51
|
Agostinone J, Alarcon-Martinez L, Gamlin C, Yu WQ, Wong ROL, Di Polo A. Insulin signalling promotes dendrite and synapse regeneration and restores circuit function after axonal injury. Brain 2019; 141:1963-1980. [PMID: 29931057 PMCID: PMC6022605 DOI: 10.1093/brain/awy142] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 04/06/2018] [Indexed: 01/07/2023] Open
Abstract
Dendrite pathology and synapse disassembly are critical features of chronic neurodegenerative diseases. In spite of this, the capacity of injured neurons to regenerate dendrites has been largely ignored. Here, we show that, upon axonal injury, retinal ganglion cells undergo rapid dendritic retraction and massive synapse loss that preceded neuronal death. Human recombinant insulin, administered as eye drops or systemically after dendritic arbour shrinkage and prior to cell loss, promoted robust regeneration of dendrites and successful reconnection with presynaptic targets. Insulin-mediated regeneration of excitatory postsynaptic sites on retinal ganglion cell dendritic processes increased neuronal survival and rescued light-triggered retinal responses. Further, we show that axotomy-induced dendrite retraction triggered substantial loss of the mammalian target of rapamycin (mTOR) activity exclusively in retinal ganglion cells, and that insulin fully reversed this response. Targeted loss-of-function experiments revealed that insulin-dependent activation of mTOR complex 1 (mTORC1) is required for new dendritic branching to restore arbour complexity, while complex 2 (mTORC2) drives dendritic process extension thus re-establishing field area. Our findings demonstrate that neurons in the mammalian central nervous system have the intrinsic capacity to regenerate dendrites and synapses after injury, and provide a strong rationale for the use of insulin and/or its analogues as pro-regenerative therapeutics for intractable neurodegenerative diseases including glaucoma.
Collapse
Affiliation(s)
- Jessica Agostinone
- Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada.,University of Montreal Hospital Research Center (CR-CHUM), University of Montreal, Montreal, Quebec, Canada
| | - Luis Alarcon-Martinez
- Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada.,University of Montreal Hospital Research Center (CR-CHUM), University of Montreal, Montreal, Quebec, Canada
| | - Clare Gamlin
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, Washington, USA
| | - Wan-Qing Yu
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, Washington, USA
| | - Rachel O L Wong
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, Washington, USA
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada.,University of Montreal Hospital Research Center (CR-CHUM), University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
52
|
Berry M, Ahmed Z, Logan A. Return of function after CNS axon regeneration: Lessons from injury-responsive intrinsically photosensitive and alpha retinal ganglion cells. Prog Retin Eye Res 2019; 71:57-67. [DOI: 10.1016/j.preteyeres.2018.11.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/26/2018] [Accepted: 11/16/2018] [Indexed: 12/16/2022]
|
53
|
Mellough CB, Collin J, Queen R, Hilgen G, Dorgau B, Zerti D, Felemban M, White K, Sernagor E, Lako M. Systematic Comparison of Retinal Organoid Differentiation from Human Pluripotent Stem Cells Reveals Stage Specific, Cell Line, and Methodological Differences. Stem Cells Transl Med 2019; 8:694-706. [PMID: 30916455 PMCID: PMC6591558 DOI: 10.1002/sctm.18-0267] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
A major goal in the stem cell field is to generate tissues that can be utilized as a universal tool for in vitro models of development and disease, drug development, or as a resource for patients suffering from disease or injury. Great efforts are being made to differentiate human pluripotent stem cells in vitro toward retinal tissue, which is akin to native human retina in its cytoarchitecture and function, yet the numerous existing retinal induction protocols remain variable in their efficiency and do not routinely produce morphologically or functionally mature photoreceptors. Herein, we determine the impact that the method of embryoid body (EB) formation and maintenance as well as cell line background has on retinal organoid differentiation from human embryonic stem cells and human induced pluripotent stem cells. Our data indicate that cell line-specific differences dominate the variables that underline the differentiation efficiency in the early stages of differentiation. In contrast, the EB generation method and maintenance conditions determine the later differentiation and maturation of retinal organoids. Of the latter, the mechanical method of EB generation under static conditions, accompanied by media supplementation with Y27632 for the first 48 hours of differentiation, results in the most consistent formation of laminated retinal neuroepithelium containing mature and electrophysiologically responsive photoreceptors. Collectively, our data provide substantive evidence for stage-specific differences in the ability to give rise to laminated retinae, which is determined by cell line-specific differences in the early stages of differentiation and EB generation/organoid maintenance methods at later stages.
Collapse
Affiliation(s)
- Carla B. Mellough
- Institute of Genetic MedicineNewcastle UniversityNewcastleUnited Kingdom
- Centre for Ophthalmology and Visual ScienceLions Eye Institute, University of Western AustraliaPerthWestern AustraliaAustralia
| | - Joseph Collin
- Institute of Genetic MedicineNewcastle UniversityNewcastleUnited Kingdom
| | - Rachel Queen
- Institute of Genetic MedicineNewcastle UniversityNewcastleUnited Kingdom
| | - Gerrit Hilgen
- Institute of NeuroscienceNewcastle UniversityNewcastleUnited Kingdom
| | - Birthe Dorgau
- Institute of Genetic MedicineNewcastle UniversityNewcastleUnited Kingdom
| | - Darin Zerti
- Institute of Genetic MedicineNewcastle UniversityNewcastleUnited Kingdom
| | - Majed Felemban
- Institute of Genetic MedicineNewcastle UniversityNewcastleUnited Kingdom
| | - Kathryn White
- EM Research ServicesNewcastle UniversityNewcastleUnited Kingdom
| | - Evelyne Sernagor
- Institute of NeuroscienceNewcastle UniversityNewcastleUnited Kingdom
| | - Majlinda Lako
- Institute of Genetic MedicineNewcastle UniversityNewcastleUnited Kingdom
| |
Collapse
|
54
|
Kovács-Öller T, Szarka G, Ganczer A, Tengölics Á, Balogh B, Völgyi B. Expression of Ca 2+-Binding Buffer Proteins in the Human and Mouse Retinal Neurons. Int J Mol Sci 2019; 20:E2229. [PMID: 31067641 PMCID: PMC6539911 DOI: 10.3390/ijms20092229] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/31/2022] Open
Abstract
Ca2+-binding buffer proteins (CaBPs) are widely expressed by various neurons throughout the central nervous system (CNS), including the retina. While the expression of CaBPs by photoreceptors, retinal interneurons and the output ganglion cells in the mammalian retina has been extensively studied, a general description is still missing due to the differences between species, developmental expression patterns and study-to-study discrepancies. Furthermore, CaBPs are occasionally located in a compartment-specific manner and two or more CaBPs can be expressed by the same neuron, thereby sharing the labor of Ca2+ buffering in the intracellular milieu. This article reviews this topic by providing a framework on CaBP functional expression by neurons of the mammalian retina with an emphasis on human and mouse retinas and the three most abundant and extensively studied buffer proteins: parvalbumin, calretinin and calbindin.
Collapse
Affiliation(s)
- Tamás Kovács-Öller
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
- Medical School, University of Pécs, 7624 Pécs, Hungary.
| | - Gergely Szarka
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Alma Ganczer
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Ádám Tengölics
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Boglárka Balogh
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Béla Völgyi
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
- Medical School, University of Pécs, 7624 Pécs, Hungary.
| |
Collapse
|
55
|
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
|
56
|
Digital Museum of Retinal Ganglion Cells with Dense Anatomy and Physiology. Cell 2019; 173:1293-1306.e19. [PMID: 29775596 DOI: 10.1016/j.cell.2018.04.040] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 02/09/2018] [Accepted: 04/27/2018] [Indexed: 11/23/2022]
Abstract
When 3D electron microscopy and calcium imaging are used to investigate the structure and function of neural circuits, the resulting datasets pose new challenges of visualization and interpretation. Here, we present a new kind of digital resource that encompasses almost 400 ganglion cells from a single patch of mouse retina. An online "museum" provides a 3D interactive view of each cell's anatomy, as well as graphs of its visual responses. The resource reveals two aspects of the retina's inner plexiform layer: an arbor segregation principle governing structure along the light axis and a density conservation principle governing structure in the tangential plane. Structure is related to visual function; ganglion cells with arbors near the layer of ganglion cell somas are more sustained in their visual responses on average. Our methods are potentially applicable to dense maps of neuronal anatomy and physiology in other parts of the nervous system.
Collapse
|
57
|
Quattrochi LE, Stabio ME, Kim I, Ilardi MC, Michelle Fogerson P, Leyrer ML, Berson DM. The M6 cell: A small-field bistratified photosensitive retinal ganglion cell. J Comp Neurol 2019; 527:297-311. [PMID: 30311650 PMCID: PMC6594700 DOI: 10.1002/cne.24556] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 09/07/2018] [Accepted: 10/01/2018] [Indexed: 01/26/2023]
Abstract
We have identified a novel, sixth type of intrinsically photosensitive retinal ganglion cell (ipRGC) in the mouse-the M6 cell. Its spiny, highly branched dendritic arbor is bistratified, with dendrites restricted to the inner and outer margins of the inner plexiform layer, co-stratifying with the processes of other ipRGC types. We show that M6 cells are by far the most abundant ganglion cell type labeled in adult pigmented Cdh3-GFP BAC transgenic mice. A few M5 ipRGCs are also labeled, but no other RGC types were encountered. Several distinct subnuclei in the geniculate complex and the pretectum contain labeled retinofugal axons in the Cdh3-GFP mouse. These are presumably the principle central targets of M6 cells (as well as M5 cells). Projections from M6 cells to the dorsal lateral geniculate nucleus were confirmed by retrograde tracing, suggesting they contribute to pattern vision. M6 cells have low levels of melanopsin expression and relatively weak melanopsin-dependent light responses. They also exhibit strong synaptically driven light responses. Their dendritic fields are the smallest and most abundantly branched of all ipRGCs. They have small receptive fields and strong antagonistic surrounds. Despite deploying dendrites partly in the OFF sublamina, M6 cells appear to be driven exclusively by the ON pathway, suggesting that their OFF arbor, like those of certain other ipRGCs, may receive ectopic input from passing ON bipolar cells axons in the OFF sublayer.
Collapse
Affiliation(s)
- Lauren E Quattrochi
- Department of Neuroscience and Carney Institute for Brain Science, Brown University, Providence, Rhode Island
| | - Maureen E Stabio
- Department of Cell & Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado
| | - Inkyu Kim
- Department of Neuroscience and Carney Institute for Brain Science, Brown University, Providence, Rhode Island
| | - Marissa C Ilardi
- Department of Neuroscience and Carney Institute for Brain Science, Brown University, Providence, Rhode Island
| | - P Michelle Fogerson
- Department of Neuroscience and Carney Institute for Brain Science, Brown University, Providence, Rhode Island
| | - Megan L Leyrer
- Department of Neuroscience and Carney Institute for Brain Science, Brown University, Providence, Rhode Island
| | - David M Berson
- Department of Neuroscience and Carney Institute for Brain Science, Brown University, Providence, Rhode Island
| |
Collapse
|
58
|
El-Danaf RN, Huberman AD. Sub-topographic maps for regionally enhanced analysis of visual space in the mouse retina. J Comp Neurol 2018; 527:259-269. [PMID: 29675855 DOI: 10.1002/cne.24457] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/01/2017] [Accepted: 06/02/2017] [Indexed: 01/28/2023]
Abstract
In many species, neurons are unevenly distributed across the retina, leading to nonuniform analysis of specific visual features at certain locations in visual space. In recent years, the mouse has emerged as a premiere model for probing visual system function, development, and disease. Thus, achieving a detailed understanding of mouse visual circuit architecture is of paramount importance. The general belief is that mice possess a relatively even topographic distribution of retinal ganglion cells (RGCs)-the output neurons of the eye. However, mouse RGCs include ∼30 subtypes; each responds best to a specific feature in the visual scene and conveys that information to central targets. Given the crucial role of RGCs and the prominence of the mouse as a model, we asked how different RGC subtypes are distributed across the retina. We targeted and filled individual fluorescently tagged RGC subtypes from across the retinal surface and evaluated the dendritic arbor extent and soma size of each cell according to its specific retinotopic position. Three prominent RGC subtypes: On-Off direction selective RGCs, object-motion-sensitive RGCs, and a specialized subclass of nonimage-forming RGCs each had marked topographic variations in their dendritic arbor sizes. Moreover, the pattern of variation was distinct for each RGC subtype. Thus, there is increasing evidence that the mouse retina encodes visual space in a region-specific manner. As a consequence, some visual features are sampled far more densely at certain retinal locations than others. These findings have implications for central visual processing, perception, and behavior in this prominent model species.
Collapse
Affiliation(s)
- Rana N El-Danaf
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California
| | - Andrew D Huberman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California.,Department of Ophthalmology, Stanford University School of Medicine, Stanford, California.,Stanford Neurosciences Institute, Stanford, California.,BioX, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
59
|
Lee ES, Lee JY, Kim GH, Jeon CJ. Identification of calretinin-expressing retinal ganglion cells projecting to the mouse superior colliculus. Cell Tissue Res 2018; 376:153-163. [PMID: 30506393 DOI: 10.1007/s00441-018-2964-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 11/12/2018] [Indexed: 11/29/2022]
Abstract
In mice, retinal ganglion cells (RGCs), which consist of around 30 subtypes, exclusively transmit retinal information to the relevant brain systems through parallel visual pathways. The superior colliculus (SC) receives the vast majority of this information from several RGC subtypes. The objective of the current study is to identify the types of calretinin (CR)-expressing RGCs that project to the SC in mice. To label RGCs, we performed CR immunoreactivity in the mouse retina after injections of fluorescent dye, dextran into mouse SC. Subsequently, the neurons double-labeled for dextran and CR were iontophoretically injected with the lipophilic dye, DiI, to characterize the detailed morphological properties of these cells. The analysis of various morphological parameters, including dendritic arborization, dendritic field size and stratification, indicated that, of the ten different types of CR-expressing RGCs in the retina, the double-labeled cells consisted of at least eight types of RGCs that projected to the SC. These cells tended to have small-medium field sizes. However, except for dendritic field size, the cells did not exhibit consistent characteristics for the other morphometric parameters examined. The combination of a tracer and single-cell injections after immunohistochemistry for a particular molecule provided valuable data that confirmed the presence of distinct subtypes of RGCs within multiple-labeled RGCs that projected to specific brain regions.
Collapse
Affiliation(s)
- Eun-Shil Lee
- Department of Biology, School of Life Sciences, BK 21 Plus KNU Creative BioResearch Group, College of Natural Sciences, and Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41566, South Korea
| | - Jea-Young Lee
- Center of Excellence for Aging and Brain Repair, USF Health, University of South Florida, Tampa, FL, 33612, USA
| | - Gil Hyun Kim
- Department of Biology, School of Life Sciences, BK 21 Plus KNU Creative BioResearch Group, College of Natural Sciences, and Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41566, South Korea
| | - Chang-Jin Jeon
- Department of Biology, School of Life Sciences, BK 21 Plus KNU Creative BioResearch Group, College of Natural Sciences, and Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41566, South Korea.
| |
Collapse
|
60
|
Yee CW, Ivanova E, Toychiev AH, Willis DE, Sagdullaev BT. Atypical Expression and Activation of GluN2A- and GluN2B-Containing NMDA Receptors at Ganglion Cells during Retinal Degeneration. Neuroscience 2018; 393:61-72. [PMID: 30312782 DOI: 10.1016/j.neuroscience.2018.09.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/21/2018] [Accepted: 09/30/2018] [Indexed: 11/26/2022]
Abstract
Cellular communication through chemical synapses is determined by the nature of the neurotransmitter and the composition of postsynaptic receptors. In the excitatory synapse between bipolar and ganglion cells of the retina, postsynaptic AMPA receptors mediate resting activity. During evoked response, however, more abundant and sustained levels of glutamate also activate GluN2B-containing NMDA receptors (NMDARs). This phasic recruitment of distinct glutamate receptors is essential for visual discrimination; however, the fidelity of this basic mechanism under elevated glutamate levels due to aberrant activity, a common pathophysiology, is not known. Here, in both male and female mice with retinal degeneration (rd10), a condition associated with elevated synaptic activity, we reveal that changes in synaptic input to ganglion cells altered both composition and activation of NMDARs. We found that, in contrast to wild type, the spontaneous activity of rd10 cells was largely NMDAR-dependent. Surprisingly, this activity was driven primarily by atypical activation of GluN2A -containing NMDARs, not GluN2B-NMDARs. Indeed, immunohistochemical analyses and Western blot showed greater levels of the GluN2A-NMDAR subunit expression in rd10 retina compared to wild type. Overall, these results demonstrate how aberrant signaling leads to pathway-specific alterations in NMDAR expression and function.
Collapse
Affiliation(s)
- Christopher W Yee
- Weill Cornell Medicine at Burke Neurological Institute, White Plains, NY 10605, United States
| | - Elena Ivanova
- Weill Cornell Medicine at Burke Neurological Institute, White Plains, NY 10605, United States
| | - Abduqodir H Toychiev
- Weill Cornell Medicine at Burke Neurological Institute, White Plains, NY 10605, United States
| | - Dianna E Willis
- Weill Cornell Medicine at Burke Neurological Institute, White Plains, NY 10605, United States
| | - Botir T Sagdullaev
- Weill Cornell Medicine at Burke Neurological Institute, White Plains, NY 10605, United States.
| |
Collapse
|
61
|
Lakk M, Young D, Baumann JM, Jo AO, Hu H, Križaj D. Polymodal TRPV1 and TRPV4 Sensors Colocalize but Do Not Functionally Interact in a Subpopulation of Mouse Retinal Ganglion Cells. Front Cell Neurosci 2018; 12:353. [PMID: 30386208 PMCID: PMC6198093 DOI: 10.3389/fncel.2018.00353] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/20/2018] [Indexed: 01/23/2023] Open
Abstract
Retinal ganglion cells (RGCs) are projection neurons that transmit the visual signal from the retina to the brain. Their excitability and survival can be strongly influenced by mechanical stressors, temperature, lipid metabolites, and inflammatory mediators but the transduction mechanisms for these non-synaptic sensory inputs are not well characterized. Here, we investigate the distribution, functional expression, and localization of two polymodal transducers of mechanical, lipid, and inflammatory signals, TRPV1 and TRPV4 cation channels, in mouse RGCs. The most abundant vanilloid mRNA species was Trpv4, followed by Trpv2 and residual expression of Trpv3 and Trpv1. Immunohistochemical and functional analyses showed that TRPV1 and TRPV4 channels are expressed as separate molecular entities, with TRPV1-only (∼10%), TRPV4-only (∼40%), and TRPV1 + TRPV4 (∼10%) expressing RGC subpopulations. The TRPV1 + TRPV4 cohort included SMI-32-immunopositive alpha RGCs, suggesting potential roles for polymodal signal transduction in modulation of fast visual signaling. Arguing against obligatory heteromerization, optical imaging showed that activation and desensitization of TRPV1 and TRPV4 responses evoked by capsaicin and GSK1016790A are independent of each other. Overall, these data predict that RGC subpopulations will be differentially sensitive to mechanical and inflammatory stressors.
Collapse
Affiliation(s)
- Monika Lakk
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, United States
| | - Derek Young
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, United States
| | - Jackson M Baumann
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, United States.,Department of Bioengineering, University of Utah, Salt Lake City, UT, United States
| | - Andrew O Jo
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, United States
| | - Hongzhen Hu
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
| | - David Križaj
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, United States.,Department of Bioengineering, University of Utah, Salt Lake City, UT, United States.,Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
62
|
Muzyka VV, Brooks M, Badea TC. Postnatal developmental dynamics of cell type specification genes in Brn3a/Pou4f1 Retinal Ganglion Cells. Neural Dev 2018; 13:15. [PMID: 29958540 PMCID: PMC6025728 DOI: 10.1186/s13064-018-0110-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 06/06/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND About 20-30 distinct Retinal Ganglion Cell (RGC) types transmit visual information from the retina to the brain. The developmental mechanisms by which RGCs are specified are still largely unknown. Brn3a is a member of the Brn3/Pou4f transcription factor family, which contains key regulators of RGC postmitotic specification. In particular, Brn3a ablation results in the loss of RGCs with small, thick and dense dendritic arbors ('midget-like' RGCs), and morphological changes in other RGC subpopulations. To identify downstream molecular mechanisms underlying Brn3a effects on RGC numbers and morphology, our group recently performed a RNA deep sequencing screen for Brn3a transcriptional targets in mouse RGCs and identified 180 candidate transcripts. METHODS We now focus on a subset of 28 candidate genes encoding potential cell type determinant proteins. We validate and further define their retinal expression profile at five postnatal developmental time points between birth and adult stage, using in situ hybridization (ISH), RT-PCR and fluorescent immunodetection (IIF). RESULTS We find that a majority of candidate genes are enriched in the ganglion cell layer during early stages of postnatal development, but dynamically change their expression profile. We also document transcript-specific expression differences for two example candidates, using RT-PCR and ISH. Brn3a dependency could be confirmed by ISH and IIF only for a fraction of our candidates. CONCLUSIONS Amongst our candidate Brn3a target genes, a majority demonstrated ganglion cell layer specificity, however only around two thirds showed Brn3a dependency. Some were previously implicated in RGC type specification, while others have known physiological functions in RGCs. Only three genes were found to be consistently regulated by Brn3a throughout postnatal retina development - Mapk10, Tusc5 and Cdh4.
Collapse
Affiliation(s)
| | - Matthew Brooks
- Genomics Core, Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, NIH, Building 6, Room 331B Center Drive, Bethesda, MD, 20892-0610, USA
| | - Tudor Constantin Badea
- Retinal Circuit Development & Genetics Unit, Building 6, Room 331B Center Drive, Bethesda, MD, 20892-0610, USA.
| |
Collapse
|
63
|
Mörschel K, Breit M, Queisser G. Generating Neuron Geometries for Detailed Three-Dimensional Simulations Using AnaMorph. Neuroinformatics 2018; 15:247-269. [PMID: 28447297 DOI: 10.1007/s12021-017-9329-x] [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: 11/29/2022]
Abstract
Generating realistic and complex computational domains for numerical simulations is often a challenging task. In neuroscientific research, more and more one-dimensional morphology data is becoming publicly available through databases. This data, however, only contains point and diameter information not suitable for detailed three-dimensional simulations. In this paper, we present a novel framework, AnaMorph, that automatically generates water-tight surface meshes from one-dimensional point-diameter files. These surface triangulations can be used to simulate the electrical and biochemical behavior of the underlying cell. In addition to morphology generation, AnaMorph also performs quality control of the semi-automatically reconstructed cells coming from anatomical reconstructions. This toolset allows an extension from the classical dimension-reduced modeling and simulation of cellular processes to a full three-dimensional and morphology-including method, leading to novel structure-function interplay studies in the medical field. The developed numerical methods can further be employed in other areas where complex geometries are an essential component of numerical simulations.
Collapse
Affiliation(s)
- Konstantin Mörschel
- Goethe Center for Scientific Computing, Goethe University Frankfurt, Kettenhofweg 139, 60325, Frankfurt am Main, Germany
| | - Markus Breit
- Goethe Center for Scientific Computing, Goethe University Frankfurt, Kettenhofweg 139, 60325, Frankfurt am Main, Germany
| | - Gillian Queisser
- Department of Mathematics, Temple University, 1805 N Broad Street, Philadelphia, PA, 19122-6094, USA.
| |
Collapse
|
64
|
Esler TB, Kerr RR, Tahayori B, Grayden DB, Meffin H, Burkitt AN. Minimizing activation of overlying axons with epiretinal stimulation: The role of fiber orientation and electrode configuration. PLoS One 2018; 13:e0193598. [PMID: 29494655 PMCID: PMC5833203 DOI: 10.1371/journal.pone.0193598] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 02/14/2018] [Indexed: 12/19/2022] Open
Abstract
Currently, a challenge in electrical stimulation of the retina with a visual prosthesis (bionic eye) is to excite only the cells lying directly under the electrode in the ganglion cell layer, while avoiding excitation of axon bundles that pass over the surface of the retina in the nerve fiber layer. Stimulation of overlying axons results in irregular visual percepts, limiting perceptual efficacy. This research explores how differences in fiber orientation between the nerve fiber layer and ganglion cell layer leads to differences in the electrical activation of the axon initial segment and axons of passage. Approach. Axons of passage of retinal ganglion cells in the nerve fiber layer are characterized by a narrow distribution of fiber orientations, causing highly anisotropic spread of applied current. In contrast, proximal axons in the ganglion cell layer have a wider distribution of orientations. A four-layer computational model of epiretinal extracellular stimulation that captures the effect of neurite orientation in anisotropic tissue has been developed using a volume conductor model known as the cellular composite model. Simulations are conducted to investigate the interaction of neural tissue orientation, stimulating electrode configuration, and stimulation pulse duration and amplitude. Main results. Our model shows that simultaneous stimulation with multiple electrodes aligned with the nerve fiber layer can be used to achieve selective activation of axon initial segments rather than passing fibers. This result can be achieved while reducing required stimulus charge density and with only modest increases in the spread of activation in the ganglion cell layer, and is shown to extend to the general case of arbitrary electrode array positioning and arbitrary target volume. Significance. These results elucidate a strategy for more targeted stimulation of retinal ganglion cells with experimentally-relevant multi-electrode geometries and achievable stimulation requirements.
Collapse
Affiliation(s)
- Timothy B. Esler
- Department of Biomedical Engineering, The University of Melbourne, Parkville, Victoria, Australia
- * E-mail:
| | - Robert R. Kerr
- Department of Biomedical Engineering, The University of Melbourne, Parkville, Victoria, Australia
- Seer Medical, Melbourne, Victoria, Australia
| | - Bahman Tahayori
- Monash Institute of Medical Engineering, Monash University, Clayton, Victoria, Australia
| | - David B. Grayden
- Department of Biomedical Engineering, The University of Melbourne, Parkville, Victoria, Australia
- Centre for Neural Engineering, The University of Melbourne, Parkville, Victoria, Australia
| | - Hamish Meffin
- National Vision Research Institute, Australian College of Optometry, Carlton, Victoria, Australia
- ARC Centre of Excellence for Integrative Brain Function, Optometry & Vision Science, The University of Melbourne, Parkville, Victoria, Australia
| | - Anthony N. Burkitt
- Department of Biomedical Engineering, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
65
|
Axogenic mechanism enhances retinal ganglion cell excitability during early progression in glaucoma. Proc Natl Acad Sci U S A 2018; 115:E2393-E2402. [PMID: 29463759 PMCID: PMC5877940 DOI: 10.1073/pnas.1714888115] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Identifying new therapies for neurodegenerative disease requires understanding how neurons respond to stress and whether this response includes adaptation to slow progression. Because neurodegeneration affects both axons and dendrites, with their synaptic contacts, adaptation could involve both compartments. We investigated this question in experimental glaucoma, the world’s leading cause of irreversible vision loss. Glaucoma attacks retinal ganglion cell neurons and their axons, which comprise the optic nerve. We found that elevations in ocular pressure, a prominent risk factor for glaucoma, caused a paradoxical increase in ganglion cell excitability, including response to light, even in cells with substantial dendritic pruning. This adaptation arose from voltage-dependent mechanisms in the axon and may help maintain signaling to the brain to preserve vision. Diseases of the brain involve early axon dysfunction that often precedes outright degeneration. Pruning of dendrites and their synapses represents a potential driver of axonopathy by reducing activity. Optic nerve degeneration in glaucoma, the world’s leading cause of irreversible blindness, involves early stress to retinal ganglion cell (RGC) axons from sensitivity to intraocular pressure (IOP). This sensitivity also influences survival of RGC dendrites and excitatory synapses in the retina. Here we tested in individual RGCs identified by type the relationship between dendritic organization and axon signaling to light following modest, short-term elevations in pressure. We found dendritic pruning occurred early, by 2 wk of elevation, and independent of whether the RGC responded to light onset (ON cells) or offset (OFF cells). Pruning was similarly independent of ON and OFF in the DBA/2J mouse, a chronic glaucoma model. Paradoxically, all RGCs, even those with significant pruning, demonstrated a transient increase in axon firing in response to the preferred light stimulus that occurred on a backdrop of generally enhanced excitability. The increased response was not through conventional presynaptic signaling, but rather depended on voltage-sensitive sodium channels that increased transiently in the axon. Pruning, axon dysfunction, and deficits in visual acuity did not progress between 2 and 4 wk of elevation. These results suggest neurodegeneration in glaucoma involves an early axogenic response that counters IOP-related stress to excitatory dendritic architecture to slow progression and maintain signaling to the brain. Thus, short-term exposure to elevated IOP may precondition the neural system to further insult.
Collapse
|
66
|
Warwick RA, Kaushansky N, Sarid N, Golan A, Rivlin-Etzion M. Inhomogeneous Encoding of the Visual Field in the Mouse Retina. Curr Biol 2018; 28:655-665.e3. [PMID: 29456141 DOI: 10.1016/j.cub.2018.01.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/12/2017] [Accepted: 01/08/2018] [Indexed: 12/30/2022]
Abstract
Stimulus characteristics of the mouse's visual field differ above and below the skyline. Here, we show for the first time that retinal ganglion cells (RGCs), the output neurons of the retina, gradually change their functional properties along the ventral-dorsal axis to allow better representation of the different stimulus characteristics. We conducted two-photon targeted recordings of transient-Offα-RGCs and found that they gradually became more sustained along the ventral-dorsal axis, revealing >5-fold-longer duration responses in the dorsal retina. Using voltage-clamp recordings, pharmacology, and genetic manipulation, we demonstrated that the primary rod pathway underlies this variance. Our findings challenge the current belief that RGCs of the same subtype exhibit the same light responses, regardless of retinal location, and suggest that networks underlying RGC responses may change with retinal location to enable optimized sampling of the visual image.
Collapse
Affiliation(s)
- Rebekah A Warwick
- Department of Neurobiology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel
| | - Nathali Kaushansky
- Department of Neurobiology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel
| | - Nimrod Sarid
- Department of Neurobiology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel
| | - Amir Golan
- Department of Neurobiology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel
| | - Michal Rivlin-Etzion
- Department of Neurobiology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel.
| |
Collapse
|
67
|
Abstract
The mouse retina has a layered structure that is composed of five classes of neurons supported by Müller glial and pigment epithelial cells. Recent studies have made progress in the classification of bipolar and ganglion cells, and also in the wiring of rod-driven signaling, color coding, and directional selectivity. Molecular biological techniques, such as genetic manipulation, transcriptomics, and fluorescence imaging, have contributed a lot to these advancements. The mouse retina has consistently been an important experimental system for both basic and clinical neurosciences.
Collapse
Affiliation(s)
- Yoshihiko Tsukamoto
- Department of Biology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan.
| |
Collapse
|
68
|
Abstract
Recent technological advances have extended the range of analytic tools to very small samples. It is now possible to assay the transcriptome, and in some cases even the proteome, of single cells reliably. This allows addressing novel questions, such as the genotype/phenotype relationships of single neurons, heterogeneity within individual cells of the same type, or the basis of differential vulnerability to injury. An important prerequisite for these kinds of studies is the ability to isolate well-defined individual cells without contamination by adjacent tissue. In the retina and optic nerve, cells of different types and functions are closely intermingled, limiting the use of standard methods such as laser capture microdissection. Here, we describe a simple method to isolate morphologically intact cells from the retina and the optic nerve and discuss considerations in recognizing and isolating different cell types after dissociation.
Collapse
|
69
|
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
|
70
|
Liu J, Sanes JR. Cellular and Molecular Analysis of Dendritic Morphogenesis in a Retinal Cell Type That Senses Color Contrast and Ventral Motion. J Neurosci 2017; 37:12247-12262. [PMID: 29114073 PMCID: PMC5729193 DOI: 10.1523/jneurosci.2098-17.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/27/2017] [Accepted: 10/17/2017] [Indexed: 01/08/2023] Open
Abstract
As neuronal dendrites develop, they acquire cell-type-specific features including characteristic size, shape, arborization, location and synaptic patterns. These features, in turn, are major determinants of type-specific neuronal function. Because neuronal diversity complicates the task of relating developmental programs to adult structure and function, we analyzed dendritic morphogenesis in a single retinal ganglion cell (RGC) type in mouse called J-RGC. We documented the emergence of five dendritic features that underlie J-RGC physiology: (1) dendritic field size, which approximate receptive field size; (2) dendritic complexity, which affects how J-RGCs sample space; (3) asymmetry, which contributes to direction-selectivity; (4) restricted lamination within the inner plexiform layer (IPL), which renders J-RGCs responsive to light decrements; and (5) distribution of synaptic inputs, which generate a color-opponent receptive field. We found dendritic growth in J-RGCs is accompanied by a refinement in dendritic self-crossing. Asymmetry arises by a combination of selective pruning and elaboration, whereas laminar restriction results from biased outgrowth toward the outermost IPL. Interestingly, asymmetry develops in a protracted dorsoventral wave, whereas lamination does so in a rapid centrifugal wave. As arbors mature, they acquire excitatory and inhibitory synapses, with the latter forming first and being concentrated in proximal dendrites. Thus, distinct mechanisms operate in different spatiotemporal dimensions of J-RGC dendritic patterning to generate the substrate for specific patterns of synaptogenesis. Finally, we asked whether the defining molecular signature of J-RGCs, the adhesion molecule JAM-B, regulates morphogenesis, and showed that it promotes dendro-dendritic interactions. Our results reveal multiple mechanisms that shape a dendritic arbor.SIGNIFICANCE STATEMENT Visual perception begins in the retina, where distinct types of retinal ganglion cells (RGCs) are tuned to specific visual features such as direction of motion. The features to which each RGC type responds are determined largely by the number and type of synaptic inputs it receives, and these, in turn, are greatly influenced by the size, shape, arborization pattern, and location of its dendrites. We analyzed dendritic morphogenesis in a functionally characterized RGC type, the J-RGC, demonstrating distinct mechanisms that operate in different dimensions to generate the dendritic scaffold and synaptic patterns for feature detection. Our work elucidates cellular and molecular mechanisms that shape dendritic arbors and synaptic distribution, enabling J-RGC connectivity and thus, function.
Collapse
Affiliation(s)
- Jinyue Liu
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138, and
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts 02115
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138, and
| |
Collapse
|
71
|
Hardcastle K, Ganguli S, Giocomo LM. Cell types for our sense of location: where we are and where we are going. Nat Neurosci 2017; 20:1474-1482. [PMID: 29073649 PMCID: PMC6175666 DOI: 10.1038/nn.4654] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/23/2017] [Indexed: 12/15/2022]
Abstract
Technological advances in profiling cells along genetic, anatomical and physiological axes have fomented interest in identifying all neuronal cell types. This goal nears completion in specialized circuits such as the retina, while remaining more elusive in higher order cortical regions. We propose that this differential success of cell type identification may not simply reflect technological gaps in co-registering genetic, anatomical and physiological features in the cortex. Rather, we hypothesize it reflects evolutionarily driven differences in the computational principles governing specialized circuits versus more general-purpose learning machines. In this framework, we consider the question of cell types in medial entorhinal cortex (MEC), a region likely to be involved in memory and navigation. While MEC contains subsets of identifiable functionally defined cell types, recent work employing unbiased statistical methods and more diverse tasks reveals unsuspected heterogeneity and adaptivity in MEC firing patterns. This suggests MEC may operate more as a generalist circuit, obeying computational design principles resembling those governing other higher cortical regions.
Collapse
Affiliation(s)
| | - Surya Ganguli
- Department of Neurobiology, Stanford University
- Department of Applied Physics, Stanford University
| | | |
Collapse
|
72
|
Cowan CS, Sabharwal J, Wu SM. Space-time codependence of retinal ganglion cells can be explained by novel and separable components of their receptive fields. Physiol Rep 2017; 4:4/17/e12952. [PMID: 27604400 PMCID: PMC5027358 DOI: 10.14814/phy2.12952] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 08/10/2016] [Indexed: 11/24/2022] Open
Abstract
Reverse correlation methods such as spike‐triggered averaging consistently identify the spatial center in the linear receptive fields (RFs) of retinal ganglion cells (GCs). However, the spatial antagonistic surround observed in classical experiments has proven more elusive. Tests for the antagonistic surround have heretofore relied on models that make questionable simplifying assumptions such as space–time separability and radial homogeneity/symmetry. We circumvented these, along with other common assumptions, and observed a linear antagonistic surround in 754 of 805 mouse GCs. By characterizing the RF's space–time structure, we found the overall linear RF's inseparability could be accounted for both by tuning differences between the center and surround and differences within the surround. Finally, we applied this approach to characterize spatial asymmetry in the RF surround. These results shed new light on the spatiotemporal organization of GC linear RFs and highlight a major contributor to its inseparability.
Collapse
Affiliation(s)
- Cameron S Cowan
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas Department of Neuroscience, Baylor College of Medicine, Houston, Texas
| | - Jasdeep Sabharwal
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas Department of Neuroscience, Baylor College of Medicine, Houston, Texas Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas
| | - Samuel M Wu
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas Department of Neuroscience, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
73
|
Kay RB, Triplett JW. Visual Neurons in the Superior Colliculus Innervated by Islet2 + or Islet2 - Retinal Ganglion Cells Display Distinct Tuning Properties. Front Neural Circuits 2017; 11:73. [PMID: 29066954 PMCID: PMC5641327 DOI: 10.3389/fncir.2017.00073] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/19/2017] [Indexed: 11/20/2022] Open
Abstract
Throughout the visual system, different subtypes of neurons are tuned to distinct aspects of the visual scene, establishing parallel circuits. Defining the mechanisms by which such tuning arises has been a long-standing challenge for neuroscience. To investigate this, we have focused on the retina’s projection to the superior colliculus (SC), where multiple visual neuron subtypes have been described. The SC receives inputs from a variety of retinal ganglion cell (RGC) subtypes; however, which RGCs drive the tuning of different SC neurons remains unclear. Here, we pursued a genetic approach that allowed us to determine the tuning properties of neurons innervated by molecularly defined subpopulations of RGCs. In homozygous Islet2-EphA3 knock-in (Isl2EA3/EA3) mice, Isl2+ and Isl2− RGCs project to non-overlapping sub-regions of the SC. Based on molecular and anatomic data, we show that significantly more Isl2− RGCs are direction-selective (DS) in comparison with Isl2+ RGCs. Targeted recordings of visual responses from each SC sub-region in Isl2EA3/EA3 mice revealed that Isl2− RGC-innervated neurons were significantly more DS than those innervated by Isl2+ RGCs. Axis-selective (AS) neurons were found in both sub-regions, though AS neurons innervated by Isl2+ RGCs were more tightly tuned. Despite this segregation, DS and AS neurons innervated by Isl2+ or Isl2− RGCs did not differ in their spatial summation or spatial frequency (SF) tuning. Further, we did not observe alterations in receptive field (RF) size or structure of SC neurons innervated by Isl2+ or Isl2− RGCs. Together, these data show that innervation by Isl2+ and Isl2− RGCs results in distinct tuning in the SC and set the stage for future studies investigating the mechanisms by which these circuits are built.
Collapse
Affiliation(s)
- Rachel B Kay
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, United States
| | - Jason W Triplett
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, United States.,Department of Pediatrics, The George Washington University School of Medicine and Health Science, Washington, DC, United States
| |
Collapse
|
74
|
Groman-Lupa S, Adewumi J, Park KU, Brzezinski JA. The Transcription Factor Prdm16 Marks a Single Retinal Ganglion Cell Subtype in the Mouse Retina. Invest Ophthalmol Vis Sci 2017; 58:5421-5433. [PMID: 29053761 PMCID: PMC5656415 DOI: 10.1167/iovs.17-22442] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 09/20/2017] [Indexed: 12/04/2022] Open
Abstract
Purpose Retinal ganglion cells (RGC) can be categorized into roughly 30 distinct subtypes. How these subtypes develop is poorly understood, in part because few unique subtype markers have been characterized. We tested whether the Prdm16 transcription factor is expressed by RGCs as a class or within particular ganglion cell subtypes. Methods Embryonic and mature retinal sections and flatmount preparations were examined by immunohistochemistry for Prdm16 and several other cell type-specific markers. To visualize the morphology of Prdm16+ cells, we utilized Thy1-YFP-H transgenic mice, where a small random population of RGCs expresses yellow fluorescent protein (YFP) throughout the cytoplasm. Results Prdm16 was expressed in the retina starting late in embryogenesis. Prdm16+ cells coexpressed the RGC marker Brn3a. These cells were arranged in an evenly spaced pattern and accounted for 2% of all ganglion cells. Prdm16+ cells coexpressed parvalbumin, but not calretinin, melanopsin, Smi32, or CART. This combination of marker expression and morphology data from Thy1-YFP-H mice suggested that the Prdm16+ cells represented a single ganglion cell subtype. Prdm16 also marked vascular endothelial cells and mural cells of retinal arterioles. Conclusions A single subtype of ganglion cell appears to be uniquely marked by Prdm16 expression. While the precise identity of these ganglion cells is unclear, they most resemble the G9 subtype described by Völgyi and colleagues in 2009. Future studies are needed to determine the function of these ganglion cells and whether Prdm16 regulates their development.
Collapse
Affiliation(s)
- Sergio Groman-Lupa
- Department of Ophthalmology, University of Colorado Denver, Aurora, Colorado, United States
| | - Joseph Adewumi
- Department of Ophthalmology, University of Colorado Denver, Aurora, Colorado, United States
| | - Ko Uoon Park
- Department of Ophthalmology, University of Colorado Denver, Aurora, Colorado, United States
| | - Joseph A. Brzezinski
- Department of Ophthalmology, University of Colorado Denver, Aurora, Colorado, United States
| |
Collapse
|
75
|
3D Visualization of Individual Regenerating Retinal Ganglion Cell Axons Reveals Surprisingly Complex Growth Paths. eNeuro 2017; 4:eN-NWR-0093-17. [PMID: 28856242 PMCID: PMC5575138 DOI: 10.1523/eneuro.0093-17.2017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 07/28/2017] [Accepted: 08/01/2017] [Indexed: 01/28/2023] Open
Abstract
Retinal ganglion cells (RGCs), the sole output cells of the retina, are a heterogeneous population of neurons that project axons to visual targets in the brain. Like most CNS neurons, RGCs are considered incapable of mounting long distance axon regeneration. Using immunolabeling-enabled 3D imaging of solvent-cleared organs (iDISCO) in transgenic mice, we tracked the entire paths of individual RGC axons and show that adult RGCs are highly capable of spontaneous long-distance regeneration, even without any treatment. Our results show that the Thy1-H-YFP mouse sparsely labels RGCs, consisting predominantly of regeneration-competent α-type RGCs (αRGCs). Following optic nerve crush, many of the YFP-labeled RGC axons extend considerable distances proximal to the injury site with only a few penetrating through the lesion. This tortuous axon growth proximal to the lesion site is even more striking with intravitreal ciliary neurotrophic factor (CNTF) treatment. We further demonstrate that despite traveling more than 5 mm (i.e., a distance equal to the length of mouse optic nerve), many of these circuitous axons are confined to the injury area and fail to reach the brain. Our results re-evaluate the view that RGCs are naturally incapable of re-extending long axons, and shift the focus from promoting axon elongation, to understanding factors that prevent direct growth of axons through the lesion and the injured nerve.
Collapse
|
76
|
Selective Vulnerability of Specific Retinal Ganglion Cell Types and Synapses after Transient Ocular Hypertension. J Neurosci 2017; 36:9240-52. [PMID: 27581463 DOI: 10.1523/jneurosci.0940-16.2016] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 07/20/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Key issues concerning ganglion cell type-specific loss and synaptic changes in animal models of experimental glaucoma remain highly debated. Importantly, changes in the structure and function of various RGC types that occur early, within 14 d after acute, transient intraocular pressure elevation, have not been previously assessed. Using biolistic transfection of individual RGCs and multielectrode array recordings to measure light responses in mice, we examined the effects of laser-induced ocular hypertension on the structure and function of a subset of RGCs. Among the α-like RGCs studied, αOFF-transient RGCs exhibited higher rates of cell death, with corresponding reductions in dendritic area, dendritic complexity, and synapse density. Functionally, OFF-transient RGCs displayed decreases in spontaneous activity and receptive field size. In contrast, neither αOFF-sustained nor αON-sustained RGCs displayed decreases in light responses, although they did exhibit a decrease in excitatory postsynaptic sites, suggesting that synapse loss may be one of the earliest signs of degeneration. Interestingly, presynaptic ribbon density decreased to a greater degree in the OFF sublamina of the inner plexiform layer, corroborating the hypothesis that RGCs with dendrites stratifying in the OFF sublamina may be damaged early. Indeed, OFF arbors of ON-OFF RGCs lose complexity more rapidly than ON arbors. Our results reveal type-specific differences in RGC responses to injury with a selective vulnerability of αOFF-transient RGCs, and furthermore, an increased susceptibility of synapses in the OFF sublamina. The selective vulnerability of specific RGC types offers new avenues for the design of more sensitive functional tests and targeted neuroprotection. SIGNIFICANCE STATEMENT Conflicting reports regarding the selective vulnerability of specific retinal ganglion cell (RGC) types in glaucoma exist. We examine, for the first time, the effects of transient intraocular pressure elevation on the structure and function of various RGC types. Among the α-like RGCs studied, αOFF-transient RGCs are the most vulnerable to transient transient intraocular pressure elevation as measured by rates of cell death, morphologic alterations in dendrites and synapses, and physiological dysfunction. Specifically, we found that presynaptic ribbon density decreased to a greater degree in the OFF sublamina of the inner plexiform layer. Our results suggest selective vulnerability both of specific types of RGCs and of specific inner plexiform layer sublaminae, opening new avenues for identifying novel diagnostic and treatment targets in glaucoma.
Collapse
|
77
|
Peng YR, Tran NM, Krishnaswamy A, Kostadinov D, Martersteck EM, Sanes JR. Satb1 Regulates Contactin 5 to Pattern Dendrites of a Mammalian Retinal Ganglion Cell. Neuron 2017; 95:869-883.e6. [PMID: 28781169 DOI: 10.1016/j.neuron.2017.07.019] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 06/16/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022]
Abstract
The size and shape of dendritic arbors are prime determinants of neuronal connectivity and function. We asked how ON-OFF direction-selective ganglion cells (ooDSGCs) in mouse retina acquire their bistratified dendrites, in which responses to light onset and light offset are segregated to distinct strata. We found that the transcriptional regulator Satb1 is selectively expressed by ooDSGCs. In Satb1 mutant mice, ooDSGC dendrites lack ON arbors, and the cells selectively lose ON responses. Satb1 regulates expression of a homophilic adhesion molecule, Contactin 5 (Cntn5). Both Cntn5 and its co-receptor Caspr4 are expressed not only by ooDSGCs, but also by interneurons that form a scaffold on which ooDSGC ON dendrites fasciculate. Removing Cntn5 from either ooDSGCs or interneurons partially phenocopies Satb1 mutants, demonstrating that Satb1-dependent Cntn5 expression in ooDSGCs leads to branch-specific homophilic interactions with interneurons. Thus, Satb1 directs formation of a morphologically and functionally specialized compartment within a complex dendritic arbor.
Collapse
Affiliation(s)
- Yi-Rong Peng
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Nicholas M Tran
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Arjun Krishnaswamy
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Dimitar Kostadinov
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Emily M Martersteck
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
78
|
Krieger B, Qiao M, Rousso DL, Sanes JR, Meister M. Four alpha ganglion cell types in mouse retina: Function, structure, and molecular signatures. PLoS One 2017; 12:e0180091. [PMID: 28753612 PMCID: PMC5533432 DOI: 10.1371/journal.pone.0180091] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 06/11/2017] [Indexed: 11/18/2022] Open
Abstract
The retina communicates with the brain using ≥30 parallel channels, each carried by axons of distinct types of retinal ganglion cells. In every mammalian retina one finds so-called "alpha" ganglion cells (αRGCs), identified by their large cell bodies, stout axons, wide and mono-stratified dendritic fields, and high levels of neurofilament protein. In the mouse, three αRGC types have been described based on responses to light steps: On-sustained, Off-sustained, and Off-transient. Here we employed a transgenic mouse line that labels αRGCs in the live retina, allowing systematic targeted recordings. We characterize the three known types and identify a fourth, with On-transient responses. All four αRGC types share basic aspects of visual signaling, including a large receptive field center, a weak antagonistic surround, and absence of any direction selectivity. They also share a distinctive waveform of the action potential, faster than that of other RGC types. Morphologically, they differ in the level of dendritic stratification within the IPL, which accounts for their response properties. Molecularly, each type has a distinct signature. A comparison across mammals suggests a common theme, in which four large-bodied ganglion cell types split the visual signal into four channels arranged symmetrically with respect to polarity and kinetics.
Collapse
Affiliation(s)
- Brenna Krieger
- Harvard Biophysics Program, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mu Qiao
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - David L. Rousso
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Joshua R. Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, United States of America
- * E-mail: (JRS); (MM)
| | - Markus Meister
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
- * E-mail: (JRS); (MM)
| |
Collapse
|
79
|
Neuillé M, Cao Y, Caplette R, Guerrero-Given D, Thomas C, Kamasawa N, Sahel JA, Hamel CP, Audo I, Picaud S, Martemyanov KA, Zeitz C. LRIT3 Differentially Affects Connectivity and Synaptic Transmission of Cones to ON- and OFF-Bipolar Cells. Invest Ophthalmol Vis Sci 2017; 58:1768-1778. [PMID: 28334377 PMCID: PMC5374884 DOI: 10.1167/iovs.16-20745] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose Mutations in LRIT3 lead to complete congenital stationary night blindness (cCSNB). Using a cCSNB mouse model lacking Lrit3 (nob6), we recently have shown that LRIT3 has a role in the correct localization of TRPM1 (transient receptor potential melastatin 1) to the dendritic tips of ON-bipolar cells (BCs), contacting both rod and cone photoreceptors. Furthermore, postsynaptic clustering of other mGluR6 cascade components is selectively eliminated at the dendritic tips of cone ON-BCs. The purpose of this study was to further define the role of LRIT3 in structural and functional organization of cone synapses. Methods Exhaustive electroretinogram analysis was performed in a patient with LRIT3 mutations. Multielectrode array recordings were performed at the level of retinal ganglion cells in nob6 mice. Targeting of GluR1 and GluR5 at the dendritic tips of OFF-BCs in nob6 retinas was assessed by immunostaining and confocal microscopy. The ultrastructure of photoreceptor synapses was evaluated by electron microscopy in nob6 mice. Results The patient with LRIT3 mutations had a selective ON-BC dysfunction with relatively preserved OFF-BC responses. In nob6 mice, complete lack of ON-pathway function with robust, yet altered signaling processing in OFF-pathways was detected. Consistent with these observations, molecules essential for the OFF-BC signaling were normally targeted to the synapse. Finally, synaptic contacts made by ON-BC but not OFF-BC neurons with the cone pedicles were disorganized without ultrastructural alterations in cone terminals, horizontal cell processes, or synaptic ribbons. Conclusions These results suggest that LRIT3 is likely involved in coordination of the transsynaptic communication between cones and ON-BCs during synapse formation and function.
Collapse
Affiliation(s)
- Marion Neuillé
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| | - Yan Cao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida United States
| | - Romain Caplette
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| | | | - Connon Thomas
- Max Planck Florida Institute for Neuroscience, Jupiter, Florida United States
| | - Naomi Kamasawa
- Max Planck Florida Institute for Neuroscience, Jupiter, Florida United States
| | - José-Alain Sahel
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France 4CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC1423, Paris, France 5Institute of Ophthalmology, University College of London, London, United Kingdom 6Fondation Ophtalmologique Adolphe de Rothschild, Paris, France 8Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Christian P Hamel
- INSERM U583, Physiopathologie et thérapie des déficits sensoriels et moteurs, Institut des Neurosciences de Montpellier, Hôpital Saint-Eloi, Montpellier, France
| | - Isabelle Audo
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France 4CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC1423, Paris, France 5Institute of Ophthalmology, University College of London, London, United Kingdom
| | - Serge Picaud
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida United States
| | - Christina Zeitz
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| |
Collapse
|
80
|
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
|
81
|
Molecular codes for cell type specification in Brn3 retinal ganglion cells. Proc Natl Acad Sci U S A 2017; 114:E3974-E3983. [PMID: 28465430 DOI: 10.1073/pnas.1618551114] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Visual information is conveyed from the eye to the brain by distinct types of retinal ganglion cells (RGCs). It is largely unknown how RGCs acquire their defining morphological and physiological features and connect to upstream and downstream synaptic partners. The three Brn3/Pou4f transcription factors (TFs) participate in a combinatorial code for RGC type specification, but their exact molecular roles are still unclear. We use deep sequencing to define (i) transcriptomes of Brn3a- and/or Brn3b-positive RGCs, (ii) Brn3a- and/or Brn3b-dependent RGC transcripts, and (iii) transcriptomes of retinorecipient areas of the brain at developmental stages relevant for axon guidance, dendrite formation, and synaptogenesis. We reveal a combinatorial code of TFs, cell surface molecules, and determinants of neuronal morphology that is differentially expressed in specific RGC populations and selectively regulated by Brn3a and/or Brn3b. This comprehensive molecular code provides a basis for understanding neuronal cell type specification in RGCs.
Collapse
|
82
|
Li K, Zhong X, Yang S, Luo Z, Li K, Liu Y, Cai S, Gu H, Lu S, Zhang H, Wei Y, Zhuang J, Zhuo Y, Fan Z, Ge J. HiPSC-derived retinal ganglion cells grow dendritic arbors and functional axons on a tissue-engineered scaffold. Acta Biomater 2017; 54:117-127. [PMID: 28216299 DOI: 10.1016/j.actbio.2017.02.032] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 02/08/2017] [Accepted: 02/15/2017] [Indexed: 12/21/2022]
Abstract
Numerous therapeutic procedures in modern medical research rely on the use of tissue engineering for the treatment of retinal diseases. However, the cell source and the transplantation method are still a limitation. Previously, it was reported that a self-organizing three-dimensional neural retina can be induced from human-induced pluripotent stem cells (hiPSCs). In this study, we disclose the generation of retinal ganglion cells (RGCs) from the neural retina and their seeding on a biodegradable poly (lactic-co-glycolic acid) (PLGA) scaffold to create an engineered RGC-scaffold biomaterial. Moreover, we explored the dendritic arbor, branching point, functional axon and action potential of the biomaterial. Finally, the cell-scaffold was transplanted into the intraocular environment of rabbits and rhesus monkeys. STATEMENT OF SIGNIFICANCE As a part of the mammalian central nervous system (CNS), the retinal ganglion cell (RGC) shows little regenerative capacity. With the use of medical biomaterial for cells seeding and deliver, a new domain is now emerging that uses tissue engineering therapy for retinal disease. However, previous studies utilized RGCs from rodent model, which has limitations for human disease treatment. In the present study, we generated RGCs from hiPSCs-3D neural retina and then seeded these RGCs on PLGA scaffold to create an engineered RGC-scaffold biomaterial. Moreover, we assessed the transplantation method for biomaterial in vivo. Our study provides a technique to produce the engineered human RGC-scaffold biomaterial.
Collapse
Affiliation(s)
- Kangjun Li
- State Key Laboratory of Ophthalmology, Key Lab of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yet-Sen University, Guangzhou, Guangdong, China
| | - Xiufeng Zhong
- State Key Laboratory of Ophthalmology, Key Lab of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yet-Sen University, Guangzhou, Guangdong, China
| | - Sijing Yang
- State Key Laboratory of Ophthalmology, Key Lab of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yet-Sen University, Guangzhou, Guangdong, China
| | - Ziming Luo
- State Key Laboratory of Ophthalmology, Key Lab of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yet-Sen University, Guangzhou, Guangdong, China
| | - Kang Li
- State Key Laboratory of Ophthalmology, Key Lab of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yet-Sen University, Guangzhou, Guangdong, China
| | - Ying Liu
- State Key Laboratory of Ophthalmology, Key Lab of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yet-Sen University, Guangzhou, Guangdong, China
| | - Song Cai
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Yet-Sen University, Guangzhou, Guangdong, China
| | - Huaiyu Gu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Yet-Sen University, Guangzhou, Guangdong, China
| | - Shoutao Lu
- Bai Duoan Medical Equipment Company, Qihe, Shandong, China
| | - Haijun Zhang
- Bai Duoan Medical Equipment Company, Qihe, Shandong, China
| | - Yantao Wei
- State Key Laboratory of Ophthalmology, Key Lab of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yet-Sen University, Guangzhou, Guangdong, China
| | - Jing Zhuang
- State Key Laboratory of Ophthalmology, Key Lab of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yet-Sen University, Guangzhou, Guangdong, China
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Key Lab of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yet-Sen University, Guangzhou, Guangdong, China
| | - Zhigang Fan
- State Key Laboratory of Ophthalmology, Key Lab of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yet-Sen University, Guangzhou, Guangdong, China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Key Lab of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yet-Sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
83
|
Vidal-Sanz M, Galindo-Romero C, Valiente-Soriano FJ, Nadal-Nicolás FM, Ortin-Martinez A, Rovere G, Salinas-Navarro M, Lucas-Ruiz F, Sanchez-Migallon MC, Sobrado-Calvo P, Aviles-Trigueros M, Villegas-Pérez MP, Agudo-Barriuso M. Shared and Differential Retinal Responses against Optic Nerve Injury and Ocular Hypertension. Front Neurosci 2017; 11:235. [PMID: 28491019 PMCID: PMC5405145 DOI: 10.3389/fnins.2017.00235] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 04/07/2017] [Indexed: 12/05/2022] Open
Abstract
Glaucoma, one of the leading causes of blindness worldwide, affects primarily retinal ganglion cells (RGCs) and their axons. The pathophysiology of glaucoma is not fully understood, but it is currently believed that damage to RGC axons at the optic nerve head plays a major role. Rodent models to study glaucoma include those that mimic either ocular hypertension or optic nerve injury. Here we review the anatomical loss of the general population of RGCs (that express Brn3a; Brn3a+RGCs) and of the intrinsically photosensitive RGCs (that express melanopsin; m+RGCs) after chronic (LP-OHT) or acute (A-OHT) ocular hypertension and after complete intraorbital optic nerve transection (ONT) or crush (ONC). Our studies show that all of these insults trigger RGC death. Compared to Brn3a+RGCs, m+RGCs are more resilient to ONT, ONC, and A-OHT but not to LP-OHT. There are differences in the course of RGC loss both between these RGC types and among injuries. An important difference between the damage caused by ocular hypertension or optic nerve injury appears in the outer retina. Both axotomy and LP-OHT induce selective loss of RGCs but LP-OHT also induces a protracted loss of cone photoreceptors. This review outlines our current understanding of the anatomical changes occurring in rodent models of glaucoma and discusses the advantages of each one and their translational value.
Collapse
Affiliation(s)
- Manuel Vidal-Sanz
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la ArrixacaMurcia, Spain
| | - Caridad Galindo-Romero
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la ArrixacaMurcia, Spain
| | - Francisco J Valiente-Soriano
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la ArrixacaMurcia, Spain
| | - Francisco M Nadal-Nicolás
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la ArrixacaMurcia, Spain
| | - Arturo Ortin-Martinez
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la ArrixacaMurcia, Spain
| | - Giuseppe Rovere
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la ArrixacaMurcia, Spain
| | - Manuel Salinas-Navarro
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la ArrixacaMurcia, Spain
| | - Fernando Lucas-Ruiz
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la ArrixacaMurcia, Spain
| | - Maria C Sanchez-Migallon
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la ArrixacaMurcia, Spain
| | - Paloma Sobrado-Calvo
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la ArrixacaMurcia, Spain
| | - Marcelino Aviles-Trigueros
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la ArrixacaMurcia, Spain
| | - María P Villegas-Pérez
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la ArrixacaMurcia, Spain
| | - Marta Agudo-Barriuso
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la ArrixacaMurcia, Spain
| |
Collapse
|
84
|
Inman DM, Harun-Or-Rashid M. Metabolic Vulnerability in the Neurodegenerative Disease Glaucoma. Front Neurosci 2017; 11:146. [PMID: 28424571 PMCID: PMC5371671 DOI: 10.3389/fnins.2017.00146] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/08/2017] [Indexed: 12/14/2022] Open
Abstract
Axons can be several orders of magnitude longer than neural somas, presenting logistical difficulties in cargo trafficking and structural maintenance. Keeping the axon compartment well supplied with energy also presents a considerable challenge; even seemingly subtle modifications of metabolism can result in functional deficits and degeneration. Axons require a great deal of energy, up to 70% of all energy used by a neuron, just to maintain the resting membrane potential. Axonal energy, in the form of ATP, is generated primarily through oxidative phosphorylation in the mitochondria. In addition, glial cells contribute metabolic intermediates to axons at moments of high activity or according to need. Recent evidence suggests energy disruption is an early contributor to pathology in a wide variety of neurodegenerative disorders characterized by axonopathy. However, the degree to which the energy disruption is intrinsic to the axon vs. associated glia is not clear. This paper will review the role of energy availability and utilization in axon degeneration in glaucoma, a chronic axonopathy of the retinal projection.
Collapse
Affiliation(s)
- Denise M Inman
- Department of Pharmaceutical Sciences, Northeast Ohio Medical UniversityRootstown, OH, USA
| | | |
Collapse
|
85
|
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
|
86
|
Sabharwal J, Seilheimer RL, Cowan CS, Wu SM. The ON Crossover Circuitry Shapes Spatiotemporal Profile in the Center and Surround of Mouse OFF Retinal Ganglion Cells. Front Neural Circuits 2016; 10:106. [PMID: 28066192 PMCID: PMC5177742 DOI: 10.3389/fncir.2016.00106] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/07/2016] [Indexed: 11/22/2022] Open
Abstract
Retinal ganglion cells (RGCs) are often grouped based on their functional properties. Many of these functional properties, such as receptive field (RF) size, are driven by specific retinal circuits. In this report, we determined the role of the ON bipolar cell (BC) mediated crossover circuitry in shaping the center and surround of OFF RGCs. We recorded from a large population of mouse RGCs using a multielectrode array (MEA) while pharmacologically removing the ON BC-mediated crossover circuit. OFF sustained and transient responses to whole field stimuli are lost under scotopic conditions, but maintained under photopic conditions. Though photopic light responses were grossly maintained, we found that photopic light response properties were altered. Using linear RF mapping, we found a significant reduction in the antagonistic surround and a decrease in size of the RF center. Using a novel approach to separate the distinct temporal filters present in the RF center, we see that the crossover pathway contributes specifically to the sluggish antagonistic filter in the center. These results provide new insight into the role of crossover pathways in driving RGCs and also demonstrate that the distinct inputs driving the RF center can be isolated and assayed by RGC activity.
Collapse
Affiliation(s)
- Jasdeep Sabharwal
- Medical Scientist Training Program, Baylor College of MedicineHouston, TX, USA; Department of Neuroscience, Baylor College of MedicineHouston, TX, USA; Department of Ophthalmology, Baylor College of MedicineHouston, TX, USA
| | - Robert L Seilheimer
- Medical Scientist Training Program, Baylor College of MedicineHouston, TX, USA; Department of Ophthalmology, Baylor College of MedicineHouston, TX, USA
| | - Cameron S Cowan
- Department of Ophthalmology, Baylor College of Medicine Houston, TX, USA
| | - Samuel M Wu
- Department of Neuroscience, Baylor College of MedicineHouston, TX, USA; Department of Ophthalmology, Baylor College of MedicineHouston, TX, USA
| |
Collapse
|
87
|
Cell type-specific expression of FoxP2 in the ferret and mouse retina. Neurosci Res 2016; 117:1-13. [PMID: 27888071 DOI: 10.1016/j.neures.2016.11.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 11/16/2016] [Accepted: 11/18/2016] [Indexed: 12/25/2022]
Abstract
Although the anatomical and physiological properties of subtypes of retinal ganglion cells (RGCs) have been extensively investigated, their molecular properties are still unclear. Here, we examined the expression patterns of FoxP2 in the retina of ferrets and mice. We found that FoxP2 was expressed in small subsets of neurons in the adult ferret retina. FoxP2-positive neurons in the ganglion cell layer were divided into two groups. Large FoxP2-positive neurons expressed Brn3a and were retrogradely labeled with cholera toxin subunit B injected into the optic nerve, indicating that they are RGCs. The soma size and the projection pattern of FoxP2-positive RGCs were consistent with those of X cells. Because we previously reported that FoxP2 was selectively expressed in X cells in the ferret lateral geniculate nucleus (LGN), our findings indicate that FoxP2 is specifically expressed in the parvocellular pathway from the retina to the LGN. Small FoxP2-positive neurons were positive for GAD65/67, suggesting that they are GABAergic amacrine cells. Most Foxp2-positive cells were RGCs in the adult mouse retina. Dendritic morphological analyses suggested that Foxp2-positive RGCs included direction-selective RGCs in mice. Thus, our findings suggest that FoxP2 is expressed in specific subtypes of RGCs in the retina of ferrets and mice.
Collapse
|
88
|
Smith BJ, Côté PD, Tremblay F. Contribution of Na v1.8 sodium channels to retinal function. Neuroscience 2016; 340:279-290. [PMID: 27984182 DOI: 10.1016/j.neuroscience.2016.10.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 10/07/2016] [Accepted: 10/21/2016] [Indexed: 12/28/2022]
Abstract
We examined the contribution of the sodium channel isoform Nav1.8 to retinal function using the specific blocker A803467. We found that A803467 has little influence on the electroretinogram (ERG) a- and b-waves, but significantly reduces the oscillatory potentials (OPs) to 40-60% of their original amplitude, with significant changes in implicit time in the rod-driven range. To date, only two cell types were found in mouse to express Nav1.8; the starburst amacrine cells (SBACs), and a subtype of retinal ganglion cells (RGCs). When we recorded light responses from ganglion cells using a multielectrode array we found significant and opposing changes in two physiological groups of RGCs. ON-sustained cells showed significant decreases while transient ON-OFF cells showed significant increases. The effects on ON-OFF transient cells but not ON-sustained cells disappeared in the presence of an inhibitory cocktail. We have previously shown that RGCs have only a minor contribution to the OPs (Smith et al., 2014), therefore suggesting that SBACs might be a significant contributor to this ERG component. Targeting SBACs with the cholinergic neurotoxin ethylcholine mustard aziridinium (AF64A) caused a reduction in the amplitude of the OPs similar to A803467. Our results, both using the ERG and MEA recordings from RGCs, suggest that Nav1.8 plays a role in modulating specific aspects of the retinal physiology and that SBACs are a fundamental cellular contributor to the OPs in mice, a clear demonstration of the dichotomy between ERG b-wave and OPs.
Collapse
Affiliation(s)
- Benjamin J Smith
- Department of Biology, Dalhousie University, 1355 Oxford St., PO Box 15000, Halifax, NS B3H 4R2, Canada.
| | - Patrice D Côté
- Department of Biology, Dalhousie University, 1355 Oxford St., PO Box 15000, Halifax, NS B3H 4R2, Canada; Department of Ophthalmology and Visual Sciences, Dalhousie University, 1276 South Park St., PO Box 15000, Halifax, NS B3H 4R2, Canada.
| | - François Tremblay
- Department of Ophthalmology and Visual Sciences, Dalhousie University, 1276 South Park St., PO Box 15000, Halifax, NS B3H 4R2, Canada; Department of Physiology and Biophysics, Dalhousie University, 5850 College Street, PO Box 15000, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
89
|
Pan F, Toychiev A, Zhang Y, Atlasz T, Ramakrishnan H, Roy K, Völgyi B, Akopian A, Bloomfield SA. Inhibitory masking controls the threshold sensitivity of retinal ganglion cells. J Physiol 2016; 594:6679-6699. [PMID: 27350405 DOI: 10.1113/jp272267] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 06/23/2016] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Retinal ganglion cells (RGCs) in dark-adapted retinas show a range of threshold sensitivities spanning ∼3 log units of illuminance. Here, we show that the different threshold sensitivities of RGCs reflect an inhibitory mechanism that masks inputs from certain rod pathways. The masking inhibition is subserved by GABAC receptors, probably on bipolar cell axon terminals. The GABAergic masking inhibition appears independent of dopaminergic circuitry that has been shown also to affect RGC sensitivity. The results indicate a novel mechanism whereby inhibition controls the sensitivity of different cohorts of RGCs. This can limit and thereby ensure that appropriate signals are carried centrally in scotopic conditions when sensitivity rather than acuity is crucial. ABSTRACT The responses of rod photoreceptors, which subserve dim light vision, are carried through the retina by three independent pathways. These pathways carry signals with largely different sensitivities. Retinal ganglion cells (RGCs), the output neurons of the retina, show a wide range of sensitivities in the same dark-adapted conditions, suggesting a divergence of the rod pathways. However, this organization is not supported by the known synaptic morphology of the retina. Here, we tested an alternative idea that the rod pathways converge onto single RGCs, but inhibitory circuits selectively mask signals so that one pathway predominates. Indeed, we found that application of GABA receptor blockers increased the sensitivity of most RGCs by unmasking rod signals, which were suppressed. Our results indicate that inhibition controls the threshold responses of RGCs under dim ambient light. This mechanism can ensure that appropriate signals cross the bottleneck of the optic nerve in changing stimulus conditions.
Collapse
Affiliation(s)
- Feng Pan
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, NY, USA.,Current address: School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Abduqodir Toychiev
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, NY, USA
| | - Yi Zhang
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tamas Atlasz
- Department of Sport Biology, Janos Szentagothai Research Center, University of Pécs, Pécs, Hungary
| | | | - Kaushambi Roy
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, NY, USA
| | - Béla Völgyi
- Department of Sport Biology, Janos Szentagothai Research Center, University of Pécs, Pécs, Hungary.,Department of Experimental Zoology and Neurobiology, Janos Szentagothai Research Center, University of Pécs, Pécs, Hungary
| | - Abram Akopian
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, NY, USA
| | - Stewart A Bloomfield
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, NY, USA
| |
Collapse
|
90
|
Binley KE, Ng WS, Barde YA, Song B, Morgan JE. Brain-derived neurotrophic factor prevents dendritic retraction of adult mouse retinal ganglion cells. Eur J Neurosci 2016; 44:2028-39. [PMID: 27285957 PMCID: PMC4988502 DOI: 10.1111/ejn.13295] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 06/02/2016] [Accepted: 06/06/2016] [Indexed: 01/24/2023]
Abstract
We used cultured adult mouse retinae as a model system to follow and quantify the retraction of dendrites using diolistic labelling of retinal ganglion cells (RGCs) following explantation. Cell death was monitored in parallel by nuclear staining as ‘labelling’ with RGC and apoptotic markers was inconsistent and exceedingly difficult to quantify reliably. Nuclear staining allowed us to delineate a lengthy time window during which dendrite retraction can be monitored in the absence of RGC death. The addition of brain‐derived neurotrophic factor (BDNF) produced a marked reduction in dendritic degeneration, even when application was delayed for 3 days after retinal explantation. These results suggest that the delayed addition of trophic factors may be functionally beneficial before the loss of cell bodies in the course of conditions such as glaucoma.
Collapse
Affiliation(s)
- Kate E Binley
- School of Optometry and Vision Sciences, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Wai S Ng
- School of Optometry and Vision Sciences, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Yves-Alain Barde
- School of Biosciences, Sir Martin Evans Building, Cardiff University, Cardiff, UK
| | - Bing Song
- School of Dentistry, Cardiff University, Heath Park, Cardiff, UK
| | - James E Morgan
- School of Optometry and Vision Sciences, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, UK
| |
Collapse
|
91
|
Joly S, Pernet V. Sphingosine 1-phosphate receptor 1 is required for retinal ganglion cell survival after optic nerve trauma. J Neurochem 2016; 138:571-86. [PMID: 27309795 DOI: 10.1111/jnc.13701] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 06/11/2016] [Accepted: 06/12/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Sandrine Joly
- CUO-Recherche; Centre de recherche du CHU de Québec and Département d'ophtalmologie; Faculté de médecine; Université Laval; Quebec City Quebec Canada
| | - Vincent Pernet
- CUO-Recherche; Centre de recherche du CHU de Québec and Département d'ophtalmologie; Faculté de médecine; Université Laval; Quebec City Quebec Canada
| |
Collapse
|
92
|
Hedberg-Buenz A, Christopher MA, Lewis CJ, Fernandes KA, Dutca LM, Wang K, Scheetz TE, Abràmoff MD, Libby RT, Garvin MK, Anderson MG. Quantitative measurement of retinal ganglion cell populations via histology-based random forest classification. Exp Eye Res 2016; 146:370-385. [PMID: 26474494 PMCID: PMC4841761 DOI: 10.1016/j.exer.2015.09.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/02/2015] [Accepted: 09/20/2015] [Indexed: 01/27/2023]
Abstract
The inner surface of the retina contains a complex mixture of neurons, glia, and vasculature, including retinal ganglion cells (RGCs), the final output neurons of the retina and primary neurons that are damaged in several blinding diseases. The goal of the current work was two-fold: to assess the feasibility of using computer-assisted detection of nuclei and random forest classification to automate the quantification of RGCs in hematoxylin/eosin (H&E)-stained retinal whole-mounts; and if possible, to use the approach to examine how nuclear size influences disease susceptibility among RGC populations. To achieve this, data from RetFM-J, a semi-automated ImageJ-based module that detects, counts, and collects quantitative data on nuclei of H&E-stained whole-mounted retinas, were used in conjunction with a manually curated set of images to train a random forest classifier. To test performance, computer-derived outputs were compared to previously published features of several well-characterized mouse models of ophthalmic disease and their controls: normal C57BL/6J mice; Jun-sufficient and Jun-deficient mice subjected to controlled optic nerve crush (CONC); and DBA/2J mice with naturally occurring glaucoma. The result of these efforts was development of RetFM-Class, a command-line-based tool that uses data output from RetFM-J to perform random forest classification of cell type. Comparative testing revealed that manual and automated classifications by RetFM-Class correlated well, with 83.2% classification accuracy for RGCs. Automated characterization of C57BL/6J retinas predicted 54,642 RGCs per normal retina, and identified a 48.3% Jun-dependent loss of cells at 35 days post CONC and a 71.2% loss of RGCs among 16-month-old DBA/2J mice with glaucoma. Output from automated analyses was used to compare nuclear area among large numbers of RGCs from DBA/2J mice (n = 127,361). In aged DBA/2J mice with glaucoma, RetFM-Class detected a decrease in median and mean nucleus size of cells classified into the RGC category, as did an independent confirmation study using manual measurements of nuclear area demarcated by BRN3A-immunoreactivity. In conclusion, we have demonstrated that histology-based random forest classification is feasible and can be utilized to study RGCs in a high-throughput fashion. Despite having some limitations, this approach demonstrated a significant association between the size of the RGC nucleus and the DBA/2J form of glaucoma.
Collapse
Affiliation(s)
- Adam Hedberg-Buenz
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA; Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Mark A Christopher
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Carly J Lewis
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Kimberly A Fernandes
- Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Laura M Dutca
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA; Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA
| | - Kai Wang
- Department of Biostatistics, University of Iowa, Iowa City, IA 52242, USA
| | - Todd E Scheetz
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA; Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA
| | - Michael D Abràmoff
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA; Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA; Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA; Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Richard T Libby
- Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Mona K Garvin
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA; Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Michael G Anderson
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA; Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
93
|
Haeussner E, Schmitz C, Frank HG, Edler von Koch F. Novel 3D light microscopic analysis of IUGR placentas points to a morphological correlate of compensated ischemic placental disease in humans. Sci Rep 2016; 6:24004. [PMID: 27045698 PMCID: PMC4820778 DOI: 10.1038/srep24004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 03/18/2016] [Indexed: 11/30/2022] Open
Abstract
The villous tree of the human placenta is a complex three-dimensional (3D) structure with branches and nodes at the feto-maternal border in the key area of gas and nutrient exchange. Recently we introduced a novel, computer-assisted 3D light microscopic method that enables 3D topological analysis of branching patterns of the human placental villous tree. In the present study we applied this novel method to the 3D architecture of peripheral villous trees of placentas from patients with intrauterine growth retardation (IUGR placentas), a severe obstetric syndrome. We found that the mean branching angle of branches in terminal positions of the villous trees was significantly different statistically between IUGR placentas and clinically normal placentas. Furthermore, the mean tortuosity of branches of villous trees in directly preterminal positions was significantly different statistically between IUGR placentas and clinically normal placentas. We show that these differences can be interpreted as consequences of morphological adaptation of villous trees between IUGR placentas and clinically normal placentas, and may have important consequences for the understanding of the morphological correlates of the efficiency of the placental villous tree and their influence on fetal development.
Collapse
Affiliation(s)
- Eva Haeussner
- Department of Anatomy II, LMU Munich, Pettenkoferstr. 11, 80336 Munich, Germany
| | - Christoph Schmitz
- Department of Anatomy II, LMU Munich, Pettenkoferstr. 11, 80336 Munich, Germany
| | - Hans-Georg Frank
- Department of Anatomy II, LMU Munich, Pettenkoferstr. 11, 80336 Munich, Germany
| | - Franz Edler von Koch
- Clinic for Obstetrics and Gynecology Dritter Orden, Menzinger Str. 44, 80638 Munich, Germany
| |
Collapse
|
94
|
Gu YN, Lee ES, Jeon CJ. Types and density of calbindin D28k-immunoreactive ganglion cells in mouse retina. Exp Eye Res 2016; 145:327-336. [PMID: 26874036 DOI: 10.1016/j.exer.2016.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 02/04/2016] [Accepted: 02/04/2016] [Indexed: 10/22/2022]
Abstract
Single-cell injection after immunocytochemistry is a reliable technique for classifying neurons by their morphological structure and their expression of a particular protein. The aim of the present study was to classify the morphological types of calbindin D28k-immunoreactive retinal ganglion cells in the mouse using single-cell injection after immunocytochemistry, to estimate the density of calbindin D28k-immunoreactive retinal ganglion cells in the mouse retina. Calbindin D28k is an important calcium-binding protein that is widely expressed in the central nervous system. Calbindin D28k-immunoreactive retinal ganglion cells were identified by immunocytochemistry and then iontophoretically injected with the lipophilic dye, DiI. Subsequently, the injected cells were imaged by confocal microscopy to classify calbindin D28k-immunoreactive retinal ganglion cells based on their dendritic ramification depth within the inner plexiform layer, field size, and morphology. The cells were heterogeneous in morphology: monostratified or bistratified, with small to large dendritic field size and sparse to dense dendritic arbors. At least 10 different morphological types (CB1-CB10) of calbindin D28k-immunoreactive retinal ganglion cells were found in the mouse retina. The density of each cell type was quite variable (1.98-23.76%). The density of calbindin D28k-immunoreactive cells in the ganglion cell layer of the mouse retina was 562 cells/mm(2), 8.18% of calbindin D28k-immunoreactive cells were axon-less displaced amacrine cells, 91.82% were retinal ganglion cells, and approximately 18.17% of mouse retinal ganglion cells expressed calbindin D28k. The selective expression of calbindin D28k in cells with different morphologies may provide important data for further physiological studies of the mouse retina.
Collapse
Affiliation(s)
- Ya-Nan Gu
- Department of Biology, School of Life Sciences, BK 21 Plus KNU Creative BioResearch Group, College of Natural Sciences, and Brain Science and Engineering Institute, Kyungpook National University, Deagu, 41566, South Korea
| | - Eun-Shil Lee
- Department of Biology, School of Life Sciences, BK 21 Plus KNU Creative BioResearch Group, College of Natural Sciences, and Brain Science and Engineering Institute, Kyungpook National University, Deagu, 41566, South Korea
| | - Chang-Jin Jeon
- Department of Biology, School of Life Sciences, BK 21 Plus KNU Creative BioResearch Group, College of Natural Sciences, and Brain Science and Engineering Institute, Kyungpook National University, Deagu, 41566, South Korea.
| |
Collapse
|
95
|
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
|
96
|
Venugopalan P, Wang Y, Nguyen T, Huang A, Muller KJ, Goldberg JL. Transplanted neurons integrate into adult retinas and respond to light. Nat Commun 2016; 7:10472. [PMID: 26843334 PMCID: PMC4742891 DOI: 10.1038/ncomms10472] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 12/16/2015] [Indexed: 12/12/2022] Open
Abstract
Retinal ganglion cells (RGCs) degenerate in diseases like glaucoma and are not replaced in adult mammals. Here we investigate whether transplanted RGCs can integrate into the mature retina. We have transplanted GFP-labelled RGCs into uninjured rat retinas in vivo by intravitreal injection. Transplanted RGCs acquire the general morphology of endogenous RGCs, with axons orienting towards the optic nerve head of the host retina and dendrites growing into the inner plexiform layer. Preliminary data show in some cases GFP(+) axons extending within the host optic nerves and optic tract, reaching usual synaptic targets in the brain, including the lateral geniculate nucleus and superior colliculus. Electrophysiological recordings from transplanted RGCs demonstrate the cells' electrical excitability and light responses similar to host ON, ON-OFF and OFF RGCs, although less rapid and with greater adaptation. These data present a promising approach to develop cell replacement strategies in diseased retinas with degenerating RGCs.
Collapse
Affiliation(s)
- Praseeda Venugopalan
- Neuroscience Program, University of Miami, Miami, Florida 33136, USA.,Shiley Eye Center, University of California, San Diego, California 92093, USA
| | - Yan Wang
- Shiley Eye Center, University of California, San Diego, California 92093, USA
| | - Tu Nguyen
- Shiley Eye Center, University of California, San Diego, California 92093, USA
| | - Abigail Huang
- Shiley Eye Center, University of California, San Diego, California 92093, USA
| | - Kenneth J Muller
- Neuroscience Program, University of Miami, Miami, Florida 33136, USA.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Jeffrey L Goldberg
- Neuroscience Program, University of Miami, Miami, Florida 33136, USA.,Shiley Eye Center, University of California, San Diego, California 92093, USA.,Byers Eye Institute, Department of Ophthalmology, Stanford University, Stanford, California 94303, USA
| |
Collapse
|
97
|
Wang J, Jacoby R, Wu SM. Physiological and morphological characterization of ganglion cells in the salamander retina. Vision Res 2016; 119:60-72. [PMID: 26731645 PMCID: PMC4774266 DOI: 10.1016/j.visres.2015.12.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 10/21/2015] [Accepted: 12/23/2015] [Indexed: 11/26/2022]
Abstract
Retinal ganglion cells (RGCs) integrate visual information from the retina and transmit collective signals to the brain. A systematic investigation of functional and morphological characteristics of various types of RGCs is important to comprehensively understand how the visual system encodes and transmits information via various RGC pathways. This study evaluated both physiological and morphological properties of 67 RGCs in dark-adapted flat-mounted salamander retina by examining light-evoked cation and chloride current responses via voltage-clamp recordings and visualizing morphology by Lucifer yellow fluorescence with a confocal microscope. Six groups of RGCs were described: asymmetrical ON-OFF RGCs, symmetrical ON RGCs, OFF RGCs, and narrow-, medium- and wide-field ON-OFF RGCs. Dendritic field diameters of RGCs ranged 102-490 μm: narrow field (<200 μm, 31% of RGCs), medium field (200-300 μm, 45%) and wide field (>300 μm, 24%). Dendritic ramification patterns of RGCs agree with the sublamina A/B rule. 34% of RGCs were monostratified, 24% bistratified and 42% diffusely stratified. 70% of ON RGCs and OFF RGCs were monostratified. Wide-field RGCs were diffusely stratified. 82% of RGCs generated light-evoked ON-OFF responses, while 11% generated ON responses and 7% OFF responses. Response sensitivity analysis suggested that some RGCs obtained separated rod/cone bipolar cell inputs whereas others obtained mixed bipolar cell inputs. 25% of neurons in the RGC layer were displaced amacrine cells. Although more types may be defined by more refined classification criteria, this report is to incorporate more physiological properties into RGC classification.
Collapse
Affiliation(s)
- Jing Wang
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, United States.
| | - Roy Jacoby
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, United States
| | - Samuel M Wu
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, United States
| |
Collapse
|
98
|
A randomized, placebo-controlled trial of the benzoquinone idebenone in a mouse model of OPA1-related dominant optic atrophy reveals a limited therapeutic effect on retinal ganglion cell dendropathy and visual function. Neuroscience 2016; 319:92-106. [PMID: 26820596 DOI: 10.1016/j.neuroscience.2016.01.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 01/15/2016] [Accepted: 01/19/2016] [Indexed: 12/19/2022]
Abstract
Dominant optic atrophy (DOA) arises from mutations in the OPA1 gene that promotes fusion of the inner mitochondrial membrane and plays a role in maintaining ATP levels. Patients display optic disc pallor, retinal ganglion cell (RGC) loss and bilaterally reduced vision. We report a randomized, placebo-controlled trial of idebenone at 2000 mg/kg/day in 56 Opa1 mutant mice (B6;C3-Opa1(Q285STOP)), with RGC dendropathy and visual loss, and 63 wildtype mice. We assessed cellular responses in the retina, brain and liver and RGC morphology, by diolistic labeling, Sholl analysis and quantification of dendritic morphometric features. Vision was assessed by optokinetic responses. ATP levels were raised by 0.57 nmol/mg (97.73%, p=0.035) in brain from idebenone-treated Opa1 mutant mice, but in the liver there was an 80.35% (p=0.011) increase in oxidative damage. NQO1 expression in Opa1 mutant mice was reduced in the brain (to 30.5%, p=0.002) but not in retina, and neither expression level was induced by idebenone. ON-center RGCs failed to show major recovery, other than improvements in secondary dendritic length (by 53.89%, p=0.052) and dendritic territory (by 2.22 × 10(4) μm(2) or 90.24%, p=0.074). An improvement in optokinetic response was observed (by 12.2 ± 3.2s, p=0.003), but this effect was not sustained over time. OFF-center RGCs from idebenone-treated wildtype mice showed shrinkage in total dendritic length by 2.40 mm (48.05%, p=0.025) and a 47.37% diminished Sholl profile (p=0.029). Visual function in wildtype idebenone-treated mice was impaired (2.9 fewer head turns than placebo, p=0.007). Idebenone appears largely ineffective in protecting Opa1 heterozygous RGCs from dendropathy. The detrimental effect of idebenone in wildtype mice has not been previously observed and raises some concerns.
Collapse
|
99
|
The functional diversity of retinal ganglion cells in the mouse. Nature 2016; 529:345-50. [PMID: 26735013 PMCID: PMC4724341 DOI: 10.1038/nature16468] [Citation(s) in RCA: 625] [Impact Index Per Article: 69.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 11/17/2015] [Indexed: 01/26/2023]
Abstract
In the vertebrate visual system, all output of the retina is carried by retinal ganglion cells. Each type encodes distinct visual features in parallel for transmission to the brain. How many such “output channels” exist and what each encodes is an area of intense debate. In mouse, anatomical estimates range between 15–20 channels, and only a handful are functionally understood. Combining two-photon calcium imaging to obtain dense retinal recordings and unsupervised clustering of the resulting sample of >11,000 cells, we here show that the mouse retina harbours substantially more than 30 functional output channels. These include all known and several new ganglion cell types, as verified by genetic and anatomical criteria. Therefore, information channels from the mouse’s eye to the mouse’s brain are considerably more diverse than shown thus far by anatomical studies, suggesting an encoding strategy resembling that used in state-of-the-art artificial vision systems.
Collapse
|
100
|
Kameneva T, Maturana MI, Hadjinicolaou AE, Cloherty SL, Ibbotson MR, Grayden DB, Burkitt AN, Meffin H. Retinal ganglion cells: mechanisms underlying depolarization block and differential responses to high frequency electrical stimulation of ON and OFF cells. J Neural Eng 2016; 13:016017. [DOI: 10.1088/1741-2560/13/1/016017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|