1
|
Shen J, Xue T. Neural-circuit architecture underlying non-image-forming visual functions. Curr Opin Neurobiol 2025; 93:103052. [PMID: 40414167 DOI: 10.1016/j.conb.2025.103052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/27/2025]
Abstract
Perceiving and responding to environmental cues underpins survival and cognition. Light, emerging as one of the most ancient and powerful signals, has shaped life on Earth for billions of years. In mammals, light information is primarily detected by retinal photoreceptors: rods, cones, and intrinsically photosensitive retinal ganglion cells. While rods and cones enable image-forming vision, evolution has preserved and extended evolutionarily ancient yet critical non-image-forming visual functions, including circadian photoentrainment, pupillary light reflexes, and light-mediated modulation of metabolism, mood, and neurodevelopment. Although non-image-forming visual functions have been partially characterized in humans and model organisms, our understanding of the neural circuit mechanisms by which light orchestrates diverse behavior remains fragmented. The discovery of ipRGCs, combined with recent advances in systems neuroscience tools, has yielded critical breakthroughs in three domains: (1) light information encoding within photoreceptors, (2) systematic mapping of retinofugal pathways, and (3) central mechanisms of light-regulated physiological functions. These advances have progressively unraveled causal relationships between non-image-forming visual functions and their underlying eye-brain circuitry. This review summarizes groundbreaking progress in the three domains discussed above, highlighting key unresolved questions in the field.
Collapse
Affiliation(s)
- Jiawei Shen
- Hefei National Research Center for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Tian Xue
- Hefei National Research Center for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; State Key Laboratory of Eye Health, Institute of Advanced Technology, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
2
|
Tanaka R, Portugues R. On analogies in vertebrate and insect visual systems. Nat Rev Neurosci 2025:10.1038/s41583-025-00932-3. [PMID: 40410391 DOI: 10.1038/s41583-025-00932-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2025] [Indexed: 05/25/2025]
Abstract
Despite the large evolutionary distance between vertebrates and insects, the visual systems of these two taxa bear remarkable similarities that have been noted repeatedly, including by pioneering neuroanatomists such as Ramón y Cajal. Fuelled by the advent of transgenic approaches in neuroscience, studies of visual system anatomy and function in both vertebrates and insects have made dramatic progress during the past two decades, revealing even deeper analogies between their visual systems than were noted by earlier observers. Such across-taxa comparisons have tended to focus on either elementary motion detection or relatively peripheral layers of the visual systems. By contrast, the aims of this Review are to expand the scope of this comparison to pathways outside visual motion detection, as well as to deeper visual structures. To achieve these aims, we primarily discuss examples from recent work in larval zebrafish (Danio rerio) and the fruitfly (Drosophila melanogaster), a pair of genetically tractable model organisms with comparatively sized, small brains. In particular, we argue that the brains of both vertebrates and insects are equipped with third-order visual structures that specialize in shared behavioural tasks, including postural and course stabilization, approach and avoidance, and some other behaviours. These wider analogies between the two distant taxa highlight shared behavioural goals and associated evolutionary constraints and suggest that studies on vertebrate and insect vision have a lot to inspire each other.
Collapse
Affiliation(s)
- Ryosuke Tanaka
- Institute of Neuroscience, Technical University of Munich, Munich, Germany.
| | - Ruben Portugues
- Institute of Neuroscience, Technical University of Munich, Munich, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
- Max Planck Fellow Group - Mechanisms of Cognition, MPI Psychiatry, Munich, Germany.
- Bernstein Center for Computational Neuroscience Munich, Munich, Germany.
| |
Collapse
|
3
|
Korkmaz H, Anstötz M, Wellinghof T, Fazari B, Hallenberger A, Bergmann AK, Niggetiedt E, Güven FD, Tundo-Lavalle F, Purath FFA, Bochinsky K, Gremer L, Willbold D, von Gall C, Ali AAH. Loss of Bmal1 impairs the glutamatergic light input to the SCN in mice. Front Cell Neurosci 2025; 19:1538985. [PMID: 40083633 PMCID: PMC11903712 DOI: 10.3389/fncel.2025.1538985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction Glutamate represents the dominant neurotransmitter that conveys the light information to the brain, including the suprachiasmatic nucleus (SCN), the central pacemaker for the circadian system. The neuronal and astrocytic glutamate transporters are crucial for maintaining efficient glutamatergic signaling. In the SCN, glutamatergic nerve terminals from the retina terminate on vasoactive intestinal polypeptide (VIP) neurons, which are essential for circadian functions. To date, little is known about the role of the core circadian clock gene, Bmal1, in glutamatergic neurotransmission of light signal to various brain regions. Methods The aim of this study was to further elucidate the role of Bmal1 in glutamatergic neurotransmission from the retina to the SCN. We therefore examined the spontaneous rhythmic locomotor activity, neuronal and glial glutamate transporters, as well as the ultrastructure of the synapse between the retinal ganglion cells (RGCs) and the SCN in adult male Bmal1-/- mice. Results We found that the deletion of Bmal1 affects the light-mediated behavior in mice, decreases the retinal thickness and affects the vesicular glutamate transporters (vGLUT1, 2) in the retina. Within the SCN, the immunoreaction of vGLUT1, 2, glial glutamate transporters (GLAST) and VIP was decreased while the glutamate concentration was elevated. At the ultrastructure level, the presynaptic terminals were enlarged and the distance between the synaptic vesicles and the synaptic cleft was increased, indicative of a decrease in the readily releasable pool at the excitatory synapses in Bmal1-/-. Conclusion Our data suggests that Bmal1 deletion affects the glutamate transmission in the retina and the SCN and affects the behavioral responses to light.
Collapse
Affiliation(s)
- Hüseyin Korkmaz
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Max Anstötz
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Tim Wellinghof
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Benedetta Fazari
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Angelika Hallenberger
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Ann Kathrin Bergmann
- Core Facility for Electron Microscopy, Faculty of Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Elena Niggetiedt
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Fatma Delâl Güven
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Federica Tundo-Lavalle
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Fathima Faiba A. Purath
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Kevin Bochinsky
- Jülich Research Center, Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Jülich, Germany
| | - Lothar Gremer
- Jülich Research Center, Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Jülich, Germany
- Institute of Physical Biology, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Dieter Willbold
- Jülich Research Center, Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Jülich, Germany
- Institute of Physical Biology, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Charlotte von Gall
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Amira A. H. Ali
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
4
|
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
|
5
|
Herrera E, Chédotal A, Mason C. Development of the Binocular Circuit. Annu Rev Neurosci 2024; 47:303-322. [PMID: 38635868 DOI: 10.1146/annurev-neuro-111020-093230] [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] [Indexed: 04/20/2024]
Abstract
Seeing in three dimensions is a major property of the visual system in mammals. The circuit underlying this property begins in the retina, from which retinal ganglion cells (RGCs) extend to the same or opposite side of the brain. RGC axons decussate to form the optic chiasm, then grow to targets in the thalamus and midbrain, where they synapse with neurons that project to the visual cortex. Here we review the cellular and molecular mechanisms of RGC axonal growth cone guidance across or away from the midline via receptors to cues in the midline environment. We present new views on the specification of ipsi- and contralateral RGC subpopulations and factors implementing their organization in the optic tract and termination in subregions of their targets. Lastly, we describe the functional and behavioral aspects of binocular vision, focusing on the mouse, and discuss recent discoveries in the evolution of the binocular circuit.
