Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Vis Neurosci ; 38: E014, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34652269

RESUMO

Intrinsically photosensitive retinal ganglion cells (ipRGCs) signal not only anterogradely to drive behavioral responses, but also retrogradely to some amacrine interneurons to modulate retinal physiology. We previously found that all displaced amacrine cells with spiking, tonic excitatory photoresponses receive gap-junction input from ipRGCs, but the connectivity patterns and functional roles of ipRGC-amacrine coupling remained largely unknown. Here, we injected PoPro1 fluorescent tracer into all six types of mouse ipRGCs to identify coupled amacrine cells, and analyzed the latter's morphological and electrophysiological properties. We also examined how genetically disrupting ipRGC-amacrine coupling affected ipRGC photoresponses. Results showed that ipRGCs couple with not just ON- and ON/OFF-stratified amacrine cells in the ganglion-cell layer as previously reported, but also OFF-stratified amacrine cells in both ganglion-cell and inner nuclear layers. M1- and M3-type ipRGCs couple mainly with ON/OFF-stratified amacrine cells, whereas the other ipRGC types couple almost exclusively with ON-stratified ones. ipRGCs transmit melanopsin-based light responses to at least 93% of the coupled amacrine cells. Some of the ON-stratifying ipRGC-coupled amacrine cells exhibit transient hyperpolarizing light responses. We detected bidirectional electrical transmission between an ipRGC and a coupled amacrine cell, although transmission was asymmetric for this particular cell pair, favoring the ipRGC-to-amacrine direction. We also observed electrical transmission between two amacrine cells coupled to the same ipRGC. In both scenarios of coupling, the coupled cells often spiked synchronously. While ipRGC-amacrine coupling somewhat reduces the peak firing rates of ipRGCs' intrinsic melanopsin-based photoresponses, it renders these responses more sustained and longer-lasting. In summary, ipRGCs' gap junctional network involves more amacrine cell types and plays more roles than previously appreciated.


Assuntos
Células Amácrinas , Opsinas de Bastonetes , Animais , Junções Comunicantes , Interneurônios , Camundongos , Retina , Células Ganglionares da Retina
2.
J Neurosci ; 36(15): 4209-17, 2016 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-27076420

RESUMO

In addition to rods and cones, mammals have inner retinal photoreceptors called intrinsically photosensitive retinal ganglion cells (ipRGCs), which use the photopigment melanopsin and mediate nonimage-forming visual responses, such as pupil reflexes and circadian entrainment. After photic activation, photopigments must be reverted to their dark state to be light-sensitive again. For rods and to some extent cones, photopigment regeneration depends on the retinoid cycle in the adjacent retinal pigment epithelium (RPE). By contrast, ipRGCs are far from the RPE, and previous work suggests that melanopsin is capable of light-dependent self-regeneration. Here, we used in vitro ipRGC recording and in vivo pupillometry to show that the RPE is required for normal melanopsin-based responses to prolonged light, especially at high stimulus intensities. Melanopsin-based photoresponses of rat ipRGCs were remarkably sustained when a functional RPE was attached to the retina, but became far more transient if the RPE was removed, or if the retinoid cycle was inhibited, or when Müller glia were poisoned. Similarly, retinoid cycle inhibition markedly reduced the steady-state amplitude of melanopsin-driven pupil reflexes in both mice and rats. However, melanopsin photoresponses in RPE-separated rat retinas became more sustained in the presence of an 11-cis-retinal analog. In conclusion, during prolonged illumination, melanopsin regeneration depends partly on 11-cis-retinal from the RPE, possibly imported via Müller cells. Implications for RPE-related eye diseases and the acne drug isotretinoin (a retinoid cycle inhibitor) are discussed. SIGNIFICANCE STATEMENT: Intrinsically photosensitive retinal ganglion cells (ipRGCs) contain the photopigment melanopsin and drive subconscious physiological responses to light, e.g., pupillary constriction and neuroendocrine regulation. In darkness, each photopigment molecule in ipRGCs, as well as rod/cone photoreceptors, contains 11-cis-retinal (a vitamin A derivative) and light isomerizes it to all-trans-retinal, which activates the photopigment. To make this photopigment excitable again,all-trans-retinal must be reisomerized to 11-cis-retinal. For rods and to some extent cones, this reisomerization occurs in the adjacent retinal pigment epithelium (RPE), but because ipRGCs are far from the RPE, they are thought to regenerate excitable melanopsin exclusively through RPE-independent means. Here, we present electrophysiological and behavioral evidence that ipRGCs depend on the RPE to continuously regenerate melanopsin during intense prolonged photostimulation.


