Cargando…

Osteopontin accumulates in basal deposits of human eyes with age-related macular degeneration and may serve as a biomarker of aging

A common clinical phenotype of several neurodegenerative and systemic disorders including Alzheimer’s disease and atherosclerosis is the abnormal accumulation of extracellular material, which interferes with routine cellular functions. Similarly, patients with age-related macular degeneration (AMD),...

Descripción completa

Detalles Bibliográficos
Autores principales: Lekwuwa, Michael, Choudhary, Mayur, Lad, Eleonora M., Malek, Goldis
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group US 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8786662/
https://www.ncbi.nlm.nih.gov/pubmed/34389792
http://dx.doi.org/10.1038/s41379-021-00887-7
_version_ 1784639162723336192
author Lekwuwa, Michael
Choudhary, Mayur
Lad, Eleonora M.
Malek, Goldis
author_facet Lekwuwa, Michael
Choudhary, Mayur
Lad, Eleonora M.
Malek, Goldis
author_sort Lekwuwa, Michael
collection PubMed
description A common clinical phenotype of several neurodegenerative and systemic disorders including Alzheimer’s disease and atherosclerosis is the abnormal accumulation of extracellular material, which interferes with routine cellular functions. Similarly, patients with age-related macular degeneration (AMD), the leading cause of vision loss among the aged population, present with extracellular lipid- and protein-filled basal deposits in the back of the eye. While the exact mechanism of growth and formation of these deposits is poorly understood, much has been learned from investigating their composition, providing critical insights into AMD pathogenesis, prevention, and therapeutics. We identified human osteopontin (OPN), a phosphoprotein expressed in a variety of tissues in the body, as a newly discovered component of basal deposits in AMD patients, with a distinctive punctate staining pattern. OPN expression within these lesions, which are associated with AMD disease progression, were found to co-localize with abnormal calcium deposition. Additionally, OPN puncta colocalized with an AMD risk-associated complement pathway protein, but not with apolipoprotein E or vitronectin, two other well-established basal deposit components. Mechanistically, we found that retinal pigment epithelial cells, cells vulnerable in AMD, will secrete OPN into the extracellular space, under oxidative stress conditions, supporting OPN biosynthesis locally within the outer retina. Finally, we report that OPN levels in plasma of aged (non-AMD) human donors were significantly higher than levels in young (non-AMD) donors, but were not significantly different from donors with the different clinical subtypes of AMD. Collectively, our study defines the expression pattern of OPN in the posterior pole as a function of disease, and its local expression as a potential histopathologic biomarker of AMD.
format Online
Article
Text
id pubmed-8786662
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Nature Publishing Group US
record_format MEDLINE/PubMed
spelling pubmed-87866622022-02-04 Osteopontin accumulates in basal deposits of human eyes with age-related macular degeneration and may serve as a biomarker of aging Lekwuwa, Michael Choudhary, Mayur Lad, Eleonora M. Malek, Goldis Mod Pathol Article A common clinical phenotype of several neurodegenerative and systemic disorders including Alzheimer’s disease and atherosclerosis is the abnormal accumulation of extracellular material, which interferes with routine cellular functions. Similarly, patients with age-related macular degeneration (AMD), the leading cause of vision loss among the aged population, present with extracellular lipid- and protein-filled basal deposits in the back of the eye. While the exact mechanism of growth and formation of these deposits is poorly understood, much has been learned from investigating their composition, providing critical insights into AMD pathogenesis, prevention, and therapeutics. We identified human osteopontin (OPN), a phosphoprotein expressed in a variety of tissues in the body, as a newly discovered component of basal deposits in AMD patients, with a distinctive punctate staining pattern. OPN expression within these lesions, which are associated with AMD disease progression, were found to co-localize with abnormal calcium deposition. Additionally, OPN puncta colocalized with an AMD risk-associated complement pathway protein, but not with apolipoprotein E or vitronectin, two other well-established basal deposit components. Mechanistically, we found that retinal pigment epithelial cells, cells vulnerable in AMD, will secrete OPN into the extracellular space, under oxidative stress conditions, supporting OPN biosynthesis locally within the outer retina. Finally, we report that OPN levels in plasma of aged (non-AMD) human donors were significantly higher than levels in young (non-AMD) donors, but were not significantly different from donors with the different clinical subtypes of AMD. Collectively, our study defines the expression pattern of OPN in the posterior pole as a function of disease, and its local expression as a potential histopathologic biomarker of AMD. Nature Publishing Group US 2021-08-13 2022 /pmc/articles/PMC8786662/ /pubmed/34389792 http://dx.doi.org/10.1038/s41379-021-00887-7 Text en © The Author(s) 2021 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Lekwuwa, Michael
Choudhary, Mayur
Lad, Eleonora M.
Malek, Goldis
Osteopontin accumulates in basal deposits of human eyes with age-related macular degeneration and may serve as a biomarker of aging
title Osteopontin accumulates in basal deposits of human eyes with age-related macular degeneration and may serve as a biomarker of aging
title_full Osteopontin accumulates in basal deposits of human eyes with age-related macular degeneration and may serve as a biomarker of aging
title_fullStr Osteopontin accumulates in basal deposits of human eyes with age-related macular degeneration and may serve as a biomarker of aging
title_full_unstemmed Osteopontin accumulates in basal deposits of human eyes with age-related macular degeneration and may serve as a biomarker of aging
title_short Osteopontin accumulates in basal deposits of human eyes with age-related macular degeneration and may serve as a biomarker of aging
title_sort osteopontin accumulates in basal deposits of human eyes with age-related macular degeneration and may serve as a biomarker of aging
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8786662/
https://www.ncbi.nlm.nih.gov/pubmed/34389792
http://dx.doi.org/10.1038/s41379-021-00887-7
work_keys_str_mv AT lekwuwamichael osteopontinaccumulatesinbasaldepositsofhumaneyeswithagerelatedmaculardegenerationandmayserveasabiomarkerofaging
AT choudharymayur osteopontinaccumulatesinbasaldepositsofhumaneyeswithagerelatedmaculardegenerationandmayserveasabiomarkerofaging
AT ladeleonoram osteopontinaccumulatesinbasaldepositsofhumaneyeswithagerelatedmaculardegenerationandmayserveasabiomarkerofaging
AT malekgoldis osteopontinaccumulatesinbasaldepositsofhumaneyeswithagerelatedmaculardegenerationandmayserveasabiomarkerofaging