Collapse
Affiliation(s)
- Eloísa Herrera
- Instituto de Neurociencias (CSIC-UMH), Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Alicante, Spain;
| | - Alain Chédotal
- Université Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France
- Institut de Pathologie, Groupe Hospitalier Est, Hospices Civils de Lyon, Lyon, France
- Institut de la Vision, INSERM, Sorbonne Université, Paris, France;
| | - Carol Mason
- Departments of Pathology and Cell Biology, Neuroscience, and Ophthalmology, Zuckerman Institute, Columbia University, New York, NY, USA;
| |
Collapse
|
6
|
Maddaloni G, Chang YJ, Senft RA, Dymecki SM. Adaptation to photoperiod via dynamic neurotransmitter segregation. Nature 2024; 632:147-156. [PMID: 39020173 DOI: 10.1038/s41586-024-07692-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/07/2024] [Indexed: 07/19/2024]
Abstract
Changes in the amount of daylight (photoperiod) alter physiology and behaviour1,2. Adaptive responses to seasonal photoperiods are vital to all organisms-dysregulation associates with disease, including affective disorders3 and metabolic syndromes4. The circadian rhythm circuitry is implicated in such responses5,6, yet little is known about the precise cellular substrates that underlie phase synchronization to photoperiod change. Here we identify a brain circuit and system of axon branch-specific and reversible neurotransmitter deployment that are critical for behavioural and sleep adaptation to photoperiod. A type of neuron called mrEn1-Pet17 in the mouse brainstem median raphe nucleus segregates serotonin from VGLUT3 (also known as SLC17A8, a proxy for glutamate) to different axonal branches that innervate specific brain regions involved in circadian rhythm and sleep-wake timing8,9. This branch-specific neurotransmitter deployment did not distinguish between daylight and dark phase; however, it reorganized with change in photoperiod. Axonal boutons, but not cell soma, changed neurochemical phenotype upon a shift away from equinox light/dark conditions, and these changes were reversed upon return to equinox conditions. When we genetically disabled Vglut3 in mrEn1-Pet1 neurons, sleep-wake periods, voluntary activity and clock gene expression did not synchronize to the new photoperiod or were delayed. Combining intersectional rabies virus tracing and projection-specific neuronal silencing, we delineated a preoptic area-to-mrEn1Pet1 connection that was responsible for decoding the photoperiodic inputs, driving the neurotransmitter reorganization and promoting behavioural synchronization. Our results reveal a brain circuit and periodic, branch-specific neurotransmitter deployment that regulates organismal adaptation to photoperiod change.
Collapse
Affiliation(s)
- G Maddaloni
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Y J Chang
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - R A Senft
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - S M Dymecki
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Fitzpatrick MJ, Krizan J, Hsiang JC, Shen N, Kerschensteiner D. A pupillary contrast response in mice and humans: Neural mechanisms and visual functions. Neuron 2024; 112:2404-2422.e9. [PMID: 38697114 PMCID: PMC11257825 DOI: 10.1016/j.neuron.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 12/21/2023] [Accepted: 04/10/2024] [Indexed: 05/04/2024]
Abstract
In the pupillary light response (PLR), increases in ambient light constrict the pupil to dampen increases in retinal illuminance. Here, we report that the pupillary reflex arc implements a second input-output transformation; it senses temporal contrast to enhance spatial contrast in the retinal image and increase visual acuity. The pupillary contrast response (PCoR) is driven by rod photoreceptors via type 6 bipolar cells and M1 ganglion cells. Temporal contrast is transformed into sustained pupil constriction by the M1's conversion of excitatory input into spike output. Computational modeling explains how the PCoR shapes retinal images. Pupil constriction improves acuity in gaze stabilization and predation in mice. Humans exhibit a PCoR with similar tuning properties to mice, which interacts with eye movements to optimize the statistics of the visual input for retinal encoding. Thus, we uncover a conserved component of active vision, its cell-type-specific pathway, computational mechanisms, and optical and behavioral significance.
Collapse
Affiliation(s)
- Michael J Fitzpatrick
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Graduate Program in Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Medical Scientist Training Program, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jenna Krizan
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Graduate Program in Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jen-Chun Hsiang
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Ning Shen
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
8
|
Maddaloni G, Barsotti N, Migliarini S, Giordano M, Nazzi S, Picchi M, Errico F, Usiello A, Pasqualetti M. Impact of Serotonin Deficiency on Circadian Dopaminergic Rhythms. Int J Mol Sci 2024; 25:6475. [PMID: 38928178 PMCID: PMC11203511 DOI: 10.3390/ijms25126475] [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: 05/14/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Physiology and behavior are structured temporally to anticipate daily cycles of light and dark, ensuring fitness and survival. Neuromodulatory systems in the brain-including those involving serotonin and dopamine-exhibit daily oscillations in neural activity and help shape circadian rhythms. Disrupted neuromodulation can cause circadian abnormalities that are thought to underlie several neuropsychiatric disorders, including bipolar mania and schizophrenia, for which a mechanistic understanding is still lacking. Here, we show that genetically depleting serotonin in Tph2 knockout mice promotes manic-like behaviors and disrupts daily oscillations of the dopamine biosynthetic enzyme tyrosine hydroxylase (TH) in midbrain dopaminergic nuclei. Specifically, while TH mRNA and protein levels in the Substantia Nigra (SN) and Ventral Tegmental Area (VTA) of wild-type mice doubled between the light and dark phase, TH levels were high throughout the day in Tph2 knockout mice, suggesting a hyperdopaminergic state. Analysis of TH expression in striatal terminal fields also showed blunted rhythms. Additionally, we found low abundance and blunted rhythmicity of the neuropeptide cholecystokinin (Cck) in the VTA of knockout mice, a neuropeptide whose downregulation has been implicated in manic-like states in both rodents and humans. Altogether, our results point to a previously unappreciated serotonergic control of circadian dopamine signaling and propose serotonergic dysfunction as an upstream mechanism underlying dopaminergic deregulation and ultimately maladaptive behaviors.
Collapse
Affiliation(s)
- Giacomo Maddaloni
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, 56127 Pisa, Italy (M.P.)
- Harvard Medical School, Department of Genetics, Harvard University, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Noemi Barsotti
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, 56127 Pisa, Italy (M.P.)
- Centro per l’Integrazione della Strumentazione Scientifica dell’Università di Pisa (CISUP), 56126 Pisa, Italy
| | - Sara Migliarini
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, 56127 Pisa, Italy (M.P.)
| | - Martina Giordano
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, 56127 Pisa, Italy (M.P.)
| | - Serena Nazzi
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, 56127 Pisa, Italy (M.P.)
| | - Marta Picchi
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, 56127 Pisa, Italy (M.P.)
| | - Francesco Errico
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy
- Department of Agricultural Sciences, University of Naples “Federico II”, 80055 Portici, Italy
| | - Alessandro Usiello
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Caserta, Italy
| | - Massimo Pasqualetti
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, 56127 Pisa, Italy (M.P.)
- Centro per l’Integrazione della Strumentazione Scientifica dell’Università di Pisa (CISUP), 56126 Pisa, Italy
- Center for Neuroscience and Cognitive Systems@UniTn, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy
| |
Collapse
|
9
|
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
|
10
|
Li H, Cui F, Wang T, Wang W, Zhang D. The impact of sunlight exposure on brain structural markers in the UK Biobank. Sci Rep 2024; 14:10313. [PMID: 38705875 PMCID: PMC11070413 DOI: 10.1038/s41598-024-59633-z] [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: 01/13/2024] [Accepted: 04/12/2024] [Indexed: 05/07/2024] Open
Abstract
Sunlight is closely intertwined with daily life. It remains unclear whether there are associations between sunlight exposure and brain structural markers. General linear regression analysis was used to compare the differences in brain structural markers among different sunlight exposure time groups. Stratification analyses were performed based on sex, age, and diseases (hypertension, stroke, diabetes). Restricted cubic spline was performed to examine the dose-response relationship between natural sunlight exposure and brain structural markers, with further stratification by season. A negative association of sunlight exposure time with brain structural markers was found in the upper tertile compared to the lower tertile. Prolonged natural sunlight exposure was associated with the volumes of total brain (β: - 0.051, P < 0.001), white matter (β: - 0.031, P = 0.023), gray matter (β: - 0.067, P < 0.001), and white matter hyperintensities (β: 0.059, P < 0.001). These associations were more pronounced in males and individuals under the age of 60. The results of the restricted cubic spline analysis showed a nonlinear relationship between sunlight exposure and brain structural markers, with the direction changing around 2 h of sunlight exposure. This study demonstrates that prolonged exposure to natural sunlight is associated with brain structural markers change.
Collapse
Affiliation(s)
- Huihui Li
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 308 Ningxia Road, QingdaoShandong Province, 266071, China
| | - Fusheng Cui
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 308 Ningxia Road, QingdaoShandong Province, 266071, China
| | - Tong Wang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 308 Ningxia Road, QingdaoShandong Province, 266071, China
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 308 Ningxia Road, QingdaoShandong Province, 266071, China.
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 308 Ningxia Road, QingdaoShandong Province, 266071, China.
| |
Collapse
|
11
|
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
|
12
|
Mahoney HL, Schmidt TM. The cognitive impact of light: illuminating ipRGC circuit mechanisms. Nat Rev Neurosci 2024; 25:159-175. [PMID: 38279030 DOI: 10.1038/s41583-023-00788-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 01/28/2024]
Abstract
Ever-present in our environments, light entrains circadian rhythms over long timescales, influencing daily activity patterns, health and performance. Increasing evidence indicates that light also acts independently of the circadian system to directly impact physiology and behaviour, including cognition. Exposure to light stimulates brain areas involved in cognition and appears to improve a broad range of cognitive functions. However, the extent of these effects and their mechanisms are unknown. Intrinsically photosensitive retinal ganglion cells (ipRGCs) have emerged as the primary conduit through which light impacts non-image-forming behaviours and are a prime candidate for mediating the direct effects of light on cognition. Here, we review the current state of understanding of these effects in humans and mice, and the tools available to uncover circuit-level and photoreceptor-specific mechanisms. We also address current barriers to progress in this area. Current and future efforts to unravel the circuits through which light influences cognitive functions may inform the tailoring of lighting landscapes to optimize health and cognitive function.
Collapse
Affiliation(s)
- Heather L Mahoney
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
| | - Tiffany M Schmidt
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
13
|
Liao CC, Gabi M, Qi HX, Kaas JH. The postnatal development of retinal projections in strepsirrhine galagos (Otolemur garnettii). J Comp Neurol 2024; 532:e25565. [PMID: 38047381 PMCID: PMC10922899 DOI: 10.1002/cne.25565] [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: 07/13/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 12/05/2023]
Abstract
Here, we describe the postnatal development of retinal projections in galagos. Galagos are of special interest as they represent the understudied strepsirrhine branch (galagos, pottos, lorises, and lemurs) of the primate radiations. The projections of both eyes were revealed in each galago by injecting red or green cholera toxin subunit B (CTB) tracers into different eyes of galagos ranging from postnatal day 5 to adult. In the dorsal lateral geniculate nucleus, the magnocellular, parvocellular, and koniocellular layers were clearly labeled and identified by having inputs from the ipsilateral or contralateral eye at all ages. In the superficial layers of the superior colliculus, the terminations from the ipsilateral eye were just ventral to those from the contralateral eye at all ages. Other terminations at postnatal day 5 and later were in the pregeniculate nucleus, the accessory optic system, and the pretectum. As in other primates, a small retinal projection terminated in the posterior part of the pulvinar, which is known to project to the temporal visual cortex. This small projection from both eyes was most apparent on day 5 and absent in mature galagos. A similar reduction over postnatal maturation has been reported in marmosets, leading to the speculation that early retinal inputs to the pulvinar are responsible for the activation and early maturation of the middle temporal visual area, MT.
Collapse
Affiliation(s)
- Chia-Chi Liao
- Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA
| | - Mariana Gabi
- Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA
| | - Hui-Xin Qi
- Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA
| | - Jon H Kaas
- Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
14
|
Kerschensteiner D, Feller MB. Mapping the Retina onto the Brain. Cold Spring Harb Perspect Biol 2024; 16:a041512. [PMID: 38052498 PMCID: PMC10835620 DOI: 10.1101/cshperspect.a041512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Vision begins in the retina, which extracts salient features from the environment and encodes them in the spike trains of retinal ganglion cells (RGCs), the output neurons of the eye. RGC axons innervate diverse brain areas (>50 in mice) to support perception, guide behavior, and mediate influences of light on physiology and internal states. In recent years, complete lists of RGC types (∼45 in mice) have been compiled, detailed maps of their dendritic connections drawn, and their light responses surveyed at scale. We know less about the RGCs' axonal projection patterns, which map retinal information onto the brain. However, some organizing principles have emerged. Here, we review the strategies and mechanisms that govern developing RGC axons and organize their innervation of retinorecipient brain areas.
Collapse
Affiliation(s)
- Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences
- Department of Neuroscience
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Marla B Feller
- Department of Molecular and Cell Biology
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California 94720, USA
| |
Collapse
|
15
|
Prakash N, Matos HY, Sebaoui S, Tsai L, Tran T, Aromolaran A, Atrachji I, Campbell N, Goodrich M, Hernandez-Pineda D, Jesus Herrero M, Hirata T, Lischinsky J, Martinez W, Torii S, Yamashita S, Hosseini H, Sokolowski K, Esumi S, Kawasawa YI, Hashimoto-Torii K, Jones KS, Corbin JG. Connectivity and molecular profiles of Foxp2- and Dbx1-lineage neurons in the accessory olfactory bulb and medial amygdala. J Comp Neurol 2024; 532:e25545. [PMID: 37849047 PMCID: PMC10922300 DOI: 10.1002/cne.25545] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023]
Abstract
In terrestrial vertebrates, the olfactory system is divided into main (MOS) and accessory (AOS) components that process both volatile and nonvolatile cues to generate appropriate behavioral responses. While much is known regarding the molecular diversity of neurons that comprise the MOS, less is known about the AOS. Here, focusing on the vomeronasal organ (VNO), the accessory olfactory bulb (AOB), and the medial amygdala (MeA), we reveal that populations of neurons in the AOS can be molecularly subdivided based on their ongoing or prior expression of the transcription factors Foxp2 or Dbx1, which delineate separate populations of GABAergic output neurons in the MeA. We show that a majority of AOB neurons that project directly to the MeA are of the Foxp2 lineage. Using single-neuron patch-clamp electrophysiology, we further reveal that in addition to sex-specific differences across lineage, the frequency of excitatory input to MeA Dbx1- and Foxp2-lineage neurons differs between sexes. Together, this work uncovers a novel molecular diversity of AOS neurons, and lineage and sex differences in patterns of connectivity.
