Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 300(6): 107344, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38705389

RESUMEN

MicroRNAs (miRs) are short, evolutionarily conserved noncoding RNAs that canonically downregulate expression of target genes. The miR family composed of miR-204 and miR-211 is among the most highly expressed miRs in the retinal pigment epithelium (RPE) in both mouse and human and also retains high sequence identity. To assess the role of this miR family in the developed mouse eye, we generated two floxed conditional KO mouse lines crossed to the RPE65-ERT2-Cre driver mouse line to perform an RPE-specific conditional KO of this miR family in adult mice. After Cre-mediated deletion, we observed retinal structural changes by optical coherence tomography; dysfunction and loss of photoreceptors by retinal imaging; and retinal inflammation marked by subretinal infiltration of immune cells by imaging and immunostaining. Single-cell RNA sequencing of diseased RPE and retinas showed potential miR-regulated target genes, as well as changes in noncoding RNAs in the RPE, rod photoreceptors, and Müller glia. This work thus highlights the role of miR-204 and miR-211 in maintaining RPE function and how the loss of miRs in the RPE exerts effects on the neural retina, leading to inflammation and retinal degeneration.


Asunto(s)
Ratones Noqueados , MicroARNs , Degeneración Retiniana , Epitelio Pigmentado de la Retina , Animales , MicroARNs/genética , MicroARNs/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Degeneración Retiniana/metabolismo , Ratones , Eliminación de Gen , Tomografía de Coherencia Óptica
2.
FASEB J ; 38(11): e23720, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38837708

RESUMEN

Recessive Stargardt disease (STGD1) is an inherited juvenile maculopathy caused by mutations in the ABCA4 gene, for which there is no suitable treatment. Loss of functional ABCA4 in the retinal pigment epithelium (RPE) alone, without contribution from photoreceptor cells, was shown to induce STGD1 pathology. Here, we identified cathepsin D (CatD), the primary RPE lysosomal protease, as a key molecular player contributing to endo-lysosomal dysfunction in STGD1 using a newly developed "disease-in-a-dish" RPE model from confirmed STGD1 patients. Induced pluripotent stem cell (iPSC)-derived RPE originating from three STGD1 patients exhibited elevated lysosomal pH, as previously reported in Abca4-/- mice. CatD protein maturation and activity were impaired in RPE from STGD1 patients and Abca4-/- mice. Consequently, STGD1 RPE cells have reduced photoreceptor outer segment degradation and abnormal accumulation of α-synuclein, the natural substrate of CatD. Furthermore, dysfunctional ABCA4 in STGD1 RPE cells results in intracellular accumulation of autofluorescent material and phosphatidylethanolamine (PE). The altered distribution of PE associated with the internal membranes of STGD1 RPE cells presumably compromises LC3-associated phagocytosis, contributing to delayed endo-lysosomal degradation activity. Drug-mediated re-acidification of lysosomes in the RPE of STGD1 restores CatD functional activity and reduces the accumulation of immature CatD protein loads. This preclinical study validates the contribution of CatD deficiencies to STGD1 pathology and provides evidence for an efficacious therapeutic approach targeting RPE cells. Our findings support a cell-autonomous RPE-driven pathology, informing future research aimed at targeting RPE cells to treat ABCA4-mediated retinopathies.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Catepsina D , Lisosomas , Epitelio Pigmentado de la Retina , Enfermedad de Stargardt , Catepsina D/metabolismo , Catepsina D/genética , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Enfermedad de Stargardt/metabolismo , Enfermedad de Stargardt/patología , Enfermedad de Stargardt/genética , Animales , Humanos , Ratones , Lisosomas/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Células Madre Pluripotentes Inducidas/metabolismo , Ratones Noqueados , Degeneración Macular/metabolismo , Degeneración Macular/patología , Degeneración Macular/genética
3.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34782457

RESUMEN

Lipofuscin granules enclose mixtures of cross-linked proteins and lipids in proportions that depend on the tissue analyzed. Retinal lipofuscin is unique in that it contains mostly lipids with very little proteins. However, retinal lipofuscin also presents biological and physicochemical characteristics indistinguishable from conventional granules, including indigestibility, tendency to cause lysosome swelling that results in rupture or defective functions, and ability to trigger NLRP3 inflammation, a symptom of low-level disruption of lysosomes. In addition, like conventional lipofuscins, it appears as an autofluorescent pigment, considered toxic waste, and a biomarker of aging. Ocular lipofuscin accumulates in the retinal pigment epithelium (RPE), whereby it interferes with the support of the neuroretina. RPE cell death is the primary cause of blindness in the most prevalent incurable genetic and age-related human disorders, Stargardt disease and age-related macular degeneration (AMD), respectively. Although retinal lipofuscin is directly linked to the cell death of the RPE in Stargardt, the extent to which it contributes to AMD is a matter of debate. Nonetheless, the number of AMD clinical trials that target lipofuscin formation speaks for the potential relevance for AMD as well. Here, we show that retinal lipofuscin triggers an atypical necroptotic cascade, amenable to pharmacological intervention. This pathway is distinct from canonic necroptosis and is instead dependent on the destabilization of lysosomes. We also provide evidence that necroptosis is activated in aged human retinas with AMD. Overall, this cytotoxicity mechanism may offer therapeutic targets and markers for genetic and age-related diseases associated with lipofuscin buildups.


