1
|
Cang J, Chen C, Li C, Liu Y. Genetically defined neuron types underlying visuomotor transformation in the superior colliculus. Nat Rev Neurosci 2024; 25:726-739. [PMID: 39333418 DOI: 10.1038/s41583-024-00856-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2024] [Indexed: 09/29/2024]
Abstract
The superior colliculus (SC) is a conserved midbrain structure that is important for transforming visual and other sensory information into motor actions. Decades of investigations in numerous species have made the SC and its nonmammalian homologue, the optic tectum, one of the best studied structures in the brain, with rich information now available regarding its anatomical organization, its extensive inputs and outputs and its important functions in many reflexive and cognitive behaviours. Excitingly, recent studies using modern genomic and physiological approaches have begun to reveal the diverse neuronal subtypes in the SC, as well as their unique functions in visuomotor transformation. Studies have also started to uncover how subtypes of SC neurons form intricate circuits to mediate visual processing and visually guided behaviours. Here, we review these recent discoveries on the cell types and neuronal circuits underlying visuomotor transformations mediated by the SC. We also highlight the important future directions made possible by these new developments.
Collapse
Affiliation(s)
- Jianhua Cang
- Department of Biology, University of Virginia, Charlottesville, VA, USA.
- Department of Psychology, University of Virginia, Charlottesville, VA, USA.
| | - Chen Chen
- Department of Psychology, University of Virginia, Charlottesville, VA, USA
| | - Chuiwen Li
- Department of Psychology, University of Virginia, Charlottesville, VA, USA
| | - Yuanming Liu
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
2
|
Tipado Z, Kuypers KPC, Sorger B, Ramaekers JG. Visual hallucinations originating in the retinofugal pathway under clinical and psychedelic conditions. Eur Neuropsychopharmacol 2024; 85:10-20. [PMID: 38648694 DOI: 10.1016/j.euroneuro.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 04/25/2024]
Abstract
Psychedelics like LSD (Lysergic acid diethylamide) and psilocybin are known to modulate perceptual modalities due to the activation of mostly serotonin receptors in specific cortical (e.g., visual cortex) and subcortical (e.g., thalamus) regions of the brain. In the visual domain, these psychedelic modulations often result in peculiar disturbances of viewed objects and light and sometimes even in hallucinations of non-existent environments, objects, and creatures. Although the underlying processes are poorly understood, research conducted over the past twenty years on the subjective experience of psychedelics details theories that attempt to explain these perceptual alterations due to a disruption of communication between cortical and subcortical regions. However, rare medical conditions in the visual system like Charles Bonnet syndrome that cause perceptual distortions may shed new light on the additional importance of the retinofugal pathway in psychedelic subjective experiences. Interneurons in the retina called amacrine cells could be the first site of visual psychedelic modulation and aid in disrupting the hierarchical structure of how humans perceive visual information. This paper presents an understanding of how the retinofugal pathway communicates and modulates visual information in psychedelic and clinical conditions. Therefore, we elucidate a new theory of psychedelic modulation in the retinofugal pathway.
Collapse
Affiliation(s)
- Zeus Tipado
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, the Netherlands; Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, the Netherlands.
| | - Kim P C Kuypers
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, the Netherlands
| | - Bettina Sorger
- Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, the Netherlands
| | - Johannes G Ramaekers
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, the Netherlands
| |
Collapse
|
3
|
Tzanou A, Theodorou E, Mantas I, Dalezios Y. Excitatory Projections of Wide Field Collicular Neurons to the Nucleus of the Optic Tract in the Rat. J Comp Neurol 2024; 532:e25651. [PMID: 38961597 DOI: 10.1002/cne.25651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 05/20/2024] [Accepted: 06/07/2024] [Indexed: 07/05/2024]
Abstract
The superficial layers of the mammalian superior colliculus (SC) contain neurons that are generally responsive to visual stimuli but can differ considerably in morphology and response properties. To elucidate the structure and function of these neurons, we combined extracellular recording and juxtacellular labeling, detailed anatomical reconstruction, and ultrastructural analysis of the synaptic contacts of labeled neurons, using transmission electron microscopy. Our labeled neurons project to different brainstem nuclei. Of particular importance are neurons that fit the morphological criteria of the wide field (WF) neurons and whose dendrites are horizontally oriented. They display a rather characteristic axonal projection pattern to the nucleus of optic tract (NOT); thus, we call them superior collicular WF projecting to the NOT (SCWFNOT) neurons. We corroborated the morphological characterization of this neuronal type as a distinct neuronal class with the help of unsupervised hierarchical cluster analysis. Our ultrastructural data demonstrate that SCWFNOT neurons establish excitatory connections with their targets in the NOT. Although, in rodents, the literature about the WF neurons has focused on their extensive projection to the lateral posterior nucleus of the thalamus, as a conduit for information to reach the visual association areas of the cortex, our data suggest that this subclass of WF neurons may participate in the optokinetic nystagmus.
Collapse
Affiliation(s)
- Athanasia Tzanou
- School of Medicine, University of Crete, Heraklion, Greece
- Institute of Applied and Computational Mathematics (IACM), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece
| | - Eirini Theodorou
- School of Medicine, University of Crete, Heraklion, Greece
- Institute of Applied and Computational Mathematics (IACM), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece
| | - Ioannis Mantas
- School of Medicine, University of Crete, Heraklion, Greece
| | - Yannis Dalezios
- School of Medicine, University of Crete, Heraklion, Greece
- Institute of Applied and Computational Mathematics (IACM), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece
| |
Collapse
|
4
|
Zhang K, Zhang T, He Q, Liang H, Guo J, Zeng M, Chen S. Shootin1 Regulates Retinal Ganglion Cell Neurite Development: Insights From an RGC Direct Somatic Cell Reprogramming Model. Invest Ophthalmol Vis Sci 2024; 65:41. [PMID: 38935030 PMCID: PMC11216252 DOI: 10.1167/iovs.65.6.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Purpose Retinal ganglion cells (RGCs) connect the retina to the brain. Proper development of the axons and dendrites of RGCs is the basis for these cells to function as projection neurons to deliver visual information to the brain. The purpose of this study was to investigate the function of Shtn1 (which encodes shootin1) in RGC neurite development. Methods Immunofluorescence (IF) was used to characterize the expression pattern of marker genes. An in vitro direct somatic cell reprogramming system was used to generate RGC-like neurons (iRGCs), which was subsequently used to study the function of Shtn1. Short-hairpin RNAs (shRNAs) were used to knock down Shtn1, and the coding sequence (CDS) of Shtn1 was used to overexpress the gene. Lentiviruses were used to deliver shRNAs or CDSs into iRGCs. The patch clamp technique was used to measure the electrophysiological properties of the iRGCs. RNA sequencing (RNA-seq) was used to examine transcriptome expression. Results Using IF, we demonstrated that shootin1 is distinctively expressed in RGCs during the period in which RGCs actively develop and adjust the connections of their neurites with upstream and downstream neurons. Using the iRGC system, we demonstrated that Shtn1 promotes the growth and complexity of neurites and thus the electrophysiological maturation, of iRGCs. RNA-seq analyses showed that Shtn1 may also regulate gene expression and neurogenesis in iRGCs. Conclusions Shtn1 promotes RGC neurite development. These findings improve our understanding of the molecular machinery governing RGC neurite development and may help to optimize future RGC regeneration methods.
Collapse
Affiliation(s)
- Ke Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Tingting Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Qinghai He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Huilin Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jingyi Guo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Mingbing Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
- Hainan Eye Hospital and Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Haikou, China
| | - Shuyi Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
5
|
Golmohammadi M, Meibodi SAA, Al-Hawary SIS, Gupta J, Sapaev IB, Najm MAA, Alwave M, Nazifi M, Rahmani M, Zamanian MY, Moriasi G. Neuroprotective effects of resveratrol on retinal ganglion cells in glaucoma in rodents: A narrative review. Animal Model Exp Med 2024; 7:195-207. [PMID: 38808561 PMCID: PMC11228121 DOI: 10.1002/ame2.12438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/22/2024] [Indexed: 05/30/2024] Open
Abstract
Glaucoma, an irreversible optic neuropathy, primarily affects retinal ganglion cells (RGC) and causes vision loss and blindness. The damage to RGCs in glaucoma occurs by various mechanisms, including elevated intraocular pressure, oxidative stress, inflammation, and other neurodegenerative processes. As the disease progresses, the loss of RGCs leads to vision loss. Therefore, protecting RGCs from damage and promoting their survival are important goals in managing glaucoma. In this regard, resveratrol (RES), a polyphenolic phytoalexin, exerts antioxidant effects and slows down the evolution and progression of glaucoma. The present review shows that RES plays a protective role in RGCs in cases of ischemic injury and hypoxia as well as in ErbB2 protein expression in the retina. Additionally, RES plays protective roles in RGCs by promoting cell growth, reducing apoptosis, and decreasing oxidative stress in H2O2-exposed RGCs. RES was also found to inhibit oxidative stress damage in RGCs and suppress the activation of mitogen-activated protein kinase signaling pathways. RES could alleviate retinal function impairment by suppressing the hypoxia-inducible factor-1 alpha/vascular endothelial growth factor and p38/p53 axes while stimulating the PI3K/Akt pathway. Therefore, RES might exert potential therapeutic effects for managing glaucoma by protecting RGCs from damage and promoting their survival.
Collapse
Affiliation(s)
- Maryam Golmohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Ibrohim B Sapaev
- Tashkent Institute of Irrigation and Agricultural Mechanization Engineers, Tashkent, Uzbekistan
- New Uzbekistan University, Tashkent, Uzbekistan
| | - Mazin A A Najm
- Pharmaceutical Chemistry Department, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Marim Alwave
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Mozhgan Nazifi
- Department of Neurology, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammadreza Rahmani
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Yasin Zamanian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Gervason Moriasi
- Department of Medical Biochemistry, School of Medicine, College of Health Sciences, Mount Kenya University, Thika, Kenya
| |
Collapse
|
6
|
Ambrad Giovannetti E, Rancz E. Behind mouse eyes: The function and control of eye movements in mice. Neurosci Biobehav Rev 2024; 161:105671. [PMID: 38604571 DOI: 10.1016/j.neubiorev.2024.105671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/12/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
The mouse visual system has become the most popular model to study the cellular and circuit mechanisms of sensory processing. However, the importance of eye movements only started to be appreciated recently. Eye movements provide a basis for predictive sensing and deliver insights into various brain functions and dysfunctions. A plethora of knowledge on the central control of eye movements and their role in perception and behaviour arose from work on primates. However, an overview of various eye movements in mice and a comparison to primates is missing. Here, we review the eye movement types described to date in mice and compare them to those observed in primates. We discuss the central neuronal mechanisms for their generation and control. Furthermore, we review the mounting literature on eye movements in mice during head-fixed and freely moving behaviours. Finally, we highlight gaps in our understanding and suggest future directions for research.
Collapse
Affiliation(s)
| | - Ede Rancz
- INMED, INSERM, Aix-Marseille University, Marseille, France.
| |
Collapse
|
7
|
Yang Q, Liu L, He F, Zhao W, Chen Z, Wu X, Rao B, Lin X, Mao F, Qu J, Zhang J. Retinal ganglion cell type-specific expression of synuclein family members revealed by scRNA-sequencing. Int J Med Sci 2024; 21:1472-1490. [PMID: 38903914 PMCID: PMC11186421 DOI: 10.7150/ijms.95598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/30/2024] [Indexed: 06/22/2024] Open
Abstract
Synuclein family members (Snca, Sncb, and Scng) are expressed in the retina, but their precise locations and roles are poorly understood. We performed an extensive analysis of the single-cell transcriptome in healthy and injured retinas to investigate their expression patterns and roles. We observed the expression of all synuclein family members in retinal ganglion cells (RGCs), which remained consistent across species (human, mouse, and chicken). We unveiled differential expression of Snca across distinct clusters (highly expressed in most), while Sncb and Sncg displayed uniform expression across all clusters. Further, we observed a decreased expression in RGCs following traumatic axonal injury. However, the proportion of α-Syn-positive RGCs in all RGCs and α-Syn-positive intrinsically photosensitive retinal ganglion cells (ipRGCs) in all ipRGCs remained unaltered. Lastly, we identified changes in communication patterns preceding cell death, with particular significance in the pleiotrophin-nucleolin (Ptn-Ncl) and neural cell adhesion molecule signaling pathways, where communication differences were pronounced between cells with varying expression levels of Snca. Our study employs an innovative approach using scRNA-seq to characterize synuclein expression in health retinal cells, specifically focusing on RGC subtypes, advances our knowledge of retinal physiology and pathology.
Collapse
Affiliation(s)
- Qingwen Yang
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Lin Liu
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Fang He
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wenna Zhao
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhongqun Chen
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaotian Wu
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Bilin Rao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xin Lin
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Fangyuan Mao
- Alberta Institute, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jia Qu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jun Zhang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| |
Collapse
|
8
|
Roy S, Yao X, Rathinavelu J, Field GD. GABAergic Inhibition Controls Receptive Field Size, Sensitivity, and Contrast Preference of Direction Selective Retinal Ganglion Cells Near the Threshold of Vision. J Neurosci 2024; 44:e1979232023. [PMID: 38182419 PMCID: PMC10941243 DOI: 10.1523/jneurosci.1979-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/07/2024] Open
Abstract
Information about motion is encoded by direction-selective retinal ganglion cells (DSGCs). These cells reliably transmit this information across a broad range of light levels, spanning moonlight to sunlight. Previous work indicates that adaptation to low light levels causes heterogeneous changes to the direction tuning of ON-OFF (oo)DSGCs and suggests that superior-preferring ON-OFF DSGCs (s-DSGCs) are biased toward detecting stimuli rather than precisely signaling direction. Using a large-scale multielectrode array, we measured the absolute sensitivity of ooDSGCs and found that s-DSGCs are 10-fold more sensitive to dim flashes of light than other ooDSGCs. We measured their receptive field (RF) sizes and found that s-DSGCs also have larger receptive fields than other ooDSGCs; however, the size difference does not fully explain the sensitivity difference. Using a conditional knock-out of gap junctions and pharmacological manipulations, we demonstrate that GABA-mediated inhibition contributes to the difference in absolute sensitivity and receptive field size at low light levels, while the connexin36-mediated gap junction coupling plays a minor role. We further show that under scotopic conditions, ooDSGCs exhibit only an ON response, but pharmacologically removing GABA-mediated inhibition unmasks an OFF response. These results reveal that GABAergic inhibition controls and differentially modulates the responses of ooDSGCs under scotopic conditions.
Collapse
Affiliation(s)
- Suva Roy
- Department of Ophthalmology, Jules Stein Eye Institute, University of California, Los Angeles, California 90095
| | - Xiaoyang Yao
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Jay Rathinavelu
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Greg D Field
- Department of Ophthalmology, Jules Stein Eye Institute, University of California, Los Angeles, California 90095
| |
Collapse
|
9
|
Cortes N, Ladret HJ, Abbas-Farishta R, Casanova C. The pulvinar as a hub of visual processing and cortical integration. Trends Neurosci 2024; 47:120-134. [PMID: 38143202 DOI: 10.1016/j.tins.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/26/2023] [Accepted: 11/26/2023] [Indexed: 12/26/2023]
Abstract
The pulvinar nucleus of the thalamus is a crucial component of the visual system and plays significant roles in sensory processing and cognitive integration. The pulvinar's extensive connectivity with cortical regions allows for bidirectional communication, contributing to the integration of sensory information across the visual hierarchy. Recent findings underscore the pulvinar's involvement in attentional modulation, feature binding, and predictive coding. In this review, we highlight recent advances in clarifying the pulvinar's circuitry and function. We discuss the contributions of the pulvinar to signal modulation across the global cortical network and place these findings within theoretical frameworks of cortical processing, particularly the global neuronal workspace (GNW) theory and predictive coding.
Collapse
Affiliation(s)
- Nelson Cortes
- Visual Neuroscience Laboratory, School of Optometry, Université de Montréal, Montreal, QC, Canada
| | - Hugo J Ladret
- Visual Neuroscience Laboratory, School of Optometry, Université de Montréal, Montreal, QC, Canada; Institut de Neurosciences de la Timone, UMR 7289, CNRS and Aix-Marseille Université, Marseille, 13005, France
| | - Reza Abbas-Farishta
- Visual Neuroscience Laboratory, School of Optometry, Université de Montréal, Montreal, QC, Canada
| | - Christian Casanova
- Visual Neuroscience Laboratory, School of Optometry, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
10
|
Zhu S, Xie T, Lv Z, Leng YB, Zhang YQ, Xu R, Qin J, Zhou Y, Roy VAL, Han ST. Hierarchies in Visual Pathway: Functions and Inspired Artificial Vision. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2301986. [PMID: 37435995 DOI: 10.1002/adma.202301986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/28/2023] [Accepted: 07/10/2023] [Indexed: 07/13/2023]
Abstract
The development of artificial intelligence has posed a challenge to machine vision based on conventional complementary metal-oxide semiconductor (CMOS) circuits owing to its high latency and inefficient power consumption originating from the data shuffling between memory and computation units. Gaining more insights into the function of every part of the visual pathway for visual perception can bring the capabilities of machine vision in terms of robustness and generality. Hardware acceleration of more energy-efficient and biorealistic artificial vision highly necessitates neuromorphic devices and circuits that are able to mimic the function of each part of the visual pathway. In this paper, we review the structure and function of the entire class of visual neurons from the retina to the primate visual cortex within reach (Chapter 2) are reviewed. Based on the extraction of biological principles, the recent hardware-implemented visual neurons located in different parts of the visual pathway are discussed in detail in Chapters 3 and 4. Furthermore, valuable applications of inspired artificial vision in different scenarios (Chapter 5) are provided. The functional description of the visual pathway and its inspired neuromorphic devices/circuits are expected to provide valuable insights for the design of next-generation artificial visual perception systems.
Collapse
Affiliation(s)
- Shirui Zhu
- Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Tao Xie
- Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Ziyu Lv
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Yan-Bing Leng
- Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Yu-Qi Zhang
- Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Runze Xu
- Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Jingrun Qin
- Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Ye Zhou
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Vellaisamy A L Roy
- School of Science and Technology, Hong Kong Metropolitan University, Hong Kong, 999077, P. R. China
| | - Su-Ting Han
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| |
Collapse
|
11
|
Matcham AC, Toma K, Tsai NY, Sze CJ, Lin PY, Stewart IF, Duan X. Cadherin-13 Maintains Retinotectal Synapses via Transneuronal Interactions. J Neurosci 2024; 44:e1310232023. [PMID: 38123991 PMCID: PMC10860569 DOI: 10.1523/jneurosci.1310-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 12/03/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
Maintaining precise synaptic contacts between neuronal partners is critical to ensure the proper functioning of the mammalian central nervous system (CNS). Diverse cell recognition molecules, such as classic cadherins (Cdhs), are part of the molecular machinery mediating synaptic choices during development and synaptic maintenance. Yet, the principles governing neuron-neuron wiring across diverse CNS neuron types remain largely unknown. The retinotectal synapses, connections from the retinal ganglion cells (RGCs) to the superior collicular (SC) neurons, offer an ideal experimental system to reveal molecular logic underlying synaptic choices and formation. This is due to the retina's unidirectional and laminar-restricted projections to the SC and the large databases of presynaptic RGC subtypes and postsynaptic SC neuronal types. Here, we focused on determining the role of Type II Cdhs in wiring the retinotectal synapses. We surveyed Cdhs expression patterns at neuronal resolution and revealed that Cdh13 is enriched in the wide-field neurons in the superficial SC (sSC). In either the Cdh13 null mutant or selective adult deletion within the wide-field neurons, there is a significant reduction of spine densities in the distal dendrites of these neurons in both sexes. Additionally, Cdh13 removal from presynaptic RGCs reduced dendritic spines in the postsynaptic wide-field neurons. Cdh13-expressing RGCs use differential mechanisms than αRGCs and On-Off Direction-Selective Ganglion Cells (ooDSGCs) to form specific retinotectal synapses. The results revealed a selective transneuronal interaction mediated by Cdh13 to maintain proper retinotectal synapses in vivo.
Collapse
Affiliation(s)
- Angela C Matcham
- Neuroscience Graduate Program, Department of Ophthalmology, Kavli Institute for Fundamental Neuroscience, University of California SanFrancisco, San Francisco 94143-2811, California
| | - Kenichi Toma
- Neuroscience Graduate Program, Department of Ophthalmology, Kavli Institute for Fundamental Neuroscience, University of California SanFrancisco, San Francisco 94143-2811, California
| | - Nicole Y Tsai
- Neuroscience Graduate Program, Department of Ophthalmology, Kavli Institute for Fundamental Neuroscience, University of California SanFrancisco, San Francisco 94143-2811, California
| | - Christina J Sze
- Neuroscience Graduate Program, Department of Ophthalmology, Kavli Institute for Fundamental Neuroscience, University of California SanFrancisco, San Francisco 94143-2811, California
| | - Pin-Yeh Lin
- Neuroscience Graduate Program, Department of Ophthalmology, Kavli Institute for Fundamental Neuroscience, University of California SanFrancisco, San Francisco 94143-2811, California
| | - Ilaria F Stewart
- Neuroscience Graduate Program, Department of Ophthalmology, Kavli Institute for Fundamental Neuroscience, University of California SanFrancisco, San Francisco 94143-2811, California
| | - Xin Duan
- Neuroscience Graduate Program, Department of Ophthalmology, Kavli Institute for Fundamental Neuroscience, University of California SanFrancisco, San Francisco 94143-2811, California
| |
Collapse
|
12
|
Tribble JR, Hui F, Quintero H, El Hajji S, Bell K, Di Polo A, Williams PA. Neuroprotection in glaucoma: Mechanisms beyond intraocular pressure lowering. Mol Aspects Med 2023; 92:101193. [PMID: 37331129 DOI: 10.1016/j.mam.2023.101193] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/25/2023] [Accepted: 06/04/2023] [Indexed: 06/20/2023]
Abstract
Glaucoma is a common, complex, multifactorial neurodegenerative disease characterized by progressive dysfunction and then loss of retinal ganglion cells, the output neurons of the retina. Glaucoma is the most common cause of irreversible blindness and affects ∼80 million people worldwide with many more undiagnosed. The major risk factors for glaucoma are genetics, age, and elevated intraocular pressure. Current strategies only target intraocular pressure management and do not directly target the neurodegenerative processes occurring at the level of the retinal ganglion cell. Despite strategies to manage intraocular pressure, as many as 40% of glaucoma patients progress to blindness in at least one eye during their lifetime. As such, neuroprotective strategies that target the retinal ganglion cell and these neurodegenerative processes directly are of great therapeutic need. This review will cover the recent advances from basic biology to on-going clinical trials for neuroprotection in glaucoma covering degenerative mechanisms, metabolism, insulin signaling, mTOR, axon transport, apoptosis, autophagy, and neuroinflammation. With an increased understanding of both the basic and clinical mechanisms of the disease, we are closer than ever to a neuroprotective strategy for glaucoma.
Collapse
Affiliation(s)
- James R Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Flora Hui
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia; Department of Optometry & Vision Sciences, The University of Melbourne, Melbourne, Australia
| | - Heberto Quintero
- Department of Neuroscience, University of Montreal, Montreal, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Sana El Hajji
- Department of Neuroscience, University of Montreal, Montreal, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Katharina Bell
- NHMRC Clinical Trials Centre, University of Sydney, Australia; Eye ACP Duke-NUS, Singapore
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
13
|
Liu Y, Savier EL, DePiero VJ, Chen C, Schwalbe DC, Abraham-Fan RJ, Chen H, Campbell JN, Cang J. Mapping visual functions onto molecular cell types in the mouse superior colliculus. Neuron 2023; 111:1876-1886.e5. [PMID: 37086721 PMCID: PMC10330256 DOI: 10.1016/j.neuron.2023.03.036] [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: 12/14/2022] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 04/24/2023]
Abstract
The superficial superior colliculus (sSC) carries out diverse roles in visual processing and behaviors, but how these functions are delegated among collicular neurons remains unclear. Here, using single-cell transcriptomics, we identified 28 neuron subtypes and subtype-enriched marker genes from tens of thousands of adult mouse sSC neurons. We then asked whether the sSC's molecular subtypes are tuned to different visual stimuli. Specifically, we imaged calcium dynamics in single sSC neurons in vivo during visual stimulation and then mapped marker gene transcripts onto the same neurons ex vivo. Our results identify a molecular subtype of inhibitory neuron accounting for ∼50% of the sSC's direction-selective cells, suggesting a genetic logic for the functional organization of the sSC. In addition, our studies provide a comprehensive molecular atlas of sSC neuron subtypes and a multimodal mapping method that will facilitate investigation of their respective functions, connectivity, and development.
Collapse
Affiliation(s)
- Yuanming Liu
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Elise L Savier
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Victor J DePiero
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Chen Chen
- Department of Psychology, University of Virginia, Charlottesville, VA 22904, USA
| | - Dana C Schwalbe
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | | | - Hui Chen
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - John N Campbell
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA.
| | - Jianhua Cang
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA; Department of Psychology, University of Virginia, Charlottesville, VA 22904, USA.
| |
Collapse
|
14
|
Teh KL, Sibille J, Gehr C, Kremkow J. Retinal waves align the concentric orientation map in mouse superior colliculus to the center of vision. SCIENCE ADVANCES 2023; 9:eadf4240. [PMID: 37172095 PMCID: PMC10181181 DOI: 10.1126/sciadv.adf4240] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Neurons in the mouse superior colliculus (SC) are arranged in a concentric orientation map, which is aligned to the center of vision and the optic flow experienced by the mouse. The origin of this map remains unclear. Here, we propose that spontaneous retinal waves during development provide a scaffold to establish the concentric orientation map within the SC and its alignment to the optic flow. We test this hypothesis by modeling the orientation-tuned SC neurons that receive ON/OFF retinal inputs. Our model suggests that the propagation direction bias of stage III retinal waves, together with OFF-delayed responses, shapes the spatial organization of the orientation map. The OFF delay establishes orientation-tuned neurons by segregating their ON/OFF receptive subfields, the wave-like activities form the concentric pattern, and the direction biases align the map to the center of vision. Together, retinal waves may play an instructive role in establishing functional properties of single SC neurons and their spatial organization within maps.
Collapse
Affiliation(s)
- Kai Lun Teh
- Neuroscience Research Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, Berlin 10117, Germany
- Bernstein Center for Computational Neuroscience Berlin, Philippstraße 13, Berlin 10115, Germany
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Philippstraße 13, Berlin 10115, Germany
- Einstein Center for Neurosciences Berlin, Charitéplatz 1, Berlin, Germany
| | - Jérémie Sibille
- Neuroscience Research Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, Berlin 10117, Germany
- Bernstein Center for Computational Neuroscience Berlin, Philippstraße 13, Berlin 10115, Germany
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Philippstraße 13, Berlin 10115, Germany
- Einstein Center for Neurosciences Berlin, Charitéplatz 1, Berlin, Germany
| | - Carolin Gehr
- Neuroscience Research Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, Berlin 10117, Germany
- Bernstein Center for Computational Neuroscience Berlin, Philippstraße 13, Berlin 10115, Germany
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Philippstraße 13, Berlin 10115, Germany
- Einstein Center for Neurosciences Berlin, Charitéplatz 1, Berlin, Germany
| | - Jens Kremkow
- Neuroscience Research Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, Berlin 10117, Germany
- Bernstein Center for Computational Neuroscience Berlin, Philippstraße 13, Berlin 10115, Germany
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Philippstraße 13, Berlin 10115, Germany
- Einstein Center for Neurosciences Berlin, Charitéplatz 1, Berlin, Germany
| |
Collapse
|
15
|
Romano GL, Gozzo L, Maurel OM, Di Martino S, Riolo V, Micale V, Drago F, Bucolo C. Fluoxetine Protects Retinal Ischemic Damage in Mice. Pharmaceutics 2023; 15:pharmaceutics15051370. [PMID: 37242611 DOI: 10.3390/pharmaceutics15051370] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND To evaluate the neuroprotective effect of the topical ocular administration of fluoxetine (FLX) in a mouse model of acute retinal damage. METHODS Ocular ischemia/reperfusion (I/R) injury in C57BL/6J mice was used to elicit retinal damage. Mice were divided into three groups: control group, I/R group, and I/R group treated with topical FLX. A pattern electroretinogram (PERG) was used as a sensitive measure of retinal ganglion cell (RGC) function. Finally, we analyzed the retinal mRNA expression of inflammatory markers (IL-6, TNF-α, Iba-1, IL-1β, and S100β) through Digital Droplet PCR. RESULTS PERG amplitude values were significantly (p < 0.05) higher in the I/R-FLX group compared to the I/R group, whereas PERG latency values were significantly (p < 0.05) reduced in I/R-FLX-treated mice compared to the I/R group. Retinal inflammatory markers increased significantly (p < 0.05) after I/R injury. FLX treatment was able to significantly (p < 0.05) attenuate the expression of inflammatory markers after I/R damage. CONCLUSIONS Topical treatment with FLX was effective in counteracting the damage of RGCs and preserving retinal function. Moreover, FLX treatment attenuates the production of pro-inflammatory molecules elicited by retinal I/R damage. Further studies need to be performed to support the use of FLX as neuroprotective agent in retinal degenerative diseases.
Collapse
Affiliation(s)
- Giovanni Luca Romano
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95100 Catania, Italy
- Center for Research in Ocular Pharmacology-CERFO, University of Catania, 95100 Catania, Italy
| | - Lucia Gozzo
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95100 Catania, Italy
| | - Oriana Maria Maurel
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95100 Catania, Italy
| | - Serena Di Martino
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95100 Catania, Italy
| | - Valentina Riolo
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95100 Catania, Italy
| | - Vincenzo Micale
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95100 Catania, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95100 Catania, Italy
- Center for Research in Ocular Pharmacology-CERFO, University of Catania, 95100 Catania, Italy
| | - Claudio Bucolo
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95100 Catania, Italy
- Center for Research in Ocular Pharmacology-CERFO, University of Catania, 95100 Catania, Italy
| |
Collapse
|
16
|
Ge Y, Chen X, Nan N, Bard J, Wu F, Yergeau D, Liu T, Wang J, Mu X. Key transcription factors influence the epigenetic landscape to regulate retinal cell differentiation. Nucleic Acids Res 2023; 51:2151-2176. [PMID: 36715342 PMCID: PMC10018358 DOI: 10.1093/nar/gkad026] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/31/2023] Open
Abstract
How the diverse neural cell types emerge from multipotent neural progenitor cells during central nervous system development remains poorly understood. Recent scRNA-seq studies have delineated the developmental trajectories of individual neural cell types in many neural systems including the neural retina. Further understanding of the formation of neural cell diversity requires knowledge about how the epigenetic landscape shifts along individual cell lineages and how key transcription factors regulate these changes. In this study, we dissect the changes in the epigenetic landscape during early retinal cell differentiation by scATAC-seq and identify globally the enhancers, enriched motifs, and potential interacting transcription factors underlying the cell state/type specific gene expression in individual lineages. Using CUT&Tag, we further identify the enhancers bound directly by four key transcription factors, Otx2, Atoh7, Pou4f2 and Isl1, including those dependent on Atoh7, and uncover the sequential and combinatorial interactions of these factors with the epigenetic landscape to control gene expression along individual retinal cell lineages such as retinal ganglion cells (RGCs). Our results reveal a general paradigm in which transcription factors collaborate and compete to regulate the emergence of distinct retinal cell types such as RGCs from multipotent retinal progenitor cells (RPCs).
Collapse
Affiliation(s)
- Yichen Ge
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Xushen Chen
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Nan Nan
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| | - Jonathan Bard
- New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY, USA
| | - Fuguo Wu
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Donald Yergeau
- New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY, USA
| | - Tao Liu
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jie Wang
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Xiuqian Mu
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
17
|
Santana NNM, Silva EHA, dos Santos SF, Costa MSMO, Nascimento Junior ES, Engelberth RCJG, Cavalcante JS. Retinorecipient areas in the common marmoset ( Callithrix jacchus): An image-forming and non-image forming circuitry. Front Neural Circuits 2023; 17:1088686. [PMID: 36817647 PMCID: PMC9932520 DOI: 10.3389/fncir.2023.1088686] [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: 11/03/2022] [Accepted: 01/10/2023] [Indexed: 02/05/2023] Open
Abstract
The mammalian retina captures a multitude of diverse features from the external environment and conveys them via the optic nerve to a myriad of retinorecipient nuclei. Understanding how retinal signals act in distinct brain functions is one of the most central and established goals of neuroscience. Using the common marmoset (Callithrix jacchus), a monkey from Northeastern Brazil, as an animal model for parsing how retinal innervation works in the brain, started decades ago due to their marmoset's small bodies, rapid reproduction rate, and brain features. In the course of that research, a large amount of new and sophisticated neuroanatomical techniques was developed and employed to explain retinal connectivity. As a consequence, image and non-image-forming regions, functions, and pathways, as well as retinal cell types were described. Image-forming circuits give rise directly to vision, while the non-image-forming territories support circadian physiological processes, although part of their functional significance is uncertain. Here, we reviewed the current state of knowledge concerning retinal circuitry in marmosets from neuroanatomical investigations. We have also highlighted the aspects of marmoset retinal circuitry that remain obscure, in addition, to identify what further research is needed to better understand the connections and functions of retinorecipient structures.
Collapse
Affiliation(s)
- Nelyane Nayara M. Santana
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Eryck H. A. Silva
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Sâmarah F. dos Santos
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Miriam S. M. O. Costa
- Laboratory of Neuroanatomy, Department of Morphology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Expedito S. Nascimento Junior
- Laboratory of Neuroanatomy, Department of Morphology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Rovena Clara J. G. Engelberth
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Jeferson S. Cavalcante
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil,*Correspondence: Jeferson S. Cavalcante,
| |
Collapse
|
18
|
Li VJ, Chorghay Z, Ruthazer ES. A Guide for the Multiplexed: The Development of Visual Feature Maps in the Brain. Neuroscience 2023; 508:62-75. [PMID: 35952996 DOI: 10.1016/j.neuroscience.2022.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 01/17/2023]
Abstract
Neural maps are found ubiquitously in the brain, where they encode a wide range of behaviourally relevant features into neural space. Developmental studies have shown that animals devote a great deal of resources to establish consistently patterned organization in neural circuits throughout the nervous system, but what purposes maps serve beneath their often intricate appearance and composition is a topic of active debate and exploration. In this article, we review the general mechanisms of map formation, with a focus on the visual system, and then survey notable organizational properties of neural maps: the multiplexing of feature representations through a nested architecture, the interspersing of fine-scale heterogeneity within a globally smooth organization, and the complex integration at the microcircuit level that enables a high dimensionality of information encoding. Finally, we discuss the roles of maps in cortical functions, including input segregation, feature extraction and routing of circuit outputs for higher order processing, as well as the evolutionary basis for the properties we observe in neural maps.
Collapse
Affiliation(s)
- Vanessa J Li
- Montreal Neurological Institute-Hospital, McGill University, 3801 University St. Montreal, Quebec H3A 2B4, Canada
| | - Zahraa Chorghay
- Montreal Neurological Institute-Hospital, McGill University, 3801 University St. Montreal, Quebec H3A 2B4, Canada
| | - Edward S Ruthazer
- Montreal Neurological Institute-Hospital, McGill University, 3801 University St. Montreal, Quebec H3A 2B4, Canada
| |
Collapse
|
19
|
Agadagba SK, Lim LW, Chan LLH. Advances in transcorneal electrical stimulation: From the eye to the brain. Front Cell Neurosci 2023; 17:1134857. [PMID: 36937185 PMCID: PMC10019785 DOI: 10.3389/fncel.2023.1134857] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/07/2023] [Indexed: 03/06/2023] Open
Abstract
The mammalian brain is reported to contain about 106-109 neurons linked together to form complex networks. Physiologically, the neuronal networks interact in a rhythmic oscillatory pattern to coordinate the brain's functions. Neuromodulation covers a broad range of techniques that can alter neuronal network activity through the targeted delivery of electrical or chemical stimuli. Neuromodulation can be used to potentially treat medical conditions and can serve as a research tool for studying neural functions. Typically, the main method of neuromodulation is to electrically stimulate specific structures in both the central and peripheral nervous systems via surgically implanted electrodes. Therefore, it is imperative to explore novel and safer methods for altering neuronal network activity. Transcorneal electrical stimulation (TES) has rapidly emerged as a non-invasive neuromodulatory technique that can exert beneficial effects on the brain through the eyes. There is substantial evidence to show that TES can change the brain oscillations in rodents. Moreover, the molecular data clearly shows that TES can also activate non-visual brain regions. In this review, we first summarize the use of TES in the retina and then discuss its effects in the brain through the eye-brain connection. We then comprehensively review the substantial evidence from electrophysiological, behavioral, and molecular studies on the role of TES on modulating neurons in the brain. Lastly, we discuss the implications and possible future directions of the research on TES as a non-invasive tool for neuromodulation of the brain via directly stimulating the mammalian eye.
Collapse
Affiliation(s)
| | - Lee Wei Lim
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Leanne Lai Hang Chan
- Department of Electrical Engineering, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
- *Correspondence: Leanne Lai Hang Chan
| |
Collapse
|
20
|
Cline HT, Lau M, Hiramoto M. Activity-dependent Organization of Topographic Neural Circuits. Neuroscience 2023; 508:3-18. [PMID: 36470479 PMCID: PMC9839526 DOI: 10.1016/j.neuroscience.2022.11.032] [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: 03/24/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Sensory information in the brain is organized into spatial representations, including retinotopic, somatotopic, and tonotopic maps, as well as ocular dominance columns. The spatial representation of sensory inputs is thought to be a fundamental organizational principle that is important for information processing. Topographic maps are plastic throughout an animal's life, reflecting changes in development and aging of brain circuitry, changes in the periphery and sensory input, and changes in circuitry, for instance in response to experience and learning. Here, we review mechanisms underlying the role of activity in the development, stability and plasticity of topographic maps, focusing on recent work suggesting that the spatial information in the visual field, and the resulting spatiotemporal patterns of activity, provide instructive cues that organize visual projections.
Collapse
Affiliation(s)
- Hollis T Cline
- Department of Neuroscience and the Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA.
| | - Melissa Lau
- Department of Neuroscience and the Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Masaki Hiramoto
- Department of Neuroscience and the Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
21
|
Burton EA, Burgess HA. A Critical Review of Zebrafish Neurological Disease Models-2. Application: Functional and Neuroanatomical Phenotyping Strategies and Chemical Screens. OXFORD OPEN NEUROSCIENCE 2022; 2:kvac019. [PMID: 37637775 PMCID: PMC10455049 DOI: 10.1093/oons/kvac019] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/28/2022] [Indexed: 08/29/2023]
Abstract
Extensive phylogenetic conservation of molecular pathways and neuroanatomical structures, associated with efficient methods for genetic modification, have been exploited increasingly to generate zebrafish models of human disease. A range of powerful approaches can be deployed to analyze these models with the ultimate goal of elucidating pathogenic mechanisms and accelerating efforts to find effective treatments. Unbiased neurobehavioral assays can provide readouts that parallel clinical abnormalities found in patients, although some of the most useful assays quantify responses that are not routinely evaluated clinically, and differences between zebrafish and human brains preclude expression of the full range of neurobehavioral abnormalities seen in disease. Imaging approaches that use fluorescent reporters and standardized brain atlases coupled with quantitative measurements of brain structure offer an unbiased means to link experimental manipulations to changes in neural architecture. Together, quantitative structural and functional analyses allow dissection of the cellular and physiological basis underlying neurological phenotypes. These approaches can be used as outputs in chemical modifier screens, which provide a major opportunity to exploit zebrafish models to identify small molecule modulators of pathophysiology that may be informative for understanding disease mechanisms and possible therapeutic approaches.
Collapse
Affiliation(s)
- Edward A Burton
- Pittsburgh Institute of Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Geriatric Research, Education, and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA 15240, USA
| | - Harold A Burgess
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| |
Collapse
|
22
|
Sibille J, Gehr C, Benichov JI, Balasubramanian H, Teh KL, Lupashina T, Vallentin D, Kremkow J. High-density electrode recordings reveal strong and specific connections between retinal ganglion cells and midbrain neurons. Nat Commun 2022; 13:5218. [PMID: 36064789 PMCID: PMC9445019 DOI: 10.1038/s41467-022-32775-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 08/16/2022] [Indexed: 11/29/2022] Open
Abstract
The superior colliculus is a midbrain structure that plays important roles in visually guided behaviors in mammals. Neurons in the superior colliculus receive inputs from retinal ganglion cells but how these inputs are integrated in vivo is unknown. Here, we discovered that high-density electrodes simultaneously capture the activity of retinal axons and their postsynaptic target neurons in the superior colliculus, in vivo. We show that retinal ganglion cell axons in the mouse provide a single cell precise representation of the retina as input to superior colliculus. This isomorphic mapping builds the scaffold for precise retinotopic wiring and functionally specific connection strength. Our methods are broadly applicable, which we demonstrate by recording retinal inputs in the optic tectum in zebra finches. We find common wiring rules in mice and zebra finches that provide a precise representation of the visual world encoded in retinal ganglion cells connections to neurons in retinorecipient areas.
Collapse
Affiliation(s)
- Jérémie Sibille
- Neuroscience Research Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Philippstraße 13, 10115, Berlin, Germany
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Philippstraße 13, 10115, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Carolin Gehr
- Neuroscience Research Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Philippstraße 13, 10115, Berlin, Germany
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Philippstraße 13, 10115, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Jonathan I Benichov
- Max Planck Institute for Ornithology, Eberhard-Gwinner Straße, 82319, Seewiesen, Germany
- Max Planck Institute for Biological Intelligence (in foundation), Eberhard-Gwinner Straße, 82319, Seewiesen, Germany
| | - Hymavathy Balasubramanian
- Neuroscience Research Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Philippstraße 13, 10115, Berlin, Germany
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Philippstraße 13, 10115, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Kai Lun Teh
- Neuroscience Research Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Philippstraße 13, 10115, Berlin, Germany
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Philippstraße 13, 10115, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Tatiana Lupashina
- Neuroscience Research Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Philippstraße 13, 10115, Berlin, Germany
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Philippstraße 13, 10115, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Daniela Vallentin
- Max Planck Institute for Ornithology, Eberhard-Gwinner Straße, 82319, Seewiesen, Germany
- Max Planck Institute for Biological Intelligence (in foundation), Eberhard-Gwinner Straße, 82319, Seewiesen, Germany
| | - Jens Kremkow
- Neuroscience Research Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- Bernstein Center for Computational Neuroscience Berlin, Philippstraße 13, 10115, Berlin, Germany.
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Philippstraße 13, 10115, Berlin, Germany.
- Einstein Center for Neurosciences Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
23
|
Tapia ML, Nascimento-Dos-Santos G, Park KK. Subtype-specific survival and regeneration of retinal ganglion cells in response to injury. Front Cell Dev Biol 2022; 10:956279. [PMID: 36035999 PMCID: PMC9411869 DOI: 10.3389/fcell.2022.956279] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/28/2022] [Indexed: 11/19/2022] Open
Abstract
Retinal ganglion cells (RGCs) are a heterogeneous population of neurons that function synchronously to convey visual information through the optic nerve to retinorecipient target areas in the brain. Injury or disease to the optic nerve results in RGC degeneration and loss of visual function, as few RGCs survive, and even fewer can be provoked to regenerate their axons. Despite causative insults being broadly shared, regeneration studies demonstrate that RGC types exhibit differential resilience to injury and undergo selective survival and regeneration of their axons. While most early studies have identified these RGC types based their morphological and physiological characteristics, recent advances in transgenic and gene sequencing technologies have further enabled type identification based on unique molecular features. In this review, we provide an overview of the well characterized RGC types and identify those shown to preferentially survive and regenerate in various regeneration models. Furthermore, we discuss cellular characteristics of both the resilient and susceptible RGC types including the combinatorial expression of different molecular markers that identify these specific populations. Lastly, we discuss potential molecular mechanisms and genes found to be selectively expressed by specific types that may contribute to their reparative capacity. Together, we describe the studies that lay the important groundwork for identifying factors that promote neural regeneration and help advance the development of targeted therapy for the treatment of RGC degeneration as well as neurodegenerative diseases in general.
Collapse
Affiliation(s)
- Mary L Tapia
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Gabriel Nascimento-Dos-Santos
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Kevin K Park
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
24
|
Sharma P, Ramachandran R. Retina regeneration: lessons from vertebrates. OXFORD OPEN NEUROSCIENCE 2022; 1:kvac012. [PMID: 38596712 PMCID: PMC10913848 DOI: 10.1093/oons/kvac012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/24/2022] [Accepted: 06/25/2022] [Indexed: 04/11/2024]
Abstract
Unlike mammals, vertebrates such as fishes and frogs exhibit remarkable tissue regeneration including the central nervous system. Retina being part of the central nervous system has attracted the interest of several research groups to explore its regenerative ability in different vertebrate models including mice. Fishes and frogs completely restore the size, shape and tissue structure of an injured retina. Several studies have unraveled molecular mechanisms underlying retina regeneration. In teleosts, soon after injury, the Müller glial cells of the retina reprogram to form a proliferating population of Müller glia-derived progenitor cells capable of differentiating into various neural cell types and Müller glia. In amphibians, the transdifferentiation of retinal pigment epithelium and differentiation of ciliary marginal zone cells contribute to retina regeneration. In chicks and mice, supplementation with external growth factors or genetic modifications cause a partial regenerative response in the damaged retina. The initiation of retina regeneration is achieved through sequential orchestration of gene expression through controlled modulations in the genetic and epigenetic landscape of the progenitor cells. Several developmental biology pathways are turned on during the Müller glia reprogramming, retinal pigment epithelium transdifferentiation and ciliary marginal zone differentiation. Further, several tumorigenic pathways and gene expression events also contribute to the complete regeneration cascade of events. In this review, we address the various retinal injury paradigms and subsequent gene expression events governed in different vertebrate species. Further, we compared how vertebrates such as teleost fishes and amphibians can achieve excellent regenerative responses in the retina compared with their mammalian counterparts.
Collapse
Affiliation(s)
- Poonam Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Knowledge City, SAS Nagar, Sector 81, Manauli PO, 140306 Mohali, Punjab, India
| | - Rajesh Ramachandran
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Knowledge City, SAS Nagar, Sector 81, Manauli PO, 140306 Mohali, Punjab, India
| |
Collapse
|
25
|
Kowal TJ, Dhande OS, Wang B, Wang Q, Ning K, Liu W, Berbari NF, Hu Y, Sun Y. Distribution of prototypical primary cilia markers in subtypes of retinal ganglion cells. J Comp Neurol 2022; 530:2176-2187. [PMID: 35434813 PMCID: PMC9219574 DOI: 10.1002/cne.25326] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/27/2022] [Accepted: 03/21/2022] [Indexed: 11/07/2022]
Abstract
Loss of retinal ganglion cells (RGCs) underlies several forms of retinal disease including glaucomatous optic neuropathy, a leading cause of irreversible blindness. Several rare genetic disorders associated with cilia dysfunction have retinal degeneration as a clinical hallmark. Much of the focus of ciliopathy associated blindness is on the connecting cilium of photoreceptors; however, RGCs also possess primary cilia. It is unclear what roles RGC cilia play, what proteins and signaling machinery localize to RGC cilia, or how RGC cilia are differentiated across the subtypes of RGCs. To better understand these questions, we assessed the presence or absence of a prototypical cilia marker Arl13b and a widely distributed neuronal cilia marker AC3 in different subtypes of mouse RGCs. Interestingly, not all RGC subtype cilia are the same and there are significant differences even among these standard cilia markers. Alpha-RGCs positive for osteopontin, calretinin, and SMI32 primarily possess AC3-positive cilia. Directionally selective RGCs that are CART positive or Trhr positive localize either Arl13b or AC3, respectively, in cilia. Intrinsically photosensitive RGCs differentially localize Arl13b and AC3 based on melanopsin expression. Taken together, we characterized the localization of gold standard cilia markers in different subtypes of RGCs and conclude that cilia within RGC subtypes may be differentially organized. Future studies aimed at understanding RGC cilia function will require a fundamental ability to observe the cilia across subtypes as their signaling protein composition is elucidated. A comprehensive understanding of RGC cilia may reveal opportunities to understanding how their dysfunction leads to retinal degeneration.
Collapse
Affiliation(s)
- Tia J. Kowal
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Onkar S. Dhande
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Biao Wang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Qing Wang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Ke Ning
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Wendy Liu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Nicolas F. Berbari
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis IN 46202 USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
- Palo Alto Veterans Administration, Palo Alto, CA 94304
| |
Collapse
|
26
|
Molins B, Mesquida M, Adan A. Bioengineering approaches for modelling retinal pathologies of the outer blood-retinal barrier. Prog Retin Eye Res 2022:101097. [PMID: 35840488 DOI: 10.1016/j.preteyeres.2022.101097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 05/31/2022] [Accepted: 06/29/2022] [Indexed: 11/18/2022]
Abstract
Alterations of the junctional complex of the outer blood-retinal barrier (oBRB), which is integrated by the close interaction of the retinal pigment epithelium, the Bruch's membrane, and the choriocapillaris, contribute to the loss of neuronal signalling and subsequent vision impairment in several retinal inflammatory disorders such as age-related macular degeneration and diabetic retinopathy. Reductionist approaches into the mechanisms that underlie such diseases have been hindered by the absence of adequate in vitro models using human cells to provide the 3D dynamic architecture that enables expression of the in vivo phenotype of the oBRB. Conventional in vitro cell models are based on 2D monolayer cellular cultures, unable to properly recapitulate the complexity of living systems. The main drawbacks of conventional oBRB models also emerge from the cell sourcing, the lack of an appropriate Bruch's membrane analogue, and the lack of choroidal microvasculature with flow. In the last years, the advent of organ-on-a-chip, bioengineering, and stem cell technologies is providing more advanced 3D models with flow, multicellularity, and external control over microenvironmental properties. By incorporating additional biological complexity, organ-on-a-chip devices can mirror physiologically relevant properties of the native tissue while offering additional set ups to model and study disease. In this review we first examine the current understanding of oBRB biology as a functional unit, highlighting the coordinated contribution of the different components to barrier function in health and disease. Then we describe recent advances in the use of pluripotent stem cells-derived retinal cells, Bruch's membrane analogues, and co-culture techniques to recapitulate the oBRB. We finally discuss current advances and challenges of oBRB-on-a-chip technologies for disease modelling.
Collapse
Affiliation(s)
- Blanca Molins
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d'Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), C/ Sabino de Arana 1, 08028, Barcelona, Spain.
| | - Marina Mesquida
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d'Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), C/ Sabino de Arana 1, 08028, Barcelona, Spain; Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Alfredo Adan
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d'Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), C/ Sabino de Arana 1, 08028, Barcelona, Spain; Instituto Clínic de Oftalmología, Hospital Clínic Barcelona, C/ Sabino de Arana 1, 08028, Barcelona, Spain
| |
Collapse
|
27
|
Wang Y, Liu X, Quan X, Qin X, Zhou Y, Liu Z, Chao Z, Jia C, Qin H, Zhang H. Pigment epithelium-derived factor and its role in microvascular-related diseases. Biochimie 2022; 200:153-171. [DOI: 10.1016/j.biochi.2022.05.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 05/19/2022] [Accepted: 05/30/2022] [Indexed: 01/02/2023]
|
28
|
Summers MT, Feller MB. Distinct inhibitory pathways control velocity and directional tuning in the mouse retina. Curr Biol 2022; 32:2130-2143.e3. [PMID: 35395192 PMCID: PMC9133153 DOI: 10.1016/j.cub.2022.03.054] [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: 01/21/2022] [Revised: 03/09/2022] [Accepted: 03/17/2022] [Indexed: 11/30/2022]
Abstract
The sensory periphery is responsible for detecting ethologically relevant features of the external world, using compact, predominantly feedforward circuits. Visual motion is a particularly prevalent sensory feature, the presence of which can be a signal to enact diverse behaviors ranging from gaze stabilization reflexes to predator avoidance or prey capture. To understand how the retina constructs the distinct neural representations required for these behaviors, we investigated two circuits responsible for encoding different aspects of image motion: ON and ON-OFF direction-selective ganglion cells (DSGCs). Using a combination of two-photon targeted whole-cell electrophysiology, pharmacology, and conditional knockout mice, we show that distinct inhibitory pathways independently control tuning for motion velocity and motion direction in these two cell types. We further employ dynamic clamp and numerical modeling techniques to show that asymmetric inhibition provides a velocity-invariant mechanism of directional tuning, despite the strong velocity dependence of classical models of direction selectivity. We therefore demonstrate that invariant representations of motion features by inhibitory interneurons act as computational building blocks to construct distinct, behaviorally relevant signals at the earliest stages of the visual system.
Collapse
Affiliation(s)
- Mathew T Summers
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Marla B Feller
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
29
|
Sokhadze G, Campbell PW, Charalambakis N, Govindaiah G, Guido W, McGee AW. Cre driver mouse lines for thalamocortical circuit mapping. J Comp Neurol 2022; 530:1049-1063. [PMID: 34545582 PMCID: PMC9891227 DOI: 10.1002/cne.25248] [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: 05/23/2021] [Revised: 08/16/2021] [Accepted: 09/07/2021] [Indexed: 02/04/2023]
Abstract
Subpopulations of neurons and associated neural circuits can be targeted in mice with genetic tools in a highly selective manner for visualization and manipulation. However, there are not well-defined Cre "driver" lines that target the expression of Cre recombinase to thalamocortical (TC) neurons. Here, we characterize three Cre driver lines for the nuclei of the dorsal thalamus: Oligodendrocyte transcription factor 3 (Olig3)-Cre, histidine decarboxylase (HDC)-Cre, and corticotropin-releasing hormone (CRH)-Cre. We examined the postnatal distribution of Cre expression for each of these lines with the Cre-dependent reporter CAG-tdTomato (Ai9). Cre-dependent expression of tdTomato reveals that Olig3-Cre expresses broadly within the thalamus, including TC neurons and interneurons, while HDC-Cre and CRH-Cre each have unique patterns of expression restricted to TC neurons within and across the sensory relay nuclei of the dorsal thalamus. Cre expression is present by the time of natural birth in all three lines, underscoring their utility for developmental studies. To demonstrate the utility of these Cre drivers for studying sensory TC circuitry, we targeted the expression of channelrhodopsin-2 to thalamus from the CAG-COP4*H134R/EYFP (Ai32) allele with either HDC-Cre or CRH-Cre. Optogenetic activation of TC afferents in primary visual cortex was sufficient to measure frequency-dependent depression. Thus, these Cre drivers provide selective Cre-dependent gene expression in thalamus suitable for both anatomical and functional studies.
Collapse
Affiliation(s)
- Guela Sokhadze
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Peter W Campbell
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Naomi Charalambakis
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Gubbi Govindaiah
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - William Guido
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Aaron W McGee
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
30
|
Sedigh-Sarvestani M, Fitzpatrick D. What and Where: Location-Dependent Feature Sensitivity as a Canonical Organizing Principle of the Visual System. Front Neural Circuits 2022; 16:834876. [PMID: 35498372 PMCID: PMC9039279 DOI: 10.3389/fncir.2022.834876] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/01/2022] [Indexed: 11/13/2022] Open
Abstract
Traditionally, functional representations in early visual areas are conceived as retinotopic maps preserving ego-centric spatial location information while ensuring that other stimulus features are uniformly represented for all locations in space. Recent results challenge this framework of relatively independent encoding of location and features in the early visual system, emphasizing location-dependent feature sensitivities that reflect specialization of cortical circuits for different locations in visual space. Here we review the evidence for such location-specific encoding including: (1) systematic variation of functional properties within conventional retinotopic maps in the cortex; (2) novel periodic retinotopic transforms that dramatically illustrate the tight linkage of feature sensitivity, spatial location, and cortical circuitry; and (3) retinotopic biases in cortical areas, and groups of areas, that have been defined by their functional specializations. We propose that location-dependent feature sensitivity is a fundamental organizing principle of the visual system that achieves efficient representation of positional regularities in visual experience, and reflects the evolutionary selection of sensory and motor circuits to optimally represent behaviorally relevant information. Future studies are necessary to discover mechanisms underlying joint encoding of location and functional information, how this relates to behavior, emerges during development, and varies across species.
Collapse
|
31
|
Chronic Monocular Deprivation Reveals MMP9-Dependent and -Independent Aspects of Murine Visual System Plasticity. Int J Mol Sci 2022; 23:ijms23052438. [PMID: 35269580 PMCID: PMC8909986 DOI: 10.3390/ijms23052438] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/17/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
The deletion of matrix metalloproteinase MMP9 is combined here with chronic monocular deprivation (cMD) to identify the contributions of this proteinase to plasticity in the visual system. Calcium imaging of supragranular neurons of the binocular region of primary visual cortex (V1b) of wild-type mice revealed that cMD initiated at eye opening significantly decreased the strength of deprived-eye visual responses to all stimulus contrasts and spatial frequencies. cMD did not change the selectivity of V1b neurons for the spatial frequency, but orientation selectivity was higher in low spatial frequency-tuned neurons, and orientation and direction selectivity were lower in high spatial frequency-tuned neurons. Constitutive deletion of MMP9 did not impact the stimulus selectivity of V1b neurons, including ocular preference and tuning for spatial frequency, orientation, and direction. However, MMP9-/- mice were completely insensitive to plasticity engaged by cMD, such that the strength of the visual responses evoked by deprived-eye stimulation was maintained across all stimulus contrasts, orientations, directions, and spatial frequencies. Other forms of experience-dependent plasticity, including stimulus selective response potentiation, were normal in MMP9-/- mice. Thus, MMP9 activity is dispensable for many forms of activity-dependent plasticity in the mouse visual system, but is obligatory for the plasticity engaged by cMD.
Collapse
|
32
|
Raghuram V, Werginz P, Fried SI, Timko BP. Morphological Factors that Underlie Neural Sensitivity to Stimulation in the Retina. ADVANCED NANOBIOMED RESEARCH 2021; 1:2100069. [PMID: 35399546 PMCID: PMC8993153 DOI: 10.1002/anbr.202100069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Retinal prostheses are a promising therapeutic intervention for patients afflicted by outer retinal degenerative diseases like retinitis pigmentosa and age-related macular degeneration. While significant advances in the development of retinal implants have been made, the quality of vision elicited by these devices remains largely sub-optimal. The variability in the responses produced by retinal devices is most likely due to the differences between the natural cell type-specific signaling that occur in the healthy retina vs. the non-specific activation of multiple cell types arising from artificial stimulation. In order to replicate these natural signaling patterns, stimulation strategies must be capable of preferentially activating specific RGC types. To design more selective stimulation strategies, a better understanding of the morphological factors that underlie the sensitivity to prosthetic stimulation must be developed. This review will focus on the role that different anatomical components play in driving the direct activation of RGCs by extracellular stimulation. Briefly, it will (1) characterize the variability in morphological properties of α-RGCs, (2) detail the influence of morphology on the direct activation of RGCs by electric stimulation, and (3) describe some of the potential biophysical mechanisms that could explain differences in activation thresholds and electrically evoked responses between RGC types.
Collapse
Affiliation(s)
- Vineeth Raghuram
- Boston VA Healthcare System, 150 S Huntington Ave, Boston, MA 02130, USA
- Dept. of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
- Dept. of Neurosurgery, Massachusetts General Hospital - Harvard Medical School, 50 Blossom Street, Boston, MA, 02114
| | - Paul Werginz
- Institute for Analysis and Scientific Computing, Vienna University of Technology, Wiedner Hauptstrasse 8-10, Vienna, Austria
- Dept. of Neurosurgery, Massachusetts General Hospital - Harvard Medical School, 50 Blossom Street, Boston, MA, 02114
| | - Shelley I. Fried
- Boston VA Healthcare System, 150 S Huntington Ave, Boston, MA 02130, USA
- Dept. of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
- Dept. of Neurosurgery, Massachusetts General Hospital - Harvard Medical School, 50 Blossom Street, Boston, MA, 02114
| | - Brian P. Timko
- Dept. of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| |
Collapse
|
33
|
Li T, Yu D, Oak HC, Zhu B, Wang L, Jiang X, Molday RS, Kriegstein A, Piao X. Phospholipid-flippase chaperone CDC50A is required for synapse maintenance by regulating phosphatidylserine exposure. EMBO J 2021; 40:e107915. [PMID: 34585770 PMCID: PMC8561630 DOI: 10.15252/embj.2021107915] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 08/25/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022] Open
Abstract
Synaptic refinement is a critical physiological process that removes excess synapses to establish and maintain functional neuronal circuits. Recent studies have shown that focal exposure of phosphatidylserine (PS) on synapses acts as an "eat me" signal to mediate synaptic pruning. However, the molecular mechanism underlying PS externalization at synapses remains elusive. Here, we find that murine CDC50A, a chaperone of phospholipid flippases, localizes to synapses, and that its expression depends on neuronal activity. Cdc50a knockdown leads to phosphatidylserine exposure at synapses and subsequent erroneous synapse removal by microglia partly via the GPR56 pathway. Taken together, our data support that CDC50A safeguards synapse maintenance by regulating focal phosphatidylserine exposure at synapses.
Collapse
Affiliation(s)
- Tao Li
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Newborn Brain Research InstituteUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
| | - Diankun Yu
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Newborn Brain Research InstituteUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
| | - Hayeon C Oak
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Newborn Brain Research InstituteUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
| | - Beika Zhu
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Newborn Brain Research InstituteUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
| | - Li Wang
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Department of NeurologyUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Xueqiao Jiang
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Newborn Brain Research InstituteUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
| | - Robert S Molday
- Department of Biochemistry and Molecular BiologyUniversity of British ColumbiaVancouverBCCanada
| | - Arnold Kriegstein
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Department of NeurologyUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Xianhua Piao
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Newborn Brain Research InstituteUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Division of NeonatologyDepartment of PediatricsUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
| |
Collapse
|
34
|
Sabbagh U, Govindaiah G, Somaiya RD, Ha RV, Wei JC, Guido W, Fox MA. Diverse GABAergic neurons organize into subtype-specific sublaminae in the ventral lateral geniculate nucleus. J Neurochem 2021; 159:479-497. [PMID: 32497303 PMCID: PMC8210463 DOI: 10.1111/jnc.15101] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 12/16/2022]
Abstract
In the visual system, retinal axons convey visual information from the outside world to dozens of distinct retinorecipient brain regions and organize that information at several levels, including either at the level of retinal afferents, cytoarchitecture of intrinsic retinorecipient neurons, or a combination of the two. Two major retinorecipient nuclei which are densely innervated by retinal axons are the dorsal lateral geniculate nucleus, which is important for classical image-forming vision, and ventral LGN (vLGN), which is associated with non-image-forming vision. The neurochemistry, cytoarchitecture, and retinothalamic connectivity in vLGN remain unresolved, raising fundamental questions of how it receives and processes visual information. To shed light on these important questions, used in situ hybridization, immunohistochemistry, and genetic reporter lines to identify and characterize novel neuronal cell types in mouse vLGN. Not only were a high percentage of these cells GABAergic, we discovered transcriptomically distinct GABAergic cell types reside in the two major laminae of vLGN, the retinorecipient, external vLGN (vLGNe) and the non-retinorecipient, internal vLGN (vLGNi). Furthermore, within vLGNe, we identified transcriptionally distinct subtypes of GABAergic cells that are distributed into four adjacent sublaminae. Using trans-synaptic viral tracing and in vitro electrophysiology, we found cells in each these vLGNe sublaminae receive monosynaptic inputs from retina. These results not only identify novel subtypes of GABAergic cells in vLGN, they suggest the subtype-specific laminar distribution of retinorecipient cells in vLGNe may be important for receiving, processing, and transmitting light-derived signals in parallel channels of the subcortical visual system.
Collapse
Affiliation(s)
- Ubadah Sabbagh
- Center for Neurobiology ResearchFralin Biomedical Research Institute at Virginia Tech CarilionRoanokeVAUSA
- Graduate Program in Translational Biology, Medicine, and HealthVirginia TechBlacksburgVAUSA
| | - Gubbi Govindaiah
- Department of Anatomical Sciences and NeurobiologyUniversity of Louisville School of MedicineLouisvilleKYUSA
| | - Rachana D. Somaiya
- Center for Neurobiology ResearchFralin Biomedical Research Institute at Virginia Tech CarilionRoanokeVAUSA
- Graduate Program in Translational Biology, Medicine, and HealthVirginia TechBlacksburgVAUSA
| | - Ryan V. Ha
- School of NeuroscienceVirginia TechBlacksburgVAUSA
| | - Jessica C. Wei
- NeuroSURFFralin Biomedical Research Institute at Virginia Tech CarilionRoanokeVAUSA
| | - William Guido
- Department of Anatomical Sciences and NeurobiologyUniversity of Louisville School of MedicineLouisvilleKYUSA
| | - Michael A. Fox
- Center for Neurobiology ResearchFralin Biomedical Research Institute at Virginia Tech CarilionRoanokeVAUSA
- School of NeuroscienceVirginia TechBlacksburgVAUSA
- Department of Biological SciencesVirginia TechBlacksburgVAUSA
- Department of PediatricsVirginia Tech Carilion School of MedicineRoanokeVAUSA
| |
Collapse
|
35
|
Zhu F, Shao J, Tian Y, Xu Z. Sulfiredoxin-1 protects retinal ganglion cells from high glucose-induced oxidative stress and inflammatory injury by potentiating Nrf2 signaling via the Akt/GSK-3β pathway. Int Immunopharmacol 2021; 101:108221. [PMID: 34653733 DOI: 10.1016/j.intimp.2021.108221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/17/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022]
Abstract
Sulfiredoxin-1 (Srxn1) has been acknowledged as a remarkable pro-survival factor in the protection of cells against stress-induced damage. The persistent exposure of retinal ganglion cells (RGCs) to high glucose (HG) in diabetes induces cellular damage, which contributes to the onset of diabetic retinopathy, a severe complication of diabetes. So far, little is known about the role of Srxn1 in regulating HG-induced injury of RGCs. The goals of this work were to evaluate the possible relevance of Srxn1 in the modulation of HG-induced apoptosis, oxidative stress and inflammation of RGCs in vitro. Our data showed that HG exposure caused a marked decrease in Srxn1 expression in RGCs. The up-regulation of Srxn1 markedly decreased HG-evoked apoptosis, reactive oxygen species (ROS) generation and pro-inflammatory cytokine release in RGCs. On the contrary, the depletion of Srxn1 rendered RGCs more susceptible to HG-induced injury. Further data demonstrated that Srnx1 enhanced the activation of nuclear factor erythroid-2 (E2)-related factor 2 (Nrf2) signaling in HG-exposed RGCs associated with up-regulating the phosphorylation of Akt and glucogen synthase kinase-3β (GSK-3β). Notably, the inhibition of Akt abolished Srnx1-overexpression-mediated Nrf2 activation, while GSK-3β inhibition reversed Srnx1-depletion-mediated inactivation of Nrf2. In addition, Nrf2 inhibition partially abrogated Srnx1-mediated protective effects against HG-induced injury of RGCs. In summary, these data demonstrate that the overexpression of Srxn1 protects RGCs from the HG-induced injury of RGCs by enhancing Nrf2 signaling via modulation of Akt/GSK-3β axis. Our work highlights that the Srxn1-mediated Akt/GSK-3β/Nrf2 axis may exert a possible role in regulating RGC injury of diabetic retinopathy.
Collapse
Affiliation(s)
- Fei Zhu
- Ophthalmology, Yulin Hospital of Traditional Chinese Medicine, Yulin 719000, China
| | - Juan Shao
- Ophthalmology, Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated Guangren Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, China.
| | - Yunlin Tian
- Ophthalmology, Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated Guangren Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, China
| | - Zhiguo Xu
- Ophthalmology, Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated Guangren Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
36
|
Visual Disfunction due to the Selective Effect of Glutamate Agonists on Retinal Cells. Int J Mol Sci 2021; 22:ijms22126245. [PMID: 34200611 PMCID: PMC8230349 DOI: 10.3390/ijms22126245] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/25/2021] [Accepted: 05/30/2021] [Indexed: 02/07/2023] Open
Abstract
One of the causes of nervous system degeneration is an excess of glutamate released upon several diseases. Glutamate analogs, like N-methyl-DL-aspartate (NMDA) and kainic acid (KA), have been shown to induce experimental retinal neurotoxicity. Previous results have shown that NMDA/KA neurotoxicity induces significant changes in the full field electroretinogram response, a thinning on the inner retinal layers, and retinal ganglion cell death. However, not all types of retinal neurons experience the same degree of injury in response to the excitotoxic stimulus. The goal of the present work is to address the effect of intraocular injection of different doses of NMDA/KA on the structure and function of several types of retinal cells and their functionality. To globally analyze the effect of glutamate receptor activation in the retina after the intraocular injection of excitotoxic agents, a combination of histological, electrophysiological, and functional tools has been employed to assess the changes in the retinal structure and function. Retinal excitotoxicity caused by the intraocular injection of a mixture of NMDA/KA causes a harmful effect characterized by a great loss of bipolar, amacrine, and retinal ganglion cells, as well as the degeneration of the inner retina. This process leads to a loss of retinal cell functionality characterized by an impairment of light sensitivity and visual acuity, with a strong effect on the retinal OFF pathway. The structural and functional injury suffered by the retina suggests the importance of the glutamate receptors expressed by different types of retinal cells. The effect of glutamate agonists on the OFF pathway represents one of the main findings of the study, as the evaluation of the retinal lesions caused by excitotoxicity could be specifically explored using tests that evaluate the OFF pathway.
Collapse
|
37
|
Meier AM, Wang Q, Ji W, Ganachaud J, Burkhalter A. Modular Network between Postrhinal Visual Cortex, Amygdala, and Entorhinal Cortex. J Neurosci 2021; 41:4809-4825. [PMID: 33849948 PMCID: PMC8260166 DOI: 10.1523/jneurosci.2185-20.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 11/21/2022] Open
Abstract
The postrhinal area (POR) is a known center for integrating spatial with nonspatial visual information and a possible hub for influencing landmark navigation by affective input from the amygdala. This may involve specific circuits within muscarinic acetylcholine receptor 2 (M2)-positive (M2+) or M2- modules of POR that associate inputs from the thalamus, cortex, and amygdala, and send outputs to the entorhinal cortex. Using anterograde and retrograde labeling with conventional and viral tracers in male and female mice, we found that all higher visual areas of the ventral cortical stream project to the amygdala, while such inputs are absent from primary visual cortex and dorsal stream areas. Unexpectedly for the presumed salt-and-pepper organization of mouse extrastriate cortex, tracing results show that inputs from the dorsal lateral geniculate nucleus and lateral posterior nucleus were spatially clustered in layer 1 (L1) and overlapped with M2+ patches of POR. In contrast, input from the amygdala to L1 of POR terminated in M2- interpatches. Importantly, the amygdalocortical input to M2- interpatches in L1 overlapped preferentially with spatially clustered apical dendrites of POR neurons projecting to amygdala and entorhinal area lateral, medial (ENTm). The results suggest that subnetworks in POR, used to build spatial maps for navigation, do not receive direct thalamocortical M2+ patch-targeting inputs. Instead, they involve local networks of M2- interpatches, which are influenced by affective information from the amygdala and project to ENTm, whose cells respond to visual landmark cues for navigation.SIGNIFICANCE STATEMENT A central purpose of visual object recognition is identifying the salience of objects and approaching or avoiding them. However, it is not currently known how the visual cortex integrates the multiple streams of information, including affective and navigational cues, which are required to accomplish this task. We find that in a higher visual area, the postrhinal cortex, the cortical sheet is divided into interdigitating modules receiving distinct inputs from visual and emotion-related sources. One of these modules is preferentially connected with the amygdala and provides outputs to entorhinal cortex, constituting a processing stream that may assign emotional salience to objects and landmarks for the guidance of goal-directed navigation.
Collapse
Affiliation(s)
- Andrew M Meier
- Department of Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110
| | - Quanxin Wang
- Department of Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110
| | - Weiqing Ji
- Department of Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110
| | - Jehan Ganachaud
- Department of Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110
| | - Andreas Burkhalter
- Department of Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110
| |
Collapse
|
38
|
Isa T, Marquez-Legorreta E, Grillner S, Scott EK. The tectum/superior colliculus as the vertebrate solution for spatial sensory integration and action. Curr Biol 2021; 31:R741-R762. [PMID: 34102128 DOI: 10.1016/j.cub.2021.04.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The superior colliculus, or tectum in the case of non-mammalian vertebrates, is a part of the brain that registers events in the surrounding space, often through vision and hearing, but also through electrosensation, infrared detection, and other sensory modalities in diverse vertebrate lineages. This information is used to form maps of the surrounding space and the positions of different salient stimuli in relation to the individual. The sensory maps are arranged in layers with visual input in the uppermost layer, other senses in deeper positions, and a spatially aligned motor map in the deepest layer. Here, we will review the organization and intrinsic function of the tectum/superior colliculus and the information that is processed within tectal circuits. We will also discuss tectal/superior colliculus outputs that are conveyed directly to downstream motor circuits or via the thalamus to cortical areas to control various aspects of behavior. The tectum/superior colliculus is evolutionarily conserved among all vertebrates, but tailored to the sensory specialties of each lineage, and its roles have shifted with the emergence of the cerebral cortex in mammals. We will illustrate both the conserved and divergent properties of the tectum/superior colliculus through vertebrate evolution by comparing tectal processing in lampreys belonging to the oldest group of extant vertebrates, larval zebrafish, rodents, and other vertebrates including primates.
Collapse
Affiliation(s)
- Tadashi Isa
- Department of Neuroscience, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan; Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, 606-8501, Japan
| | | | - Sten Grillner
- Department of Neuroscience, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Ethan K Scott
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
39
|
Choi BJ, Chen YCD, Desplan C. Building a circuit through correlated spontaneous neuronal activity in the developing vertebrate and invertebrate visual systems. Genes Dev 2021; 35:677-691. [PMID: 33888564 PMCID: PMC8091978 DOI: 10.1101/gad.348241.121] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
During the development of the vertebrate nervous systems, genetic programs assemble an immature circuit that is subsequently refined by neuronal activity evoked by external stimuli. However, prior to sensory experience, the intrinsic property of the developing nervous system also triggers correlated network-level neuronal activity, with retinal waves in the developing vertebrate retina being the best documented example. Spontaneous activity has also been found in the visual system of Drosophila Here, we compare the spontaneous activity of the developing visual system between mammalian and Drosophila and suggest that Drosophila is an emerging model for mechanistic and functional studies of correlated spontaneous activity.
Collapse
Affiliation(s)
- Ben Jiwon Choi
- Department of Biology, New York University, New York, New York 10003, USA
| | | | - Claude Desplan
- Department of Biology, New York University, New York, New York 10003, USA
| |
Collapse
|
40
|
Liu H, Prokosch V. Energy Metabolism in the Inner Retina in Health and Glaucoma. Int J Mol Sci 2021; 22:ijms22073689. [PMID: 33916246 PMCID: PMC8036449 DOI: 10.3390/ijms22073689] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023] Open
Abstract
Glaucoma, the leading cause of irreversible blindness, is a heterogeneous group of diseases characterized by progressive loss of retinal ganglion cells (RGCs) and their axons and leads to visual loss and blindness. Risk factors for the onset and progression of glaucoma include systemic and ocular factors such as older age, lower ocular perfusion pressure, and intraocular pressure (IOP). Early signs of RGC damage comprise impairment of axonal transport, downregulation of specific genes and metabolic changes. The brain is often cited to be the highest energy-demanding tissue of the human body. The retina is estimated to have equally high demands. RGCs are particularly active in metabolism and vulnerable to energy insufficiency. Understanding the energy metabolism of the inner retina, especially of the RGCs, is pivotal for understanding glaucoma’s pathophysiology. Here we review the key contributors to the high energy demands in the retina and the distinguishing features of energy metabolism of the inner retina. The major features of glaucoma include progressive cell death of retinal ganglions and optic nerve damage. Therefore, this review focuses on the energetic budget of the retinal ganglion cells, optic nerve and the relevant cells that surround them.
Collapse
|
41
|
Lyu J, Mu X. Genetic control of retinal ganglion cell genesis. Cell Mol Life Sci 2021; 78:4417-4433. [PMID: 33782712 DOI: 10.1007/s00018-021-03814-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 02/27/2021] [Accepted: 03/18/2021] [Indexed: 12/18/2022]
Abstract
Retinal ganglion cells (RGCs) are the only projection neurons in the neural retina. They receive and integrate visual signals from upstream retinal neurons in the visual circuitry and transmit them to the brain. The function of RGCs is performed by the approximately 40 RGC types projecting to various central brain targets. RGCs are the first cell type to form during retinogenesis. The specification and differentiation of the RGC lineage is a stepwise process; a hierarchical gene regulatory network controlling the RGC lineage has been identified and continues to be elaborated. Recent studies with single-cell transcriptomics have led to unprecedented new insights into their types and developmental trajectory. In this review, we summarize our current understanding of the functions and relationships of the many regulators of the specification and differentiation of the RGC lineage. We emphasize the roles of these key transcription factors and pathways in different developmental steps, including the transition from retinal progenitor cells (RPCs) to RGCs, RGC differentiation, generation of diverse RGC types, and central projection of the RGC axons. We discuss critical issues that remain to be addressed for a comprehensive understanding of these different aspects of RGC genesis and emerging technologies, including single-cell techniques, novel genetic tools and resources, and high-throughput genome editing and screening assays, which can be leveraged in future studies.
Collapse
Affiliation(s)
- Jianyi Lyu
- Department of Ophthalmology/Ross Eye Institute, State University of New York At Buffalo, Buffalo, NY, 14203, USA
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xiuqian Mu
- Department of Ophthalmology/Ross Eye Institute, State University of New York At Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
42
|
Rasmussen RN, Matsumoto A, Arvin S, Yonehara K. Binocular integration of retinal motion information underlies optic flow processing by the cortex. Curr Biol 2021; 31:1165-1174.e6. [PMID: 33484637 PMCID: PMC7987724 DOI: 10.1016/j.cub.2020.12.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/10/2020] [Accepted: 12/21/2020] [Indexed: 11/17/2022]
Abstract
Locomotion creates various patterns of optic flow on the retina, which provide the observer with information about their movement relative to the environment. However, it is unclear how these optic flow patterns are encoded by the cortex. Here, we use two-photon calcium imaging in awake mice to systematically map monocular and binocular responses to horizontal motion in four areas of the visual cortex. We find that neurons selective to translational or rotational optic flow are abundant in higher visual areas, whereas neurons suppressed by binocular motion are more common in the primary visual cortex. Disruption of retinal direction selectivity in Frmd7 mutant mice reduces the number of translation-selective neurons in the primary visual cortex and translation- and rotation-selective neurons as well as binocular direction-selective neurons in the rostrolateral and anterior visual cortex, blurring the functional distinction between primary and higher visual areas. Thus, optic flow representations in specific areas of the visual cortex rely on binocular integration of motion information from the retina. Translation- and rotation-selective neurons are abundant in higher visual areas Optic-flow-selective neurons in V1 and RL/A rely on retinal direction selectivity Retinal direction selectivity controls functional segregation between V1 and RL/A Binocular integration of retinal motion information underlies optic flow selectivity
Collapse
Affiliation(s)
- Rune Nguyen Rasmussen
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Ole Worms Allé 8, 8000 Aarhus C, Denmark
| | - Akihiro Matsumoto
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Ole Worms Allé 8, 8000 Aarhus C, Denmark
| | - Simon Arvin
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Ole Worms Allé 8, 8000 Aarhus C, Denmark
| | - Keisuke Yonehara
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Ole Worms Allé 8, 8000 Aarhus C, Denmark.
| |
Collapse
|
43
|
Posttranslational Modification of Sox11 Regulates RGC Survival and Axon Regeneration. eNeuro 2021; 8:ENEURO.0358-20.2020. [PMID: 33441400 PMCID: PMC7890524 DOI: 10.1523/eneuro.0358-20.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/28/2022] Open
Abstract
The failure of adult CNS neurons to survive and regenerate their axons after injury or in neurodegenerative disease remains a major target for basic and clinical neuroscience. Recent data demonstrated in the adult mouse that exogenous expression of Sry-related high-mobility-box 11 (Sox11) promotes optic nerve regeneration after optic nerve injury but exacerbates the death of a subset of retinal ganglion cells (RGCs), α-RGCs. During development, Sox11 is required for RGC differentiation from retinal progenitor cells (RPCs), and we found that mutation of a single residue to prevent SUMOylation at lysine 91 (K91) increased Sox11 nuclear localization and RGC differentiation in vitro. Here, we explored whether this Sox11 manipulation similarly has stronger effects on RGC survival and optic nerve regeneration. In vitro, we found that non-SUMOylatable Sox11K91A leads to RGC death and suppresses axon outgrowth in primary neurons. We furthermore found that Sox11K91A more strongly promotes axon regeneration but also increases RGC death after optic nerve injury in vivo in the adult mouse. RNA sequence (RNA-seq) data showed that Sox11 and Sox11K91A increase the expression of key signaling pathway genes associated with axon growth and regeneration but downregulated Spp1 and Opn4 expression in RGC cultures, consistent with negatively regulating the survival of α-RGCs and ipRGCs. Thus, Sox11 and its SUMOylation site at K91 regulate gene expression, survival and axon growth in RGCs, and may be explored further as potential regenerative therapies for optic neuropathy.
Collapse
|
44
|
Abstract
Safe driving demands the coordination of multiple sensory and cognitive functions, such as vision and attention. Patients with neurologic or ophthalmic disease are exposed to selective pathophysiologic insults to driving-critical systems, placing them at a higher risk for unsafe driving and restricted driving privileges. Here, we evaluate how vision and attention contribute to unsafe driving across different patient populations. In ophthalmic disease, we focus on macular degeneration, glaucoma, diabetic retinopathy, and cataract; in neurologic disease, we focus on Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Unsafe driving is generally associated with impaired vision and attention in ophthalmic and neurologic patients, respectively. Furthermore, patients with ophthalmic disease experience some degree of impairment in attention. Similarly, patients with neurologic disease experience some degree of impairment in vision. While numerous studies have demonstrated a relationship between impaired vision and unsafe driving in neurologic disease, there remains a dearth of knowledge regarding the relationship between impaired attention and unsafe driving in ophthalmic disease. In summary, this chapter confirms-and offers opportunities for future research into-the contribution of vision and attention to safe driving.
