Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Cell ; 186(2): 398-412.e17, 2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36669474

RESUMO

Public health studies indicate that artificial light is a high-risk factor for metabolic disorders. However, the neural mechanism underlying metabolic modulation by light remains elusive. Here, we found that light can acutely decrease glucose tolerance (GT) in mice by activation of intrinsically photosensitive retinal ganglion cells (ipRGCs) innervating the hypothalamic supraoptic nucleus (SON). Vasopressin neurons in the SON project to the paraventricular nucleus, then to the GABAergic neurons in the solitary tract nucleus, and eventually to brown adipose tissue (BAT). Light activation of this neural circuit directly blocks adaptive thermogenesis in BAT, thereby decreasing GT. In humans, light also modulates GT at the temperature where BAT is active. Thus, our work unveils a retina-SON-BAT axis that mediates the effect of light on glucose metabolism, which may explain the connection between artificial light and metabolic dysregulation, suggesting a potential prevention and treatment strategy for managing glucose metabolic disorders.


Assuntos
Tecido Adiposo Marrom , Hipotálamo , Camundongos , Animais , Humanos , Tecido Adiposo Marrom/metabolismo , Hipotálamo/metabolismo , Termogênese/fisiologia , Retina , Células Ganglionares da Retina , Glucose/metabolismo
2.
Cell ; 185(17): 3124-3137.e15, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35944541

RESUMO

During development, melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs) become light sensitive much earlier than rods and cones. IpRGCs project to many subcortical areas, whereas physiological functions of these projections are yet to be fully elucidated. Here, we found that ipRGC-mediated light sensation promotes synaptogenesis of pyramidal neurons in various cortices and the hippocampus. This phenomenon depends on activation of ipRGCs and is mediated by the release of oxytocin from the supraoptic nucleus (SON) and the paraventricular nucleus (PVN) into cerebral-spinal fluid. We further characterized a direct connection between ipRGCs and oxytocin neurons in the SON and mutual projections between oxytocin neurons in the SON and PVN. Moreover, we showed that the lack of ipRGC-mediated, light-promoted early cortical synaptogenesis compromised learning ability in adult mice. Our results highlight the importance of light sensation early in life on the development of learning ability and therefore call attention to suitable light environment for infant care.


Assuntos
Ocitocina , Células Ganglionares da Retina , Animais , Encéfalo/metabolismo , Humanos , Camundongos , Células Ganglionares da Retina/fisiologia , Opsinas de Bastonetes/metabolismo
3.
Cell ; 175(1): 71-84.e18, 2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30173913

RESUMO

Light exerts a range of powerful biological effects beyond image vision, including mood and learning regulation. While the source of photic information affecting mood and cognitive functions is well established, viz. intrinsically photosensitive retinal ganglion cells (ipRGCs), the central mediators are unknown. Here, we reveal that the direct effects of light on learning and mood utilize distinct ipRGC output streams. ipRGCs that project to the suprachiasmatic nucleus (SCN) mediate the effects of light on learning, independently of the SCN's pacemaker function. Mood regulation by light, on the other hand, requires an SCN-independent pathway linking ipRGCs to a previously unrecognized thalamic region, termed perihabenular nucleus (PHb). The PHb is integrated in a distinctive circuitry with mood-regulating centers and is both necessary and sufficient for driving the effects of light on affective behavior. Together, these results provide new insights into the neural basis required for light to influence mood and learning.


Assuntos
Afeto/efeitos da radiação , Aprendizagem/efeitos da radiação , Luz , Afeto/fisiologia , Animais , Encéfalo/fisiologia , Ritmo Circadiano , Aprendizagem/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Fototerapia/métodos , Retina/metabolismo , Retina/fisiologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/fisiologia , Células Ganglionares da Retina/efeitos da radiação , Transdução de Sinais/fisiologia , Núcleo Supraquiasmático/metabolismo , Visão Ocular/fisiologia , Vias Visuais/metabolismo , Percepção Visual/fisiologia
4.
Cell ; 175(3): 652-664.e12, 2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-30270038

RESUMO

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 PLCß4 (phospholipase C-ß4) 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.


Assuntos
Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Células Ganglionares da Retina/metabolismo , Visão Ocular , Animais , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nucleotídeos Cíclicos/metabolismo , Células Ganglionares da Retina/fisiologia , Canais de Cátion TRPC/metabolismo
5.
Cereb Cortex ; 34(3)2024 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-38521995

RESUMO

In brightness, the pupil constricts, while in darkness, the pupil dilates; this is known as the pupillary light response (PLR). The PLR is driven by all photoreceptors: rods and cones, which contribute to image-forming vision, and intrinsically photosensitive retinal ganglion cells (ipRGCs), which mainly contribute to non-image-forming vision. Rods and cones cause immediate pupil constriction upon light exposure, whereas ipRGCs cause sustained constriction throughout light exposure. Recent studies have shown that covert attention modulated the initial PLR; however, it remains unclear whether the same holds for the sustained PLR. We tested this by leveraging ipRGCs' responsiveness to blue light, causing the most prominent sustained constriction. While replicating previous studies by showing that pupils constricted more when either directly looking at, or covertly attending to, bright as compared to dim stimuli (with the same color), we also found that the pupil constricted more when directly looking at blue as compared to red stimuli (with the same luminosity). Crucially, however, in two high-powered studies (n = 60), we did not find any pupil-size difference when covertly attending to blue as compared to red stimuli. This suggests that ipRGC-mediated pupil constriction, and possibly non-image-forming vision more generally, is not modulated by covert attention.


Assuntos
Células Ganglionares da Retina , Visão Ocular , Constrição , Células Ganglionares da Retina/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Luz , Estimulação Luminosa
6.
Exp Eye Res ; 245: 109976, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38897270

RESUMO

This review examines the pivotal role of photoreceptor cells in ocular refraction development, focusing on dopamine (DA) as a key neurotransmitter. Contrary to the earlier view favoring cone cells, recent studies have highlighted the substantial contributions of both rod and cone cells to the visual signaling pathways that influence ocular refractive development. Notably, rod cells appeared to play a central role. Photoreceptor cells interact intricately with circadian rhythms, color vision pathways, and other neurotransmitters, all of which are crucial for the complex mechanisms driving the development of myopia. This review emphasizes that ocular refractive development results from a coordinated interplay between diverse cell types, signaling pathways, and neurotransmitters. This perspective has significant implications for unraveling the complex mechanisms underlying myopia and aiding in the development of more effective prevention and treatment strategies.


Assuntos
Miopia , Refração Ocular , Miopia/fisiopatologia , Miopia/metabolismo , Miopia/etiologia , Humanos , Refração Ocular/fisiologia , Animais , Dopamina/metabolismo , Células Fotorreceptoras Retinianas Cones/fisiologia , Células Fotorreceptoras Retinianas Cones/patologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Ritmo Circadiano/fisiologia , Transdução de Sinais/fisiologia , Células Fotorreceptoras de Vertebrados/fisiologia , Células Fotorreceptoras de Vertebrados/patologia
7.
Graefes Arch Clin Exp Ophthalmol ; 262(3): 983-990, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37864638

RESUMO

Myopia, a common ophthalmic disorder, places a high economic burden on individuals and society. Genetic and environmental factors influence myopia progression; however, the underlying mechanisms remain unelucidated. This paper reviews recent advances in circadian rhythm, intrinsically photosensitive retinal ganglion cells (ipRGCs), and dopamine (DA) signalling in myopia and proposes the hypothesis of a circadian rhythm brain retinal circuit in myopia progression. The search of relevant English articles was conducted in the PubMed databases until June 2023. Based on the search, emerging evidence indicated that circadian rhythm was associated with myopia, including circadian genes Bmal1, Cycle, and Per. In both humans and animals, the ocular morphology and physiology show rhythmic oscillations. Theoretically, such ocular rhythms are regulated locally and indirectly via the suprachiasmatic nucleus, which receives signal from the ipRGCs. Compared with the conventional retinal ganglion cells, ipRGCs can sense the presence of light because of specific expression of melanopsin. Light, together with ipRGCs and DA signalling, plays a crucial role in both circadian rhythm and myopia. In summary, regarding myopia progression, a circadian rhythm brain retinal circuit involving ipRGCs and DA signalling has not been well established. However, based on the relationship between circadian rhythm, ipRGCs, and DA signalling in myopia, we hypothesised a circadian rhythm brain retinal circuit.


Assuntos
Miopia , Células Ganglionares da Retina , Animais , Humanos , Dopamina , Miopia/genética , Retina , Ritmo Circadiano
8.
Development ; 147(23)2020 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-33288502

RESUMO

Retinal ganglion cells (RGCs) serve as a crucial communication channel from the retina to the brain. In the adult, these cells receive input from defined sets of presynaptic partners and communicate with postsynaptic brain regions to convey features of the visual scene. However, in the developing visual system, RGC interactions extend beyond their synaptic partners such that they guide development before the onset of vision. In this Review, we summarize our current understanding of how interactions between RGCs and their environment influence cellular targeting, migration and circuit maturation during visual system development. We describe the roles of RGC subclasses in shaping unique developmental responses within the retina and at central targets. Finally, we highlight the utility of RNA sequencing and genetic tools in uncovering RGC type-specific roles during the development of the visual system.


Assuntos
Encéfalo/crescimento & desenvolvimento , Retina/crescimento & desenvolvimento , Células Ganglionares da Retina/fisiologia , Visão Ocular/genética , Animais , Sequência de Bases , Diferenciação Celular/genética , Movimento Celular/genética , Humanos , Retina/metabolismo , Células Ganglionares da Retina/metabolismo
9.
Proc Natl Acad Sci U S A ; 117(29): 17320-17329, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32632006

RESUMO

Second only to headache, photophobia is the most debilitating symptom reported by people with migraine. While the melanopsin-containing intrinsically photosensitive retinal ganglion cells (ipRGCs) are thought to play a role, how cone and melanopsin signals are integrated in this pathway to produce visual discomfort is poorly understood. We studied 60 people: 20 without headache and 20 each with interictal photophobia from migraine with or without visual aura. Participants viewed pulses of spectral change that selectively targeted melanopsin, the cones, or both and rated the degree of visual discomfort produced by these stimuli while we recorded pupil responses. We examined the data within a model that describes how cone and melanopsin signals are weighted and combined at the level of the retina and how this combined signal is transformed into a rating of discomfort or pupil response. Our results indicate that people with migraine do not differ from headache-free controls in the manner in which melanopsin and cone signals are combined. Instead, people with migraine demonstrate an enhanced response to integrated ipRGC signals for discomfort. This effect of migraine is selective for ratings of visual discomfort, in that an enhancement of pupil responses was not seen in the migraine group, nor were group differences found in surveys of other behaviors putatively linked to ipRGC function (chronotype, seasonal sensitivity, presence of a photic sneeze reflex). By revealing a dissociation in the amplification of discomfort vs. pupil response, our findings suggest a postretinal alteration in processing of ipRGC signals for photophobia in migraine.


Assuntos
Transtornos de Enxaqueca/metabolismo , Fotofobia/metabolismo , Células Ganglionares da Retina/fisiologia , Adulto , Feminino , Humanos , Masculino , Estimulação Luminosa , Pupila/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Opsinas de Bastonetes/fisiologia
10.
J Pineal Res ; 71(2): e12711, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33326640

RESUMO

In mammals, the suprachiasmatic nuclei (SCN) constitute the main circadian clock, receiving input from the retina which allows synchronization of endogenous biological rhythms with the daily light/dark cycle. Over the year, the SCN encodes photoperiodic variations through duration of melatonin secretion, with abundant nocturnal levels in winter and lower levels in summer. Thus, light information is critical to regulate seasonal reproduction in many species and is part of the central photoperiodic integration. Since intrinsically photosensitive retinal ganglion cells (ipRGCs) are vital for circadian photoentrainment and other nonvisual functions, we studied the contribution of ipRGCs in photoperiod integration in C3H retinal degeneration 1 (rd1) mice. We assessed locomotor activity and melatonin secretion in mice exposed to short or long photoperiods. Our results showed that rd1 mice are still responsive to photoperiod variations in term of locomotor activity, melatonin secretion, and regulation of the reproductive axis. In addition, retinas of animals exposed to short photoperiod exhibit higher melanopsin labeling intensity compared with the long photoperiod condition, suggesting seasonal-dependent changes within this photoreceptive system. These results show that ipRGCs in rd1 mice can still measure photoperiod and suggest a key role of melanopsin cells in photoperiod integration and the regulation of seasonal physiology.


Assuntos
Melatonina , Degeneração Retiniana , Animais , Ritmo Circadiano/fisiologia , Camundongos , Camundongos Endogâmicos C3H , Fotoperíodo , Núcleo Supraquiasmático/fisiologia
11.
Mol Cell Neurosci ; 103: 103468, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32027966

RESUMO

The zebrafish pineal organ is a photoreceptive structure containing two main neuronal populations (photoreceptors and projections neurons). Here we describe a subpopulation of projection neurons that expresses the melanopsin gene, opn4xa. This new pineal cell type, that displays characteristics of both projection neurons and photoreceptors, share a similar dependency for BMP and Notch signalling pathways with classical non-photosensitive projection neurons (PN). Functionally, however, whereas classical, opn4xa-negative PNs display an achromatic LIGHT OFF response, the novel cell type we describe exhibit a LIGHT ON character that is elicited by green and blue light. Taken together, our data suggest a previously unanticipated heterogeneity in the projection neuron population in the zebrafish pineal organ raising the question of the importance of these differences in pineal function.


Assuntos
Interneurônios/metabolismo , Neurônios/metabolismo , Células Fotorreceptoras/fisiologia , Glândula Pineal/metabolismo , Animais , Glândula Pineal/patologia , Transdução de Sinais/fisiologia , Peixe-Zebra/genética
12.
Int J Mol Sci ; 22(16)2021 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-34445064

RESUMO

Photoreceptors are critical components of the retina and play a role in the first step of the conversion of light to electric signals. With the discovery of the intrinsically photosensitive retinal ganglion cells, which regulate non-image-forming visual processes, our knowledge of the photosensitive cell family in the retina has deepened. Photoreceptor development is regulated by specific genes and proteins and involves a series of molecular processes including DNA transcription, post-transcriptional modification, protein translation, and post-translational modification. Single-cell sequencing is a promising technology for the study of photoreceptor development. This review presents an overview of the types of human photoreceptors, summarizes recent discoveries in the regulatory mechanisms underlying their development at single-cell resolution, and outlines the prospects in this field.


Assuntos
Células Fotorreceptoras de Vertebrados/citologia , Retina/crescimento & desenvolvimento , Análise de Célula Única/métodos , Animais , Humanos , Organoides/citologia , Organoides/embriologia , Organoides/crescimento & desenvolvimento , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/citologia , Retina/embriologia
13.
Proc Natl Acad Sci U S A ; 114(10): 2741-2746, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28223508

RESUMO

Intrinsically photosensitive retinal ganglion cells (ipRGCs) express the photopigment melanopsin and mediate several non-image-forming visual functions, including circadian photoentrainment and the pupillary light reflex (PLR). ipRGCs act as autonomous photoreceptors via the intrinsic melanopsin-based phototransduction pathway and as a relay for rod/cone input via synaptically driven responses. Under low light intensities, where only synaptically driven rod/cone input activates ipRGCs, the duration of the ipRGC response will be determined by the termination kinetics of the rod/cone circuits. Little is known, however, about the termination kinetics of the intrinsic melanopsin-based phototransduction pathway and its contribution to several melanopsin-mediated behaviors. Here, we show that C-terminal phosphorylation of melanopsin determines the recovery kinetics of the intrinsic melanopsin-based photoresponse in ipRGCs, the duration of the PLR, and the speed of reentrainment. In contrast, circadian phase alignment and direct effects of light on activity (masking) are not influenced by C-terminal phosphorylation of melanopsin. Electrophysiological measurements demonstrate that expression of a virally encoded melanopsin lacking all C-terminal phosphorylation sites (C terminus phosphonull) leads to a prolonged intrinsic light response. In addition, mice expressing the C terminus phosphonull in ipRGCs reentrain faster to a delayed light/dark cycle compared with mice expressing virally encoded WT melanopsin; however, the phase angle of entrainment and masking were indistinguishable. Importantly, a sustained PLR in the phosphonull animals is only observed at brighter light intensities that activate melanopsin phototransduction, but not at dimmer light intensities that activate only the rod/cone pathway. Taken together, our results highlight how the kinetics of the melanopsin photoresponse differentially regulate distinct light-mediated behaviors.


Assuntos
Comportamento Animal , Transdução de Sinal Luminoso/genética , Células Ganglionares da Retina/metabolismo , Opsinas de Bastonetes/metabolismo , Animais , Ritmo Circadiano/genética , Cinética , Luz , Transdução de Sinal Luminoso/fisiologia , Camundongos , Técnicas de Patch-Clamp , Fosforilação/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/fisiologia , Reflexo Pupilar/genética , Reflexo Pupilar/fisiologia , Retina/metabolismo , Retina/fisiologia , Células Ganglionares da Retina/fisiologia , Opsinas de Bastonetes/química , Opsinas de Bastonetes/genética , Sinapses/genética , Sinapses/metabolismo , Visão Ocular/genética , Visão Ocular/fisiologia
14.
Proc Natl Acad Sci U S A ; 114(46): 12291-12296, 2017 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-29087940

RESUMO

The photopigment melanopsin supports reflexive visual functions in people, such as pupil constriction and circadian photoentrainment. What contribution melanopsin makes to conscious visual perception is less studied. We devised a stimulus that targeted melanopsin separately from the cones using pulsed (3-s) spectral modulations around a photopic background. Pupillometry confirmed that the melanopsin stimulus evokes a response different from that produced by cone stimulation. In each of four subjects, a functional MRI response in area V1 was found. This response scaled with melanopic contrast and was not easily explained by imprecision in the silencing of the cones. Twenty additional subjects then observed melanopsin pulses and provided a structured rating of the perceptual experience. Melanopsin stimulation was described as an unpleasant, blurry, minimal brightening that quickly faded. We conclude that isolated stimulation of melanopsin is likely associated with a response within the cortical visual pathway and with an evoked conscious percept.


Assuntos
Visão de Cores/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Células Ganglionares da Retina/fisiologia , Opsinas de Bastonetes/fisiologia , Córtex Visual/fisiologia , Percepção Visual/fisiologia , Adulto , Feminino , Humanos , Luz , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Estimulação Luminosa , Pupila/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Córtex Visual/diagnóstico por imagem , Vias Visuais
15.
Ophthalmic Physiol Opt ; 39(6): 459-468, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31696535

RESUMO

PURPOSE: Optical filters and tints manipulating short-wavelength light (sometimes called 'blue-blocking' or 'blue-attenuating' filters) are used in the management of a range of ocular, retinal, neurological and psychiatric disorders. In many cases, the only available quantification of the optical effects of a given optical filter is the spectral transmittance, which specifies the amount of light transmitted as a function of wavelength. METHODS: We propose a novel physiologically relevant and retinally referenced framework for quantifying the visual and non-visual effects of these filters, incorporating the attenuation of luminance (luminous transmittance), the attenuation of melanopsin activation (melanopsin transmittance), the colour shift, and the reduction of the colour gamut (gamut reduction). Using these criteria, we examined a novel database of spectral transmittance functions of optical filters (n = 121) which were digitally extracted from a variety of sources. RESULTS: We find a large diversity in the alteration of visual and non-visual properties. The spectral transmittance properties of the examined filters vary widely, in terms of shapes and cut-off wavelengths. All filters show relatively more melanopsin attenuation than luminance attenuation (lower melanopsin transmittance than luminous transmittance). Across the data set, we find that melanopsin transmittance and luminous transmittance are correlated. CONCLUSIONS: We suggest that future studies and examinations of the physiological effects of optical filters quantify the visual and non-visual effects of the filters beyond the spectral transmittance, which will eventually aid in developing a mechanistic understanding of how different filters affect physiology. We strongly discourage comparing the downstream effects of different filters on, e.g. sleep or circadian responses, without considering their effects on the retinal stimulus.


Assuntos
Luz , Óptica e Fotônica , Estimulação Luminosa/métodos , Retina/fisiologia , Visão Ocular/fisiologia , Humanos , Opsinas de Bastonetes/metabolismo , Opsinas de Bastonetes/efeitos da radiação
16.
Proc Natl Acad Sci U S A ; 113(21): 6047-52, 2016 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-27162356

RESUMO

The suprachiasmatic nucleus (SCN) receives direct retinal input from the intrinsically photosensitive retinal ganglion cells (ipRGCs) for circadian photoentrainment. Interestingly, the SCN is the only brain region that receives equal inputs from the left and right eyes. Despite morphological assessments showing that axonal fibers originating from ipRGCs cover the entire SCN, physiological evidence suggests that only vasoactive intestinal polypeptide (VIP)/gastrin-releasing peptide (GRP) cells located ventrally in the SCN receive retinal input. It is still unclear, therefore, which subpopulation of SCN neurons receives synaptic input from the retina and how the SCN receives equal inputs from both eyes. Here, using single ipRGC axonal tracing and a confocal microscopic analysis in mice, we show that ipRGCs have elaborate innervation patterns throughout the entire SCN. Unlike conventional retinal ganglion cells (RGCs) that innervate visual targets either ipsilaterally or contralaterally, a single ipRGC can bilaterally innervate the SCN. ipRGCs form synaptic contacts with major peptidergic cells of the SCN, including VIP, GRP, and arginine vasopressin (AVP) neurons, with each ipRGC innervating specific subdomains of the SCN. Furthermore, a single SCN-projecting ipRGC can send collateral inputs to many other brain regions. However, the size and complexity of the axonal arborizations in non-SCN regions are less elaborate than those in the SCN. Our results provide a better understanding of how retinal neurons connect to the central circadian pacemaker to synchronize endogenous circadian clocks with the solar day.


Assuntos
Relógios Circadianos/fisiologia , Células Ganglionares da Retina/metabolismo , Núcleo Supraquiasmático/metabolismo , Sinapses/metabolismo , Visão Ocular/fisiologia , Animais , Camundongos , Camundongos Transgênicos , Células Ganglionares da Retina/citologia , Núcleo Supraquiasmático/citologia , Sinapses/genética
17.
Int J Mol Sci ; 20(13)2019 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-31261700

RESUMO

Melanopsin-containing retinal ganglion cells (mRGCs) represent a third class of retinal photoreceptors involved in regulating the pupillary light reflex and circadian photoentrainment, among other things. The functional integrity of the circadian system and melanopsin cells is an essential component of well-being and health, being both impaired in aging and disease. Here we review evidence of melanopsin-expressing cell alterations in aging and neurodegenerative diseases and their correlation with the development of circadian rhythm disorders. In healthy humans, the average density of melanopsin-positive cells falls after age 70, accompanied by age-dependent atrophy of dendritic arborization. In addition to aging, inner and outer retinal diseases also involve progressive deterioration and loss of mRGCs that positively correlates with progressive alterations in circadian rhythms. Among others, mRGC number and plexus complexity are impaired in Parkinson's disease patients; changes that may explain sleep and circadian rhythm disorders in this pathology. The key role of mRGCs in circadian photoentrainment and their loss in age and disease endorse the importance of eye care, even if vision is lost, to preserve melanopsin ganglion cells and their essential functions in the maintenance of an adequate quality of life.


Assuntos
Envelhecimento/patologia , Ritmo Circadiano , Doenças Neurodegenerativas/etiologia , Células Ganglionares da Retina/metabolismo , Opsinas de Bastonetes/metabolismo , Envelhecimento/metabolismo , Animais , Humanos , Células Ganglionares da Retina/patologia
18.
Neuroophthalmology ; 42(2): 65-72, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29563950

RESUMO

Previously, it has been reported that melanopsin-mediated pupillary light response (PLR), measured with pupillometry, is reduced in patients with idiopathic intracranial hypertension (IIH), indicating the clinical utility of the tool in the diagnosis of IIH. In the current study, the authors aimed to measure the PLR in 13 treatment-naive patients with new-onset IIH and 13 healthy controls. In contrast to the previous report, which was based on patients with longstanding IIH (n = 13), the authors found no significant difference in the melanopsin-mediated PLR (p = 0.48).

19.
J Neurosci ; 36(5): 1471-82, 2016 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-26843631

RESUMO

Signals from cones are recombined in postreceptoral channels [luminance, L + M; red-green, L - M; blue-yellow, S - (L + M)]. The melanopsin-containing retinal ganglion cells are also active at daytime light levels and recent psychophysical results suggest that melanopsin contributes to conscious vision in humans. Here, we measured BOLD fMRI responses to spectral modulations that separately targeted the postreceptoral cone channels and melanopsin. Responses to spatially uniform (27.5° field size, central 5° obscured) flicker at 0.5, 1, 2, 4, 8, 16, 32, and 64 Hz were recorded from areas V1, V2/V3, motion-sensitive area MT, and the lateral occipital complex. In V1 and V2/V3, higher temporal sensitivity was observed to L + M + S (16 Hz) compared with L - M flicker (8 Hz), consistent with psychophysical findings. Area MT was most sensitive to rapid (32 Hz) flicker of either L + M + S or L - M. We found S cone responses only in areas V1 and V2/V3 (peak frequency: 4-8 Hz). In addition, we studied an L + M modulation and found responses that were effectively identical at all temporal frequencies to those recorded for the L + M + S modulation. Finally, we measured the cortical response to melanopsin-directed flicker and compared this response with control modulations that addressed stimulus imprecision and the possibility of stimulation of cones in the shadow of retinal blood vessels (penumbral cones). For our stimulus conditions, melanopsin flicker did not elicit a cortical response exceeding that of the control modulations. We note that failure to control for penumbral cone stimulation could be mistaken for a melanopsin response. SIGNIFICANCE STATEMENT: The retina contains cone photoreceptors and ganglion cells that contain the photopigment melanopsin. Cones provide brightness and color signals to visual cortex. Melanopsin influences circadian rhythm and the pupil, but its contribution to cortex and perception is less clear. We measured the response of human visual cortex with fMRI using spectral modulations tailored to stimulate the cones and melanopsin separately. We found that cortical responses to cone signals vary systematically across visual areas. Differences in temporal sensitivity for achromatic, red-green, and blue-yellow stimuli generally reflect the known perceptual properties of vision. We found that melanopsin signals do not produce a measurable response in visual cortex at temporal frequencies between 0.5 and 64 Hz at daytime light levels.


Assuntos
Fusão Flicker/fisiologia , Estimulação Luminosa/métodos , Pupila/fisiologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Opsinas de Bastonetes/metabolismo , Córtex Visual/metabolismo , Adulto , Feminino , Humanos , Imageamento por Ressonância Magnética/métodos , Masculino
20.
FASEB J ; 30(11): 3690-3701, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27440795

RESUMO

The circadian clock is thought to adjust retinal sensitivity to ambient light levels, yet the involvement of specific clock genes is poorly understood. We explored the potential role of the nuclear receptor subfamily 1, group D, member 1 (REV-ERBα; or NR1D1) in this respect. In light-evoked behavioral tests, compared with wild-type littermates, Rev-Erbα-/- mice showed enhanced negative masking at low light levels (0.1 lx). Rev-Erbα-/- mouse retinas displayed significantly higher numbers of intrinsically photosensitive retinal ganglion cells (ipRGCs; 62% more compared with wild-type) and more intense melanopsin immunostaining of individual ipRGCs. In agreement with a pivotal role for melanopsin, negative masking at low light intensities was abolished in Rev-Erbα-/- Opn4-/- (melanopsin gene) double-null mice. Rev-Erbα-/- mice showed shortened latencies of both a and b electroretinogram waves, modified scotopic and photopic b-wave and scotopic threshold responses, and increased pupillary constriction, all of which suggested increased light sensitivity. However, wild-type and Rev-Erbα-/- mice displayed no detectable differences by in vivo fundus imaging, retinal histology, or expression of cell type-specific markers for major retinal cell populations. We conclude that REV-ERBα plays a major role in retinal information processing, and we speculate that REV-ERBα and melanopsin set sensitivity levels of the rod-mediated ipRGC pathway to coordinate activity with ambient light.-Ait-Hmyed Hakkari, O., Acar, N., Savier, E., Spinnhirny, P., Bennis, M., Felder-Schmittbuhl, M.-P., Mendoza, J., Hicks, D. Rev-Erbα modulates retinal visual processing and behavioral responses to light.


Assuntos
Comportamento Animal/fisiologia , Luz , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Retina/fisiologia , Opsinas de Bastonetes/metabolismo , Animais , Relógios Circadianos/genética , Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Camundongos Knockout , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/deficiência , Estimulação Luminosa/métodos , Células Ganglionares da Retina/fisiologia , Opsinas de Bastonetes/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa