Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Commun Biol ; 4(1): 161, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33547385

ABSTRACT

Mutations in CLN3 lead to photoreceptor cell loss in CLN3 disease, a lysosomal storage disorder characterized by childhood-onset vision loss, neurological impairment, and premature death. However, how CLN3 mutations cause photoreceptor cell death is not known. Here, we show that CLN3 is required for phagocytosis of photoreceptor outer segment (POS) by retinal pigment epithelium (RPE) cells, a cellular process essential for photoreceptor survival. Specifically, a proportion of CLN3 in human, mouse, and iPSC-RPE cells localized to RPE microvilli, the site of POS phagocytosis. Furthermore, patient-derived CLN3 disease iPSC-RPE cells showed decreased RPE microvilli density and reduced POS binding and ingestion. Notably, POS phagocytosis defect in CLN3 disease iPSC-RPE cells could be rescued by wild-type CLN3 gene supplementation. Altogether, these results illustrate a novel role of CLN3 in regulating POS phagocytosis and suggest a contribution of primary RPE dysfunction for photoreceptor cell loss in CLN3 disease that can be targeted by gene therapy.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Membrane Glycoproteins/metabolism , Molecular Chaperones/metabolism , Neuronal Ceroid-Lipofuscinoses/metabolism , Phagocytosis , Retinal Photoreceptor Cell Outer Segment/metabolism , Retinal Pigment Epithelium/metabolism , Cell Line , Genetic Therapy , Humans , Induced Pluripotent Stem Cells/pathology , Membrane Glycoproteins/genetics , Microvilli/metabolism , Microvilli/pathology , Molecular Chaperones/genetics , Mutation , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/pathology , Neuronal Ceroid-Lipofuscinoses/therapy , Retinal Photoreceptor Cell Outer Segment/pathology , Retinal Pigment Epithelium/pathology , Signal Transduction
2.
JAMA Ophthalmol ; 137(6): 603-609, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30896765

ABSTRACT

Importance: Targeting the early pathogenic steps in Stargardt disease type 1 (STGD1) is critical to advance our understanding of this condition and to develop potential therapies. Lipofuscin precursors may accumulate within photoreceptors, leading to photoreceptor damage and preceding retinal pigment epithelial (RPE) cell death. Fluorescence adaptive optics scanning light ophthalmoscopy can provide autofluorescence (AF) images in vivo with microscopic resolution to elucidate the cellular origin of AF abnormalities in STGD1. Objective: To study the spatial distribution of photoreceptor, RPE, and AF abnormalities in patients with STGD1 at a cellular level. Design, Setting, and Participants: Cross-sectional study using fluorescence adaptive optics scanning light ophthalmoscopy to compare the cones, rods, and RPE cells between 3 patients with STGD1 and 1 control individual. Imaging sessions were conducted at the University of Rochester. Further image analyses were performed at Beijing Tongren Eye Center and the University of Pittsburgh. Data were collected from August 2015 to February 2016, and analysis began in March 2016. Main Outcomes and Measures: Structural appearance of cones, rods, and AF structures at different retinal locations. Results: Two women and 1 man with macular atrophy phenotype of STGD1 and visual acuity loss ranging from 20/30 to 20/150 and 1 woman without STGD1 with 20/20 visual acuity were analyzed. Cone and rod spacing was increased in all 3 patients at all locations where photoreceptors were detectable; most cones had a dark appearance. Autofluorescence was low contrast but contained structures consistent with RPE cells in the periphery. In the transition zone peripheral to the foveal atrophic lesion, the structural pattern of AF was more consistent with photoreceptors than RPE cells. The microscopic AF was disrupted within areas of clinically detectable atrophy. Conclusions and Relevance: Adaptive optics high-resolution images of cones, rods, and RPE cells at the leading disease front of STGD1 macular atrophy show an AF pattern that appears to colocalize with photoreceptors or may result from a combination of AF signals from both RPE cells and photoreceptors. This in vivo observation is consistent with histologic reports of fluorescence arising from photoreceptors in STGD1. The detection of bisretinoid accumulation in the photoreceptors may represent an early pathologic step in STGD1 and can provide an in vivo imaging tool to act as a biomarker of disease progression.


Subject(s)
Photoreceptor Cells, Vertebrate/pathology , Retinal Pigment Epithelium/pathology , Stargardt Disease/diagnosis , Adult , Cross-Sectional Studies , Female , Fluorescein Angiography , Fundus Oculi , Humans , Male , Ophthalmoscopy , Optical Imaging , Optics and Photonics , Young Adult
3.
Invest Ophthalmol Vis Sci ; 59(15): 5705-5716, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30513531

ABSTRACT

Purpose: To characterize in vivo morphometry and multispectral autofluorescence of the retinal pigment epithelial (RPE) cell mosaic and its relationship to cone cell topography across the macula. Methods: RPE cell morphometrics were computed in regularly spaced regions of interest (ROIs) from contiguous short-wavelength autofluorescence (SWAF) and photoreceptor reflectance images collected across the macula in one eye of 10 normal participants (23-65 years) by using adaptive optics scanning light ophthalmoscopy (AOSLO). Infrared autofluorescence (IRAF) images of the RPE were collected with AOSLO in seven normal participants (22-65 years), with participant overlap, and compared to SWAF quantitatively and qualitatively. Results: RPE cell statistics could be analyzed in 84% of SWAF ROIs. RPE cell density consistently decreased with eccentricity from the fovea (participant mean ± SD: 6026 ± 1590 cells/mm2 at fovea; 4552 ± 1370 cells/mm2 and 3757 ± 1290 cells/mm2 at 3.5 mm temporally and nasally, respectively). Mean cone-to-RPE cell ratio decreased rapidly from 16.6 at the foveal center to <5 by 1 mm. IRAF revealed cells in six of seven participants, in agreement with SWAF RPE cell size and location. Differences in cell fluorescent structure, contrast, and visibility beneath vasculature were observed between modalities. Conclusions: Improvements in AOSLO autofluorescence imaging permit efficient visualization of RPE cells with safe light exposures, allowing individual characterization of RPE cell morphometry that is variable between participants. The normative dataset and analysis of RPE cell IRAF and SWAF herein are essential for understanding microscopic characteristics of cell fluorescence and may assist in interpreting disease progression in RPE cells.


Subject(s)
Retinal Cone Photoreceptor Cells/cytology , Retinal Pigment Epithelium/cytology , Adult , Aged , Cell Count , Female , Healthy Volunteers , Humans , Male , Middle Aged , Mosaicism , Ophthalmoscopy/methods , Optical Imaging , Optics and Photonics , Retinal Pigment Epithelium/diagnostic imaging , Tomography, Optical Coherence , Young Adult
4.
Br J Ophthalmol ; 102(1): 136-141, 2018 01.
Article in English | MEDLINE | ID: mdl-29074494

ABSTRACT

PURPOSE: Several genes causing autosomal-dominant cone-rod dystrophy (AD-CRD) have been identified. However, the mechanisms by which genetic mutations lead to cellular loss in human disease remain poorly understood. Here we combine genotyping with high-resolution adaptive optics retinal imaging to elucidate the retinal phenotype at a cellular level in patients with AD-CRD harbouring a defect in the GUCA1A gene. METHODS: Nine affected members of a four-generation AD-CRD pedigree and three unaffected first-degree relatives underwent clinical examinations including visual acuity, fundus examination, Goldmann perimetry, spectral domain optical coherence tomography and electroretinography. Genome-wide scan followed by bidirectional sequencing was performed on all affected participants. High-resolution imaging using a custom adaptive optics scanning light ophthalmoscope (AOSLO) was performed for selected participants. RESULTS: Clinical evaluations showed a range of disease severity from normal fundus appearance in teenaged patients to pronounced macular atrophy in older patients. Molecular genetic testing showed a mutation in in GUCA1A segregating with disease. AOSLO imaging revealed that of the two teenage patients with mild disease, one had severe disruption of the photoreceptor mosaic while the other had a normal cone mosaic. CONCLUSIONS: AOSLO imaging demonstrated variability in the pattern of cone and rod cell loss between two teenage cousins with early AD-CRD, who had similar clinical features and had the identical disease-causing mutation in GUCA1A. This finding suggests that a mutation in GUCA1A does not lead to the same degree of AD-CRD in all patients. Modifying factors may mitigate or augment disease severity, leading to different retinal cellular phenotypes.


Subject(s)
Cone-Rod Dystrophies/genetics , Electroretinography/instrumentation , Fluorescein Angiography/instrumentation , Optics and Photonics , Tomography, Optical Coherence/instrumentation , Visual Acuity , Adolescent , Adult , Aged , Child , Cone-Rod Dystrophies/diagnosis , Cone-Rod Dystrophies/therapy , Equipment Design , Female , Fundus Oculi , Genotype , Humans , Male , Middle Aged , Pedigree , Phenotype , Young Adult
5.
Proc Natl Acad Sci U S A ; 114(39): E8214-E8223, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28878022

ABSTRACT

Age-related macular degeneration (AMD) and related macular dystrophies (MDs) are a major cause of vision loss. However, the mechanisms underlying their progression remain ill-defined. This is partly due to the lack of disease models recapitulating the human pathology. Furthermore, in vivo studies have yielded limited understanding of the role of specific cell types in the eye vs. systemic influences (e.g., serum) on the disease pathology. Here, we use human induced pluripotent stem cell-retinal pigment epithelium (hiPSC-RPE) derived from patients with three dominant MDs, Sorsby's fundus dystrophy (SFD), Doyne honeycomb retinal dystrophy/malattia Leventinese (DHRD), and autosomal dominant radial drusen (ADRD), and demonstrate that dysfunction of RPE cells alone is sufficient for the initiation of sub-RPE lipoproteinaceous deposit (drusen) formation and extracellular matrix (ECM) alteration in these diseases. Consistent with clinical studies, sub-RPE basal deposits were present beneath both control (unaffected) and patient hiPSC-RPE cells. Importantly basal deposits in patient hiPSC-RPE cultures were more abundant and displayed a lipid- and protein-rich "drusen-like" composition. Furthermore, increased accumulation of COL4 was observed in ECM isolated from control vs. patient hiPSC-RPE cultures. Interestingly, RPE-specific up-regulation in the expression of several complement genes was also seen in patient hiPSC-RPE cultures of all three MDs (SFD, DHRD, and ADRD). Finally, although serum exposure was not necessary for drusen formation, COL4 accumulation in ECM, and complement pathway gene alteration, it impacted the composition of drusen-like deposits in patient hiPSC-RPE cultures. Together, the drusen model(s) of MDs described here provide fundamental insights into the unique biology of maculopathies affecting the RPE-ECM interface.


Subject(s)
Bruch Membrane/pathology , Eye Diseases, Hereditary/pathology , Induced Pluripotent Stem Cells/cytology , Macular Degeneration/pathology , Retinal Drusen/pathology , Retinal Pigment Epithelium/cytology , Blindness/genetics , Blindness/pathology , Cells, Cultured , Collagen Type IV/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Humans , Optic Disk Drusen/congenital , Optic Disk Drusen/pathology , Retinal Pigment Epithelium/pathology , Tissue Inhibitor of Metalloproteinase-3/genetics
6.
Proc Natl Acad Sci U S A ; 114(3): 586-591, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28049835

ABSTRACT

Although imaging of the living retina with adaptive optics scanning light ophthalmoscopy (AOSLO) provides microscopic access to individual cells, such as photoreceptors, retinal pigment epithelial cells, and blood cells in the retinal vasculature, other important cell classes, such as retinal ganglion cells, have proven much more challenging to image. The near transparency of inner retinal cells is advantageous for vision, as light must pass through them to reach the photoreceptors, but it has prevented them from being directly imaged in vivo. Here we show that the individual somas of neurons within the retinal ganglion cell (RGC) layer can be imaged with a modification of confocal AOSLO, in both monkeys and humans. Human images of RGC layer neurons did not match the quality of monkey images for several reasons, including safety concerns that limited the light levels permissible for human imaging. We also show that the same technique applied to the photoreceptor layer can resolve ambiguity about cone survival in age-related macular degeneration. The capability to noninvasively image RGC layer neurons in the living eye may one day allow for a better understanding of diseases, such as glaucoma, and accelerate the development of therapeutic strategies that aim to protect these cells. This method may also prove useful for imaging other structures, such as neurons in the brain.


Subject(s)
Ophthalmoscopy/methods , Retinal Ganglion Cells/cytology , Animals , Female , Glaucoma/diagnostic imaging , Humans , Macaca fascicularis/anatomy & histology , Macaca mulatta/anatomy & histology , Macular Degeneration/diagnostic imaging , Macular Degeneration/pathology , Male , Optical Phenomena , Retinal Cone Photoreceptor Cells/cytology , Species Specificity
7.
JAMA Ophthalmol ; 133(10): 1198-203, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26247787

ABSTRACT

IMPORTANCE: Stargardt disease (STGD1) is characterized by macular atrophy and flecks in the retinal pigment epithelium. The causative ABCA4 gene encodes a protein localizing to photoreceptor outer segments. The pathologic steps by which ABCA4 mutations lead to clinically detectable retinal pigment epithelium changes remain unclear. We investigated early STGD1 using adaptive optics scanning light ophthalmoscopy. OBSERVATIONS: Adaptive optics scanning light ophthalmoscopy imaging of 2 brothers with early STGD1 and their unaffected parents was compared with conventional imaging. Cone and rod spacing were increased in both patients (P < .001) with a dark cone appearance. No foveal cones were detected in the older brother. In the younger brother, foveal cones were enlarged with low density (peak cone density, 48.3 × 103 cones/mm2). The ratio of cone to rod spacing was increased in both patients, with greater divergence from normal approaching the foveal center, indicating that cone loss predominates centrally and rod loss increases peripherally. Both parents had normal photoreceptor mosaics. Genetic testing revealed 3 disease-causing mutations. CONCLUSIONS AND RELEVANCE: This study provides in vivo images of rods and cones in STGD1. Although the primary clinical features of STGD1 are retinal pigment epithelial lesions, adaptive optics scanning light ophthalmoscopy reveals increased cone and rod spacing in areas that appear normal in conventional images, suggesting that photoreceptor loss precedes clinically detectable retinal pigment epithelial disease in STGD1.


Subject(s)
Macular Degeneration/congenital , Photoreceptor Cells, Vertebrate/pathology , ATP-Binding Cassette Transporters/genetics , Female , Humans , Macular Degeneration/diagnosis , Macular Degeneration/genetics , Male , Mutation , Ophthalmoscopy/methods , Pedigree , Stargardt Disease , Tomography, Optical Coherence , Visual Acuity
8.
JAMA Ophthalmol ; 132(9): 1099-104, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24922193

ABSTRACT

IMPORTANCE: Fundus albipunctatus (FA) is a form of congenital stationary night blindness characterized by yellow-white spots, which were classically described as subretinal. Although night blindness and delayed dark adaptation are hallmarks of this condition, recent studies have described a macular phenotype, particularly among older patients. Using a fluorescence adaptive optics scanning laser ophthalmoscope (FAOSLO), this study provides in vivo morphologic data at the cellular level in FA. OBJECTIVE: To study the cone photoreceptors and the albipunctate spots in FA at single-cell resolution. DESIGN, SETTING, AND PARTICIPANT: A woman in her 30s with FA underwent a complete ophthalmic examination, including conventional imaging tests, at the University of Rochester. A FAOSLO was used to obtain infrared reflectance images of the cone mosaic at the central fovea and along the superior and temporal meridians to 10° eccentricity. Cone density was measured at the foveal center, and cone spacing was calculated in sampling windows eccentrically. In the area of the albipunctate spots, autofluorescence FAOSLO images (excitation, 561 nm; emission, 624 Δ 40 nm) were simultaneously obtained. MAIN OUTCOMES AND MEASURES: Structural appearance of cones, cone density and spacing, and reflectance and autofluorescence of albipunctate spots. RESULTS: Cone density was reduced to 70% of the lower limit of the normal range at the foveal center (78.7 × 10(3) cones/mm(2); mean [SD] reference range, 199 [87] × 10(3) cones/mm(2)), and cone spacing was increased eccentrically to 10° (sign test, P = .045). Individual cone central core reflectances appeared dim, suggesting loss of photoreceptor outer segments. The albipunctate spots were hypoautofluorescent. No photoreceptors or retinal pigment epithelium cells were identified at the locations of the albipunctate spots. CONCLUSIONS AND RELEVANCE: Although the predominant clinical symptom of night blindness and the electroretinography results suggest a primary rod dysfunction, examination with a FAOSLO demonstrates that cone density is also reduced. This finding may represent an early sign of progression to macular phenotype in FA. The hypoautofluorescence suggests that the albipunctate spots do not represent lipofuscin.


Subject(s)
Fluorescein Angiography , Ophthalmoscopes , Retinal Cone Photoreceptor Cells/pathology , Retinal Diseases/diagnosis , Adult , Cell Count , Disease Progression , Electroretinography , Female , Fovea Centralis , Humans , Tomography, Optical Coherence/methods , Visual Fields
9.
Biomed Opt Express ; 4(11): 2527-39, 2013.
Article in English | MEDLINE | ID: mdl-24298413

ABSTRACT

Morgan and colleagues demonstrated that the RPE cell mosaic can be resolved in the living human eye non-invasively by imaging the short-wavelength autofluorescence using an adaptive optics (AO) ophthalmoscope. This method, based on the assumption that all subjects have the same longitudinal chromatic aberration (LCA) correction, has proved difficult to use in diseased eyes, and in particular those affected by age-related macular degeneration (AMD). In this work, we improve Morgan's method by accounting for chromatic aberration variations by optimizing the confocal aperture axial and transverse placement through an automated iterative maximization of image intensity. The increase in image intensity after algorithmic aperture placement varied depending upon patient and aperture position prior to optimization but increases as large as a factor of 10 were observed. When using a confocal aperture of 3.4 Airy disks in diameter, images were obtained using retinal radiant exposures of less than 2.44 J/cm(2), which is ~22 times below the current ANSI maximum permissible exposure. RPE cell morphologies that were strikingly similar to those seen in postmortem histological studies were observed in AMD eyes, even in areas where the pattern of fluorescence appeared normal in commercial fundus autofluorescence (FAF) images. This new method can be used to study RPE morphology in AMD and other diseases, providing a powerful tool for understanding disease pathogenesis and progression, and offering a new means to assess the efficacy of treatments designed to restore RPE health.

10.
J Biol Chem ; 288(21): 14742-55, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23580649

ABSTRACT

The autosomal recessive mutation, sld, attenuates mucous cell expression in murine sublingual glands with corresponding effects on mucin 19 (Muc19). We conducted a systematic study including genetic mapping, sequencing, and functional analyses to elucidate a mutation to explain the sld phenotype in neonatal mice. Genetic mapping and gene expression analyses localized the sld mutation within the gene Muc19/Smgc, specifically attenuating Muc19 transcripts, and Muc19 knock-out mice mimic the sld phenotype in neonates. Muc19 transcription is unaffected in sld mice, whereas mRNA stability is markedly decreased. Decreased mRNA stability is not due to a defect in 3'-end processing nor to sequence differences in Muc19 transcripts. Comparative sequencing of the Muc19/Smgc gene identified four candidate intronic mutations within the Muc19 coding region. Minigene splicing assays revealed a novel splicing event in which insertion of two additional repeats within a CA repeat region of intron 53 of the sld genome enhances retention of intron 54, decreasing the levels of correctly spliced transcripts. Moreover, pateamine A, an inhibitor of nonsense-mediated mRNA decay, inhibits degradation of aberrant Muc19 transcripts. The mutation in intron 53 thus enhances aberrant splicing leading to degradation of aberrant transcripts and decreased Muc19 message stability, consistent with the sld phenotype. We propose a working model of the unique splicing event enhanced by the mutation, as well as putative explanations for the gradual but limited increase in Muc19 glycoprotein expression and its restricted localization to subpopulations of mucous cells in sld mice during postnatal gland development.


Subject(s)
Introns/physiology , Models, Biological , Mucins/biosynthesis , Mutation , RNA Stability/physiology , RNA, Messenger/metabolism , Sublingual Gland/metabolism , Alternative Splicing/physiology , Animals , Gene Expression Regulation/physiology , Mice , Mice, Knockout , Mucins/genetics , Open Reading Frames/physiology , RNA, Messenger/genetics , Sublingual Gland/cytology , Sublingual Gland/growth & development
11.
J Histochem Cytochem ; 52(5): 671-81, 2004 May.
Article in English | MEDLINE | ID: mdl-15100244

ABSTRACT

Human fractalkine (CX3CL1), a delta-chemokine, is implicated in the mediation of multiple cell functions. In addition to serving as a chemotactic factor for mononuclear cell subtypes, membrane-bound fractalkine may promote viral infection by interacting with virions that encode putative fractalkine-binding proteins. Fractalkine expression in normal epithelial tissues studied to date is either constitutive or is upregulated with inflammation. In salivary glands, the expression of fractalkine is unknown. Moreover, salivary glands are a major site for the persistent and productive infection by human herpesvirus (HHV)-7, which encodes two putative fractalkine-binding gene products, U12 and U51. Surprisingly, the cellular distribution of HHV-7 in major salivary glands has not been explored. We therefore determined by immunohistochemistry the cellular localization of fractalkine in three different salivary glands: parotid, submandibular, and labial glands. Fractalkine expression was highly variable, ranging from high to undetectable levels. We further examined the association of fractalkine with inflammatory cell infiltration or HHV-7 infection of salivary epithelial cells. Inflammatory cells were always adjacent to epithelial cells expressing fractalkine, consistent with a function of fractalkine in inflammatory cell recruitment and/or retention in salivary glands. In contrast, HHV-7-infected epithelial cells did not always express fractalkine, suggesting that fractalkine may not be an absolute requirement for viral entry.


Subject(s)
Chemokines, CX3C/metabolism , Herpesvirus 7, Human/metabolism , Inflammation/immunology , Inflammation/metabolism , Membrane Proteins/metabolism , Salivary Glands/metabolism , Adult , Aged , CD3 Complex/metabolism , Chemokine CX3CL1 , Epithelial Cells/metabolism , Epithelial Cells/virology , Female , Herpesvirus 7, Human/immunology , Humans , Immunohistochemistry , In Vitro Techniques , Inflammation/pathology , Male , Middle Aged , Parotid Gland/metabolism , Parotid Gland/pathology , Parotid Gland/virology , Salivary Glands/pathology , Salivary Glands/virology , Submandibular Gland/metabolism , Submandibular Gland/pathology , Submandibular Gland/virology
SELECTION OF CITATIONS
SEARCH DETAIL