1
|
Rusciano D, Bagnoli P. Multimodality in the Collicular Pathway: Towards Compensatory Visual Processes. Cells 2025; 14:635. [PMID: 40358159 PMCID: PMC12071832 DOI: 10.3390/cells14090635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/08/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
The integration of multisensory inputs plays a crucial role in shaping perception and behavior, particularly in the visual system. The collicular pathway, encompassing the optic tectum in non-mammalian vertebrates and the superior colliculus (SC) in mammals, is a key hub for integrating sensory information and mediating adaptive motor responses. Comparative studies across species reveal evolutionary adaptations that enhance sensory processing and contribute to compensatory mechanisms following neuronal injury. The present review outlines the structure and function of the multisensory visual pathways, emphasizing the retinocollicular projections, and their multisensory integration, which depends on synaptic convergence of afferents conveying information from different sensory modalities. The cellular mechanisms underlying multimodal integration remain to be fully clarified, and further investigations are needed to clarify the link between neuronal activity in response to multisensory stimulation and behavioral response involving motor activity. By exploring the interplay between fundamental neuroscience and translational applications, we aim to address multisensory integration as a pivotal target for its potential role in visual rehabilitation strategies.
Collapse
Affiliation(s)
| | - Paola Bagnoli
- Department of Biology, University of Pisa, 56123 Pisa, Italy
| |
Collapse
|
2
|
Yeh PT, Jhan KC, Chua EP, Chen WC, Chu SW, Wu SC, Chen SK. Discrete photoentrainment of mammalian central clock is regulated by bi-stable dynamic network in the suprachiasmatic nucleus. Nat Commun 2025; 16:3331. [PMID: 40199869 PMCID: PMC11978930 DOI: 10.1038/s41467-025-58661-1] [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: 06/11/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
The biological clock synchronizes with the environmental light-dark cycle through circadian photoentrainment. While intracellular pathways regulating clock gene expression after light exposure in the suprachiasmatic nucleus are well studied in mammals, the neuronal circuits driving phase shifts remain unclear. Here, using a mouse model, we show that chemogenetic activation of early-night light-responsive neurons induces phase delays at any circadian time, potentially breaking the photoentrainment dead zone. In contrast, activating late-night light-responsive neurons mimics light-induced phase shifts. Using in vivo two-photon microscopy, we found that most neurons in the suprachiasmatic nucleus exhibit stochastic light responses, while a small subset is consistently activated in the early subjective night and another is inhibited in the late subjective night. Our findings suggest a dynamic bi-stable network model for circadian photoentrainment, where phase shifts arise from a functional circuit integrating signals to groups of outcome neurons, rather than a labeled-line principle seen in sensory systems.
Collapse
Affiliation(s)
- Po-Ting Yeh
- Department of Life Science, National Taiwan University, Taipei, 10617, Taiwan
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Taiwan University and Academia Sinica, Taipei, 11529, Taiwan
| | - Kai-Chun Jhan
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Ern-Pei Chua
- Department of Life Science, National Taiwan University, Taipei, 10617, Taiwan
| | - Wun-Ci Chen
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shi-Wei Chu
- Department of Physics, National Taiwan University, Taipei, 10617, Taiwan
- Brain Research Center, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shun-Chi Wu
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Brain Research Center, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shih-Kuo Chen
- Department of Life Science, National Taiwan University, Taipei, 10617, Taiwan.
- Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, 10617, Taiwan.
- Center for Biotechnology, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
3
|
Renteria CA, Kahng J, Tibble B, Iyer RR, Shi J, Algrain H, Chaney EJ, Aksamitiene E, Liu YZ, Robinson P, Schmidt T, Boppart SA. Two-photon activation, deactivation, and coherent control of melanopsin in live cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645437. [PMID: 40196647 PMCID: PMC11974792 DOI: 10.1101/2025.03.26.645437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Intrinsically photosensitive retinal ganglion cells are photoreceptors discovered in the last 20 years. These cells project to the suprachiasmatic nucleus of the brain to drive circadian rhythms, regulated by ambient light levels. The photopigment responsible for photoactivation in these cells, melanopsin, has been shown to exhibit many unique activation features among opsins. Notably, the photopigment can exist in three states dependent on the intensity and spectrum of ambient light, which affects its function. Despite increasing knowledge about these cells and melanopsin, tools that can manipulate their three states, and do so with single-cell precision, are limited. This reduces the extent to which circuit-level phenomena, and studying the implications of melanopsin tri-stability in living systems, can be pursued. In this report, we evoke and modulate calcium transients in live cells and intrinsically photosensitive retinal ganglion cells from isolated retinal tissues following two-photon excitation using near-infrared light pulses. We demonstrate that two-photon activation of melanopsin can successfully stimulate melanopsin-expressing cells with high spatio-temporal precision. Moreover, we demonstrate that the functional tri-stability of the photopigment can be interrogated by multiphoton excitation using spectral-temporal modulation of a broadband, ultrafast laser source.
Collapse
Affiliation(s)
- Carlos A. Renteria
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL
- NIH/NIBIB P41 Center for Label-free Imaging and Multiscale Biophotonics (CLIMB), University of Illinois Urbana-Champaign, Urbana, IL
| | - Jiho Kahng
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL
- Department of Engineering Physics, University of Illinois Urbana-Champaign, Urbana, IL
| | - Brian Tibble
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL
- Department of Molecular and Cellular Biology, University of Illinois Urbana-Champaign, Urbana, IL
| | - Rishyashring R. Iyer
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL
- NIH/NIBIB P41 Center for Label-free Imaging and Multiscale Biophotonics (CLIMB), University of Illinois Urbana-Champaign, Urbana, IL
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL
| | - Jindou Shi
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL
| | - Haya Algrain
- College of Natural and Mathematical Sciences, University of Maryland, Baltimore County, Baltimore, MD
| | - Eric J. Chaney
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL
| | - Edita Aksamitiene
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL
| | - Yuan-Zhi Liu
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL
| | - Phyllis Robinson
- College of Natural and Mathematical Sciences, University of Maryland, Baltimore County, Baltimore, MD
| | - Tiffany Schmidt
- Department of Neurobiology, Northwestern University, Evanston, IL
| | - Stephen A. Boppart
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL
- NIH/NIBIB P41 Center for Label-free Imaging and Multiscale Biophotonics (CLIMB), University of Illinois Urbana-Champaign, Urbana, IL
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL
| |
Collapse
|
4
|
Geng D, Li Y, Yang B, Zhang L, Gu H, Zhang T, Zhao Z, Liu H, Cui Q, Zheng R, Cao P, Zhang F. Cholecystokinin neurons in the spinal trigeminal nucleus interpolaris regulate mechanically evoked predatory hunting in male mice. Nat Commun 2025; 16:2544. [PMID: 40087271 PMCID: PMC11909130 DOI: 10.1038/s41467-025-57771-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Predatory hunting plays a critical role in animal survival. Motion-related vibrissal somatosensory signaling is essential for prey detection and hunting in mice. However, little is known about the neural circuits that convert vibrissal somatosensory cues to trigger predatory hunting. Here, we report that mechanical force onto the vibrissal area of the male mice is a key stimulus for predatory hunting. Mechanically evoked predatory hunting was abrogated by the chemogenetic inactivation of cholecystokinin-positive (Cck+) neurons in the spinal trigeminal nucleus interpolaris (Sp5I). The Cck+ Sp5I neurons responded to the intensity of mechanical stimulus and sent neural signals to the superior colliculus that were relevant to stereotypical predatory hunting motor actions. Synaptic inactivation of the projections from Cck+ Sp5I neurons to the superior colliculus impaired mechanically evoked predatory attacks. Together, these data reveal a spinal trigeminal nucleus neural circuit that is specifically engaged in translating vibrissal somatosensory cues to provoke predatory hunting.
Collapse
Affiliation(s)
- Dandan Geng
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Yaning Li
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Bo Yang
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Li Zhang
- National Institute of Biological Sciences, Beijing, China
| | - Huating Gu
- National Institute of Biological Sciences, Beijing, China
| | - Tianyun Zhang
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Zijie Zhao
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Hui Liu
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Qingzhuo Cui
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Rong Zheng
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Peng Cao
- National Institute of Biological Sciences, Beijing, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| | - Fan Zhang
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China.
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
5
|
Delpech C, Schaeffer J, Vilallongue N, Delaunay A, Benadjal A, Blot B, Excoffier B, Plissonnier E, Gascon E, Albert F, Paccard A, Saintpierre A, Gasnier C, Zagar Y, Castellani V, Belin S, Chédotal A, Nawabi H. Axon guidance during mouse central nervous system regeneration is required for specific brain innervation. Dev Cell 2024; 59:3213-3228.e8. [PMID: 39353435 DOI: 10.1016/j.devcel.2024.09.005] [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: 10/10/2023] [Revised: 07/11/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024]
Abstract
Reconstructing functional neuronal circuits is one major challenge of central nervous system repair. Through activation of pro-growth signaling pathways, some neurons achieve long-distance axon regrowth. Yet, functional reconnection has hardly been obtained, as these regenerating axons fail to resume their initial trajectory and reinnervate their proper target. Axon guidance is considered to be active only during development. Here, using the mouse visual system, we show that axon guidance is still active in the adult brain in regenerative conditions. We highlight that regenerating retinal ganglion cell axons avoid one of their primary targets, the suprachiasmatic nucleus (SCN), due to Slit/Robo repulsive signaling. Together with promoting regeneration, silencing Slit/Robo in vivo enables regenerating axons to enter the SCN and form active synapses. The newly formed circuit is associated with neuronal activation and functional recovery. Our results provide evidence that axon guidance mechanisms are required to reconnect regenerating axons to specific brain nuclei.
Collapse
Affiliation(s)
- Céline Delpech
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Julia Schaeffer
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Noemie Vilallongue
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Apolline Delaunay
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Amin Benadjal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Beatrice Blot
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Blandine Excoffier
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Elise Plissonnier
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Eduardo Gascon
- Aix Marseille University, CNRS, INT, Institute of Neurosci Timone, Marseille, France
| | - Floriane Albert
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Antoine Paccard
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Ana Saintpierre
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Celestin Gasnier
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Yvrick Zagar
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Valérie Castellani
- University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France
| | - Stephane Belin
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France; Institut de pathologie, groupe hospitalier Est, Hospices Civils de Lyon, Lyon, France
| | - Homaira Nawabi
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| |
Collapse
|
6
|
Klett N, Gompf HS, Allen CN, Cravetchi O, Hablitz LM, Gunesch AN, Irwin RP, Todd WD, Saper CB, Fuller PM. GABAergic signalling in the suprachiasmatic nucleus is required for coherent circadian rhythmicity. Eur J Neurosci 2024; 60:6652-6667. [PMID: 39558544 PMCID: PMC11612841 DOI: 10.1111/ejn.16582] [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: 02/20/2024] [Revised: 09/11/2024] [Accepted: 10/07/2024] [Indexed: 11/20/2024]
Abstract
The suprachiasmatic nucleus is the circadian pacemaker of the mammalian brain. Suprachiasmatic nucleus neurons display synchronization of their firing frequency on a circadian timescale, which is required for the pacemaker function of the suprachiasmatic nucleus. However, the mechanisms by which suprachiasmatic nucleus neurons remain synchronized in vivo are poorly understood, although synaptic communication is considered indispensable. Suprachiasmatic nucleus neurons contain the neurotransmitter GABA and express GABA receptors. This has inspired the hypothesis that GABA signalling may play a central role in network synchronization, although this remains untested in vivo. Here, using local genetic deletion, we show that disruption of GABA synaptic transmission within the suprachiasmatic nucleus of adult mice results in the eventual deterioration of physiological and behavioural rhythmicity in vivo and concomitant cellular desynchrony in vitro. These findings suggest that intercellular GABA signalling is essential for behavioural rhythmicity and cellular synchrony of the suprachiasmatic nucleus neural network.