Asunto(s)
Membranas Intracelulares/metabolismo , Lipofuscina/farmacología , Lisosomas/metabolismo , Necroptosis/efectos de los fármacos , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Envejecimiento , Oxidorreductasas de Alcohol , Animales , Muerte Celular , Humanos , Lipofuscina/metabolismo , Degeneración Macular/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo
4.
J Neurosci ; 42(11): 2180-2189, 2022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-35091503

RESUMEN

The high sensitivity of night vision requires that rod photoreceptors reliably and reproducibly signal the absorption of single photons, a process that depends on tight regulation of intracellular cGMP concentration through the phototransduction cascade. Here in the mouse (Mus musculus), we studied a single-site D167A mutation of the gene for the α subunit of rod photoreceptor phosphodiesterase (PDEA), made with the aim of removing a noncatalytic binding site for cGMP. This mutation unexpectedly eliminated nearly all PDEA expression and reduced expression of the ß subunit (PDEB) to ∼5%-10% of WT. The remaining PDE had nearly normal specific activity; degeneration was slow, with 50%-60% of rods remaining after 6 months. Responses were larger and more sensitive than normal but slower in rise and decay, probably from slower dark turnover of cGMP. Remarkably, responses became much less reproducible than WT, with response variance increasing for amplitude by over 10-fold, and for latency and time-to-peak by >100-fold. We hypothesize that the increase in variance is the result of greater variability in the dark-resting concentration of cGMP, produced by spatial and temporal nonuniformity in spontaneous PDE activity. This variability decreased as stimuli were made brighter, presumably because of greater spatial uniformity of phototransduction and the approach to saturation. We conclude that the constancy of the rod response depends critically on PDE expression to maintain adequate spontaneous PDE activity, so that the concentration of second messenger is relatively uniform throughout the outer segment.SIGNIFICANCE STATEMENT Rod photoreceptors in the vertebrate retina reliably signal the absorption of single photons of light by generating responses that are remarkably reproducible in amplitude and waveform. We show that this reproducibility depends critically on the concentration of the effector enzyme phosphodiesterase (PDE), which metabolizes the second messenger cGMP and generates rod light responses. In rods with the D167A mutation of the α subunit of PDE, only 5%-10% of PDE is expressed. Single-photon responses then become much more variable than in WT rods. We think this variability is caused by spatial and temporal inhomogeneity in the concentration of cGMP in darkness, so that photons absorbed in different parts of the cell produce responses of greatly varying amplitude and waveform.


Asunto(s)
GMP Cíclico , Hidrolasas Diéster Fosfóricas , Animales , GMP Cíclico/metabolismo , Ratones , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Reproducibilidad de los Resultados , Retina/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo
5.
FASEB J ; 36(5): e22309, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35471581

RESUMEN

RAB28 is a farnesylated, ciliary G-protein. Patient variants in RAB28 are causative of autosomal recessive cone-rod dystrophy (CRD), an inherited human blindness. In rodent and zebrafish models, the absence of Rab28 results in diminished dawn, photoreceptor, outer segment phagocytosis (OSP). Here, we demonstrate that Rab28 is also required for dusk peaks of OSP, but not for basal OSP levels. This study further elucidated the molecular mechanisms by which Rab28 controls OSP and inherited blindness. Proteomic profiling identified factors whose expression in the eye or whose expression at dawn and dusk peaks of OSP is dysregulated by loss of Rab28. Notably, transgenic overexpression of Rab28, solely in zebrafish cones, rescues the OSP defect in rab28 KO fish, suggesting rab28 gene replacement in cone photoreceptors is sufficient to regulate Rab28-OSP. Rab28 loss also perturbs function of the visual cycle as retinoid levels of 11-cRAL, 11cRP, and atRP are significantly reduced in larval and adult rab28 KO retinae (p < .05). These data give further understanding on the molecular mechanisms of RAB28-associated CRD, highlighting roles of Rab28 in both peaks of OSP, in vitamin A metabolism and in retinoid recycling.


