1
|
Greer D, Lei T, Kryshtal A, Jessen ZF, Schwartz GW. Visual identification of conspecifics shapes social behavior in mice. Curr Biol 2024:S0960-9822(24)01582-3. [PMID: 39706174 DOI: 10.1016/j.cub.2024.11.041] [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: 05/28/2024] [Revised: 10/07/2024] [Accepted: 11/20/2024] [Indexed: 12/23/2024]
Abstract
Recognizing conspecifics-others of the same species-in order to determine how to interact with them appropriately is a fundamental goal of animal sensory systems. It has undergone selective pressure in nearly all species. Mice have a large repertoire of social behaviors that are the subject of a rapidly growing field of study in neuroscience. Mouse social interactions likely incorporate all available sensory modalities, and the vast majority of studies have not attempted to isolate them. Our understanding of the role of vision in mouse social interactions remains overlooked, given the prominence of olfactory research in this area. To address this, we developed a behavioral platform that allowed us to present a subject mouse with the visual information of stimulus mice in isolation from olfactory, acoustic, and tactile cues. Our results indicate that the visual identification of the sex or individual identity of other mice influences behavior. These findings highlight the underappreciated role of vision in mouse social interactions and open new avenues to study the visual circuits underlying social behavior.
Collapse
Affiliation(s)
- Devon Greer
- Northwestern Interdepartmental Neuroscience Graduate Program, Northwestern University, Chicago, IL 60611, USA.
| | - Tianhao Lei
- Northwestern Interdepartmental Neuroscience Graduate Program, Northwestern University, Chicago, IL 60611, USA
| | - Anna Kryshtal
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Zachary F Jessen
- Northwestern Interdepartmental Neuroscience Graduate Program, Northwestern University, Chicago, IL 60611, USA; Medical Scientist Training Program, Northwestern University, Chicago, IL 60611, USA.
| | - Gregory William Schwartz
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
2
|
Zhu F, Zhang W, Li L, Wang W, Liu S, Zhao Y, Ji X, Yang Y, Kang Z, Guo X, Deng F. Short-term exposure to indoor artificial light at night during sleep impairs cardiac autonomic function of young healthy adults in China. ENVIRONMENTAL RESEARCH 2024; 262:119786. [PMID: 39142452 DOI: 10.1016/j.envres.2024.119786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/16/2024]
Abstract
The artificial light at night (ALAN) exposure has emerged as a significant environmental and public health concern globally. However, there is far less evidence on the health effects of indoor ALAN than on outdoor ALAN. Moreover, evidence on cardiovascular effects of indoor ALAN is more limited. To evaluate the association between short-term exposure to ALAN during sleep with heart rate variability (HRV) in young healthy adults, as well as the mediating role of blood oxygen saturation (SpO2), and to further explore the intervention effects of shading habits, this prospective repeated measurement study was conducted among 81 adults with 150 nights (1324h) of HRV monitoring. HRV and SpO2 were monitored during sleep, concurrently with the measurement of indoor and outdoor ALAN. Shading habits were defined as whether to wear blindfolds or draw bed curtains during sleep, and were collected by questionnaires. Linear mixed-effect model was conducted to assess the association between ALAN exposure and HRV indices. The role of SpO2 in the association was analyzed using mediation analyses. We found that indoor ALAN exposure reduced parasympathetic activity and imbalanced cardiac autonomic function. We also found that the use of outdoor ALAN may underestimate or misestimate the potential health effects of ALAN. A significant mediation effects were observed on standard deviation of normal-to-normal intervals (SDNN; p-value for ACME = 0.014) and the ratio of low frequency power to high frequency power (LF/HF; p-value for ACME = 0.026) through minimum SpO2 after indoor ALAN exposure. The association between indoor ALAN and HRV was more pronounced among participants without shading habits during sleep. This study provides general population-based evidence that short-term exposure to indoor ALAN was significantly associated with impaired HRV, and SpO2 partially mediated the association. Improve shading habits during sleep may mitigate the adverse effects of indoor ALAN.
Collapse
Affiliation(s)
- Fengrui Zhu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China; Eye Hospital, China Academy of Chinese Medical Sciences, Shijingshan, 100040, Beijing, China
| | - Wenlou Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China
| | - Luyi Li
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China
| | - Wanzhou Wang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China
| | - Shan Liu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China
| | - Yetong Zhao
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China
| | - Xuezhao Ji
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China
| | - Yingxin Yang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Zefeng Kang
- Eye Hospital, China Academy of Chinese Medical Sciences, Shijingshan, 100040, Beijing, China
| | - Xinbiao Guo
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China
| | - Furong Deng
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China; Center for Environment and Health, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.
| |
Collapse
|
3
|
Dobrovinskaya O, Alamilla J, Olivas-Aguirre M. Impact of Modern Lifestyle on Circadian Health and Its Contribution to Adipogenesis and Cancer Risk. Cancers (Basel) 2024; 16:3706. [PMID: 39518143 PMCID: PMC11545514 DOI: 10.3390/cancers16213706] [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: 09/27/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Recent research underscores a crucial connection between circadian rhythm disruption and cancer promotion, highlighting an urgent need for attention. OBJECTIVES Explore the molecular mechanisms by which modern lifestyle factors-such as artificial light exposure, shift work, and dietary patterns-affect cortisol/melatonin regulation and cancer risk. METHODS Employing a narrative review approach, we synthesized findings from Scopus, Google Scholar, and PubMed to analyze lifestyle impacts on circadian health, focusing on cortisol and melatonin chronobiology as molecular markers. We included studies that documented quantitative changes in these markers due to modern lifestyle habits, excluding those lacking quantitative data or presenting inconclusive results. Subsequent sections focused solely on articles that quantified the effects of circadian disruption on adipogenesis and tumor microenvironment modifications. RESULTS This review shows how modern habits lead to molecular changes in cortisol and melatonin, creating adipose microenvironments that support cancer development. These disruptions facilitate immune evasion, chemotherapy resistance, and tumor growth, highlighting the critical roles of cortisol dysregulation and melatonin imbalance. CONCLUSIONS Through the presented findings, we establish a causal link between circadian rhythm dysregulation and the promotion of certain cancer types. By elucidating this relationship, the study emphasizes the importance of addressing lifestyle factors that contribute to circadian misalignment, suggesting that targeted interventions could play a crucial role in mitigating cancer risk and improving overall health outcomes.
Collapse
Affiliation(s)
- Oxana Dobrovinskaya
- Laboratory of Immunobiology and Ionic Transport Regulation, University Center for Biomedical Research, University of Colima, Colima 28040, Mexico;
| | - Javier Alamilla
- Consejo Nacional de Humanidades, Ciencia y Tecnología (CONAHCYT), Programa de Investigadores e Investigadoras por México, México City 03940, Mexico;
- Centro Universitario de Investigaciones Biomédicas (CUIB), Universidad de Colima, Colima 28040, Mexico
| | - Miguel Olivas-Aguirre
- Consejo Nacional de Humanidades, Ciencia y Tecnología (CONAHCYT), Programa de Investigadores e Investigadoras por México, México City 03940, Mexico;
- Laboratory of Cancer Pathophysiology, University Center for Biomedical Research, University of Colima, Colima 28040, Mexico
| |
Collapse
|
4
|
Barsanele PS, de Assis LVM, da Silva JJ, Furtado EMDO, Fernandes P, Cipolla-Neto J, Poletini MO, Moraes MN. Glaucoma-inducing retinal ganglion cell degeneration alters diurnal rhythm of key molecular components of the central clock and locomotor activity in mice. FASEB J 2024; 38:e70109. [PMID: 39441606 DOI: 10.1096/fj.202401105r] [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: 05/24/2024] [Revised: 09/06/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024]
Abstract
Glaucoma is a chronic optic neuropathy characterized by the progressive degeneration of retinal ganglion cells (RGC). These cells play a crucial role in transmitting visual and non-visual information to brain regions, including the suprachiasmatic nucleus (SCN), responsible for synchronizing biological rhythms. To understand how glaucoma affects circadian rhythm synchronization, we investigated potential changes in the molecular clock machinery in the SCN. We found that the progressive increase in intraocular pressure (IOP) negatively correlated with spontaneous locomotor activity (SLA). Transcriptome analysis revealed significant alterations in the SCN of glaucomatous mice, including downregulation of genes associated with circadian rhythms. In fact, we showed a loss of diurnal oscillation in the expression of vasoactive intestinal peptide (Vip), its receptor (Vipr2), and period 1 (Per1) in the SCN of glaucomatous mice. These findings were supported by the 7-h phase shift in the peak expression of arginine vasopressin (Avp) in the SCN of mice with glaucoma. Despite maintaining a 24-h period under both light/dark (LD) and constant dark (DD) conditions, glaucomatous mice exhibited altered SLA rhythms, characterized by decreased amplitude. Taken altogether, our findings provide evidence of how glaucoma affects the regulation of the central circadian clock and its consequence on the regulation of circadian rhythms.
Collapse
Affiliation(s)
- Pietra Souza Barsanele
- Laboratório de Cronobiologia Molecular, Departamento de Ciências Biológicas, Instituto de Ciências Ambientais Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, Brazil
- Programa de Pós-graduação em Fisiologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Neurobiologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | | | - Juliano Jefferson da Silva
- Laboratório de Neurobiologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Eliz Maria de Oliveira Furtado
- Laboratório de Cronobiologia Molecular, Departamento de Ciências Biológicas, Instituto de Ciências Ambientais Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, Brazil
- Programa de Pós-graduação em Fisiologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Paola Fernandes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - José Cipolla-Neto
- Laboratório de Neurobiologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Maristela Oliveira Poletini
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria Nathália Moraes
- Laboratório de Cronobiologia Molecular, Departamento de Ciências Biológicas, Instituto de Ciências Ambientais Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, Brazil
- Programa de Pós-graduação em Fisiologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Neurobiologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
5
|
Aranda ML, Min E, Liu LT, Schipma AE, Schmidt TM. Light tunes a novel long-term threat avoidance behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620706. [PMID: 39554108 PMCID: PMC11565844 DOI: 10.1101/2024.10.28.620706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Animals must constantly scan their environment for imminent threats to their safety. However, they must also integrate their past experiences across long timescales to assess the potential recurrence of new threats. Though visual inputs are critical for the detection of environmental danger, whether and how visual information shapes an animal's assessment of whether a new threat is likely to reappear in a given context is unknown. Using a novel behavioral assessment of long-term threat avoidance behavior, we find that animals will avoid a familiar location where they previously experienced a single exposure to an innately threatening visual stimulus. This avoidance behavior is highly sensitive and lasts for multiple days. Intriguingly, we find that the melanopsin-expressing, intrinsically photosensitive retinal ganglion cells tune this behavior via a perihabenular-nucleus accumbens circuit distinct from the canonical visual threat detection circuits. These findings define a specific retinal cell type driving a new long-term threat avoidance behavior driven by prior visual experience.
Collapse
|
6
|
Chan JWY, Li CT, Chau SWH, Chan NY, Li TMH, Huang B, Tsoh J, Li SX, Chong KKL, Roecklein KA, Wing YK. Attenuated melanopsin-mediated post-illumination pupillary response (PIPR) is associated with reduced actigraphic amplitude and mesor in older adults. Sleep 2024:zsae239. [PMID: 39383299 DOI: 10.1093/sleep/zsae239] [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: 06/24/2024] [Indexed: 10/11/2024] Open
Abstract
STUDY OBJECTIVES This study aimed to explore the relationship between post-illumination pupillary response (PIPR) with sleep and circadian measures in a community sample of healthy older adults. METHODS Eligible participants were invited to complete a one-week sleep diary, actigraphy and provide an overnight urine sample to measure urinary 6-sulfatoxymelatonin (aMT6s). PIPR was defined as the as the pupil constriction at 6s post-stimulus (PIPR-6s), and ii) for 30s beginning 10s after stimulus (PIPR-30s) normalized as a percentage to the baseline pupil diameter, after 1s of blue and 1s of red-light stimulus, respectively. The Net-PIPRs were reported by subtracting the PIPR to red stimulus from the PIPR to blue stimulus. The relationship between PIPR metrics to aMT6s and actigraphic rest-activity rhythm parameters was examined by generalized linear models. RESULTS A total of 48 participants were recruited (Mean age: 62.6 ± 7.1 years, Male: 44%). Both Net PIPR-6s and Net PIPR-30s were significantly associated with actigraphic rest-activity amplitude (B=0.03, p=0.001 and B=0.03, p=0.01, respectively), and actigraphic rest-activity mesor (B=0.02, p=0.001 and B=0.03, p=0.004, respectively). Additionally, the Net PIPR-30s were positively associated with overnight aMT6s level (B=0.04, p=0.03), and negatively associated with actigraphic rest-activity acrophase (B=-0.01, p=0.004) in the fully adjusted models. CONCLUSION Attenuated PIPR is associated with a reduced actigraphic amplitude and mesor. The reduced retinal light responsivity may be a potential pathway contributing to impaired photic input to the circadian clock and resulted in the age-related circadian changes in older adults.
Collapse
Affiliation(s)
- Joey W Y Chan
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chun-Tung Li
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Steven Wai Ho Chau
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ngan Yin Chan
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tim Man-Ho Li
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bei Huang
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Joshua Tsoh
- Department of Psychiatry, Prince of Wales Hospital, Hong Kong SAR, China
| | - Shirley X Li
- Department of Psychology, The University of Hong Kong, Hong Kong SAR, China
| | - Kelvin K L Chong
- Department of Ophthalmology and Visual Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | | | - Yun Kwok Wing
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
7
|
Negueruela S, Morenilla-Palao C, Sala S, Ordoño P, Herrera M, Coca Y, López-Cascales MT, Florez-Paz D, Gomis A, Herrera E. Proper Frequency of Perinatal Retinal Waves Is Essential for the Precise Wiring of Visual Axons in Nonimage-Forming Nuclei. J Neurosci 2024; 44:e1408232024. [PMID: 39151955 PMCID: PMC11450533 DOI: 10.1523/jneurosci.1408-23.2024] [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: 07/11/2023] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024] Open
Abstract
The development of the visual system is a complex and multistep process characterized by the precise wiring of retinal ganglion cell (RGC) axon terminals with their corresponding neurons in the visual nuclei of the brain. Upon reaching primary image-forming nuclei (IFN), such as the superior colliculus and the lateral geniculate nucleus, RGC axons undergo extensive arborization that refines over the first few postnatal weeks. The molecular mechanisms driving this activity-dependent remodeling process, which is influenced by waves of spontaneous activity in the developing retina, are still not well understood. In this study, by manipulating the activity of RGCs in mice from either sex and analyzing their transcriptomic profiles before eye-opening, we identified the Type I membrane protein synaptotagmin 13 (Syt13) as involved in spontaneous activity-dependent remodeling. Using these mice, we also explored the impact of spontaneous retinal activity on the development of other RGC recipient targets such as nonimage-forming (NIF) nuclei and demonstrated that proper frequency and duration of retinal waves occurring prior to visual experience are essential for shaping the connectivity of the NIF circuit. Together, these findings contribute to a deeper understanding of the molecular and physiological mechanisms governing activity-dependent axon refinement during the assembly of the visual circuit.
Collapse
Affiliation(s)
- Santiago Negueruela
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), San Juan de Alicante 03550, Spain
| | - Cruz Morenilla-Palao
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), San Juan de Alicante 03550, Spain
| | - Salvador Sala
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), San Juan de Alicante 03550, Spain
| | - Patricia Ordoño
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), San Juan de Alicante 03550, Spain
| | - Macarena Herrera
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), San Juan de Alicante 03550, Spain
| | - Yaiza Coca
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), San Juan de Alicante 03550, Spain
| | - Maria Teresa López-Cascales
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), San Juan de Alicante 03550, Spain
| | - Danny Florez-Paz
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), San Juan de Alicante 03550, Spain
| | - Ana Gomis
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), San Juan de Alicante 03550, Spain
| | - Eloísa Herrera
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), San Juan de Alicante 03550, Spain
| |
Collapse
|
8
|
Ling Y, Wang Y, Ye J, Luan C, Bi A, Gu Y, Shi X. Changes in Intrinsically Photosensitive Retinal Ganglion Cells, Dopaminergic Amacrine Cells, and Their Connectivity in the Retinas of Lid Suture Myopia. Invest Ophthalmol Vis Sci 2024; 65:8. [PMID: 39230992 PMCID: PMC11379095 DOI: 10.1167/iovs.65.11.8] [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: 09/06/2024] Open
Abstract
Purpose This study investigates alterations in intrinsically photosensitive retinal ganglion cells (ipRGCs) and dopaminergic amacrine cells (DACs) in lid suture myopia (LSM) rats. Methods LSM was induced in rats by suturing the right eyes for 4 weeks. Double immunofluorescence staining of ipRGCs and DACs in whole-mount retinas was performed to analyze changes in the density and morphology of control, LSM, and fellow eyes. Real-time quantitative PCR and Western blotting were used to detect related genes and protein expression levels. Results Significant myopia was induced in the lid-sutured eye, but the fellow eye was not different to control. Decreased ipRGC density with paradoxically increased overall melanopsin expression and enlarged dendritic beads was observed in both the LSM and fellow eyes of the LSM rat retinas. In contrast, DAC changes occurred only in the LSM eyes, with reduced DAC density and tyrosine hydroxylase (TH) expression, sparser dendritic processes, and fewer varicosities. Interestingly, contacts between ipRGCs and DACs in the inner plexiform layer (IPL) and the expression of pituitary adenylate cyclase-activating polypeptide (PACAP) and vesicular monoamine transporter protein 2 (VMAT2) mRNA were decreased in the LSM eyes. Conclusions The ipRGCs and DACs in LSM rat retinas undergo multiple alterations in density, morphology, and related molecule expressions. However, the ipRGC changes alone appear not to be required for the development of myopia, given that myopia is only induced in the lid-sutured eye, and they are unlikely alone to drive the DAC changes. Reduced contacts between ipRGCs and DACs in the LSM eyes may be the structural foundation for the impaired signaling between them. PACAP and VMAT2, strongly associated with ipRGCs and DACs, may play important roles in LSM through complex mechanisms.
Collapse
Affiliation(s)
- Ying Ling
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yao Wang
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Jingjing Ye
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changlin Luan
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Ailing Bi
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Gu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xuefeng Shi
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Institute of Ophthalmology, Nankai University, Tianjin, China
| |
Collapse
|
9
|
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
|
10
|
Bergum N, Berezin CT, Vigh J. Dopamine enhances GABA A receptor-mediated current amplitude in a subset of intrinsically photosensitive retinal ganglion cells. J Neurophysiol 2024; 132:501-513. [PMID: 38958282 PMCID: PMC11427049 DOI: 10.1152/jn.00457.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/05/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
Neuromodulation in the retina is crucial for effective processing of retinal signal at different levels of illuminance. Intrinsically photosensitive retinal ganglion cells (ipRGCs), the neurons that drive nonimage-forming visual functions, express a variety of neuromodulatory receptors that tune intrinsic excitability as well as synaptic inputs. Past research has examined actions of neuromodulators on light responsiveness of ipRGCs, but less is known about how neuromodulation affects synaptic currents in ipRGCs. To better understand how neuromodulators affect synaptic processing in ipRGC, we examine actions of opioid and dopamine agonists have on inhibitory synaptic currents in ipRGCs. Although µ-opioid receptor (MOR) activation had no effect on γ-aminobutyric acid (GABA) currents, dopamine [via the D1-type dopamine receptor (D1R)]) amplified GABAergic currents in a subset of ipRGCs. Furthermore, this D1R-mediated facilitation of the GABA conductance in ipRGCs was mediated by a cAMP/PKA-dependent mechanism. Taken together, these findings reinforce the idea that dopamine's modulatory role in retinal adaptation affects both nonimage-forming and image-forming visual functions.NEW & NOTEWORTHY Neuromodulators such as dopamine are important regulators of retinal function. Here, we demonstrate that dopamine increases inhibitory inputs to intrinsically photosensitive retinal ganglion cells (ipRGCs), in addition to its previously established effect on intrinsic light responsiveness. This indicates that dopamine, in addition to its ability to intrinsically modulate ipRGC activity, can also affect synaptic inputs to ipRGCs, thereby tuning retina circuits involved in nonimage-forming visual functions.
Collapse
Affiliation(s)
- Nikolas Bergum
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
| | - Casey-Tyler Berezin
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins,Colorado, United States
| | - Jozsef Vigh
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins,Colorado, United States
| |
Collapse
|
11
|
Kumar D, Khan B, Okcay Y, Sis ÇÖ, Abdallah A, Murray F, Sharma A, Uemura M, Taliyan R, Heinbockel T, Rahman S, Goyal R. Dynamic endocannabinoid-mediated neuromodulation of retinal circadian circuitry. Ageing Res Rev 2024; 99:102401. [PMID: 38964508 DOI: 10.1016/j.arr.2024.102401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 06/05/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
Circadian rhythms are biological rhythms that originate from the "master circadian clock," called the suprachiasmatic nucleus (SCN). SCN orchestrates the circadian rhythms using light as a chief zeitgeber, enabling humans to synchronize their daily physio-behavioral activities with the Earth's light-dark cycle. However, chronic/ irregular photic disturbances from the retina via the retinohypothalamic tract (RHT) can disrupt the amplitude and the expression of clock genes, such as the period circadian clock 2, causing circadian rhythm disruption (CRd) and associated neuropathologies. The present review discusses neuromodulation across the RHT originating from retinal photic inputs and modulation offered by endocannabinoids as a function of mitigation of the CRd and associated neuro-dysfunction. Literature indicates that cannabinoid agonists alleviate the SCN's ability to get entrained to light by modulating the activity of its chief neurotransmitter, i.e., γ-aminobutyric acid, thus preventing light-induced disruption of activity rhythms in laboratory animals. In the retina, endocannabinoid signaling modulates the overall gain of the retinal ganglion cells by regulating the membrane currents (Ca2+, K+, and Cl- channels) and glutamatergic neurotransmission of photoreceptors and bipolar cells. Additionally, endocannabinoids signalling also regulate the high-voltage-activated Ca2+ channels to mitigate the retinal ganglion cells and intrinsically photosensitive retinal ganglion cells-mediated glutamate release in the SCN, thus regulating the RHT-mediated light stimulation of SCN neurons to prevent excitotoxicity. As per the literature, cannabinoid receptors 1 and 2 are becoming newer targets in drug discovery paradigms, and the involvement of endocannabinoids in light-induced CRd through the RHT may possibly mitigate severe neuropathologies.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Neuropharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, HP 173229, India.
| | - Bareera Khan
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, HP 173229, India
| | - Yagmur Okcay
- University of Health Sciences Gulhane Faculty of Pharmacy Department of Pharmacology, Turkey.
| | - Çağıl Önal Sis
- University of Health Sciences Gulhane Faculty of Pharmacy Department of Pharmacology, Turkey.
| | - Aya Abdallah
- Institute of Medical Science, University of Aberdeen, Aberdeen, Scotland.
| | - Fiona Murray
- Institute of Medical Science, University of Aberdeen, Aberdeen, Scotland.