Collapse
Affiliation(s)
- David E Anderson
- Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Deepta A Ghate
- Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Matthew Rizzo
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States.
| |
Collapse
|
45
|
Ge Y, Wu F, Cheng M, Bard J, Mu X. Two new genetically modified mouse alleles labeling distinct phases of retinal ganglion cell development by fluorescent proteins. Dev Dyn 2020; 249:1514-1528. [PMID: 32741043 PMCID: PMC7855626 DOI: 10.1002/dvdy.233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/14/2020] [Accepted: 07/29/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND During development, all retinal cell types arise from retinal progenitor cells (RPCs) in a step-wise fashion. Atoh7 and Pou4f2 mark, and function in, two phases of retinal ganglion cell (RGC) genesis; Atoh7 functions in a subpopulation of RPCs to render them competent for the RGC fate, whereas Pou4f2 participates in RGC fate specification and RGC differentiation. Despite extensive research on their roles, the properties of the two phases represented by these two factors have not been well studied, likely due to the retinal cellular heterogeneity. RESULTS In this report, we describe two novel knock-in mouse alleles, Atoh7zsGreenCreERT2 and Pou4f2FlagtdTomato , which labeled retinal cells in the two phases of RGC development by fluorescent proteins. Also, the Atoh7zsGreenCreERT2 allele allowed for indirect labeling of RGCs and other cell types upon tamoxifen induction in a dose-dependent manner. Further, these alleles could be used to purify retinal cells in the different phases by fluorescence assisted cell sorting (FACS). Single cell RNA-seq analysis of purified cells from Atoh7zsGreenCreERT2 retinas further validated that this allele labeled both transitional/competent RPCs and their progenies including RGCs. CONCLUSIONS Thus, these two alleles are very useful tools for studying the molecular and genetic mechanisms underlying RGC formation.
Collapse
Affiliation(s)
- Yichen Ge
- Department of Ophthalmology/Ross Eye Institute, University at Buffalo, Buffalo, NY
| | - Fuguo Wu
- Department of Ophthalmology/Ross Eye Institute, University at Buffalo, Buffalo, NY
| | - Mobin Cheng
- Department of Ophthalmology/Ross Eye Institute, University at Buffalo, Buffalo, NY
| | - Jonathan Bard
- New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY
| | - Xiuqian Mu
- Department of Ophthalmology/Ross Eye Institute, University at Buffalo, Buffalo, NY
- New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY
| |
Collapse
|
46
|
Muthuswamy A, Pardo ID, Rao DB, Switzer RC, Sharma AK, Bolon B. Neuroanatomy and Sampling of Central Projections for the Visual System in Mammals Used in Toxicity Testing. Toxicol Pathol 2020; 49:455-471. [PMID: 33243077 DOI: 10.1177/0192623320967279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Visual system toxicity may manifest anywhere in the visual system, from the eye proper to the visual brain. Therefore, effective screening for visual system toxicity must evaluate not only ocular structures (ie, eye and optic nerve) but also multiple key brain regions involved in vision (eg, optic tract, subcortical relay nuclei, and primary and secondary visual cortices). Despite a generally comparable pattern across species, the neuroanatomic organization and function of the visual brain in rodents and rabbits exhibit appreciable differences relative to nonrodents. Currently recognized sampling practices for general toxicity studies in animals, which are based on easily discerned external neuroanatomic landmarks and guided by extant stereotaxic brain atlases, typically will permit histopathologic evaluation of many brain centers involved in visual sensation (eg, optic chiasm, optic tract, dorsal lateral geniculate nucleus, primary and secondary visual cortices) and often some subcortical brain nuclei involved in light-modulated nonvisual activities needed for visual attention and orientation (eg, rostral colliculus in quadrupeds, termed the superior colliculus in bipeds; several cranial nerve nuclei). Pathologic findings induced by toxicants in the visual brain centers are similar to those that are produced in other brain regions.
Collapse
Affiliation(s)
| | - Ingrid D Pardo
- 390190Pfizer Inc, Global Pathology and Investigative Toxicology, Groton, CT, USA
| | - Deepa B Rao
- ToxPath Specialists LLC [a StageBio Company], Frederick, MD, USA
| | | | | | - Brad Bolon
- GEMpath Inc., Longmont, CO, USA * Deceased
| |
Collapse
|
47
|
Bell BJ, Wang AA, Kim DW, Xiong J, Blackshaw S, Wu MN. Characterization of mWake expression in the murine brain. J Comp Neurol 2020; 529:1954-1987. [PMID: 33140455 DOI: 10.1002/cne.25066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 01/24/2023]
Abstract
Structure-function analyses of the mammalian brain have historically relied on anatomically-based approaches. In these investigations, physical, chemical, or electrolytic lesions of anatomical structures are applied, and the resulting behavioral or physiological responses assayed. An alternative approach is to focus on the expression pattern of a molecule whose function has been characterized and then use genetic intersectional methods to optogenetically or chemogenetically manipulate distinct circuits. We previously identified WIDE AWAKE (WAKE) in Drosophila, a clock output molecule that mediates the temporal regulation of sleep onset and sleep maintenance. More recently, we have studied the mouse homolog, mWAKE/ANKFN1, and our data suggest that its basic role in the circadian regulation of arousal is conserved. Here, we perform a systematic analysis of the expression pattern of mWake mRNA, protein, and cells throughout the adult mouse brain. We find that mWAKE labels neurons in a restricted, but distributed manner, in multiple regions of the hypothalamus (including the suprachiasmatic nucleus, dorsomedial hypothalamus, and tuberomammillary nucleus region), the limbic system, sensory processing nuclei, and additional specific brainstem, subcortical, and cortical areas. Interestingly, mWAKE is also observed in non-neuronal ependymal cells. In addition, to describe the molecular identities and clustering of mWake+ cells, we provide detailed analyses of single cell RNA sequencing data from the hypothalamus, a region with particularly significant mWAKE expression. These findings lay the groundwork for future studies into the potential role of mWAKE+ cells in the rhythmic control of diverse behaviors and physiological processes.
Collapse
Affiliation(s)
- Benjamin J Bell
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Annette A Wang
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Dong Won Kim
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jiali Xiong
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University, Baltimore, Maryland, USA
| | - Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mark N Wu
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
48
|
Alvarado JA, Dhande OS, Prosseda PP, Kowal TJ, Ning K, Jabbehdari S, Hu Y, Sun Y. Developmental distribution of primary cilia in the retinofugal visual pathway. J Comp Neurol 2020; 529:1442-1455. [PMID: 32939774 DOI: 10.1002/cne.25029] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/21/2020] [Accepted: 08/17/2020] [Indexed: 12/21/2022]
Abstract
The mammalian visual system is composed of circuitry connecting sensory input from the retina to the processing core of the visual cortex. The two main retinorecipient brain targets, the superior colliculus (SC) and dorsal lateral geniculate nucleus (dLGN), bridge retinal input and visual output. The primary cilium is a conserved organelle increasingly viewed as a critical sensor for the regulation of developmental and homeostatic pathways in most mammalian cell types. Moreover, cilia have been described as crucial for neurogenesis, neuronal maturation, and survival in the cortex and retina. However, cilia in the visual relay center remain to be fully described. In this study, we characterized the ciliation profile of the SC and dLGN and found that the overall number of ciliated cells declined during development. Interestingly, shorter ciliated cells in both regions were identified as neurons, whose numbers remained stable over time, suggesting that cilia retention is a critical feature for optimal neuronal function in SC and dLGN. Our study suggests that primary cilia are important for neuronal maturation and function in cells of the SC and dLGN.
Collapse
Affiliation(s)
- Jorge A Alvarado
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Onkar S Dhande
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Philipp P Prosseda
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Tia J Kowal
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Ke Ning
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Sayena Jabbehdari
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA.,Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA.,Palo Alto Veterans Administration, Palo Alto, California, USA
| |
Collapse
|
49
|
Singh RK, Nasonkin IO. Limitations and Promise of Retinal Tissue From Human Pluripotent Stem Cells for Developing Therapies of Blindness. Front Cell Neurosci 2020; 14:179. [PMID: 33132839 PMCID: PMC7513806 DOI: 10.3389/fncel.2020.00179] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/25/2020] [Indexed: 12/13/2022] Open
Abstract
The self-formation of retinal tissue from pluripotent stem cells generated a tremendous promise for developing new therapies of retinal degenerative diseases, which previously seemed unattainable. Together with use of induced pluripotent stem cells or/and CRISPR-based recombineering the retinal organoid technology provided an avenue for developing models of human retinal degenerative diseases "in a dish" for studying the pathology, delineating the mechanisms and also establishing a platform for large-scale drug screening. At the same time, retinal organoids, highly resembling developing human fetal retinal tissue, are viewed as source of multipotential retinal progenitors, young photoreceptors and just the whole retinal tissue, which may be transplanted into the subretinal space with a goal of replacing patient's degenerated retina with a new retinal "patch." Both approaches (transplantation and modeling/drug screening) were projected when Yoshiki Sasai demonstrated the feasibility of deriving mammalian retinal tissue from pluripotent stem cells, and generated a lot of excitement. With further work and testing of both approaches in vitro and in vivo, a major implicit limitation has become apparent pretty quickly: the absence of the uniform layer of Retinal Pigment Epithelium (RPE) cells, which is normally present in mammalian retina, surrounds photoreceptor layer and develops and matures first. The RPE layer polarize into apical and basal sides during development and establish microvilli on the apical side, interacting with photoreceptors, nurturing photoreceptor outer segments and participating in the visual cycle by recycling 11-trans retinal (bleached pigment) back to 11-cis retinal. Retinal organoids, however, either do not have RPE layer or carry patches of RPE mostly on one side, thus directly exposing most photoreceptors in the developing organoids to neural medium. Recreation of the critical retinal niche between the apical RPE and photoreceptors, where many retinal disease mechanisms originate, is so far unattainable, imposes clear limitations on both modeling/drug screening and transplantation approaches and is a focus of investigation in many labs. Here we dissect different retinal degenerative diseases and analyze how and where retinal organoid technology can contribute the most to developing therapies even with a current limitation and absence of long and functional outer segments, supported by RPE.
Collapse
|
50
|
Korshunov KS, Blakemore LJ, Trombley PQ. Illuminating and Sniffing Out the Neuromodulatory Roles of Dopamine in the Retina and Olfactory Bulb. Front Cell Neurosci 2020; 14:275. [PMID: 33110404 PMCID: PMC7488387 DOI: 10.3389/fncel.2020.00275] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/04/2020] [Indexed: 01/28/2023] Open
Abstract
In the central nervous system, dopamine is well-known as the neuromodulator that is involved with regulating reward, addiction, motivation, and fine motor control. Yet, decades of findings are revealing another crucial function of dopamine: modulating sensory systems. Dopamine is endogenous to subsets of neurons in the retina and olfactory bulb (OB), where it sharpens sensory processing of visual and olfactory information. For example, dopamine modulation allows the neural circuity in the retina to transition from processing dim light to daylight and the neural circuity in the OB to regulate odor discrimination and detection. Dopamine accomplishes these tasks through numerous, complex mechanisms in both neural structures. In this review, we provide an overview of the established and emerging research on these mechanisms and describe similarities and differences in dopamine expression and modulation of synaptic transmission in the retinas and OBs of various vertebrate organisms. This includes discussion of dopamine neurons’ morphologies, potential identities, and biophysical properties along with their contributions to circadian rhythms and stimulus-driven synthesis, activation, and release of dopamine. As dysregulation of some of these mechanisms may occur in patients with Parkinson’s disease, these symptoms are also discussed. The exploration and comparison of these two separate dopamine populations shows just how remarkably similar the retina and OB are, even though they are functionally distinct. It also shows that the modulatory properties of dopamine neurons are just as important to vision and olfaction as they are to motor coordination and neuropsychiatric/neurodegenerative conditions, thus, we hope this review encourages further research to elucidate these mechanisms.
Collapse
Affiliation(s)
- Kirill S Korshunov
- Department of Biological Science, Florida State University, Tallahassee, FL, United States.,Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Laura J Blakemore
- Department of Biological Science, Florida State University, Tallahassee, FL, United States.,Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Paul Q Trombley
- Department of Biological Science, Florida State University, Tallahassee, FL, United States.,Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| |
Collapse
|