Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Hum Mol Genet ; 31(7): 1035-1050, 2022 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-34652420

RESUMO

Heteromeric Kv2.1/Kv8.2 channels are voltage-gated potassium channels localized to the photoreceptor inner segment. They carry IKx, which is largely responsible for setting the photoreceptor resting membrane potential. Mutations in Kv8.2 result in childhood-onset cone dystrophy with supernormal rod response (CDSRR). We generated a Kv8.2 knockout (KO) mouse and examined retinal signaling and photoreceptor degeneration to gain deeper insight into the complex phenotypes of this disease. Using electroretinograms, we show that there were delayed or reduced signaling from rods depending on the intensity of the light stimulus, consistent with reduced capacity for light-evoked changes in membrane potential. The delayed response was not seen ex vivo where extracellular potassium levels were controlled by the perfusion buffer, so we propose the in vivo alteration is influenced by genotype-associated ionic imbalance. We observed mild retinal degeneration. Signaling from cones was reduced but there was no loss of cone density. Loss of Kv8.2 altered responses to flickering light with responses attenuated at high frequencies and altered in shape at low frequencies. The Kv8.2 KO line on an all-cone retina background had reduced cone-driven ERG b wave amplitudes and underwent degeneration. Altogether, we provide insight into how a deficit in the dark current affects the health and function of photoreceptors.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana , Degeneração Retiniana , Doenças Retinianas , Animais , Eletrorretinografia , Camundongos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Retina/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Degeneração Retiniana/genética
2.
J Neurosci ; 42(21): 4231-4249, 2022 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-35437278

RESUMO

Signal integration of converging neural circuits is poorly understood. One example is in the retina where the integration of rod and cone signaling is responsible for the large dynamic range of vision. The relative contribution of rods versus cones is dictated by a complex function involving background light intensity and stimulus temporal frequency. One understudied mechanism involved in coordinating rod and cone signaling onto the shared retinal circuit is the hyperpolarization activated current (Ih) mediated by hyperpolarization-activated cyclic nucleotide-gated 1 (HCN1) channels expressed in rods and cones. Ih opposes membrane hyperpolarization driven by activation of the phototransduction cascade and modulates the strength and kinetics of the photoreceptor voltage response. We examined conditional knock-out (KO) of HCN1 from mouse rods using electroretinography (ERG). In the absence of HCN1, rod responses are prolonged in dim light which altered the response to slow modulation of light intensity both at the level of retinal signaling and behavior. Under brighter intensities, cone-driven signaling was suppressed. To our surprise, conditional KO of HCN1 from mouse cones had no effect on cone-mediated signaling. We propose that Ih is dispensable in cones because of the high level of temporal control of cone phototransduction. Thus, HCN1 is required for cone-driven retinal signaling only indirectly by modulating the voltage response of rods to limit their output.SIGNIFICANCE STATEMENT Hyperpolarization gated hyperpolarization-activated cyclic nucleotide-gated 1 (HCN1) channels carry a feedback current that helps to reset light-activated photoreceptors. Using conditional HCN1 knock-out (KO) mice we show that ablating HCN1 from rods allows rods to signal in bright light when they are normally shut down. Instead of enhancing vision this results in suppressing cone signaling. Conversely, ablating HCN1 from cones was of no consequence. This work provides novel insights into the integration of rod and cone signaling in the retina and challenges our assumptions about the role of HCN1 in cones.


Assuntos
Nucleotídeos Cíclicos , Células Fotorreceptoras Retinianas Bastonetes , Animais , Eletrorretinografia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Camundongos , Camundongos Knockout , Canais de Potássio/genética , Retina/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia
3.
Adv Exp Med Biol ; 1415: 269-276, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37440044

RESUMO

Photoreceptors (PRs) in the neural retina convert photon capture into an electrical signal that is communicated across a chemical synapse to second-order neurons in the retina and on through the rest of the visual pathway. This information is decoded in the visual cortex to create images. The activity of PRs depends on the concerted action of several voltage-gated ion channels that will be discussed in this chapter.