| | - Ashish Sharma
- School of Medicine, Washington University, St. Louis, USA
| | - Maiko Uemura
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology Science, Pilani, Rajasthan 333301, India.
| | - Thomas Heinbockel
- Howard University College of Medicine, Department of Anatomy, Washington, DC 20059, USA
| | - Shafiqur Rahman
- Department of Pharmaceutical Sciences, College of Pharmacy South Dakota State University, Brookings, SD, USA.
| | - Rohit Goyal
- Department of Neuropharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, HP 173229, India.
| |
Collapse
|
12
|
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
|
13
|
Khezri MR, Hsueh H, Mohammadipanah S, Khalili Fard J, Ghasemnejad‐Berenji M. The interplay between the PI3K/AKT pathway and circadian clock in physiologic and cancer-related pathologic conditions. Cell Prolif 2024; 57:e13608. [PMID: 38336976 PMCID: PMC11216939 DOI: 10.1111/cpr.13608] [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: 09/01/2023] [Revised: 12/15/2023] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
The circadian clock is responsible for the regulation of different cellular processes, and its disturbance has been linked to the development of different diseases, such as cancer. The main molecular mechanism for this issue has been linked to the crosstalk between core clock regulators and intracellular pathways responsible for cell survival. The PI3K/AKT signalling pathway is one of the most known intracellular pathways in the case of cancer initiation and progression. This pathway regulates different aspects of cell survival including proliferation, apoptosis, metabolism, and response to environmental stimuli. Accumulating evidence indicates that there is a link between the PI3K/AKT pathway activity and circadian rhythm in physiologic and cancer-related pathogenesis. Different classes of PI3Ks and AKT isoforms are involved in regulating circadian clock components in a transcriptional and functional manner. Reversely, core clock components induce a rhythmic fashion in PI3K and AKT activity in physiologic and pathogenic conditions. The aim of this review is to re-examine the interplay between this pathway and circadian clock components in normal condition and cancer pathogenesis, which provides a better understanding of how circadian rhythms may be involved in cancer progression.
Collapse
Affiliation(s)
- Mohammad Rafi Khezri
- Reproductive Health Research Center, Clinical Research InstituteUrmia University of Medical SciencesUrmiaIran
| | - Hsiang‐Yin Hsueh
- The Ohio State University Graduate Program in Molecular, Cellular and Developmental BiologyThe Ohio State UniversityColumbusOhioUSA
| | - Somayeh Mohammadipanah
- Reproductive Health Research Center, Clinical Research InstituteUrmia University of Medical SciencesUrmiaIran
| | - Javad Khalili Fard
- Department of Pharmacology and Toxicology, Faculty of PharmacyTabriz University of Medical SciencesTabrizIran
| | - Morteza Ghasemnejad‐Berenji
- Department of Pharmacology and Toxicology, Faculty of PharmacyUrmia University of Medical SciencesUrmiaIran
- Research Center for Experimental and Applied Pharmaceutical SciencesUrmia University of Medical SciencesUrmiaIran
| |
Collapse
|
14
|
Wescott DL, Hasler BP, Franzen PL, Taylor ML, Klevens AM, Gamlin P, Siegle GJ, Roecklein KA. Circadian photoentrainment varies by season and depressed state: associations between light sensitivity and sleep and circadian timing. Sleep 2024; 47:zsae066. [PMID: 38530635 PMCID: PMC11168757 DOI: 10.1093/sleep/zsae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/08/2024] [Indexed: 03/28/2024] Open
Abstract
STUDY OBJECTIVES Altered light sensitivity may be an underlying vulnerability for disrupted circadian photoentrainment. The photic information necessary for circadian photoentrainment is sent to the circadian clock from melanopsin-containing intrinsically photosensitive retinal ganglion cells (ipRGCs). The current study tested whether the responsivity of ipRGCs measured using the post-illumination pupil response (PIPR) was associated with circadian phase, sleep timing, and circadian alignment, and if these relationships varied by season or depression severity. METHODS Adult participants (N = 323, agem = 40.5, agesd = 13.5) with varying depression severity were recruited during the summer (n = 154) and winter (n = 169) months. Light sensitivity was measured using the PIPR. Circadian phase was assessed using Dim Light Melatonin Onset (DLMO) on Friday evenings. Midsleep was measured using actigraphy. Circadian alignment was calculated as the DLMO-midsleep phase angle. Multilevel regression models covaried for age, gender, and time since wake of PIPR assessment. RESULTS Greater light sensitivity was associated with later circadian phase in summer but not in winter (β = 0.23; p = 0.03). Greater light sensitivity was associated with shorter DLMO-midsleep phase angles (β = 0.20; p = 0.03) in minimal depression but not in moderate depression (SIGHSAD < 6.6; Johnson-Neyman region of significance). CONCLUSIONS Light sensitivity measured by the PIPR was associated with circadian phase during the summer but not in winter, suggesting ipRGC functioning in humans may affect circadian entrainment when external zeitgebers are robust. Light sensitivity was associated with circadian alignment only in participants with minimal depression, suggesting circadian photoentrainment, a possible driver of mood, may be decreased in depression year-round, similar to decreased photoentrainment in winter.
Collapse
Affiliation(s)
| | - Brant P Hasler
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peter L Franzen
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Maddison L Taylor
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alison M Klevens
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Paul Gamlin
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Greg J Siegle
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | |
Collapse
|
15
|
Tang WSW, Lau NXM, Krishnan MN, Chin YC, Ho CSH. Depression and Eye Disease-A Narrative Review of Common Underlying Pathophysiological Mechanisms and their Potential Applications. J Clin Med 2024; 13:3081. [PMID: 38892791 PMCID: PMC11172702 DOI: 10.3390/jcm13113081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Background: Depression has been shown to be associated with eye diseases, including dry eye disease (DED), cataracts, glaucoma, age-related macular degeneration (AMD), and diabetic retinopathy (DR). This narrative review explores potential pathophysiological connections between depression and eye disease, as well as its potential correlations with ocular parameters. Methods: A literature search was conducted in August 2022 in PUBMED, EMBASE, and PsycINFO. Published articles related to the subject were consolidated and classified according to respective eye diseases and pathophysiological mechanisms. Results: The literature reviewed suggests that common pathophysiological states like inflammation and neurodegeneration may contribute to both depression and certain eye diseases, while somatic symptoms and altered physiology, such as disruptions in circadian rhythm due to eye diseases, can also influence patients' mood states. Grounded in the shared embryological, anatomical, and physiological features between the eye and the brain, depression is also correlated to changes observed in non-invasive ophthalmological imaging modalities, such as changes in the retinal nerve fibre layer and retinal microvasculature. Conclusions: There is substantial evidence of a close association between depression and eye diseases. Understanding the underlying concepts can inform further research on treatment options and monitoring of depression based on ocular parameters.
Collapse
Affiliation(s)
- Wymann Shao Wen Tang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Nicole Xer Min Lau
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | | | - You Chuen Chin
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Raffles Medical Group, Singapore 188770, Singapore
| | - Cyrus Su Hui Ho
- Department of Psychological Medicine, National University of Singapore, Singapore 119077, Singapore
- Department of Psychological Medicine, National University Hospital, Singapore 119228, Singapore
| |
Collapse
|
16
|
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
|
17
|
Aranda ML, Bhoi JD, Parra OAP, Lee SK, Yamada T, Yang Y, Schmidt TM. Genetic tuning of intrinsically photosensitive retinal ganglion cell subtype identity to drive visual behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.590656. [PMID: 38712084 PMCID: PMC11071530 DOI: 10.1101/2024.04.25.590656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The melanopsin-expressing, intrinsically photosensitive retinal ganglion cells (ipRGCs) comprise a subset of the ∼40 retinal ganglion cell types in the mouse retina and drive a diverse array of light-evoked behaviors from circadian photoentrainment to pupil constriction to contrast sensitivity for visual perception. Central to the ability of ipRGCs to control this diverse array of behaviors is the distinct complement of morphophysiological features and gene expression patterns found in the M1-M6 ipRGC subtypes. However, the genetic regulatory programs that give rise to subtypes of ipRGCs are unknown. Here, we identify the transcription factor Brn3b (Pou4f2) as a key genetic regulator that shapes the unique functions of ipRGC subtypes and their diverse downstream visual behaviors.
Collapse
|
18
|
Dyer B, Yu SO, Lane Brown R, Lang RA, D’Souza SP. A new Opn4cre recombinase mouse line to target intrinsically photosensitive retinal ganglion cells (ipRGCs). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.589750. [PMID: 38659888 PMCID: PMC11042346 DOI: 10.1101/2024.04.16.589750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) play a crucial role in several physiological light responses. In this study we generate a new Opn4cre knock-in allele (Opn4cre(DSO)), in which cre is placed immediately downstream of the Opn4 start codon. This approach aims to faithfully reproduce endogenous Opn4 expression and improve compatibility with widely used reporters. We evaluated the efficacy and sensitivity of Opn4cre(DSO) for labeling in retina and brain, and provide an in-depth comparison with the extensively utilized Opn4cre(Saha) line. Through this characterization, Opn4cre(DSO) demonstrated higher specificity in labeling ipRGCs, with minimal recombination escape. Leveraging a combination of electrophysiological, molecular, and morphological analyses, we confirmed its sensitivity in detecting all ipRGC types (M1-M6). Using this new tool, we describe the topographical distributions of ipRGC types across the retinal landscape, uncovering distinct ventronasal biases for M5 and M6 types, setting them apart from their M1-M4 counterparts. In the brain, we find vastly different labeling patterns between lines, with Opn4cre(DSO) only labeling ipRGC axonal projections to their targets. The combination of off-target effects of Opn4cre(Saha) across the retina and brain, coupled with diminished efficiencies of both Cre lines when coupled to less sensitive reporters, underscores the need for careful consideration in experimental design and validation with any Opn4cre driver. Overall, the Opn4cre(DSO) mouse line represents an improved tool for studying ipRGC function and distribution, offering a means to selectively target these cells to study light-regulated behaviors and physiology.
Collapse
Affiliation(s)
- Brannen Dyer
- Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, OH
- Science of Light Center, Cincinnati Children’s Hospital Medical Center, OH
- Abrahamson Pediatric Eye Institute, Cincinnati Children’s Hospital Medical Center, OH
| | - Sue O. Yu
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA
| | - R. Lane Brown
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA
| | - Richard A. Lang
- Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, OH
- Science of Light Center, Cincinnati Children’s Hospital Medical Center, OH
- Abrahamson Pediatric Eye Institute, Cincinnati Children’s Hospital Medical Center, OH
- Department of Ophthalmology, University of Cincinnati, OH
| | - Shane P. D’Souza
- Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, OH
- Science of Light Center, Cincinnati Children’s Hospital Medical Center, OH
- Abrahamson Pediatric Eye Institute, Cincinnati Children’s Hospital Medical Center, OH
| |
Collapse
|
19
|
Lazzerini Ospri L, Zhan JJ, Thomsen MB, Wang H, Komal R, Tang Q, Messanvi F, du Hoffmann J, Cravedi K, Chudasama Y, Hattar S, Zhao H. Light affects the prefrontal cortex via intrinsically photosensitive retinal ganglion cells. SCIENCE ADVANCES 2024; 10:eadh9251. [PMID: 38552022 PMCID: PMC10980283 DOI: 10.1126/sciadv.adh9251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 02/23/2024] [Indexed: 04/01/2024]
Abstract
The ventromedial prefrontal cortex (vmPFC) is a part of the limbic system engaged in the regulation of social, emotional, and cognitive states, which are characteristically impaired in disorders of the brain such as schizophrenia and depression. Here, we show that intrinsically photosensitive retinal ganglion cells (ipRGCs) modulate, through light, the integrity, activity, and function of the vmPFC. This regulatory role, which is independent of circadian and mood alterations, is mediated by an ipRGC-thalamic-corticolimbic pathway. Lack of ipRGC signaling in mice causes dendritic degeneration, dysregulation of genes involved in synaptic plasticity, and depressed neuronal activity in the vmPFC. These alterations primarily undermine the ability of the vmPFC to regulate emotions. Our discovery provides a potential light-dependent mechanism for certain PFC-centric disorders in humans.
Collapse
Affiliation(s)
| | - Jesse J. Zhan
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael B. Thomsen
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hui Wang
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ruchi Komal
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qijun Tang
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Fany Messanvi
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Johann du Hoffmann
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kevin Cravedi
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yogita Chudasama
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Samer Hattar
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
20
|
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
|
21
|
Lazzerini-Ospri L, Zhan JJ, Thomsen MB, Wang H, Messanvi F, du Hoffmann J, Cravedi K, Komal R, Chudasama Y, Zhao H, Hattar S. WITHDRAWN: Light affects the prefrontal cortex via intrinsically photosensitive retinal ganglion cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.22.556752. [PMID: 37808740 PMCID: PMC10557604 DOI: 10.1101/2023.09.22.556752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
This manuscript has been withdrawn by bioRxiv following a formal request by the NIH Intramural Research Integrity Office owing to lack of author consent.
Collapse
|
22
|
Hunyara JL, Daly KM, Torres K, Yurgel ME, Komal R, Hattar S, Kolodkin AL. Teneurin-3 regulates the generation of non-image-forming visual circuitry and responsiveness to light in the suprachiasmatic nucleus. PLoS Biol 2023; 21:e3002412. [PMID: 38048352 PMCID: PMC10729976 DOI: 10.1371/journal.pbio.3002412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 12/19/2023] [Accepted: 10/31/2023] [Indexed: 12/06/2023] Open
Abstract
Visual system function depends upon the elaboration of precise connections between retinal ganglion cell (RGC) axons and their central targets in the brain. Though some progress has been made in defining the molecules that regulate RGC connectivity required for the assembly and function of image-forming circuitry, surprisingly little is known about factors required for intrinsically photosensitive RGCs (ipRGCs) to target a principal component of the non-image-forming circuitry: the suprachiasmatic nucleus (SCN). Furthermore, the molecules required for forming circuits critical for circadian behaviors within the SCN are not known. We observe here that the adhesion molecule teneurin-3 (Tenm3) is highly expressed in vasoactive intestinal peptide (VIP) neurons located in the core region of the SCN. Since Tenm3 is required for other aspects of mammalian visual system development, we investigate roles for Tenm3 in regulating ipRGC-SCN connectivity and function. Our results show that Tenm3 negatively regulates association between VIP and arginine vasopressin (AVP) neurons within the SCN and is essential for M1 ipRGC axon innervation to the SCN. Specifically, in Tenm3-/- mice, we find a reduction in ventro-medial innervation to the SCN. Despite this reduction, Tenm3-/- mice have higher sensitivity to light and faster re-entrainment to phase advances, probably due to the increased association between VIP and AVP neurons. These data show that Tenm3 plays key roles in elaborating non-image-forming visual system circuitry and that it influences murine responses to phase-advancing light stimuli.
Collapse
Affiliation(s)
- John L. Hunyara
- The Solomon H. Snyder Department of Neuroscience, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - K. M. Daly
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Katherine Torres
- The Solomon H. Snyder Department of Neuroscience, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Maria E. Yurgel
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ruchi Komal
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Samer Hattar
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alex L. Kolodkin
- The Solomon H. Snyder Department of Neuroscience, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
23
|
Li Y, Androulakis IP. The SCN-HPA-Periphery Circadian Timing System: Mathematical Modeling of Clock Synchronization and the Effects of Photoperiod on Jetlag Adaptation. J Biol Rhythms 2023; 38:601-616. [PMID: 37529986 PMCID: PMC10615703 DOI: 10.1177/07487304231188541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
Synchronizing the circadian timing system (CTS) to external light/dark cycles is crucial for homeostasis maintenance and environmental adaptation. The CTS is organized hierarchically, with the central pacemaker located in the suprachiasmatic nuclei (SCN) generating coherent oscillations that are entrained to light/dark cycles. These oscillations regulate the release of glucocorticoids by the hypothalamus-pituitary-adrenal (HPA) axis, which acts as a systemic entrainer of peripheral clocks throughout the body. The SCN adjusts its network plasticity in response to variations in photoperiod, leading to changes in the rhythmic release of glucocorticoids and ultimately impacting peripheral clocks. However, the effects of photoperiod-induced variations of glucocorticoids on the synchronization of peripheral clocks are not fully understood, and the interaction between jetlag adaption and photoperiod changes is unclear. This study presents a semi-mechanistic mathematical model to investigate how the CTS responds to changes in photoperiod. Specifically, the study focuses on the entrainment properties of a system composed of the SCN, HPA axis, and peripheral clocks. The results show that high-amplitude glucocorticoid rhythms lead to a more coherent phase distribution in the periphery. In addition, our study investigates the effect of photoperiod exposure on jetlag recovery time and phase shift, proposing different interventional strategies for eastward and westward jetlag. The findings suggest that decreasing photic exposure before jetlag during eastward traveling and after jetlag during westward traveling can accelerate jetlag readaptation. The study provides insights into the mechanisms of CTS organization and potential recovery strategies for transitions between time zones and lighting zones.
Collapse
Affiliation(s)
- Yannuo Li
- Department of Chemical & Biochemical Engineering, Rutgers University-New Brunswick, New Brunswick, New Jersey, USA
| | - Ioannis P Androulakis
- Department of Chemical & Biochemical Engineering, Rutgers University-New Brunswick, New Brunswick, New Jersey, USA
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, USA
- Department of Surgery, Robert Wood Johnson Medical School, Rutgers University-New Brunswick, New Brunswick, New Jersey, USA
| |
Collapse
|
24
|
Lee H, Weinberg-Wolf H, Lee HL, Lee T, Conte J, Godoy-Parejo C, Demb JB, Rudenko A, Kim IJ. Brn3b regulates the formation of fear-related midbrain circuits and defensive responses to visual threat. PLoS Biol 2023; 21:e3002386. [PMID: 37983249 PMCID: PMC10695396 DOI: 10.1371/journal.pbio.3002386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 12/04/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023] Open
Abstract
Defensive responses to visually threatening stimuli represent an essential fear-related survival instinct, widely detected across species. The neural circuitry mediating visually triggered defensive responses has been delineated in the midbrain. However, the molecular mechanisms regulating the development and function of these circuits remain unresolved. Here, we show that midbrain-specific deletion of the transcription factor Brn3b causes a loss of neurons projecting to the lateral posterior nucleus of the thalamus. Brn3b deletion also down-regulates the expression of the neuropeptide tachykinin 2 (Tac2). Furthermore, Brn3b mutant mice display impaired defensive freezing responses to visual threat precipitated by social isolation. This behavioral phenotype could be ameliorated by overexpressing Tac2, suggesting that Tac2 acts downstream of Brn3b in regulating defensive responses to threat. Together, our experiments identify specific genetic components critical for the functional organization of midbrain fear-related visual circuits. Similar mechanisms may contribute to the development and function of additional long-range brain circuits underlying fear-associated behavior.
Collapse
Affiliation(s)
- Hyoseo Lee
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Hannah Weinberg-Wolf
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Hae-Lim Lee
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Tracy Lee
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Joseph Conte
- Department of Biology, City College of New York, New York, New York, United States of America
| | - Carlos Godoy-Parejo
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Jonathan B. Demb
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Wu Tsai Institute, Yale University, New Haven, Connecticut, United States of America
| | - Andrii Rudenko
- Department of Biology, City College of New York, New York, New York, United States of America
- Graduate Programs in Biology and Biochemistry, City University of New York, New York, New York, United States of America
| | - In-Jung Kim
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Wu Tsai Institute, Yale University, New Haven, Connecticut, United States of America
| |
Collapse
|
25
|
Pan D, Wang Z, Chen Y, Cao J. Melanopsin-mediated optical entrainment regulates circadian rhythms in vertebrates. Commun Biol 2023; 6:1054. [PMID: 37853054 PMCID: PMC10584931 DOI: 10.1038/s42003-023-05432-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/09/2023] [Indexed: 10/20/2023] Open
Abstract
Melanopsin (OPN4) is a light-sensitive protein that plays a vital role in the regulation of circadian rhythms and other nonvisual functions. Current research on OPN4 has focused on mammals; more evidence is needed from non-mammalian vertebrates to fully assess the significance of the non-visual photosensitization of OPN4 for circadian rhythm regulation. There are species differences in the regulatory mechanisms of OPN4 for vertebrate circadian rhythms, which may be due to the differences in the cutting variants, tissue localization, and photosensitive activation pathway of OPN4. We here summarize the distribution of OPN4 in mammals, birds, and teleost fish, and the classical excitation mode for the non-visual photosensitive function of OPN4 in mammals is discussed. In addition, the role of OPN4-expressing cells in regulating circadian rhythm in different vertebrates is highlighted, and the potential rhythmic regulatory effects of various neuropeptides or neurotransmitters expressed in mammalian OPN4-expressing ganglion cells are summarized among them.
Collapse
Affiliation(s)
- Deng Pan
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China
| | - Zixu Wang
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China
| | - Yaoxing Chen
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China
| | - Jing Cao
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China.
| |
Collapse
|
26
|
Huang Y, Liao P, Yu J, Chen S. Light disrupts social memory via a retina-to-supraoptic nucleus circuit. EMBO Rep 2023; 24:e56839. [PMID: 37531065 PMCID: PMC10561173 DOI: 10.15252/embr.202356839] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/03/2023] Open
Abstract
The formation of social memory between individuals of the opposite sex is crucial for expanding mating options or establishing monogamous pair bonding. A specialized neuronal circuit that regulates social memory could enhance an individual's mating opportunities and provide a parallel pathway for computing social behaviors. While the influence of light exposure on various forms of memory, such as fear and object memory, has been studied, its modulation of social recognition memory remains unclear. Here, we demonstrate that acute exposure to light impairs social recognition memory (SRM) in mice. Unlike sound and touch stimuli, light inhibits oxytocin neurons in the supraoptic nucleus (SON) via M1 SON-projecting intrinsically photosensitive retinal ganglion cells (ipRGCs) and GABAergic neurons in the perinuclear zone of the SON (pSON). We further show that optogenetic activation of SON oxytocin neurons using channelrhodopsin is sufficient to enhance SRM performance, even under light conditions. Our findings unveil a dedicated neuronal circuit through which luminance affects SRM, utilizing a non-image-forming visual pathway, distinct from the canonical modulatory role of the oxytocin system.
Collapse
Affiliation(s)
- Yu‐Fan Huang
- Department of Life ScienceNational Taiwan UniversityTaipeiTaiwan
| | - Po‐Yu Liao
- Department of Life ScienceNational Taiwan UniversityTaipeiTaiwan
| | - Jo‐Hsien Yu
- Department of Life ScienceNational Taiwan UniversityTaipeiTaiwan
| | - Shih‐Kuo Chen
- Department of Life ScienceNational Taiwan UniversityTaipeiTaiwan
| |
Collapse
|
27
|
Abstract
Because the central nervous system is largely nonrenewing, neurons and their synapses must be maintained over the lifetime of an individual to ensure circuit function. Age is a dominant risk factor for neural diseases, and declines in nervous system function are a common feature of aging even in the absence of disease. These alterations extend to the visual system and, in particular, to the retina. The retina is a site of clinically relevant age-related alterations but has also proven to be a uniquely approachable system for discovering principles that govern neural aging because it is well mapped, contains diverse neuron types, and is experimentally accessible. In this article, we review the structural and molecular impacts of aging on neurons within the inner and outer retina circuits. We further discuss the contribution of non-neuronal cell types and systems to retinal aging outcomes. Understanding how and why the retina ages is critical to efforts aimed at preventing age-related neural decline and restoring neural function.
Collapse
Affiliation(s)
- Jeffrey D Zhu
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| | - Sharma Pooja Tarachand
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| | - Qudrat Abdulwahab
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| | - Melanie A Samuel
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| |
Collapse
|
28
|
She Z, Ward AH, Gawne TJ. The effects of ambient narrowband long-wavelength light on lens-induced myopia and form-deprivation myopia in tree shrews. Exp Eye Res 2023; 234:109593. [PMID: 37482282 PMCID: PMC10529043 DOI: 10.1016/j.exer.2023.109593] [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: 05/13/2023] [Revised: 06/21/2023] [Accepted: 07/11/2023] [Indexed: 07/25/2023]
Abstract
Here we examine the effects of ambient red light on lens-induced myopia and diffuser-induced myopia in tree shrews, small diurnal mammals closely related to primates. Starting at 24 days of visual experience (DVE), seventeen tree shrews were reared in red light (624 ± 10 or 634 ± 10 nm, 527-749 human lux) for 12-14 days wearing either a -5D lens (RL-5D, n = 5) or a diffuser (RLFD, n = 5) monocularly, or without visual restriction (RL-Control, n = 7). Refractive errors and ocular dimensions were compared to those obtained from tree shrews raised in broad-spectrum white light (WL-5D, n = 5; WLFD, n = 10; WL Control, n = 7). The RL-5D tree shrews developed less myopia in their lens-treated eyes than WL-5D tree shrews at the end of the experiment (-1.1 ± 0.9D vs. -3.8 ± 0.3D, p = 0.007). The diffuser-treated eyes of the RLFD tree shrews were near-emmetropic (-0.3 ± 0.6D, vs. -5.4 ± 0.7D in the WLFD group). Red light induced hyperopia in control animals (RL-vs. WL-Control, +3.0 ± 0.7 vs. +1.0 ± 0.2D, p = 0.02), the no-lens eyes of the RL-5D animals, and the no-diffuser eyes of the RLFD animals (+2.5 ± 0.5D and +2.3 ± 0.3D, respectively). The refractive alterations were consistent with the alterations in vitreous chamber depth. The lens-induced myopia developed in red light suggests that a non-chromatic cue could signal defocus to a less accurate extent, although it could also be a result of "form-deprivation" caused by defocus blur. As with previous studies in rhesus monkeys, the ability of red light to promote hyperopia appears to correlate with its ability to retard lens-induced myopia and form-deprivation myopia, the latter of which might be related to non-visual ocular mechanisms.
Collapse
Affiliation(s)
- Zhihui She
- Department of Optometry and Vision Science, University of Alabama at Birmingham, 1716 University Blvd, HPB 528, Birmingham, AL, 35294, UK
| | - Alexander H Ward
- Georgia Cancer Center, Augusta University. Dr. Ward Contributed to This Work During His Graduate Training at the University of Alabama at Birmingham, UK
| | - Timothy J Gawne
- Department of Optometry and Vision Science, University of Alabama at Birmingham, 1716 University Blvd, HPB 528, Birmingham, AL, 35294, UK.
| |
Collapse
|
29
|
Berry MH, Leffler J, Allen CN, Sivyer B. Functional subtypes of rodent melanopsin ganglion cells switch roles between night and day illumination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.26.554902. [PMID: 38168436 PMCID: PMC10760181 DOI: 10.1101/2023.08.26.554902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs), contain the photopigment melanopsin, and influence both image and non-image forming behaviors. Despite being categorized into multiple types (M1-M6), physiological variability within these types suggests our current understanding of ipRGCs is incomplete. We used multi-electrode array (MEA) recordings and unbiased cluster analysis under synaptic blockade to identify 8 functional clusters of ipRGCs, each with distinct photosensitivity and response timing. We used Cre mice to drive the expression of channelrhodopsin in SON-ipRGCs, enabling the localization of distinct ipRGCs in the dorsal retina. Additionally, we conducted a retrospective unbiased cluster analysis of ipRGC photoresponses to light stimuli across scotopic, mesopic, and photopic intensities, aimed at activating both rod and cone inputs to ipRGCs. Our results revealed shared and distinct synaptic inputs to the identified functional clusters, demonstrating that ipRGCs encode visual information with high fidelity at low light intensities, but poorly at photopic light intensities, when melanopsin activation is highest. Collectively, our findings support a framework with at least 8 functional subtypes of ipRGCs, each encoding luminance with distinct spike outputs, highlighting the inherent functional diversity and complexity of ipRGCs and suggesting a reevaluation of their contributions to retinal function and visual perception under varying light conditions.
Collapse
Affiliation(s)
- Michael H. Berry
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239
- Medical Scientist Training program, Oregon Health & Science University, Portland, OR, 97239
| | - Joseph Leffler
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239
| | - Charles N. Allen
- Oregon Institute of Occupational Health Sciences, Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239
| | - Benjamin Sivyer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239
| |
Collapse
|
30
|
Wang H, Ma X, Yu Z, Hu N, Du Y, He X, Pan D, Pu L, Zhang X, Sun X, Li J. Exposure to outdoor artificial light at night increases risk and burden of metabolic disease in Ningxia, China. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:87517-87526. [PMID: 37428317 DOI: 10.1007/s11356-023-28684-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 07/04/2023] [Indexed: 07/11/2023]
Abstract
Nearly a quarter of the world's land has already been polluted by artificial light. And numerous human and animal studies have corroborated that light at night can disrupt metabolism. Therefore, we aimed to estimate the association between outdoor artificial light at night (ALAN) and the presence of metabolic disease. Daily hospital admission cases from Ningxia, China, between 2014 and 2020 were included. Cumulative associations between outdoor ALAN and metabolic disease were estimated using logistic regression and distributed lagged non-linear models (DLNM) with lags of 0-30 days and stratified analysis by age groups and gender. The results suggest that 26.80% of metabolic disease cases in Ningxia can be attributed to outdoor ALAN and that men, especially in men aged 46-59 years, are more susceptible to lighting. Policymakers need to develop measures and facilities in corresponding areas, such as universal access to indoor blackout curtains. In particular, men should be urged to minimize going outside at night and to develop protective measures specifically for men.
Collapse
Affiliation(s)
- Huihui Wang
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China
| | - Xiaohong Ma
- Medical Insurance Office, Traditional Chinese Medicine Hospital of Yinchuan, Hui Autonomous Region, Yinchuan, 750001, Ningxia, China
| | - Zhenfan Yu
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China
| | - Naifan Hu
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China
| | - Yurun Du
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China
| | - Xiaoxue He
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China
| | - Degong Pan
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China
| | - Lining Pu
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China
| | - Xue Zhang
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China
| | - Xian Sun
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China
| | - Jiangping Li
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China.
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
31
|
Jones AA, Arble DM. In light of breathing: environmental light is an important modulator of breathing with clinical implications. Front Neurosci 2023; 17:1217799. [PMID: 37521684 PMCID: PMC10373889 DOI: 10.3389/fnins.2023.1217799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
In vertebrate animals, the automatic, rhythmic pattern of breathing is a highly regulated process that can be modulated by various behavioral and physiological factors such as metabolism, sleep-wake state, activity level, and endocrine signaling. Environmental light influences many of these modulating factors both indirectly by organizing daily and seasonal rhythms of behavior and directly through acute changes in neural signaling. While several observations from rodent and human studies suggest that environmental light affects breathing, few have systematically evaluated the underlying mechanisms and clinical relevance of environmental light on the regulation of respiratory behavior. Here, we provide new evidence and discuss the potential neurobiological mechanisms by which light modulates breathing. We conclude that environmental light should be considered, from bench to bedside, as a clinically relevant modulator of respiratory health and disease.
Collapse
|
32
|
Quan Y, Duan H, Zhan Z, Shen Y, Lin R, Liu T, Zhang T, Wu J, Huang J, Zhai G, Song X, Zhou Y, Sun X. Binocular head-mounted chromatic pupillometry can detect structural and functional loss in glaucoma. Front Neurosci 2023; 17:1187619. [PMID: 37456990 PMCID: PMC10346847 DOI: 10.3389/fnins.2023.1187619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/07/2023] [Indexed: 07/18/2023] Open
Abstract
Aim The aim of this study is to evaluate the utility of binocular chromatic pupillometry in detecting impaired pupillary light response (PLR) in patients with primary open-angle glaucoma (POAG) and to assess the feasibility of using binocular chromatic pupillometer in opportunistic POAG diagnosis in community-based or telemedicine-based services. Methods In this prospective, cross-sectional study, 74 patients with POAG and 23 healthy controls were enrolled. All participants underwent comprehensive ophthalmologic examinations including optical coherence tomography (OCT) and standard automated perimetry (SAP). The PLR tests included sequential tests of full-field chromatic stimuli weighted by rods, intrinsically photosensitive retinal ganglion cells (ipRGCs), and cones (Experiment 1), as well as alternating chromatic light flash-induced relative afferent pupillary defect (RAPD) test (Experiment 2). In Experiment 1, the constricting amplitude, velocity, and time to maximum constriction/dilation were calculated in three cell type-weighted responses, and the post-illumination response of ipRGC-weighted response was evaluated. In Experiment 2, infrared pupillary asymmetry (IPA) amplitude and anisocoria duration induced by intermittent blue or red light flashes were calculated. Results In Experiment 1, the PLR of POAG patients was significantly reduced in all conditions, reflecting the defect in photoreception through rods, cones, and ipRGCs. The variable with the highest area under the receiver operating characteristic curve (AUC) was time to max dilation under ipRGC-weighted stimulus, followed by the constriction amplitude under cone-weighted stimulus and the constriction amplitude response to ipRGC-weighted stimuli. The impaired PLR features were associated with greater visual field loss, thinner retinal nerve fiber layer (RNFL) thickness, and cupping of the optic disk. In Experiment 2, IPA and anisocoria duration induced by intermittent blue or red light flashes were significantly greater in participants with POAG than in controls. IPA and anisocoria duration had good diagnostic value, correlating with the inter-eye asymmetry of visual field loss. Conclusion We demonstrate that binocular chromatic pupillometry could potentially serve as an objective clinical tool for opportunistic glaucoma diagnosis in community-based or telemedicine-based services. Binocular chromatic pupillometry allows an accurate, objective, and rapid assessment of retinal structural impairment and functional loss in glaucomatous eyes of different severity levels.
Collapse
Affiliation(s)
- Yadan Quan
- Department of Ophthalmology and Visual Science, Shanghai Medical College, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
- NHC and Chinese Academy of Medical Sciences Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Huiyu Duan
- Institute of Image Communication and Network Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zongyi Zhan
- Department of Retinal Disease, Shenzhen Eye Institute, Shenzhen Eye Hospital, Shenzhen Eye Hospital Affiliated to Jinan University, Shenzhen, China
| | - Yuening Shen
- Department of Ophthalmology and Visual Science, Shanghai Medical College, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Rui Lin
- Department of Ophthalmology and Visual Science, Shanghai Medical College, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
- NHC and Chinese Academy of Medical Sciences Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Tingting Liu
- Department of Ophthalmology and Visual Science, Shanghai Medical College, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
- NHC and Chinese Academy of Medical Sciences Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Ting Zhang
- Department of Ophthalmology and Visual Science, Shanghai Medical College, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
- NHC and Chinese Academy of Medical Sciences Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Jihong Wu
- Department of Ophthalmology and Visual Science, Shanghai Medical College, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
- NHC and Chinese Academy of Medical Sciences Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Jing Huang
- Institute of Image Communication and Network Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guangtao Zhai
- Institute of Image Communication and Network Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xuefei Song
- Department of Ophthalmology, Ninth People's Hospital of Shanghai, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yixiong Zhou
- Department of Ophthalmology, Ninth People's Hospital of Shanghai, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Xinghuai Sun
- Department of Ophthalmology and Visual Science, Shanghai Medical College, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
- NHC and Chinese Academy of Medical Sciences Key Laboratory of Myopia, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
33
|
Eto T, Higuchi S. Review on age-related differences in non-visual effects of light: melatonin suppression, circadian phase shift and pupillary light reflex in children to older adults. J Physiol Anthropol 2023; 42:11. [PMID: 37355647 DOI: 10.1186/s40101-023-00328-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 06/15/2023] [Indexed: 06/26/2023] Open
Abstract
Physiological effects of light exposure in humans are diverse. Among them, the circadian rhythm phase shift effect in order to maintain a 24-h cycle of the biological clock is referred to as non-visual effects of light collectively with melatonin suppression and pupillary light reflex. The non-visual effects of light may differ depending on age, and clarifying age-related differences in the non-visual effects of light is important for providing appropriate light environments for people of different ages. Therefore, in various research fields, including physiological anthropology, many studies on the effects of age on non-visual functions have been carried out in older people, children and adolescents by comparing the effects with young adults. However, whether the non-visual effects of light vary depending on age and, if so, what factors contribute to the differences have remained unclear. In this review, results of past and recent studies on age-related differences in the non-visual effects of light are presented and discussed in order to provide clues for answering the question of whether non-visual effects of light actually vary depending on age. Some studies, especially studies focusing on older people, have shown age-related differences in non-visual functions including differences in melatonin suppression, circadian phase shift and pupillary light reflex, while other studies have shown no differences. Studies showing age-related differences in the non-visual effects of light have suspected senile constriction and crystalline lens opacity as factors contributing to the differences, while studies showing no age-related differences have suspected the presence of a compensatory mechanism. Some studies in children and adolescents have shown that children's non-visual functions may be highly sensitive to light, but the studies comparing with other age groups seem to have been limited. In order to study age-related differences in non-visual effects in detail, comparative studies should be conducted using subjects having a wide range of ages and with as much control as possible for intensity, wavelength component, duration, circadian timing, illumination method of light exposure, and other factors (mydriasis or non-mydriasis, cataracts or not in the older adults, etc.).
Collapse
Affiliation(s)
- Taisuke Eto
- Research Fellow of the Japan Society for the Promotion of Science, Kodaira, Japan
- Department of Sleep-Wake Disorders, National Center of Neurology and Psychiatry, National Institute of Mental Health, Kodaira, Japan
| | - Shigekazu Higuchi
- Department of Human Life Design and Science, Faculty of Design, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
34
|
Feord RC, Gomoliszewska A, Pienaar A, Mouland JW, Brown TM. Colour opponency is widespread across the mouse subcortical visual system and differentially targets GABAergic and non-GABAergic neurons. Sci Rep 2023; 13:9313. [PMID: 37291239 PMCID: PMC10250360 DOI: 10.1038/s41598-023-35885-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: 12/16/2022] [Accepted: 05/25/2023] [Indexed: 06/10/2023] Open
Abstract
Colour vision plays many important roles in animal behaviour but the brain pathways processing colour remain surprisingly poorly understood, including in the most commonly used laboratory mammal, mice. Indeed, particular features of mouse retinal organisation present challenges in defining the mechanisms underlying colour vision in mice and have led to suggestions that this may substantially rely on 'non-classical' rod-cone opponency. By contrast, studies using mice with altered cone spectral sensitivity, to facilitate application of photoreceptor-selective stimuli, have revealed widespread cone-opponency across the subcortical visual system. To determine the extent to which such findings are truly reflective of wildtype mouse colour vision, and facilitate neural circuit mapping of colour-processing pathways using intersectional genetic approaches, we here establish and validate stimuli for selectively manipulating excitation of the native mouse S- and M-cone opsin classes. We then use these to confirm the widespread appearance of cone-opponency (> 25% of neurons) across the mouse visual thalamus and pretectum. We further extend these approaches to map the occurrence of colour-opponency across optogenetically identified GABAergic (GAD2-expressing) cells in key non-image forming visual centres (pretectum and intergeniculate leaflet/ventral lateral geniculate; IGL/vLGN). Strikingly, throughout, we find S-ON/M-OFF opponency is specifically enriched in non-GABAergic cells, with identified GABAergic cells in the IGL/VLGN entirely lacking this property. Collectively, therefore, we establish an important new approach for studying cone function in mice, confirming a surprisingly extensive appearance of cone-opponent processing in the mouse visual system and providing new insight into functional specialisation of the pathways processing such signals.
Collapse
Affiliation(s)
- R C Feord
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - A Gomoliszewska
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - A Pienaar
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - J W Mouland
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - T M Brown
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
35
|
Schilling T, Soltanlou M, Nuerk HC, Bahmani H. Blue-light stimulation of the blind-spot constricts the pupil and enhances contrast sensitivity. PLoS One 2023; 18:e0286503. [PMID: 37256905 DOI: 10.1371/journal.pone.0286503] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 05/17/2023] [Indexed: 06/02/2023] Open
Abstract
Short- and long-wavelength light can alter pupillary responses differently, allowing inferences to be made about the contribution of different photoreceptors on pupillary constriction. In addition to classical retinal photoreceptors, the pupillary light response is formed by the activity of melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGC). It has been shown in rodents that melanopsin is expressed in the axons of ipRGCs that bundle at the optic nerve head, which forms the perceptual blind-spot. Hence, the first aim of this study was to investigate if blind-spot stimulation induces a pupillary response. The second aim was to investigate the effect of blind-spot stimulation by using the contrast sensitivity tests. Fifteen individuals participated in the pupil response experiment and thirty-two individuals in the contrast sensitivity experiment. The pupillary change was quantified using the post-illumination pupil response (PIPR) amplitudes after blue-light (experimental condition) and red-light (control condition) pulses in the time window between 2 s and 6 s post-illumination. The contrast sensitivity was assessed using two different tests: the Freiburg Visual Acuity Test and Contrast Test and the Tuebingen Contrast Sensitivity Test, respectively. Contrast sensitivity was measured before and 20 minutes after binocular blue-light stimulation of the blind-spot at spatial frequencies higher than or equal to 3 cycles per degree (cpd) and at spatial frequencies lower than 3 cpd (control condition). Blue-light blind-spot stimulation induced a significantly larger PIPR compared to red-light, confirming a melanopsin-mediated pupil-response in the blind-spot. Furthermore, contrast sensitivity was increased after blind-spot stimulation, confirmed by both contrast sensitivity tests. Only spatial frequencies of at least 3 cpd were enhanced. This study demonstrates that stimulating the blind-spot with blue-light constricts the pupil and increases the contrast sensitivity at higher spatial frequencies.
Collapse
Affiliation(s)
| | - Mojtaba Soltanlou
- Department of Psychology, University of Tuebingen, Tuebingen, Germany
- School of Psychology, University of Surrey, Guildford, United Kingdom
| | - Hans-Christoph Nuerk
- Department of Psychology, University of Tuebingen, Tuebingen, Germany
- LEAD Graduate School & Research Network, University of Tuebingen, Tuebingen, Germany
| | - Hamed Bahmani
- Dopavision GmbH, Berlin, Germany
- Bernstein Center for Computational Neuroscience, Tuebingen, Germany
| |
Collapse
|
36
|
Linne C, Mon KY, D’Souza S, Jeong H, Jiang X, Brown DM, Zhang K, Vemaraju S, Tsubota K, Kurihara T, Pardue MT, Lang RA. Encephalopsin (OPN3) is required for normal refractive development and the GO/GROW response to induced myopia. Mol Vis 2023; 29:39-57. [PMID: 37287644 PMCID: PMC10243678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/05/2023] [Indexed: 06/09/2023] Open
Abstract
Purpose Myopia, or nearsightedness, is the most common form of refractive error and is increasing in prevalence. While significant efforts have been made to identify genetic variants that predispose individuals to myopia, these variants are believed to account for only a small portion of the myopia prevalence, leading to a feedback theory of emmetropization, which depends on the active perception of environmental visual cues. Consequently, there has been renewed interest in studying myopia in the context of light perception, beginning with the opsin family of G-protein coupled receptors (GPCRs). Refractive phenotypes have been characterized in every opsin signaling pathway studied, leaving only Opsin 3 (OPN3), the most widely expressed and blue-light sensing noncanonical opsin, to be investigated for function in the eye and refraction. Methods Opn3 expression was assessed in various ocular tissues using an Opn3eGFP reporter. Weekly refractive development in Opn3 retinal and germline mutants from 3 to 9 weeks of age was measured using an infrared photorefractor and spectral domain optical coherence tomography (SD-OCT). Susceptibility to lens-induced myopia was then assessed using skull-mounted goggles with a -30 diopter experimental and a 0 diopter control lens. Mouse eye biometry was similarly tracked from 3 to 6 weeks. A myopia gene expression signature was assessed 24 h after lens induction for germline mutants to further assess myopia-induced changes. Results Opn3 was found to be expressed in a subset of retinal ganglion cells and a limited number of choroidal cells. Based on an assessment of Opn3 mutants, the OPN3 germline, but not retina conditional Opn3 knockout, exhibits a refractive myopia phenotype, which manifests in decreased lens thickness, shallower aqueous compartment depth, and shorter axial length, atypical of traditional axial myopias. Despite the short axial length, Opn3 null eyes demonstrate normal axial elongation in response to myopia induction and mild changes in choroidal thinning and myopic shift, suggesting that susceptibility to lens-induced myopia is largely unchanged. Additionally, the Opn3 null retinal gene expression signature in response to induced myopia after 24 h is distinct, with opposing Ctgf, Cx43, and Egr1 polarity compared to controls. Conclusions The data suggest that an OPN3 expression domain outside the retina can control lens shape and thus the refractive performance of the eye. Prior to this study, the role of Opn3 in the eye had not been investigated. This work adds OPN3 to the list of opsin family GPCRs that are implicated in emmetropization and myopia. Further, the work to exclude retinal OPN3 as the contributing domain in this refractive phenotype is unique and suggests a distinct mechanism when compared to other opsins.
Collapse
Affiliation(s)
- Courtney Linne
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Molecular & Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Khine Yin Mon
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Shane D’Souza
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Molecular & Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Heonuk Jeong
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
| | - Xiaoyan Jiang
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
| | - Dillon M. Brown
- Department of Ophthalmology and Neuroscience Program, Emory University School of Medicine, Atlanta, GA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, GA
| | - Kevin Zhang
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Molecular & Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Shruti Vemaraju
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, OH
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Tsubota Laboratory, Inc., Tokyo, Japan
| | - Toshihide Kurihara
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
| | - Machelle T. Pardue
- Department of Ophthalmology and Neuroscience Program, Emory University School of Medicine, Atlanta, GA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, GA
| | - Richard A. Lang
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, OH
| |
Collapse
|
37
|
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: 3] [Impact Index Per Article: 1.5] [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
|
38
|
Berry MH, Moldavan M, Garrett T, Meadows M, Cravetchi O, White E, Leffler J, von Gersdorff H, Wright KM, Allen CN, Sivyer B. A melanopsin ganglion cell subtype forms a dorsal retinal mosaic projecting to the supraoptic nucleus. Nat Commun 2023; 14:1492. [PMID: 36932080 PMCID: PMC10023714 DOI: 10.1038/s41467-023-36955-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 02/24/2023] [Indexed: 03/19/2023] Open
Abstract
Visual input to the hypothalamus from intrinsically photosensitive retinal ganglion cells (ipRGCs) influences several functions including circadian entrainment, body temperature, and sleep. ipRGCs also project to nuclei such as the supraoptic nucleus (SON), which is involved in systemic fluid homeostasis, maternal behavior, social behaviors, and appetite. However, little is known about the SON-projecting ipRGCs or their relationship to well-characterized ipRGC subtypes. Using a GlyT2Cre mouse line, we show a subtype of ipRGCs restricted to the dorsal retina that selectively projects to the SON. These ipRGCs tile a dorsal region of the retina, forming a substrate for encoding ground luminance. Optogenetic activation of their axons demonstrates they release the neurotransmitter glutamate in multiple regions, including the suprachiasmatic nucleus (SCN) and SON. Our results challenge the idea that ipRGC dendrites overlap to optimize photon capture and suggests non-image forming vision operates to sample local regions of the visual field to influence diverse behaviors.
