101
|
Haj-Yasein NN, Bugge CE, Jensen V, Østby I, Ottersen OP, Hvalby Ø, Nagelhus EA. Deletion of aquaporin-4 increases extracellular K(+) concentration during synaptic stimulation in mouse hippocampus. Brain Struct Funct 2014; 220:2469-74. [PMID: 24744149 PMCID: PMC4481334 DOI: 10.1007/s00429-014-0767-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 03/28/2014] [Indexed: 11/28/2022]
Abstract
The coupling between the water channel aquaporin-4 (AQP4) and K+ transport has attracted much interest. In this study, we assessed the effect of Aqp4 deletion on activity-induced [K+]o changes in acute slices from hippocampus and corpus callosum of adult mice. We show that Aqp4 deletion has a layer-specific effect on [K+]o that precisely mirrors the known effect on extracellular volume dynamics. In CA1, the peak [K+]o in stratum radiatum during 20 Hz stimulation of Schaffer collateral/commissural fibers was significantly higher in Aqp4−/− mice than in wild types, whereas no differences were observed throughout the [K+]o recovery phase. In stratum pyramidale and corpus callosum, neither peak [K+]o nor post-stimulus [K+]o recovery was affected by Aqp4 deletion. Our data suggest that AQP4 modulates [K+]o during synaptic stimulation through its effect on extracellular space volume.
Collapse
Affiliation(s)
- Nadia Nabil Haj-Yasein
- Letten Centre, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| | | | | | | | | | | | | |
Collapse
|
102
|
Abstract
The aquaporins (AQPs) are a family of small, integral membrane proteins that facilitate water transport across the plasma membranes of cells in response to osmotic gradients. Data from knockout mice support the involvement of AQPs in epithelial fluid secretion, cell migration, brain oedema and adipocyte metabolism, which suggests that modulation of AQP function or expression could have therapeutic potential in oedema, cancer, obesity, brain injury, glaucoma and several other conditions. Moreover, loss-of-function mutations in human AQPs cause congenital cataracts (AQP0) and nephrogenic diabetes insipidus (AQP2), and autoantibodies against AQP4 cause the autoimmune demyelinating disease neuromyelitis optica. Although some potential AQP modulators have been identified, challenges associated with the development of better modulators include the druggability of the target and the suitability of the assay methods used to identify modulators.
Collapse
|
103
|
Heuser K, Szokol K, Taubøll E. The role of glial cells in epilepsy. TIDSSKRIFT FOR DEN NORSKE LEGEFORENING 2014; 134:37-41. [PMID: 24429754 DOI: 10.4045/tidsskr.12.1344] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND Brain research in the last century was mainly directed at neurons, with the role of glia assumed to be limited to repair, supplying nutrients and above all acting as a packing material between neurons. In recent years, the importance of glial cells for normal brain function has been recognised. This article summarizes knowledge of glial cells of relevance to epilepsy. METHOD The article is based on a literature search in PubMed as well as the authors' clinical and research experience. RESULTS Astrocytes are the largest subgroup of glial cells and, in common with neurons, have diverse membrane transporters, ion channels and receptors. Among the most important roles of astrocytes are the uptake and redistribution of ions and water, glucose metabolism and communication with nerve cells. Disturbances in all of these functions have been associated with epilepsy. INTERPRETATION Epilepsy has previously been regarded as exclusively a disturbance in the functioning of neurons and especially of their contact points, the synapses. The mechanisms of action of today's anti-epileptic drugs are therefore primarily directed at neuronal channels and receptors. New knowledge of the role played by glial cells could increase our understanding of how epilepsy arises and could lead to new treatment strategies.
Collapse
|
104
|
Sahoo S, Aurich MK, Jonsson JJ, Thiele I. Membrane transporters in a human genome-scale metabolic knowledgebase and their implications for disease. Front Physiol 2014; 5:91. [PMID: 24653705 PMCID: PMC3949408 DOI: 10.3389/fphys.2014.00091] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 02/17/2014] [Indexed: 01/18/2023] Open
Abstract
Membrane transporters enable efficient cellular metabolism, aid in nutrient sensing, and have been associated with various diseases, such as obesity and cancer. Genome-scale metabolic network reconstructions capture genomic, physiological, and biochemical knowledge of a target organism, along with a detailed representation of the cellular metabolite transport mechanisms. Since the first reconstruction of human metabolism, Recon 1, published in 2007, progress has been made in the field of metabolite transport. Recently, we published an updated reconstruction, Recon 2, which significantly improved the metabolic coverage and functionality. Human metabolic reconstructions have been used to investigate the role of metabolism in disease and to predict biomarkers and drug targets. Given the importance of cellular transport systems in understanding human metabolism in health and disease, we analyzed the coverage of transport systems for various metabolite classes in Recon 2. We will review the current knowledge on transporters (i.e., their preferred substrates, transport mechanisms, metabolic relevance, and disease association for each metabolite class). We will assess missing coverage and propose modifications and additions through a transport module that is functional when combined with Recon 2. This information will be valuable for further refinements. These data will also provide starting points for further experiments by highlighting areas of incomplete knowledge. This review represents the first comprehensive overview of the transporters involved in central metabolism and their transport mechanisms, thus serving as a compendium of metabolite transporters specific for human metabolic reconstructions.
Collapse
Affiliation(s)
- Swagatika Sahoo
- Center for Systems Biology, University of Iceland Reykjavik, Iceland ; Molecular Systems Physiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg Belval, Luxembourg
| | - Maike K Aurich
- Center for Systems Biology, University of Iceland Reykjavik, Iceland ; Molecular Systems Physiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg Belval, Luxembourg
| | - Jon J Jonsson
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Iceland Reykjavik, Iceland ; Department of Genetics and Molecular Medicine, Landspitali, National University Hospital of Iceland Reykjavik, Iceland
| | - Ines Thiele
- Center for Systems Biology, University of Iceland Reykjavik, Iceland ; Molecular Systems Physiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg Belval, Luxembourg
| |
Collapse
|
105
|
Smith AJ, Jin BJ, Ratelade J, Verkman AS. Aggregation state determines the localization and function of M1- and M23-aquaporin-4 in astrocytes. ACTA ACUST UNITED AC 2014; 204:559-73. [PMID: 24515349 PMCID: PMC3926963 DOI: 10.1083/jcb.201308118] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
An aggregation state–dependent mechanism for segregation of plasma membrane protein complexes confers specific functional roles to the M1 and M23 isoforms of the water channel AQP4. The astrocyte water channel aquaporin-4 (AQP4) is expressed as heterotetramers of M1 and M23 isoforms in which the presence of M23–AQP4 promotes formation of large macromolecular aggregates termed orthogonal arrays. Here, we demonstrate that the AQP4 aggregation state determines its subcellular localization and cellular functions. Individually expressed M1–AQP4 was freely mobile in the plasma membrane and could diffuse into rapidly extending lamellipodial regions to support cell migration. In contrast, M23–AQP4 formed large arrays that did not diffuse rapidly enough to enter lamellipodia and instead stably bound adhesion complexes and polarized to astrocyte end-feet in vivo. Co-expressed M1– and M23–AQP4 formed aggregates of variable size that segregated due to diffusional sieving of small, mobile M1–AQP4-enriched arrays into lamellipodia and preferential interaction of large, M23–AQP4-enriched arrays with the extracellular matrix. Our results therefore demonstrate an aggregation state–dependent mechanism for segregation of plasma membrane protein complexes that confers specific functional roles to M1– and M23–AQP4.
Collapse
Affiliation(s)
- Alex J Smith
- Departments of Medicine and Physiology, University of California, San Francisco, San Francisco, CA 94143
| | | | | | | |
Collapse
|
106
|
Hauser KF, Knapp PE. Interactions of HIV and drugs of abuse: the importance of glia, neural progenitors, and host genetic factors. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 118:231-313. [PMID: 25175867 PMCID: PMC4304845 DOI: 10.1016/b978-0-12-801284-0.00009-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Considerable insight has been gained into the comorbid, interactive effects of HIV and drug abuse in the brain using experimental models. This review, which considers opiates, methamphetamine, and cocaine, emphasizes the importance of host genetics and glial plasticity in driving the pathogenic neuron remodeling underlying neuro-acquired immunodeficiency syndrome and drug abuse comorbidity. Clinical findings are less concordant than experimental work, and the response of individuals to HIV and to drug abuse can vary tremendously. Host-genetic variability is important in determining viral tropism, neuropathogenesis, drug responses, and addictive behavior. However, genetic differences alone cannot account for individual variability in the brain "connectome." Environment and experience are critical determinants in the evolution of synaptic circuitry throughout life. Neurons and glia both exercise control over determinants of synaptic plasticity that are disrupted by HIV and drug abuse. Perivascular macrophages, microglia, and to a lesser extent astroglia can harbor the infection. Uninfected bystanders, especially astroglia, propagate and amplify inflammatory signals. Drug abuse by itself derails neuronal and glial function, and the outcome of chronic exposure is maladaptive plasticity. The negative consequences of coexposure to HIV and drug abuse are determined by numerous factors including genetics, sex, age, and multidrug exposure. Glia and some neurons are generated throughout life, and their progenitors appear to be targets of HIV and opiates/psychostimulants. The chronic nature of HIV and drug abuse appears to result in sustained alterations in the maturation and fate of neural progenitors, which may affect the balance of glial populations within multiple brain regions.
Collapse
Affiliation(s)
- Kurt F Hauser
- Department of Pharmacology & Toxicology, Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia, USA.
| | - Pamela E Knapp
- Department of Pharmacology & Toxicology, Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia, USA; Department of Anatomy & Neurobiology, Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
107
|
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.
Collapse
|
108
|
Hubbard JA, Hsu MS, Fiacco TA, Binder DK. Glial cell changes in epilepsy: Overview of the clinical problem and therapeutic opportunities. Neurochem Int 2013; 63:638-51. [DOI: 10.1016/j.neuint.2013.01.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 01/14/2013] [Accepted: 01/18/2013] [Indexed: 12/20/2022]
|
109
|
Unraveling aquaporin interaction partners. Biochim Biophys Acta Gen Subj 2013; 1840:1614-23. [PMID: 24252279 DOI: 10.1016/j.bbagen.2013.11.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 11/07/2013] [Accepted: 11/12/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND Insight into protein-protein interactions (PPIs) is highly desirable in order to understand the physiology of cellular events. This understanding is one of the challenges in biochemistry and molecular biology today, especially for eukaryotic membrane proteins where hurdles of production, purification and structural determination must be passed. SCOPE OF REVIEW We have explored the common strategies used to find medically relevant interaction partners of aquaporins (AQPs). The most frequently used methods to detect direct contact, yeast two-hybrid interaction assay and co-precipitation, are described together with interactions specifically found for the selected targets AQP0, AQP2, AQP4 and AQP5. MAJOR CONCLUSIONS The vast majority of interactions involve the aquaporin C-terminus and the characteristics of the interaction partners are strikingly diverse. While the well-established methods for PPIs are robust, a novel approach like bimolecular fluorescence complementation (BiFC) is attractive for screening many conditions as well as transient interactions. The ultimate goal is structural evaluation of protein complexes in order to get mechanistic insight into how proteins communicate at a molecular level. GENERAL SIGNIFICANCE What we learn from the human aquaporin field in terms of method development and communication between proteins can be of major use for any integral membrane protein of eukaryotic origin. This article is part of a Special Issue entitled Aquaporins.
Collapse
|
110
|
Dzamba D, Honsa P, Anderova M. NMDA Receptors in Glial Cells: Pending Questions. Curr Neuropharmacol 2013; 11:250-62. [PMID: 24179462 PMCID: PMC3648778 DOI: 10.2174/1570159x11311030002] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 01/08/2013] [Accepted: 02/02/2013] [Indexed: 01/28/2023] Open
Abstract
Glutamate receptors of the N-methyl-D-aspartate (NMDA) type are involved in many cognitive processes, including behavior, learning and synaptic plasticity. For a long time NMDA receptors were thought to be the privileged domain of neurons; however, discoveries of the last 25 years have demonstrated their active role in glial cells as well. Despite the large number of studies in the field, there are many unresolved questions connected with NMDA receptors in glia that are still a matter of debate. The main objective of this review is to shed light on these controversies by summarizing results from all relevant works concerning astrocytes, oligodendrocytes and polydendrocytes (also known as NG2 glial cells) in experimental animals, further extended by studies performed on human glia. The results are divided according to the study approach to enable a better comparison of how findings obtained at the mRNA level correspond with protein expression or functionality. Furthermore, special attention is focused on the NMDA receptor subunits present in the particular glial cell types, which give them special characteristics different from those of neurons – for example, the absence of Mg2+ block and decreased Ca2+ permeability. Since glial cells are implicated in important physiological and pathophysiological roles in the central nervous system (CNS), the last part of this review provides an overview of glial NMDA receptors with respect to ischemic brain injury.
Collapse
Affiliation(s)
- David Dzamba
- Department of Cellular Neurophysiology, Institute of Experimental Medicine AS CR, Prague, Czech Republic and Second Medical Faculty, Charles University, Prague, Czech Republic
| | | | | |
Collapse
|
111
|
Transient OGG1, APE1, PARP1 and Polβ expression in an Alzheimer's disease mouse model. Mech Ageing Dev 2013; 134:467-77. [PMID: 24121118 DOI: 10.1016/j.mad.2013.09.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 09/23/2013] [Accepted: 09/28/2013] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is a disease of major public health significance, whose pathogenesis is strongly linked to the presence of fibrillar aggregates of amyloid-beta (Aβ) in the aging human brain. We exploited the transgenic (Tg)-ArcSwe mouse model for human AD to explore whether oxidative stress and the capacity to repair oxidative DNA damage via base excision repair (BER) are related to Aβ pathology in AD. Tg-ArcSwe mice express variants of Aβ, accumulating senile plaques at 4-6 months of age, and develop AD-like neuropathology as adult animals. The relative mRNA levels of genes encoding BER enzymes, including 8-oxoguanine glycosylase (OGG1), AP endonuclease 1 (APE1), polymerase β (Polβ) and poly(ADP-ribose) polymerase 1 (PARP1), were quantified in various brain regions of 6 weeks, 4 months and 12 months old mice. The results show that OGG1 transcriptional expression was higher, and APE1 expression lower, in 4 months old Tg-ArcSwe than in wildtype (wt) mice. Furthermore, Polβ transcriptional expression was significantly lower in transgenic 12 months old mice than in wt. Transcriptional profiling also showed that BER repair capacity vary during the lifespan in Tg-ArcSwe and wt mice. The BER expression pattern in Tg-ArcSwe mice thus reflects responses to oxidative stress in vulnerable brain structures.
Collapse
|
112
|
Huang J, Sun SQ, Lu WT, Xu J, Gan SW, Chen Z, Qiu GP, Huang SQ, Zhuo F, Liu Q, Xu SY. The internalization and lysosomal degradation of brain AQP4 after ischemic injury. Brain Res 2013; 1539:61-72. [PMID: 24070677 DOI: 10.1016/j.brainres.2013.09.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 08/19/2013] [Accepted: 09/18/2013] [Indexed: 11/25/2022]
Abstract
The membrane-bound water channel aquaporin-4 (AQP4) plays a significant role in maintaining brain water homeostasis. In ischemic brain, changes in the expression level of AQP4 have been reported. Previous studies suggest that the internalization of several membrane-bound proteins, including AQP4, may occur with or without lysosomal degradation. In this study, the internalization of AQP4 was detected in the ischemic rat brain via double immunofluorescence labeling. Specifically, AQP4 and early endosome antigen-1 (EEA1) co-localized after 1 h post-ischemic injury. Moreover, the co-expression of AQP4 and lysosomal-associated membrane protein-1 (LAMP1) was observed after 3 h post-ischemia. These findings suggest that AQP4 is internalized and the lysosome is involved in degrading the internalized AQP4 in the ischemic brain. AQP4 is known to be downregulated by the protein kinase C activator phorbol 12-myristate 13-acetate (PMA) in vivo and in vitro. The results in this study displayed that PMA infusion could decrease brain edema accompanied by AQP4 downregulation in ischemic brain. However, compared with vehicle infusion, PKC activator infusion did not increase the ratio of internalized or lysosomal degraded AQP4. That is, we have not found out evidence to prove protein kinase C activator PMA can promote the internalization or lysosomal degradation of AQP4 in the ischemic brain.
Collapse
Affiliation(s)
- Juan Huang
- Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, People's Republic of China; Department of Anatomy, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Popescu BFG, Bunyan RF, Guo Y, Parisi JE, Lennon VA, Lucchinetti CF. Evidence of aquaporin involvement in human central pontine myelinolysis. Acta Neuropathol Commun 2013; 1:40. [PMID: 24252214 PMCID: PMC3893459 DOI: 10.1186/2051-5960-1-40] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 07/17/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Central pontine myelinolysis (CPM) is a demyelinating disorder of the central basis pontis that is often associated with osmotic stress. The aquaporin water channels (AQPs) have been pathogenically implicated because serum osmolarity changes redistribute water and osmolytes among various central nervous system compartments. RESULTS We characterized the immunoreactivity of aquaporin-1 and aquaporin-4 (AQP1 and AQP4) and associated neuropathology in microscopic transverse sections from archival autopsied pontine tissue from 6 patients with pathologically confirmed CPM. Loss of both AQP1 and AQP4 was evident within demyelinating lesions in four of the six cases, despite the presence of glial fibrillary acidic protein (GFAP)-positive astrocytes. Lesional astrocytes were small, and exhibited fewer and shorter processes than perilesional astrocytes. In two of the six cases, astrocytes within demyelinating lesions exhibited increased AQP1 and AQP4 immunoreactivities, and gemistocytes and mitotic astrocytes were numerous. Blinded review of medical records revealed that all four cases lacking lesional AQP1 and AQP4 immunoreactivities were male, whereas the two cases with enhanced lesional AQP1 and AQP4 immunoreactivities were female. CONCLUSIONS This report is the first to establish astrocytic AQP loss in a subset of human CPM cases and suggests AQP1 and AQP4 may be involved in the pathogenesis of CPM. Further studies are required to determine whether the loss of AQP1 and AQP4 is restricted to male CPM patients, or rather may be a feature associated with specific underlying precipitants of CPM that may be more common among men. Non-rodent experimental models are needed to better clarify the complex and dynamic mechanisms involved in the regulation of AQPs in CPM, in order to determine whether it occurs secondary to the destructive disease process, or represents a compensatory mechanism protecting the astrocyte against apoptosis.
Collapse
|
114
|
Dmytrenko L, Cicanic M, Anderova M, Vorisek I, Ottersen OP, Sykova E, Vargova L. The impact of alpha-syntrophin deletion on the changes in tissue structure and extracellular diffusion associated with cell swelling under physiological and pathological conditions. PLoS One 2013; 8:e68044. [PMID: 23861848 PMCID: PMC3702576 DOI: 10.1371/journal.pone.0068044] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 05/23/2013] [Indexed: 01/08/2023] Open
Abstract
Aquaporin-4 (AQP4) is the primary cellular water channel in the brain and is abundantly expressed by astrocytes along the blood-brain barrier and brain-cerebrospinal fluid interfaces. Water transport via AQP4 contributes to the activity-dependent volume changes of the extracellular space (ECS), which affect extracellular solute concentrations and neuronal excitability. AQP4 is anchored by α-syntrophin (α-syn), the deletion of which leads to reduced AQP4 levels in perivascular and subpial membranes. We used the real-time iontophoretic method and/or diffusion-weighted magnetic resonance imaging to clarify the impact of α-syn deletion on astrocyte morphology and changes in extracellular diffusion associated with cell swelling in vitro and in vivo. In mice lacking α-syn, we found higher resting values of the apparent diffusion coefficient of water (ADCW) and the extracellular volume fraction (α). No significant differences in tortuosity (λ) or non-specific uptake (k'), were found between α-syn-negative (α-syn -/-) and α-syn-positive (α-syn +/+) mice. The deletion of α-syn resulted in a significantly smaller relative decrease in α observed during elevated K(+) (10 mM) and severe hypotonic stress (-100 mOsmol/l), but not during mild hypotonic stress (-50 mOsmol/l). After the induction of terminal ischemia/anoxia, the final values of ADCW as well as of the ECS volume fraction α indicate milder cell swelling in α-syn -/- in comparison with α-syn +/+ mice. Shortly after terminal ischemia/anoxia induction, the onset of a steep rise in the extracellular potassium concentration and an increase in λ was faster in α-syn -/- mice, but the final values did not differ between α-syn -/- and α-syn +/+ mice. This study reveals that water transport through AQP4 channels enhances and accelerates astrocyte swelling. The substantially altered ECS diffusion parameters will likely affect the movement of neuroactive substances and/or trophic factors, which in turn may modulate the extent of tissue damage and/or drug distribution.
Collapse
Affiliation(s)
- Lesia Dmytrenko
- Institute of Experimental Medicine AS CR, v.v.i., Prague, Czech Republic
| | - Michal Cicanic
- Institute of Experimental Medicine AS CR, v.v.i., Prague, Czech Republic
- Charles University, 2nd Faculty of Medicine, Prague, Czech Republic
| | - Miroslava Anderova
- Institute of Experimental Medicine AS CR, v.v.i., Prague, Czech Republic
- Charles University, 2nd Faculty of Medicine, Prague, Czech Republic
| | - Ivan Vorisek
- Institute of Experimental Medicine AS CR, v.v.i., Prague, Czech Republic
- Charles University, 2nd Faculty of Medicine, Prague, Czech Republic
| | - Ole Petter Ottersen
- Center for Molecular Biology and Neuroscience and Department of Anatomy, University of Oslo, Oslo, Norway
| | - Eva Sykova
- Institute of Experimental Medicine AS CR, v.v.i., Prague, Czech Republic
- Charles University, 2nd Faculty of Medicine, Prague, Czech Republic
| | - Lydia Vargova
- Institute of Experimental Medicine AS CR, v.v.i., Prague, Czech Republic
- Charles University, 2nd Faculty of Medicine, Prague, Czech Republic
| |
Collapse
|
115
|
Jin BJ, Zhang H, Binder DK, Verkman AS. Aquaporin-4-dependent K(+) and water transport modeled in brain extracellular space following neuroexcitation. ACTA ACUST UNITED AC 2013; 141:119-32. [PMID: 23277478 PMCID: PMC3536523 DOI: 10.1085/jgp.201210883] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Potassium (K(+)) ions released into brain extracellular space (ECS) during neuroexcitation are efficiently taken up by astrocytes. Deletion of astrocyte water channel aquaporin-4 (AQP4) in mice alters neuroexcitation by reducing ECS [K(+)] accumulation and slowing K(+) reuptake. These effects could involve AQP4-dependent: (a) K(+) permeability, (b) resting ECS volume, (c) ECS contraction during K(+) reuptake, and (d) diffusion-limited water/K(+) transport coupling. To investigate the role of these mechanisms, we compared experimental data to predictions of a model of K(+) and water uptake into astrocytes after neuronal release of K(+) into the ECS. The model computed the kinetics of ECS [K(+)] and volume, with input parameters including initial ECS volume, astrocyte K(+) conductance and water permeability, and diffusion in astrocyte cytoplasm. Numerical methods were developed to compute transport and diffusion for a nonstationary astrocyte-ECS interface. The modeling showed that mechanisms b-d, together, can predict experimentally observed impairment in K(+) reuptake from the ECS in AQP4 deficiency, as well as altered K(+) accumulation in the ECS after neuroexcitation, provided that astrocyte water permeability is sufficiently reduced in AQP4 deficiency and that solute diffusion in astrocyte cytoplasm is sufficiently low. The modeling thus provides a potential explanation for AQP4-dependent K(+)/water coupling in the ECS without requiring AQP4-dependent astrocyte K(+) permeability. Our model links the physical and ion/water transport properties of brain cells with the dynamics of neuroexcitation, and supports the conclusion that reduced AQP4-dependent water transport is responsible for defective neuroexcitation in AQP4 deficiency.
Collapse
Affiliation(s)
- Byung-Ju Jin
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | |
Collapse
|
116
|
Alvestad S, Hammer J, Hoddevik EH, Skare Ø, Sonnewald U, Amiry-Moghaddam M, Ottersen OP. Mislocalization of AQP4 precedes chronic seizures in the kainate model of temporal lobe epilepsy. Epilepsy Res 2013; 105:30-41. [DOI: 10.1016/j.eplepsyres.2013.01.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 01/04/2013] [Accepted: 01/06/2013] [Indexed: 10/27/2022]
|
117
|
Eid T, Tu N, Lee TSW, Lai JCK. Regulation of astrocyte glutamine synthetase in epilepsy. Neurochem Int 2013; 63:670-81. [PMID: 23791709 DOI: 10.1016/j.neuint.2013.06.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 05/28/2013] [Accepted: 06/08/2013] [Indexed: 11/20/2022]
Abstract
Astrocytes play a crucial role in regulating and maintaining the extracellular chemical milieu of the central nervous system under physiological conditions. Moreover, proliferation of phenotypically altered astrocytes (a.k.a. reactive astrogliosis) has been associated with many neurologic and psychiatric disorders, including mesial temporal lobe epilepsy (MTLE). Glutamine synthetase (GS), which is found in astrocytes, is the only enzyme known to date that is capable of converting glutamate and ammonia to glutamine in the mammalian brain. This reaction is important, because a continuous supply of glutamine is necessary for the synthesis of glutamate and GABA in neurons. The known stoichiometry of glutamate transport across the astrocyte plasma membrane also suggests that rapid metabolism of intracellular glutamate via GS is a prerequisite for efficient glutamate clearance from the extracellular space. Several studies have indicated that the activity of GS in astrocytes is diminished in several brain disorders, including MTLE. It has been hypothesized that the loss of GS activity in MTLE leads to increased extracellular glutamate concentrations and epileptic seizures. Understanding the mechanisms by which GS is regulated may lead to novel therapeutic approaches to MTLE, which is frequently refractory to antiepileptic drugs. This review discusses several known mechanisms by which GS expression and function are influenced, from transcriptional control to enzyme modification.
Collapse
Affiliation(s)
- Tore Eid
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, United States.
| | | | | | | |
Collapse
|
118
|
Filter gate closure inhibits ion but not water transport through potassium channels. Proc Natl Acad Sci U S A 2013; 110:10842-7. [PMID: 23754382 DOI: 10.1073/pnas.1304714110] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The selectivity filter of K(+) channels is conserved throughout all kingdoms of life. Carbonyl groups of highly conserved amino acids point toward the lumen to act as surrogates for the water molecules of K(+) hydration. Ion conductivity is abrogated if some of these carbonyl groups flip out of the lumen, which happens (i) in the process of C-type inactivation or (ii) during filter collapse in the absence of K(+). Here, we show that K(+) channels remain permeable to water, even after entering such an electrically silent conformation. We reconstituted fluorescently labeled and constitutively open mutants of the bacterial K(+) channel KcsA into lipid vesicles that were either C-type inactivating or noninactivating. Fluorescence correlation spectroscopy allowed us to count both the number of proteoliposomes and the number of protein-containing micelles after solubilization, providing the number of reconstituted channels per proteoliposome. Quantification of the per-channel increment in proteoliposome water permeability with the aid of stopped-flow experiments yielded a unitary water permeability pf of (6.9 ± 0.6) × 10(-13) cm(3)⋅s(-1) for both mutants. "Collapse" of the selectivity filter upon K(+) removal did not alter pf and was fully reversible, as demonstrated by current measurements through planar bilayers in a K(+)-containing medium to which K(+)-free proteoliposomes were fused. Water flow through KcsA is halved by 200 mM K(+) in the aqueous solution, which indicates an effective K(+) dissociation constant in that range for a singly occupied channel. This questions the widely accepted hypothesis that multiple K(+) ions in the selectivity filter act to mutually destabilize binding.
Collapse
|
119
|
Ren Z, Iliff JJ, Yang L, Yang J, Chen X, Chen MJ, Giese RN, Wang B, Shi X, Nedergaard M. 'Hit & Run' model of closed-skull traumatic brain injury (TBI) reveals complex patterns of post-traumatic AQP4 dysregulation. J Cereb Blood Flow Metab 2013; 33:834-45. [PMID: 23443171 PMCID: PMC3677112 DOI: 10.1038/jcbfm.2013.30] [Citation(s) in RCA: 240] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cerebral edema is a major contributor to morbidity associated with traumatic brain injury (TBI). The methods involved in most rodent models of TBI, including head fixation, opening of the skull, and prolonged anesthesia, likely alter TBI development and reduce secondary injury. We report the development of a closed-skull model of murine TBI, which minimizes time of anesthesia, allows the monitoring of intracranial pressure (ICP), and can be modulated to produce mild and moderate grade TBI. In this model, we characterized changes in aquaporin-4 (AQP4) expression and localization after mild and moderate TBI. We found that global AQP4 expression after TBI was generally increased; however, analysis of AQP4 localization revealed that the most prominent effect of TBI on AQP4 was the loss of polarized localization at endfoot processes of reactive astrocytes. This AQP4 dysregulation peaked at 7 days after injury and was largely indistinguishable between mild and moderate grade TBI for the first 2 weeks after injury. Within the same model, blood-brain barrieranalysis of variance permeability, cerebral edema, and ICP largely normalized within 7 days after moderate TBI. These findings suggest that changes in AQP4 expression and localization may not contribute to cerebral edema formation, but rather may represent a compensatory mechanism to facilitate its resolution.
Collapse
Affiliation(s)
- Zeguang Ren
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Seifert G, Steinhäuser C. Neuron–astrocyte signaling and epilepsy. Exp Neurol 2013; 244:4-10. [DOI: 10.1016/j.expneurol.2011.08.024] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 08/16/2011] [Accepted: 08/25/2011] [Indexed: 12/30/2022]
|
121
|
Tong J, Briggs MM, McIntosh TJ. Water permeability of aquaporin-4 channel depends on bilayer composition, thickness, and elasticity. Biophys J 2013. [PMID: 23199918 DOI: 10.1016/j.bpj.2012.09.025] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Aquaporin-4 (AQP4) is the primary water channel in the mammalian brain, particularly abundant in astrocytes, whose plasma membranes normally contain high concentrations of cholesterol. Here we test the hypothesis that the water permeabilities of two naturally occurring isoforms (AQP4-M1 and AQP4-M23) depend on bilayer mechanical/structural properties modulated by cholesterol and phospholipid composition. Osmotic stress measurements were performed with proteoliposomes containing AQP4 and three different lipid mixtures: 1), phosphatidylcholine (PC) and phosphatidylglycerol (PG); 2), PC, PG, with 40 mol % cholesterol; and 3), sphingomyelin (SM), PG, with 40 mol % cholesterol. The unit permeabilities of AQP4-M1 were 3.3 ± 0.4 × 10(-13) cm(3)/s (mean ± SE), 1.2 ± 0.1 × 10(-13) cm(3)/s, and 0.4 ± 0.1 × 10(-13) cm(3)/s in PC:PG, PC:PG:cholesterol, and SM:PG:cholesterol, respectively. The unit permeabilities of AQP4-M23 were 2.1 ± 0.2 × 10(-13) cm(3)/s, 0.8 ± 0.1 × 10(-13) cm(3)/s, and 0.3 ± 0.1 × 10(-13) cm(3)/s in PC:PG, PC:PG:cholesterol, and SM:PG:cholesterol, respectively. Thus, for each isoform the unit permeabilities strongly depended on bilayer composition and systematically decreased with increasing bilayer compressibility modulus and bilayer thickness. These observations suggest that altering lipid environment provides a means of regulating water channel permeability. Such permeability changes could have physiological consequences, because AQP4 water permeability would be reduced by its sequestration into SM:cholesterol-enriched raft microdomains. Conversely, under ischemic conditions astrocyte membrane cholesterol content decreases, which could increase AQP4 permeability.
Collapse
Affiliation(s)
- Jihong Tong
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | | | | |
Collapse
|
122
|
Abstract
The aquaporins (AQPs) are plasma membrane water-transporting proteins. AQP4 is the principal member of this protein family in the CNS, where it is expressed in astrocytes and is involved in water movement, cell migration and neuroexcitation. AQP1 is expressed in the choroid plexus, where it facilitates cerebrospinal fluid secretion, and in dorsal root ganglion neurons, where it tunes pain perception. The AQPs are potential drug targets for several neurological conditions. Astrocytoma cells strongly express AQP4, which may facilitate their infiltration into the brain, and the neuroinflammatory disease neuromyelitis optica is caused by AQP4-specific autoantibodies that produce complement-mediated astrocytic damage.
Collapse
Affiliation(s)
- Marios C Papadopoulos
- Academic Neurosurgery Unit, St. George's, University of London, Tooting, London, SW17 0RE, UK. mpapadop@sgul. ac.uk
| | | |
Collapse
|
123
|
Cruz NF, Ball KK, Froehner SC, Adams ME, Dienel GA. Regional registration of [6-(14)C]glucose metabolism during brain activation of α-syntrophin knockout mice. J Neurochem 2013; 125:247-59. [PMID: 23346911 DOI: 10.1111/jnc.12166] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 01/02/2013] [Accepted: 01/20/2013] [Indexed: 12/16/2022]
Abstract
α-Syntrophin is a component of the dystrophin scaffold-protein complex that serves as an adaptor for recruitment of key proteins to the cytoplasmic side of plasma membranes. α-Syntrophin knockout (KO) causes loss of the polarized localization of aquaporin4 (AQP4) at astrocytic endfeet and interferes with water and K(+) homeostasis. During brain activation, release of ions and metabolites from endfeet is anticipated to increase perivascular fluid osmolarity, AQP4-mediated osmotic water flow from endfeet, and metabolite washout from brain. This study tests the hypothesis that reduced levels of endfoot AQP4 increase retention of [(14)C]metabolites during sensory stimulation. Conscious KO and wild-type mice were pulse-labeled with [6-(14)C] glucose during unilateral acoustic stimulation or bilateral acoustic plus whisker stimulation, and label retention was assayed by computer-assisted brain imaging or analysis of [(14)C]metabolites in extracts, respectively. High-resolution autoradiographic assays detected a 17% side-to-side difference (p < 0.05) in inferior colliculus of KO mice, not wild-type mice. However, there were no labeling differences between KO and wild-type mice for five major HPLC fractions from four dissected regions, presumably because of insufficient anatomical resolution. The results suggest a role for AQP4-mediated water flow in support of washout of metabolites, and underscore the need for greater understanding of astrocytic water and metabolite fluxes.
Collapse
Affiliation(s)
- Nancy F Cruz
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kelly K Ball
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Stanley C Froehner
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - Marvin E Adams
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
124
|
Kleindienst A, Dunbar JG, Glisson R, Marmarou A. The role of vasopressin V1A receptors in cytotoxic brain edema formation following brain injury. Acta Neurochir (Wien) 2013. [PMID: 23188468 DOI: 10.1007/s00701-012-1558-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The hormone and neuropeptide arginine-vasopressin is designated to the maintenance of osmotic homoeostasis and blood pressure regulation. While experimental data show vasopressin V(1A) receptors to regulate aquaporin (AQP)4 water channel dependent brain water movement, the specific role in vasogenic and cytotoxic edema formation remains unclear. The present study was designed to quantify the V(1A) receptor mediated regional brain edema formation in two clinically relevant experimental models, brain injury combined with secondary insult and focal ischemia. METHODS Male Sprague-Dawley rats were randomly assigned to a continuous infusion of vehicle (1 % DMSO) or the selective non-peptide V(1A) antagonist SR49059 (83nM = 1 mg/kg) starting before controlled cortical impact (CCI) injury plus hypoxia and hypotension (HH, 30 min), or middle cerebral artery (MCA) occlusion (2 h + 2 h reperfusion). RESULTS A global analysis of brain water content by the wet/dry weight method allowed optimizing the SR49059 dosage, and demonstrated the down-regulation of brain AQP4 expression by immunoblotting. Microgravimetrical quantification in 64 one mm(3) samples per animal (n = 6 per group) from bregma +2.7 to -6.3 mm analysis demonstrated brain edema to be reduced at 4 h by SR49059 treatment in the injured and contralateral cortex following CCI + HH (p = 0.007, p < 0.001) and in the infarct area following MCA occlusion (p = 0.013, p = 0.002, p = 0.004). CONCLUSIONS Our findings demonstrate that an early cytotoxic brain edema component following brain injury plus secondary insult or focal ischemia results from a vasopressin V(1A) receptor mediated response, and occurs most likely through AQP4 up-regulation. The V(1A) antagonist SR49059 offers a new avenue in brain edema treatment and prompts further study into the role of vasopressin following brain injury.
Collapse
Affiliation(s)
- Andrea Kleindienst
- Department of Neurosurgery, Virginia Commonwealth University, 1101 E. Marshall St., 23298, Richmond, VA, USA.
| | | | | | | |
Collapse
|
125
|
Zanotto C, Abib RT, Batassini C, Tortorelli LS, Biasibetti R, Rodrigues L, Nardin P, Hansen F, Gottfried C, Leite MC, Gonçalves CA. Non-specific inhibitors of aquaporin-4 stimulate S100B secretion in acute hippocampal slices of rats. Brain Res 2013; 1491:14-22. [DOI: 10.1016/j.brainres.2012.10.065] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 10/29/2012] [Accepted: 10/31/2012] [Indexed: 10/27/2022]
|
126
|
Protective role of brain water channel AQP4 in murine cerebral malaria. Proc Natl Acad Sci U S A 2012; 110:1035-40. [PMID: 23277579 DOI: 10.1073/pnas.1220566110] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tragically common among children in sub-Saharan Africa, cerebral malaria is characterized by rapid progression to coma and death. In this study, we used a model of cerebral malaria appearing in C57BL/6 WT mice after infection with the rodent malaria parasite Plasmodium berghei ANKA. Expression and cellular localization of the brain water channel aquaporin-4 (AQP4) was investigated during the neurological syndrome. Semiquantitative real-time PCR comparing uninfected and infected mice showed a reduction of brain AQP4 transcript in cerebral malaria, and immunoblots revealed reduction of brain AQP4 protein. Reduction of brain AQP4 protein was confirmed in cerebral malaria by quantitative immunogold EM; however, polarized distribution of AQP4 at the perivascular and subpial astrocyte membranes was not altered. To further examine the role of AQP4 in cerebral malaria, WT mice and littermates genetically deficient in AQP4 were infected with P. berghei. Upon development of cerebral malaria, WT and AQP4-null mice exhibited similar increases in width of perivascular astroglial end-feet in brain. Nevertheless, the AQP4-null mice exhibited more severe signs of cerebral malaria with greater brain edema, although disruption of the blood-brain barrier was similar in both groups. In longitudinal studies, cerebral malaria appeared nearly 1 d earlier in the AQP4-null mice, and reduced survival was noted when chloroquine rescue was attempted. We conclude that the water channel AQP4 confers partial protection against cerebral malaria.
Collapse
|
127
|
Florence CM, Baillie LD, Mulligan SJ. Dynamic volume changes in astrocytes are an intrinsic phenomenon mediated by bicarbonate ion flux. PLoS One 2012; 7:e51124. [PMID: 23226475 PMCID: PMC3511399 DOI: 10.1371/journal.pone.0051124] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 10/30/2012] [Indexed: 11/18/2022] Open
Abstract
Astrocytes, the major type of non-neuronal cells in the brain, play an important functional role in extracellular potassium ([K(+)](o)) and pH homeostasis. Pathological brain states that result in [K(+)](o) and pH dysregulation have been shown to cause astrocyte swelling. However, whether astrocyte volume changes occur under physiological conditions is not known. In this study we used two-photon imaging to visualize real-time astrocyte volume changes in the stratum radiatum of the hippocampus CA1 region. Astrocytes were observed to swell by 19.0±0.9% in response to a small physiological increase in the concentration of [K(+)](o) (3 mM). Astrocyte swelling was mediated by the influx of bicarbonate (HCO(3-)) ions as swelling was significantly decreased when the influx of HCO(3-) was reduced. We found: 1) in HCO(3-) free extracellular solution astrocytes swelled by 5.4±0.7%, 2) when the activity of the sodium-bicarbonate cotransporter (NBC) was blocked the astrocytes swelled by 8.3±0.7%, and 3) in the presence of an extracellular carbonic anhydrase (CA) inhibitor astrocytes swelled by 11.4±0.6%. Because a significant HCO(3-) efflux is known to occur through the γ-amino-butyric acid (GABA) channel, we performed a series of experiments to determine if astrocytes were capable of HCO(3-) mediated volume shrinkage with GABA channel activation. Astrocytes were found to shrink -7.7±0.5% of control in response to the GABA(A) channel agonist muscimol. Astrocyte shrinkage from GABA(A) channel activation was significantly decreased to -5.0±0.6% of control in the presence of the membrane-permeant CA inhibitor acetazolamide (ACTZ). These dynamic astrocyte volume changes may represent a previously unappreciated yet fundamental mechanism by which astrocytes regulate physiological brain functioning.
Collapse
Affiliation(s)
- Clare M. Florence
- Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Landon D. Baillie
- Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Sean J. Mulligan
- Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
128
|
Loss of perivascular Kir4.1 potassium channels in the sclerotic hippocampus of patients with mesial temporal lobe epilepsy. J Neuropathol Exp Neurol 2012; 71:814-25. [PMID: 22878665 DOI: 10.1097/nen.0b013e318267b5af] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Recent experimental data in mice have shown that the inwardly rectifying K channel Kir4.1 mediates K spatial buffering in the hippocampus. Here we used immunohistochemistry to examine the distribution of Kir4.1 in hippocampi from patients with medication-refractory temporal lobe epilepsy. The selectivity of the antibody was confirmed in mice with a glial conditional deletion of the gene encoding Kir4.1. These mice showed a complete loss of labeled cells, indicating that Kir4.1 is restricted to glia. In human cases, Kir4.1 immunoreactivity observed in cells morphologically consistent with astrocytes was significantly reduced in 12 patients with hippocampal sclerosis versus 11 patients without sclerosis and 4 normal autopsy controls. Loss of astrocytic Kir4.1 immunoreactivity was most pronounced around vessels and was restricted to gliotic areas. Loss of Kir4.1 expression was associated with loss of dystrophin and α-syntrophin, but not with loss of β-dystroglycan, suggesting partial disruption of the dystrophin-associated protein complex. The changes identified in patients with hippocampal sclerosis likely interfere with K homeostasis and may contribute to the epileptogenicity of the sclerotic hippocampus.
Collapse
|
129
|
Aquaporins in drug discovery and pharmacotherapy. Mol Aspects Med 2012; 33:691-703. [DOI: 10.1016/j.mam.2012.01.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 01/13/2012] [Accepted: 01/15/2012] [Indexed: 11/18/2022]
|
130
|
Bataveljić D, Nikolić L, Milosević M, Todorović N, Andjus PR. Changes in the astrocytic aquaporin-4 and inwardly rectifying potassium channel expression in the brain of the amyotrophic lateral sclerosis SOD1(G93A) rat model. Glia 2012; 60:1991-2003. [PMID: 22987392 DOI: 10.1002/glia.22414] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Accepted: 08/10/2012] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease affecting upper and lower motor neurons. Dysfunction and death of motor neurons are closely related to the modified astrocytic environment. Astrocytic endfeet, lining the blood-brain barrier (BBB), are enriched in two proteins, aquaporin-4 (AQP4) and inwardly rectifying potassium channel (Kir) 4.1. Both channels are important for the maintainance of a functional BBB astrocytic lining. In this study, expression levels of AQP4 and Kir4.1 were for the first time examined in the brainstem and cortex, along with the functional properties of Kir channels in cultured cortical astrocytes of the SOD1(G93A) rat model of ALS. Western blot analysis showed increased expression of AQP4 and decreased expression of Kir4.1 in the brainstem and cortex of the ALS rat. In addition, higher immunoreactivity of AQP4 and reduced immunolabeling of Kir4.1 in facial and trigeminal nuclei as well as in the motor cortex were also observed. Particularly, the observed changes in the expression of both channels were retained in cultured astrocytes. Furthermore, whole-cell patch-clamp recordings from cultured ALS cortical astrocytes showed a significantly lower Kir current density. Importantly, the potassium uptake current in ALS astrocytes was significantly reduced at all extracellular potassium concentrations. Consequently, the Kir-specific Cs(+)- and Ba(2+)-sensitive currents were also decreased. The changes in the studied channels, notably at the upper CNS level, could underline the hampered ability of astrocytes to maintain water and potassium homeostasis, thus affecting the BBB, disturbing the neuronal microenvironment, and causing motoneuronal dysfunction and death.
Collapse
Affiliation(s)
- Danijela Bataveljić
- Center for Laser Microscopy, Faculty of Biology, University of Belgrade, Studentski trg 3, POB 52, Belgrade, Serbia
| | | | | | | | | |
Collapse
|
131
|
Volman V, Bazhenov M, Sejnowski TJ. Computational models of neuron-astrocyte interaction in epilepsy. Front Comput Neurosci 2012; 6:58. [PMID: 23060780 PMCID: PMC3459315 DOI: 10.3389/fncom.2012.00058] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Accepted: 07/23/2012] [Indexed: 01/30/2023] Open
Abstract
Astrocytes actively shape the dynamics of neurons and neuronal ensembles by affecting several aspects critical to neuronal function, such as regulating synaptic plasticity, modulating neuronal excitability, and maintaining extracellular ion balance. These pathways for astrocyte-neuron interaction can also enhance the information-processing capabilities of brains, but in other circumstances may lead the brain on the road to pathological ruin. In this article, we review the existing computational models of astrocytic involvement in epileptogenesis, focusing on their relevance to existing physiological data.
Collapse
Affiliation(s)
- Vladislav Volman
- Computational Neurobiology Laboratory, Howard Hughes Medical Institute, The Salk Institute for Biological Studies La Jolla, CA, USA ; Center for Theoretical Biological Physics, University of California at San Diego La Jolla, CA, USA ; L-3 Applied Technologies/Simulation, Engineering, and Testing San Diego, CA, USA
| | | | | |
Collapse
|
132
|
Losi G, Cammarota M, Carmignoto G. The role of astroglia in the epileptic brain. Front Pharmacol 2012; 3:132. [PMID: 22807916 PMCID: PMC3395023 DOI: 10.3389/fphar.2012.00132] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 06/23/2012] [Indexed: 11/13/2022] Open
Abstract
Epilepsies comprise a family of multifactorial neurological disorders that affect at least 50 million people worldwide. Despite a long history of neurobiological and clinical studies the mechanisms that lead the brain network to a hyperexcitable state and to the intense, massive neuronal discharges reflecting a seizure episode are only partially defined. Most epilepsies of genetic origin are related to mutations in ionic channels that cause neuronal hyperexcitability. However, idiopathic epilepsies of unclear origin represent the majority of these brain disorders. A large body of evidence suggests that in the epileptic brain neurons are not the only players. Indeed, the glial cell astrocyte is known to be morphologically and functionally altered in different types of epilepsy. Although it is unclear whether these astrocyte dysfunctions can have a causative role in epileptogenesis, the hypothesis that astrocytes contribute to epileptiform activities recently received a considerable experimental support. Notably, currently used antiepileptic drugs, that act mainly on neuronal ion channels, are ineffective in a large group of patients. Clarifying astrocyte functions in the epileptic brain tissue could unveil astrocytes as novel therapeutic targets. In this review we present first a short overview on the role of astrocytes in the epileptic brain starting from the "historical" observations on their fundamental modulation of brain homeostasis, such as the control of water content, ionic equilibrium, and neurotransmitters concentrations. We then focus our review on most recent studies that hint at a distinct contribution of these cells in the generation of focal epileptiform activities.
Collapse
Affiliation(s)
- Gabriele Losi
- Institute of Neuroscience of the National Research Council and Department of Biomedical Sciences, University of Padova Padova, Italy
| | | | | |
Collapse
|
133
|
Srivastava R, Aslam M, Kalluri SR, Schirmer L, Buck D, Tackenberg B, Rothhammer V, Chan A, Gold R, Berthele A, Bennett JL, Korn T, Hemmer B. Potassium channel KIR4.1 as an immune target in multiple sclerosis. N Engl J Med 2012; 367:115-23. [PMID: 22784115 PMCID: PMC5131800 DOI: 10.1056/nejmoa1110740] [Citation(s) in RCA: 253] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Multiple sclerosis is a chronic inflammatory demyelinating disease of the central nervous system. Many findings suggest that the disease has an autoimmune pathogenesis; the target of the immune response is not yet known. METHODS We screened serum IgG from persons with multiple sclerosis to identify antibodies that are capable of binding to brain tissue and observed specific binding of IgG to glial cells in a subgroup of patients. Using a proteomic approach focusing on membrane proteins, we identified the ATP-sensitive inward rectifying potassium channel KIR4.1 as the target of the IgG antibodies. We used a multifaceted validation strategy to confirm KIR4.1 as a target of the autoantibody response in multiple sclerosis and to show its potential pathogenicity in vivo. RESULTS Serum levels of antibodies to KIR4.1 were higher in persons with multiple sclerosis than in persons with other neurologic diseases and healthy donors (P<0.001 for both comparisons). We replicated this finding in two independent groups of persons with multiple sclerosis or other neurologic diseases (P<0.001 for both comparisons). Analysis of the combined data sets indicated the presence of serum antibodies to KIR4.1 in 186 of 397 persons with multiple sclerosis (46.9%), in 3 of 329 persons with other neurologic diseases (0.9%), and in none of the 59 healthy donors. These antibodies bound to the first extracellular loop of KIR4.1. Injection of KIR4.1 serum IgG into the cisternae magnae of mice led to a profound loss of KIR4.1 expression, altered expression of glial fibrillary acidic protein in astrocytes, and activation of the complement cascade at sites of KIR4.1 expression in the cerebellum. CONCLUSIONS KIR4.1 is a target of the autoantibody response in a subgroup of persons with multiple sclerosis. (Funded by the German Ministry for Education and Research and Deutsche Forschungsgemeinschaft.).
Collapse
Affiliation(s)
- Rajneesh Srivastava
- Department of Neurology, Klinikum rechts der Isar, Technische Universität, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Lucero MS, Mirarchi F, Goldstein J, Silberstein C. Intraperitoneal administration of Shiga toxin 2 induced neuronal alterations and reduced the expression levels of aquaporin 1 and aquaporin 4 in rat brain. Microb Pathog 2012; 53:87-94. [PMID: 22610042 DOI: 10.1016/j.micpath.2012.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 05/03/2012] [Accepted: 05/10/2012] [Indexed: 11/28/2022]
Abstract
Shiga toxin-producing Escherichia coli produces watery and hemorrhagic diarrhea, and hemolytic uremic syndrome (HUS) characterized by thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure. Central nervous system (CNS) complications are observed in around 30% of infant population with HUS. Common signs of severe CNS involvement leading to death include seizures, alteration of consciousness, hemiparesis, visual disturbances, and brain stem symptoms. The purpose of the present work was to study the effects of Shiga toxin 2 (Stx2) in the brain of rats intraperitoneally (i.p.) injected with a supernatant from recombinant E. coli expressing Stx2 (sStx2). Neurological alterations such as postural and motor abnormalities including lethargy, abnormal walking, and paralysis of hind legs, were observed in this experimental model of HUS in rats. Neuronal damage, as well as significant decrease in aquaporin 1 (AQP1) and aquaporin 4 (AQP4) expression levels were observed in the brain of rats, 2 days after sStx2 injection, compared to controls. Downregulation of aquaporin protein levels, and neuronal alterations, observed in brain of rats injected with sStx2, may be involved in edema formation and in neurological manifestations characteristic of HUS.
Collapse
Affiliation(s)
- María Soledad Lucero
- Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | | | | |
Collapse
|
135
|
Song Y, Gunnarson E. Potassium dependent regulation of astrocyte water permeability is mediated by cAMP signaling. PLoS One 2012; 7:e34936. [PMID: 22493723 PMCID: PMC3321040 DOI: 10.1371/journal.pone.0034936] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 03/11/2012] [Indexed: 11/18/2022] Open
Abstract
Astrocytes express potassium and water channels to support dynamic regulation of potassium homeostasis. Potassium kinetics can be modulated by aquaporin-4 (AQP4), the essential water channel for astrocyte water permeability regulation. We investigated whether extracellular potassium ([K(+)](o)) can regulate astrocyte water permeability and the mechanisms of such an effect. Studies were performed on rat primary astrocytes and a rat astrocyte cell line transfected with AQP4. We found that 10 mM [K(+)](o) caused an immediate, more than 40%, increase in astrocyte water permeability which was sustained in 5 min. The water channel AQP4 was a target for this regulation. Potassium induced a significant increase in intracellular cAMP as measured with a FRET based method and with enzyme immunoassay. We found that protein kinase A (PKA) could phosphorylate AQP4 in vitro. Further elevation of [K(+)](o) to 35 mM induced a global intracellular calcium response and a transient water permeability increase that was abolished in 5 min. When inwardly rectifying potassium (Kir)-channels were blocked, 10 mM [K(+)](o) also induced a calcium increase and the water permeability increase no longer persisted. In conclusion, we find that elevation of extracellular potassium regulates AQP4 and astrocyte water permeability via intracellular signaling involving cAMP. A prolonged increase of astrocyte water permeability is Kir-channel dependent and this response can be impeded by intracellular calcium signaling. Our results support the concept of coupling between AQP4 and potassium handling in astrocytes.
Collapse
Affiliation(s)
- Yutong Song
- Department of Women’s and Children’s Health, Karolinska Institutet, Astrid Lindgren Children’s Hospital, Stockholm, Sweden
| | - Eli Gunnarson
- Department of Women’s and Children’s Health, Karolinska Institutet, Astrid Lindgren Children’s Hospital, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
136
|
Binder DK, Nagelhus EA, Ottersen OP. Aquaporin-4 and epilepsy. Glia 2012; 60:1203-14. [DOI: 10.1002/glia.22317] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 02/09/2012] [Indexed: 12/17/2022]
|
137
|
Glial K+ Clearance and Cell Swelling: Key Roles for Cotransporters and Pumps. Neurochem Res 2012; 37:2299-309. [DOI: 10.1007/s11064-012-0731-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 01/13/2012] [Accepted: 02/11/2012] [Indexed: 10/28/2022]
|
138
|
Lee DJ, Hsu MS, Seldin MM, Arellano JL, Binder DK. Decreased expression of the glial water channel aquaporin-4 in the intrahippocampal kainic acid model of epileptogenesis. Exp Neurol 2012; 235:246-55. [PMID: 22361023 DOI: 10.1016/j.expneurol.2012.02.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 12/24/2011] [Accepted: 02/06/2012] [Indexed: 01/28/2023]
Abstract
Recent evidence suggests that astrocytes may be a potential new target for the treatment of epilepsy. The glial water channel aquaporin-4 (AQP4) is expressed in astrocytes, and along with the inwardly-rectifying K(+) channel K(ir)4.1 is thought to underlie the reuptake of H(2)O and K(+) into glial cells during neural activity. Previous studies have demonstrated increased seizure duration and slowed potassium kinetics in AQP4(-/-) mice, and redistribution of AQP4 in hippocampal specimens from patients with chronic epilepsy. However, the regulation and role of AQP4 during epileptogenesis remain to be defined. In this study, we examined the expression of AQP4 and other glial molecules (GFAP, K(ir)4.1, glutamine synthetase) in the intrahippocampal kainic acid (KA) model of epilepsy and compared behavioral and histologic outcomes in wild-type mice vs. AQP4(-/-) mice. Marked and prolonged reduction in AQP4 immunoreactivity on both astrocytic fine processes and endfeet was observed following KA status epilepticus in multiple hippocampal layers. In addition, AQP4(-/-) mice had more spontaneous recurrent seizures than wild-type mice during the first week after KA SE as assessed by chronic video-EEG monitoring and blinded EEG analysis. While both genotypes exhibited similar reactive astrocytic changes, granule cell dispersion and CA1 pyramidal neuron loss, there were an increased number of fluorojade-positive cells early after KA SE in AQP4(-/-) mice. These results indicate a marked reduction of AQP4 following KA SE and suggest that dysregulation of water and potassium homeostasis occurs during early epileptogenesis. Restoration of astrocytic water and ion homeostasis may represent a novel therapeutic strategy.
Collapse
Affiliation(s)
- Darrin J Lee
- Department of Neurological Surgery, University of California, Davis, CA, USA
| | | | | | | | | |
Collapse
|
139
|
Astrocyte dysfunction in temporal lobe epilepsy: K+ channels and gap junction coupling. Glia 2012; 60:1192-202. [DOI: 10.1002/glia.22313] [Citation(s) in RCA: 138] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 01/27/2012] [Accepted: 01/27/2012] [Indexed: 12/11/2022]
|
140
|
Cesetti T, Ciccolini F, Li Y. GABA Not Only a Neurotransmitter: Osmotic Regulation by GABA(A)R Signaling. Front Cell Neurosci 2012; 6:3. [PMID: 22319472 PMCID: PMC3268181 DOI: 10.3389/fncel.2012.00003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 01/10/2012] [Indexed: 12/05/2022] Open
Abstract
Mature macroglia and almost all neural progenitor types express γ-aminobutyric (GABA) A receptors (GABAARs), whose activation by ambient or synaptic GABA, leads to influx or efflux of chloride (Cl−) depending on its electro-chemical gradient (ECl). Since the flux of Cl− is indissolubly associated to that of osmotically obliged water, GABAARs regulate water movements by modulating ion gradients. In addition, since water movements also occur through specialized water channels and transporters, GABAAR signaling could affect the movement of water by regulating the function of the channels and transporters involved, thereby affecting not only the direction of the water fluxes but also their dynamics. We will here review recent observations indicating that in neural cells GABAAR-mediated osmotic regulation affects the cellular volume thereby activating multiple intracellular signaling mechanisms important for cell proliferation, maturation, and survival. In addition, we will discuss evidence that the osmotic regulation exerted by GABA may contribute to brain water homeostasis in physiological and in pathological conditions causing brain edema, in which the GABAergic transmission is often altered.
Collapse
Affiliation(s)
- Tiziana Cesetti
- Department of Physiology and Pathophysiology, Interdisciplinary Center for Neurosciences, University of Heidelberg Heidelberg, Germany
| | | | | |
Collapse
|
141
|
Iacovetta C, Rudloff E, Kirby R. The role of aquaporin 4 in the brain. Vet Clin Pathol 2012; 41:32-44. [PMID: 22250904 DOI: 10.1111/j.1939-165x.2011.00390.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2011] [Revised: 11/14/2011] [Accepted: 12/10/2011] [Indexed: 11/26/2022]
Abstract
Emerging evidence suggests that aquaporin (AQP) 4 water channels play an important role in water homeostasis in the brain. These water channels are most abundant in the cell membrane of astrocytes, but are also present within ependymal cell membranes and in osmosensory areas of the hypothalamus. Water transport through AQP4 depends on concentration gradients across the membrane, but the rate of transport is determined by the capacity of astrocytes to up- and down-regulate AQP4 numbers, their location within the membrane, and the overall permeability of the channel. Other functions of brain AQP4 involve potassium uptake and release by astrocytes, migration of glial cells, glial scarring, and astrocyte-to-astrocyte cell communication. AQP water channels are involved in formation and control of edema in the brain and in multiple disease processes in the brain, such as seizures and tumors. There is abundant scientific literature on AQP4 describing its structure, function, location, and role in water homeostasis and edema in the brain. Investigation of AQP expression in the canine and feline brain should be pursued so that clinically relevant comparisons between findings in mice, rats, and people and animal patients can be made.
Collapse
|
142
|
Nakamura Y, Suzuki Y, Tsujita M, Huber VJ, Yamada K, Nakada T. Development of a Novel Ligand, [C]TGN-020, for Aquaporin 4 Positron Emission Tomography Imaging. ACS Chem Neurosci 2011; 2:568-571. [PMID: 22022637 PMCID: PMC3198134 DOI: 10.1021/cn2000525] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 06/27/2011] [Indexed: 12/02/2022] Open
Abstract
![]()
Aquaporin 4 (AQP4), the most abundant isozyme of the water specific membrane transporter aquaporin family, has now been implicated to play a significant role in the pathogenesis of various disease processes of the nervous system from epilepsy to Alzheimer’s disease. Considering its clinical relevance, it is highly desirable to develop a noninvasive method for the quantitative analysis of AQP distribution in humans under clinical settings. Currently, the method of choice for such diagnostic examinations continues to be positron emission tomography (PET). Here, we report the successful development of a PET ligand for AQP4 imaging based on TGN-020, a potent AQP4 inhibitor developed previously in our laboratory. Utilizing [11C]-TGN-020, PET images were successfully generated in wild type and AQP4 null mice, providing a basis for future evaluation regarding its suitability for clinical studies.
Collapse
Affiliation(s)
- Yukihiro Nakamura
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, 1 Asahimachi, Niigata 951-8585, Japan
| | - Yuji Suzuki
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, 1 Asahimachi, Niigata 951-8585, Japan
| | - Mika Tsujita
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, 1 Asahimachi, Niigata 951-8585, Japan
| | - Vincent J. Huber
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, 1 Asahimachi, Niigata 951-8585, Japan
| | - Kenichi Yamada
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, 1 Asahimachi, Niigata 951-8585, Japan
| | - Tsutomu Nakada
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, 1 Asahimachi, Niigata 951-8585, Japan
| |
Collapse
|
143
|
Steier R, Aradi M, Pál J, Bukovics P, Perlaki G, Orsi G, Janszky J, Schwarcz A, Sulyok E, Dóczi T. The influence of benzamil hydrochloride on the evolution of hyponatremic brain edema as assessed by in vivo MRI study in rats. Acta Neurochir (Wien) 2011; 153:2091-7; discussion 2097. [PMID: 21445679 DOI: 10.1007/s00701-011-0996-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 03/09/2011] [Indexed: 11/29/2022]
Abstract
OBJECTIVE The present study was undertaken to reveal the influence of intracerebroventricular (ICV) benzamil on the dynamics of brain water accumulation in hyponatremic rats. Parameters of brain water homeostasis were continuously monitored, using in vivo magnetic resonance imaging (MRI) methods. The results were compared with those obtained in a previous study by tissue desiccation. METHODS A 3-T MRI instrument was applied to perform serial diffusion-weighted imaging to measure the apparent diffusion coefficient (ADC) and MR spectroscopy to determine water signal. A decrease of ADC is thought to represent an increase of intracellular water, whereas water signal is used to quantify brain water content. Five groups of male Wistar rats were studied as follows: normonatremic, native animals (group NN, n = 7), hyponatremic animals (group HN, n = 8), hyponatremic animals treated with ICV benzamil (group HNB, n = 8), hyponatremic animals treated with ICV saline (group HNS, n = 5) and normonatremic animals treated with ICV benzamil (group NNB, n = 5). Hyponatremia was induced by intraperitoneal administration of 140 mmol/l dextrose solution in a dose of 20% of body weight. Benzamil hydrochloride (4 μg) was injected ICV to the treated animals. RESULTS During the course of hyponatemia, ADC declined steadily from the baseline (100%) to reach a minimum of 92.32 ± 3.20% at 90 min (p < 0.0005). This process was associated with an increase in water signal to a maximum of 5.95 ± 2.62% at 100 min (p < 0.0005). After pretreatment with benzamil, no consistent changes occurred either in ADC or in water signal. CONCLUSIONS These findings suggest that sodium channel blockade with ICV benzamil has an immediate protective effect against the development of hyponatremic brain edema. Sodium channels, therefore, appear to be intimately involved in the initiation and progression of brain water accumulation in severe hyponatremia.
Collapse
Affiliation(s)
- Roy Steier
- Department of Neurosurgery, Faculty of Medicine, University of Pécs, Rét Street 2, 7623, Pécs, Hungary.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Feig SL, Haberly LB. Surface-associated astrocytes, not endfeet, form the glia limitans in posterior piriform cortex and have a spatially distributed, not a domain, organization. J Comp Neurol 2011; 519:1952-69. [PMID: 21452238 DOI: 10.1002/cne.22615] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
"Surface-associated astrocytes" (SAAs) in posterior piriform cortex (PPC) are unique by virtue of a direct apposition to the cortical surface and large-caliber processes that descend into layer I. In this study additional unique and functionally relevant features of SAAs in PPC of the rat were identified by light and electron microscopy. Examination of sections cut parallel to the surface of PPC and stained for glial fibrillar acidic protein revealed that, in addition to descending processes, SAAs give rise to an extensive matrix of "superficial processes." Electron microscopy revealed that these superficial processes, together with cell bodies, form a continuous sheet at the surface of PPC with features in common with the glia limitans that is formed by endfeet in other cortical areas. These include a glia limiting membrane with basal lamina and similar associated organelles, including a striking array of mitochondria. Of particular interest, SAAs lack the domain organization observed in neocortex and hippocampus. Rather, superficial processes overlap extensively with gap junctions between their proximal regions as well as between cell bodies. Study of the descending processes revealed thin extensions, many of which appose synaptic profiles. We conclude that SAAs provide a potential substrate for bidirectional signaling and transport between brain and the pial arteries and cerebrospinal fluid in the subarachnoid space. We postulate that the spatially distributed character of SAAs in PPC reflects and supports the spatially distributed circuitry and sensory representation that are also unique features of this area.
Collapse
Affiliation(s)
- S L Feig
- Department of Anatomy and Neuroscience Training Program, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53706, USA.
| | | |
Collapse
|
145
|
Dibaj P, Kaiser M, Hirrlinger J, Kirchhoff F, Neusch C. Kir4.1 channels regulate swelling of astroglial processes in experimental spinal cord edema. J Neurochem 2011; 103:2620-8. [PMID: 17953658 DOI: 10.1111/j.1471-4159.2007.04979.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In glial cells, inwardly rectifying K(+) channels (Kir) control extracellular [K(+) ](o) homeostasis by uptake of K(+) from the extracellular space and release of K(+) into the microvasculature. Kir channels were also recently implicated in K(+) -associated water influx and cell swelling. We studied the time-dependent expression and functional implication of the glial Kir4.1 channel for astroglial swelling in a spinal cord edema model. In this CNS region, Kir4.1 is expressed on astrocytes from the second postnatal week on and co-localizes with aquaporin 4 (AQP4). Swelling of individual astrocytes in response to osmotic stress and to pharmacological Kir blockade were analyzed by time-lapse-two-photon laser-scanning microscopy in situ. Application of 30% hypotonic solution induced astroglial soma swelling whereas no swelling was observed on astroglial processes or endfeet. Co-application of hypotonic solution and Ba(2+) , a Kir channel blocker, induced prominent swelling of astroglial processes. In Kir4.1-/- mice, however, somatic as well as process swelling was observed upon application of 30% hypotonic solutions. No additional effect was provoked upon co-application with Ba(2+) . Our experiments show that Kir channels prevent glial process swelling under osmotic stress. The underlying Kir channel subunit that controls glial process swelling is Kir4.1, whereas changes of the glial soma are not substantially related to Kir4.1.
Collapse
Affiliation(s)
- Payam Dibaj
- Department of Neurology, Georg-August-University Göttingen, Göttingen, Germany
| | | | | | | | | |
Collapse
|
146
|
Haj-Yasein NN, Jensen V, Vindedal GF, Gundersen GA, Klungland A, Ottersen OP, Hvalby O, Nagelhus EA. Evidence that compromised K+ spatial buffering contributes to the epileptogenic effect of mutations in the human Kir4.1 gene (KCNJ10). Glia 2011; 59:1635-42. [PMID: 21748805 DOI: 10.1002/glia.21205] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 05/27/2011] [Accepted: 06/01/2011] [Indexed: 12/21/2022]
Abstract
Mutations in the human Kir4.1 potassium channel gene (KCNJ10) are associated with epilepsy. Using a mouse model with glia-specific deletion of Kcnj10, we have explored the mechanistic underpinning of the epilepsy phenotype. The gene deletion was shown to delay K(+) clearance after synaptic activation in stratum radiatum of hippocampal slices. The activity-dependent changes in extracellular space volume did not differ between Kcnj10 mutant and wild-type mice, indicating that the Kcnj10 gene product Kir4.1 mediates osmotically neutral K(+) clearance. Combined, our K(+) and extracellular volume recordings indicate that compromised K(+) spatial buffering in brain underlies the epilepsy phenotype associated with human KCNJ10 mutations.
Collapse
Affiliation(s)
- Nadia Nabil Haj-Yasein
- Centre for Molecular Biology and Neuroscience, Letten Centre, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | | | |
Collapse
|
147
|
Borsani E. Aquaporins in sensory and pain transmission. Curr Neuropharmacol 2011; 8:122-7. [PMID: 21119883 PMCID: PMC2923366 DOI: 10.2174/157015910791233187] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Revised: 03/01/2010] [Accepted: 04/07/2010] [Indexed: 12/31/2022] Open
Abstract
Recent data suggest a possible involvement of Aquaporins (AQPs) in pain transmission. AQPs are small membrane channel proteins involved in osmoregulation and, to date, AQP1, AQP2, AQP3, AQP4, AQP5, AQP8 and AQP9 have been found in the nervous system. Nevertheless only AQP1, AQP2 and AQP4 seem to be involved in nociception. In this review, direct and indirect evidences of the role of AQPs in pain processing will be reported.
Collapse
Affiliation(s)
- Elisa Borsani
- Division of Human Anatomy, Department of Biomedical Sciences and Biotecnologies, University of Brescia, V.le Europa 11, 25123 Brescia, Italy.
| |
Collapse
|
148
|
Bonomini F, Francesca B, Rezzani R. Aquaporin and blood brain barrier. Curr Neuropharmacol 2011; 8:92-6. [PMID: 21119879 PMCID: PMC2923372 DOI: 10.2174/157015910791233132] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Revised: 03/01/2010] [Accepted: 04/07/2010] [Indexed: 12/25/2022] Open
Abstract
Large water fluxes continuously take place between the different compartments of the brain as well as between the brain parenchyma and the blood or cerebrospinal fluid. Disturbances in this well-regulated water homeostasis may have deleterious effects on brain function and may be fatal in cases where water accumulates in the brain following pathologies such as ischemia, haemorrhage, or brain trauma. The molecular pathways by which water molecules cross the blood brain barrier are not well-understood, although the discovery of Aquaporin 4 (AQP4) in the brain improved the understanding of some of these transport processes, particularly under pathological conditions.
Collapse
Affiliation(s)
- Francesca Bonomini
- Division of Human Anatomy, Department of Biomedical Sciences and Biotechnologies, University of Brescia, V.le Europa 11, 25123 Brescia, Italy
| | | | | |
Collapse
|
149
|
Aquaporin-4: orthogonal array assembly, CNS functions, and role in neuromyelitis optica. Acta Pharmacol Sin 2011; 32:702-10. [PMID: 21552296 DOI: 10.1038/aps.2011.27] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aquaporin-4 (AQP4) is a water-selective transporter expressed in astrocytes throughout the central nervous system, as well as in kidney, lung, stomach and skeletal muscle. The two AQP4 isoforms produced by alternative spicing, M1 and M23 AQP4, form heterotetramers that assemble in cell plasma membranes in supramolecular structures called orthogonal arrays of particles (OAPs). Phenotype analysis of AQP4-null mice indicates the involvement of AQP4 in brain and spinal cord water balance, astrocyte migration, neural signal transduction and neuroinflammation. AQP4-null mice manifest reduced brain swelling in cytotoxic cerebral edema, but increased brain swelling in vasogenic edema and hydrocephalus. AQP4 deficiency also increases seizure duration, impairs glial scarring, and reduces the severity of autoimmune neuroinflammation. Each of these phenotypes is likely explicable on the basis of reduced astrocyte water permeability in AQP4 deficiency. AQP4 is also involved in the neuroinflammatory demyelinating disease neuromyelitis optica (NMO), where autoantibodies (NMO-IgG) targeting AQP4 produce astrocyte damage and inflammation. Mice administered NMO-IgG and human complement by intracerebral injection develop characteristic NMO lesions with neuroinflammation, demyelination, perivascular complement deposition and loss of glial fibrillary acidic protein and AQP4 immunoreactivity. Our findings suggest the potential utility of AQP4-based therapeutics, including small-molecule modulators of AQP4 water transport function for therapy of brain swelling, injury and epilepsy, as well as small-molecule or monoclonal antibody blockers of NMO-IgG binding to AQP4 for therapy of NMO.
Collapse
|
150
|
Strohschein S, Hüttmann K, Gabriel S, Binder DK, Heinemann U, Steinhäuser C. Impact of aquaporin-4 channels on K+ buffering and gap junction coupling in the hippocampus. Glia 2011; 59:973-80. [PMID: 21446052 DOI: 10.1002/glia.21169] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 02/23/2011] [Indexed: 11/08/2022]
Abstract
Aquaporin-4 (AQP4) is the main water channel in the brain and primarily localized to astrocytes where the channels are thought to contribute to water and K(+) homeostasis. The close apposition of AQP4 and inward rectifier K(+) channels (Kir4.1) led to the hypothesis of direct functional interactions between both channels. We investigated the impact of AQP4 on stimulus-induced alterations of the extracellular K(+) concentration ([K(+)](o)) in murine hippocampal slices. Recordings with K(+)-selective microelectrodes combined with field potential analyses were compared in wild type (wt) and AQP4 knockout (AQP4(-/-)) mice. Astrocyte gap junction coupling was assessed with tracer filling during patch clamp recording. Antidromic fiber stimulation in the alveus evoked smaller increases and slower recovery of [K(+)](o) in the stratum pyramidale of AQP4(-/-) mice indicating reduced glial swelling and a larger extracellular space when compared with control tissue. Moreover, the data hint at an impairment of the glial Na(+)/K(+) ATPase in AQP4-deficient astrocytes. In a next step, we investigated the laminar profile of [K(+)](o) by moving the recording electrode from the stratum pyramidale toward the hippocampal fissure. At distances beyond 300 μm from the pyramidal layer, the stimulation-induced, normalized increases of [K(+)](o) in AQP4(-/-) mice exceeded the corresponding values of wt mice, indicating facilitated spatial buffering. Astrocytes in AQP4(-/-) mice also displayed enhanced tracer coupling, which might underlie the improved spatial re- distribution of [K(+)](o) in the hippocampus. These findings highlight the role of AQP4 channels in the regulation of K(+) homeostasis.
Collapse
Affiliation(s)
- Susan Strohschein
- Institute of Cellular Neurosciences, University of Bonn, Bonn, Germany.
| | | | | | | | | | | |
Collapse
|