Asunto(s)
Proteómica , Pez Cebra , Animales , Ceguera/metabolismo , Humanos , Fagocitosis , Células Fotorreceptoras Retinianas Conos/metabolismo , Retinoides/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
6.
FASEB J ; 34(3): 3693-3714, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31989709

RESUMEN

Stargardt disease (STGD1), known as inherited retinal dystrophy, is caused by ABCA4 mutations. The pigmented Abca4-/- mouse strain only reflects the early stage of STGD1 since it is devoid of retinal degeneration. This blue light-illuminated pigmented Abca4-/- mouse model presented retinal pigment epithelium (RPE) and photoreceptor degeneration which was similar to the advanced STGD1 phenotype. In contrast, wild-type mice showed no RPE degeneration after blue light illumination. In Abca4-/- mice, the acute blue light diminished the mean autofluorescence (AF) intensity in both fundus short-wavelength autofluorescence (SW-AF) and near-infrared autofluorescence (NIR-AF) modalities correlating with reduced levels of bisretinoid-fluorophores. Blue light-induced RPE cellular damage preceded the photoreceptors loss. In late-stage STGD1-like patient and blue light-illuminated Abca4-/- mice, lipofuscin and melanolipofuscin granules were found to contribute to NIR-AF, indicated by the colocalization of lipofuscin-AF and NIR-AF under the fluorescence microscope. In this mouse model, the correlation between in vivo and ex vivo assessments revealed histological characteristics of fundus AF abnormalities. The flecks which are hyper AF in both SW-AF and NIR-AF corresponded to the subretinal macrophages fully packed with pigment granules (lipofuscin, melanin, and melanolipofuscin). This mouse model, which has the phenotype of advanced STGD1, is important to understand the histopathology of Stargardt disease.


Asunto(s)
Retina/diagnóstico por imagen , Enfermedad de Stargardt/diagnóstico por imagen , Enfermedad de Stargardt/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Electrorretinografía , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas In Vitro , Lipofuscina/metabolismo , Masculino , Melaninas/metabolismo , Ratones , Microscopía Fluorescente , Retina/metabolismo , Tomografía de Coherencia Óptica
7.
Proc Natl Acad Sci U S A ; 115(47): E11120-E11127, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30397118

RESUMEN

Recessive Stargardt disease (STGD1) is an inherited blinding disorder caused by mutations in the Abca4 gene. ABCA4 is a flippase in photoreceptor outer segments (OS) that translocates retinaldehyde conjugated to phosphatidylethanolamine across OS disc membranes. Loss of ABCA4 in Abca4-/- mice and STGD1 patients causes buildup of lipofuscin in the retinal pigment epithelium (RPE) and degeneration of photoreceptors, leading to blindness. No effective treatment currently exists for STGD1. Here we show by several approaches that ABCA4 is additionally expressed in RPE cells. (i) By in situ hybridization analysis and by RNA-sequencing analysis, we show the Abca4 mRNA is expressed in human and mouse RPE cells. (ii) By quantitative immunoblotting, we show that the level of ABCA4 protein in homogenates of wild-type mouse RPE is about 1% of the level in neural retina homogenates. (iii) ABCA4 immunofluorescence is present in RPE cells of wild-type and Mertk-/- but not Abca4-/- mouse retina sections, where it colocalizes with endolysosomal proteins. To elucidate the role of ABCA4 in RPE cells, we generated a line of genetically modified mice that express ABCA4 in RPE cells but not in photoreceptors. Mice from this line on the Abca4-/- background showed partial rescue of photoreceptor degeneration and decreased lipofuscin accumulation compared with nontransgenic Abca4-/- mice. We propose that ABCA4 functions to recycle retinaldehyde released during proteolysis of rhodopsin in RPE endolysosomes following daily phagocytosis of distal photoreceptor OS. ABCA4 deficiency in the RPE may play a role in the pathogenesis of STGD1.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Degeneración Macular/congénito , Células Fotorreceptoras/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Retinaldehído/metabolismo , Transportadoras de Casetes de Unión a ATP/biosíntesis , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Lipofuscina/metabolismo , Lisosomas/metabolismo , Degeneración Macular/genética , Degeneración Macular/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Fagocitosis/inmunología , Retina/patología , Degeneración Retiniana/patología , Rodopsina/metabolismo , Enfermedad de Stargardt , Tirosina Quinasa c-Mer/genética
8.
Proc Natl Acad Sci U S A ; 114(15): 3987-3992, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28348233

RESUMEN

Recessive Stargardt macular degeneration (STGD1) is caused by mutations in the gene for the ABCA4 transporter in photoreceptor outer segments. STGD1 patients and Abca4-/- (STGD1) mice exhibit buildup of bisretinoid-containing lipofuscin pigments in the retinal pigment epithelium (RPE), increased oxidative stress, augmented complement activation and slow degeneration of photoreceptors. A reduction in complement negative regulatory proteins (CRPs), possibly owing to bisretinoid accumulation, may be responsible for the increased complement activation seen on the RPE of STGD1 mice. CRPs prevent attack on host cells by the complement system, and complement receptor 1-like protein y (CRRY) is an important CRP in mice. Here we attempted to rescue the phenotype in STGD1 mice by increasing expression of CRRY in the RPE using a gene therapy approach. We injected recombinant adeno-associated virus containing the CRRY coding sequence (AAV-CRRY) into the subretinal space of 4-wk-old Abca4-/- mice. This resulted in sustained, several-fold increased expression of CRRY in the RPE, which significantly reduced the complement factors C3/C3b in the RPE. Unexpectedly, AAV-CRRY-treated STGD1 mice also showed reduced accumulation of bisretinoids compared with sham-injected STGD1 control mice. Furthermore, we observed slower photoreceptor degeneration and increased visual chromophore in 1-y-old AAV-CRRY-treated STGD1 mice. Rescue of the STGD1 phenotype by AAV-CRRY gene therapy suggests that complement attack on the RPE is an important etiologic factor in STGD1. Modulation of the complement system by locally increasing CRP expression using targeted gene therapy represents a potential treatment strategy for STGD1 and other retinopathies associated with complement dysregulation.


Asunto(s)
Complemento C3/metabolismo , Degeneración Macular/congénito , Células Fotorreceptoras de Vertebrados/patología , Receptores de Complemento/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Animales , Autofagia , Dependovirus/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Inyecciones Intraoculares , Lipofuscina/metabolismo , Degeneración Macular/metabolismo , Degeneración Macular/patología , Ratones Endogámicos BALB C , Ratones Mutantes , Estrés Oxidativo , Células Fotorreceptoras de Vertebrados/metabolismo , Receptores de Complemento/genética , Receptores de Complemento 3b , Epitelio Pigmentado de la Retina/patología , Retinoides/metabolismo , Enfermedad de Stargardt
9.
J Biol Chem ; 292(52): 21407-21416, 2017 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-29109151

RESUMEN

Peropsin is a non-visual opsin in both vertebrate and invertebrate species. In mammals, peropsin is present in the apical microvilli of retinal pigment epithelial (RPE) cells. These structures interdigitate with the outer segments of rod and cone photoreceptor cells. RPE cells play critical roles in the maintenance of photoreceptors, including the recycling of visual chromophore for the opsin visual pigments. Here, we sought to identify the function of peropsin in the mouse eye. To this end, we generated mice with a null mutation in the peropsin gene (Rrh). These mice exhibited normal retinal histology, normal morphology of outer segments and RPE cells, and no evidence of photoreceptor degeneration. Biochemically, Rrh-/- mice had ∼2-fold higher vitamin A (all-trans-retinol (all-trans-ROL)) in the neural retina following a photobleach and 5-fold lower retinyl esters in the RPE. This phenotype was similar to those reported in mice that lack interphotoreceptor retinoid-binding protein (IRBP) or cellular retinol-binding protein, suggesting that peropsin plays a role in the movement of all-trans-ROL from photoreceptors to the RPE. We compared the phenotypes in mice lacking both peropsin and IRBP with those of mice lacking peropsin or IRBP alone and found that the retinoid phenotype was similarly severe in each of these knock-out mice. We conclude that peropsin controls all-trans-ROL movement from the retina to the RPE or may regulate all-trans-ROL storage within the RPE. We propose that peropsin affects light-dependent regulation of all-trans-ROL uptake from photoreceptors into RPE cells through an as yet undefined mechanism.


Asunto(s)
Rodopsina/metabolismo , Vitamina A/fisiología , Animales , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Ratones , Ratones Noqueados , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Pigmentos Retinianos/metabolismo , Retinaldehído/metabolismo , Retinoides/metabolismo , Proteínas de Unión al Retinol/genética , Proteínas de Unión al Retinol/metabolismo , Proteínas Celulares de Unión al Retinol/metabolismo , Rodopsina/genética , Rodopsina/fisiología , Opsinas de Bastones/metabolismo , Vitamina A/metabolismo
10.
Proc Natl Acad Sci U S A ; 111(14): E1402-8, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24706818

RESUMEN

Accumulation of lipofuscin bisretinoids (LBs) in the retinal pigment epithelium (RPE) is the alleged cause of retinal degeneration in genetic blinding diseases (e.g., Stargardt) and a possible etiological agent for age-related macular degeneration. Currently, there are no approved treatments for these diseases; hence, agents that efficiently remove LBs from RPE would be valuable therapeutic candidates. Here, we show that beta cyclodextrins (ß-CDs) bind LBs and protect them against oxidation. Computer modeling and biochemical data are consistent with the encapsulation of the retinoid arms of LBs within the hydrophobic cavity of ß-CD. Importantly, ß-CD treatment reduced by 73% and 48% the LB content of RPE cell cultures and of eyecups obtained from Abca4-Rdh8 double knock-out (DKO) mice, respectively. Furthermore, intravitreal administration of ß-CDs reduced significantly the content of bisretinoids in the RPE of DKO animals. Thus, our results demonstrate the effectiveness of ß-CDs to complex and remove LB deposits from RPE cells and provide crucial data to develop novel prophylactic approaches for retinal disorders elicited by LBs.


Asunto(s)
Lipofuscina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Retinoides/metabolismo , beta-Ciclodextrinas/metabolismo , Animales , Sitios de Unión , Cromatografía Líquida de Alta Presión , Simulación por Computador , Fluorescencia , Técnicas In Vitro , Lipofuscina/aislamiento & purificación , Ratones , Ratones Noqueados , Oxidación-Reducción , Retinoides/aislamiento & purificación
11.
J Biol Chem ; 289(13): 9113-20, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24550392

RESUMEN

Age-related macular degeneration (AMD) is a common central blinding disease of the elderly. Homozygosity for a sequence variant causing Y402H and I62V substitutions in the gene for complement factor H (CFH) is strongly associated with risk of AMD. CFH, secreted by many cell types, including those of the retinal pigment epithelium (RPE), is a regulatory protein that inhibits complement activation. Recessive Stargardt maculopathy is another central blinding disease caused by mutations in the gene for ABCA4, a transporter in photoreceptor outer segments (OS) that clears retinaldehyde and prevents formation of toxic bisretinoids. Photoreceptors daily shed their distal OS, which are phagocytosed by the RPE cells. Here, we investigated the relationship between the CFH haplotype of human RPE (hRPE) cells, exposure to OS containing bisretinoids, and complement activation. We show that hRPE cells of the AMD-predisposing CFH haplotype (HH402/VV62) are attacked by complement following exposure to bisretinoid-containing Abca4(-/-) OS. This activation was dependent on factor B, indicating involvement of the alternative pathway. In contrast, hRPE cells of the AMD-protective CFH haplotype (YY402/II62) showed no complement activation following exposure to either Abca4(-/-) or wild-type OS. The AMD-protective YY402/II62 hRPE cells were more resistant to the membrane attack complex, whereas HH402/VV62 hRPE cells showed significant membrane attack complex deposition following ingestion of Abca4(-/-) OS. These results suggest that bisretinoid accumulation in hRPE cells stimulates activation and dysregulation of complement. Cells with an intact complement negative regulatory system are protected from complement attack, whereas cells with reduced CFH synthesis because of the Y402H and I62V substitutions are vulnerable to disease.


Asunto(s)
Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Haplotipos , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/metabolismo , Retinoides/metabolismo , Transportadoras de Casetes de Unión a ATP/deficiencia , Animales , Membrana Celular/metabolismo , Complemento C3b/metabolismo , Factor H de Complemento/biosíntesis , Predisposición Genética a la Enfermedad/genética , Humanos , Degeneración Macular/genética , Degeneración Macular/metabolismo , Degeneración Macular/patología , Ratones , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Segmento Externo de las Células Fotorreceptoras Retinianas/patología , Epitelio Pigmentado de la Retina/patología
12.
Mol Vis ; 21: 110-23, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25684976

RESUMEN

PURPOSE: To determine the localization of complement factor H (Cfh) mRNA and its protein in the mouse outer retina. METHODS: Quantitative real-time PCR (qPCR) was used to determine the expression of Cfh and Cfh-related (Cfhr) transcripts in the RPE/choroid. In situ hybridization (ISH) was performed using the novel RNAscope 2.0 FFPE assay to localize the expression of Cfh mRNA in the mouse outer retina. Immunohistochemistry (IHC) was used to localize Cfh protein expression, and western blots were used to characterize CFH antibodies used for IHC. RESULTS: Cfh and Cfhr2 transcripts were detected in the mouse RPE/choroid using qPCR, while Cfhr1, Cfhr3, and Cfhrc (Gm4788) were not detected. ISH showed abundant Cfh mRNA in the RPE of all mouse strains (C57BL/6, BALB/c, 129/Sv) tested, with the exception of the Cfh(-/-) eye. Surprisingly, the Cfh protein was detected by immunohistochemistry in photoreceptors rather than in RPE cells. The specificity of the CFH antibodies was tested by western blotting. Our CFH antibodies recognized purified mouse Cfh protein, serum Cfh protein in wild-type C57BL/6, BALB/c, and 129/Sv, and showed an absence of the Cfh protein in the serum of Cfh(-/-) mice. Greatly reduced Cfh protein immunohistological signals in the Cfh(-/-) eyes also supported the specificity of the Cfh protein distribution results. CONCLUSIONS: Only Cfh and Cfhr2 genes are expressed in the mouse outer retina. Only Cfh mRNA was detected in the RPE, but no protein. We hypothesize that the steady-state concentration of Cfh protein is low in the cells due to secretion, and therefore is below the detection level for IHC.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/genética , Factor H de Complemento/genética , Células Epiteliales/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , ARN Mensajero/genética , Epitelio Pigmentado de la Retina/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Inactivadoras del Complemento C3b/metabolismo , Factor H de Complemento/metabolismo , Células Epiteliales/citología , Femenino , Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Especificidad de Órganos , Células Fotorreceptoras de Vertebrados/citología , ARN Mensajero/metabolismo , Epitelio Pigmentado de la Retina/citología , Alineación de Secuencia , Homología de Secuencia de Aminoácido
13.
PLoS One ; 19(5): e0300584, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38709779

RESUMEN

Though rod and cone photoreceptors use similar phototransduction mechanisms, previous model calculations have indicated that the most important differences in their light responses are likely to be differences in amplification of the G-protein cascade, different decay rates of phosphodiesterase (PDE) and pigment phosphorylation, and different rates of turnover of cGMP in darkness. To test this hypothesis, we constructed TrUx;GapOx rods by crossing mice with decreased transduction gain from decreased transducin expression, with mice displaying an increased rate of PDE decay from increased expression of GTPase-activating proteins (GAPs). These two manipulations brought the sensitivity of TrUx;GapOx rods to within a factor of 2 of WT cone sensitivity, after correcting for outer-segment dimensions. These alterations did not, however, change photoreceptor adaptation: rods continued to show increment saturation though at a higher background intensity. These experiments confirm model calculations that rod responses can mimic some (though not all) of the features of cone responses after only a few changes in the properties of transduction proteins.


Asunto(s)
Células Fotorreceptoras Retinianas Conos , Células Fotorreceptoras Retinianas Bastones , Transducina , Animales , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Ratones , Transducina/metabolismo , Transducina/genética , Retina/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Hidrolasas Diéster Fosfóricas/genética
14.
Nat Commun ; 15(1): 5970, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39043666

RESUMEN

Vacuolar protein sorting 35 (VPS35), the core component of the retromer complex which regulates endosomal trafficking, is genetically linked with Parkinson's disease (PD). Impaired vision is a common non-motor manifestation of PD. Here, we show mouse retinas with VPS35-deficient rods exhibit synapse loss and visual deficit, followed by progressive degeneration concomitant with the emergence of Lewy body-like inclusions and phospho-α-synuclein (P-αSyn) aggregation. Ultrastructural analyses reveal VPS35-deficient rods accumulate aggregates in late endosomes, deposited as lipofuscins bound to P-αSyn. Mechanistically, we uncover a protein network of VPS35 and its interaction with HSC70. VPS35 deficiency promotes sequestration of HSC70 and P-αSyn aggregation in late endosomes. Microglia which engulf lipofuscins and P-αSyn aggregates are activated, displaying autofluorescence, observed as bright dots in fundus imaging of live animals, coinciding with pathology onset and progression. The Rod∆Vps35 mouse line is a valuable tool for further mechanistic investigation of αSyn lesions and retinal degenerative diseases.


Asunto(s)
Degeneración Retiniana , Proteínas de Transporte Vesicular , alfa-Sinucleína , Animales , alfa-Sinucleína/metabolismo , alfa-Sinucleína/genética , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/genética , Ratones , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Endosomas/metabolismo , Microglía/metabolismo , Microglía/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Retina/metabolismo , Retina/patología , Ratones Noqueados , Modelos Animales de Enfermedad , Humanos , Sinapsis/metabolismo , Sinapsis/patología , Masculino
15.
Nat Commun ; 15(1): 1244, 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38336975

RESUMEN

A major limitation to developing chimeric antigen receptor (CAR)-T cell therapies for solid tumors is identifying surface proteins highly expressed in tumors but not in normal tissues. Here, we identify Tyrosinase Related Protein 1 (TYRP1) as a CAR-T cell therapy target to treat patients with cutaneous and rare melanoma subtypes unresponsive to immune checkpoint blockade. TYRP1 is primarily located intracellularly in the melanosomes, with a small fraction being trafficked to the cell surface via vesicular transport. We develop a highly sensitive CAR-T cell therapy that detects surface TYRP1 in tumor cells with high TYRP1 overexpression and presents antitumor activity in vitro and in vivo in murine and patient-derived cutaneous, acral and uveal melanoma models. Furthermore, no systemic or off-tumor severe toxicities are observed in an immunocompetent murine model. The efficacy and safety profile of the TYRP1 CAR-T cell therapy supports the ongoing preparation of a phase I clinical trial.


Asunto(s)
Melanoma , Receptores Quiméricos de Antígenos , Neoplasias de la Úvea , Humanos , Ratones , Animales , Melanoma/terapia , Melanoma/tratamiento farmacológico , Inmunoterapia Adoptiva , Neoplasias de la Úvea/terapia , Neoplasias de la Úvea/tratamiento farmacológico , Tratamiento Basado en Trasplante de Células y Tejidos , Glicoproteínas de Membrana , Oxidorreductasas
16.
Nutrients ; 16(12)2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38931295

RESUMEN

The use of natural products as alternatives to traditional pharmacological treatments in orthodontics is gaining interest due to their anti-inflammatory, antibacterial, and antioxidant properties. This systematic review synthesizes evidence from clinical trials to evaluate the efficacy of natural products in reducing inflammation and bacterial presence in orthodontic and orthognathic treatment settings. The database search was conducted across PubMed, Scopus, and Embase up to January 2024. The review focused on randomized controlled trials only. The selected studies centered on the anti-inflammatory, antibacterial, and antioxidant effects of natural products, adhering to the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines for data extraction. Nine studies, totaling 358 participants, were included. Significant findings demonstrated a reduction in gingival inflammation by over 40% with the use of Aloe vera compared to chlorhexidine. Another study noted a decrease in bleeding on probing by 13.6 points in the treatment group over placebo. Additionally, honey showed a rapid modulation of plaque pH and significantly reduced bacterial counts of Streptococcus mutans. Furthermore, the use of resveratrol emulgel was linked to substantial improvements in gingival health, with a reduction in the gingival index and probing pocket depth. The results indicate that natural products can significantly enhance orthodontic treatment outcomes by reducing inflammation and bacterial levels. These products offer effective alternatives to traditional treatments and show potential for integration into routine orthodontic care protocols. Further research is encouraged to standardize application methods and dosages to maximize clinical benefits and patient satisfaction.


Asunto(s)
Antioxidantes , Productos Biológicos , Deformidades Dentofaciales , Humanos , Aloe , Antibacterianos/uso terapéutico , Antiinflamatorios , Productos Biológicos/uso terapéutico , Clorhexidina , Deformidades Dentofaciales/cirugía , Deformidades Dentofaciales/tratamiento farmacológico , Gingivitis/tratamiento farmacológico , Miel , Ortodoncia/métodos , Preparaciones de Plantas , Ensayos Clínicos Controlados Aleatorios como Asunto , Resveratrol/farmacología , Streptococcus mutans/efectos de los fármacos , Resultado del Tratamiento
17.
Hum Mol Genet ; 20(13): 2560-70, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21493626

RESUMEN

Mutations in the MYO7A gene cause a deaf-blindness disorder, known as Usher syndrome 1B.  In the retina, the majority of MYO7A is in the retinal pigmented epithelium (RPE), where many of the reactions of the visual retinoid cycle take place.  We have observed that the retinas of Myo7a-mutant mice are resistant to acute light damage. In exploring the basis of this resistance, we found that Myo7a-mutant mice have lower levels of RPE65, the RPE isomerase that has a key role in the retinoid cycle.  We show for the first time that RPE65 normally undergoes a light-dependent translocation to become more concentrated in the central region of the RPE cells.  This translocation requires MYO7A, so that, in Myo7a-mutant mice, RPE65 is partly mislocalized in the light.  RPE65 is degraded more quickly in Myo7a-mutant mice, perhaps due to its mislocalization, providing a plausible explanation for its lower levels.  Following a 50-60% photobleach, Myo7a-mutant retinas exhibited increased all-trans-retinyl ester levels during the initial stages of dark recovery, consistent with a deficiency in RPE65 activity.  Lastly, MYO7A and RPE65 were co-immunoprecipitated from RPE cell lysate by antibodies against either of the proteins, and the two proteins were partly colocalized, suggesting a direct or indirect interaction.  Together, the results support a role for MYO7A in the translocation of RPE65, illustrating the involvement of a molecular motor in the spatiotemporal organization of the retinoid cycle in vision.


Asunto(s)
Proteínas del Ojo/metabolismo , Miosinas/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Humanos , Espacio Intracelular/metabolismo , Luz/efectos adversos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miosina VIIa , Miosinas/genética , Unión Proteica/fisiología , Transporte de Proteínas/genética , Transporte de Proteínas/efectos de la radiación , Retina/metabolismo , Retina/patología , Retina/efectos de la radiación , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Síndromes de Usher/genética , Síndromes de Usher/metabolismo , Síndromes de Usher/patología
18.
Proc Natl Acad Sci U S A ; 107(19): 8599-604, 2010 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-20445106

RESUMEN

Structural features of neurons create challenges for effective production and distribution of essential metabolic energy. We investigated how metabolic energy is distributed between cellular compartments in photoreceptors. In avascular retinas, aerobic production of energy occurs only in mitochondria that are located centrally within the photoreceptor. Our findings indicate that metabolic energy flows from these central mitochondria as phosphocreatine toward the photoreceptor's synaptic terminal in darkness. In light, it flows in the opposite direction as ATP toward the outer segment. Consistent with this model, inhibition of creatine kinase in avascular retinas blocks synaptic transmission without influencing outer segment activity. Our findings also reveal how vascularization of neuronal tissue can influence the strategies neurons use for energy management. In vascularized retinas, mitochondria in the synaptic terminals of photoreceptors make neurotransmission less dependent on creatine kinase. Thus, vasculature of the tissue and the intracellular distribution of mitochondria can play key roles in setting the strategy for energy distribution in neurons.


Asunto(s)
Oscuridad , Metabolismo Energético/fisiología , Retina/fisiología , Animales , Creatina Quinasa/antagonistas & inhibidores , Creatina Quinasa/metabolismo , Dinitrofluorobenceno/farmacología , Electrorretinografía , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/efectos de la radiación , Glutamatos/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/efectos de la radiación , Modelos Biológicos , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/enzimología , Terminales Presinápticos/efectos de la radiación , Inhibidores de Proteínas Quinasas/farmacología , Retina/efectos de los fármacos , Retina/enzimología , Retina/efectos de la radiación , Células Fotorreceptoras Retinianas Conos/citología , Células Fotorreceptoras Retinianas Conos/efectos de los fármacos , Células Fotorreceptoras Retinianas Conos/enzimología , Células Fotorreceptoras Retinianas Conos/efectos de la radiación , Segmento Externo de las Células Fotorreceptoras Retinianas/efectos de los fármacos , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Segmento Externo de las Células Fotorreceptoras Retinianas/efectos de la radiación , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/enzimología , Vasos Retinianos/efectos de la radiación , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/efectos de la radiación , Urodelos/fisiología
19.
J Clin Med ; 12(22)2023 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-38002773

RESUMEN

With an increasing understanding of the differences between men and women's psychological experiences, this study aimed to probe the sex-based differences in anxiety, depressive symptoms, and coping strategies among orthognathic patients. The study hypothesis was that female patients would show higher levels of anxiety and depressive symptoms than males, and that coping mechanisms would differ between male and female sexes. A cross-sectional design was adopted, examining orthognathic patients from the Department of Oral and Maxillo-Facial Surgery at the Emergency Clinical Municipal Hospital in Timisoara, Romania, from 2020 to 2023. Eligible participants (18+ years with no prior orthognathic treatment) completed a comprehensive online questionnaire 6 weeks before scheduled surgery. This was composed of validated self-report instruments comprising the SF-36, GAD-7, and the PHQ-9, and the COPE-60, along with additional sociodemographic data. Of the 127 orthognathic patients analyzed (68 men and 59 women, aged 18 to 65 years, mean age 32), men rated their physical health status slightly better on the SF-36 scale. However, the most notable difference was in mental health, with females scoring higher on both the PHQ-9 (indicative of depression) and the GAD-7 (indicative of anxiety) scales. Specifically, female participants exhibited average PHQ-9 scores 1.8 points higher and GAD-7 scores 1.5 points higher than their male counterparts. Coping mechanisms also varied: 42% of male patients primarily employed "Disengagement" strategies, while 58% of females predominantly used "Engagement" and "Emotion Focused" strategies. Emotion-focused coping was associated with a 1.6-fold increased risk of depressive symptoms. Sex differences play a crucial role in the psychological experiences of orthognathic patients, evident in anxiety and depression levels, perceived health status, and coping strategies. This underlines the importance of sex-tailored psychological support in the preoperative phase for orthognathic surgery patients.

20.
J Biol Chem ; 286(21): 18593-601, 2011 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-21464132

RESUMEN

Accumulation of vitamin A-derived lipofuscin fluorophores in the retinal pigment epithelium (RPE) is a pathologic feature of recessive Stargardt macular dystrophy, a blinding disease caused by dysfunction or loss of the ABCA4 transporter in rods and cones. Age-related macular degeneration, a prevalent blinding disease of the elderly, is strongly associated with mutations in the genes for complement regulatory proteins (CRP), causing chronic inflammation of the RPE. Here we explore the possible relationship between lipofuscin accumulation and complement activation in vivo. Using the abca4(-/-) mouse model for recessive Stargardt, we investigated the role of lipofuscin fluorophores (A2E-lipofuscin) on oxidative stress and complement activation. We observed higher expression of oxidative-stress genes and elevated products of lipid peroxidation in eyes from abca4(-/-) versus wild-type mice. We also observed higher levels of complement-activation products in abca4(-/-) RPE cells. Unexpectedly, expression of multiple CRPs, which protect cells from attack by the complement system, were lower in abca4(-/-) versus wild-type RPE. To test whether acute exposure of healthy RPE cells to A2E-lipofuscin affects oxidative stress and expression of CRPs, we fed cultured fetal-derived human RPE cells with rod outer segments from wild-type or abca4(-/-) retinas. In contrast to RPE cells in abca4(-/-) mice, human RPE cells exposed to abca4(-/-) rod outer segments adaptively increased expression of both oxidative-stress and CRP genes. These results suggest that A2E accumulation causes oxidative stress, complement activation, and down-regulation of protective CRP in the Stargardt mouse model. Thus, Stargardt disease and age-related macular degeneration may both be caused by chronic inflammation of the RPE.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Activación de Complemento , Proteínas del Sistema Complemento/metabolismo , Degeneración Macular/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Proteínas del Sistema Complemento/genética , Humanos , Lipofuscina/genética , Lipofuscina/metabolismo , Degeneración Macular/genética , Degeneración Macular/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Estrés Oxidativo/genética , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Epitelio Pigmentado de la Retina/patología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA