1
|
Miao Y, Zhao GL, Cheng S, Wang Z, Yang XL. Activation of retinal glial cells contributes to the degeneration of ganglion cells in experimental glaucoma. Prog Retin Eye Res 2023; 93:101169. [PMID: 36736070 DOI: 10.1016/j.preteyeres.2023.101169] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/12/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023]
Abstract
Elevation of intraocular pressure (IOP) is a major risk factor for neurodegeneration in glaucoma. Glial cells, which play an important role in normal functioning of retinal neurons, are well involved into retinal ganglion cell (RGC) degeneration in experimental glaucoma animal models generated by elevated IOP. In response to elevated IOP, mGluR I is first activated and Kir4.1 channels are subsequently inhibited, which leads to the activation of Müller cells. Müller cell activation is followed by a complex process, including proliferation, release of inflammatory and growth factors (gliosis). Gliosis is further regulated by several factors. Activated Müller cells contribute to RGC degeneration through generating glutamate receptor-mediated excitotoxicity, releasing cytotoxic factors and inducing microglia activation. Elevated IOP activates microglia, and following morphological and functional changes, these cells, as resident immune cells in the retina, show adaptive immune responses, including an enhanced release of pro-inflammatory factors (tumor neurosis factor-α, interleukins, etc.). These ATP and Toll-like receptor-mediated responses are further regulated by heat shock proteins, CD200R, chemokine receptors, and metabotropic purinergic receptors, may aggravate RGC loss. In the optic nerve head, astrogliosis is initiated and regulated by a complex reaction process, including purines, transmitters, chemokines, growth factors and cytokines, which contributes to RGC axon injury through releasing pro-inflammatory factors and changing extracellular matrix in glaucoma. The effects of activated glial cells on RGCs are further modified by the interplay among different types of glial cells. This review is concluded by presenting an in-depth discussion of possible research directions in this field in the future.
Collapse
Affiliation(s)
- Yanying Miao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Guo-Li Zhao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Shuo Cheng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhongfeng Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Xiong-Li Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
2
|
Goldberg Z, Sher I, Qassim L, Chapman J, Rotenstreich Y, Shavit-Stein E. Intrinsic Expression of Coagulation Factors and Protease Activated Receptor 1 (PAR1) in Photoreceptors and Inner Retinal Layers. Int J Mol Sci 2022; 23:ijms23020984. [PMID: 35055169 PMCID: PMC8778890 DOI: 10.3390/ijms23020984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 12/19/2022] Open
Abstract
The aim of this study was to characterize the distribution of the thrombin receptor, protease activated receptor 1 (PAR1), in the neuroretina. Neuroretina samples of wild-type C57BL/6J and PAR1−/− mice were processed for indirect immunofluorescence and Western blot analysis. Reverse transcription quantitative real-time PCR (RT-qPCR) was used to determine mRNA expression of coagulation Factor X (FX), prothrombin (PT), and PAR1 in the isolated neuroretina. Thrombin activity following KCl depolarization was assessed in mouse neuroretinas ex vivo. PAR1 staining was observed in the retinal ganglion cells, inner nuclear layer cells, and photoreceptors in mouse retinal cross sections by indirect immunofluorescence. PAR1 co-localized with rhodopsin in rod outer segments but was not expressed in cone outer segments. Western blot analysis confirmed PAR1 expression in the neuroretina. Factor X, prothrombin, and PAR1 mRNA expression was detected in isolated neuroretinas. Thrombin activity was elevated by nearly four-fold in mouse neuroretinas following KCl depolarization (0.012 vs. 0.044 mu/mL, p = 0.0497). The intrinsic expression of coagulation factors in the isolated neuroretina together with a functional increase in thrombin activity following KCl depolarization may suggest a role for the PAR1/thrombin pathway in retinal function.
Collapse
Affiliation(s)
- Zehavit Goldberg
- Goldschleger Eye Institute, Sheba Medical Center, Ramat Gan 5266202, Israel; (Z.G.); (I.S.); (Y.R.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ifat Sher
- Goldschleger Eye Institute, Sheba Medical Center, Ramat Gan 5266202, Israel; (Z.G.); (I.S.); (Y.R.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Lamis Qassim
- Department of Neurology, Sheba Medical Center, Ramat Gan 5266202, Israel; (L.Q.); (J.C.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Joab Chapman
- Department of Neurology, Sheba Medical Center, Ramat Gan 5266202, Israel; (L.Q.); (J.C.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Robert and Martha Harden Chair in Mental and Neurological Diseases, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ygal Rotenstreich
- Goldschleger Eye Institute, Sheba Medical Center, Ramat Gan 5266202, Israel; (Z.G.); (I.S.); (Y.R.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Efrat Shavit-Stein
- Department of Neurology, Sheba Medical Center, Ramat Gan 5266202, Israel; (L.Q.); (J.C.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Correspondence: ; Fax: +972-3-530-4409
| |
Collapse
|
3
|
Li Q, Cheng Y, Zhang S, Sun X, Wu J. TRPV4-induced Müller cell gliosis and TNF-α elevation-mediated retinal ganglion cell apoptosis in glaucomatous rats via JAK2/STAT3/NF-κB pathway. J Neuroinflammation 2021; 18:271. [PMID: 34789280 PMCID: PMC8596927 DOI: 10.1186/s12974-021-02315-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 11/01/2021] [Indexed: 01/22/2023] Open
Abstract
Background Glaucoma, the leading cause of irreversible blindness worldwide, is a type of retinal disease characterized by the selective death of retinal ganglion cells (RGCs). However, the pathogenesis of glaucoma has not been fully elucidated. Transient receptor potential vanilloid 4 (TRPV4) is a pressure-sensitive and calcium-permeable cation channel. TRPV4 is widely distributed in the retina and its sustained activation leads to RGC death; indicating that TRPV4 may be a possible target for glaucoma treatment. Here, we investigated the effects of TRPV4 on RGC apoptosis in a rat model of chronic ocular hypertension (COH), then examined the mechanism underlying these effects. Methods The COH model was established by injection of micro-magnetic beads into the anterior chamber of adult male rats. The expression levels of TRPV4, glial fibrillary acidic protein, and inflammatory factors were assessed by immunohistochemistry and immunoblotting. RGC apoptosis and visual dysfunction were evaluated by TUNEL assay and photopic negative response. Functional expression of TRPV4 was examined by electrophysiology and calcium imaging. Real-time polymerase chain reaction and immunoblotting were employed to investigate the molecular mechanism underlying the effects of TRPV4 on tumor necrosis factor-α (TNF-α) release. Results We found that TRPV4 played an essential role in glaucoma, such that high levels of TRPV4 expression were associated with elevated intraocular pressure. Furthermore, TRPV4 activation was involved in glaucoma-induced RGC apoptosis and RGC-related reductions in visual function. Mechanistic investigation demonstrated that TRPV4 activation led to enhanced Müller cell gliosis and TNF-α release via the JAK2/STAT3/NF-kB pathway, while TRPV4 inhibition could reverse these effects. Finally, TRPV4 activation could lead to elevated expression of TNF receptor 1 in RGCs, while inhibition of TNF-α could reduce TRPV4-mediated RGC apoptosis. Conclusions TRPV4 activation induces Müller cell gliosis and TNF-α elevation via the JAK2/STAT3/NF-κB pathway, which may exacerbate RGC apoptosis in glaucoma; these results suggest that TRPV4 can serve as a therapeutic target in glaucoma treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02315-8.
Collapse
Affiliation(s)
- Qian Li
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, National Health Commission, #83 Fenyang Road, 200031, Shanghai, China
| | - Yun Cheng
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, National Health Commission, #83 Fenyang Road, 200031, Shanghai, China
| | - Shenghai Zhang
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, National Health Commission, #83 Fenyang Road, 200031, Shanghai, China
| | - Xinghuai Sun
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China. .,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China. .,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China. .,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, National Health Commission, #83 Fenyang Road, 200031, Shanghai, China.
| | - Jihong Wu
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China. .,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China. .,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China. .,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, National Health Commission, #83 Fenyang Road, 200031, Shanghai, China.
| |
Collapse
|
4
|
Reichenbach A, Bringmann A. Glia of the human retina. Glia 2019; 68:768-796. [PMID: 31793693 DOI: 10.1002/glia.23727] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 12/22/2022]
Abstract
The human retina contains three types of glial cells: microglia and two types of macroglia, astrocytes and Müller cells. Macroglia provide homeostatic and metabolic support to photoreceptors and neurons required for neuronal activity. The fovea, the site of the sharpest vision which is astrocyte- and microglia-free, contains two populations of Müller glia: cells which form the Müller cell cone in the foveola and z-shaped Müller cells of the foveal walls. Both populations are characterized by morphological and functional differences. Müller cells of the foveola do not support the activity of photoreceptors and neurons, but provide the structural stability of the foveal tissue and improve the light transmission through the tissue to the photoreceptors. This article gives overviews of the glia of the human retina and the structure and function of both Müller cell types in the fovea, and describes the contributions of astrocytes and Müller cells to the ontogenetic development of the fovea.
Collapse
Affiliation(s)
- Andreas Reichenbach
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| |
Collapse
|
5
|
Wu HJ, Li XY, Qian WJ, Li Q, Wang SY, Ji M, Ma YY, Gao F, Sun XH, Wang X, Miao Y, Yang XL, Wang Z. Dopamine D1 receptor-mediated upregulation of BKCa
currents modifies Müller cell gliosis in a rat chronic ocular hypertension model. Glia 2018; 66:1507-1519. [PMID: 29508439 DOI: 10.1002/glia.23321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 02/15/2018] [Accepted: 02/19/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Hang-Jing Wu
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Xue-Yan Li
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Wen-Jing Qian
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Qian Li
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Shu-Yue Wang
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Min Ji
- Department of Ophthalmology at Eye & ENT Hospital; Fudan University; Shanghai 200031 China
| | - Yuan-Yuan Ma
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Feng Gao
- Department of Ophthalmology at Eye & ENT Hospital; Fudan University; Shanghai 200031 China
| | - Xing-Huai Sun
- Department of Ophthalmology at Eye & ENT Hospital; Fudan University; Shanghai 200031 China
| | - Xin Wang
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Yanying Miao
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Xiong-Li Yang
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Zhongfeng Wang
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| |
Collapse
|
6
|
Pannicke T, Wagner L, Reichenbach A, Grosche A. Electrophysiological characterization of Müller cells from the ischemic retina of mice deficient in the leukemia inhibitory factor. Neurosci Lett 2018; 670:69-74. [PMID: 29391217 DOI: 10.1016/j.neulet.2018.01.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/23/2018] [Accepted: 01/24/2018] [Indexed: 12/24/2022]
Abstract
Leukemia inhibitory factor (LIF) is a cytokine that exerts different effects in the nervous system. It is involved in neuronal injuries and diseases and is assumed to be neuroprotective and to regulate reactive gliosis. In LIF-deficient (LIF-/-) mice, expression of glial fibrillary acidic protein in retinal Müller glial cells as a hallmark of reactive gliosis is suppressed during retinal degenerations. Here, we detected expression of LIF and its receptors in Müller cells of the murine retina. Moreover, electrophysiological alterations of Müller cells 7 days after transient retinal ischemia were studied by the patch-clamp technique. The amplitude of inward currents in Müller cells from the postischemic retina was reduced to 51% in wild type and to 70% in LIF-/- mice. This demonstrates that decrease of inward currents takes place in reactive Müller cells even in the absence of LIF.
Collapse
Affiliation(s)
- Thomas Pannicke
- Paul-Flechsig-Institut für Hirnforschung, Universität Leipzig, Liebigstr. 19, D-04103 Leipzig, Germany.
| | - Lysann Wagner
- Paul-Flechsig-Institut für Hirnforschung, Universität Leipzig, Liebigstr. 19, D-04103 Leipzig, Germany
| | - Andreas Reichenbach
- Paul-Flechsig-Institut für Hirnforschung, Universität Leipzig, Liebigstr. 19, D-04103 Leipzig, Germany
| | - Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Großhaderner Str. 9, D-82152 Planegg-Martinsried, Germany
| |
Collapse
|
7
|
Evangelho K, Mogilevskaya M, Losada-Barragan M, Vargas-Sanchez JK. Pathophysiology of primary open-angle glaucoma from a neuroinflammatory and neurotoxicity perspective: a review of the literature. Int Ophthalmol 2017; 39:259-271. [PMID: 29290065 DOI: 10.1007/s10792-017-0795-9] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/11/2017] [Indexed: 11/29/2022]
Abstract
PURPOSE Glaucoma is the leading cause of blindness in humans, affecting 2% of the population. This disorder can be classified into various types including primary, secondary, glaucoma with angle closure and with open angle. The prevalence of distinct types of glaucoma differs for each particular region of the world. One of the most common types of this disease is primary open-angle glaucoma (POAG), which is a complex inherited disorder characterized by progressive retinal ganglion cell death, optic nerve head excavation and visual field loss. Nowadays, POAG is considered an optic neuropathy, while intraocular pressure is proposed to play a fundamental role in its pathophysiology and especially in optic disk damage. However, the exact mechanism of optic nerve head damage remains a topic of debate. This literature review aims to bring together the information on the pathophysiology of primary open-angle glaucoma, particularly focusing on neuroinflammatory mechanisms leading to the death of the retinal ganglion cell. METHODS A literature search was done on PubMed using key words including primary open-angle glaucoma, retinal ganglion cells, Müller cells, glutamate, glial cells, ischemia, hypoxia, exitotoxicity, neuroinflammation, axotomy and neurotrophic factors. The literature was reviewed to collect the information published about the pathophysiologic mechanisms of RGC death in the POAG, from a neuroinflammatory and neurotoxicity perspective. RESULTS Proposed mechanisms for glaucomatous damage are a result of pressure in RGC followed by ischemia, hypoxia of the ONH, and consequently death due to glutamate-induced excitotoxicity, deprivation of energy and oxygen, increase in levels of inflammatory mediators and alteration of trophic factors flow. These events lead to blockage of anterograde and retrograde axonal transport with ensuing axotomy and eventually blindness. CONCLUSIONS The damage to ganglion cells and eventually glaucomatous injury can occur via various mechanisms including baric trauma, ischemia and impact of metabolic toxins, which triggers an inflammatory process and secondary degeneration in the ONH.
Collapse
Affiliation(s)
- Karine Evangelho
- Grupo de Investigación en Ciencias Biomédicas GRINCIBIO, Facultad de medicina, Sede Bogotá, Universidad Antonio Nariño, Bogotá, Colombia
| | - Maria Mogilevskaya
- Grupo de Investigación en Ingeniería Clínica - Hospital Universitario la Samaritana GINIC-HUS, Sede Bogotá, ECCI, Bogotá, Colombia
| | - Monica Losada-Barragan
- Grupo de Biología Celular y Funcional e Ingeniería de Biomoléculas, Facultad de Ciencias, Sede Bogotá, Universidad Antonio Nariño, Bogotá, Colombia
| | - Jeinny Karina Vargas-Sanchez
- Grupo de Investigación en Ciencias Biomédicas GRINCIBIO, Facultad de medicina, Sede Bogotá, Universidad Antonio Nariño, Bogotá, Colombia.
| |
Collapse
|
8
|
Abstract
Retinal Müller glial cells have been shown to undergo reactive gliosis in a variety of retinal diseases. Upregulation of glial fibrillary acidic protein (GFAP) is a hallmark of Müller cell activation. Reactive gliosis after retinal detachment or ischemia/reperfusion is characterized by hypertrophy and downregulation of inwardly rectifying K+ (Kir) currents. However, this kind of physiological alteration could not be detected in slowly progressing retinal degenerations. The photoreceptor toxin N-methyl-N-nitrosourea (MNU) leads to the rapid loss of cells in the outer nuclear layer and subsequent Müller cell activation. Here, we investigated whether Müller cells from MNU-treated mice exhibit reactive gliosis. We found that Müller cells showed increased GFAP expression and increased membrane capacitance, indicating hypertrophy. Membrane potential and Kir channel-mediated K+ currents were not significantly altered whereas Kir4.1 mRNA expression and Kir-mediated inward current densities were markedly decreased. This suggests that MNU-induced Müller cell gliosis is characterized by plasma membrane increase without alteration in the membrane content of Kir channels. Taken together, our findings show that Müller cells of MNU-treated mice are reactive and respond with a form of gliosis which is characterized by cellular hypertrophy but no changes in Kir current amplitudes.
Collapse
|
9
|
Involvement of the MEK-ERK/p38-CREB/c-fos signaling pathway in Kir channel inhibition-induced rat retinal Müller cell gliosis. Sci Rep 2017; 7:1480. [PMID: 28469203 PMCID: PMC5431154 DOI: 10.1038/s41598-017-01557-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/28/2017] [Indexed: 11/07/2022] Open
Abstract
Our previous studies have demonstrated that activation of group I metabotropic glutamate receptors downregulated Kir channels in chronic ocular hypertension (COH) rats, thus contributing to Müller cell gliosis, characterized by upregulated expression of glial fibrillary acidic protein (GFAP). In the present study, we explored possible signaling pathways linking Kir channel inhibition and GFAP upregulation. In normal retinas, intravitreal injection of BaCl2 significantly increased GFAP expression in Müller cells, which was eliminated by co-injecting mitogen-activated protein kinase (MAPK) inhibitor U0126. The protein levels of phosphorylated extracellular signal-regulated protein kinase1/2 (p-ERK1/2) and its upstream regulator, p-MEK, were significantly increased, while the levels of phosphorylated c-Jun N-terminal kinase (p-JNK) and p38 kinase (p-p38) remained unchanged. Furthermore, the protein levels of phosphorylated cAMP response element binding protein (p-CREB) and c-fos were also increased, which were blocked by co-injecting ERK inhibitor FR180204. In purified cultured rat Müller cells, BaCl2 treatment induced similar changes in these protein levels apart from p-p38 levels and the p-p38:p38 ratio showing significant upregulation. Moreover, intravitreal injection of U0126 eliminated the upregulated GFAP expression in COH retinas. Together, these results suggest that Kir channel inhibition-induced Müller cell gliosis is mediated by the MEK-ERK/p38-CREB/c-fos signaling pathway.
Collapse
|
10
|
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: 1.8] [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
|
11
|
Köller H, Fischer HG. Cytokines and Virus Proteins: Modulators of Glial Electrophysiological Properties. Neuroscientist 2016. [DOI: 10.1177/107385849900500310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cytokines are released during acute and chronic inflammatory diseases of the CNS and activate receptors on glial cells, thereby inducing various effects such as proliferation, expression of major histocompatibility complex genes or secretion of growth factors. Here, we summarize current evidence indicating that K+ currents, Ca 2+ currents, and the activity of ion transporters on astrocytes, microglial cells, and oligodendrocytes are also affected by cytokines. In disease states with associated elevated cytokine titers, such alterations in electrophysiological properties of glial cells might contribute to the patho genesis of neurological symptoms. NEUROSCIENTIST 5:142-146, 1999
Collapse
|
12
|
Reichenbach A, Bringmann A. Purinergic signaling in retinal degeneration and regeneration. Neuropharmacology 2015; 104:194-211. [PMID: 25998275 DOI: 10.1016/j.neuropharm.2015.05.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/07/2015] [Accepted: 05/07/2015] [Indexed: 02/01/2023]
Abstract
Purinergic signaling is centrally involved in mediating the degeneration of the injured and diseased retina, the induction of retinal gliosis, and the protection of the retinal tissue from degeneration. Dysregulated calcium signaling triggered by overactivation of P2X7 receptors is a crucial step in the induction of neuronal and microvascular cell death under pathogenic conditions like ischemia-hypoxia, elevated intraocular pressure, and diabetes, respectively. Overactivation of P2X7 plays also a pathogenic role in inherited and age-related photoreceptor cell death and in the age-related dysfunction and degeneration of the retinal pigment epithelium. Gliosis of micro- and macroglial cells, which is induced and/or modulated by purinergic signaling and associated with an impaired homeostatic support to neurons, and the ATP-mediated propagation of retinal gliosis from a focal injury into the surrounding noninjured tissue are involved in inducing secondary cell death in the retina. On the other hand, alterations in the glial metabolism of extracellular nucleotides, resulting in a decreased level of ATP and an increased level of adenosine, may be neuroprotective in the diseased retina. Purinergic signals stimulate the proliferation of retinal glial cells which contributes to glial scarring which has protective effects on retinal degeneration and adverse effects on retinal regeneration. Pharmacological modulation of purinergic receptors, e.g., inhibition of P2X and activation of adenosine receptors, may have clinical importance for the prevention of photoreceptor, neuronal, and microvascular cell death in diabetic retinopathy, retinitis pigmentosa, age-related macular degeneration, and glaucoma, respectively, for the clearance of retinal edema, and the inhibition of dysregulated cell proliferation in proliferative retinopathies. This article is part of a Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- Andreas Reichenbach
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany.
| | - Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| |
Collapse
|
13
|
Gao L, Chen X, Tang Y, Zhao J, Li Q, Fan X, Xu H, Yin ZQ. Neuroprotective effect of memantine on the retinal ganglion cells of APPswe/PS1ΔE9 mice and its immunomodulatory mechanisms. Exp Eye Res 2015; 135:47-58. [PMID: 25912193 DOI: 10.1016/j.exer.2015.04.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/20/2015] [Accepted: 04/21/2015] [Indexed: 01/16/2023]
Abstract
Besides the cognitive impairment and degeneration in the brain, vision dysfunction and retina damage are always prevalent in patients with Alzheimer's disease (AD). The uncompetitive antagonist of the N-methyl-d-aspartate receptor, memantine (MEM), has been proven to improve the cognition of patients with AD. However, limited information exists regarding the mechanism of neurodegeneration and the possible neuroprotective mechanisms of MEM on the retinas of patients with AD. In the present study, by using APPswe/PS1ΔE9 double transgenic (dtg) mice, we found that MEM rescued the loss of retinal ganglion cells (RGCs), as well as improved visual impairments, including improving the P50 component in pattern electroretinograms and the latency delay of the P2 component in flash visual evoked potentials of APPswe/PS1ΔE9 dtg mice. The activated microglia in the retinas of APPswe/PS1ΔE9 dtg mice were also inhibited by MEM. Additionally, the level of glutamine synthetase expressed by Müller cells within the RGC layer was upregulated in APPswe/PS1ΔE9 dtg mice, which was inhibited by MEM. Simultaneously, MEM also reduced the apoptosis of choline acetyl transferase-immunoreactive cholinergic amacrine cells within the RGC layer of AD mice. Moreover, the phosphorylation level of extracellular regulated protein kinases 1 and 2 was increased in APPswe/PS1ΔE9 dtg mice, which was blocked by MEM treatment. These findings suggest that MEM protects RGCs in the retinas of APPswe/PS1ΔE9 dtg mice by modulating the immune response of microglia and the adapted response of Müller cells, making MEM a potential ophthalmic treatment alternative in patients with AD.
Collapse
Affiliation(s)
- Lixiong Gao
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China; Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing 400038, China.
| | - Xi Chen
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China; School of Medicine, Nankai University, Tianjin 300071, China; Department of Ophthalmology, Chinese People's Liberation Army General Hospital, Beijing 100853, China.
| | - Yongping Tang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China.
| | - Jinghui Zhao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing 400038, China.
| | - Qiyou Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China.
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing 400038, China.
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China.
| | - Zheng Qin Yin
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China.
| |
Collapse
|
14
|
|
15
|
Gao F, Li F, Miao Y, Dong LD, Zhang SH, Wu J, Sun XH, Wang Z. Group I metabotropic glutamate receptor agonist DHPG modulates Kir4.1 protein and mRNA in cultured rat retinal Müller cells. Neurosci Lett 2014; 588:12-7. [PMID: 25549543 DOI: 10.1016/j.neulet.2014.12.048] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 12/20/2014] [Accepted: 12/23/2014] [Indexed: 11/30/2022]
Abstract
Müller cell gliosis is a general response in a variety of pathological alternations of the retina, which is characterized by the upregulated expression of glial fibrillary acidic protein (GFAP) and the downregulation of membrane K(+) conductance. We have demonstrated that downregulation of Kir K(+) currents in Müller cells in an experimental glaucoma model is due to activation of group I metabotropic glutamate receptor (mGluR I) by glutamate, which contributes to Müller cell gliosis. Here, whether and how activation of mGluR I modulate membrane Kir4.1 protein internalization and Kir4.1 mRNA expression were investigated in purified cultured rat retinal Müller cells using immunocytochemistry, Western blot and real-time PCR techniques. DHPG (10μM, a selective mGluR I agonist) treatment induced Müller cell gliosis, as evidenced by enhanced GFAP expression. Although total Kir4.1 proteins extracted from the DHPG-treated cells kept unchanged, Kir4.1 proteins in the cell membrane compartment were significantly decreased, which was prior to the change of GFAP in time course. In addition, DHPG (10 and 100μM) treatment induced a transient decrease in Kir4.1 mRNA expression in the cells. All these results suggest that activation of mGluR I by DHPG may decrease the number of functional Kir4.1 channels in purified cultured rat retinal Müller cells through modulating Kir4.1 protein and mRNA, thus contributing to Müller cell gliosis.
Collapse
Affiliation(s)
- Feng Gao
- Department of Ophthalmology at Eye and ENT hospital, Fudan University, Shanghai 200031, China; Institutes of Brain Science, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China
| | - Fang Li
- Institutes of Brain Science, Fudan University, Shanghai 200031, China; Institute of Neurobiology, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Yanying Miao
- Institutes of Brain Science, Fudan University, Shanghai 200031, China; Institute of Neurobiology, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Ling-Dan Dong
- Institutes of Brain Science, Fudan University, Shanghai 200031, China; Institute of Neurobiology, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Sheng-Hai Zhang
- Department of Ophthalmology at Eye and ENT hospital, Fudan University, Shanghai 200031, China; Institutes of Brain Science, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China
| | - Jihong Wu
- Department of Ophthalmology at Eye and ENT hospital, Fudan University, Shanghai 200031, China; Institutes of Brain Science, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China.
| | - Xing-Huai Sun
- Department of Ophthalmology at Eye and ENT hospital, Fudan University, Shanghai 200031, China; Institutes of Brain Science, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China
| | - Zhongfeng Wang
- Department of Ophthalmology at Eye and ENT hospital, Fudan University, Shanghai 200031, China; Institutes of Brain Science, Fudan University, Shanghai 200031, China; Institute of Neurobiology, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China.
| |
Collapse
|
16
|
Zayas-Santiago A, Agte S, Rivera Y, Benedikt J, Ulbricht E, Karl A, Dávila J, Savvinov A, Kucheryavykh Y, Inyushin M, Cubano LA, Pannicke T, Veh RW, Francke M, Verkhratsky A, Eaton MJ, Reichenbach A, Skatchkov SN. Unidirectional photoreceptor-to-Müller glia coupling and unique K+ channel expression in Caiman retina. PLoS One 2014; 9:e97155. [PMID: 24831221 PMCID: PMC4022631 DOI: 10.1371/journal.pone.0097155] [Citation(s) in RCA: 19] [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: 02/13/2014] [Accepted: 04/15/2014] [Indexed: 02/07/2023] Open
Abstract
Background Müller cells, the principal glial cells of the vertebrate retina, are fundamental for the maintenance and function of neuronal cells. In most vertebrates, including humans, Müller cells abundantly express Kir4.1 inwardly rectifying potassium channels responsible for hyperpolarized membrane potential and for various vital functions such as potassium buffering and glutamate clearance; inter-species differences in Kir4.1 expression were, however, observed. Localization and function of potassium channels in Müller cells from the retina of crocodiles remain, hitherto, unknown. Methods We studied retinae of the Spectacled caiman (Caiman crocodilus fuscus), endowed with both diurnal and nocturnal vision, by (i) immunohistochemistry, (ii) whole-cell voltage-clamp, and (iii) fluorescent dye tracing to investigate K+ channel distribution and glia-to-neuron communications. Results Immunohistochemistry revealed that caiman Müller cells, similarly to other vertebrates, express vimentin, GFAP, S100β, and glutamine synthetase. In contrast, Kir4.1 channel protein was not found in Müller cells but was localized in photoreceptor cells. Instead, 2P-domain TASK-1 channels were expressed in Müller cells. Electrophysiological properties of enzymatically dissociated Müller cells without photoreceptors and isolated Müller cells with adhering photoreceptors were significantly different. This suggests ion coupling between Müller cells and photoreceptors in the caiman retina. Sulforhodamine-B injected into cones permeated to adhering Müller cells thus revealing a uni-directional dye coupling. Conclusion Our data indicate that caiman Müller glial cells are unique among vertebrates studied so far by predominantly expressing TASK-1 rather than Kir4.1 K+ channels and by bi-directional ion and uni-directional dye coupling to photoreceptor cells. This coupling may play an important role in specific glia-neuron signaling pathways and in a new type of K+ buffering.
Collapse
Affiliation(s)
- Astrid Zayas-Santiago
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Silke Agte
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- Division of Soft Matter Physics, Department of Physics, University of Leipzig, Leipzig, Germany
| | - Yomarie Rivera
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Jan Benedikt
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Elke Ulbricht
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Anett Karl
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - José Dávila
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Alexey Savvinov
- Department of Physical Sciences, Universidad de Puerto Rico, Recinto de Río Piedras, Río Piedras, Puerto Rico, United States of America
| | - Yuriy Kucheryavykh
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Mikhail Inyushin
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Luis A. Cubano
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Thomas Pannicke
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | | | - Mike Francke
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- Translational Centre for Regenerative Medicine (TRM) University of Leipzig, Leipzig, Germany
| | - Alexei Verkhratsky
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Misty J. Eaton
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Andreas Reichenbach
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Serguei N. Skatchkov
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
- * E-mail:
| |
Collapse
|
17
|
Morphological Analyses on Retinal Glial Responses to Glaucomatous Injury Evoked by Venous Cauterization. Appl Microsc 2014. [DOI: 10.9729/am.2014.44.1.21] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
18
|
Gupta RK, Prasad S. Early down regulation of the glial Kir4.1 and GLT-1 expression in pericontusional cortex of the old male mice subjected to traumatic brain injury. Biogerontology 2013; 14:531-41. [PMID: 24026668 DOI: 10.1007/s10522-013-9459-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 08/27/2013] [Indexed: 10/26/2022]
Abstract
Astroglia play multiple roles in brain function by providing matrix to neurons, secreting neurotrophic factors, maintaining K(+) and glutamate homeostasis and thereby controlling synaptic plasticity which undergoes alterations during aging. K(+) and glutamate homeostasis is maintained by astrocytes membrane bound inwardly rectifying K(+) channel (Kir4.1) and glutamate transporter-1 (GLT-1 or EAAT-2) proteins, respectively in the synapse and their expression may be altered due to traumatic brain injury (TBI). Also, it is not well understood whether this change is age dependent. To find out this, TBI was experimentally induced in adult and old male AKR strain mice using CHI technique, and expression of the Kir4.1 and GLT-1 in the pericontusional cortex at various time intervals was studied by Western blotting and semi quantitative RT-PCR techniques. Here, we report that expression of both Kir4.1 and GLT-1 genes at transcript and protein levels is significantly down regulated in the pericontusional ipsi-lateral cortex of old TBI mice as compared to that in the adult TBI mice as function of time after injury. Further, expression of both the genes starts decreasing early in old mice i.e., from the first hour after TBI as compared to that starts from fourth hour in adult TBI mice. Thus TBI affects expression of Kir4.1 and GLT-1 genes in age- and time dependent manner and it may lead to accumulations of more K(+) and glutamate early in the synapse of old mice as compared to adult. This may be implicated in the TBI induced early and severe neuronal depolarization and excito-neurotoxicity in old age.
Collapse
Affiliation(s)
- R K Gupta
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | | |
Collapse
|
19
|
|
20
|
Group I mGluR-mediated inhibition of Kir channels contributes to retinal Müller cell gliosis in a rat chronic ocular hypertension model. J Neurosci 2012; 32:12744-55. [PMID: 22972998 DOI: 10.1523/jneurosci.1291-12.2012] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Müller cell gliosis, which is characterized by upregulated expression of glial fibrillary acidic protein (GFAP), is a universal response in many retinal pathological conditions. Whether down-regulation of inward rectifying K+ (Kir) channels, which commonly accompanies the enhanced GFAP expression, could contribute to Müller cell gliosis is poorly understood. We investigated changes of Kir currents, GFAP and Kir4.1 protein expression in Müller cells in a rat chronic ocular hypertension (COH) model, and explored the mechanisms underlying Müller cell gliosis. We show that Kir currents and Kir4.1 protein expression in Müller cells were reduced significantly, while GFAP expression was increased in COH rats, and these changes were eliminated by MPEP, a group I metabotropic glutamate receptors (mGluR I) subtype mGluR5 antagonist. In normal isolated Müller cells, the mGluR I agonist (S)-3,5-dihydroxyphenylglycine (DHPG) suppressed the Kir currents and the suppression was blocked by MPEP. The DHPG effect was mediated by the intracellular Ca2+ -dependent PLC/IP3-ryanodine/PKC signaling pathway, but the cAMP-PKA pathway was not involved. Moreover, intravitreal injection of DHPG in normal rats induced changes in Müller cells, similar to those observed in COH rats. The DHPG-induced increase of GFAP expression in Müller cells was obstructed by Ba2+, suggesting the involvement of Kir channels. We conclude that overactivation of mGluR5 by excessive extracellular glutamate in COH rats could contribute to Müller cell gliosis by suppressing Kir channels.
Collapse
|
21
|
Zhao T, Li Y, Weng C, Yin Z. The changes of potassium currents in RCS rat Müller cell during retinal degeneration. Brain Res 2011; 1427:78-87. [PMID: 22055109 DOI: 10.1016/j.brainres.2011.10.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2011] [Revised: 09/12/2011] [Accepted: 10/05/2011] [Indexed: 11/19/2022]
Abstract
Müller cells are the principal glial cells expressing membrane-bound potassium channel and predominantly mediating the homeostatic regulation of extracellular K+ produced by neuronal activity in retina. It's well known that Müller cells can be activated in many pathological conditions, but little is known about the change of potassium currents of Müller cells during the progression of retinitis pigmentosa. Herein, the Royal College of Surgeons rats (RCS rat) were employed to investigate some phenotypic and functional changes of Müller cells during retinal degeneration such as the expression of Kir4.1, membrane properties and K+ channel currents by using immunohistochemistry, RT-PCR, western blot and whole-cell patch clamping respectively. Compared with Müller cells in control retina, increased glutamine synthetase (GS) mRNA levels were seen at P30 and P60, and then decreased gradually in RCS rat retina. Morphologically, Müller cells showed significant hypertrophy and proliferation after p60. The increased expression of intermediate filament, glial fibrillary acidic protein (GFAP) and vimentin began at P30 and reached a peak at p60. Kir4.1 channels presented a peak expression at P30. Concomitantly, K(+) currents of Müller cells increased at P30 and decreased at P90 significantly. We concluded that retinal Müller cells of RCS rats underwent an activation initiated by the onset of retinal degeneration before p60 and then an obvious reactive gliosis, which led the basic membrane properties to suffer marked changes, and caused the Kir4.1 channels of Müller cells to occur a clear functional shift, even lose their normal electrophysiological properties. This process aggravates the impairment caused by the initial photoreceptor degeneration.
Collapse
Affiliation(s)
- TongTao Zhao
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China
| | | | | | | |
Collapse
|
22
|
Bringmann A, Wiedemann P. Müller glial cells in retinal disease. ACTA ACUST UNITED AC 2011; 227:1-19. [PMID: 21921569 DOI: 10.1159/000328979] [Citation(s) in RCA: 291] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 04/27/2011] [Indexed: 11/19/2022]
Abstract
Virtually all pathogenic stimuli activate Müller cells. Reactive Müller cells exert protective and toxic effects on photoreceptors and neurons. They contribute to oxidative stress and glutamate toxicity due to malfunctions of glutamate uptake and glutathione synthesis. Downregulation of potassium conductance disrupts transcellular potassium and water transport, resulting in neuronal hyperexcitability and edema. Protective effects of reactive Müller cells include upregulation of adenosine 5'-triphosphate (ATP)-degrading ectoenzymes, which enhances the extracellular availability of the neuroprotectant adenosine, abrogation of the osmotic release of ATP, which might protect retinal ganglion cells from apoptosis, and the release of antioxidants and neurotrophic factors. The dedifferentiation of reactive Müller cells to progenitor-like cells might have an impact on future therapeutic approaches. A better understanding of the gliotic mechanisms will be helpful in developing efficient therapeutic strategies aiming at increased protective and regenerative properties and decreased toxicity of reactive Müller cells.
Collapse
Affiliation(s)
- Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| | | |
Collapse
|
23
|
Wurm A, Pannicke T, Iandiev I, Francke M, Hollborn M, Wiedemann P, Reichenbach A, Osborne NN, Bringmann A. Purinergic signaling involved in Müller cell function in the mammalian retina. Prog Retin Eye Res 2011; 30:324-42. [DOI: 10.1016/j.preteyeres.2011.06.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 06/06/2011] [Accepted: 06/06/2011] [Indexed: 10/18/2022]
|
24
|
Takeda M, Takahashi M, Nasu M, Matsumoto S. Peripheral inflammation suppresses inward rectifying potassium currents of satellite glial cells in the trigeminal ganglia. Pain 2011; 152:2147-2156. [PMID: 21680091 DOI: 10.1016/j.pain.2011.05.023] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 05/13/2011] [Accepted: 05/18/2011] [Indexed: 11/28/2022]
Abstract
Previous studies indicate that silencing Kir4.1, a specific inward rectifying K(+) (Kir) channel subunit, in sensory ganglionic satellite glial cells (SGCs) induces behavioral hyperalgesia. However, the function of Kir4.1 channels in SGCs in vivo under pathophysiological conditions remains to be determined. The aim of the present study was to examine whether peripheral inflammation in anesthetized rats alters the SGC Kir4.1 current using in vivo patch clamp and immunohistochemical techniques. Inflammation was induced by injection of complete Freund's adjuvant into the whisker pad. The threshold of escape from mechanical stimulation applied to the orofacial area in inflamed rats was significantly lower than in naïve rats. The mean percentage of small/medium diameter trigeminal ganglion (TRG) neurons encircled by Kir4.1-immunoreactive SGCs in inflamed rats was also significantly lower than in naïve rats. In vivo whole-cell recordings were made using SGCs in the trigeminal ganglia (TRGs). Increasing extracellular K(+) concentrations resulted in significantly smaller potentiation of the mean peak amplitude of the Kir current in inflamed compared with naïve rats. In addition, the density of the Ba(2+)-sensitive Kir current associated with small-diameter TRG neurons was significantly lower in inflamed rats compared with naïve rats. Mean membrane potential in inflamed rats was more depolarized than in naïve rats. These results suggest that inflammation could suppress Kir4.1 currents of SGCs in the TRGs and that this impairment of glial potassium homeostasis in the TRGs contributes to trigeminal pain. Therefore, the Kir4.1 channel in SGCs may be a new molecular target for the treatment of trigeminal inflammatory pain.