Collapse
Affiliation(s)
- Nathan Klett
- Oregon Institute for Occupational Health SciencesUSA
- Neuroscience Graduate ProgramUSA
| | - Heinrich S. Gompf
- Department of Neurological SurgeryUniversity of California, DavisDavisCAUSA
| | - Charles N. Allen
- Oregon Institute for Occupational Health SciencesUSA
- Department of Behavioral NeuroscienceOregon Health & Science UniversityPortlandORUSA
| | | | - Lauren M. Hablitz
- Oregon Institute for Occupational Health SciencesUSA
- Department of Behavioral NeuroscienceOregon Health & Science UniversityPortlandORUSA
- Present address:
Center for Translational NeuromedicineUniversity of Rochester Medical CenterRochesterNYUSA
| | | | | | - William D. Todd
- Department of Neurology, Division of Sleep Medicine, and Program in NeuroscienceBeth Israel Deaconess Medical Center, Harvard Medical School BostonMAUSA
- Present address:
Department of Zoology and PhysiologyUniversity of WyomingLaramieWYUSA
| | - Clifford B. Saper
- Department of Neurology, Division of Sleep Medicine, and Program in NeuroscienceBeth Israel Deaconess Medical Center, Harvard Medical School BostonMAUSA
| | - Patrick M. Fuller
- Department of Neurological SurgeryUniversity of California, DavisDavisCAUSA
| |
Collapse
|
7
|
Yuan M, Tan G, Cai D, Luo X, Shen K, Deng Q, Lei X, Zeng WB, Luo MH, Huang L, Ren C, Shen Y. GABAergic Retinal Ganglion Cells Projecting to the Superior Colliculus Mediate the Looming-Evoked Flight Response. Neurosci Bull 2024; 40:1886-1900. [PMID: 39285154 PMCID: PMC11625033 DOI: 10.1007/s12264-024-01295-y] [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: 12/20/2023] [Accepted: 05/05/2024] [Indexed: 12/08/2024] Open
Abstract
The looming stimulus-evoked flight response to approaching predators is a defensive behavior in most animals. However, how looming stimuli are detected in the retina and transmitted to the brain remains unclear. Here, we report that a group of GABAergic retinal ganglion cells (RGCs) projecting to the superior colliculus (SC) transmit looming signals from the retina to the brain, mediating the looming-evoked flight behavior by releasing GABA. GAD2-Cre and vGAT-Cre transgenic mice were used in combination with Cre-activated anterograde or retrograde tracer viruses to map the inputs to specific GABAergic RGC circuits. Optogenetic technology was used to assess the function of SC-projecting GABAergic RGCs (scpgRGCs) in the SC. FDIO-DTA (Flp-dependent Double-Floxed Inverted Open reading frame-Diphtheria toxin) combined with the FLP (Florfenicol, Lincomycin & Prednisolone) approach was used to ablate or silence scpgRGCs. In the mouse retina, GABAergic RGCs project to different brain areas, including the SC. ScpgRGCs are monosynaptically connected to parvalbumin-positive SC neurons known to be required for the looming-evoked flight response. Optogenetic activation of scpgRGCs triggers GABA-mediated inhibition in SC neurons. Ablation or silencing of scpgRGCs compromises looming-evoked flight responses without affecting image-forming functions. Our study reveals that scpgRGCs control the looming-evoked flight response by regulating SC neurons via GABA, providing novel insight into the regulation of innate defensive behaviors.
Collapse
Affiliation(s)
- Man Yuan
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei, China
| | - Gao Tan
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei, China
| | - Danrui Cai
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei, China
| | - Xue Luo
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei, China
| | - Kejiong Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei, China
| | - Qinqin Deng
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei, China
| | - Xinlan Lei
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei, China
| | - Wen-Bo Zeng
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Wuhan, 430071, China
| | - Min-Hua Luo
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Wuhan, 430071, China
| | - Lu Huang
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, 510632, China
| | - Chaoran Ren
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, 510632, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510530, China
| | - Yin Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei, China.
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
8
|
Korympidou MM, Strauss S, Schubert T, Franke K, Berens P, Euler T, Vlasits AL. GABAergic amacrine cells balance biased chromatic information in the mouse retina. Cell Rep 2024; 43:114953. [PMID: 39509269 DOI: 10.1016/j.celrep.2024.114953] [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: 03/15/2024] [Revised: 07/30/2024] [Accepted: 10/21/2024] [Indexed: 11/15/2024] Open
Abstract
The retina extracts chromatic information present in an animal's environment. How this information is processed in the retina is not well understood. In the mouse, chromatic information is not collected equally throughout the retina. Green and UV signals are primarily detected in the dorsal and ventral retina, respectively. However, at the output of the retina, chromatic tuning is more mixed throughout the retina. This suggests that lateral processing by inhibitory amacrine cells shapes chromatic information at the retinal output. We systematically surveyed the chromatic responses of dendritic processes of the 40+ GABAergic amacrine cell types. We identified 25 functional types with distinct chromatic and achromatic properties. We used pharmacology and a biologically inspired deep learning model to explore how inhibition and excitation shape the properties of functional types. Our data suggest that amacrine cells balance the biased spectral tuning of excitation, thereby supporting diversity of chromatic information at the retinal output.
Collapse
Affiliation(s)
- Maria M Korympidou
- Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience (GTC), University of Tübingen, 72076 Tübingen, Germany
| | - Sarah Strauss
- Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience (GTC), University of Tübingen, 72076 Tübingen, Germany; Hertie Institute for AI in Brain Health, University of Tübingen, 72076 Tübingen, Germany
| | - Timm Schubert
- Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Katrin Franke
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Stanford, CA 94303, USA
| | - Philipp Berens
- Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Hertie Institute for AI in Brain Health, University of Tübingen, 72076 Tübingen, Germany; Tübingen AI Center, University of Tübingen, 72076 Tübingen, Germany
| | - Thomas Euler
- Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany.
| | - Anna L Vlasits
- Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Department of Ophthalmology & Visual Sciences, University of Illinois, Chicago, IL 60603, USA.
| |
Collapse
|
9
|
D'Souza SP, Upton BA, Eldred KC, Glass I, Nayak G, Grover K, Ahmed A, Nguyen MT, Hu YC, Gamlin P, Lang RA. Developmental control of rod number via a light-dependent retrograde pathway from intrinsically photosensitive retinal ganglion cells. Dev Cell 2024; 59:2897-2911.e6. [PMID: 39142280 PMCID: PMC11537824 DOI: 10.1016/j.devcel.2024.07.018] [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: 02/27/2024] [Revised: 06/07/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024]
Abstract
Photoreception is essential for the development of the visual system, shaping vision's first synapse to cortical development. Here, we find that the lighting environment controls developmental rod apoptosis via Opn4-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs). Using genetics, sensory environment manipulations, and computational approaches, we establish a pathway where light-dependent glutamate released from ipRGCs is detected via a transiently expressed glutamate receptor (Grik3) on rod precursors within the inner retina. Communication between these cells is mediated by hybrid neurites on ipRGCs that sense light before eye opening. These structures span the ipRGC-rod precursor distance over development and contain the machinery for photoreception (Opn4) and neurotransmitter release (Vglut2 & Syp). Assessment of the human gestational retina identifies conserved hallmarks of an ipRGC-to-rod axis, including displaced rod precursors, transient GRIK3 expression, and ipRGCs with deep-projecting neurites. This analysis defines an adaptive retrograde pathway linking the sensory environment to rod precursors via ipRGCs prior to eye opening.
Collapse
Affiliation(s)
- Shane P D'Souza
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Brian A Upton
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kiara C Eldred
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Ian Glass
- Birth Defects Research Laboratory, Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98195, USA
| | - Gowri Nayak
- Transgenic Animal and Genome Editing Core, Department of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kassidy Grover
- Division of Hematology and Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Abdulla Ahmed
- Medical Doctor (M.D.) Training Program, George Washington University School of Medicine, Washington, DC 20052, USA
| | - Minh-Thanh Nguyen
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yueh-Chiang Hu
- Transgenic Animal and Genome Editing Core, Department of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Paul Gamlin
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Richard A Lang
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Ophthalmology, University of Cincinnati, Cincinnati, OH 45229, USA.
| |
Collapse
|
10
|
Campbell I, Sharifpour R, Balda Aizpurua JF, Beckers E, Paparella I, Berger A, Koshmanova E, Mortazavi N, Read J, Zubkov M, Talwar P, Collette F, Sherif S, Phillips C, Lamalle L, Vandewalle G. Regional response to light illuminance across the human hypothalamus. eLife 2024; 13:RP96576. [PMID: 39466317 PMCID: PMC11517251 DOI: 10.7554/elife.96576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024] Open
Abstract
Light exerts multiple non-image-forming biological effects on physiology including the stimulation of alertness and cognition. However, the subcortical circuitry underlying the stimulating impact of light is not established in humans. We used 7 Tesla functional magnetic resonance imaging to assess the impact of variations in light illuminance on the regional activity of the hypothalamus while healthy young adults (N=26; 16 women; 24.3±2.9 y) were completing two auditory cognitive tasks. We find that, during both the executive and emotional tasks, higher illuminance triggered an activity increase over the posterior part of the hypothalamus, which includes part of the tuberomamillary nucleus and the posterior part of the lateral hypothalamus. In contrast, increasing illuminance evoked a decrease in activity over the anterior and ventral parts of the hypothalamus, encompassing notably the suprachiasmatic nucleus and another part of the tuberomammillary nucleus. Critically, the performance of the executive task was improved under higher illuminance and was negatively correlated with the activity of the posterior hypothalamus area. These findings reveal the distinct local dynamics of different hypothalamus regions that underlie the impact of light on cognition.
Collapse
Affiliation(s)
| | | | | | - Elise Beckers
- GIGA-CRC Human Imaging, University of LiègeLiègeBelgium
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht UniversityMaastrichtNetherlands
| | | | - Alexandre Berger
- GIGA-CRC Human Imaging, University of LiègeLiègeBelgium
- Synergia Medical SAMont-Saint-GuibertBelgium
- Institute of Neuroscience (IoNS), Department of Clinical Neuroscience, Université Catholique de Louvain (UCLouvain)Woluwe-Saint-LambertBelgium
| | | | | | - John Read
- GIGA-CRC Human Imaging, University of LiègeLiègeBelgium
| | | | - Puneet Talwar
- GIGA-CRC Human Imaging, University of LiègeLiègeBelgium
| | | | - Siya Sherif
- GIGA-CRC Human Imaging, University of LiègeLiègeBelgium
| | | | | | | |
Collapse
|
11
|
Saito M, Miyamoto K, Murakami I. Illumination by short-wavelength light inside the blind spot decreases light detectability. iScience 2024; 27:110612. [PMID: 39220265 PMCID: PMC11363485 DOI: 10.1016/j.isci.2024.110612] [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: 02/06/2024] [Revised: 04/29/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Although the optic disk corresponding to the blind spot contains no classical photoreceptors, it contains photopigment melanopsin. To clarify whether melanopsin is involved in light detection, we conducted detection tasks for light stimuli presented in the normal visual field, with and without another illumination inside the blind spot. We found that a blue blind-spot illumination decreased the light detectability on a dark background. This effect was replicable when it was determined immediately after the blind-spot illumination was turned off, suggesting the contribution of a sluggish system rather than scattering. Moreover, the aforementioned effect was not observed when the blind-spot illumination was in red, indicating wavelength specificity in favor of melanopsin's sensitivity profile. These findings suggest that melanopsin is activated by the blind-spot illumination and thereby interferes with light detection near the absolute threshold. Light detection originating from conventional photoreceptors is modulated by melanopsin-based computation presumably estimating a baseline noise level.