Assuntos
Células Fotorreceptoras , Retina , Células Fotorreceptoras/metabolismo , Retina/metabolismo , Transdução de Sinais , Sinapses/metabolismo , Canais Iônicos/metabolismo , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Células Fotorreceptoras Retinianas Cones/fisiologia
4.
J Neurosci ; 38(27): 6145-6160, 2018 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-29875267

RESUMO

α2δ-4 is an auxiliary subunit of voltage-gated Cav1.4 L-type channels that regulate the development and mature exocytotic function of the photoreceptor ribbon synapse. In humans, mutations in the CACNA2D4 gene encoding α2δ-4 cause heterogeneous forms of vision impairment in humans, the underlying pathogenic mechanisms of which remain unclear. To investigate the retinal function of α2δ-4, we used genome editing to generate an α2δ-4 knock-out (α2δ-4 KO) mouse. In male and female α2δ-4 KO mice, rod spherules lack ribbons and other synaptic hallmarks early in development. Although the molecular organization of cone synapses is less affected than rod synapses, horizontal and cone bipolar processes extend abnormally in the outer nuclear layer in α2δ-4 KO retina. In reconstructions of α2δ-4 KO cone pedicles by serial block face scanning electron microscopy, ribbons appear normal, except that less than one-third show the expected triadic organization of processes at ribbon sites. The severity of the synaptic defects in α2δ-4 KO mice correlates with a progressive loss of Cav1.4 channels, first in terminals of rods and later cones. Despite the absence of b-waves in electroretinograms, visually guided behavior is evident in α2δ-4 KO mice and better under photopic than scotopic conditions. We conclude that α2δ-4 plays an essential role in maintaining the structural and functional integrity of rod and cone synapses, the disruption of which may contribute to visual impairment in humans with CACNA2D4 mutations.SIGNIFICANCE STATEMENT In the retina, visual information is first communicated by the synapse formed between photoreceptors and second-order neurons. The mechanisms that regulate the structural integrity of this synapse are poorly understood. Here we demonstrate a role for α2δ-4, a subunit of voltage-gated Ca2+ channels, in organizing the structure and function of photoreceptor synapses. We find that presynaptic Ca2+ channels are progressively lost and that rod and cone synapses are disrupted in mice that lack α2δ-4. Our results suggest that alterations in presynaptic Ca2+ signaling and photoreceptor synapse structure may contribute to vision impairment in humans with mutations in the CACNA2D4 gene encoding α2δ-4.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/ultraestrutura , Sinapses/metabolismo , Sinapses/ultraestrutura , Animais , Feminino , Humanos , Macaca fascicularis , Masculino , Camundongos , Camundongos Knockout
5.
Exp Eye Res ; 170: 108-116, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29486162

RESUMO

The 14-3-3 family of proteins has undergone considerable expansion in higher eukaryotes with humans and mice expressing seven isoforms (ß, ε, η, γ, θ, ζ, and σ) from seven distinct genes (YWHAB, YWAHE, YWHAH, YWHAG, YWHAQ, YWHAZ, and SFN). Growing evidence indicates that while highly conserved, these isoforms are not entirely functionally redundant as they exhibit unique tissue expression profiles, subcellular localization, and biochemical functions. A key limitation in our understanding of 14-3-3 biology lies in our limited knowledge of cell-type specific 14-3-3 expression. Here we provide a characterization of 14-3-3 expression in whole retina and isolated rod photoreceptors using reverse-transcriptase digital droplet PCR. We find that all 14-3-3 genes with the exception of SFN are expressed in mouse retina with YWHAQ and YWHAE being the most highly expressed. Rod photoreceptors are enriched in YWHAE (14-3-3 ε). Immunohistochemistry revealed that 14-3-3 ε and 14-3-3 ζ exhibit unique distributions in photoreceptors with 14-3-3 ε restricted to the inner segment and 14-3-3 ζ localized to the outer segment. Our data demonstrates that, in the retina, 14-3-3 isoforms likely serve specific functions as they exhibit unique expression levels and cell-type specificity. As such, future investigations into 14-3-3 function in rod photoreceptors should be centered on 14-3-3 ε and 14-3-3 ζ, depending on the subcellular region of question.


