1
|
Thoreson WB, Bartol TM, Conoan NH, Diamond JS. Geometric tortuosity at invaginating rod synapses slows glutamate diffusion and shapes synaptic responses: insights from anatomically realistic Monte Carlo simulations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.30.621088. [PMID: 39554003 PMCID: PMC11565802 DOI: 10.1101/2024.10.30.621088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
At the first synapse in the vertebrate retina, rod photoreceptor terminals form deep invaginations occupied by multiple second-order rod bipolar and horizontal cell (RBP and HC) dendrites. Synaptic vesicles are released into this invagination at multiple sites beneath an elongated presynaptic ribbon. We investigated the impact of this complex architecture on the diffusion of synaptic glutamate and activity of postsynaptic receptors. We obtained serial electron micrographs of mouse retina and reconstructed four rod terminals along with their postsynaptic RBP and HC dendrites. We incorporated these structures into an anatomically realistic Monte Carlo simulation of neurotransmitter diffusion and receptor activation. We compared passive diffusion of glutamate in these realistic structures to existing, geometrically simplified models of the synapse and found that glutamate exits anatomically realistic synapses ten times more slowly than previously predicted. By comparing simulations with electrophysiological recordings, we modeled synaptic activation of EAAT5 glutamate transporters in rods, AMPA receptors on HC dendrites, and metabotropic glutamate receptors (mGluR6) on RRBP dendrites. Our simulations suggested that ~3,000 EAAT5 transporters populate the rod presynaptic membrane and that, while uptake by surrounding glial Müller cells retrieves much of the glutamate released by rods, binding and uptake by EAAT5 influences RBP response kinetics. The long lifetime of glutamate within the cleft allows mGluR6 on RBP dendrites to temporally integrate the steady stream of vesicles released at this synapse in darkness. Glutamate's tortuous diffusional path through realistic synaptic geometry confers quantal variability, as release from nearby ribbon sites exerts larger effects on RBP and HC receptors than release from more distant sites. While greater integration may allow slower sustained release rates, added quantal variability complicates the challenging task of detecting brief decreases in release produced by rod light responses at scotopic threshold.
Collapse
|
2
|
Mesnard CS, Hays CL, Townsend LE, Barta CL, Gurumurthy CB, Thoreson WB. SYNAPTOTAGMIN-9 IN MOUSE RETINA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.27.546758. [PMID: 37425946 PMCID: PMC10327071 DOI: 10.1101/2023.06.27.546758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Synaptotagmin-9 (Syt9) is a Ca2+ sensor mediating fast synaptic release expressed in various parts of the brain. The presence and role of Syt9 in retina is unknown. We found evidence for Syt9 expression throughout the retina and created mice to conditionally eliminate Syt9 in a cre-dependent manner. We crossed Syt9fl/fl mice with Rho-iCre, HRGP-Cre, and CMV-cre mice to generate mice in which Syt9 was eliminated from rods (rodSyt9CKO), cones (coneSyt9CKO), or whole animals (CMVSyt9). CMVSyt9 mice showed an increase in scotopic electroretinogram (ERG) b-waves evoked by bright flashes with no change in a-waves. Cone-driven photopic ERG b-waves were not significantly different in CMVSyt9 knockout mice and selective elimination of Syt9 from cones had no effect on ERGs. However, selective elimination from rods decreased scotopic and photopic b-waves as well as oscillatory potentials. These changes occurred only with bright flashes where cone responses contribute. Synaptic release was measured in individual rods by recording anion currents activated by glutamate binding to presynaptic glutamate transporters. Loss of Syt9 from rods had no effect on spontaneous or depolarization-evoked release. Our data show that Syt9 is acts at multiple sites in the retina and suggest that it may play a role in regulating transmission of cone signals by rods.
Collapse
Affiliation(s)
- Chris S. Mesnard
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68106, USA
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68106, USA
| | - Cassandra L. Hays
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68106, USA
- Department of Medical Education, Creighton University, Omaha, NE 68178
| | - Lou E. Townsend
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68106, USA
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68106, USA
| | - Cody L. Barta
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68106, USA
| | | | - Wallace B. Thoreson
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68106, USA
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68106, USA
| |
Collapse
|
3
|
Lee SJ, Emery D, Vukmanic E, Wang Y, Lu X, Wang W, Fortuny E, James R, Kaplan HJ, Liu Y, Du J, Dean DC. Metabolic transcriptomics dictate responses of cone photoreceptors to retinitis pigmentosa. Cell Rep 2023; 42:113054. [PMID: 37656622 PMCID: PMC10591869 DOI: 10.1016/j.celrep.2023.113054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/21/2023] [Accepted: 08/15/2023] [Indexed: 09/03/2023] Open
Abstract
Most mutations in retinitis pigmentosa (RP) arise in rod photoreceptors, but cone photoreceptors, responsible for high-resolution daylight and color vision, are subsequently affected, causing the most debilitating features of the disease. We used mass spectroscopy to follow 13C metabolites delivered to the outer retina and single-cell RNA sequencing to assess photoreceptor transcriptomes. The S cone metabolic transcriptome suggests engagement of the TCA cycle and ongoing response to ROS characteristic of oxidative phosphorylation, which we link to their histone modification transcriptome. Tumor necrosis factor (TNF) and its downstream effector RIP3, which drive ROS generation via mitochondrial dysfunction, are induced and activated as S cones undergo early apoptosis in RP. The long/medium-wavelength (L/M) cone transcriptome shows enhanced glycolytic capacity, which maintains their function as RP progresses. Then, as extracellular glucose eventually diminishes, L/M cones are sustained in long-term dormancy by lactate metabolism.
Collapse
Affiliation(s)
- Sang Joon Lee
- Department of Medicine, Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA; Department of Ophthalmology, Kosin University College of Medicine, #262 Gamcheon-ro, Seo-gu, Busan 49267, Korea
| | - Douglas Emery
- Department of Medicine, Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Eric Vukmanic
- Department of Medicine, Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Yekai Wang
- Departments of Ophthalmology and Visual Sciences and Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Xiaoqin Lu
- Department of Medicine, Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Wei Wang
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Enzo Fortuny
- Department of Neurosurgery, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Robert James
- Department of Neurosurgery, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Henry J Kaplan
- Department of Ophthalmology, St. Louis University School of Medicine, St. Louis MO 63110, USA
| | - Yongqing Liu
- Department of Medicine, Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Jianhai Du
- Departments of Ophthalmology and Visual Sciences and Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA.
| | - Douglas C Dean
- Department of Medicine, Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA.
| |
Collapse
|
4
|
Thoreson WB, Chhunchha B. EAAT5 glutamate transporter rapidly binds glutamate with micromolar affinity in mouse rods. J Gen Physiol 2023; 155:e202313349. [PMID: 37477643 PMCID: PMC10359920 DOI: 10.1085/jgp.202313349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/17/2023] [Accepted: 07/05/2023] [Indexed: 07/22/2023] Open
Abstract
Light responses of rod photoreceptor cells in the retina are encoded by changes in synaptic glutamate release that is in turn shaped by reuptake involving EAAT5 plasma membrane glutamate transporters. Heterologously expressed EAAT5 activates too slowly upon glutamate binding to support significant uptake. We tested EAAT5 activation in mouse rods in vivo by stimulating glutamate transporter anion currents (IA(glu)) with UV flash photolysis of MNI-glutamate, varying flash intensity to vary glutamate levels. Responses to uncaging rose rapidly with time constants of 2-3 ms, similar to IA(glu) events arising from spontaneous release. Spontaneous release events and IA(glu) evoked by weak flashes also declined with similar time constants of 40-50 ms. Stronger flashes evoked responses that decayed more slowly. Time constants were twofold faster at 35°C, suggesting that they reflect transporter kinetics, not diffusion. Selective EAAT1 and EAAT2 inhibitors had no significant effect, suggesting IA(glu) in rods arises solely from EAAT5. We calibrated glutamate levels attained during flash photolysis by expressing a fluorescent glutamate sensor iGluSnFr in cultured epithelial cells. We compared fluorescence at different glutamate concentrations to fluorescence evoked by photolytic uncaging of MNI-glutamate. The relationship between flash intensity and glutamate yielded EC50 values for EAAT5 amplitude, decay time, and rise time of ∼10 μM. Micromolar affinity and rapid activation of EAAT5 in rods show it can rapidly bind synaptic glutamate. However, we also found that EAAT5 currents are saturated by the synchronous release of only a few vesicles, suggesting limited capacity and a role for glial uptake at higher release rates.
Collapse
Affiliation(s)
- Wallace B. Thoreson
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Institute, Omaha, NE, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bhavana Chhunchha
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Institute, Omaha, NE, USA
| |
Collapse
|
5
|
Schwarz K, Schmitz F. Synapse Dysfunctions in Multiple Sclerosis. Int J Mol Sci 2023; 24:ijms24021639. [PMID: 36675155 PMCID: PMC9862173 DOI: 10.3390/ijms24021639] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic neuroinflammatory disease of the central nervous system (CNS) affecting nearly three million humans worldwide. In MS, cells of an auto-reactive immune system invade the brain and cause neuroinflammation. Neuroinflammation triggers a complex, multi-faceted harmful process not only in the white matter but also in the grey matter of the brain. In the grey matter, neuroinflammation causes synapse dysfunctions. Synapse dysfunctions in MS occur early and independent from white matter demyelination and are likely correlates of cognitive and mental symptoms in MS. Disturbed synapse/glia interactions and elevated neuroinflammatory signals play a central role. Glutamatergic excitotoxic synapse damage emerges as a major mechanism. We review synapse/glia communication under normal conditions and summarize how this communication becomes malfunctional during neuroinflammation in MS. We discuss mechanisms of how disturbed glia/synapse communication can lead to synapse dysfunctions, signaling dysbalance, and neurodegeneration in MS.
Collapse
|
6
|
Ohkuma M, Maruyama T, Ishii T, Igarashi N, Azuma K, Inoue T, Obata R, Miyachi EI, Kaneda M. Effects of Progesterone and Other Gonadal Hormones on Glutamatergic Circuits in the Retina. J NIPPON MED SCH 2023; 90:333-345. [PMID: 37690823 DOI: 10.1272/jnms.jnms.2023_90-405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
BACKGROUND Gonadal hormones function in the retina; however, their targets have not yet been identified. Therefore, the present study examined the effects of progesterone and other gonadal hormones on glutamatergic circuits in the retina. METHODS Extracellular glutamate concentrations, which correspond to the amount of glutamate released, were examined using an enzyme-linked fluorescent assay system. The activity of glutamatergic synapses between bipolar cells and ganglion cells was investigated using a patch clamp technique. Changes in retinal thickness during pregnancy were assessed using optical coherence tomography (OCT) images. RESULTS Progesterone and pregnenolone sulfate increased extracellular glutamate concentrations, whereas estrogen and testosterone did not. Progesterone increased the activity of glutamatergic synapses between bipolar cells and ganglion cells. A temporal decrease in the thickness of the peripheral retina was observed in the 1st trimester. CONCLUSIONS Progesterone, but not estrogen or testosterone, activated glutamate release in the mouse retina. Increases in the concentration of progesterone during pregnancy did not induce any detectable change in retinal thickness.
Collapse
Affiliation(s)
| | - Takuma Maruyama
- Department of Physiology, Nippon Medical School
- Present address: Department of Physiology, Division of Neurophysiology, School of Medicine, Tokyo Women's Medical University
| | | | - Nozomi Igarashi
- Department of Ophthalmology, Faculty of Medicine, Tokyo University
| | - Keiko Azuma
- Department of Ophthalmology, Faculty of Medicine, Tokyo University
| | - Tatsuya Inoue
- Department of Ophthalmology, School of Medicine, Yokohama City University
| | - Ryo Obata
- Department of Ophthalmology, Faculty of Medicine, Tokyo University
| | | | | |
Collapse
|
7
|
Mesnard CS, Hays CL, Barta CL, Sladek AL, Grassmeyer JJ, Hinz KK, Quadros RM, Gurumurthy CB, Thoreson WB. Synaptotagmins 1 and 7 in vesicle release from rods of mouse retina. Exp Eye Res 2022; 225:109279. [PMID: 36280223 PMCID: PMC9830644 DOI: 10.1016/j.exer.2022.109279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/14/2022] [Accepted: 10/10/2022] [Indexed: 01/13/2023]
Abstract
Synaptotagmins are the primary Ca2+ sensors for synaptic exocytosis. Previous work suggested synaptotagmin-1 (Syt1) mediates evoked vesicle release from cone photoreceptor cells in the vertebrate retina whereas release from rods may involve another sensor in addition to Syt1. We found immunohistochemical evidence for syntaptotagmin-7 (Syt7) in mouse rod terminals and so performed electroretinograms (ERG) and single-cell recordings using mice in which Syt1 and/or Syt7 were conditionally removed from rods and/or cones. Synaptic release was measured in mouse rods by recording presynaptic anion currents activated during glutamate re-uptake and from exocytotic membrane capacitance changes. Deleting Syt1 from rods reduced glutamate release evoked by short depolarizing steps but not long steps whereas deleting Syt7 from rods reduced release evoked by long but not short steps. Deleting both sensors completely abolished depolarization-evoked release from rods. Effects of various intracellular Ca2+ buffers showed that Syt1-mediated release from rods involves vesicles close to ribbon-associated Ca2+ channels whereas Syt7-mediated release evoked by longer steps involves more distant release sites. Spontaneous release from rods was unaffected by eliminating Syt7. While whole animal knockout of Syt7 slightly reduced ERG b-waves and oscillatory potentials, selective elimination of Syt7 from rods had no effect on ERGs. Furthermore, eliminating Syt1 from rods and cones abolished ERG b-waves and additional elimination of Syt7 had no further effect. These results show that while Syt7 contributes to slow non-ribbon release from rods, Syt1 is the principal sensor shaping rod and cone inputs to bipolar cells in response to light flashes.
Collapse
Affiliation(s)
- C S Mesnard
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, USA; Pharmacology and Experimental Neuroscience, USA
| | - C L Hays
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, USA
| | - C L Barta
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, USA
| | - A L Sladek
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, USA
| | - J J Grassmeyer
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, USA; Pharmacology and Experimental Neuroscience, USA
| | - K K Hinz
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, USA
| | - R M Quadros
- Pharmacology and Experimental Neuroscience, USA; Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68106, USA
| | - C B Gurumurthy
- Pharmacology and Experimental Neuroscience, USA; Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68106, USA
| | - W B Thoreson
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, USA; Pharmacology and Experimental Neuroscience, USA.
| |
Collapse
|
8
|
Mesnard CS, Barta CL, Sladek AL, Zenisek D, Thoreson WB. Eliminating Synaptic Ribbons from Rods and Cones Halves the Releasable Vesicle Pool and Slows Down Replenishment. Int J Mol Sci 2022; 23:6429. [PMID: 35742873 PMCID: PMC9223732 DOI: 10.3390/ijms23126429] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/03/2022] [Accepted: 06/05/2022] [Indexed: 01/25/2023] Open
Abstract
Glutamate release from rod and cone photoreceptor cells involves presynaptic ribbons composed largely of the protein RIBEYE. To examine roles of ribbons in rods and cones, we studied mice in which GCamP3 replaced the B-domain of RIBEYE. We discovered that ribbons were absent from rods and cones of both knock-in mice possessing GCamP3 and conditional RIBEYE knockout mice. The mice lacking ribbons showed reduced temporal resolution and contrast sensitivity assessed with optomotor reflexes. ERG recordings showed 50% reduction in scotopic and photopic b-waves. The readily releasable pool (RRP) of vesicles in rods and cones measured using glutamate transporter anion currents (IA(glu)) was also halved. We also studied the release from cones by stimulating them optogenetically with ChannelRhodopsin2 (ChR2) while recording postsynaptic currents in horizontal cells. Recovery of the release from paired pulse depression was twofold slower in the rods and cones lacking ribbons. The release from rods at -40 mV in darkness involves regularly spaced multivesicular fusion events. While the regular pattern of release remained in the rods lacking ribbons, the number of vesicles comprising each multivesicular event was halved. Our results support conclusions that synaptic ribbons in rods and cones expand the RRP, speed up vesicle replenishment, and augment some forms of multivesicular release. Slower replenishment and a smaller RRP in photoreceptors lacking ribbons may contribute to diminished temporal frequency responses and weaker contrast sensitivity.
Collapse
Affiliation(s)
- Chris S. Mesnard
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.S.M.); (C.L.B.); (A.L.S.)
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Cody L. Barta
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.S.M.); (C.L.B.); (A.L.S.)
| | - Asia L. Sladek
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.S.M.); (C.L.B.); (A.L.S.)
| | - David Zenisek
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06510, USA;
| | - Wallace B. Thoreson
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.S.M.); (C.L.B.); (A.L.S.)
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
9
|
Tang FS, Yuan HL, Liu JB, Zhang G, Chen SY, Ke JB. Glutamate Transporters EAAT2 and EAAT5 Differentially Shape Synaptic Transmission from Rod Bipolar Cell Terminals. eNeuro 2022; 9:ENEURO.0074-22.2022. [PMID: 35523583 PMCID: PMC9121915 DOI: 10.1523/eneuro.0074-22.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 11/21/2022] Open
Abstract
Excitatory amino acid transporters (EAATs) control visual signal transmission in the retina by rapidly removing glutamate released from photoreceptors and bipolar cells (BCs). Although it has been reported that EAAT2 and EAAT5 are expressed at presynaptic terminals of photoreceptors and some BCs in mammals, the distinct functions of these two glutamate transporters in retinal synaptic transmission, especially at a single synapse, remain elusive. In this study, we found that EAAT2 was expressed in all BC types while coexisting with EAAT5 in rod bipolar (RB) cells and several types of cone BCs from mice of either sex. Our immunohistochemical study, together with a recently published literature (Gehlen et al., 2021), showed that EAAT2 and EAAT5 were both located in RB axon terminals near release sites. Optogenetic, electrophysiological and pharmacological analyses, however, demonstrated that EAAT2 and EAAT5 regulated neurotransmission at RB→AII amacrine cell synapses in significantly different ways: EAAT5 dramatically affected both the peak amplitude and kinetics of postsynaptic responses in AIIs, whereas EAAT2 had either relatively small or opposite effects. By contrast, blockade of EAAT1/GLAST, which was exclusively expressed in Müller cells, showed no obvious effect on AII responses, indicating that glutamate uptake by Müller cells did not influence synaptic transmission from RB terminals. Furthermore, we found that temporal resolution at RB→AII synapses was reduced substantially by blockade of EAAT5 but not EAAT2. Taken together, our work reveals the distinct functions of EAAT2 and EAAT5 in signal transmission at RB ribbon synapses.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiang-Bin Ke
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| |
Collapse
|
10
|
Kovermann P, Engels M, Müller F, Fahlke C. Cellular Physiology and Pathophysiology of EAAT Anion Channels. Front Cell Neurosci 2022; 15:815279. [PMID: 35087380 PMCID: PMC8787812 DOI: 10.3389/fncel.2021.815279] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/13/2021] [Indexed: 11/17/2022] Open
Abstract
Excitatory amino acid transporters (EAATs) optimize the temporal resolution and energy demand of mammalian excitatory synapses by quickly removing glutamate from the synaptic cleft into surrounding neuronal and glial cells and ensuring low resting glutamate concentrations. In addition to secondary active glutamate transport, EAATs also function as anion channels. The channel function of these transporters is conserved in all homologs ranging from archaebacteria to mammals; however, its physiological roles are insufficiently understood. There are five human EAATs, which differ in their glutamate transport rates. Until recently the high-capacity transporters EAAT1, EAAT2, and EAAT3 were believed to conduct only negligible anion currents, with no obvious function in cell physiology. In contrast, the low-capacity glutamate transporters EAAT4 and EAAT5 are thought to regulate neuronal signaling as glutamate-gated channels. In recent years, new experimental approaches and novel animal models, together with the discovery of a human genetic disease caused by gain-of-function mutations in EAAT anion channels have enabled identification of the first physiological and pathophysiological roles of EAAT anion channels.
Collapse
|
11
|
Excitatory Amino Acid Transporter EAAT5 Improves Temporal Resolution in the Retina. eNeuro 2021; 8:ENEURO.0406-21.2021. [PMID: 34772693 PMCID: PMC8670604 DOI: 10.1523/eneuro.0406-21.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/16/2021] [Indexed: 11/21/2022] Open
Abstract
Excitatory amino acid transporters (EAATs) remove glutamate from the synaptic cleft. In the retina, EAAT1 and EAAT2 are considered the major glutamate transporters. However, it has not yet been possible to determine how EAAT5 shapes the retinal light responses because of the lack of a selective EAAT5 blocker or EAAT5 knock-out (KO) animal model. In this study, EAAT5 was found to be expressed in a punctate manner close to release sites of glutamatergic synapses in the mouse retina. Light responses from retinae of wild-type (WT) and of a newly generated model with a targeted deletion of EAAT5 (EAAT5-/-) were recorded in vitro using multielectrode arrays (MEAs). Flicker resolution was considerably lower in EAAT5-/- retinae than in WT retinae. The close proximity to the glutamate release site makes EAAT5 an ideal tool to improve temporal information processing in the retina by controlling information transfer at glutamatergic synapses.
Collapse
|
12
|
Grabner CP, Moser T. The mammalian rod synaptic ribbon is essential for Ca v channel facilitation and ultrafast synaptic vesicle fusion. eLife 2021; 10:63844. [PMID: 34617508 PMCID: PMC8594941 DOI: 10.7554/elife.63844] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 10/06/2021] [Indexed: 12/19/2022] Open
Abstract
Rod photoreceptors (PRs) use ribbon synapses to transmit visual information. To signal ‘no light detected’ they release glutamate continually to activate post-synaptic receptors. When light is detected glutamate release pauses. How a rod’s individual ribbon enables this process was studied here by recording evoked changes in whole-cell membrane capacitance from wild-type and ribbonless (Ribeye-ko) mice. Wild-type rods filled with high (10 mM) or low (0.5 mM) concentrations of the Ca2+-buffer EGTA created a readily releasable pool (RRP) of 87 synaptic vesicles (SVs) that emptied as a single kinetic phase with a τ<0.4 ms. The lower concentration of EGTA accelerated Cav channel opening and facilitated release kinetics. In contrast, ribbonless rods created a much smaller RRP of 22 SVs, and they lacked Cav channel facilitation; however, Ca2+ channel-release coupling remained tight. These release deficits caused a sharp attenuation of rod-driven scotopic light responses. We conclude that the synaptic ribbon facilitates Ca2+-influx and establishes a large RRP of SVs.
Collapse
Affiliation(s)
- Chad Paul Grabner
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute of Biophysical Chemistry, Göttingen, Germany.,Collaborative Research Center 1286 'Quantitative Synaptology', University of Göttingen, Göttingen, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute of Biophysical Chemistry, Göttingen, Germany.,Collaborative Research Center 1286 'Quantitative Synaptology', University of Göttingen, Göttingen, Germany.,Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
13
|
Hays CL, Sladek AL, Thoreson WB. Resting and stimulated mouse rod photoreceptors show distinct patterns of vesicle release at ribbon synapses. J Gen Physiol 2021; 152:211528. [PMID: 33175961 PMCID: PMC7664508 DOI: 10.1085/jgp.202012716] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/01/2020] [Accepted: 10/19/2020] [Indexed: 01/04/2023] Open
Abstract
The vertebrate visual system can detect and transmit signals from single photons. To understand how single-photon responses are transmitted, we characterized voltage-dependent properties of glutamate release in mouse rods. We measured presynaptic glutamate transporter anion current and found that rates of synaptic vesicle release increased with voltage-dependent Ca2+ current. Ca2+ influx and release rate also rose with temperature, attaining a rate of ∼11 vesicles/s/ribbon at -40 mV (35°C). By contrast, spontaneous release events at hyperpolarized potentials (-60 to -70 mV) were univesicular and occurred at random intervals. However, when rods were voltage clamped at -40 mV for many seconds to simulate maintained darkness, release occurred in coordinated bursts of 17 ± 7 quanta (mean ± SD; n = 22). Like fast release evoked by brief depolarizing stimuli, these bursts involved vesicles in the readily releasable pool of vesicles and were triggered by the opening of nearby ribbon-associated Ca2+ channels. Spontaneous release rates were elevated and bursts were absent after genetic elimination of the Ca2+ sensor synaptotagmin 1 (Syt1). This study shows that at the resting potential in darkness, rods release glutamate-filled vesicles from a pool at the base of synaptic ribbons at low rates but in Syt1-dependent bursts. The absence of bursting in cones suggests that this behavior may have a role in transmitting scotopic responses.
Collapse
Affiliation(s)
- Cassandra L Hays
- Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE.,Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Asia L Sladek
- Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Wallace B Thoreson
- Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE.,Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
14
|
Alam M, Yadav RK, Minj E, Tiwari A, Mehan S. Exploring Molecular Approaches in Amyotrophic Lateral Sclerosis: Drug Targets from Clinical and Pre-Clinical Findings. Curr Mol Pharmacol 2021; 14:263-280. [PMID: 32342825 DOI: 10.2174/1566524020666200427214356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/24/2019] [Accepted: 12/26/2019] [Indexed: 11/22/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease (MND) characterized by the death of upper and lower motor neurons (corticospinal tract) in the motor cortex, basal ganglia, brain stem, and spinal cord. The patient experiences the sign and symptoms between 55 to 75 years of age, which include impaired motor movement, difficulty in speaking and swallowing, grip loss, muscle atrophy, spasticity, and sometimes associated with memory and cognitive impairments. Median survival is 3 to 5 years after diagnosis and 5 to 10% of the patients live for more than 10 years. The limited intervention of pharmacologically active compounds, that are used clinically, is majorly associated with the narrow therapeutic index. Pre-clinically established experimental models, where neurotoxin methyl mercury mimics the ALS like behavioural and neurochemical alterations in rodents associated with neuronal mitochondrial dysfunctions and downregulation of adenyl cyclase mediated cAMP/CREB, is the main pathological hallmark for the progression of ALS in central as well in the peripheral nervous system. Despite the considerable investigation into neuroprotection, it still constrains treatment choices to strong care and organization of ALS complications. Therefore, this current review specially targeted the investigation of clinical and pre-clinical features available for ALS to understand the pathogenic mechanisms and to explore the pharmacological interventions associated with the up-regulation of intracellular adenyl cyclase/cAMP/ CREB and activation of mitochondrial-ETC coenzyme-Q10 as a future drug target in the amelioration of ALS mediated motor neuronal dysfunctions.
Collapse
Affiliation(s)
- Mamtaj Alam
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Rajeshwar K Yadav
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Elizabeth Minj
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Aarti Tiwari
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Sidharth Mehan
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| |
Collapse
|
15
|
Thoreson WB. Transmission at rod and cone ribbon synapses in the retina. Pflugers Arch 2021; 473:1469-1491. [PMID: 33779813 DOI: 10.1007/s00424-021-02548-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/29/2022]
Abstract
Light-evoked voltage responses of rod and cone photoreceptor cells in the vertebrate retina must be converted to a train of synaptic vesicle release events for transmission to downstream neurons. This review discusses the processes, proteins, and structures that shape this critical early step in vision, focusing on studies from salamander retina with comparisons to other experimental animals. Many mechanisms are conserved across species. In cones, glutamate release is confined to ribbon release sites although rods are also capable of release at non-ribbon sites. The role of non-ribbon release in rods remains unclear. Release from synaptic ribbons in rods and cones involves at least three vesicle pools: a readily releasable pool (RRP) matching the number of membrane-associated vesicles along the ribbon base, a ribbon reserve pool matching the number of additional vesicles on the ribbon, and an enormous cytoplasmic reserve. Vesicle release increases in parallel with Ca2+ channel activity. While the opening of only a few Ca2+ channels beneath each ribbon can trigger fusion of a single vesicle, sustained release rates in darkness are governed by the rate at which the RRP can be replenished. The number of vacant release sites, their functional status, and the rate of vesicle delivery in turn govern replenishment. Along with an overview of the mechanisms of exocytosis and endocytosis, we consider specific properties of ribbon-associated proteins and pose a number of remaining questions about this first synapse in the visual system.
Collapse
Affiliation(s)
- Wallace B Thoreson
- Truhlsen Eye Institute, Departments of Ophthalmology & Visual Sciences and Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
16
|
Hays CL, Sladek AL, Field GD, Thoreson WB. Properties of multivesicular release from mouse rod photoreceptors support transmission of single-photon responses. eLife 2021; 10:67446. [PMID: 33769285 PMCID: PMC8032395 DOI: 10.7554/elife.67446] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/20/2021] [Indexed: 01/18/2023] Open
Abstract
Vision under starlight requires rod photoreceptors to transduce and transmit single-photon responses to the visual system. Small single-photon voltage changes must therefore cause detectable reductions in glutamate release. We found that rods achieve this by employing mechanisms that enhance release regularity and its sensitivity to small voltage changes. At the resting membrane potential in darkness, mouse rods exhibit coordinated and regularly timed multivesicular release events, each consisting of ~17 vesicles and occurring two to three times more regularly than predicted by Poisson statistics. Hyperpolarizing rods to mimic the voltage change produced by a single photon abruptly reduced the probability of multivesicular release nearly to zero with a rebound increase at stimulus offset. Simulations of these release dynamics indicate that this regularly timed, multivesicular release promotes transmission of single-photon responses to post-synaptic rod-bipolar cells. Furthermore, the mechanism is efficient, requiring lower overall release rates than uniquantal release governed by Poisson statistics.
Collapse
Affiliation(s)
- Cassandra L Hays
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, United States.,Cellular and Integrative Physiology, Omaha, United States
| | - Asia L Sladek
- Pharmacology and Experimental Neuroscience, Omaha, United States
| | - Greg D Field
- Department of Neurobiology, Duke University School of Medicine, Durham, United States
| | - Wallace B Thoreson
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, United States.,Pharmacology and Experimental Neuroscience, Omaha, United States
| |
Collapse
|
17
|
Ghosh I, Sankhe R, Mudgal J, Arora D, Nampoothiri M. Spermidine, an autophagy inducer, as a therapeutic strategy in neurological disorders. Neuropeptides 2020; 83:102083. [PMID: 32873420 DOI: 10.1016/j.npep.2020.102083] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/18/2020] [Accepted: 07/26/2020] [Indexed: 02/06/2023]
Abstract
Spermidine is a naturally occurring endogenous polyamine synthesized from diamine putrescine. It is a well-known autophagy inducer that maintains cellular and neuronal homeostasis. Healthy brain development and function are dependent on brain polyamine concentration. Polyamines interact with the opioid system, glutamatergic signaling and neuroinflammation in the neuronal and glial compartments. Among the polyamines, spermidine is found highest in the human brain. Age-linked fluctuations in the spermidine levels may possibly contribute to the impairments in neural network and neurogenesis. Exogenously administered spermidine helps in the treatment of brain diseases. Further, current studies highlight the ability of spermidine to promote longevity by inducing autophagy. Still, the causal neuroprotective mechanism of spermidine in neuronal dysfunction remains unidentified. This review aims to summarize various neuroprotective effects of spermidine related to anti-aging/ anti-inflammatory properties and the prevention of neurotoxicity that helps in achieving beneficial effects in age-related neurological disorder. We also expose the signaling cascades modulated by spermidine which might result in therapeutic action. The present review highlights clinical studies along with in-vivo and in-vitro preclinical studies to provide a new dimension for the therapeutic potential of spermidine in neurological disorders.
Collapse
Affiliation(s)
- Indrani Ghosh
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Runali Sankhe
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Devinder Arora
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; School of Pharmacy and Pharmacology, MHIQ, QUM Network, Griffith University, Gold Coast, Queensland, Australia
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India.
| |
Collapse
|
18
|
Pfeiffer RL, Marc RE, Jones BW. Müller Cell Metabolic Signatures: Evolutionary Conservation and Disruption in Disease. Trends Endocrinol Metab 2020; 31:320-329. [PMID: 32187524 PMCID: PMC7188339 DOI: 10.1016/j.tem.2020.01.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/05/2019] [Accepted: 01/09/2020] [Indexed: 01/21/2023]
Abstract
Müller cells are glia that play important regulatory roles in retinal metabolism. These roles have been evolutionarily conserved across at least 300 million years. Müller cells have a tightly locked metabolic signature in the healthy retina, which rapidly degrades in response to insult and disease. This variation in metabolic signature occurs in a chaotic fashion, involving some central metabolic pathways. The cause of this divergence of Müller cells, from a single class with a unique metabolic signature to numerous separable metabolic classes, is currently unknown and illuminates potential alternative metabolic pathways that may be revealed in disease. Understanding the impacts of this heterogeneity on degenerate retinas and the implications for the metabolic support of surrounding neurons will be critical to long-term integration of retinal therapeutics for the restoration of visual perception following photoreceptor degeneration.
Collapse
Affiliation(s)
- Rebecca L Pfeiffer
- Moran Eye Center, University of Utah Department of Ophthalmology and Visual Sciences, 65 Mario Capecchi Drive, Salt Lake City, UT 84132, USA. *
| | - Robert E Marc
- Moran Eye Center, University of Utah Department of Ophthalmology and Visual Sciences, 65 Mario Capecchi Drive, Salt Lake City, UT 84132, USA; Signature Immunologics, Torrey, UT, USA
| | - Bryan W Jones
- Moran Eye Center, University of Utah Department of Ophthalmology and Visual Sciences, 65 Mario Capecchi Drive, Salt Lake City, UT 84132, USA.
| |
Collapse
|
19
|
Ingram NT, Sampath AP, Fain GL. Membrane conductances of mouse cone photoreceptors. J Gen Physiol 2020; 152:e201912520. [PMID: 31986199 PMCID: PMC7054858 DOI: 10.1085/jgp.201912520] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 12/19/2022] Open
Abstract
Vertebrate photoreceptor cells respond to light through a closure of CNG channels located in the outer segment. Multiple voltage-sensitive channels in the photoreceptor inner segment serve to transform and transmit the light-induced outer-segment current response. Despite extensive studies in lower vertebrates, we do not know how these channels produce the photoresponse of mammalian photoreceptors. Here we examined these ionic conductances recorded from single mouse cones in unlabeled, dark-adapted retinal slices. First, we show measurements of the voltage dependence of the light response. After block of voltage-gated Ca2+ channels, the light-dependent current was nearly linear within the physiological range of voltages with constant chord conductance and a reversal potential similar to that previously determined in lower vertebrate photoreceptors. At a dark resting membrane potential of -45 mV, cones maintain a standing Ca2+ current (iCa) between 15 and 20 pA. We characterized the time and voltage dependence of iCa and a calcium-activated anion channel. After constitutive closure of the CNG channels by the nonhydrolysable analogue GTP-γ-S, we observed a light-dependent increase in iCa followed by a Ca2+-activated K+ current, both probably the result of feedback from horizontal cells. We also recorded the hyperpolarization-activated cyclic nucleotide-gated (HCN) conductance (ih) and measured its current-voltage relationship and reversal potential. With small hyperpolarizations, ih activated with a time constant of 25 ms; activation was speeded with larger hyperpolarizations. Finally, we characterized two voltage-gated K+-conductances (iK). Depolarizing steps beginning at -10 mV activated a transient, outwardly rectifying iK blocked by 4-AP and insensitive to TEA. A sustained iK isolated through subtraction was blocked by TEA but was insensitive to 4-AP. The sustained iK had a nearly linear voltage dependence throughout the physiological voltage range of the cone. Together these data constitute the first comprehensive study of the channel conductances of mouse photoreceptors.
Collapse
Affiliation(s)
- Norianne T. Ingram
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, CA
| | - Alapakkam P. Sampath
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, CA
| | - Gordon L. Fain
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, CA
| |
Collapse
|
20
|
Grassmeyer JJ, Cahill AL, Hays CL, Barta C, Quadros RM, Gurumurthy CB, Thoreson WB. Ca 2+ sensor synaptotagmin-1 mediates exocytosis in mammalian photoreceptors. eLife 2019; 8:e45946. [PMID: 31172949 PMCID: PMC6588344 DOI: 10.7554/elife.45946] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 06/06/2019] [Indexed: 11/24/2022] Open
Abstract
To encode light-dependent changes in membrane potential, rod and cone photoreceptors utilize synaptic ribbons to sustain continuous exocytosis while making rapid, fine adjustments to release rate. Release kinetics are shaped by vesicle delivery down ribbons and by properties of exocytotic Ca2+ sensors. We tested the role for synaptotagmin-1 (Syt1) in photoreceptor exocytosis by using novel mouse lines in which Syt1 was conditionally removed from rods or cones. Photoreceptors lacking Syt1 exhibited marked reductions in exocytosis as measured by electroretinography and single-cell recordings. Syt1 mediated all evoked release in cones, whereas rods appeared capable of some slow Syt1-independent release. Spontaneous release frequency was unchanged in cones but increased in rods lacking Syt1. Loss of Syt1 did not alter synaptic anatomy or reduce Ca2+ currents. These results suggest that Syt1 mediates both phasic and tonic release at photoreceptor synapses, revealing unexpected flexibility in the ability of Syt1 to regulate Ca2+-dependent synaptic transmission.
Collapse
Affiliation(s)
- Justin J Grassmeyer
- Truhlsen Eye Institute, Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaUnited States
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaUnited States
| | - Asia L Cahill
- Truhlsen Eye Institute, Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaUnited States
| | - Cassandra L Hays
- Truhlsen Eye Institute, Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaUnited States
- Department of Cellular and Integrative PhysiologyUniversity of Nebraska Medical CenterOmahaUnited States
| | - Cody Barta
- Truhlsen Eye Institute, Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaUnited States
| | - Rolen M Quadros
- Mouse Genome Engineering Core Facility, Vice Chancellor for Research OfficeUniversity of Nebraska Medical CenterOmahaUnited States
| | - Channabasavaiah B Gurumurthy
- Mouse Genome Engineering Core Facility, Vice Chancellor for Research OfficeUniversity of Nebraska Medical CenterOmahaUnited States
- Developmental Neuroscience, Munroe Meyer Institute for Genetics and RehabilitationUniversity of Nebraska Medical CenterOmahaUnited States
| | - Wallace B Thoreson
- Truhlsen Eye Institute, Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaUnited States
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaUnited States
| |
Collapse
|
21
|
Hellmer CB, Clemons MR, Nawy S, Ichinose T. A group I metabotropic glutamate receptor controls synaptic gain between rods and rod bipolar cells in the mouse retina. Physiol Rep 2018; 6:e13885. [PMID: 30338673 PMCID: PMC6194217 DOI: 10.14814/phy2.13885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 09/10/2018] [Accepted: 09/16/2018] [Indexed: 11/24/2022] Open
Abstract
The canonical mGluR6-Trpm1 pathway that generates the sign-inverting signal between photoreceptors and ON bipolar cells has been well described. However, one type of ON bipolar cell, the rod bipolar cell (RBC), additionally is thought to express the group I mGluRs whose function is unknown. We examined the role of group I mGluRs in mouse RBCs and here provide evidence that it controls synaptic gain between rods and RBCs. In dark-adapted conditions, the mGluR1 antagonists LY367385 and (RS)-1-Aminoindan-1,5-dicarboxylic acid, but not the mGluR5 antagonist 2-Methyl-6-(phenylethynyl)pyridine hydrochloride reduced the light-evoked responses in RBCs indicating that mGluR1, but not mGluR5, serves to potentiate RBC responses. Perturbing the downstream phospholipase C (PLC)-protein kinase C (PKC) pathway by inhibiting PLC, tightly buffering intracellular Ca2+ , or preventing its release from intracellular stores reduced the synaptic potentiation by mGluR1. The effect of mGluR1 activation was dependent upon adaptation state, strongly increasing the synaptic gain in dark-, but not in light-adapted retinas, or in the presence of a moderate background light, consistent with the idea that mGluR1 activation requires light-dependent glutamate release from rods. Moreover, immunostaining revealed that protein kinase Cα (PKCα) is more strongly expressed in RBC dendrites in dark-adapted conditions, revealing an additional mechanism behind the loss of mGluR1 potentiation. In light-adapted conditions, exogenous activation of mGluR1 with the agonist 3,5-Dihydroxyphenylglycine increased the mGluR6 currents in some RBCs and decreased it in others, suggesting an additional action of mGluR1 that is unmasked in the light-adapted state. Elevating intracellular free Ca2+ , consistently resulted in a decrease in synaptic gain. Our results provide evidence that mGluR1 controls the synaptic gain in RBCs.
Collapse
Affiliation(s)
- Chase B. Hellmer
- Department of Ophthalmology, Visual and Anatomical SciencesWayne State University School of MedicineDetroitMichigan48201
| | - Melissa Rampino Clemons
- Dominic P Purpura Dept. of NeuroscienceAlbert Einstein College of Medicine BronxBronxNew York10461
| | - Scott Nawy
- Dominic P Purpura Dept. of NeuroscienceAlbert Einstein College of Medicine BronxBronxNew York10461
- Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaNebraska68198
| | - Tomomi Ichinose
- Department of Ophthalmology, Visual and Anatomical SciencesWayne State University School of MedicineDetroitMichigan48201
| |
Collapse
|
22
|
Ohkuma M, Kaneda M, Yoshida S, Fukuda A, Miyachi E. Optical measurement of glutamate in slice preparations of the mouse retina. Neurosci Res 2018. [PMID: 29522783 DOI: 10.1016/j.neures.2018.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Signaling by glutamatergic synapses plays an important role in visual processing in the retina. In this study, we used an enzyme-linked fluorescence assay system to monitor the dynamics of extracellular glutamate in a slice preparation from the mouse retina. High K stimulation induced an elevation of fluorescence in the inner plexiform layer (IPL) of the retina when glutamate transporters were inhibited by dl-threo-β-benzyloxyaspartic acid (TBOA). The high K-induced fluorescence signals in the IPL were inhibited by the calcium channel blocker Cd2+. Blockade of GABAergic and glycinergic circuits by picrotoxin and strychnine also elevated the fluorescence signals in the IPL. Thus, the enzyme-linked fluorescence assay system might be useful for monitoring the bulk concentration of extracellular glutamate released by synapses in the inner retina.
Collapse
Affiliation(s)
- M Ohkuma
- Department of Physiology, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - M Kaneda
- Department of Physiology, Nippon Medical School, Sendagi 1-1-5, Bunkyo-ku, Tokyo 160-8602, Japan.
| | - S Yoshida
- Department of Environmental and Life Sciences, Toyohashi University of Technology, Toyohashi, Aichi 441-8580, Japan
| | - A Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - E Miyachi
- Department of Physiology, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| |
Collapse
|
23
|
O'Donovan SM, Sullivan CR, McCullumsmith RE. The role of glutamate transporters in the pathophysiology of neuropsychiatric disorders. NPJ SCHIZOPHRENIA 2017; 3:32. [PMID: 28935880 PMCID: PMC5608761 DOI: 10.1038/s41537-017-0037-1] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 08/24/2017] [Accepted: 09/01/2017] [Indexed: 02/08/2023]
Abstract
Altered glutamate transporter expression is a common feature of many neuropsychiatric conditions, including schizophrenia. Excitatory amino acid transporters (EAATs) are responsible for the reuptake of glutamate, preventing non-physiological spillover from the synapse. Postmortem studies have revealed significant dysregulation of EAAT expression in various brain regions at the cellular and subcellular level. Recent animal studies have also demonstrated a role for glutamate spillover as a mechanism of disease. In this review, we describe current evidence for the role of glutamate transporters in regulating synaptic plasticity and transmission. In neuropsychiatric conditions, EAAT splice variant expression is altered. There are changes in the localization of the transporters and disruption of the metabolic and structural protein network that supports EAAT activity. This results in aberrant neuroplasticity and excitatory signaling, contributing to the symptoms associated with neuropsychiatric disease. Understanding the complex functions of glutamate transporters will clarify the relevance of their role in the pathophysiology of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sinead M O'Donovan
- Department of Psychiatry, University of Cincinnati, Cincinnati, OH, 45221, USA.
| | - Courtney R Sullivan
- Department of Psychiatry, University of Cincinnati, Cincinnati, OH, 45221, USA
| | | |
Collapse
|
24
|
Functional Roles of Complexin 3 and Complexin 4 at Mouse Photoreceptor Ribbon Synapses. J Neurosci 2017; 36:6651-67. [PMID: 27335398 DOI: 10.1523/jneurosci.4335-15.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 05/10/2016] [Indexed: 01/22/2023] Open
Abstract
UNLABELLED Complexins (Cplxs) are SNARE complex regulators controlling the speed and Ca(2+) sensitivity of SNARE-mediated synaptic vesicle fusion. We have shown previously that photoreceptor ribbon synapses in mouse retina are equipped with Cplx3 and Cplx4 and that lack of both Cplxs perturbs photoreceptor ribbon synaptic function; however, Cplx3/4 function in photoreceptor synaptic transmission remained elusive. To investigate Cplx3/4 function in photoreceptor ribbon synapses, voltage-clamp recordings from postsynaptic horizontal cells were performed in horizontal slice preparations of Cplx3/4 wild-type (WT) and Cplx3/4 double knock-out (DKO) mice. We measured tonic activity in light and dark, current responses to changes in luminous intensity, and electrically evoked postsynaptic responses. Cplx3/4 decreased the frequency of tonic events and shifted their amplitude distribution to smaller values. Light responses were sustained in the presence of Cplx3/4, but transient in their absence. Finally, Cplx3/4 increased synaptic vesicle release evoked by electrical stimulation. Using electron microscopy, we quantified the number of synaptic vesicles at presynaptic ribbons after light or dark adaptation. In Cplx3/4 WT photoreceptors, the number of synaptic vesicles associated with the ribbon base close to the release site was significantly lower in light than in dark. This is in contrast to Cplx3/4 DKO photoreceptors, in which the number of ribbon-associated synaptic vesicles remained unchanged regardless of the adaptational state. Our results indicate a suppressing and a facilitating action of Cplx3/4 on Ca(2+)-dependent tonic and evoked neurotransmitter release, respectively, and a regulatory role in the adaptation-dependent availability of synaptic vesicles for release at photoreceptor ribbon synapses. SIGNIFICANCE STATEMENT Synaptic vesicle fusion at active zones of chemical synapses is executed by SNARE complexes. Complexins (Cplxs) are SNARE complex regulators and photoreceptor ribbon synapses are equipped with Cplx3 and Cplx4. The absence of both Cplxs perturbs ribbon synaptic function. Because we lack information on Cplx function in photoreceptor synaptic transmission, we investigated Cplx function using voltage-clamp recordings from postsynaptic horizontal cells of Cplx3/4 wild-type and Cplx3/4 double knock-out mice and quantified synaptic vesicle number at the ribbon after light and dark adaptation using electron microscopy. The findings reveal a suppressing action of Cplx3/4 on tonic neurotransmitter release, a facilitating action on evoked release, and a regulatory role of Cplx3/4 in the adaptation-dependent availability of synaptic vesicles at mouse photoreceptor ribbon synapses.
Collapse
|
25
|
Dai J, Fu Y, Zeng Y, Li S, Qin Yin Z. Improved retinal function in RCS rats after suppressing the over-activation of mGluR5. Sci Rep 2017; 7:3546. [PMID: 28615682 PMCID: PMC5471183 DOI: 10.1038/s41598-017-03702-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/03/2017] [Indexed: 01/09/2023] Open
Abstract
Müller cells maintain retinal synaptic homeostasis by taking up glutamate from the synaptic cleft and transporting glutamine back to the neurons. To study the interaction between Müller cells and photoreceptors, we injected either DL-α-aminoadipate or L-methionine sulfoximine–both inhibitors of glutamine synthetase–subretinally in rats. Following injection, the a-wave of the electroretinogram (ERG) was attenuated, and metabotropic glutamate receptor 5 (mGluR5) was activated. Selective antagonism of mGluR5 by 2-methyl-6-(phenylethynyl)-pyridine increased the ERG a-wave amplitude and also increased rhodopsin expression. Conversely, activation of mGluR5 by the agonist, (R,S)-2-chloro-5-hydroxyphenylglycine, decreased both the a-wave amplitude and rhodopsin expression, but upregulated expression of Gq alpha subunit and phospholipase C βIII. Overexpression of mGluR5 reduced the inward-rectifying potassium ion channel (Kir) current and decreased the expression of Kir4.1 and aquaporin-4 (AQP4). Further experiments indicated that mGluR5 formed a macromolecular complex with these two membrane channels. Lastly, increased expression of mGluR5 was found in Royal College of Surgeons rats–a model of retinitis pigmentosa (RP). Inhibition of mGluR5 in this model restored the amplitude of ERG features, and reduced the expression of glial fibrillary acidic protein. These results suggest that mGluR5 may be worth considering as a potential therapeutic target in RP.
Collapse
Affiliation(s)
- Jiaman Dai
- Bioengineering College, Chongqing University, Chongqing, 400040, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Yan Fu
- 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
| | - Yuxiao Zeng
- 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
| | - Shiying 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.
| | - Zheng Qin Yin
- Bioengineering College, Chongqing University, Chongqing, 400040, China. .,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
|
26
|
Shaping of Signal Transmission at the Photoreceptor Synapse by EAAT2 Glutamate Transporters. eNeuro 2017; 4:eN-NWR-0339-16. [PMID: 28612046 PMCID: PMC5467398 DOI: 10.1523/eneuro.0339-16.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 05/12/2017] [Accepted: 05/25/2017] [Indexed: 12/30/2022] Open
Abstract
Photoreceptor ribbon synapses tonically release glutamate. To ensure efficient signal transmission and prevent glutamate toxicity, a highly efficient glutamate removal system provided by members of the SLC1 gene family is required. By using a combination of biophysical and in vivo studies, we elucidate the role of excitatory amino acid transporter 2 (EAAT2) proteins in synaptic glutamate homeostasis at the zebrafish photoreceptor synapse. The main glutamate sink is provided by the glial EAAT2a, reflected by reduced electroretinographic responses in EAAT2a-depleted larvae. EAAT2b is located on the tips of cone pedicles and contributes little to glutamate reuptake. However, this transporter displays both a large chloride conductance and leak current, being important in stabilizing the cone resting potential. This work demonstrates not only how proteins originating from the same gene family can complement each other’s expression profiles and biophysical properties, but also how presynaptic and glial transporters are coordinated to ensure efficient synaptic transmission at glutamatergic synapses of the central nervous system.
Collapse
|
27
|
Rose CR, Ziemens D, Untiet V, Fahlke C. Molecular and cellular physiology of sodium-dependent glutamate transporters. Brain Res Bull 2016; 136:3-16. [PMID: 28040508 DOI: 10.1016/j.brainresbull.2016.12.013] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/20/2016] [Accepted: 12/21/2016] [Indexed: 02/04/2023]
Abstract
Glutamate is the major excitatory transmitter in the vertebrate brain. After its release from presynaptic nerve terminals, it is rapidly taken up by high-affinity sodium-dependent plasma membrane transporters. While both neurons and glial cells express these excitatory amino acid transporters (EAATs), the majority of glutamate uptake is accomplished by astrocytes, which convert synaptically-released glutamate to glutamine or feed it into their own metabolism. Glutamate uptake by astrocytes not only shapes synaptic transmission by regulating the availability of glutamate to postsynaptic neuronal receptors, but also protects neurons from hyper-excitability and subsequent excitotoxic damage. In the present review, we provide an overview of the molecular and cellular characteristics of sodium-dependent glutamate transporters and their associated anion permeation pathways, with a focus on astrocytic glutamate transport. We summarize their functional properties and roles within tripartite synapses under physiological and pathophysiological conditions, exemplifying the intricate interactions and interrelationships between neurons and glial cells in the brain.
Collapse
Affiliation(s)
- Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Germany.
| | - Daniel Ziemens
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Germany
| | - Verena Untiet
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, Germany
| | - Christoph Fahlke
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, Germany
| |
Collapse
|
28
|
Stereotyped initiation of retinal waves by bipolar cells via presynaptic NMDA autoreceptors. Nat Commun 2016; 7:12650. [PMID: 27586999 PMCID: PMC5025778 DOI: 10.1038/ncomms12650] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 07/18/2016] [Indexed: 02/07/2023] Open
Abstract
Glutamatergic retinal waves, the spontaneous patterned neural activities propagating among developing retinal ganglion cells (RGCs), instruct the activity-dependent refinement of visuotopic maps. However, its initiation and underlying mechanism remain largely elusive. Here using larval zebrafish and multiple in vivo approaches, we discover that bipolar cells (BCs) are responsible for the generation of glutamatergic retinal waves. The wave originates from BC axon terminals (ATs) and propagates laterally to nearby BCs and vertically to downstream RGCs and the optic tectum. Its initiation is triggered by the activation of and consequent glutamate release from BC ATs, and is mediated by the N-methyl-D-aspartate subtype of glutamate receptors (NMDARs) expressed at these ATs. Intercellular asymmetry of NMDAR expression at BC ATs enables the preferential initiation of waves at the temporal retina, where BC ATs express more NMDARs. Thus, our findings indicate that glutamatergic retinal waves are initiated by BCs through a presynaptic NMDA autoreceptor-dependent process. Retinal waves are important for visual system development. However, the mechanism involved in their generation remains largely unknown. Here using in vivo two-photon imaging the authors identify the presence of retinal waves in zebrafish larvae and find that they are initiated at bipolar cells via presynaptic NMDARs.
Collapse
|
29
|
Cork KM, Van Hook MJ, Thoreson WB. Mechanisms, pools, and sites of spontaneous vesicle release at synapses of rod and cone photoreceptors. Eur J Neurosci 2016; 44:2015-27. [PMID: 27255664 DOI: 10.1111/ejn.13288] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 05/21/2016] [Accepted: 05/24/2016] [Indexed: 01/31/2023]
Abstract
Photoreceptors have depolarized resting potentials that stimulate calcium-dependent release continuously from a large vesicle pool but neurons can also release vesicles without stimulation. We characterized the Ca(2+) dependence, vesicle pools, and release sites involved in spontaneous release at photoreceptor ribbon synapses. In whole-cell recordings from light-adapted horizontal cells (HCs) of tiger salamander retina, we detected miniature excitatory post-synaptic currents (mEPSCs) when no stimulation was applied to promote exocytosis. Blocking Ca(2+) influx by lowering extracellular Ca(2+) , by application of Cd(2+) and other agents reduced the frequency of mEPSCs but did not eliminate them, indicating that mEPSCs can occur independently of Ca(2+) . We also measured release presynaptically from rods and cones by examining quantal glutamate transporter anion currents. Presynaptic quantal event frequency was reduced by Cd(2+) or by increased intracellular Ca(2+) buffering in rods, but not in cones, that were voltage clamped at -70 mV. By inhibiting the vesicle cycle with bafilomycin, we found the frequency of mEPSCs declined more rapidly than the amplitude of evoked excitatory post-synaptic currents (EPSCs) suggesting a possible separation between vesicle pools in evoked and spontaneous exocytosis. We mapped sites of Ca(2+) -independent release using total internal reflectance fluorescence (TIRF) microscopy to visualize fusion of individual vesicles loaded with dextran-conjugated pHrodo. Spontaneous release in rods occurred more frequently at non-ribbon sites than evoked release events. The function of Ca(2+) -independent spontaneous release at continuously active photoreceptor synapses remains unclear, but the low frequency of spontaneous quanta limits their impact on noise.
Collapse
Affiliation(s)
- Karlene M Cork
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, 4050 Durham Research Center, University of Nebraska Medical Center, Omaha, NE, 68198-5840, USA.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Matthew J Van Hook
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, 4050 Durham Research Center, University of Nebraska Medical Center, Omaha, NE, 68198-5840, USA
| | - Wallace B Thoreson
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, 4050 Durham Research Center, University of Nebraska Medical Center, Omaha, NE, 68198-5840, USA.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
30
|
RIM1/2-Mediated Facilitation of Cav1.4 Channel Opening Is Required for Ca2+-Stimulated Release in Mouse Rod Photoreceptors. J Neurosci 2015; 35:13133-47. [PMID: 26400943 DOI: 10.1523/jneurosci.0658-15.2015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Night blindness can result from impaired photoreceptor function and a subset of cases have been linked to dysfunction of Cav1.4 calcium channels and in turn compromised synaptic transmission. Here, we show that active zone proteins RIM1/2 are important regulators of Cav1.4 channel function in mouse rod photoreceptors and thus synaptic activity. The conditional double knock-out (cdko) of RIM1 and RIM2 from rods starting a few weeks after birth did not change Cav1.4 protein expression at rod ribbon synapses nor was the morphology of the ribbon altered. Heterologous overexpression of RIM2 with Cav1.4 had no significant influence on current density when examined with BaCl2 as the charge carrier. Nonetheless, whole-cell voltage-clamp recordings from cdko rods revealed a profound reduction in Ca(2+) currents. Concomitantly, we observed a 4-fold reduction in spontaneous miniature release events from the cdko rod terminals and an almost complete absence of evoked responses when monitoring changes in membrane incorporation after strong step depolarizations. Under control conditions, 49 and 83 vesicles were released with 0.2 and 1 s depolarizations, respectively, which is close to the maximal number of vesicles estimated to be docked at the base of the ribbon active zone, but without RIM1/2, only a few vesicles were stimulated for release after a 1 s stimulation. In conclusion, our study shows that RIM1/2 potently enhance the influx of Ca(2+) into rod terminals through Cav1.4 channels, which is vitally important for the release of vesicles from the rod ribbon. Significance statement: Active zone scaffolding proteins are thought to bring multiple components involved in Ca(2+)-dependent exocytosis into functional interactions. We show that removal of scaffolding proteins RIM1/2 from rod photoreceptor ribbon synapses causes a dramatic loss of Ca(2+) influx through Cav1.4 channels and a correlated reduction in evoked release, yet the channels remain localized to synaptic ribbons in a normal fashion. Our findings strongly argue that RIM1/2 facilitate Ca(2+) entry and in turn Ca(2+) evoked release by modulating Cav1.4 channel openings; however, RIM1/2 are not needed for the retention of Cav1.4 at the synapse. In summary, a key function of RIM1/2 at rod ribbons is to enhance Cav1.4 channel activity, possibly through direct or indirect modulation of the channel.
Collapse
|
31
|
Codocedo JF, Inestrosa NC. Environmental control of microRNAs in the nervous system: Implications in plasticity and behavior. Neurosci Biobehav Rev 2015; 60:121-38. [PMID: 26593111 DOI: 10.1016/j.neubiorev.2015.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 10/24/2015] [Accepted: 10/26/2015] [Indexed: 02/07/2023]
Abstract
The discovery of microRNAs (miRNAs) a little over 20 years ago was revolutionary given that miRNAs are essential to numerous physiological and physiopathological processes. Currently, several aspects of the biogenic process of miRNAs and of the translational repression mechanism exerted on their targets mRNAs are known in detail. In fact, the development of bioinformatics tools for predicting miRNA targets has established that miRNAs have the potential to regulate almost all known biological processes. Therefore, the identification of the signals and molecular mechanisms that regulate miRNA function is relevant to understanding the role of miRNAs in both pathological and adaptive processes. Recently, a series of studies has focused on miRNA expression in the brain, establishing that their levels are altered in response to various environmental factors (EFs), such as light, sound, odorants, nutrients, drugs and stress. In this review, we discuss how exposure to various EFs modulates the expression and function of several miRNAs in the nervous system and how this control determines adaptation to their environment, behavior and disease state.
Collapse
Affiliation(s)
- Juan F Codocedo
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia; Centro UC Síndrome de Down, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
32
|
Contreras L. Role of AGC1/aralar in the metabolic synergies between neuron and glia. Neurochem Int 2015; 88:38-46. [DOI: 10.1016/j.neuint.2015.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 03/30/2015] [Accepted: 04/07/2015] [Indexed: 10/23/2022]
|
33
|
Hurley JB, Lindsay KJ, Du J. Glucose, lactate, and shuttling of metabolites in vertebrate retinas. J Neurosci Res 2015; 93:1079-92. [PMID: 25801286 PMCID: PMC4720126 DOI: 10.1002/jnr.23583] [Citation(s) in RCA: 175] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 02/11/2015] [Accepted: 02/12/2015] [Indexed: 02/06/2023]
Abstract
The vertebrate retina has specific functions and structures that give it a unique set of constraints on the way in which it can produce and use metabolic energy. The retina's response to illumination influences its energy requirements, and the retina's laminated structure influences the extent to which neurons and glia can access metabolic fuels. There are fundamental differences between energy metabolism in retina and that in brain. The retina relies on aerobic glycolysis much more than the brain does, and morphological differences between retina and brain limit the types of metabolic relationships that are possible between neurons and glia. This Mini-Review summarizes the unique metabolic features of the retina with a focus on the role of lactate shuttling.
Collapse
Affiliation(s)
- James B. Hurley
- Departments of Biochemistry and Ophthalmology, University of Washington, Seattle, Washington
| | - Kenneth J. Lindsay
- Departments of Biochemistry and Ophthalmology, University of Washington, Seattle, Washington
| | - Jianhai Du
- Departments of Biochemistry and Ophthalmology, University of Washington, Seattle, Washington
| |
Collapse
|
34
|
Brandli A. Remote Limb Ischemic Preconditioning: A Neuroprotective Technique in Rodents. J Vis Exp 2015:e52213. [PMID: 26065365 DOI: 10.3791/52213] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Sublethal ischemia protects tissues against subsequent, more severe ischemia through the upregulation of endogenous mechanisms in the affected tissue. Sublethal ischemia has also been shown to upregulate protective mechanisms in remote tissues. A brief period of ischemia (5-10 min) in the hind limb of mammals induces self-protective responses in the brain, lung, heart and retina. The effect is known as remote ischemic preconditioning (RIP). It is a therapeutically promising way of protecting vital organs, and is already under clinical trials for heart and brain injuries. This publication demonstrates a controlled, minimally invasive method of making a limb - specifically the hind limb of a rat - ischemic. A blood pressure cuff developed for use in human neonates is connected to a manual sphygmomanometer and used to apply 160 mmHg pressure around the upper part of the hind limb. A probe designed to detect skin temperature is used to verify the ischemia, by recording the drop in skin temperature caused by pressure-induced occlusion of the leg arteries, and the rise in temperature which follows release of the cuff. This method of RIP affords protection to the rat retina against bright light-induced damage and degeneration.
Collapse
Affiliation(s)
- Alice Brandli
- Discipline of Physiology and Bosch Institute, Sydney Medical School, University of Sydney;
| |
Collapse
|
35
|
|
36
|
Marx MC, Billups D, Billups B. Maintaining the presynaptic glutamate supply for excitatory neurotransmission. J Neurosci Res 2015; 93:1031-44. [PMID: 25648608 DOI: 10.1002/jnr.23561] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/04/2015] [Accepted: 01/05/2015] [Indexed: 01/09/2023]
Abstract
Glutamate released from synapses during excitatory neurotransmission must be rapidly recycled to maintain neuronal communication. This review evaluates data from physiological experiments at hippocampal CA3 to CA1 synapses and the calyx of Held synapse in the brainstem to analyze quantitatively the rates of release and resupply of glutamate required to sustain neurotransmission. We calculate that, without efficient recycling, the presynaptic glutamate supply will be exhausted within about a minute of normal synaptic activity. We also discuss replenishment of the presynaptic pool by diffusion from the soma, direct uptake of glutamate back into the presynaptic terminal, and uptake of glutamate precursor molecules. Diffusion of glutamate from the soma is calculated to be fast enough to resupply presynaptic glutamate in the hippocampus but not at the calyx of Held. However, because the somatic cytoplasm will also quickly run out of glutamate and synapses can function continually even if the presynaptic axon is severed, mechanisms other than diffusion must be present to resupply glutamate for release. Direct presynaptic uptake of glutamate is not present at the calyx of Held but may play a role in glutamate recycling in the hippocampus. Alternatively, glutamine or tricarboxylic acid cycle intermediates released from glia can serve as a precursor for glutamate in synaptic terminals, and we calculate that the magnitude of presynaptic glutamine uptake is sufficient to supply enough glutamate to sustain neurotransmission. The nature of these mechanisms, their relative abundance, and the co-ordination between them remain areas of intensive investigation.
Collapse
Affiliation(s)
- Mari-Carmen Marx
- Eccles Institute of Neuroscience, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Daniela Billups
- Eccles Institute of Neuroscience, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Brian Billups
- Eccles Institute of Neuroscience, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
37
|
The excitatory amino acid carrier 1 (EAAC1) in the rat nucleus of the solitary tract: subcellular localization suggests no major role in glutamate clearance. Brain Struct Funct 2014; 221:1113-24. [DOI: 10.1007/s00429-014-0958-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 12/03/2014] [Indexed: 11/26/2022]
|
38
|
Pyruvate kinase and aspartate-glutamate carrier distributions reveal key metabolic links between neurons and glia in retina. Proc Natl Acad Sci U S A 2014; 111:15579-84. [PMID: 25313047 DOI: 10.1073/pnas.1412441111] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Symbiotic relationships between neurons and glia must adapt to structures, functions, and metabolic roles of the tissues they are in. We show here that Müller glia in retinas have specific enzyme deficiencies that can enhance their ability to synthesize Gln. The metabolic cost of these deficiencies is that they impair the Müller cell's ability to metabolize Glc. We show here that the cells can compensate for this deficiency by using metabolites produced by neurons. Müller glia are deficient for pyruvate kinase (PK) and for aspartate/glutamate carrier 1 (AGC1), a key component of the malate-aspartate shuttle. In contrast, photoreceptor neurons express AGC1 and the M2 isoform of pyruvate kinase, which is commonly associated with aerobic glycolysis in tumors, proliferating cells, and some other cell types. Our findings reveal a previously unidentified type of metabolic relationship between neurons and glia. Müller glia compensate for their unique metabolic adaptations by using lactate and aspartate from neurons as surrogates for their missing PK and AGC1.
Collapse
|
39
|
Tse DY, Chung I, Wu SM. Pharmacological inhibitions of glutamate transporters EAAT1 and EAAT2 compromise glutamate transport in photoreceptor to ON-bipolar cell synapses. Vision Res 2014; 103:49-62. [PMID: 25152321 DOI: 10.1016/j.visres.2014.07.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 07/18/2014] [Accepted: 07/22/2014] [Indexed: 11/30/2022]
Abstract
To maintain reliable signal transmission across a synapse, free synaptic neurotransmitters must be removed from the cleft in a timely manner. In the first visual synapse, this critical task is mainly undertaken by glutamate transporters (EAATs). Here we study the differential roles of the EAAT1, EAAT2 and EAAT5 subtypes in glutamate (GLU) uptake at the photoreceptor-to-depolarizing bipolar cell synapse in intact dark-adapted retina. Various doses of EAAT blockers and/or GLU were injected into the eye before the electroretinogram (ERG) was measured. Their effectiveness and potency in inhibiting the ERG b-wave were studied to determine their relative contributions to the GLU clearing activity at the synapse. The results showed that EAAT1 and EAAT2 plays different roles. Selectively blocking glial EAAT1 alone using UCPH101 inhibited the b-wave 2-24h following injection, suggesting a dominating role of EAAT1 in the overall GLU clearing capacity in the synaptic cleft. Selectively blocking EAAT2 on photoreceptor terminals had no significant effect on the b-wave, but increased the potency of exogenous GLU in inhibiting the b-wave. These suggest that EAAT2 play a secondary yet significant role in the GLU reuptake activity at the rod and the cone output synapses. Additionally, we have verified our electrophysiological findings with double-label immunohistochemistry, and extend the literature on the spatial distribution of EAAT2 splice variants in the mouse retina.
Collapse
Affiliation(s)
- Dennis Y Tse
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA.
| | - Inyoung Chung
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA; Department of Ophthalmology, Gyeongsang National University, Jinju, Republic of Korea
| | - Samuel M Wu
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
40
|
Tse DY, Chung I, Wu SM. Possible roles of glutamate transporter EAAT5 in mouse cone depolarizing bipolar cell light responses. Vision Res 2014; 103:63-74. [PMID: 24972005 DOI: 10.1016/j.visres.2014.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 06/11/2014] [Accepted: 06/18/2014] [Indexed: 10/25/2022]
Abstract
A remarkable feature of neuronal glutamate transporters (EAATs) is their dual functions of classical carriers and ligand-gated chloride (Cl(-)) channels. Cl(-) conductance is rapidly activated by glutamate in subtype EAAT5, which mediates light responses in depolarizing bipolar cells (DBC) in retinae of lower vertebrates. In this study, we examine whether EAAT5 also mediates the DBC light response in mouse. We took advantage of an infrared illuminated micro-injection system, and studied the effects of the EAAT blocker (TBOA) and a glutamate receptor agonist (LAP4) on the mouse electroretinogram (ERG) b-wave responses. Our results showed that TBOA and LAP4 shared similar temporal patterns of inhibition: both inhibited the ERG b-wave shortly after injection and recovered with similar time courses. TBOA inhibited the b-wave completely at mesopic light intensity with an IC50 value about 1 log unit higher than that of LAP4. The inhibitory effects of TBOA and LAP4 were found to be additive in the photopic range. Furthermore, TBOA alone inhibited the b-wave in the cone operative range in knockout mice lacking DBCRs at a low concentration that did not alter synaptic glutamate clearance activity. It also produced a stronger inhibition than that of LAP4 on the cone-driven b-wave measured with a double flash method in wildtype mice. These electrophysiological data suggest a significant role for EAAT5 in mediating cone-driven DBC light responses. Our immunohistochemistry data indicated the presence of postsynaptic EAAT5 on some DBCCs and some DBCRs, providing an anatomical basis for EAAT5's role in DBC light responses.
Collapse
Affiliation(s)
- Dennis Y Tse
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA.
| | - Inyoung Chung
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA; Department of Ophthalmology, Gyeongsang National University, Jinju, Republic of Korea
| | - Samuel M Wu
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
41
|
Kainate receptors mediate signaling in both transient and sustained OFF bipolar cell pathways in mouse retina. J Neurosci 2014; 34:6128-39. [PMID: 24790183 DOI: 10.1523/jneurosci.4941-13.2014] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
A fundamental question in sensory neuroscience is how parallel processing is implemented at the level of molecular and circuit mechanisms. In the retina, it has been proposed that distinct OFF cone bipolar cell types generate fast/transient and slow/sustained pathways by the differential expression of AMPA- and kainate-type glutamate receptors, respectively. However, the functional significance of these receptors in the intact circuit during light stimulation remains unclear. Here, we measured glutamate release from mouse bipolar cells by two-photon imaging of a glutamate sensor (iGluSnFR) expressed on postsynaptic amacrine and ganglion cell dendrites. In both transient and sustained OFF layers, cone-driven glutamate release from bipolar cells was blocked by antagonists to kainate receptors but not AMPA receptors. Electrophysiological recordings from bipolar and ganglion cells confirmed the essential role of kainate receptors for signaling in both transient and sustained OFF pathways. Kainate receptors mediated responses to contrast modulation up to 20 Hz. Light-evoked responses in all mouse OFF bipolar pathways depend on kainate, not AMPA, receptors.
Collapse
|
42
|
Khatri N, Man HY. Synaptic activity and bioenergy homeostasis: implications in brain trauma and neurodegenerative diseases. Front Neurol 2013; 4:199. [PMID: 24376435 PMCID: PMC3858785 DOI: 10.3389/fneur.2013.00199] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 11/26/2013] [Indexed: 12/14/2022] Open
Abstract
Powered by glucose metabolism, the brain is the most energy-demanding organ in our body. Adequate ATP production and regulation of the metabolic processes are essential for the maintenance of synaptic transmission and neuronal function. Glutamatergic synaptic activity utilizes the largest portion of bioenergy for synaptic events including neurotransmitter synthesis, vesicle recycling, and most importantly, the postsynaptic activities leading to channel activation and rebalancing of ionic gradients. Bioenergy homeostasis is coupled with synaptic function via activities of the sodium pumps, glutamate transporters, glucose transport, and mitochondria translocation. Energy insufficiency is sensed by the AMP-activated protein kinase (AMPK), a master metabolic regulator that stimulates the catalytic process to enhance energy production. A decline in energy supply and a disruption in bioenergy homeostasis play a critical role in multiple neuropathological conditions including ischemia, stroke, and neurodegenerative diseases including Alzheimer’s disease and traumatic brain injuries.
Collapse
Affiliation(s)
- Natasha Khatri
- Department of Biology, Boston University , Boston, MA , USA ; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine , Boston, MA , USA
| | - Heng-Ye Man
- Department of Biology, Boston University , Boston, MA , USA ; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine , Boston, MA , USA
| |
Collapse
|
43
|
Abstract
Glutamate in neurons is an important excitatory neurotransmitter, but it also is a key metabolite. We investigated how glutamate in a neural tissue is protected from catabolism. Flux analysis using (13)C-labeled fuels revealed that retinas use activities of the malate aspartate shuttle to protect >98% of their glutamate from oxidation in mitochondria. Isolation of glutamate from the oxidative pathway relies on cytosolic NADH/NAD(+), which is influenced by extracellular glucose, lactate, and pyruvate.
Collapse
|
44
|
Reichenbach A, Bringmann A. New functions of Müller cells. Glia 2013; 61:651-78. [PMID: 23440929 DOI: 10.1002/glia.22477] [Citation(s) in RCA: 475] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 01/10/2012] [Indexed: 12/12/2022]
Abstract
Müller cells, the major type of glial cells in the retina, are responsible for the homeostatic and metabolic support of retinal neurons. By mediating transcellular ion, water, and bicarbonate transport, Müller cells control the composition of the extracellular space fluid. Müller cells provide trophic and anti-oxidative support of photoreceptors and neurons and regulate the tightness of the blood-retinal barrier. By the uptake of glutamate, Müller cells are more directly involved in the regulation of the synaptic activity in the inner retina. This review gives a survey of recently discoved new functions of Müller cells. Müller cells are living optical fibers that guide light through the inner retinal tissue. Thereby they enhance the signal/noise ratio by minimizing intraretinal light scattering and conserve the spatial distribution of light patterns in the propagating image. Müller cells act as soft, compliant embedding for neurons, protecting them in case of mechanical trauma, and also as soft substrate required for neurite growth and neuronal plasticity. Müller cells release neuroactive signaling molecules which modulate neuronal activity, are implicated in the mediation of neurovascular coupling, and mediate the homeostasis of the extracellular space volume under hypoosmotic conditions which are a characteristic of intense neuronal activity. Under pathological conditions, a subset of Müller cells may differentiate to neural progenitor/stem cells which regenerate lost photoreceptors and neurons. Increasing knowledge of Müller cell function and responses in the normal and diseased retina will have great impact for the development of new therapeutic approaches for retinal diseases.
Collapse
Affiliation(s)
- Andreas Reichenbach
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany.
| | | |
Collapse
|
45
|
Firl A, Sack GS, Newman ZL, Tani H, Feller MB. Extrasynaptic glutamate and inhibitory neurotransmission modulate ganglion cell participation during glutamatergic retinal waves. J Neurophysiol 2013; 109:1969-78. [PMID: 23343894 DOI: 10.1152/jn.00039.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
During the first 2 wk of mouse postnatal development, transient retinal circuits give rise to the spontaneous initiation and lateral propagation of depolarizations across the ganglion cell layer (GCL). Glutamatergic retinal waves occur during the second postnatal week, when GCL depolarizations are mediated by ionotropic glutamate receptors. Bipolar cells are the primary source of glutamate in the inner retina, indicating that the propagation of waves depends on their activation. Using the fluorescence resonance energy transfer-based optical sensor of glutamate FLII81E-1μ, we found that retinal waves are accompanied by a large transient increase in extrasynaptic glutamate throughout the inner plexiform layer. Using two-photon Ca(2+) imaging to record spontaneous Ca(2+) transients in large populations of cells, we found that despite this spatially diffuse source of depolarization, only a subset of neurons in the GCL and inner nuclear layer (INL) are robustly depolarized during retinal waves. Application of the glutamate transporter blocker dl-threo-β-benzyloxyaspartate (25 μM) led to a significant increase in cell participation in both layers, indicating that the concentration of extrasynaptic glutamate affects cell participation in both the INL and GCL. In contrast, blocking inhibitory transmission with the GABAA receptor antagonist gabazine and the glycine receptor antagonist strychnine increased cell participation in the GCL without significantly affecting the INL. These data indicate that during development, glutamate spillover provides a spatially diffuse source of depolarization, but that inhibitory circuits dictate which neurons within the GCL participate in retinal waves.
Collapse
Affiliation(s)
- Alana Firl
- Vision Sciences Graduate Program, Department of Optometry, University of California, Berkeley, CA, USA
| | | | | | | | | |
Collapse
|
46
|
Inactivation of the microRNA-183/96/182 cluster results in syndromic retinal degeneration. Proc Natl Acad Sci U S A 2013; 110:E507-16. [PMID: 23341629 DOI: 10.1073/pnas.1212655110] [Citation(s) in RCA: 148] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The microRNA-183/96/182 cluster is highly expressed in the retina and other sensory organs. To uncover its in vivo functions in the retina, we generated a knockout mouse model, designated "miR-183C(GT/GT)," using a gene-trap embryonic stem cell clone. We provide evidence that inactivation of the cluster results in early-onset and progressive synaptic defects of the photoreceptors, leading to abnormalities of scotopic and photopic electroretinograms with decreased b-wave amplitude as the primary defect and progressive retinal degeneration. In addition, inactivation of the miR-183/96/182 cluster resulted in global changes in retinal gene expression, with enrichment of genes important for synaptogenesis, synaptic transmission, photoreceptor morphogenesis, and phototransduction, suggesting that the miR-183/96/182 cluster plays important roles in postnatal functional differentiation and synaptic connectivity of photoreceptors.
Collapse
|
47
|
Bringmann A, Grosche A, Pannicke T, Reichenbach A. GABA and Glutamate Uptake and Metabolism in Retinal Glial (Müller) Cells. Front Endocrinol (Lausanne) 2013; 4:48. [PMID: 23616782 PMCID: PMC3627989 DOI: 10.3389/fendo.2013.00048] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 04/04/2013] [Indexed: 12/11/2022] Open
Abstract
Müller cells, the principal glial cells of the retina, support the synaptic activity by the uptake and metabolization of extracellular neurotransmitters. Müller cells express uptake and exchange systems for various neurotransmitters including glutamate and γ-aminobutyric acid (GABA). Müller cells remove the bulk of extracellular glutamate in the inner retina and contribute to the glutamate clearance around photoreceptor terminals. By the uptake of glutamate, Müller cells are involved in the shaping and termination of the synaptic activity, particularly in the inner retina. Reactive Müller cells are neuroprotective, e.g., by the clearance of excess extracellular glutamate, but may also contribute to neuronal degeneration by a malfunctioning or even reversal of glial glutamate transporters, or by a downregulation of the key enzyme, glutamine synthetase. This review summarizes the present knowledge about the role of Müller cells in the clearance and metabolization of extracellular glutamate and GABA. Some major pathways of GABA and glutamate metabolism in Müller cells are described; these pathways are involved in the glutamate-glutamine cycle of the retina, in the defense against oxidative stress via the production of glutathione, and in the production of substrates for the neuronal energy metabolism.
Collapse
Affiliation(s)
- Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, Faculty of Medicine, University of LeipzigLeipzig, Germany
| | - Antje Grosche
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of LeipzigLeipzig, Germany
| | - Thomas Pannicke
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of LeipzigLeipzig, Germany
| | - Andreas Reichenbach
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of LeipzigLeipzig, Germany
- *Correspondence: Andreas Reichenbach, Paul Flechsig Institute of Brain Research, University of Leipzig, Jahnallee 59, D-04109 Leipzig, Germany. e-mail:
| |
Collapse
|
48
|
Reichenbach A, Bringmann A. Cell Biology of the Müller Cell. Retina 2013. [DOI: 10.1016/b978-1-4557-0737-9.00017-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
49
|
Abstract
Photoreceptors are exquisitely adapted to transform light stimuli into electrical signals that modulate neurotransmitter release. These cells are organized into several compartments including the unique outer segment (OS). Its whole function is to absorb light and transduce this signal into a change of membrane potential. Another compartment is the inner segment where much of metabolism and regulation of membrane potential takes place and that connects the OS and synapse. The synapse is the compartment where changes in membrane potentials are relayed to other neurons in the retina via release of neurotransmitter. The composition of the plasma membrane surrounding these compartments varies to accommodate their specific functions. In this chapter, we discuss the organization of the plasma membrane emphasizing the protein composition of each region as it relates to visual signaling. We also point out examples where mutations in these proteins cause visual impairment.
Collapse
Affiliation(s)
- Sheila A Baker
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.
| | | |
Collapse
|
50
|
Ablation of retinal horizontal cells from adult mice leads to rod degeneration and remodeling in the outer retina. J Neurosci 2012; 32:10713-24. [PMID: 22855819 DOI: 10.1523/jneurosci.0442-12.2012] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In the brain, including the retina, interneurons show an enormous structural and functional diversity. Retinal horizontal cells represent a class of interneurons that form triad synapses with photoreceptors and ON bipolar cells. At this first retinal synapse, horizontal cells modulate signal transmission from photoreceptors to bipolar cells by feedback and feedforward inhibition. To test how the fully developed retina reacts to the specific loss of horizontal cells, these interneurons were specifically ablated from adult mice using the diphtheria toxin (DT)/DT-receptor system and the connexin57 promoter. Following ablation, the retinal network responded with extensive remodeling: rods retracted their axons from the outer plexiform layer and partially degenerated, whereas cones survived. Cone pedicles remained in the outer plexiform layer and preserved synaptic contacts with OFF but not with ON bipolar cells. Consistently, the retinal ON pathway was impaired, leading to reduced amplitudes and prolonged latencies in electroretinograms. However, ganglion cell responses showed only slight changes in time course, presumably because ON bipolar cells formed multiple ectopic synapses with photoreceptors, and visual performance, assessed with an optomotor system, was only mildly affected. Thus, the loss of an entire interneuron class can be largely compensated even by the adult retinal network.
Collapse
|