Collapse
Affiliation(s)
- Marina Saito
- Department of Psychology, the University of Tokyo, Tokyo 113-0033, Japan
- Japan Society for the Promotion of Science, Tokyo 102-0083, Japan
- Faculty of Design and Architecture, Nagoya City University, Nagoya 467-8501, Japan
| | - Kentaro Miyamoto
- Laboratory for Imagination and Executive Functions, RIKEN Center for Brain Science, Wako 351-0198, Japan
| | - Ikuya Murakami
- Department of Psychology, the University of Tokyo, Tokyo 113-0033, Japan
| |
Collapse
|
12
|
Carrero L, Antequera D, Municio C, Carro E. Circadian rhythm disruption and retinal dysfunction: a bidirectional link in Alzheimer's disease? Neural Regen Res 2024; 19:1967-1972. [PMID: 38227523 DOI: 10.4103/1673-5374.390962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/07/2023] [Indexed: 01/17/2024] Open
Abstract
Dysfunction in circadian rhythms is a common occurrence in patients with Alzheimer's disease. A predominant function of the retina is circadian synchronization, carrying information to the brain through the retinohypothalamic tract, which projects to the suprachiasmatic nucleus. Notably, Alzheimer's disease hallmarks, including amyloid-β, are present in the retinas of Alzheimer's disease patients, followed/associated by structural and functional disturbances. However, the mechanistic link between circadian dysfunction and the pathological changes affecting the retina in Alzheimer's disease is not fully understood, although some studies point to the possibility that retinal dysfunction could be considered an early pathological process that directly modulates the circadian rhythm.
Collapse
Affiliation(s)
- Laura Carrero
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
- PhD Program in Neuroscience, Autonoma de Madrid University, Madrid, Spain
| | - Desireé Antequera
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain; Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| | - Cristina Municio
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| | - Eva Carro
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain; Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| |
Collapse
|
13
|
Müllner FE, Roska B. Individual thalamic inhibitory interneurons are functionally specialized toward distinct visual features. Neuron 2024; 112:2765-2782.e9. [PMID: 38917805 PMCID: PMC11348917 DOI: 10.1016/j.neuron.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 03/22/2024] [Accepted: 06/03/2024] [Indexed: 06/27/2024]
Abstract
Inhibitory interneurons in the dorsolateral geniculate nucleus (dLGN) are situated at the first central synapse of the image-forming visual pathway, but little is known about their function. Given their anatomy, they are expected to be multiplexors, integrating many different retinal channels along their dendrites. Here, using targeted single-cell-initiated rabies tracing, we found that mouse dLGN interneurons exhibit a degree of retinal input specialization similar to thalamocortical neurons. Some are anatomically highly specialized, for example, toward motion-selective information. Two-photon calcium imaging performed in vivo revealed that interneurons are also functionally specialized. In mice lacking retinal horizontal direction selectivity, horizontal direction selectivity is reduced in interneurons, suggesting a causal link between input and functional specialization. Functional specialization is not only present at interneuron somata but also extends into their dendrites. Altogether, inhibitory interneurons globally display distinct visual features which reflect their retinal input specialization and are ideally suited to perform feature-selective inhibition.
Collapse
Affiliation(s)
- Fiona E Müllner
- Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland; Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland; Friedrich Miescher Institute for Biomedical Research, 4056 Basel, Switzerland
| | - Botond Roska
- Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland; Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland.
| |
Collapse
|
14
|
Dyer B, Yu SO, Brown RL, Lang RA, D'Souza SP. Defining spatial nonuniformities of all ipRGC types using an improved Opn4 cre recombinase mouse line. CELL REPORTS METHODS 2024; 4:100837. [PMID: 39127043 PMCID: PMC11384080 DOI: 10.1016/j.crmeth.2024.100837] [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: 04/25/2024] [Revised: 06/18/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024]
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) play a crucial role in several physiological light responses. In this study, we generate an improved Opn4cre knockin allele (Opn4cre(DSO)), which faithfully reproduces endogenous Opn4 expression and improves compatibility with widely used reporters. We evaluated the efficacy and sensitivity of Opn4cre(DSO) for labeling in retina and brain and provide an in-depth comparison with the extensively utilized Opn4cre(Saha) line. Through this characterization, Opn4cre(DSO) demonstrated higher specificity in labeling ipRGCs with minimal recombination escape. Leveraging a combination of electrophysiological, molecular, and morphological analyses, we confirmed its sensitivity in detecting all ipRGC types (M1-M6) and defined their unique topographical distribution across the retina. In the brain, the Opn4cre(DSO) line labels ipRGC projections with minimal labeling of cell bodies. Overall, the Opn4cre(DSO) mouse line represents an improved tool for studying ipRGC function and distribution, offering a means to selectively target these cells to study light-regulated behaviors and physiology.
Collapse
Affiliation(s)
- Brannen Dyer
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sue O Yu
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA, USA
| | - R Lane Brown
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA, USA
| | - Richard A Lang
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Ophthalmology, University of Cincinnati, Cincinnati, OH, USA
| | - Shane P D'Souza
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
15
|
Contreras E, Liang C, Mahoney HL, Javier JL, Luce ML, Labastida Medina K, Bozza T, Schmidt TM. Flp-recombinase mouse line for genetic manipulation of ipRGCs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592761. [PMID: 38766000 PMCID: PMC11100754 DOI: 10.1101/2024.05.06.592761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Light has myriad impacts on behavior, health, and physiology. These signals originate in the retina and are relayed to the brain by more than 40 types of retinal ganglion cells (RGCs). Despite a growing appreciation for the diversity of RGCs, how these diverse channels of light information are ultimately integrated by the ~50 retinorecipient brain targets to drive these light-evoked effects is a major open question. This gap in understanding primarily stems from a lack of genetic tools that specifically label, manipulate, or ablate specific RGC types. Here, we report the generation and characterization of a new mouse line (Opn4FlpO), in which FlpO is expressed from the Opn4 locus, to manipulate the melanopsin-expressing, intrinsically photosensitive retinal ganglion cells. We find that the Opn4FlpO line, when crossed to multiple reporters, drives expression that is confined to ipRGCs and primarily labels the M1-M3 subtypes. Labeled cells in this mouse line show the expected intrinsic, melanopsin-based light response and morphological features consistent with the M1-M3 subtypes. In alignment with the morphological and physiological findings, we see strong innervation of non-image forming brain targets by ipRGC axons, and weaker innervation of image forming targets in Opn4FlpO mice labeled using AAV-based and FlpO-reporter lines. Consistent with the FlpO insertion disrupting the endogenous Opn4 transcript, we find that Opn4FlpO/FlpO mice show deficits in the pupillary light reflex, demonstrating their utility for behavioral research in future experiments. Overall, the Opn4FlpO mouse line drives Flp-recombinase expression that is confined to ipRGCs and most effectively drives recombination in M1-M3 ipRGCs. This mouse line will be of broad use to those interested in manipulating ipRGCs through a Flp-based recombinase for intersectional studies or in combination with other, non-Opn4 Cre driver lines.
Collapse
Affiliation(s)
- E Contreras
- Department of Neurobiology, Northwestern University, Evanston, IL
- Northwestern University Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, United States
| | - C Liang
- Department of Neurobiology, Northwestern University, Evanston, IL
| | - H L Mahoney
- Department of Neurobiology, Northwestern University, Evanston, IL
| | - J L Javier
- Department of Neurobiology, Northwestern University, Evanston, IL
| | - M L Luce
- Department of Neurobiology, Northwestern University, Evanston, IL
| | | | - T Bozza
- Department of Neurobiology, Northwestern University, Evanston, IL
| | - T M Schmidt
- Department of Neurobiology, Northwestern University, Evanston, IL
- Department of Ophthalmology, Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
16
|
Shang W, Xie S, Feng W, Li Z, Jia J, Cao X, Shen Y, Li J, Shi H, Gu Y, Weng SJ, Lin L, Pan YH, Yuan XB. A non-image-forming visual circuit mediates the innate fear of heights in male mice. Nat Commun 2024; 15:3746. [PMID: 38702319 PMCID: PMC11068790 DOI: 10.1038/s41467-024-48147-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 04/19/2024] [Indexed: 05/06/2024] Open
Abstract
The neural basis of fear of heights remains largely unknown. In this study, we investigated the fear response to heights in male mice and observed characteristic aversive behaviors resembling human height vertigo. We identified visual input as a critical factor in mouse reactions to heights, while peripheral vestibular input was found to be nonessential for fear of heights. Unexpectedly, we found that fear of heights in naïve mice does not rely on image-forming visual processing by the primary visual cortex. Instead, a subset of neurons in the ventral lateral geniculate nucleus (vLGN), which connects to the lateral/ventrolateral periaqueductal gray (l/vlPAG), drives the expression of fear associated with heights. Additionally, we observed that a subcortical visual pathway linking the superior colliculus to the lateral posterior thalamic nucleus inhibits the defensive response to height threats. These findings highlight a rapid fear response to height threats through a subcortical visual and defensive pathway from the vLGN to the l/vlPAG.
Collapse
Affiliation(s)
- Wei Shang
- Key Laboratory of Brain Functional Genomics of Shanghai and Ministry of Education, Institute of Brain Functional Genomics, School of Life Science and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, 200062, China
| | - Shuangyi Xie
- Key Laboratory of Brain Functional Genomics of Shanghai and Ministry of Education, Institute of Brain Functional Genomics, School of Life Science and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, 200062, China
| | - Wenbo Feng
- Key Laboratory of Brain Functional Genomics of Shanghai and Ministry of Education, Institute of Brain Functional Genomics, School of Life Science and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, 200062, China
| | - Zhuangzhuang Li
- Department of Otolaryngology Head & Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Jingyan Jia
- Key Laboratory of Brain Functional Genomics of Shanghai and Ministry of Education, Institute of Brain Functional Genomics, School of Life Science and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, 200062, China
| | - Xiaoxiao Cao
- Key Laboratory of Brain Functional Genomics of Shanghai and Ministry of Education, Institute of Brain Functional Genomics, School of Life Science and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, 200062, China
| | - Yanting Shen
- Key Laboratory of Brain Functional Genomics of Shanghai and Ministry of Education, Institute of Brain Functional Genomics, School of Life Science and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, 200062, China
| | - Jing Li
- Key Laboratory of Brain Functional Genomics of Shanghai and Ministry of Education, Institute of Brain Functional Genomics, School of Life Science and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, 200062, China
| | - Haibo Shi
- Department of Otolaryngology Head & Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Yiran Gu
- Key Laboratory of Brain Functional Genomics of Shanghai and Ministry of Education, Institute of Brain Functional Genomics, School of Life Science and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, 200062, China
| | - Shi-Jun Weng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Longnian Lin
- Key Laboratory of Brain Functional Genomics of Shanghai and Ministry of Education, Institute of Brain Functional Genomics, School of Life Science and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, 200062, China
| | - Yi-Hsuan Pan
- Key Laboratory of Brain Functional Genomics of Shanghai and Ministry of Education, Institute of Brain Functional Genomics, School of Life Science and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, 200062, China.
| | - Xiao-Bing Yuan
- Key Laboratory of Brain Functional Genomics of Shanghai and Ministry of Education, Institute of Brain Functional Genomics, School of Life Science and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, 200062, China.
| |
Collapse
|
17
|
Aranda ML, Bhoi JD, Parra OAP, Lee SK, Yamada T, Yang Y, Schmidt TM. Genetic tuning of intrinsically photosensitive retinal ganglion cell subtype identity to drive visual behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.590656. [PMID: 38712084 PMCID: PMC11071530 DOI: 10.1101/2024.04.25.590656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The melanopsin-expressing, intrinsically photosensitive retinal ganglion cells (ipRGCs) comprise a subset of the ∼40 retinal ganglion cell types in the mouse retina and drive a diverse array of light-evoked behaviors from circadian photoentrainment to pupil constriction to contrast sensitivity for visual perception. Central to the ability of ipRGCs to control this diverse array of behaviors is the distinct complement of morphophysiological features and gene expression patterns found in the M1-M6 ipRGC subtypes. However, the genetic regulatory programs that give rise to subtypes of ipRGCs are unknown. Here, we identify the transcription factor Brn3b (Pou4f2) as a key genetic regulator that shapes the unique functions of ipRGC subtypes and their diverse downstream visual behaviors.