Assuntos
Células Fotorreceptoras de Vertebrados/metabolismo , Retina/fisiologia , Retinoides/metabolismo , Animais , Eletrorretinografia , Isotretinoína/farmacologia , Camundongos , Neuroglia/efeitos dos fármacos , Neuroglia/fisiologia , Técnicas de Patch-Clamp , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Ratos , Ratos Long-Evans , Reflexo Pupilar/efeitos dos fármacos , Reflexo Pupilar/fisiologia , Retina/citologia , Retina/efeitos dos fármacos , Células Fotorreceptoras Retinianas Cones/efeitos dos fármacos , Células Fotorreceptoras Retinianas Cones/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/efeitos dos fármacos , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Retinaldeído/metabolismo , Opsinas de Bastonetes/metabolismo
3.
J Biol Chem ; 290(36): 22030-48, 2015 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-26100636

RESUMO

It is widely accepted that overactivation of NMDA receptors, resulting in calcium overload and consequent mitochondrial dysfunction in retinal ganglion neurons, plays a significant role in promoting neurodegenerative disorders such as glaucoma. Calcium has been shown to initiate a transient hyperpolarization of the mitochondrial membrane potential triggering a burst of reactive oxygen species leading to apoptosis. Strategies that enhance cell survival signaling pathways aimed at preventing this adverse hyperpolarization of the mitochondrial membrane potential may provide a novel therapeutic intervention in retinal disease. In the retina, brain-derived neurotrophic factor has been shown to be neuroprotective, and our group previously reported a PSD-95/PDZ-binding cyclic peptide (CN2097) that augments brain-derived neurotrophic factor-induced pro-survival signaling. Here, we examined the neuroprotective properties of CN2097 using an established retinal in vivo NMDA toxicity model. CN2097 completely attenuated NMDA-induced caspase 3-dependent and -independent cell death and PARP-1 activation pathways, blocked necrosis, and fully prevented the loss of long term ganglion cell viability. Although neuroprotection was partially dependent upon CN2097 binding to the PDZ domain of PSD-95, our results show that the polyarginine-rich transport moiety C-R(7), linked to the PDZ-PSD-95-binding cyclic peptide, was sufficient to mediate short and long term protection via a mitochondrial targeting mechanism. C-R(7) localized to mitochondria and was found to reduce mitochondrial respiration, mitochondrial membrane hyperpolarization, and the generation of reactive oxygen species, promoting survival of retinal neurons.


Assuntos
Potencial da Membrana Mitocondrial/efeitos dos fármacos , N-Metilaspartato/farmacologia , Peptídeos/farmacologia , Neurônios Retinianos/efeitos dos fármacos , Animais , Western Blotting , Morte Celular/efeitos dos fármacos , Proteína 4 Homóloga a Disks-Large , Agonistas de Aminoácidos Excitatórios/farmacologia , Guanilato Quinases/metabolismo , Células HEK293 , Humanos , Masculino , Proteínas de Membrana/metabolismo , Microscopia de Fluorescência , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Membranas Mitocondriais/efeitos dos fármacos , Membranas Mitocondriais/fisiologia , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Peptídeos/metabolismo , Peptídeos Cíclicos/metabolismo , Peptídeos Cíclicos/farmacologia , Ligação Proteica , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Retina/citologia , Retina/efeitos dos fármacos , Retina/metabolismo , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo , Neurônios Retinianos/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/fisiologia
4.
J Neurosci ; 34(5): 1942-8, 2014 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-24478373

RESUMO

In the developing mouse retina, spontaneous and light-driven activity shapes bipolar→ganglion cell glutamatergic synapse formation, beginning around the time of eye-opening (P12-P14) and extending through the first postnatal month. During this time, glutamate release can spill outside the synaptic cleft and possibly stimulate extrasynaptic NMDA-type glutamate receptors (NMDARs) on ganglion cells. Furthermore, the role of NMDARs during development may differ between ON and OFF bipolar synapses as in mature retina, where ON synapses reportedly include extrasynaptic NMDARs with GluN2B subunits. To better understand the function of glutamatergic synapses during development, we made whole-cell recordings of NMDAR-mediated responses, in vitro, from two types of genetically identified direction-selective ganglion cells (dsGCs): TRHR (thyrotropin-releasing hormone receptor) and Drd4 (dopamine receptor 4). Both dsGC types responded to puffed NMDA between P7 and P28; and both types exhibited robust light-evoked NMDAR-mediated responses at P14 and P28 that were quantified by conductance analysis during nicotinic and GABA(A) receptor blockade. For a given cell type and at a given age, ON and OFF bipolar cell inputs evoked similar NMDAR-mediated responses, suggesting that ON-versus-OFF differences in mature retina do not apply to the cell types or ages studied here. At P14, puff- and light-evoked NMDAR-mediated responses in both dsGCs were partially blocked by the GluN2B antagonist ifenprodil, whereas at P28 only TRHR cells remained ifenprodil-sensitive. NMDARs contribute at both ON and OFF bipolar cell synapses during a period of robust activity-dependent synaptic development, with declining GluN2B involvement over time in specific ganglion cell types.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Células Bipolares da Retina/metabolismo , Células Ganglionares da Retina/fisiologia , Sinapses/fisiologia , Fatores Etários , Animais , Animais Recém-Nascidos , Agonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Proteínas de Fluorescência Verde/genética , Técnicas In Vitro , Luz , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , N-Metilaspartato/farmacologia , Neurotransmissores/farmacologia , Técnicas de Patch-Clamp , Retina/citologia , Células Ganglionares da Retina/citologia , Vias Visuais/efeitos dos fármacos , Vias Visuais/fisiologia
5.
J Neurophysiol ; 114(5): 2955-66, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26400257

RESUMO

Intrinsically photosensitive retinal ganglion cells (ipRGCs) mediate both image-forming vision and non-image-forming visual responses such as pupillary constriction and circadian photoentrainment. Five types of ipRGCs, named M1-M5, have been discovered in rodents. To further investigate their photoresponse properties, we made multielectrode array spike recordings from rat ipRGCs, classified them into M1, M2/M4, and M3/M5 clusters, and measured their intrinsic, melanopsin-based responses to single and flickering light pulses. Results showed that ipRGC spiking can track flickers up to ∼0.2 Hz in frequency and that flicker intervals between 5 and 14 s evoke the most spikes. We also learned that melanopsin's integration time is intensity and cluster dependent. Using these data, we constructed a mathematical model for each cluster's intrinsic photoresponse. We found that the data for the M1 cluster are best fit by a model that assumes a large photoresponse, causing the cell to enter depolarization block. Our models also led us to hypothesize that the M2/M4 and M3/M5 clusters experience comparable photoexcitation but that the M3/M5 cascade decays significantly faster than the M2/M4 cascade, resulting in different response waveforms between these clusters. These mathematical models will help predict how each ipRGC cluster might respond to stimuli of any waveform and could inform the invention of lighting technologies that promote health through melanopsin stimulation.


Assuntos
Transdução de Sinal Luminoso , Modelos Neurológicos , Células Fotorreceptoras de Vertebrados/fisiologia , Células Ganglionares da Retina/fisiologia , Potenciais de Ação , Animais , Estimulação Luminosa , Ratos
6.
Exp Eye Res ; 130: 17-28, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25450063

RESUMO

Intrinsically photosensitive retinal ganglion cells (ipRGCs) are inner retinal photoreceptors that mediate non-image-forming visual functions, e.g. pupillary constriction, regulation of pineal melatonin release, and circadian photoentrainment. Five types of ipRGCs were recently discovered in mouse, but whether they exist in other mammals remained unknown. We report that the rat also has five types of ipRGCs, whose morphologies match those of mouse ipRGCs; this is the first demonstration of all five cell types in a non-mouse species. Through immunostaining and λmax measurements, we showed that melanopsin is likely the photopigment of all rat ipRGCs. The various cell types exhibited diverse spontaneous spike rates, with the M1 type spiking the least and M4 spiking the most, just like we had observed for their mouse counterparts. Also similar to mouse, all ipRGCs in rat generated not only sluggish intrinsic photoresponses but also fast, synaptically driven ones. However, we noticed two significant differences between these species. First, whereas we learned previously that all mouse ipRGCs had equally sustained synaptic light responses, rat M1 cells' synaptic photoresponses were far more transient than those of M2-M5. Since M1 cells provide all input to the circadian clock, this rat-versus-mouse discrepancy could explain the difference in photoentrainment threshold between mouse and other species. Second, rat ipRGCs' melanopsin-based spiking photoresponses could be classified into three varieties, but only two were discerned for mouse ipRGCs. This correlation of spiking photoresponses with cell types will help researchers classify ipRGCs in multielectrode-array (MEA) spike recordings.


Assuntos
Células Ganglionares da Retina/citologia , Animais , Animais Recém-Nascidos , Axônios/fisiologia , Ritmo Circadiano/fisiologia , Eletrofisiologia , Luz , Potenciais da Membrana/fisiologia , Camundongos , Estimulação Luminosa , Ratos , Ratos Sprague-Dawley , Reflexo Pupilar/fisiologia , Células Ganglionares da Retina/fisiologia , Células Ganglionares da Retina/efeitos da radiação , Opsinas de Bastonetes/metabolismo , Visão Ocular/fisiologia
7.
J Physiol ; 592(7): 1619-36, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24396062

RESUMO

Intrinsically photosensitive retinal ganglion cells (ipRGCs) mediate non-image-forming visual responses, including pupillary constriction, circadian photoentrainment and suppression of pineal melatonin secretion. Five morphological types of ipRGCs, M1-M5, have been identified in mice. In order to understand their functions better, we studied the photoresponses of all five cell types, by whole-cell recording from fluorescently labelled ipRGCs visualized using multiphoton microscopy. All ipRGC types generated melanopsin-based ('intrinsic') as well as synaptically driven ('extrinsic') light responses. The intrinsic photoresponses of M1 cells were lower threshold, higher amplitude and faster than those of M2-M5. The peak amplitudes of extrinsic light responses differed among the ipRGC types; however, the responses of all cell types had comparable thresholds, kinetics and waveforms, and all cells received rod input. While all five types exhibited inhibitory amacrine-cell and excitatory bipolar-cell inputs from the 'on' channel, M1 and M3 received additional 'off'-channel inhibition, possibly through their 'off'-sublamina dendrites. The M2-M5 ipRGCs had centre-surround-organized receptive fields, implicating a capacity to detect spatial contrast. In contrast, the receptive fields of M1 cells lacked surround antagonism, which might be caused by the surround of the inhibitory input nullifying the surround of the excitatory input. All ipRGCs responded robustly to a wide range of motion speeds, and M1-M4 cells appeared tuned to different speeds, suggesting that they might analyse the speed of motion. Retrograde labelling revealed that M1-M4 cells project to the superior colliculus, suggesting that the contrast and motion information signalled by these cells could be used by this sensorimotor area to detect novel objects and motion in the visual field.


Assuntos
Transdução de Sinal Luminoso/efeitos da radiação , Luz , Células Ganglionares da Retina/efeitos da radiação , Percepção Visual/efeitos da radiação , Animais , Sensibilidades de Contraste/efeitos da radiação , Potenciais Evocados , Feminino , Subunidades alfa de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Proteínas Heterotriméricas de Ligação ao GTP/deficiência , Proteínas Heterotriméricas de Ligação ao GTP/genética , Cinética , Masculino , Camundongos , Camundongos Knockout , Microscopia de Fluorescência por Excitação Multifotônica , Percepção de Movimento/efeitos da radiação , Reconhecimento Visual de Modelos/efeitos da radiação , Estimulação Luminosa , Células Ganglionares da Retina/classificação , Células Ganglionares da Retina/metabolismo , Percepção Espacial/efeitos da radiação , Colículos Superiores/metabolismo , Colículos Superiores/efeitos da radiação , Transducina/deficiência , Transducina/genética , Visão Ocular/efeitos da radiação , Campos Visuais/efeitos da radiação , Vias Visuais/metabolismo , Vias Visuais/efeitos da radiação
8.
J Neurosci ; 32(33): 11478-85, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22895730

RESUMO

A recently discovered type of mammalian retinal ganglion cell encodes environmental light intensity and mediates non-image-forming visual behaviors, such as the pupillary reflex and circadian photoentrainment. These intrinsically photosensitive retinal ganglion cells (ipRGCs) generate endogenous, melanopsin-based photoresponses as well as extrinsic, rod/cone-driven responses. Because the ipRGCs' light responses and the behaviors they control are both remarkably tonic, these cells have been hypothesized to be capable of irradiance detection lasting throughout the day. I tested this hypothesis by obtaining multielectrode-array recordings from ipRGCs in a novel rat eyecup preparation that enhances the regeneration of rod/cone photopigments. I found that 10 h constant light could continuously evoke action potentials in these ganglion cells under conditions that stimulated (1) only melanopsin, (2) mainly the rod input, and (3) both intrinsic and extrinsic responses. In response to a 10 h stimulus with gradual intensity changes to simulate sunrise and sunset, ipRGC firing rates slowly increased during the "sunrise" phase and slowly decreased during the "sunset" phase. Furthermore, I recorded from putative ipRGCs of melanopsin-knock-out mice and found that these cells retained the ability to respond in a sustained fashion to 20 min light steps, indicating that melanopsin is not required for such tonic responses. In conclusion, ipRGCs can signal light continuously for at least 10 h and can probably track gradual irradiance changes over the course of the day. These results further suggest that the photoreceptors and ON bipolar cells presynaptic to ipRGCs may be able to respond to light continuously for 10 h.


Assuntos
Transdução de Sinal Luminoso/fisiologia , Iluminação/métodos , Retina/citologia , Células Ganglionares da Retina/fisiologia , Células Ganglionares da Retina/efeitos da radiação , 2-Amino-5-fosfonovalerato/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Potenciais de Ação/fisiologia , Potenciais de Ação/efeitos da radiação , Aminobutiratos/farmacologia , Animais , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Técnicas In Vitro , Luz , Transdução de Sinal Luminoso/efeitos dos fármacos , Transdução de Sinal Luminoso/genética , Masculino , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Estimulação Luminosa , Quinoxalinas/farmacologia , Ratos , Ratos Long-Evans , Células Ganglionares da Retina/efeitos dos fármacos , Opsinas de Bastonetes/deficiência , Opsinas de Bastonetes/metabolismo , Fatores de Tempo , Vias Visuais/efeitos dos fármacos , Vias Visuais/efeitos da radiação
9.
J Neurophysiol ; 109(7): 1876-89, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23343892

RESUMO

In the mammalian retina, some ganglion cells express the photopigment melanopsin and function as photoreceptors. Five morphological types of these intrinsically photosensitive retinal ganglion cells (ipRGCs), M1-M5, have been identified in mice. Whereas M1 specializes in non-image-forming visual functions and drives such behaviors as the pupillary light reflex and circadian photoentrainment, the other types appear to contribute to image-forming as well as non-image-forming vision. Recent work has begun to reveal physiological diversity among some of the ipRGC types, including differences in photosensitivity, firing rate, and membrane resistance. To gain further insights into these neurons' functional differences, we conducted a comprehensive survey of the electrophysiological properties of all five morphological types. Compared with the other types, M1 had the highest membrane resistance, longest membrane time constant, lowest spike frequencies, widest action potentials, most positive spike thresholds, smallest hyperpolarization-activated inwardly-rectifying current-induced "sagging" responses to hyperpolarizing currents, and the largest effects of voltage-gated K(+) currents on membrane potentials. M4 and M5 were at the other end of the spectrum for most of these measures, while M2 and M3 tended to be in the middle of this spectrum. Additionally, M1 and M2 cells generated more diverse voltage-gated Ca(2+) currents than M3-M5. In conclusion, M1 cells are significantly different from all other ipRGCs in most respects, possibly reflecting the unique physiological requirements of non-image-forming vision. Furthermore, the non-M1 ipRGCs are electrophysiologically heterogeneous, implicating these cells' diverse functional roles in both non-image-forming vision and pattern vision.


Assuntos
Potenciais de Ação , Células Fotorreceptoras de Vertebrados/fisiologia , Células Ganglionares da Retina/fisiologia , Animais , Canais de Cálcio/metabolismo , Potenciais da Membrana , Camundongos , Células Fotorreceptoras de Vertebrados/classificação , Células Fotorreceptoras de Vertebrados/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Células Ganglionares da Retina/classificação , Células Ganglionares da Retina/metabolismo
10.
J Neurosci Methods ; 384: 109762, 2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-36470470

RESUMO

BACKGROUND: Constituting about 5 % of mouse retinal ganglion cells (RGCs), intrinsically photosensitive retinal ganglion cells (ipRGCs) express the photopigment melanopsin (gene symbol Opn4) and drive such photoresponses as pupil constriction, melatonin suppression, and circadian photoentrainment. Opn4Cre mice with Cre recombinase-expressing ipRGCs have enabled genetic manipulation of ipRGCs; unfortunately, while Cre expression within the inner retina is ipRGC-specific, leaky expression also occurs in some outer retinal photoreceptors, so Cre-induced alterations in the latter cells may confound certain studies of ipRGC function. Methods that express Cre in ipRGCs but not rods or cones are needed. NEW METHOD: We have constructed a recombinant serotype-2 adeno-associated virus, rAAV2-Opn4-Cre, with the improved Cre recombinase (iCre) gene under the control of a ∼3kbp Opn4 promoter sequence, and injected it intravitreally into mice to transduce inner retinal neurons while sparing the outer retina. RESULTS: We introduced rAAV2-Opn4-Cre into Cre reporter mice in which enhanced green fluorescent protein (EGFP) expression indicates Cre expression. Single-cell electrophysiological recordings and intracellular dye fills showed that 84 % of the EGFP+ cells were ipRGCs including M1-M6 types, while 16 % were conventional RGCs. COMPARISON WITH EXISTING METHODS: Whereas Opn4Cre mice express Cre in some rod/cone photoreceptors, intravitreally applied rAAV2-Opn4-Cre induces Cre only in the inner retina, albeit with leaky expression in some conventional RGCs. CONCLUSIONS: rAAV2-Opn4-Cre has overcome a significant limitation of Opn4Cre mice. We recommend usage scenarios where the Cre-expressing conventional RGCs should not pose a problem.


Assuntos
Dependovirus , Células Fotorreceptoras Retinianas Cones , Camundongos , Animais , Dependovirus/genética , Células Fotorreceptoras de Vertebrados , Luz
11.
Biomed Opt Express ; 14(12): 6283-6290, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-38420324

RESUMO

This study investigates the feasibility of capturing visually evoked hemodynamic responses in the mouse brain using photoacoustic tomography (PAT) and ultrasound (US) dual-modality imaging. A commercial piezoelectric transducer array and a capacitive micromachined ultrasonic transducer (CMUT) array were compared using a programmable PAT-US imaging system. The system resolution was measured by imaging phantoms. We also tested the ability of the system to capture visually evoked hemodynamic responses in the superior colliculus as well as the primary visual cortex in wild-type mice. Results show that the piezoelectric transducer array and the CMUT array exhibit comparable imaging performance, and both arrays can capture visually evoked hemodynamic responses in subcortical as well as cortical regions of the mouse brain.

12.
J Biol Rhythms ; 38(4): 366-378, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37222434

RESUMO

Beyond visual perception, light has non-image-forming effects mediated by melanopsin-expressing, intrinsically photosensitive retinal ganglion cells (ipRGCs). The present study first used multielectrode array recordings to show that in a diurnal rodent, Nile grass rats (Arvicanthis niloticus), ipRGCs generate rod/cone-driven and melanopsin-based photoresponses that stably encode irradiance. Subsequently, two ipRGC-mediated non-image-forming effects, namely entrainment of daily rhythms and light-induced arousal, were examined. Animals were first housed under a 12:12 h light/dark cycle (lights-on at 0600 h) with the light phase generated by a low-irradiance fluorescent light (F12), a daylight spectrum (D65) stimulating all photoreceptors, or a narrowband 480 nm spectrum (480) that maximized melanopsin stimulation and minimized S-cone stimulation (λmax 360 nm) compared to D65. Daily rhythms of locomotor activities showed onset and offset closer to lights-on and lights-off, respectively, in D65 and 480 than in F12, and higher day/night activity ratio under D65 versus 480 and F12, suggesting the importance of S-cone stimulation. To assess light-induced arousal, 3-h light exposures using 4 spectra that stimulated melanopsin equally but S-cones differentially were superimposed on F12 background lighting: D65, 480, 480 + 365 (narrowband 365 nm), and D65 - 365. Compared to the F12-only condition, all four pulses increased in-cage activity and promoted wakefulness, with 480 + 365 having the greatest and longest-lasting wakefulness-promoting effects, again indicating the importance of stimulating S-cones as well as melanopsin. These findings provide insights into the temporal dynamics of photoreceptor contributions to non-image-forming photoresponses in a diurnal rodent that may help guide future studies of lighting environments and phototherapy protocols that promote human health and productivity.


Assuntos
Murinae , Células Fotorreceptoras Retinianas Cones , Humanos , Animais , Células Fotorreceptoras Retinianas Cones/fisiologia , Vigília , Ritmo Circadiano/fisiologia , Células Ganglionares da Retina , Opsinas de Bastonetes , Luz , Estimulação Luminosa
13.
Eur J Neurosci ; 35(4): 507-18, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22304466

RESUMO

A novel class of photoreceptors, the intrinsically photosensitive retinal ganglion cells (ipRGCs), express the photopigment melanopsin and drive non-image-forming responses to light such as circadian photoentrainment, the pupillary light reflex and suppression of nocturnal melatonin production in the pineal. Because dendrites from one subclass of these cells - the M1-type ipRGCs - make presumptive synaptic contacts at sites of dopamine release from dopaminergic amacrine cells, they are prime targets for modulation by dopamine, a neuromodulator implicated in retinal circadian rhythms and light adaptation. In patch-clamp recordings from ipRGCs in intact rat retinas, dopamine attenuated the melanopsin-based photocurrent. We confirmed that this was the result of direct action on ipRGCs by replicating the effect in dissociated ipRGCs that were isolated from influences of other retinal neurons. In these recordings, the D1-family dopamine receptor agonist SKF38393 attenuated the photocurrent, caused a modest depolarization, and reduced the input resistance of ipRGCs. The D2-family agonist quinpirole had no effect on the photocurrent. Single-cell reverse-transcriptase polymerase chain reaction revealed that the majority of ipRGCs tested expressed drd1a, the gene coding for the D1a dopamine receptor. This finding was supported by immunohistochemical localization of D1a receptor protein in melanopsin-expressing ganglion cells. Finally, the adenylate cyclase activator forskolin, applied in combination with the phosphodiesterase inhibitor IBMX (isobutylmethylxanthine), mimicked the effects of SKF38393 on the ipRGC photocurrent, membrane potential and input resistance, consistent with a D1-receptor signaling pathway. These data suggest that dopamine, acting via D1-family receptors, alters the responses of ipRGCs and thus of non-image-forming vision.


Assuntos
Dopamina/metabolismo , Transdução de Sinal Luminoso/fisiologia , Células Fotorreceptoras/fisiologia , Células Ganglionares da Retina/fisiologia , 1-Metil-3-Isobutilxantina/farmacologia , 2,3,4,5-Tetra-Hidro-7,8-Di-Hidroxi-1-Fenil-1H-3-Benzazepina/farmacologia , Actinas/genética , Actinas/metabolismo , Animais , Carbazóis/farmacologia , Células Cultivadas , Colforsina/farmacologia , AMP Cíclico/metabolismo , Dopamina/farmacologia , Agonistas de Dopamina/farmacologia , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Técnicas In Vitro , Luz , Transdução de Sinal Luminoso/efeitos dos fármacos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Estimulação Luminosa , Pirróis/farmacologia , Quimpirol/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Dopaminérgicos/genética , Receptores Dopaminérgicos/metabolismo , Retina/citologia , Opsinas de Bastonetes/genética , Opsinas de Bastonetes/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia , Vias Visuais/efeitos dos fármacos , Vias Visuais/fisiologia
14.
Biomed Opt Express ; 13(2): 1017-1025, 2022 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-35284169

RESUMO

The lack of a non-invasive or minimally invasive imaging technique has long been a challenge to investigating brain activities in mice. Functional magnetic resonance imaging and the more recently developed diffuse optical imaging both suffer from limited spatial resolution. Photoacoustic (PA) imaging combines the sensitivity of optical excitation to hemodynamic changes and ultrasound detection's relatively high spatial resolution. In this study, we evaluated the feasibility of using a label-free, real-time PA computed tomography (PACT) system to measure visually evoked hemodynamic responses within the primary visual cortex (V1) in mice. Photostimulation of the retinas evoked significantly faster and stronger V1 responses in wild-type mice than in age-matched rod/cone-degenerate mice, consistent with known differences between rod/cone- vs. melanopsin-mediated photoreception. In conclusion, the PACT system in this study has sufficient sensitivity and spatial resolution to resolve visual cortical hemodynamics during retinal photostimulation, and PACT is a potential tool for investigating visually evoked brain activities in mouse models of retinal diseases.

15.
Nature ; 433(7027): 745-9, 2005 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-15674243

RESUMO

Melanopsin has been proposed to be the photopigment of the intrinsically photosensitive retinal ganglion cells (ipRGCs); these photoreceptors of the mammalian eye drive circadian and pupillary adjustments through direct projections to the brain. Their action spectrum (lambda(max) approximately 480 nm) implicates an opsin and melanopsin is the only opsin known to exist in these cells. Melanopsin is required for ipRGC photosensitivity and for behavioural photoresponses that survive disrupted rod and cone function. Heterologously expressed melanopsin apparently binds retinaldehyde and mediates photic activation of G proteins. However, its amino-acid sequence differs from vertebrate photosensory opsins and some have suggested that melanopsin may be a photoisomerase, providing retinoid chromophore to an unidentified opsin. To determine whether melanopsin is a functional sensory photopigment, here we transiently expressed it in HEK293 cells that stably expressed TRPC3 channels. Light triggered a membrane depolarization in these cells and increased intracellular calcium. The light response resembled that of ipRGCs, with almost identical spectral sensitivity (lambda(max) approximately 479 nm). The phototransduction pathway included Gq or a related G protein, phospholipase C and TRPC3 channels. We conclude that mammalian melanopsin is a functional sensory photopigment, that it is the photopigment of ganglion-cell photoreceptors, and that these photoreceptors may use an invertebrate-like phototransduction cascade.


Assuntos
Transdução de Sinal Luminoso/efeitos da radiação , Luz , Opsinas de Bastonetes/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio/efeitos da radiação , Linhagem Celular , Eletrofisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Expressão Gênica , Humanos , Canais Iônicos/metabolismo , Camundongos , Opsinas de Bastonetes/genética , Canais de Cátion TRPC , Fosfolipases Tipo C/metabolismo
16.
Proc Natl Acad Sci U S A ; 105(37): 14181-6, 2008 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-18779590

RESUMO

Retinal dopaminergic amacrine neurons (DA neurons) play a central role in reconfiguring retinal function according to prevailing illumination conditions, yet the mechanisms by which light regulates their activity are poorly understood. We investigated the means by which sustained light responses are evoked in DA neurons. Sustained light responses were driven by cationic currents and persisted in vitro and in vivo in the presence of L-AP4, a blocker of retinal ON-bipolar cells. Several characteristics of these L-AP4-resistant light responses suggested that they were driven by melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs), including long latencies, marked poststimulus persistence, and a peak spectral sensitivity of 478 nm. Furthermore, sustained DA neuron light responses, but not transient DA neuron responses, persisted in rod/cone degenerate retinas, in which ipRGCs account for virtually all remaining retinal phototransduction. Thus, ganglion-cell photoreceptors provide excitatory drive to DA neurons, most likely by way of the coramification of their dendrites and the processes of DA neurons in the inner plexiform layer. This unprecedented centrifugal outflow of ganglion-cell signals within the retina provides a novel basis for the restructuring of retinal circuits by light.


Assuntos
Células Amácrinas/metabolismo , Dopamina/metabolismo , Células Ganglionares da Retina/metabolismo , Transdução de Sinais , Animais , Eletrofisiologia , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Degeneração Neural/metabolismo , Técnicas de Patch-Clamp , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Opsinas de Bastonetes/metabolismo
17.
Front Neurol ; 12: 627550, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33643205

RESUMO

A growing number of studies document circadian phase-shifting after exposure to millisecond light flashes. When strung together by intervening periods of darkness, these stimuli evoke pacemaker responses rivaling or outmatching those created by steady luminance, suggesting that the circadian system's relationship to light can be contextualized outside the principle of simple dose-dependence. In the current review, we present a brief chronology of this work. We then develop a conceptual model around it that attempts to relate the circadian effects of flashes to a natural integrative process the pacemaker uses to intermittently sample the photic information available at dawn and dusk. Presumably, these snapshots are employed as building blocks in the construction of a coherent representation of twilight the pacemaker consults to orient the next day's physiology (in that way, flash-resetting of pacemaker rhythms might be less an example of a circadian visual illusion and more an example of the kinds of gestalt inferences that the image-forming system routinely makes when identifying objects within the visual field; i.e., closure). We conclude our review with a discussion on the role of cones in the pacemaker's twilight predictions, providing new electrophysiological data suggesting that classical photoreceptors-but not melanopsin-are necessary for millisecond, intermediate-intensity flash responses in ipRGCs (intrinsically photosensitive retinal ganglion cells). Future investigations are necessary to confirm this "Cone Sentinel Model" of circadian flash-integration and twilight-prediction, and to further define the contribution of cones vs. rods in transducing pacemaker flash signals.

18.
Invest Ophthalmol Vis Sci ; 62(1): 10, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33410914

RESUMO

Purpose: Intrinsically photosensitive retinal ganglion cells (ipRGCs) signal not only centrally to non-image-forming visual centers of the brain but also intraretinally to amacrine interneurons through gap junction electrical coupling, potentially modulating image-forming retinal processing. We aimed to determine (1) which ipRGC types couple with amacrine cells, (2) the neuromodulator contents of ipRGC-coupled amacrine cells, and (3) whether connexin36 (Cx36) contributes to ipRGC-amacrine coupling. Methods: Gap junction-permeable Neurobiotin tracer was injected into green fluorescent protein (GFP)-labeled ipRGCs in Opn4Cre/+; Z/EG mice to stain coupled amacrine cells, and immunohistochemistry was performed to reveal the neuromodulator contents of the Neurobiotin-stained amacrine cells. We also created Opn4Cre/+; Cx36flox/flox; Z/EG mice to knock out Cx36 in GFP-labeled ipRGCs and looked for changes in the number of ipRGC-coupled amacrine cells. Results: Seventy-three percent of ipRGCs, including all six types (M1-M6), were tracer-coupled with amacrine somas 5.7 to 16.5 µm in diameter but not with ganglion cells. Ninety-two percent of the ipRGC-coupled somas were in the ganglion cell layer and the rest in the inner nuclear layer. Some ipRGC-coupled amacrine cells were found to accumulate serotonin or to contain nitric oxide synthase or neuropeptide Y. Knocking out Cx36 in M2 and M4 dramatically reduced the number of coupled somas. Conclusions: Heterologous gap junction coupling with amacrine cells is widespread across mouse ipRGC types. ipRGC-coupled amacrine cells probably comprise multiple morphologic types and use multiple neuromodulators, suggesting that gap junctional ipRGC-to-amacrine signaling likely exerts diverse modulatory effects on retinal physiology. ipRGC-amacrine coupling is mediated partly, but not solely, by Cx36.


Assuntos
Células Amácrinas/citologia , Conexinas/metabolismo , Junções Comunicantes/fisiologia , Neuropeptídeo Y/metabolismo , Óxido Nítrico Sintase/metabolismo , Células Ganglionares da Retina/citologia , Serotonina/metabolismo , Células Amácrinas/metabolismo , Animais , Biotina/administração & dosagem , Biotina/análogos & derivados , Comunicação Celular/fisiologia , Feminino , Proteínas de Fluorescência Verde/administração & dosagem , Substâncias Luminescentes/administração & dosagem , Masculino , Camundongos , Camundongos Knockout , Isoformas de Proteínas , Células Ganglionares da Retina/metabolismo , Opsinas de Bastonetes , Proteína delta-2 de Junções Comunicantes
19.
Int IEEE EMBS Conf Neural Eng ; 2021: 263-266, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34646429

RESUMO

For epiretinal prostheses, disc electrodes stimulate retinal ganglion cells (RGCs) with electric current to create visual percepts. Prior studies have determined that the sodium channel band (SOCB), located on the RGC axon (30-50 µm from the soma) is the most sensitive site to extracellular stimulation because of its high sodium channel density. Biophysical cable models used to study RGC activation in silico often rely on simplified axon trajectories, disregarding the non-uniform paths that axons follow to the optic disc. However, since axonal activation is a critical mechanism in epiretinal stimulation, it is important to investigate variable RGC axon trajectories. In this study, we use a computational model to perform a sensitivity analysis examining how the morphology of an RGC axon affects predictions of retinal activation. We determine that RGC cable models are sensitive to changes in the ascending axon trajectory between the soma and nerve fiber layer. On the other hand, RGC cable models are relatively robust to trajectory deviations in the plane parallel to the disc electrode's surface. Overall, our results suggest that incorporating natural variations of soma depth and nerve fiber layer entry angle could result in a more realistic model of the retina's response to epiretinal stimulation and a better understanding of elicited visual percepts.

20.
Curr Eye Res ; 46(4): 515-523, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32841098

RESUMO

PURPOSE: Intrinsically photosensitive retinal ganglion cells (ipRGCs) contain the photopigment melanopsin and can signal light continuously for many hours. Melanopsin is excited when its chromophore 11-cis-retinal absorbs a photon and becomes all-trans-retinal, which must be reisomerized to 11-cis-retinal to regenerate photoexcitable melanopsin. Due to the great distance separating ipRGCs from the retinal pigment epithelium (RPE) whose retinoid cycle produces 11-cis-retinal, ipRGCs had been assumed to regenerate all melanopsin molecules autonomously. Surprisingly, we previously found that pharmacologically inhibiting the retinoid cycle rendered melanopsin-based responses to prolonged illumination less sustained, suggesting that the RPE may supply retinoids to help ipRGCs regenerate melanopsin during extended photostimulation. However, the specificity of those drugs is unclear. Here, we reexamined the role of the retinoid cycle, and tested whether the RPE-to-ipRGC transport of retinoids utilizes cellular retinaldehyde-binding protein (CRALBP), present throughout the RPE and Müller glia. METHODS: To measure melanopsin-mediated photoresponses in isolation, all animals were 8- to 12-month-old rod/cone-degenerate mice. We genetically knocked out RPE-specific 65 kDa protein (RPE65), a critical enzyme in the retinoid cycle. We also knocked out the CRALBP gene rlbp1 mainly in Foxg1-expressing Müller cells. We obtained multielectrode-array recordings from ipRGCs in a novel RPE-attached mouse retina preparation, and imaged pupillary light reflexes in vivo. RESULTS: Melanopsin-based ipRGC responses to prolonged light became less tonic in both knockout lines, and pupillary light reflexes were also less sustained in RPE65-knockout than control mice. CONCLUSIONS: These results confirm that ipRGCs rely partly on the retinoid cycle to continuously regenerate melanopsin during prolonged photostimulation, and suggest that CRALBP in Müller glia likely transports 11-cis-retinal from the RPE to ipRGCs - this is the first proposed functional role for CRALBP in the inner retina.


Assuntos
Proteínas de Transporte/metabolismo , Reflexo Pupilar/fisiologia , Células Ganglionares da Retina/metabolismo , Opsinas de Bastonetes/fisiologia , cis-trans-Isomerases/metabolismo , Animais , Inativação Gênica , Imuno-Histoquímica , Luz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Estimulação Luminosa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA