1
|
Mattam S, Thomas RH, Akansha EO, Jathanna JS, Poojary RR, Sarpangala S, Jose J, Theruveethi N. Influence of white-light-emitting diodes on primary visual cortex layer 5 pyramidal neurons (V1L5PNs) and remodeling by blue-light-blocking lenses. Int Ophthalmol 2024; 44:118. [PMID: 38416231 PMCID: PMC10901925 DOI: 10.1007/s10792-024-03036-6] [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: 11/24/2022] [Accepted: 01/12/2024] [Indexed: 02/29/2024]
Abstract
Studies have explored the consequences of excessive exposure to white-light-emitting diodes (LEDs) in the retina. Hence, we aimed to assess the implications of such exposure on structural alterations of the visual cortex, learning and memory, and amelioration by blue-light-blocking lenses (BBLs). Eight-week-old Wistar rats (n = 24) were used for the experiment and divided into four groups (n = 6 in each group) as control, white LED light exposure (LE), BBL Crizal Prevencia-1 (CP), and DuraVision Blue-2 (DB). Animals in the exposure group were exposed to white LED directly for 28 days (12:12-h light/dark cycle), whereas animals in the BBL groups were exposed to similar light with BBLs attached to the LEDs. Post-exposure, a Morris water maze was performed for memory retention, followed by structural analysis of layer 5 pyramidal neurons in the visual cortex. We observed a significant difference (P < 0.001) in the functional test on day 1 and day 2 of training in the LE group. Structural analysis of Golgi-Cox-stained visual cortex layer 5 pyramidal neurons showed significant alterations in the apical and basal branching points (p < 0.001) and basal intersection points (p < 0.001) in the LE group. Post hoc analysis revealed significant changes between (p < 0.001) LE and CP and (p < 0.001) CP and DB groups. Constant and cumulative exposure to white LEDs presented with structural and functional alterations in the visual cortex, which are partly remodeled by BBLs.
Collapse
Affiliation(s)
- Susmitha Mattam
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, India
- Sankara College of Optometry, Hyderabad, 500032, India
| | - R Huban Thomas
- Department of Anatomy, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Elizebeth O Akansha
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, India
- University of Houston College of Optometry, Houston, USA
| | - Judith S Jathanna
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Radhika R Poojary
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Shailaja Sarpangala
- Department of Ophthalmology, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Judy Jose
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Nagarajan Theruveethi
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
2
|
Zhu S, Xie T, Lv Z, Leng YB, Zhang YQ, Xu R, Qin J, Zhou Y, Roy VAL, Han ST. Hierarchies in Visual Pathway: Functions and Inspired Artificial Vision. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2301986. [PMID: 37435995 DOI: 10.1002/adma.202301986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/28/2023] [Accepted: 07/10/2023] [Indexed: 07/13/2023]
Abstract
The development of artificial intelligence has posed a challenge to machine vision based on conventional complementary metal-oxide semiconductor (CMOS) circuits owing to its high latency and inefficient power consumption originating from the data shuffling between memory and computation units. Gaining more insights into the function of every part of the visual pathway for visual perception can bring the capabilities of machine vision in terms of robustness and generality. Hardware acceleration of more energy-efficient and biorealistic artificial vision highly necessitates neuromorphic devices and circuits that are able to mimic the function of each part of the visual pathway. In this paper, we review the structure and function of the entire class of visual neurons from the retina to the primate visual cortex within reach (Chapter 2) are reviewed. Based on the extraction of biological principles, the recent hardware-implemented visual neurons located in different parts of the visual pathway are discussed in detail in Chapters 3 and 4. Furthermore, valuable applications of inspired artificial vision in different scenarios (Chapter 5) are provided. The functional description of the visual pathway and its inspired neuromorphic devices/circuits are expected to provide valuable insights for the design of next-generation artificial visual perception systems.
Collapse
Affiliation(s)
- Shirui Zhu
- Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Tao Xie
- Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Ziyu Lv
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Yan-Bing Leng
- Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Yu-Qi Zhang
- Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Runze Xu
- Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Jingrun Qin
- Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Ye Zhou
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Vellaisamy A L Roy
- School of Science and Technology, Hong Kong Metropolitan University, Hong Kong, 999077, P. R. China
| | - Su-Ting Han
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Lu K, Wu J, Tang S, Wang Y, Zhang L, Chai F, Liang XF. Altered Visual Function in Short-Wave-Sensitive 1 ( sws1) Gene Knockout Japanese Medaka ( Oryzias latipes) Larvae. Cells 2023; 12:2157. [PMID: 37681889 PMCID: PMC10486665 DOI: 10.3390/cells12172157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/29/2023] [Accepted: 08/21/2023] [Indexed: 09/09/2023] Open
Abstract
Visual perception plays a crucial role in foraging, avoiding predators, mate selection, and communication. The regulation of color vision is largely dependent on opsin, which is the first step in the formation of the visual transduction cascade in photoreceptor cells. Short-wave-sensitive 1 (sws1) is a visual pigment that mediates short-wavelength light transduction in vertebrates. The depletion of sws1 resulted in increased M-opsin in mice. However, there is still no report on the visual function of sws1 in teleost fish. Here, we constructed the sws1 knockout medaka using CRISPR/Cas9 technology. The 6 dph (days post-hatching) medaka sws1-/- larvae exhibited significantly decreased food intake and total length at the first feeding stage, and the mRNA levels of orexigenic genes (npy and agrp) were significantly upregulated after feeding. The swimming speed was significantly reduced during the period of dark-light transition stimulation in the sws1-mutant larvae. Histological analysis showed that the thickness of the lens was reduced, whereas the thickness of the ganglion cell layer (GCL) was significantly increased in sws1-/- medaka larvae. Additionally, the deletion of sws1 decreased the mRNA levels of genes involved in phototransduction (gnb3b, grk7a, grk7b, and pde6c). We also observed increased retinal cell apoptosis and oxidative stress in sws1 knockout medaka larvae. Collectively, these results suggest that sws1 deficiency in medaka larvae may impair visual function and cause retinal cell apoptosis, which is associated with the downregulation of photoconduction expression and oxidative stress.
Collapse
Affiliation(s)
- Ke Lu
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China; (K.L.); (Y.W.)
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| | - Jiaqi Wu
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China; (K.L.); (Y.W.)
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| | - Shulin Tang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China; (K.L.); (Y.W.)
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| | - Yuye Wang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China; (K.L.); (Y.W.)
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| | - Lixin Zhang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China; (K.L.); (Y.W.)
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| | - Farui Chai
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China; (K.L.); (Y.W.)
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| | - Xu-Fang Liang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China; (K.L.); (Y.W.)
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| |
Collapse
|
6
|
Mouland JW, Watson AJ, Martial FP, Lucas RJ, Brown TM. Colour and melanopsin mediated responses in the murine retina. Front Cell Neurosci 2023; 17:1114634. [PMID: 36993934 PMCID: PMC10040579 DOI: 10.3389/fncel.2023.1114634] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/17/2023] [Indexed: 03/15/2023] Open
Abstract
Introduction: Intrinsically photosensitive retinal ganglion cells (ipRGCs) integrate melanopsin and rod/cone-mediated inputs to signal to the brain. Whilst originally identified as a cell type specialised for encoding ambient illumination, several lines of evidence indicate a strong association between colour discrimination and ipRGC-driven responses. Thus, cone-mediated colour opponent responses have been widely found across ipRGC target regions in the mouse brain and influence a key ipRGC-dependent function, circadian photoentrainment. Although ipRGCs exhibiting spectrally opponent responses have also been identified, the prevalence of such properties have not been systematically evaluated across the mouse retina or yet been found in ipRGC subtypes known to influence the circadian system. Indeed, there is still uncertainty around the overall prevalence of cone-dependent colour opponency across the mouse retina, given the strong retinal gradient in S and M-cone opsin (co)-expression and overlapping spectral sensitivities of most mouse opsins.Methods: To address this, we use photoreceptor isolating stimuli in multielectrode recordings from human red cone opsin knock-in mouse (Opn1mwR) retinas to systematically survey cone mediated responses and the occurrence of colour opponency across ganglion cell layer (GCL) neurons and identify ipRGCs based on spectral comparisons and/or the persistence of light responses under synaptic blockade.Results: Despite detecting robust cone-mediated responses across the retina, we find cone opponency is rare, especially outside of the central retina (overall ~3% of GCL neurons). In keeping with previous suggestions we also see some evidence of rod-cone opponency (albeit even more rare under our experimental conditions), but find no evidence for any enrichment of cone (or rod) opponent responses among functionally identified ipRGCs.Conclusion: In summary, these data suggest the widespread appearance of cone-opponency across the mouse early visual system and ipRGC-related responses may be an emergent feature of central visual processing mechanisms.
Collapse
Affiliation(s)
- Joshua W. Mouland
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- *Correspondence: Joshua W. Mouland
| | - Alex J. Watson
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Franck P. Martial
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Robert J. Lucas
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Timothy M. Brown
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
7
|
Pupillary Light Response Deficits in 4-Week-Old Piglets and Adolescent Children after Low-Velocity Head Rotations and Sports-Related Concussions. Biomedicines 2023; 11:biomedicines11020587. [PMID: 36831121 PMCID: PMC9952885 DOI: 10.3390/biomedicines11020587] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Neurological disorders and traumatic brain injury (TBI) are among the leading causes of death and disability. The pupillary light reflex (PLR) is an emerging diagnostic tool for concussion in humans. We compared PLR obtained with a commercially available pupillometer in the 4 week old piglet model of the adolescent brain subject to rapid nonimpact head rotation (RNR), and in human adolescents with and without sports-related concussion (SRC). The 95% PLR reference ranges (RR, for maximum and minimum pupil diameter, latency, and average and peak constriction velocities) were established in healthy piglets (N = 13), and response reliability was validated in nine additional healthy piglets. PLR assessments were obtained in female piglets allocated to anesthetized sham (N = 10), single (sRNR, N = 13), and repeated (rRNR, N = 14) sagittal low-velocity RNR at pre-injury, as well as days 1, 4, and 7 post injury, and evaluated against RRs. In parallel, we established human PLR RRs in healthy adolescents (both sexes, N = 167) and compared healthy PLR to values obtained <28 days from a SRC (N = 177). In piglets, maximum and minimum diameter deficits were greater in rRNR than sRNR. Alterations peaked on day 1 post sRNR and rRNR, and remained altered at day 4 and 7. In SRC adolescents, the proportion of adolescents within the RR was significantly lower for maximum pupil diameter only (85.8%). We show that PLR deficits may persist in humans and piglets after low-velocity head rotations. Differences in timing of assessment after injury, developmental response to injury, and the number and magnitude of impacts may contribute to the differences observed between species. We conclude that PLR is a feasible, quantifiable involuntary physiological metric of neurological dysfunction in pigs, as well as humans. Healthy PLR porcine and human reference ranges established can be used for neurofunctional assessments after TBI or hypoxic exposures (e.g., stroke, apnea, or cardiac arrest).
Collapse
|
8
|
Chakraborty R, Collins MJ, Kricancic H, Davis B, Alonso-Caneiro D, Yi F, Baskaran K. The effect of intrinsically photosensitive retinal ganglion cell (ipRGC) stimulation on axial length changes to imposed optical defocus in young adults. JOURNAL OF OPTOMETRY 2023; 16:53-63. [PMID: 35589503 PMCID: PMC9811374 DOI: 10.1016/j.optom.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/08/2022] [Accepted: 04/21/2022] [Indexed: 06/15/2023]
Abstract
PURPOSE The intrinsically photosensitive retinal ganglion cells (ipRGCs) regulate pupil size and circadian rhythms. Stimulation of the ipRGCs using short-wavelength blue light causes a sustained pupil constriction known as the post-illumination pupil response (PIPR). Here we examined the effects of ipRGC stimulation on axial length changes to imposed optical defocus in young adults. MATERIALS AND METHODS Nearly emmetropic young participants were given either myopic (+3 D, n = 16) or hyperopic (-3 D, n = 17) defocus in their right eye for 2 h. Before and after defocus, a series of axial length measurements for up to 180 s were performed in the right eye using the IOL Master following exposure to 5 s red (625 nm, 3.74 × 1014 photons/cm2/s) and blue (470 nm, 3.29 × 1014 photons/cm2/s) stimuli. The pupil measurements were collected from the left eye to track the ipRGC activity. The 6 s and 30 s PIPR, early and late area under the curve (AUC), and time to return to baseline were calculated. RESULTS The PIPR with blue light was significantly stronger after 2 h of hyperopic defocus as indicated by a lower 6 and 30 s PIPR and a larger early and late AUC (all p<0.05). Short-wavelength ipRGC stimulation also significantly exaggerated the ocular response to hyperopic defocus, causing a significantly greater increase in axial length than that resulting from the hyperopic defocus alone (p = 0.017). Neither wavelength had any effect on axial length with myopic defocus. CONCLUSIONS These findings suggest an interaction between myopiagenic hyperopic defocus and ipRGC signaling.
Collapse
Affiliation(s)
- Ranjay Chakraborty
- Caring Futures Institute, Flinders University, Bedford Park, SA 5042, Australia; College of Nursing and Health Sciences, Optometry and Vision Science, Sturt North, Flinders University, Bedford Park, SA 5042, Australia.
| | - Michael J Collins
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Victoria Park Road, Kelvin Grove 4059, Brisbane, QLD, Australia
| | - Henry Kricancic
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Victoria Park Road, Kelvin Grove 4059, Brisbane, QLD, Australia
| | - Brett Davis
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Victoria Park Road, Kelvin Grove 4059, Brisbane, QLD, Australia
| | - David Alonso-Caneiro
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Victoria Park Road, Kelvin Grove 4059, Brisbane, QLD, Australia
| | - Fan Yi
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Victoria Park Road, Kelvin Grove 4059, Brisbane, QLD, Australia
| | | |
Collapse
|
9
|
Differential susceptibility of retinal ganglion cell subtypes against neurodegenerative diseases. Graefes Arch Clin Exp Ophthalmol 2022; 260:1807-1821. [PMID: 35038014 DOI: 10.1007/s00417-022-05556-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/27/2021] [Accepted: 01/06/2022] [Indexed: 12/15/2022] Open
Abstract
Retinal ganglion cells (RGCs) are essential to propagate external visual information from the retina to the brain. Death of RGCs is speculated to be closely correlated with blinding retinal diseases, such as glaucoma and traumatic optic neuropathy (TON). Emerging innovative technologies have helped refine and standardize the classification of RGCs; at present, they are classified into more than 40 subpopulations in mammals. These RGC subtypes are identified by a combination of anatomical morphologies, electrophysiological functions, and genetic profiles. Increasing evidence suggests that neurodegenerative diseases do not collectively affect the RGCs. In fact, which RGC subtype exhibits the strongest or weakest susceptibility is hotly debated. Although a consensus has not yet been reached, it is certain that assorted RGCs display differential susceptibility against irreversible degeneration. Interestingly, a single RGC subtype can exhibit various vulnerabilities to optic nerve damage in diverse injury models. Thus, elucidating how susceptible RGC subtypes are to various injuries can protect vulnerable RGCs from damage and improve the possibility of preventing and treating visual impairment caused by neurodegenerative diseases. In this review, we summarize in detail the progress and status quo of research on the type-specific susceptibility of RGCs and point out current limitations and the possible directions for future research in this field.
Collapse
|
10
|
Chakraborty R, Landis EG, Mazade R, Yang V, Strickland R, Hattar S, Stone RA, Iuvone PM, Pardue MT. Melanopsin modulates refractive development and myopia. Exp Eye Res 2022; 214:108866. [PMID: 34838844 PMCID: PMC8792255 DOI: 10.1016/j.exer.2021.108866] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 01/03/2023]
Abstract
Myopia, or nearsightedness, is the most common form of refractive abnormality and is characterized by excessive ocular elongation in relation to ocular power. Retinal neurotransmitter signaling, including dopamine, is implicated in myopic ocular growth, but the visual pathways that initiate and sustain myopia remain unclear. Melanopsin-expressing retinal ganglion cells (mRGCs), which detect light, are important for visual function, and have connections with retinal dopamine cells. Here, we investigated how mRGCs influence normal and myopic refractive development using two mutant mouse models: Opn4-/- mice that lack functional melanopsin photopigments and intrinsic mRGC responses but still receive other photoreceptor-mediated input to these cells; and Opn4DTA/DTA mice that lack intrinsic and photoreceptor-mediated mRGC responses due to mRGC cell death. In mice with intact vision or form-deprivation, we measured refractive error, ocular properties including axial length and corneal curvature, and the levels of retinal dopamine and its primary metabolite, L-3,4-dihydroxyphenylalanine (DOPAC). Myopia was measured as a myopic shift, or the difference in refractive error between the form-deprived and contralateral eyes. We found that Opn4-/- mice had altered normal refractive development compared to Opn4+/+ wildtype mice, starting ∼4D more myopic but developing ∼2D greater hyperopia by 16 weeks of age. Consistent with hyperopia at older ages, 16 week-old Opn4-/- mice also had shorter eyes compared to Opn4+/+ mice (3.34 vs 3.42 mm). Opn4DTA/DTA mice, however, were more hyperopic than both Opn4+/+ and Opn4-/- mice across development ending with even shorter axial lengths. Despite these differences, both Opn4-/- and Opn4DTA/DTA mice had ∼2D greater myopic shifts in response to form-deprivation compared to Opn4+/+ mice. Furthermore, when vision was intact, dopamine and DOPAC levels were similar between Opn4-/- and Opn4+/+ mice, but higher in Opn4DTA/DTA mice, which differed with age. However, form-deprivation reduced retinal dopamine and DOAPC by ∼20% in Opn4-/- compared to Opn4+/+ mice but did not affect retinal dopamine and DOPAC in Opn4DTA/DTA mice. Lastly, systemically treating Opn4-/- mice with the dopamine precursor L-DOPA reduced their form-deprivation myopia by half compared to non-treated mice. Collectively our findings show that disruption of retinal melanopsin signaling alters the rate and magnitude of normal refractive development, yields greater susceptibility to form-deprivation myopia, and changes dopamine signaling. Our results suggest that mRGCs participate in the eye's response to myopigenic stimuli, acting partly through dopaminergic mechanisms, and provide a potential therapeutic target underling myopia progression. We conclude that proper mRGC function is necessary for correct refractive development and protection from myopia progression.
Collapse
Affiliation(s)
- Ranjay Chakraborty
- Department of Ophthalmology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta, GA, 30322, United States; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, United States; College of Nursing and Health Sciences, Optometry and Vision Science, Flinders University, Bedford Park, SA, 5001, Adelaide, Australia; Caring Futures Institute, Flinders University, Bedford Park, SA, 5042, Adelaide, Australia
| | - Erica G Landis
- Department of Ophthalmology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta, GA, 30322, United States; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, United States; Neuroscience Program, Emory University School of Medicine, 1365 Clifton Rd NE, Atlanta, GA, 30322, United States
| | - Reece Mazade
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, United States; Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr, Atlanta, GA, 30332, United States
| | - Victoria Yang
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, United States; Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr, Atlanta, GA, 30332, United States
| | - Ryan Strickland
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, United States; Neuroscience Program, Emory University School of Medicine, 1365 Clifton Rd NE, Atlanta, GA, 30322, United States
| | - Samer Hattar
- Section on Light and Circadian Rhythms, NIMH, NIH, 9000 Rockville Pike, Bethesda, MD, USA, 20892
| | - Richard A Stone
- Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - P Michael Iuvone
- Department of Ophthalmology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta, GA, 30322, United States; Department of Pharmacology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta, GA, 30322, United States
| | - Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, United States; Neuroscience Program, Emory University School of Medicine, 1365 Clifton Rd NE, Atlanta, GA, 30322, United States; Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr, Atlanta, GA, 30332, United States.
| |
Collapse
|
11
|
Recent Advances in Chronotherapy Targeting Respiratory Diseases. Pharmaceutics 2021; 13:pharmaceutics13122008. [PMID: 34959290 PMCID: PMC8704788 DOI: 10.3390/pharmaceutics13122008] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 11/17/2022] Open
Abstract
Respiratory diseases contribute to a significant percentage of mortality and morbidity worldwide. The circadian rhythm is a natural biological process where our bodily functions align with the 24 h oscillation (sleep-wake cycle) process and are controlled by the circadian clock protein/gene. Disruption of the circadian rhythm could alter normal lung function. Chronotherapy is a type of therapy provided at specific time intervals based on an individual's circadian rhythm. This would allow the drug to show optimum action, and thereby modulate its pharmacokinetics to lessen unwanted or unintended effects. In this review, we deliberated on the recent advances employed in chrono-targeted therapeutics for chronic respiratory diseases.
Collapse
|
12
|
Colman K, Andrews RN, Atkins H, Boulineau T, Bradley A, Braendli-Baiocco A, Capobianco R, Caudell D, Cline M, Doi T, Ernst R, van Esch E, Everitt J, Fant P, Gruebbel MM, Mecklenburg L, Miller AD, Nikula KJ, Satake S, Schwartz J, Sharma A, Shimoi A, Sobry C, Taylor I, Vemireddi V, Vidal J, Wood C, Vahle JL. International Harmonization of Nomenclature and Diagnostic Criteria (INHAND): Non-proliferative and Proliferative Lesions of the Non-human Primate ( M. fascicularis). J Toxicol Pathol 2021; 34:1S-182S. [PMID: 34712008 PMCID: PMC8544165 DOI: 10.1293/tox.34.1s] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The INHAND (International Harmonization of Nomenclature and Diagnostic Criteria for Lesions Project (www.toxpath.org/inhand.asp) is a joint initiative of the Societies of Toxicologic Pathology from Europe (ESTP), Great Britain (BSTP), Japan (JSTP) and North America (STP) to develop an internationally accepted nomenclature for proliferative and nonproliferative lesions in laboratory animals. The purpose of this publication is to provide a standardized nomenclature for classifying microscopic lesions observed in most tissues and organs from the nonhuman primate used in nonclinical safety studies. Some of the lesions are illustrated by color photomicrographs. The standardized nomenclature presented in this document is also available electronically on the internet (http://www.goreni.org/). Sources of material included histopathology databases from government, academia, and industrial laboratories throughout the world. Content includes spontaneous lesions as well as lesions induced by exposure to test materials. Relevant infectious and parasitic lesions are included as well. A widely accepted and utilized international harmonization of nomenclature for lesions in laboratory animals will provide a common language among regulatory and scientific research organizations in different countries and increase and enrich international exchanges of information among toxicologists and pathologists.
Collapse
Affiliation(s)
- Karyn Colman
- Novartis Institutes for BioMedical Research, Cambridge, MA,
USA
| | - Rachel N. Andrews
- Wake Forest School of Medicine, Department of Radiation
Oncology, Winston-Salem, NC, USA
| | - Hannah Atkins
- Penn State College of Medicine, Department of Comparative
Medicine, Hershey, PA, USA
| | | | - Alys Bradley
- Charles River Laboratories Edinburgh Ltd., Tranent,
Scotland, UK
| | - Annamaria Braendli-Baiocco
- Roche Pharma Research and Early Development, Pharmaceutical
Sciences, Roche Innovation Center Basel, Switzerland
| | - Raffaella Capobianco
- Janssen Research & Development, a Division of Janssen
Pharmaceutica NV, Beerse, Belgium
| | - David Caudell
- Department of Pathology, Section on Comparative Medicine,
Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Mark Cline
- Department of Pathology, Section on Comparative Medicine,
Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Takuya Doi
- LSIM Safety Institute Corporation, Ibaraki, Japan
| | | | | | - Jeffrey Everitt
- Department of Pathology, Duke University School of
Medicine, Durham, NC, USA
| | | | | | | | - Andew D. Miller
- Cornell University College of Veterinary Medicine, Ithaca,
NY, USA
| | | | - Shigeru Satake
- Shin Nippon Biomedical Laboratories, Ltd., Kagoshima and
Tokyo, Japan
| | | | - Alok Sharma
- Covance Laboratories, Inc., Madison, WI, USA
| | | | | | | | | | | | - Charles Wood
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT,
USA
| | | |
Collapse
|
13
|
Siqueira RC, Belíssimo LM, Pinho TS, Dourado LFN, Alves AP, de Paiva MRB, Ajero U, Cunha ADS. Short-Term Results of Photobiomodulation Using Light-Emitting Diode Light of 670 nm in Eyes with Age-Related Macular Degeneration. PHOTOBIOMODULATION PHOTOMEDICINE AND LASER SURGERY 2021; 39:581-586. [PMID: 34546108 DOI: 10.1089/photob.2021.0005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective: To evaluate the short-term result of retinal functional behavior in patients with dry age-related macular degeneration (AMD) corrected by photobiomodulation (PBM) with 670 nm light-emitting diode (LED) light. Materials and methods: Ten patients with dry AMD underwent a treatment consisting of nine PBM sessions with LED light of 670 nm with two cycles of 50 mW/cm2, producing 4 J/cm2 per dose in 88 sec. The studied eye was compared with the baseline (before therapy), and after nine PBM sessions, the following aspects were evaluated: best-corrected visual acuity (VA), retinal sensitivity, and characteristics of the correction area by the fundus automated perimetry using the Compass system. A functional and structural assessment of the retina was also performed using the multifocal electroretinography (ERG), optical coherence tomography (OCT), fluorescence retinography (FR), and autofluorescence (AF). All examinations were performed 1, 4, and 16 weeks after the therapy. The Chi-square and Student's t-tests were used for comparisons. The analyses followed the 95% confidence level (p-value ≤0.05). Results: The BCVA significantly improved, from an average of 1.1 to 0.98 LogMAR (p = 0.01). The visual field examination, according to the parameters of mean deviation, standard deviation, and index of deviation of background perimeter, showed a significant improvement of -12.6% to -10.6%, 10.54% to 9.89%, and 56% to 60%, respectively (p = 0.02, 0.03, and 0.02, respectively). No participant had an adverse effect during the follow-up period; neither did any participant experience abnormalities in OCT, ERG, FR, and AF findings. Conclusions: In this short-term study, the PBM technique in patients with dry AMD showed the potential to improve VA and macular perimetry without causing significant adverse events. A larger number of patients and a longer follow-up will be necessary to further assess the success of this technique in these patients.
Collapse
Affiliation(s)
- Rubens Camargo Siqueira
- Rubens Siqueira Research Center, São Jose do Rio Preto, Brazil.,Faculty of Medicine of São José do Rio Preto-FAMERP, São Jose do Rio Preto, Brazil
| | | | | | | | - Ana Paula Alves
- Physics Department, Institute of Exact Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Ubirajara Ajero
- Physics Department, Institute of Exact Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | |
Collapse
|
14
|
Evangelisti S, La Morgia C, Testa C, Manners DN, Brizi L, Bianchini C, Carbonelli M, Barboni P, Sadun AA, Tonon C, Carelli V, Vandewalle G, Lodi R. Brain functional MRI responses to blue light stimulation in Leber’s hereditary optic neuropathy. Biochem Pharmacol 2021; 191:114488. [DOI: 10.1016/j.bcp.2021.114488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/20/2022]
|
15
|
Brinks J, van Dijk EHC, Klaassen I, Schlingemann RO, Kielbasa SM, Emri E, Quax PHA, Bergen AA, Meijer OC, Boon CJF. Exploring the choroidal vascular labyrinth and its molecular and structural roles in health and disease. Prog Retin Eye Res 2021; 87:100994. [PMID: 34280556 DOI: 10.1016/j.preteyeres.2021.100994] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/04/2021] [Accepted: 07/07/2021] [Indexed: 12/14/2022]
Abstract
The choroid is a key player in maintaining ocular homeostasis and plays a role in a variety of chorioretinal diseases, many of which are poorly understood. Recent advances in the field of single-cell RNA sequencing have yielded valuable insights into the properties of choroidal endothelial cells (CECs). Here, we review the role of the choroid in various physiological and pathophysiological mechanisms, focusing on the role of CECs. We also discuss new insights regarding the phenotypic properties of CECs, CEC subpopulations, and the value of measuring transcriptomics in primary CEC cultures derived from post-mortem eyes. In addition, we discuss key phenotypic, structural, and functional differences that distinguish CECs from other endothelial cells such as retinal vascular endothelial cells. Understanding the specific clinical and molecular properties of the choroid will shed new light on the pathogenesis of the broad clinical range of chorioretinal diseases such as age-related macular degeneration, central serous chorioretinopathy and other diseases within the pachychoroid spectrum, uveitis, and diabetic choroidopathy. Although our knowledge is still relatively limited with respect to the clinical features and molecular pathways that underlie these chorioretinal diseases, we summarise new approaches and discuss future directions for gaining new insights into these sight-threatening diseases and highlight new therapeutic strategies such as pluripotent stem cell‒based technologies and gene therapy.
Collapse
Affiliation(s)
- J Brinks
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - E H C van Dijk
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - I Klaassen
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - R O Schlingemann
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Ophthalmology, University of Lausanne, Jules Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - S M Kielbasa
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, the Netherlands
| | - E Emri
- Department of Clinical Genetics, Section of Ophthalmogenetics, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - P H A Quax
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - A A Bergen
- Department of Clinical Genetics, Section of Ophthalmogenetics, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - O C Meijer
- Department of Medicine, Division of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, the Netherlands
| | - C J F Boon
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands; Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
16
|
Hannibal J. Comparative Neurology of Circadian Photoreception: The Retinohypothalamic Tract (RHT) in Sighted and Naturally Blind Mammals. Front Neurosci 2021; 15:640113. [PMID: 34054403 PMCID: PMC8160255 DOI: 10.3389/fnins.2021.640113] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/29/2021] [Indexed: 11/13/2022] Open
Abstract
The mammalian eye contains two systems for light perception: an image detecting system constituted primarily of the classical photoreceptors, rods and cones, and a non-image forming system (NIF) constituted of a small group of intrinsically photosensitive retinal ganglion cells driven by melanopsin (mRGCs). The mRGCs receive input from the outer retina and NIF mediates light entrainment of circadian rhythms, masking behavior, light induced inhibition of nocturnal melatonin secretion, pupillary reflex (PLR), and affect the sleep/wake cycle. This review focuses on the mammalian NIF and its anatomy in the eye as well as its neuronal projection to the brain. This pathway is known as the retinohypothalamic tract (RHT). The development and functions of the NIF as well as the knowledge gained from studying gene modified mice is highlighted. Furthermore, the similarities of the NIF between sighted (nocturnal and diurnal rodent species, monkeys, humans) and naturally blind mammals (blind mole rats Spalax ehrenbergi and the Iberian mole, Talpa occidentalis) are discussed in relation to a changing world where increasing exposure to artificial light at night (ALAN) is becoming a challenge for humans and animals in the modern society.
Collapse
Affiliation(s)
- Jens Hannibal
- Department of Clinical Biochemistry, Bispebjerg Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
Abstract
Melanopsin retinal ganglion cells (mRGCs) are the third class of retinal photoreceptors with unique anatomical, electrophysiological, and biological features. There are different mRGC subtypes with differential projections to the brain. These cells contribute to many nonimage-forming functions of the eye, the most relevant being the photoentrainment of circadian rhythms through the projections to the suprachiasmatic nucleus of the hypothalamus. Other relevant biological functions include the regulation of the pupillary light reflex, mood, alertness, and sleep, as well as a possible role in formed vision. The relevance of the mRGC-related pathways in the brain is highlighted by the role that the dysfunction and/or loss of these cells may play in affecting circadian rhythms and sleep in many neurodegenerative disorders including Alzheimer's, Parkinson's and Huntington's disease and in aging. Moreover, the occurrence of circadian dysfunction is a known risk factor for dementia. In this chapter, the anatomy, physiology, and functions of these cells as well as their resistance to neurodegeneration in mitochondrial optic neuropathies or their predilection to be lost in other neurodegenerative disorders will be discussed.
Collapse
|
18
|
Rezende MTC, Figueiredo BGD, de Souza Bonifácio TA, Santos N, Andrade MJO. Variability of chromatic visual sensitivity: discrimination according to daily shifts. BIOL RHYTHM RES 2020. [DOI: 10.1080/09291016.2020.1837563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Maria Thalita Cardoso Rezende
- Department of Psychology, Perception, Neurosciences and Behavior Laboratory, Federal University of Paraíba, João Pessoa, Brazil
| | | | | | - Natanael Santos
- Department of Psychology, Perception, Neurosciences and Behavior Laboratory, Federal University of Paraíba, João Pessoa, Brazil
| | - Michael Jackson Oliveirade Andrade
- Department of Psychology, Laboratory of Neuroscience, Chronobiology and Sleep Psychology, State University of Minas Gerais, Divinópolis, Brazil
| |
Collapse
|
19
|
Duda M, Domagalik A, Orlowska-Feuer P, Krzysztynska-Kuleta O, Beldzik E, Smyk MK, Stachurska A, Oginska H, Jeczmien-Lazur JS, Fafrowicz M, Marek T, Lewandowski MH, Sarna T. Melanopsin: From a small molecule to brain functions. Neurosci Biobehav Rev 2020; 113:190-203. [DOI: 10.1016/j.neubiorev.2020.03.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/29/2022]
|
20
|
Berg DJ, Kartheiser K, Leyrer M, Saali A, Berson DM. Transcriptomic Signatures of Postnatal and Adult Intrinsically Photosensitive Ganglion Cells. eNeuro 2019; 6:ENEURO.0022-19.2019. [PMID: 31387875 PMCID: PMC6712207 DOI: 10.1523/eneuro.0022-19.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 07/19/2019] [Accepted: 07/23/2019] [Indexed: 11/21/2022] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) are rare mammalian photoreceptors essential for non-image-forming vision functions, such as circadian photoentrainment and the pupillary light reflex. They comprise multiple subtypes distinguishable by morphology, physiology, projections, and levels of expression of melanopsin (Opn4), their photopigment. The molecular programs that distinguish ipRGCs from other ganglion cells and ipRGC subtypes from one another remain elusive. Here, we present comprehensive gene expression profiles of early postnatal and adult mouse ipRGCs purified from two lines of reporter mice that mark different sets of ipRGC subtypes. We find dozens of novel genes highly enriched in ipRGCs. We reveal that Rasgrp1 and Tbx20 are selectively expressed in subsets of ipRGCs, though these molecularly defined groups imperfectly match established ipRGC subtypes. We demonstrate that the ipRGCs regulating circadian photoentrainment are diverse at the molecular level. Our findings reveal unexpected complexity in gene expression patterns across mammalian ipRGC subtypes.
Collapse
Affiliation(s)
- Daniel J Berg
- Molecular Biology, Cellular Biology, and Biochemistry Program, Brown University, Providence, Rhode Island 02912
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912
| | | | - Megan Leyrer
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912
| | - Alexandra Saali
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912
| | - David M Berson
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912
| |
Collapse
|
21
|
Abstract
Detection and discrimination of spatial patterns is thought to originate with photoreception by rods and cones. Here, we investigated whether the inner-retinal photoreceptor melanopsin could represent a third origin for form vision. We developed a 4-primary visual display capable of presenting patterns differing in contrast for melanopsin vs cones, and generated spectrally distinct stimuli that were indistinguishable for cones (metamers) but presented contrast for melanopsin. Healthy observers could detect sinusoidal gratings formed by these metamers when presented in the peripheral retina at low spatial (≤0.8 cpd) and temporal (≤0.45 Hz) frequencies, and Michelson contrasts ≥14% for melanopsin. Metameric gratings became invisible at lower light levels (<1013 melanopsin photons cm−2 sr−1 s−1) when rods are more active. The addition of metameric increases in melanopsin contrast altered appearance of greyscale representations of coarse gratings and a range of everyday images. These data identify melanopsin as a new potential origin for aspects of spatial vision in humans. The perception of spatial patterns (form vision) is thought to rely on rod and cone cells in the retina. Here, the authors show that a third kind of retinal cell, melanopsin-expressing ganglion cells, can also detect form in humans, under particular conditions.
Collapse
|
22
|
La Morgia C, Carelli V, Carbonelli M. Melanopsin Retinal Ganglion Cells and Pupil: Clinical Implications for Neuro-Ophthalmology. Front Neurol 2018; 9:1047. [PMID: 30581410 PMCID: PMC6292931 DOI: 10.3389/fneur.2018.01047] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 11/19/2018] [Indexed: 02/02/2023] Open
Abstract
Melanopsin retinal ganglion cells (mRGCs) are intrinsically photosensitive RGCs that mediate many relevant non-image forming functions of the eye, including the pupillary light reflex, through the projections to the olivary pretectal nucleus. In particular, the post-illumination pupil response (PIPR), as evaluated by chromatic pupillometry, can be used as a reliable marker of mRGC function in vivo. In the last years, pupillometry has become a promising tool to assess mRGC dysfunction in various neurological and neuro-ophthalmological conditions. In this review we will present the most relevant findings of pupillometric studies in glaucoma, hereditary optic neuropathies, ischemic optic neuropathies, idiopathic intracranial hypertension, multiple sclerosis, Parkinson's disease, and mood disorders. The use of PIPR as a marker for mRGC function is also proposed for other neurodegenerative disorders in which circadian dysfunction is documented.
Collapse
Affiliation(s)
- Chiara La Morgia
- Unità Operativa Complessa Clinica Neurologica, IRCCS Istituto delle Scienze Neurologiche di Bologna, Ospedale Bellaria, Bologna, Italy.,Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Valerio Carelli
- Unità Operativa Complessa Clinica Neurologica, IRCCS Istituto delle Scienze Neurologiche di Bologna, Ospedale Bellaria, Bologna, Italy.,Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Michele Carbonelli
- Unità Operativa Complessa Clinica Neurologica, IRCCS Istituto delle Scienze Neurologiche di Bologna, Ospedale Bellaria, Bologna, Italy
| |
Collapse
|
23
|
Jiang Z, Yue WWS, Chen L, Sheng Y, Yau KW. Cyclic-Nucleotide- and HCN-Channel-Mediated Phototransduction in Intrinsically Photosensitive Retinal Ganglion Cells. Cell 2018; 175:652-664.e12. [PMID: 30270038 PMCID: PMC6203304 DOI: 10.1016/j.cell.2018.08.055] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 03/19/2018] [Accepted: 08/21/2018] [Indexed: 02/06/2023]
Abstract
Non-image-forming vision in mammals is mediated primarily by melanopsin-expressing, intrinsically photosensitive retinal ganglion cells (ipRGCs). In mouse M1-ipRGCs, by far the best-studied subtype, melanopsin activates PLCβ4 (phospholipase C-β4) to open TRPC6,7 channels, mechanistically similar to phototransduction in fly rhabdomeric (microvillous) photoreceptors. We report here that, surprisingly, mouse M4-ipRGCs rely on a different and hitherto undescribed melanopsin-driven, ciliary phototransduction mechanism involving cyclic nucleotide as the second messenger and HCN channels rather than CNG channels as the ion channel for phototransduction. Even more surprisingly, within an individual mouse M2-ipRGC, this HCN-channel-dependent, ciliary phototransduction pathway operates in parallel with the TRPC6,7-dependent rhabdomeric pathway. These findings reveal a complex heterogeneity in phototransduction among ipRGCs and, more importantly, break a general dogma about segregation of the two phototransduction motifs, likely with strong evolutionary implications.
Collapse
Affiliation(s)
- Zheng Jiang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Wendy W S Yue
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lujing Chen
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Neuroscience Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yanghui Sheng
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Neuroscience Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - King-Wai Yau
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
24
|
Katz B, Minke B. The Drosophila light-activated TRP and TRPL channels - Targets of the phosphoinositide signaling cascade. Prog Retin Eye Res 2018; 66:200-219. [DOI: 10.1016/j.preteyeres.2018.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/29/2018] [Accepted: 05/02/2018] [Indexed: 01/28/2023]
|
25
|
Chakraborty R, Ostrin LA, Nickla DL, Iuvone PM, Pardue MT, Stone RA. Circadian rhythms, refractive development, and myopia. Ophthalmic Physiol Opt 2018; 38:217-245. [PMID: 29691928 PMCID: PMC6038122 DOI: 10.1111/opo.12453] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/11/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE Despite extensive research, mechanisms regulating postnatal eye growth and those responsible for ametropias are poorly understood. With the marked recent increases in myopia prevalence, robust and biologically-based clinical therapies to normalize refractive development in childhood are needed. Here, we review classic and contemporary literature about how circadian biology might provide clues to develop a framework to improve the understanding of myopia etiology, and possibly lead to rational approaches to ameliorate refractive errors developing in children. RECENT FINDINGS Increasing evidence implicates diurnal and circadian rhythms in eye growth and refractive error development. In both humans and animals, ocular length and other anatomical and physiological features of the eye undergo diurnal oscillations. Systemically, such rhythms are primarily generated by the 'master clock' in the surpachiasmatic nucleus, which receives input from the intrinsically photosensitive retinal ganglion cells (ipRGCs) through the activation of the photopigment melanopsin. The retina also has an endogenous circadian clock. In laboratory animals developing experimental myopia, oscillations of ocular parameters are perturbed. Retinal signaling is now believed to influence refractive development; dopamine, an important neurotransmitter found in the retina, not only entrains intrinsic retinal rhythms to the light:dark cycle, but it also modulates refractive development. Circadian clocks comprise a transcription/translation feedback control mechanism utilizing so-called clock genes that have now been associated with experimental ametropias. Contemporary clinical research is also reviving ideas first proposed in the nineteenth century that light exposures might impact refraction in children. As a result, properties of ambient lighting are being investigated in refractive development. In other areas of medical science, circadian dysregulation is now thought to impact many non-ocular disorders, likely because the patterns of modern artificial lighting exert adverse physiological effects on circadian pacemakers. How, or if, such modern light exposures and circadian dysregulation contribute to refractive development is not known. SUMMARY The premise of this review is that circadian biology could be a productive area worthy of increased investigation, which might lead to the improved understanding of refractive development and improved therapeutic interventions.
Collapse
Affiliation(s)
- Ranjay Chakraborty
- College of Nursing and Health Sciences, Flinders University, Adelaide, Australia
| | | | | | | | - Machelle T. Pardue
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Decatur
| | - Richard A. Stone
- University of Pennsylvania School of Medicine, Philadelphia, USA
| |
Collapse
|
26
|
Andrade MJOD, Neto AC, Oliveira ARD, Santana JB, Santos NAD. Daily variation of visual sensitivity to luminance contrast: Effects of time of measurement and circadian typology. Chronobiol Int 2018; 35:996-1007. [PMID: 29565681 DOI: 10.1080/07420528.2018.1450753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
This study analyzed the fluctuation of the achromatic visual contrast sensitivity (CS) of adult males (M = 23.42 ± 2.6 years) during a daily period. Twenty-eight volunteers were divided into three groups according to circadian typology (CT): moderate morning (MM; n = 8); intermediate (I; n = 10) and moderate evening (ME; n = 10). The Pittsburgh Sleep Quality Index was used to evaluate sleep quality, and the Horne and Ostberg Morningness-Eveningness Questionnaire was used to measure CT. To measure CS, we used Metropsis software version 11.0 with vertical sinusoidal grids of 0.2, 0.6, 1, 3.1, 6.1, 8.8, 13.2 and 15.6 cycles per degree of visual angle (cpd). The stimuli were presented on a cathode ray tube (CRT) color video monitor with a 19-inch flat screen, a 1024 × 786 pixel resolution, a 100 Hz refresh rate and a photopic luminance of 39.6 cd/m2. It was inferred that there is a tendency for visual contrast to vary according to daily rhythmicity and CT, mainly for the median spatial frequencies (1.0 cpd, χ2 = 9.93, p < 0.05 and 3.1 cpd, χ2 = 10.33, p < 0.05) and high spatial frequencies (13.2 cpd, χ2 = 11.54, p < 0.05) of ME participants. ME participants had minimal visual contrast sensitivity during the morning shift and a progressive increase from afternoon to night.
Collapse
Affiliation(s)
| | - Armindo Campos Neto
- a Department of Psychology , Federal University of Paraíba , João Pessoa , Brazil
| | - Ana Raquel de Oliveira
- b Department of Psychology , Federal University of Campina Grande , Campina Grande , Brazil
| | | | | |
Collapse
|
27
|
Abstract
The eye is susceptible to adverse toxic effects by direct application, inadvertent ocular contact, or systemic exposure to chemicals or their metabolites. Although the albino rat is a less than ideal model for ocular toxicity studies, it has gained popularity for specific applications and may be the first species in which the ocular toxicity of a systemically administered xenobiotic becomes evident. This chapter reviews the embryology, anatomy, and physiology of the eye and associated glands and describes common nonneoplastic and neoplastic lesions encountered in laboratory rats.
Collapse
|
28
|
Johnson EN, Westbrook T, Shayesteh R, Chen EL, Schumacher JW, Fitzpatrick D, Field GD. Distribution and diversity of intrinsically photosensitive retinal ganglion cells in tree shrew. J Comp Neurol 2017; 527:328-344. [PMID: 29238991 DOI: 10.1002/cne.24377] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 12/03/2017] [Accepted: 12/04/2017] [Indexed: 12/24/2022]
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) mediate the pupillary light reflex, circadian entrainment, and may contribute to luminance and color perception. The diversity of ipRGCs varies from rodents to primates, suggesting differences in their contributions to retinal output. To further understand the variability in their organization and diversity across species, we used immunohistochemical methods to examine ipRGCs in tree shrew (Tupaia belangeri). Tree shrews share membership in the same clade, or evolutionary branch, as rodents and primates. They are highly visual, diurnal animals with a cone-dominated retina and a geniculo-cortical organization resembling that of primates. We identified cells with morphological similarities to M1 and M2 cells described previously in rodents and primates. M1-like cells typically had somas in the ganglion cell layer, with 23% displaced to the inner nuclear layer (INL). However, unlike M1 cells, they had bistratified dendritic fields ramifying in S1 and S5 that collectively tiled space. M2-like cells had dendritic fields restricted to S5 that were smaller and more densely branching. A novel third type of melanopsin immunopositive cell was identified. These cells had somata exclusively in the INL and monostratified dendritic fields restricted to S1 that tiled space. Surprisingly, these cells immunolabeled for tyrosine hydroxylase, a key component in dopamine synthesis. These cells immunolabeled for an RGC marker, not amacrine cell markers, suggesting that they are dopaminergic ipRGCs. We found no evidence for M4 or M5 ipRGCs, described previously in rodents. These results identify some organizational features of the ipRGC system that are canonical versus species-specific.
Collapse
Affiliation(s)
- Elizabeth N Johnson
- Neurobiology Department, Duke University School of Medicine, Durham, North Carolina.,Wharton Neuroscience Initiative, The Wharton School, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Teleza Westbrook
- Neurobiology Department, Duke University School of Medicine, Durham, North Carolina
| | - Rod Shayesteh
- Neurobiology Department, Duke University School of Medicine, Durham, North Carolina
| | - Emily L Chen
- Neurobiology Department, Duke University School of Medicine, Durham, North Carolina
| | | | | | - Greg D Field
- Neurobiology Department, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
29
|
Salari V, Scholkmann F, Vimal RLP, Császár N, Aslani M, Bókkon I. Phosphenes, retinal discrete dark noise, negative afterimages and retinogeniculate projections: A new explanatory framework based on endogenous ocular luminescence. Prog Retin Eye Res 2017; 60:101-119. [DOI: 10.1016/j.preteyeres.2017.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/13/2017] [Accepted: 07/15/2017] [Indexed: 02/07/2023]
|
30
|
Psychophysical Measurements of Luminance Contrast Sensitivity and Color Discrimination with Transparent and Blue-Light Filter Intraocular Lenses. Ophthalmol Ther 2017; 6:301-312. [PMID: 28795358 PMCID: PMC5693818 DOI: 10.1007/s40123-017-0101-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Indexed: 12/03/2022] Open
Abstract
Introduction The purpose of this study was to measure luminance contrast sensitivity and color vision thresholdfs in normal subjects using a blue light filter lens and transparent intraocular lens material. Methods Monocular luminance grating contrast sensitivity was measured with Psycho for Windows (version 2.36; Cambridge Research Systems) at 3.0, 6.0, 12.0, 20.0, and 30.0 cycles per degree of visual angle (cpd) in 15 normal subjects (eight female), with a mean age of 21.6 years (SD = 3.8 years). Chromatic discrimination was assessed with the Cambridge colour test (CCT) along the protan, deutan, and tritan color confusion axes. Both tests were performed in a darkened room under two situations: with a transparent lens and with blue light filter lens. Subjective impressions were taken by subjects regarding their visual experience under both conditions. Results No difference was found between the luminance contrast sensitivity measured with transparent and blue light filter. However, 13/15 (87%) of the subjects reported more comfortable vision with the blue filter. In the color vision test, tritan thresholds were significantly higher for the blue filter compared with the transparent filter (p = 0.003). For protan and deutan thresholds no differences were found. Conclusion Blue-yellow color vision is impaired with the blue light filter, and no impairment occurs with the transparent filter. No significant differences in thresholds were found in the luminance contrast sensitivity comparing the blue light and transparent filters. The impact of short wavelength light filtering on intrinsically photosensitive retinal ganglion cells is also discussed.
Collapse
|
31
|
Guo ZZ, Jiang SM, Zeng LP, Tang L, Li N, Xu ZP, Wei X. ipRGCs: possible causation accounts for the higher prevalence of sleep disorders in glaucoma patients. Int J Ophthalmol 2017; 10:1163-1167. [PMID: 28730123 DOI: 10.18240/ijo.2017.07.22] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 02/06/2017] [Indexed: 02/05/2023] Open
Abstract
Sleep accounts for a third of one's lifetime, partial or complete deprivation of sleep could elicit sever disorders of body function. Previous studies have reported the higher prevalence of sleep disorders in glaucoma patients, but the definite mechanism for this phenomenon is unknown. On the other hand, it is well known by us that the intrinsically photosensitive retinal ganglion cells (ipRGCs) serve additional ocular functions, called non-image-forming (NIF) functions, in the regulation of circadian rhythm, melatonin secretion, sleep, mood and others. Specifically, ipRGCs can directly or indirectly innervate the central areas such as suprachiasmatic nucleus (SCN), downstream pineal gland (the origin of melatonin), sleep and wake-inducing centers and mood regulation areas, making NIF functions of ipRGCs relate to sleep. The more interesting thing is that previous research showed glaucoma not only affected visual functions such as the degeneration of classical retinal ganglion cells (RGCs), but also affected ipRGCs. Therefore, we hypothesize that higher prevalence of sleep disorders in glaucoma patients maybe result from the underlying glaucomatous injuries of ipRGCs leading to the abnormalities of diverse NIF functions corresponding to sleep.
Collapse
Affiliation(s)
- Zhen-Zhen Guo
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Shan-Ming Jiang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Li-Ping Zeng
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Li Tang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ni Li
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Zhu-Ping Xu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xin Wei
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
32
|
Schwitzer T, Schwan R, Bubl E, Lalanne L, Angioi-Duprez K, Laprevote V. Looking into the brain through the retinal ganglion cells in psychiatric disorders: A review of evidences. Prog Neuropsychopharmacol Biol Psychiatry 2017; 76:155-162. [PMID: 28336492 DOI: 10.1016/j.pnpbp.2017.03.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/06/2017] [Accepted: 03/19/2017] [Indexed: 01/11/2023]
Abstract
Psychiatry and neuroscience research need novel approaches to indirectly investigate brain function. As the retina is an anatomical and developmental extension of the central nervous system (CNS), changes in retinal function may reflect neurological dysfunctions in psychiatric disorders. The last and most integrated retinal relay before visual information transfer to the brain is the ganglion cell layer. Here, based on collected arguments, we argue that these cells offer a crucial site for indirectly investigating brain function. We describe the anatomical and physiological properties of these cells together with measurements of their functional properties named pattern electroretinogram (PERG). Based on ganglion cell dysfunctions measured with PERG in neurological disorders, we argue for the relevance of studying ganglion cell function in psychiatric research. We review studies that have evaluated ganglion cell function in psychiatric and addictive disorders and discuss how changes in PERG measurements could be functional markers of pathophysiological mechanisms of psychiatric disorders.
Collapse
Affiliation(s)
- Thomas Schwitzer
- Pôle Hospitalo-Universitaire de Psychiatrie d'Adulte du Grand Nancy, Centre Psychothérapique de Nancy, Laxou, France; EA7298, INGRES, Université de Lorraine, Vandœuvre-lès-Nancy, France; INSERM U1114, Fédération de Médecine Translationnelle de Strasbourg, Département de Psychiatrie, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France.
| | - Raymund Schwan
- Pôle Hospitalo-Universitaire de Psychiatrie d'Adulte du Grand Nancy, Centre Psychothérapique de Nancy, Laxou, France; EA7298, INGRES, Université de Lorraine, Vandœuvre-lès-Nancy, France; Maison des Addictions, CHRU Nancy, Nancy, France
| | - Emanuel Bubl
- Saarland University Medical Center, Department for Psychiatry and Psychotherapy, Homburg, Germany
| | - Laurence Lalanne
- INSERM U1114, Fédération de Médecine Translationnelle de Strasbourg, Département de Psychiatrie, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France
| | | | - Vincent Laprevote
- Pôle Hospitalo-Universitaire de Psychiatrie d'Adulte du Grand Nancy, Centre Psychothérapique de Nancy, Laxou, France; EA7298, INGRES, Université de Lorraine, Vandœuvre-lès-Nancy, France; Maison des Addictions, CHRU Nancy, Nancy, France
| |
Collapse
|
33
|
García-Ayuso D, Galindo-Romero C, Di Pierdomenico J, Vidal-Sanz M, Agudo-Barriuso M, Villegas Pérez MP. Light-induced retinal degeneration causes a transient downregulation of melanopsin in the rat retina. Exp Eye Res 2017; 161:10-16. [PMID: 28552384 DOI: 10.1016/j.exer.2017.05.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/03/2017] [Accepted: 05/18/2017] [Indexed: 12/16/2022]
Abstract
In this work we study the effects of an acute light-induced retinal degeneration on the population of melanopsin positive retinal ganglion cells (m+RGCs) and the expression of the melanopsin protein in the retina. The m+RGCs may be more resistant than other RGCs to lesion, but the effects of an acute light exposure in this population are unknown. Albino rats were exposed to white light (3000 lux) continuously for 48 h and processed 0, 3, 7 or 30 days after light exposure (ALE). Whole-mounted retinas were immunodetected with antibodies against melanopsin, Brn3a, and rhodopsin to study the populations of m+RGC, Brn3a+RGC and rods (which are the most abundant photoreceptors in the rat retina). Three days ALE there was substantial rod loss in an arciform area of the superior retina and with time this loss expanded in the form of rings all throughout the retina. Light exposure did not affect the number of Brn3a+RGCs but diminished the numbers of m+RGCs. Immediately ALE there was a significant decrease in the mean number of immunodetected m+RGCs that was more marked in the superior retina. Later, the number of m+RGCs increased progressively and reached normal values one month ALE. Western blot analysis showed that melanopsin expression down-regulates shortly ALE and recovers thereafter, in accordance with the anatomical data. This study demonstrates that there is a transient downregulation of melanopsin expression in the RGCs during the first month ALE. Further studies would be needed to clarify the long-term effect of light exposure on the m+RGC population.
Collapse
Affiliation(s)
- Diego García-Ayuso
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria Hospital Virgen de la Arrixaca (IMIB-Virgen de la Arrixaca), Spain.
| | - Caridad Galindo-Romero
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria Hospital Virgen de la Arrixaca (IMIB-Virgen de la Arrixaca), Spain
| | - Johnny Di Pierdomenico
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria Hospital Virgen de la Arrixaca (IMIB-Virgen de la Arrixaca), Spain
| | - Manuel Vidal-Sanz
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria Hospital Virgen de la Arrixaca (IMIB-Virgen de la Arrixaca), Spain
| | - Marta Agudo-Barriuso
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria Hospital Virgen de la Arrixaca (IMIB-Virgen de la Arrixaca), Spain
| | - María P Villegas Pérez
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria Hospital Virgen de la Arrixaca (IMIB-Virgen de la Arrixaca), Spain.
| |
Collapse
|
34
|
Nasir-Ahmad S, Lee SCS, Martin PR, Grünert U. Melanopsin-expressing ganglion cells in human retina: Morphology, distribution, and synaptic connections. J Comp Neurol 2017; 527:312-327. [PMID: 28097654 DOI: 10.1002/cne.24176] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 11/05/2022]
Abstract
Melanopsin-expressing retinal ganglion cells are intrinsically photosensitive cells that are involved in non-image forming visual processes such as the pupillary light reflex and circadian entrainment but also contribute to visual perception. Here we used immunohistochemistry to study the morphology, density, distribution, and synaptic connectivity of melanopsin-expressing ganglion cells in four post mortem human donor retinas. Two types of melanopsin-expressing ganglion cells were distinguished based on their dendritic stratification near either the outer or the inner border of the inner plexiform layer. Outer stratifying cells make up on average 60% of the melanopsin-expressing cells. About half of the melanopsin-expressing cells (or 80% of the outer stratifying cells) have their soma displaced to the inner nuclear layer. Inner stratifying cells have their soma exclusively in the ganglion cell layer and include a small proportion of bistratified cells. The dendritic field diameter of melanopsin-expressing cells ranges from 250 (near the fovea) to 1,000 µm in peripheral retina. The dendritic trees of outer stratifying cells cover the retina independent of soma location. The dendritic fields of both outer and inner stratifying cells show a high degree of overlap with a coverage factor of approximately two. Melanopsin-expressing cells occur at an average peak density of between ∼20 and ∼40 cells/mm2 at about 2 mm eccentricity, the density drops to below ∼10 cells/mm2 at about 8 mm eccentricity. Both the outer and inner stratifying dendrites express postsynaptic density (PSD95) immunoreactive puncta suggesting that they receive synaptic input from bipolar cells.
Collapse
Affiliation(s)
- Subha Nasir-Ahmad
- Save Sight Institute and Department of Clinical Ophthalmology, The University of Sydney, Sydney, New South Wales, 2000, Australia
| | - Sammy C S Lee
- Save Sight Institute and Department of Clinical Ophthalmology, The University of Sydney, Sydney, New South Wales, 2000, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, The University of Sydney, Sydney, New South Wales, 2000, Australia
| | - Paul R Martin
- Save Sight Institute and Department of Clinical Ophthalmology, The University of Sydney, Sydney, New South Wales, 2000, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, The University of Sydney, Sydney, New South Wales, 2000, Australia.,School of Medical Sciences, The University of Sydney, Sydney, New South Wales, 2000, Australia
| | - Ulrike Grünert
- Save Sight Institute and Department of Clinical Ophthalmology, The University of Sydney, Sydney, New South Wales, 2000, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, The University of Sydney, Sydney, New South Wales, 2000, Australia.,School of Medical Sciences, The University of Sydney, Sydney, New South Wales, 2000, Australia
| |
Collapse
|
35
|
Sabbah S, Berg D, Papendorp C, Briggman KL, Berson DM. A Cre Mouse Line for Probing Irradiance- and Direction-Encoding Retinal Networks. eNeuro 2017; 4:ENEURO.0065-17.2017. [PMID: 28466070 PMCID: PMC5411164 DOI: 10.1523/eneuro.0065-17.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/31/2017] [Accepted: 04/10/2017] [Indexed: 01/18/2023] Open
Abstract
Cell type-specific Cre driver lines have revolutionized the analysis of retinal cell types and circuits. We show that the transgenic mouse Rbp4-Cre selectively labels several retinal neuronal types relevant to the encoding of absolute light intensity (irradiance) and visual motion. In the ganglion cell layer (GCL), most marked cells are wide-field spiking polyaxonal amacrine cells (ACs) with sustained irradiance-encoding ON responses that persist during chemical synaptic blockade. Their arbors spread about 1 mm across the retina and are restricted to the inner half of the ON sublamina of the inner plexiform layer (IPL). There, they costratify with dendrites of M2 intrinsically photosensitive retinal ganglion cells (ipRGCs), to which they are tracer coupled. We propose that synaptically driven and intrinsic photocurrents of M2 cells pass through gap junctions to drive AC light responses. Also marked in this mouse are two types of RGCs. R-cells have a bistratified dendritic arbor, weak directional tuning, and irradiance-encoding ON responses. However, they also receive excitatory OFF input, revealed during ON-channel blockade. Serial blockface electron microscopic (SBEM) reconstruction confirms OFF bipolar input, and reveals that some OFF input derives from a novel type of OFF bipolar cell (BC). R-cells innervate specific layers of the dorsal lateral geniculate nucleus (dLGN) and superior colliculus (SC). The other marked RGC type (RDS) is bistratified, transient, and ON-OFF direction selective (DS). It apparently innervates the nucleus of the optic tract (NOT). The Rbp4-Cre mouse will be valuable for targeting these cell types for further study and for selectively manipulating them for circuit analysis.
Collapse
Affiliation(s)
- Shai Sabbah
- Department of Neuroscience, Brown University, Providence, RI 02912
| | - Daniel Berg
- Department of Neuroscience, Brown University, Providence, RI 02912
| | - Carin Papendorp
- Department of Neuroscience, Brown University, Providence, RI 02912
| | - Kevin L. Briggman
- National Institute for Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - David M. Berson
- Department of Neuroscience, Brown University, Providence, RI 02912
| |
Collapse
|
36
|
Agudo-Barriuso M, Nadal-Nicolás FM, Madeira MH, Rovere G, Vidal-Villegas B, Vidal-Sanz M. Melanopsin expression is an indicator of the well-being of melanopsin-expressing retinal ganglion cells but not of their viability. Neural Regen Res 2016; 11:1243-4. [PMID: 27651769 PMCID: PMC5020820 DOI: 10.4103/1673-5374.189182] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Marta Agudo-Barriuso
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Francisco M Nadal-Nicolás
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - María H Madeira
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Giuseppe Rovere
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Beatriz Vidal-Villegas
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Manuel Vidal-Sanz
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| |
Collapse
|
37
|
Yan SS, Wang W. The effect of lens aging and cataract surgery on circadian rhythm. Int J Ophthalmol 2016; 9:1066-74. [PMID: 27500118 DOI: 10.18240/ijo.2016.07.21] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 02/14/2016] [Indexed: 12/31/2022] Open
Abstract
Many organisms have evolved an approximately 24-hour circadian rhythm that allows them to achieve internal physiological homeostasis with external environment. Suprachiasmatic nucleus (SCN) is the central pacemaker of circadian rhythm, and its activity is entrained to the external light-dark cycle. The SCN controls circadian rhythm through regulating the synthesis of melatonin by pineal gland via a multisynaptic pathway. Light, especially short-wavelength blue light, is the most potent environmental time cue in circadian photoentrainment. Recently, the discovery of a novel type of retinal photoreceptors, intrinsically photosensitive retinal ganglion cells, sheds light on the mechanism of circadian photoentrainment and raises concerns about the effect of ocular diseases on circadian system. With age, light transmittance is significantly decreased due to the aging of crystalline lens, thus possibly resulting in progressive loss of circadian photoreception. In the current review, we summarize the circadian physiology, highlight the important role of light in circadian rhythm regulation, discuss about the correlation between age-related cataract and sleep disorders, and compare the effect of blue light- filtering intraocular lenses (IOLs) and ultraviolet only filtering IOLs on circadian rhythm.
Collapse
Affiliation(s)
- Shen-Shen Yan
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China
| | - Wei Wang
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
38
|
Meyer A, Tetenborg S, Greb H, Segelken J, Dorgau B, Weiler R, Hormuzdi SG, Janssen-Bienhold U, Dedek K. Connexin30.2: In Vitro Interaction with Connexin36 in HeLa Cells and Expression in AII Amacrine Cells and Intrinsically Photosensitive Ganglion Cells in the Mouse Retina. Front Mol Neurosci 2016; 9:36. [PMID: 27303262 PMCID: PMC4882342 DOI: 10.3389/fnmol.2016.00036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 05/09/2016] [Indexed: 11/13/2022] Open
Abstract
Electrical coupling via gap junctions is an abundant phenomenon in the mammalian retina and occurs in all major cell types. Gap junction channels are assembled from different connexin subunits, and the connexin composition of the channel confers specific properties to the electrical synapse. In the mouse retina, gap junctions were demonstrated between intrinsically photosensitive ganglion cells and displaced amacrine cells but the underlying connexin remained undetermined. In the primary rod pathway, gap junctions play a crucial role, coupling AII amacrine cells among each other and to ON cone bipolar cells. Although it has long been known that connexin36 and connexin45 are necessary for the proper functioning of this most sensitive rod pathway, differences between homocellular AII/AII gap junctions and AII/ON bipolar cell gap junctions suggested the presence of an additional connexin in AII amacrine cells. Here, we used a connexin30.2-lacZ mouse line to study the expression of connexin30.2 in the retina. We show that connexin30.2 is expressed in intrinsically photosensitive ganglion cells and AII amacrine cells. Moreover, we tested whether connexin30.2 and connexin36-both expressed in AII amacrine cells-are able to interact with each other and are deposited in the same gap junctional plaques. Using newly generated anti-connexin30.2 antibodies, we show in HeLa cells that both connexins are indeed able to interact and may form heteromeric channels: both connexins were co-immunoprecipitated from transiently transfected HeLa cells and connexin30.2 gap junction plaques became significantly larger when co-expressed with connexin36. These data suggest that connexin36 is able to form heteromeric gap junctions with another connexin. We hypothesize that co-expression of connexin30.2 and connexin36 may endow AII amacrine cells with the means to differentially regulate its electrical coupling to different synaptic partners.
Collapse
Affiliation(s)
- Arndt Meyer
- Department of Neuroscience and Neurobiology, University of Oldenburg Oldenburg, Germany
| | - Stephan Tetenborg
- Department of Neuroscience and Neurobiology, University of Oldenburg Oldenburg, Germany
| | - Helena Greb
- Department of Neuroscience and Neurobiology, University of Oldenburg Oldenburg, Germany
| | - Jasmin Segelken
- Department of Neuroscience and Neurobiology, University of Oldenburg Oldenburg, Germany
| | - Birthe Dorgau
- Department of Neuroscience and Neurobiology, University of Oldenburg Oldenburg, Germany
| | - Reto Weiler
- Department of Neuroscience and Neurobiology, University of OldenburgOldenburg, Germany; Research Center Neurosensory Science, University of OldenburgOldenburg, Germany
| | | | - Ulrike Janssen-Bienhold
- Department of Neuroscience and Neurobiology, University of OldenburgOldenburg, Germany; Research Center Neurosensory Science, University of OldenburgOldenburg, Germany
| | - Karin Dedek
- Department of Neuroscience and Neurobiology, University of OldenburgOldenburg, Germany; Research Center Neurosensory Science, University of OldenburgOldenburg, Germany
| |
Collapse
|
39
|
Shirzad-Wasei N, DeGrip WJ. Heterologous expression of melanopsin: Present, problems and prospects. Prog Retin Eye Res 2016; 52:1-21. [DOI: 10.1016/j.preteyeres.2016.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 01/25/2016] [Accepted: 02/01/2016] [Indexed: 12/12/2022]
|
40
|
Ohlemacher SK, Sridhar A, Xiao Y, Hochstetler AE, Sarfarazi M, Cummins TR, Meyer JS. Stepwise Differentiation of Retinal Ganglion Cells from Human Pluripotent Stem Cells Enables Analysis of Glaucomatous Neurodegeneration. Stem Cells 2016; 34:1553-62. [PMID: 26996528 DOI: 10.1002/stem.2356] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 12/18/2015] [Accepted: 01/06/2016] [Indexed: 12/14/2022]
Abstract
Human pluripotent stem cells (hPSCs), including both embryonic and induced pluripotent stem cells, possess the unique ability to readily differentiate into any cell type of the body, including cells of the retina. Although previous studies have demonstrated the ability to differentiate hPSCs to a retinal lineage, the ability to derive retinal ganglion cells (RGCs) from hPSCs has been complicated by the lack of specific markers with which to identify these cells from a pluripotent source. In the current study, the definitive identification of hPSC-derived RGCs was accomplished by their directed, stepwise differentiation through an enriched retinal progenitor intermediary, with resultant RGCs expressing a full complement of associated features and proper functional characteristics. These results served as the basis for the establishment of induced pluripotent stem cells (iPSCs) from a patient with a genetically inherited form of glaucoma, which results in damage and loss of RGCs. Patient-derived RGCs specifically exhibited a dramatic increase in apoptosis, similar to the targeted loss of RGCs in glaucoma, which was significantly rescued by the addition of candidate neuroprotective factors. Thus, the current study serves to establish a method by which to definitively acquire and identify RGCs from hPSCs and demonstrates the ability of hPSCs to serve as an effective in vitro model of disease progression. Moreover, iPSC-derived RGCs can be utilized for future drug screening approaches to identify targets for the treatment of glaucoma and other optic neuropathies. Stem Cells 2016;34:1553-1562.
Collapse
Affiliation(s)
- Sarah K Ohlemacher
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Akshayalakshmi Sridhar
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Yucheng Xiao
- Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, USA
| | - Alexandra E Hochstetler
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Mansoor Sarfarazi
- Molecular Ophthalmic Genetics Laboratory, University of Connecticut Health Center, Farmington, CT, USA
| | - Theodore R Cummins
- Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA
| | - Jason S Meyer
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA.,Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, USA.,Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
41
|
Daneault V, Dumont M, Massé É, Vandewalle G, Carrier J. Light-sensitive brain pathways and aging. J Physiol Anthropol 2016; 35:9. [PMID: 26980095 PMCID: PMC4791759 DOI: 10.1186/s40101-016-0091-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/01/2016] [Indexed: 01/08/2023] Open
Abstract
Notwithstanding its effects on the classical visual system allowing image formation, light acts upon several non-image-forming (NIF) functions including body temperature, hormonal secretions, sleep-wake cycle, alertness, and cognitive performance. Studies have shown that NIF functions are maximally sensitive to blue wavelengths (460–480 nm), in comparison to longer light wavelengths. Higher blue light sensitivity has been reported for melatonin suppression, pupillary constriction, vigilance, and performance improvement but also for modulation of cognitive brain functions. Studies investigating acute stimulating effects of light on brain activity during the execution of cognitive tasks have suggested that brain activations progress from subcortical regions involved in alertness, such as the thalamus, the hypothalamus, and the brainstem, before reaching cortical regions associated with the ongoing task. In the course of aging, lower blue light sensitivity of some NIF functions has been reported. Here, we first describe neural pathways underlying effects of light on NIF functions and we discuss eye and cerebral mechanisms associated with aging which may affect NIF light sensitivity. Thereafter, we report results of investigations on pupillary constriction and cognitive brain sensitivity to light in the course of aging. Whereas the impact of light on cognitive brain responses appears to decrease substantially, pupillary constriction seems to remain more intact over the lifespan. Altogether, these results demonstrate that aging research should take into account the diversity of the pathways underlying the effects of light on specific NIF functions which may explain their differences in light sensitivity.
Collapse
Affiliation(s)
- V Daneault
- Functional Neuroimaging Unit, University of Montreal Geriatric Institute, Montreal, QC, Canada. .,Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montreal, QC, Canada. .,Department of Psychology, University of Montreal, Montreal, QC, Canada.
| | - M Dumont
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montreal, QC, Canada
| | - É Massé
- Functional Neuroimaging Unit, University of Montreal Geriatric Institute, Montreal, QC, Canada.,Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montreal, QC, Canada
| | - G Vandewalle
- Department of Psychology, University of Montreal, Montreal, QC, Canada.,Cyclotron Research Centre, University of Liège, Liège, Belgium
| | - J Carrier
- Functional Neuroimaging Unit, University of Montreal Geriatric Institute, Montreal, QC, Canada.,Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montreal, QC, Canada.,Department of Psychology, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
42
|
Light therapy and chronobiology in critical illness. THE LANCET RESPIRATORY MEDICINE 2016; 4:167-8. [PMID: 26895653 DOI: 10.1016/s2213-2600(16)00055-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 02/02/2016] [Indexed: 11/23/2022]
|
43
|
Hannibal J, Georg B, Fahrenkrug J. Altered Circadian Food Anticipatory Activity Rhythms in PACAP Receptor 1 (PAC1) Deficient Mice. PLoS One 2016; 11:e0146981. [PMID: 26757053 PMCID: PMC4710526 DOI: 10.1371/journal.pone.0146981] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/23/2015] [Indexed: 11/19/2022] Open
Abstract
Light signals from intrinsically photosensitive retinal ganglion cells (ipRGCs) entrain the circadian clock and regulate negative masking. Two neurotransmitters, glutamate and Pituitary Adenylate Cyclase Activating Polypeptide (PACAP), found in the ipRGCs transmit light signals to the brain via glutamate receptors and the specific PACAP type 1 (PAC1) receptor. Light entrainment occurs during the twilight zones and has little effect on clock phase during daytime. When nocturnal animals have access to food only for a few hours during the resting phase at daytime, they adapt behavior to the restricted feeding (RF) paradigm and show food anticipatory activity (FAA). A recent study in mice and rats demonstrating that light regulates FAA prompted us to investigate the role of PACAP/PAC1 signaling in the light mediated regulation of FAA. PAC1 receptor knock out (PAC1-/-) and wild type (PAC1+/+) mice placed in running wheels were examined in a full photoperiod (FPP) of 12:12 h light/dark (LD) and a skeleton photoperiod (SPP) 1:11:1:11 h L:DD:L:DD at 300 and 10 lux light intensity. Both PAC1-/- mice and PAC1+/+ littermates entrained to FPP and SPP at both light intensities. However, when placed in RF with access to food for 4–5 h during the subjective day, a significant change in behavior was observed in PAC1-/- mice compared to PAC1+/+ mice. While PAC1-/- mice showed similar FAA as PAC1+/+ animals in FPP at 300 lux, PAC1-/- mice demonstrated an advanced onset of FAA with a nearly 3-fold increase in amplitude compared to PAC1+/+ mice when placed in SPP at 300 lux. The same pattern of FAA was observed at 10 lux during both FPP and SPP. The present study indicates a role of PACAP/PAC1 signaling during light regulated FAA. Most likely, PACAP found in ipRGCs mediating non-image forming light information to the brain is involved.
Collapse
Affiliation(s)
- Jens Hannibal
- Department of Clinical Biochemistry, Faculty of Health Sciences, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| | - Birgitte Georg
- Department of Clinical Biochemistry, Faculty of Health Sciences, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jan Fahrenkrug
- Department of Clinical Biochemistry, Faculty of Health Sciences, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
44
|
La Morgia C, Ross-Cisneros FN, Koronyo Y, Hannibal J, Gallassi R, Cantalupo G, Sambati L, Pan BX, Tozer KR, Barboni P, Provini F, Avanzini P, Carbonelli M, Pelosi A, Chui H, Liguori R, Baruzzi A, Koronyo-Hamaoui M, Sadun AA, Carelli V. Melanopsin retinal ganglion cell loss in Alzheimer disease. Ann Neurol 2015; 79:90-109. [PMID: 26505992 PMCID: PMC4737313 DOI: 10.1002/ana.24548] [Citation(s) in RCA: 279] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 10/16/2015] [Accepted: 10/16/2015] [Indexed: 12/24/2022]
Abstract
Objective Melanopsin retinal ganglion cells (mRGCs) are photoreceptors driving circadian photoentrainment, and circadian dysfunction characterizes Alzheimer disease (AD). We investigated mRGCs in AD, hypothesizing that they contribute to circadian dysfunction. Methods We assessed retinal nerve fiber layer (RNFL) thickness by optical coherence tomography (OCT) in 21 mild‐moderate AD patients, and in a subgroup of 16 we evaluated rest–activity circadian rhythm by actigraphy. We studied postmortem mRGCs by immunohistochemistry in retinas, and axons in optic nerve cross‐sections of 14 neuropathologically confirmed AD patients. We coimmunostained for retinal amyloid β (Aβ) deposition and melanopsin to locate mRGCs. All AD cohorts were compared with age‐matched controls. Results We demonstrated an age‐related optic neuropathy in AD by OCT, with a significant reduction of RNFL thickness (p = 0.038), more evident in the superior quadrant (p = 0.006). Axonal loss was confirmed in postmortem AD optic nerves. Abnormal circadian function characterized only a subgroup of AD patients. Sleep efficiency was significantly reduced in AD patients (p = 0.001). We also found a significant loss of mRGCs in postmortem AD retinal specimens (p = 0.003) across all ages and abnormal mRGC dendritic morphology and size (p = 0.003). In flat‐mounted AD retinas, Aβ accumulation was remarkably evident inside and around mRGCs. Interpretation We show variable degrees of rest–activity circadian dysfunction in AD patients. We also demonstrate age‐related loss of optic nerve axons and specifically mRGC loss and pathology in postmortem AD retinal specimens, associated with Aβ deposition. These results all support the concept that mRGC degeneration is a contributor to circadian rhythm dysfunction in AD. ANN NEUROL 2016;79:90–109
Collapse
Affiliation(s)
- Chiara La Morgia
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy
- Neurology Unit, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | | | - Yosef Koronyo
- Department of Neurosurgery and Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jens Hannibal
- Department of Clinical Biochemistry, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Roberto Gallassi
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy
- Neurology Unit, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Gaetano Cantalupo
- Child Neuropsychiatry, Department of Life and Reproduction Sciences, University of Verona, Verona, Italy
| | - Luisa Sambati
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy
- Neurology Unit, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Billy X Pan
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Kevin R Tozer
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | | | - Federica Provini
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy
- Neurology Unit, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Pietro Avanzini
- Department of Neurosciences, University of Parma, Parma, Italy
| | - Michele Carbonelli
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy
| | - Annalisa Pelosi
- Department of Neurosciences, University of Parma, Parma, Italy
| | - Helena Chui
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Rocco Liguori
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy
- Neurology Unit, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Agostino Baruzzi
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy
- Neurology Unit, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery and Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Alfredo A Sadun
- Doheny Eye Institute, Los Angeles, CA
- Department of Ophthalmology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA
| | - Valerio Carelli
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy
- Neurology Unit, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
45
|
Abstract
There is a growing recognition that the coordinated timing of behavioral, physiologic, and metabolic circadian rhythms is a requirement for a healthy body and mind. In mammals, the primary circadian oscillator is the hypothalamic suprachiasmatic nucleus (SCN), which is responsible for circadian coordination throughout the organism. Temporal homeostasis is recognized as a complex interplay between rhythmic clock gene expression in brain regions outside the SCN and in peripheral organs. Abnormalities in this intricate circadian orchestration may alter sleep patterns and contribute to the pathophysiology of affective disorders.
Collapse
|
46
|
Münch M, Léon L, Collomb S, Kawasaki A. Comparison of acute non-visual bright light responses in patients with optic nerve disease, glaucoma and healthy controls. Sci Rep 2015; 5:15185. [PMID: 26478261 PMCID: PMC4609937 DOI: 10.1038/srep15185] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 09/22/2015] [Indexed: 11/30/2022] Open
Abstract
This study examined the effect of optic nerve disease, hence retinal ganglion cell loss, on non-visual functions related to melanopsin signalling. Test subjects were patients with bilateral visual loss and optic atrophy from either hereditary optic neuropathy (n = 11) or glaucoma (n = 11). We measured melatonin suppression, subjective sleepiness and cognitive functions in response to bright light exposure in the evening. We also quantified the post-illumination pupil response to a blue light stimulus. All results were compared to age-matched controls (n = 22). Both groups of patients showed similar melatonin suppression when compared to their controls. Greater melatonin suppression was intra-individually correlated to larger post-illumination pupil response in patients and controls. Only the glaucoma patients demonstrated a relative attenuation of their pupil response. In addition, they were sleepier with slower reaction times during nocturnal light exposure. In conclusion, glaucomatous, but not hereditary, optic neuropathy is associated with reduced acute light effects. At mild to moderate stages of disease, this is detected only in the pupil function and not in responses conveyed via the retinohypothalamic tract such as melatonin suppression.
Collapse
Affiliation(s)
- M Münch
- Solar Energy and Building Physics Laboratory, Environmental and Civil Engineering Institute, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - L Léon
- University of Lausanne, Hôpital Ophtalmique Jules-Gonin, Lausanne, Switzerland
| | - S Collomb
- University of Lausanne, Hôpital Ophtalmique Jules-Gonin, Lausanne, Switzerland
| | - A Kawasaki
- University of Lausanne, Hôpital Ophtalmique Jules-Gonin, Lausanne, Switzerland
| |
Collapse
|
47
|
Jeong MJ, Jeon CJ. Localization of melanopsin-immunoreactive cells in the Mongolian gerbil retina. Neurosci Res 2015; 100:6-16. [PMID: 26083722 DOI: 10.1016/j.neures.2015.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/19/2015] [Accepted: 06/04/2015] [Indexed: 12/20/2022]
Abstract
Melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs) are involved in circadian rhythm and pupil responses. The purpose of this study was to reveal the organization of melanopsin-immunoreactive (IR) neurons in the Mongolian gerbil retina using immunocytochemistry. Melanopsin-IR cells were primarily located in the ganglion cell layer (GCL; M1c; 75.15%). Many melanopsin-IR cells were also observed in the inner nuclear layer (INL; M1d; 22.28%). The M1c and M1d cell types extended their dendritic processes into the OFF sublayer of the inner plexiform layer (IPL). We rarely observed bistratified cells (M3; 2.56%) with dendrites in both the ON and OFF sublayers of the IPL. Surprisingly, we did not observe M2 cells which are well observed in other rodents. Melanopsin-IR cell somas were small to medium in size and had large dendritic fields. They had 2-5 primary dendrites that branched sparingly and had varicosities. Melanopsin-IR cell density was very low: they comprised 0.50% of the total ganglion cell population. Moreover, none of the melanopsin-IR cells expressed calbindin-D28K, calretinin, or parvalbumin. These results suggest that in the Mongolian gerbil, melanopsin-IR cells are expressed in a very small RGC subpopulation, and are independent of calcium-binding proteins-containing RGCs.
Collapse
Affiliation(s)
- Mi-Jin Jeong
- Department of Biology, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, and Brain Science and Engineering Institute, Kyungpook National University, Daegu 702-701, South Korea
| | - Chang-Jin Jeon
- Department of Biology, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, and Brain Science and Engineering Institute, Kyungpook National University, Daegu 702-701, South Korea.
| |
Collapse
|
48
|
Co-expression of two subtypes of melatonin receptor on rat M1-type intrinsically photosensitive retinal ganglion cells. PLoS One 2015; 10:e0117967. [PMID: 25714375 PMCID: PMC4340921 DOI: 10.1371/journal.pone.0117967] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 12/15/2014] [Indexed: 12/15/2022] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) are involved in circadian and other non-image forming visual responses. An open question is whether the activity of these neurons may also be under the regulation mediated by the neurohormone melatonin. In the present work, by double-staining immunohistochemical technique, we studied the expression of MT1 and MT2, two known subtypes of mammalian melatonin receptors, in rat ipRGCs. A single subset of retinal ganglion cells labeled by the specific antibody against melanopsin exhibited the morphology typical of M1-type ipRGCs. Immunoreactivity for both MT1 and MT2 receptors was clearly seen in the cytoplasm of all labeled ipRGCs, indicating that these two receptors were co-expressed in each of these neurons. Furthermore, labeling for both the receptors were found in neonatal M1 cells as early as the day of birth. It is therefore highly plausible that retinal melatonin may directly modulate the activity of ipRGCs, thus regulating non-image forming visual functions.
Collapse
|
49
|
Müller K, Naumann S, Weber W, Zurbriggen MD. Optogenetics for gene expression in mammalian cells. Biol Chem 2015; 396:145-52. [DOI: 10.1515/hsz-2014-0199] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 07/29/2014] [Indexed: 12/14/2022]
Abstract
Abstract
Molecular switches that are controlled by chemicals have evolved as central research instruments in mammalian cell biology. However, these tools are limited in terms of their spatiotemporal resolution due to freely diffusing inducers. These limitations have recently been addressed by the development of optogenetic, genetically encoded, and light-responsive tools that can be controlled with the unprecedented spatiotemporal precision of light. In this article, we first provide a brief overview of currently available optogenetic tools that have been designed to control diverse cellular processes. Then, we focus on recent developments in light-controlled gene expression technologies and provide the reader with a guideline for choosing the most suitable gene expression system.
Collapse
|
50
|
Cameron EG, Robinson PR. β-Arrestin-dependent deactivation of mouse melanopsin. PLoS One 2014; 9:e113138. [PMID: 25401926 PMCID: PMC4234672 DOI: 10.1371/journal.pone.0113138] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 10/21/2014] [Indexed: 11/25/2022] Open
Abstract
In mammals, the expression of the unusual visual pigment, melanopsin, is restricted to a small subset of intrinsically photosensitive retinal ganglion cells (ipRGCs), whose signaling regulate numerous non-visual functions including sleep, circadian photoentrainment and pupillary constriction. IpRGCs exhibit attenuated electrical responses following sequential and prolonged light exposures indicative of an adaptational response. The molecular mechanisms underlying deactivation and adaptation in ipRGCs however, have yet to be fully elucidated. The role of melanopsin phosphorylation and β-arrestin binding in this adaptive process is suggested by the phosphorylation-dependent reduction of melanopsin signaling in vitro and the ubiquitous expression of β-arrestin in the retina. These observations, along with the conspicuous absence of visual arrestin in ipRGCs, suggest that a β-arrestin terminates melanopsin signaling. Here, we describe a light- and phosphorylation- dependent reduction in melanopsin signaling mediated by both β-arrestin 1 and β-arrestin 2. Using an in vitro calcium imaging assay, we demonstrate that increasing the cellular concentration of β-arrestin 1 and β-arrestin 2 significantly increases the rate of deactivation of light-activated melanopsin in HEK293 cells. Furthermore, we show that this response is dependent on melanopsin carboxyl-tail phosphorylation. Crosslinking and co-immunoprecipitation experiments confirm β-arrestin 1 and β-arrestin 2 bind to melanopsin in a light- and phosphorylation- dependent manner. These data are further supported by proximity ligation assays (PLA), which demonstrate a melanopsin/β-arrestin interaction in HEK293 cells and ipRGCs. Together, these results suggest that melanopsin signaling is terminated in a light- and phosphorylation-dependent manner through the binding of a β-arrestin within the retina.
Collapse
Affiliation(s)
- Evan G. Cameron
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, Maryland, United States of America
| | - Phyllis R. Robinson
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|