Collapse
|
18
|
Dyer B, Yu SO, Lane Brown R, Lang RA, D’Souza SP. A new Opn4cre recombinase mouse line to target intrinsically photosensitive retinal ganglion cells (ipRGCs). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.589750. [PMID: 38659888 PMCID: PMC11042346 DOI: 10.1101/2024.04.16.589750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) play a crucial role in several physiological light responses. In this study we generate a new Opn4cre knock-in allele (Opn4cre(DSO)), in which cre is placed immediately downstream of the Opn4 start codon. This approach aims to faithfully reproduce endogenous Opn4 expression and improve compatibility with widely used reporters. We evaluated the efficacy and sensitivity of Opn4cre(DSO) for labeling in retina and brain, and provide an in-depth comparison with the extensively utilized Opn4cre(Saha) line. Through this characterization, Opn4cre(DSO) demonstrated higher specificity in labeling ipRGCs, with minimal recombination escape. Leveraging a combination of electrophysiological, molecular, and morphological analyses, we confirmed its sensitivity in detecting all ipRGC types (M1-M6). Using this new tool, we describe the topographical distributions of ipRGC types across the retinal landscape, uncovering distinct ventronasal biases for M5 and M6 types, setting them apart from their M1-M4 counterparts. In the brain, we find vastly different labeling patterns between lines, with Opn4cre(DSO) only labeling ipRGC axonal projections to their targets. The combination of off-target effects of Opn4cre(Saha) across the retina and brain, coupled with diminished efficiencies of both Cre lines when coupled to less sensitive reporters, underscores the need for careful consideration in experimental design and validation with any Opn4cre driver. Overall, the Opn4cre(DSO) mouse line represents an improved tool for studying ipRGC function and distribution, offering a means to selectively target these cells to study light-regulated behaviors and physiology.
Collapse
Affiliation(s)
- Brannen Dyer
- Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, OH
- Science of Light Center, Cincinnati Children’s Hospital Medical Center, OH
- Abrahamson Pediatric Eye Institute, Cincinnati Children’s Hospital Medical Center, OH
| | - Sue O. Yu
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA
| | - R. Lane Brown
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA
| | - Richard A. Lang
- Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, OH
- Science of Light Center, Cincinnati Children’s Hospital Medical Center, OH
- Abrahamson Pediatric Eye Institute, Cincinnati Children’s Hospital Medical Center, OH
- Department of Ophthalmology, University of Cincinnati, OH
| | - Shane P. D’Souza
- Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, OH
- Science of Light Center, Cincinnati Children’s Hospital Medical Center, OH
- Abrahamson Pediatric Eye Institute, Cincinnati Children’s Hospital Medical Center, OH
| |
Collapse
|
19
|
Yuan M, Jin S, Tan G, Song S, Liu Y, Wang H, Shen Y. A Non-canonical Excitatory PV RGC-PV SC Visual Pathway for Mediating the Looming-evoked Innate Defensive Response. Neurosci Bull 2024; 40:310-324. [PMID: 37302108 PMCID: PMC10912393 DOI: 10.1007/s12264-023-01076-z] [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: 02/23/2023] [Accepted: 05/04/2023] [Indexed: 06/13/2023] Open
Abstract
Parvalbumin-positive retinal ganglion cells (PV+ RGCs) are an essential subset of RGCs found in various species. However, their role in transmitting visual information remains unclear. Here, we characterized PV+ RGCs in the retina and explored the functions of the PV+ RGC-mediated visual pathway. By applying multiple viral tracing strategies, we investigated the downstream of PV+ RGCs across the whole brain. Interestingly, we found that the PV+ RGCs provided direct monosynaptic input to PV+ excitatory neurons in the superficial layers of the superior colliculus (SC). Ablation or suppression of SC-projecting PV+ RGCs abolished or severely impaired the flight response to looming visual stimuli in mice without affecting visual acuity. Furthermore, using transcriptome expression profiling of individual cells and immunofluorescence colocalization for RGCs, we found that PV+ RGCs are predominant glutamatergic neurons. Thus, our findings indicate the critical role of PV+ RGCs in an innate defensive response and suggest a non-canonical subcortical visual pathway from excitatory PV+ RGCs to PV+ SC neurons that regulates looming visual stimuli. These results provide a potential target for intervening and treating diseases related to this circuit, such as schizophrenia and autism.
Collapse
Affiliation(s)
- Man Yuan
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Sen Jin
- The Brain Cognition and Brain Disease Institute, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, National Medical Products Administration Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Shenzhen, 518055, China
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan, 430071, China
| | - Gao Tan
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Siyuan Song
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, 77030, USA
| | - Yizong Liu
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Huadong Wang
- The Brain Cognition and Brain Disease Institute, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, National Medical Products Administration Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Shenzhen, 518055, China
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan, 430071, China
| | - Yin Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China.
- Frontier Science Center of Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
20
|
Pan D, Wang Z, Chen Y, Cao J. Melanopsin-mediated optical entrainment regulates circadian rhythms in vertebrates. Commun Biol 2023; 6:1054. [PMID: 37853054 PMCID: PMC10584931 DOI: 10.1038/s42003-023-05432-7] [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] [Accepted: 10/09/2023] [Indexed: 10/20/2023] Open
Abstract
Melanopsin (OPN4) is a light-sensitive protein that plays a vital role in the regulation of circadian rhythms and other nonvisual functions. Current research on OPN4 has focused on mammals; more evidence is needed from non-mammalian vertebrates to fully assess the significance of the non-visual photosensitization of OPN4 for circadian rhythm regulation. There are species differences in the regulatory mechanisms of OPN4 for vertebrate circadian rhythms, which may be due to the differences in the cutting variants, tissue localization, and photosensitive activation pathway of OPN4. We here summarize the distribution of OPN4 in mammals, birds, and teleost fish, and the classical excitation mode for the non-visual photosensitive function of OPN4 in mammals is discussed. In addition, the role of OPN4-expressing cells in regulating circadian rhythm in different vertebrates is highlighted, and the potential rhythmic regulatory effects of various neuropeptides or neurotransmitters expressed in mammalian OPN4-expressing ganglion cells are summarized among them.
Collapse
Affiliation(s)
- Deng Pan
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China
| | - Zixu Wang
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China
| | - Yaoxing Chen
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China
| | - Jing Cao
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China.
| |
Collapse
|
21
|
Huang Y, Liao P, Yu J, Chen S. Light disrupts social memory via a retina-to-supraoptic nucleus circuit. EMBO Rep 2023; 24:e56839. [PMID: 37531065 PMCID: PMC10561173 DOI: 10.15252/embr.202356839] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/03/2023] Open
Abstract
The formation of social memory between individuals of the opposite sex is crucial for expanding mating options or establishing monogamous pair bonding. A specialized neuronal circuit that regulates social memory could enhance an individual's mating opportunities and provide a parallel pathway for computing social behaviors. While the influence of light exposure on various forms of memory, such as fear and object memory, has been studied, its modulation of social recognition memory remains unclear. Here, we demonstrate that acute exposure to light impairs social recognition memory (SRM) in mice. Unlike sound and touch stimuli, light inhibits oxytocin neurons in the supraoptic nucleus (SON) via M1 SON-projecting intrinsically photosensitive retinal ganglion cells (ipRGCs) and GABAergic neurons in the perinuclear zone of the SON (pSON). We further show that optogenetic activation of SON oxytocin neurons using channelrhodopsin is sufficient to enhance SRM performance, even under light conditions. Our findings unveil a dedicated neuronal circuit through which luminance affects SRM, utilizing a non-image-forming visual pathway, distinct from the canonical modulatory role of the oxytocin system.
Collapse
Affiliation(s)
- Yu‐Fan Huang
- Department of Life ScienceNational Taiwan UniversityTaipeiTaiwan
| | - Po‐Yu Liao
- Department of Life ScienceNational Taiwan UniversityTaipeiTaiwan
| | - Jo‐Hsien Yu
- Department of Life ScienceNational Taiwan UniversityTaipeiTaiwan
| | - Shih‐Kuo Chen
- Department of Life ScienceNational Taiwan UniversityTaipeiTaiwan
| |
Collapse
|
22
|
Wang F, Zhong W, Yang Q, Zhao W, Liu X, Rao B, Lin X, Zhang J. Distribution and synaptic organization of substance P-like immunoreactive neurons in the mouse retina. Brain Struct Funct 2023; 228:1703-1724. [PMID: 37481742 DOI: 10.1007/s00429-023-02688-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
Substance P (SP), a neuroprotective peptidergic neurotransmitter, is known to have immunoreactivity (IR) localized to amacrine and/or ganglion cells in a variety of species' retinas, but it has not yet been studied in the mouse retina. Thus, we investigated the distribution and synaptic organization of SP-IR by confocal and electron microscopy immunocytochemistry in the mouse retina. SP-IR was distributed in the inner nuclear layer (INL), inner plexiform layer (IPL), and ganglion cell layer (GCL). Most of the SP-IR somas belonged to amacrine cells (2.5% of all) in the INL and their processes stratified into the S1, S3, and S5 layers of the IPL, with the most intense band in the S5 layer. Some SP-IR somas can also be observed in the GCL, which were identified as displaced amacrine cells (82%, 1269/1550) and ganglion cells (18%, 281/1550) by antibodies against AP2α and RBPMS, respectively. Such SP-IR ganglion cells (1.2% of all RGCs) can be further divided into 3 subgroups expressing SP/α-Synuclein (α-Syn), SP/GAD67, and/or SP/GAD67/α-Syn. Possible physiological and pathological roles of these ganglion cells are discussed. Further, electron microscopy evidence demonstrates that SP-IR amacrine cells receive major inputs from other SP-IR amacrine cell processes (146/242 inputs) and output mostly to SP-negative amacrine cell processes (291/673 outputs), suggesting series inhibition among amacrine cells. These results reveal for the first time an explicit distribution, novel ganglion cell features, and synaptic organization of SP-IR in the mouse retina, which is important for the future use of mouse models to study the roles of SP in healthy and diseased (including Parkinson's disease) retinal states.
Collapse
Affiliation(s)
- Fenglan Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wenhui Zhong
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Qingwen Yang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wenna Zhao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaoqing Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Bilin Rao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Xin Lin
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Jun Zhang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
23
|
Berry MH, Leffler J, Allen CN, Sivyer B. Functional subtypes of rodent melanopsin ganglion cells switch roles between night and day illumination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.26.554902. [PMID: 38168436 PMCID: PMC10760181 DOI: 10.1101/2023.08.26.554902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs), contain the photopigment melanopsin, and influence both image and non-image forming behaviors. Despite being categorized into multiple types (M1-M6), physiological variability within these types suggests our current understanding of ipRGCs is incomplete. We used multi-electrode array (MEA) recordings and unbiased cluster analysis under synaptic blockade to identify 8 functional clusters of ipRGCs, each with distinct photosensitivity and response timing. We used Cre mice to drive the expression of channelrhodopsin in SON-ipRGCs, enabling the localization of distinct ipRGCs in the dorsal retina. Additionally, we conducted a retrospective unbiased cluster analysis of ipRGC photoresponses to light stimuli across scotopic, mesopic, and photopic intensities, aimed at activating both rod and cone inputs to ipRGCs. Our results revealed shared and distinct synaptic inputs to the identified functional clusters, demonstrating that ipRGCs encode visual information with high fidelity at low light intensities, but poorly at photopic light intensities, when melanopsin activation is highest. Collectively, our findings support a framework with at least 8 functional subtypes of ipRGCs, each encoding luminance with distinct spike outputs, highlighting the inherent functional diversity and complexity of ipRGCs and suggesting a reevaluation of their contributions to retinal function and visual perception under varying light conditions.
