1
|
Zhao W, Lin Z, Zhang L, Lin X, Wang J, Xu S, Chen E, Wu T, Ye Y, Chen H. Bioinspired Three-Mode Photosensitive Synaptic LED for Optical Information Processing. NANO LETTERS 2024; 24:14109-14117. [PMID: 39466915 DOI: 10.1021/acs.nanolett.4c04444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Inspired by human sense organs, AI is advancing toward multimodal perception, with display technology evolving into intelligent human-computer interaction tools. However, hardware networks with multimodal responses connected by different devices bring problems such as delayed information transfer and inefficiency. Thus, an innovative three-mode photosensitive synaptic LED (PSSL) is first proposed by adding a photosensitive layer indacenodithiophene-benzothiadiazole (IDTBT) to the quantum-dot light-emitting diode (QLED), switched by changing the bias voltage. The self-powered PSSL has a photoresponse range from 310 nm to 808 nm (ultraviolet-near-infrared, UV-NIR). The device exhibits a bipolar response under red and UV light at 1 V. When the voltage reaches the turn-on voltage, the PSSL device turns into a neuromorphic LED, exhibiting conductivity enhancement under red-light irradiation and suppression under UV-light irradiation. As a result, the PSSLs are expected to be applied in the field of optical encryption communication and in neuromorphic display.
Collapse
Affiliation(s)
- Wenxiao Zhao
- Institute of Optoelectronic Display, National & Local United Engineering Lab of Flat Panel Display Technology, Fuzhou University, Fuzhou 350002, China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350100, China
| | - Zexi Lin
- Institute of Optoelectronic Display, National & Local United Engineering Lab of Flat Panel Display Technology, Fuzhou University, Fuzhou 350002, China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350100, China
| | - Liyan Zhang
- Institute of Optoelectronic Display, National & Local United Engineering Lab of Flat Panel Display Technology, Fuzhou University, Fuzhou 350002, China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350100, China
| | - Xing Lin
- Institute of Optoelectronic Display, National & Local United Engineering Lab of Flat Panel Display Technology, Fuzhou University, Fuzhou 350002, China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350100, China
| | - Jiawei Wang
- Institute of Optoelectronic Display, National & Local United Engineering Lab of Flat Panel Display Technology, Fuzhou University, Fuzhou 350002, China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350100, China
| | - Sheng Xu
- Institute of Optoelectronic Display, National & Local United Engineering Lab of Flat Panel Display Technology, Fuzhou University, Fuzhou 350002, China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350100, China
| | - Enguo Chen
- Institute of Optoelectronic Display, National & Local United Engineering Lab of Flat Panel Display Technology, Fuzhou University, Fuzhou 350002, China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350100, China
| | - Teng Wu
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350100, China
| | - Yun Ye
- Institute of Optoelectronic Display, National & Local United Engineering Lab of Flat Panel Display Technology, Fuzhou University, Fuzhou 350002, China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350100, China
| | - Huipeng Chen
- Institute of Optoelectronic Display, National & Local United Engineering Lab of Flat Panel Display Technology, Fuzhou University, Fuzhou 350002, China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350100, China
| |
Collapse
|
2
|
Toma K, Zhao M, Zhang S, Wang F, Graham HK, Zou J, Modgil S, Shang WH, Tsai NY, Cai Z, Liu L, Hong G, Kriegstein AR, Hu Y, Körbelin J, Zhang R, Liao YJ, Kim TN, Ye X, Duan X. Perivascular neurons instruct 3D vascular lattice formation via neurovascular contact. Cell 2024; 187:2767-2784.e23. [PMID: 38733989 PMCID: PMC11223890 DOI: 10.1016/j.cell.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/15/2024] [Accepted: 04/11/2024] [Indexed: 05/13/2024]
Abstract
The vasculature of the central nervous system is a 3D lattice composed of laminar vascular beds interconnected by penetrating vessels. The mechanisms controlling 3D lattice network formation remain largely unknown. Combining viral labeling, genetic marking, and single-cell profiling in the mouse retina, we discovered a perivascular neuronal subset, annotated as Fam19a4/Nts-positive retinal ganglion cells (Fam19a4/Nts-RGCs), directly contacting the vasculature with perisomatic endfeet. Developmental ablation of Fam19a4/Nts-RGCs led to disoriented growth of penetrating vessels near the ganglion cell layer (GCL), leading to a disorganized 3D vascular lattice. We identified enriched PIEZO2 expression in Fam19a4/Nts-RGCs. Piezo2 loss from all retinal neurons or Fam19a4/Nts-RGCs abolished the direct neurovascular contacts and phenocopied the Fam19a4/Nts-RGC ablation deficits. The defective vascular structure led to reduced capillary perfusion and sensitized the retina to ischemic insults. Furthermore, we uncovered a Piezo2-dependent perivascular granule cell subset for cerebellar vascular patterning, indicating neuronal Piezo2-dependent 3D vascular patterning in the brain.
Collapse
Affiliation(s)
- Kenichi Toma
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Mengya Zhao
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Shaobo Zhang
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Fei Wang
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Hannah K Graham
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Jun Zou
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| | - Shweta Modgil
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Wenhao H Shang
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Nicole Y Tsai
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Zhishun Cai
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Liping Liu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Guiying Hong
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Arnold R Kriegstein
- Department of Neurology and The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jakob Körbelin
- ENDomics Lab, Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ruobing Zhang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Yaping Joyce Liao
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Tyson N Kim
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Xin Ye
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA.
| | - Xin Duan
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA; Department of Physiology and Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
3
|
James RE, Hamilton NR, Huffman LN, Pasterkamp J, Goff LA, Kolodkin AL. Semaphorin 6A in Retinal Ganglion Cells Regulates Functional Specialization of the Inner Retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.18.567662. [PMID: 38014224 PMCID: PMC10680864 DOI: 10.1101/2023.11.18.567662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
To form functional circuits, neurons must settle in their appropriate cellular locations and then project and elaborate neurites to contact their target synaptic neuropils. Laminar organization within the vertebrate retinal inner plexiform layer (IPL) facilitates pre- and postsynaptic neurite targeting, yet, the precise mechanisms underlying establishment of functional IPL subdomains are not well understood. Here we explore mechanisms defining the compartmentalization of OFF and ON neurites generally, and OFF and ON direction-selective neurites specifically, within the developing IPL. We show that semaphorin 6A (Sema6A), a repulsive axon guidance cue, is required for delineation of OFF versus ON circuits within the IPL: in the Sema6a null IPL, the boundary between OFF and ON domains is blurred. Furthermore, Sema6A expressed by retinal ganglion cells (RGCs) directs laminar segregation of OFF and ON starburst amacrine cell (SAC) dendritic scaffolds, which themselves serve as a substrate upon which other retinal neurites elaborate. These results demonstrate for the first time that RGCs, the first neuron-type born within the retina, play an active role in functional specialization of the IPL. Retinal ganglion cell-dependent regulation of OFF and ON starburst amacrine cell dendritic scaffold segregation prevents blurring of OFF versus ON functional domains in the murine inner plexiform layer.