Collapse
Affiliation(s)
- Michael H Berry
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
- Medical Scientist Training Program, Oregon Health & Science University, Portland, OR, USA
| | - Michael Moldavan
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Tavita Garrett
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
- Neuroscience Graduate program, Oregon Health & Science University, Portland, OR, USA
| | - Marc Meadows
- Neuroscience Graduate program, Oregon Health & Science University, Portland, OR, USA
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Olga Cravetchi
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Elizabeth White
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Joseph Leffler
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Henrique von Gersdorff
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
| | - Kevin M Wright
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Charles N Allen
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Benjamin Sivyer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA.
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
39
|
Meng JJ, Shen JW, Li G, Ouyang CJ, Hu JX, Li ZS, Zhao H, Shi YM, Zhang M, Liu R, Chen JT, Ma YQ, Zhao H, Xue T. Light modulates glucose metabolism by a retina-hypothalamus-brown adipose tissue axis. Cell 2023; 186:398-412.e17. [PMID: 36669474 DOI: 10.1016/j.cell.2022.12.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/22/2022] [Accepted: 12/13/2022] [Indexed: 01/20/2023]
Abstract
Public health studies indicate that artificial light is a high-risk factor for metabolic disorders. However, the neural mechanism underlying metabolic modulation by light remains elusive. Here, we found that light can acutely decrease glucose tolerance (GT) in mice by activation of intrinsically photosensitive retinal ganglion cells (ipRGCs) innervating the hypothalamic supraoptic nucleus (SON). Vasopressin neurons in the SON project to the paraventricular nucleus, then to the GABAergic neurons in the solitary tract nucleus, and eventually to brown adipose tissue (BAT). Light activation of this neural circuit directly blocks adaptive thermogenesis in BAT, thereby decreasing GT. In humans, light also modulates GT at the temperature where BAT is active. Thus, our work unveils a retina-SON-BAT axis that mediates the effect of light on glucose metabolism, which may explain the connection between artificial light and metabolic dysregulation, suggesting a potential prevention and treatment strategy for managing glucose metabolic disorders.
Collapse
Affiliation(s)
- Jian-Jun Meng
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, 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
| | - Jia-Wei Shen
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, 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
| | - Guang Li
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, 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
| | - Chang-Jie Ouyang
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, 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
| | - Jia-Xi Hu
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, 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
| | - Zi-Shuo Li
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, 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
| | - Hang Zhao
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, 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
| | - Yi-Ming Shi
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, 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
| | - Mei Zhang
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, 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
| | - Rong Liu
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, 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
| | - Ju-Tao Chen
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, 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
| | - Yu-Qian Ma
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, 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
| | - Huan Zhao
- College of Biology, Food and Environment, Hefei University, Hefei 230601, China
| | - Tian Xue
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, 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; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
40
|
Nadal-Nicolás FM, Galindo-Romero C, Lucas-Ruiz F, Marsh-Amstrong N, Li W, Vidal-Sanz M, Agudo-Barriuso M. Pan-retinal ganglion cell markers in mice, rats, and rhesus macaques. Zool Res 2023; 44:226-248. [PMID: 36594396 PMCID: PMC9841181 DOI: 10.24272/j.issn.2095-8137.2022.308] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Univocal identification of retinal ganglion cells (RGCs) is an essential prerequisite for studying their degeneration and neuroprotection. Before the advent of phenotypic markers, RGCs were normally identified using retrograde tracing of retinorecipient areas. This is an invasive technique, and its use is precluded in higher mammals such as monkeys. In the past decade, several RGC markers have been described. Here, we reviewed and analyzed the specificity of nine markers used to identify all or most RGCs, i.e., pan-RGC markers, in rats, mice, and macaques. The best markers in the three species in terms of specificity, proportion of RGCs labeled, and indicators of viability were BRN3A, expressed by vision-forming RGCs, and RBPMS, expressed by vision- and non-vision-forming RGCs. NEUN, often used to identify RGCs, was expressed by non-RGCs in the ganglion cell layer, and therefore was not RGC-specific. γ-SYN, TUJ1, and NF-L labeled the RGC axons, which impaired the detection of their somas in the central retina but would be good for studying RGC morphology. In rats, TUJ1 and NF-L were also expressed by non-RGCs. BM88, ERRβ, and PGP9.5 are rarely used as markers, but they identified most RGCs in the rats and macaques and ERRβ in mice. However, PGP9.5 was also expressed by non-RGCs in rats and macaques and BM88 and ERRβ were not suitable markers of viability.
Collapse
Affiliation(s)
- Francisco M Nadal-Nicolás
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Murcia 30120, Spain
- Dpto. Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia 30120, Spain
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892-2510, USA
| | - Caridad Galindo-Romero
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Murcia 30120, Spain
- Dpto. Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia 30120, Spain
| | - Fernando Lucas-Ruiz
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Murcia 30120, Spain
- Dpto. Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia 30120, Spain
| | - Nicholas Marsh-Amstrong
- Department of Ophthalmology and Vision Science, University of California, Davis, CA 95817, USA
| | - Wei Li
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892-2510, USA
| | - Manuel Vidal-Sanz
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Murcia 30120, Spain
- Dpto. Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia 30120, Spain. E-mail:
| | - Marta Agudo-Barriuso
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Murcia 30120, Spain
- Dpto. Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia 30120, Spain. E-mail:
| |
Collapse
|
41
|
Raja S, Milosavljevic N, Allen AE, Cameron MA. Burning the candle at both ends: Intraretinal signaling of intrinsically photosensitive retinal ganglion cells. Front Cell Neurosci 2023; 16:1095787. [PMID: 36687522 PMCID: PMC9853061 DOI: 10.3389/fncel.2022.1095787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/13/2022] [Indexed: 01/09/2023] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) are photoreceptors located in the ganglion cell layer. They project to brain regions involved in predominately non-image-forming functions including entrainment of circadian rhythms, control of the pupil light reflex, and modulation of mood and behavior. In addition to possessing intrinsic photosensitivity via the photopigment melanopsin, these cells receive inputs originating in rods and cones. While most research in the last two decades has focused on the downstream influence of ipRGC signaling, recent studies have shown that ipRGCs also act retrogradely within the retina itself as intraretinal signaling neurons. In this article, we review studies examining intraretinal and, in addition, intraocular signaling pathways of ipRGCs. Through these pathways, ipRGCs regulate inner and outer retinal circuitry through both chemical and electrical synapses, modulate the outputs of ganglion cells (both ipRGCs and non-ipRGCs), and influence arrangement of the correct retinal circuitry and vasculature during development. These data suggest that ipRGC function plays a significant role in the processing of image-forming vision at its earliest stage, positioning these photoreceptors to exert a vital role in perceptual vision. This research will have important implications for lighting design to optimize the best chromatic lighting environments for humans, both in adults and potentially even during fetal and postnatal development. Further studies into these unique ipRGC signaling pathways could also lead to a better understanding of the development of ocular dysfunctions such as myopia.
Collapse
Affiliation(s)
- Sushmitha Raja
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Nina Milosavljevic
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Annette E. Allen
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Morven A. Cameron
- School of Medicine, Western Sydney University, Sydney, NSW, Australia,*Correspondence: Morven A. Cameron,
| |
Collapse
|
42
|
Portengen BL, Porro GL, Imhof SM, Naber M. The Trade-Off Between Luminance and Color Contrast Assessed With Pupil Responses. Transl Vis Sci Technol 2023; 12:15. [PMID: 36622687 PMCID: PMC9838585 DOI: 10.1167/tvst.12.1.15] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/06/2022] [Indexed: 01/10/2023] Open
Abstract
Purpose A scene consisting of a white stimulus on a black background incorporates strong luminance contrast. When both stimulus and background receive different colors, luminance contrast decreases but color contrast increases. Here, we sought to characterize the pattern of stimulus salience across varying trade-offs of color and luminance contrasts by using the pupil light response. Methods Three experiments were conducted with 17, 16, and 17 healthy adults. For all experiments, a flickering stimulus (2 Hz; alternating color to black) was presented superimposed on a background with a complementary color to the stimulus (i.e., opponency colors in human color perception: blue and yellow for Experiment 1, red and green for Experiment 2, and equiluminant red and green for Experiment 3). Background luminance varied between 0% and 45% to trade off luminance and color contrast with the stimulus. By comparing the locus of the optimal trade-off between color and luminance across different color axes, we explored the generality of the trade-off. Results The strongest pupil responses were found when a substantial amount of color contrast was present (at the expense of luminance contrast). Pupil response amplitudes increased by 15% to 30% after the addition of color contrast. An optimal pupillary responsiveness was reached at a background luminance setting of 20% to 35% color contrast across several color axes. Conclusions These findings suggest that a substantial component of pupil light responses incorporates color processing. More sensitive pupil responses and more salient stimulus designs can be achieved by adding subtle levels of color contrast between stimulus and background. Translational Relevance More robust pupil responses will enhance tests of the visual field with pupil perimetry.
Collapse
Affiliation(s)
- Brendan L. Portengen
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Experimental Psychology, Helmholtz Institute, Utrecht University, Utrecht, The Netherlands
| | - Giorgio L. Porro
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Saskia M. Imhof
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marnix Naber
- Department of Experimental Psychology, Helmholtz Institute, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
43
|
Arévalo-López C, Gleitze S, Madariaga S, Plaza-Rosales I. Pupillary response to chromatic light stimuli as a possible biomarker at the early stage of glaucoma: a review. Int Ophthalmol 2023; 43:343-356. [PMID: 35781599 DOI: 10.1007/s10792-022-02381-8] [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: 09/10/2021] [Accepted: 06/14/2022] [Indexed: 02/07/2023]
Abstract
Glaucoma is a multifactorial neurodegenerative disease of the optic nerve currently considered a severe health problem because of its high prevalence, being the primary cause of irreversible blindness worldwide. The most common type corresponds to Primary Open-Angle Glaucoma. Glaucoma produces, among other alterations, a progressive loss of retinal ganglion cells (RGC) and its axons which are the key contributors to generate action potentials that reach the visual cortex to create the visual image. Glaucoma is characterized by Visual Field loss whose main feature is to be painless and therefore makes early detection difficult, causing a late diagnosis and a delayed treatment indication that slows down its progression. Intrinsically photosensitive retinal ganglion cells, which represent a subgroup of RGCs are characterized by their response to short-wave light stimulation close to 480 nm, their non-visual function, and their role in the generation of the pupillary reflex. Currently, the sensitivity of clinical examinations correlates to RGC damage; however, the need for an early damage biomarker is still relevant. It is an urgent task to create new diagnostic approaches to detect an early stage of glaucoma in a prompt, quick, and economical manner. We summarize the pathology of glaucoma and its current clinical detection methods, and we suggest evaluating the pupillary response to chromatic light as a potential biomarker of disease, due to its diagnostic benefit and its cost-effectiveness in clinical practice in order to reduce irreversible damage caused by glaucoma.
Collapse
Affiliation(s)
- Carla Arévalo-López
- Department of Medical Technology, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Silvia Gleitze
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Samuel Madariaga
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Laboratorio de Neurosistemas, Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Ecological Cognitive Neuroscience Group, Santiago, Chile
| | - Iván Plaza-Rosales
- Department of Medical Technology, Faculty of Medicine, Universidad de Chile, Santiago, Chile. .,Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile. .,Laboratorio de Neurosistemas, Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile. .,Ecological Cognitive Neuroscience Group, Santiago, Chile.
| |
Collapse
|
44
|
Qiu L, Wei S, Yang Y, Zhang R, Ru S, Zhang X. Mechanism of bisphenol S exposure on color sensitivity of zebrafish larvae. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 316:120670. [PMID: 36395908 DOI: 10.1016/j.envpol.2022.120670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/03/2022] [Accepted: 11/13/2022] [Indexed: 06/16/2023]
Abstract
Color vision, initiated from cone cells, is vitally essential for identifying environmental information in vertebrate. Although the retinotoxicity of bisphenol S (BPS) has been reported, data on the influence of BPS treatment on cone cells are scarce. In the present study, transgenic zebrafish (Danio rerio) labeling red and ultraviolet (UV) cones were exposed to BPS (0, 1, 10, and 100 μg/L) during the early stages of retinal development, to elucidate the mechanism underlying its retinal cone toxicity of BPS. The results showed that 10 and 100 μg/L BPS induced oxidative DNA damage, structural damage (decreased number of ribbon synapses), mosaic patterning disorder, and altered expression of genes involved in the phototransduction pathway in red and UV cones. Furthermore, BPS exposure also caused abnormal development of key neurons (retinal ganglion cells, optic nerve, and hypothalamus), responsible for transmitting the light-electrical signal to brain, and thereby resulted in inhibition of light-electrical signal transduction, finally diminishing the spectral sensitivity of zebrafish larvae to long- and short-type light signal at 5 day post fertilization. This study highlights the cone-toxicity of environmental relevant concentrations of BPS, and clarifies the mechanism of color vision impairment induced by BPS at the cellular level, updating the understanding of visual behavior driven by environmental factors.
Collapse
Affiliation(s)
- Liguo Qiu
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
| | - Shuhui Wei
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Yixin Yang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Rui Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Xiaona Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
| |
Collapse
|
45
|
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
|
46
|
Hunyara JL, Foshe S, Varadarajan SG, Gribble KD, Huberman AD, Kolodkin AL. Characterization of non-alpha retinal ganglion cell injury responses reveals a possible block to restoring ipRGC function. Exp Neurol 2022; 357:114176. [PMID: 35870522 PMCID: PMC9549754 DOI: 10.1016/j.expneurol.2022.114176] [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: 05/02/2022] [Revised: 07/04/2022] [Accepted: 07/16/2022] [Indexed: 11/04/2022]
Abstract
Visual impairment caused by retinal ganglion cell (RGC) axon damage or degeneration affects millions of individuals throughout the world. While some progress has been made in promoting long-distance RGC axon regrowth following injury, it remains unclear whether RGC axons can properly reconnect with their central targets to restore visual function. Additionally, the regenerative capacity of many RGC subtypes remains unknown in part due to a lack of available genetic tools. Here, we use a new mouse line, Sema6ACreERT2, that labels On direction-selective RGCs (oDSGCs) and characterize the survival and regenerative potential of these cells following optic nerve crush (ONC). In parallel, we use a previously characterized mouse line, Opn4CreERT2, to answer these same questions for M1 intrinsically photosensitive RGCs (ipRGCs). We find that both M1 ipRGCs and oDSGCs are resilient to injury but do not display long-distance axon regrowth following Lin28a overexpression. Unexpectedly, we found that M1 ipRGC, but not oDSGC, intraretinal axons exhibit ectopic branching and are misaligned near the optic disc between one- and three-weeks following injury. Additionally, we observe that numerous ectopic presynaptic specializations associate with misguided ipRGC intraretinal axons. Taken together, these results reveal insights into the injury response of M1 ipRGCs and oDSGCs, providing a foundation for future efforts seeking to restore visual system function following injury.
Collapse
Affiliation(s)
- John L Hunyara
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sierra Foshe
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | - Katherine D Gribble
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Andrew D Huberman
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA; Department of Ophthalmology, Stanford University, Stanford, CA 94305, USA
| | - Alex L Kolodkin
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
47
|
A small molecule M1 promotes optic nerve regeneration to restore target-specific neural activity and visual function. Proc Natl Acad Sci U S A 2022; 119:e2121273119. [PMID: 36306327 PMCID: PMC9636930 DOI: 10.1073/pnas.2121273119] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Axon regeneration is an energy-demanding process that requires active mitochondrial transport. In contrast to the central nervous system (CNS), axonal mitochondrial transport in regenerating axons of the peripheral nervous system (PNS) increases within hours and sustains for weeks after injury. Yet, little is known about targeting mitochondria in nervous system repair. Here, we report the induction of sustained axon regeneration, neural activities in the superior colliculus (SC), and visual function recovery after optic nerve crush (ONC) by M1, a small molecule that promotes mitochondrial fusion and transport. We demonstrated that M1 enhanced mitochondrial dynamics in cultured neurons and accelerated in vivo axon regeneration in the PNS. Ex vivo time-lapse imaging and kymograph analysis showed that M1 greatly increased mitochondrial length, axonal mitochondrial motility, and transport velocity in peripheral axons of the sciatic nerves. Following ONC, M1 increased the number of axons regenerating through the optic chiasm into multiple subcortical areas and promoted the recovery of local field potentials in the SC after optogenetic stimulation of retinal ganglion cells, resulting in complete recovery of the pupillary light reflex, and restoration of the response to looming visual stimuli was detected. M1 increased the gene expression of mitochondrial fusion proteins and major axonal transport machinery in both the PNS and CNS neurons without inducing inflammatory responses. The knockdown of two key mitochondrial genes,
Opa1
or
Mfn2
, abolished the growth-promoting effects of M1 after ONC, suggesting that maintaining a highly dynamic mitochondrial population in axons is required for successful CNS axon regeneration.
Collapse
|
48
|
Checa-Ros A, D’Marco L. Role of Omega-3 Fatty Acids as Non-Photic Zeitgebers and Circadian Clock Synchronizers. Int J Mol Sci 2022; 23:12162. [PMID: 36293015 PMCID: PMC9603208 DOI: 10.3390/ijms232012162] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 10/23/2024] Open
Abstract
Omega-3 fatty acids (ω-3 FAs) are well-known for their actions on immune/inflammatory and neurological pathways, functions that are also under circadian clock regulation. The daily photoperiod represents the primary circadian synchronizer ('zeitgeber'), although diverse studies have pointed towards an influence of dietary FAs on the biological clock. A comprehensive literature review was conducted following predefined selection criteria with the aim of updating the evidence on the molecular mechanisms behind circadian rhythm regulation by ω-3 FAs. We collected preclinical and clinical studies, systematic reviews, and metanalyses focused on the effect of ω-3 FAs on circadian rhythms. Twenty animal (conducted on rodents and piglets) and human trials and one observational study providing evidence on the regulation of neurological, inflammatory/immune, metabolic, reproductive, cardiovascular, and biochemical processes by ω-3 FAs via clock genes were discussed. The evidence suggests that ω-3 FAs may serve as non-photic zeitgebers and prove therapeutically beneficial for circadian disruption-related pathologies. Future work should focus on the role of clock genes as a target for the therapeutic use of ω-3 FAs in inflammatory and neurological disorders, as well as on the bidirectional association between the molecular clock and ω-3 FAs.
Collapse
Affiliation(s)
- Ana Checa-Ros
- Department of Medicine and Surgery, Faculty of Health Sciences, Universidad Cardenal Herrera—CEU, CEU Universities, 46115 Valencia, Spain
- Aston Institute of Health and Neurosciences, School of Life & Health Sciences, Aston University, Birmingham B4 7ET, UK
| | - Luis D’Marco
- Department of Medicine and Surgery, Faculty of Health Sciences, Universidad Cardenal Herrera—CEU, CEU Universities, 46115 Valencia, Spain
- Department of Nephrology, Hospital General Universitario de Valencia, 46014 Valencia, Spain
| |
Collapse
|
49
|
Fitzpatrick MJ, Kerschensteiner D. Homeostatic plasticity in the retina. Prog Retin Eye Res 2022; 94:101131. [PMID: 36244950 DOI: 10.1016/j.preteyeres.2022.101131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 02/07/2023]
Abstract
Vision begins in the retina, whose intricate neural circuits extract salient features of the environment from the light entering our eyes. Neurodegenerative diseases of the retina (e.g., inherited retinal degenerations, age-related macular degeneration, and glaucoma) impair vision and cause blindness in a growing number of people worldwide. Increasing evidence indicates that homeostatic plasticity (i.e., the drive of a neural system to stabilize its function) can, in principle, preserve retinal function in the face of major perturbations, including neurodegeneration. Here, we review the circumstances and events that trigger homeostatic plasticity in the retina during development, sensory experience, and disease. We discuss the diverse mechanisms that cooperate to compensate and the set points and outcomes that homeostatic retinal plasticity stabilizes. Finally, we summarize the opportunities and challenges for unlocking the therapeutic potential of homeostatic plasticity. Homeostatic plasticity is fundamental to understanding retinal development and function and could be an important tool in the fight to preserve and restore vision.
Collapse
|
50
|
Ladakis DC, Gould J, Khazen JM, Lefelar JM, Tarpey S, Bies CJ, Salky R, Fitzgerald KC, Bhargava P, Nourbakhsh B, Sotirchos ES. Fatigue is a common symptom in myelin oligodendrocyte glycoprotein antibody disease. Mult Scler J Exp Transl Clin 2022; 8:20552173221131235. [DOI: 10.1177/20552173221131235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Background Unlike multiple sclerosis and neuromyelitis optica, the burden of fatigue in myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) is unclear. Objective To compare fatigue levels between people with MOGAD and household controls (HC) and explore factors associated with fatigue severity. Methods In a cross-sectional survey, data were collected from people with MOGAD and HC by utilizing an online questionnaire. Data elements included demographics, sleep quality measures, comorbidities, MOGAD characteristics, and fatigue severity measured by the Modified Fatigue Impact Scale (MFIS). We compared fatigue severity between MOGAD participants and HC and assessed the associations between demographic and disease characteristics and fatigue severity. Results There were 180/283 MOGAD and 61/126 HC respondents. Compared to HC, people with MOGAD reported more severe fatigue, as measured by the MFIS total score (49.3 vs. 36.5; p < 0.001), and a larger proportion of MOGAD participants (75.6% vs. 44.3%; p < 0.001) were classified as fatigued. Among MOGAD participants, higher age ( p = 0.04), history of bilateral optic neuritis ( p = 0.02), and current use of acute treatment ( p = 0.04) were independently associated with higher fatigue. Conclusions Fatigue is common in people with MOGAD, and a history of bilateral optic neuritis, comorbid conditions, and ongoing disease activity appear to contribute to fatigue severity.
Collapse
Affiliation(s)
- Dimitrios C Ladakis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | - Rebecca Salky
- The MOG project, Olney, MD, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Kathryn C Fitzgerald
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bardia Nourbakhsh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elias S Sotirchos
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|