Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(21): e2404763121, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38743626

RESUMEN

Congenital stationary night blindness (CSNB) is an inherited retinal disease that causes a profound loss of rod sensitivity without severe retinal degeneration. One well-studied rhodopsin point mutant, G90D-Rho, is thought to cause CSNB because of its constitutive activity in darkness causing rod desensitization. However, the nature of this constitutive activity and its precise molecular source have not been resolved for almost 30 y. In this study, we made a knock-in (KI) mouse line with a very low expression of G90D-Rho (equal in amount to ~0.1% of normal rhodopsin, WT-Rho, in WT rods), with the remaining WT-Rho replaced by REY-Rho, a mutant with a very low efficiency of activating transducin due to a charge reversal of the highly conserved ERY motif to REY. We observed two kinds of constitutive noise: one being spontaneous isomerization (R*) of G90D-Rho at a molecular rate (R* s-1) 175-fold higher than WT-Rho and the other being G90D-Rho-generated dark continuous noise comprising low-amplitude unitary events occurring at a very high molecular rate equivalent in effect to ~40,000-fold of R* s-1 from WT-Rho. Neither noise type originated from G90D-Opsin because exogenous 11-cis-retinal had no effect. Extrapolating the above observations at low (0.1%) expression of G90D-Rho to normal disease exhibited by a KI mouse model with RhoG90D/WTand RhoG90D/G90D genotypes predicts the disease condition very well quantitatively. Overall, the continuous noise from G90D-Rho therefore predominates, constituting the major equivalent background light causing rod desensitization in CSNB.


Asunto(s)
Enfermedades Hereditarias del Ojo , Enfermedades Genéticas Ligadas al Cromosoma X , Miopía , Ceguera Nocturna , Rodopsina , Animales , Ceguera Nocturna/genética , Ceguera Nocturna/metabolismo , Enfermedades Hereditarias del Ojo/genética , Enfermedades Hereditarias del Ojo/metabolismo , Ratones , Rodopsina/genética , Rodopsina/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Miopía/genética , Miopía/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Oscuridad , Transducina/genética , Transducina/metabolismo , Técnicas de Sustitución del Gen , Modelos Animales de Enfermedad
2.
Proc Natl Acad Sci U S A ; 120(52): e2315282120, 2023 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-38109525

RESUMEN

Intrinsically photosensitive retinal ganglion cells (ipRGCs) serve as primary photoceptors by expressing the photopigment, melanopsin, and also as retinal relay neurons for rod and cone signals en route to the brain, in both cases for the purpose of non-image vision as well as aspects of image vision. So far, six subtypes of ipRGCs (M1 through M6) have been characterized. Regarding their phototransduction mechanisms, we have previously found that, unconventionally, rhabdomeric (microvillous) and ciliary signaling motifs co-exist within a given M1-, M2-, and M4-ipRGC, with the first mechanism involving PLCß4 and TRPC6,7 channels and the second involving cAMP and HCN channels. We have now examined M3-, M5-, and M6-cells and found that each cell likewise uses both signaling pathways for phototransduction, despite differences in the percentage representation by each pathway in a given ipRGC subtype for bright-flash responses (and saturated except for M6-cells). Generally, M3- and M5-cells show responses quite similar in kinetics to M2-responses, and M6-cell responses resemble broadly those of M1-cells although much lower in absolute sensitivity and amplitude. Therefore, similar to rod and cone subtypes in image vision, ipRGC subtypes possess the same phototransduction mechanism(s) even though they do not show microvilli or cilia morphologically.


Asunto(s)
Neuronas Retinianas , Visión Ocular , Fototransducción/fisiología , Células Ganglionares de la Retina/fisiología , Células Fotorreceptoras Retinianas Conos/metabolismo , Neuronas Retinianas/metabolismo , Opsinas de Bastones/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(1): e2216599120, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36584299

RESUMEN

Nonimage-forming vision in mammals is mediated primarily by melanopsin (OPN4)-expressing, intrinsically photosensitive retinal ganglion cells (ipRGCs). In mouse M1-ipRGCs, melanopsin predominantly activates, via Gαq,11,14, phospholipase C-ß4 to open transient receptor 6 (TRPC6) and TRPC7 channels. In M2- and M4-ipRGCs, however, a prominent phototransduction mechanism involves the opening of hyperpolarization- and cyclic nucleotide-gated channels via cyclic nucleotide, although the upstream steps remain uncertain. We report here experiments, primarily on M4-ipRGCs, with photo-uncaging of cyclic nucleotides and virally expressed CNGA2 channels to conclude that the second messenger is cyclic adenosine monophosphate (cAMP) - very surprising considering that cyclic guanosine monophosphate (cGMP) is used in almost all cyclic nucleotide-mediated phototransduction mechanisms across the animal kingdom. We further found that the upstream G protein is likewise Gq, which via its Gßγ subunits directly activates adenylyl cyclase (AC). Our findings are a demonstration in a native cell of a cross-motif GPCR signaling pathway from Gq directly to AC with a specific function.


Asunto(s)
Adenilil Ciclasas , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Fototransducción , Células Ganglionares de la Retina , Animales , Ratones , Adenilil Ciclasas/genética , Adenilil Ciclasas/metabolismo , Fototransducción/fisiología , Mamíferos/metabolismo , Nucleótidos Cíclicos/metabolismo , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/fisiología , Opsinas de Bastones/metabolismo , Transducción de Señal/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(32): e2121225119, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35914143

RESUMEN

G protein-coupled receptor (GPCR) signaling is ubiquitous. As an archetype of this signaling motif, rod phototransduction has provided many fundamental, quantitative details, including a dogma that one active GPCR molecule activates a substantial number of downstream G protein/enzyme effector complexes. However, rod phototransduction is light-activated, whereas GPCR pathways are predominantly ligand-activated. Here, we report a detailed study of the ligand-triggered GPCR pathway in mammalian olfactory transduction, finding that an odorant-receptor molecule when (one-time) complexed with its most effective odorants produces on average much less than one downstream effector. Further experiments gave a nominal success probability of tentatively ∼10-4 (more conservatively, ∼10-2 to ∼10-5). This picture is potentially more generally representative of GPCR signaling than is rod phototransduction, constituting a paradigm shift.


Asunto(s)
Ligandos , Odorantes , Receptores Acoplados a Proteínas G , Receptores Odorantes , Transducción de Señal , Olfato , Animales , Fototransducción , Mamíferos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Odorantes/metabolismo , Células Fotorreceptoras Retinianas Bastones
5.
Nat Commun ; 13(1): 2862, 2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35606344

RESUMEN

From mouse to primate, there is a striking discontinuity in our current understanding of the neural coding of motion direction. In non-primate mammals, directionally selective cell types and circuits are a signature feature of the retina, situated at the earliest stage of the visual process. In primates, by contrast, direction selectivity is a hallmark of motion processing areas in visual cortex, but has not been found in the retina, despite significant effort. Here we combined functional recordings of light-evoked responses and connectomic reconstruction to identify diverse direction-selective cell types in the macaque monkey retina with distinctive physiological properties and synaptic motifs. This circuitry includes an ON-OFF ganglion cell type, a spiking, ON-OFF polyaxonal amacrine cell and the starburst amacrine cell, all of which show direction selectivity. Moreover, we discovered that macaque starburst cells possess a strong, non-GABAergic, antagonistic surround mediated by input from excitatory bipolar cells that is critical for the generation of radial motion sensitivity in these cells. Our findings open a door to investigation of a precortical circuitry that computes motion direction in the primate visual system.


Asunto(s)
Conectoma , Macaca , Retina , Células Amacrinas/fisiología , Animales , Potenciales Evocados Visuales/fisiología , Macaca/fisiología , Mamíferos , Ratones , Primates/fisiología , Retina/fisiología , Células Ganglionares de la Retina/fisiología , Sinapsis/fisiología
6.
Sci Rep ; 11(1): 23424, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34873237

RESUMEN

Intrinsically-photosensitive retinal ganglion cells (ipRGCs) are non-rod/non-cone retinal photoreceptors expressing the visual pigment, melanopsin, to detect ambient irradiance for various non-image-forming visual functions. The M1-subtype, amongst the best studied, mediates primarily circadian photoentrainment and pupillary light reflex. Their intrinsic light responses are more prolonged than those of rods and cones even at the single-photon level, in accordance with the typically slower time course of non-image-forming vision. The short (OPN4S) and long (OPN4L) alternatively-spliced forms of melanopsin proteins are both present in M1-ipRGCs, but their functional difference is unclear. We have examined this point by genetically removing the Opn4 gene (Opn4-/-) in mouse and re-expressing either OPN4S or OPN4L singly in Opn4-/- mice by using adeno-associated virus, but found no obvious difference in their intrinsic dim-flash responses. Previous studies have indicated that two dominant slow steps in M1-ipRGC phototransduction dictate these cells' intrinsic dim-flash-response kinetics, with time constants (τ1 and τ2) at room temperature of ~ 2 s and ~ 20 s, respectively. Here we found that melanopsin inactivation by phosphorylation or by ß-arrestins may not be one of these two steps, because their genetic disruptions did not prolong the two time constants or affect the response waveform. Disruption of GAP (GTPase-Activating-Protein) activity on the effector enzyme, PLCß4, in M1-ipRGC phototransduction to slow down G-protein deactivation also did not prolong the response decay, but caused its rising phase to become slightly sigmoidal by giving rise to a third time constant, τ3, of ~ 2 s (room temperature). This last observation suggests that GAP-mediated G-protein deactivation does partake in the flash-response termination, although normally with a time constant too short to be visible in the response waveform.


Asunto(s)
Células Fotorreceptoras Retinianas Conos/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Ritmo Circadiano/fisiología , Dependovirus , Inyecciones Intravítreas , Cinética , Luz , Fototransducción , Ratones , Ratones Transgénicos , Mutación , Neurociencias , Fosforilación , Opsinas de Bastones/química , Transducción de Señal , Visión Ocular , beta-Arrestinas/química
7.
Sci Adv ; 6(51)2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33328242

RESUMEN

Almost a century ago, Stiles and Crawford reported that the human eye is more sensitive to light entering through the pupil center than through its periphery (Stiles-Crawford effect). This psychophysical phenomenon, later found to correlate with photoreceptor orientation toward the pupil, was dynamically phototropic, adjustable within days to an eccentrically displaced pupil. For decades, this phototropism has been speculated to involve coordinated movements of the rectilinear photoreceptor outer and inner segments. We report here that, unexpectedly, the murine photoreceptor outer segment has a seemingly light-independent orientation, but the inner segment's orientation undergoes light-dependent movement, giving rise to nonrectilinear outer and inner segments in adult mice born and reared in darkness. Light during an early critical period (~P0 to P8), however, largely sets the correct photoreceptor orientation permanently afterward. Unexpectedly, abolishing rod and cone phototransductions did not mimic darkness in early life, suggesting photosignaling extrinsic to rods and cones is involved.

8.
Curr Biol ; 30(24): 4921-4931.e5, 2020 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-33065015

RESUMEN

Retinal rod and cone photoreceptors mediate vision in dim and bright light, respectively, by transducing absorbed photons into neural electrical signals. Their phototransduction mechanisms are essentially identical. However, one difference is that, whereas a rod visual pigment remains stable in darkness, a cone pigment has some tendency to dissociate spontaneously into apo-opsin and retinal (the chromophore) without isomerization. This cone-pigment property is long known but has mostly been overlooked. Importantly, because apo-opsin has weak constitutive activity, it triggers transduction to produce electrical noise even in darkness. Currently, the precise dark apo-opsin contents across cone subtypes are mostly unknown, as are their dark activities. We report here a study of goldfish red (L), green (M), and blue (S) cones, finding with microspectrophotometry widely different apo-opsin percentages in darkness, being ∼30% in L cones, ∼3% in M cones, and negligible in S cones. L and M cones also had higher dark apo-opsin noise than holo-pigment thermal isomerization activity. As such, given the most likely low signal amplification at the pigment-to-transducin/phosphodiesterase phototransduction step, especially in L cones, apo-opsin noise may not be easily distinguishable from light responses and thus may affect cone vision near threshold.


Asunto(s)
Oscuridad , Fototransducción/fisiología , Opsinas/metabolismo , Células Fotorreceptoras Retinianas Conos/fisiología , Animales , Carpa Dorada , Modelos Animales , Técnicas de Placa-Clamp , Estimulación Luminosa/métodos , Células Fotorreceptoras Retinianas Conos/efectos de la radiación , Análisis de la Célula Individual
9.
Proc Natl Acad Sci U S A ; 117(37): 23033-23043, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32873651

RESUMEN

Numerous rhodopsin mutations have been implicated in night blindness and retinal degeneration, often with unclear etiology. D190N-rhodopsin (D190N-Rho) is a well-known inherited human mutation causing retinitis pigmentosa. Both higher-than-normal spontaneous-isomerization activity and misfolding/mistargeting of the mutant protein have been proposed as causes of the disease, but neither explanation has been thoroughly examined. We replaced wild-type rhodopsin (WT-Rho) in RhoD190N/WT mouse rods with a largely "functionally silenced" rhodopsin mutant to isolate electrical responses triggered by D190N-Rho activity, and found that D190N-Rho at the single-molecule level indeed isomerizes more frequently than WT-Rho by over an order of magnitude. Importantly, however, this higher molecular dark activity does not translate into an overall higher cellular dark noise, owing to diminished D190N-Rho content in the rod outer segment. Separately, we found that much of the degeneration and shortened outer-segment length of RhoD190N/WT mouse rods was not averted by ablating rod transducin in phototransduction-also consistent with D190N-Rho's higher isomerization activity not being the primary cause of disease. Instead, the low pigment content, shortened outer-segment length, and a moderate unfolded protein response implicate protein misfolding as the major pathogenic problem. Finally, D190N-Rho also provided some insight into the mechanism of spontaneous pigment excitation.


Asunto(s)
Degeneración Retiniana/metabolismo , Rodopsina/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Fototransducción/fisiología , Ratones , Mutación/fisiología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Retinitis Pigmentosa/metabolismo , Segmento Externo de la Célula en Bastón/metabolismo
10.
PLoS Biol ; 18(2): e3000630, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32040503

RESUMEN

Opsin3 (Opn3) is a transmembrane heptahelical G protein-coupled receptor (GPCR) with the potential to produce a nonvisual photoreceptive effect. Interestingly, anatomical profiling of GPCRs reveals that Opn3 mRNA is highly expressed in adipose tissue. The photosensitive functions of Opn3 in mammals are poorly understood, and whether Opn3 has a role in fat is entirely unknown. In this study, we found that Opn3-knockout (Opn3-KO) mice were prone to diet-induced obesity and insulin resistance. At the cellular level, Opn3-KO brown adipocytes cultured in darkness had decreased glucose uptake and lower nutrient-induced mitochondrial respiration than wild-type (WT) cells. Light exposure promoted mitochondrial activity and glucose uptake in WT adipocytes but not in Opn3-KO cells. Brown adipocytes carrying a defective mutation in Opn3's putative G protein-binding domain also exhibited a reduction in glucose uptake and mitochondrial respiration in darkness. Using RNA-sequencing, we identified several novel light-sensitive and Opn3-dependent molecular signatures in brown adipocytes. Importantly, direct exposure of brown adipose tissue (BAT) to light in living mice significantly enhanced thermogenic capacity of BAT, and this effect was diminished in Opn3-KO animals. These results uncover a previously unrecognized cell-autonomous, light-sensing mechanism in brown adipocytes via Opn3-GPCR signaling that can regulate fuel metabolism and mitochondrial respiration. Our work also provides a molecular basis for developing light-based treatments for obesity and its related metabolic disorders.


Asunto(s)
Adipocitos Marrones/metabolismo , Metabolismo Energético , Opsinas de Bastones/metabolismo , Tejido Adiposo Pardo/inervación , Animales , Dieta Alta en Grasa/efectos adversos , Regulación de la Expresión Génica , Glucosa/metabolismo , Resistencia a la Insulina , Luz , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mutación , Obesidad/genética , Obesidad/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Opsinas de Bastones/genética , Transducción de Señal , Termogénesis
11.
Proc Natl Acad Sci U S A ; 116(11): 5144-5153, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30796193

RESUMEN

G protein-coupled receptor (GPCR) signaling is crucial for many physiological processes. A signature of such pathways is high amplification, a concept originating from retinal rod phototransduction, whereby one photoactivated rhodopsin molecule (Rho*) was long reported to activate several hundred transducins (GT*s), each then activating a cGMP-phosphodiesterase catalytic subunit (GT*·PDE*). This high gain at the Rho*-to-GT* step has been challenged more recently, but estimates remain dispersed and rely on some nonintact rod measurements. With two independent approaches, one with an extremely inefficient mutant rhodopsin and the other with WT bleached rhodopsin, which has exceedingly weak constitutive activity in darkness, we obtained an estimate for the electrical effect from a single GT*·PDE* molecular complex in intact mouse rods. Comparing the single-GT*·PDE* effect to the WT single-photon response, both in Gcaps-/- background, gives an effective gain of only ∼12-14 GT*·PDE*s produced per Rho*. Our findings have finally dispelled the entrenched concept of very high gain at the receptor-to-G protein/effector step in GPCR systems.


Asunto(s)
Células Fotorreceptoras Retinianas Bastones/metabolismo , Transducina/metabolismo , Secuencias de Aminoácidos , Animales , GMP Cíclico/metabolismo , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Fototransducción , Ratones Transgénicos , Mutación/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Fotones , Rodopsina/química , Rodopsina/metabolismo
12.
Cell ; 175(3): 652-664.e12, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30270038

RESUMEN

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.


Asunto(s)
Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Células Ganglionares de la Retina/metabolismo , Visión Ocular , Animales , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Nucleótidos Cíclicos/metabolismo , Células Ganglionares de la Retina/fisiología , Canales Catiónicos TRPC/metabolismo
13.
Proc Natl Acad Sci U S A ; 115(21): 5570-5575, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29735665

RESUMEN

In mammalian olfactory transduction, odorants activate a cAMP-mediated signaling pathway that leads to the opening of cyclic nucleotide-gated (CNG), nonselective cation channels and depolarization. The Ca2+ influx through open CNG channels triggers an inward current through Ca2+-activated Cl channels (ANO2), which is expected to produce signal amplification. However, a study on an Ano2-/- mouse line reported no elevation in the behavioral threshold of odorant detection compared with wild type (WT). Subsequent studies by others on the same Ano2-/- line, nonetheless, found subtle defects in olfactory behavior and some abnormal axonal projections from the olfactory receptor neurons (ORNs) to the olfactory bulb. As such, the question regarding signal amplification by the Cl current in WT mouse remains unsettled. Recently, with suction-pipette recording, we have successfully separated in frog ORNs the CNG and Cl currents during olfactory transduction and found the Cl current to predominate in the response down to the threshold of action-potential signaling to the brain. For better comparison with the mouse data by others, we have now carried out similar current-separation experiments on mouse ORNs. We found that the Cl current clearly also predominated in the mouse olfactory response at signaling threshold, accounting for ∼80% of the response. In the absence of the Cl current, we expect the threshold stimulus to increase by approximately sevenfold.


Asunto(s)
Anoctaminas/fisiología , Encéfalo/fisiología , Calcio/farmacología , Cloruros/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/fisiología , Neuronas Receptoras Olfatorias/fisiología , Animales , Encéfalo/citología , AMP Cíclico/farmacología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Noqueados , Neuronas Receptoras Olfatorias/citología , Neuronas Receptoras Olfatorias/efectos de los fármacos , Técnicas de Placa-Clamp , Transducción de Señal/efectos de los fármacos , Olfato/efectos de los fármacos
14.
Curr Biol ; 27(12): 1791-1800.e5, 2017 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-28578927

RESUMEN

The mammalian pupillary light reflex (PLR) involves a bilateral brain circuit whereby afferent light signals in the optic nerve ultimately drive iris-sphincter-muscle contraction via excitatory cholinergic parasympathetic innervation [1, 2]. Additionally, the PLR in nocturnal and crepuscular sub-primate mammals has a "local" component in the isolated sphincter muscle [3-5], as in amphibians, fish, and bird [6-10]. In mouse, this local PLR requires the pigment melanopsin [5], originally found in intrinsically photosensitive retinal ganglion cells (ipRGCs) [11-19]. However, melanopsin's presence and effector pathway locally in the iris remain uncertain. The sphincter muscle itself may express melanopsin [5], or its cholinergic parasympathetic innervation may be modulated by suggested intraocular axonal collaterals of ipRGCs traveling to the eye's ciliary body or even to the iris [20-22]. Here, we show that the muscarinic receptor antagonist, atropine, eliminated the effect of acetylcholine (ACh), but not of light, on isolated mouse sphincter muscle. Conversely, selective genetic deletion of melanopsin in smooth muscle mostly removed the light-induced, but not the ACh-triggered, increase in isolated sphincter muscle's tension and largely suppressed the local PLR in vivo. Thus, sphincter muscle cells are bona fide, albeit unconventional, photoreceptors. We found melanopsin expression in a small subset of mouse iris sphincter muscle cells, with the light-induced contractile signal apparently spreading through gap junctions into neighboring muscle cells. Light and ACh share a common signaling pathway in sphincter muscle. In summary, our experiments have provided details of a photosignaling process in the eye occurring entirely outside the retina.


Asunto(s)
Acetilcolina/metabolismo , Atropina/farmacología , Iris/efectos de los fármacos , Luz , Antagonistas Muscarínicos/farmacología , Transducción de Señal , Animales , Ratones , Ratones Transgénicos , Estimulación Luminosa , Transducción de Señal/efectos de los fármacos
15.
Elife ; 62017 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-28186874

RESUMEN

Visual pigments can be spontaneously activated by internal thermal energy, generating noise that interferes with real-light detection. Recently, we developed a physicochemical theory that successfully predicts the rate of spontaneous activity of representative rod and cone pigments from their peak-absorption wavelength (λmax), with pigments having longer λmax being noisier. Interestingly, cone pigments may generally be ~25 fold noisier than rod pigments of the same λmax, possibly ascribed to an 'open' chromophore-binding pocket in cone pigments defined by the capability of chromophore-exchange in darkness. Here, we show in mice that the λmax-dependence of pigment noise could be extended even to a mutant pigment, E122Q-rhodopsin. Moreover, although E122Q-rhodopsin shows some cone-pigment-like characteristics, its noise remained quantitatively predictable by the 'non-open' nature of its chromophore-binding pocket as in wild-type rhodopsin. The openness/closedness of the chromophore-binding pocket is potentially a useful indicator of whether a pigment is intended for detecting dim or bright light.


Asunto(s)
Células Fotorreceptoras Retinianas Conos/fisiología , Pigmentos Retinianos/química , Pigmentos Retinianos/metabolismo , Células Fotorreceptoras Retinianas Bastones/fisiología , Animales , Sitios de Unión , Fenómenos Químicos , Ratones
16.
Proc Natl Acad Sci U S A ; 113(40): 11078-11087, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27647918

RESUMEN

Olfactory transduction in vertebrate olfactory receptor neurons (ORNs) involves primarily a cAMP-signaling cascade that leads to the opening of cyclic-nucleotide-gated (CNG), nonselective cation channels. The consequent Ca2+ influx triggers adaptation but also signal amplification, the latter by opening a Ca2+-activated Cl channel (ANO2) to elicit, unusually, an inward Cl current. Hence the olfactory response has inward CNG and Cl components that are in rapid succession and not easily separable. We report here success in quantitatively separating these two currents with respect to amplitude and time course over a broad range of odorant strengths. Importantly, we found that the Cl current is the predominant component throughout the olfactory dose-response relation, down to the threshold of signaling to the brain. This observation is very surprising given a recent report by others that the olfactory-signal amplification effected by the Ca2+-activated Cl current does not influence the behavioral olfactory threshold in mice.


Asunto(s)
Calcio/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Neuronas Receptoras Olfatorias/metabolismo , Olfato/fisiología , Animales , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Cloruros/metabolismo , AMP Cíclico/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Ratones , Odorantes/análisis , Neuronas Receptoras Olfatorias/fisiología , Transducción de Señal , Olfato/genética
17.
Proc Natl Acad Sci U S A ; 113(32): 9093-8, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27462111

RESUMEN

It is a deeply engrained notion that the visual pigment rhodopsin signals light as a monomer, even though many G protein-coupled receptors are now known to exist and function as dimers. Nonetheless, recent studies (albeit all in vitro) have suggested that rhodopsin and its chromophore-free apoprotein, R-opsin, may indeed exist as a homodimer in rod disk membranes. Given the overwhelmingly strong historical context, the crucial remaining question, therefore, is whether pigment dimerization truly exists naturally and what function this dimerization may serve. We addressed this question in vivo with a unique mouse line (S-opsin(+)Lrat(-/-)) expressing, transgenically, short-wavelength-sensitive cone opsin (S-opsin) in rods and also lacking chromophore to exploit the fact that cone opsins, but not R-opsin, require chromophore for proper folding and trafficking to the photoreceptor's outer segment. In R-opsin's absence, S-opsin in these transgenic rods without chromophore was mislocalized; in R-opsin's presence, however, S-opsin trafficked normally to the rod outer segment and produced functional S-pigment upon subsequent chromophore restoration. Introducing a competing R-opsin transmembrane helix H1 or helix H8 peptide, but not helix H4 or helix H5 peptide, into these transgenic rods caused mislocalization of R-opsin and S-opsin to the perinuclear endoplasmic reticulum. Importantly, a similar peptide-competition effect was observed even in WT rods. Our work provides convincing evidence for visual pigment dimerization in vivo under physiological conditions and for its role in pigment maturation and targeting. Our work raises new questions regarding a potential mechanistic role of dimerization in rhodopsin signaling.


Asunto(s)
Multimerización de Proteína , Pigmentos Retinianos/química , Animales , Retículo Endoplásmico/metabolismo , Ratones , Opsinas/química , Especies Reactivas de Oxígeno/metabolismo , Pigmentos Retinianos/fisiología
18.
J Comp Neurol ; 524(14): 2845-72, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-26972791

RESUMEN

The long-term goal of this research is to understand how retinal ganglion cells that express the photopigment melanopsin, also known as OPN4, contribute to vision in humans and other primates. Here we report the results of anatomical studies using our polyclonal antibody specifically against human melanopsin that confirm and extend previous descriptions of melanopsin cells in primates. In macaque and human retina, two distinct populations of melanopsin cells were identified based on dendritic stratification in either the inner or the outer portion of the inner plexiform layer (IPL). Variation in dendritic field size and cell density with eccentricity was confirmed, and dendritic spines, a new feature of melanopsin cells, were described. The spines were the sites of input from DB6 diffuse bipolar cell axon terminals to the inner stratifying type of melanopsin cells. The outer stratifying melanopsin type received inputs from DB6 bipolar cells via a sparse outer axonal arbor. Outer stratifying melanopsin cells also received inputs from axon terminals of dopaminergic amacrine cells. On the outer stratifying melanopsin cells, ribbon synapses from bipolar cells and conventional synapses from amacrine cells were identified in electron microscopic immunolabeling experiments. Both inner and outer stratifying melanopsin cell types were retrogradely labeled following tracer injection in the lateral geniculate nucleus (LGN). In addition, a method for targeting melanopsin cells for intracellular injection using their intrinsic fluorescence was developed. This technique was used to demonstrate that melanopsin cells were tracer coupled to amacrine cells and would be applicable to electrophysiological experiments in the future. J. Comp. Neurol. 524:2845-2872, 2016. © 2016 The Authors The Journal of Comparative Neurology Published by Wiley Periodicals, Inc.


Asunto(s)
Retina/citología , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Opsinas de Bastones/biosíntesis , Opsinas de Bastones/genética , Secuencia de Aminoácidos , Animales , Recuento de Células/métodos , Humanos , Macaca , Macaca fascicularis , Macaca nemestrina , Persona de Mediana Edad , Especificidad de la Especie
19.
Proc Natl Acad Sci U S A ; 112(42): 13093-8, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26392540

RESUMEN

The molecular circadian clocks in the mammalian retina are locally synchronized by environmental light cycles independent of the suprachiasmatic nuclei (SCN) in the brain. Unexpectedly, this entrainment does not require rods, cones, or melanopsin (OPN4), possibly suggesting the involvement of another retinal photopigment. Here, we show that the ex vivo mouse retinal rhythm is most sensitive to short-wavelength light but that this photoentrainment requires neither the short-wavelength-sensitive cone pigment [S-pigment or cone opsin (OPN1SW)] nor encephalopsin (OPN3). However, retinas lacking neuropsin (OPN5) fail to photoentrain, even though other visual functions appear largely normal. Initial evidence suggests that OPN5 is expressed in select retinal ganglion cells. Remarkably, the mouse corneal circadian rhythm is also photoentrainable ex vivo, and this photoentrainment likewise requires OPN5. Our findings reveal a light-sensing function for mammalian OPN5, until now an orphan opsin.


Asunto(s)
Córnea/fisiología , Proteínas de la Membrana/fisiología , Opsinas/fisiología , Retina/fisiología , Núcleo Supraquiasmático/fisiología , Animales , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Opsinas/genética , Rayos Ultravioleta
20.
Nat Commun ; 5: 4047, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24915161

RESUMEN

Many forms of blindness result from the dysfunction or loss of retinal photoreceptors. Induced pluripotent stem cells (iPSCs) hold great potential for the modelling of these diseases or as potential therapeutic agents. However, to fulfill this promise, a remaining challenge is to induce human iPSC to recreate in vitro key structural and functional features of the native retina, in particular the presence of photoreceptors with outer-segment discs and light sensitivity. Here we report that hiPSC can, in a highly autonomous manner, recapitulate spatiotemporally each of the main steps of retinal development observed in vivo and form three-dimensional retinal cups that contain all major retinal cell types arranged in their proper layers. Moreover, the photoreceptors in our hiPSC-derived retinal tissue achieve advanced maturation, showing the beginning of outer-segment disc formation and photosensitivity. This success brings us one step closer to the anticipated use of hiPSC for disease modelling and open possibilities for future therapies.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Fotorreceptoras de Vertebrados/fisiología , Retina/citología , Diferenciación Celular , Línea Celular , Humanos , Retina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...