brain Edema/etiology , Brain Edema/pathology , Edema
3.
Brain ; 143(8): 2576-2593, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32705145

ABSTRACT

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.


Subject(s)
Alzheimer Disease/metabolism , Aquaporin 4/metabolism , Brain/metabolism , Glymphatic System/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Animals , Brain/pathology , Cerebrospinal Fluid/metabolism , Disease Models, Animal , Extracellular Fluid/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
4.
Physiol Rev ; 93(4): 1543-62, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24137016

ABSTRACT

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.


Subject(s)
Aquaporin 4/physiology , Brain/physiology , Animals , Aquaporin 4/analysis , Aquaporin 4/chemistry , Astrocytes/physiology , Blood-Brain Barrier/physiology , Calcium Signaling/physiology , Homeostasis/physiology , Humans , Neuronal Plasticity/physiology
5.
Glia ; 67(1): 91-100, 2019 01.
Article in English | MEDLINE | ID: mdl-30306658

ABSTRACT

Idiopathic normal pressure hydrocephalus (iNPH) is a subtype of dementia that may be successfully treated with cerebrospinal fluid (CSF) diversion. Recently, magnetic resonance imaging (MRI) using a MRI contrast agent as a CSF tracer revealed impaired clearance of the CSF tracer from various brain regions such as the entorhinal cortex of iNPH patients. Hampered clearance of waste solutes, for example, soluble amyloid-ß, may underlie neurodegeneration and dementia in iNPH. The goal of the present study was to explore whether iNPH is associated with altered subcellular distribution of aquaporin-4 (AQP4) water channels, which is reported to facilitate CSF circulation and paravascular glymphatic drainage of metabolites from the brain parenchyma. Cortical brain biopsies of 30 iNPH patients and 12 reference individuals were subjected to AQP4 immunogold cytochemistry. Electron microscopy revealed significantly reduced density of AQP4 water channels in astrocytic endfoot membranes along cortical microvessels in patients with iNPH versus reference subjects. There was a significant positive correlation between density of AQP4 toward endothelial cells (perivascular) and toward parenchyma, but the reduced density of AQP4 toward parenchyma was not significant in iNPH. We conclude that perivascular AQP4 expression is attenuated in iNPH, potentially contributing to impaired glymphatic circulation, and waste clearance, and subsequent neurodegeneration. Hence, restoring normal perivascular AQP4 distribution may emerge as a novel treatment strategy for iNPH.


Subject(s)
Aquaporin 4/metabolism , Astrocytes/metabolism , Glymphatic System/metabolism , Hydrocephalus, Normal Pressure/metabolism , Adult , Aged , Aged, 80 and over , Aquaporin 4/analysis , Aquaporin 4/ultrastructure , Astrocytes/chemistry , Astrocytes/ultrastructure , Cohort Studies , Female , Glymphatic System/chemistry , Glymphatic System/ultrastructure , Humans , Hydrocephalus, Normal Pressure/pathology , Male , Middle Aged , Prospective Studies
6.
Glia ; 67(12): 2399-2409, 2019 12.
Article in English | MEDLINE | ID: mdl-31350866

ABSTRACT

Astrocytic endfeet cover the brain surface and form a sheath around the cerebral vasculature. An emerging concept is that endfeet control blood-brain water transport and drainage of interstitial fluid and waste along paravascular pathways. Little is known about the signaling mechanisms that regulate endfoot volume and hence the width of these drainage pathways. Here, we used the genetically encoded fluorescent Ca2+ indicator GCaMP6f to study Ca2+ signaling within astrocytic somata, processes, and endfeet in response to an osmotic challenge known to induce cell swelling. Acute cortical slices were subjected to artificial cerebrospinal fluid with 20% reduction in osmolarity while GCaMP6f fluorescence was imaged with two-photon microscopy. Ca2+ signals induced by hypoosmotic conditions were observed in all astrocytic compartments except the soma. The Ca2+ response was most prominent in subpial and perivascular endfeet and included spikes with single peaks, plateau-type elevations, and rapid oscillations, the latter restricted to subpial endfeet. Genetic removal of the type 2 inositol 1,4,5-triphosphate receptor (IP3R2) severely suppressed the Ca2+ responses in endfeet but failed to affect brain water accumulation in vivo after water intoxication. Furthermore, the increase in endfoot Ca2+ spike rate during hypoosmotic conditions was attenuated in mutant mice lacking the aquaporin-4 anchoring molecule dystrophin and after blockage of transient receptor potential vanilloid 4 channels. We conclude that the characteristics and underpinning of Ca2+ responses to hypoosmotic stress differ within the astrocytic territory and that IP3R2 is essential for the Ca2+ signals only in subpial and perivascular endfeet.


Subject(s)
Astrocytes/metabolism , Brain Edema/metabolism , Calcium Signaling/physiology , Cerebral Cortex/metabolism , Osmosis/physiology , Animals , Astrocytes/pathology , Brain Edema/pathology , Cerebral Cortex/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques
7.
Glia ; 67(6): 1113-1121, 2019 06.
Article in English | MEDLINE | ID: mdl-30791140

ABSTRACT

Cortical spreading depression (CSD) is a slowly propagating wave of depolarization of gray matter. This phenomenon is believed to underlie the migraine aura and similar waves of depolarization may exacerbate injury in a number of neurological disease states. CSD is characterized by massive ion dyshomeostasis, cell swelling, and multiphasic blood flow changes. Recently, it was shown that CSD is associated with a closure of the paravascular space (PVS), a proposed exit route for brain interstitial fluid and solutes, including excitatory and inflammatory substances that increase in the wake of CSD. The PVS closure was hypothesized to rely on swelling of astrocytic endfeet due to their high expression of aquaporin-4 (AQP4) water channels. We investigated whether CSD is associated with swelling of endfeet around penetrating arterioles in the cortex of living mice. Endfoot cross-sectional area was assessed by two-photon microscopy of mice expressing enhanced green fluorescent protein in astrocytes and related to the degree of arteriolar constriction. In anesthetized mice CSD triggered pronounced endfoot swelling that was short-lasting and coincided with the initial arteriolar constriction. Mice lacking AQP4 displayed volume changes of similar magnitude. CSD-induced endfoot swelling and arteriolar constriction also occurred in awake mice, albeit with faster kinetics than in anesthetized mice. We conclude that swelling of astrocytic endfeet is a robust event in CSD. The early onset and magnitude of the endfoot swelling is such that it may significantly delay perivascular drainage of interstitial solutes in neurological conditions where CSD plays a pathophysiological role.


Subject(s)
Aquaporin 4/deficiency , Astrocytes/metabolism , Cell Size , Cortical Spreading Depression/physiology , Visual Cortex/metabolism , Animals , Aquaporin 4/genetics , Astrocytes/pathology , Mice , Mice, Transgenic , Visual Cortex/pathology
8.
Neuroimage ; 188: 515-523, 2019 03.
Article in English | MEDLINE | ID: mdl-30557661

ABSTRACT

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.


Subject(s)
Aquaporin 4/physiology , Biological Transport/physiology , Blood-Brain Barrier/physiology , Body Water , Glymphatic System/physiology , Magnetic Resonance Imaging/methods , Neuroimaging/methods , Animals , Blood-Brain Barrier/diagnostic imaging , Female , Glymphatic System/diagnostic imaging , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spin Labels
9.
Cereb Cortex ; 28(11): 4036-4048, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30169757

ABSTRACT

Epileptic seizures are associated with increased astrocytic Ca2+ signaling, but the fine spatiotemporal kinetics of the ictal astrocyte-neuron interplay remains elusive. By using 2-photon imaging of awake head-fixed mice with chronic hippocampal windows we demonstrate that astrocytic Ca2+ signals precede neuronal Ca2+ elevations during the initial bout of kainate-induced seizures. On average, astrocytic Ca2+ elevations preceded neuronal activity in CA1 by about 8 s. In subsequent bouts of epileptic seizures, astrocytes and neurons were activated simultaneously. The initial astrocytic Ca2+ elevation was abolished in mice lacking the type 2 inositol-1,4,5-trisphosphate-receptor (Itpr2-/-). Furthermore, we found that Itpr2-/- mice exhibited 60% less epileptiform activity compared with wild-type mice when assessed by telemetric EEG monitoring. In both genotypes we also demonstrate that spreading depression waves may play a part in seizure termination. Our findings imply a role for astrocytic Ca2+ signals in ictogenesis.


Subject(s)
Astrocytes/physiology , Calcium Signaling , Epilepsy/physiopathology , Hippocampus/physiopathology , Neurons/physiology , Seizures/physiopathology , Animals , Epilepsy/chemically induced , Excitatory Amino Acid Agonists/administration & dosage , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/physiology , Kainic Acid/administration & dosage , Male , Mice, Inbred C57BL , Mice, Knockout , Seizures/chemically induced
10.
Cereb Cortex ; 27(1): 24-33, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28365776

ABSTRACT

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.


Subject(s)
Aquaporin 4/metabolism , Astrocytes/physiology , Cortical Spreading Depression/physiology , Extracellular Fluid/metabolism , Glutamic Acid/metabolism , Wakefulness/physiology , Animals , Aquaporin 4/genetics , Calcium Signaling/physiology , Down-Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Glia ; 65(6): 964-973, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28317216

ABSTRACT

Aquaporin-4 (AQP4), the predominant water channel in the brain, is expressed in astrocytes and ependymal cells. In rodents AQP4 is highly polarized to perivascular astrocytic endfeet and loss of AQP4 polarization is associated with disease. The present study was undertaken to compare the expression pattern of AQP4 in human and mouse cortical astrocytes. Cortical tissue specimens were sampled from 11 individuals undergoing neurosurgery wherein brain tissue was removed as part of the procedure, and compared with cortical tissue from 5 adult wild-type mice processed similarly. The tissue samples were immersion-fixed and prepared for AQP4 immunogold electron microscopy, allowing quantitative assessment of AQP4's subcellular distribution. In mouse we found that AQP4 water channels were prominently clustered around vessels, being 5 to 10-fold more abundant in astrocytic endfoot membranes facing the capillary endothelium than in parenchymal astrocytic membranes. In contrast, AQP4 was markedly less polarized in human astrocytes, being only two to three-fold enriched in astrocytic endfoot membranes adjacent to capillaries. The lower degree of AQP4 polarization in human subjects (1/3 of that in mice) was mainly due to higher AQP4 expression in parenchymal astrocytic membranes. We conclude that there are hitherto unrecognized species differences in AQP4 polarization toward microvessels in the cerebral cortex.


Subject(s)
Aquaporin 4/metabolism , Astrocytes/metabolism , Cerebral Cortex/blood supply , Cerebral Cortex/metabolism , Microvessels/metabolism , Adult , Aged , Animals , Astrocytes/ultrastructure , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/surgery , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cerebral Cortex/surgery , Cerebral Cortex/ultrastructure , Cohort Studies , Epilepsy/metabolism , Epilepsy/pathology , Epilepsy/surgery , Female , Humans , Immunohistochemistry , Intracranial Aneurysm/metabolism , Intracranial Aneurysm/pathology , Intracranial Aneurysm/surgery , Male , Mice, Inbred C57BL , Microscopy, Electron , Microvessels/ultrastructure , Middle Aged , Young Adult
12.
Mol Cell Neurosci ; 77: 47-52, 2016 12.
Article in English | MEDLINE | ID: mdl-27751903

ABSTRACT

There is a constitutive production of water in brain. The efflux routes of this excess water remain to be identified. We used basal brain water content as a proxy for the capacity of water exit routes. Basal brain water content was increased in mice with a complete loss of aquaporin-4 (AQP4) water channels (global Aqp4-/- mice), but not in mice with a selective removal of perivascular AQP4 or in a novel mouse line with a selective deletion of ependymal AQP4 (Foxj1-Cre:Aqp4flox/flox mice). Unique for the global Aqp4-/- mice is the loss of the AQP4 pool subjacent to the pial membrane. Our data suggest that water accumulates in brain when subpial AQP4 is missing, pointing to a critical role of this pool of water channels in brain water exit.


Subject(s)
Aquaporin 4/metabolism , Ependyma/metabolism , Animals , Aquaporin 4/genetics , Astrocytes/metabolism , Ependyma/cytology , Ependymoglial Cells/metabolism , Mice , Mice, Inbred C57BL , Water/metabolism
13.
J Neurosci ; 35(7): 3016-21, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25698739

ABSTRACT

To date, it has been difficult to reveal physiological Ca(2+) events occurring within the fine astrocytic processes of mature animals. The objective of the study was to explore whether neuronal activity evokes astrocytic Ca(2+) signals at glutamatergic synapses of adult mice. We stimulated the Schaffer collateral/commissural fibers in acute hippocampal slices from adult mice transduced with the genetically encoded Ca(2+) indicator GCaMP5E driven by the glial fibrillary acidic protein promoter. Two-photon imaging revealed global stimulation-evoked astrocytic Ca(2+) signals with distinct latencies, rise rates, and amplitudes in fine processes and somata. Specifically, the Ca(2+) signals in the processes were faster and of higher amplitude than those in the somata. A combination of P2 purinergic and group I/II metabotropic glutamate receptor (mGluR) antagonists reduced the amplitude of the Ca(2+) transients by 30-40% in both astrocytic compartments. Blockage of the mGluRs alone only modestly reduced the magnitude of the stimulation-evoked Ca(2+) signals in processes and failed to affect the somatic Ca(2+) response. Local application of group I or I/II mGluR agonists or adenosine triphosphate (ATP) elicited global astrocytic Ca(2+) signals that mimicked the stimulation-evoked astrocytic Ca(2+) responses. We conclude that stimulation-evoked Ca(2+) signals in astrocytic processes at CA3-CA1 synapses of adult mice (1) differ from those in astrocytic somata and (2) are modulated by glutamate and ATP.


Subject(s)
Adenosine Triphosphate/pharmacology , Astrocytes/metabolism , Calcium Signaling/drug effects , Glutamic Acid/pharmacology , Hippocampus/cytology , Synapses/drug effects , Animals , Astrocytes/drug effects , Calcium/metabolism , Calcium Signaling/physiology , Calmodulin/genetics , Calmodulin/metabolism , Dioxolanes/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Humans , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenylacetates/pharmacology , Purines/pharmacology , Synapses/physiology , Synapsins/genetics , Synapsins/metabolism , Time Factors
14.
Cereb Cortex ; 25(11): 4469-76, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25840424

ABSTRACT

Cortical spreading depression is a slowly propagating wave of near-complete depolarization of brain cells followed by temporary suppression of neuronal activity. Accumulating evidence indicates that cortical spreading depression underlies the migraine aura and that similar waves promote tissue damage in stroke, trauma, and hemorrhage. Cortical spreading depression is characterized by neuronal swelling, profound elevation of extracellular potassium and glutamate, multiphasic blood flow changes, and drop in tissue oxygen tension. The slow speed of the cortical spreading depression wave implies that it is mediated by diffusion of a chemical substance, yet the identity of this substance and the pathway it follows are unknown. Intercellular spread between gap junction-coupled neurons or glial cells and interstitial diffusion of K(+) or glutamate have been proposed. Here we use extracellular direct current potential recordings, K(+)-sensitive microelectrodes, and 2-photon imaging with ultrasensitive Ca(2+) and glutamate fluorescent probes to elucidate the spatiotemporal dynamics of ionic shifts associated with the propagation of cortical spreading depression in the visual cortex of adult living mice. Our data argue against intercellular spread of Ca(2+) carrying the cortical spreading depression wavefront and are in favor of interstitial K(+) diffusion, rather than glutamate diffusion, as the leading event in cortical spreading depression.


Subject(s)
Cerebral Cortex/physiology , Cortical Spreading Depression/physiology , Ions/metabolism , Neurons/physiology , Nonlinear Dynamics , Analysis of Variance , Animals , Cortical Spreading Depression/drug effects , Dose-Response Relationship, Drug , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/pharmacology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Patch-Clamp Techniques , Potassium/pharmacology , Synapsins/genetics , Synapsins/metabolism , Transduction, Genetic
15.
Proc Natl Acad Sci U S A ; 108(2): 846-51, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21187412

ABSTRACT

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.


Subject(s)
Aquaporin 4/chemistry , Aquaporin 4/genetics , Astrocytes/metabolism , Brain Edema/metabolism , Calcium/metabolism , Adenosine Triphosphate/chemistry , Animals , Brain/pathology , Edema/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osmosis , Photons , Signal Transduction , Water/chemistry
16.
Fluids Barriers CNS ; 21(1): 28, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38532513

ABSTRACT

Waste from the brain has been shown to be cleared via the perivascular spaces through the so-called glymphatic system. According to this model the cerebrospinal fluid (CSF) enters the brain in perivascular spaces of arteries, crosses the astrocyte endfoot layer, flows through the parenchyma collecting waste that is subsequently drained along veins. Glymphatic clearance is dependent on astrocytic aquaporin-4 (AQP4) water channels that are highly enriched in the endfeet. Even though the polarized expression of AQP4 in endfeet is thought to be of crucial importance for glymphatic CSF influx, its role in extracellular solute clearance has only been evaluated using non-quantitative fluorescence measurements. Here we have quantitatively evaluated clearance of intrastriatally infused small and large radioactively labeled solutes in mice lacking AQP4 (Aqp4-/-) or lacking the endfoot pool of AQP4 (Snta1-/-). We confirm that Aqp4-/- mice show reduced clearance of both small and large extracellular solutes. Moreover, we find that the Snta1-/- mice have reduced clearance only for the 500 kDa [3H]dextran, but not 0.18 kDa [3H]mannitol suggesting that polarization of AQP4 to the endfeet is primarily important for clearance of large, but not small molecules. Lastly, we observed that clearance of 500 kDa [3H]dextran increased with age in adult mice. Based on our quantitative measurements, we confirm that presence of AQP4 is important for clearance of extracellular solutes, while the perivascular AQP4 localization seems to have a greater impact on clearance of large versus small molecules.


MAIN POINTS: Solute clearance is reduced in mice lacking AQP4 Polarization of AQP4 to the endfeet may have a greater impact on clearance of large versus small molecules Clearance of large but not small solutes is correlated with age within adult age.


Subject(s)
Dextrans , Glymphatic System , Animals , Mice , Aquaporin 4/metabolism , Astrocytes/metabolism , Brain/metabolism , Dextrans/metabolism , Glymphatic System/metabolism
17.
Glia ; 60(8): 1203-14, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22378467

ABSTRACT

Recent studies have implicated glial cells in modulation of synaptic transmission, so it is plausible that glial cells may have a functional role in the hyperexcitability characteristic of epilepsy. Indeed, alterations in distinct astrocyte membrane channels, receptors, and transporters have all been associated with the epileptic state. This review focuses on the potential roles of the glial water channel aquaporin-4 (AQP4) in modulation of brain excitability and in epilepsy. We will review studies of mice lacking AQP4 (Aqp4(-/-) mice) or α-syntrophin (an AQP4 anchoring protein) and discuss the available human studies demonstrating alterations of AQP4 in human epilepsy tissue specimens. We will conclude with new studies of AQP4 regulation and discuss the potential role of AQP4 in the development of epilepsy (epileptogenesis). While many questions remain unanswered, the available data indicate that AQP4 and its molecular partners may represent important new therapeutic targets.


Subject(s)
Aquaporin 4/metabolism , Epilepsy/metabolism , Epilepsy/pathology , Neuroglia/metabolism , Animals , Aquaporin 4/genetics , Disease Models, Animal , Humans , Mice , Mice, Transgenic
18.
Glia ; 60(6): 867-74, 2012 May.
Article in English | MEDLINE | ID: mdl-22419561

ABSTRACT

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.


Subject(s)
Aquaporin 4/deficiency , Astrocytes/physiology , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , Excitatory Postsynaptic Potentials/genetics , Extracellular Space/genetics , Animals , Astrocytes/ultrastructure , Biophysical Phenomena , CA1 Region, Hippocampal/ultrastructure , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Ganglionic Stimulants/pharmacology , Glial Fibrillary Acidic Protein/metabolism , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Patch-Clamp Techniques , Phosphopyruvate Hydratase/metabolism , Pyramidal Cells/physiology , Quaternary Ammonium Compounds/pharmacology , Synapses/genetics , Synapses/ultrastructure
19.
Glia ; 60(12): 2018-26, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22987438

ABSTRACT

Key roles of macroglia are inextricably coupled to specialized membrane domains. The perivascular endfoot membrane has drawn particular attention, as this domain contains a unique complement of aquaporin-4 (AQP4) and other channel proteins that distinguishes it from perisynaptic membranes. Recent studies indicate that the polarization of macroglia is lost in a number of diseases, including temporal lobe epilepsy and Alzheimer's disease. A better understanding is required of the molecular underpinning of astroglial polarization, particularly when it comes to the significance of the dystrophin associated protein complex (DAPC). Here, we employ immunofluorescence and immunogold cytochemistry to analyze the molecular scaffolding in perivascular endfeet in macroglia of retina and three regions of brain (cortex, dentate gyrus, and cerebellum), using AQP4 as a marker. Compared with brain astrocytes, Müller cells (a class of retinal macroglia) exhibit lower densities of the scaffold proteins dystrophin and α-syntrophin (a DAPC protein), but higher levels of AQP4. In agreement, depletion of dystrophin or α-syntrophin--while causing a dramatic loss of AQP4 from endfoot membranes of brain astrocytes--had only modest or insignificant effect, respectively, on the AQP4 pool in endfoot membranes of Müller cells. In addition, while polarization of brain macroglia was less affected by dystrophin depletion than by targeted deletion of α-syntrophin, the reverse was true for retinal macroglia. These data indicate that the molecular scaffolding in perivascular endfeet is more complex than previously assumed and that macroglia are heterogeneous with respect to the mechanisms that dictate their polarization.


Subject(s)
Astrocytes/metabolism , Brain Chemistry/genetics , Brain/metabolism , Cell Polarity/genetics , Neuroglia/metabolism , Retina/metabolism , Animals , Aquaporin 4/metabolism , Astrocytes/chemistry , Astrocytes/ultrastructure , Brain/ultrastructure , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Dystrophin/metabolism , Dystrophin-Associated Proteins/biosynthesis , Dystrophin-Associated Proteins/deficiency , Dystrophin-Associated Proteins/genetics , Immunohistochemistry , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Knockout , Mice, Transgenic , Muscle Proteins/deficiency , Muscle Proteins/genetics , Muscle Proteins/metabolism , Neuroglia/chemistry , Neuroglia/ultrastructure , Retina/chemistry , Retina/ultrastructure
20.
Glia ; 60(3): 432-40, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22131281

ABSTRACT

Expression of the water channel aquaporin-4 (AQP4) at the blood-brain interface is dependent upon the dystrophin associated protein complex. Here we investigated whether deletion of the Aqp4 gene affects the molecular composition of this protein scaffold and the integrity of the blood-brain barrier. High-resolution immunogold cytochemistry revealed that perivascular expression of α-syntrophin was reduced by 60% in Aqp4(-/-) mice. Additionally, perivascular AQP4 expression was reduced by 88% in α-syn(-/-) mice, in accordance with earlier reports. Immunofluorescence showed that Aqp4 deletion also caused a modest reduction in perivascular dystrophin, whereas ß-dystroglycan labeling was unaltered. Perivascular microglia were devoid of AQP4 immunoreactivity. Deletion of Aqp4 did not alter the ultrastructure of capillary endothelial cells, the expression of tight junction proteins (claudin-5, occludin, and zonula occludens 1), or the vascular permeability to horseradish peroxidase and Evans blue albumin dye. We conclude that Aqp4 deletion reduces the expression of perivascular glial scaffolding proteins without affecting the endothelial barrier. Our data also indicate that AQP4 and α-syntrophin are mutually dependent upon each other for proper perivascular expression.


Subject(s)
Aquaporin 4/deficiency , Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Endothelium/metabolism , Glial Fibrillary Acidic Protein/metabolism , Neuroglia/metabolism , Animals , Aquaporin 4/genetics , Blood Vessels/metabolism , Blood Vessels/ultrastructure , Calcium-Binding Proteins/metabolism , Capillary Permeability/genetics , Cerebral Cortex/cytology , Endothelium/ultrastructure , Evans Blue , Gene Expression Regulation/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Microscopy, Immunoelectron , Muscle Proteins/metabolism , Neuroglia/ultrastructure , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL