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








Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(7)2024 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-38612421

RESUMO

The retinal pigment epithelium (RPE), which ensures the normal functioning of the neural retina, is a pigmented single-cell layer that separates the retina from the Bruch's membrane and the choroid. There are three main types of pigment granules in the RPE cells of the human eye: lipofuscin granules (LG) containing the fluorescent "age pigment" lipofuscin, melanoprotein granules (melanosomes, melanolysosomes) containing the screening pigment melanin and complex melanolipofuscin granules (MLG) containing both types of pigments simultaneously-melanin and lipofuscin. This review examines the functional role of pigment granules in the aging process and in the development of oxidative stress and associated pathologies in RPE cells. The focus is on the process of light-induced oxidative degradation of pigment granules caused by reactive oxygen species. The reasons leading to increased oxidative stress in RPE cells as a result of the oxidative degradation of pigment granules are considered. A mechanism is proposed to explain the phenomenon of age-related decline in melanin content in RPE cells. The essence of the mechanism is that when the lipofuscin part of the melanolipofuscin granule is exposed to light, reactive oxygen species are formed, which destroy the melanin part. As more melanolipofuscin granules are formed with age and the development of degenerative diseases, the melanin in pigmented epithelial cells ultimately disappears.


Assuntos
Melaninas , Epitélio Pigmentado da Retina , Humanos , Lipofuscina , Espécies Reativas de Oxigênio , Retina
2.
Adv Exp Med Biol ; 1415: 15-20, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37440008

RESUMO

Lutein (L), zeaxanthin (Z), and meso-zeaxanthin (MZ) are the three macular pigments (MP) carotenoids that uniquely accumulate in the macula lutea region of the human retina. L and Z are obtained by humans through dietary intake. The third MP, MZ, is rarely present in diet, and its abundance in the human fovea is due to the metabolic conversion of dietary L by the retinal pigment epithelium's RPE65 enzyme. The major functions of MP in ocular health are to filter high-intensity, phototoxic blue light and to act as effective antioxidants for scavenging free radicals. The pyridinium bisretinoid, N-retinylidene-N-retinylethanolamine (A2E), contributes to drusen formation in dry age-related macular degeneration (AMD) and to the autofluorescent flecks in autosomal recessive Stargardt disease (STGD1). Retinal carotenoids attenuate A2E formation and can directly and indirectly alleviate A2E-mediated oxidative damage. In this chapter, we review these more recently recognized interconnections between MP carotenoids and A2E bisretinoids.


Assuntos
Macula Lutea , Degeneração Macular , Pigmento Macular , Humanos , Luteína , Degeneração Macular/genética , Degeneração Macular/metabolismo , Pigmento Macular/metabolismo , Retina/metabolismo , Retinoides/farmacologia
3.
Biophys Rev ; 14(4): 1051-1065, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36124271

RESUMO

Lipofuscin granules (LGs) are accumulated in the retinal pigment epithelium (RPE) cells. The progressive LG accumulation can somehow lead to pathology and accelerate the aging process. The review examines composition, spectral properties and photoactivity of LGs isolated from the human cadaver eyes. By use of atomic force microscopy and near-field microscopy, we have revealed the fluorescent heterogeneity of LGs. We have discovered the generation of reactive oxygen species by LGs, and found that LGs and melanolipofuscin granules are capable of photoinduced oxidation of lipids. It was shown that A2E, as the main fluorophore (bisretinoid) of LGs, is much less active as an oxidation photosensitizer than other fluorophores (bisretinoids) of LGs. Photooxidized products of bisretinoids pose a much greater danger to the cell than non-oxidized one. Our studies of the fluorescent properties of LGs and their fluorophores (bisretinoids) showed for the first time that their spectral characteristics change (shift to the short-wavelength region) in pathology and after exposure to ionizing radiation. By recording the fluorescence spectra and fluorescence decay kinetics of oxidized products of LG fluorophores, it is possible to improve the methods of early diagnosis of degenerative diseases. Lipofuscin ("aging pigment") is not an inert "slag". The photoactivity of LGs can pose a significant danger to the RPE cells. Fluorescence characteristics of LGs are a tool to detect early stages of degeneration in the retina and RPE.

4.
Redox Biol ; 54: 102386, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35809434

RESUMO

To facilitate the movement of retinoids through the visual cycle and to limit nonspecific chemical reaction, multiple mechanisms are utilized to handle these molecules when not contained within the binding pocket of opsin. Vitamin A aldehyde is sequestered by reversible Schiff base formation with phosphatidylethanolamine (PE) and subsequently undergoes NADPH-dependent reduction. Otherwise inefficient handling of retinaldehyde can lead to the formation of fluorescent di-retinal compounds within the outer segments of photoreceptor cells. These bisretinoid fluorophores initiate photooxidative processes having adverse consequences for retina. Various carrier proteins confer water solubility and maintain the 11-cis-retinoid configuration. Mechanisms for sequestration of retinoid include the formation of a reversible Schiff base between retinaldehyde and taurine (A1-taurine, A1T), the most abundant amino acid in photoreceptor cells. Here we have undertaken to examine the effects of taurine depletion using the transport inhibitors guanidinoethyl sulfonate (GES) and ß-alanine. Oral treatment of BALB/cJ mice with ß-alanine reduced ocular A1T and the mice exhibited significantly lower scotopic and photopic a-wave amplitudes. As a secondary effect of retinal degeneration, A1T was not detected and taurine was significantly reduced in mice carrying a P23H opsin mutation. The thinning of ONL that is indicative of reduced photoreceptor cell viability in albino Abca4-/- mice was more pronounced in ß-alanine treated mice. Treatment of agouti and albino Abca4-/- mice with ß-alanine and GES was associated with reduced bisretinoid measured chromatographically. Consistent with a reduction in carbonyl scavenging activity by taurine, methylglyoxal-adducts were also increased in the presence of ß-alanine. Taken together these findings support the postulate that A1T serves as a reservoir of vitamin A aldehyde, with diminished A1T explaining reduced photoreceptor light-sensitivity, accentuated ONL thinning in Abca4-/- mice and attenuated bisretinoid formation.


Assuntos
Retinaldeído , Bases de Schiff , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Camundongos , Opsinas/análise , Opsinas/genética , Opsinas/metabolismo , Células Fotorreceptoras/metabolismo , Retina/metabolismo , Retinaldeído/análise , Retinaldeído/metabolismo , Retinoides/análise , Retinoides/química , Retinoides/metabolismo , Bases de Schiff/análise , Bases de Schiff/metabolismo , Taurina , beta-Alanina/metabolismo
5.
FASEB J ; 36(5): e22309, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35471581

RESUMO

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.


Assuntos
Proteômica , Peixe-Zebra , Animais , Cegueira/metabolismo , Humanos , Fagocitose , Células Fotorreceptoras Retinianas Cones/metabolismo , Retinoides/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo
6.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34782457

RESUMO

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.


Assuntos
Membranas Intracelulares/metabolismo , Lipofuscina/farmacologia , Lisossomos/metabolismo , Necroptose/efeitos dos fármacos , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Envelhecimento , Oxirredutases do Álcool , Animais , Morte Celular , Humanos , Lipofuscina/metabolismo , Degeneração Macular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Retina/metabolismo , Epitélio Pigmentado da Retina/metabolismo
7.
Exp Eye Res ; 209: 108680, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34161819

RESUMO

Accumulation of bisretinoids such as A2E and its isomer iso-A2E is thought to mediate blue light-induced oxidative damage associated with age-related macular degeneration (AMD) and autosomal recessive Stargardt disease (STGD1). We hypothesize that increasing dietary intake of the macular carotenoids lutein and zeaxanthin in individuals at risk of AMD and STGD1 can inhibit the formation of bisretinoids A2E and iso-A2E, which can potentially ameliorate macular degenerative diseases. To study the beneficial effect of macular carotenoids in a retinal degenerative diseases model, we used ATP-binding cassette, sub-family A member 4 (Abca4-/-)/ß,ß-carotene-9',10'-oxygenase 2 (Bco2-/-) double knockout (KO) mice that accumulate elevated levels of A2E and iso-A2E in the retinal pigment epithelium (RPE) and macular carotenoids in the retina. Abca4-/-/Bco2-/- and Abca4-/- mice were fed a lutein-supplemented chow, zeaxanthin-supplemented chow or placebo chow (~2.6 mg of carotenoid/mouse/day) for three months. Visual function and electroretinography (ERG) were measured after one month and three months of carotenoid supplementation. The lutein and zeaxanthin supplemented Abca4-/-/Bco2-/- mice had significantly lower levels of RPE/choroid A2E and iso-A2E compared to control mice fed with placebo chow and improved visual performance. Carotenoid supplementation in Abca4-/- mice minimally raised retinal carotenoid levels and did not show much difference in bisretinoid levels or visual function compared to the control diet group. There was a statistically significant inverse correlation between carotenoid levels in the retina and A2E and iso-A2E levels in the RPE/choroid. Supplementation with retinal carotenoids, especially zeaxanthin, effectively inhibits bisretinoid formation in a mouse model of STGD1 genetically enhanced to accumulate carotenoids in the retina. These results provide further impetus to pursue oral carotenoids as therapeutic interventions for STGD1 and AMD.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Dioxigenases/genética , Regulação da Expressão Gênica , Luteína/farmacocinética , Degeneração Macular/tratamento farmacológico , Epitélio Pigmentado da Retina/efeitos dos fármacos , Zeaxantinas/farmacocinética , Transportadores de Cassetes de Ligação de ATP/biossíntese , Animais , Dioxigenases/biossíntese , Modelos Animais de Doenças , Eletrorretinografia , Degeneração Macular/metabolismo , Degeneração Macular/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Epitélio Pigmentado da Retina/metabolismo , Visão Ocular/efeitos dos fármacos
8.
J Biol Chem ; 295(19): 6767-6780, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32188692

RESUMO

Mutations in retinaldehyde-binding protein 1 (RLBP1), encoding the visual cycle protein cellular retinaldehyde-binding protein (CRALBP), cause an autosomal recessive form of retinal degeneration. By binding to 11-cis-retinoid, CRALBP augments the isomerase activity of retinoid isomerohydrolase RPE65 (RPE65) and facilitates 11-cis-retinol oxidation to 11-cis-retinal. CRALBP also maintains the 11-cis configuration and protects against unwanted retinaldehyde activity. Studying a sibling pair that is compound heterozygous for mutations in RLBP1/CRALBP, here we expand the phenotype of affected individuals, elucidate a previously unreported phenotype in RLBP1/CRALBP carriers, and demonstrate consistencies between the affected individuals and Rlbp1/Cralbp-/- mice. In the RLBP1/CRALBP-affected individuals, nonrecordable rod-specific electroretinogram traces were recovered after prolonged dark adaptation. In ultrawide-field fundus images, we observed radially arranged puncta typical of RLBP1/CRALBP-associated disease. Spectral domain-optical coherence tomography (SD-OCT) revealed hyperreflective aberrations within photoreceptor-associated bands. In short-wavelength fundus autofluorescence (SW-AF) images, speckled hyperautofluorescence and mottling indicated macular involvement. In both the affected individuals and their asymptomatic carrier parents, reduced SW-AF intensities, measured as quantitative fundus autofluorescence (qAF), indicated chronic impairment in 11-cis-retinal availability and provided information on mutation severity. Hypertransmission of the SD-OCT signal into the choroid together with decreased near-infrared autofluorescence (NIR-AF) provided evidence for retinal pigment epithelial cell (RPE) involvement. In Rlbp1/Cralbp-/- mice, reduced 11-cis-retinal levels, qAF and NIR-AF intensities, and photoreceptor loss were consistent with the clinical presentation of the affected siblings. These findings indicate that RLBP1 mutations are associated with progressive disease involving RPE atrophy and photoreceptor cell degeneration. In asymptomatic carriers, qAF disclosed previously undetected visual cycle deficiency.


Assuntos
Proteínas de Transporte/genética , Mutação , Transtornos da Visão/genética , Adolescente , Adulto , Animais , Doenças Assintomáticas , Criança , Feminino , Fundo de Olho , Heterozigoto , Humanos , Masculino , Camundongos , Fenótipo , Retinoides/metabolismo , Transtornos da Visão/metabolismo , Transtornos da Visão/patologia
9.
Adv Exp Med Biol ; 1185: 341-346, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31884635

RESUMO

Bisretinoid fluorophores are the major constituents of the lipofuscin of retinal pigment epithelium (RPE) that accumulates with age and contributes to retina disease. Knowledge of the burden placed on the RPE cell by the accumulation of these phototoxic retinaldehyde-adducts depends on the identification and quantitation of the various bisretinoid species that constitute this family of fluorophores. Here we report a previously unidentified fluorescent bisretinoid by UPLC coupled to photodiode array detection, fluorescence, and electrospray ionization mass spectrometry (UPLC-PDA-FLR-ESI-MS) (Kim HJ, Sparrow JR, J Lipid Res 59:1620-1629, 2018). This novel bisretinoid is 1-octadecyl-2-lyso-sn-glycero A2PE (alkyl ether lysoA2PE). The structural assignment was based on molecular weight (m/z 998), UV-visible absorbance maxima (340, 440 nm), and retention time (73 minutes) and was corroborated by biomimetic synthesis using all-trans-retinal and glycerophosphoethanolamine analogues as starting materials. In mechanistic studies, A2PE was hydrolyzed by PLA2, and plasmalogen lysoA2PE was cleaved under acidic conditions. Unprecedented UPLC detection of the bisretinoid alkyl ether lysoA2PE in human RPE but not in neural retina indicates that the phospholipase A2 activity that generates the latter bisretinoid resides in RPE.


Assuntos
Epitélio Pigmentado da Retina/enzimologia , Retinoides/química , Humanos , Lipofuscina/metabolismo , Fosfolipases A2/metabolismo , Retinaldeído/química
10.
J Lipid Res ; 59(9): 1620-1629, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29986955

RESUMO

Bisretinoids are a family of fluorophores that form in photoreceptor cells' outer segments by nonenzymatic reaction of two vitamin A aldehydes (A2) with phosphatidylethanolamine (PE). Bisretinoid fluorophores are the major constituents of the lipofuscin of retinal pigment epithelium (RPE) that accumulate with age and contribute to some retinal diseases. Here, we report the identification of a previously unknown fluorescent bisretinoid. By ultra-performance LC (UPLC) coupled to photodiode array detection, fluorescence (FLR), and ESI-MS, we determined that this novel bisretinoid is 1-octadecyl-2-lyso-sn-glycero A2PE (alkyl ether lysoA2PE). This structural assignment was based on molecular mass (m/z 998), UV-visible absorbance maxima (340 and 440 nm), and retention time (73 min) and was corroborated by biomimetic synthesis using all-trans-retinal and glycerophosphoethanolamine analogs as starting materials. UPLC profiles of ocular extracts acquired from human donor eyes revealed that alkyl ether lysoA2PE was detectable in RPE, but not neural retina. LysoA2PE FLR spectra exhibited a significant hyperchromic shift in hydrophobic environments. The propensity for lysoA2PE to undergo photooxidation/degradation was less pronounced than A2E. In mechanistic studies, A2PE was hydrolyzed by phospholipase A2 and plasmalogen lysoA2PE was cleaved under acidic conditions. The characterization of these additional members of the bisretinoid family advances our understanding of the mechanisms underlying bisretinoid biogenesis.


Assuntos
Fosfatidiletanolaminas/química , Fosfatidiletanolaminas/metabolismo , Retina/metabolismo , Retinaldeído/química , Humanos , Oxirredução , Processos Fotoquímicos
11.
ChemPhotoChem ; 1(6): 256-259, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29057298

RESUMO

Vitamin A based bisretinoid accumulation is a major focus in the study of macular degeneration. Whether specific endogenous lysosomal proteins can bind A2E, a pronounced bisretinoid in lipofuscin granules in retinal pigment epithelial cells, and interfere with enzymatic or photoinduced oxidation of such, has not been explored. Herein, using fluorescence and electronic absorption spectroscopy and mass spectrometry, we demonstrate that Saposin B, a critical protein in the degradation of sulfatides and "flushing" of lipids, can bind A2E, preventing its H2O2-dependent enzymatic oxidation by horseradish peroxidase and photooxidation by blue light (λ=450-460 nm).

12.
Proc Natl Acad Sci U S A ; 114(15): 3987-3992, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28348233

RESUMO

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.


Assuntos
Complemento C3/metabolismo , Degeneração Macular/congênito , Células Fotorreceptoras de Vertebrados/patologia , Receptores de Complemento/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Autofagia , Dependovirus/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Injeções Intraoculares , Lipofuscina/metabolismo , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Camundongos Endogâmicos BALB C , Camundongos Mutantes , Estresse Oxidativo , Células Fotorreceptoras de Vertebrados/metabolismo , Receptores de Complemento/genética , Receptores de Complemento 3b , Epitélio Pigmentado da Retina/patologia , Retinoides/metabolismo , Doença de Stargardt
13.
World J Exp Med ; 6(4): 63-71, 2016 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-27909686

RESUMO

AIM: To investigate the effect of two ways of lipofuscin production (lipid peroxidation and glycation) on lipofuscin fluorescence characteristics and phototoxicity and to compare them with the properties of natural lipofuscin. METHODS: Model lipofuscins were prepared on the basis of bovine photoreceptor outer segments (POS) with bisretinoid A2E addition. One set of samples was prepared from POS modified by lipid peroxidation, while another set from POS modified by glycation with fructose. Fluorescent properties and kinetics of photoinduced superoxide generation of model lipofuscins and human retinal pigment epithelium (RPE) lipofuscin were compared. The fluorescence spectra of samples were measured at 365 nm excitation wavelength and 380-650 emission wavelength. RESULTS: The fluorescence spectra of model lipofuscins are almost the same as the spectrum of natural lipofuscin. Visible light irradiation of both model lipofuscins and natural lipofuscin isolated from RPE cells leads to decrease of a fluorescence maximum at 550 nm and to appearance of a distinct, new maximum at 445-460 nm. The rate of photogeneration of reactive oxygen forms by both model lipofuscins was almost the same and approximately two times less than that of RPE lipofuscin granules. CONCLUSION: These data suggest that fluorescent characteristics and phototoxicity of lipofuscin granules depend only to an insignificant degree on the oxidative modification of POS proteins and lipids, and generally are defined by the bisretinoid fluorophores contained in them.

14.
Transl Vis Sci Technol ; 5(3): 5, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27226929

RESUMO

PURPOSE: Discovery of candidate spectra for abundant fluorophore families in human retinal pigment epithelium (RPE) by ex vivo hyperspectral imaging. METHODS: Hyperspectral autofluorescence emission images were captured between 420 and 720 nm (10-nm intervals), at two excitation bands (436-460, 480-510 nm), from three locations (fovea, perifovea, near-periphery) in 20 normal RPE/Bruch's membrane (BrM) flatmounts. Mathematical factorization extracted a BrM spectrum (S0) and abundant lipofuscin/melanolipofuscin (LF/ML) spectra of RPE origin (S1, S2, S3) from each tissue. RESULTS: Smooth spectra S1 to S3, with perinuclear localization consistent with LF/ML at all three retinal locations and both excitations in 14 eyes (84 datasets), were included in the analysis. The mean peak emissions of S0, S1, and S2 at λex 436 nm were, respectively, 495 ± 14, 535 ± 17, and 576 ± 20 nm. S3 was generally trimodal, with peaks at either 580, 620, or 650 nm (peak mode, 650 nm). At λex 480 nm, S0, S1, and S2 were red-shifted to 526 ± 9, 553 ± 10, and 588 ± 23 nm, and S3 was again trimodal (peak mode, 620 nm). S1 often split into two spectra, S1A and S1B. S3 strongly colocalized with melanin. There were no significant differences across age, sex, or retinal location. CONCLUSIONS: There appear to be at least three families of abundant RPE fluorophores that are ubiquitous across age, retinal location, and sex in this sample of healthy eyes. Further molecular characterization by imaging mass spectrometry and localization via super-resolution microscopy should elucidate normal and abnormal RPE physiology involving fluorophores. TRANSLATIONAL RELEVANCE: Our results help establish hyperspectral autofluorescence imaging of the human retinal pigment epithelium as a useful tool for investigating retinal health and disease.

15.
Proc Natl Acad Sci U S A ; 113(17): 4564-9, 2016 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-27071115

RESUMO

Although currently available treatment options for age-related macular degeneration (AMD) are limited, particularly for atrophic AMD, the identification of predisposing genetic variations has informed clinical studies addressing therapeutic options such as complement inhibitors and anti-inflammatory agents. To lower risk of early AMD, recommended lifestyle interventions such as the avoidance of smoking and the intake of low glycemic antioxidant-rich diets have largely followed from the identification of nongenetic modifiable factors. On the other hand, the challenge of understanding the complex relationship between aging and cumulative damage leading to AMD has fueled investigations of the visual cycle adducts that accumulate in retinal pigment epithelial (RPE) cells and are a hallmark of aging retina. These studies have revealed properties of these compounds that provide insights into processes that may compromise RPE and could contribute to disease mechanisms in AMD. This work has also led to the design of targeted therapeutics that are currently under investigation.


Assuntos
Degeneração Macular/metabolismo , Retinaldeído/metabolismo , Animais , Humanos , Degeneração Macular/tratamento farmacológico , Terapia de Alvo Molecular , Epitélio Pigmentado da Retina/metabolismo , Fatores de Risco
16.
Adv Exp Med Biol ; 854: 347-53, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26427431

RESUMO

One of the earliest events preceding several forms of retinal degeneration is the formation and accumulation of vitamin A dimers in the retinal pigment epithelium (RPE) and underlying Bruch's membrane (BM). Such degenerations include Stargardt disease, Best disease, forms of retinitis pigmentosa, and age-related macular degeneration (AMD). Since their discovery in the 1990's, dimers of vitamin A, have been postulated as chemical triggers driving retinal senescence and degeneration. There is evidence to suggest that the rate at which vitamin A dimerizes and the eye's response to the dimerization products may dictate the retina's lifespan. Here, we present outstanding questions, finding the answers to which may help to elucidate the role of vitamin A dimerization in retinal degeneration.


Assuntos
Fluorescência , Fundo de Olho , Lipofuscina/química , Retina/patologia , Degeneração Retiniana/patologia , Vitamina A/química , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Lâmina Basilar da Corioide/química , Lâmina Basilar da Corioide/metabolismo , Lâmina Basilar da Corioide/patologia , Dimerização , Humanos , Lipofuscina/metabolismo , Modelos Químicos , Estrutura Molecular , Retina/metabolismo , Degeneração Retiniana/metabolismo , Epitélio Pigmentado da Retina/química , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Vitamina A/metabolismo
17.
Adv Exp Med Biol ; 854: 355-61, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26427432

RESUMO

We discuss how an imperfect visual cycle results in the formation of vitamin A dimers, thought to be involved in the pathogenesis of various retinal diseases, and summarize how slowing vitamin A dimerization has been a therapeutic target of interest to prevent blindness. To elucidate the molecular mechanism of vitamin A dimerization, an alternative form of vitamin A, one that forms dimers more slowly yet maneuvers effortlessly through the visual cycle, was developed. Such a vitamin A, reinforced with deuterium (C20-D3-vitamin A), can be used as a non-disruptive tool to understand the contribution of vitamin A dimers to vision loss. Eventually, C20-D3-vitamin A could become a disease-modifying therapy to slow or stop vision loss associated with dry age-related macular degeneration (AMD), Stargardt disease and retinal diseases marked by such vitamin A dimers. Human clinical trials of C20-D3-vitamin A (ALK-001) are underway.


Assuntos
Cegueira/prevenção & controle , Degeneração Macular/congênito , Degeneração Macular/prevenção & controle , Distrofias Retinianas/prevenção & controle , Vitamina A/uso terapêutico , Cegueira/etiologia , Ensaios Clínicos como Assunto , Deutério/química , Dimerização , Humanos , Degeneração Macular/complicações , Modelos Químicos , Conformação Molecular/efeitos dos fármacos , Estrutura Molecular , Éteres Fenílicos/uso terapêutico , Propanolaminas/uso terapêutico , Distrofias Retinianas/complicações , Doença de Stargardt , Vitamina A/química , Vitaminas/química , Vitaminas/uso terapêutico
18.
Prog Mol Biol Transl Sci ; 134: 449-63, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26310170

RESUMO

Lipofuscin is highly fluorescent material, formed in several tissues but best studied in the eye. The accumulation of lipofuscin in the retinal pigment epithelium (RPE) is a hallmark of aging in the eye and has been implicated in various retinal degenerations, including age-related macular degeneration. The bis-retinoid N-retinyl-N-retinylidene ethanolamine (A2E), formed from retinal, has been identified as a byproduct of the visual cycle, and numerous in vitro studies have found toxicity associated with this compound. The compound is known to accumulate in the RPE with age and was the first identified compound extracted from lipofuscin. Our studies have correlated the distribution of lipofuscin and A2E across the human and mouse RPE. Lipofuscin fluorescence was imaged in the RPE from human donors of various ages and from assorted mouse models. The spatial distribution of A2E was determined using matrix-assisted laser desorption-ionization imaging mass spectrometry on both flat-mounted and transversally sectioned RPE tissue. Our data support the clinical observations in humans of strong RPE fluorescence, increasing with age, in the central area of the RPE. However, there was no correlation between the distribution of A2E and lipofuscin, as the levels of A2E were highest in the far periphery and decreased toward the central region. Interestingly, in all the mouse models, A2E distribution and lipofuscin fluorescence correlate well. These data demonstrate that the accumulation of A2E is not responsible for the increase in lipofuscin fluorescence observed in the central RPE with aging in humans.


Assuntos
Lipofuscina/metabolismo , Retinoides/metabolismo , Animais , Humanos , Imageamento Tridimensional , Modelos Biológicos , Retinoides/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
19.
Dis Model Mech ; 8(2): 131-8, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25504631

RESUMO

The eye uses vitamin A as a cofactor to sense light and, during this process, some vitamin A molecules dimerize, forming vitamin A dimers. A striking chemical signature of retinas undergoing degeneration in major eye diseases such as age-related macular degeneration (AMD) and Stargardt disease is the accumulation of these dimers in the retinal pigment epithelium (RPE) and Bruch's membrane (BM). However, it is not known whether dimers of vitamin A are secondary symptoms or primary insults that drive degeneration. Here, we present a chromatography-free method to prepare gram quantities of the vitamin A dimer, A2E, and show that intravenous administration of A2E to the rabbit results in retinal degeneration. A2E-damaged photoreceptors and RPE cells triggered inflammation, induced remolding of the choroidal vasculature and triggered a decline in the retina's response to light. Data suggest that vitamin A dimers are not bystanders, but can be primary drivers of retinal degeneration. Thus, preventing dimer formation could be a preemptive strategy to address serious forms of blindness.


Assuntos
Dimerização , Degeneração Retiniana/patologia , Degeneração Retiniana/fisiopatologia , Vitamina A/administração & dosagem , Vitamina A/efeitos adversos , Animais , Corioide/patologia , Corioide/fisiopatologia , Eletrorretinografia , Injeções Intravenosas , Células Fotorreceptoras de Vertebrados/patologia , Coelhos , Degeneração Retiniana/induzido quimicamente , Degeneração Retiniana/complicações , Neovascularização Retiniana/complicações , Neovascularização Retiniana/patologia , Neovascularização Retiniana/fisiopatologia , Epitélio Pigmentado da Retina/patologia , Epitélio Pigmentado da Retina/fisiopatologia
20.
J Biol Chem ; 289(13): 9113-20, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24550392

RESUMO

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.


Assuntos
Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Haplótipos , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/metabolismo , Retinoides/metabolismo , Transportadores de Cassetes de Ligação de ATP/deficiência , Animais , Membrana Celular/metabolismo , Complemento C3b/metabolismo , Fator H do Complemento/biossíntese , Predisposição Genética para Doença/genética , Humanos , Degeneração Macular/genética , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Camundongos , Segmento Externo das Células Fotorreceptoras da Retina/metabolismo , Segmento Externo das Células Fotorreceptoras da Retina/patologia , Epitélio Pigmentado da Retina/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA