Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 296: 100259, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33837742

RESUMEN

The ability of iron to transfer electrons enables the contribution of this metal to a variety of cellular activities even as the redox properties of iron are also responsible for the generation of hydroxyl radicals (•OH), the most destructive of the reactive oxygen species. We previously showed that iron can promote the oxidation of bisretinoid by generating highly reactive hydroxyl radical (•OH). Now we report that preservation of iron regulation in the retina is not sufficient to prevent iron-induced bisretinoid oxidative degradation when blood iron levels are elevated in liver-specific hepcidin knockout mice. We obtained evidence for the perpetuation of Fenton reactions in the presence of the bisretinoid A2E and visible light. On the other hand, iron chelation by deferiprone was not associated with changes in postbleaching recovery of 11-cis-retinal or dark-adapted ERG b-wave amplitudes indicating that the activity of Rpe65, a rate-determining visual cycle protein that carries an iron-binding domain, is not affected. Notably, iron levels were elevated in the neural retina and retinal pigment epithelial (RPE) cells of Abca4-/- mice. Consistent with higher iron content, ferritin-L immunostaining was elevated in RPE of a patient diagnosed with ABCA4-associated disease and in RPE and photoreceptor cells of Abca4-/- mice. In neural retina of the mutant mice, reduced Tfrc mRNA was also an indicator of retinal iron overload. Thus iron chelation may defend retina when bisretinoid toxicity is implicated in disease processes.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Genes Recesivos , Retinaldehído/metabolismo , Retinoides/metabolismo , Enfermedad de Stargardt/metabolismo , cis-trans-Isomerasas/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Animales , Ratones , Ratones Noqueados , Oxidación-Reducción , Retinaldehído/genética , Retinoides/genética , Enfermedad de Stargardt/genética , Enfermedad de Stargardt/patología , cis-trans-Isomerasas/genética
2.
J Mol Biol ; 432(19): 5378-5389, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32795534

RESUMEN

Rhodopsin is the photosensitive protein, which binds to 11-cis-retinal as its chromophore. In the dark, rhodopsin exists as a stable complex between the opsin moiety and 11-cis-retinal. The absorption of a light photon converts 11-cis-retinal to all-trans-retinal and initiates our vision. As a result, the increase in the rate of dark activation of rhodopsin reduces its photosensitivity resulting in night blindness. The mutations, G90D and T94I are night blindness-causing mutations that exhibit completely different physicochemical characteristics associated with the dark activation of rhodopsin, such as a high rate of thermal isomerization of 11-cis-retinal and a slow pigment regeneration. To elucidate the molecular mechanism by which G90D and T94I mutations affect rhodopsin dark activation and regeneration, we performed light-induced difference FTIR spectroscopy on dark and primary photo-intermediate states of G90D and T94I mutants. The FTIR spectra clearly show that both charged G90D and hydrophobic T94I mutants alter the H-bond network at the Schiff base region of the chromophore, which weakens the electrostatic interaction with Glu113 counterion. Our results further show an altered water-mediated H-bond network around the central transmembrane region of mutant rhodopsin, which is reminiscent of the active Meta-II state. This altered water-mediated H-bond network may cause thermal isomerization of the chromophore and facilitate rhodopsin dark activation.


Asunto(s)
Ceguera Nocturna/genética , Rodopsina/genética , Animales , Bovinos , Enlace de Hidrógeno , Isomerismo , Modelos Moleculares , Ceguera Nocturna/metabolismo , Mutación Puntual , Conformación Proteica , Retinaldehído/química , Retinaldehído/genética , Retinaldehído/metabolismo , Rodopsina/química , Rodopsina/metabolismo
3.
Appl Environ Microbiol ; 84(24)2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30315080

RESUMEN

Freshwater lakes harbor complex microbial communities, but these ecosystems are often dominated by acI Actinobacteria Members of this cosmopolitan lineage are proposed to bolster heterotrophic growth using phototrophy because their genomes encode actino-opsins (actR). This model has been difficult to validate experimentally because acI Actinobacteria are not consistently culturable. Based primarily on genomes from single cells and metagenomes, we provide a detailed biosynthetic route for members of acI clades A and B to synthesize retinal and its carotenoid precursors. Consequently, acI cells should be able to natively assemble light-driven actinorhodopsins (holo-ActR) to pump protons, unlike many bacteria that encode opsins but may need to exogenously obtain retinal because they lack retinal machinery. Moreover, we show that all acI clades contain genes for a secondary branch of the carotenoid pathway, implying synthesis of a complex carotenoid. Transcription analysis of acI Actinobacteria in a eutrophic lake shows that all retinal and carotenoid pathway operons are transcribed and that actR is among the most highly transcribed of all acI genes. Furthermore, heterologous expression of acI retinal pathway genes showed that lycopene, retinal, and ActR can be made using the genes encoded in these organisms. Model cells producing ActR and the key acI retinal-producing ß-carotene oxygenase formed holo-ActR and acidified solution during illumination. Taken together, our results prove that acI Actinobacteria containing both ActR and acI retinal production machinery have the capacity to natively synthesize a green light-dependent outward proton-pumping rhodopsin.IMPORTANCE Microbes play critical roles in determining the quality of freshwater ecosystems, which are vital to human civilization. Because acI Actinobacteria are ubiquitous and abundant in freshwater lakes, clarifying their ecophysiology is a major step in determining the contributions that they make to nitrogen and carbon cycling. Without accurate knowledge of these cycles, freshwater systems cannot be incorporated into climate change models, ecosystem imbalances cannot be predicted, and policy for service disruption cannot be planned. Our work fills major gaps in microbial light utilization, secondary metabolite production, and energy cycling in freshwater habitats.


Asunto(s)
Actinobacteria/genética , Actinobacteria/metabolismo , Genes Bacterianos/genética , Lagos/microbiología , Retinaldehído/biosíntesis , Retinaldehído/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Carotenoides/genética , Carotenoides/metabolismo , Ecosistema , Redes y Vías Metabólicas/genética , Modelos Moleculares , Opsinas/genética , Opsinas/metabolismo , Procesos Fototróficos , Bombas de Protones , Rodopsina , Análisis de Secuencia de Proteína
4.
Biochem J ; 475(20): 3171-3188, 2018 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-30352831

RESUMEN

The vitamin A derivative 11-cis-retinaldehyde plays a pivotal role in vertebrate vision by serving as the chromophore of rod and cone visual pigments. In the initial step of vision, a photon is absorbed by this chromophore resulting in its isomerization to an all-trans state and consequent activation of the visual pigment and phototransduction cascade. Spent chromophore is released from the pigments through hydrolysis. Subsequent photon detection requires the delivery of regenerated 11-cis-retinaldehyde to the visual pigment. This trans-cis conversion is achieved through a process known as the visual cycle. In this review, we will discuss the enzymes, binding proteins and transporters that enable the visual pigment renewal process with a focus on advances made during the past decade in our understanding of their structural biology.


Asunto(s)
Epitelio Pigmentado de la Retina/metabolismo , Pigmentos Retinianos/metabolismo , Retinaldehído/metabolismo , Visión Ocular/fisiología , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , Humanos , Estructura Secundaria de Proteína , Pigmentos Retinianos/química , Pigmentos Retinianos/genética , Retinaldehído/química , Retinaldehído/genética
5.
Proc Natl Acad Sci U S A ; 115(19): 4963-4968, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29686088

RESUMEN

Intracellular Fe plays a key role in redox active energy and electron transfer. We sought to understand how Fe levels impact the retina, given that retinal pigment epithelial (RPE) cells are also challenged by accumulations of vitamin A aldehyde adducts (bisretinoid lipofuscin) that photogenerate reactive oxygen species and photodecompose into damaging aldehyde- and dicarbonyl-bearing species. In mice treated with the Fe chelator deferiprone (DFP), intracellular Fe levels, as reflected in transferrin receptor mRNA expression, were reduced. DFP-treated albino Abca4-/- and agouti wild-type mice exhibited elevated bisretinoid levels as measured by high-performance liquid chromatography or noninvasively by quantitative fundus autofluorescence. Thinning of the outer nuclear layer, a parameter indicative of the loss of photoreceptor cell viability, was also reduced in DFP-treated albino Abca4-/- In contrast to the effects of the Fe chelator, mice burdened with increased intracellular Fe in RPE due to deficiency in the Fe export proteins hephaestin and ceruloplasmin, presented with reduced bisretinoid levels. These findings indicate that intracellular Fe promotes bisretinoid oxidation and degradation. This interpretation was supported by experiments showing that DFP decreased the oxidative/degradation of the bisretinoid A2E in the presence of light and reduced cell death in cell-based experiments. Moreover, light-independent oxidation and degradation of A2E by Fenton chemistry products were evidenced by the consumption of A2E, release of dicarbonyls, and generation of oxidized A2E species in cell-free assays.


Asunto(s)
Epitelio Pigmentado de la Retina/metabolismo , Retinoides/metabolismo , Transportadoras de Casetes de Unión a ATP/deficiencia , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Deferiprona , Hierro , Quelantes del Hierro/farmacología , Lipofuscina/genética , Lipofuscina/metabolismo , Ratones , Ratones Noqueados , Piridonas/farmacología , Epitelio Pigmentado de la Retina/patología , Retinaldehído/genética , Retinaldehído/metabolismo
6.
J Biol Chem ; 292(26): 10983-10997, 2017 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-28487362