Assuntos
Proteínas 14-3-3/genética , Regulação da Expressão Gênica/fisiologia , Retina/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Animais , Western Blotting , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Plasmídeos , Isoformas de Proteínas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Traffic ; 16(12): 1239-53, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26373354

RESUMO

Na(+) /K(+) -ATPase (NKA) participates in setting electrochemical gradients, cardiotonic steroid signaling and cellular adhesion. Distinct isoforms of NKA are found in different tissues and subcellular localization patterns. For example, NKA α1 is widely expressed, NKA α3 is enriched in neurons and NKA α4 is a testes-specific isoform found in sperm flagella. In some tissues, ankyrin, a key component of the membrane cytoskeleton, can regulate the trafficking of NKA. In the retina, NKA and ankyrin-B are expressed in multiple cell types and immunostaining for each is striking in the synaptic layers. Labeling for NKA is also prominent along the inner segment plasma membrane (ISPM) of photoreceptors. NKA co-immunoprecipitates with ankyrin-B, but on a subcellular level colocalization of these two proteins varies dependent on the cell type. We used transgenic Xenopus laevis tadpoles to evaluate the subcellular trafficking of NKA in photoreceptors. GFP-NKA α3 and α1 are localized to the ISPM, but α4 is localized to outer segments (OSs). We identified a VxP motif responsible for the OS targeting by using a series of chimeric and mutant NKA constructs. This motif is similar to previously identified ciliary targeting motifs. Given the structural similarities between OSs and flagella, our findings shed light on the subcellular targeting of this testes-specific NKA isoform.


Assuntos
Anquirinas/metabolismo , Flagelos/enzimologia , Retina/enzimologia , Segmento Interno das Células Fotorreceptoras da Retina/enzimologia , Segmento Externo das Células Fotorreceptoras da Retina/enzimologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Motivos de Aminoácidos , Animais , Anquirinas/genética , Bovinos , Membrana Celular/enzimologia , Proteínas de Fluorescência Verde/genética , Humanos , Imunoprecipitação , Técnicas In Vitro , Larva/enzimologia , Camundongos Endogâmicos C57BL , Organismos Geneticamente Modificados , Subunidades Proteicas , Transporte Proteico , Transdução de Sinais , ATPase Trocadora de Sódio-Potássio/genética , Especificidade da Espécie , Xenopus laevis/genética
7.
Cell Mol Life Sci ; 72(4): 833-43, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25142030

RESUMO

Hyperpolarization-activated cyclic nucleotide-gated 1 (HCN1) channels carry Ih, which contributes to neuronal excitability and signal transmission in the nervous system. Controlling the trafficking of HCN1 is an important aspect of its regulation, yet the details of this process are poorly understood. Here, we investigated how the C-terminus of HCN1 regulates trafficking by testing for its ability to redirect the localization of a non-targeted reporter in transgenic Xenopus laevis photoreceptors. We found that HCN1 contains an ER localization signal and through a series of deletion constructs, identified the responsible di-arginine ER retention signal. This signal is located in the intrinsically disordered region of the C-terminus of HCN1. To test the function of the ER retention signal in intact channels, we expressed wild type and mutant HCN1 in HEK293 cells and found this signal negatively regulates surface expression of HCN1. In summary, we report a new mode of regulating HCN1 trafficking: through the use of a di-arginine ER retention signal that monitors processing of the channel in the early secretory pathway.


Assuntos
Arginina/metabolismo , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Animais Geneticamente Modificados/metabolismo , Arginina/química , Células HEK293 , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Dados de Sequência Molecular , Células Fotorreceptoras/metabolismo , Via Secretória , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Xenopus laevis/metabolismo
8.
Curr Top Membr ; 72: 231-65, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24210432

RESUMO

Photoreceptors are exquisitely adapted to transform light stimuli into electrical signals that modulate neurotransmitter release. These cells are organized into several compartments including the unique outer segment (OS). Its whole function is to absorb light and transduce this signal into a change of membrane potential. Another compartment is the inner segment where much of metabolism and regulation of membrane potential takes place and that connects the OS and synapse. The synapse is the compartment where changes in membrane potentials are relayed to other neurons in the retina via release of neurotransmitter. The composition of the plasma membrane surrounding these compartments varies to accommodate their specific functions. In this chapter, we discuss the organization of the plasma membrane emphasizing the protein composition of each region as it relates to visual signaling. We also point out examples where mutations in these proteins cause visual impairment.


Assuntos
Membrana Celular/metabolismo , Células Fotorreceptoras/metabolismo , Animais , Canais de Cálcio/deficiência , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Células Fotorreceptoras/química , Retina/anatomia & histologia , Rodopsina/metabolismo , Proteínas SNARE/metabolismo , Transdução de Sinais , Trocador de Sódio e Cálcio/metabolismo , Vertebrados/metabolismo
9.
Front Mol Neurosci ; 16: 1155955, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37181655

RESUMO

The voltage-gated calcium channel, Cav1.4 is localized to photoreceptor ribbon synapses and functions both in molecular organization of the synapse and in regulating release of synaptic vesicles. Mutations in Cav1.4 subunits typically present as either incomplete congenital stationary night blindness or a progressive cone-rod dystrophy in humans. We developed a cone-rich mammalian model system to further study how different Cav1.4 mutations affect cones. RPE65 R91W KI; Nrl KO "Conefull" mice were crossed to Cav1.4 α1F or α2δ4 KO mice to generate the "Conefull:α1F KO" and "Conefull:α2δ4 KO" lines. Animals were assessed using a visually guided water maze, electroretinogram (ERG), optical coherence tomography (OCT), and histology. Mice of both sexes and up to six-months of age were used. Conefull: α1F KO mice could not navigate the visually guided water maze, had no b-wave in the ERG, and the developing all-cone outer nuclear layer reorganized into rosettes at the time of eye opening with degeneration progressing to 30% loss by 2-months of age. In comparison, the Conefull: α2δ4 KO mice successfully navigated the visually guided water maze, had a reduced amplitude b-wave ERG, and the development of the all-cone outer nuclear layer appeared normal although progressive degeneration with 10% loss by 2-months of age was observed. In summary, new disease models for studying congenital synaptic diseases due to loss of Cav1.4 function have been created.

10.
Saudi J Ophthalmol ; 37(4): 313-320, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38155679

RESUMO

PURPOSE: The purpose of this study was to develop a visually guided swim assay (VGSA) for measuring vision in mouse retinal disease models comparable to the multi-luminance mobility test (MLMT) utilized in human clinical trials. METHODS: Three mouse retinal disease models were studied: Bardet-Biedl syndrome type 1 (Bbs1M390R/M390R), n = 5; Bardet-Biedl syndrome type 10 (Bbs10-/-), n = 11; and X linked retinoschisis (retinoschisin knockout; Rs1-KO), n = 5. Controls were normally-sighted mice, n = 10. Eyeless Pax6Sey-Dey mice, n = 4, were used to determine the performance of animals without vision in VGSA. RESULTS: Eyeless Pax6Sey-Dey mice had a VGSA time-to-platform (TTP) 7X longer than normally-sighted controls (P < 0.0001). Controls demonstrated no difference in their TTP in both lighting conditions; the same was true for Pax6Sey-Dey. At 4-6 M, Rs1-KO and Bbs10-/- had longer TTP in the dark than controls (P = 0.0156 and P = 1.23 × 10-8, respectively). At 9-11 M, both BBS models had longer TTP than controls in light and dark with times similar to Pax6Sey-Dey (P < 0.0001), demonstrating progressive vision loss in BBS models, but not in controls nor in Rs1-KO. At 1 M, Bbs10-/- ERG light-adapted (cone) amplitudes were nonrecordable, resulting in a floor effect. VGSA did not reach a floor until 9-11 M. ERG combined rod/cone b-wave amplitudes were nonrecordable in all three mutant groups at 9-11 M, but VGSA still showed differences in visual function. ERG values correlate non-linearly with VGSA, and VGSA measured the continual decline of vision. CONCLUSION: ERG is no longer a useful endpoint once the nonrecordable level is reached. VGSA differentiates between different levels of vision, different ages, and different disease models even after ERG is nonrecordable, similar to the MLMT in humans.

11.
J Neurosci ; 31(41): 14660-8, 2011 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-21994382

RESUMO

The members of the R7 regulator of G-protein signaling (RGS) protein subfamily are versatile regulators of G-protein signaling throughout the nervous system. Recent studies indicate that they are often found in complexes with membrane anchor proteins that serve as versatile modulators of their activity, intracellular targeting, and stability. One striking example is the interplay between the membrane anchor R9AP and the RGS9-1 · Gß5 GTPase-activating complex responsible for the rapid inactivation of the G-protein transducin in vertebrate photoreceptor cells during their recovery from light excitation. The amount of this complex in photoreceptors sets their temporal resolution and is precisely regulated by the expression level of R9AP, which serves to protect the RGS9-1 and Gß5 subunits from intracellular proteolysis. In this study, we investigated the mechanism by which R9AP performs its protective function in mouse rods and found that it is entirely confined to recruiting RGS9-1 · Gß5 to cellular membranes. Furthermore, membrane attachment of RGS9-1 · Gß5 is sufficient for its stable expression in rods even in the absence of R9AP. Our second finding is that RGS9-1 · Gß5 possesses targeting information that specifies its exclusion from the outer segment and that this information is neutralized by association with R9AP to allow outer segment targeting. Finally, we demonstrate that the ability of R9AP · RGS9-1 · Gß5 to accelerate GTP hydrolysis on transducin is independent of its means of membrane attachment, since replacing the transmembrane domain of R9AP with a site for lipid modification did not impair the catalytic activity of this complex.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Líquido Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Células Fotorreceptoras de Vertebrados/citologia , Células Fotorreceptoras de Vertebrados/metabolismo , Proteólise , Proteínas RGS/metabolismo , Retina/citologia , Transdução de Sinais/fisiologia , Animais , Membrana Celular/metabolismo , Adaptação à Escuridão/genética , Relação Dose-Resposta à Radiação , Eletroporação/métodos , Subunidades beta da Proteína de Ligação ao GTP/genética , Regulação da Expressão Gênica/genética , Luz , Proteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Proteínas RGS/genética , Proteínas SNARE/genética , Proteínas SNARE/metabolismo
12.
J Cell Sci ; 123(Pt 21): 3639-44, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20923839

RESUMO

Photoreceptors are among the most metabolically active cells in the body, relying on both oxidative phosphorylation and glycolysis to satisfy their high energy needs. Local glycolysis is thought to be particularly crucial in supporting the function of the photoreceptor's light-sensitive outer segment compartment, which is devoid of mitochondria. Accordingly, it has been commonly accepted that the facilitative glucose transporter Glut1 responsible for glucose entry into photoreceptors is localized in part to the outer segment plasma membrane. However, we now demonstrate that Glut1 is entirely absent from the rod outer segment and is actively excluded from this compartment by targeting information present in its cytosolic C-terminal tail. Our data indicate that glucose metabolized in the outer segment must first enter through other parts of the photoreceptor cell. Consequently, the entire energy supply of the outer segment is dependent on diffusion of energy-rich substrates through the thin connecting cilium that links this compartment to the rest of the cell.


Assuntos
Transportador de Glucose Tipo 1/metabolismo , Glucose/metabolismo , Segmento Interno das Células Fotorreceptoras da Retina/metabolismo , Segmento Externo da Célula Bastonete/metabolismo , Deleção de Sequência/genética , Animais , Animais Geneticamente Modificados , Difusão Facilitada , Transportador de Glucose Tipo 1/genética , Camundongos , Camundongos Endogâmicos , Mutagênese Sítio-Dirigida , Cílio Conector dos Fotorreceptores/ultraestrutura , Engenharia de Proteínas , Sinais Direcionadores de Proteínas/genética , Transporte Proteico , Ratos , Segmento Interno das Células Fotorreceptoras da Retina/ultraestrutura , Segmento Externo da Célula Bastonete/ultraestrutura , Transgenes/genética , Xenopus laevis/metabolismo
13.
PLoS One ; 17(6): e0268335, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35679272

RESUMO

Hyperpolarization activated cyclic nucleotide-gated channel 1 (HCN1) is expressed throughout the nervous system and is critical for regulating neuronal excitability, with mutations being associated with multiple forms of epilepsy. Adaptive modulation of HCN1 has been observed, as has pathogenic dysregulation. While the mechanisms underlying this modulation remain incompletely understood, regulation of HCN1 has been shown to include phosphorylation. A candidate phosphorylation-dependent regulator of HCN1 channels is 14-3-3. We used bioinformatics to identify three potential 14-3-3 binding sites in HCN1. We confirmed that 14-3-3 could pull down HCN1 from multiple tissue sources and used HEK293 cells to detail the interaction. Two sites in the intrinsically disordered C-terminus of HCN1 were necessary and sufficient for a phosphorylation-dependent interaction with 14-3-3. The same region of HCN1 containing the 14-3-3 binding peptides is required for phosphorylation-independent protein degradation. We propose a model in which phosphorylation of mouse S810 and S867 (human S789 and S846) recruits 14-3-3 to inhibit a yet unidentified factor signaling for protein degradation, thus increasing the half-life of HCN1.


Assuntos
Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Canais de Potássio , Animais , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Células HEK293 , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Camundongos , Neurônios/metabolismo , Canais de Potássio/genética , Canais de Potássio/metabolismo
14.
PLoS One ; 17(12): e0276298, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36477475

RESUMO

OBJECTIVE: To evaluate efficacy of a novel adeno-associated virus (AAV) vector, AAV2/4-RS1, for retinal rescue in the retinoschisin knockout (Rs1-KO) mouse model of X-linked retinoschisis (XLRS). Brinzolamide (Azopt®), a carbonic anhydrase inhibitor, was tested for its ability to potentiate the effects of AAV2/4-RS1. METHODS: AAV2/4-RS1 with a cytomegalovirus (CMV) promoter (2x1012 viral genomes/mL) was delivered to Rs1-KO mice via intravitreal (N = 5; 1µL) or subretinal (N = 21; 2µL) injections at postnatal day 60-90. Eleven mice treated with subretinal therapy also received topical Azopt® twice a day. Serial full field electroretinography (ERG) was performed starting at day 50-60 post-injection. Mice were evaluated using a visually guided swim assay (VGSA) in light and dark conditions. The experimental groups were compared to untreated Rs1-KO (N = 11), wild-type (N = 12), and Rs1-KO mice receiving only Azopt® (N = 5). Immunofluorescence staining was performed to assess RS1 protein expression following treatment. RESULTS: The ERG b/a ratio was significantly higher in the subretinal plus Azopt® (p<0.0001), subretinal without Azopt® (p = 0.0002), and intravitreal (p = 0.01) treated eyes compared to untreated eyes. There was a highly significant subretinal treatment effect on ERG amplitudes collectively at 7-9 months post-injection (p = 0.0003). Cones showed more effect than rods. The subretinal group showed improved time to platform in the dark VGSA compared to untreated mice (p<0.0001). RS1 protein expression was detected in the outer retina in subretinal treated mice and in the inner retina in intravitreal treated mice. CONCLUSIONS: AAV2/4-RS1 shows promise for improving retinal phenotype in the Rs1-KO mouse model. Subretinal delivery was superior to intravitreal. Topical brinzolamide did not improve efficacy. AAV2/4-RS1 may be considered as a potential treatment for XLRS patients.


Assuntos
Retinosquise , Camundongos , Animais , Retinosquise/genética , Retinosquise/terapia , Camundongos Knockout , Terapia Genética
15.
Traffic ; 10(6): 648-63, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19302411

RESUMO

Intraflagellar transport (IFT) provides a mechanism for the transport of cilium-specific proteins, but the mechanisms for linkage of cargo and IFT proteins have not been identified. Using the sensory outer segments (OS) of photoreceptors, which are derived from sensory cilia, we have identified IFT-cargo complexes containing IFT proteins, kinesin 2 family proteins, two photoreceptor-specific membrane proteins, guanylyl cyclase 1 (GC1, Gucy2e) and rhodopsin (RHO), and the chaperones, mammalian relative of DNAJ, DnajB6 (MRJ), and HSC70 (Hspa8). Analysis of these complexes leads to a model in which MRJ through its binding to IFT88 and GC1 plays a critical role in formation or stabilization of the IFT-cargo complexes. Consistent with the function of MRJ in the activation of HSC70 ATPase activity, Mg-ATP enhances the co-IP of GC1, RHO, and MRJ with IFT proteins. Furthermore, RNAi knockdown of MRJ in IMCD3 cells expressing GC1-green fluorescent protein (GFP) reduces cilium membrane targeting of GC1-GFP without apparent effect on cilium elongation.


Assuntos
Guanilato Ciclase/metabolismo , Chaperonas Moleculares/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Rodopsina/metabolismo , Sequência de Aminoácidos , Animais , Imuno-Histoquímica , Camundongos , Dados de Sequência Molecular , Técnicas do Sistema de Duplo-Híbrido
16.
Front Cell Neurosci ; 14: 595523, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33250719

RESUMO

Vision, hearing, smell, taste, and touch are the tools used to perceive and navigate the world. They enable us to obtain essential resources such as food and highly desired resources such as mates. Thanks to the investments in biomedical research the molecular unpinning's of human sensation are rivaled only by our knowledge of sensation in the laboratory mouse. Humans rely heavily on vision whereas mice use smell as their dominant sense. Both modalities have many features in common, starting with signal detection by highly specialized primary sensory neurons-rod and cone photoreceptors (PR) for vision, and olfactory sensory neurons (OSN) for the smell. In this chapter, we provide an overview of how these two types of primary sensory neurons operate while highlighting the similarities and distinctions.

17.
Elife ; 92020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32940604

RESUMO

Synapses are fundamental information processing units that rely on voltage-gated Ca2+ (Cav) channels to trigger Ca2+-dependent neurotransmitter release. Cav channels also play Ca2+-independent roles in other biological contexts, but whether they do so in axon terminals is unknown. Here, we addressed this unknown with respect to the requirement for Cav1.4 L-type channels for the formation of rod photoreceptor synapses in the retina. Using a mouse strain expressing a non-conducting mutant form of Cav1.4, we report that the Cav1.4 protein, but not its Ca2+ conductance, is required for the molecular assembly of rod synapses; however, Cav1.4 Ca2+ signals are needed for the appropriate recruitment of postsynaptic partners. Our results support a model in which presynaptic Cav channels serve both as organizers of synaptic building blocks and as sources of Ca2+ ions in building the first synapse of the visual pathway and perhaps more broadly in the nervous system.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Terminações Pré-Sinápticas/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Sinapses/fisiologia , Transmissão Sináptica , Animais , Masculino , Camundongos
18.
J Cell Biol ; 157(1): 103-13, 2002 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11916979

RESUMO

Approximately 10% of the photoreceptor outer segment (OS) is turned over each day, requiring large amounts of lipid and protein to be moved from the inner segment to the OS. Defects in intraphotoreceptor transport can lead to retinal degeneration and blindness. The transport mechanisms are unknown, but because the OS is a modified cilium, intraflagellar transport (IFT) is a candidate mechanism. IFT involves movement of large protein complexes along ciliary microtubules and is required for assembly and maintenance of cilia. We show that IFT particle proteins are localized to photoreceptor connecting cilia. We further find that mice with a mutation in the IFT particle protein gene, Tg737/IFT88, have abnormal OS development and retinal degeneration. Thus, IFT is important for assembly and maintenance of the vertebrate OS.


Assuntos
Proteínas de Protozoários/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/citologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Proteínas Supressoras de Tumor , Animais , Cegueira/etiologia , Bovinos , Chlamydomonas , Cílios/química , Cílios/metabolismo , Flagelos/metabolismo , Masculino , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Mutagênese Insercional/fisiologia , Proteínas de Plantas , Proteínas/genética , Proteínas de Protozoários/análise , Proteínas de Protozoários/genética , Coelhos , Células Fotorreceptoras Retinianas Bastonetes/química , Opsinas de Bastonetes/metabolismo , Testículo/química , Vesículas Transportadoras/química , Vesículas Transportadoras/metabolismo
19.
Invest Ophthalmol Vis Sci ; 60(8): 3150-3161, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31335952

RESUMO

Purpose: Cav1.4 is a voltage-gated calcium channel clustered at the presynaptic active zones of photoreceptors. Cav1.4 functions in communication by mediating the Ca2+ influx that triggers neurotransmitter release. It also aids in development since rod ribbon synapses do not form in Cav1.4 knock-out mice. Here we used a rescue strategy to investigate the ability of Cav1.4 to trigger synaptogenesis in both immature and mature mouse rods. Methods: In vivo electroporation was used to transiently express Cav α1F or tamoxifen-inducible Cav α1F in a subset of Cav1.4 knock-out mouse rods. Synaptogenesis was assayed using morphologic markers and a vision-guided water maze. Results: We found that introduction of Cav α1F to knock-out terminals rescued synaptic development as indicated by PSD-95 expression and elongated ribbons. When expression of Cav α1F was induced in mature animals, we again found restoration of PSD-95 and elongated ribbons. However, the induced expression of Cav α1F led to diffuse distribution of Cav α1F in the terminal instead of being clustered beneath the ribbon. Approximately a quarter of treated animals passed the water maze test, suggesting the rescue of retinal signaling in these mice. Conclusions: These data confirm that Cav α1F expression is necessary for rod synaptic terminal development and demonstrate that rescue is robust even in adult animals with late stages of synaptic disease. The degree of rod synaptic plasticity seen here should be sufficient to support future vision-restoring treatments such as gene or cell replacement that will require photoreceptor synaptic rewiring.


Assuntos
Canais de Cálcio Tipo L/genética , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Transmissão Sináptica/genética , Animais , Animais Recém-Nascidos , Canais de Cálcio Tipo L/metabolismo , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Terminações Pré-Sinápticas/metabolismo , Sinapses/metabolismo
20.
Vision Res ; 48(3): 413-23, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17931679

RESUMO

Intraflagellar transport (IFT) of a approximately 17S particle containing at least 16 distinct polypeptides is required for the assembly and maintenance of cilia and flagella. Although both genetic and biochemical evidence suggest a role for IFT in vertebrate photoreceptors, the spatial distribution of IFT proteins within photoreceptors remains poorly defined. We have evaluated the distribution of 4 IFT proteins using a combination of immunocytochemistry and rod-specific overexpression of GFP tagged IFT proteins. Endogenous IFT proteins are most highly concentrated within the inner segment, around the basal body, and within the outer segment IFT proteins are localized in discrete particles along the entire length of the axoneme. IFT52-GFP and IFT57-GFP mimicked this pattern in transgenic Xenopus.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas do Olho/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Animais , Animais Geneticamente Modificados , Embrião não Mamífero/metabolismo , Camundongos , Retina/diagnóstico por imagem , Retina/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Ultrassonografia , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA