1
|
Gonschorek D, Goldin MA, Oesterle J, Schwerd-Kleine T, Arlinghaus R, Zhao Z, Schubert T, Marre O, Euler T. Nitric oxide modulates contrast suppression in a subset of mouse retinal ganglion cells. eLife 2025; 13:RP98742. [PMID: 39783858 PMCID: PMC11717361 DOI: 10.7554/elife.98742] [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] [Indexed: 01/12/2025] Open
Abstract
Neuromodulators have major influences on the regulation of neural circuit activity across the nervous system. Nitric oxide (NO) has been shown to be a prominent neuromodulator in many circuits and has been extensively studied in the retina. Here, it has been associated with the regulation of light adaptation, gain control, and gap junctional coupling, but its effect on the retinal output, specifically on the different types of retinal ganglion cells (RGCs), is still poorly understood. In this study, we used two-photon Ca2+ imaging and multi-electrode array (MEA) recordings to measure light-evoked activity of RGCs in the ganglion cell layer in the ex vivo mouse retina. This approach allowed us to investigate the neuromodulatory effects of NO on a cell type-level. Our findings reveal that NO selectively modulates the suppression of temporal responses in a distinct subset of contrast-suppressed RGC types, increasing their activity without altering the spatial properties of their receptive fields. Given that under photopic conditions, NO release is triggered by quick changes in light levels, we propose that these RGC types signal fast contrast changes to higher visual regions. Remarkably, we found that about one-third of the RGC types, recorded using two-photon Ca2+ imaging, exhibited consistent, cell type-specific adaptational response changes throughout an experiment, independent of NO. By employing a sequential-recording paradigm, we could disentangle those additional adaptational response changes from drug-induced modulations. Taken together, our research highlights the selective neuromodulatory effects of NO on RGCs and emphasizes the need of considering non-pharmacological activity changes, like adaptation, in such study designs.
Collapse
Affiliation(s)
- Dominic Gonschorek
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- GRK 2381 ’cGMP: From Bedside to Bench’, University of TübingenTübingenGermany
| | - Matías A Goldin
- Institut de la Vision, Sorbonne Université, INSERM, CNRSParisFrance
| | - Jonathan Oesterle
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- Hertie Institute for AI in Brain Health, Tübingen AI Center, University of TübingenTübingenGermany
| | - Tom Schwerd-Kleine
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- GRK 2381 ’cGMP: From Bedside to Bench’, University of TübingenTübingenGermany
| | - Ryan Arlinghaus
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
| | - Zhijian Zhao
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
| | - Timm Schubert
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
| | - Olivier Marre
- Institut de la Vision, Sorbonne Université, INSERM, CNRSParisFrance
| | - Thomas Euler
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- GRK 2381 ’cGMP: From Bedside to Bench’, University of TübingenTübingenGermany
- Bernstein Center for Computational Neuroscience, University of TübingenTübingenGermany
| |
Collapse
|
2
|
Du Y, Pang M, Chen H, Zhou X, Geng R, Zhang Y, Yang L, Li J, Han Y, Liu J, Zhang R, Bi H, Guo D. Inhibitory effect of Zhujing Pill on myopia progression: Mechanistic insights based on metabonomics and network pharmacology. PLoS One 2024; 19:e0312379. [PMID: 39625993 PMCID: PMC11614212 DOI: 10.1371/journal.pone.0312379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 10/02/2024] [Indexed: 12/06/2024] Open
Abstract
OBJECTIVES This study endeavored to uncover the mechanisms by which Zhujing pill (ZJP) slows myopia progression. METHODS We employed biometric analyses to track diopter and axial length changes in guinea pigs with negative lens-induced myopia (LIM). Through integrating metabonomics and network pharmacology, we aimed to predict the anti-myopic targets and active ingredients of ZJP. Subsequent analysis, including real-time fluorescent quantitative PCR (qPCR) and Western blotting (WB), assessed the expression levels of CHRNA7, LPCAT1, and NOS2 in retinal tissues. KEY FINDINGS Our findings demonstrate that ZJP significantly mitigates diopter increase and axial elongation in LIM guinea pigs. Metabonomic analysis revealed significant changes in 13 serum metabolites, with ZJP reversing the expression of 5 key metabolites. By integrating metabonomics with network pharmacology, we identified core targets of ZJP against myopia and constructed a compound-gene-disease-metabolite network. The expressions of LPCAT1 and CHRNA7 were found to decrease in the LIM group but increase with ZJP treatment, whereas NOS2 expression showed the opposite pattern. CONCLUSIONS This investigation provides the first evidence of ZJP's multifaceted effectiveness in managing myopia, highlighting its impact on multiple components, targets, and pathways, including the novel involvement of LPCAT1 and CHRNA7 in myopia pathogenesis.
Collapse
Affiliation(s)
- Yongle Du
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengran Pang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haoyu Chen
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiangkun Zhou
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruyue Geng
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yanan Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Linqi Yang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiawen Li
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yufeng Han
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinpeng Liu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruixue Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hongsheng Bi
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, China
| | - Dadong Guo
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, China
- Shandong Academy of Eye Disease Prevention and Therapy, Jinan, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
3
|
Biswas S, Busoy JMF, Barathi VA, Muralidharan AR, Schmetterer L, Kathrani BK, Brennan NA, Najjar RP. Interactions Between High-Intensity Light and Unrestricted Vision in the Drive for Hyperopia. Invest Ophthalmol Vis Sci 2024; 65:22. [PMID: 39656469 PMCID: PMC11645742 DOI: 10.1167/iovs.65.14.22] [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] [Received: 06/13/2024] [Accepted: 11/13/2024] [Indexed: 12/16/2024] Open
Abstract
Purpose To evaluate the impact of optical vs. illuminance factors and their duration-dependency on lens-induced hyperopia (LIH) in chick eyes. Methods Hyperopia was induced in one eye in chicks (10 groups; n = 126) from day 1 after hatching until day 8 using +10-diopter lenses with fellow eyes as controls. One group (LIH) served as the control without any interventions. The remaining groups were exposed to 2, 4, or 6 hours of unrestricted vision (UnV), high-intensity light (HL; 15,000 lux), or both (HL + UnV). Ocular axial length (AL), refractive error, and choroidal thickness were measured on days 1, 4, and 8. Inter-ocular difference (IOD = experimental - contralateral control eye) ± SEM was used to express outcome measures. Results By day 8, LIH decreased AL (-0.42 ± 0.03 mm) and produced hyperopic refraction (+3.48 ± 0.32 diopters) and choroidal thickening (+85.81 ± 35.23 µm) in the LIH group (all P < 0.001). Exposure to UnV reduced LIH (i.e., hyperopic refraction, axial shortening, and choroidal thickening) in a duration-dependent manner, whereas HL potentiated the development of LIH in a duration-dependent manner. When combined, UnV overpowered HL, with resultant impact on refraction and AL being close to UnV alone, except at 6 hours, when HL + UnV induced shorter AL compared with UnV alone (P = 0.03). Conclusions Daily exposure to HL, UnV, and HL + UnV altered LIH in a duration-dependent manner with UnV and LIH producing competing signals. The signal generated by UnV was generally stronger than HL in combined exposure, yet longer durations of HL affected the drive for emmetropization in eyes with UnV.
Collapse
Affiliation(s)
- Sayantan Biswas
- Singapore Eye Research Institute, Singapore, Singapore
- School of Optometry, College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | | | - Veluchamy A. Barathi
- Singapore Eye Research Institute, Singapore, Singapore
- Ophthalmology and Visual Science Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Arumugam R. Muralidharan
- Singapore Eye Research Institute, Singapore, Singapore
- Ophthalmology and Visual Science Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Leopold Schmetterer
- Singapore Eye Research Institute, Singapore, Singapore
- Nanyang Technological University, Singapore, Singapore
| | | | | | - Raymond P. Najjar
- Singapore Eye Research Institute, Singapore, Singapore
- Ophthalmology and Visual Science Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
- Eye N' Brain Research Group, Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| |
Collapse
|
4
|
Özçete ÖD, Banerjee A, Kaeser PS. Mechanisms of neuromodulatory volume transmission. Mol Psychiatry 2024; 29:3680-3693. [PMID: 38789677 PMCID: PMC11540752 DOI: 10.1038/s41380-024-02608-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024]
Abstract
A wealth of neuromodulatory transmitters regulate synaptic circuits in the brain. Their mode of signaling, often called volume transmission, differs from classical synaptic transmission in important ways. In synaptic transmission, vesicles rapidly fuse in response to action potentials and release their transmitter content. The transmitters are then sensed by nearby receptors on select target cells with minimal delay. Signal transmission is restricted to synaptic contacts and typically occurs within ~1 ms. Volume transmission doesn't rely on synaptic contact sites and is the main mode of monoamines and neuropeptides, important neuromodulators in the brain. It is less precise than synaptic transmission, and the underlying molecular mechanisms and spatiotemporal scales are often not well understood. Here, we review literature on mechanisms of volume transmission and raise scientific questions that should be addressed in the years ahead. We define five domains by which volume transmission systems can differ from synaptic transmission and from one another. These domains are (1) innervation patterns and firing properties, (2) transmitter synthesis and loading into different types of vesicles, (3) architecture and distribution of release sites, (4) transmitter diffusion, degradation, and reuptake, and (5) receptor types and their positioning on target cells. We discuss these five domains for dopamine, a well-studied monoamine, and then compare the literature on dopamine with that on norepinephrine and serotonin. We include assessments of neuropeptide signaling and of central acetylcholine transmission. Through this review, we provide a molecular and cellular framework for volume transmission. This mechanistic knowledge is essential to define how neuromodulatory systems control behavior in health and disease and to understand how they are modulated by medical treatments and by drugs of abuse.
Collapse
Affiliation(s)
- Özge D Özçete
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Aditi Banerjee
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
5
|
Ling Y, Wang Y, Ye J, Luan C, Bi A, Gu Y, Shi X. Changes in Intrinsically Photosensitive Retinal Ganglion Cells, Dopaminergic Amacrine Cells, and Their Connectivity in the Retinas of Lid Suture Myopia. Invest Ophthalmol Vis Sci 2024; 65:8. [PMID: 39230992 PMCID: PMC11379095 DOI: 10.1167/iovs.65.11.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024] Open
Abstract
Purpose This study investigates alterations in intrinsically photosensitive retinal ganglion cells (ipRGCs) and dopaminergic amacrine cells (DACs) in lid suture myopia (LSM) rats. Methods LSM was induced in rats by suturing the right eyes for 4 weeks. Double immunofluorescence staining of ipRGCs and DACs in whole-mount retinas was performed to analyze changes in the density and morphology of control, LSM, and fellow eyes. Real-time quantitative PCR and Western blotting were used to detect related genes and protein expression levels. Results Significant myopia was induced in the lid-sutured eye, but the fellow eye was not different to control. Decreased ipRGC density with paradoxically increased overall melanopsin expression and enlarged dendritic beads was observed in both the LSM and fellow eyes of the LSM rat retinas. In contrast, DAC changes occurred only in the LSM eyes, with reduced DAC density and tyrosine hydroxylase (TH) expression, sparser dendritic processes, and fewer varicosities. Interestingly, contacts between ipRGCs and DACs in the inner plexiform layer (IPL) and the expression of pituitary adenylate cyclase-activating polypeptide (PACAP) and vesicular monoamine transporter protein 2 (VMAT2) mRNA were decreased in the LSM eyes. Conclusions The ipRGCs and DACs in LSM rat retinas undergo multiple alterations in density, morphology, and related molecule expressions. However, the ipRGC changes alone appear not to be required for the development of myopia, given that myopia is only induced in the lid-sutured eye, and they are unlikely alone to drive the DAC changes. Reduced contacts between ipRGCs and DACs in the LSM eyes may be the structural foundation for the impaired signaling between them. PACAP and VMAT2, strongly associated with ipRGCs and DACs, may play important roles in LSM through complex mechanisms.
Collapse
Affiliation(s)
- Ying Ling
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yao Wang
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Jingjing Ye
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changlin Luan
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Ailing Bi
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Gu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xuefeng Shi
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Institute of Ophthalmology, Nankai University, Tianjin, China
| |
Collapse
|
6
|
Bergum N, Berezin CT, Vigh J. Dopamine enhances GABA A receptor-mediated current amplitude in a subset of intrinsically photosensitive retinal ganglion cells. J Neurophysiol 2024; 132:501-513. [PMID: 38958282 PMCID: PMC11427049 DOI: 10.1152/jn.00457.2023] [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: 12/11/2023] [Revised: 06/05/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
Neuromodulation in the retina is crucial for effective processing of retinal signal at different levels of illuminance. Intrinsically photosensitive retinal ganglion cells (ipRGCs), the neurons that drive nonimage-forming visual functions, express a variety of neuromodulatory receptors that tune intrinsic excitability as well as synaptic inputs. Past research has examined actions of neuromodulators on light responsiveness of ipRGCs, but less is known about how neuromodulation affects synaptic currents in ipRGCs. To better understand how neuromodulators affect synaptic processing in ipRGC, we examine actions of opioid and dopamine agonists have on inhibitory synaptic currents in ipRGCs. Although µ-opioid receptor (MOR) activation had no effect on γ-aminobutyric acid (GABA) currents, dopamine [via the D1-type dopamine receptor (D1R)]) amplified GABAergic currents in a subset of ipRGCs. Furthermore, this D1R-mediated facilitation of the GABA conductance in ipRGCs was mediated by a cAMP/PKA-dependent mechanism. Taken together, these findings reinforce the idea that dopamine's modulatory role in retinal adaptation affects both nonimage-forming and image-forming visual functions.NEW & NOTEWORTHY Neuromodulators such as dopamine are important regulators of retinal function. Here, we demonstrate that dopamine increases inhibitory inputs to intrinsically photosensitive retinal ganglion cells (ipRGCs), in addition to its previously established effect on intrinsic light responsiveness. This indicates that dopamine, in addition to its ability to intrinsically modulate ipRGC activity, can also affect synaptic inputs to ipRGCs, thereby tuning retina circuits involved in nonimage-forming visual functions.
Collapse
Affiliation(s)
- Nikolas Bergum
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
| | - Casey-Tyler Berezin
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins,Colorado, United States
| | - Jozsef Vigh
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins,Colorado, United States
| |
Collapse
|
7
|
Patel JC, Sherpa AD, Melani R, Witkovsky P, Wiseman MR, O'Neill B, Aoki C, Tritsch NX, Rice ME. GABA co-released from striatal dopamine axons dampens phasic dopamine release through autoregulatory GABA A receptors. Cell Rep 2024; 43:113834. [PMID: 38431842 PMCID: PMC11089423 DOI: 10.1016/j.celrep.2024.113834] [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: 10/05/2022] [Revised: 11/29/2023] [Accepted: 02/05/2024] [Indexed: 03/05/2024] Open
Abstract
Striatal dopamine axons co-release dopamine and gamma-aminobutyric acid (GABA), using GABA provided by uptake via GABA transporter-1 (GAT1). Functions of GABA co-release are poorly understood. We asked whether co-released GABA autoinhibits dopamine release via axonal GABA type A receptors (GABAARs), complementing established inhibition by dopamine acting at axonal D2 autoreceptors. We show that dopamine axons express α3-GABAAR subunits in mouse striatum. Enhanced dopamine release evoked by single-pulse optical stimulation in striatal slices with GABAAR antagonism confirms that an endogenous GABA tone limits dopamine release. Strikingly, an additional inhibitory component is seen when multiple pulses are used to mimic phasic axonal activity, revealing the role of GABAAR-mediated autoinhibition of dopamine release. This autoregulation is lost in conditional GAT1-knockout mice lacking GABA co-release. Given the faster kinetics of ionotropic GABAARs than G-protein-coupled D2 autoreceptors, our data reveal a mechanism whereby co-released GABA acts as a first responder to dampen phasic-to-tonic dopamine signaling.
Collapse
Affiliation(s)
- Jyoti C Patel
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| | - Ang D Sherpa
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA; Center for Neural Science New York University, 4 Washington Place, New York, NY 10003, USA
| | - Riccardo Melani
- NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Paul Witkovsky
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Madeline R Wiseman
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Brian O'Neill
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Chiye Aoki
- NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA; Center for Neural Science New York University, 4 Washington Place, New York, NY 10003, USA
| | - Nicolas X Tritsch
- NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Margaret E Rice
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA; NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
8
|
Lyons-Warren AM, Tantry EK, Moss EH, Kochukov MY, Belfort BDW, Ortiz-Guzman J, Freyberg Z, Arenkiel BR. Co-transmitting interneurons in the mouse olfactory bulb regulate olfactory detection and discrimination. Cell Rep 2023; 42:113471. [PMID: 37980561 PMCID: PMC10872518 DOI: 10.1016/j.celrep.2023.113471] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 10/20/2023] [Accepted: 11/03/2023] [Indexed: 11/21/2023] Open
Abstract
Co-transmission of multiple neurotransmitters from a single neuron increases the complexity of signaling information within defined neuronal circuits. Superficial short-axon cells in the olfactory bulb release both dopamine and γ-aminobutyric acid (GABA), yet the specific targets of these neurotransmitters and their respective roles in olfaction have remained unknown. Here, we implement intersectional genetics in mice to selectively block GABA or dopamine release from superficial short-axon cells to identify their distinct cellular targets, impact on circuit function, and behavioral contribution of each neurotransmitter toward olfactory behaviors. We provide functional and anatomical evidence for divergent superficial short-axon cell signaling onto downstream neurons to shape patterns of mitral cell firing that contribute to olfactory-related behaviors.
Collapse
Affiliation(s)
- Ariel M Lyons-Warren
- Department of Pediatrics, Section of Pediatric Neurology and Developmental Neuroscience, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, USA
| | - Evelyne K Tantry
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, USA
| | - Elizabeth H Moss
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, USA
| | - Mikhail Y Kochukov
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, USA
| | - Benjamin D W Belfort
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Joshua Ortiz-Guzman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, USA.
| |
Collapse
|
9
|
Bergum N, Berezin CT, Vigh J. Dopamine enhances GABA A receptor-mediated current amplitude in a subset of intrinsically photosensitive retinal ganglion cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.11.571141. [PMID: 38168350 PMCID: PMC10760026 DOI: 10.1101/2023.12.11.571141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Neuromodulation in the retina is crucial for effective processing of retinal signal at different levels of illuminance. Intrinsically photosensitive retinal ganglion cells (ipRGCs), the neurons that drive non-image forming visual functions, express a variety of neuromodulatory receptors that tune intrinsic excitability as well as synaptic inputs. Past research has examined actions of neuromodulators on light responsiveness of ipRGCs, but less is known about how neuromodulation affects synaptic currents in ipRGCs. To better understand how neuromodulators affect synaptic processing in ipRGC, we examine actions of opioid and dopamine agonists have on inhibitory synaptic currents in ipRGCs. Although μ-opioid receptor (MOR) activation had no effect on γ-aminobutyric acid (GABA) currents, dopamine (via the D1R) amplified GABAergic currents in a subset of ipRGCs. Furthermore, this D1R-mediated facilitation of the GABA conductance in ipRGCs was mediated by a cAMP/PKA-dependent mechanism. Taken together, these findings reinforce the idea that dopamine's modulatory role in retinal adaptation affects both non-image forming as well as image forming visual functions.
Collapse
Affiliation(s)
- Nikolas Bergum
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Casey-Tyler Berezin
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, USA
| | - Jozsef Vigh
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
10
|
Wang F, Zhong W, Yang Q, Zhao W, Liu X, Rao B, Lin X, Zhang J. Distribution and synaptic organization of substance P-like immunoreactive neurons in the mouse retina. Brain Struct Funct 2023; 228:1703-1724. [PMID: 37481742 DOI: 10.1007/s00429-023-02688-x] [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: 01/17/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
Substance P (SP), a neuroprotective peptidergic neurotransmitter, is known to have immunoreactivity (IR) localized to amacrine and/or ganglion cells in a variety of species' retinas, but it has not yet been studied in the mouse retina. Thus, we investigated the distribution and synaptic organization of SP-IR by confocal and electron microscopy immunocytochemistry in the mouse retina. SP-IR was distributed in the inner nuclear layer (INL), inner plexiform layer (IPL), and ganglion cell layer (GCL). Most of the SP-IR somas belonged to amacrine cells (2.5% of all) in the INL and their processes stratified into the S1, S3, and S5 layers of the IPL, with the most intense band in the S5 layer. Some SP-IR somas can also be observed in the GCL, which were identified as displaced amacrine cells (82%, 1269/1550) and ganglion cells (18%, 281/1550) by antibodies against AP2α and RBPMS, respectively. Such SP-IR ganglion cells (1.2% of all RGCs) can be further divided into 3 subgroups expressing SP/α-Synuclein (α-Syn), SP/GAD67, and/or SP/GAD67/α-Syn. Possible physiological and pathological roles of these ganglion cells are discussed. Further, electron microscopy evidence demonstrates that SP-IR amacrine cells receive major inputs from other SP-IR amacrine cell processes (146/242 inputs) and output mostly to SP-negative amacrine cell processes (291/673 outputs), suggesting series inhibition among amacrine cells. These results reveal for the first time an explicit distribution, novel ganglion cell features, and synaptic organization of SP-IR in the mouse retina, which is important for the future use of mouse models to study the roles of SP in healthy and diseased (including Parkinson's disease) retinal states.
Collapse
Affiliation(s)
- Fenglan Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wenhui Zhong
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Qingwen Yang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wenna Zhao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaoqing Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Bilin Rao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Xin Lin
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Jun Zhang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
11
|
Beltrán-Matas P, Hartveit E, Veruki ML. Functional properties of GABA A receptors of AII amacrine cells of the rat retina. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1134765. [PMID: 38983040 PMCID: PMC11182327 DOI: 10.3389/fopht.2023.1134765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/08/2023] [Indexed: 07/11/2024]
Abstract
Amacrine cells are a highly diverse group of inhibitory retinal interneurons that sculpt the responses of bipolar cells, ganglion cells, and other amacrine cells. They integrate excitatory inputs from bipolar cells and inhibitory inputs from other amacrine cells, but for most amacrine cells, little is known about the specificity and functional properties of their inhibitory inputs. Here, we have investigated GABAA receptors of the AII amacrine, a critical neuron in the rod pathway microcircuit, using patch-clamp recording in rat retinal slices. Puffer application of GABA evoked robust responses, but, surprisingly, spontaneous GABAA receptor-mediated postsynaptic currents were not observed, neither under control conditions nor following application of high-K+ solution to facilitate release. To investigate the biophysical and pharmacological properties of GABAA receptors in AIIs, we therefore used nucleated patches and a fast application system. Both brief and long pulses of GABA (3 mM) evoked GABAA receptor-mediated currents with slow, multi-exponential decay kinetics. The average weighted time constant (τw) of deactivation was ~163 ms. Desensitization was even slower, with τw ~330 ms. Non-stationary noise analysis of patch responses and directly observed channel gating yielded a single-channel conductance of ~23 pS. Pharmacological investigation suggested the presence of α2 and/or α3 subunits, as well as the γ2 subunit. Such subunit combinations are typical of GABAA receptors with slow kinetics. If synaptic GABAA receptors of AII amacrines have similar functional properties, the slow deactivation and desensitization kinetics will facilitate temporal summation of GABAergic inputs, allowing effective summation and synaptic integration to occur even for relatively low frequencies of inhibitory inputs.
Collapse
Affiliation(s)
| | - Espen Hartveit
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | |
Collapse
|
12
|
Liu H, Schaeffel F, Yang Z, Feldkaemper MP. GABAB Receptor Activation Affects Eye Growth in Chickens with Visually Induced Refractive Errors. Biomolecules 2023; 13:biom13030434. [PMID: 36979369 PMCID: PMC10046083 DOI: 10.3390/biom13030434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 03/02/2023] Open
Abstract
This study aims to explore the role of GABAB receptors in the development of deprivation myopia (DM), lens-induced myopia (LIM) and lens-induced hyperopia (LIH). Chicks were intravitreally injected with 25 µg baclofen (GABABR agonist) in one eye and saline into the fellow eye. Choroidal thickness (ChT) was measured via OCT before and 2, 4, 6, 8, 24 h after injection. ChT decreased strongly at 6 and 8 h after baclofen injection and returned back to baseline level after 24 h. Moreover, chicks were monocularly treated with translucent diffusers, −7D or +7D lenses and randomly assigned to baclofen or saline treatment. DM chicks were injected daily into both eyes, while LIM and LIH chicks were monocularly injected into the lens-wearing eyes, for 4 days. Refractive error, axial length and ChT were measured before and after treatment. Dopamine and its metabolites were analyzed via HPLC. Baclofen significantly reduced the myopic shift and eye growth in DM and LIM eyes. However, it did not change ChT compared to respective saline-injected eyes. On the other hand, baclofen inhibited the hyperopic shift and choroidal thickening in LIH eyes. All the baclofen-injected eyes showed significantly lower vitreal DOPAC content. Since GABA is an inhibitory ubiquitous neurotransmitter, interfering with its signaling affects spatial retinal processing and therefore refractive error development with both diffusers and lenses.
Collapse
Affiliation(s)
- Hong Liu
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
- Aier Institute of Optometry and Vision Science, Aier Eye Hospital Group, Changsha 410000, China
| | - Frank Schaeffel
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
- Myopia Research Group, Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4031 Basel, Switzerland
| | - Zhikuan Yang
- Aier Institute of Optometry and Vision Science, Aier Eye Hospital Group, Changsha 410000, China
- Hunan Province Optometry Engineering and Technology Research Center, Changsha 410000, China
- Hunan Province International Cooperation Base for Optometry Science and Technology, Changsha 410000, China
- Correspondence: (Z.Y.); (M.P.F.)
| | - Marita Pauline Feldkaemper
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
- Correspondence: (Z.Y.); (M.P.F.)
| |
Collapse
|
13
|
Huang W, Xu Q, Liu F, Su J, Xiao D, Tang L, Hao ZZ, Liu R, Xiang K, Bi Y, Miao Z, Liu X, Liu Y, Liu S. Identification of TPBG-Expressing Amacrine Cells in DAT-tdTomato Mouse. Invest Ophthalmol Vis Sci 2022; 63:13. [PMID: 35551574 PMCID: PMC9123489 DOI: 10.1167/iovs.63.5.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/25/2022] [Indexed: 11/29/2022] Open
Abstract
Purpose Neurons are the bricks of the neuronal system and experimental access to certain neuron subtypes will be of great help to decipher neuronal circuits. Here, we identified trophoblast glycoprotein (TPBG)-expressing GABAergic amacrine cells (ACs) that were selectively labeled in DAT-tdTomato transgenic mice. Methods Retina and brain sections were prepared for immunostaining with antibodies against various biomarkers. Patch-sequencing was performed to obtain the transcriptomes of tdTomato-positive cells in DAT-tdTomato mice. Whole-cell recordings were conducted to identify responses to light stimulation. Results Tyrosine hydroxylase immunoreactive cells were colocalized with tdTomato-positive cells in substantia nigra pars compacta, but not in the retina. Transcriptomes collected from tdTomato-positive cells in retinas via Patch-sequencing exhibited the expression of marker genes of ACs (Pax6 and Slc32a1) and marker genes of GABAergic neurons (Gad1, Gad2, and Slc6a1). Immunostaining with antibodies against relevant proteins (GAD67, GAD65, and GABA) also confirmed transcriptomic results. Furthermore, tdTomato-positive cells in retinas selectively expressed Tpbg, a marker gene for distinct clusters molecularly defined, which was proved with TPBG immunoreactivity in fluorescently labeled cells. Finally, tdTomato-positive cells recorded showed ON-OFF responses to light stimulation. Conclusions Ectopic expression occurs in the retina but not in the substantia nigra pars compacta in the DAT-tdTomato mouse, and fluorescently labeled cells in the retina are TPBG-expressing GABAergic ACs. This type of transgenic mice has been proved as an ideal tool to achieve efficient labeling of a distinct subset of ACs that selectively express Tpbg.
Collapse
Affiliation(s)
- Wanjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Qiang Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Feng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jing Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Dongchang Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Lei Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Zhao-Zhe Hao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Ruifeng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Kangjian Xiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yalan Bi
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge CB10 1SD, United Kingdom
| | - Zhichao Miao
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge CB10 1SD, United Kingdom
- Translational Research Institute of Brain and Brain-Like Intelligence and Department of Anesthesiology, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Xialin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
- Research Unit of Ocular Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Sheng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou, China
| |
Collapse
|
14
|
Knoflach F, Bertrand D. Pharmacological modulation of GABA A receptors. Curr Opin Pharmacol 2021; 59:3-10. [PMID: 34020139 DOI: 10.1016/j.coph.2021.04.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 04/13/2021] [Indexed: 10/21/2022]
Abstract
Ligand-gated ion channels are integral membrane proteins that activate through a change in conformation upon transmitter binding and were identified as key players of brain function. GABAA receptors are major inhibitory ligand-gated ion channels of this protein family. They are the target of many therapeutic compounds widely used in the clinic and continue to attract the attention of academic and pharmaceutical laboratories. Advances in the knowledge of the structure of GABAA receptors at the molecular level with unprecedented resolution enabled the determination of the binding sites of many allosteric modulators revealing the nature of their interactions with the receptors. Herein, we review the latest findings on allosteric modulation of GABAA receptors and their relevance to drug discovery.
Collapse
Affiliation(s)
- Frédéric Knoflach
- F. Hoffmann-La Roche Ltd., Neuroscience & Rare Diseases (NRD) Research, Roche Innovation Center Basel, Basel, 4070, Switzerland
| | - Daniel Bertrand
- HiQScreen Sàrl, 6 rte de Compois, Vésenaz, Geneva, 1222, Switzerland.
| |
Collapse
|
15
|
Sawant A, Ebbinghaus BN, Bleckert A, Gamlin C, Yu WQ, Berson D, Rudolph U, Sinha R, Hoon M. Organization and emergence of a mixed GABA-glycine retinal circuit that provides inhibition to mouse ON-sustained alpha retinal ganglion cells. Cell Rep 2021; 34:108858. [PMID: 33730586 PMCID: PMC8030271 DOI: 10.1016/j.celrep.2021.108858] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 12/07/2020] [Accepted: 02/19/2021] [Indexed: 12/04/2022] Open
Abstract
In the retina, amacrine interneurons inhibit retinal ganglion cell (RGC) dendrites to shape retinal output. Amacrine cells typically use either GABA or glycine to exert synaptic inhibition. Here, we combined transgenic tools with immunohistochemistry, electrophysiology, and 3D electron microscopy to determine the composition and organization of inhibitory synapses across the dendritic arbor of a well-characterized RGC type in the mouse retina: the ON-sustained alpha RGC. We find mixed GABA-glycine receptor synapses across this RGC type, unveiling the existence of "mixed" inhibitory synapses in the retinal circuit. Presynaptic amacrine boutons with dual release sites are apposed to ON-sustained alpha RGC postsynapses. We further reveal the sequence of postsynaptic assembly for these mixed synapses: GABA receptors precede glycine receptors, and a lack of early GABA receptor expression impedes the recruitment of glycine receptors. Together our findings uncover the organization and developmental profile of an additional motif of inhibition in the mammalian retina.
Collapse
Affiliation(s)
- Abhilash Sawant
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA; Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Briana N Ebbinghaus
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - Adam Bleckert
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Clare Gamlin
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Wan-Qing Yu
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - David Berson
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Uwe Rudolph
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Champaign, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Raunak Sinha
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA; Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - Mrinalini Hoon
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA; Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
16
|
Auer F, Franco Taveras E, Klein U, Kesenheimer C, Fleischhauer D, Möhrlen F, Frings S. Anoctamin 2-chloride channels reduce simple spike activity and mediate inhibition at elevated calcium concentration in cerebellar Purkinje cells. PLoS One 2021; 16:e0247801. [PMID: 33651839 PMCID: PMC7924762 DOI: 10.1371/journal.pone.0247801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/12/2021] [Indexed: 11/19/2022] Open
Abstract
Modulation of neuronal excitability is a prominent way of shaping the activity of neuronal networks. Recent studies highlight the role of calcium-activated chloride currents in this context, as they can both increase or decrease excitability. The calcium-activated chloride channel Anoctamin 2 (ANO2 alias TMEM16B) has been described in several regions of the mouse brain, including the olivo-cerebellar system. In inferior olivary neurons, ANO2 was proposed to increase excitability by facilitating the generation of high-threshold calcium spikes. An expression of ANO2 in cerebellar Purkinje cells was suggested, but its role in these neurons remains unclear. In the present study, we confirmed the expression of Ano2 mRNA in Purkinje cells and performed electrophysiological recordings to examine the influence of ANO2-chloride channels on the excitability of Purkinje cells by comparing wildtype mice to mice lacking ANO2. Recordings were performed in acute cerebellar slices of adult mice, which provided the possibility to study the role of ANO2 within the cerebellar cortex. Purkinje cells were uncoupled from climbing fiber input to assess specifically the effect of ANO2 channels on Purkinje cell activity. We identified an attenuating effect of ANO2-mediated chloride currents on the instantaneous simple spike activity both during strong current injections and during current injections close to the simple spike threshold. Moreover, we report a reduction of inhibitory currents from GABAergic interneurons upon depolarization, lasting for several seconds. Together with the role of ANO2-chloride channels in inferior olivary neurons, our data extend the evidence for a role of chloride-dependent modulation in the olivo-cerebellar system that might be important for proper cerebellum-dependent motor coordination and learning.
Collapse
Affiliation(s)
- Friederike Auer
- Department of Animal Physiology, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Eliana Franco Taveras
- Department of Animal Physiology, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Uli Klein
- Department of Animal Physiology, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Céline Kesenheimer
- Department of Animal Physiology, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Dana Fleischhauer
- Department of Animal Physiology, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Frank Möhrlen
- Department of Animal Physiology, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Stephan Frings
- Department of Animal Physiology, Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
17
|
Hirano AA, Vuong HE, Kornmann HL, Schietroma C, Stella SL, Barnes S, Brecha NC. Vesicular Release of GABA by Mammalian Horizontal Cells Mediates Inhibitory Output to Photoreceptors. Front Cell Neurosci 2020; 14:600777. [PMID: 33335476 PMCID: PMC7735995 DOI: 10.3389/fncel.2020.600777] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
Feedback inhibition by horizontal cells regulates rod and cone photoreceptor calcium channels that control their release of the neurotransmitter glutamate. This inhibition contributes to synaptic gain control and the formation of the center-surround antagonistic receptive fields passed on to all downstream neurons, which is important for contrast sensitivity and color opponency in vision. In contrast to the plasmalemmal GABA transporter found in non-mammalian horizontal cells, there is evidence that the mechanism by which mammalian horizontal cells inhibit photoreceptors involves the vesicular release of the inhibitory neurotransmitter GABA. Historically, inconsistent findings of GABA and its biosynthetic enzyme, L-glutamate decarboxylase (GAD) in horizontal cells, and the apparent lack of surround response block by GABAergic agents diminished support for GABA's role in feedback inhibition. However, the immunolocalization of the vesicular GABA transporter (VGAT) in the dendritic and axonal endings of horizontal cells that innervate photoreceptor terminals suggested GABA was released via vesicular exocytosis. To test the idea that GABA is released from vesicles, we localized GABA and GAD, multiple SNARE complex proteins, synaptic vesicle proteins, and Cav channels that mediate exocytosis to horizontal cell dendritic tips and axonal terminals. To address the perceived relative paucity of synaptic vesicles in horizontal cell endings, we used conical electron tomography on mouse and guinea pig retinas that revealed small, clear-core vesicles, along with a few clathrin-coated vesicles and endosomes in horizontal cell processes within photoreceptor terminals. Some small-diameter vesicles were adjacent to the plasma membrane and plasma membrane specializations. To assess vesicular release, a functional assay involving incubation of retinal slices in luminal VGAT-C antibodies demonstrated vesicles fused with the membrane in a depolarization- and calcium-dependent manner, and these labeled vesicles can fuse multiple times. Finally, targeted elimination of VGAT in horizontal cells resulted in a loss of tonic, autaptic GABA currents, and of inhibitory feedback modulation of the cone photoreceptor Cai, consistent with the elimination of GABA release from horizontal cell endings. These results in mammalian retina identify the central role of vesicular release of GABA from horizontal cells in the feedback inhibition of photoreceptors.
Collapse
Affiliation(s)
- Arlene A. Hirano
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Helen E. Vuong
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Helen L. Kornmann
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Cataldo Schietroma
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Salvatore L. Stella
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Steven Barnes
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Doheny Eye Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nicholas C. Brecha
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
18
|
Fougère M, van der Zouwen CI, Boutin J, Ryczko D. Heterogeneous expression of dopaminergic markers and Vglut2 in mouse mesodiencephalic dopaminergic nuclei A8-A13. J Comp Neurol 2020; 529:1273-1292. [PMID: 32869307 DOI: 10.1002/cne.25020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 12/17/2022]
Abstract
Co-transmission of glutamate by brain dopaminergic (DA) neurons was recently proposed as a potential factor influencing cell survival in models of Parkinson's disease. Intriguingly, brain DA nuclei are differentially affected in Parkinson's disease. Whether this is associated with different patterns of co-expression of the glutamatergic phenotype along the rostrocaudal brain axis is unknown in mammals. We hypothesized that, as in zebrafish, the glutamatergic phenotype is present preferentially in the caudal mesodiencephalic DA nuclei. Here, we used in mice a cell fate mapping strategy based on reporter protein expression (ZsGreen) consecutive to previous expression of the vesicular glutamate transporter 2 (Vglut2) gene, coupled with immunofluorescence experiments against tyrosine hydroxylase (TH) or dopamine transporter (DAT). We found three expression patterns in DA cells, organized along the rostrocaudal brain axis. The first pattern (TH-positive, DAT-positive, ZsGreen-positive) was found in A8-A10. The second pattern (TH-positive, DAT-negative, ZsGreen-positive) was found in A11. The third pattern (TH-positive, DAT-negative, ZsGreen-negative) was found in A12-A13. These patterns should help to refine the establishment of the homology of DA nuclei between vertebrate species. Our results also uncover that Vglut2 is expressed at some point during cell lifetime in DA nuclei known to degenerate in Parkinson's disease and largely absent from those that are preserved, suggesting that co-expression of the glutamatergic phenotype in DA cells influences their survival in Parkinson's disease.
Collapse
Affiliation(s)
- Maxime Fougère
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de La Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Cornelis Immanuel van der Zouwen
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de La Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Joël Boutin
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de La Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Dimitri Ryczko
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de La Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
- Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada
- Centre d'Excellence en Neurosciences de l'Université de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
19
|
Hellmer CB, Bohl JM, Hall LM, Koehler CC, Ichinose T. Dopaminergic Modulation of Signal Processing in a Subset of Retinal Bipolar Cells. Front Cell Neurosci 2020; 14:253. [PMID: 32922266 PMCID: PMC7456991 DOI: 10.3389/fncel.2020.00253] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/23/2020] [Indexed: 11/13/2022] Open
Abstract
The retina and the olfactory bulb are the gateways to the visual and olfactory systems, respectively, similarly using neural networks to initiate sensory signal processing. Sensory receptors receive signals that are transmitted to neural networks before projecting to primary cortices. These networks filter sensory signals based on their unique features and adjust their sensitivities by gain control systems. Interestingly, dopamine modulates sensory signal transduction in both systems. In the retina, dopamine adjusts the retinal network for daylight conditions (“light adaptation”). In the olfactory system, dopamine mediates lateral inhibition between the glomeruli, resulting in odorant signal decorrelation and discrimination. While dopamine is essential for signal discrimination in the olfactory system, it is not understood whether dopamine has similar roles in visual signal processing in the retina. To elucidate dopaminergic effects on visual processing, we conducted patch-clamp recording from second-order retinal bipolar cells, which exhibit multiple types that can convey different temporal features of light. We recorded excitatory postsynaptic potentials (EPSPs) evoked by various frequencies of sinusoidal light in the absence and presence of a dopamine receptor 1 (D1R) agonist or antagonist. Application of a D1R agonist, SKF-38393, shifted the peak temporal responses toward higher frequencies in a subset of bipolar cells. In contrast, a D1R antagonist, SCH-23390, reversed the effects of SKF on these types of bipolar cells. To examine the mechanism of dopaminergic modulation, we recorded voltage-gated currents, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, and low-voltage activated (LVA) Ca2+ channels. SKF modulated HCN and LVA currents, suggesting that these channels are the target of D1R signaling to modulate visual signaling in these bipolar cells. Taken together, we found that dopamine modulates the temporal tuning of a subset of retinal bipolar cells. Consequently, we determined that dopamine plays a role in visual signal processing, which is similar to its role in signal decorrelation in the olfactory bulb.
Collapse
Affiliation(s)
- Chase B Hellmer
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Jeremy M Bohl
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Leo M Hall
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Christina C Koehler
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Tomomi Ichinose
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
20
|
Cameron MA, Morley JW, Pérez-Fernández V. Seeing the light: different photoreceptor classes work together to drive adaptation in the mammalian retina. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
21
|
Grünert U, Martin PR. Cell types and cell circuits in human and non-human primate retina. Prog Retin Eye Res 2020; 78:100844. [PMID: 32032773 DOI: 10.1016/j.preteyeres.2020.100844] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/28/2020] [Accepted: 01/31/2020] [Indexed: 12/12/2022]
Abstract
This review summarizes our current knowledge of primate including human retina focusing on bipolar, amacrine and ganglion cells and their connectivity. We have two main motivations in writing. Firstly, recent progress in non-invasive imaging methods to study retinal diseases mean that better understanding of the primate retina is becoming an important goal both for basic and for clinical sciences. Secondly, genetically modified mice are increasingly used as animal models for human retinal diseases. Thus, it is important to understand to which extent the retinas of primates and rodents are comparable. We first compare cell populations in primate and rodent retinas, with emphasis on how the fovea (despite its small size) dominates the neural landscape of primate retina. We next summarise what is known, and what is not known, about the postreceptoral neurone populations in primate retina. The inventories of bipolar and ganglion cells in primates are now nearing completion, comprising ~12 types of bipolar cell and at least 17 types of ganglion cell. Primate ganglion cells show clear differences in dendritic field size across the retina, and their morphology differs clearly from that of mouse retinal ganglion cells. Compared to bipolar and ganglion cells, amacrine cells show even higher morphological diversity: they could comprise over 40 types. Many amacrine types appear conserved between primates and mice, but functions of only a few types are understood in any primate or non-primate retina. Amacrine cells appear as the final frontier for retinal research in monkeys and mice alike.
Collapse
Affiliation(s)
- Ulrike Grünert
- The University of Sydney, Save Sight Institute, Faculty of Medicine and Health, Sydney, NSW, 2000, Australia; Australian Research Council Centre of Excellence for Integrative Brain Function, Sydney Node, The University of Sydney, Sydney, NSW, 2000, Australia.
| | - Paul R Martin
- The University of Sydney, Save Sight Institute, Faculty of Medicine and Health, Sydney, NSW, 2000, Australia; Australian Research Council Centre of Excellence for Integrative Brain Function, Sydney Node, The University of Sydney, Sydney, NSW, 2000, Australia
| |
Collapse
|
22
|
Increased endogenous dopamine prevents myopia in mice. Exp Eye Res 2020; 193:107956. [PMID: 32032629 DOI: 10.1016/j.exer.2020.107956] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 01/31/2020] [Accepted: 02/01/2020] [Indexed: 02/01/2023]
Abstract
Experimental evidence suggests that dopamine (DA) modulates refractive eye growth. We evaluated whether increasing endogenous DA activity using pharmacological or genetic approaches decreased myopia susceptibility in mice. First, we assessed the effects of systemic L-3,4-dihydroxyphenylalanine (L-DOPA) injections on form deprivation myopia (FDM) in C57BL/6 J (WTC57) mice. WTC57 mice received daily systemic injections of L-DOPA (n = 11), L-DOPA + ascorbic acid (AA, n = 22), AA (n = 20), or Saline (n = 16). Second, we tested transgenic mice with increased or decreased expression of vesicular monoamine transporter 2 (VMAT2HI, n = 22; WTHI, n = 18; VMAT2LO, n = 18; or WTLO, n = 9) under normal and form deprivation conditions. VMAT2 packages DA into vesicles, affecting DA release. At post-natal day 28 (P28), monocular FD was induced in each genotype. Weekly measurements of refractive error, corneal curvature, and ocular biometry were performed until P42 or P49. WTC57 mice exposed to FD developed a significant myopic shift (treated-contralateral eye) in AA (-3.27 ± 0.73D) or saline (-3.71 ± 0.80D) treated groups that was significantly attenuated by L-DOPA (-0.73 ± 0.90D, p = 0.0002) or L-DOPA + AA (-0.11 ± 0.46D, p = 0.0103). The VMAT2LO mice, with under-expression of VMAT2, were most susceptible to FDM. VMAT2LO mice developed significant myopic shifts to FD after one week compared to VMAT2HI and WT mice (VMAT2LO: -5.48 ± 0.54D; VMAT2HI: -0.52 ± 0.92D, p < 0.05; WT: -2.13 ± 0.78D, p < 0.05; ungoggled control: -0.22 ± 0.24D, p < 0.001). These results indicate that endogenously increasing DA synthesis and release by genetic and pharmacological methods prevents FDM in mice.
Collapse
|
23
|
Caravaggio F, Worhunsky P, Graff-Guerrero A, Matuskey D. Further in vivo characterization of [ 11 C]-(+)-PHNO uptake into a retina-like region of interest in humans. Synapse 2019; 74:e22135. [PMID: 31553807 DOI: 10.1002/syn.22135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 09/20/2019] [Indexed: 11/12/2022]
Abstract
The neurotransmitter dopamine is present in the retina and is involved in several modulatory functions. Unlike in rodents, dopamine D3 receptors are expressed in the retina of humans. Recently, uptake of the D3 receptor-preferring radiotracer [11 C]-(+)-PHNO has been observed in a retina-like region of interest (ROI) in humans. Here, we attempted to quantify [11 C]-(+)-PHNO uptake into this ROI using an independent sample, employing an extended scan acquisition time (120 min) and arterial kinetic modeling. Data from 14 healthy controls were analyzed (Mean Age: 38.41 ± 9.55, 3 female), 8 of which provided arterial line input function data (Mean Age: 41.07 ± 7.82, 3 female). Using Ichise's multilinear analysis (MA1) method, it was possible to quantify the volume of distribution (VT ) of [11 C]-(+)-PHNO in this retina-like region (Mean VT = 13.56 ± 3.52; Mean χ2 = 2.08 ± 2.20). Notably, the shape of the time activity curve resembled closely that of the globus pallidus. Moreover, the VT values in the retina correlated well with binding potential (BPND ) values calculated using the simplified reference tissue model (Mean BPND = 2.11 ± .94; Mean χ2 = 5.76 ± 2.56), employing the cerebellum as the reference region (r = .76, r2 = .58). In summary, we provide evidence that the in vivo uptake of [11 C]-(+)-PHNO into a retina-like ROI in humans can be quantified using both arterial blood sampling (VT ) and simplified reference tissue methods (BPND ).
Collapse
Affiliation(s)
- Fernando Caravaggio
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Patrick Worhunsky
- Department of Psychiatry, Yale University, New Haven, Connecticut, USA
| | - Ariel Graff-Guerrero
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - David Matuskey
- Department of Psychiatry, Yale University, New Haven, Connecticut, USA.,PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut, USA.,Department of Neurology, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
24
|
Van Hook MJ, Nawy S, Thoreson WB. Voltage- and calcium-gated ion channels of neurons in the vertebrate retina. Prog Retin Eye Res 2019; 72:100760. [PMID: 31078724 PMCID: PMC6739185 DOI: 10.1016/j.preteyeres.2019.05.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/25/2019] [Accepted: 05/01/2019] [Indexed: 02/06/2023]
Abstract
In this review, we summarize studies investigating the types and distribution of voltage- and calcium-gated ion channels in the different classes of retinal neurons: rods, cones, horizontal cells, bipolar cells, amacrine cells, interplexiform cells, and ganglion cells. We discuss differences among cell subtypes within these major cell classes, as well as differences among species, and consider how different ion channels shape the responses of different neurons. For example, even though second-order bipolar and horizontal cells do not typically generate fast sodium-dependent action potentials, many of these cells nevertheless possess fast sodium currents that can enhance their kinetic response capabilities. Ca2+ channel activity can also shape response kinetics as well as regulating synaptic release. The L-type Ca2+ channel subtype, CaV1.4, expressed in photoreceptor cells exhibits specific properties matching the particular needs of these cells such as limited inactivation which allows sustained channel activity and maintained synaptic release in darkness. The particular properties of K+ and Cl- channels in different retinal neurons shape resting membrane potentials, response kinetics and spiking behavior. A remaining challenge is to characterize the specific distributions of ion channels in the more than 100 individual cell types that have been identified in the retina and to describe how these particular ion channels sculpt neuronal responses to assist in the processing of visual information by the retina.
Collapse
Affiliation(s)
- Matthew J Van Hook
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Scott Nawy
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA; Department Pharmacology & Experimental Neuroscience(2), University of Nebraska Medical Center, Omaha, NE, USA
| | - Wallace B Thoreson
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA; Department Pharmacology & Experimental Neuroscience(2), University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
25
|
Deng B, Li Q, Liu X, Cao Y, Li B, Qian Y, Xu R, Mao R, Zhou E, Zhang W, Huang J, Rao Y. Chemoconnectomics: Mapping Chemical Transmission in Drosophila. Neuron 2019; 101:876-893.e4. [PMID: 30799021 DOI: 10.1016/j.neuron.2019.01.045] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 11/02/2018] [Accepted: 01/17/2019] [Indexed: 12/27/2022]
Abstract
We define the chemoconnectome (CCT) as the entire set of neurotransmitters, neuromodulators, neuropeptides, and their receptors underlying chemotransmission in an animal. We have generated knockout lines of Drosophila CCT genes for functional investigations and knockin lines containing Gal4 and other tools for examining gene expression and manipulating neuronal activities, with a versatile platform allowing genetic intersections and logic gates. CCT reveals the coexistence of specific transmitters but mutual exclusion of the major inhibitory and excitatory transmitters in the same neurons. One neuropeptide and five receptors were detected in glia, with octopamine β2 receptor functioning in glia. A pilot screen implicated 41 genes in sleep regulation, with the dopamine receptor Dop2R functioning in neurons expressing the peptides Dilp2 and SIFa. Thus, CCT is a novel concept, chemoconnectomics a new approach, and CCT tool lines a powerful resource for systematic investigations of chemical-transmission-mediated neural signaling circuits underlying behavior and cognition.
Collapse
Affiliation(s)
- Bowen Deng
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Qi Li
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Xinxing Liu
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Yue Cao
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Bingfeng Li
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Yongjun Qian
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Rui Xu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Renbo Mao
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Enxing Zhou
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Wenxia Zhang
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Juan Huang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yi Rao
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China.
| |
Collapse
|
26
|
Kim MK, Aung MH, Mees L, Olson DE, Pozdeyev N, Iuvone PM, Thule PM, Pardue MT. Dopamine Deficiency Mediates Early Rod-Driven Inner Retinal Dysfunction in Diabetic Mice. Invest Ophthalmol Vis Sci 2018; 59:572-581. [PMID: 29372256 PMCID: PMC5788047 DOI: 10.1167/iovs.17-22692] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Purpose Electroretinograms (ERGs) are abnormal in diabetic retinas before the appearance of vascular lesions, providing a possible biomarker for diabetic vision loss. Previously, we reported that decreased retinal dopamine (DA) levels in diabetic rodents contributed to early visual and retinal dysfunction. In the current study, we examined whether oscillatory potentials (OPs) could serve as a potential marker for detecting early inner retinal dysfunction due to retinal DA deficiency. Methods Retinal function was tested with dark-adapted ERGs, taken at 3, 4, and 5 weeks after diabetes induction with streptozotocin. Electrical responses were analyzed and correlations were made with previously reported retinal DA levels. The effect of restoring systemic DA levels or removing DA from the retina in diabetic mice on OPs was assessed using L-3,4-dihydroxyphenylalanine (L-DOPA) treatments and retina-specific tyrosine hydroxylase (Th) knockout mice (rTHKO), respectively. Results Diabetic animals had significantly delayed OPs compared to control animals in response to dim, but not bright, flash stimuli. L-DOPA treatment preserved OP implicit time in diabetic mice. Diabetic rTHKO mice had further delayed OPs compared to diabetic mice with normal retinal Th, with L-DOPA treatment also providing benefit. Decreasing retinal DA levels significantly correlated with increasing OP delays mediated by rod pathways. Conclusions Our data suggest that inner retinal dysfunction in early-stage diabetes is mediated by rod-pathway deficits and DA deficiencies. OP delays may be used to determine the earliest functional deficits in diabetic retinopathy and to establish an early treatment window for DA therapies that may prevent progressive vision loss.
Collapse
Affiliation(s)
- Moon K Kim
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Moe H Aung
- Neuroscience, Emory University, Atlanta, Georgia, United States
| | - Lukas Mees
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States.,Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States
| | - Darin E Olson
- Division of Endocrinology, Metabolism and Lipids, Emory University, Atlanta, Georgia, United States.,Medical Service, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States
| | - Nikita Pozdeyev
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States.,Department of Pharmacology, Emory University, Atlanta, Georgia, United States
| | - P Michael Iuvone
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States.,Neuroscience, Emory University, Atlanta, Georgia, United States.,Department of Pharmacology, Emory University, Atlanta, Georgia, United States
| | - Peter M Thule
- Division of Endocrinology, Metabolism and Lipids, Emory University, Atlanta, Georgia, United States.,Medical Service, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States
| | - Machelle T Pardue
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States.,Neuroscience, Emory University, Atlanta, Georgia, United States.,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States.,Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States
| |
Collapse
|
27
|
Hoshi H, Sato F. The morphological characterization of orientation-biased displaced large-field ganglion cells in the central part of goldfish retina. J Comp Neurol 2018; 526:243-261. [PMID: 28921532 DOI: 10.1002/cne.24331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/01/2017] [Accepted: 09/04/2017] [Indexed: 11/10/2022]
Abstract
The vertebrate retina has about 30 subtypes of ganglion cells. Each ganglion cell receives synaptic inputs from specific types of bipolar and amacrine cells ramifying at the same depth of the inner plexiform layer (IPL), each of which is thought to process a specific aspect of visual information. Here, we identified one type of displaced ganglion cell in the goldfish retina which had a large and elongated dendritic field. As a population, all of these ganglion cells were oriented in the horizontal axis and perpendicular to the dorsal-ventral axis of the goldfish eye in the central part of retina. This ganglion cell has previously been classified as Type 1.2. However, the circuit elements which synapse with this ganglion cell are not yet characterized. We found that this displaced ganglion cell was directly tracer-coupled only with homologous ganglion cells at sites containing Cx35/36 puncta. We further illustrated that the processes of dopaminergic neurons often terminated next to intersections between processes of ganglion cells, close to where dopamine D1 receptors were localized. Finally, we showed that Mb1 ON bipolar cells had ribbon synapses in the axonal processes passing through the IPL and made ectopic synapses with this displaced ganglion cell that stratified into stratum 1 of the IPL. These results suggest that the displaced ganglion cell may synapse with both Mb1 cells using ectopic ribbon synapses and OFF cone bipolar cells with regular ribbon synapses in the IPL to function in both scotopic and photopic light conditions.
Collapse
Affiliation(s)
- Hideo Hoshi
- Department of Anatomy, School of Medicine, Toho University, Tokyo, Japan
| | - Fumi Sato
- Department of Anatomy, School of Medicine, Toho University, Tokyo, Japan
| |
Collapse
|
28
|
Antagonistic effect of dopamine structural analogues on human GABAρ1 receptor. Sci Rep 2017; 7:17385. [PMID: 29234054 PMCID: PMC5727059 DOI: 10.1038/s41598-017-17530-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 11/24/2017] [Indexed: 12/12/2022] Open
Abstract
GABAergic and dopaminergic pathways are co-localized in several areas of the central nervous system and recently several reports have shown co-release of both neurotransmitters. The GABA-A receptor (β and ρ1 subunits) is modulated by dopamine (DA) and, interestingly, GABAρ1 can be modulated by several biogenic amines. Here we explored the effects of the metabolites of the dopaminergic pathway and other structural analogues of DA on GABAρ1 and the DA gated ion channel (LGC-53) from Caenorhabditis elegans expressed in Xenopus laevis oocytes. Our findings show an antagonistic effect of the metabolite 3-Methoxytyramine (3-MT, IC50 = 285 ± 30 µM) with similar potency compared to DA on induced GABA currents; however, it was inactive on LGC-53. The structural DA analogues and metabolites, 3, 4-dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA), 2-phenylethylamine (β-PEA) and 4-amino-1-butanol (4-AM-1-OH), antagonized GABAρ1 currents, whereas β-PEA acted as partial agonists on LGC-53, indicating that the putative binding sites of both receptors may share structural characteristics. These results suggest that the DA metabolites 3-MT, DOPAC and HVA modulate GABAρ1 and possibly affect the activity of the receptors that include this subunit in vivo.
Collapse
|
29
|
Affiliation(s)
- Jeffrey S. Diamond
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892-3701
| |
Collapse
|
30
|
Shigemoto R, Joesch M. The genetic encoded toolbox for electron microscopy and connectomics. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2017; 6. [DOI: 10.1002/wdev.288] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/02/2017] [Accepted: 07/05/2017] [Indexed: 11/08/2022]
|
31
|
Granger AJ, Wallace ML, Sabatini BL. Multi-transmitter neurons in the mammalian central nervous system. Curr Opin Neurobiol 2017; 45:85-91. [PMID: 28500992 DOI: 10.1016/j.conb.2017.04.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/17/2017] [Indexed: 01/22/2023]
Abstract
It is firmly established that many mammalian neurons release various combinations of amino acids, their derivatives, and other small molecules from presynaptic terminals in order to signal to their postsynaptic targets. Here we discuss recent findings about four types of multi-transmitter neurons-those that release GABA and acetylcholine (Ach); dopamine (DA) and GABA or glutamate; and glutamate and GABA. The mechanisms of co-release in each class differ and highlight the complex and dynamic nature of neurotransmitter release. Furthermore, identifying the neurotransmitter signature of each neuron and the post-synaptic targets of each neurotransmitter remain challenging. The existence of multi-transmitter neurons complicates the interpretation of connectomic wiring diagrams and poses interesting challenges for our understanding of circuit function in the brain.
Collapse
Affiliation(s)
- Adam J Granger
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, United States
| | - Michael L Wallace
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, United States
| | - Bernardo L Sabatini
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, United States.
| |
Collapse
|
32
|
Circadian Plasticity of Mammalian Inhibitory Interneurons. Neural Plast 2017; 2017:6373412. [PMID: 28367335 PMCID: PMC5358450 DOI: 10.1155/2017/6373412] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 01/15/2017] [Accepted: 02/19/2017] [Indexed: 12/11/2022] Open
Abstract
Inhibitory interneurons participate in all neuronal circuits in the mammalian brain, including the circadian clock system, and are indispensable for their effective function. Although the clock neurons have different molecular and electrical properties, their main function is the generation of circadian oscillations. Here we review the circadian plasticity of GABAergic interneurons in several areas of the mammalian brain, suprachiasmatic nucleus, neocortex, hippocampus, olfactory bulb, cerebellum, striatum, and in the retina.
Collapse
|
33
|
Trakhtenberg EF, Pita-Thomas W, Fernandez SG, Patel KH, Venugopalan P, Shechter JM, Morkin MI, Galvao J, Liu X, Dombrowski SM, Goldberg JL. Serotonin receptor 2C regulates neurite growth and is necessary for normal retinal processing of visual information. Dev Neurobiol 2016; 77:419-437. [PMID: 26999672 DOI: 10.1002/dneu.22391] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 01/25/2016] [Accepted: 03/12/2016] [Indexed: 12/21/2022]
Abstract
Serotonin (5HT) is present in a subpopulation of amacrine cells, which form synapses with retinal ganglion cells (RGCs), but little is known about the physiological role of retinal serotonergic circuitry. We found that the 5HT receptor 2C (5HTR2C) is upregulated in RGCs after birth. Amacrine cells generate 5HT and about half of RGCs respond to 5HTR2C agonism with calcium elevation. We found that there are on average 83 5HT+ amacrine cells randomly distributed across the adult mouse retina, all negative for choline acetyltransferase and 90% positive for tyrosine hydroxylase. We also investigated whether 5HTR2C and 5HTR5A affect RGC neurite growth. We found that both suppress neurite growth, and that RGCs from the 5HTR2C knockout (KO) mice grow longer neurites. Furthermore, 5HTR2C is subject to post-transcriptional editing, and we found that only the edited isoform's suppressive effect on neurite growth could be reversed by a 5HTR2C inverse agonist. Next, we investigated the physiological role of 5HTR2C in the retina, and found that 5HTR2C KO mice showed increased amplitude on pattern electroretinogram. Finally, RGC transcriptional profiling and pathways analysis suggested partial developmental compensation for 5HTR2C absence. Taken together, our findings demonstrate that 5HTR2C regulates neurite growth and RGC activity and is necessary for normal amplitude of RGC response to physiologic stimuli, and raise the hypothesis that these functions are modulated by a subset of 5HT+/ChAT-/TH+ amacrine cells as part of retinal serotonergic circuitry. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 419-437, 2017.
Collapse
Affiliation(s)
- Ephraim F Trakhtenberg
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Department of Neurosurgery, Harvard Medical School, Boston, Massachusetts.,Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Wolfgang Pita-Thomas
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida.,Department of Anatomy and Neurobiology, Washington University, St. Louis, Missouri
| | - Stephanie G Fernandez
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Karan H Patel
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Praseeda Venugopalan
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Department of Neurosurgery, Harvard Medical School, Boston, Massachusetts.,Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Jesse M Shechter
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Melina I Morkin
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Joana Galvao
- Shiley Eye Center, University of California, San Diego, California
| | - Xiongfei Liu
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Susan M Dombrowski
- Genomatix Software, Ann Arbor, Michigan.,Department of Obstetrics & Gynecology, Wayne State University School of Medicine, Detroit, Michigan
| | - Jeffrey L Goldberg
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, Florida.,Shiley Eye Center, University of California, San Diego, California.,Byers Eye Institute, Stanford University, Palo Alto, California
| |
Collapse
|
34
|
Barker DJ, Root DH, Zhang S, Morales M. Multiplexed neurochemical signaling by neurons of the ventral tegmental area. J Chem Neuroanat 2016; 73:33-42. [PMID: 26763116 PMCID: PMC4818729 DOI: 10.1016/j.jchemneu.2015.12.016] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 12/31/2015] [Accepted: 12/31/2015] [Indexed: 12/15/2022]
Abstract
The ventral tegmental area (VTA) is an evolutionarily conserved structure that has roles in reward-seeking, safety-seeking, learning, motivation, and neuropsychiatric disorders such as addiction and depression. The involvement of the VTA in these various behaviors and disorders is paralleled by its diverse signaling mechanisms. Here we review recent advances in our understanding of neuronal diversity in the VTA with a focus on cell phenotypes that participate in 'multiplexed' neurotransmission involving distinct signaling mechanisms. First, we describe the cellular diversity within the VTA, including neurons capable of transmitting dopamine, glutamate or GABA as well as neurons capable of multiplexing combinations of these neurotransmitters. Next, we describe the complex synaptic architecture used by VTA neurons in order to accommodate the transmission of multiple transmitters. We specifically cover recent findings showing that VTA multiplexed neurotransmission may be mediated by either the segregation of dopamine and glutamate into distinct microdomains within a single axon or by the integration of glutamate and GABA into a single axon terminal. In addition, we discuss our current understanding of the functional role that these multiplexed signaling pathways have in the lateral habenula and the nucleus accumbens. Finally, we consider the putative roles of VTA multiplexed neurotransmission in synaptic plasticity and discuss how changes in VTA multiplexed neurons may relate to various psychopathologies including drug addiction and depression.
Collapse
Affiliation(s)
- David J Barker
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Blvd Suite 200, Baltimore, MD 21224, United States
| | - David H Root
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Blvd Suite 200, Baltimore, MD 21224, United States
| | - Shiliang Zhang
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Blvd Suite 200, Baltimore, MD 21224, United States
| | - Marisela Morales
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Blvd Suite 200, Baltimore, MD 21224, United States.
| |
Collapse
|
35
|
Gjorgjieva J, Drion G, Marder E. Computational implications of biophysical diversity and multiple timescales in neurons and synapses for circuit performance. Curr Opin Neurobiol 2016; 37:44-52. [PMID: 26774694 PMCID: PMC4860045 DOI: 10.1016/j.conb.2015.12.008] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/17/2015] [Accepted: 12/22/2015] [Indexed: 12/27/2022]
Abstract
Despite advances in experimental and theoretical neuroscience, we are still trying to identify key biophysical details that are important for characterizing the operation of brain circuits. Biological mechanisms at the level of single neurons and synapses can be combined as 'building blocks' to generate circuit function. We focus on the importance of capturing multiple timescales when describing these intrinsic and synaptic components. Whether inherent in the ionic currents, the neuron's complex morphology, or the neurotransmitter composition of synapses, these multiple timescales prove crucial for capturing the variability and richness of circuit output and enhancing the information-carrying capacity observed across nervous systems.
Collapse
Affiliation(s)
- Julijana Gjorgjieva
- Volen Center and Biology Department, Brandeis University, Waltham, MA 02454, United States
| | - Guillaume Drion
- Volen Center and Biology Department, Brandeis University, Waltham, MA 02454, United States; Department of Electrical Engineering and Computer Science, University of Liège, Liège B-4000, Belgium
| | - Eve Marder
- Volen Center and Biology Department, Brandeis University, Waltham, MA 02454, United States.
| |
Collapse
|
36
|
Hirasawa H, Contini M, Raviola E. Extrasynaptic release of GABA and dopamine by retinal dopaminergic neurons. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0186. [PMID: 26009765 DOI: 10.1098/rstb.2014.0186] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In the mouse retina, dopaminergic amacrine (DA) cells synthesize both dopamine and GABA. Both transmitters are released extrasynaptically and act on neighbouring and distant retinal neurons by volume transmission. In simultaneous recordings of dopamine and GABA release from isolated perikarya of DA cells, a proportion of the events of dopamine and GABA exocytosis were simultaneous, suggesting co-release. In addition, DA cells establish GABAergic synapses onto AII amacrine cells, the neurons that transfer rod bipolar signals to cone bipolars. GABAA but not dopamine receptors are clustered in the postsynaptic membrane. Therefore, dopamine, irrespective of its site of release-synaptic or extrasynaptic-exclusively acts by volume transmission. Dopamine is released upon illumination and sets the gain of retinal neurons for vision in bright light. The GABA released at DA cells' synapses probably prevents signals from the saturated rods from entering the cone pathway when the dark-adapted retina is exposed to bright illumination. The GABA released extrasynaptically by DA and other amacrine cells may set a 'GABAergic tone' in the inner plexiform layer and thus counteract the effects of a spillover of glutamate released at the bipolar cell synapses of adjacent OFF and ON strata, thus preserving segregation of signals between ON and OFF pathways.
Collapse
Affiliation(s)
- Hajime Hirasawa
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA Department of Physiology, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama, Saitama 350-0495, Japan
| | - Massimo Contini
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA Dipartimento di Medicina Sperimentale e Clinica, Viale Morgagni, 63, Firenze 50134, Italy
| | - Elio Raviola
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
37
|
Abstract
The 'one neuron, one neurotransmitter' doctrine states that synaptic communication between two neurons occurs through the release of a single chemical transmitter. However, recent findings suggest that neurons that communicate using more than one classical neurotransmitter are prevalent throughout the adult mammalian CNS. In particular, several populations of neurons previously thought to release only glutamate, acetylcholine, dopamine or histamine also release the major inhibitory neurotransmitter GABA. Here, we review these findings and discuss the implications of GABA co-release for synaptic transmission and plasticity.
Collapse
|
38
|
Firsov ML, Astakhova LA. The Role of Dopamine in Controlling Retinal Photoreceptor Function in Vertebrates. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/s11055-015-0210-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
39
|
Vuong HE, Hardi CN, Barnes S, Brecha NC. Parallel Inhibition of Dopamine Amacrine Cells and Intrinsically Photosensitive Retinal Ganglion Cells in a Non-Image-Forming Visual Circuit of the Mouse Retina. J Neurosci 2015; 35:15955-70. [PMID: 26631476 PMCID: PMC4666919 DOI: 10.1523/jneurosci.3382-15.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/20/2015] [Accepted: 10/30/2015] [Indexed: 12/12/2022] Open
Abstract
An inner retinal microcircuit composed of dopamine (DA)-containing amacrine cells and melanopsin-containing, intrinsically photosensitive retinal ganglion cells (M1 ipRGCs) process information about the duration and intensity of light exposures, mediating light adaptation, circadian entrainment, pupillary reflexes, and other aspects of non-image-forming vision. The neural interaction is reciprocal: M1 ipRGCs excite DA amacrine cells, and these, in turn, feed inhibition back onto M1 ipRGCs. We found that the neuropeptide somatostatin [somatotropin release inhibiting factor (SRIF)] also inhibits the intrinsic light response of M1 ipRGCs and postulated that, to tune the bidirectional interaction of M1 ipRGCs and DA amacrine cells, SRIF amacrine cells would provide inhibitory modulation to both cell types. SRIF amacrine cells, DA amacrine cells, and M1 ipRGCs form numerous contacts. DA amacrine cells and M1 ipRGCs express the SRIF receptor subtypes sst(2A) and sst4 respectively. SRIF modulation of the microcircuit was investigated with targeted patch-clamp recordings of DA amacrine cells in TH-RFP mice and M1 ipRGCs in OPN4-EGFP mice. SRIF increases K(+) currents, decreases Ca(2+) currents, and inhibits spike activity in both cell types, actions reproduced by the selective sst(2A) agonist L-054,264 (N-[(1R)-2-[[[(1S*,3R*)-3-(aminomethyl)cyclohexyl]methyl]amino]-1-(1H-indol-3-ylmethyl)-2-oxoethyl]spiro[1H-indene-1,4'-piperidine]-1'-carboxamide) in DA amacrine cells and the selective sst4 agonist L-803,087 (N(2)-[4-(5,7-difluoro-2-phenyl-1H-indol-3-yl)-1-oxobutyl]-L-arginine methyl ester trifluoroacetate) in M1 ipRGCs. These parallel actions of SRIF may serve to counteract the disinhibition of M1 ipRGCs caused by SRIF inhibition of DA amacrine cells. This allows the actions of SRIF on DA amacrine cells to proceed with adjusting retinal DA levels without destabilizing light responses by M1 ipRGCs, which project to non-image-forming targets in the brain.
Collapse
Affiliation(s)
- Helen E Vuong
- Departments of Neurobiology and Molecular, Cellular, and Integrative Physiology, Stein Eye Institute, and
| | | | - Steven Barnes
- Departments of Neurobiology and Departments of Physiology and Biophysics and Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada, and Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California 90073
| | - Nicholas C Brecha
- Departments of Neurobiology and Molecular, Cellular, and Integrative Physiology, Stein Eye Institute, and CURE: Digestive Diseases Research Center, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California 90095, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California 90073
| |
Collapse
|
40
|
Vuong HE, Pérez de Sevilla Müller L, Hardi CN, McMahon DG, Brecha NC. Heterogeneous transgene expression in the retinas of the TH-RFP, TH-Cre, TH-BAC-Cre and DAT-Cre mouse lines. Neuroscience 2015; 307:319-37. [PMID: 26335381 PMCID: PMC4603663 DOI: 10.1016/j.neuroscience.2015.08.060] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 08/21/2015] [Accepted: 08/24/2015] [Indexed: 11/29/2022]
Abstract
Transgenic mouse lines are essential tools for understanding the connectivity, physiology and function of neuronal circuits, including those in the retina. This report compares transgene expression in the retina of a tyrosine hydroxylase (TH)-red fluorescent protein (RFP) mouse line with three catecholamine-related Cre recombinase mouse lines [TH-bacterial artificial chromosome (BAC)-, TH-, and dopamine transporter (DAT)-Cre] that were crossed with a ROSA26-tdTomato reporter line. Retinas were evaluated and immunostained with commonly used antibodies including those directed to TH, GABA and glycine to characterize the RFP or tdTomato fluorescent-labeled amacrine cells, and an antibody directed to RNA-binding protein with multiple splicing to identify ganglion cells. In TH-RFP retinas, types 1 and 2 dopamine (DA) amacrine cells were identified by their characteristic cellular morphology and type 1 DA cells by their expression of TH immunoreactivity. In the TH-BAC-, TH-, and DAT-tdTomato retinas, less than 1%, ∼ 6%, and 0%, respectively, of the fluorescent cells were the expected type 1 DA amacrine cells. Instead, in the TH-BAC-tdTomato retinas, fluorescently labeled AII amacrine cells were predominant, with some medium diameter ganglion cells. In TH-tdTomato retinas, fluorescence was in multiple neurochemical amacrine cell types, including four types of polyaxonal amacrine cells. In DAT-tdTomato retinas, fluorescence was in GABA immunoreactive amacrine cells, including two types of bistratified and two types of monostratified amacrine cells. Although each of the Cre lines was generated with the intent to specifically label DA cells, our findings show a cellular diversity in Cre expression in the adult retina and indicate the importance of careful characterization of transgene labeling patterns. These mouse lines with their distinctive cellular labeling patterns will be useful tools for future studies of retinal function and visual processing.
Collapse
Affiliation(s)
- H E Vuong
- Molecular, Cellular, and Integrative Physiology Program, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
| | - L Pérez de Sevilla Müller
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
| | - C N Hardi
- Department of Psychology, College of Letters and Science, UCLA, Los Angeles, CA 90095, United States
| | - D G McMahon
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, United States
| | - N C Brecha
- Molecular, Cellular, and Integrative Physiology Program, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; CURE-Digestive Diseases Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90095, United States.
| |
Collapse
|
41
|
Ho T, Jobling AI, Greferath U, Chuang T, Ramesh A, Fletcher EL, Vessey KA. Vesicular expression and release of ATP from dopaminergic neurons of the mouse retina and midbrain. Front Cell Neurosci 2015; 9:389. [PMID: 26500494 PMCID: PMC4593860 DOI: 10.3389/fncel.2015.00389] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 09/17/2015] [Indexed: 12/20/2022] Open
Abstract
Vesicular nucleotide transporter (VNUT) is required for active accumulation of adenosine tri-phosphate (ATP) into vesicles for purinergic neurotransmission, however, the cell types that express VNUT in the central nervous system remain unknown. This study characterized VNUT expression within the mammalian retina and brain and assessed a possible functional role in purinergic signaling. Two native isoforms of VNUT were detected in mouse retina and brain based on RNA transcript and protein analysis. Using immunohistochemistry, VNUT was found to co-localize with tyrosine hydroxylase (TH) positive, dopaminergic (DA) neurons of the substantia nigra and ventral tegmental area, however, VNUT expression in extranigral non-DA neurons was also observed. In the retina, VNUT labeling was found to co-localize solely with TH-positive DA-cells. In the outer retina, VNUT-positive interplexiform cell processes were in close contact with horizontal cells and cone photoreceptor terminals, which are known to express P2 purinergic-receptors. In order to assess function, dissociated retinal neurons were loaded with fluorescent ATP markers (Quinacrine or Mant-ATP) and the DA marker FFN102, co-labeled with a VNUT antibody and imaged in real time. Fluorescent ATP markers and FFN102 puncta were found to co-localize in VNUT positive neurons and upon stimulation with high potassium, ATP marker fluorescence at the cell membrane was reduced. This response was blocked in the presence of cadmium. These data suggest DA neurons co-release ATP via calcium dependent exocytosis and in the retina this may modulate the visual response by activating purine receptors on closely associated neurons.
Collapse
Affiliation(s)
- Tracy Ho
- Visual Neuroscience Laboratory, Department of Anatomy and Neuroscience, The University of Melbourne Parkville, VIC, Australia
| | - Andrew I Jobling
- Visual Neuroscience Laboratory, Department of Anatomy and Neuroscience, The University of Melbourne Parkville, VIC, Australia
| | - Ursula Greferath
- Visual Neuroscience Laboratory, Department of Anatomy and Neuroscience, The University of Melbourne Parkville, VIC, Australia
| | - Trinette Chuang
- Polyclonal Antibody Development, R&D Antibody Development, EMD Millipore Temecula, CA, USA
| | - Archana Ramesh
- Polyclonal Antibody Development, R&D Antibody Development, EMD Millipore Temecula, CA, USA
| | - Erica L Fletcher
- Visual Neuroscience Laboratory, Department of Anatomy and Neuroscience, The University of Melbourne Parkville, VIC, Australia
| | - Kirstan A Vessey
- Visual Neuroscience Laboratory, Department of Anatomy and Neuroscience, The University of Melbourne Parkville, VIC, Australia
| |
Collapse
|
42
|
Fyk-Kolodziej BE, Shimano T, Gafoor D, Mirza N, Griffith RD, Gong TW, Holt AG. Dopamine in the auditory brainstem and midbrain: co-localization with amino acid neurotransmitters and gene expression following cochlear trauma. Front Neuroanat 2015; 9:88. [PMID: 26257610 PMCID: PMC4510424 DOI: 10.3389/fnana.2015.00088] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 06/19/2015] [Indexed: 11/13/2022] Open
Abstract
Dopamine (DA) modulates the effects of amino acid neurotransmitters (AANs), including GABA and glutamate, in motor, visual, olfactory, and reward systems (Hnasko et al., 2010; Stuber et al., 2010; Hnasko and Edwards, 2012). The results suggest that DA may play a similar modulatory role in the auditory pathways. Previous studies have shown that deafness results in decreased GABA release, changes in excitatory neurotransmitter levels, and increased spontaneous neuronal activity within brainstem regions related to auditory function. Modulation of the expression and localization of tyrosine hydroxylase (TH; the rate limiting enzyme in the production of DA) in the IC following cochlear trauma has been previously reported (Tong et al., 2005). In the current study the possibility of co-localization of TH with AANs was examined. Changes in the gene expression of TH were compared with changes in the gene expression of markers for AANs in the cochlear nucleus (CN) and inferior colliculus (IC) to determine whether those deafness related changes occur concurrently. The results indicate that bilateral cochlear ablation significantly reduced TH gene expression in the CN after 2 months while in the IC the reduction in TH was observed at both 3 days and 2 months following ablation. Furthermore, in the CN, glycine transporter 2 (GLYT2) and the GABA transporter (GABAtp) were also significantly reduced only after 2 months. However, in the IC, DA receptor 1 (DRDA1), vesicular glutamate transporters 2 and 3 (VGLUT2, VGLUT3), GABAtp and GAD67 were reduced in expression both at the 3 days and 2 months time points. A close relationship between the distribution of TH and several of the AANs was determined in both the CN and the IC. In addition, GLYT2 and VGLUT3 each co-localized with TH within IC somata and dendrites. Therefore, the results of the current study suggest that DA is spatially well positioned to influence the effects of AANs on auditory neurons.
Collapse
Affiliation(s)
- Bozena E Fyk-Kolodziej
- Molecular Anatomy of Auditory-related Central Systems, Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit MI, USA
| | - Takashi Shimano
- Department of Otolaryngology, Kansai Medical University Osaka, Japan
| | - Dana Gafoor
- Molecular Anatomy of Auditory-related Central Systems, Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit MI, USA
| | - Najab Mirza
- Molecular Anatomy of Auditory-related Central Systems, Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit MI, USA
| | - Ronald D Griffith
- Molecular Anatomy of Auditory-related Central Systems, Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit MI, USA
| | - Tzy-Wen Gong
- Kresge Hearing Research Institute, University of Michigan School of Medicine, Ann Arbor MI, USA
| | - Avril Genene Holt
- Molecular Anatomy of Auditory-related Central Systems, Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit MI, USA
| |
Collapse
|
43
|
Debertin G, Kántor O, Kovács-Öller T, Balogh L, Szabó-Meleg E, Orbán J, Nyitrai M, Völgyi B. Tyrosine hydroxylase positive perisomatic rings are formed around various amacrine cell types in the mammalian retina. J Neurochem 2015; 134:416-28. [DOI: 10.1111/jnc.13144] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 04/10/2015] [Indexed: 01/21/2023]
Affiliation(s)
- Gábor Debertin
- Department of Experimental Zoology and Neurobiology; University of Pécs; Pécs Hungary
- János Szentágothai Research Center; Pécs Hungary
- MTA-PTE NAP B Retinal Electrical Synapses Research Group; Pécs Hungary
| | - Orsolya Kántor
- Department of Anatomy, Histology and Embryology; Semmelweis University; Budapest Hungary
| | - Tamás Kovács-Öller
- Department of Experimental Zoology and Neurobiology; University of Pécs; Pécs Hungary
- János Szentágothai Research Center; Pécs Hungary
- MTA-PTE NAP B Retinal Electrical Synapses Research Group; Pécs Hungary
| | - Lajos Balogh
- National “F. J. C.” Research Institute for Radiobiology and Radiohygiene; Budapest Hungary
| | | | - József Orbán
- Department of Biophysics; University of Pécs; Pécs Hungary
| | - Miklós Nyitrai
- János Szentágothai Research Center; Pécs Hungary
- Department of Biophysics; University of Pécs; Pécs Hungary
| | - Béla Völgyi
- Department of Experimental Zoology and Neurobiology; University of Pécs; Pécs Hungary
- János Szentágothai Research Center; Pécs Hungary
- MTA-PTE NAP B Retinal Electrical Synapses Research Group; Pécs Hungary
- Department of Ophthalmology; New York University Langone Medical Center; New York New York USA
| |
Collapse
|
44
|
Affiliation(s)
- Brigham J. Hartley
- Departments of Psychiatry and Neuroscience, Mount Sinai School of
Medicine, 1425 Madison Avenue, New York, NY 10029
| | - Ngoc Tran
- Departments of Psychiatry and Neuroscience, Mount Sinai School of
Medicine, 1425 Madison Avenue, New York, NY 10029
| | - Ian Ladran
- Departments of Psychiatry and Neuroscience, Mount Sinai School of
Medicine, 1425 Madison Avenue, New York, NY 10029
| | - Kathryn Reggio
- Departments of Psychiatry and Neuroscience, Mount Sinai School of
Medicine, 1425 Madison Avenue, New York, NY 10029
| | - Kristen J. Brennand
- Departments of Psychiatry and Neuroscience, Mount Sinai School of
Medicine, 1425 Madison Avenue, New York, NY 10029
| |
Collapse
|
45
|
Filippi A, Mueller T, Driever W. vglut2 and gad expression reveal distinct patterns of dual GABAergic versus glutamatergic cotransmitter phenotypes of dopaminergic and noradrenergic neurons in the zebrafish brain. J Comp Neurol 2015; 522:2019-37. [PMID: 24374659 PMCID: PMC4288968 DOI: 10.1002/cne.23524] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 11/27/2013] [Accepted: 11/27/2013] [Indexed: 01/22/2023]
Abstract
Throughout the vertebrate lineage, dopaminergic neurons form important neuromodulatory systems that influence motor behavior, mood, cognition, and physiology. Studies in mammals have established that dopaminergic neurons often use γ-aminobutyric acid (GABA) or glutamatergic cotransmission during development and physiological function. Here, we analyze vglut2, gad1b and gad2 expression in combination with tyrosine hydroxylase immunoreactivity in 4-day-old larval and 30-day-old juvenile zebrafish brains to determine which dopaminergic and noradrenergic groups may use GABA or glutamate as a second transmitter. Our results show that most dopaminergic neurons also express GABAergic markers, including the dopaminergic groups of the olfactory bulb (homologous to mammalian A16) and the subpallium, the hypothalamic groups (A12, A14), the prethalamic zona incerta group (A13), the preoptic groups (A15), and the pretectal group. Thus, the majority of catecholaminergic neurons are gad1b/2-positive and coexpress GABA. A very few gad1/2-negative dopaminergic groups, however, express vglut2 instead and use glutamate as a second transmitter. These glutamatergic dual transmitter phenotypes are the Orthopedia transcription factor–dependent, A11-type dopaminergic neurons of the posterior tuberculum. All together, our results demonstrate that all catecholaminergic groups in zebrafish are either GABAergic or glutamatergic. Thus, cotransmission of dopamine and noradrenaline with either GABA or glutamate appears to be a regular feature of zebrafish catecholaminergic systems. We compare our results with those that have been described for mammalian systems, discuss the phenomenon of transmitter dualism in the context of developmental specification of GABAergic and glutamatergic regions in the brain, and put this phenomenon in an evolutionary perspective. J. Comp. Neurol. 522:2019–2037, 2014.
Collapse
Affiliation(s)
- Alida Filippi
- Developmental Biology, Institute of Biology I, Faculty of Biology, Albert-Ludwigs-University Freiburg, 79104, Freiburg, Germany
| | | | | |
Collapse
|
46
|
Immunolocalization of the P2X4 receptor on neurons and glia in the mammalian retina. Neuroscience 2014; 277:55-71. [DOI: 10.1016/j.neuroscience.2014.06.055] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 06/23/2014] [Accepted: 06/24/2014] [Indexed: 02/07/2023]
|
47
|
Tritsch NX, Oh WJ, Gu C, Sabatini BL. Midbrain dopamine neurons sustain inhibitory transmission using plasma membrane uptake of GABA, not synthesis. eLife 2014; 3:e01936. [PMID: 24843012 PMCID: PMC4001323 DOI: 10.7554/elife.01936] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Synaptic transmission between midbrain dopamine neurons and target neurons in the striatum is essential for the selection and reinforcement of movements. Recent evidence indicates that nigrostriatal dopamine neurons inhibit striatal projection neurons by releasing a neurotransmitter that activates GABAA receptors. Here, we demonstrate that this phenomenon extends to mesolimbic afferents, and confirm that the released neurotransmitter is GABA. However, the GABA synthetic enzymes GAD65 and GAD67 are not detected in midbrain dopamine neurons. Instead, these cells express the membrane GABA transporters mGAT1 (Slc6a1) and mGAT4 (Slc6a11) and inhibition of these transporters prevents GABA co-release. These findings therefore indicate that GABA co-release is a general feature of midbrain dopaminergic neurons that relies on GABA uptake from the extracellular milieu as opposed to de novo synthesis. This atypical mechanism may confer dopaminergic neurons the flexibility to differentially control GABAergic transmission in a target-dependent manner across their extensive axonal arbors. DOI:http://dx.doi.org/10.7554/eLife.01936.001 The electrical signals that are fired along neurons cannot be transmitted across the small gaps, called synapses that are found between neurons. Instead, the neuron sending the signal releases chemicals called neurotransmitters into the synapse. These neurotransmitters bind to receptor proteins on the surface of the second neuron and control how it fires. A neurotransmitter called dopamine plays a key role in the circuits of the brain that control how we learn certain tasks involving movement. In particular, two populations of neurons from the midbrain that release dopamine target the striatum, an area of the brain that is responsible for motor control. These neurons also release other neurotransmitters, but the identity of these other chemicals is not known, and the details of the interaction between the neurons and the striatum are poorly understood. Previous research showed that some of the midbrain neurons activate receptors that normally respond to a neurotransmitter called gamma-aminobutyric acid (GABA). However, several different chemicals can trigger this receptor. Using a range of techniques, Tritsch et al. now confirm that dopamine neurons release GABA alongside dopamine, and that this applies to both sets of the dopamine-producing neurons that feed into the striatum. Some neurons can manufacture GABA from amino acids found in their internal fluid. However, Tritsch et al. could not detect the enzymes needed for this reaction in dopamine-producing neurons. Instead, these neurons contain proteins that can transport GABA across the cell membrane, which suggests that the neurons collect GABA from the extracellular fluid that surrounds them. DOI:http://dx.doi.org/10.7554/eLife.01936.002
Collapse
Affiliation(s)
- Nicolas X Tritsch
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| | - Won-Jong Oh
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Chenghua Gu
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Bernardo L Sabatini
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| |
Collapse
|
48
|
Svensson E, Proekt A, Jing J, Weiss KR. PKC-mediated GABAergic enhancement of dopaminergic responses: implication for short-term potentiation at a dual-transmitter synapse. J Neurophysiol 2014; 112:22-9. [PMID: 24717352 DOI: 10.1152/jn.00794.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Transmitter-mediated homosynaptic potentiation is generally implemented by the same transmitter that mediates the excitatory postsynaptic potentials (EPSPs), e.g., glutamate. When a presynaptic neuron contains more than one transmitter, however, potentiation can in principle be implemented by a transmitter different from that which elicits the EPSPs. Neuron B20 in Aplysia contains both dopamine and GABA. Although only dopamine acts as the fast excitatory transmitter at the B20-to-B8 synapse, GABA increases the size of these dopaminergic EPSPs. We now provide evidence that repeated stimulation of B20 potentiates B20-evoked dopaminergic EPSPs in B8 apparently via a postsynaptic mechanism, and short-term potentiation of this synapse is critical for the establishment and maintenance of an egestive network state. We show that GABA can act postsynaptically to increase dopamine currents that are elicited by direct applications of dopamine to B8 and that dopamine is acting on a 5-HT3-like receptor. This potentiation is mediated by GABAB-like receptors as GABAB-receptor agonists and antagonists, respectively, mimicked and blocked the potentiating actions of GABA. The postsynaptic actions of GABA rely on a G protein-mediated activation of PKC. Our results suggest that the postsynaptic action of cotransmitter-mediated potentiation may contribute to the maintenance of the egestive state of Aplysia feeding network and, in more general terms, may participate in the plasticity of networks that mediate complex behaviors.
Collapse
Affiliation(s)
- Erik Svensson
- Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, New York, New York
| | - Alex Proekt
- Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, New York, New York
| | - Jian Jing
- Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, New York, New York
| | - Klaudiusz R Weiss
- Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, New York, New York
| |
Collapse
|
49
|
Trudeau LE, Hnasko TS, Wallén-Mackenzie A, Morales M, Rayport S, Sulzer D. The multilingual nature of dopamine neurons. PROGRESS IN BRAIN RESEARCH 2014; 211:141-64. [PMID: 24968779 DOI: 10.1016/b978-0-444-63425-2.00006-4] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The ability of dopamine (DA) neurons to release other transmitters in addition to DA itself has been increasingly recognized, hence the concept of their multilingual nature. A subset of DA neurons, mainly found in the ventral tegmental area, express VGLUT2, allowing them to package and release glutamate onto striatal spiny projection neurons and cholinergic interneurons. Some dopaminergic axon terminals release GABA. Glutamate release by DA neurons has a developmental role, facilitating axonal growth and survival, and may determine in part the critical contribution of the ventral striatum to psychostimulant-induced behavior. Vesicular glutamate coentry may have synergistic effects on vesicular DA filling. The multilingual transmission of DA neurons across multiple striatal domains and the increasing insight into the role of glutamate cotransmission in the ventral striatum highlight the importance of analyzing DA neuron transmission at the synaptic level.
Collapse
Affiliation(s)
- Louis-Eric Trudeau
- Department of Pharmacology, Neuroscience Research Group, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada; Department of Neurosciences, Neuroscience Research Group, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.
| | - Thomas S Hnasko
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Asa Wallén-Mackenzie
- Unit of Functional Neurobiology, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Marisela Morales
- National Institute on Drug Abuse, Intramural Research Program, Neuronal Networks Section, Baltimore, MD, USA
| | - Steven Rayport
- Department of Psychiatry, Columbia University, New York, NY, USA; Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, NY, USA
| | - David Sulzer
- Department of Psychiatry, Columbia University, New York, NY, USA; Department of Neurology, Columbia University, New York, NY, USA; Department of Pharmacology, Columbia University, New York, NY, USA; Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, NY, USA
| |
Collapse
|
50
|
Stensrud M, Chaudhry F, Leergaard T, Bjaalie J, Gundersen V. Vesicular glutamate transporter-3 in the rodent brain: Vesicular colocalization with vesicular γ-aminobutyric acid transporter. J Comp Neurol 2013; 521:3042-56. [DOI: 10.1002/cne.23331] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 03/08/2013] [Accepted: 03/13/2013] [Indexed: 01/12/2023]
Affiliation(s)
- M.J. Stensrud
- Department of Anatomy; Institute of Basic Medical Sciences, and Centre for Molecular Biology and Neuroscience (CMBN), University of Oslo; 0317 Oslo; Norway
| | - F.A. Chaudhry
- The Biotechnology Centre of Oslo and The Centre for Molecular Biology and Neuroscience (CMBN); University of Oslo; 0317 Oslo; Norway
| | - T.B. Leergaard
- Department of Anatomy; Institute of Basic Medical Sciences, and Centre for Molecular Biology and Neuroscience (CMBN), University of Oslo; 0317 Oslo; Norway
| | - J.G. Bjaalie
- Department of Anatomy; Institute of Basic Medical Sciences, and Centre for Molecular Biology and Neuroscience (CMBN), University of Oslo; 0317 Oslo; Norway
| | | |
Collapse
|