Collapse
Affiliation(s)
- Mamoru Takeda
- Department of Physiology, School of Life Dentistry at Tokyo, Nippon Dental University, 1-9-20, Fujimi-cho, Chiyoda-ku, Tokyo 102-8159, Japan Research Center for Odontology, School of Life Dentistry at Tokyo, Nippon Dental University, 1-9-20, Fujimi-cho, Chiyoda-ku, Tokyo 102-8159, Japan
| | | | | | | |
Collapse
|
25
|
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: 2.0] [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
|
26
|
Hirrlinger PG, Ulbricht E, Iandiev I, Reichenbach A, Pannicke T. Alterations in protein expression and membrane properties during Müller cell gliosis in a murine model of transient retinal ischemia. Neurosci Lett 2010; 472:73-8. [PMID: 20132867 DOI: 10.1016/j.neulet.2010.01.062] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Revised: 01/06/2010] [Accepted: 01/27/2010] [Indexed: 01/26/2023]
Abstract
Retinal Müller glial cells are involved in K+ ion homeostasis of the tissue. Inwardly rectifying K(+) (Kir) channels play a decisive role in the process of spatial K+ buffering. It has been demonstrated that Kir-mediated currents of Müller cells are downregulated in various cases of retinal neurodegeneration. However, this has not yet been verified for any murine animal model. The aim of the present study was to investigate Müller cells after transient retinal ischemia in mice. High intraocular pressure was applied for 1h; the retina was analysed 1 week later. We studied protein expression in the tissue by immunohistochemistry, and membrane currents of isolated cells by patch-clamp experiments. We found the typical indicators of reactive gliosis such as upregulation of glial fibrillary acidic protein. Moreover, the membrane capacitance of isolated Müller cells was increased and the amplitudes of Kir-mediated currents were slightly, but significantly decreased. This murine high intraocular pressure model of transient retinal ischemia is proposed as a versatile tool for further studies on Müller cell functions in retinal degeneration.
Collapse
Affiliation(s)
- Petra G Hirrlinger
- Paul-Flechsig-Institut für Hirnforschung, Universität Leipzig, Jahnallee 59, D-04109 Leipzig, Germany
| | | | | | | | | |
Collapse
|
27
|
Zhang H, Mei X, Zhang P, Ma C, White FA, Donnelly DF, Lamotte RH. Altered functional properties of satellite glial cells in compressed spinal ganglia. Glia 2009; 57:1588-99. [PMID: 19330845 DOI: 10.1002/glia.20872] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The cell bodies of sensory neurons in the dorsal root ganglion (DRG) are enveloped by satellite glial cells (SGCs). In an animal model of intervertebral foraminal stenosis and low-back pain, a chronic compression of the DRG (CCD) increases the excitability of neuronal cell bodies in the compressed ganglion. The morphological and electrophysiological properties of SGCs were investigated in both CCD and uninjured, control lumbar DRGs. SGCs responded within 12 h of the onset of CCD as indicated by an increased expression of glial fibrillary acidic protein (GFAP) in the compressed DRG but to lesser extent in neighboring or contralateral DRGs. Within 1 week, coupling through gap junctions between SGCs was significantly enhanced in the compressed ganglion. Under whole-cell patch clamp recordings, inward and outward potassium currents, but not sodium currents, were detected in individual SGCs. SGCs enveloping differently sized neurons had similar electrophysiological properties. SGCs in the compressed vs. control DRG exhibited significantly reduced inwardly rectifying potassium currents (Kir), increased input resistances and positively shifted resting membrane potentials. The reduction in Kir was greater for nociceptive medium-sized neurons compared to non-nociceptive neurons. Kir currents of SGCs around spontaneously active neurons were significantly reduced 1 day after compression but recovered by 7 days. These data demonstrate rapid alterations in glial membrane currents and GFAP expression in close temporal association with the development of neuronal hyperexcitability in the CCD model of neuropathic pain. However, these alterations are not fully sustained and suggest other mechanisms for the maintenance of the hyperexcitable state.
Collapse
Affiliation(s)
- Haijun Zhang
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Bringmann A, Iandiev I, Pannicke T, Wurm A, Hollborn M, Wiedemann P, Osborne NN, Reichenbach A. Cellular signaling and factors involved in Müller cell gliosis: neuroprotective and detrimental effects. Prog Retin Eye Res 2009; 28:423-51. [PMID: 19660572 DOI: 10.1016/j.preteyeres.2009.07.001] [Citation(s) in RCA: 522] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Müller cells are active players in normal retinal function and in virtually all forms of retinal injury and disease. Reactive Müller cells protect the tissue from further damage and preserve tissue function by the release of antioxidants and neurotrophic factors, and may contribute to retinal regeneration by the generation of neural progenitor/stem cells. However, Müller cell gliosis can also contribute to neurodegeneration and impedes regenerative processes in the retinal tissue by the formation of glial scars. This article provides an overview of the neuroprotective and detrimental effects of Müller cell gliosis, with accounts on the cellular signal transduction mechanisms and factors which are implicated in Müller cell-mediated neuroprotection, immunomodulation, regulation of Müller cell proliferation, upregulation of intermediate filaments, glial scar formation, and the generation of neural progenitor/stem cells. A proper understanding of the signaling mechanisms implicated in gliotic alterations of Müller cells is essential for the development of efficient therapeutic strategies that increase the supportive/protective and decrease the destructive roles of gliosis.
Collapse
Affiliation(s)
- Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Liebigstrasse 10-14, D-04103 Leipzig, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Bringmann A, Iandiev I, Pannicke T, Wurm A, Bühner E, Reichenbach A, Wiedemann P, Uhlmann S. Porcine Müller Glial Cells Increase Expression of BKCaChannels in Retinal Detachment. Curr Eye Res 2009; 32:143-51. [PMID: 17364747 DOI: 10.1080/02713680601139333] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE To determine whether experimental retinal detachment causes an alteration in Ca2 +-activated, big conductance K+ (BK) currents of Müller glial cells. METHODS Rhegmatogenous retinal detachment was induced in porcine eyes. Müller cells were acutely isolated from control retinas and from retinas that were detached for 7 days. BK currents were detected by using the BK channel opener and the blocker phloretin and tetraethylammonium, respectively. RESULTS In addition to cellular hypertrophy and a decrease in inward rectifier K+ currents, Müller cells from detached retinas showed an increase in the amplitude of currents mediated by BK channels (850 +/- 105 pA) when compared with cells from control retinas (228 +/- 60 pA; p < 0.001). Similarly, the density of the BK channel-mediated currents was greater in cells from detached retinas (12.32 +/- 1.52 pA/pF) compared with control cells (4.07 +/- 1.07 pA/pF; p < 0.001). The increase in BK currents was correlated with the decrease of the inward rectifier K+ currents. CONCLUSIONS It is suggested that an increase in the expression of functional BK channels may be involved in gliotic responses of Müller cells after retinal detachment (e.g., in mitogen-induced Ca2+ responses and cellular proliferation).
Collapse
Affiliation(s)
- Andreas Bringmann
- Department of Ophthalmology and Eye Clinic, Faculty of Medicine, University of Leipzig, Liebigstrasse 10-14, D-04103 Leipzig, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Bringmann A, Wiedemann P. Involvement of Müller glial cells in epiretinal membrane formation. Graefes Arch Clin Exp Ophthalmol 2009; 247:865-83. [PMID: 19415318 DOI: 10.1007/s00417-009-1082-x] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2008] [Revised: 02/10/2009] [Accepted: 04/06/2009] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Proliferative retinopathies are considered to represent maladapted retinal wound repair processes driven by growth factor- and cytokine-induced overstimulation of proliferation, migration, extracellular matrix production and contraction of retinal cells. The formation of neovascular membranes represents an attempt to reoxygenize non-perfused retinal areas. Müller glial cells play a crucial role in the pathogenesis of proliferative retinopathies. This review summarizes the present knowledge regarding the role of Müller cells in periretinal membrane formation, especially in the early steps of epiretinal membrane formation, which involve an interaction of inflammatory and glial cells, and gives a survey of the factors which are suggested to be implicated in the induction of Müller cell gliosis and proliferation. CONCLUSIONS Alterations in the membrane conductance of Müller cells suggest that Müller cells may alter their phenotype into progenitor-like cells in the course of proliferative retinopathies; transdifferentiated Müller cells may have great impact for the development of new cell-based therapies.
Collapse
Affiliation(s)
- Andreas Bringmann
- Department of Ophthalmology, Faculty of Medicine, University of Leipzig, Eye Hospital, Leipzig, Germany.
| | | |
Collapse
|
31
|
K+ currents fail to change in reactive retinal glial cells in a mouse model of glaucoma. Graefes Arch Clin Exp Ophthalmol 2008; 246:1249-54. [DOI: 10.1007/s00417-008-0872-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Revised: 03/26/2008] [Accepted: 05/11/2008] [Indexed: 10/22/2022] Open
|
32
|
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: 1.6] [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
|
33
|
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: 1.3] [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]
|
34
|
Reichenbach A, Wurm A, Pannicke T, Iandiev I, Wiedemann P, Bringmann A. Müller cells as players in retinal degeneration and edema. Graefes Arch Clin Exp Ophthalmol 2007; 245:627-36. [PMID: 17219109 DOI: 10.1007/s00417-006-0516-y] [Citation(s) in RCA: 187] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Accepted: 12/02/2006] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Under normal conditions, Müller cells support neuronal activity and the integrity of the blood-retinal barrier, whereas gliotic alterations of Müller cells under pathological conditions may contribute to retinal degeneration and edema formation. A major function of Müller cells is the fluid absorption from the retinal tissue, which is mediated by transcellular water transport coupled to currents through potassium channels. METHODS Alterations of retinal Müller cells under pathological conditions were investigated by immunohistochemistry and recording their behavior under osmotic stress. RESULTS In animal models of various retinopathies, e.g., retinal ischemia, ocular inflammation, retinal detachment, and diabetes, it was found that Müller cells decrease the expression of their major potassium channel (Kir4.1). This alteration is associated with an impairment of the rapid water transport across Müller cell membranes, as recognizable in the induction of cellular swelling under hypoosmolar conditions. Osmotic swelling of Müller cells is also induced by oxidative stress and by inflammatory mediators such as arachidonic acid and prostaglandins. CONCLUSIONS The data suggest that a disturbed fluid transport through Müller cells is (in addition to vascular leakage) a pathogenic factor contributing to the development of retinal edema. Pharmacological re-activation of the retinal water clearance by Müller cells may represent an approach to the development of new edema-resolving drugs. Triamcinolone acetonide, which is clinically used to resolve edema, prevents osmotic swelling of Müller cells as it induces the release of endogenous adenosine and subsequent A1 receptor activation which results in the opening of ion channels. Apparently, triamcinolone resolves edema by both inhibition of vascular leakage and stimulation of retinal fluid clearance by Müller cells.
Collapse
Affiliation(s)
- Andreas Reichenbach
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of Leipzig, Jahnallee 59, 04109, Leipzig, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Iandiev I, Biedermann B, Bringmann A, Reichel MB, Reichenbach A, Pannicke T. Atypical gliosis in Müller cells of the slowly degenerating rds mutant mouse retina. Exp Eye Res 2006; 82:449-57. [PMID: 16154566 DOI: 10.1016/j.exer.2005.07.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2005] [Revised: 04/26/2005] [Accepted: 07/31/2005] [Indexed: 10/25/2022]
Abstract
Retinal Müller glial cells are known to undergo reactive changes (gliosis) in various retinal diseases. In virtually all cases studied, an upregulation of glial fibrillary acidic protein (GFAP) and a hypertrophy can be observed. Physiological alterations, such as a strong downregulation of inwardly rectifying K+ (Kir) currents, were found after retinal detachment (man, rabbit) and after ischemia/reperfusion (rat) but not in more slowly progressing retinal degenerations (Borna Disease Virus-infected rats, RCS rats). This led us to hypothesize that Müller cells respond with 'typical' reactive gliosis only to rapid but not to slow retinal degeneration. To test this hypothesis, we studied Müller cells from rds mutant mice (PrphRd2), which show a retinal degeneration of early onset and slow progression, resulting in a complete loss of photoreceptors after 9-12 months. In Müller cells of rds mice, we found immunoreactivity for GFAP, a marker of gliosis in Müller cells, from postnatal day 21 on, accompanied by a moderately increased membrane capacitance (taken as an indicator of hypertrophy), whereas no change in the expression of the Kir4.1 protein occurred in adult rds mice. We failed to observe significant changes in the membrane resistance and the membrane potential of cells from rds mice from first week after birth until 1 year of age. Current densities were decreased in cells from 3- and 5-week old rds mice. Furthermore, as in control cells from wildtype animals, these cells displayed dominant Kir currents, voltage-dependent Na+ currents, and glutamate uptake currents. These data support the idea that in mice as well as previously shown in rats, slow retinal degeneration induces an atypical gliosis of Müller cells.
Collapse
Affiliation(s)
- Ianors Iandiev
- Paul-Flechsig-Institut für Hirnforschung, Universität Leipzig, Jahnallee 59, D-04109 Leipzig, Germany.
| | | | | | | | | | | |
Collapse
|
36
|
Pannicke T, Biedermann B, Uckermann O, Weick M, Bringmann A, Wolf S, Wiedemann P, Habermann G, Buse E, Reichenbach A. Physiological properties of retinal Muller glial cells from the cynomolgus monkey, Macaca fascicularis--a comparison to human Muller cells. Vision Res 2005; 45:1781-91. [PMID: 15797768 DOI: 10.1016/j.visres.2005.01.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2004] [Revised: 01/11/2005] [Accepted: 01/19/2005] [Indexed: 11/16/2022]
Abstract
Retinae from rabbits and laboratory rodents are often used as 'models' of the human retina, although there are anatomical differences. To test whether monkey eyes provide a better model, a physiological study of Muller glial cells was performed comparing isolated cells and retinal wholemounts from the cynomolgus monkey, Macaca fascicularis and from man. The membrane conductance of Muller cells from both species was dominated by inward and outward K(+) currents. Cells displayed glutamate uptake currents and responded to nucleotides by intracellular Ca(2+) increases. However, there were also species differences, such as a lack of GABA(A) receptors and of Ca(2+)-dependent K(+) currents in monkey cells. Thus, the use of Muller cells from cynomolgus monkeys may be advantageous for investigating a few specific properties; in general, monkey cells are no more similar to human cells than those from standard laboratory animals.
Collapse
Affiliation(s)
- Thomas Pannicke
- Paul-Flechsig-Institut für Hirnforschung, Universität Leipzig, Jahnallee 59, D-04109 Leipzig, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Segev F, Mor O, Segev A, Belkin M, Assia EI. Downregulation of gene expression in the ageing lens: a possible contributory factor in senile cataract. Eye (Lond) 2005; 19:80-5. [PMID: 15105821 DOI: 10.1038/sj.eye.6701423] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
PURPOSE To study the molecular characteristics of lens epithelial cells from patients with senile cataract by cDNA microarray technique. METHODS Lens epithelial cells adhering to anterior capsules taken during cataract surgery collected from 108 patients, aged 56-92 years (senile cataract group), were pooled. Pooled epithelial cells of normal, noncataractous lenses from one patient with ocular trauma, one patient with lens subluxation, and 25 cadaveric eyes, all under the age of 55 years, served as a control. Total RNA was extracted by conventional methods from the two groups of cells, and a fluorescent probe was prepared for each group. The probes were hybridized on 9700 known human cDNA clones. Hybridized clones were analysed using a scanning laser and the results were processed by GEMTools (Incyte Genomics) software. RESULTS A total of 1827 clones hybridized with the two probes. Of these, 400 showed differences of more than two-fold in gene expression between the two probes. Relative to controls, gene expression in the senile cataract lenses was upregulated in 318 clones and downregulated in 82. Three genes-filensin, inwardly rectifying potassium channel (IRPC), and pigment epithelium-derived factor (PEDF) were strongly downregulated (by 41.3-, 6.8-, and 5.9-fold, respectively) in senile cataract. CONCLUSIONS Cataractogenesis is associated with numerous changes in the genetic profile of the lens epithelial cells. Since filensin, IRPC, and PEDF genes are known to have important roles in the physiology and morphology of the transparent lens, substantial downregulation of their expression might contribute to the formation of senile cataract.
Collapse
Affiliation(s)
- F Segev
- Department of Ophthalmology, Meir Hospital, Sapir Medical Center, Kfar-Saba, Israel.
| | | | | | | | | |
Collapse
|
38
|
Tenckhoff S, Hollborn M, Kohen L, Wolf S, Wiedemann P, Bringmann A. Diversity of aquaporin mRNA expressed by rat and human retinas. Neuroreport 2005; 16:53-6. [PMID: 15618890 DOI: 10.1097/00001756-200501190-00013] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Muller glial cells of the sensory retina mediate K+ and water fluxes that are facilitated by aquaporin-4 (AQP4) water channels and by Kir4.1-K+ channels. However, it is not known which subtypes of aquaporins are expressed in the mammalian retina. Using RT-PCR, we found that both human and rat retinas express mRNA for a diversity of water channel proteins. The human retina expresses mRNAs for AQP0 to AQP12 proteins. Using real-time PCR, we found that the mRNAs for AQP4 and Kir4.1 are downregulated in retinas that were obtained from patients with proliferative retinopathy compared with post-mortem controls. The data suggest that the development of proliferative gliosis is accompanied by disturbed transglial water and ion movements.
Collapse
Affiliation(s)
- Solveig Tenckhoff
- Department of Ophthalmology and Eye Clinic, University of Leipzig Medical Faculty, Liebigstr. 10-14, D-04103 Leipzig, Germany
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
Nervous systems are generally composed of two cell types-neurons and glia. Early studies of neurons revealed that these cells can conduct electrical currents, immediately implying that they have roles in the relay of information throughout the nervous system. Roles for glia have, until recently, remained obscure. The importance of glia in regulating neuronal survival had been long recognized. However, this trophic support function has hampered attempts to address additional, more active functions of these cells in the nervous system. In this chapter, recent efforts to reveal some of these additional functions are described. Evidence supporting a role for glia in synaptic development and activity is presented, as well as experiments suggesting glial guidance of neuronal migration and process outgrowth. Roles for glia in influencing the electrical activity of neurons are also discussed. Finally, an exciting system is described for studying glial cells in the nematode C. elegans, in which recent studies suggest that glia are not required for neuronal viability.
Collapse
Affiliation(s)
- Shai Shaham
- The Rockefeller University, New York, New York 10021, USA
| |
Collapse
|
40
|
Pannicke T, Uckermann O, Iandiev I, Biedermann B, Wiedemann P, Perlman I, Reichenbach A, Bringmann A. Altered membrane physiology in Müller glial cells after transient ischemia of the rat retina. Glia 2004; 50:1-11. [PMID: 15593100 DOI: 10.1002/glia.20151] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Inwardly rectifying K+ (Kir) channels have been implicated in the mediation of retinal K+ homeostasis by Muller glial cells. To assess possible involvement of altered glial K+ channel expression in ischemia-reperfusion injury, transient retinal ischemia was induced in rat eyes. Acutely isolated Muller cells from postischemic retinae displayed a fast downregulation of their Kir currents, which began within 1 day and reached a maximum at 3 days of reperfusion, with a peak decrease to 20% as compared with control. This strong decrease of Kir currents was accompanied by an increase of the incidence of cells which displayed depolarization-evoked fast transient (A-type) K+ currents. While no cell from untreated control rats expressed A-type K+ currents, all cells investigated from 3- and 7-day postischemic retinae displayed such currents. An increased incidence of cells displaying fast transient Na+ currents was observed at 7 days after ischemia. These results suggest a role of altered glial Kir channel expression in postischemic neuronal degeneration via disturbance of retinal K+ siphoning.
Collapse
Affiliation(s)
- Thomas Pannicke
- Paul-Flechsig-Institut für Hirnforschung, Abteilung Neurophysiologie, Universität Leipzig, Leipzig, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Cystoid macular edema (CME) is a well-known endpoint of various ocular diseases, but the relative pathogenic impact of extra- and intracellular fluid accumulation within the retinal tissue still remains uncertain. While most authors favor an extracellular fluid accumulation as the main causative factor of cyst formation, there are indications that Müller cell swelling may also contribute to CME development (particularly in cases without significant angiographic vascular leakage). Vascular leakage occurs after a breakdown of the blood-retinal barrier during traumatic, vascular, and inflammatory ocular diseases, and allows the serum to get into the retinal interstitium. Since intraretinal fluid distribution is restricted by two diffusion barriers, the inner and outer plexiform layers, serum leakage from intraretinal vessels causes cysts mainly in the inner nuclear layer while leakage from choroid/pigment epithelium generates (in addition to subretinal fluid accumulation) cyst formation in the Henle fiber layer. In the normal healthy retina, the transretinal water fluxes are mediated by glial and pigment epithelial cells. These water fluxes are inevitably coupled to fluxes of osmolytes; in the case of glial (Müller) cells, to K(+) clearance currents. For this purpose, the cells express a complex, microtopographically optimized pattern of transporters and channels for osmolytes and water in their plasma membrane. Ischemic/hypoxic alterations of the retinal microvasculature result in gliotic responses which involve down-regulation of K(+) channels in the perivascular Müller cell end-feet. This means a closure of the main pathway which normally generates the osmotic drive for the redistribution of water from the inner retina into the blood. The result is an intracellular K(+) accumulation which, then, osmotically drives water from the blood into the glial cells (i.e., in the opposite direction) and causes glial cell swelling, edema, and cyst formation. While the underlying mechanisms await further research, it is expected that their improved knowledge will stimulate the development of novel therapeutic approaches to resolve edema in retinal tissue.
Collapse
Affiliation(s)
- Andreas Bringmann
- Department of Ophthalmology, Eye Clinic, University of Leipzig, Germany.
| | | | | |
Collapse
|
42
|
Eichler W, Yafai Y, Keller T, Wiedemann P, Reichenbach A. PEDF derived from glial Müller cells: a possible regulator of retinal angiogenesis. Exp Cell Res 2004; 299:68-78. [PMID: 15302574 DOI: 10.1016/j.yexcr.2004.05.020] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2003] [Revised: 05/17/2004] [Indexed: 11/24/2022]
Abstract
A precise balance between stimulators and inhibitors of angiogenesis, such as vascular endothelial growth factor (VEGF) and pigment epithelium-derived factor (PEDF), respectively, is essential for angiogenic homeostasis in ocular tissues. Retinal hypoxia is accompanied by some pathological conditions that may promote intraocular neovascularization. Here we demonstrate that retinal glial (Müller) cells express and release pigment epithelium-derived factor (PEDF). Decreasing oxygen concentrations cause strong attenuation of PEDF release resulting in enhanced VEGF/PEDF ratios. Exposure of Müller cells to VEGF suppressed PEDF release in a dose-dependent manner. This may represent a novel mechanism of ocular angiogenic homeostasis sufficient in the control of PEDF levels during normoxia or mild hypoxia but supplemented by other (hitherto unknown) mechanisms in cases of strong hypoxia. In spite of the enhanced VEGF/PEDF ratios resulting from hypoxia, conditioned media of Müller cells failed to stimulate additional proliferation of retinal endothelial cells. These findings suggest that in the ischemic retina, Müller cells generate a permissive condition for angiogenesis by secreting more VEGF and less PEDF, but the onset of retinal endothelial cell proliferation requires another triggering signal that remains to be identified.
Collapse
MESH Headings
- Animals
- Cattle
- Cells, Cultured
- Culture Media, Conditioned/pharmacology
- Dose-Response Relationship, Drug
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Eye Proteins
- Guinea Pigs
- Humans
- Hypoxia/metabolism
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/physiopathology
- Neovascularization, Physiologic/drug effects
- Neovascularization, Physiologic/physiology
- Nerve Growth Factors
- Neuroglia/cytology
- Neuroglia/drug effects
- Neuroglia/metabolism
- Proteins/metabolism
- Retina/cytology
- Retina/metabolism
- Retina/physiopathology
- Retinal Artery/drug effects
- Retinal Artery/growth & development
- Retinal Artery/metabolism
- Retinal Neovascularization/metabolism
- Retinal Neovascularization/physiopathology
- Serpins/metabolism
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor A/pharmacology
Collapse
Affiliation(s)
- Wolfram Eichler
- Eye Hospital, University of Leipzig, D-04103 Leipzig, Germany.
| | | | | | | | | |
Collapse
|
43
|
Schopf S, Ruge H, Bringmann A, Reichenbach A, Skatchkov SN. Switch of K+ buffering conditions in rabbit retinal Müller glial cells during postnatal development. Neurosci Lett 2004; 365:167-70. [PMID: 15246541 DOI: 10.1016/j.neulet.2004.04.074] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2004] [Revised: 04/27/2004] [Accepted: 04/28/2004] [Indexed: 11/17/2022]
Abstract
Although spatial buffering of excess extracellular K+ by K+ channels is a main function of retinal glial (Müller) cells, there are severe limitations to long distance K+-spatial buffering that have been predicted for (immature) glial cells: (i) a lack of inwardly rectifying K+ (Kir) channels [Glia 21(1997) 46]; and (ii) high internal resistance of outgrowing (cable like) processes [W. Rall, Handbook of Physiology, Section 1, vol. 1, Part 1, American Physiological Society, Bethesda, 1977, pp. 39-97]. In order to determine if changes in developing Müller cells improve or worsen their capability of carrying K+ spatial buffering currents, we compared the whole-cell currents of acutely isolated Müller cells at 5, 11 and 28 postnatal days of rabbits. Both K+-spatial buffer limitations described above were found in early postnatal stage (5 days), however, the cells overcome these limitations shortly after 11 days. During the period of 11-28 days, rabbit Müller cells simultaneously increase stalk axial conductance and express Kir channels. Both processes take place during the critical stage of retinal maturation, and should dramatically improve "cable" K+-spatial buffering.
Collapse
Affiliation(s)
- Stefan Schopf
- Department of Neurophysiology, Paul Flechsig Institute of Brain Research, University of Leipzig, D-04109 Leipzig, Germany
| | | | | | | | | |
Collapse
|
44
|
Abstract
Epilepsy is one of the most common neurological disorders, but the cellular basis of human epilepsy remains largely a mystery, and about 30% of all epilepsies remain uncontrolled. The vast bulk of epilepsy research has focused on neuronal and synaptic mechanisms, but the hypersynchronous firing that is the hallmark of epilepsy could also result from the abnormal function of glial cells by virtue of their critical role in the homeostasis of the brain's extracellular milieu. Therefore, increasing our understanding of glial pro-epileptic and epileptogenic mechanisms holds promise for the development of improved pharmacological treatments for epilepsy. Reactive astrocytes, a prominent feature of the human epileptic brain, undergo changes in their membrane properties and electrophysiology, in particular in the expression of membrane K(+) and Na(+) channels, which result in pro-epileptic changes in their homeostatic control of the extracellular space. Nonetheless, a causal role for reactive astrocytosis in epilepsy has been difficult to determine because glial reactivity can be induced by a wide range of central nervous system insults, including epileptic seizures themselves. A complicating factor is that different insults to the central nervous system result in reactive astrocytes with different membrane properties. Therefore, most animal models of epilepsy preselect the properties of the reactive glia studied. Finally, a causal role for reactive glia in epilepsy cannot be firmly established by examining human epileptic tissue because of its chronic and pharmacoresistant pathological condition that warranted the surgical intervention. Therefore, the development of clinically relevant models of reactive astrocytosis, and of symptomatic epileptogenesis, is needed to investigate the issue. A recently developed model of post-traumatic epileptogenesis in the rat, where chronic spontaneous recurrent seizures develop after a single event of a clinically relevant form of closed head injury, the fluid percussion injury, offers hope to help understand the role of reactive glia in seizures and epileptogenesis and lead to the development of improved therapies.
Collapse
Affiliation(s)
- Raimondo D'Ambrosio
- Department of Neurological Surgery and Regional Epilepsy Center, University of Washington, Box 359914, 325 Ninth Avenue, Seattle, WA 98104, USA.
| |
Collapse
|
45
|
Abstract
Inwardly rectifying potassium (K(ir)) channels are a prominent feature of mature, postmitotic astrocytes. These channels are believed to set the resting membrane potential near the potassium equilibrium potential (E(K)) and are implicated in potassium buffering. A number of previous studies suggest that K(ir) channel expression is indicative of cell differentiation. We therefore set out to examine K(ir) channel expression in malignant glia, which are incapable of differentiation. We used two established and widely used glioma cell lines, D54MG (a WHO grade 4 glioma) and STTG-1 (a WHO grade 3 glioma), and compared them to immature and differentiated astrocytes. Both glioma cell lines were characterized by large outward K(+) currents, depolarized resting membrane potentials (V(m)) (-38.5 +/- 4.2 mV, D54 and -28.1 +/- 3.5 mV, STTG1), and relatively high input resistances (R(m)) (260.6 +/- 64.7 MOmega, D54 and 687.2 +/- 160.3 MOmega, STTG1). These features were reminiscent of immature astrocytes, which also displayed large outward K(+) currents, had a mean V(m) of -51.1 +/- 3.7 and a mean R(m) value of 627.5 +/- 164 MOmega. In contrast, mature astrocytes had a significantly more negative resting membrane potential (-75.2 +/- 0.56 mV), and a mean R(m) of 25.4 +/- 7.4 MOmega. Barium (Ba(2+)) sensitive K(ir) currents were >20-fold larger in mature astrocytes (4.06 +/- 1.1 nS/pF) than in glioma cells (0.169 +/- 0.033 nS/pF D54, 0.244 +/- 0.04 nS/pF STTG1), which had current densities closer to those of dividing, immature astrocytes (0.474 +/- 0.12 nS/pF). Surprisingly, Western blot analysis shows expression of several K(ir) channel subunits in glioma cells (K(ir)2.3, 3.1, and 4.1). However, while in astrocytes these channels localize diffusely throughout the cell, in glioma cells they are found almost exclusively in either the cell nucleus (K(ir)2.3 and 4.1) or ER/Golgi (3.1). These data suggest that mislocalization of K(ir) channel proteins to intracellular compartments is responsible for a lack of appreciable K(ir) currents in glioma cells.
Collapse
Affiliation(s)
- M L Olsen
- Department of Neurobiology and Civitan International Research Center, University of Alabama, Birmingham, Alabama 35294, USA
| | | |
Collapse
|
46
|
Bordey A, Spencer DD. Distinct electrophysiological alterations in dentate gyrus versus CA1 glial cells from epileptic humans with temporal lobe sclerosis. Epilepsy Res 2004; 59:107-22. [PMID: 15246115 DOI: 10.1016/j.eplepsyres.2004.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2003] [Revised: 11/11/2003] [Accepted: 04/05/2004] [Indexed: 11/28/2022]
Abstract
Previous studies have characterized the electrophysiological properties of astrocytes in the CA1 region of hippocampi resected from patients with intractable temporal lobe epilepsy (TLE). However, the properties of hilar astrocytes from such patients have not been studied although astrocytes display regional heterogeneity and a non-uniform response to injury. Thus, we performed patch-clamp recordings of putative astrocytes in hilar and CA1 regions of surgically removed epileptic hippocampi with and without sclerosis (mesial TLE, MTLE patients, and paradoxical TLE, PTLE patients, respectively), and non-epileptic, non-sclerotic hippocampi (tumor patients). Our data show that the current profile of hilar astrocytes undergoes significant changes in MTLE but not in PTLE or tumor hippocampi. In particular, inwardly rectifying K(+) (K(IR)) and outwardly rectifying K(+) currents were reduced, inward Na(+) currents and membrane resistances were increased in putative astrocytes from MLTE cases compared to PTLE and tumor cases. Because the conductance of K(IR) channels in cell-attached patches (approximately 34pS) from MTLE tissue was not altered, a reduction in the number of K(IR) channels likely accounts for the decrease in whole-cell K(IR) conductance. Presumed astrocytes in the CA1 region from each patient group displayed intercellular coupling and a passive current profile; these characteristics were never observed in hilar glial cells. No apparent changes in the current profile of coupled CA1 glial cells could be detected between MTLE, PTLE and tumor tissues. Additionally, CA1 glial cells expressed a high density of 34pS K(IR) channels. These data suggest that K(+) buffering via K(IR) channels may be functionally compromised in hilar astrocytes of epileptic and sclerotic (MTLE) human hippocampi. By contrast, CA1 astrocytes retained their intercellular coupling and K(IR) channel expression necessary for K(+) buffering.
Collapse
Affiliation(s)
- A Bordey
- Department of Neurosurgery and Cellular and Molecular Physiology, Yale University, 333 Cedar Street, LSOG 228, New Haven, CT 06520-8082, USA.
| | | |
Collapse
|
47
|
Chávez AE, Pannicke T, Roncagliolo M, Reichenbach A, Palacios AG. Electrophysiological properties of retinal Müller glial cells from myelin mutant rat. Glia 2003; 45:338-45. [PMID: 14966865 DOI: 10.1002/glia.10330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The structural and functional similarities between Müller cells and oligodendrocytes prompted the present study of the electrophysiological properties of Müller (glia) cells obtained from the retinae of control and myelin mutant taiep rats during the postnatal developmental period (P12-P180). The whole-cell configuration of the patch-clamp technique was used to characterize the general properties and the K+ currents from dissociated Müller cells. During the first 3 weeks of life, a decrease of the membrane resistance and an increase of the membrane potential were observed in Müller cells from both control and taiep rats. However, Müller cells from taiep rats never achieved the very negative membrane potential (-50 mV vs -80 mV) and the low membrane resistance characteristic for control cells. Furthermore, Müller cells displayed increased inward and outward K+ currents during postnatal development up to P30/60 in controls; however, in taiep rats, this increase ceased at P20/30, and low-amplitude currents persisted into adulthood. These results provide first evidence of physiological changes in retinal Müller cells as a consequence of a myelin mutation causing a progressive deterioration of the central nervous system (CNS) due to a disturbance of the microtubule network of oligodendrocytes. We hypothesize that the progressive dysmyelination process of the optic nerve, accompanied by functional deficits of retinal neurons (e.g., ganglion cells), induces physiological alterations of Müller cells.
Collapse
Affiliation(s)
- Andrés E Chávez
- Department of Physiology, Faculty of Science, Neuroscience Center of Valparaiso, University of Valparaíso, Valparaíso, Chile
| | | | | | | | | |
Collapse
|
48
|
Francke M, Uhlmann S, Pannicke T, Goczalik I, Uckermann O, Weick M, Härtig W, Wiedemann P, Reichenbach A, Bringmann A. Experimental dispase-induced retinopathy causes up-regulation of P2Y receptor-mediated calcium responses in Müller glial cells. Ophthalmic Res 2003; 35:30-41. [PMID: 12566861 DOI: 10.1159/000068192] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2002] [Indexed: 11/19/2022]
Abstract
During proliferative vitreoretinopathy (PVR) Müller glial cells show an up-regulation of their responsiveness to extracellular adenosine 5'-triphosphate (ATP). In the present study, we investigated if such a glial cell response is also a feature for other retinopathies besides PVR. To this aim, the proteolytic enzyme, dispase (0.1 U), was injected into the vitreous of rabbit eyes. After 3 weeks, a distinct retinopathy had developed which showed no signs of PVR. The retinopathy was characterized by strong alterations of the retinal vasculature in the medullary rays, by photoreceptor degeneration, retinal atrophy, and activation of microglial cells. Müller cells became reactive, as indicated by up-regulation of glial fibrillary acidic protein immunoreactivity and by hypertrophy involving subretinal fibrosis. Müller cell reactivity was also evidenced electrophysiologically by a down-regulation of their inwardly rectifying potassium currents and by an up-regulation of their responsiveness to extracellular ATP. Significantly more Müller cells from dispase-treated eyes showed ATP-evoked calcium (83%) and current responses (69%) when compared with cells from control eyes (13 and 9%, respectively). The results indicate that increased responsiveness to extracellular ATP may be a more general feature of Müller cell gliosis, and is also observed in retinopathies besides PVR.
Collapse
Affiliation(s)
- M Francke
- Department of Neurophysiology, Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Potassium homeostasis in the brain at the organ and cell level. ACTA ACUST UNITED AC 2003. [DOI: 10.1016/s1569-2558(03)31027-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
50
|
Biedermann B, Wolf S, Kohen L, Wiedemann P, Buse E, Reichenbach A, Pannicke T. Patch-clamp recording of Müller glial cells after cryopreservation. J Neurosci Methods 2002; 120:173-8. [PMID: 12385767 DOI: 10.1016/s0165-0270(02)00195-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Human and other primate retinal Müller cells display dominating K(+) currents as well as other membrane conductances that may change in cases of retinal pathology. Because the use of human and primate tissue is limited by reasons of availability, a method for long-term storage of these cells is desirable. We describe a cryopreservation method in which isolated Müller cells are stored in liquid nitrogen. After thawing, the cells can be used for patch-clamp experiments immediately, without culturing. We show that the main electrophysiological properties are not altered by this method and that voltage- and ligand-gated currents can be recorded from cryopreserved cells even after 2-years storage.
Collapse
Affiliation(s)
- Bernd Biedermann
- Paul-Flechsig-Institut für Hirnforschung, Universität Leipzig, Jahnallee 59, D-04109 Leipzig, Germany
| | | | | | | | | | | | | |
Collapse
|