Collapse
Affiliation(s)
- Nandkishore Prakash
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Heidi Y Matos
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Sonia Sebaoui
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Luke Tsai
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Tuyen Tran
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Adejimi Aromolaran
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Isabella Atrachji
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Nya Campbell
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Meredith Goodrich
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - David Hernandez-Pineda
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Maria Jesus Herrero
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Tsutomu Hirata
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Julieta Lischinsky
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Wendolin Martinez
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Shisui Torii
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Satoshi Yamashita
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Hassan Hosseini
- Department of Pharmacology, University of Michigan Medical
School, Ann Arbor, MI, USA; Neuroscience Graduate Program, University of Michigan
Medical School, Ann Arbor, MI 48109, USA
| | - Katie Sokolowski
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Shigeyuki Esumi
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Yuka Imamura Kawasawa
- Department of Pharmacology, Pennsylvania State University
College of Medicine, Hershey, PA, USA
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Kevin S Jones
- Department of Pharmacology, University of Michigan Medical
School, Ann Arbor, MI, USA; Neuroscience Graduate Program, University of Michigan
Medical School, Ann Arbor, MI 48109, USA
| | - Joshua G Corbin
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| |
Collapse
|
16
|
Li Z, Peng B, Huang JJ, Zhang Y, Seo MB, Fang Q, Zhang GW, Zhang X, Zhang LI, Tao HW. Enhancement and contextual modulation of visuospatial processing by thalamocollicular projections from ventral lateral geniculate nucleus. Nat Commun 2023; 14:7278. [PMID: 37949869 PMCID: PMC10638288 DOI: 10.1038/s41467-023-43147-9] [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: 03/05/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
In the mammalian visual system, the ventral lateral geniculate nucleus (vLGN) of the thalamus receives salient visual input from the retina and sends prominent GABAergic axons to the superior colliculus (SC). However, whether and how vLGN contributes to fundamental visual information processing remains largely unclear. Here, we report in mice that vLGN facilitates visually-guided approaching behavior mediated by the lateral SC and enhances the sensitivity of visual object detection. This can be attributed to the extremely broad spatial integration of vLGN neurons, as reflected in their much lower preferred spatial frequencies and broader spatial receptive fields than SC neurons. Through GABAergic thalamocollicular projections, vLGN specifically exerts prominent surround suppression of visuospatial processing in SC, leading to a fine tuning of SC preferences to higher spatial frequencies and smaller objects in a context-dependent manner. Thus, as an essential component of the central visual processing pathway, vLGN serves to refine and contextually modulate visuospatial processing in SC-mediated visuomotor behaviors via visually-driven long-range feedforward inhibition.
Collapse
Affiliation(s)
- Zhong Li
- Center for Neural Circuits and Sensory Processing Disorders, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Bo Peng
- Center for Neural Circuits and Sensory Processing Disorders, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Graduate Program in Neuroscience, University of Southern California, Los Angeles, CA, 90033, USA
| | - Junxiang J Huang
- Center for Neural Circuits and Sensory Processing Disorders, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Graduate Program in Biological and Biomedical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Yuan Zhang
- Center for Neural Circuits and Sensory Processing Disorders, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Michelle B Seo
- Center for Neural Circuits and Sensory Processing Disorders, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Graduate Program in Neuroscience, University of Southern California, Los Angeles, CA, 90033, USA
| | - Qi Fang
- Center for Neural Circuits and Sensory Processing Disorders, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Graduate Program in Neuroscience, University of Southern California, Los Angeles, CA, 90033, USA
| | - Guang-Wei Zhang
- Center for Neural Circuits and Sensory Processing Disorders, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Xiaohui Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Li I Zhang
- Center for Neural Circuits and Sensory Processing Disorders, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| | - Huizhong Whit Tao
- Center for Neural Circuits and Sensory Processing Disorders, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
17
|
Kahan A, Mahe K, Dutta S, Kassraian P, Wang A, Gradinaru V. Immediate responses to ambient light in vivo reveal distinct subpopulations of suprachiasmatic VIP neurons. iScience 2023; 26:107865. [PMID: 37766975 PMCID: PMC10520357 DOI: 10.1016/j.isci.2023.107865] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/21/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
The circadian rhythm pacemaker, the suprachiasmatic nucleus (SCN), mediates light entrainment via vasoactive intestinal peptide (VIP) neurons (SCNVIP). Yet, how these neurons uniquely respond and connect to intrinsically photosensitive retinal ganglion cells (ipRGCs) expressing melanopsin (Opn4) has not been determined functionally in freely behaving animals. To address this, we first used monosynaptic tracing from SCNVIP neurons in mice and identified two SCNVIP subpopulations. Second, we recorded calcium changes in response to ambient light, at both bulk and single-cell levels, and found two unique activity patterns in response to high- and low-intensity blue light. The activity patterns of both subpopulations could be manipulated by application of an Opn4 antagonist. These results suggest that the two SCNVIP subpopulations connect to two types of Opn4-expressing ipRGCs, likely M1 and M2, but only one is responsive to red light. These findings have important implications for our basic understanding of non-image-forming circadian light processing.
Collapse
Affiliation(s)
- Anat Kahan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Karan Mahe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Sayan Dutta
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Pegah Kassraian
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Alexander Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
18
|
Maddaloni G, Chang YJ, Senft RA, Dymecki SM. A brain circuit and neuronal mechanism for decoding and adapting to change in daylength. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.11.557218. [PMID: 37745319 PMCID: PMC10515809 DOI: 10.1101/2023.09.11.557218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Changes in daylight amount (photoperiod) drive pronounced alterations in physiology and behaviour1,2. Adaptive responses to seasonal photoperiods are vital to all organisms - dysregulation is associated with disease, from affective disorders3 to metabolic syndromes4. Circadian rhythm circuitry has been implicated5,6 yet little is known about the precise neural and cellular substrates that underlie phase synchronization to photoperiod change. Here we present a previously unknown brain circuit and novel system of axon branch-specific and reversible neurotransmitter deployment that together prove critical for behavioural and sleep adaptation to photoperiod change. We found that the recently defined neuron type called mrEn1-Pet17 located in the mouse brainstem Median Raphe Nucleus (MRN) segregates serotonin versus VGLUT3 (here proxy for the neurotransmitter glutamate) to different axonal branches innervating specific brain regions involved in circadian rhythm and sleep/wake timing8,9. We found that whether measured during the light or dark phase of the day this branch-specific neurotransmitter deployment in mrEn1-Pet1 neurons was indistinguishable; however, it strikingly reorganizes on photoperiod change. Specifically, axonal boutons but not cell soma show a shift in neurochemical phenotype upon change away from equinox light/dark conditions that reverses upon return to equinox. When we genetically disabled the deployment of VGLUT3 in mrEn1-Pet1 neurons, we found that sleep/wake periods and voluntary activity failed to synchronize to the new photoperiod or was significantly delayed. Combining intersectional rabies virus tracing and projection-specific neuronal silencing in vivo, we delineated a Preoptic Area-to-mrEn1Pet1 connection responsible for decoding the photoperiodic inputs, driving the neurochemical shift and promoting behavioural synchronization. Our results reveal a previously unrecognized brain circuit along with a novel form of periodic, branch-specific neurotransmitter deployment that together regulate organismal adaptation to photoperiod changes.
Collapse
Affiliation(s)
- G Maddaloni
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston 02115 MA, USA
| | - Y J Chang
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston 02115 MA, USA
| | - R A Senft
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston 02115 MA, USA
| | - S M Dymecki
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston 02115 MA, USA
| |
Collapse
|
19
|
Makateb A, Rashidinia A, Khosravifard K, Dabaghi P. Investigating the effects of a blue-blocking software on the daily rhythm of sleep, melatonin, cortisol, positive and negative emotions. Chronobiol Int 2023; 40:896-902. [PMID: 37302816 DOI: 10.1080/07420528.2023.2222816] [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: 02/22/2023] [Revised: 05/10/2023] [Accepted: 06/04/2023] [Indexed: 06/13/2023]
Abstract
Since the use of light and electronic devices is inevitable, the use of blue light filters (in various light sources, electronic devices or optical devices including intraocular lenses) has been shown to improve sleep quality, especially in later hours of the day and during night time. In this study, we examine the effect of the blue light on sleep and wakefulness rhythms and positive and negative emotions. This randomized clinical trial was conducted with 80 AJA University of Medical Sciences employees who use computers at least 2 h a day. All subjects were employees of the discharge unit of Imam Reza Hospital, which is located next to AJA University. The subjects were divided into two groups of 40 people, blue light filter software intervention and sham treatment. Pittsburgh Sleep Quality Index (PSQI), Positive and Negative Affect Schedule (PANAS), Visual Function Questionnaire (VFQ), Epworth Sleepiness Scale (ESS) and salivary melatonin and cortisol levels were assessed for both groups before and 3 months after the intervention. Data analysis was performed using IBM SPSS statistics for windows, version 21.0 (Armonk, NY: IBM Corporation). P value ≤ 0.05 was considered as statistically significant. The results showed that the Pittsburgh sleep scale after the intervention was significantly lower in the intervention group than in the control group. After the intervention, the VFQ was significantly lower in the intervention group than in the control group (P = 0.018). There was no significant difference in the Epworth Sleepiness Scale (ESS) between the two study groups after the intervention (P = 0.370). There was no significant difference in Positive and Negative Affect Schedule (PANAS) in the two study groups after the intervention (P = 0.140). After the intervention, cortisol levels were significantly higher in the intervention group than in the control group (P = 0.006). Also, the amount of cortisol increased significantly in the intervention group (P = 0.028). The amount of melatonin decreased significantly in the intervention group (P = 0.034). The sleep quality score after the intervention was significantly lower in the intervention group than in the control group. This indicates better sleep quality in the intervention group. The results also show that the level of visual fatigue in the intervention group decreased significantly. However, no significant change was detected regarding positive and negative emotions. After the intervention, cortisol levels were significantly higher in the intervention group than the control group. In addition, cortisol levels increased significantly and melatonin levels decreased significantly in the intervention group during the course of study.
Collapse
Affiliation(s)
- Ali Makateb
- Opthalmology Department, AJA University of Medical Sciences, Tehran, Iran
| | - Ali Rashidinia
- Opthalmology Department, AJA University of Medical Sciences, Tehran, Iran
| | | | - Parviz Dabaghi
- Department of Clinical Psychology, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Wang G, Liu YF, Yang Z, Yu CX, Tong Q, Tang YL, Shao YQ, Wang LQ, Xu X, Cao H, Zhang YQ, Zhong YM, Weng SJ, Yang XL. Short-term acute bright light exposure induces a prolonged anxiogenic effect in mice via a retinal ipRGC-CeA circuit. SCIENCE ADVANCES 2023; 9:eadf4651. [PMID: 36947616 PMCID: PMC10032603 DOI: 10.1126/sciadv.adf4651] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/17/2023] [Indexed: 06/18/2023]
Abstract
Light modulates mood through various retina-brain pathways. We showed that mice treated with short-term acute bright light exposure displayed anxiety-related phenotypes in a prolonged manner even after the termination of the exposure. Such a postexposure anxiogenic effect depended upon melanopsin-based intrinsically photosensitive retinal ganglion cell (ipRGC) activities rather than rod/cone photoreceptor inputs. Chemogenetic manipulation of specific central nuclei demonstrated that the ipRGC-central amygdala (CeA) visual circuit played a key role in this effect. The corticosterone system was likely to be involved in this effect, as evidenced by enhanced expression of the glucocorticoid receptor (GR) protein in the CeA and the bed nucleus of the stria terminalis and by the absence of this effect in animals treated with the GR antagonist. Together, our findings reveal a non-image forming visual circuit specifically designed for "the delayed" extinction of anxiety against potential threats, thus conferring a survival advantage.
Collapse
Affiliation(s)
- Ge Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yun-Feng Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Zhe Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Chen-Xi Yu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Qiuping Tong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yu-Long Tang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yu-Qi Shao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Li-Qin Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xun Xu
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Hong Cao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yu-Qiu Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yong-Mei Zhong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Shi-Jun Weng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xiong-Li Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
21
|
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
|
22
|
Jeczmien-Lazur JS, Sanetra AM, Pradel K, Izowit G, Chrobok L, Palus-Chramiec K, Piggins HD, Lewandowski MH. Metabolic cues impact non-oscillatory intergeniculate leaflet and ventral lateral geniculate nucleus: standard versus high-fat diet comparative study. J Physiol 2023; 601:979-1016. [PMID: 36661095 DOI: 10.1113/jp283757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/12/2023] [Indexed: 01/21/2023] Open
Abstract
The intergeniculate leaflet and ventral lateral geniculate nucleus (IGL/VLG) are subcortical structures involved in entrainment of the brain's circadian system to photic and non-photic (e.g. metabolic and arousal) cues. Both receive information about environmental light from photoreceptors, exhibit infra-slow oscillations (ISO) in vivo, and connect to the master circadian clock. Although current evidence demonstrates that the IGL/VLG communicate metabolic information and are crucial for entrainment of circadian rhythms to time-restricted feeding, their sensitivity to food intake-related peptides has not been investigated yet. We examined the effect of metabolically relevant peptides on the spontaneous activity of IGL/VLG neurons. Using ex vivo and in vivo electrophysiological recordings as well as in situ hybridisation, we tested potential sensitivity of the IGL/VLG to anorexigenic and orexigenic peptides, such as cholecystokinin, glucagon-like peptide 1, oxyntomodulin, peptide YY, orexin A and ghrelin. We explored neuronal responses to these drugs during day and night, and in standard vs. high-fat diet conditions. We found that IGL/VLG neurons responded to all the substances tested, except peptide YY. Moreover, more neurons responded to anorexigenic drugs at night, while a high-fat diet affected the IGL/VLG sensitivity to orexigenic peptides. Interestingly, ISO neurons responded to light and orexin A, but did not respond to the other food intake-related peptides. In contrast, non-ISO cells were activated by metabolic peptides, with only some being responsive to light. Our results show for the first time that peptides involved in the body's energy homeostasis stimulate the thalamus and suggest functional separation of the IGL/VLG cells. KEY POINTS: The intergeniculate leaflet and ventral lateral geniculate nucleus (IGL/VLG) of the rodent thalamus process various signals and participate in circadian entrainment. In both structures, cells exhibiting infra-slow oscillatory activity as well as non-rhythmically firing neurons being observed. Here, we reveal that only one of these two groups of cells responds to anorexigenic (cholecystokinin, glucagon-like peptide 1 and oxyntomodulin) and orexigenic (ghrelin and orexin A) peptides. Neuronal responses vary depending on the time of day (day vs. night) and on the diet (standard vs. high-fat diet). Additionally, we visualised receptors to the tested peptides in the IGL/VLG using in situ hybridisation. Our results suggest that two electrophysiologically different subpopulations of IGL/VLG neurons are involved in two separate functions: one related to the body's energy homeostasis and one associated with the subcortical visual system.
Collapse
Affiliation(s)
- Jagoda S Jeczmien-Lazur
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Anna M Sanetra
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Kamil Pradel
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Gabriela Izowit
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Lukasz Chrobok
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland.,School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Katarzyna Palus-Chramiec
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Hugh D Piggins
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Marian H Lewandowski
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
23
|
Govindaiah G, Fox MA, Guido W. Pattern of Driver-Like Input onto Neurons of the Mouse Ventral Lateral Geniculate Nucleus. eNeuro 2023; 10:ENEURO.0386-22.2022. [PMID: 36609305 PMCID: PMC9850909 DOI: 10.1523/eneuro.0386-22.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/06/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
The ventral lateral geniculate nucleus (vLGN) is a retinorecipient region of thalamus that contributes to a number of complex visual behaviors. Retinal axons that target vLGN terminate exclusively in the external subdivision (vLGNe), which is also transcriptionally and cytoarchitectonically distinct from the internal subdivision (vLGNi). While recent studies shed light on the cell types and efferent projections of vLGNe and vLGNi, we have a crude understanding of the source and nature of the excitatory inputs driving postsynaptic activity in these regions. Here, we address this by conducting in vitro whole-cell recordings in acutely prepared thalamic slices and using electrical and optical stimulation techniques to examine the postsynaptic excitatory activity evoked by the activation of retinal or cortical layer V input onto neurons in vLGNe and vLGNi. Activation of retinal afferents by electrical stimulation of optic tract or optical stimulation of retinal terminals resulted in robust driver-like excitatory activity in vLGNe. Optical activation of corticothalamic terminals from layer V resulted in similar driver-like activity in both vLGNe and vLGNi. Using a dual-color optogenetic approach, we found that many vLGNe neurons received convergent input from these two sources. Both individual pathways displayed similar driver-like properties, with corticothalamic stimulation leading to a stronger form of synaptic depression than retinogeniculate stimulation. We found no evidence of convergence in vLGNi, with neurons only responding to corticothalamic stimulation. These data provide insight into the influence of excitatory inputs to vLGN and reveal that only neurons in vLGNe receive convergent input from both sources.
Collapse
Affiliation(s)
- Gubbi Govindaiah
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky 40202
| | - Michael A. Fox
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
- School of Neuroscience, Virginia Tech, Blacksburg, Virginia 24061
| | - William Guido
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky 40202
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Brock O, Gelegen C, Sully P, Salgarella I, Jager P, Menage L, Mehta I, Jęczmień-Łazur J, Djama D, Strother L, Coculla A, Vernon AC, Brickley S, Holland P, Cooke SF, Delogu A. A Role for Thalamic Projection GABAergic Neurons in Circadian Responses to Light. J Neurosci 2022; 42:9158-9179. [PMID: 36280260 PMCID: PMC9761691 DOI: 10.1523/jneurosci.0112-21.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/07/2022] Open
Abstract
The thalamus is an important hub for sensory information and participates in sensory perception, regulation of attention, arousal and sleep. These functions are executed primarily by glutamatergic thalamocortical neurons that extend axons to the cortex and initiate cortico-thalamocortical connectional loops. However, the thalamus also contains projection GABAergic neurons that do not extend axons toward the cortex. Here, we have harnessed recent insight into the development of the intergeniculate leaflet (IGL) and the ventral lateral geniculate nucleus (LGv) to specifically target and manipulate thalamic projection GABAergic neurons in female and male mice. Our results show that thalamic GABAergic neurons of the IGL and LGv receive retinal input from diverse classes of retinal ganglion cells (RGCs) but not from the M1 intrinsically photosensitive retinal ganglion cell (ipRGC) type. We describe the synergistic role of the photoreceptor melanopsin and the thalamic neurons of the IGL/LGv in circadian entrainment to dim light. We identify a requirement for the thalamic IGL/LGv neurons in the rapid changes in vigilance states associated with circadian light transitions.SIGNIFICANCE STATEMENT The intergeniculate leaflet (IGL) and ventral lateral geniculate nucleus (LGv) are part of the extended circadian system and mediate some nonimage-forming visual functions. Here, we show that each of these structures has a thalamic (dorsal) as well as prethalamic (ventral) developmental origin. We map the retinal input to thalamus-derived cells in the IGL/LGv complex and discover that while RGC input is dominant, this is not likely to originate from M1ipRGCs. We implicate thalamic cells in the IGL/LGv in vigilance state transitions at circadian light changes and in overt behavioral entrainment to dim light, the latter exacerbated by concomitant loss of melanopsin expression.
Collapse
Affiliation(s)
- Olivier Brock
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
| | - Cigdem Gelegen
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
| | - Peter Sully
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
| | - Irene Salgarella
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
| | - Polona Jager
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
| | - Lucy Menage
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
| | - Ishita Mehta
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
| | - Jagoda Jęczmień-Łazur
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
| | - Deyl Djama
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
- Department of Life Sciences and Centre for Neurotechnology, Imperial College London, London SW7 2AZ, United Kingdom
| | - Lauren Strother
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
| | - Angelica Coculla
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, United Kingdom
| | - Stephen Brickley
- Department of Life Sciences and Centre for Neurotechnology, Imperial College London, London SW7 2AZ, United Kingdom
| | - Philip Holland
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
- Wolfson Centre for Age Related Disease, King's College London, London SE1 1UL, United Kingdom
| | - Samuel F Cooke
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, United Kingdom
| | - Alessio Delogu
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, United Kingdom
| |
Collapse
|
27
|
Endogenous opioid signaling in the retina modulates sleep/wake activity in mice. Neurobiol Sleep Circadian Rhythms 2022; 13:100078. [PMID: 35800978 PMCID: PMC9254600 DOI: 10.1016/j.nbscr.2022.100078] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 12/26/2022] Open
|
28
|
Divergent outer retinal circuits drive image and non-image visual behaviors. Cell Rep 2022; 39:111003. [PMID: 35767957 PMCID: PMC9400924 DOI: 10.1016/j.celrep.2022.111003] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/25/2022] [Accepted: 06/03/2022] [Indexed: 11/22/2022] Open
Abstract
Image- and non-image-forming vision are essential for animal behavior. Here we use genetically modified mouse lines to examine retinal circuits driving image- and non-image-functions. We describe the outer retinal circuits underlying the pupillary light response (PLR) and circadian photoentrainment, two non-image-forming behaviors. Rods and cones signal light increments and decrements through the ON and OFF pathways, respectively. We find that the OFF pathway drives image-forming vision but cannot drive circadian photoentrainment or the PLR. Cone light responses drive image formation but fail to drive the PLR. At photopic levels, rods use the primary and secondary rod pathways to drive the PLR, whereas at the scotopic and mesopic levels, rods use the primary pathway to drive the PLR, and the secondary pathway is insufficient. Circuit dynamics allow rod ON pathways to drive two non-image-forming behaviors across a wide range of light intensities, whereas the OFF pathway is potentially restricted to image formation.
Collapse
|
29
|
Theis J. Differential diagnosis and theories of pathophysiology of post-traumatic photophobia: A review. NeuroRehabilitation 2022; 50:309-319. [DOI: 10.3233/nre-228014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Photophobia is a common sensory symptom after traumatic brain injury (TBI) that may have a grave impact on a patient’s functional independence, neurorehabilitation, and activities of daily living. Post-TBI photophobia can be difficult to treat and the majority of patients can suffer chronically up to and beyond one year after their injury. OBJECTIVES: This review evaluates the current theories of the pathophysiology of photophobia and the most-common co-morbid etiologies of light sensitivity in TBI to help guide the differential diagnosis and individualized management of post-TBI photophobia. METHODS: Primary articles were found via PubMed and Google Scholar search of key terms including “photophobia” “light sensitivity” “photosensitivity” “photo-oculodynia” “intrinsically photosensitive retinal ganglion cells” “ipRGC” and “concussion” “brain injury” “dry eye”. Due to paucity of literature papers were reviewed from 1900 to present in English. RESULTS: Recent advances in understanding the pathophysiology of photophobia in dry eye and migraine and their connection to intrinsically photosensitive retinal ganglion cells (ipRGC) have revealed complex and multifaceted trigeminovascular and trigeminoautonomic pathways underlying photophobia. Patients who suffer a TBI often have co-morbidities like dry eye and migraine that may influence the patient’s photophobia. CONCLUSION: Post-traumatic photophobia is a complex multi-disciplinary complaint that can severely impact a patient’s quality of life. Exploration of underlying etiology may allow for improved treatment and symptomatic relief for these patients beyond tinted lenses alone.
Collapse
Affiliation(s)
- Jacqueline Theis
- Concussion Care Centre of Virginia, Richmond, VA, USA
- Virginia Neuro-Optometry, Richmond, VA, USA Tel.: +1 804 387 2902; Fax: +1 804 509 0543; E-mail:
| |
Collapse
|
30
|
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]
|
31
|
Chrobok L, Belle MDC, Myung J. From Fast Oscillations to Circadian Rhythms: Coupling at Multiscale Frequency Bands in the Rodent Subcortical Visual System. Front Physiol 2021; 12:738229. [PMID: 34899375 PMCID: PMC8662821 DOI: 10.3389/fphys.2021.738229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
The subcortical visual system (SVS) is a unique collection of brain structures localised in the thalamus, hypothalamus and midbrain. The SVS receives ambient light inputs from retinal ganglion cells and integrates this signal with internal homeostatic demands to influence physiology. During this processing, a multitude of oscillatory frequency bands coalesces, with some originating from the retinas, while others are intrinsically generated in the SVS. Collectively, these rhythms are further modulated by the day and night cycle. The multiplexing of these diverse frequency bands (from circadian to infra-slow and gamma oscillations) makes the SVS an interesting system to study coupling at multiscale frequencies. We review the functional organisation of the SVS, and the various frequencies generated and processed by its neurons. We propose a perspective on how these different frequency bands couple with one another to synchronise the activity of the SVS to control physiology and behaviour.
Collapse
Affiliation(s)
- Lukasz Chrobok
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Mino D C Belle
- Institute of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Jihwan Myung
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, Taipei, Taiwan.,Brain and Consciousness Research Centre, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| |
Collapse
|
32
|
Zhao N, Shi J, Xu H, Luo Q, Li Q, Liu M. Baicalin suppresses glaucoma pathogenesis by regulating the PI3K/AKT signaling in vitro and in vivo. Bioengineered 2021; 12:10187-10198. [PMID: 34860641 PMCID: PMC8810108 DOI: 10.1080/21655979.2021.2001217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/28/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Glaucoma, characterized with progressive degeneration of retinal ganglion cells (RGCs), is the second frequently leading cause of sight loss in the word after cataract. Baicalin plays a protective role in age-related macular degeneration, retinopathy of prematurity, branch retinal vein occlusion, and ischemia-induced neurodegeneration in the retina. The present study aimed to investigate the role of baicalin in glaucoma. RGCs were stimulated with N-methyl-D-aspartate (NMDA) to mimic the in vitro model of glaucoma. A mouse model of glaucoma induced by chronic elevated intraocular pressure was also established. The apoptosis, oxidative stress, and autophagy of RGCs were detected by flow cytometry analysis, 2,7-dichlorodihydrofluorescein diacetate staining, and Western blotting, respectively. Retinal pathological changes were exhibited by hemotoxylin and eosin staining. Baicalin restrained the NMDA-induced cell apoptosis, autophagy, and oxidative stress of RGCs by activating the PI3K/AKT signaling in vitro. The elevated intraocular pressure-induced pathological changes in retinas of glaucoma mice were attenuated by baicalin. Moreover, the number of RGCs was significantly decreased in glaucoma mice, and then increased by baicalin treatment. Baicalin also inhibited autophagy and activated PI3K/AKT signaling in vivo. In conclusion, baicalin suppresses glaucoma pathogenesis by regulating the PI3K/AKT signaling in vitro and in vivo.
Collapse
Affiliation(s)
- Ningmin Zhao
- Department of Pharmacy, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Jieran Shi
- Department of Pharmacy, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Haohang Xu
- Department of Pharmacy, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Qing Luo
- Department of Pharmacy, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Qiaoyan Li
- Department of Pharmacy, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Mingzhou Liu
- Department of Pharmacy, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
| |
Collapse
|
33
|
Chrobok L, Pradel K, Janik ME, Sanetra AM, Bubka M, Myung J, Ridla Rahim A, Klich JD, Jeczmien-Lazur JS, Palus-Chramiec K, Lewandowski MH. Intrinsic circadian timekeeping properties of the thalamic lateral geniculate nucleus. J Neurosci Res 2021; 99:3306-3324. [PMID: 34758124 DOI: 10.1002/jnr.24973] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/07/2021] [Accepted: 09/17/2021] [Indexed: 01/08/2023]
Abstract
Circadian rhythmicity in mammals is sustained by the central brain clock-the suprachiasmatic nucleus of the hypothalamus (SCN), entrained to the ambient light-dark conditions through a dense retinal input. However, recent discoveries of autonomous clock gene expression cast doubt on the supremacy of the SCN and suggest circadian timekeeping mechanisms devolve to local brain clocks. Here, we use a combination of molecular, electrophysiological, and optogenetic tools to evaluate intrinsic clock properties of the main retinorecipient thalamic center-the lateral geniculate nucleus (LGN) in male rats and mice. We identify the dorsolateral geniculate nucleus as a slave oscillator, which exhibits core clock gene expression exclusively in vivo. Additionally, we provide compelling evidence for intrinsic clock gene expression accompanied by circadian variation in neuronal activity in the intergeniculate leaflet and ventrolateral geniculate nucleus (VLG). Finally, our optogenetic experiments propose the VLG as a light-entrainable oscillator, whose phase may be advanced by retinal input at the beginning of the projected night. Altogether, this study for the first time demonstrates autonomous timekeeping mechanisms shaping circadian physiology of the LGN.
Collapse
Affiliation(s)
- Lukasz Chrobok
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Kamil Pradel
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Marcelina Elzbieta Janik
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Anna Magdalena Sanetra
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Monika Bubka
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Jihwan Myung
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, Taipei, Taiwan.,Brain and Consciousness Research Centre, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| | - Amalia Ridla Rahim
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, Taipei, Taiwan.,Brain and Consciousness Research Centre, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| | - Jasmin Daniela Klich
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Jagoda Stanislawa Jeczmien-Lazur
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Katarzyna Palus-Chramiec
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Marian Henryk Lewandowski
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
34
|
D'Souza SP, Swygart DI, Wienbar SR, Upton BA, Zhang KX, Mackin RD, Casasent AK, Samuel MA, Schwartz GW, Lang RA. Retinal patterns and the cellular repertoire of neuropsin (Opn5) retinal ganglion cells. J Comp Neurol 2021; 530:1247-1262. [PMID: 34743323 PMCID: PMC8969148 DOI: 10.1002/cne.25272] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 11/08/2022]
Abstract
Obtaining a parts list of the sensory components of the retina is vital to understanding the effects of light in behavior, health, and disease. Rods, cones, and intrinsically photosensitive retinal ganglion cells (ipRGCs) are the best described photoreceptors in the mammalian retina, but recent functional roles have been proposed for retinal neuropsin (Opn5) - an atypical opsin. However, little is known about the pattern of Opn5 expression in the retina. Using cre (Opn5cre ) and cre-dependent reporters, we uncover patterns of Opn5 expression and find that Opn5 is restricted to retinal ganglion cells (RGCs). Opn5-RGCs are non-homogenously distributed through the retina, with greater densities of cells located in the dorsotemporal quadrant. In addition to local topology of these cells, using cre-dependent AAV viral tracing, we surveyed their central targets and found that they are biased towards image-forming and image-stabilizing regions. Finally, molecular and electrophysiological profiling reveal that Opn5-RGCs comprise previously defined RGC types which respond optimally to edges and object-motion (F-mini-ONs, HD2, HD1, LEDs, ooDSRGCs, etc.). Together, these data describe the second collection of RGCs that express atypical opsins in the mouse, and expand the roles of image-forming cells in retinal physiology and function. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Shane P D'Souza
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH, 45229, USA.,The Visual Systems Group.,Center for Chronobiology, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology
| | - David I Swygart
- Departments of Ophthalmology and Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Sophia R Wienbar
- Departments of Ophthalmology and Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Brian A Upton
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH, 45229, USA.,The Visual Systems Group.,Center for Chronobiology, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology.,Medical Scientist Training Program, College of Medicine, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Kevin X Zhang
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH, 45229, USA.,The Visual Systems Group.,Center for Chronobiology, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology.,Medical Scientist Training Program, College of Medicine, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Robert D Mackin
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Anna K Casasent
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Melanie A Samuel
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Gregory W Schwartz
- Departments of Ophthalmology and Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.,Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, 60201, USA
| | - Richard A Lang
- The Visual Systems Group.,Center for Chronobiology, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology.,Division of Developmental Biology, Cincinnati Children's Hospital, Cincinnati, OH, 45229, USA.,Department of Ophthalmology, University of Cincinnati, College of Medicine, Cincinnati, OH, 45229, USA
| |
Collapse
|
35
|
Salay LD, Huberman AD. Divergent outputs of the ventral lateral geniculate nucleus mediate visually evoked defensive behaviors. Cell Rep 2021; 37:109792. [PMID: 34610302 PMCID: PMC10954303 DOI: 10.1016/j.celrep.2021.109792] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/24/2021] [Accepted: 09/12/2021] [Indexed: 11/21/2022] Open
Abstract
Rapid alternations between exploration and defensive reactions require ongoing risk assessment. How visual cues and internal states flexibly modulate the selection of behaviors remains incompletely understood. Here, we show that the ventral lateral geniculate nucleus (vLGN)-a major retinorecipient structure-is a critical node in the network controlling defensive behaviors to visual threats. We find that vLGNGABA neuron activity scales with the intensity of environmental illumination and is modulated by behavioral state. Chemogenetic activation of vLGNGABA neurons reduces freezing, whereas inactivation dramatically extends the duration of freezing to visual threats. Perturbations of vLGN activity disrupt exploration in brightly illuminated environments. We describe both a vLGN→nucleus reuniens (Re) circuit and a vLGN→superior colliculus (SC) circuit, which exert opposite influences on defensive responses. These findings reveal roles for genetic- and projection-defined vLGN subpopulations in modulating the expression of behavioral threat responses according to internal state.
Collapse
Affiliation(s)
- Lindsey D Salay
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andrew D Huberman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA 94305, USA; BioX, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
36
|
Melanopic Limits of Metamer Spectral Optimisation in Multi-Channel Smart Lighting Systems. ENERGIES 2021. [DOI: 10.3390/en14030527] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Modern indoor lighting faces the challenge of finding an appropriate balance between energy consumption, legal requirements, visual performance, and the circadian effectiveness of a spectrum. Multi-channel LED luminaires have the option of keeping image-forming metrics steady while varying the melanopic radiance through metamer spectra for non-visual purposes. Here, we propose the theoretical concept of an automated smart lighting system that is designed to satisfy the user’s visual preference through neural networks while triggering the non-visual pathway via metamers. To quantify the melanopic limits of metamers at a steady chromaticity point, we have used 561 chromaticity coordinates along the Planckian locus (2700 K to 7443 K, ±Duv 0 to 0.048) as optimisation targets and generated the spectra by using a 6-channel, 8-channel, and 11-channel LED combination at three different luminance levels. We have found that in a best-case scenario, the melanopic radiance can be varied up to 65% while keeping the chromaticity coordinates constant (Δu′v′≤7.05×10−5) by using metamer spectra. The highest melanopic metamer contrast can be reached near the Planckian locus between 3292 and 4717 K within a Duv range of −0.009 to 0.006. Additionally, we publish over 1.2 million optimised spectra generated by multichannel LED luminaires as an open-source dataset along with this work.
Collapse
|