RESUMEN

Phototransduction is initiated when the absorption of light converts the 11-cis-retinal chromophore to its all-trans configuration in both rod and cone vertebrate photoreceptors. To sustain vision, 11-cis-retinal is continuously regenerated from its all-trans conformation through a series of enzymatic steps comprising the "visual or retinoid" cycle. Abnormalities in this cycle can compromise vision because of the diminished supply of 11-cis-retinal and the accumulation of toxic, constitutively active opsin. As shown previously for rod cells, attenuation of constitutively active opsin can be achieved with the unbleachable analogue, 11-cis-6-membered ring (11-cis-6mr)-retinal, which has therapeutic effects against certain degenerative retinal diseases. However, to discern the molecular mechanisms responsible for this action, pigment regeneration with this locked retinal analogue requires delineation also in cone cells. Here, we compared the regenerative properties of rod and green cone opsins with 11-cis-6mr-retinal and demonstrated that this retinal analogue could regenerate rod pigment but not green cone pigment. Based on structural modeling suggesting that Pro-205 in green cone opsin could prevent entry and binding of 11-cis-6mr-retinal, we initially mutated this residue to Ile, the corresponding residue in rhodopsin. However, this substitution did not enable green cone opsin to regenerate with 11-cis-6mr-retinal. Interestingly, deletion of 16 N-terminal amino acids in green cone opsin partially restored the binding of 11-cis-6mr-retinal. These results and our structural modeling indicate that a more complex binding pathway determines the regeneration of mammalian green cone opsin with chromophore analogues such as 11-cis-6mr-retinal.


Asunto(s)
Modelos Moleculares , Opsinas/química , Retinaldehído/química , Animales , Humanos , Opsinas/genética , Opsinas/metabolismo , Retinaldehído/genética , Retinaldehído/metabolismo , Células Sf9 , Spodoptera
7.
Biochemistry ; 55(16): 2371-80, 2016 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-27039989

RESUMEN

Optogenetics relies on the expression of specific microbial rhodopsins in the neuronal plasma membrane. Most notably, this includes channelrhodopsins, which when heterologously expressed in neurons function as light-gated cation channels. Recently, a new class of microbial rhodopsins, termed anion channel rhodopsins (ACRs), has been discovered. These proteins function as efficient light-activated channels strictly selective for anions. They exclude the flow of protons and other cations and cause hyperpolarization of the membrane potential in neurons by allowing the inward flow of chloride ions. In this study, confocal near-infrared resonance Raman spectroscopy (RRS) along with hydrogen/deuterium exchange, retinal analogue substitution, and site-directed mutagenesis were used to study the retinal structure as well as its interactions with the protein in the unphotolyzed state of an ACR from Guillardia theta (GtACR1). These measurements reveal that (i) the retinal chromophore exists as an all-trans configuration with a protonated Schiff base (PSB) very similar to that of bacteriorhodopsin (BR), (ii) the chromophore RRS spectrum is insensitive to changes in pH from 3 to 11, whereas above this pH the Schiff base (SB) is deprotonated, (iii) when Ser97, the homologue to Asp85 in BR, is replaced with a Glu, it remains in a neutral form (i.e., as a carboxylic acid) but is deprotonated at higher pH to form a blue-shifted species, (iv) Asp234, the homologue of the protonated retinylidene SB counterion Asp212 in BR, does not serve as the primary counteranion for the protonated SB, and (v) substitution of Glu68 with an Gln increases the pH at which SB deprotonation is observed. These results suggest that Glu68 and Asp234 located near the SB exist in a neutral state in unphotolyzed GtACR1 and indicate that other unidentified negative charges stabilize the protonated state of the GtACR1 SB.


Asunto(s)
Criptófitas/química , Rodopsina/química , Sustitución de Aminoácidos , Criptófitas/genética , Conformación Proteica , Retinaldehído/química , Retinaldehído/genética , Retinoides/química , Retinoides/genética , Rodopsina/genética , Bases de Schiff/química , Espectrometría Raman
8.
FASEB J ; 29(11): 4579-88, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26187344

RESUMEN

Age-related macular degeneration (AMD) is a neurodegenerative disease that causes adult-onset blindness. There are 2 forms of this progressive disease: wet and dry. Currently there is no cure for AMD, but several treatment options have started to emerge making early detection critical for therapeutic success. Analysis of the eyes of Abca4(-/-)Rdh8(-/-) mice that display light-induced retinal degeneration indicates that 11-cis-retinal and docosahexaenoic acid (DHA) levels were significantly decreased as compared with the eyes of control dark-adapted C57BL/6J mice. In addition, exposure to intense light correlated with higher levels of prostaglandin G2 in the eyes of Abca4(-/-)Rdh8(-/-) mice. Intense light exposure also lowered DHA levels in the eyes of wild-type C57BL/6J mice without discernible retinal degeneration. Analysis of human serum from patients with AMD recapitulated these dysregulated DHA levels and revealed dysregulation of arachidonic acid (AA) levels as well (∼32% increase in patients with AMD compared with average levels in healthy individuals). From these observations, we then built a statistical model that included levels of DHA and AA from human serum. This model had a 74% probability of correctly identifying patients with AMD from controls. Addition of a genetic analysis for one of the most prevalent amino acid substitutions in the age-related maculopathy susceptibility 2 gene linked to AMD, Ala(69)→Ser, did not improve the statistical model. Thus, we have characterized a reliable method with the potential to detect AMD without a genetic component, paving the way for a larger-scale clinical evaluation. Our studies on mouse models along with the analysis of human serum suggest that our small molecule-based model may serve as an effective tool to estimate the risk of developing AMD.


Asunto(s)
Ácidos Docosahexaenoicos/sangre , Degeneración Macular/sangre , Modelos Biológicos , Retinaldehído/sangre , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Adulto , Anciano , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Sustitución de Aminoácidos , Animales , Ácidos Docosahexaenoicos/genética , Femenino , Humanos , Degeneración Macular/genética , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Mutación Missense , Retinaldehído/genética
9.
Hum Mol Genet ; 24(15): 4417-28, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25972377

RESUMEN

Human RPE65 mutations cause a spectrum of blinding retinal dystrophies from severe early-onset disease to milder manifestations. The RPE65 P25L missense mutation, though having <10% of wild-type (WT) activity, causes relatively mild retinal degeneration. To better understand these mild forms of RPE65-related retinal degeneration, and their effect on cone photoreceptor survival, we generated an Rpe65/P25L knock-in (KI/KI) mouse model. We found that, when subject to the low-light regime (∼100 lux) of regular mouse housing, homozygous Rpe65/P25L KI/KI mice are morphologically and functionally very similar to WT siblings. While mutant protein expression is decreased by over 80%, KI/KI mice retinae retain comparable 11-cis-retinal levels with WT. Consistently, the scotopic and photopic electroretinographic (ERG) responses to single-flash stimuli also show no difference between KI/KI and WT mice. However, the recovery of a-wave response following moderate visual pigment bleach is delayed in KI/KI mice. Importantly, KI/KI mice show significantly increased resistance to high-intensity (20 000 lux for 30 min) light-induced retinal damage (LIRD) as compared with WT, indicating impaired rhodopsin regeneration in KI/KI. Taken together, the Rpe65/P25L mutant produces sufficient chromophore under normal conditions to keep opsins replete and thus manifests a minimal phenotype. Only when exposed to intensive light is this hypomorphic mutation manifested physiologically, as its reduced expression and catalytic activity protects against the successive cycles of opsin regeneration underlying LIRD. These data also help define minimal requirements of chromophore for photoreceptor survival in vivo and may be useful in assessing a beneficial therapeutic dose for RPE65 gene therapy in humans.


Asunto(s)
Retina/metabolismo , Degeneración Retiniana/genética , Retinaldehído/genética , cis-trans-Isomerasas/genética , Animales , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Humanos , Luz , Ratones , Mutación Missense , Opsinas/genética , Opsinas/metabolismo , Retina/patología , Células Fotorreceptoras Retinianas Conos/patología , Degeneración Retiniana/fisiopatología , Retinaldehído/biosíntesis , cis-trans-Isomerasas/metabolismo
10.
FEBS Lett ; 589(10): 1067-71, 2015 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-25836735

RESUMEN

Histidine kinase rhodopsin 1 is a photoreceptor in green algae functioning as a UV-light sensor. It switches between a UV-absorbing state (Rh-UV) and a blue-absorbing state (Rh-Bl) with a protonated retinal Schiff base (RSB) cofactor in a mixture of 13-trans,15-anti and 13-cis,15-syn isomers. The present spectroscopic study now shows that cofactor-protein assembly stabilizes the protonated 13-trans,15-anti RSB isomer. Formation of the active photoswitch requires the photoinduced conversion to Rh-UV. The transitions between the Rh-Bl isomers and the deprotonated 13-cis,15-anti and 13-trans,15-syn isomers of Rh-UV proceed via multiple photoisomerizations of one or simultaneously two double bonds.


Asunto(s)
Chlamydomonas reinhardtii/enzimología , Proteínas de Plantas/química , Proteínas Quinasas/química , Retinaldehído/química , Rodopsina/química , Rayos Ultravioleta , Chlamydomonas reinhardtii/genética , Histidina Quinasa , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Retinaldehído/genética , Retinaldehído/metabolismo , Rodopsina/genética , Rodopsina/metabolismo , Bases de Schiff
11.
Biochim Biophys Acta ; 1842(7): 1109-20, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24717912

RESUMEN

Toll-like receptor (TLR) signaling plays a fundamental role in the induction and progression of autoimmune disease. In the present study, we showed that lipopolysaccharide (LPS), a TLR4 ligand, functions as an antagonist of peroxisome proliferator-activated receptor alpha (PPARα), a nuclear transcription factor. Using endotoxin induced uveitis (EIU) as a model, we found that TLR was negatively regulated by PPARα. Our data revealed that treatment with the PPARα agonist fenofibrate dramatically prevented LPS-induced uveitis and inhibited TLR/ Nuclear factor-kappaB (NF-κB) signaling during inflammation. Evaluation of the severity of anterior uveitis further showed that PPARα agonist treatment significantly decreased inflammatory cell infiltration, total protein concentration, vessel density, inflammatory cytokine production, and clinical scores in the anterior section of the eye during EIU. Moreover, fenofibrate administration recovered retinal function and decreased the production of inflammatory cytokines, retinal vascular leukostasis, and inflammatory cell infiltration into the posterior section of the eyes during EIU. In vitro studies further showed that down-regulation or deletion of PPARα led to increased TLR4 levels and the activation of NF-κB signaling in RPE cells and also blocked the anti-inflammatory effects of fenofibrate. Furthermore, activation or up-regulation of PPARα decreased TLR4 levels and inhibited the NF-κB signaling pathway induced by LPS in RPE cells. In TLR4-expressing reporter cells, activation or up-regulation of PPARα partially inhibited the activation of NF-κB and also decreased TLR4 transcriptional activity. In conclusion, the activation of PPARα represents a novel therapeutic strategy for human uveitis, as PPARα negatively regulates TLR4 activity and therefore exerts anti-inflammatory actions.


Asunto(s)
Endotoxinas/inmunología , FN-kappa B/metabolismo , PPAR gamma/metabolismo , Receptor Toll-Like 4/metabolismo , Uveítis/metabolismo , Animales , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Fenofibrato/inmunología , Fenofibrato/metabolismo , Leucostasis/genética , Leucostasis/inmunología , Leucostasis/metabolismo , Lipopolisacáridos/inmunología , Masculino , FN-kappa B/genética , FN-kappa B/inmunología , PPAR gamma/genética , PPAR gamma/inmunología , Distribución Aleatoria , Ratas , Ratas Endogámicas Lew , Retinaldehído/genética , Retinaldehído/metabolismo , Transducción de Señal , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Transcripción Genética/genética , Transcripción Genética/inmunología , Uveítis/genética , Uveítis/inmunología
12.
J Biol Chem ; 289(19): 13661-6, 2014 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-24668807

RESUMEN

ß-Carotene 15-15'-oxygenase (BCO1) catalyzes the oxidative cleavage of dietary provitamin A carotenoids to retinal (vitamin A aldehyde). Aldehydes readily exchange their carbonyl oxygen with water, making oxygen labeling experiments challenging. BCO1 has been thought to be a monooxygenase, incorporating oxygen from O2 and H2O into its cleavage products. This was based on a study that used conditions that favored oxygen exchange with water. We incubated purified recombinant human BCO1 and ß-carotene in either (16)O2-H2(18)O or (18)O2-H2(16)O medium for 15 min at 37 °C, and the relative amounts of (18)O-retinal and (16)O-retinal were measured by liquid chromatography-tandem mass spectrometry. At least 79% of the retinal produced by the reaction has the same oxygen isotope as the O2 gas used. Together with the data from (18)O-retinal-H2(16)O and (16)O-retinal-H2(18)O incubations to account for nonenzymatic oxygen exchange, our results show that BCO1 incorporates only oxygen from O2 into retinal. Thus, BCO1 is a dioxygenase.


Asunto(s)
Dioxigenasas/química , Oxígeno/química , Retinaldehído/química , Vitamina A/biosíntesis , Dioxigenasas/genética , Dioxigenasas/metabolismo , Humanos , Oxígeno/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Retinaldehído/genética , Retinaldehído/metabolismo , Vitamina A/química , Vitamina A/genética
13.
J Biol Chem ; 289(7): 3991-4000, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24403072

RESUMEN

Opn5 is one of the recently identified opsin groups that is responsible for nonvisual photoreception in animals. We previously showed that a chicken homolog of mammalian Opn5 (Opn5m) is a Gi-coupled UV sensor having molecular properties typical of bistable pigments. Here we demonstrated that mammalian Opn5m evolved to be a more specialized photosensor by losing one of the characteristics of bistable pigments, direct binding of all-trans-retinal. We first confirmed that Opn5m proteins in zebrafish, Xenopus tropicalis, mouse, and human are also UV-sensitive pigments. Then we found that only mammalian Opn5m proteins lack the ability to directly bind all-trans-retinal. Mutational analysis showed that these characteristics were acquired by a single amino acid replacement at position 168. By comparing the expression patterns of Opn5m between mammals and chicken, we found that, like chicken Opn5m, mammalian Opn5m was localized in the ganglion cell layer and inner nuclear layer of the retina. However, the mouse and primate (common marmoset) opsins were distributed not in the posterior hypothalamus (including the region along the third ventricle) where chicken Opn5m is localized, but in the preoptic hypothalamus. Interestingly, RPE65, an essential enzyme for forming 11-cis-retinal in the visual cycle is expressed near the preoptic hypothalamus of the mouse and common marmoset brain but not near the region of the chicken brain where chicken Opn5m is expressed. Therefore, mammalian Opn5m may work exclusively as a short wavelength sensor in the brain as well as in the retina with the assistance of an 11-cis-retinal-supplying system.


Asunto(s)
Encéfalo/metabolismo , Evolución Molecular , Proteínas de la Membrana/metabolismo , Mutación Missense , Opsinas/metabolismo , Retina/metabolismo , Rayos Ultravioleta , Sustitución de Aminoácidos , Animales , Callithrix , Embrión de Pollo , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos ICR , Opsinas/genética , Retinaldehído/genética , Retinaldehído/metabolismo , Xenopus , Pez Cebra , cis-trans-Isomerasas/genética , cis-trans-Isomerasas/metabolismo
14.
J Biol Chem ; 288(47): 33912-33926, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24106275

RESUMEN

Autosomal dominant retinitis pigmentosa (ADRP) mutants (T4K, N15S, T17M, V20G, P23A/H/L, and Q28H) in the N-terminal cap of rhodopsin misfold when expressed in mammalian cells. To gain insight into the causes of misfolding and to define the contributions of specific residues to receptor stability and function, we evaluated the responses of these mutants to 11-cis-retinal pharmacological chaperone rescue or disulfide bond-mediated repair. Pharmacological rescue restored folding in all mutants, but the purified mutant pigments in all cases were thermo-unstable and exhibited abnormal photobleaching, metarhodopsin II decay, and G protein activation. As a complementary approach, we superimposed this panel of ADRP mutants onto a rhodopsin background containing a juxtaposed cysteine pair (N2C/D282C) that forms a disulfide bond. This approach restored folding in T4K, N15S, V20G, P23A, and Q28H but not T17M, P23H, or P23L. ADRP mutant pigments obtained by disulfide bond repair exhibited enhanced stability, and some also displayed markedly improved photobleaching and signal transduction properties. Our major conclusion is that the N-terminal cap stabilizes opsin during biosynthesis and contributes to the dark-state stability of rhodopsin. Comparison of these two restorative approaches revealed that the correct position of the cap relative to the extracellular loops is also required for optimal photochemistry and efficient G protein activation.


Asunto(s)
Mutación Missense , Pliegue de Proteína , Retinitis Pigmentosa/metabolismo , Rodopsina/metabolismo , Sustitución de Aminoácidos , Animales , Bovinos , Células HEK293 , Humanos , Estabilidad Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Retinaldehído/genética , Retinaldehído/metabolismo , Retinitis Pigmentosa/genética , Rodopsina/genética
15.
J Biol Chem ; 288(48): 34484-93, 2013 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-24129572

RESUMEN

Differentiated retinal pigmented epithelial (RPE) cells have been obtained from human induced pluripotent stem (hiPS) cells. However, the visual (retinoid) cycle in hiPS-RPE cells has not been adequately examined. Here we determined the expression of functional visual cycle enzymes in hiPS-RPE cells compared with that of isolated wild-type mouse primary RPE (mpRPE) cells in vitro and in vivo. hiPS-RPE cells appeared morphologically similar to mpRPE cells. Notably, expression of certain visual cycle proteins was maintained during cell culture of hiPS-RPE cells, whereas expression of these same molecules rapidly decreased in mpRPE cells. Production of the visual chromophore, 11-cis-retinal, and retinosome formation also were documented in hiPS-RPE cells in vitro. When mpRPE cells with luciferase activity were transplanted into the subretinal space of mice, bioluminance intensity was preserved for >3 months. Additionally, transplantation of mpRPE into blind Lrat(-/-) and Rpe65(-/-) mice resulted in the recovery of visual function, including increased electrographic signaling and endogenous 11-cis-retinal production. Finally, when hiPS-RPE cells were transplanted into the subretinal space of Lrat(-/-) and Rpe65(-/-) mice, their vision improved as well. Moreover, histological analyses of these eyes displayed replacement of dysfunctional RPE cells by hiPS-RPE cells. Together, our results show that hiPS-RPE cells can exhibit a functional visual cycle in vitro and in vivo. These cells could provide potential treatment options for certain blinding retinal degenerative diseases.


Asunto(s)
Células Madre Pluripotentes Inducidas/trasplante , Degeneración Retiniana/genética , Degeneración Retiniana/terapia , Epitelio Pigmentado de la Retina/trasplante , cis-trans-Isomerasas/genética , Animales , Diferenciación Celular , Células Cultivadas , Regulación Enzimológica de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/enzimología , Ratones , Degeneración Retiniana/patología , Epitelio Pigmentado de la Retina/enzimología , Retinaldehído/biosíntesis , Retinaldehído/genética , Visión Ocular/genética , Visión Ocular/fisiología , cis-trans-Isomerasas/deficiencia
16.
Dev Dyn ; 242(9): 1056-65, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23765990

RESUMEN

BACKGROUND: Retinoic acid (RA) signaling controls patterning and neuronal differentiation within the hindbrain, but forebrain RA function remains controversial. RA is produced from metabolism of retinol to retinaldehyde by retinol dehydrogenase (RDH), followed by metabolism of retinaldehyde to RA by retinaldehyde dehydrogenase (RALDH). Previous studies on Raldh2-/- and Raldh3-/- mice demonstrated an RA requirement for γ-aminobutyric acid (GABA)ergic and dopaminergic differentiation in forebrain basal ganglia, but no RA requirement was observed during early forebrain patterning or subsequent forebrain cortical expansion. However, other studies suggested that RA controls forebrain patterning, and analysis of ethylnitrosourea-induced Rdh10 mutants suggested that RA synthesized in the meninges stimulates forebrain cortical expansion. RESULTS: We generated Rdh10-/- mouse embryos that lack RA activity early in the head and later in the meninges. We observed defects in hindbrain patterning and eye RA signaling, but early forebrain patterning was unaffected. Retinaldehyde treatment of Rdh10-/- embryos from E7-E9 rescues a cranial skeletal defect, resulting in E14.5 embryos lacking meningeal RA activity but maintaining normal forebrain shape and cortical expansion. CONCLUSIONS: Rdh10-/- embryos demonstrate that RA controls hindbrain but not early forebrain patterning, while studies on retinaldehyde-rescued Rdh10-/- embryos show that meningeal RA synthesis is unnecessary to stimulate forebrain cortical expansion.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Embrión de Mamíferos/embriología , Neurogénesis/fisiología , Prosencéfalo/embriología , Retinaldehído/metabolismo , Tretinoina/metabolismo , Oxidorreductasas de Alcohol/genética , Animales , Embrión de Mamíferos/citología , Ojo/citología , Ojo/embriología , Ratones , Ratones Noqueados , Prosencéfalo/citología , Retinaldehído/genética , Transducción de Señal/fisiología
17.
J Biol Chem ; 288(21): 15326-41, 2013 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-23572532

RESUMEN

Although several genetic and biochemical factors are associated with the pathogenesis of retinal degeneration, it has yet to be determined how these different impairments can cause similar degenerative phenotypes. Here, we report microglial/macrophage activation in both a Stargardt disease and age-related macular degeneration mouse model caused by delayed clearance of all-trans-retinal from the retina, and in a retinitis pigmentosa mouse model with impaired retinal pigment epithelium (RPE) phagocytosis. Mouse microglia displayed RPE cytotoxicity and increased production of inflammatory chemokines/cytokines, Ccl2, Il1b, and Tnf, after coincubation with ligands that activate innate immunity. Notably, phagocytosis of photoreceptor proteins increased the activation of microglia/macrophages and RPE cells isolated from model mice as well as wild-type mice. The mRNA levels of Tlr2 and Tlr4, which can recognize proteins as their ligands, were elevated in mice with retinal degeneration. Bone marrow-derived macrophages from Tlr4-deficient mice did not increase Ccl2 after coincubation with photoreceptor proteins. Tlr4(-/-)Abca4(-/-)Rdh8(-/-) mice displayed milder retinal degenerative phenotypes than Abca4(-/-)Rdh8(-/-) mice. Additionally, inactivation of microglia/macrophages by pharmacological approaches attenuated mouse retinal degeneration. This study demonstrates an important contribution of TLR4-mediated microglial activation by endogenous photoreceptor proteins in retinal inflammation that aggravates retinal cell death. This pathway is likely to represent an underlying common pathology in degenerative retinal disorders.


Asunto(s)
Proteínas del Ojo/metabolismo , Degeneración Macular/metabolismo , Microglía/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Retinaldehído/metabolismo , Retinitis Pigmentosa/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Proteínas del Ojo/genética , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Degeneración Macular/genética , Degeneración Macular/patología , Ratones , Ratones Noqueados , Microglía/patología , Células Fotorreceptoras de Vertebrados/patología , Retinaldehído/genética , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Receptor Toll-Like 4/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
18.
Proc Natl Acad Sci U S A ; 110(13): 4998-5003, 2013 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-23479626

RESUMEN

Most opsins selectively bind 11-cis retinal as a chromophore to form a photosensitive pigment, which underlies various physiological functions, such as vision and circadian photoentrainment. Recently, opsin 3 (Opn3), originally called encephalopsin or panopsin, and its homologs were identified in various tissues including brain, eye, and liver in both vertebrates and invertebrates, including human. Because Opn3s are mainly expressed in tissues that are not considered to contain sufficient amounts of 11-cis retinal to form pigments, the photopigment formation ability of Opn3 has been of interest. Here, we report the successful expression of Opn3 homologs, pufferfish teleost multiple tissue opsin (PufTMT) and mosquito Opn3 (MosOpn3) and show that these proteins formed functional photopigments with 11-cis and 9-cis retinals. The PufTMT- and MosOpn3-based pigments have absorption maxima in the blue-to-green region and exhibit a bistable nature. These Opn3 homolog-based pigments activate Gi-type and Go-type G proteins light dependently, indicating that they potentially serve as light-sensitive Gi/Go-coupled receptors. We also demonstrated that mammalian cultured cells transfected with the MosOpn3 or PufTMT became light sensitive without the addition of 11-cis retinal and the photosensitivity retained after the continuous light exposure, showing a reusable pigment formation with retinal endogenously contained in culture medium. Interestingly, we found that the MosOpn3 also acts as a light sensor when constituted with 13-cis retinal, a ubiquitously present retinal isomer. Our findings suggest that homologs of vertebrate Opn3 might function as photoreceptors in various tissues; furthermore, these Opn3s, particularly the mosquito homolog, could provide a promising optogenetic tool for regulating cAMP-related G protein-coupled receptor signalings.


Asunto(s)
Anopheles , Proteínas de Peces/biosíntesis , Proteínas de Insectos/biosíntesis , Opsinas/biosíntesis , Receptores Acoplados a Proteínas G/biosíntesis , Retinaldehído/metabolismo , Tetraodontiformes , Animales , Secuencia de Bases , Proteínas de Peces/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Células HEK293 , Humanos , Proteínas de Insectos/genética , Luz , Datos de Secuencia Molecular , Opsinas/genética , Receptores Acoplados a Proteínas G/genética , Retinaldehído/genética , Homología de Secuencia de Aminoácido , Transducción de Señal/fisiología , Transducción de Señal/efectos de la radiación
19.
PLoS One ; 8(3): e57487, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23526944

RESUMEN

Proteorhodopsin (PR) photoheterotrophy in the marine flavobacterium Dokdonia sp. PRO95 has previously been investigated, showing no growth stimulation in the light at intermediate carbon concentrations. Here we report the genome sequence of strain PRO95 and compare it to two other PR encoding Dokdonia genomes: that of strain 4H-3-7-5 which shows the most similar genome, and that of strain MED134 which grows better in the light under oligotrophic conditions. Our genome analysis revealed that the PRO95 genome as well as the 4H-3-7-5 genome encode a protein related to xanthorhodopsins. The genomic environment and phylogenetic distribution of this gene suggest that it may have frequently been recruited by lateral gene transfer. Expression analyses by RT-PCR and direct mRNA-sequencing showed that both rhodopsins and the complete ß-carotene pathway necessary for retinal production are transcribed in PRO95. Proton translocation measurements showed enhanced proton pump activity in response to light, supporting that one or both rhodopsins are functional. Genomic information and carbon source respiration data were used to develop a defined cultivation medium for PRO95, but reproducible growth always required small amounts of yeast extract. Although PRO95 contains and expresses two rhodopsin genes, light did not stimulate its growth as determined by cell numbers in a nutrient poor seawater medium that mimics its natural environment, confirming previous experiments at intermediate carbon concentrations. Starvation or stress conditions might be needed to observe the physiological effect of light induced energy acquisition.


Asunto(s)
Proteínas Bacterianas/genética , Flavobacteriaceae/genética , Flavobacteriaceae/fisiología , Rodopsina/genética , Rodopsinas Microbianas/genética , Carotenoides/biosíntesis , Carotenoides/genética , Flavobacteriaceae/clasificación , Transferencia de Gen Horizontal , Genoma Bacteriano , Filogenia , Retinaldehído/biosíntesis , Retinaldehído/genética , Agua de Mar/microbiología , Especificidad de la Especie
20.
J Biol Chem ; 288(15): 10451-8, 2013 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-23439646

RESUMEN

Channelrhodopsins are microbial type rhodopsins that operate as light-gated ion channels. Largely prolonged lifetimes of the conducting state of channelrhodopsin-2 may be achieved by mutations of crucial single amino acids, i.e. cysteine 128. Such mutants are of great scientific interest in the field of neurophysiology because they allow neurons to be switched on and off on demand (step function rhodopsins). Due to their slow photocycle, structural alterations of these proteins can be studied by vibrational spectroscopy in more detail than possible with wild type. Here, we present spectroscopic evidence that the photocycle of the C128T mutant involves three different dark-adapted states that are populated according to the wavelength and duration of the preceding illumination. Our results suggest an important role of multiphoton reactions and the previously described side reaction for dark state regeneration. Structural changes that cause formation and depletion of the assumed ion conducting state P520 are only small and follow larger changes that occur early and late in the photocycle, respectively. They require only minor structural rearrangements of amino acids near the retinal binding pocket and are triggered by all-trans/13-cis retinal isomerization, although additional isomerizations are also involved in the photocycle. We will discuss an extended photocycle model of this mutant on the basis of spectroscopic and electrophysiological data.


Asunto(s)
Adaptación a la Oscuridad/fisiología , Mutación Missense , Retinaldehído/metabolismo , Rodopsina/metabolismo , Sustitución de Aminoácidos , Animales , Sitios de Unión , Células COS , Chlorocebus aethiops , Retinaldehído/genética , Rodopsina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA