Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
eNeuro ; 9(5)2022.
Article in English | MEDLINE | ID: mdl-36041828

ABSTRACT

Opsin-3 (Opn3, encephalopsin) was the first nonvisual opsin gene discovered in mammals. Since then, several Opn3 functions have been described, and in two cases (adipose tissue, smooth muscle) light sensing activity is implicated. In addition to peripheral tissues, Opn3 is robustly expressed within the central nervous system, for which it derives its name. Despite this expression, no studies have investigated developmental or adult CNS consequences of Opn3 loss-of-function. Here, the behavioral consequences of mice deficient in Opn3 were investigated. Opn3-deficient mice perform comparably to wild-type mice in measures of motor coordination, socialization, anxiety-like behavior, and various aspects of learning and memory. However, Opn3-deficient mice have an attenuated acoustic startle reflex (ASR) relative to littermates. This deficit is not because of changes in hearing sensitivity, although Opn3 was shown to be expressed in auditory and vestibular structures, including cochlear outer hair cells. Interestingly, the ASR was not acutely light-dependent and did not vary between daytime and nighttime trials, despite known functions of Opn3 in photoreception and circadian gene amplitude. Together, these results demonstrate the first role of Opn3 on behavior, although the role of this opsin in the CNS remains largely elusive.


Subject(s)
Reflex, Startle , Rod Opsins , Acoustic Stimulation , Animals , Mammals/metabolism , Mice , Opsins , Rod Opsins/genetics , Rod Opsins/metabolism
2.
Biochem Pharmacol ; 191: 114304, 2021 09.
Article in English | MEDLINE | ID: mdl-33129807

ABSTRACT

Daylight is ubiquitous and is crucial for mammalian vision as well as for non-visual input to the brain via the intrinsically photosensitive retinal ganglion cells (ipRGCs) that express the photopigment melanopsin. The ipRGCs project to the circadian clock in the suprachiasmatic nuclei and thereby ensure entrainment to the 24-hour day-night cycle, and changes in daylength trigger the appropriate seasonal behaviours. The ipRGCs also project to the perihabenular nucleus and surrounding brain regions that modulate mood, stress and learning in animals and humans. Given that light has strong direct effects on mood, cognition, alertness, performance, and sleep, light can be considered a "drug" to treat many clinical conditions. Light therapy is already well established for winter and other depressions and circadian sleep disorders. Beyond visual and non-visual effects via the retina, daylight contributes to prevent myopia in the young by its impact on eye development, and is important for Vitamin D synthesis and bone health via the skin. The sun is the most powerful light source and, dependent on dose, its ultraviolet radiance is toxic for living organisms and can be used as a disinfectant. Most research involves laboratory-based electric light, without the dynamic and spectral changes that daylight undergoes moment by moment. There is a gap between the importance of daylight for human beings and the amount of research being done on this subject. Daylight is taken for granted as an environmental factor, to be enjoyed or avoided, according to conditions. More daylight awareness in architecture and urban design beyond aesthetic values and visual comfort may lead to higher quality work and living environments. Although we do not yet have a factual basis for the assumption that natural daylight is overall "better" than electric light, the environmental debate mandates serious consideration of sunlight not just for solar power but also as biologically necessary for sustainable and healthy living.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Light , Photoperiod , Humans , Mood Disorders/etiology , Mood Disorders/metabolism , Mood Disorders/prevention & control , Myopia/etiology , Myopia/metabolism , Myopia/prevention & control , Retina/metabolism , Retinal Ganglion Cells/metabolism , Rod Opsins/metabolism , Suprachiasmatic Nucleus/metabolism , Vitamin D/metabolism
3.
Neuron ; 102(1): 128-142.e8, 2019 04 03.
Article in English | MEDLINE | ID: mdl-30795900

ABSTRACT

Light plays a pivotal role in the regulation of affective behaviors. However, the precise circuits that mediate the impact of light on depressive-like behaviors are not well understood. Here, we show that light influences depressive-like behaviors through a disynaptic circuit linking the retina and the lateral habenula (LHb). Specifically, M4-type melanopsin-expressing retinal ganglion cells (RGCs) innervate GABA neurons in the thalamic ventral lateral geniculate nucleus and intergeniculate leaflet (vLGN/IGL), which in turn inhibit CaMKIIα neurons in the LHb. Specific activation of vLGN/IGL-projecting RGCs, activation of LHb-projecting vLGN/IGL neurons, or inhibition of postsynaptic LHb neurons is sufficient to decrease the depressive-like behaviors evoked by long-term exposure to aversive stimuli or chronic social defeat stress. Furthermore, we demonstrate that the antidepressive effects of light therapy require activation of the retina-vLGN/IGL-LHb pathway. These results reveal a dedicated retina-vLGN/IGL-LHb circuit that regulates depressive-like behaviors and provide a potential mechanistic explanation for light treatment of depression.


Subject(s)
Depression , Depressive Disorder/therapy , GABAergic Neurons/physiology , Geniculate Bodies/physiology , Habenula/physiology , Phototherapy , Retinal Ganglion Cells/physiology , Visual Pathways/physiology , Animals , Behavior, Animal , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Disease Models, Animal , GABAergic Neurons/metabolism , Male , Neural Inhibition/physiology , Neurons/metabolism , Neurons/physiology , Retina/physiology , Rod Opsins/metabolism , Stress, Psychological , Thalamus/physiology
4.
Mol Vis ; 24: 621-632, 2018.
Article in English | MEDLINE | ID: mdl-30294201

ABSTRACT

Purpose: The purpose of this study was to investigate the effects of bilberry extract with its anthocyanins on retinal photoreceptor cell damage and on the endoplasmic reticulum (ER) stress induced by exposure to blue light-emitting diode (LED) light. Methods: Cultured murine photoreceptor cells (661W) were exposed to blue LED light with or without bilberry extract or its anthocyanins in the culture media. Aggregated short-wavelength opsin (S-opsin) in murine photoreceptor cells was observed with immunostaining. The expression of factors involved in the unfolded protein response was examined with immunoblot analysis and quantitative real-time reverse transcription (RT)-PCR. Furthermore, cell death was observed with double staining with Hoechst 33342 and propidium iodide after dithiothreitol (DTT) treatment. Results: Bilberry extract and anthocyanins suppressed the aggregation of S-opsin, activation of ATF4, and expression of the mRNA of the factors associated with the unfolded protein response (UPR). In addition, bilberry extract and the anthocyanins inhibited the death of photoreceptor cells induced by DTT, an ER stress inducer. Conclusions: These findings suggest that bilberry extract containing anthocyanins can alter the effects of blue LED light and DTT-induced retinal photoreceptor cell damage. These effects were achieved by modulating the activation of ATF4 and through the suppression of the abnormal aggregation of S-opsin.


Subject(s)
Anthocyanins/pharmacology , Endoplasmic Reticulum Stress/drug effects , Light/adverse effects , Photoreceptor Cells, Vertebrate/radiation effects , Plant Extracts/pharmacology , Unfolded Protein Response/drug effects , Vaccinium myrtillus/chemistry , Animals , Apoptosis , Blotting, Western , Cell Line , Dithiothreitol/pharmacology , Immunoblotting , Mice , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Protein Aggregation, Pathological , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/prevention & control , Real-Time Polymerase Chain Reaction , Retinal Degeneration/etiology , Retinal Degeneration/metabolism , Retinal Degeneration/prevention & control , Rod Opsins/metabolism
5.
Phytomedicine ; 42: 51-55, 2018 Mar 15.
Article in English | MEDLINE | ID: mdl-29655697

ABSTRACT

BACKGROUND: In the East Asia, the genus Acer (Aceraceae) is a herbal medicine that is used to treat various diseases, including hemostasis, hepatic disorders, traumatic bleeding and poor eyesight. However, the effects of Acer palmatum thumb. on retinal degeneration are unknown. AIM: In this study, we investigated whether Acer palmatum thumb.ethanol extract (KIOM-2015E) can protect eyes from retinal degeneration. Our research investigated whether KIOM-2015E could have a protective effect in the retinal degenerating mouse model induced by N-ethyl-N-nitrosourea (ENU). MATERIALS AND METHODS: Retinal degeneration was induced by a single intraperitoneal injection of ENU in ICR mice. KIOM-2015E (100, 200 mg/kg) was orally administered once per day. The eyeballs were embedded and lysed after drug administration to examine the histological changed and protein expression levels. RESULTS: The ENU-induced retinal degeneration model exhibited increased photoreceptor cell death and a loss of the outer nuclear layer. Additionally, the expression of PKCα and OPN1SW was reduced, and that of GFAP and Nestin was increased in ENU-treated retinal tissues. CONCLUSION: KIOM-2015E treatment ameliorated the ENU-induced retinal degeneration. KIOM-2015E prevents ENU-induced retinal degeneration by modulating protein expression and the thickness of the outer nuclear layer in the retina.


Subject(s)
Acer/chemistry , Plant Extracts/pharmacology , Retinal Degeneration/drug therapy , Administration, Oral , Animals , Disease Models, Animal , Ethylnitrosourea/administration & dosage , Ethylnitrosourea/toxicity , Glial Fibrillary Acidic Protein/metabolism , Injections, Intraperitoneal , Male , Mice, Inbred ICR , Nestin/metabolism , Plant Extracts/administration & dosage , Plant Leaves/chemistry , Plants, Medicinal/chemistry , Protective Agents/pharmacology , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinal Degeneration/chemically induced , Retinal Degeneration/metabolism , Rod Opsins/metabolism
6.
Exp Brain Res ; 236(4): 955-961, 2018 04.
Article in English | MEDLINE | ID: mdl-29379995

ABSTRACT

In this study, we examined the cellular distribution of encephalopsin (opsin 3; OPN3) expression in the striatum of non-human primates. In addition, because of our long standing interest in Parkinson's disease and neuroprotection, we examined whether parkinsonian (MPTP; 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) insult and/or photobiomodulation (670 nm) had any impact on encephalopsin expression in this key area of the basal ganglia. Striatal sections of control naïve monkeys, together with those that were either MPTP- and/or photobiomodulation-treated were processed for immunohistochemistry. Our results revealed two populations of striatal interneurones that expressed encephalopsin, one of which was the giant, choline acetyltransferase-containing, cholinergic interneurones. The other population had smaller somata and was not cholinergic. Neither cell group expressed the calcium-binding protein, parvalbumin. There was also rich encephalopsin expression in a set of terminals forming striosome-like patches across the striatum. Finally, we found that neither parkinsonian (MPTP) insult nor photobiomodulation had any effect on encephalopsin expression in the striatum. In summary, our results revealed an extensive network of encephalopsin containing structures throughout the striatum, indicating that external light is in a position to influence a range of striatal activities at both the interneurone and striosome level.


Subject(s)
Corpus Striatum/metabolism , Interneurons/metabolism , Low-Level Light Therapy , MPTP Poisoning/metabolism , Rod Opsins/metabolism , Animals , Immunohistochemistry , MPTP Poisoning/therapy , Macaca fascicularis
7.
Lasers Surg Med ; 49(7): 705-718, 2017 09.
Article in English | MEDLINE | ID: mdl-28418107

ABSTRACT

BACKGROUND AND OBJECTIVE: Though devices for hair growth based on low levels of light have shown encouraging results, further improvements of their efficacy is impeded by a lack of knowledge on the exact molecular targets that mediate physiological response in skin and hair follicle. The aim of this study was to investigate the expression of selected light-sensitive receptors in the human hair follicle and to study the impact of UV-free blue light on hair growth ex vivo. MATERIAL AND METHODS: The expression of Opsin receptors in human skin and hair follicles has been characterized using RT-qPCR and immunofluorescence approaches. The functional significance of Opsin 3 was assessed by silencing its expression in the hair follicle cells followed by a transcriptomic profiling. Proprietary LED-based devices emitting two discrete visible wavelengths were used to access the effects of selected optical parameters on hair growth ex vivo and outer root sheath cells in vitro. RESULTS: The expression of OPN2 (Rhodopsin) and OPN3 (Panopsin, Encephalopsin) was detected in the distinct compartments of skin and anagen hair follicle. Treatment with 3.2 J/cm2 of blue light with 453 nm central wavelength significantly prolonged anagen phase in hair follicles ex vivo that was correlated with sustained proliferation in the light-treated samples. In contrast, hair follicle treatment with 3.2 J/cm2 of 689 nm light (red light) did not significantly affect hair growth ex vivo. Silencing of OPN3 in the hair follicle outer root sheath cells resulted in the altered expression of genes involved in the control of proliferation and apoptosis, and abrogated stimulatory effects of blue light (3.2 J/cm2 ; 453 nm) on proliferation in the outer root sheath cells. CONCLUSIONS: We provide the first evidence that (i) OPN2 and OPN3 are expressed in human hair follicle, and (ii) A 453 nm blue light at low radiant exposure exerts a positive effect on hair growth ex vivo, potentially via interaction with OPN3. Lasers Surg. Med. 49:705-718, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Alopecia/radiotherapy , Hair Follicle/metabolism , Hair/growth & development , Light , Low-Level Light Therapy/methods , Rhodopsin/metabolism , Rod Opsins/metabolism , Adult , Aged , Alopecia/physiopathology , Apoptosis , Biomarkers/metabolism , Cell Proliferation , Female , Hair Follicle/physiology , Humans , In Vitro Techniques , Male , Middle Aged
8.
PLoS One ; 11(11): e0163925, 2016.
Article in English | MEDLINE | ID: mdl-27861495

ABSTRACT

Opsin family genes encode G protein-coupled seven-transmembrane proteins that bind a retinaldehyde chromophore in photoreception. Here, we sought potential as yet undescribed avian retinal photoreceptors, focusing on Opsin 3 homologs in the chicken. We found two Opsin 3-related genes in the chicken genome: one corresponding to encephalopsin/panopsin (Opn3) in mammals, and the other belonging to the teleost multiple tissue opsin (TMT) 2 group. Bioluminescence imaging and G protein activation assays demonstrated that the chicken TMT opsin (cTMT) functions as a blue light sensor when forced-expressed in mammalian cultured cells. We did not detect evidence of light sensitivity for the chicken Opn3 (cOpn3). In situ hybridization demonstrated expression of cTMT in subsets of differentiating cells in the inner retina and, as development progressed, predominant localization to retinal horizontal cells (HCs). Immunohistochemistry (IHC) revealed cTMT in HCs as well as in small numbers of cells in the ganglion and inner nuclear layers of the post-hatch chicken retina. In contrast, cOpn3-IR cells were found in distinct subsets of cells in the inner nuclear layer. cTMT-IR cells were also found in subsets of cells in the hypothalamus. Finally, we found differential distribution of cOpn3 and cTMT proteins in specific cells of the cerebellum. The present results suggest that a novel TMT-type opsin 3 may function as a photoreceptor in the chicken retina and brain.


Subject(s)
Brain/metabolism , Retina/metabolism , Rod Opsins/metabolism , Animals , Brain/cytology , Calcium/metabolism , Cerebellum/cytology , Cerebellum/metabolism , Chickens , Exons , Gene Expression , Genomics , Hypothalamus/cytology , Hypothalamus/metabolism , Introns , Light , Multigene Family , Phylogeny , Protein Transport , RNA Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/cytology , Retinal Horizontal Cells/physiology , Rod Opsins/genetics
9.
Invest Ophthalmol Vis Sci ; 56(8): 4680-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26207303

ABSTRACT

PURPOSE: Intrinsically photosensitive retinal ganglion cells (ipRGCs) mediate nonimage-forming visual functions such as pupillary constriction and circadian photoentrainment. Optimizing daytime nonimage-forming photostimulation has health benefits. We aimed to enhance ipRGC excitation using flickering instead of steady light. METHODS: Human subjects were tested with a three-dimensional matrix of flickering 463-nm stimuli: three photon counts (13.7, 14.7 and 15.7 log photons cm(-2)), three duty cycles (12%, 47%, and 93%) and seven flicker frequencies (0.1, 0.25, 0.5, 1, 2, 4, and 7 Hz). Steady-state pupil constrictions were measured. RESULTS: Among stimuli containing 13.7 log photons cm-2, the one flickering at 2 Hz with a 12% duty cycle evoked the greatest pupil constriction of 48% ± 4%, 71% greater than that evoked by an equal-intensity (12.3 log photons cm(-2) s(-1)) continuous light. This frequency and duty cycle were also best for 14.7 log photons cm-2 stimuli, inducing a 58% ± 4% constriction which was 38% more than that caused by an equal-intensity (13.3 log photons cm(-2) s(-1)) constant light. For 15.7 log photons cm-2 stimuli, the 1-Hz, 47% duty cycle flicker was optimal although it evoked the same constriction as the best 14.7 log photons cm(-2) flicker. CONCLUSIONS: Pupillary constriction depends on flicker frequency and duty cycle besides intensity. Among the stimuli tested, the one with the lowest photon count inducing a maximal response is 13.3 log photons cm(-2) s(-1) flickering at 2 Hz with 12% duty cycle. Our data could guide the design of healthier architectural lighting and better phototherapy devices for treating seasonal affective disorder and jet lag.


Subject(s)
Dark Adaptation/physiology , Light , Photic Stimulation/methods , Pupil/physiology , Retinal Ganglion Cells/metabolism , Rod Opsins/radiation effects , Vision, Ocular/physiology , Adult , Animals , Electrophysiological Phenomena , Female , Humans , Male , Mice , Pupil/radiation effects , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/radiation effects , Rod Opsins/metabolism , Young Adult
10.
PLoS One ; 10(3): e0123424, 2015.
Article in English | MEDLINE | ID: mdl-25822371

ABSTRACT

A direct projection from melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs) reaches the primary visual thalamus (dorsal lateral geniculate nucleus; dLGN). The significance of this melanopsin input to the visual system is only recently being investigated. One unresolved question is the degree to which neurons in the dLGN could use melanopsin to track dynamic changes in light intensity under light adapted conditions. Here we set out to address this question. We were able to present full field steps visible only to melanopsin by switching between rod-isoluminant 'yellow' and 'blue' lights in a mouse lacking cone function (Cnga3-/-). In the retina these stimuli elicited melanopsin-like responses from a subset of ganglion cells. When presented to anaesthetised mice, we found that ~25-30% of visually responsive neurones in the contralateral dLGN responded to these melanopsin-isolating steps with small increases in firing rate. Such responses could be elicited even with fairly modest increases in effective irradiance (32% Michelson contrast for melanopsin). These melanopsin-driven responses were apparent at bright backgrounds (corresponding to twilight-daylight conditions), but their threshold irradiance was strongly dependent upon prior light exposure when stimuli were superimposed on a spectrally neutral ramping background light. While both onset and offset latencies were long for melanopsin-derived responses compared to those evoked by rods, there was great variability in these parameters with some cells responding to melanopsin steps in <1 s. These data indicate that a subset of dLGN units can employ melanopsin signals to detect modest changes in irradiance under photopic conditions.


Subject(s)
Geniculate Bodies/metabolism , Geniculate Bodies/physiology , Light Signal Transduction/physiology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/physiology , Rod Opsins/metabolism , Animals , Light , Mice , Photic Stimulation/methods , Retina/metabolism , Retina/physiology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/physiology , Thalamus/metabolism , Thalamus/physiology
11.
PLoS One ; 9(9): e108209, 2014.
Article in English | MEDLINE | ID: mdl-25251771

ABSTRACT

Mammals contain 1 melanopsin (Opn4) gene that is expressed in a subset of retinal ganglion cells to serve as a photopigment involved in non-image-forming vision such as photoentrainment of circadian rhythms. In contrast, most nonmammalian vertebrates possess multiple melanopsins that are distributed in various types of retinal cells; however, their functions remain unclear. We previously found that the lamprey has only 1 type of mammalian-like melanopsin gene, which is similar to that observed in mammals. Here we investigated the molecular properties and localization of melanopsin in the lamprey and other cyclostome hagfish retinas, which contribute to visual functions including image-forming vision and mainly to non-image-forming vision, respectively. We isolated 1 type of mammalian-like melanopsin cDNA from the eyes of each species. We showed that the recombinant lamprey melanopsin was a blue light-sensitive pigment and that both the lamprey and hagfish melanopsins caused light-dependent increases in calcium ion concentration in cultured cells in a manner that was similar to that observed for mammalian melanopsins. We observed that melanopsin was distributed in several types of retinal cells, including horizontal cells and ganglion cells, in the lamprey retina, despite the existence of only 1 melanopsin gene in the lamprey. In contrast, melanopsin was almost specifically distributed to retinal ganglion cells in the hagfish retina. Furthermore, we found that the melanopsin-expressing horizontal cells connected to the rhodopsin-containing short photoreceptor cells in the lamprey. Taken together, our findings suggest that in cyclostomes, the global distribution of melanopsin in retinal cells might not be related to the melanopsin gene number but to the extent of retinal contribution to visual function.


Subject(s)
Hagfishes/physiology , Lampreys/physiology , Retina/ultrastructure , Rod Opsins/analysis , Animals , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Hagfishes/genetics , Lampreys/genetics , Retina/metabolism , Rod Opsins/genetics , Rod Opsins/metabolism , Vision, Ocular
12.
Curr Eye Res ; 39(12): 1221-31, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25006880

ABSTRACT

PURPOSE: RPE65, a retinal pigment epithelium-specific 65-kDa protein, plays a critical role in the visual cycle of the eye. Rpe65(-/-) mice develop vision loss due to a lack of 11-cis-retinal, degradation of M-opsin and mislocalization of S-opsin. Several studies have suggested that 9-cis-ß-carotene, a precursor of 9-cis-retinal and all-trans-retinal, could have therapeutic applications in vision loss. We therefore examined whether Dunaliella bardawil, a 9-cis-ß-carotene-rich alga, protects against the degradation of M-opsin using Rpe65(-/-) mouse retinal explant cultures. METHODS: The eyes of three-week-old Rpe65(-/-) and C57BL/6 J mice were enucleated, and the corneas were removed. The eyecups were incubated with culture medium in the absence or presence of D. bardawil for 6 h to 4 days. Localizations of M-opsin proteins in the retina were observed immunohistochemically. Expression levels of M-opsin, S-opsin and rhodopsin proteins were evaluated by Western blot analysis. RESULTS: In C57BL/6 J mouse retina, no change was observed in localization and expression levels of M-opsin in the explant culture system. In Rpe65(-/-) mouse retina, the amount of M-opsin protein was decreased in the photoreceptor outer segment after 6 h to 4 days of culture. However, the presence of D. bardawil significantly ameliorated this decrease. In contrast, expression levels of S-opsin and rhodopsin were unchanged in the presence of the explant culture. CONCLUSIONS: These results demonstrate that D. bardawil treatment protects against M-opsin degradation in Rpe65(-/-) mouse retina and suggest that D. bardawil has therapeutic potential for retinal degeneration caused by Rpe65 gene mutation, such as Leber congenital amaurosis and retinitis pigmentosa.


Subject(s)
Chlorophyta/chemistry , Plant Extracts/pharmacology , Retina/metabolism , Retinal Degeneration/prevention & control , Rod Opsins/metabolism , beta Carotene/chemistry , cis-trans-Isomerases/physiology , Animals , Blotting, Western , Cone Opsins/metabolism , Fluorescent Antibody Technique, Indirect , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/metabolism , Rhodopsin/metabolism
13.
Exp Eye Res ; 122: 65-76, 2014 May.
Article in English | MEDLINE | ID: mdl-24698764

ABSTRACT

Mutations in myosin VIIa (MYO7A) cause Usher Syndrome 1B (USH1B), a disease characterized by the combination of sensorineural hearing loss and visual impairment termed retinitis pigmentosa (RP). Although the shaker-1 mouse model of USH1B exists, only minor defects in the retina have been observed during its lifespan. Previous studies of the zebrafish mariner mutant, which also carries a mutation in myo7aa, revealed balance and hearing defects in the mutants but the retinal phenotype has not been described. We found elevated cell death in the outer nuclear layer (ONL) of myo7aa(-/-) mutants. While myo7aa(-/-) mutants retained visual behaviors in the optokinetic reflex (OKR) assay, electroretinogram (ERG) recordings revealed a significant decrease in both a- and b-wave amplitudes in mutant animals, but not a change in ERG threshold sensitivity. Immunohistochemistry showed mislocalization of rod and blue cone opsins and reduced expression of rod-specific markers in the myo7aa(-/-) ONL, providing further evidence that the photoreceptor degeneration observed represents the initial stages of the RP. Further, constant light exposure resulted in widespread photoreceptor degeneration and the appearance of large holes in the retinal pigment epithelium (RPE). No differences were observed in the retinomotor movements of the photoreceptors or in melanosome migration within the RPE, suggesting that myo7aa(-/-) does not function in these processes in teleosts. These results indicate that the zebrafish myo7aa(-/-) mutant is a useful animal model for the RP seen in humans with USH1B.


Subject(s)
Codon, Nonsense , Myosins/genetics , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Cell Death , Dark Adaptation , Disease Models, Animal , Electroretinography , Immunohistochemistry , In Situ Nick-End Labeling , Light , Melanosomes/physiology , Microscopy, Electron, Transmission , Myosin VIIa , Nystagmus, Optokinetic/physiology , Photoreceptor Cells, Vertebrate/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/physiopathology , Retinal Rod Photoreceptor Cells/metabolism , Rod Opsins/metabolism , Usher Syndromes/genetics , Usher Syndromes/metabolism , Usher Syndromes/pathology
14.
PLoS One ; 7(9): e43787, 2012.
Article in English | MEDLINE | ID: mdl-23028470

ABSTRACT

Melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs) are the only functional photoreceptive cells in the eye of newborn mice. Through postnatal day 9, in the absence of functional rods and cones, these ipRGCs mediate a robust avoidance behavior to a light source, termed negative phototaxis. To determine whether this behavior is associated with an aversive experience in neonatal mice, we characterized light-induced vocalizations and patterns of neuronal activation in regions of the brain involved in the processing of aversive and painful stimuli. Light evoked distinct melanopsin-dependent ultrasonic vocalizations identical to those emitted under stressful conditions, such as isolation from the litter. In contrast, light did not evoke the broad-spectrum calls elicited by acute mechanical pain. Using markers of neuronal activation, we found that light induced the immediate-early gene product Fos in the posterior thalamus, a brain region associated with the enhancement of responses to mechanical stimulation of the dura by light, and thought to be the basis for migrainous photophobia. Additionally, light induced the phosphorylation of extracellular-related kinase (pERK) in neurons of the central amygdala, an intracellular signal associated with the processing of the aversive aspects of pain. However, light did not activate Fos expression in the spinal trigeminal nucleus caudalis, the primary receptive field for painful stimulation to the head. We conclude that these light-evoked vocalizations and the distinct pattern of brain activation in neonatal mice are consistent with a melanopsin-dependent neural pathway involved in processing light as an aversive but not acutely painful stimulus.


Subject(s)
Light , Retinal Ganglion Cells/metabolism , Rod Opsins/metabolism , Vocalization, Animal/physiology , Amygdala/metabolism , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Photic Stimulation , Proto-Oncogene Proteins c-fos/metabolism , Thalamus/metabolism , Trigeminal Ganglion/metabolism , Vision, Ocular/physiology
16.
Nature ; 470(7333): 259-63, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21270798

ABSTRACT

In the vertebrate retina, establishment of precise synaptic connections among distinct retinal neuron cell types is critical for processing visual information and for accurate visual perception. Retinal ganglion cells (RGCs), amacrine cells and bipolar cells establish stereotypic neurite arborization patterns to form functional neural circuits in the inner plexiform layer (IPL), a laminar region that is conventionally divided into five major parallel sublaminae. However, the molecular mechanisms governing distinct retinal subtype targeting to specific sublaminae within the IPL remain to be elucidated. Here we show that the transmembrane semaphorin Sema6A signals through its receptor PlexinA4 (PlexA4) to control lamina-specific neuronal stratification in the mouse retina. Expression analyses demonstrate that Sema6A and PlexA4 proteins are expressed in a complementary fashion in the developing retina: Sema6A in most ON sublaminae and PlexA4 in OFF sublaminae of the IPL. Mice with null mutations in PlexA4 or Sema6A exhibit severe defects in stereotypic lamina-specific neurite arborization of tyrosine hydroxylase (TH)-expressing dopaminergic amacrine cells, intrinsically photosensitive RGCs (ipRGCs) and calbindin-positive cells in the IPL. Sema6A and PlexA4 genetically interact in vivo for the regulation of dopaminergic amacrine cell laminar targeting. Therefore, neuronal targeting to subdivisions of the IPL in the mammalian retina is directed by repulsive transmembrane guidance cues present on neuronal processes.


Subject(s)
Cell Membrane/metabolism , Neurons/cytology , Neurons/metabolism , Retina/cytology , Retina/metabolism , Semaphorins/metabolism , Signal Transduction , Amacrine Cells/enzymology , Amacrine Cells/metabolism , Animals , Calbindins , Dopamine/metabolism , Gene Expression Profiling , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nerve Tissue Proteins , Neurites/metabolism , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Retina/embryology , Retinal Ganglion Cells/metabolism , Rod Opsins/metabolism , S100 Calcium Binding Protein G/metabolism , Semaphorins/deficiency , Semaphorins/genetics , Tyrosine 3-Monooxygenase/metabolism
18.
Brain ; 133(Pt 8): 2426-38, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20659957

ABSTRACT

Mitochondrial optic neuropathies, that is, Leber hereditary optic neuropathy and dominant optic atrophy, selectively affect retinal ganglion cells, causing visual loss with relatively preserved pupillary light reflex. The mammalian eye contains a light detection system based on a subset of retinal ganglion cells containing the photopigment melanopsin. These cells give origin to the retinohypothalamic tract and support the non-image-forming visual functions of the eye, which include the photoentrainment of circadian rhythms, light-induced suppression of melatonin secretion and pupillary light reflex. We studied the integrity of the retinohypothalamic tract in five patients with Leber hereditary optic neuropathy, in four with dominant optic atrophy and in nine controls by testing the light-induced suppression of nocturnal melatonin secretion. This response was maintained in optic neuropathy subjects as in controls, indicating that the retinohypothalamic tract is sufficiently preserved to drive light information detected by melanopsin retinal ganglion cells. We then investigated the histology of post-mortem eyes from two patients with Leber hereditary optic neuropathy and one case with dominant optic atrophy, compared with three age-matched controls. On these retinas, melanopsin retinal ganglion cells were characterized by immunohistochemistry and their number and distribution evaluated by a new protocol. In control retinas, we show that melanopsin retinal ganglion cells are lost with age and are more represented in the parafoveal region. In patients, we demonstrate a relative sparing of these cells compared with the massive loss of total retinal ganglion cells, even in the most affected areas of the retina. Our results demonstrate that melanopsin retinal ganglion cells resist neurodegeneration due to mitochondrial dysfunction and maintain non-image-forming functions of the eye in these visually impaired patients. We also show that in normal human retinas, these cells are more concentrated around the fovea and are lost with ageing. The current results provide a plausible explanation for the preservation of pupillary light reaction despite profound visual loss in patients with mitochondrial optic neuropathy, revealing the robustness of melanopsin retinal ganglion cells to a metabolic insult and opening the question of mechanisms that might protect these cells.


Subject(s)
Nerve Degeneration/physiopathology , Optic Atrophy, Autosomal Dominant/physiopathology , Optic Atrophy, Hereditary, Leber/physiopathology , Retinal Ganglion Cells/physiology , Rod Opsins/metabolism , Visual Pathways/physiopathology , Adult , Aged, 80 and over , Aging/pathology , Aging/physiology , Case-Control Studies , Female , Humans , Hypothalamus/pathology , Hypothalamus/physiopathology , Male , Middle Aged , Mitochondrial Diseases/pathology , Mitochondrial Diseases/physiopathology , Nerve Degeneration/pathology , Optic Atrophy, Autosomal Dominant/pathology , Optic Atrophy, Hereditary, Leber/pathology , Retina/pathology , Retina/physiopathology , Retinal Ganglion Cells/pathology , Visual Pathways/pathology
19.
Invest Ophthalmol Vis Sci ; 49(3): 1126-35, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18326740

ABSTRACT

PURPOSE: To define rod and cone function further in terms of visual cycle mechanism, the retinal phenotype resulting from Rpe65 (retinoid isomerase I) deficiency in Nrl(-)(/)(-) mice having a single class of photoreceptors resembling wild-type cones was characterized and outcomes of retinoid supplementation evaluated. METHODS: Rpe65(-)(/)(-)/Nrl(-)(/)(-) mice were generated by breeding Rpe65(-)(/)(-) and Nrl(-)(/)(-) strains. Retinal histology, protein expression, retinoid content, and electroretinographic (ERG) responses were evaluated before and after treatment with 11-cis retinal by intraperitoneal injection. Results Retinas of young Rpe65(-)(/-)/Nrl(-)(/-) mice exhibited normal lamination, but lacked intact photoreceptor outer segments at all ages examined. Rpe65, Nrl, and rhodopsin were not detected, and S-opsin and M/L-opsin levels were reduced. Retinyl esters were the only retinoids present. In contrast, Nrl(-)(/)(-) mice exhibited decreased levels of retinaldehydes and retinyl esters, and elevated levels of retinols. ERG responses were elicited from Rpe65(-)(/-)/Nrl(-)(/-) mice only at the two highest intensities over a 4-log-unit range. Significant retinal thinning and outer nuclear layer loss occurred in Rpe65(-)(/-)/Nrl(-)(/-) mice with aging. Administration of exogenous 11-cis retinal did not rescue retinal morphology or markedly improve ERG responses. CONCLUSIONS: The findings provide clarification of reported cone loss of function in Rpe65(-)(/-)/Nrl(-)(/-) mice, now showing that chromophore absence results in destabilized cone outer segments and rapid retinal degeneration. The data support the view that rod-dominant retinas do not have a cone-specific mechanism for 11-cis retinal synthesis and have potential significance for therapeutic strategies for rescue of cone-rich retinal regions affected by disease in the aging human population.


Subject(s)
Basic-Leucine Zipper Transcription Factors/physiology , Carrier Proteins/physiology , Eye Proteins/physiology , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Degeneration/metabolism , Retinaldehyde/biosynthesis , Animals , Blotting, Western , Chromatography, High Pressure Liquid , Dark Adaptation , Electroretinography , Female , Fluorescent Antibody Technique, Indirect , Genotype , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/physiopathology , Retinal Degeneration/drug therapy , Retinal Degeneration/physiopathology , Retinaldehyde/administration & dosage , Retinoids/metabolism , Rod Opsins/metabolism , cis-trans-Isomerases
20.
Invest Ophthalmol Vis Sci ; 48(11): 5168-77, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17962470

ABSTRACT

PURPOSE: Oxidative stress has been proposed as a major pathogenic factor in age-related macular degeneration (AMD), the leading cause of vision loss among elderly people of western European ancestry. Lutein (LUT) and zeaxanthin (ZEA), major components in macular pigment, are among the retinal antioxidants. Though xanthophyll intake may reduce the likelihood of having advanced AMD, direct evidence of neuroprotection is lacking. Prior work has shown that docosahexaenoic acid (DHA), the major polyunsaturated fatty acid in the retina, delays apoptosis and promotes differentiation of photoreceptors. This study was conducted to investigate whether LUT, ZEA, and beta-carotene (BC), major dietary carotenoids protect photoreceptors from oxidative stress and whether this protection is synergistic with that of DHA. METHODS: Pure rat retinal neurons in culture, supplemented with LUT, ZEA, or BC, with or without DHA, were subjected to oxidative stress induced with paraquat and hydrogen peroxide. Apoptosis, preservation of mitochondrial membrane potential, cytochrome c translocation, and opsin expression were evaluated. RESULTS: Pretreatment with DHA, LUT, ZEA, and BC reduced oxidative stress-induced apoptosis in photoreceptors, preserved mitochondrial potential, and prevented cytochrome c release from mitochondria. ZEA and LUT also enhanced photoreceptor differentiation. In control cultures, photoreceptors failed to grow their characteristic outer segments; addition of DHA, ZEA, or LUT increased opsin expression and promoted the development of outer-segment-like processes. CONCLUSIONS: These results show for the first time the direct neuroprotection of photoreceptors by xanthophylls and suggest that ZEA and LUT, along with DHA, are important environmental influences that together promote photoreceptor survival and differentiation.


Subject(s)
Apoptosis/drug effects , Docosahexaenoic Acids/pharmacology , Lutein/pharmacology , Oxidative Stress/drug effects , Photoreceptor Cells, Vertebrate/cytology , Xanthophylls/pharmacology , Animals , Cell Differentiation/drug effects , Cell Survival , Cytochromes c/metabolism , Cytoprotection , Fluorescent Antibody Technique, Indirect , Hydrogen Peroxide/toxicity , Membrane Potentials/drug effects , Mitochondria/physiology , Paraquat/toxicity , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Rats , Rats, Wistar , Rod Opsins/metabolism , Zeaxanthins
SELECTION OF CITATIONS
SEARCH DETAIL