Collapse
|
4
|
Wang B, Zhang Y. Asymmetric connections with starburst amacrine cells underlie the upward motion selectivity of J-type retinal ganglion cells. PLoS Biol 2023; 21:e3002301. [PMID: 37721959 PMCID: PMC10538761 DOI: 10.1371/journal.pbio.3002301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 09/28/2023] [Accepted: 08/17/2023] [Indexed: 09/20/2023] Open
Abstract
Motion is an important aspect of visual information. The directions of visual motion are encoded in the retina by direction-selective ganglion cells (DSGCs). ON-OFF DSGCs and ON DSGCs co-stratify with starburst amacrine cells (SACs) in the inner plexiform layer and depend on SACs for their direction selectivity. J-type retinal ganglion cells (J-RGCs), a type of OFF DSGCs in the mouse retina, on the other hand, do not co-stratify with SACs, and how direction selectivity in J-RGCs emerges has not been understood. Here, we report that both the excitatory and inhibitory synaptic inputs to J-RGCs are direction-selective (DS), with the inhibitory inputs playing a more important role for direction selectivity. The DS inhibitory inputs come from SACs, and the functional connections between J-RGCs and SACs are spatially asymmetric. Thus, J-RGCs and SACs form functionally important synaptic contacts even though their dendritic arbors show little overlap. These findings underscore the need to look beyond the neurons' stratification patterns in retinal circuit studies. Our results also highlight the critical role of SACs for retinal direction selectivity.
Collapse
Affiliation(s)
- Bo Wang
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yifeng Zhang
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
5
|
Prieur DS, Francius C, Gaspar P, Mason CA, Rebsam A. Semaphorin-6D and Plexin-A1 Act in a Non-Cell-Autonomous Manner to Position and Target Retinal Ganglion Cell Axons. J Neurosci 2023; 43:5769-5778. [PMID: 37344233 PMCID: PMC10423046 DOI: 10.1523/jneurosci.0072-22.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/04/2023] [Accepted: 05/01/2023] [Indexed: 06/23/2023] Open
Abstract
Semaphorins and Plexins form ligand/receptor pairs that are crucial for a wide range of developmental processes from cell proliferation to axon guidance. The ability of semaphorins to act both as signaling receptors and ligands yields a multitude of responses. Here, we describe a novel role for Semaphorin-6D (Sema6D) and Plexin-A1 in the positioning and targeting of retinogeniculate axons. In Plexin-A1 or Sema6D mutant mice of either sex, the optic tract courses through, rather than along, the border of the dorsal lateral geniculate nucleus (dLGN), and some retinal axons ectopically arborize adjacent and lateral to the optic tract rather than defasciculating and entering the target region. We find that Sema6D and Plexin-A1 act together in a dose-dependent manner, as the number of the ectopic retinal projections is altered in proportion to the level of Sema6D or Plexin-A1 expression. Moreover, using retinal in utero electroporation of Sema6D or Plexin-A1 shRNA, we show that Sema6D and Plexin-A1 are both required in retinal ganglion cells for axon positioning and targeting. Strikingly, nonelectroporated retinal ganglion cell axons also mistarget in the tract region, indicating that Sema6D and Plexin-A1 can act non-cell-autonomously, potentially through axon-axon interactions. These data provide novel evidence for a dose-dependent and non-cell-autonomous role for Sema6D and Plexin-A1 in retinal axon organization in the optic tract and dLGN.SIGNIFICANCE STATEMENT Before innervating their central brain targets, retinal ganglion cell axons fasciculate in the optic tract and then branch and arborize in their target areas. Upon deletion of the guidance molecules Plexin-A1 or Semaphorin-6D, the optic tract becomes disorganized near and extends within the dorsal lateral geniculate nucleus. In addition, some retinal axons form ectopic aggregates within the defasciculated tract. Sema6D and Plexin-A1 act together as a receptor-ligand pair in a dose-dependent manner, and non-cell-autonomously, to produce this developmental aberration. Such a phenotype highlights an underappreciated role for axon guidance molecules in tract cohesion and appropriate defasciculation near, and arborization within, targets.
Collapse
Affiliation(s)
- Delphine S Prieur
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-S 839, Paris, 75005, France
- Sorbonne Université, Paris, 75005, France
- Institut du Fer à Moulin, Paris, 75005, France
| | - Cédric Francius
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-S 839, Paris, 75005, France
- Sorbonne Université, Paris, 75005, France
- Institut du Fer à Moulin, Paris, 75005, France
| | - Patricia Gaspar
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-S 839, Paris, 75005, France
- Sorbonne Université, Paris, 75005, France
- Institut du Fer à Moulin, Paris, 75005, France
| | - Carol A Mason
- Departments of Pathology and Cell Biology, Neuroscience, and Ophthalmology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027
| | - Alexandra Rebsam
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-S 839, Paris, 75005, France
- Sorbonne Université, Paris, 75005, France
- Institut du Fer à Moulin, Paris, 75005, France
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Institut de la Vision, Paris, F-75012, France
| |
Collapse
|
6
|
Goswami-Sewell D, Bagnetto C, Gomez CC, Anderson JT, Maheshwari A, Zuniga-Sanchez E. βII-Spectrin Is Required for Synaptic Positioning during Retinal Development. J Neurosci 2023; 43:5277-5289. [PMID: 37369589 PMCID: PMC10359034 DOI: 10.1523/jneurosci.0063-23.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 06/29/2023] Open
Abstract
Neural circuit assembly is a multistep process where synaptic partners are often born at distinct developmental stages, and yet they must find each other and form precise synaptic connections with one another. This developmental process often relies on late-born neurons extending their processes to the appropriate layer to find and make synaptic connections to their early-born targets. The molecular mechanism responsible for the integration of late-born neurons into an emerging neural circuit remains unclear. Here, we uncovered a new role for the cytoskeletal protein βII-spectrin in properly positioning presynaptic and postsynaptic neurons to the developing synaptic layer. Loss of βII-spectrin disrupts retinal lamination, leads to synaptic connectivity defects, and results in impaired visual function in both male and female mice. Together, these findings highlight a new function of βII-spectrin in assembling neural circuits in the mouse outer retina.SIGNIFICANCE STATEMENT Neurons that assemble into a functional circuit are often integrated at different developmental time points. However, the molecular mechanism that guides the precise positioning of neuronal processes to the correct layer for synapse formation is relatively unknown. Here, we show a new role for the cytoskeletal scaffolding protein, βII-spectrin in the developing retina. βII-spectrin is required to position presynaptic and postsynaptic neurons to the nascent synaptic layer in the mouse outer retina. Loss of βII-spectrin disrupts positioning of neuronal processes, alters synaptic connectivity, and impairs visual function.
Collapse
Affiliation(s)
| | - Caitlin Bagnetto
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas 77030
| | - Cesiah C Gomez
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas 77030
| | - Joseph T Anderson
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas 77030
| | - Akash Maheshwari
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas 77030
| | - Elizabeth Zuniga-Sanchez
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas 77030
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
7
|
Min KW, Kim N, Lee JH, Sung Y, Kim M, Lee EJ, Kim JM, Kim JH, Lee J, Cho W, Yang JM, Kim N, Kim J, Lee CJ, Park YG, Lee SH, Lee HW, Kim JW. Visuomotor anomalies in achiasmatic mice expressing a transfer-defective Vax1 mutant. Exp Mol Med 2023; 55:385-400. [PMID: 36737666 PMCID: PMC9981622 DOI: 10.1038/s12276-023-00930-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 02/05/2023] Open
Abstract
In binocular animals that exhibit stereoscopic visual responses, the axons of retinal ganglion cells (RGCs) connect to brain areas bilaterally by forming a commissure called the optic chiasm (OC). Ventral anterior homeobox 1 (Vax1) contributes to the formation of the OC, acting endogenously in optic pathway cells and exogenously in growing RGC axons. Here, we generated Vax1AA/AA mice expressing the Vax1AA mutant, which is incapable of intercellular transfer. We found that RGC axons cannot take up Vax1AA protein from the Vax1AA/AA mouse optic stalk (OS) and grow slowly to arrive at the hypothalamus at a late stage. The RGC axons of Vax1AA/AA mice connect exclusively to ipsilateral brain areas after failing to access the midline, resulting in reduced visual acuity and abnormal oculomotor responses. Overall, our study provides physiological evidence for the necessity of intercellular transfer of Vax1 and the importance of the bilateral RGC axon projection in proper visuomotor responses.
Collapse
Affiliation(s)
- Kwang Wook Min
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Namsuk Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea.,Neurovascular Unit, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Jae Hoon Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Younghoon Sung
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Museong Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Eun Jung Lee
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Jong-Myeong Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Jae-Hyun Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Jaeyoung Lee
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Wonjin Cho
- Department of Bio & Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Jee Myung Yang
- Department of Ophthalmology, Dongguk University Ilsan Hospital, Dongguk-ro 27, Ilsandong-gu, Goyang, Gyeong-gi, Republic of Korea
| | - Nury Kim
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Jaehoon Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Young-Gyun Park
- Department of Bio & Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Seung-Hee Lee
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jin Woo Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
8
|
Whitney IE, Butrus S, Dyer MA, Rieke F, Sanes JR, Shekhar K. Vision-Dependent and -Independent Molecular Maturation of Mouse Retinal Ganglion Cells. Neuroscience 2023; 508:153-173. [PMID: 35870562 PMCID: PMC10809145 DOI: 10.1016/j.neuroscience.2022.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/20/2022] [Accepted: 07/13/2022] [Indexed: 01/17/2023]
Abstract
The development and connectivity of retinal ganglion cells (RGCs), the retina's sole output neurons, are patterned by activity-independent transcriptional programs and activity-dependent remodeling. To inventory the molecular correlates of these influences, we applied high-throughput single-cell RNA sequencing (scRNA-seq) to mouse RGCs at six embryonic and postnatal ages. We identified temporally regulated modules of genes that correlate with, and likely regulate, multiple phases of RGC development, ranging from differentiation and axon guidance to synaptic recognition and refinement. Some of these genes are expressed broadly while others, including key transcription factors and recognition molecules, are selectively expressed by one or a few of the 45 transcriptomically distinct types defined previously in adult mice. Next, we used these results as a foundation to analyze the transcriptomes of RGCs in mice lacking visual experience due to dark rearing from birth or to mutations that ablate either bipolar or photoreceptor cells. 98.5% of visually deprived (VD) RGCs could be unequivocally assigned to a single RGC type based on their transcriptional profiles, demonstrating that visual activity is dispensable for acquisition and maintenance of RGC type identity. However, visual deprivation significantly reduced the transcriptomic distinctions among RGC types, implying that activity is required for complete RGC maturation or maintenance. Consistent with this notion, transcriptomic alternations in VD RGCs significantly overlapped with gene modules found in developing RGCs. Our results provide a resource for mechanistic analyses of RGC differentiation and maturation, and for investigating the role of activity in these processes.
Collapse
Affiliation(s)
- Irene E Whitney
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Salwan Butrus
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Karthik Shekhar
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Helen Wills Neuroscience Institute, California Institute for Quantitative Biosciences, QB3, Center for Computational Biology, University of California, Berkeley, CA 94720, USA; Biological Systems Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
9
|
Cao Y, Fajardo D, Guerrero-Given D, Samuel MA, Ohtsuka T, Boye SE, Kamasawa N, Martemyanov KA. Post-developmental plasticity of the primary rod pathway allows restoration of visually guided behaviors. Curr Biol 2022; 32:4783-4796.e3. [PMID: 36179691 PMCID: PMC9691582 DOI: 10.1016/j.cub.2022.09.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/17/2022] [Accepted: 09/08/2022] [Indexed: 01/24/2023]
Abstract
The formation of neural circuits occurs in a programmed fashion, but proper activity in the circuit is essential for refining the organization necessary for driving complex behavioral tasks. In the retina, sensory deprivation during the critical period of development is well known to perturb the organization of the visual circuit making the animals unable to use vision for behavior. However, the extent of plasticity, molecular factors involved, and malleability of individual channels in the circuit to manipulations outside of the critical period are not well understood. In this study, we selectively disconnected and reconnected rod photoreceptors in mature animals after completion of the retina circuit development. We found that introducing synaptic rod photoreceptor input post-developmentally allowed their integration into the circuit both anatomically and functionally. Remarkably, adult mice with newly integrated rod photoreceptors gained high-sensitivity vision, even when it was absent from birth. These observations reveal plasticity of the retina circuit organization after closure of the critical period and encourage the development of vision restoration strategies for congenital blinding disorders.
Collapse
Affiliation(s)
- Yan Cao
- Department of Neuroscience, UF Scripps Biomedical Research, Jupiter, FL 33458, USA
| | - Diego Fajardo
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Debbie Guerrero-Given
- The Imaging Center, Electron Microscopy Core Facility, Max Planck Florida Institute, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Melanie A Samuel
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Toshihisa Ohtsuka
- Department of Biochemistry, Graduate School of Medicine, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Shannon E Boye
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Naomi Kamasawa
- The Imaging Center, Electron Microscopy Core Facility, Max Planck Florida Institute, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, UF Scripps Biomedical Research, Jupiter, FL 33458, USA.
| |
Collapse
|
10
|
Activation of HIF-1α/VEGF-A pathway by deferoxamine ameliorates retinal hypoxia in a rat subarachnoid hemorrhage model. Neuroreport 2022; 33:690-696. [DOI: 10.1097/wnr.0000000000001835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
11
|
Zhang C, Hellevik A, Takeuchi S, Wong RO. Hierarchical partner selection shapes rod-cone pathway specificity in the inner retina. iScience 2022; 25:105032. [PMID: 36117987 PMCID: PMC9474917 DOI: 10.1016/j.isci.2022.105032] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/11/2022] [Accepted: 08/23/2022] [Indexed: 12/04/2022] Open
Abstract
Neurons form stereotyped microcircuits that underlie specific functions. In the vertebrate retina, the primary rod and cone pathways that convey dim and bright light signals, respectively, exhibit distinct wiring patterns. Rod and cone pathways are thought to be assembled separately during development. However, using correlative fluorescence imaging and serial electron microscopy, we show here that cross-pathway interactions are involved to achieve pathway-specific connectivity within the inner retina. We found that A17 amacrine cells, a rod pathway-specific cellular component, heavily bias their synaptogenesis with rod bipolar cells (RBCs) but increase their connectivity with cone bipolar cells (CBCs) when RBCs are largely ablated. This cross-pathway synaptic plasticity occurs during synaptogenesis and is triggered even on partial loss of RBCs. Thus, A17 cells adopt a hierarchical approach in selecting postsynaptic partners from functionally distinct pathways (RBC>CBC), in which contact and/or synaptogenesis with preferred partners (RBCs) influences connectivity with less-preferred partners (CBCs).
Collapse
Affiliation(s)
- Chi Zhang
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Ayana Hellevik
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Shunsuke Takeuchi
- Department of Biological Sciences, University of Tokyo, Tokyo 113-0033, Japan
| | - Rachel O. Wong
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
12
|
Gao S, Xiang SY, Song ZW, Han YN, Zhang YN, Hao Y. Motion detection and direction recognition in a photonic spiking neural network consisting of VCSELs-SA. OPTICS EXPRESS 2022; 30:31701-31713. [PMID: 36242247 DOI: 10.1364/oe.465653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/04/2022] [Indexed: 06/16/2023]
Abstract
Motion detection and direction recognition are two important fundamental visual functions among the many cognitive functions performed by the human visual system. The retina and visual cortex are indispensable for composing the visual nervous system. The retina is responsible for transmitting electrical signals converted from light signals to the visual cortex of the brain. We propose a photonic spiking neural network (SNN) based on vertical-cavity surface-emitting lasers with an embedding saturable absorber (VCSELs-SA) with temporal integration effects, and demonstrate that the motion detection and direction recognition tasks can be solved by mimicking the visual nervous system. Simulation results reveal that the proposed photonic SNN with a modified supervised algorithm combining the tempotron and the STDP rule can correctly detect the motion and recognize the direction angles, and is robust to time jitter and the current difference between VCSEL-SAs. The proposed approach adopts a low-power photonic neuromorphic system for real-time information processing, which provides theoretical support for the large-scale application of hardware photonic SNN in the future.
Collapse
|
13
|
Abstract
Neuroplasticity, i.e., the modifiability of the brain, is different in development and adulthood. The first includes changes in: (i) neurogenesis and control of neuron number; (ii) neuronal migration; (iii) differentiation of the somato-dendritic and axonal phenotypes; (iv) formation of connections; (v) cytoarchitectonic differentiation. These changes are often interrelated and can lead to: (vi) system-wide modifications of brain structure as well as to (vii) acquisition of specific functions such as ocular dominance or language. Myelination appears to be plastic both in development and adulthood, at least, in rodents. Adult neuroplasticity is limited, and is mainly expressed as changes in the strength of excitatory and inhibitory synapses while the attempts to regenerate connections have met with limited success. The outcomes of neuroplasticity are not necessarily adaptive, but can also be the cause of neurological and psychiatric pathologies.
Collapse
|
14
|
A cell-ECM mechanism for connecting the ipsilateral eye to the brain. Proc Natl Acad Sci U S A 2021; 118:2104343118. [PMID: 34654745 PMCID: PMC8545493 DOI: 10.1073/pnas.2104343118] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Abstract
Distinct features of the visual world are transmitted from the retina to the brain through anatomically segregated circuits. Despite this being an organizing principle of visual pathways in mammals, we lack an understanding of the signaling mechanisms guiding axons of different types of retinal neurons into segregated layers of brain regions. We explore this question by identifying how axons from the ipsilateral retina innervate a specific lamina of the superior colliculus. Our studies reveal a unique cell–extracellular matrix recognition mechanism that specifies precise targeting of these axons to the superior colliculus. Loss of this mechanism not only resulted in the absence of this eye-specific visual circuit, but it led to an impairment of innate predatory visual behavior as well. Information about features in the visual world is parsed by circuits in the retina and is then transmitted to the brain by distinct subtypes of retinal ganglion cells (RGCs). Axons from RGC subtypes are stratified in retinorecipient brain nuclei, such as the superior colliculus (SC), to provide a segregated relay of parallel and feature-specific visual streams. Here, we sought to identify the molecular mechanisms that direct the stereotyped laminar targeting of these axons. We focused on ipsilateral-projecting subtypes of RGCs (ipsiRGCs) whose axons target a deep SC sublamina. We identified an extracellular glycoprotein, Nephronectin (NPNT), whose expression is restricted to this ipsiRGC-targeted sublamina. SC-derived NPNT and integrin receptors expressed by ipsiRGCs are both required for the targeting of ipsiRGC axons to the deep sublamina of SC. Thus, a cell–extracellular matrix (ECM) recognition mechanism specifies precise laminar targeting of ipsiRGC axons and the assembly of eye-specific parallel visual pathways.
Collapse
|
15
|
Rochon PL, Theriault C, Rangel Olguin AG, Krishnaswamy A. The cell adhesion molecule Sdk1 shapes assembly of a retinal circuit that detects localized edges. eLife 2021; 10:e70870. [PMID: 34545809 PMCID: PMC8514235 DOI: 10.7554/elife.70870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/11/2021] [Indexed: 01/10/2023] Open
Abstract
Nearly 50 different mouse retinal ganglion cell (RGC) types sample the visual scene for distinct features. RGC feature selectivity arises from their synapses with a specific subset of amacrine (AC) and bipolar cell (BC) types, but how RGC dendrites arborize and collect input from these specific subsets remains poorly understood. Here we examine the hypothesis that RGCs employ molecular recognition systems to meet this challenge. By combining calcium imaging and type-specific histological stains, we define a family of circuits that express the recognition molecule Sidekick-1 (Sdk1), which include a novel RGC type (S1-RGC) that responds to local edges. Genetic and physiological studies revealed that Sdk1 loss selectively disrupts S1-RGC visual responses, which result from a loss of excitatory and inhibitory inputs and selective dendritic deficits on this neuron. We conclude that Sdk1 shapes dendrite growth and wiring to help S1-RGCs become feature selective.
Collapse
|
16
|
Development of the vertebrate retinal direction-selective circuit. Dev Biol 2021; 477:273-283. [PMID: 34118273 DOI: 10.1016/j.ydbio.2021.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/17/2021] [Accepted: 06/04/2021] [Indexed: 01/13/2023]
Abstract
The vertebrate retina contains an array of neural circuits that detect distinct features in visual space. Direction-selective (DS) circuits are an evolutionarily conserved retinal circuit motif - from zebrafish to rodents to primates - specialized for motion detection. During retinal development, neuronal subtypes that wire DS circuits form exquisitely precise connections with each other to shape the output of retinal ganglion cells tuned for specific speeds and directions of motion. In this review, we follow the chronology of DS circuit development in the vertebrate retina, including the cellular, molecular, and activity-dependent mechanisms that regulate the formation of DS circuits, from cell birth and migration to synapse formation and refinement. We highlight recent findings that identify genetic programs critical for specifying neuronal subtypes within DS circuits and molecular interactions essential for responses along the cardinal axes of motion. Finally, we discuss the roles of DS circuits in visual behavior and in certain human visual disease conditions. As one of the best-characterized circuits in the vertebrate retina, DS circuits represent an ideal model system for studying the development of neural connectivity at the level of individual genes, cells, and behavior.
Collapse
|
17
|
Pourhoseini S, Goswami-Sewell D, Zuniga-Sanchez E. Neurofascin Is a Novel Component of Rod Photoreceptor Synapses in the Outer Retina. Front Neural Circuits 2021; 15:635849. [PMID: 33643000 PMCID: PMC7902911 DOI: 10.3389/fncir.2021.635849] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/20/2021] [Indexed: 11/13/2022] Open
Abstract
Neural circuit formation is an intricate and complex process where multiple neuron types must come together to form synaptic connections at a precise location and time. How this process is orchestrated during development remains poorly understood. Cell adhesion molecules are known to play a pivotal role in assembling neural circuits. They serve as recognition molecules between corresponding synaptic partners. In this study, we identified a new player in assembling neural circuits in the outer retina, the L1-family cell adhesion molecule Neurofascin (Nfasc). Our data reveals Nfasc is expressed in the synaptic layer where photoreceptors make synaptic connections to their respective partners. A closer examination of Nfasc expression shows high levels of expression in rod bipolars but not in cone bipolars. Disruption of Nfasc using a conditional knockout allele results in selective loss of pre- and post-synaptic proteins in the rod synaptic layer but not in the cone synaptic layer. Electron microscopic analysis confirms that indeed there are abnormal synaptic structures with less dendrites of rod bipolars innervating rod terminals in loss of Nfasc animals. Consistent with these findings, we also observe a decrease in rod-driven retinal responses with disruption of Nfasc function but not in cone-driven responses. Taken together, our data suggest a new role of Nfasc in rod synapses within the mouse outer retina.
Collapse
Affiliation(s)
- Sahar Pourhoseini
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | | | - Elizabeth Zuniga-Sanchez
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
18
|
Zhaba WD, Deji QZ, Deng HJ, Han YL, Gao SQ, Liu XL, Zhou ML. Retinal hypoxia after experimental subarachnoid hemorrhage. Neurosci Lett 2020; 742:135554. [PMID: 33352284 DOI: 10.1016/j.neulet.2020.135554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/17/2020] [Accepted: 12/02/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND AND PURPOSE The patients who survive subarachnoid hemorrhage (SAH) often have long-term neurological complications. There are no reports about the pathological change of retina after SAH. METHODS An experimental model of SAH was established by injecting autologous blood into the prechiasmatic cistern of Sprague-Dawley rats. Hematoxylin and eosin (HE) staining was performed to show the alternation of morphology in retina after SAH. To detect the retinal new vessels (NVs), CD31 was labelled by immunofluorescence and immunohistochemistry. The time-course expressions of vascular endothelial growth factor (VEGF)-A and hypoxia-inducible factor-1α (HIF-1 α) was also revealed by Western blot analysis. RESULTS A clear reduction of retinal ganglion cells (RGCs) was noticed after SAH. The immunofluorescence and immunohistochemical staining of CD31 reveals a large number of NVs in RGC layer after SAH compared with the normal controls. The level of VEGF-A in the retina after SAH was increased and peaked at 12h and 14 d. The expression of HIF-1α in the retina increased as early as 3 h after SAH, reached a peak at 12 h after SAH. CONCLUSIONS The results showed that SAH induced the retina hypoxia resulting in the reduction of RGCs, increase of NVs and activation of NVs related HIF-1α/VEGF-A pathway.
Collapse
Affiliation(s)
- Wang-Dui Zhaba
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qu-Zhen Deji
- Department of Ophthalmology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hong-Ji Deng
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Yan-Ling Han
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Sheng-Qing Gao
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xi-Lin Liu
- Graduate School of Wannan Medical College, Wuhu, Anhui, China
| | - Meng-Liang Zhou
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
19
|
Rangel Olguin AG, Rochon PL, Krishnaswamy A. New Optical Tools to Study Neural Circuit Assembly in the Retina. Front Neural Circuits 2020; 14:44. [PMID: 32848633 PMCID: PMC7424070 DOI: 10.3389/fncir.2020.00044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/23/2020] [Indexed: 12/17/2022] Open
Abstract
During development, neurons navigate a tangled thicket of thousands of axons and dendrites to synapse with just a few specific targets. This phenomenon termed wiring specificity, is critical to the assembly of neural circuits and the way neurons manage this feat is only now becoming clear. Recent studies in the mouse retina are shedding new insight into this process. They show that specific wiring arises through a series of stages that include: directed axonal and dendritic growth, the formation of neuropil layers, positioning of such layers, and matching of co-laminar synaptic partners. Each stage appears to be directed by a distinct family of recognition molecules, suggesting that the combinatorial expression of such family members might act as a blueprint for retinal connectivity. By reviewing the evidence in support of each stage, and by considering their underlying molecular mechanisms, we attempt to synthesize these results into a wiring model which generates testable predictions for future studies. Finally, we conclude by highlighting new optical methods that could be used to address such predictions and gain further insight into this fundamental process.
Collapse
|
20
|
Graham HK, Duan X. Molecular mechanisms regulating synaptic specificity and retinal circuit formation. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e379. [PMID: 32267095 DOI: 10.1002/wdev.379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 03/13/2020] [Accepted: 03/19/2020] [Indexed: 12/28/2022]
Abstract
The central nervous system (CNS) is composed of precisely assembled circuits which support a variety of physiological functions and behaviors. These circuits include multiple subtypes of neurons with unique morphologies, electrical properties, and molecular identities. How these component parts are precisely wired-up has been a topic of great interest to the field of developmental neurobiology and has implications for our understanding of the etiology of many neurological disorders and mental illnesses. To date, many molecules involved in synaptic choice and specificity have been identified, including members of several families of cell-adhesion molecules (CAMs), which are cell-surface molecules that mediate cell-cell contacts and subsequent intracellular signaling. One favored hypothesis is that unique expression patterns of CAMs define specific neuronal subtype populations and determine compatible pre- and postsynaptic neuronal partners based on the expression of these unique CAMs. The mouse retina has served as a beautiful model for investigations into mammalian CAM interactions due to its well-defined neuronal subtypes and distinct circuits. Moreover, the retina is readily amenable to visualization of circuit organization and electrophysiological measurement of circuit function. The advent of recent genetic, genomic, and imaging technologies has opened the field up to large-scale, unbiased approaches for identification of new molecular determinants of synaptic specificity. Thus, building on the foundation of work reviewed here, we can expect rapid expansion of the field, harnessing the mouse retina as a model to understand the molecular basis for synaptic specificity and functional circuit assembly. This article is categorized under: Nervous System Development > Vertebrates: General Principles Nervous System Development > Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Hannah K Graham
- Department of Ophthalmology, University of California San Francisco, San Francisco, California, USA.,Neuroscience Graduate Program, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Xin Duan
- Department of Ophthalmology, University of California San Francisco, San Francisco, California, USA.,Neuroscience Graduate Program, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA.,Department of Physiology, University of California San Francisco, San Francisco, California, USA.,Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
21
|
Wakeham CM, Ren G, Morgans CW. Expression and distribution of trophoblast glycoprotein in the mouse retina. J Comp Neurol 2020; 528:1660-1671. [PMID: 31891182 DOI: 10.1002/cne.24850] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 02/03/2023]
Abstract
We recently identified the leucine-rich repeat (LRR) adhesion protein, trophoblast glycoprotein (TPBG), as a novel PKCα-dependent phosphoprotein in retinal rod bipolar cells (RBCs). Since TPBG has not been thoroughly examined in the retina, this study characterizes the localization and expression patterns of TPBG in the developing and adult mouse retina using two antibodies, one against the N-terminal LRR domain and the other against the C-terminal PDZ-interacting motif. Both antibodies labeled RBC dendrites in the outer plexiform layer and axon terminals in the IPL, as well as a putative amacrine cell with their cell bodies in the inner nuclear layer (INL) and a dense layer in the middle of the inner plexiform layer (IPL). In live transfected HEK293 cells, TPBG was localized to the plasma membrane with the N-terminal LRR domain facing the extracellular space. TPBG immunofluorescence in RBCs was strongly altered by the loss of TRPM1 in the adult retina, with significantly less dendritic and axon terminal labeling in TRPM1 knockout compared to wild type, despite no change in total TPBG detected by immunoblotting. During retinal development, TPBG expression increases dramatically just prior to eye opening with a time course closely correlated with that of TRPM1 expression. In the retina, LRR proteins have been implicated in the development and maintenance of functional bipolar cell synapses, and TPBG may play a similar role in RBCs.
Collapse
Affiliation(s)
- Colin M Wakeham
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon
| | - Gaoying Ren
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon
| | - Catherine W Morgans
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
22
|
Coate TM, Scott MK, Gurjar MC. Current concepts in cochlear ribbon synapse formation. Synapse 2019; 73:e22087. [PMID: 30592086 PMCID: PMC6573016 DOI: 10.1002/syn.22087] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022]
Abstract
In mammals, hair cells and spiral ganglion neurons (SGNs) in the cochlea together are sophisticated "sensorineural" structures that transduce auditory information from the outside world into the brain. Hair cells and SGNs are joined by glutamatergic ribbon-type synapses composed of a molecular machinery rivaling in complexity the mechanoelectric transduction components found at the apical side of the hair cell. The cochlear hair cell ribbon synapse has received much attention lately because of recent and important findings related to its damage (sometimes termed "synaptopathy") as a result of noise overexposure. During development, ribbon synapses between type I SGNs and inner hair cells form in the time window between birth and hearing onset and is a process coordinated with type I SGN myelination, spontaneous activity, synaptic pruning, and innervation by efferents. In this review, we highlight new findings regarding the diversity of type I SGNs and inner hair cell synapses, and the molecular mechanisms of selective hair cell targeting. Also discussed are cell adhesion molecules and protein constituents of the ribbon synapse, and how these factors participate in ribbon synapse formation. We also note interesting new insights into the morphological development of type II SGNs, and the potential for cochlear macrophages as important players in protecting SGNs. We also address recent studies demonstrating that the structural and physiological profiles of the type I SGNs do not reach full maturity until weeks after hearing onset, suggesting a protracted development that is likely modulated by activity.
Collapse
Affiliation(s)
- Thomas M. Coate
- Georgetown University, Department of Biology, 37th and O St. NW. Washington, DC. 20007. USA
| | - M. Katie Scott
- Department of Biological Sciences and Purdue Institute of Integrative Neuroscience, Purdue University, West Lafayette, Indiana 47907. USA
| | - Mansa C. Gurjar
- Georgetown University, Department of Biology, 37th and O St. NW. Washington, DC. 20007. USA
| |
Collapse
|
23
|
Fritzsch B, Elliott KL, Pavlinkova G. Primary sensory map formations reflect unique needs and molecular cues specific to each sensory system. F1000Res 2019; 8:F1000 Faculty Rev-345. [PMID: 30984379 PMCID: PMC6439788 DOI: 10.12688/f1000research.17717.1] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/21/2019] [Indexed: 12/21/2022] Open
Abstract
Interaction with the world around us requires extracting meaningful signals to guide behavior. Each of the six mammalian senses (olfaction, vision, somatosensation, hearing, balance, and taste) has a unique primary map that extracts sense-specific information. Sensory systems in the periphery and their target neurons in the central nervous system develop independently and must develop specific connections for proper sensory processing. In addition, the regulation of sensory map formation is independent of and prior to central target neuronal development in several maps. This review provides an overview of the current level of understanding of primary map formation of the six mammalian senses. Cell cycle exit, combined with incompletely understood molecules and their regulation, provides chemoaffinity-mediated primary maps that are further refined by activity. The interplay between cell cycle exit, molecular guidance, and activity-mediated refinement is the basis of dominance stripes after redundant organ transplantations in the visual and balance system. A more advanced level of understanding of primary map formation could benefit ongoing restoration attempts of impaired senses by guiding proper functional connection formations of restored sensory organs with their central nervous system targets.
Collapse
Affiliation(s)
- Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, USA
| | | | - Gabriela Pavlinkova
- Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic
| |
Collapse
|
24
|
Yamagata M, Sanes JR. Expression and Roles of the Immunoglobulin Superfamily Recognition Molecule Sidekick1 in Mouse Retina. Front Mol Neurosci 2019; 11:485. [PMID: 30687002 PMCID: PMC6333872 DOI: 10.3389/fnmol.2018.00485] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/13/2018] [Indexed: 12/24/2022] Open
Abstract
Processes of >100 types of interneurons (bipolar and amacrine cells) and projection neurons (retinal ganglion cells, RGCs) form specific and stereotyped patterns of connections in the inner plexiform layer (IPL) of the mouse retina. Four closely related homophilic immunoglobulin superfamily recognition molecules (Sidekick [Sdk] 1, Sdk 2, Dscam, and DscamL1) have been shown to play roles in patterning neuronal arbors and connections in chick retina, and all but Sdk1 have been shown to play related roles in mice. Here, we compare patterns of Sdk1 and Sdk2 expression in mouse retina and use genetic methods to assess roles of Sdk1. In adult retina, 3 neuronal types express sdk1 but not sdk2 at detectable levels, 5 express sdk2 but not sdk1 and 3 express both. Patterns of gene expression and protein localization at or near synapses are established during the first postnatal week. Dendrites of amacrine cells and RGCs that express sdk1 but not sdk2 arborize in the same narrow stratum in the center of the IPL. In the absence of Sdk1, this laminar restriction is degraded. Overexpression of sdk1 in developing cells that normally express sdk2 reorients their dendrites to resemble those of endogenously Sdk1-positive cells, indicating that Sdk1 plays an instructive role in patterning the IPL. Sdk1 fails to affect arbors when introduced after they are mature, suggesting that it is required to form but not maintain laminar restrictions. The effect of ectopically expressed sdk1 requires the presence of endogenous Sdk1, suggesting that the effect requires homophilic interactions among Sdk1-positive neurites. Together with previous results on Sdk2, Dscam, DscamL1, as well as the related Contactins, our results support the idea that an elaborate immunoglobulin superfamily code plays a prominent role in establishing neural circuits in the retina by means of tightly regulated cell type-specific expression and homophilically restricted intercellular interactions.
Collapse
Affiliation(s)
- Masahito Yamagata
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, United States
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, United States
| |
Collapse
|
25
|
Lilley BN, Sabbah S, Hunyara JL, Gribble KD, Al-Khindi T, Xiong J, Wu Z, Berson DM, Kolodkin AL. Genetic access to neurons in the accessory optic system reveals a role for Sema6A in midbrain circuitry mediating motion perception. J Comp Neurol 2019; 527:282-296. [PMID: 30076594 PMCID: PMC6312510 DOI: 10.1002/cne.24507] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/18/2018] [Accepted: 07/19/2018] [Indexed: 01/24/2023]
Abstract
The accessory optic system (AOS) detects retinal image slip and reports it to the oculomotor system for reflexive image stabilization. Here, we characterize two Cre lines that permit genetic access to AOS circuits responding to vertical motion. The first (Pcdh9-Cre) labels only one of the four subtypes of ON direction-selective retinal ganglion cells (ON-DS RGCs), those preferring ventral retinal motion. Their axons diverge from the optic tract just behind the chiasm and selectively innervate the medial terminal nucleus (MTN) of the AOS. Unlike most RGC subtypes examined, they survive after optic nerve crush. The second Cre-driver line (Pdzk1ip1-Cre) labels postsynaptic neurons in the MTN. These project predominantly to the other major terminal nucleus of the AOS, the nucleus of the optic tract (NOT). We find that the transmembrane protein semaphorin 6A (Sema6A) is required for the formation of axonal projections from the MTN to the NOT, just as it is for the retinal innervation of the MTN. These new tools permit manipulation of specific circuits in the AOS and show that Sema6A is required for establishing AOS connections in multiple locations.
Collapse
Affiliation(s)
- Brendan N. Lilley
- Solomon Snyder Department of Neuroscience, Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Current address: The Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Shai Sabbah
- Department of Neuroscience, Brown University, Providence, RI 02912
- Current address: Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - John L. Hunyara
- Solomon Snyder Department of Neuroscience, Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Katherine D. Gribble
- Solomon Snyder Department of Neuroscience, Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Timour Al-Khindi
- Solomon Snyder Department of Neuroscience, Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jiali Xiong
- Solomon Snyder Department of Neuroscience, Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Zhuhao Wu
- Laboratory of Brain Development and Repair, Rockefeller University, New York, NY 10065
| | - David M. Berson
- Department of Neuroscience, Brown University, Providence, RI 02912
| | - Alex L. Kolodkin
- Solomon Snyder Department of Neuroscience, Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
26
|
Yu WQ, El-Danaf RN, Okawa H, Pacholec JM, Matti U, Schwarz K, Odermatt B, Dunn FA, Lagnado L, Schmitz F, Huberman AD, Wong ROL. Synaptic Convergence Patterns onto Retinal Ganglion Cells Are Preserved despite Topographic Variation in Pre- and Postsynaptic Territories. Cell Rep 2018; 25:2017-2026.e3. [PMID: 30463000 PMCID: PMC6317877 DOI: 10.1016/j.celrep.2018.10.089] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/13/2018] [Accepted: 10/24/2018] [Indexed: 11/25/2022] Open
Abstract
Sensory processing can be tuned by a neuron's integration area, the types of inputs, and the proportion and number of connections with those inputs. Integration areas often vary topographically to sample space differentially across regions. Here, we highlight two visual circuits in which topographic changes in the postsynaptic retinal ganglion cell (RGC) dendritic territories and their presynaptic bipolar cell (BC) axonal territories are either matched or unmatched. Despite this difference, in both circuits, the proportion of inputs from each BC type, i.e., synaptic convergence between specific BCs and RGCs, remained constant across varying dendritic territory sizes. Furthermore, synapse density between BCs and RGCs was invariant across topography. Our results demonstrate a wiring design, likely engaging homotypic axonal tiling of BCs, that ensures consistency in synaptic convergence between specific BC types onto their target RGCs while enabling independent regulation of pre- and postsynaptic territory sizes and synapse number between cell pairs.
Collapse
Affiliation(s)
- Wan-Qing Yu
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Rana N El-Danaf
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Haruhisa Okawa
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Justin M Pacholec
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Ulf Matti
- Department of Neuroanatomy, Medical School Homburg/Saar, Institute for Anatomy and Cell Biology, Saarland University, 66421 Homburg/Saar, Germany
| | - Karin Schwarz
- Department of Neuroanatomy, Medical School Homburg/Saar, Institute for Anatomy and Cell Biology, Saarland University, 66421 Homburg/Saar, Germany
| | | | - Felice A Dunn
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Leon Lagnado
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Frank Schmitz
- Department of Neuroanatomy, Medical School Homburg/Saar, Institute for Anatomy and Cell Biology, Saarland University, 66421 Homburg/Saar, Germany
| | - Andrew D Huberman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Neurobiology and Ophthalmology, Stanford Neurosciences Institute, and BioX, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rachel O L Wong
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
27
|
Angueyra JM, Kindt KS. Leveraging Zebrafish to Study Retinal Degenerations. Front Cell Dev Biol 2018; 6:110. [PMID: 30283779 PMCID: PMC6156122 DOI: 10.3389/fcell.2018.00110] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/20/2018] [Indexed: 12/11/2022] Open
Abstract
Retinal degenerations are a heterogeneous group of diseases characterized by death of photoreceptors and progressive loss of vision. Retinal degenerations are a major cause of blindness in developed countries (Bourne et al., 2017; De Bode, 2017) and currently have no cure. In this review, we will briefly review the latest advances in therapies for retinal degenerations, highlighting the current barriers to study and develop therapies that promote photoreceptor regeneration in mammals. In light of these barriers, we present zebrafish as a powerful model to study photoreceptor regeneration and their integration into retinal circuits after regeneration. We outline why zebrafish is well suited for these analyses and summarize the powerful tools available in zebrafish that could be used to further uncover the mechanisms underlying photoreceptor regeneration and rewiring. In particular, we highlight that it is critical to understand how rewiring occurs after regeneration and how it differs from development. Insights derived from photoreceptor regeneration and rewiring in zebrafish may provide leverage to develop therapeutic targets to treat retinal degenerations.
Collapse
Affiliation(s)
- Juan M. Angueyra
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Katie S. Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
28
|
Retinal ganglion cell axon sorting at the optic chiasm requires dystroglycan. Dev Biol 2018; 442:210-219. [PMID: 30149005 DOI: 10.1016/j.ydbio.2018.08.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 08/21/2018] [Accepted: 08/23/2018] [Indexed: 01/19/2023]
Abstract
In the developing visual system, retinal ganglion cell (RGC) axons project from the retina to several distal retinorecipient regions in the brain. Several molecules have been implicated in guiding RGC axons in vivo, but the role of extracellular matrix molecules in this process remains poorly understood. Dystroglycan is a laminin-binding transmembrane protein important for formation and maintenance of the extracellular matrix and basement membranes and has previously been implicated in axon guidance in the developing spinal cord. Using two genetic models of functional dystroglycan loss, we show that dystroglycan is necessary for correct sorting of contralateral and ipsilateral RGC axons at the optic chiasm. Mis-sorted axons still target retinorecipient brain regions and persist in adult mice, even after axon pruning is complete. Our results highlight the importance of the extracellular matrix for axon sorting at an intermediate choice point in the developing visual circuit.
Collapse
|
29
|
Beier C, Palanker D, Sher A. Stereotyped Synaptic Connectivity Is Restored during Circuit Repair in the Adult Mammalian Retina. Curr Biol 2018; 28:1818-1824.e2. [PMID: 29804805 PMCID: PMC6550309 DOI: 10.1016/j.cub.2018.04.063] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/14/2018] [Accepted: 04/18/2018] [Indexed: 01/03/2023]
Abstract
Proper function of the central nervous system (CNS) depends on the specificity of synaptic connections between cells of various types. Cellular and molecular mechanisms responsible for the establishment and refinement of these connections during development are the subject of an active area of research [1-6]. However, it is unknown if the adult mammalian CNS can form new type-selective synapses following neural injury or disease. Here, we assess whether selective synaptic connections can be reestablished after circuit disruption in the adult mammalian retina. The stereotyped circuitry at the first synapse in the retina, as well as the relatively short distances new neurites must travel compared to other areas of the CNS, make the retina well suited to probing for synaptic specificity during circuit reassembly. Selective connections between short-wavelength sensitive cone photoreceptors (S-cones) and S-cone bipolar cells provides the foundation of the primordial blue-yellow vision, common to all mammals [7-18]. We take advantage of the ground squirrel retina, which has a one-to-one S-cone-to-S-cone-bipolar-cell connection, to test if this connectivity can be reestablished following local photoreceptor loss [8, 19]. We find that after in vivo selective photoreceptor ablation, deafferented S-cone bipolar cells expand their dendritic trees. The new dendrites randomly explore the proper synaptic layer, bypass medium-wavelength sensitive cone photoreceptors (M-cones), and selectively synapse with S-cones. However, non-connected dendrites are not pruned back to resemble unperturbed S-cone bipolar cells. We show, for the first time, that circuit repair in the adult mammalian retina can recreate stereotypic selective wiring.
Collapse
Affiliation(s)
- Corinne Beier
- Department of Electrical Engineering, University of California, Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Daniel Palanker
- Department of Ophthalmology, Stanford University, Stanford, CA 94305, USA; Hansen Experimental Physics Laboratory, Stanford University, Stanford, CA 94305, USA
| | - Alexander Sher
- Santa Cruz Institute for Particle Physics, University of California, Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA.
| |
Collapse
|
30
|
Sarin S, Zuniga-Sanchez E, Kurmangaliyev YZ, Cousins H, Patel M, Hernandez J, Zhang KX, Samuel MA, Morey M, Sanes JR, Zipursky SL. Role for Wnt Signaling in Retinal Neuropil Development: Analysis via RNA-Seq and In Vivo Somatic CRISPR Mutagenesis. Neuron 2018; 98:109-126.e8. [PMID: 29576390 DOI: 10.1016/j.neuron.2018.03.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 01/16/2018] [Accepted: 03/02/2018] [Indexed: 12/22/2022]
Abstract
Screens for genes that orchestrate neural circuit formation in mammals have been hindered by practical constraints of germline mutagenesis. To overcome these limitations, we combined RNA-seq with somatic CRISPR mutagenesis to study synapse development in the mouse retina. Here synapses occur between cellular layers, forming two multilayered neuropils. The outer neuropil, the outer plexiform layer (OPL), contains synapses made by rod and cone photoreceptor axons on rod and cone bipolar dendrites, respectively. We used RNA-seq to identify selectively expressed genes encoding cell surface and secreted proteins and CRISPR-Cas9 electroporation with cell-specific promoters to assess their roles in OPL development. Among the genes identified in this way are Wnt5a and Wnt5b. They are produced by rod bipolars and activate a non-canonical signaling pathway in rods to regulate early OPL patterning. The approach we use here can be applied to other parts of the brain.
Collapse
Affiliation(s)
- Sumeet Sarin
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Elizabeth Zuniga-Sanchez
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yerbol Z Kurmangaliyev
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Henry Cousins
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Mili Patel
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Jeanette Hernandez
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kelvin X Zhang
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Melanie A Samuel
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Marta Morey
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA.
| | - S Lawrence Zipursky
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
31
|
Seabrook TA, Dhande OS, Ishiko N, Wooley VP, Nguyen PL, Huberman AD. Strict Independence of Parallel and Poly-synaptic Axon-Target Matching during Visual Reflex Circuit Assembly. Cell Rep 2017; 21:3049-3064. [PMID: 29241535 PMCID: PMC6333306 DOI: 10.1016/j.celrep.2017.11.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 10/04/2017] [Accepted: 11/10/2017] [Indexed: 12/19/2022] Open
Abstract
The use of sensory information to drive specific behaviors relies on circuits spanning long distances that wire up through a range of axon-target recognition events. Mechanisms assembling poly-synaptic circuits and the extent to which parallel pathways can "cross-wire" to compensate for loss of one another remain unclear and are crucial to our understanding of brain development and models of regeneration. In the visual system, specific retinal ganglion cells (RGCs) project to designated midbrain targets connected to downstream circuits driving visuomotor reflexes. Here, we deleted RGCs connecting to pupillary light reflex (PLR) midbrain targets and discovered that axon-target matching is tightly regulated. RGC axons of the eye-reflex pathway avoided vacated PLR targets. Moreover, downstream PLR circuitry is maintained; hindbrain and peripheral components retained their proper connectivity and function. These findings point to a model in which poly-synaptic circuit development reflects independent, highly stringent wiring of each parallel pathway and downstream station.
Collapse
Affiliation(s)
- Tania A Seabrook
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Onkar S Dhande
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Nao Ishiko
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Victoria P Wooley
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Phong L Nguyen
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Andrew D Huberman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94304, USA; Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94303, USA; Bio-X, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
32
|
Xie X, Tabuchi M, Brown MP, Mitchell SP, Wu MN, Kolodkin AL. The laminar organization of the Drosophila ellipsoid body is semaphorin-dependent and prevents the formation of ectopic synaptic connections. eLife 2017. [PMID: 28632130 PMCID: PMC5511011 DOI: 10.7554/elife.25328] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The ellipsoid body (EB) in the Drosophila brain is a central complex (CX) substructure that harbors circumferentially laminated ring (R) neuron axons and mediates multifaceted sensory integration and motor coordination functions. However, what regulates R axon lamination and how lamination affects R neuron function remain unknown. We show here that the EB is sequentially innervated by small-field and large-field neurons and that early developing EB neurons play an important regulatory role in EB laminae formation. The transmembrane proteins semaphorin-1a (Sema-1a) and plexin A function together to regulate R axon lamination. R neurons recruit both GABA and GABA-A receptors to their axon terminals in the EB, and optogenetic stimulation coupled with electrophysiological recordings show that Sema-1a-dependent R axon lamination is required for preventing the spread of synaptic inhibition between adjacent EB lamina. These results provide direct evidence that EB lamination is critical for local pre-synaptic inhibitory circuit organization. DOI:http://dx.doi.org/10.7554/eLife.25328.001 The human brain contains around one hundred billion nerve cells, or neurons, which are interconnected and organized into distinct layers within different brain regions. Electrical impulses pass along a cable-like part of each neuron, known as the axon, to reach other neurons in different layers of various brain structures. The brain of a fruit fly contains fewer neurons – about 100 thousand in total – but it still establishes precise connections among neurons in different brain layers. In both flies and humans, axons grow along set paths to reach their targets by following guidance cues. Many of these cues are conserved between insects and mammals, including proteins belonging to the semaphorin family. These proteins work together to steer growing axons towards their proper targets and repel them away from the incorrect ones. However, how neurons establish connections in specific layers remains poorly understood. In the middle of the fruit fly brain lies a donut-shaped structure called the ellipsoid body, which the fly needs to navigate the world around it. The ellipsoid body contains a group of neurons that extend their axons to form multiple concentric rings. Xie et al. have now asked how the different “ring neurons” are organized in the ellipsoid body and how this sort of organization affects the connections between the neurons. Imaging techniques were used to visualize the layered organization of different ring neurons and to track their growing axons. Further work showed that this organization depends on semaphorin signaling, because when this pathway was disrupted, the layered pattern did not develop properly. This in turn, caused the axons of the ring neuron to wander out of their correct concentric ring and connect with the wrong targets in adjacent rings. Together these findings show that neurons rely on evolutionarily conserved semaphorins to correctly organize themselves into layers and connect with the appropriate targets. Further work is now needed to identify additional proteins that are critical for fly brains to form layered structures, and to understand how this layered organization influences how an animal behaves. DOI:http://dx.doi.org/10.7554/eLife.25328.002
Collapse
Affiliation(s)
- Xiaojun Xie
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, United States.,Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, United States
| | - Masashi Tabuchi
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, United States
| | - Matthew P Brown
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, United States.,Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, United States
| | - Sarah P Mitchell
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, United States.,Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, United States
| | - Mark N Wu
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, United States
| | - Alex L Kolodkin
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, United States.,Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|