1
|
Matsumoto A, Morris J, Looger LL, Yonehara K. Functionally distinct GABAergic amacrine cell types regulate spatiotemporal encoding in the mouse retina. Nat Neurosci 2025:10.1038/s41593-025-01935-0. [PMID: 40234708 DOI: 10.1038/s41593-025-01935-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025]
Abstract
GABA (γ-aminobutyric acid) is the primary inhibitory neurotransmitter in the mammalian central nervous system. GABAergic neuronal types play important roles in neural processing and the etiology of neurological disorders; however, there is no comprehensive understanding of their functional diversity. Here we perform two-photon imaging of GABA release in the inner plexiform layer of male and female mice retinae (8-16 weeks old) using the GABA sensor iGABASnFR2. By applying varied light stimuli to isolated retinae, we reveal over 40 different GABA-releasing neuron types. Individual types show layer-specific visual encoding within inner plexiform layer sublayers. Synaptic input and output sites are aligned along specific retinal orientations. The combination of cell type-specific spatial structure and unique release kinetics enables inhibitory neurons to sculpt excitatory signals in response to a wide range of behaviorally relevant motion structures. Our findings emphasize the importance of functional diversity and intricate specialization of GABAergic neurons in the central nervous system.
Collapse
Affiliation(s)
- Akihiro Matsumoto
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus, Denmark.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Japan.
- Graduate Institute for Advanced Studies, SOKENDAI, Hayama, Japan.
| | - Jacqueline Morris
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA, USA
| | - Loren L Looger
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA, USA
| | - Keisuke Yonehara
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus, Denmark.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Japan.
- Graduate Institute for Advanced Studies, SOKENDAI, Hayama, Japan.
| |
Collapse
|
2
|
Zloh M, Kutilek P, Hejda J, Fiserova I, Kubovciak J, Murakami M, Stofkova A. Visual stimulation and brain-derived neurotrophic factor (BDNF) have protective effects in experimental autoimmune uveoretinitis. Life Sci 2024; 355:122996. [PMID: 39173995 DOI: 10.1016/j.lfs.2024.122996] [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: 05/08/2024] [Revised: 07/26/2024] [Accepted: 08/18/2024] [Indexed: 08/24/2024]
Abstract
AIMS To investigate the therapeutic potential of visual stimulation (VS) and BDNF in murine experimental autoimmune uveoretinitis (EAU). MAIN METHODS Mice were immunized by subcutaneous injection of interphotoreceptor retinoid-binding protein in Freund's complete adjuvant and intravenous injection of pertussis toxin, and were then exposed to high-contrast VS 12 h/day (days 1-14 post-immunization). EAU severity was assessed by examining clinical score, visual acuity, inflammatory markers, and immune cells in the retina. The transcriptome of activated retinal cells was determined by RNA-seq using RNA immunoprecipitated in complex with phosphorylated ribosomal protein S6. The retinal levels of protein products of relevant upregulated genes were quantified. The effect of BDNF on EAU was tested in unstimulated mice by its daily topical ocular administration (days 8-14 post-immunization). KEY FINDINGS VS attenuated EAU development and decreased the expression of pro-inflammatory cytokines/chemokines and numbers of immune cells in the retina (n = 10-20 eyes/group for each analysis). In activated retinal cells of control mice (n = 30 eyes/group), VS upregulated genes encoding immunomodulatory neuropeptides, of which BDNF and vasoactive intestinal peptide (VIP) also showed increased mRNA and protein levels in the retina of VS-treated EAU mice (n = 6-10 eyes/group for each analysis). In unstimulated EAU mice, BDNF treatment mimicked the protective effects of VS by modulating the inflammatory and stem cell properties of Müller cells (n = 5 eyes/group for each analysis). SIGNIFICANCE VS effectively suppresses EAU, at least through enhancing retinal levels of anti-inflammatory and neuroprotective factors, VIP and BDNF. Our findings also suggest BDNF as a promising therapeutic agent for uveitis treatment.
Collapse
Affiliation(s)
- Miloslav Zloh
- Department of Physiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Patrik Kutilek
- Department of Health Care and Population Protection, Faculty of Biomedical Engineering, Czech Technical University in Prague, Kladno, Czech Republic
| | - Jan Hejda
- Department of Health Care and Population Protection, Faculty of Biomedical Engineering, Czech Technical University in Prague, Kladno, Czech Republic
| | - Ivana Fiserova
- Department of Physiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic; Department of Biochemistry, Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Kubovciak
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Masaaki Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Group of Quantum Immunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology (QST), Chiba, Japan; Division of Molecular Neuroimmunology, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi, Japan; Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
| | - Andrea Stofkova
- Department of Physiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
3
|
Ganzen L, Yadav SC, Wei M, Ma H, Nawy S, Kramer RH. Retinoic Acid-Dependent Loss of Synaptic Output from Bipolar Cells Impairs Visual Information Processing in Inherited Retinal Degeneration. J Neurosci 2024; 44:e0129242024. [PMID: 39060177 PMCID: PMC11358532 DOI: 10.1523/jneurosci.0129-24.2024] [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: 01/18/2024] [Revised: 07/09/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
In retinitis pigmentosa (RP), rod and cone photoreceptors degenerate, depriving downstream neurons of light-sensitive input, leading to vision impairment or blindness. Although downstream neurons survive, some undergo morphological and physiological remodeling. Bipolar cells (BCs) link photoreceptors, which sense light, to retinal ganglion cells (RGCs), which send information to the brain. While photoreceptor loss disrupts input synapses to BCs, whether BC output synapses remodel has remained unknown. Here we report that synaptic output from BCs plummets in RP mouse models of both sexes owing to loss of voltage-gated Ca2+ channels. Remodeling reduces the reliability of synaptic output to repeated optogenetic stimuli, causing RGC firing to fail at high-stimulus frequencies. Fortunately, functional remodeling of BCs can be reversed by inhibiting the retinoic acid receptor (RAR). RAR inhibitors targeted to BCs present a new therapeutic opportunity for mitigating detrimental effects of remodeling on signals initiated either by surviving photoreceptors or by vision-restoring tools.
Collapse
Affiliation(s)
- Logan Ganzen
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Shubhash Chandra Yadav
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Mingxiao Wei
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Hong Ma
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Scott Nawy
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Richard H Kramer
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| |
Collapse
|
4
|
Fitzpatrick MJ, Krizan J, Hsiang JC, Shen N, Kerschensteiner D. A pupillary contrast response in mice and humans: Neural mechanisms and visual functions. Neuron 2024; 112:2404-2422.e9. [PMID: 38697114 PMCID: PMC11257825 DOI: 10.1016/j.neuron.2024.04.012] [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: 04/03/2023] [Revised: 12/21/2023] [Accepted: 04/10/2024] [Indexed: 05/04/2024]
Abstract
In the pupillary light response (PLR), increases in ambient light constrict the pupil to dampen increases in retinal illuminance. Here, we report that the pupillary reflex arc implements a second input-output transformation; it senses temporal contrast to enhance spatial contrast in the retinal image and increase visual acuity. The pupillary contrast response (PCoR) is driven by rod photoreceptors via type 6 bipolar cells and M1 ganglion cells. Temporal contrast is transformed into sustained pupil constriction by the M1's conversion of excitatory input into spike output. Computational modeling explains how the PCoR shapes retinal images. Pupil constriction improves acuity in gaze stabilization and predation in mice. Humans exhibit a PCoR with similar tuning properties to mice, which interacts with eye movements to optimize the statistics of the visual input for retinal encoding. Thus, we uncover a conserved component of active vision, its cell-type-specific pathway, computational mechanisms, and optical and behavioral significance.
Collapse
Affiliation(s)
- Michael J Fitzpatrick
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Graduate Program in Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Medical Scientist Training Program, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jenna Krizan
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Graduate Program in Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jen-Chun Hsiang
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Ning Shen
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
5
|
Zhang X, Wang X, Zhu J, Chen K, Ullah R, Tong J, Shen Y. Retinal VIP-amacrine cells: their development, structure, and function. Eye (Lond) 2024; 38:1065-1076. [PMID: 38066110 PMCID: PMC11009269 DOI: 10.1038/s41433-023-02844-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: 07/18/2022] [Revised: 10/20/2023] [Accepted: 11/13/2023] [Indexed: 04/13/2024] Open
Abstract
Amacrine cells (ACs) are the most structurally and functionally diverse neuron type in the retina. Different ACs have distinct functions, such as neuropeptide secretion and inhibitory connection. Vasoactive intestinal peptide (VIP) -ergic -ACs are retina gamma-aminobutyric acid (GABA) -ergic -ACs that were discovered long ago. They secrete VIP and form connections with bipolar cells (BCs), other ACs, and retinal ganglion cells (RGCs). They have a specific structure, density, distribution, and function. They play an important role in myopia, light stimulated responses, retinal vascular disease and other ocular diseases. Their significance in the study of refractive development and disease is increasing daily. However, a systematic review of the structure and function of retinal VIP-ACs is lacking. We discussed the detailed characteristics of VIP-ACs from every aspect across species and providing systematic knowledge base for future studies. Our review led to the main conclusion that retinal VIP-ACs develop early, and although their morphology and distribution across species are not the same, they have similar functions in a wide range of ocular diseases based on their function of secreting neuropeptides and forming inhibitory connections with other cells.
Collapse
Affiliation(s)
- Xuhong Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Medical School, Zhejiang University, Hangzhou, China
| | - Xiaoyu Wang
- Department of Ophthalmology, The First Affiliated Hospital of Medical School, Zhejiang University, Hangzhou, China
- Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Jiru Zhu
- Department of Ophthalmology, The First Affiliated Hospital of Medical School, Zhejiang University, Hangzhou, China
| | - Kuangqi Chen
- Department of Ophthalmology, The First Affiliated Hospital of Medical School, Zhejiang University, Hangzhou, China
| | - Rahim Ullah
- Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianping Tong
- Department of Ophthalmology, The First Affiliated Hospital of Medical School, Zhejiang University, Hangzhou, China.
| | - Ye Shen
- Department of Ophthalmology, The First Affiliated Hospital of Medical School, Zhejiang University, Hangzhou, China.
| |
Collapse
|
6
|
Park SJ, Lei W, Pisano J, Orpia A, Minehart J, Pottackal J, Hanke-Gogokhia C, Zapadka TE, Clarkson-Paredes C, Popratiloff A, Ross SE, Singer JH, Demb JB. Molecular identification of wide-field amacrine cells in mouse retina that encode stimulus orientation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.28.573580. [PMID: 38234775 PMCID: PMC10793454 DOI: 10.1101/2023.12.28.573580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Visual information processing is sculpted by a diverse group of inhibitory interneurons in the retina called amacrine cells. Yet, for most of the >60 amacrine cell types, molecular identities and specialized functional attributes remain elusive. Here, we developed an intersectional genetic strategy to target a group of wide-field amacrine cells (WACs) in mouse retina that co-express the transcription factor Bhlhe22 and the Kappa Opioid Receptor (KOR; B/K WACs). B/K WACs feature straight, unbranched dendrites spanning over 0.5 mm (∼15° visual angle) and produce non-spiking responses to either light increments or decrements. Two-photon dendritic population imaging reveals Ca 2+ signals tuned to the physical orientations of B/K WAC dendrites, signifying a robust structure-function alignment. B/K WACs establish divergent connections with multiple retinal neurons, including unexpected connections with non-orientation-tuned ganglion cells and bipolar cells. Our work sets the stage for future comprehensive investigations of the most enigmatic group of retinal neurons: WACs.
Collapse
|
7
|
Nath A, Grimes WN, Diamond JS. Layers of inhibitory networks shape receptive field properties of AII amacrine cells. Cell Rep 2023; 42:113390. [PMID: 37930888 PMCID: PMC10769003 DOI: 10.1016/j.celrep.2023.113390] [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: 04/21/2023] [Revised: 09/10/2023] [Accepted: 10/20/2023] [Indexed: 11/08/2023] Open
Abstract
In the retina, rod and cone pathways mediate visual signals over a billion-fold range in luminance. AII ("A-two") amacrine cells (ACs) receive signals from both pathways via different bipolar cells, enabling AIIs to operate at night and during the day. Previous work has examined luminance-dependent changes in AII gap junction connectivity, but less is known about how surrounding circuitry shapes AII receptive fields across light levels. Here, we report that moderate contrast stimuli elicit surround inhibition in AIIs under all but the dimmest visual conditions, due to actions of horizontal cells and at least two ACs that inhibit presynaptic bipolar cells. Under photopic (daylight) conditions, surround inhibition transforms AII response kinetics, which are inherited by downstream ganglion cells. Ablating neuronal nitric oxide synthase type-1 (nNOS-1) ACs removes AII surround inhibition under mesopic (dusk/dawn), but not photopic, conditions. Our findings demonstrate how multiple layers of neural circuitry interact to encode signals across a wide physiological range.
Collapse
Affiliation(s)
- Amurta Nath
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - William N Grimes
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeffrey S Diamond
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
8
|
Choi J, Li J, Ferdous S, Liang Q, Moffitt JR, Chen R. Spatial organization of the mouse retina at single cell resolution by MERFISH. Nat Commun 2023; 14:4929. [PMID: 37582959 PMCID: PMC10427710 DOI: 10.1038/s41467-023-40674-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 08/07/2023] [Indexed: 08/17/2023] Open
Abstract
The visual signal processing in the retina requires the precise organization of diverse neuronal types working in concert. While single-cell omics studies have identified more than 120 different neuronal subtypes in the mouse retina, little is known about their spatial organization. Here, we generated the single-cell spatial atlas of the mouse retina using multiplexed error-robust fluorescence in situ hybridization (MERFISH). We profiled over 390,000 cells and identified all major cell types and nearly all subtypes through the integration with reference single-cell RNA sequencing (scRNA-seq) data. Our spatial atlas allowed simultaneous examination of nearly all cell subtypes in the retina, revealing 8 previously unknown displaced amacrine cell subtypes and establishing the connection between the molecular classification of many cell subtypes and their spatial arrangement. Furthermore, we identified spatially dependent differential gene expression between subtypes, suggesting the possibility of functional tuning of neuronal types based on location.
Collapse
Affiliation(s)
- Jongsu Choi
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jin Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Salma Ferdous
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Qingnan Liang
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jeffrey R Moffitt
- Program in Cellular and Molecular Medicine, Boston Children's Hospital; Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Rui Chen
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
9
|
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
|
10
|
Meah A, Boodram V, Bucinca-Cupallari F, Lim H. Axonal architecture of the mouse inner retina revealed by second harmonic generation. PNAS NEXUS 2022; 1:pgac160. [PMID: 36106183 PMCID: PMC9463061 DOI: 10.1093/pnasnexus/pgac160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/11/2022] [Indexed: 01/29/2023]
Abstract
We describe a novel method for visualizing the network of axons in the unlabeled fresh wholemount retina. The intrinsic radiation of second harmonic generation (SHG) was utilized to visualize single axons of all major retinal neurons, i.e., photoreceptors, horizontal cells, bipolar cells, amacrine cells, and the retinal ganglion cells. The cell types of SHG+ axons were determined using transgenic GFP/YFP mice. New findings were obtained with retinal SHG imaging: Müller cells do not maintain uniformly polarized microtubules in the processes; SHG+ axons of bipolar cells terminate in the inner plexiform layer (IPL) in a subtype-specific manner; a subset of amacrine cells, presumably the axon-bearing types, emits SHG; and the axon-like neurites of amacrine cells provide a cytoskeletal scaffolding for the IPL stratification. To demonstrate the utility, retinal SHG imaging was applied to testing whether the inner retina is preserved in glaucoma, using DBA/2 mice as a model of glaucoma and DBA/2-Gpnmb+ as the nonglaucomatous control. It was found that the morphology of the inner retina was largely intact in glaucoma and the presynaptic compartments to the retinal ganglion cells were uncompromised. It proves retinal SHG imaging as a promising technology for studying the physiological and diseased retinas in 3D.
Collapse
Affiliation(s)
- Arafat Meah
- Department of Physics and Astronomy, Hunter College, New York, NY 10065, USA
| | - Vinessia Boodram
- Department of Physics and Astronomy, Hunter College, New York, NY 10065, USA
| | - Festa Bucinca-Cupallari
- Department of Physics and Astronomy, Hunter College, New York, NY 10065, USA,The Graduate Centre of the City University of New York, New York, NY 10065, USA
| | | |
Collapse
|
11
|
Xu J, Jo A, DeVries RP, Deniz S, Cherian S, Sunmola I, Song X, Marshall JJ, Gruner KA, Daigle TL, Contractor A, Lerner TN, Zeng H, Zhu Y. Intersectional mapping of multi-transmitter neurons and other cell types in the brain. Cell Rep 2022; 40:111036. [PMID: 35793636 PMCID: PMC9290751 DOI: 10.1016/j.celrep.2022.111036] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 04/04/2022] [Accepted: 06/13/2022] [Indexed: 01/04/2023] Open
Abstract
Recent developments in intersectional strategies have greatly advanced our ability to precisely target brain cell types based on unique co-expression patterns. To accelerate the application of intersectional genetics, we perform a brain-wide characterization of 13 Flp and tTA mouse driver lines and selected seven for further analysis based on expression of vesicular neurotransmitter transporters. Using selective Cre driver lines, we created more than 10 Cre/tTA combinational lines for cell type targeting and circuit analysis. We then used VGLUT-Cre/VGAT-Flp combinational lines to identify and map 30 brain regions containing neurons that co-express vesicular glutamate and gamma-aminobutyric acid (GABA) transporters, followed by tracing their projections with intersectional viral vectors. Focusing on the lateral habenula (LHb) as a target, we identified glutamatergic, GABAergic, or co-glutamatergic/GABAergic innervations from ∼40 brain regions. These data provide an important resource for the future application of intersectional strategies and expand our understanding of the neuronal subtypes in the brain.
Collapse
Affiliation(s)
- Jian Xu
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Andrew Jo
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Raina P DeVries
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL 60637, USA
| | - Sercan Deniz
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Suraj Cherian
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Idris Sunmola
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Xingqi Song
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - John J Marshall
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Katherine A Gruner
- Mouse Histology and Phenotyping Laboratory, Northwestern University, Chicago, IL 60611, USA
| | - Tanya L Daigle
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Anis Contractor
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Talia N Lerner
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Yongling Zhu
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
12
|
Abstract
Retinal circuits transform the pixel representation of photoreceptors into the feature representations of ganglion cells, whose axons transmit these representations to the brain. Functional, morphological, and transcriptomic surveys have identified more than 40 retinal ganglion cell (RGC) types in mice. RGCs extract features of varying complexity; some simply signal local differences in brightness (i.e., luminance contrast), whereas others detect specific motion trajectories. To understand the retina, we need to know how retinal circuits give rise to the diverse RGC feature representations. A catalog of the RGC feature set, in turn, is fundamental to understanding visual processing in the brain. Anterograde tracing indicates that RGCs innervate more than 50 areas in the mouse brain. Current maps connecting RGC types to brain areas are rudimentary, as is our understanding of how retinal signals are transformed downstream to guide behavior. In this article, I review the feature selectivities of mouse RGCs, how they arise, and how they are utilized downstream. Not only is knowledge of the behavioral purpose of RGC signals critical for understanding the retinal contributions to vision; it can also guide us to the most relevant areas of visual feature space. Expected final online publication date for the Annual Review of Vision Science, Volume 8 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Daniel Kerschensteiner
- John F. Hardesty, MD, Department of Ophthalmology and Visual Sciences; Department of Neuroscience; Department of Biomedical Engineering; and Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, Missouri, USA;
| |
Collapse
|
13
|
Genç B, Jara JH, Sanchez SS, Lagrimas AKB, Gözütok Ö, Koçak N, Zhu Y, Hande Özdinler P. Upper motor neurons are a target for gene therapy and UCHL1 is necessary and sufficient to improve cellular integrity of diseased upper motor neurons. Gene Ther 2022; 29:178-192. [PMID: 34853443 PMCID: PMC9018479 DOI: 10.1038/s41434-021-00303-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 12/15/2022]
Abstract
There are no effective cures for upper motor neuron (UMN) diseases, such as amyotrophic lateral sclerosis (ALS), primary lateral sclerosis, and hereditary spastic paraplegia. Here, we show UMN loss occurs independent of spinal motor neuron degeneration and that UMNs are indeed effective cellular targets for gene therapy, which offers a potential solution especially for UMN disease patients. UCHL1 (ubiquitin C-terminal hydrolase-L1) is a deubiquitinating enzyme crucial for maintaining free ubiquitin levels. Corticospinal motor neurons (CSMN, a.k.a UMNs in mice) show early, selective, and profound degeneration in Uchl1nm3419 (UCHL1-/-) mice, which lack all UCHL1 function. When UCHL1 activity is ablated only from spinal motor neurons, CSMN remained intact. However, restoring UCHL1 specifically in CSMN of UCHL1-/- mice via directed gene delivery was sufficient to improve CSMN integrity to the healthy control levels. In addition, when UCHL1 gene was delivered selectively to CSMN that are diseased due to misfolded SOD1 toxicity and TDP-43 pathology via AAV-mediated retrograde transduction, the disease causing misfolded SOD1 and mutant human TDP-43 were reduced in hSOD1G93A and prpTDP-43A315T models, respectively. Diseased CSMN retained their neuronal integrity and cytoarchitectural stability in two different mouse models that represent two distinct causes of neurodegeneration in ALS.
Collapse
Affiliation(s)
- Barış Genç
- Davee Department of Neurology and Clinical Neurological Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Javier H Jara
- Davee Department of Neurology and Clinical Neurological Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Santana S Sanchez
- Davee Department of Neurology and Clinical Neurological Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Amiko K B Lagrimas
- Davee Department of Neurology and Clinical Neurological Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Öge Gözütok
- Davee Department of Neurology and Clinical Neurological Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Nuran Koçak
- Davee Department of Neurology and Clinical Neurological Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Yongling Zhu
- Departments of Ophthalmology and Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - P Hande Özdinler
- Davee Department of Neurology and Clinical Neurological Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
14
|
Liu Y, Zhang FF, Song Y, Wang R, Zhang Q, Shen ZS, Zhang FF, Zhong DY, Wang XH, Guo Q, Tang QY, Zhang Z. The Slack Channel Deletion Causes Mechanical Pain Hypersensitivity in Mice. Front Mol Neurosci 2022; 15:811441. [PMID: 35359569 PMCID: PMC8963359 DOI: 10.3389/fnmol.2022.811441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
The role of the Slack (also known as Slo2.2, KNa1.1, or KCNT1) channel in pain-sensing is still in debate on which kind of pain it regulates. In the present study, we found that the Slack–/– mice exhibited decreased mechanical pain threshold but normal heat and cold pain sensitivity. Subsequently, X-gal staining, in situ hybridization, and immunofluorescence staining revealed high expression of the Slack channel in Isolectin B4 positive (IB4+) neurons in the dorsal root ganglion (DRG) and somatostatin-positive (SOM+) neurons in the spinal cord. Patch-clamp recordings indicated the firing frequency was increased in both small neurons in DRG and spinal SOM+ neurons in the Slack–/– mice whereas no obvious slow afterhyperpolarization was observed in both WT mice and Slack–/– mice. Furthermore, we found Kcnt1 gene expression in spinal SOM+ neurons in Slack–/– mice partially relieved the mechanical pain hypersensitivity of Slack–/– mice and decreased AP firing rates of the spinal SOM+ neurons. Finally, deletion of the Slack channel in spinal SOM+ neurons is sufficient to result in mechanical pain hypersensitivity in mice. In summary, our results suggest the important role of the Slack channel in the regulation of mechanical pain-sensing both in small neurons in DRG and SOM+ neurons in the spinal dorsal horn.
Collapse
Affiliation(s)
- Ye Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Fang-Fang Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Ying Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Ran Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Qi Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Zhong-Shan Shen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Fei-Fei Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Dan-Ya Zhong
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Xiao-Hui Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Qing Guo
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Qiong-Yao Tang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Qiong-Yao Tang,
| | - Zhe Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Zhe Zhang,
| |
Collapse
|
15
|
Mouse Lines with Cre-Mediated Recombination in Retinal Amacrine Cells. eNeuro 2022; 9:ENEURO.0255-21.2021. [PMID: 35045975 PMCID: PMC8856716 DOI: 10.1523/eneuro.0255-21.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 11/21/2022] Open
Abstract
Amacrine cells (ACs) are the most diverse neuronal cell type in the vertebrate retina. Yet little is known about the contribution of ACs to visual processing and retinal disease. A major challenge in evaluating AC function is genetic accessibility. A classic tool of mouse genetics, Cre-mediated recombination, can provide such access. We have screened existing genetically-modified mouse strains and identified multiple candidates that express Cre-recombinase in subsets of retinal ACs. The Cre-expressing mice were crossed to fluorescent-reporter mice to assay Cre expression. In addition, a Cre-dependent fluorescent reporter plasmid was electroporated into the subretinal space of Cre strains. Herein, we report three mouse lines (Tac1::IRES-cre, Camk2a-cre, and Scx-cre) that express Cre recombinase in sub-populations of ACs. In two of these lines, recombination occurred in multiple AC types and a small number of other retinal cell types, while recombination in the Camk2a-cre line appears specific to a morphologically distinct AC. We anticipate that these characterized mouse lines will be valuable tools to the community of researchers who study retinal biology and disease.
Collapse
|
16
|
Pottackal J, Walsh HL, Rahmani P, Zhang K, Justice NJ, Demb JB. Photoreceptive Ganglion Cells Drive Circuits for Local Inhibition in the Mouse Retina. J Neurosci 2021; 41:1489-1504. [PMID: 33397711 PMCID: PMC7896016 DOI: 10.1523/jneurosci.0674-20.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 11/11/2020] [Accepted: 12/18/2020] [Indexed: 12/22/2022] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) exhibit melanopsin-dependent light responses that persist in the absence of rod and cone photoreceptor-mediated input. In addition to signaling anterogradely to the brain, ipRGCs signal retrogradely to intraretinal circuitry via gap junction-mediated electrical synapses with amacrine cells (ACs). However, the targets and functions of these intraretinal signals remain largely unknown. Here, in mice of both sexes, we identify circuitry that enables M5 ipRGCs to locally inhibit retinal neurons via electrical synapses with a nonspiking GABAergic AC. During pharmacological blockade of rod- and cone-mediated input, whole-cell recordings of corticotropin-releasing hormone-expressing (CRH+) ACs reveal persistent visual responses that require both melanopsin expression and gap junctions. In the developing retina, ipRGC-mediated input to CRH+ ACs is weak or absent before eye opening, indicating a primary role for this input in the mature retina (i.e., in parallel with rod- and cone-mediated input). Among several ipRGC types, only M5 ipRGCs exhibit consistent anatomical and physiological coupling to CRH+ ACs. Optogenetic stimulation of local CRH+ ACs directly drives IPSCs in M4 and M5, but not M1-M3, ipRGCs. CRH+ ACs also inhibit M2 ipRGC-coupled spiking ACs, demonstrating direct interaction between discrete networks of ipRGC-coupled interneurons. Together, these results demonstrate a functional role for electrical synapses in translating ipRGC activity into feedforward and feedback inhibition of local retinal circuits.SIGNIFICANCE STATEMENT Melanopsin directly generates light responses in intrinsically photosensitive retinal ganglion cells (ipRGCs). Through gap junction-mediated electrical synapses with retinal interneurons, these uniquely photoreceptive RGCs may also influence the activity and output of neuronal circuits within the retina. Here, we identified and studied an electrical synaptic circuit that, in principle, could couple ipRGC activity to the chemical output of an identified retinal interneuron. Specifically, we found that M5 ipRGCs form electrical synapses with corticotropin-releasing hormone-expressing amacrine cells, which locally release GABA to inhibit specific RGC types. Thus, ipRGCs are poised to influence the output of diverse retinal circuits via electrical synapses with interneurons.
Collapse
Affiliation(s)
| | | | | | | | - Nicholas J Justice
- Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas 77030
| | - Jonathan B Demb
- Interdepartmental Neuroscience Program
- Department of Ophthalmology and Visual Science
- Department of Cellular and Molecular Physiology
- Department of Neuroscience, Yale University, New Haven, Connecticut 06511
| |
Collapse
|
17
|
Optimized culture of retinal ganglion cells and amacrine cells from adult mice. PLoS One 2020; 15:e0242426. [PMID: 33284815 PMCID: PMC7721191 DOI: 10.1371/journal.pone.0242426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 11/02/2020] [Indexed: 01/22/2023] Open
Abstract
Cell culture is widely utilized to study the cellular and molecular biology of different neuronal cell populations. Current techniques to study enriched neurons in vitro are primarily limited to embryonic/neonatal animals and induced pluripotent stem cells (iPSCs). Although the use of these cultures is valuable, the accessibility of purified primary adult neuronal cultures would allow for improved assessment of certain neurological diseases and pathways at the cellular level. Using a modified 7-step immunopanning technique to isolate for retinal ganglion cells (RGCs) and amacrine cells (ACs) from adult mouse retinas, we have successfully developed a model of neuronal culture that maintains for at least one week. Isolations of Thy1.2+ cells are enriched for RGCs, with the isolation cell yield being congruent to the theoretical yield of RGCs in a mouse retina. ACs of two different populations (CD15+ and CD57+) can also be isolated. The populations of these three adult neurons in culture are healthy, with neurite outgrowths in some cases greater than 500μm in length. Optimization of culture conditions for RGCs and CD15+ cells revealed that neuronal survival and the likelihood of neurite outgrowth respond inversely to different culture media. Serially diluted concentrations of puromycin decreased cultured adult RGCs in a dose-dependent manner, demonstrating the potential usefulness of these adult neuronal cultures in screening assays. This novel culture system can be used to model in vivo neuronal behaviors. Studies can now be expanded in conjunction with other methodologies to study the neurobiology of function, aging, and diseases.
Collapse
|
18
|
Parmhans N, Fuller AD, Nguyen E, Chuang K, Swygart D, Wienbar SR, Lin T, Kozmik Z, Dong L, Schwartz GW, Badea TC. Identification of retinal ganglion cell types and brain nuclei expressing the transcription factor Brn3c/Pou4f3 using a Cre recombinase knock-in allele. J Comp Neurol 2020; 529:1926-1953. [PMID: 33135183 DOI: 10.1002/cne.25065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022]
Abstract
Members of the POU4F/Brn3 transcription factor family have an established role in the development of retinal ganglion cell (RGCs) types, the main transducers of visual information from the mammalian eye to the brain. Our previous work using sparse random recombination of a conditional knock-in reporter allele expressing alkaline phosphatase (AP) and intersectional genetics had identified three types of Brn3c positive (Brn3c+ ) RGCs. Here, we describe a novel Brn3cCre mouse allele generated by serial Dre to Cre recombination and use it to explore the expression overlap of Brn3c with Brn3a and Brn3b and the dendritic arbor morphologies and visual stimulus response properties of Brn3c+ RGC types. Furthermore, we explore brain nuclei that express Brn3c or receive input from Brn3c+ neurons. Our analysis reveals a much larger number of Brn3c+ RGCs and more diverse set of RGC types than previously reported. Most RGCs expressing Brn3c during development are still Brn3c positive in the adult, and all express Brn3a while only about half express Brn3b. Genetic Brn3c-Brn3b intersection reveals an area of increased RGC density, extending from dorsotemporal to ventrolateral across the retina and overlapping with the mouse binocular field of view. In addition, we report a Brn3c+ RGC projection to the thalamic reticular nucleus, a visual nucleus that was not previously shown to receive retinal input. Furthermore, Brn3c+ neurons highlight a previously unknown subdivision of the deep mesencephalic nucleus. Thus, our newly generated allele provides novel biological insights into RGC type classification, brain connectivity, and cytoarchitectonic.
Collapse
Affiliation(s)
- Nadia Parmhans
- Retinal Circuit Development and Genetics Unit, Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, NIH, Bethesda, Maryland, USA
| | - Anne Drury Fuller
- Retinal Circuit Development and Genetics Unit, Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, NIH, Bethesda, Maryland, USA
| | - Eileen Nguyen
- Retinal Circuit Development and Genetics Unit, Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, NIH, Bethesda, Maryland, USA
| | - Katherine Chuang
- Retinal Circuit Development and Genetics Unit, Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, NIH, Bethesda, Maryland, USA
| | - David Swygart
- Departments of Ophthalmology and Physiology Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Sophia Rose Wienbar
- Departments of Ophthalmology and Physiology Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Tyger Lin
- Retinal Circuit Development and Genetics Unit, Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, NIH, Bethesda, Maryland, USA
| | - Zbynek Kozmik
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Lijin Dong
- Genetic Engineering Facility, National Eye Institute, NIH, Bethesda, Maryland, USA
| | - Gregory William Schwartz
- Departments of Ophthalmology and Physiology Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Tudor Constantin Badea
- Retinal Circuit Development and Genetics Unit, Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, NIH, Bethesda, Maryland, USA
| |
Collapse
|
19
|
Kerstein PC, Leffler J, Sivyer B, Taylor WR, Wright KM. Gbx2 Identifies Two Amacrine Cell Subtypes with Distinct Molecular, Morphological, and Physiological Properties. Cell Rep 2020; 33:108382. [PMID: 33207201 PMCID: PMC7713908 DOI: 10.1016/j.celrep.2020.108382] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/21/2020] [Accepted: 10/22/2020] [Indexed: 01/21/2023] Open
Abstract
Our understanding of nervous system function is limited by our ability to identify and manipulate neuronal subtypes within intact circuits. We show that the Gbx2CreERT2-IRES-EGFP mouse line labels two amacrine cell (AC) subtypes in the mouse retina that have distinct morphological, physiological, and molecular properties. Using a combination of RNA-seq, genetic labeling, and patch clamp recordings, we show that one subtype is GABAergic that receives excitatory input from On bipolar cells. The other population is a non-GABAergic, non-glycinergic (nGnG) AC subtype that lacks the expression of standard neurotransmitter markers. Gbx2+ nGnG ACs have smaller, asymmetric dendritic arbors that receive excitatory input from both On and Off bipolar cells. Gbx2+ nGnG ACs also exhibit spatially restricted tracer coupling to bipolar cells (BCs) through gap junctions. This study identifies a genetic tool for investigating the two distinct AC subtypes, and it provides a model for studying synaptic communication and visual circuit function. Investigations into neural circuit development and function are limited by the lack of genetic tools to label and perturb individual neuronal subtypes. Using the Gbx2CreERT2 mouse line, Kerstein et al. identify two amacrine cell subtypes in the mouse retina and explore their distinct molecular, morphological, and physiological characteristics.
Collapse
Affiliation(s)
- Patrick C Kerstein
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Joseph Leffler
- School of Optometry and Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Neuroscience Graduate Program, Oregon Health and Science University, Portland, OR 97239, USA
| | - Benjamin Sivyer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Portland, OR 97239, USA; Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR 97239, USA
| | - W Rowland Taylor
- School of Optometry and Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kevin M Wright
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA; Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
20
|
Xu J, DeVries SH, Zhu Y. Quantification of Adeno-Associated Virus with Safe Nucleic Acid Dyes. Hum Gene Ther 2020; 31:1086-1099. [PMID: 32368927 DOI: 10.1089/hum.2020.063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Adeno-associated virus (AAV) is the most commonly used viral vector for both biological and gene therapeutic applications. Although many methods have been developed to measure quantity attributes of AAV, they are often technically challenging and time-consuming. Here, we report a method to titer AAV with GelGreen® dye, a safe green fluorescence nucleic acid dye recently engineered by Biotium company (Fremont, CA). This method, hereinafter referred to as GelGreen method, provides a fast (∼30 min) and reliable strategy for AAV titration. To validate GelGreen method, we measured genome titer of an AAV reference material AAV8RSM and compared our titration results with those determined by Reference Material Working Group (ARMWG). We showed that GelGreen results and capsid enzyme-linked immunosorbent assay results are comparable with each other. We also showed that GelRed® dye, a red fluorescence dye from Biotium, can be used to directly "visualize" AAV genome titer on a conventional gel imager, presenting an especially direct approach to estimate viral quantity. Finally, we showed that GelGreen and GelRed dyes can also be used to quantify self-complementary AAV (scAAV) and crudely purified AAV samples. In summary, we described a technique to titer AAV by using new generation of safe DNA dyes. This technique is simple, safe, reliable, and cost efficient. It has potential to be broadly applied for quantifying and normalizing AAV viral vectors.
Collapse
Affiliation(s)
- Jian Xu
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Steven H DeVries
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yongling Zhu
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
21
|
Yan W, Laboulaye MA, Tran NM, Whitney IE, Benhar I, Sanes JR. Mouse Retinal Cell Atlas: Molecular Identification of over Sixty Amacrine Cell Types. J Neurosci 2020; 40:5177-5195. [PMID: 32457074 PMCID: PMC7329304 DOI: 10.1523/jneurosci.0471-20.2020] [Citation(s) in RCA: 192] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/07/2020] [Accepted: 05/13/2020] [Indexed: 02/01/2023] Open
Abstract
Amacrine cells (ACs) are a diverse class of interneurons that modulate input from photoreceptors to retinal ganglion cells (RGCs), rendering each RGC type selectively sensitive to particular visual features, which are then relayed to the brain. While many AC types have been identified morphologically and physiologically, they have not been comprehensively classified or molecularly characterized. We used high-throughput single-cell RNA sequencing to profile >32,000 ACs from mice of both sexes and applied computational methods to identify 63 AC types. We identified molecular markers for each type and used them to characterize the morphology of multiple types. We show that they include nearly all previously known AC types as well as many that had not been described. Consistent with previous studies, most of the AC types expressed markers for the canonical inhibitory neurotransmitters GABA or glycine, but several expressed neither or both. In addition, many expressed one or more neuropeptides, and two expressed glutamatergic markers. We also explored transcriptomic relationships among AC types and identified transcription factors expressed by individual or multiple closely related types. Noteworthy among these were Meis2 and Tcf4, expressed by most GABAergic and most glycinergic types, respectively. Together, these results provide a foundation for developmental and functional studies of ACs, as well as means for genetically accessing them. Along with previous molecular, physiological, and morphologic analyses, they establish the existence of at least 130 neuronal types and nearly 140 cell types in the mouse retina.SIGNIFICANCE STATEMENT The mouse retina is a leading model for analyzing the development, structure, function, and pathology of neural circuits. A complete molecular atlas of retinal cell types provides an important foundation for these studies. We used high-throughput single-cell RNA sequencing to characterize the most heterogeneous class of retinal interneurons, amacrine cells, identifying 63 distinct types. The atlas includes types identified previously as well as many novel types. We provide evidence for the use of multiple neurotransmitters and neuropeptides, and identify transcription factors expressed by groups of closely related types. Combining these results with those obtained previously, we proposed that the mouse retina contains ∼130 neuronal types and is therefore comparable in complexity to other regions of the brain.
Collapse
Affiliation(s)
- Wenjun Yan
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138
| | - Mallory A Laboulaye
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138
| | - Nicholas M Tran
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138
| | - Irene E Whitney
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138
| | - Inbal Benhar
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138
| |
Collapse
|
22
|
Chen D, Ren K, Liu H, Mao H, Li Z, Mo H, Xie S, Shi Y, Chen Q, Wang W. A Whole-Brain Cell-Type-Specific Sparse Neuron Labeling Method and Its Application in a Shank3 Autistic Mouse Model. Front Cell Neurosci 2020; 14:145. [PMID: 32581718 PMCID: PMC7291601 DOI: 10.3389/fncel.2020.00145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
Single neurons, as the basic unit of the brain, consist of a cell body and processes, including dendrites and axons. Even neurons of the same type show various subtle process characteristics to fit into the diverse neural circuits. Different cell types of neurons form complicated circuits in the brain. Therefore, detailed neuronal morphology is required to understand normal neuronal function and pathological mechanisms, such as those that occur in autism. Here, we developed a strategy to sparsely label the same type of neurons throughout the whole brain and tested its application in an autistic animal model—Shank3 knockout (KO) mice. To achieve this, we designed an adeno-associated virus (AAV) that expresses Cre recombinase-dependent regular and membrane-targeted enhanced green fluorescent protein (EGFP) under a human synapsin 1 promoter and verified it in several Cre transgenic mice. We could sparsely label the projection neurons in multiple brain areas by retro-ocular injection of the virus into CaMKIIα-Cre mice. Then, we analyzed the morphology of the projection neurons in Shank3 KO mice with this method. We found differential dendritic complexity and dendritic spine changes in projection neurons in Shank3 KO mice crossed with CaMKIIα-Cre mice compared with littermate control mice in the striatum, cortex, and hippocampus. By combining this method with various Cre mouse lines crossed with mouse models of disease, we can screen the morphological traits of distinct types of neurons throughout the whole brain that will help us to understand the exact role of the specific cell types of neurons not only in autism spectrum disorder (ASD) mouse models but also in other psychiatric disorder mouse models.
Collapse
Affiliation(s)
- Di Chen
- Institute of Neuroscience, Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Keke Ren
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Haiying Liu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Honghui Mao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Zongyan Li
- Institute of Neuroscience, Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huiming Mo
- Department of Physiology, Medical College of Yan'an University, Yan'an, China
| | - Shengjun Xie
- Institute of Neuroscience, Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yiwu Shi
- Institute of Neuroscience, Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qian Chen
- Institute of Neuroscience, Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
23
|
Park SJH, Lieberman EE, Ke JB, Rho N, Ghorbani P, Rahmani P, Jun NY, Lee HL, Kim IJ, Briggman KL, Demb JB, Singer JH. Connectomic analysis reveals an interneuron with an integral role in the retinal circuit for night vision. eLife 2020; 9:e56077. [PMID: 32412412 PMCID: PMC7228767 DOI: 10.7554/elife.56077] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 04/27/2020] [Indexed: 12/28/2022] Open
Abstract
Night vision in mammals depends fundamentally on rod photoreceptors and the well-studied rod bipolar (RB) cell pathway. The central neuron in this pathway, the AII amacrine cell (AC), exhibits a spatially tuned receptive field, composed of an excitatory center and an inhibitory surround, that propagates to ganglion cells, the retina's projection neurons. The circuitry underlying the surround of the AII, however, remains unresolved. Here, we combined structural, functional and optogenetic analyses of the mouse retina to discover that surround inhibition of the AII depends primarily on a single interneuron type, the NOS-1 AC: a multistratified, axon-bearing GABAergic cell, with dendrites in both ON and OFF synaptic layers, but with a pure ON (depolarizing) response to light. Our study demonstrates generally that novel neural circuits can be identified from targeted connectomic analyses and specifically that the NOS-1 AC mediates long-range inhibition during night vision and is a major element of the RB pathway.
Collapse
Affiliation(s)
- Silvia JH Park
- Department of Ophthalmology & Visual Science, Yale UniversityNew HavenUnited States
| | - Evan E Lieberman
- Department of Biology, University of MarylandCollege ParkUnited States
| | - Jiang-Bin Ke
- Department of Biology, University of MarylandCollege ParkUnited States
| | - Nao Rho
- Department of Biology, University of MarylandCollege ParkUnited States
| | - Padideh Ghorbani
- Department of Biology, University of MarylandCollege ParkUnited States
| | - Pouyan Rahmani
- Department of Ophthalmology & Visual Science, Yale UniversityNew HavenUnited States
| | - Na Young Jun
- Department of Ophthalmology & Visual Science, Yale UniversityNew HavenUnited States
| | - Hae-Lim Lee
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
| | - In-Jung Kim
- Department of Ophthalmology & Visual Science, Yale UniversityNew HavenUnited States
| | - Kevin L Briggman
- Circuit Dynamics and Connectivity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaUnited States
| | - Jonathan B Demb
- Department of Ophthalmology & Visual Science, Yale UniversityNew HavenUnited States
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Neuroscience, Yale UniversityNew HavenUnited States
| | - Joshua H Singer
- Department of Biology, University of MarylandCollege ParkUnited States
| |
Collapse
|
24
|
Yamakawa M, Santosa SM, Chawla N, Ivakhnitskaia E, Del Pino M, Giakas S, Nadel A, Bontu S, Tambe A, Guo K, Han KY, Cortina MS, Yu C, Rosenblatt MI, Chang JH, Azar DT. Transgenic models for investigating the nervous system: Currently available neurofluorescent reporters and potential neuronal markers. Biochim Biophys Acta Gen Subj 2020; 1864:129595. [PMID: 32173376 DOI: 10.1016/j.bbagen.2020.129595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/24/2020] [Accepted: 03/03/2020] [Indexed: 02/06/2023]
Abstract
Recombinant DNA technologies have enabled the development of transgenic animal models for use in studying a myriad of diseases and biological states. By placing fluorescent reporters under the direct regulation of the promoter region of specific marker proteins, these models can localize and characterize very specific cell types. One important application of transgenic species is the study of the cytoarchitecture of the nervous system. Neurofluorescent reporters can be used to study the structural patterns of nerves in the central or peripheral nervous system in vivo, as well as phenomena involving embryologic or adult neurogenesis, injury, degeneration, and recovery. Furthermore, crucial molecular factors can also be screened via the transgenic approach, which may eventually play a major role in the development of therapeutic strategies against diseases like Alzheimer's or Parkinson's. This review describes currently available reporters and their uses in the literature as well as potential neural markers that can be leveraged to create additional, robust transgenic models for future studies.
Collapse
Affiliation(s)
- Michael Yamakawa
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Samuel M Santosa
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Neeraj Chawla
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Evguenia Ivakhnitskaia
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Matthew Del Pino
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Sebastian Giakas
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Arnold Nadel
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Sneha Bontu
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Arjun Tambe
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Kai Guo
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Kyu-Yeon Han
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Maria Soledad Cortina
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Charles Yu
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Mark I Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America.
| | - Dimitri T Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America.
| |
Collapse
|
25
|
Cui LJ, Chen WH, Liu AL, Han X, Jiang SX, Yuan F, Zhong YM, Yang XL, Weng SJ. nGnG Amacrine Cells and Brn3b-negative M1 ipRGCs are Specifically Labeled in the ChAT-ChR2-EYFP Mouse. Invest Ophthalmol Vis Sci 2020; 61:14. [PMID: 32049344 PMCID: PMC7326507 DOI: 10.1167/iovs.61.2.14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Experimental access to specific cell subtypes is essential for deciphering the complexity of retinal networks. Here, we characterized the selective labeling, caused by ectopic transgene expression, of two atypical retinal neurons in the ChAT-Channelrhodopsin-2 (ChR2)-EYFP mouse. Methods Retinal sections and flat-mounts were prepared for double-staining immunohistochemistry with antibodies against EYFP and various neuronal markers. Sagittal/coronal brain slices were made to visualize EYFP signals in central nuclei. Whole-cell recordings were conducted to test the functionality of ChR2. Results Two populations of EYFP-positive retinal cells were observed. The inner nuclear layer (INL)-located one (type I cell) distributed regularly throughout the entire retina, whereas the ganglion cell layer (GCL)-residing one (type II cell) was restricted ventrally. None of them was cholinergic, as evidenced by the complete absence of ChAT immunoreactivity. Type I cells were immunolabeled by the amacrine marker syntaxin. However, the vast majority of them were neither positive to GABA/GAD65, nor to GlyT1/glycine, suggesting that they were non-GABAergic non-glycinergic amacrine cells (nGnG ACs), which was confirmed by double-labeling with the nGnG AC marker PPP1R17. Type II cells were immunopositive to melanopsin, but not to Brn3a or Brn3b. They possessed dendrites stratifying in the outermost inner plexiform layer (IPL) and axons projecting to the suprachiasmatic nucleus (SCN) rather than the olivary pretectal nucleus (OPN), suggesting that they belonged to a Brn3b-negative subset of M1-type intrinsically photosensitive retinal ganglion cells (ipRGCs). Glutamatergic transmission-independent photocurrents were elicited in EYFP-positive cells, indicating the functional expression of ChR2. Conclusions The ChAT-ChR2-EYFP retina exhibits ectopic, but functional, transgene expression in nGnG ACs and SCN-innervating M1 ipRGCs, thus providing an ideal tool to achieve efficient labeling and optogenetic manipulation of these cells.
Collapse
|
26
|
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
|
27
|
Suzuki T, Morimoto N, Akaike A, Osakada F. Multiplex Neural Circuit Tracing With G-Deleted Rabies Viral Vectors. Front Neural Circuits 2020; 13:77. [PMID: 31998081 PMCID: PMC6967742 DOI: 10.3389/fncir.2019.00077] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 11/14/2019] [Indexed: 12/26/2022] Open
Abstract
Neural circuits interconnect to organize large-scale networks that generate perception, cognition, memory, and behavior. Information in the nervous system is processed both through parallel, independent circuits and through intermixing circuits. Analyzing the interaction between circuits is particularly indispensable for elucidating how the brain functions. Monosynaptic circuit tracing with glycoprotein (G) gene-deleted rabies viral vectors (RVΔG) comprises a powerful approach for studying the structure and function of neural circuits. Pseudotyping of RVΔG with the foreign envelope EnvA permits expression of transgenes such as fluorescent proteins, genetically-encoded sensors, or optogenetic tools in cells expressing TVA, a cognate receptor for EnvA. Trans-complementation with rabies virus glycoproteins (RV-G) enables trans-synaptic labeling of input neurons directly connected to the starter neurons expressing both TVA and RV-G. However, it remains challenging to simultaneously map neuronal connections from multiple cell populations and their interactions between intermixing circuits solely with the EnvA/TVA-mediated RV tracing system in a single animal. To overcome this limitation, here, we multiplexed RVΔG circuit tracing by optimizing distinct viral envelopes (oEnvX) and their corresponding receptors (oTVX). Based on the EnvB/TVB and EnvE/DR46-TVB systems derived from the avian sarcoma leukosis virus (ASLV), we developed optimized TVB receptors with lower or higher affinity (oTVB-L or oTVB-H) and the chimeric envelope oEnvB, as well as an optimized TVE receptor with higher affinity (oTVE-H) and its chimeric envelope oEnvE. We demonstrated independence of RVΔG infection between the oEnvA/oTVA, oEnvB/oTVB, and oEnvE/oTVE systems and in vivo proof-of-concept for multiplex circuit tracing from two distinct classes of layer 5 neurons targeting either other cortical or subcortical areas. We also successfully labeled common input of the lateral geniculate nucleus to both cortico-cortical layer 5 neurons and inhibitory neurons of the mouse V1 with multiplex RVΔG tracing. These oEnvA/oTVA, oEnvB/oTVB, and oEnvE/oTVE systems allow for differential labeling of distinct circuits to uncover the mechanisms underlying parallel processing through independent circuits and integrated processing through interaction between circuits in the brain.
Collapse
Affiliation(s)
- Toshiaki Suzuki
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Nao Morimoto
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan.,Laboratory of Neural Information Processing, Institute for Advanced Research, Nagoya University, Nagoya, Japan
| | - Akinori Akaike
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Fumitaka Osakada
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan.,Laboratory of Neural Information Processing, Institute for Advanced Research, Nagoya University, Nagoya, Japan.,Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Nagoya, Japan.,PRESTO/CREST, Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
28
|
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
|
29
|
Kiyama T, Long Y, Chen CK, Whitaker CM, Shay A, Wu H, Badea TC, Mohsenin A, Parker-Thornburg J, Klein WH, Mills SL, Massey SC, Mao CA. Essential Roles of Tbr1 in the Formation and Maintenance of the Orientation-Selective J-RGCs and a Group of OFF-Sustained RGCs in Mouse. Cell Rep 2019; 27:900-915.e5. [PMID: 30995485 PMCID: PMC6542366 DOI: 10.1016/j.celrep.2019.03.077] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 02/10/2019] [Accepted: 03/20/2019] [Indexed: 01/27/2023] Open
Abstract
In the mouse retina, more than 30 retinal ganglion cell (RGC) subtypes have been classified based on a combined metric of morphological and functional characteristics. RGCs arise from a common pool of retinal progenitor cells during embryonic stages and differentiate into mature subtypes in adult retinas. However, the cellular and molecular mechanisms controlling formation and maturation of such remarkable cellular diversity remain unknown. Here, we demonstrate that T-box transcription factor T-brain 1 (Tbr1) is expressed in two groups of morphologically and functionally distinct RGCs: the orientation-selective J-RGCs and a group of OFF-sustained RGCs with symmetrical dendritic arbors. When Tbr1 is genetically ablated during retinal development, these two RGC groups cannot develop. Ectopically expressing Tbr1 in M4 ipRGCs during development alters dendritic branching and density but not the inner plexiform layer stratification level. Our data indicate that Tbr1 plays critical roles in regulating the formation and dendritic morphogenesis of specific RGC types.
Collapse
Affiliation(s)
- Takae Kiyama
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Ye Long
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Ching-Kang Chen
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Christopher M Whitaker
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Allison Shay
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hongyu Wu
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Tudor C Badea
- National Eye Institute, NIH, Bethesda, MD 20892, USA
| | - Amir Mohsenin
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA; Robert Cizik Eye Clinic, Houston, TX 77030, USA
| | - Jan Parker-Thornburg
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - William H Klein
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stephen L Mills
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Stephen C Massey
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Chai-An Mao
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA.
| |
Collapse
|
30
|
Hori T, Fukutome M, Koike C. Adeno Associated Virus (AAV) as a Tool for Clinical and Experimental Delivery of Target Genes into the Mammalian Retina. Biol Pharm Bull 2019; 42:343-347. [DOI: 10.1248/bpb.b18-00913] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Tesshu Hori
- Laboratory for Systems Neuroscience and Developmental Biology, College of Pharmaceutical Sciences, Ritsumeikan University
| | | | - Chieko Koike
- Laboratory for Systems Neuroscience and Developmental Biology, College of Pharmaceutical Sciences, Ritsumeikan University
- Graduate School of Life Sciences, Ritsumeikan University
- Center for Systems Vision Science, Research Organization of Science and Technology, Ritsumeikan University
| |
Collapse
|
31
|
Kannan M, Vasan G, Pieribone VA. Optimizing Strategies for Developing Genetically Encoded Voltage Indicators. Front Cell Neurosci 2019; 13:53. [PMID: 30863283 PMCID: PMC6399427 DOI: 10.3389/fncel.2019.00053] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 02/04/2019] [Indexed: 01/23/2023] Open
Abstract
Genetically encoded optical indicators of neuronal activity enable unambiguous recordings of input-output activity patterns from identified cells in intact circuits. Among them, genetically encoded voltage indicators (GEVIs) offer additional advantages over calcium indicators as they are direct sensors of membrane potential and can adeptly report subthreshold events and hyperpolarization. Here, we outline the major GEVI designs and give an account of properties that need to be carefully optimized during indicator engineering. While designing the ideal GEVI, one should keep in mind aspects such as membrane localization, signal size, signal-to-noise ratio, kinetics and voltage dependence of optical responses. Using ArcLight and derivatives as prototypes, we delineate how a probe should be optimized for the former properties and developed along other areas in a need-based manner. Finally, we present an overview of the GEVI engineering process and lend an insight into their discovery, delivery and diagnosis.
Collapse
Affiliation(s)
- Madhuvanthi Kannan
- The John B. Pierce Laboratory, New Haven, CT, United States.,Department of Cellular and Molecular Physiology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Ganesh Vasan
- The John B. Pierce Laboratory, New Haven, CT, United States.,Department of Cellular and Molecular Physiology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Vincent A Pieribone
- The John B. Pierce Laboratory, New Haven, CT, United States.,Department of Cellular and Molecular Physiology, Yale School of Medicine, Yale University, New Haven, CT, United States.,Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
32
|
Román Rosón M, Bauer Y, Kotkat AH, Berens P, Euler T, Busse L. Mouse dLGN Receives Functional Input from a Diverse Population of Retinal Ganglion Cells with Limited Convergence. Neuron 2019; 102:462-476.e8. [PMID: 30799020 DOI: 10.1016/j.neuron.2019.01.040] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 11/08/2018] [Accepted: 01/17/2019] [Indexed: 10/27/2022]
Abstract
Mouse vision is based on the parallel output of more than 30 functional types of retinal ganglion cells (RGCs). Little is known about how representations of visual information change between retina and dorsolateral geniculate nucleus (dLGN) of the thalamus, the main relay between retina and cortex. Here, we functionally characterized responses of retrogradely labeled dLGN-projecting RGCs and dLGN neurons to the same set of visual stimuli. We found that many of the previously identified functional RGC types innervate dLGN, which maintained a high degree of functional diversity. Using a linear model to assess functional connectivity between RGC types and dLGN neurons, we found that responses of dLGN neurons could be predicted as linear combination of inputs from on average five RGC types, but only two of those had the strongest functional impact. Thus, mouse dLGN receives functional input from a diverse population of RGC types with limited convergence.
Collapse
Affiliation(s)
- Miroslav Román Rosón
- Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Institute for Ophthalmic Research, University Hospital Tübingen, 72076 Tübingen, Germany; Division of Neurobiology, Department Biology II, LMU Munich, 82151 Munich, Germany; Graduate School of Neural & Behavioural Sciences, International Max Planck Research School, University of Tübingen, 72074 Tübingen, Germany
| | - Yannik Bauer
- Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Division of Neurobiology, Department Biology II, LMU Munich, 82151 Munich, Germany; Graduate School of Systemic Neuroscience (GSN), LMU Munich, 82151 Munich, Germany
| | - Ann H Kotkat
- Division of Neurobiology, Department Biology II, LMU Munich, 82151 Munich, Germany; ENB Elite Master of Science Program in Neuroengineering, Technical University of Munich, 80333 Munich, Germany
| | - Philipp Berens
- Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Institute for Ophthalmic Research, University Hospital Tübingen, 72076 Tübingen, Germany; Bernstein Centre for Computational Neuroscience, 72076 Tübingen, Germany.
| | - Thomas Euler
- Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Institute for Ophthalmic Research, University Hospital Tübingen, 72076 Tübingen, Germany; Bernstein Centre for Computational Neuroscience, 72076 Tübingen, Germany.
| | - Laura Busse
- Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Division of Neurobiology, Department Biology II, LMU Munich, 82151 Munich, Germany; Bernstein Centre for Computational Neuroscience, 72076 Tübingen, Germany; Bernstein Centre for Computational Neuroscience, 82151 Munich, Germany.
| |
Collapse
|
33
|
Dedic N, Chen A, Deussing JM. The CRF Family of Neuropeptides and their Receptors - Mediators of the Central Stress Response. Curr Mol Pharmacol 2018; 11:4-31. [PMID: 28260504 PMCID: PMC5930453 DOI: 10.2174/1874467210666170302104053] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 11/26/2015] [Accepted: 08/03/2016] [Indexed: 12/12/2022]
Abstract
Background: Dysregulated stress neurocircuits, caused by genetic and/or environmental changes, underlie the development of many neuropsychiatric disorders. Corticotropin-releasing factor (CRF) is the major physiological activator of the hypothalamic-pituitary-adrenal (HPA) axis and conse-quently a primary regulator of the mammalian stress response. Together with its three family members, urocortins (UCNs) 1, 2, and 3, CRF integrates the neuroendocrine, autonomic, metabolic and behavioral responses to stress by activating its cognate receptors CRFR1 and CRFR2. Objective: Here we review the past and current state of the CRF/CRFR field, ranging from pharmacologi-cal studies to genetic mouse models and virus-mediated manipulations. Results: Although it is well established that CRF/CRFR1 signaling mediates aversive responses, includ-ing anxiety and depression-like behaviors, a number of recent studies have challenged this viewpoint by revealing anxiolytic and appetitive properties of specific CRF/CRFR1 circuits. In contrast, the UCN/CRFR2 system is less well understood and may possibly also exert divergent functions on physiol-ogy and behavior depending on the brain region, underlying circuit, and/or experienced stress conditions. Conclusion: A plethora of available genetic tools, including conventional and conditional mouse mutants targeting CRF system components, has greatly advanced our understanding about the endogenous mecha-nisms underlying HPA system regulation and CRF/UCN-related neuronal circuits involved in stress-related behaviors. Yet, the detailed pathways and molecular mechanisms by which the CRF/UCN-system translates negative or positive stimuli into the final, integrated biological response are not completely un-derstood. The utilization of future complementary methodologies, such as cell-type specific Cre-driver lines, viral and optogenetic tools will help to further dissect the function of genetically defined CRF/UCN neurocircuits in the context of adaptive and maladaptive stress responses.
Collapse
Affiliation(s)
- Nina Dedic
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr, 2-10, 80804 Munich. Germany
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr, 2-10, 80804 Munich. Germany
| | - Jan M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr, 2-10, 80804 Munich. Germany
| |
Collapse
|
34
|
Jacoby J, Nath A, Jessen ZF, Schwartz GW. A Self-Regulating Gap Junction Network of Amacrine Cells Controls Nitric Oxide Release in the Retina. Neuron 2018; 100:1149-1162.e5. [PMID: 30482690 PMCID: PMC6317889 DOI: 10.1016/j.neuron.2018.09.047] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/28/2018] [Accepted: 09/25/2018] [Indexed: 01/31/2023]
Abstract
Neuromodulators regulate circuits throughout the nervous system, and revealing the cell types and stimulus conditions controlling their release is vital to understanding their function. The effects of the neuromodulator nitric oxide (NO) have been studied in many circuits, including in the vertebrate retina, where it regulates synaptic release, gap junction coupling, and blood vessel dilation, but little is known about the cells that release NO. We show that a single type of amacrine cell (AC) controls NO release in the inner retina, and we report its light responses, electrical properties, and calcium dynamics. We discover that this AC forms a dense gap junction network and that the strength of electrical coupling in the network is regulated by light through NO. A model of the network offers insights into the biophysical specializations leading to auto-regulation of NO release within the network.
Collapse
Affiliation(s)
- Jason Jacoby
- Department of Ophthalmology, Northwestern University, Chicago, IL, USA
| | - Amurta Nath
- Interdepartmental Neuroscience Program, Northwestern University, Chicago, IL, USA; Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL, USA
| | - Zachary F Jessen
- Medical Scientist Training Program, Northwestern University, Chicago, IL, USA
| | - Gregory W Schwartz
- Department of Ophthalmology, Northwestern University, Chicago, IL, USA; Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
35
|
GABA release selectively regulates synapse development at distinct inputs on direction-selective retinal ganglion cells. Proc Natl Acad Sci U S A 2018; 115:E12083-E12090. [PMID: 30509993 DOI: 10.1073/pnas.1803490115] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Synaptic inhibition controls a neuron's output via functionally distinct inputs at two subcellular compartments, the cell body and the dendrites. It is unclear whether the assembly of these distinct inhibitory inputs can be regulated independently by neurotransmission. In the mammalian retina, γ-aminobutyric acid (GABA) release from starburst amacrine cells (SACs) onto the dendrites of on-off direction-selective ganglion cells (ooDSGCs) is essential for directionally selective responses. We found that ooDSGCs also receive GABAergic input on their somata from other amacrine cells (ACs), including ACs containing the vasoactive intestinal peptide (VIP). When net GABAergic transmission is reduced, somatic, but not dendritic, GABAA receptor clusters on the ooDSGC increased in number and size. Correlative fluorescence imaging and serial electron microscopy revealed that these enlarged somatic receptor clusters are localized to synapses. By contrast, selectively blocking vesicular GABA release from either SACs or VIP ACs did not alter dendritic or somatic receptor distributions on the ooDSGCs, showing that neither SAC nor VIP AC GABA release alone is required for the development of inhibitory synapses in ooDSGCs. Furthermore, a reduction in net GABAergic transmission, but not a selective reduction from SACs, increased excitatory drive onto ooDSGCs. This increased excitation may drive a homeostatic increase in ooDSGC somatic GABAA receptors. Differential regulation of GABAA receptors on the ooDSGC's soma and dendrites could facilitate homeostatic control of the ooDSGC's output while enabling the assembly of the GABAergic connectivity underlying direction selectivity to be indifferent to altered transmission.
Collapse
|
36
|
Deussing JM, Chen A. The Corticotropin-Releasing Factor Family: Physiology of the Stress Response. Physiol Rev 2018; 98:2225-2286. [DOI: 10.1152/physrev.00042.2017] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The physiological stress response is responsible for the maintenance of homeostasis in the presence of real or perceived challenges. In this function, the brain activates adaptive responses that involve numerous neural circuits and effector molecules to adapt to the current and future demands. A maladaptive stress response has been linked to the etiology of a variety of disorders, such as anxiety and mood disorders, eating disorders, and the metabolic syndrome. The neuropeptide corticotropin-releasing factor (CRF) and its relatives, the urocortins 1–3, in concert with their receptors (CRFR1, CRFR2), have emerged as central components of the physiological stress response. This central peptidergic system impinges on a broad spectrum of physiological processes that are the basis for successful adaptation and concomitantly integrate autonomic, neuroendocrine, and behavioral stress responses. This review focuses on the physiology of CRF-related peptides and their cognate receptors with the aim of providing a comprehensive up-to-date overview of the field. We describe the major molecular features covering aspects of gene expression and regulation, structural properties, and molecular interactions, as well as mechanisms of signal transduction and their surveillance. In addition, we discuss the large body of published experimental studies focusing on state-of-the-art genetic approaches with high temporal and spatial precision, which collectively aimed to dissect the contribution of CRF-related ligands and receptors to different levels of the stress response. We discuss the controversies in the field and unravel knowledge gaps that might pave the way for future research directions and open up novel opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Jan M. Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany; and Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany; and Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
37
|
Jacoby J, Schwartz GW. Typology and Circuitry of Suppressed-by-Contrast Retinal Ganglion Cells. Front Cell Neurosci 2018; 12:269. [PMID: 30210298 PMCID: PMC6119723 DOI: 10.3389/fncel.2018.00269] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 08/02/2018] [Indexed: 11/13/2022] Open
Abstract
Retinal ganglion cells (RGCs) relay ~40 parallel and independent streams of visual information, each encoding a specific feature of a visual scene, to the brain for further processing. The polarity of a visual neuron’s response to a change in contrast is generally the first characteristic used for functional classification: ON cells increase their spike rate to positive contrast; OFF cells increase their spike rate for negative contrast; ON-OFF cells increase their spike rate for both contrast polarities. Suppressed-by-Contrast (SbC) neurons represent a less well-known fourth category; they decrease firing below a baseline rate for both positive and negative contrasts. SbC RGCs were discovered over 50 years ago, and SbC visual neurons have now been found in the thalamus and primary visual cortex of several mammalian species, including primates. Recent discoveries of SbC RGCs in mice have provided new opportunities for tracing upstream circuits in the retina responsible for the SbC computation and downstream targets in the brain where this information is used. We review and clarify recent work on the circuit mechanism of the SbC computation in these RGCs. Studies of mechanism rely on precisely defined cell types, and we argue that, like ON, OFF, and ON-OFF RGCs, SbC RGCs consist of more than one type. A new appreciation of the diversity of SbC RGCs will help guide future work on their targets in the brain and their roles in visual perception and behavior.
Collapse
Affiliation(s)
- Jason Jacoby
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Gregory William Schwartz
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, United States
| |
Collapse
|
38
|
Jo A, Xu J, Deniz S, Cherian S, DeVries SH, Zhu Y. Intersectional Strategies for Targeting Amacrine and Ganglion Cell Types in the Mouse Retina. Front Neural Circuits 2018; 12:66. [PMID: 30186122 PMCID: PMC6113359 DOI: 10.3389/fncir.2018.00066] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/02/2018] [Indexed: 12/02/2022] Open
Abstract
The mammalian retina harbors over 100 different cell types. To understand how retinal circuits work, it is essential to systematically access each type. A widely used approach for achieving targeted transgene expression exploits promoter-driven Cre lines. However, Cre expression in a given transgenic line in the retina and elsewhere in the brain is rarely confined to a single cell type, contributing ambiguity to the interpretation of results from broadly applied manipulations. To obtain unambiguous information about retinal processing, it is desirable to have strategies for further restricting transgene expression to a few or even to a single cell type. We employed an intersectional strategy based on a Cre/Flp double recombinase system to target amacrine and ganglion cell types in the inner retina. We analyzed expression patterns in seven Flp drivers and then created combinational mouse lines by selective cross breeding with Cre drivers. Breeding with Flp drivers can routinely remove labeling from more than 90% of the cells in Cre drivers, leading to only a handful cell types, typically 2–3, remaining in the intersection. Cre/Flp combinatorial mouse lines enabled us to identify and anatomically characterize retinal cell types with greater ease and demonstrated the feasibility of intersectional strategies in retinal research. In addition to the retina, we examined Flp expression in the lateral geniculate nucleus and superior colliculus. Our results establish a foundation for future application of intersectional strategies in the retina and retino-recipient regions.
Collapse
Affiliation(s)
- Andrew Jo
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jian Xu
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Sercan Deniz
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Suraj Cherian
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Steven H DeVries
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Yongling Zhu
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
39
|
Park SJH, Pottackal J, Ke JB, Jun NY, Rahmani P, Kim IJ, Singer JH, Demb JB. Convergence and Divergence of CRH Amacrine Cells in Mouse Retinal Circuitry. J Neurosci 2018; 38:3753-3766. [PMID: 29572434 PMCID: PMC5895998 DOI: 10.1523/jneurosci.2518-17.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 03/01/2018] [Accepted: 03/07/2018] [Indexed: 11/21/2022] Open
Abstract
Inhibitory interneurons sculpt the outputs of excitatory circuits to expand the dynamic range of information processing. In mammalian retina, >30 types of amacrine cells provide lateral inhibition to vertical, excitatory bipolar cell circuits, but functional roles for only a few amacrine cells are well established. Here, we elucidate the function of corticotropin-releasing hormone (CRH)-expressing amacrine cells labeled in Cre-transgenic mice of either sex. CRH cells costratify with the ON alpha ganglion cell, a neuron highly sensitive to positive contrast. Electrophysiological and optogenetic analyses demonstrate that two CRH types (CRH-1 and CRH-3) make GABAergic synapses with ON alpha cells. CRH-1 cells signal via graded membrane potential changes, whereas CRH-3 cells fire action potentials. Both types show sustained ON-type responses to positive contrast over a range of stimulus conditions. Optogenetic control of transmission at CRH-1 synapses demonstrates that these synapses are tuned to low temporal frequencies, maintaining GABA release during fast hyperpolarizations during brief periods of negative contrast. CRH amacrine cell output is suppressed by prolonged negative contrast, when ON alpha ganglion cells continue to receive inhibitory input from converging OFF-pathway amacrine cells; the converging ON- and OFF-pathway inhibition balances tonic excitatory drive to ON alpha cells. Previously, it was demonstrated that CRH-1 cells inhibit firing by suppressed-by-contrast (SbC) ganglion cells during positive contrast. Therefore, divergent outputs of CRH-1 cells inhibit two ganglion cell types with opposite responses to positive contrast. The opposing responses of ON alpha and SbC ganglion cells are explained by differing excitation/inhibition balance in the two circuits.SIGNIFICANCE STATEMENT A goal of neuroscience research is to explain the function of neural circuits at the level of specific cell types. Here, we studied the function of specific types of inhibitory interneurons, corticotropin-releasing hormone (CRH) amacrine cells, in the mouse retina. Genetic tools were used to identify and manipulate CRH cells, which make GABAergic synapses with a well studied ganglion cell type, the ON alpha cell. CRH cells converge with other types of amacrine cells to tonically inhibit ON alpha cells and balance their high level of excitation. CRH cells diverge to different types of ganglion cell, the unique properties of which depend on their balance of excitation and inhibition.
Collapse
Affiliation(s)
| | | | - Jiang-Bin Ke
- Department of Biology, University of Maryland, College Park, Maryland 20742
| | | | | | - In-Jung Kim
- Department of Ophthalmology and Visual Science
- Interdepartmental Neuroscience Program
- Department of Neuroscience
| | - Joshua H Singer
- Department of Biology, University of Maryland, College Park, Maryland 20742
| | - Jonathan B Demb
- Department of Ophthalmology and Visual Science,
- Interdepartmental Neuroscience Program
- Department of Cellular and Molecular Physiology, Yale University, New Haven, Connecticut 06511, and
| |
Collapse
|
40
|
Parmhans N, Sajgo S, Niu J, Luo W, Badea TC. Characterization of retinal ganglion cell, horizontal cell, and amacrine cell types expressing the neurotrophic receptor tyrosine kinase Ret. J Comp Neurol 2017; 526:742-766. [PMID: 29218725 DOI: 10.1002/cne.24367] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 11/07/2017] [Accepted: 11/22/2017] [Indexed: 11/11/2022]
Abstract
We report the retinal expression pattern of Ret, a receptor tyrosine kinase for the glial derived neurotrophic factor (GDNF) family ligands (GFLs), during development and in the adult mouse. Ret is initially expressed in retinal ganglion cells (RGCs), followed by horizontal cells (HCs) and amacrine cells (ACs), beginning with the early stages of postmitotic development. Ret expression persists in all three classes of neurons in the adult. Using RNA sequencing, immunostaining and random sparse recombination, we show that Ret is expressed in at least three distinct types of ACs, and ten types of RGCs. Using intersectional genetics, we describe the dendritic arbor morphologies of RGC types expressing Ret in combination with each of the three members of the POU4f/Brn3 family of transcription factors. Ret expression overlaps with Brn3a in 4 RGC types, with Brn3b in 5 RGC types, and with Brn3c in one RGC type, respectively. Ret+ RGCs project to the lateral geniculate nucleus (LGN), pretectal area (PTA) and superior colliculus (SC), and avoid the suprachiasmatic nucleus and accessory optic system. Brn3a+ Ret+ and Brn3c+ Ret+ RGCs project preferentially to contralateral retinorecipient areas, while Brn3b+ Ret+ RGCs shows minor ipsilateral projections to the olivary pretectal nucleus and the LGN. Our findings establish intersectional genetic approaches for the anatomic and developmental characterization of individual Ret+ RGC types. In addition, they provide necessary information for addressing the potential interplay between GDNF neurotrophic signaling and transcriptional regulation in RGC type specification.
Collapse
Affiliation(s)
- Nadia Parmhans
- Retinal Circuit Development & Genetics Unit, Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, NIH, Bethesda, Maryland
| | - Szilard Sajgo
- Retinal Circuit Development & Genetics Unit, Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, NIH, Bethesda, Maryland
| | - Jingwen Niu
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Wenqin Luo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tudor Constantin Badea
- Retinal Circuit Development & Genetics Unit, Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, NIH, Bethesda, Maryland
| |
Collapse
|
41
|
Large scale matching of function to the genetic identity of retinal ganglion cells. Sci Rep 2017; 7:15395. [PMID: 29133846 PMCID: PMC5684394 DOI: 10.1038/s41598-017-15741-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/30/2017] [Indexed: 02/07/2023] Open
Abstract
Understanding the role of neurons in encoding and transmitting information is a major goal in neuroscience. This requires insight on the data-rich neuronal spiking patterns combined, ideally, with morphology and genetic identity. Electrophysiologists have long experienced the trade-offs between anatomically-accurate single-cell recording techniques and high-density multi-cellular recording methods with poor anatomical correlations. In this study, we present a novel technique that combines large-scale micro-electrode array recordings with genetic identification and the anatomical location of the retinal ganglion cell soma. This was obtained through optogenetic stimulation and subsequent confocal imaging of genetically targeted retinal ganglion cell sub-populations in the mouse. With the many molecular options available for optogenetic gene expression, we view this method as a versatile tool for matching function to genetic classifications, which can be extended to include morphological information if the density of labelled cells is at the correct level.
Collapse
|
42
|
Hanlon KS, Chadderton N, Palfi A, Blanco Fernandez A, Humphries P, Kenna PF, Millington-Ward S, Farrar GJ. A Novel Retinal Ganglion Cell Promoter for Utility in AAV Vectors. Front Neurosci 2017; 11:521. [PMID: 28983234 PMCID: PMC5613148 DOI: 10.3389/fnins.2017.00521] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 09/04/2017] [Indexed: 12/13/2022] Open
Abstract
Significant advances in gene therapy have enabled exploration of therapies for inherited retinal disorders, many of which are in preclinical development or clinical evaluation. Gene therapy for retinal conditions has led the way in this growing field. The loss of retinal ganglion cells (RGCs) is a hallmark of a number of retinal disorders. As the field matures innovations that aid in refining therapies and optimizing efficacy are in demand. Gene therapies under development for RGC-related disorders, when delivered with recombinant adeno associated vectors (AAV), have typically been expressed from ubiquitous promoter sequences. Here we describe how a novel promoter from the murine Nefh gene was selected to drive transgene expression in RGCs. The Nefh promoter, in an AAV2/2 vector, was shown to drive preferential EGFP expression in murine RGCs in vivo following intravitreal injection. In contrast, EGFP expression from a CMV promoter was observed not only in RGCs, but throughout the inner nuclear layer and in amacrine cells located within the ganglion cell layer (GCL). Of note, the Nefh promoter sequence is sufficiently compact to be readily accommodated in AAV vectors, where transgene size represents a significant constraint. Moreover, this promoter should in principle provide a more targeted and potentially safer alternative for RGC-directed gene therapies.
Collapse
Affiliation(s)
- Killian S Hanlon
- School of Genetics and Microbiology, Smurfit Institute of Genetics, Trinity College DublinDublin, Ireland
| | - Naomi Chadderton
- School of Genetics and Microbiology, Smurfit Institute of Genetics, Trinity College DublinDublin, Ireland
| | - Arpad Palfi
- School of Genetics and Microbiology, Smurfit Institute of Genetics, Trinity College DublinDublin, Ireland
| | | | - Peter Humphries
- School of Genetics and Microbiology, Smurfit Institute of Genetics, Trinity College DublinDublin, Ireland
| | - Paul F Kenna
- School of Genetics and Microbiology, Smurfit Institute of Genetics, Trinity College DublinDublin, Ireland.,Research Foundation, Royal Victoria Eye and Ear HospitalDublin, Ireland
| | - Sophia Millington-Ward
- School of Genetics and Microbiology, Smurfit Institute of Genetics, Trinity College DublinDublin, Ireland
| | - G Jane Farrar
- School of Genetics and Microbiology, Smurfit Institute of Genetics, Trinity College DublinDublin, Ireland
| |
Collapse
|
43
|
Affiliation(s)
- Jeffrey S. Diamond
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892-3701
| |
Collapse
|
44
|
Pérez de Sevilla Müller L, Solomon A, Sheets K, Hapukino H, Rodriguez AR, Brecha NC. Multiple cell types form the VIP amacrine cell population. J Comp Neurol 2017; 527:133-158. [PMID: 28472856 DOI: 10.1002/cne.24234] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 04/21/2017] [Accepted: 04/27/2017] [Indexed: 12/21/2022]
Abstract
Amacrine cells are a heterogeneous group of interneurons that form microcircuits with bipolar, amacrine and ganglion cells to process visual information in the inner retina. This study has characterized the morphology, neurochemistry and major cell types of a VIP-ires-Cre amacrine cell population. VIP-tdTomato and -Confetti (Brainbow2.1) mouse lines were generated by crossing a VIP-ires-Cre line with either a Cre-dependent tdTomato or Brainbow2.1 reporter line. Retinal sections and whole-mounts were evaluated by quantitative, immunohistochemical, and intracellular labeling approaches. The majority of tdTomato and Confetti fluorescent cell bodies were in the inner nuclear layer (INL) and a few cell bodies were in the ganglion cell layer (GCL). Fluorescent processes ramified in strata 1, 3, 4, and 5 of the inner plexiform layer (IPL). All tdTomato fluorescent cells expressed syntaxin 1A and GABA-immunoreactivity indicating they were amacrine cells. The average VIP-tdTomato fluorescent cell density in the INL and GCL was 535 and 24 cells/mm2 , respectively. TdTomato fluorescent cells in the INL and GCL contained VIP-immunoreactivity. The VIP-ires-Cre amacrine cell types were identified in VIP-Brainbow2.1 retinas or by intracellular labeling in VIP-tdTomato retinas. VIP-1 amacrine cells are bistratified, wide-field cells that ramify in strata 1, 4, and 5, VIP-2A and 2B amacrine cells are medium-field cells that mainly ramify in strata 3 and 4, and VIP-3 displaced amacrine cells are medium-field cells that ramify in strata 4 and 5 of the IPL. VIP-ires-Cre amacrine cells form a neuropeptide-expressing cell population with multiple cell types, which are likely to have distinct roles in visual processing.
Collapse
Affiliation(s)
- Luis Pérez de Sevilla Müller
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | - Alexander Solomon
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | - Kristopher Sheets
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | - Hinekura Hapukino
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | - Allen R Rodriguez
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | - Nicholas C Brecha
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763.,Department of Medicine, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763.,Department of Ophthalmology and the Stein Eye Institute, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763.,CURE Digestive Diseases Research Center, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763.,Veterans Administration Greater Los Angeles Health System, Los Angeles, California, 90073
| |
Collapse
|
45
|
Tien NW, Soto F, Kerschensteiner D. Homeostatic Plasticity Shapes Cell-Type-Specific Wiring in the Retina. Neuron 2017; 94:656-665.e4. [PMID: 28457596 DOI: 10.1016/j.neuron.2017.04.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/17/2017] [Accepted: 04/11/2017] [Indexed: 10/19/2022]
Abstract
Convergent input from different presynaptic partners shapes the responses of postsynaptic neurons. Whether developing postsynaptic neurons establish connections with each presynaptic partner independently or balance inputs to attain specific responses is unclear. Retinal ganglion cells (RGCs) receive convergent input from bipolar cell types with different contrast responses and temporal tuning. Here, using optogenetic activation and pharmacogenetic silencing, we found that type 6 bipolar (B6) cells dominate excitatory input to ONα-RGCs. We generated mice in which B6 cells were selectively removed from developing circuits (B6-DTA). In B6-DTA mice, ONα-RGCs adjusted connectivity with other bipolar cells in a cell-type-specific manner. They recruited new partners, increased synapses with some existing partners, and maintained constant input from others. Patch-clamp recordings revealed that anatomical rewiring precisely preserved contrast and temporal frequency response functions of ONα-RGCs, indicating that homeostatic plasticity shapes cell-type-specific wiring in the developing retina to stabilize visual information sent to the brain.
Collapse
Affiliation(s)
- Nai-Wen Tien
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Graduate Program in Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Florentina Soto
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
46
|
Abstract
The dorsal lateral geniculate nucleus (dLGN) of the thalamus is the principal conduit for visual information from retina to visual cortex. Viewed initially as a simple relay, recent studies in the mouse reveal far greater complexity in the way input from the retina is combined, transmitted, and processed in dLGN. Here we consider the structural and functional organization of the mouse retinogeniculate pathway by examining the patterns of retinal projections to dLGN and how they converge onto thalamocortical neurons to shape the flow of visual information to visual cortex.
Collapse
Affiliation(s)
- Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences,Washington University School of Medicine,Saint Louis,Missouri 63110
| | - William Guido
- Department of Anatomical Sciences and Neurobiology,University of Louisville School of Medicine,Louisville,Kentucky 40292
| |
Collapse
|
47
|
Lu Q, Ganjawala TH, Ivanova E, Cheng JG, Troilo D, Pan ZH. AAV-mediated transduction and targeting of retinal bipolar cells with improved mGluR6 promoters in rodents and primates. Gene Ther 2016; 23:680-9. [PMID: 27115727 PMCID: PMC4863234 DOI: 10.1038/gt.2016.42] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 04/14/2016] [Accepted: 04/18/2016] [Indexed: 12/20/2022]
Abstract
Adeno-associated virus (AAV) vectors have been a powerful gene delivery vehicle to the retina for basic research and gene therapy. For many of these applications, achieving cell type-specific targeting and high transduction efficiency is desired. Recently, there has been increasing interest in AAV-mediated gene targeting to specific retinal bipolar cell types. A 200-bp enhancer in combination with a basal SV40 promoter has been commonly used to target transgenes into ON-type bipolar cells. In the current study, we searched for additional cis-regulatory elements in the mGluR6 gene for improving AAV-mediated transduction efficiency into retinal bipolar cells. Our results showed that the combination of the endogenous mGluR6 promoter with additional enhancers in the introns of the mGluR6 gene markedly enhanced AAV transduction efficiency as well as made the targeting more selective for rod bipolar cells in mice. Furthermore, the AAV vectors with the improved promoter could target to ON bipolar cells with robust transduction efficiency in the parafovea and the far peripheral retina of marmoset monkeys. The improved mGluR6 promoter constructs could provide a valuable tool for genetic manipulation in rod bipolar cells in mice and facilitate clinical applications for ON bipolar cell-based gene therapies.
Collapse
Affiliation(s)
- Q Lu
- Dept. of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI
| | - TH Ganjawala
- Dept. of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI
| | - E Ivanova
- Burke Medical Research Institute, Weill Medical College of Cornell University, White Plains, NY
| | - JG Cheng
- Neuroscience Center, University of North Carolina, Chapel Hill, NC
| | - D Troilo
- State University of New York, College of Optometry, New York, NY
| | - Z-H Pan
- Dept. of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI
- Dept. of Ophthalmology, Kresge Eye Institute, Wayne State University School of Medicine, Detroit, MI
| |
Collapse
|
48
|
Katz ML, Viney TJ, Nikolic K. Receptive Field Vectors of Genetically-Identified Retinal Ganglion Cells Reveal Cell-Type-Dependent Visual Functions. PLoS One 2016; 11:e0147738. [PMID: 26845435 PMCID: PMC4742227 DOI: 10.1371/journal.pone.0147738] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 01/07/2016] [Indexed: 11/18/2022] Open
Abstract
Sensory stimuli are encoded by diverse kinds of neurons but the identities of the recorded neurons that are studied are often unknown. We explored in detail the firing patterns of eight previously defined genetically-identified retinal ganglion cell (RGC) types from a single transgenic mouse line. We first introduce a new technique of deriving receptive field vectors (RFVs) which utilises a modified form of mutual information (“Quadratic Mutual Information”). We analysed the firing patterns of RGCs during presentation of short duration (~10 second) complex visual scenes (natural movies). We probed the high dimensional space formed by the visual input for a much smaller dimensional subspace of RFVs that give the most information about the response of each cell. The new technique is very efficient and fast and the derivation of novel types of RFVs formed by the natural scene visual input was possible even with limited numbers of spikes per cell. This approach enabled us to estimate the 'visual memory' of each cell type and the corresponding receptive field area by calculating Mutual Information as a function of the number of frames and radius. Finally, we made predictions of biologically relevant functions based on the RFVs of each cell type. RGC class analysis was complemented with results for the cells’ response to simple visual input in the form of black and white spot stimulation, and their classification on several key physiological metrics. Thus RFVs lead to predictions of biological roles based on limited data and facilitate analysis of sensory-evoked spiking data from defined cell types.
Collapse
Affiliation(s)
- Matthew L. Katz
- Centre for Bio-Inspired Technology, Institute of Biomedical Engineering, Department of Electrical and Electronic Engineering, The Bessemer Building, Imperial College London, London SW7 2AZ, United Kingdom
| | - Tim J. Viney
- Neural Circuit Laboratories, Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
- University of Basel, 4003 Basel, Switzerland
| | - Konstantin Nikolic
- Centre for Bio-Inspired Technology, Institute of Biomedical Engineering, Department of Electrical and Electronic Engineering, The Bessemer Building, Imperial College London, London SW7 2AZ, United Kingdom
- * E-mail:
| |
Collapse
|
49
|
Jacoby J, Zhu Y, DeVries SH, Schwartz GW. An Amacrine Cell Circuit for Signaling Steady Illumination in the Retina. Cell Rep 2015; 13:2663-70. [PMID: 26711334 DOI: 10.1016/j.celrep.2015.11.062] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 10/13/2015] [Accepted: 11/18/2015] [Indexed: 10/22/2022] Open
Abstract
Decades of research have focused on the circuit connectivity between retinal neurons, but only a handful of amacrine cells have been described functionally and placed in the context of a specific retinal circuit. Here, we identify a circuit where inhibition from a specific amacrine cell plays a vital role in shaping the feature selectivity of a postsynaptic ganglion cell. We record from transgenically labeled CRH-1 amacrine cells and identify a postsynaptic target for CRH-1 amacrine cell inhibition in an atypical retinal ganglion cell (RGC) in mouse retina, the Suppressed-by-Contrast (SbC) RGC. Unlike other RGC types, SbC RGCs spike tonically in steady illumination and are suppressed by both increases and decreases in illumination. Inhibition from GABAergic CRH-1 amacrine cells shapes this unique contrast response profile to positive contrast. We show the existence and impact of this circuit, with both paired recordings and cell-type-specific ablation.
Collapse
Affiliation(s)
- Jason Jacoby
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611, USA
| | - Yongling Zhu
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611, USA
| | - Steven H DeVries
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611, USA; Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611, USA
| | - Gregory W Schwartz
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611, USA; Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611, USA.
| |
Collapse
|
50
|
Abstract
The mammalian retina is an important model system for studying neural circuitry: Its role in sensation is clear, its cell types are relatively well defined, and its responses to natural stimuli-light patterns-can be studied in vitro. To solve the retina, we need to understand how the circuits presynaptic to its output neurons, ganglion cells, divide the visual scene into parallel representations to be assembled and interpreted by the brain. This requires identifying the component interneurons and understanding how their intrinsic properties and synapses generate circuit behaviors. Because the cellular composition and fundamental properties of the retina are shared across species, basic mechanisms studied in the genetically modifiable mouse retina apply to primate vision. We propose that the apparent complexity of retinal computation derives from a straightforward mechanism-a dynamic balance of synaptic excitation and inhibition regulated by use-dependent synaptic depression-applied differentially to the parallel pathways that feed ganglion cells.
Collapse
Affiliation(s)
- Jonathan B Demb
- Department of Ophthalmology and Visual Science and Department of Cellular and Molecular Physiology, Yale University, New Haven, Connecticut 06511;
| | - Joshua H Singer
- Department of Biology, University of Maryland, College Park, Maryland 20742;
| |
Collapse
|