1
|
Ochoa-de la Paz LD, Gulias-Cañizo R. Glia as a key factor in cell volume regulation processes of the central nervous system. Front Cell Neurosci 2022; 16:967496. [PMID: 36090789 PMCID: PMC9453262 DOI: 10.3389/fncel.2022.967496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/03/2022] [Indexed: 11/23/2022] Open
Abstract
Brain edema is a pathological condition with potentially fatal consequences, related to cerebral injuries such as ischemia, chronic renal failure, uremia, and diabetes, among others. Under these pathological states, the cell volume control processes are fully compromised, because brain cells are unable to regulate the movement of water, mainly regulated by osmotic gradients. The processes involved in cell volume regulation are homeostatic mechanisms that depend on the mobilization of osmolytes (ions, organic molecules, and polyols) in the necessary direction to counteract changes in osmolyte concentration in response to water movement. The expression and coordinated function of proteins related to the cell volume regulation process, such as water channels, ion channels, and other cotransport systems in the glial cells, and considering the glial cell proportion compared to neuronal cells, leads to consider the astroglial network the main regulatory unit for water homeostasis in the central nervous system (CNS). In the last decade, several studies highlighted the pivotal role of glia in the cell volume regulation process and water homeostasis in the brain, including the retina; any malfunction of this astroglial network generates a lack of the ability to regulate the osmotic changes and water movements and consequently exacerbates the pathological condition.
Collapse
Affiliation(s)
- Lenin David Ochoa-de la Paz
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico
- Asociación para Evitar la Ceguera en México (APEC), Unidad de Investigación APEC-UNAM, Mexico
- *Correspondence: Lenin David Ochoa-de la Paz
| | | |
Collapse
|
2
|
Li X, Lv J, Li J, Ren X. Kir4.1 may represent a novel therapeutic target for diabetic retinopathy (Review). Exp Ther Med 2021; 22:1021. [PMID: 34373707 PMCID: PMC8343704 DOI: 10.3892/etm.2021.10453] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/28/2021] [Indexed: 12/27/2022] Open
Abstract
As the major cause of irreversible loss of vision in adults, diabetic retinopathy (DR) is one of the most serious complications of diabetes. The imbalance of the retinal microenvironment and destruction of the blood-retinal barrier have a significant role in the progression of DR. Inward rectifying potassium channel 4.1 (Kir4.1) is located on Müller cells and is closely related to potassium homeostasis, water balance and glutamate clearance in the whole retina. The present review discusses the functions of Kir4.1 in regulating the retinal microenvironment and related biological mechanisms in DR. In the future, Kir4.1 may represent a novel alternative therapeutic target for DR through affecting the retinal microenvironment.
Collapse
Affiliation(s)
- Xiaoyu Li
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Department of Radiotherapy Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Jiajun Lv
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Department of Radiotherapy Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Jiazhi Li
- Department of Radiotherapy Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Xiang Ren
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
3
|
Llorián-Salvador M, Barabas P, Byrne EM, Lechner J, Augustine J, Curtis TM, Chen M, Xu H. VEGF-B Is an Autocrine Gliotrophic Factor for Müller Cells under Pathologic Conditions. Invest Ophthalmol Vis Sci 2021; 61:35. [PMID: 32945843 PMCID: PMC7509798 DOI: 10.1167/iovs.61.11.35] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Müller glia are important in retinal health and disease and are a major source of retinal VEGF-A. Of the different VEGF family members, the role of VEGF-A in retinal health and disease has been studied extensively. The potential contribution of other VEGF family members to retinal pathophysiology, however, remains poorly defined. This study aimed to understand the role of VEGF-B in Müller cell pathophysiology. Methods The expression of different VEGFs and their receptors in human MIO-M1 and mouse QMMuC-1 Müller cell lines and primary murine Müller cells was examined by RT-PCR, ELISA, and Western blot. The effect of recombinant VEGF-B or VEGF-B neutralization on Müller cell viability and survival under normal, hypoxic, and oxidative (4-hydroxynonenal [4-HNE]) conditions was evaluated by Alamar Blue, Yo-Pro uptake, and immunocytochemistry. The expression of glial fibrillary acidic protein, aquaporin-4, inward rectifying K+ channel subtype 4.1, glutamine synthetase, and transient receptor potential vanilloid 4 under different treatment conditions was examined by RT-PCR, immunocytochemistry, and Western blot. Transient receptor potential vanilloid 4 channel activity was assessed using a Fura-2–based calcium assay. Results VEGF-B was expressed in Müller cells at the highest levels compared with other members of the VEGF family. VEGF-B neutralization did not affect Müller cell viability or functionality under normal conditions, but enhanced hypoxia– or 4-HNE–induced Müller cell death and decreased inward rectifying K+ channel subtype 4.1 and aquaporin-4 expression. Recombinant VEGF-B restored Müller cell glutamine synthetase expression under hypoxic conditions and protected Müller cells from 4-HNE–induced damage by normalizing transient receptor potential vanilloid 4 channel expression and activity. Conclusions Autocrine production of VEGF-B protects Müller cells under pathologic conditions.
Collapse
Affiliation(s)
- María Llorián-Salvador
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, BT9 7BL. Belfast, United Kingdom
| | - Peter Barabas
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, BT9 7BL. Belfast, United Kingdom
| | - Eimear M Byrne
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, BT9 7BL. Belfast, United Kingdom
| | - Judith Lechner
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, BT9 7BL. Belfast, United Kingdom
| | - Josy Augustine
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, BT9 7BL. Belfast, United Kingdom
| | - Timothy M Curtis
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, BT9 7BL. Belfast, United Kingdom
| | - Mei Chen
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, BT9 7BL. Belfast, United Kingdom
| | - Heping Xu
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, BT9 7BL. Belfast, United Kingdom
| |
Collapse
|
4
|
Zhang KY, Johnson TV. The internal limiting membrane: Roles in retinal development and implications for emerging ocular therapies. Exp Eye Res 2021; 206:108545. [PMID: 33753089 DOI: 10.1016/j.exer.2021.108545] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/02/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022]
Abstract
Basement membranes help to establish, maintain, and separate their associated tissues. They also provide growth and signaling substrates for nearby resident cells. The internal limiting membrane (ILM) is the basement membrane at the ocular vitreoretinal interface. While the ILM is essential for normal retinal development, it is dispensable in adulthood. Moreover, the ILM may constitute a significant barrier to emerging ocular therapeutics, such as viral gene therapy or stem cell transplantation. Here we take a neurodevelopmental perspective in examining how retinal neurons, glia, and vasculature interact with individual extracellular matrix constituents at the ILM. In addition, we review evidence that the ILM may impede novel ocular therapies and discuss approaches for achieving retinal parenchymal targeting of gene vectors and cell transplants delivered into the vitreous cavity by manipulating interactions with the ILM.
Collapse
Affiliation(s)
- Kevin Y Zhang
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Maumenee B-110, Baltimore, MD, 21287, USA
| | - Thomas V Johnson
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Maumenee B-110, Baltimore, MD, 21287, USA.
| |
Collapse
|
5
|
Noël G, Tham DKL, Guadagno E, MacVicar B, Moukhles H. The Laminin-Induced Phosphorylation of PKCδ Regulates AQP4 Distribution and Water Permeability in Rat Astrocytes. Cell Mol Neurobiol 2020; 41:1743-1757. [PMID: 32851539 DOI: 10.1007/s10571-020-00944-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 08/14/2020] [Indexed: 11/29/2022]
Abstract
In astrocytes, the water-permeable channel aquaporin-4 (AQP4) is concentrated at the endfeet that abut the blood vessels of the brain. The asymmetric distribution of this channel is dependent on the function of dystroglycan (DG), a co-expressed laminin receptor, and its associated protein complex. We have demonstrated that the addition of laminin to astrocytes in culture causes the clustering of AQP4, DG, and lipid rafts. The last, in particular, have been associated with the initiation of cell signaling. As laminin binding to DG in muscle cells can induce the tyrosine phosphorylation of syntrophin and laminin requires tyrosine kinases for acetylcholine receptor clustering in myotubes, we asked if signal transduction might also be involved in AQP4 clustering in astrocytes. We analyzed the timecourse of AQP4, DG, and monosialotetrahexosylganglioside (GM1) clustering in primary cultures of rat astrocytes following the addition of laminin, and determined that the clustering of DG precedes that of AQP4 and GM1. We also showed that laminin induces the formation of phosphotyrosine-rich clusters and that the tyrosine kinase inhibitor, genistein, disrupts the laminin-induced clustering of both β-DG and AQP4. Using the Kinexus antibody microarray chip, we then identified protein-serine kinase C delta (PKCδ) as one of the main proteins exhibiting high levels of tyrosine phosphorylation upon laminin treatment. Selective inhibitors of PKC and siRNA against PKCδ disrupted β-DG and AQP4 clustering, and also caused water transport to increase in astrocytes treated with laminin. Our results demonstrate that the effects of laminin on AQP4 localization and function are relayed, at least in part, through PKC signaling.
Collapse
Affiliation(s)
- Geoffroy Noël
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 1Z3, Canada
| | - Daniel Kai Long Tham
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 1Z3, Canada
| | - Eric Guadagno
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 1Z3, Canada
| | - Brian MacVicar
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, V6T 1Z3, Canada
| | - Hakima Moukhles
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 1Z3, Canada.
| |
Collapse
|
6
|
Noël G, Tham DKL, MacVicar BA, Moukhles H. Agrin plays a major role in the coalescence of the aquaporin-4 clusters induced by gamma-1-containing laminin. J Comp Neurol 2019; 528:407-418. [PMID: 31454080 DOI: 10.1002/cne.24763] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/13/2019] [Accepted: 08/22/2019] [Indexed: 01/06/2023]
Abstract
The basement membrane that seperates the endothelial cells and astrocytic endfeet that comprise the blood-brain barrier is rich in collagen, laminin, agrin, and perlecan. Previous studies have demonstrated that the proper recruitment of the water-permeable channel aquaporin-4 (AQP4) to astrocytic endfeet is dependent on interactions between laminin and the receptor dystroglycan. In this study, we conducted a deeper investigation into how the basement membrane might further regulate the expression, localization, and function of AQP4, using primary astrocytes as a model system. We found that treating these cells with laminin causes endogenous agrin to localize to the cell surface, where it co-clusters with β-dystroglycan (β-DG). Conversely, agrin sliencing profoundly disrupts β-DG clustering. As in the case of laminin111, Matrigel™, a complete basement membrane analog, also causes the clustering of AQP4 and β-DG. This clustering, whether induced by laminin111 or Matrigel™ is inhibited when the astrocytes are first incubated with an antibody against the γ1 subunit of laminin, suggesting that the latter is crucial to the process. Finally, we showed that laminin111 appears to negatively regulate AQP4-mediated water transport in astrocytes, suppressing the cell swelling that occurs following a hypoosmotic challenge. This suppression is abolished if DG expression is silenced, again demonstrating the central role of this receptor in relaying the effects of laminin.
Collapse
Affiliation(s)
- Geoffroy Noël
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daniel Kai Long Tham
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brian A MacVicar
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hakima Moukhles
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
7
|
Nickolls AR, Bönnemann CG. The roles of dystroglycan in the nervous system: insights from animal models of muscular dystrophy. Dis Model Mech 2018; 11:11/12/dmm035931. [PMID: 30578246 PMCID: PMC6307911 DOI: 10.1242/dmm.035931] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Dystroglycan is a cell membrane protein that binds to the extracellular matrix in a variety of mammalian tissues. The α-subunit of dystroglycan (αDG) is heavily glycosylated, including a special O-mannosyl glycoepitope, relying upon this unique glycosylation to bind its matrix ligands. A distinct group of muscular dystrophies results from specific hypoglycosylation of αDG, and they are frequently associated with central nervous system involvement, ranging from profound brain malformation to intellectual disability without evident morphological defects. There is an expanding literature addressing the function of αDG in the nervous system, with recent reports demonstrating important roles in brain development and in the maintenance of neuronal synapses. Much of these data are derived from an increasingly rich array of experimental animal models. This Review aims to synthesize the information from such diverse models, formulating an up-to-date understanding about the various functions of αDG in neurons and glia of the central and peripheral nervous systems. Where possible, we integrate these data with our knowledge of the human disorders to promote translation from basic mechanistic findings to clinical therapies that take the neural phenotypes into account. Summary: Dystroglycan is a ubiquitous matrix receptor linked to brain and muscle disease. Unraveling the functions of this protein will inform basic and translational research on neural development and muscular dystrophies.
Collapse
Affiliation(s)
- Alec R Nickolls
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.,Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Carsten G Bönnemann
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
8
|
Leyva-Leyva M, Sandoval A, Felix R, González-Ramírez R. Biochemical and Functional Interplay Between Ion Channels and the Components of the Dystrophin-Associated Glycoprotein Complex. J Membr Biol 2018; 251:535-550. [PMID: 29779049 DOI: 10.1007/s00232-018-0036-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 05/09/2018] [Indexed: 12/19/2022]
Abstract
Dystrophin is a cytoskeleton-linked membrane protein that binds to a larger multiprotein assembly called the dystrophin-associated glycoprotein complex (DGC). The deficiency of dystrophin or the components of the DGC results in the loss of connection between the cytoskeleton and the extracellular matrix with significant pathophysiological implications in skeletal and cardiac muscle as well as in the nervous system. Although the DGC plays an important role in maintaining membrane stability, it can also be considered as a versatile and flexible molecular complex that contribute to the cellular organization and dynamics of a variety of proteins at specific locations in the plasma membrane. This review deals with the role of the DGC in transmembrane signaling by forming supramolecular assemblies for regulating ion channel localization and activity. These interactions are relevant for cell homeostasis, and its alterations may play a significant role in the etiology and pathogenesis of various disorders affecting muscle and nerve function.
Collapse
Affiliation(s)
- Margarita Leyva-Leyva
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico
| | - Alejandro Sandoval
- Faculty of Superior Studies Iztacala, National Autonomous University of Mexico (UNAM), Tlalnepantla, Mexico
| | - Ricardo Felix
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, Mexico.
| | - Ricardo González-Ramírez
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico.
| |
Collapse
|
9
|
Thompson K, Chen J, Luo Q, Xiao Y, Cummins TR, Bhatwadekar AD. Advanced glycation end (AGE) product modification of laminin downregulates Kir4.1 in retinal Müller cells. PLoS One 2018; 13:e0193280. [PMID: 29474462 PMCID: PMC5825079 DOI: 10.1371/journal.pone.0193280] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/07/2018] [Indexed: 11/19/2022] Open
Abstract
Diabetic retinopathy (DR) is a major cause of adult blindness. Retinal Müller cells maintain water homeostasis and potassium concentration via inwardly rectifying Kir4.1 channels. Accumulation of advanced glycation end products (AGEs) is a major pathologic event in DR. While diabetes leads to a decrease in the Kir4.1 channels, it remains unknown whether AGEs-linked to the basement membrane (BM) affect normal Kir4.1 channels. For this study, we hypothesized that AGE-modification of laminin is detrimental to Kir4.1 channels, therefore, disrupting Müller cell function. The AGE-modified laminin-coated substrates were prepared by incubating Petri-dishes with laminin and methylglyoxal for seven days. The rat Müller cells (rMC-1) were propagated on AGE-modified laminin, and Kir4.1 expression and function were evaluated. Quantification of AGEs using ELISA revealed a dose-dependent increase in methylglyoxal-hydro-imidazolone adducts. The rMC-1 propagated on AGE-modified laminin demonstrated a decrease in Kir4.1 levels in immunofluorescence and western blot studies and a decrease in the Kir4.1 channel function. Kir4.1 decrease on AGE-modified laminin resulted in a disorganization of an actin cytoskeleton and disruption of α-dystroglycan-syntrophin-dystrophin complexes. Our studies suggest that AGE-modification of laminin is detrimental to Kir4.1 channels. By studying the role of AGEs in Kir4.1 channels we have identified a novel mechanism of Müller cell dysfunction and its subsequent involvement in DR.
Collapse
Affiliation(s)
- Kayla Thompson
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Jonathan Chen
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Qianyi Luo
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Yucheng Xiao
- Department of Biology, Indiana University-Purdue University, Indianapolis, Indiana, United States of America
| | - Theodore R. Cummins
- Department of Biology, Indiana University-Purdue University, Indianapolis, Indiana, United States of America
| | - Ashay D. Bhatwadekar
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
10
|
Tham DKL, Joshi B, Moukhles H. Aquaporin-4 Cell-Surface Expression and Turnover Are Regulated by Dystroglycan, Dynamin, and the Extracellular Matrix in Astrocytes. PLoS One 2016; 11:e0165439. [PMID: 27788222 PMCID: PMC5082936 DOI: 10.1371/journal.pone.0165439] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/11/2016] [Indexed: 11/19/2022] Open
Abstract
The water-permeable channel aquaporin-4 (AQP4) is highly expressed in perivascular astrocytes of the mammalian brain and represents the major conduit for water across the blood-brain barrier. Within these cells, AQP4 is found in great quantities at perivascular endfoot sites but is detected in lesser amounts at the membrane domains within the brain parenchyma. We had previously established that this polarization was regulated by the interaction between dystroglycan (DG), an extracellular matrix receptor that is co-expressed with AQP4, and the laminin that is contained within the perivascular basal lamina. In the present study, we have attempted to describe the mechanisms that underlie this regulation, using primary astrocyte cultures. Via biotinylation, we found that the cell-surface expression of AQP4 is DG-dependent and is potentiated by laminin. We also determined that this laminin-dependent increase occurs not through an upregulation of total AQP4 levels, but rather from a redirection of AQP4 from an intracellular, EEA-1-associated pool to the cell surface. We then demonstrated an association between DG and dynamin and showed that dynamin functioned in conjunction with clathrin to regulate surface AQP4 amounts. Furthermore, we observed that DG preferentially binds to the inactive forms of dynamin, suggesting that this interaction was inhibitory for AQP4 endocytosis. Finally, we showed that laminin selectively upregulates the cell-surface expression of the M23 isoform of AQP4. Our data therefore indicate that the dual interation of DG with laminin and dynamin is involved in the regulation of AQP4 internalization, leading to its asymmetric enrichment at perivascular astrocyte endfeet.
Collapse
Affiliation(s)
- Daniel Kai Long Tham
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bharat Joshi
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hakima Moukhles
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
11
|
Pannicke T, Ivo Chao T, Reisenhofer M, Francke M, Reichenbach A. Comparative electrophysiology of retinal Müller glial cells-A survey on vertebrate species. Glia 2016; 65:533-568. [PMID: 27767232 DOI: 10.1002/glia.23082] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/15/2016] [Accepted: 09/28/2016] [Indexed: 12/13/2022]
Abstract
Müller cells are the dominant macroglial cells in the retina of all vertebrates. They fulfill a variety of functions important for retinal physiology, among them spatial buffering of K+ ions and uptake of glutamate and other neurotransmitters. To this end, Müller cells express inwardly rectifying K+ channels and electrogenic glutamate transporters. Moreover, a lot of voltage- and ligand-gated ion channels, aquaporin water channels, and electrogenic transporters are expressed in Müller cells, some of them in a species-specific manner. For example, voltage-dependent Na+ channels are found exclusively in some but not all mammalian species. Whereas a lot of data exist from amphibians and mammals, the results from other vertebrates are sparse. It is the aim of this review to present a survey on Müller cell electrophysiology covering all classes of vertebrates. The focus is on functional studies, mainly performed using the whole-cell patch-clamp technique. However, data about the expression of membrane channels and transporters from immunohistochemistry are also included. Possible functional roles of membrane channels and transporters are discussed. Obviously, electrophysiological properties involved in the main functions of Müller cells developed early in vertebrate evolution. GLIA 2017;65:533-568.
Collapse
Affiliation(s)
- Thomas Pannicke
- Paul-Flechsig-Institut für Hirnforschung, Abteilung Pathophysiologie der Neuroglia, Universität Leipzig, Germany
| | - T Ivo Chao
- Institute of Anatomy and Cell Biology, Medical School Göttingen, Germany
| | - Miriam Reisenhofer
- Department of Chemistry, University of Zürich, Switzerland
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Mike Francke
- Paul-Flechsig-Institut für Hirnforschung, Abteilung Pathophysiologie der Neuroglia, Universität Leipzig, Germany
- Sächsischer Inkubator für klinische Translation (SIKT), Universität Leipzig, Germany
| | - Andreas Reichenbach
- Paul-Flechsig-Institut für Hirnforschung, Abteilung Pathophysiologie der Neuroglia, Universität Leipzig, Germany
| |
Collapse
|
12
|
Posati T, Pistone A, Saracino E, Formaggio F, Mola MG, Troni E, Sagnella A, Nocchetti M, Barbalinardo M, Valle F, Bonetti S, Caprini M, Nicchia GP, Zamboni R, Muccini M, Benfenati V. A Nanoscale Interface Promoting Molecular and Functional Differentiation of Neural Cells. Sci Rep 2016; 6:31226. [PMID: 27503424 PMCID: PMC4977496 DOI: 10.1038/srep31226] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 07/04/2016] [Indexed: 12/11/2022] Open
Abstract
Potassium channels and aquaporins expressed by astrocytes are key players in the maintenance of cerebral homeostasis and in brain pathophysiologies. One major challenge in the study of astrocyte membrane channels in vitro, is that their expression pattern does not resemble the one observed in vivo. Nanostructured interfaces represent a significant resource to control the cellular behaviour and functionalities at micro and nanoscale as well as to generate novel and more reliable models to study astrocytes in vitro. However, the potential of nanotechnologies in the manipulation of astrocytes ion channels and aquaporins has never been previously reported. Hydrotalcite-like compounds (HTlc) are layered materials with increasing potential as biocompatible nanoscale interface. Here, we evaluate the effect of the interaction of HTlc nanoparticles films with primary rat neocortical astrocytes. We show that HTlc films are biocompatible and do not promote gliotic reaction, while favouring astrocytes differentiation by induction of F-actin fibre alignment and vinculin polarization. Western Blot, Immunofluorescence and patch-clamp revealed that differentiation was accompanied by molecular and functional up-regulation of both inward rectifying potassium channel Kir 4.1 and aquaporin 4, AQP4. The reported results pave the way to engineering novel in vitro models to study astrocytes in a in vivo like condition.
Collapse
Affiliation(s)
- Tamara Posati
- Consiglio Nazionale delle Ricerche (CNR), Istituto per lo Sintesi Organica e la Fotoreattività (ISOF), via Gobetti, 101, 40129, Bologna, Italy
| | - Assunta Pistone
- Consiglio Nazionale delle Ricerche (CNR), Istituto per lo Studio dei Materiali Nanostrutturati (ISMN), via Gobetti, 101, 40129, Bologna, Italy
| | - Emanuela Saracino
- Consiglio Nazionale delle Ricerche (CNR), Istituto per lo Studio dei Materiali Nanostrutturati (ISMN), via Gobetti, 101, 40129, Bologna, Italy
| | - Francesco Formaggio
- Consiglio Nazionale delle Ricerche (CNR), Istituto per lo Studio dei Materiali Nanostrutturati (ISMN), via Gobetti, 101, 40129, Bologna, Italy
| | - Maria Grazia Mola
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università degli Studi di Bari “Aldo Moro”, Via Amendola 165/A, 70126, Bari, Italy
| | - Elisabetta Troni
- Consiglio Nazionale delle Ricerche (CNR), Istituto per lo Sintesi Organica e la Fotoreattività (ISOF), via Gobetti, 101, 40129, Bologna, Italy
| | - Anna Sagnella
- Laboratorio di Micro e Submicro Tecnologie abilitanti dell’Emilia-Romagna (MIST E-R), Via P. Gobetti 101, I-40129 Bologna, Italy
| | - Morena Nocchetti
- Dipartimento di Scienze Farmaceutiche, University of Perugia, Via del Liceo 1, 06123, Perugia, Italy
| | - Marianna Barbalinardo
- Consiglio Nazionale delle Ricerche (CNR), Istituto per lo Studio dei Materiali Nanostrutturati (ISMN), via Gobetti, 101, 40129, Bologna, Italy
| | - Francesco Valle
- Consiglio Nazionale delle Ricerche (CNR), Istituto per lo Studio dei Materiali Nanostrutturati (ISMN), via Gobetti, 101, 40129, Bologna, Italy
| | - Simone Bonetti
- Consiglio Nazionale delle Ricerche (CNR), Istituto per lo Studio dei Materiali Nanostrutturati (ISMN), via Gobetti, 101, 40129, Bologna, Italy
| | - Marco Caprini
- Department of Pharmacy and Biotechnology, via S. Donato 19/2, University of Bologna, 40127 Bologna, Italy
| | - Grazia Paola Nicchia
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università degli Studi di Bari “Aldo Moro”, Via Amendola 165/A, 70126, Bari, Italy
| | - Roberto Zamboni
- Consiglio Nazionale delle Ricerche (CNR), Istituto per lo Sintesi Organica e la Fotoreattività (ISOF), via Gobetti, 101, 40129, Bologna, Italy
| | - Michele Muccini
- Consiglio Nazionale delle Ricerche (CNR), Istituto per lo Studio dei Materiali Nanostrutturati (ISMN), via Gobetti, 101, 40129, Bologna, Italy
| | - Valentina Benfenati
- Consiglio Nazionale delle Ricerche (CNR), Istituto per lo Sintesi Organica e la Fotoreattività (ISOF), via Gobetti, 101, 40129, Bologna, Italy
| |
Collapse
|
13
|
Nwaobi SE, Cuddapah VA, Patterson KC, Randolph AC, Olsen ML. The role of glial-specific Kir4.1 in normal and pathological states of the CNS. Acta Neuropathol 2016; 132:1-21. [PMID: 26961251 PMCID: PMC6774634 DOI: 10.1007/s00401-016-1553-1] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 02/16/2016] [Accepted: 02/25/2016] [Indexed: 12/15/2022]
Abstract
Kir4.1 is an inwardly rectifying K(+) channel expressed exclusively in glial cells in the central nervous system. In glia, Kir4.1 is implicated in several functions including extracellular K(+) homeostasis, maintenance of astrocyte resting membrane potential, cell volume regulation, and facilitation of glutamate uptake. Knockout of Kir4.1 in rodent models leads to severe neurological deficits, including ataxia, seizures, sensorineural deafness, and early postnatal death. Accumulating evidence indicates that Kir4.1 plays an integral role in the central nervous system, prompting many laboratories to study the potential role that Kir4.1 plays in human disease. In this article, we review the growing evidence implicating Kir4.1 in a wide array of neurological disease. Recent literature suggests Kir4.1 dysfunction facilitates neuronal hyperexcitability and may contribute to epilepsy. Genetic screens demonstrate that mutations of KCNJ10, the gene encoding Kir4.1, causes SeSAME/EAST syndrome, which is characterized by early onset seizures, compromised verbal and motor skills, profound cognitive deficits, and salt-wasting. KCNJ10 has also been linked to developmental disorders including autism. Cerebral trauma, ischemia, and inflammation are all associated with decreased astrocytic Kir4.1 current amplitude and astrocytic dysfunction. Additionally, neurodegenerative diseases such as Alzheimer disease and amyotrophic lateral sclerosis demonstrate loss of Kir4.1. This is particularly exciting in the context of Huntington disease, another neurodegenerative disorder in which restoration of Kir4.1 ameliorated motor deficits, decreased medium spiny neuron hyperexcitability, and extended survival in mouse models. Understanding the expression and regulation of Kir4.1 will be critical in determining if this channel can be exploited for therapeutic benefit.
Collapse
Affiliation(s)
- Sinifunanya E Nwaobi
- Department of Cell, Developmental and Integrative Biology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL, 35294, UK
| | - Vishnu A Cuddapah
- Department of Cell, Developmental and Integrative Biology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL, 35294, UK
| | - Kelsey C Patterson
- Department of Cell, Developmental and Integrative Biology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL, 35294, UK
| | - Anita C Randolph
- Department of Cell, Developmental and Integrative Biology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL, 35294, UK
| | - Michelle L Olsen
- Department of Cell, Developmental and Integrative Biology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL, 35294, UK.
| |
Collapse
|
14
|
Vacca O, Charles-Messance H, El Mathari B, Sene A, Barbe P, Fouquet S, Aragón J, Darche M, Giocanti-Aurégan A, Paques M, Sahel JA, Tadayoni R, Montañez C, Dalkara D, Rendon A. AAV-mediated gene therapy in Dystrophin-Dp71 deficient mouse leads to blood-retinal barrier restoration and oedema reabsorption. Hum Mol Genet 2016; 25:3070-3079. [PMID: 27288449 DOI: 10.1093/hmg/ddw159] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 04/28/2016] [Accepted: 05/19/2016] [Indexed: 11/12/2022] Open
Abstract
Dystrophin-Dp71 being a key membrane cytoskeletal protein, expressed mainly in Müller cells that provide a mechanical link at the Müller cell membrane by direct binding to actin and a transmembrane protein complex. Its absence has been related to blood-retinal barrier (BRB) permeability through delocalization and down-regulation of the AQP4 and Kir4.1 channels (1). We have previously shown that the adeno-associated virus (AAV) variant, ShH10, transduces Müller cells in the Dp71-null mouse retina efficiently and specifically (2,3). Here, we use ShH10 to restore Dp71 expression in Müller cells of Dp71 deficient mouse to study molecular and functional effects of this restoration in an adult mouse displaying retinal permeability. We show that strong and specific expression of exogenous Dp71 in Müller cells leads to correct localization of Dp71 protein restoring all protein interactions in order to re-establish a proper functional BRB and retina homeostasis thus preventing retina from oedema. This study is the basis for the development of new therapeutic strategies in dealing with diseases with BRB breakdown and macular oedema such as diabetic retinopathy (DR).
Collapse
Affiliation(s)
- Ophélie Vacca
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 75012 Paris, France .,Neuroscience Paris-Saclay Institute (NeuroPSI)-CNRS UMR 9197-Université Paris-Sud, Cognition & Behavior, 91405 Orsay cedex, France
| | - Hugo Charles-Messance
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Brahim El Mathari
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Abdoulaye Sene
- Department of Ophthalmology, Therapeutic, Saint Louis, MO 63103, USA
| | - Peggy Barbe
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Stéphane Fouquet
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Jorge Aragón
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 75012 Paris, France.,Department of Genetics & Molecular Biology, CINVESTAV: Research Centre for Advanced Studies, IPN, C.P. 07360 México
| | - Marie Darche
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Audrey Giocanti-Aurégan
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 75012 Paris, France.,Ophthalmology Department, Avicenne Hospital, 93000 Bobigny, France
| | - Michel Paques
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 75012 Paris, France.,CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC, 75012 Paris, France
| | - José-Alain Sahel
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 75012 Paris, France.,CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC, 75012 Paris, France
| | - Ramin Tadayoni
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 75012 Paris, France.,Ophthalmology Dept, Hôpital Lariboisière, AP-HP, Univ Paris Diderot, 75010 Paris, France
| | - Cecilia Montañez
- Department of Genetics & Molecular Biology, CINVESTAV: Research Centre for Advanced Studies, IPN, C.P. 07360 México
| | - Deniz Dalkara
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Alvaro Rendon
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| |
Collapse
|
15
|
Brignone MS, Lanciotti A, Camerini S, De Nuccio C, Petrucci TC, Visentin S, Ambrosini E. MLC1 protein: a likely link between leukodystrophies and brain channelopathies. Front Cell Neurosci 2015; 9:66. [PMID: 25883547 PMCID: PMC4381631 DOI: 10.3389/fncel.2015.00106] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 03/09/2015] [Indexed: 01/12/2023] Open
Abstract
Megalencephalic leukoencephalopathy with subcortical cysts (MLCs) disease is a rare inherited, autosomal recessive form of childhood-onset spongiform leukodystrophy characterized by macrocephaly, deterioration of motor functions, epileptic seizures and mental decline. Brain edema, subcortical fluid cysts, myelin and astrocyte vacuolation are the histopathological hallmarks of MLC. Mutations in either the MLC1 gene (>75% of patients) or the GlialCAM gene (<20% of patients) are responsible for the disease. Recently, the GlialCAM adhesion protein was found essential for the membrane expression and function of the chloride channel ClC-2 indicating MLC disease caused by mutation in GlialCAM as the first channelopathy among leukodystrophies. On the contrary, the function of MLC1 protein, which binds GlialCAM, its functional relationship with ClC-2 and the molecular mechanisms underlying MLC1 mutation-induced functional defects are not fully understood yet. The human MLC1 gene encodes a 377-amino acid membrane protein with eight predicted transmembrane domains which shows very low homology with voltage-dependent potassium (K+) channel subunits. The high expression of MLC1 in brain astrocytes contacting blood vessels and meninges and brain alterations observed in MLC patients have led to hypothesize a role for MLC1 in the regulation of ion and water homeostasis. Recent studies have shown that MLC1 establishes structural and/or functional interactions with several ion/water channels and transporters and ion channel accessory proteins, and that these interactions are affected by MLC1 mutations causing MLC. Here, we review data on MLC1 functional properties obtained in in vitro and in vivo models and discuss evidence linking the effects of MLC1 mutations to brain channelopathies.
Collapse
Affiliation(s)
- Maria S Brignone
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Rome, Italy
| | - Angela Lanciotti
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Rome, Italy
| | - Serena Camerini
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Rome, Italy
| | - Chiara De Nuccio
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Rome, Italy
| | - Tamara C Petrucci
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Rome, Italy
| | - Sergio Visentin
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Rome, Italy
| | - Elena Ambrosini
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Rome, Italy
| |
Collapse
|
16
|
|
17
|
Johnson EK, Li B, Yoon JH, Flanigan KM, Martin PT, Ervasti J, Montanaro F. Identification of new dystroglycan complexes in skeletal muscle. PLoS One 2013; 8:e73224. [PMID: 23951345 PMCID: PMC3738564 DOI: 10.1371/journal.pone.0073224] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 07/18/2013] [Indexed: 01/16/2023] Open
Abstract
The dystroglycan complex contains the transmembrane protein β-dystroglycan and its interacting extracellular mucin-like protein α-dystroglycan. In skeletal muscle fibers, the dystroglycan complex plays an important structural role by linking the cytoskeletal protein dystrophin to laminin in the extracellular matrix. Mutations that affect any of the proteins involved in this structural axis lead to myofiber degeneration and are associated with muscular dystrophies and congenital myopathies. Because loss of dystrophin in Duchenne muscular dystrophy (DMD) leads to an almost complete loss of dystroglycan complexes at the myofiber membrane, it is generally assumed that the vast majority of dystroglycan complexes within skeletal muscle fibers interact with dystrophin. The residual dystroglycan present in dystrophin-deficient muscle is thought to be preserved by utrophin, a structural homolog of dystrophin that is up-regulated in dystrophic muscles. However, we found that dystroglycan complexes are still present at the myofiber membrane in the absence of both dystrophin and utrophin. Our data show that only a minority of dystroglycan complexes associate with dystrophin in wild type muscle. Furthermore, we provide evidence for at least three separate pools of dystroglycan complexes within myofibers that differ in composition and are differentially affected by loss of dystrophin. Our findings indicate a more complex role of dystroglycan in muscle than currently recognized and may help explain differences in disease pathology and severity among myopathies linked to mutations in DAPC members.
Collapse
Affiliation(s)
- Eric K. Johnson
- Center for Gene Therapy, the Research Institute at Nationwide Children’s Hospital, and The Ohio State University Biochemistry Program, Columbus, Ohio, United States of America
- Ohio State Biochemistry Program, the Ohio State University, Columbus, Ohio, United States of America
| | - Bin Li
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jung Hae Yoon
- Center for Gene Therapy, the Research Institute at Nationwide Children’s Hospital, and The Ohio State University Biochemistry Program, Columbus, Ohio, United States of America
| | - Kevin M. Flanigan
- Center for Gene Therapy, the Research Institute at Nationwide Children’s Hospital, and The Ohio State University Biochemistry Program, Columbus, Ohio, United States of America
- Department of Pediatrics, the Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Paul T. Martin
- Center for Gene Therapy, the Research Institute at Nationwide Children’s Hospital, and The Ohio State University Biochemistry Program, Columbus, Ohio, United States of America
- Department of Pediatrics, the Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - James Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Federica Montanaro
- Center for Gene Therapy, the Research Institute at Nationwide Children’s Hospital, and The Ohio State University Biochemistry Program, Columbus, Ohio, United States of America
- Ohio State Biochemistry Program, the Ohio State University, Columbus, Ohio, United States of America
- Department of Pediatrics, the Ohio State University College of Medicine, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
18
|
Effects of prednisolone on the dystrophin-associated proteins in the blood-brain barrier and skeletal muscle of dystrophic mdx mice. J Transl Med 2013; 93:592-610. [PMID: 23528847 DOI: 10.1038/labinvest.2013.46] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The mdx mouse, the most widely used animal model of Duchenne muscular dystrophy (DMD), develops a seriously impaired blood-brain barrier (BBB). As glucocorticoids are used clinically to delay the progression of DMD, we evaluated the effects of chronic treatment with α-methyl-prednisolone (PDN) on the expression of structural proteins and markers in the brain and skeletal muscle of the mdx mouse. We analyzed the immunocytochemical and biochemical expression of four BBB markers, including endothelial ZO-1 and occludin, desmin in pericytes, and glial fibrillary acidic protein (GFAP) in glial cells, and the expression of the short dystrophin isoform Dp 71, the dystrophin-associated proteins (DAPs), and aquaporin-4 (AQP4) and α-β dystroglycan (DG) in the brain. We evaluated the BBB integrity of mdx and PDN-treated mdx mice by means of intravascular injection of horseradish peroxidase (HRP). The expression of DAPs was also assessed in gastrocnemius muscles and correlated with utrophin expression, and laminin content was measured in the muscle and brain. PDN treatment induced a significant increase in the mRNA and protein content of the BBB markers; a reduction in the phosphorylation of occludin in the brain and of AQP4/β DG in both tissues; an increase of Dp71 protein content; and an increase of both mRNA and protein levels of the AQP4/α-β DG complex. The latter was associated with enhanced laminin and utrophin in the muscle. The HRP assay demonstrated functional restoration of the BBB in the PDN-treated mdx mice. Specifically, mdx mice showed extensive perivascular labeling due to escape of the marker, while HRP was exclusively intravascular in the PDN-treated mice and the controls. These data illustrate for the first time that PDN reverses the BBB alterations in the mdx mouse and re-establishes the proper expression and phosphorylation of β-DG in both the BBB and skeletal muscle. Further, PDN partially protects against muscle damage. The reduction in AQP4 and occludin phosphorylation, coupled with their anchoring to glial and endothelial membranes in PDN-treated mice, suggests that the drug may target the glial and endothelial cells. Our results suggest a novel mechanism for PDN action on cerebral and muscular function, restoring the link between DAPs and the extracellular matrix, most likely through protein kinase inactivation.
Collapse
|
19
|
Peng H, Carbonetto S. Astrocyte polarization and wound healing in culture: studying cell adhesion molecules. Methods Mol Biol 2012; 814:177-88. [PMID: 22144308 DOI: 10.1007/978-1-61779-452-0_13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Astrocytes are highly polarized cells. This is manifest not only during development and in the adult brain, but also following injury. In response to a wound, astrocytes extend processes that participate in formation of a glial scar, which walls off lesions in the brain or spinal cord. Similarly, astrocytes in culture polarize dramatically and extend processes towards a scrape wound. This simple assay has allowed much progress in understanding the cellular events and molecular pathways in astrocyte polarization (1). Cell adhesion is essential for the early response to the wound, both with respect to process extension and cell polarization. This is evident in the involvement of members of the integrin family of cell adhesion molecules at the leading edge of the wounded astrocyte. Understanding the cellular and molecular bases of these events is likely relevant to astrocyte function during development (radial glia) as well as in wound healing.
Collapse
Affiliation(s)
- H Peng
- The Centre for Research in Neuroscience, McGill University Health Centre, Montréal, QC, Canada
| | | |
Collapse
|
20
|
Karlsson T, Glogauer M, Ellen RP, Loitto VM, Magnusson KE, Magalhães MAO. Aquaporin 9 phosphorylation mediates membrane localization and neutrophil polarization. J Leukoc Biol 2011; 90:963-73. [PMID: 21873454 DOI: 10.1189/jlb.0910540] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Neutrophils are of prime importance in the host innate defense against invading microorganisms by using two primary mechanisms-locomotion toward and phagocytosis of the prey. Recent research points to pivotal roles for water channels known as AQPs in cell motility. Here, we focused on the role of AQP9 in chemoattractant-induced polarization and migration of primary mouse neutrophils and neutrophil-like HL60 cells. We found that AQP9 is phosphorylated downstream of fMLFR or PMA stimulation in primary human neutrophils. The dynamics of AQP9 were assessed using GFP-tagged AQP9 constructs and other fluorescent markers through various live-cell imaging techniques. Expression of WT or the phosphomimic S11D AQP9 changed cell volume regulation as a response to hyperosmotic changes and enhanced neutrophil polarization and chemotaxis. WT AQP9 and S11D AQP9 displayed a very dynamic distribution at the cell membrane, whereas the phosphorylation-deficient S11A AQP9 failed to localize to the plasma membrane. Furthermore, we found that Rac1 regulated the translocation of AQP9 to the plasma membrane. Our results show that AQP9 plays an active role in neutrophil volume regulation and migration. The display of AQP9 at the plasma membrane depends on AQP9 phosphorylation, which appeared to be regulated through a Rac1-dependent pathway.
Collapse
Affiliation(s)
- Thommie Karlsson
- Division of Medical Microbiology, Department of Clinical and Experimental Medicine, Linkoping University, Linkoping, Sweden
| | | | | | | | | | | |
Collapse
|
21
|
Nico B, Ribatti D. Role of aquaporins in cell migration and edema formation in human brain tumors. Exp Cell Res 2011; 317:2391-6. [PMID: 21784068 DOI: 10.1016/j.yexcr.2011.07.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 07/04/2011] [Accepted: 07/09/2011] [Indexed: 01/08/2023]
Abstract
The aquaporins (AQPs) are a family of transmembrane water channel proteins widely distributed and play a major role in transcellular and transepithelial water movement. Moreover, recent evidence indicates that AQPs may be involved in cell migration, angiogenesis, and tumor growth. This review article summarizes literature data concerning the involvement of AQP-1 and -4 in human brain tumor growth and edema formation and suggests a potential therapeutic approach by antagonizing their biological activity.
Collapse
Affiliation(s)
- Beatrice Nico
- Department of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | | |
Collapse
|
22
|
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
|
23
|
Tham DKL, Moukhles H. Regulation of Kir4.1 and AQP4 expression and stability at the basolateral domain of epithelial MDCK cells by the extracellular matrix. Am J Physiol Renal Physiol 2011; 301:F396-409. [PMID: 21543416 DOI: 10.1152/ajprenal.00315.2010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The proper targeting of ion channels to specialized domains is crucial for cell function. Kir4.1, the inwardly rectifying potassium channel, and aquaporin-4 (AQP4), the type 4 water-permeable channel, are localized at the basolateral domain of polarized epithelial cells; however, the mechanisms involved in their localization have yet to be determined. In this study, we investigated the role of the extracellular matrix in the localization of these channels in polarized Madin-Darby canine kidney (MDCK) cells. MDCK cells expressing green fluorescent protein-Kir4.1 or -AQP4 were cultured on laminin-1 or fibronectin and examined by confocal microscopy and cell surface biotinylation to assess plasma membrane expression of Kir4.1 and AQP4. Our data show that laminin-1 and fibronectin induce a significant increase in cell surface expression of both channels at the basolateral domain. Using fluorescence recovery after photobleaching, we demonstrate that laminin-1 and fibronectin reduce the diffusion rates of these channels. Finally, we show that the laminin receptor dystroglycan is important for cell surface expression of Kir4.1 but not AQP4. However, laminin-1 increases cell surface expression of both channels in cells deficient for dystroglycan, indicating that other receptors are involved. Indeed, RGD-containing peptides, which inhibit fibronectin binding to certain integrins, prevent the fibronectin-induced increase in Kir4.1 and AQP4 cell surface expression and reverse the laminin- and fibronectin-induced reduction in both channels' diffusion rates. Similarly, the αvβ3-integrin function-blocking antibody alters the reduction of AQP4 diffusion rates induced by both laminin and fibronectin, suggesting that αvβ3-integrin plays a role in the stabilization of APQ4 at the basolateral domain of epithelial cells.
Collapse
Affiliation(s)
- Daniel Kai Long Tham
- Dept of Cellular and Physiological Sciences, Univ. of British Columbia, Vancouver, Canada
| | | |
Collapse
|
24
|
Hirrlinger PG, Pannicke T, Winkler U, Claudepierre T, Varshney S, Schulze C, Reichenbach A, Brunken WJ, Hirrlinger J. Genetic deletion of laminin isoforms β2 and γ3 induces a reduction in Kir4.1 and aquaporin-4 expression and function in the retina. PLoS One 2011; 6:e16106. [PMID: 21283711 PMCID: PMC3025027 DOI: 10.1371/journal.pone.0016106] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 12/07/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Glial cells such as retinal Müller glial cells are involved in potassium ion and water homeostasis of the neural tissue. In these cells, inwardly rectifying potassium (Kir) channels and aquaporin-4 water channels play an important role in the process of spatial potassium buffering and water drainage. Moreover, Kir4.1 channels are involved in the maintenance of the negative Müller cell membrane potential. The subcellular distribution of Kir4.1 and aquaporin-4 channels appears to be maintained by interactions with extracellular and intracellular molecules. Laminins in the extracellular matrix, dystroglycan in the membrane, and dystrophins in the cytomatrix form a complex mediating the polarized expression of Kir4.1 and aquaporin-4 in Müller cells. METHODOLOGY/PRINCIPAL FINDINGS The aim of the present study was to test the function of the β2 and γ3 containing laminins in murine Müller cells. We used knockout mice with genetic deletion of both β2 and γ3 laminin genes to assay the effects on Kir4.1 and aquaporin-4. We studied protein and mRNA expression by immunohistochemistry, Western Blot, and quantitative RT-PCR, respectively, and membrane currents of isolated cells by patch-clamp experiments. We found a down-regulation of mRNA and protein of Kir4.1 as well as of aquaporin-4 protein in laminin knockout mice. Moreover, Müller cells from laminin β2 and γ3 knockout mice had reduced Kir-mediated inward currents and their membrane potentials were more positive than those in age-matched wild-type mice. CONCLUSION These findings demonstrate a strong impact of laminin β2 and γ3 subunits on the expression and function of both aquaporin-4 and Kir4.1, two important membrane proteins in Müller cells.
Collapse
Affiliation(s)
- Petra G Hirrlinger
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Wolburg H, Wolburg-Buchholz K, Fallier-Becker P, Noell S, Mack AF. Structure and functions of aquaporin-4-based orthogonal arrays of particles. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2011; 287:1-41. [PMID: 21414585 DOI: 10.1016/b978-0-12-386043-9.00001-3] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Orthogonal arrays or assemblies of intramembranous particles (OAPs) are structures in the membrane of diverse cells which were initially discovered by means of the freeze-fracturing technique. This technique, developed in the 1960s, was important for the acceptance of the fluid mosaic model of the biological membrane. OAPs were first described in liver cells, and then in parietal cells of the stomach, and most importantly, in the astrocytes of the brain. Since the discovery of the structure of OAPs and the identification of OAPs as the morphological equivalent of the water channel protein aquaporin-4 (AQP4) in the 1990s, a plethora of morphological work on OAPs in different cells was published. Now, we feel a need to balance new and old data on OAPs and AQP4 to elucidate the interrelationship of both structures and molecules. In this review, the identity of OAPs as AQP4-based structures in a diversity of cells will be described. At the same time, arguments are offered that under pathological or experimental circumstances, AQP4 can also be expressed in a non-OAP form. Thus, we attempt to project classical work on OAPs onto the molecular biology of AQP4. In particular, astrocytes and glioma cells will play the major part in this review, not only due to our own work but also due to the fact that most studies on structure and function of AQP4 were done in the nervous system.
Collapse
Affiliation(s)
- Hartwig Wolburg
- Institute of Pathology, University of Tübingen, Tübingen, Germany
| | | | | | | | | |
Collapse
|
26
|
Nico B, Tamma R, Annese T, Mangieri D, De Luca A, Corsi P, Benagiano V, Longo V, Crivellato E, Salmaggi A, Ribatti D. Glial dystrophin-associated proteins, laminin and agrin, are downregulated in the brain of mdx mouse. J Transl Med 2010; 90:1645-60. [PMID: 20714324 DOI: 10.1038/labinvest.2010.149] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In this study, we investigated the involvement of dystrophin-associated proteins (DAPs) and their relationship with the perivascular basement membrane in the brains of mdx mice and controls at the age of 2 months. We analyzed (1) the expression of glial DAPs α-β-dystroglycan (DG), α-syntrophin, aquaporin-4 (AQP4) water channel, Kir 4.1 and dystrophin isoform (Dp71) by immunocytochemistry, laser confocal microscopy, immunogold electron microscopy, immunoblotting and RT-PCR; (2) the ultrastructure of the basement membrane and expression of laminin and agrin; and (3) the dual immunofluorescence colocalization of AQP4/α-β-DG, and of Kir 4.1/agrin. The following results were observed in mdx brain as compared with controls: (1) a significant reduction in protein content and mRNA expression of DAPs; (2) ultrastructurally, a thickened and discontinuous appearance of the basement membrane and a significant reduction in laminin and agrin; and (3) a molecular rearrangment of α-β-DG, coupled with a parallel loss of agrin and Kir 4.1 on basement membrane and glial endfeet. These data indicate that in mdx brain the deficiency in dystrophin and dystrophin isoform (Dp71) is coupled with a reduction of DAP components, coupled with an altered anchoring to the basement membrane.
Collapse
Affiliation(s)
- Beatrice Nico
- Department of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Inyushin M, Kucheryavykh LY, Kucheryavykh YV, Nichols CG, Buono RJ, Ferraro TN, Skatchkov SN, Eaton MJ. Potassium channel activity and glutamate uptake are impaired in astrocytes of seizure-susceptible DBA/2 mice. Epilepsia 2010; 51:1707-13. [PMID: 20831751 DOI: 10.1111/j.1528-1167.2010.02592.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
PURPOSE KCNJ10 encodes subunits of inward rectifying potassium (Kir) channel Kir4.1 found predominantly in glial cells within the brain. Genetic inactivation of these channels in glia impairs extracellular K(+) and glutamate clearance and produces a seizure phenotype. In both mice and humans, polymorphisms and mutations in the KCNJ10 gene have been associated with seizure susceptibility. The purpose of the present study was to determine whether there are differences in Kir channel activity and potassium- and glutamate-buffering capabilities between astrocytes from seizure resistant C57BL/6 (B6) and seizure susceptible DBA/2 (D2) mice that are consistent with an altered K(+) channel activity as a result of genetic polymorphism of KCNJ10. METHODS Using cultured astrocytes and hippocampal brain slices together with whole-cell patch-clamp, we determined the electrophysiologic properties, particularly K(+) conductances, of B6 and D2 mouse astrocytes. Using a colorimetric assay, we determined glutamate clearance capacity by B6 and D2 astrocytes. RESULTS Barium-sensitive Kir currents elicited from B6 astrocytes are substantially larger than those elicited from D2 astrocytes. In addition, potassium and glutamate buffering by D2 cortical astrocytes is impaired, relative to buffering by B6 astrocytes. DISCUSSION In summary, the activity of Kir4.1 channels differs between seizure-susceptible D2 and seizure-resistant B6 mice. Reduced activity of Kir4.1 channels in astrocytes of D2 mice is associated with deficits in potassium and glutamate buffering. These deficits may, in part, explain the relatively low seizure threshold of D2 mice.
Collapse
Affiliation(s)
- Mikhail Inyushin
- Department of Physiology, Universidad Central del Caribe, School of Medicine, Bayamón, Puerto Rico 00960-6032, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Benfenati V, Toffanin S, Capelli R, Camassa LMA, Ferroni S, Kaplan DL, Omenetto FG, Muccini M, Zamboni R. A silk platform that enables electrophysiology and targeted drug delivery in brain astroglial cells. Biomaterials 2010; 31:7883-91. [PMID: 20688390 DOI: 10.1016/j.biomaterials.2010.07.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 07/04/2010] [Indexed: 01/26/2023]
Abstract
Astroglial cell survival and ion channel activity are relevant molecular targets for the mechanistic study of neural cell interactions with biomaterials and/or electronic interfaces. Astrogliosis is the most typical reaction to in vivo brain implants and needs to be avoided by developing biomaterials that preserve astroglial cell physiological function. This cellular phenomenon is characterized by a proliferative state and altered expression of astroglial potassium (K(+)) channels. Silk is a natural polymer with potential for new biomedical applications due to its ability to support in vitro growth and differentiation of many cell types. We report on silk interactions with cultured neocortical astroglial cells. Astrocytes survival is similar when plated on silk-coated glass and on poly-D-lysine (PDL), a well known polyionic substrate used to promote astroglial cell adhesion to glass surfaces. Comparative analyses of whole-cell patch-clamp experiments reveal that silk- and PDL-coated cells display depolarized resting membrane potentials (-40 mV), very high input resistance, and low specific conductance, with values similar to those of undifferentiated glial cells. Analysis of K(+) channel conductance reveals that silk-astrocytes express large outwardly delayed rectifying K(+) current (K(DR)). The magnitude of K(DR) in PDL- and silk-coated astrocytes is similar, indicating that silk does not alter the resting K(+) current. We also demonstrate that guanosine- (GUO) embedded silk enables the direct modulation of astroglial K(+) conductance in vitro. Astrocytes plated on GUO-embedded silk are more hyperpolarized and express inward rectifying K(+) conductance (K(ir)). The K(+) inward current increases and this is paralleled by upregulation and membrane polarization of K(ir)4.1 protein signal. Collectively these results indicate that silk is a suitable biomaterial platform for the in vitro studies of astroglial ion channel responses and related physiology.
Collapse
Affiliation(s)
- Valentina Benfenati
- Consiglio Nazionale delle Ricerche (CNR), Istituto per lo Studio dei Materiali Nanostrutturati (ISMN), Bologna, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Biological role of dystroglycan in Schwann cell function and its implications in peripheral nervous system diseases. J Biomed Biotechnol 2010; 2010:740403. [PMID: 20625412 PMCID: PMC2896880 DOI: 10.1155/2010/740403] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Accepted: 04/20/2010] [Indexed: 11/17/2022] Open
Abstract
Dystroglycan is a central component of the dystrophin-glycoprotein complex (DGC) that links extracellular matrix with cytoskeleton, expressed in a variety of fetal and adult tissues. Dystroglycan plays diverse roles in development and homeostasis including basement membrane formation, epithelial morphogenesis, membrane stability, cell polarization, and cell migration. In this paper, we will focus on biological role of dystroglycan in Schwann cell function, especially myelination. First, we review the molecular architecture of DGC in Schwann cell abaxonal membrane. Then, we will review the loss-of-function studies using targeted mutagenesis, which have revealed biological functions of each component of DGC in Schwann cells. Based on these findings, roles of dystroglycan in Schwann cell function, in myelination in particular, and its implications in diseases will be discussed in detail. Finally, in view of the fact that understanding the role of dystroglycan in Schwann cells is just beginning, future perspectives will be discussed.
Collapse
|
30
|
Benfenati V, Ferroni S. Water transport between CNS compartments: functional and molecular interactions between aquaporins and ion channels. Neuroscience 2009; 168:926-40. [PMID: 20026249 DOI: 10.1016/j.neuroscience.2009.12.017] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2009] [Revised: 12/04/2009] [Accepted: 12/05/2009] [Indexed: 12/16/2022]
Abstract
The physiological ability of the mammalian CNS to integrate peripheral stimuli and to convey information to the body is tightly regulated by its capacity to preserve the ion composition and volume of the perineuronal milieu. It is well known that astroglial syncytium plays a crucial role in such process by controlling the homeostasis of ions and water through the selective transmembrane movement of inorganic and organic molecules and the equilibration of osmotic gradients. Astrocytes, in fact, by contacting neurons and cells lining the fluid-filled compartments, are in a strategic position to fulfill this role. They are endowed with ion and water channel proteins that are localized in specific plasma membrane domains facing diverse liquid spaces. Recent data in rodents have demonstrated that the precise dynamics of the astroglia-mediated homeostatic regulation of the CNS is dependent on the interactions between water channels and ion channels, and their anchoring with proteins that allow the formation of macromolecular complexes in specific cellular domains. Interplay can occur with or without direct molecular interactions suggesting the existence of different regulatory mechanisms. The importance of molecular and functional interactions is pinpointed by the numerous observations that as consequence of pathological insults leading to the derangement of ion and volume homeostasis the cell surface expression and/or polarized localization of these proteins is perturbed. Here, we critically discuss the experimental evidence concerning: (1) molecular and functional interplay of aquaporin 4, the major aquaporin protein in astroglial cells, with potassium and gap-junctional channels that are involved in extracellular potassium buffering. (2) the interactions of aquaporin 4 with chloride and calcium channels regulating cell volume homeostasis. The relevance of the crosstalk between water channels and ion channels in the pathogenesis of astroglia-related acute and chronic diseases of the CNS is also briefly discussed.
Collapse
Affiliation(s)
- V Benfenati
- Istituto per lo Studio dei Materiali Nanostrutturati, ISMN, National Research Council, Via Gobetti 101, 40129 Bologna, Italy
| | | |
Collapse
|
31
|
Panagis L, Zhao X, Ge Y, Ren L, Mittag TW, Danias J. Gene expression changes in areas of focal loss of retinal ganglion cells in the retina of DBA/2J mice. Invest Ophthalmol Vis Sci 2009; 51:2024-34. [PMID: 19737878 DOI: 10.1167/iovs.09-3560] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose. To determine whether differences in gene expression occur between areas of focal retinal ganglion cell (RGC) loss and of relative RGC preservation in the DBA/2 mouse retina and whether they can provide insight into the pathophysiology of glaucoma. Methods. Areas of focal RGC loss (judged by lack of Fluorogold labeling; Fluorochrome, Denver, CO), adjacent areas with relative RGC preservation in DBA/2 retina, and Fluorogold-labeled retina from DBA/2(-pe) (pearl) mice were dissected and used for microarray analysis. RT-PCR and immunoblot analysis were used to confirm differential gene expression. Bioinformatic analysis was used to identify gene networks affected in the glaucomatous retina. Results. Microarray analysis identified 372 and 115 gene chip IDs as up- and downregulated, respectively, by 0.5-fold in areas of RGC loss. Differentially expressed genes included those coding for cytoskeletal proteins, enzymes, transport proteins, extracellular matrix (ECM) proteins, and immune response proteins. Several genes were confirmed by RT-PCR. For at least two genes, differential protein expression was verified. Bioinformatics analysis identified multiple affected functional gene networks. Pearl mice appeared to have significantly different gene expression, even when compared with relatively preserved areas of the DBA/2 retina. Conclusions. Regional gene expression changes occur in areas of focal RGC loss in the DBA/2 retina. The genes involved code for proteins with diverse cellular functions. Further investigation is needed to determine the cellular localization of the expression of these genes during the development of spontaneous glaucoma in the DBA/2 mouse and to determine whether some of these gene expression changes are causative or protective of RGC loss.
Collapse
Affiliation(s)
- Lampros Panagis
- Departments of Ophthalmology, Mount Sinai School of Medicine, New York, New York, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Noël G, Tham DKL, Moukhles H. Interdependence of laminin-mediated clustering of lipid rafts and the dystrophin complex in astrocytes. J Biol Chem 2009; 284:19694-704. [PMID: 19451651 DOI: 10.1074/jbc.m109.010090] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Astrocyte endfeet surrounding blood vessels are active domains involved in water and potassium ion transport crucial to the maintenance of water and potassium ion homeostasis in brain. A growing body of evidence points to a role for dystroglycan and its interaction with perivascular laminin in the targeting of the dystrophin complex and the water-permeable channel, aquaporin 4 (AQP4), at astrocyte endfeet. However, the mechanisms underlying such compartmentalization remain poorly understood. In the present study we found that AQP4 resided in Triton X-100-insoluble fraction, whereas dystroglycan was recovered in the soluble fraction in astrocytes. Cholesterol depletion resulted in the translocation of a pool of AQP4 to the soluble fraction indicating that its distribution is indeed associated with cholesterol-rich membrane domains. Upon laminin treatment AQP4 and the dystrophin complex, including dystroglycan, reorganized into laminin-associated clusters enriched for the lipid raft markers GM1 and flotillin-1 but not caveolin-1. Reduced diffusion rates of GM1 in the laminin-induced clusters were indicative of the reorganization of raft components in these domains. In addition, both cholesterol depletion and dystroglycan silencing reduced the number and area of laminin-induced clusters of GM1, AQP4, and dystroglycan. These findings demonstrate the interdependence between laminin binding to dystroglycan and GM1-containing lipid raft reorganization and provide novel insight into the dystrophin complex regulation of AQP4 polarization in astrocytes.
Collapse
Affiliation(s)
- Geoffroy Noël
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver V6T 1Z3, Canada
| | | | | |
Collapse
|
33
|
KUCHERYAVYKH YURIYV, SHUBA YAROSLAVM, ANTONOV SERGEIM, INYUSHIN MIKHAILY, CUBANO LUIS, PEARSON WADEL, KURATA HARLEY, REICHENBACH ANDREAS, VEH RÜDIGERW, NICHOLS COLING, EATON MISTYJ, SKATCHKOV SERGUEIN. Complex rectification of Müller cell Kir currents. Glia 2008; 56:775-90. [PMID: 18293411 PMCID: PMC9930535 DOI: 10.1002/glia.20652] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Although Kir4.1 channels are the major inwardly rectifying channels in glial cells and are widely accepted to support K+- and glutamate-uptake in the nervous system, the properties of Kir4.1 channels during vital changes of K+ and polyamines remain poorly understood. Therefore, the present study examined the voltage-dependence of K+ conductance with varying physiological and pathophysiological external [K+] and intrapipette spermine ([SP]) concentrations in Müller glial cells and in tsA201 cells expressing recombinant Kir4.1 channels. Two different types of [SP] block were characterized: "fast" and "slow." Fast block was steeply voltage-dependent, with only a low sensitivity to spermine and strong dependence on extracellular potassium concentration, [K+]o. Slow block had a strong voltage sensitivity that begins closer to resting membrane potential and was essentially [K+]o-independent, but with a higher spermine- and [K+]i-sensitivity. Using a modified Woodhull model and fitting i/V curves from whole cell recordings, we have calculated free [SP](in) in Müller glial cells as 0.81 +/- 0.24 mM. This is much higher than has been estimated previously in neurons. Biphasic block properties underlie a significantly varying extent of rectification with [K+] and [SP]. While confirming similar properties of glial Kir and recombinant Kir4.1, the results also suggest mechanisms underlying K+ buffering in glial cells: When [K+]o is rapidly increased, as would occur during neuronal excitation, "fast block" would be relieved, promoting potassium influx to glial cells. Increase in [K+]in would then lead to relief of "slow block," further promoting K+-influx.
Collapse
Affiliation(s)
- YURIY V. KUCHERYAVYKH
- Department of Biochemistry, Universidad Central del Caribe, School of Medicine, Bayamón, PR
| | - YAROSLAV M. SHUBA
- International Center of Molecular Physiology, National Academy of Sciences of Ukraine, Kiev, 01024 Ukraine
| | - SERGEI M. ANTONOV
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, 194223 Russia
| | - MIKHAIL Y. INYUSHIN
- Department of Physiology, Universidad Central del Caribe, School of Medicine, Bayamón, PR
| | - LUIS CUBANO
- Department of Anatomy and Cell Biology, Universidad Central del Caribe, School of Medicine, Bayamón, PR
| | - WADE L. PEARSON
- Department of Cell Biology and Physiology, Washington University, School of Medicine, St. Louis, MO
| | - HARLEY KURATA
- Department of Cell Biology and Physiology, Washington University, School of Medicine, St. Louis, MO
| | - ANDREAS REICHENBACH
- Paul Flechsig Institute of Brain Research, Leipzig University, D-04109 Leipzig, Germany
| | - RÜDIGER W. VEH
- Institute of Integrative Neuroanatomy, Charité, Philipstrasse 12, Berlin, Germany
| | - COLIN G. NICHOLS
- Department of Cell Biology and Physiology, Washington University, School of Medicine, St. Louis, MO
| | - MISTY J. EATON
- Department of Biochemistry, Universidad Central del Caribe, School of Medicine, Bayamón, PR
| | - SERGUEI N. SKATCHKOV
- Department of Biochemistry, Universidad Central del Caribe, School of Medicine, Bayamón, PR,Department of Physiology, Universidad Central del Caribe, School of Medicine, Bayamón, PR,Correspondence to: S. N. Skatchkov, Departments of Biochemistry and Physiology, School of Medicine, Universidad Central del Caribe, Box 60-327, Bayamón, Puerto Rico, USA 00960-6032.
| |
Collapse
|
34
|
Fort PE, Sene A, Pannicke T, Roux MJ, Forster V, Mornet D, Nudel U, Yaffe D, Reichenbach A, Sahel JA, Rendon A. Kir4.1 and AQP4 associate with Dp71- and utrophin-DAPs complexes in specific and defined microdomains of Müller retinal glial cell membrane. Glia 2008; 56:597-610. [DOI: 10.1002/glia.20633] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
35
|
Proliferative gliosis causes mislocation and inactivation of inwardly rectifying K+ (Kir) channels in rabbit retinal glial cells. Exp Eye Res 2008; 86:305-13. [DOI: 10.1016/j.exer.2007.11.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2007] [Revised: 09/26/2007] [Accepted: 11/02/2007] [Indexed: 11/20/2022]
|
36
|
Rurak J, Noel G, Lui L, Joshi B, Moukhles H. Distribution of potassium ion and water permeable channels at perivascular glia in brain and retina of the Large(myd) mouse. J Neurochem 2007; 103:1940-53. [PMID: 17803675 DOI: 10.1111/j.1471-4159.2007.04886.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The dystroglycan protein complex provides a link between the cytoskeleton and the extracellular matrix (ECM). Defective O-glycosylation of alpha-dystroglycan (alpha-DG) severs this link leading to muscular dystrophies named dystroglycanopathies. These are characterized not only by muscle degeneration, but also by brain and ocular defects. In brain and retina, alpha-DG and ECM molecules are enriched around blood vessels where they may be involved in localizing the inwardly rectifying potassium channel, Kir4.1, and aquaporin channel, AQP4, to astrocytic endfeet. To investigate in vivo the role of ECM ligand-binding to glycosylated sites on alpha-DG in the polarized distribution of these channels, we used the Large(myd) mouse, an animal model for dystroglycanopathies. We found that Kir4.1 and AQP4 are lost from astrocytic endfeet in brain whereas significant labeling for these channels is detected at similar cell domains in retina. Furthermore, while both alpha- and beta1-syntrophins are lost from perivascular astrocytes in brain, labeling for beta1-syntrophin is found in retina of the Large(myd) mouse. These findings show that while ligand-binding to the highly glycosylated isoform of alpha-DG in concert with alpha- and beta1-syntrophins is crucial for the polarized distribution of Kir4.1 and AQP4 to functional domains in brain, distinct mechanisms may contribute to their localization in retina.
Collapse
Affiliation(s)
- Jennifer Rurak
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | |
Collapse
|
37
|
Dénes V, Witkovsky P, Koch M, Hunter DD, Pinzón-Duarte G, Brunken WJ. Laminin deficits induce alterations in the development of dopaminergic neurons in the mouse retina. Vis Neurosci 2007; 24:549-62. [PMID: 17711601 PMCID: PMC2935900 DOI: 10.1017/s0952523807070514] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2007] [Accepted: 05/17/2007] [Indexed: 11/05/2022]
Abstract
Genetically modified mice lacking the beta2 laminin chain (beta2null), the gamma3 laminin chain (gamma3 null), or both beta2/gamma3 chains (compound null) were produced. The development of tyrosine hydroxylase (TH) immunoreactive neurons in these mouse lines was studied between birth and postnatal day (P) 20. Compared to wild type mice, no alterations were seen in gamma3 null mice. In beta2 null mice, however, the large, type I TH neurons appeared later in development, were at a lower density and had reduced TH immunoreactivity, although TH process number and size were not altered. In the compound null mouse, the same changes were observed together with reduced TH process outgrowth. Surprisingly, in the smaller, type II TH neurons, TH immunoreactivity was increased in laminin-deficient compared to wild type mice. Other retinal defects we observed were a patchy disruption of the inner limiting retinal basement membrane and a disoriented growth of Müller glial cells. Starburst and AII type amacrine cells were not apparently altered in laminin-deficient relative to wild type mice. We postulate that laminin-dependent developmental signals are conveyed to TH amacrine neurons through intermediate cell types, perhaps the Müller glial cell and/or the retinal ganglion cell.
Collapse
Affiliation(s)
- Viktória Dénes
- Department of Anatomy and Cellular Biology, Tufts University School of Medicine, Boston, Massachusetts
- Tufts Center for Vision Research, Boston, Massachusetts
| | - Paul Witkovsky
- Department of Ophthalmology, New York University School of Medicine, New York, New York
| | - Manuel Koch
- Center for Biochemistry and Department of Dermatology, University of Köln, Köln, Germany
| | | | - Germán Pinzón-Duarte
- Department of Anatomy and Cellular Biology, Tufts University School of Medicine, Boston, Massachusetts
- Tufts Center for Vision Research, Boston, Massachusetts
| | - William J. Brunken
- Department of Anatomy and Cellular Biology, Tufts University School of Medicine, Boston, Massachusetts
- Tufts Center for Vision Research, Boston, Massachusetts
| |
Collapse
|
38
|
Bragg AD, Amiry-Moghaddam M, Ottersen OP, Adams ME, Froehner SC. Assembly of a perivascular astrocyte protein scaffold at the mammalian blood–brain barrier is dependent on α-syntrophin. Glia 2006; 53:879-90. [PMID: 16609960 DOI: 10.1002/glia.20347] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
alpha-Syntrophin, a member of the dystrophin-associated protein complex, is required for proper localization of the water channel aquaporin-4 at the blood-brain barrier. Mice lacking alpha-syntrophin have reduced levels of aquaporin-4 in perivascular astroglial endfeet. Consequently, they exhibit reduced edema and infarct volume in brain trauma models and reduced K+ clearance from the neuropil, leading to increased seizure susceptibility. We have used the alpha-syntrophin null mice to investigate whether alpha-syntrophin is required for proper localization of other components of the dystrophin complex at the blood-brain barrier. We find that alpha-syntrophin is required for the full recruitment of gamma2-syntrophin and alpha-dystrobrevin-2 to glial endfeet in adult cerebellum. In contrast, the localization of beta1- and beta2-syntrophin and alpha-dystrobrevin-1 at the blood-brain barrier is not dependent on the presence of alpha-syntrophin. The localization patterns of alpha-dystrobrevin-1 and -2 in wild type cerebellum are strikingly different; while alpha-dystrobrevin-1 is present in glial endfeet throughout the cerebellum, alpha-dystrobrevin-2 is restricted to glial endfeet in the granular layer alone. Finally, we show that the enrichment of dystrophin in glial endfeet depends on the presence of alpha-syntrophin. This finding is the first demonstration that dystrophin localization is dependent on syntrophin. Since the localization of gamma2-syntrophin, alpha-dystrobrevin-2, and dystrophin is contingent on alpha-syntrophin, we conclude that alpha-syntrophin is a central organizer of the astrocyte dystrophin complex, an important molecular scaffold for localization of aquaporin-4 at the blood-brain barrier.
Collapse
Affiliation(s)
- April D Bragg
- Department of Physiology and Biophysics, Health Sciences Building, Rm G424, 1959 NE Pacific St, University of Washington, Seattle, 98195, USA.
| | | | | | | | | |
Collapse
|