Collapse
Affiliation(s)
- Michael H. Berry
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239
- Medical Scientist Training program, Oregon Health & Science University, Portland, OR, 97239
| | - Joseph Leffler
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239
| | - Charles N. Allen
- Oregon Institute of Occupational Health Sciences, Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239
| | - Benjamin Sivyer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239
| |
Collapse
|
24
|
D'Souza SP, Upton BA, Eldred KC, Glass I, Grover K, Ahmed A, Ngyuen MT, Gamlin P, Lang RA. Developmental adaptation of rod photoreceptor number via photoreception in melanopsin (OPN4) retinal ganglion cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554675. [PMID: 37662196 PMCID: PMC10473760 DOI: 10.1101/2023.08.24.554675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Photoreception, a form of sensory experience, is essential for normal development of the mammalian visual system. Detecting photons during development is a prerequisite for visual system function - from vision's first synapse at the cone pedicle and maturation of retinal vascular networks, to transcriptional establishment and maturation of cell types within the visual cortex. Consistent with this theme, we find that the lighting environment regulates developmental rod photoreceptor apoptosis via OPN4-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs). Using a combination of genetics, sensory environment manipulations, and computational approaches, we establish a molecular pathway in which light-dependent glutamate release from ipRGCs is detected via a transiently expressed kainate receptor (GRIK3) in immature rods localized to the inner retina. Communication between ipRGCs and nascent inner retinal rods appears to be mediated by unusual hybrid neurites projecting from ipRGCs that sense light before eye-opening. These structures, previously referred to as outer retinal dendrites (ORDs), span the ipRGC-immature rod distance over the first postnatal week and contain the machinery for sensory detection (melanopsin, OPN4) and axonal/anterograde neurotransmitter release (Synaptophysin, and VGLUT2). Histological and computational assessment of human mid-gestation development reveal conservation of several hallmarks of an ipRGC-to-immature rod pathway, including displaced immature rods, transient GRIK3 expression in the rod lineage, and the presence of ipRGCs with putative neurites projecting deep into the developing retina. Thus, this analysis defines a retinal retrograde signaling pathway that links the sensory environment to immature rods via ipRGC photoreceptors, allowing the visual system to adapt to distinct lighting environments priory to eye-opening.
Collapse
|
25
|
Calligaro H, Shoghi A, Chen X, Kim KY, Yu HL, Khov B, Finander B, Le H, Ellisman MH, Panda S. Ultrastructure of Synaptic Connectivity within Subregions of the Suprachiasmatic Nucleus Revealed by a Genetically Encoded Tag and Serial Blockface Electron Microscopy. eNeuro 2023; 10:ENEURO.0227-23.2023. [PMID: 37500494 PMCID: PMC10449486 DOI: 10.1523/eneuro.0227-23.2023] [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: 06/29/2023] [Accepted: 07/01/2023] [Indexed: 07/29/2023] Open
Abstract
The hypothalamic suprachiasmatic nucleus (SCN) is the central circadian pacemaker in vertebrates. The SCN receives photic information exclusively through melanopsin-expressing retinal ganglion cells (mRGCs) to synchronize circadian rhythms with the environmental light cycles. The SCN is composed of two major peptidergic neuron types in the core and shell regions of the SCN. Determining how mRGCs interact with the network of synaptic connections onto and between SCN neurons is key to understand how light regulates the circadian clock and to elucidate the relevant local circuits within the SCN. To map these connections, we used a newly developed Cre-dependent electron microscopy (EM) reporter, APEX2, to label the mitochondria of mRGC axons. Serial blockface scanning electron microscopy was then used to resolve the fine 3D structure of mRGC axons and synaptic boutons in the SCN of a male mouse. The resulting maps reveal patterns of connectomic organization in the core and shell of the SCN. We show that these regions are composed of different neuronal subtypes and differ with regard to the pattern of mRGC input, as the shell receives denser mRGC synaptic input compared with the core. This finding challenges the present view that photic information coming directly from the retina is received primarily by the core region of the SCN.
Collapse
Affiliation(s)
- Hugo Calligaro
- Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Azarin Shoghi
- Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Xinyue Chen
- Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Keun-Young Kim
- Department of Neurosciences, University of California at San Diego School of Medicine, La Jolla, CA 92161
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92161
| | - Hsin Liu Yu
- Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Brian Khov
- Salk Institute for Biological Studies, La Jolla, CA 92037
| | | | - Hiep Le
- Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Mark H. Ellisman
- Department of Neurosciences, University of California at San Diego School of Medicine, La Jolla, CA 92161
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92161
| | | |
Collapse
|
26
|
Tu HQ, Li S, Xu YL, Zhang YC, Li PY, Liang LY, Song GP, Jian XX, Wu M, Song ZQ, Li TT, Hu HB, Yuan JF, Shen XL, Li JN, Han QY, Wang K, Zhang T, Zhou T, Li AL, Zhang XM, Li HY. Rhythmic cilia changes support SCN neuron coherence in circadian clock. Science 2023; 380:972-979. [PMID: 37262147 DOI: 10.1126/science.abm1962] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 04/13/2023] [Indexed: 06/03/2023]
Abstract
The suprachiasmatic nucleus (SCN) drives circadian clock coherence through intercellular coupling, which is resistant to environmental perturbations. We report that primary cilia are required for intercellular coupling among SCN neurons to maintain the robustness of the internal clock in mice. Cilia in neuromedin S-producing (NMS) neurons exhibit pronounced circadian rhythmicity in abundance and length. Genetic ablation of ciliogenesis in NMS neurons enabled a rapid phase shift of the internal clock under jet-lag conditions. The circadian rhythms of individual neurons in cilia-deficient SCN slices lost their coherence after external perturbations. Rhythmic cilia changes drive oscillations of Sonic Hedgehog (Shh) signaling and clock gene expression. Inactivation of Shh signaling in NMS neurons phenocopied the effects of cilia ablation. Thus, cilia-Shh signaling in the SCN aids intercellular coupling.
Collapse
Affiliation(s)
- Hai-Qing Tu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Sen Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Ling Xu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Cheng Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Pei-Yao Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Li-Yun Liang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Guang-Ping Song
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Xiao-Xiao Jian
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Min Wu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Zeng-Qing Song
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Ting-Ting Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Huai-Bin Hu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Jin-Feng Yuan
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Xiao-Lin Shen
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Jia-Ning Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Qiu-Ying Han
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Kai Wang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Tao Zhang
- Laboratory Animal Center, Academy of Military Medical Sciences, Beijing, China
| | - Tao Zhou
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Ai-Ling Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xue-Min Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hui-Yan Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Ahern J, Chrobok Ł, Champneys AR, Piggins HD. A new phase model of the spatiotemporal relationships between three circadian oscillators in the brainstem. Sci Rep 2023; 13:5480. [PMID: 37016055 PMCID: PMC10073201 DOI: 10.1038/s41598-023-32315-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/25/2023] [Indexed: 04/06/2023] Open
Abstract
Analysis of ex vivo Per2 bioluminescent rhythm previously recorded in the mouse dorsal vagal complex reveals a characteristic phase relationship between three distinct circadian oscillators. These signals represent core clock gene expression in the area postrema (AP), the nucleus of the solitary tract (NTS) and the ependymal cells surrounding the 4th ventricle (4Vep). Initially, the data suggests a consistent phasing in which the AP peaks first, followed shortly by the NTS, with the 4Vep peaking 8-9 h later. Wavelet analysis reveals that this pattern is not consistently maintained throughout a recording, however, the phase dynamics strongly imply that oscillator interactions are present. A simple phase model of the three oscillators is developed and it suggests that realistic phase dynamics occur between three model oscillators with coupling close to a synchronisation transition. The coupling topology suggests that the AP bidirectionally communicates phase information to the NTS and the 4Vep to synchronise the three structures. A comparison of the model with previous experimental manipulations demonstrates its feasibility to explain DVC circadian phasing. Finally, we show that simulating steadily decaying coupling improves the model's ability to capture experimental phase dynamics.
Collapse
Affiliation(s)
- Jake Ahern
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
- Engineering Mathematics, University of Bristol, Bristol, BS8 1TW, UK
| | - Łukasz Chrobok
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Alan R Champneys
- Engineering Mathematics, University of Bristol, Bristol, BS8 1TW, UK
| | - Hugh D Piggins
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
28
|
Carrero L, Antequera D, Alcalde I, Megías D, Figueiro-Silva J, Merayo-Lloves J, Municio C, Carro E. Disturbed circadian rhythm and retinal degeneration in a mouse model of Alzheimer's disease. Acta Neuropathol Commun 2023; 11:55. [PMID: 37004084 PMCID: PMC10067208 DOI: 10.1186/s40478-023-01529-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/11/2023] [Indexed: 04/03/2023] Open
Abstract
The circadian clock is synchronized to the 24 h day by environmental light which is transmitted from the retina to the suprachiasmatic nucleus (SCN) primarily via the retinohypothalamic tract (RHT). Circadian rhythm abnormalities have been reported in neurodegenerative disorders such as Alzheimer's disease (AD). Whether these AD-related changes are a result of the altered clock gene expression, retina degeneration, including the dysfunction in RHT transmission, loss of retinal ganglion cells and its electrophysiological capabilities, or a combination of all of these pathological mechanisms, is not known. Here, we evaluated transgenic APP/PS1 mouse model of AD and wild-type mice at 6- and 12-month-old, as early and late pathological stage, respectively. We noticed the alteration of circadian clock gene expression not only in the hypothalamus but also in two extra-hypothalamic brain regions, cerebral cortex and hippocampus, in APP/PS1 mice. These alterations were observed in 6-month-old transgenic mice and were exacerbated at 12 months of age. This could be explained by the reduced RHT projections in the SCN of APP/PS1 mice, correlating with downregulation of hypothalamic GABAergic response in APP/PS1 mice in advanced stage of pathology. Importantly, we also report retinal degeneration in APP/PS1 mice, including Aβ deposits and reduced choline acetyltransferase levels, loss of melanopsin retinal ganglion cells and functional integrity mainly of inner retina layers. Our findings support the theory that retinal degeneration constitutes an early pathological event that directly affects the control of circadian rhythm in AD.
Collapse
Affiliation(s)
- Laura Carrero
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041, Madrid, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
- Autonoma de Madrid University, Madrid, Spain
| | - Desireé Antequera
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Ignacio Alcalde
- Instituto Universitario Fernández-Vega, Universidad de Oviedo and Fundación de Investigación Oftalmológica, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Diego Megías
- Advanced Optical Microscopy Unit, Unidades Centrales Científico-Técnicas, Instituto de Salud Carlos III, Madrid, Spain
| | - Joana Figueiro-Silva
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Science, University of Zurich, Zurich, Switzerland
| | - Jesús Merayo-Lloves
- Instituto Universitario Fernández-Vega, Universidad de Oviedo and Fundación de Investigación Oftalmológica, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Cristina Municio
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041, Madrid, Spain.
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain.
| | - Eva Carro
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain.
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
29
|
Starnes AN, Jones JR. Inputs and Outputs of the Mammalian Circadian Clock. BIOLOGY 2023; 12:508. [PMID: 37106709 PMCID: PMC10136320 DOI: 10.3390/biology12040508] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023]
Abstract
Circadian rhythms in mammals are coordinated by the central circadian pacemaker, the suprachiasmatic nucleus (SCN). Light and other environmental inputs change the timing of the SCN neural network oscillator, which, in turn, sends output signals that entrain daily behavioral and physiological rhythms. While much is known about the molecular, neuronal, and network properties of the SCN itself, the circuits linking the outside world to the SCN and the SCN to rhythmic outputs are understudied. In this article, we review our current understanding of the synaptic and non-synaptic inputs onto and outputs from the SCN. We propose that a more complete description of SCN connectivity is needed to better explain how rhythms in nearly all behaviors and physiological processes are generated and to determine how, mechanistically, these rhythms are disrupted by disease or lifestyle.
Collapse
Affiliation(s)
| | - Jeff R. Jones
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
30
|
Campbell I, Sharifpour R, Vandewalle G. Light as a Modulator of Non-Image-Forming Brain Functions—Positive and Negative Impacts of Increasing Light Availability. Clocks Sleep 2023; 5:116-140. [PMID: 36975552 PMCID: PMC10047820 DOI: 10.3390/clockssleep5010012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/17/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
Light use is rising steeply, mainly because of the advent of light-emitting diode (LED) devices. LEDs are frequently blue-enriched light sources and may have different impacts on the non-image forming (NIF) system, which is maximally sensitive to blue-wavelength light. Most importantly, the timing of LED device use is widespread, leading to novel light exposure patterns on the NIF system. The goal of this narrative review is to discuss the multiple aspects that we think should be accounted for when attempting to predict how this situation will affect the NIF impact of light on brain functions. We first cover both the image-forming and NIF pathways of the brain. We then detail our current understanding of the impact of light on human cognition, sleep, alertness, and mood. Finally, we discuss questions concerning the adoption of LED lighting and screens, which offer new opportunities to improve well-being, but also raise concerns about increasing light exposure, which may be detrimental to health, particularly in the evening.
Collapse
|
31
|
Berry MH, Moldavan M, Garrett T, Meadows M, Cravetchi O, White E, Leffler J, von Gersdorff H, Wright KM, Allen CN, Sivyer B. A melanopsin ganglion cell subtype forms a dorsal retinal mosaic projecting to the supraoptic nucleus. Nat Commun 2023; 14:1492. [PMID: 36932080 PMCID: PMC10023714 DOI: 10.1038/s41467-023-36955-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 02/24/2023] [Indexed: 03/19/2023] Open
Abstract
Visual input to the hypothalamus from intrinsically photosensitive retinal ganglion cells (ipRGCs) influences several functions including circadian entrainment, body temperature, and sleep. ipRGCs also project to nuclei such as the supraoptic nucleus (SON), which is involved in systemic fluid homeostasis, maternal behavior, social behaviors, and appetite. However, little is known about the SON-projecting ipRGCs or their relationship to well-characterized ipRGC subtypes. Using a GlyT2Cre mouse line, we show a subtype of ipRGCs restricted to the dorsal retina that selectively projects to the SON. These ipRGCs tile a dorsal region of the retina, forming a substrate for encoding ground luminance. Optogenetic activation of their axons demonstrates they release the neurotransmitter glutamate in multiple regions, including the suprachiasmatic nucleus (SCN) and SON. Our results challenge the idea that ipRGC dendrites overlap to optimize photon capture and suggests non-image forming vision operates to sample local regions of the visual field to influence diverse behaviors.
Collapse
Affiliation(s)
- Michael H Berry
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
- Medical Scientist Training Program, Oregon Health & Science University, Portland, OR, USA
| | - Michael Moldavan
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Tavita Garrett
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
- Neuroscience Graduate program, Oregon Health & Science University, Portland, OR, USA
| | - Marc Meadows
- Neuroscience Graduate program, Oregon Health & Science University, Portland, OR, USA
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Olga Cravetchi
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Elizabeth White
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Joseph Leffler
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Henrique von Gersdorff
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
| | - Kevin M Wright
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Charles N Allen
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Benjamin Sivyer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA.
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
32
|
Mouland JW, Watson AJ, Martial FP, Lucas RJ, Brown TM. Colour and melanopsin mediated responses in the murine retina. Front Cell Neurosci 2023; 17:1114634. [PMID: 36993934 PMCID: PMC10040579 DOI: 10.3389/fncel.2023.1114634] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/17/2023] [Indexed: 03/15/2023] Open
Abstract
Introduction: Intrinsically photosensitive retinal ganglion cells (ipRGCs) integrate melanopsin and rod/cone-mediated inputs to signal to the brain. Whilst originally identified as a cell type specialised for encoding ambient illumination, several lines of evidence indicate a strong association between colour discrimination and ipRGC-driven responses. Thus, cone-mediated colour opponent responses have been widely found across ipRGC target regions in the mouse brain and influence a key ipRGC-dependent function, circadian photoentrainment. Although ipRGCs exhibiting spectrally opponent responses have also been identified, the prevalence of such properties have not been systematically evaluated across the mouse retina or yet been found in ipRGC subtypes known to influence the circadian system. Indeed, there is still uncertainty around the overall prevalence of cone-dependent colour opponency across the mouse retina, given the strong retinal gradient in S and M-cone opsin (co)-expression and overlapping spectral sensitivities of most mouse opsins.Methods: To address this, we use photoreceptor isolating stimuli in multielectrode recordings from human red cone opsin knock-in mouse (Opn1mwR) retinas to systematically survey cone mediated responses and the occurrence of colour opponency across ganglion cell layer (GCL) neurons and identify ipRGCs based on spectral comparisons and/or the persistence of light responses under synaptic blockade.Results: Despite detecting robust cone-mediated responses across the retina, we find cone opponency is rare, especially outside of the central retina (overall ~3% of GCL neurons). In keeping with previous suggestions we also see some evidence of rod-cone opponency (albeit even more rare under our experimental conditions), but find no evidence for any enrichment of cone (or rod) opponent responses among functionally identified ipRGCs.Conclusion: In summary, these data suggest the widespread appearance of cone-opponency across the mouse early visual system and ipRGC-related responses may be an emergent feature of central visual processing mechanisms.
Collapse
Affiliation(s)
- Joshua W. Mouland
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- *Correspondence: Joshua W. Mouland
| | - Alex J. Watson
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Franck P. Martial
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Robert J. Lucas
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Timothy M. Brown
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
33
|
Lan YQ, Yu MB, Zhan ZY, Huang YR, Zhao LW, Quan YD, Li ZJ, Sun DF, Wu YL, Wu HY, Liu ZT, Wu KL. Use of a tissue clearing technique combined with retrograde trans-synaptic viral tracing to evaluate changes in mouse retinorecipient brain regions following optic nerve crush. Neural Regen Res 2023; 18:913-921. [DOI: 10.4103/1673-5374.353852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
34
|
Tang YL, Liu AL, Lv SS, Zhou ZR, Cao H, Weng SJ, Zhang YQ. Green light analgesia in mice is mediated by visual activation of enkephalinergic neurons in the ventrolateral geniculate nucleus. Sci Transl Med 2022; 14:eabq6474. [PMID: 36475906 DOI: 10.1126/scitranslmed.abq6474] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Green light exposure has been shown to reduce pain in animal models. Here, we report a vision-associated enkephalinergic neural circuit responsible for green light-mediated analgesia. Full-field green light exposure at an intensity of 10 lux produced analgesic effects in healthy mice and in a model of arthrosis. Ablation of cone photoreceptors completely inhibited the analgesic effect, whereas rod ablation only partially reduced pain relief. The analgesic effect was not modulated by the ablation of intrinsically photosensitive retinal ganglion cells (ipRGCs), which are atypical photoreceptors that control various nonvisual effects of light. Inhibition of the retino-ventrolateral geniculate nucleus (vLGN) pathway completely abolished the analgesic effects. Activation of this pathway reduced nociceptive behavioral responses; such activation was blocked by the inhibition of proenkephalin (Penk)-positive neurons in the vLGN (vLGNPenk). Moreover, green light analgesia was prevented by knockdown of Penk in the vLGN or by ablation of vLGNPenk neurons. In addition, activation of the projections from vLGNPenk neurons to the dorsal raphe nucleus (DRN) was sufficient to suppress nociceptive behaviors, whereas its inhibition abolished the green light analgesia. Our findings indicate that cone-dominated retinal inputs mediated green light analgesia through the vLGNPenk-DRN pathway and suggest that this signaling pathway could be exploited for reducing pain.
Collapse
Affiliation(s)
- Yu-Long Tang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Ai-Lin Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Su-Su Lv
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Zi-Rui Zhou
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Hong Cao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Shi-Jun Weng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Yu-Qiu Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
35
|
Reply to Sharifpour et al.: Light response measurement of the human SCN by 7T fMRI. Proc Natl Acad Sci U S A 2022; 119:e2215410119. [PMID: 36445962 PMCID: PMC9894227 DOI: 10.1073/pnas.2215410119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
|
36
|
van Beurden AW, Schoonderwoerd RA, Tersteeg MMH, de Torres Gutiérrez P, Michel S, Blommers R, Rohling JHT, Meijer JH. Single cell model for re-entrainment to a shifted light cycle. FASEB J 2022; 36:e22518. [PMID: 36057093 PMCID: PMC9543151 DOI: 10.1096/fj.202200478r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/25/2022] [Accepted: 08/15/2022] [Indexed: 11/11/2022]
Abstract
Our daily 24-h rhythm is synchronized to the external light-dark cycle resulting from the Earth's daily rotation. In the mammalian brain, the suprachiasmatic nucleus (SCN) serves as the master clock and receives light-mediated input via the retinohypothalamic tract. Abrupt changes in the timing of the light-dark cycle (e.g., due to jet lag) cause a phase shift in the circadian rhythms in the SCN. Here, we investigated the effects of a 6-h delay in the light-dark cycle on PERIOD2::LUCIFERASE expression at the single-cell level in mouse SCN organotypic explants. The ensemble pattern in phase shift response obtained from individual neurons in the anterior and central SCN revealed a bimodal distribution; specifically, neurons in the ventrolateral SCN responded with a rapid phase shift, while neurons in the dorsal SCN generally did not respond to the shift in the light-dark cycle. We also stimulated the hypothalamic tract in acute SCN slices to simulate light-mediated input to the SCN; interestingly, we found similarities between the distribution and fraction of rapid shifting neurons (in response to the delay) and neurons that were excited in response to electrical stimulation. These results suggest that a subpopulation of neurons in the ventral SCN that have an excitatory response to light input, shift their clock more readily than dorsal located neurons, and initiate the SCN's entrainment to the new light-dark cycle. Thus, we propose that light-excited neurons in the anterior and central SCN play an important role in the organism's ability to adjust to changes in the external light-dark cycle.
Collapse
Affiliation(s)
- Anouk W van Beurden
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Robin A Schoonderwoerd
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mayke M H Tersteeg
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Stephan Michel
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ruben Blommers
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jos H T Rohling
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Johanna H Meijer
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
37
|
Wen X, Liao P, Luo Y, Yang L, Yang H, Liu L, Jiang R. Tandem pore domain acid-sensitive K channel 3 (TASK-3) regulates visual sensitivity in healthy and aging retina. SCIENCE ADVANCES 2022; 8:eabn8785. [PMID: 36070380 PMCID: PMC9451158 DOI: 10.1126/sciadv.abn8785] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 07/21/2022] [Indexed: 06/15/2023]
Abstract
Retinal ganglion cells (RGCs) not only collect but also integrate visual signals and send them from the retina to the brain. The mechanisms underlying the RGC integration of synaptic activity within retinal circuits have not been fully explored. Here, we identified a pronounced expression of tandem pore domain acid-sensitive potassium channel 3 (TASK-3), a two-pore domain potassium channel (K2P), in RGCs. By using a specific antagonist and TASK-3 knockout mice, we found that TASK-3 regulates the intrinsic excitability and the light sensitivity of RGCs by sensing neuronal activity-dependent extracellular acidification. In vivo, the blockade or loss of TASK-3 dampened pupillary light reflex, visual acuity, and contrast sensitivity. Furthermore, overexpressing TASK-3 specifically in RGCs using an adeno-associated virus approach restored the visual function of TASK-3 knockout mice and aged mice where the expression and function of TASK-3 were reduced. Thus, our results provide evidence that implicates a critical role of K2P in visual processing in the retina.
Collapse
Affiliation(s)
- Xiangyi Wen
- Department of Ophthalmology, Department of Optometry and Visual Science, Laboratory of Optometry and Vision Sciences, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ping Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuncheng Luo
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Linghui Yang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Huaiyu Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Longqian Liu
- Department of Ophthalmology, Department of Optometry and Visual Science, Laboratory of Optometry and Vision Sciences, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ruotian Jiang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
38
|
Xiong X, Hu T, Yin Z, Zhang Y, Chen F, Lei P. Research advances in the study of sleep disorders, circadian rhythm disturbances and Alzheimer’s disease. Front Aging Neurosci 2022; 14:944283. [PMID: 36062143 PMCID: PMC9428322 DOI: 10.3389/fnagi.2022.944283] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Although there are still no satisfactory answers to the question of why we need to sleep, a better understanding of its function will help to improve societal attitudes toward sleep. Sleep disorders are very common in neurodegenerative diseases and are a key factor in the quality of life of patients and their families. Alzheimer’s disease (AD) is an insidious and irreversible neurodegenerative disease. Along with progressive cognitive impairment, sleep disorders and disturbances in circadian rhythms play a key role in the progression of AD. Sleep and circadian rhythm disturbances are more common in patients with AD than in the general population and can appear early in the course of the disease. Therefore, this review discusses the bidirectional relationships among circadian rhythm disturbances, sleep disorders, and AD. In addition, pharmacological and non-pharmacological treatment options for patients with AD and sleep disorders are outlined.
Collapse
Affiliation(s)
- Xiangyang Xiong
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Tianpeng Hu
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhenyu Yin
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yaodan Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
| | | | - Ping Lei
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Ping Lei,
| |
Collapse
|
39
|
Schoonderwoerd RA, de Torres Gutiérrez P, Blommers R, van Beurden AW, Coenen TCJJ, Klett NJ, Michel SH, Meijer JH. Inhibitory responses to retinohypothalamic tract stimulation in the circadian clock of the diurnal rodent Rhabdomys pumilio. FASEB J 2022; 36:e22415. [PMID: 35867045 PMCID: PMC9544711 DOI: 10.1096/fj.202200477r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/24/2022] [Accepted: 06/06/2022] [Indexed: 11/17/2022]
Abstract
In both diurnal and nocturnal mammals, the timing of activity is regulated by the central circadian clock of the suprachiasmatic nucleus (SCN). The SCN is synchronized to the external light cycle via the retinohypothalamic tract (RHT). To investigate potential differences in light processing between nocturnal mice and the diurnal rodent Rhabdomys pumilio, we mimicked retinal input by stimulation of the RHT ex vivo. Using Ca2+ imaging, we observed excitations as well as inhibitions of SCN neurons in response to electrical RHT stimulation. In mice, the vast majority of responses were excitatory (85%), whereas in Rhabdomys, the proportion of excitatory and inhibitory responses was similar (51% excitatory, 49% inhibitory). Glutamate blockers AP5 and CNQX blocked the excitatory responses to RHT stimulation but did not abolish the inhibitory responses in mice or Rhabdomys, indicating that the inhibitions were monosynaptically transmitted via the RHT. Simultaneous application of glutamate blockers with the GABAA antagonist gabazine blocked all inhibitory responses in mice, but not in Rhabdomys. Collectively, our results indicate that in Rhabdomys, considerably more inhibitory responses to light are present and that these responses are driven directly by the RHT. We propose that this increased proportion of inhibitory input could reflect a difference in the entrainment mechanism employed by diurnal rodents.
Collapse
Affiliation(s)
- Robin A Schoonderwoerd
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Ruben Blommers
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anouk W van Beurden
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tineke C J J Coenen
- Central Animal Facility, Leiden University Medical Center, Leiden, The Netherlands
| | - Nathan J Klett
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Stephan H Michel
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Johanna H Meijer
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
40
|
Xu J, Jo A, DeVries RP, Deniz S, Cherian S, Sunmola I, Song X, Marshall JJ, Gruner KA, Daigle TL, Contractor A, Lerner TN, Zeng H, Zhu Y. Intersectional mapping of multi-transmitter neurons and other cell types in the brain. Cell Rep 2022; 40:111036. [PMID: 35793636 PMCID: PMC9290751 DOI: 10.1016/j.celrep.2022.111036] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 04/04/2022] [Accepted: 06/13/2022] [Indexed: 01/04/2023] Open
Abstract
Recent developments in intersectional strategies have greatly advanced our ability to precisely target brain cell types based on unique co-expression patterns. To accelerate the application of intersectional genetics, we perform a brain-wide characterization of 13 Flp and tTA mouse driver lines and selected seven for further analysis based on expression of vesicular neurotransmitter transporters. Using selective Cre driver lines, we created more than 10 Cre/tTA combinational lines for cell type targeting and circuit analysis. We then used VGLUT-Cre/VGAT-Flp combinational lines to identify and map 30 brain regions containing neurons that co-express vesicular glutamate and gamma-aminobutyric acid (GABA) transporters, followed by tracing their projections with intersectional viral vectors. Focusing on the lateral habenula (LHb) as a target, we identified glutamatergic, GABAergic, or co-glutamatergic/GABAergic innervations from ∼40 brain regions. These data provide an important resource for the future application of intersectional strategies and expand our understanding of the neuronal subtypes in the brain.
Collapse
Affiliation(s)
- Jian Xu
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Andrew Jo
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Raina P DeVries
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL 60637, USA
| | - Sercan Deniz
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Suraj Cherian
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Idris Sunmola
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Xingqi Song
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - John J Marshall
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Katherine A Gruner
- Mouse Histology and Phenotyping Laboratory, Northwestern University, Chicago, IL 60611, USA
| | - Tanya L Daigle
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Anis Contractor
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Talia N Lerner
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Yongling Zhu
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
41
|
Lee C, Liang F, Lee I, Lu T, Shan Y, Jeng C, Zou Y, Yu H, Chen (Alen) S. External light‐dark cycle shapes gut microbiota through intrinsically photosensitive retinal ganglion cells. EMBO Rep 2022; 23:e52316. [PMID: 35476894 PMCID: PMC9171413 DOI: 10.15252/embr.202052316] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 12/24/2021] [Accepted: 03/18/2022] [Indexed: 01/15/2023] Open
Abstract
Gut microbiota are involved in many physiological functions such as metabolism, brain development, and neurodegenerative diseases. Many microbes in the digestive tract do not maintain a constant level of their relative abundance but show daily oscillations under normal conditions. Recent evidence indicates that chronic jetlag, constant darkness, or deletion of the circadian core gene can alter the composition of gut microbiota and dampen the daily oscillation of gut microbes. However, the neuronal circuit responsible for modulating gut microbiota remained unclear. Using genetic mouse models and 16s rRNA metagenomic analysis, we find that light-dark cycle information transmitted by the intrinsically photosensitive retinal ganglion cells (ipRGCs) is essential for daily oscillations of gut microbes under temporal restricted high-fat diet conditions. Furthermore, aberrant light exposure such as dim light at night (dLAN) can alter the composition, relative abundance, and daily oscillations of gut microbiota. Together, our results indicate that external light-dark cycle information can modulate gut microbiota in the direction from the brain to the gut via the sensory system.
Collapse
Affiliation(s)
- Chi‐Chan Lee
- Department of Life Science National Taiwan University Taipei Taiwan
| | - Feng Liang
- Department of Life Science National Taiwan University Taipei Taiwan
| | - I‐Chi Lee
- Department of Life Science National Taiwan University Taipei Taiwan
| | - Tsung‐Hao Lu
- Department of Life Science National Taiwan University Taipei Taiwan
| | - Yu‐Yau Shan
- Department of Life Science National Taiwan University Taipei Taiwan
| | - Chih‐Fan Jeng
- Department of Life Science National Taiwan University Taipei Taiwan
| | - Yan‐Fang Zou
- Department of Life Science National Taiwan University Taipei Taiwan
| | - Hon‐Tsen Yu
- Department of Life Science National Taiwan University Taipei Taiwan
- Genome and Systems Biology Degree Program National Taiwan University and Academia Sinica Taipei Taiwan
| | - Shih‐Kuo Chen (Alen)
- Department of Life Science National Taiwan University Taipei Taiwan
- Genome and Systems Biology Degree Program National Taiwan University and Academia Sinica Taipei Taiwan
| |
Collapse
|
42
|
Abstract
Retinal circuits transform the pixel representation of photoreceptors into the feature representations of ganglion cells, whose axons transmit these representations to the brain. Functional, morphological, and transcriptomic surveys have identified more than 40 retinal ganglion cell (RGC) types in mice. RGCs extract features of varying complexity; some simply signal local differences in brightness (i.e., luminance contrast), whereas others detect specific motion trajectories. To understand the retina, we need to know how retinal circuits give rise to the diverse RGC feature representations. A catalog of the RGC feature set, in turn, is fundamental to understanding visual processing in the brain. Anterograde tracing indicates that RGCs innervate more than 50 areas in the mouse brain. Current maps connecting RGC types to brain areas are rudimentary, as is our understanding of how retinal signals are transformed downstream to guide behavior. In this article, I review the feature selectivities of mouse RGCs, how they arise, and how they are utilized downstream. Not only is knowledge of the behavioral purpose of RGC signals critical for understanding the retinal contributions to vision; it can also guide us to the most relevant areas of visual feature space. Expected final online publication date for the Annual Review of Vision Science, Volume 8 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Daniel Kerschensteiner
- John F. Hardesty, MD, Department of Ophthalmology and Visual Sciences; Department of Neuroscience; Department of Biomedical Engineering; and Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, Missouri, USA;
| |
Collapse
|
43
|
Kim S, Nam Y, Kim HS, Jung H, Jeon SG, Hong SB, Moon M. Alteration of Neural Pathways and Its Implications in Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10040845. [PMID: 35453595 PMCID: PMC9025507 DOI: 10.3390/biomedicines10040845] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 02/01/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease accompanied by cognitive and behavioral symptoms. These AD-related manifestations result from the alteration of neural circuitry by aggregated forms of amyloid-β (Aβ) and hyperphosphorylated tau, which are neurotoxic. From a neuroscience perspective, identifying neural circuits that integrate various inputs and outputs to determine behaviors can provide insight into the principles of behavior. Therefore, it is crucial to understand the alterations in the neural circuits associated with AD-related behavioral and psychological symptoms. Interestingly, it is well known that the alteration of neural circuitry is prominent in the brains of patients with AD. Here, we selected specific regions in the AD brain that are associated with AD-related behavioral and psychological symptoms, and reviewed studies of healthy and altered efferent pathways to the target regions. Moreover, we propose that specific neural circuits that are altered in the AD brain can be potential targets for AD treatment. Furthermore, we provide therapeutic implications for targeting neuronal circuits through various therapeutic approaches and the appropriate timing of treatment for AD.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Hyeon soo Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Haram Jung
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Seong Gak Jeon
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Sang Bum Hong
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea
- Correspondence:
| |
Collapse
|
44
|
Abstract
SignificanceThe function of our biological clock is dependent on environmental light. Rodent studies have shown that there are multiple colors that affect the clock, but indirect measures in humans suggest blue light is key. We performed functional MRI studies in human subjects with unprecedented spatial resolution to investigate color sensitivity of our clock. Here, we show that narrowband blue, green, and orange light were all effective in changing neuronal activity of the clock. While the clock of nocturnal rodents is excited by light, the human clock responds with a decrease in neuronal activity as indicated by a negative BOLD response. The sensitivity of the clock to multiple colors should be integrated in light therapy aimed to strengthen our 24-h rhythms.
Collapse
|
45
|
Abed S, Reilly A, Arnold SJ, Feldheim DA. Adult Expression of Tbr2 Is Required for the Maintenance but Not Survival of Intrinsically Photosensitive Retinal Ganglion Cells. Front Cell Neurosci 2022; 16:826590. [PMID: 35401124 PMCID: PMC8983909 DOI: 10.3389/fncel.2022.826590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/18/2022] [Indexed: 12/14/2022] Open
Abstract
Retinal ganglion cells expressing the photopigment melanopsin are intrinsically photosensitive (ipRGCs). ipRGCs regulate subconscious non-image-forming behaviors such as circadian rhythms, pupil dilation, and light-mediated mood. Previously, we and others showed that the transcription factor Tbr2 (EOMES) is required during retinal development for the formation of ipRGCs. Tbr2 is also expressed in the adult retina leading to the hypothesis that it plays a role in adult ipRGC function. To test this, we removed Tbr2 in adult mice. We found that this results in the loss of melanopsin expression in ipRGCs but does not lead to cell death or morphological changes to their dendritic or axonal termination patterns. Additionally, we found ectopic expression of Tbr2 in conventional RGCs does not induce melanopsin expression but can increase melanopsin expression in existing ipRGCs. An interesting feature of ipRGCs is their superior survival relative to conventional RGCs after an optic nerve injury. We find that loss of Tbr2 decreases the survival rate of ipRGCs after optic nerve damage suggesting that Tbr2 plays a role in ipRGC survival after injury. Lastly, we show that the GABAergic amacrine cell marker Meis2, is expressed in the majority of Tbr2-expressing displaced amacrine cells as well as in a subset of Tbr2-expressing RGCs. These findings demonstrate that Tbr2 is necessary but not sufficient for melanopsin expression, that Tbr2 is involved in ipRGC survival after optic nerve injury, and identify a marker for Tbr2-expressing displaced amacrine cells.
Collapse
Affiliation(s)
- Sadaf Abed
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Andreea Reilly
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Sebastian J. Arnold
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - David A. Feldheim
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
- *Correspondence: David A. Feldheim,
| |
Collapse
|
46
|
Klett NJ, Cravetchi O, Allen CN. Long-Term Imaging Reveals a Circadian Rhythm of Intracellular Chloride in Neurons of the Suprachiasmatic Nucleus. J Biol Rhythms 2022; 37:110-123. [PMID: 34994231 PMCID: PMC9203244 DOI: 10.1177/07487304211059770] [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] [Indexed: 11/17/2022]
Abstract
Both inhibitory and excitatory GABA transmission exist in the mature suprachiasmatic nucleus (SCN), the master pacemaker of circadian physiology. Whether GABA is inhibitory or excitatory depends on the intracellular chloride concentration ([Cl-]i). Here, using the genetically encoded ratiometric probe Cl-Sensor, we investigated [Cl-]i in AVP and VIP-expressing SCN neurons for several days in culture. The chloride ratio (RCl) demonstrated circadian rhythmicity in AVP + neurons and VIP + neurons, but was not detected in GFAP + astrocytes. RCl peaked between ZT 7 and ZT 8 in both AVP + and VIP + neurons. RCl rhythmicity was not dependent on the activity of several transmembrane chloride carriers, action potential generation, or the L-type voltage-gated calcium channels, but was sensitive to GABA antagonists. We conclude that [Cl-]i is under circadian regulation in both AVP + and VIP + neurons.
Collapse
Affiliation(s)
- Nathan J. Klett
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR 97239
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239
| | - Olga Cravetchi
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239
| | - Charles N. Allen
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
| |
Collapse
|
47
|
Beyond irradiance: Visual signals influencing mammalian circadian function. PROGRESS IN BRAIN RESEARCH 2022; 273:145-169. [DOI: 10.1016/bs.pbr.2022.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
48
|
Guido ME, Marchese NA, Rios MN, Morera LP, Diaz NM, Garbarino-Pico E, Contin MA. Non-visual Opsins and Novel Photo-Detectors in the Vertebrate Inner Retina Mediate Light Responses Within the Blue Spectrum Region. Cell Mol Neurobiol 2022; 42:59-83. [PMID: 33231827 PMCID: PMC11441211 DOI: 10.1007/s10571-020-00997-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023]
Abstract
In recent decades, a number of novel non-visual opsin photopigments belonging to the family of G protein- coupled receptors, likely involved in a number of non-image-forming processes, have been identified and characterized in cells of the inner retina of vertebrates. It is now known that the vertebrate retina is composed of visual photoreceptor cones and rods responsible for diurnal/color and nocturnal/black and white vision, and cells like the intrinsically photosensitive retinal ganglion cells (ipRGCs) and photosensitive horizontal cells in the inner retina, both detecting blue light and expressing the photopigment melanopsin (Opn4). Remarkably, these non-visual photopigments can continue to operate even in the absence of vision under retinal degeneration. Moreover, inner retinal neurons and Müller glial cells have been shown to express other photopigments such as the photoisomerase retinal G protein-coupled receptor (RGR), encephalopsin (Opn3), and neuropsin (Opn5), all able to detect blue/violet light and implicated in chromophore recycling, retinal clock synchronization, neuron-to-glia communication, and other activities. The discovery of these new photopigments in the inner retina of vertebrates is strong evidence of novel light-regulated activities. This review focuses on the features, localization, photocascade, and putative functions of these novel non-visual opsins in an attempt to shed light on their role in the inner retina of vertebrates and in the physiology of the whole organism.
Collapse
Affiliation(s)
- Mario E Guido
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina.
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina.
| | - Natalia A Marchese
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
| | - Maximiliano N Rios
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
| | - Luis P Morera
- Instituto de Organizaciones Saludables, Universidad Siglo 21, Córdoba, Argentina
| | - Nicolás M Diaz
- Department of Ophthalmology, University of Washington School of Medicine, 750 Republican St., Campus, Box 358058, Seattle, WA, 98109, USA
| | - Eduardo Garbarino-Pico
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
| | - María Ana Contin
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
| |
Collapse
|
49
|
Bano-Otalora B, Moye MJ, Brown T, Lucas RJ, Diekman CO, Belle MD. Daily electrical activity in the master circadian clock of a diurnal mammal. eLife 2021; 10:68179. [PMID: 34845984 PMCID: PMC8631794 DOI: 10.7554/elife.68179] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 10/09/2021] [Indexed: 11/13/2022] Open
Abstract
Circadian rhythms in mammals are orchestrated by a central clock within the suprachiasmatic nuclei (SCN). Our understanding of the electrophysiological basis of SCN activity comes overwhelmingly from a small number of nocturnal rodent species, and the extent to which these are retained in day-active animals remains unclear. Here, we recorded the spontaneous and evoked electrical activity of single SCN neurons in the diurnal rodent Rhabdomys pumilio, and developed cutting-edge data assimilation and mathematical modeling approaches to uncover the underlying ionic mechanisms. As in nocturnal rodents, R. pumilio SCN neurons were more excited during daytime hours. By contrast, the evoked activity of R. pumilio neurons included a prominent suppressive response that is not present in the SCN of nocturnal rodents. Our modeling revealed and subsequent experiments confirmed transient subthreshold A-type potassium channels as the primary determinant of this response, and suggest a key role for this ionic mechanism in optimizing SCN function to accommodate R. pumilio's diurnal niche.
Collapse
Affiliation(s)
- Beatriz Bano-Otalora
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom.,Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Matthew J Moye
- Department of Mathematical Sciences, New Jersey Institute of Technology, Newark, United States.,Department of Quantitative Pharmacology and Pharmacometrics (QP2), Kenilworth, United States
| | - Timothy Brown
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom.,Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Robert J Lucas
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom.,Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Casey O Diekman
- Department of Mathematical Sciences, New Jersey Institute of Technology, Newark, United States.,EPSRC Centre for Predictive Modelling in Healthcare, Living Systems Institute, University of Exeter, Exeter, United Kingdom
| | - Mino Dc Belle
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
50
|
Lee S, Chen M, Shi Y, Zhou ZJ. Selective glycinergic input from vGluT3 amacrine cells confers a suppressed-by-contrast trigger feature in a subtype of M1 ipRGCs in the mouse retina. J Physiol 2021; 599:5047-5060. [PMID: 34292589 PMCID: PMC8741526 DOI: 10.1113/jp281717] [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: 03/31/2021] [Accepted: 07/21/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS M1 intrinsically photosensitive retinal ganglion cells (ipRGCs) are known to encode absolute light intensity (irradiance) for non-image-forming visual functions (subconscious vision), such as circadian photoentrainment and the pupillary light reflex. It remains unclear how M1 cells respond to relative light intensity (contrast) and patterned visual signals. The present study identified a special form of contrast sensitivity (suppressed-by-contrast) in M1 cells, suggesting a role of patterned visual signals in regulating non-image-forming vision and a potential role of M1 ipRGCs in encoding image-forming visual cues. The study also uncovered a synaptic mechanism and a retinal circuit mediated by vesicular glutamate transporter 3 (vGluT3) amacrine cells that underlie the suppressed-by-contrast response of M1 cells. M1 ipRGC subtypes (M1a and M1b) were revealed that are distinguishable based on synaptic connectivity with vGluT3 amacrine cells, receptive field properties, intrinsic photo sensitivity and membrane excitability, and morphological features, suggesting a division of visual tasks among discrete M1 subpopulations. ABSTRACT The M1 type ipRGC (intrinsically photosensitive retinal ganglion cell) is known to encode ambient light signals for non-image-forming visual functions such as circadian photo-entrainment and the pupillary light reflex. Here, we report that a subpopulation of M1 cells (M1a) in the mouse retina possess the suppressed-by-contrast (sbc) trigger feature that is a receptive field property previously found only in ganglion cells mediating image-forming vision. Using optogenetics and the dual patch clamp technique, we found that vesicular glutamate transporter 3 (vGluT3) (vGluT3) amacrine cells make glycinergic, but not glutamatergic, synapses specifically onto M1a cells. The spatiotemporal and pharmacological properties of visually evoked responses of M1a cells closely matched the receptive field characteristics of vGluT3 cells, suggesting a major role of the vGluT3 amacrine cell input in shaping the sbc trigger feature of M1a cells. We found that the other subpopulation of M1 cells (M1b), which did not receive a direct vGluT3 cell input, lacked the sbc trigger feature, being distinctively different from M1a cells in intrinsic photo responses, membrane excitability, receptive-field characteristics and morphological features. Together, the results reveal a retinal circuit that uses the sbc trigger feature to regulate irradiance coding and potentially send image-forming cues to non-image-forming visual centres in the brain.
Collapse
Affiliation(s)
- Seunghoon Lee
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, USA
| | - Minggang Chen
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, USA
| | - Yuelin Shi
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, USA
| | - Z Jimmy Zhou
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|