1
|
Besirli CG, Nath M, Yao J, Pawar M, Myers AM, Zacks D, Fort PE. HSPB4/CRYAA Protect Photoreceptors during Retinal Detachment in Part through FAIM2 Regulation. Neurol Int 2024; 16:905-917. [PMID: 39311341 PMCID: PMC11417767 DOI: 10.3390/neurolint16050068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/26/2024] Open
Abstract
Our previous study discussed crystallin family induction in an experimental rat model of retinal detachment. Therefore, we attempted to evaluate the role of α-crystallin in photoreceptor survival in an experimental model of retinal detachment, as well as its association with the intrinsically neuroprotective protein Fas-apoptotic inhibitory molecule 2 (FAIM2). Separation of retina and RPE was induced in rat and mouse eyes by subretinal injection of hyaluronic acid. Retinas were subsequently analyzed for the presence αA-crystallin (HSPB4) and αB-crystallin (HSPB5) proteins using immunohistochemistry and immunoblotting. Photoreceptor death was analyzed using terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling (TUNEL) staining and cell counts. The 661W cells subjected to FasL were used as a cell model of photoreceptor degeneration to assess the mechanisms of the protective effect of αA-crystallin and its dependence on its phosphorylation on T148. We further evaluated the interaction between FAIM2 and αA-crystallin using a co-immunoprecipitation assay. Our results showed that α-crystallin protein levels were rapidly induced in response to retinal detachment, with αA-crystallin playing a particularly important role in protecting photoreceptors during retinal detachment. Our data also show that the photoreceptor intrinsically neuroprotective protein FAIM2 is induced and interacts with α-crystallins following retinal detachment. Mechanistically, our work also demonstrated that the phosphorylation of αA-crystallin is important for the interaction of αA-crystallin with FAIM2 and their neuroprotective effect. Thus, αA-crystallin is involved in the regulation of photoreceptor survival during retinal detachment, playing a key role in the stabilization of FAIM2, serving as an important modulator of photoreceptor cell survival under chronic stress conditions.
Collapse
Affiliation(s)
- Cagri G. Besirli
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA; (C.G.B.)
| | - Madhu Nath
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA; (C.G.B.)
| | - Jingyu Yao
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA; (C.G.B.)
| | - Mercy Pawar
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA; (C.G.B.)
| | - Angela M. Myers
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA; (C.G.B.)
| | - David Zacks
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA; (C.G.B.)
| | - Patrice E. Fort
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA; (C.G.B.)
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
2
|
Lunding BS, Bassi MR, Christensen JP, Thomsen AR, Sørensen TL, Vorum H, Honoré B, Nissen MH, Steffensen MA. Systemic infection in aged mice causes upregulation of crystallin alpha A in the RPE/choroid. Exp Eye Res 2024; 245:109984. [PMID: 38945517 DOI: 10.1016/j.exer.2024.109984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/24/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Aging changes the responsiveness of our immune defense, and this decline in immune reactivity plays an important role in the increased susceptibility to infections that marks progressing age. Aging is also the most pronounced risk factor for development of age-related macular degeneration (AMD), a disease that is characterized by dysfunctional retinal pigment epithelial (RPE) cells and loss of central vision. We have previously shown that acute systemic viral infection has a large impact on the retina in young mice, leading to upregulation of chemokines in the RPE/choroid (RPE/c) and influx of CD8 T cells in the neuroretina. In this study, we sought to investigate the impact of systemic infection on the RPE/c in aged mice to evaluate whether infection in old age could play a role in the pathogenesis of AMD. We found that systemic infection in mice led to upregulation of genes from the crystallin family in the RPE/c from aged mice, but not in the RPE/c from young mice. Crystallin alpha A (CRYAA) was the most upregulated gene, and increased amounts of CRYAA protein were also detected in the aged RPE/c. Increased CRYAA gene and protein expression has previously been found in drusen and choroid from AMD patients, and this protein has also been linked to neovascularization. Since both drusen and neovascularization are important hallmarks of advanced AMD, it is interesting to speculate if upregulation of crystallins in response to infection in old age could be relevant for the pathogenesis of AMD.
Collapse
Affiliation(s)
| | | | | | | | - Torben Lykke Sørensen
- Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark; Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Henrik Vorum
- Department of Clinical Medicine, Aalborg University, Denmark; Department of Ophthalmology, Aalborg University Hospital, Denmark
| | - Bent Honoré
- Department of Biomedicine, Aarhus University, Denmark
| | - Mogens Holst Nissen
- Department of Immunology and Microbiology, University of Copenhagen, Denmark
| | | |
Collapse
|
3
|
Patil H, Yi H, Cho KI, Ferreira PA. Proteostatic Remodeling of Small Heat Shock Chaperones─Crystallins by Ran-Binding Protein 2─and the Peptidyl-Prolyl cis-trans Isomerase and Chaperone Activities of Its Cyclophilin Domain. ACS Chem Neurosci 2024; 15:1967-1989. [PMID: 38657106 DOI: 10.1021/acschemneuro.3c00792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Abstract
Disturbances in protein phase transitions promote protein aggregation─a neurodegeneration hallmark. The modular Ran-binding protein 2 (Ranbp2) is a cytosolic molecular hub for rate-limiting steps of phase transitions of Ran-GTP-bound protein ensembles exiting nuclear pores. Chaperones also regulate phase transitions and proteostasis by suppressing protein aggregation. Ranbp2 haploinsufficiency promotes the age-dependent neuroprotection of the chorioretina against phototoxicity by proteostatic regulations of neuroprotective substrates of Ranbp2 and by suppressing the buildup of polyubiquitylated substrates. Losses of peptidyl-prolyl cis-trans isomerase (PPIase) and chaperone activities of the cyclophilin domain (CY) of Ranbp2 recapitulate molecular effects of Ranbp2 haploinsufficiency. These CY impairments also stimulate deubiquitylation activities and phase transitions of 19S cap subunits of the 26S proteasome that associates with Ranbp2. However, links between CY moonlighting activity, substrate ubiquitylation, and proteostasis remain incomplete. Here, we reveal the Ranbp2 regulation of small heat shock chaperones─crystallins in the chorioretina by proteomics of mice with total or selective modular deficits of Ranbp2. Specifically, loss of CY PPIase of Ranbp2 upregulates αA-Crystallin, which is repressed in adult nonlenticular tissues. Conversely, impairment of CY's chaperone activity opposite to the PPIase pocket downregulates a subset of αA-Crystallin's substrates, γ-crystallins. These CY-dependent effects cause age-dependent and chorioretinal-selective declines of ubiquitylated substrates without affecting the chorioretinal morphology. A model emerges whereby inhibition of Ranbp2's CY PPIase remodels crystallins' expressions, subdues molecular aging, and preordains the chorioretina to neuroprotection by augmenting the chaperone capacity and the degradation of polyubiquitylated substrates against proteostatic impairments. Further, the druggable Ranbp2 CY holds pan-therapeutic potential against proteotoxicity and neurodegeneration.
Collapse
Affiliation(s)
- Hemangi Patil
- Department of Ophthalmology Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Haiqing Yi
- Department of Ophthalmology Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Kyoung-In Cho
- Department of Ophthalmology Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Paulo A Ferreira
- Department of Ophthalmology Duke University Medical Center, Durham, North Carolina 27710, United States
- Department of Pathology Duke University Medical Center, Durham, North Carolina 27710, United States
| |
Collapse
|
4
|
Patil H, Cho KI, Ferreira PA. Proteostatic remodeling of small heat shock chaperones - crystallins by Ran-binding protein 2 and the peptidyl-prolyl cis-trans isomerase and chaperone activities of its cyclophilin domain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.26.577462. [PMID: 38352504 PMCID: PMC10862737 DOI: 10.1101/2024.01.26.577462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Disturbances in phase transitions and intracellular partitions of nucleocytoplasmic shuttling substrates promote protein aggregation - a hallmark of neurodegenerative diseases. The modular Ran-binding protein 2 (Ranbp2) is a cytosolic molecular hub for rate-limiting steps of disassembly and phase transitions of Ran-GTP-bound protein ensembles exiting nuclear pores. Chaperones also play central roles in phase transitions and proteostasis by suppressing protein aggregation. Ranbp2 haploinsufficiency promotes the age-dependent neuroprotection of the chorioretina against photo-oxidative stress by proteostatic regulations of Ranbp2 substrates and by countering the build-up of poly-ubiquitylated substrates. Further, the peptidyl-prolyl cis-trans isomerase (PPIase) and chaperone activities of the cyclophilin domain (CY) of Ranbp2 modulate the proteostasis of selective neuroprotective substrates, such as hnRNPA2B1, STAT3, HDAC4 or L/M-opsin, while promoting a decline of ubiquitylated substrates. However, links between CY PPIase activity on client substrates and its effect(s) on ubiquitylated substrates are unclear. Here, proteomics of genetically modified mice with deficits of Ranbp2 uncovered the regulation of the small heat shock chaperones - crystallins by Ranbp2 in the chorioretina. Loss of CY PPIase of Ranbp2 up-regulates αA-crystallin proteostasis, which is repressed in non-lenticular tissues. Conversely, the αA-crystallin's substrates, γ-crystallins, are down-regulated by impairment of CY's C-terminal chaperone activity. These CY-dependent effects cause the age-dependent decline of ubiquitylated substrates without overt chorioretinal morphological changes. A model emerges whereby the Ranbp2 CY-dependent remodeling of crystallins' proteostasis subdues molecular aging and preordains chorioretinal neuroprotection by augmenting the chaperone buffering capacity and the decline of ubiquitylated substrates against proteostatic impairments. Further, CY's moonlighting activity holds pan -therapeutic potential against neurodegeneration.
Collapse
|
5
|
Manikandan SK, Logan A, Cerrada-Gimenez M, Fitzhenry L, Coffey L, Kaja S, Rani S. Immune System, Inflammation and Autoantigens in Wet Age-Related Macular Degeneration: Pathological Significance and Therapeutic Importance. Life (Basel) 2023; 13:2236. [PMID: 38137838 PMCID: PMC10744676 DOI: 10.3390/life13122236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 12/24/2023] Open
Abstract
Wet age-related macular degeneration (wAMD) is a chronic inflammation-associated neurodegenerative disease affecting the posterior part of the eye in the aging population. Aging results in the reduced functionality of cells and tissues, including the cells of the retina. Initiators of a chronic inflammatory and pathologic state in wAMD may be a result of the accumulation of inevitable metabolic injuries associated with the maintenance of tissue homeostasis from a young age to over 50. Apart from this, risk factors like smoking, genetic predisposition, and failure to repair the injuries that occur, alongside attempts to rescue the hypoxic outer retina may also contribute to the pathogenesis. Aging of the immune system (immunosenescence) and a compromised outer blood retinal barrier (BRB) result in the exposure of the privileged milieu of the retina to the systemic immune system, further increasing the severity of the disease. When immune-privileged sites like the retina are under pathological stress, certain age- and disease-related conditions may necessitate assistance from cells distant from the resident ones to help restore the functionality of the tissue. As a necessary part of tissue repair, inflammation is a major response to disease and recruits immune cells to the site of damage. We suspect that the specific reparative inflammatory responses are controlled by an autoantigen-T cell-mediated mechanism, a process that may be hindered in wAMD.
Collapse
Affiliation(s)
- Sreeraj Kuruppilakath Manikandan
- Ocular Therapeutics Research Group, Pharmaceutical and Molecular Biotechnology Research Centre, South East Technological University, Waterford Campus, X91 K0EK Waterford, Ireland; (S.K.M.); (L.F.)
| | - Ann Logan
- Department of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7HL, UK;
| | | | - Laurence Fitzhenry
- Ocular Therapeutics Research Group, Pharmaceutical and Molecular Biotechnology Research Centre, South East Technological University, Waterford Campus, X91 K0EK Waterford, Ireland; (S.K.M.); (L.F.)
| | - Lee Coffey
- Ocular Therapeutics Research Group, Pharmaceutical and Molecular Biotechnology Research Centre, South East Technological University, Waterford Campus, X91 K0EK Waterford, Ireland; (S.K.M.); (L.F.)
| | - Simon Kaja
- Departments of Ophthalmology, Molecular Pharmacology & Neuroscience, Loyola University Chicago, Maywood, IL 60153, USA
| | - Sweta Rani
- Ocular Therapeutics Research Group, Pharmaceutical and Molecular Biotechnology Research Centre, South East Technological University, Waterford Campus, X91 K0EK Waterford, Ireland; (S.K.M.); (L.F.)
| |
Collapse
|
6
|
Carmy‐Bennun T, Myer C, Bhattacharya SK, Hackam AS. Quantitative proteomic analysis after neuroprotective MyD88 inhibition in the retinal degeneration 10 mouse. J Cell Mol Med 2021; 25:9533-9542. [PMID: 34562309 PMCID: PMC8505828 DOI: 10.1111/jcmm.16893] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/13/2021] [Accepted: 08/18/2021] [Indexed: 12/18/2022] Open
Abstract
Progressive photoreceptor death occurs in blinding diseases such as retinitis pigmentosa. Myeloid differentiation primary response protein 88 (MyD88) is a central adaptor protein for innate immune system Toll-like receptors (TLR) and induces cytokine secretion during retinal disease. We recently demonstrated that inhibiting MyD88 in mouse models of retinal degeneration led to increased photoreceptor survival, which was associated with altered cytokines and increased neuroprotective microglia. However, the identity of additional molecular changes associated with MyD88 inhibitor-induced neuroprotection is not known. In this study, we used isobaric tags for relative and absolute quantification (iTRAQ) labelling followed by LC-MS/MS for quantitative proteomic analysis on the rd10 mouse model of retinal degeneration to identify protein pathways changed by MyD88 inhibition. Quantitative proteomics using iTRAQ LC-MS/MS is a high-throughput method ideal for providing insight into molecular pathways during disease and experimental treatments. Forty-two proteins were differentially expressed in retinas from mice treated with MyD88 inhibitor compared with control. Notably, increased expression of multiple crystallins and chaperones that respond to cellular stress and have anti-apoptotic properties was identified in the MyD88-inhibited mice. These data suggest that inhibiting MyD88 enhances chaperone-mediated retinal protection pathways. Therefore, this study provides insight into molecular events contributing to photoreceptor protection from modulating inflammation.
Collapse
Affiliation(s)
- Tal Carmy‐Bennun
- Bascom Palmer Eye InstituteUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Ciara Myer
- Bascom Palmer Eye InstituteUniversity of Miami Miller School of MedicineMiamiFLUSA
- Miami Integrative Metabolomics Research CenterMiamiFLUSA
| | - Sanjoy K. Bhattacharya
- Bascom Palmer Eye InstituteUniversity of Miami Miller School of MedicineMiamiFLUSA
- Miami Integrative Metabolomics Research CenterMiamiFLUSA
| | - Abigail S. Hackam
- Bascom Palmer Eye InstituteUniversity of Miami Miller School of MedicineMiamiFLUSA
- Miami Integrative Metabolomics Research CenterMiamiFLUSA
| |
Collapse
|
7
|
Wood JPM, Tahmasebi M, Casson RJ, Plunkett M, Chidlow G. Physiological response of the retinal pigmented epithelium to 3-ns pulse laser application, in vitro and in vivo. Clin Exp Ophthalmol 2021; 49:454-469. [PMID: 33904222 DOI: 10.1111/ceo.13931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/30/2021] [Accepted: 04/17/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND To treat healthy retinal pigmented epithelium (RPE) with the 3-ns retinal rejuvenation therapy (2RT) laser and to investigate the subsequent wound-healing response of these cells. METHODS Primary rat RPE cells were treated with the 2RT laser at a range of energy settings. Treated cells were fixed up to 7 days post-irradiation and assessed for expression of proteins associated with wound-healing. For in vivo treatments, eyes of Dark Agouti rats were exposed to laser and tissues collected up to 7 days post-irradiation. Isolated wholemount RPE preparations were examined for structural and protein expression changes. RESULTS Cultured RPE cells were ablated by 2RT laser in an energy-dependent manner. In all cases, the RPE cell layer repopulated completely within 7 days. Replenishment of RPE cells was associated with expression of the heat shock protein, Hsp27, the intermediate filament proteins, vimentin and nestin, and the cell cycle-associated protein, cyclin D1. Cellular tight junctions were lost in lased regions but re-expressed when cell replenishment was complete. In vivo, 2RT treatment gave rise to both an energy-dependent localised denudation of the RPE and the subsequent repopulation of lesion sites. Cell replenishment was associated with the increased expression of cyclin D1, vimentin and the heat shock proteins Hsp27 and αB-crystallin. CONCLUSIONS The 2RT laser was able to target the RPE both in vitro and in vivo, causing debridement of the cells and the consequent stimulation of a wound-healing response leading to layer reformation.
Collapse
Affiliation(s)
- John P M Wood
- Central Adelaide Local Health Network, Adelaide, South Australia, Australia.,Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Marzieh Tahmasebi
- Central Adelaide Local Health Network, Adelaide, South Australia, Australia.,Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Robert J Casson
- Central Adelaide Local Health Network, Adelaide, South Australia, Australia.,Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Malcolm Plunkett
- Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Glyn Chidlow
- Central Adelaide Local Health Network, Adelaide, South Australia, Australia.,Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
8
|
Somasundaran S, Constable IJ, Mellough CB, Carvalho LS. Retinal pigment epithelium and age-related macular degeneration: A review of major disease mechanisms. Clin Exp Ophthalmol 2020; 48:1043-1056. [PMID: 32710488 PMCID: PMC7754492 DOI: 10.1111/ceo.13834] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/05/2020] [Accepted: 07/10/2020] [Indexed: 01/18/2023]
Abstract
Age‐related macular degeneration (AMD) is a progressive degenerative disease that is the leading cause of vision loss in the elderly population. Degeneration/dysregulation of the retinal pigment epithelium (RPE), a supportive monolayer of cells underlying the photoreceptors, is commonly seen in patients with AMD. While treatment exists for the neovascular/wet form of AMD, there is currently no cure for the non‐exudative/dry form of AMD, making it imperative to understand the pathogenesis of this disease. Although our understanding of the aetiology of AMD has increased over the years, the underlying disease mechanism has not yet been identified, mainly due to the multifactorial nature of this disease. Herein, we review some of the commonly proposed degeneration pathways of RPE cells and their role in the pathogenesis of AMD; including activation of the complement cascade, oxidative stress‐induced cell death mechanisms, dysfunctional mitochondria and the role of crystallins in AMD disease progression.
Collapse
Affiliation(s)
- Shreya Somasundaran
- Centre for Ophthalmology and Visual Science/Lions Eye Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Ian J Constable
- Centre for Ophthalmology and Visual Science/Lions Eye Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Carla B Mellough
- Centre for Ophthalmology and Visual Science/Lions Eye Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Livia S Carvalho
- Centre for Ophthalmology and Visual Science/Lions Eye Institute, University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
9
|
A Re-Appraisal of Pathogenic Mechanisms Bridging Wet and Dry Age-Related Macular Degeneration Leads to Reconsider a Role for Phytochemicals. Int J Mol Sci 2020; 21:ijms21155563. [PMID: 32756487 PMCID: PMC7432893 DOI: 10.3390/ijms21155563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/31/2020] [Accepted: 08/01/2020] [Indexed: 12/14/2022] Open
Abstract
Which pathogenic mechanisms underlie age-related macular degeneration (AMD)? Are they different for dry and wet variants, or do they stem from common metabolic alterations? Where shall we look for altered metabolism? Is it the inner choroid, or is it rather the choroid–retinal border? Again, since cell-clearing pathways are crucial to degrade altered proteins, which metabolic system is likely to be the most implicated, and in which cell type? Here we describe the unique clearing activity of the retinal pigment epithelium (RPE) and the relevant role of its autophagy machinery in removing altered debris, thus centering the RPE in the pathogenesis of AMD. The cell-clearing systems within the RPE may act as a kernel to regulate the redox homeostasis and the traffic of multiple proteins and organelles toward either the choroid border or the outer segments of photoreceptors. This is expected to cope with the polarity of various domains within RPE cells, with each one owning a specific metabolic activity. A defective clearance machinery may trigger unconventional solutions to avoid intracellular substrates’ accumulation through unconventional secretions. These components may be deposited between the RPE and Bruch’s membrane, thus generating the drusen, which remains the classic hallmark of AMD. These deposits may rather represent a witness of an abnormal RPE metabolism than a real pathogenic component. The empowerment of cell clearance, antioxidant, anti-inflammatory, and anti-angiogenic activity of the RPE by specific phytochemicals is here discussed.
Collapse
|
10
|
Mammadzada P, Corredoira PM, André H. The role of hypoxia-inducible factors in neovascular age-related macular degeneration: a gene therapy perspective. Cell Mol Life Sci 2020; 77:819-833. [PMID: 31893312 PMCID: PMC7058677 DOI: 10.1007/s00018-019-03422-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/04/2019] [Accepted: 12/10/2019] [Indexed: 12/19/2022]
Abstract
Understanding the mechanisms that underlie age-related macular degeneration (AMD) has led to the identification of key molecules. Hypoxia-inducible transcription factors (HIFs) have been associated with choroidal neovascularization and the progression of AMD into the neovascular clinical phenotype (nAMD). HIFs regulate the expression of multiple growth factors and cytokines involved in angiogenesis and inflammation, hallmarks of nAMD. This knowledge has propelled the development of a new group of therapeutic strategies focused on gene therapy. The present review provides an update on current gene therapies in ocular angiogenesis, particularly nAMD, from both basic and clinical perspectives.
Collapse
Affiliation(s)
- Parviz Mammadzada
- Division of Eye and Vision, Department of Clinical Neuroscience, Karolinska Institutet, St. Erik Eye Hospital, Stockholm, Sweden
| | - Pablo M Corredoira
- Division of Eye and Vision, Department of Clinical Neuroscience, Karolinska Institutet, St. Erik Eye Hospital, Stockholm, Sweden
| | - Helder André
- Division of Eye and Vision, Department of Clinical Neuroscience, Karolinska Institutet, St. Erik Eye Hospital, Stockholm, Sweden.
| |
Collapse
|
11
|
Valdés-Sánchez L, Calado SM, de la Cerda B, Aramburu A, García-Delgado AB, Massalini S, Montero-Sánchez A, Bhatia V, Rodríguez-Bocanegra E, Diez-Lloret A, Rodríguez-Martínez D, Chakarova C, Bhattacharya SS, Díaz-Corrales FJ. Retinal pigment epithelium degeneration caused by aggregation of PRPF31 and the role of HSP70 family of proteins. Mol Med 2019; 26:1. [PMID: 31892304 PMCID: PMC6938640 DOI: 10.1186/s10020-019-0124-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 12/05/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Mutations in pre-mRNA splicing factor PRPF31 can lead to retinitis pigmentosa (RP). Although the exact disease mechanism remains unknown, it has been hypothesized that haploinsufficiency might be involved in the pathophysiology of the disease. METHODS In this study, we have analyzed a mouse model containing the p.A216P mutation in Prpf31 gene. RESULTS We found that mutant Prpf31 protein produces cytoplasmic aggregates in the retinal pigment epithelium and decreasing the protein levels of this splicing factor in the nucleus. Additionally, normal protein was recruited in insoluble aggregates when the mutant protein was overexpressed in vitro. In response to protein aggregation, Hspa4l is overexpressed. This member of the HSP70 family of chaperones might contribute to the correct folding and solubilization of the mutant protein, allowing its translocation to the nucleus. CONCLUSIONS Our data suggests that a mechanism haploinsufficiency and dominant-negative is involved in retinal degeneration due to mutations in PRPF31. HSP70 over-expression might be a new therapeutic target for the treatment of retinal degeneration due to PRPF31 mutations.
Collapse
Affiliation(s)
- Lourdes Valdés-Sánchez
- Regeneration and Cell Therapy Department, Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER (Junta de Andalucía), CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Americo Vespucio 24, 41092, Seville, Spain
| | - Sofia M Calado
- Regeneration and Cell Therapy Department, Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER (Junta de Andalucía), CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Americo Vespucio 24, 41092, Seville, Spain
- Present Address: Center for Biomedical Research (CBMR), University of Algarve, 8800-139, Faro, Portugal
| | - Berta de la Cerda
- Regeneration and Cell Therapy Department, Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER (Junta de Andalucía), CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Americo Vespucio 24, 41092, Seville, Spain
| | - Ana Aramburu
- Regeneration and Cell Therapy Department, Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER (Junta de Andalucía), CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Americo Vespucio 24, 41092, Seville, Spain
- Present Address: Clinique de l'Oeil, Avenue Bois de la Chapelle 15, 1213, Onex, Switzerland
| | - Ana Belén García-Delgado
- Regeneration and Cell Therapy Department, Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER (Junta de Andalucía), CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Americo Vespucio 24, 41092, Seville, Spain
| | - Simone Massalini
- Regeneration and Cell Therapy Department, Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER (Junta de Andalucía), CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Americo Vespucio 24, 41092, Seville, Spain
- Present Address: Center for Molecular and Cellular Bioengineering (CMCB) DFG-Research Center for Regenerative Therapies Dresden (CRTD) Cluster of Excellence, Technische Universität Dresden, Fetscherstraße, 105 01307, Dresden, Germany
| | - Adoración Montero-Sánchez
- Regeneration and Cell Therapy Department, Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER (Junta de Andalucía), CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Americo Vespucio 24, 41092, Seville, Spain
| | - Vaibhav Bhatia
- Regeneration and Cell Therapy Department, Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER (Junta de Andalucía), CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Americo Vespucio 24, 41092, Seville, Spain
| | - Eduardo Rodríguez-Bocanegra
- Regeneration and Cell Therapy Department, Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER (Junta de Andalucía), CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Americo Vespucio 24, 41092, Seville, Spain
- Present Address: Universitätsklinikum Tübingen, Forschungsinstitut für Augenheilkunde, Elfriede-Aulhorn-Str. 7, 72076, Tübingen, Germany
| | - Andrea Diez-Lloret
- Regeneration and Cell Therapy Department, Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER (Junta de Andalucía), CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Americo Vespucio 24, 41092, Seville, Spain
| | - Daniel Rodríguez-Martínez
- Regeneration and Cell Therapy Department, Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER (Junta de Andalucía), CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Americo Vespucio 24, 41092, Seville, Spain
| | - Christina Chakarova
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Shom S Bhattacharya
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| | - Francisco J Díaz-Corrales
- Regeneration and Cell Therapy Department, Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER (Junta de Andalucía), CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Americo Vespucio 24, 41092, Seville, Spain.
| |
Collapse
|
12
|
Gong J, Cai H, Noggle S, Paull D, Rizzolo LJ, Del Priore LV, Fields MA. Stem cell-derived retinal pigment epithelium from patients with age-related macular degeneration exhibit reduced metabolism and matrix interactions. Stem Cells Transl Med 2019; 9:364-376. [PMID: 31840941 PMCID: PMC7031648 DOI: 10.1002/sctm.19-0321] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022] Open
Abstract
Modeling age‐related macular degeneration (AMD) is challenging, because it is a multifactorial disease. To focus on interactions between the retinal pigment epithelium (RPE) and Bruch's membrane, we generated RPE from AMD patients and used an altered extracellular matrix (ECM) that models aged Bruch's membrane. Induced pluripotent stem cells (iPSCs) were generated from fibroblasts isolated from AMD patients or age‐matched (normal) controls. RPE derived from iPSCs were analyzed by morphology, marker expression, transepithelial electrical resistance (TER), and phagocytosis of rod photoreceptor outer segments. Cell attachment and viability was tested on nitrite‐modified ECM, a typical modification of aged Bruch's membrane. DNA microarrays with hierarchical clustering and analysis of mitochondrial function were used to elucidate possible mechanisms for the observed phenotypes. Differentiated RPE displayed cell‐specific morphology and markers. The TER and phagocytic capacity were similar among iPSC‐derived RPE cultures. However, distinct clusters were found for the transcriptomes of AMD and control iPSC‐derived RPE. AMD‐derived iPSC‐RPE downregulated genes responsible for metabolic‐related pathways and cell attachment. AMD‐derived iPSC‐RPE exhibited reduced mitochondrial respiration and ability to attach and survive on nitrite‐modified ECM. Cells that did attach induced the expression of complement genes. Despite reprogramming, iPSC derived from AMD patients yielded RPE with a transcriptome that is distinct from that of age‐matched controls. When challenged with an AMD‐like modification of Bruch's membrane, AMD‐derived iPSC‐RPE activated the complement immune system.
Collapse
Affiliation(s)
- Jie Gong
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut
| | - Hui Cai
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut
| | -
- The New York Stem Cell Foundation (NYSCF) Research Institute, New York, New York
| | - Scott Noggle
- The New York Stem Cell Foundation (NYSCF) Research Institute, New York, New York
| | - Daniel Paull
- The New York Stem Cell Foundation (NYSCF) Research Institute, New York, New York
| | - Lawrence J Rizzolo
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut.,Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Lucian V Del Priore
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut
| | - Mark A Fields
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
13
|
Wei Q, Jiang C, Ye X, Huang X, Jin H, Xu G. Vitreous Proteomics Provides New Insights into Antivascular Endothelial Growth Factor Therapy for Pathologic Myopia Choroid Neovascularization. J Interferon Cytokine Res 2019; 39:786-796. [PMID: 31718389 DOI: 10.1089/jir.2019.0030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This study aimed to investigate the protein expression profile of vitreous humor (VH) from pathologic myopic retinoschisis (PMRS) patients with or without intravitreal antivascular endothelial growth factor (anti-VEGF) therapy. VH samples from PMRS patients were subjected to proteomic analysis. Clinical data, including visual acuity, refractive error, and axial length, were recorded, and the fundus optical coherence tomography was performed. Seven PMRS patients were enrolled: 3 PMRS patients as control group, 3 PMRS patients with coexisting choroidal neovascularization (CNV) who developed retinoschisis aggravation after multiple intravitreal conbercept (IVC) injections, and one PMRS patient with coexisting CNV without leakage CNV (CNV-). A total of 310 differentially expressed proteins were identified in these VH samples. The expression of 28 proteins, related to cellular adhesion, protease inhibitors, proangiogenic factors, and antiangiogenic factors, was significantly downregulated in the IVC-treated eyes than in control- and CNV-eyes. α-smooth muscle actin (α-SMA) expression was significantly upregulated in the IVC-treated eyes. Furthermore, the expression of αA-crystallin and fibrillin-1 was significantly upregulated in both IVC and CNV-eyes than in control eyes. These suggest that multiple IVC injections may increase the VH αSMA concentration, which may contribute to posterior hyaloid membrane or retinal inner limiting membrane contraction. Label-free proteomics is an efficient method to provide further insight into the pathogenesis of vitreoretinal diseases.
Collapse
Affiliation(s)
- Qiaoling Wei
- Department of Ophthalmology, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Chen Jiang
- Department of Ophthalmology, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Xiaofeng Ye
- Department of Ophthalmology, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Xin Huang
- Department of Ophthalmology, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Hong Jin
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Gezhi Xu
- Department of Ophthalmology, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Hamadmad S, Shah MH, Kusibati R, Kim B, Erickson B, Heisler-Taylor T, Bhattacharya SK, Abdel-Rahman MH, Cebulla CM. Significant upregulation of small heat shock protein αA-crystallin in retinal detachment. Exp Eye Res 2019; 189:107811. [PMID: 31550446 DOI: 10.1016/j.exer.2019.107811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/21/2019] [Accepted: 09/18/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Sumaya Hamadmad
- The Ohio State University Department of Ophthalmology and Visual Science, Havener Eye Institute, The Ohio State University Wexner Medical Center, 915 Olentangy River Road, Columbus, OH, 43212, USA
| | - Mohd Hussain Shah
- The Ohio State University Department of Ophthalmology and Visual Science, Havener Eye Institute, The Ohio State University Wexner Medical Center, 915 Olentangy River Road, Columbus, OH, 43212, USA
| | - Rania Kusibati
- The Ohio State University Department of Ophthalmology and Visual Science, Havener Eye Institute, The Ohio State University Wexner Medical Center, 915 Olentangy River Road, Columbus, OH, 43212, USA
| | - Bongsu Kim
- The Ohio State University Department of Ophthalmology and Visual Science, Havener Eye Institute, The Ohio State University Wexner Medical Center, 915 Olentangy River Road, Columbus, OH, 43212, USA
| | - Brandon Erickson
- The Ohio State University Department of Ophthalmology and Visual Science, Havener Eye Institute, The Ohio State University Wexner Medical Center, 915 Olentangy River Road, Columbus, OH, 43212, USA
| | - Tyler Heisler-Taylor
- The Ohio State University Department of Ophthalmology and Visual Science, Havener Eye Institute, The Ohio State University Wexner Medical Center, 915 Olentangy River Road, Columbus, OH, 43212, USA
| | - Sanjoy K Bhattacharya
- Bascom Palmer Eye Institute, Department of Ophthalmology, The Miller School of Medicine, Miami, FL, 33136, USA
| | - Mohamed H Abdel-Rahman
- The Ohio State University Department of Ophthalmology and Visual Science, Havener Eye Institute, The Ohio State University Wexner Medical Center, 915 Olentangy River Road, Columbus, OH, 43212, USA; Division Human Genetics, The Ohio State University Wexner Medical Center, Columbus, OH, 43240, USA
| | | | - Colleen M Cebulla
- The Ohio State University Department of Ophthalmology and Visual Science, Havener Eye Institute, The Ohio State University Wexner Medical Center, 915 Olentangy River Road, Columbus, OH, 43212, USA.
| |
Collapse
|
15
|
Lennikov A, Saddala MS, Mukwaya A, Tang S, Huang H. Autoimmune-Mediated Retinopathy in CXCR5-Deficient Mice as the Result of Age-Related Macular Degeneration Associated Proteins Accumulation. Front Immunol 2019; 10:1903. [PMID: 31474986 PMCID: PMC6702970 DOI: 10.3389/fimmu.2019.01903] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 07/26/2019] [Indexed: 12/17/2022] Open
Abstract
Previous research has shown that CXCR5−/− mice develop retinal degeneration (RD) with age, a characteristic related to age macular degeneration (AMD). RD in these mice is not well-understood, and in this study, we sought to characterize further the RD phenotype and to gain mechanistic insights into the function of CXCR5 in the retina. CXCR5−/− and WT control mice were used. Fundus images demonstrated a significant (p < 0.001) increase of hypo-pigmented spots in the retina of aged CXCR5−/− mice compared with WT control mice. PAS staining indicated localization of deposits in the sub-retinal pigment epithelia (RPE) layer. AMD-associated proteins Cryab, amyloid beta, and C3d were detected within the RPE/sub-RPE tissues by immunofluorescence (IF). In addition, western blot analysis of COX-2, Arg1, and VEGF-a revealed an increase in the signaling of these molecules within the RPE/choroid complex. Transmission electron microscopy (TEM) indicated a drusen-like structure of sub-RPE deposits with an accumulation of vacuolated cellular debris. Loss of photoreceptors was detected by peanut lectin staining and was corroborated by a reduction in MAP2 signaling. Loss of blood-retinal barrier integrity was demonstrated by a reduction of ZO-1 expression. Inflammatory cells were detected in the sub-RPE space, with an increase in IBA-1 positive microglia cells on the surface of the RPE. Mass spectrometry analysis of CXCR5−/− mouse RPE/choroid proteins extracts, separated by SDS-page and incubated with autologous serum, identified autoantibodies against AMD-associated proteins: Cryaa, Cryab, and Anxa2. In vitro evaluations in BV-2 cell culture indicated a significant increase in production of Arg-1 (p < 0.001) and COX-2 (p < 0.01) in the presence of anti-CXCR5 antibody when compared with Igg-treated control BV-2 cells stimulated with IL-4 and TNFα/IFNγ, respectively. Anti-CXCR5 antibody treatment without stimulating agents did not affect Arg-1 and COX-2 expression; this suggests that CXCR5 may have a regulatory role in microglia cells activation. These results indicate that with age, CXCR5−/− mice develop RD characterized by microglia dysfunction, increased production of CXCL13 in the RPE progressive photoreceptor, neuronal loss, and sub-RPE deposition of cellular debris, resulting in the production of immunogenic proteins and autoimmune-mediated RD.
Collapse
Affiliation(s)
- Anton Lennikov
- Department of Ophthalmology, University of Missouri, Columbia, MO, United States.,Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Madhu Sudhana Saddala
- Department of Ophthalmology, University of Missouri, Columbia, MO, United States.,Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Anthony Mukwaya
- Department of Ophthalmology, Faculty of Health Sciences, Institute for Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Shibo Tang
- Aier School of Ophthalmology, Aier Eye Institute, Central South University, Changsha, China
| | - Hu Huang
- Department of Ophthalmology, University of Missouri, Columbia, MO, United States.,Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
16
|
On the origin of proteins in human drusen: The meet, greet and stick hypothesis. Prog Retin Eye Res 2018; 70:55-84. [PMID: 30572124 DOI: 10.1016/j.preteyeres.2018.12.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 12/12/2022]
Abstract
Retinal drusen formation is not only a clinical hallmark for the development of age-related macular degeneration (AMD) but also for other disorders, such as Alzheimer's disease and renal diseases. The initiation and growth of drusen is poorly understood. Attention has focused on lipids and minerals, but relatively little is known about the origin of drusen-associated proteins and how they are retained in the space between the basal lamina of the retinal pigment epithelium and the inner collagenous layer space (sub-RPE-BL space). While some authors suggested that drusen proteins are mainly derived from cellular debris from processed photoreceptor outer segments and the RPE, others suggest a choroidal cell or blood origin. Here, we reviewed and supplemented the existing literature on the molecular composition of the retina/choroid complex, to gain a more complete understanding of the sources of proteins in drusen. These "drusenomics" studies showed that a considerable proportion of currently identified drusen proteins is uniquely originating from the blood. A smaller, but still large fraction of drusen proteins comes from both blood and/or RPE. Only a small proportion of drusen proteins is uniquely derived from the photoreceptors or choroid. We next evaluated how drusen components may "meet, greet and stick" to each other and/or to structures like hydroxyapatite spherules to form macroscopic deposits in the sub-RPE-BL space. Finally, we discuss implications of our findings with respect to the previously proposed homology between drusenogenesis in AMD and plaque formation in atherosclerosis.
Collapse
|
17
|
Zayas-Santiago A, Ríos DS, Zueva LV, Inyushin MY. Localization of αA-Crystallin in Rat Retinal Müller Glial Cells and Photoreceptors. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2018; 24:545-552. [PMID: 30253817 PMCID: PMC6378655 DOI: 10.1017/s1431927618015118] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/17/2018] [Accepted: 08/18/2018] [Indexed: 06/01/2023]
Abstract
Transparent cells in the vertebrate optical tract, such as lens fiber cells and corneal epithelium cells, have specialized proteins that somehow permit only a low level of light scattering in their cytoplasm. It has been shown that both cell types contain (1) beaded intermediate filaments as well as (2) α-crystallin globulins. It is known that genetic and chemical alterations to these specialized proteins induce cytoplasmic opaqueness and visual complications. Crystallins were described previously in the retinal Müller cells of frogs. In the present work, using immunocytochemistry, fluorescence confocal imaging, and immuno-electron microscopy, we found that αA-crystallins are present in the cytoplasm of retinal Müller cells and in the photoreceptors of rats. Given that Müller glial cells were recently described as "living light guides" as were photoreceptors previously, we suggest that αA-crystallins, as in other highly transparent cells, allow Müller cells and photoreceptors to minimize intraretinal scattering during retinal light transmission.
Collapse
Affiliation(s)
- Astrid Zayas-Santiago
- Department of Pathology and Laboratory Medicine, Universidad Central del Caribe, Bayamón, PR 00960, USA
| | - David S. Ríos
- College of Science and Health Professions, Universidad Central de Bayamón, Bayamón, PR00960, USA
| | - Lidia V. Zueva
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St-Petersburg, Russia
| | - Mikhail Y. Inyushin
- Department of Physiology, Universidad Central del Caribe, Bayamón, PR 00960, USA
| |
Collapse
|
18
|
Ishikawa K, Sreekumar PG, Spee C, Nazari H, Zhu D, Kannan R, Hinton DR. αB-Crystallin Regulates Subretinal Fibrosis by Modulation of Epithelial-Mesenchymal Transition. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:859-73. [PMID: 26878210 PMCID: PMC4822331 DOI: 10.1016/j.ajpath.2015.11.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 10/20/2015] [Accepted: 11/19/2015] [Indexed: 01/18/2023]
Abstract
Subretinal fibrosis is an end stage of neovascular age-related macular degeneration, characterized by fibrous membrane formation after choroidal neovascularization. An initial step of the pathogenesis is an epithelial-mesenchymal transition (EMT) of retinal pigment epithelium cells. αB-crystallin plays multiple roles in age-related macular degeneration, including cytoprotection and angiogenesis. However, the role of αB-crystallin in subretinal EMT and fibrosis is unknown. Herein, we showed attenuation of subretinal fibrosis after regression of laser-induced choroidal neovascularization and a decrease in mesenchymal retinal pigment epithelium cells in αB-crystallin knockout mice compared with wild-type mice. αB-crystallin was prominently expressed in subretinal fibrotic lesions in mice. In vitro, overexpression of αB-crystallin induced EMT, whereas suppression of αB-crystallin induced a mesenchymal-epithelial transition. Transforming growth factor-β2-induced EMT was further enhanced by overexpression of αB-crystallin but was inhibited by suppression of αB-crystallin. Silencing of αB-crystallin inhibited multiple fibrotic processes, including cell proliferation, migration, and fibronectin production. Bone morphogenetic protein 4 up-regulated αB-crystallin, and its EMT induction was inhibited by knockdown of αB-crystallin. Furthermore, inhibition of αB-crystallin enhanced monotetraubiquitination of SMAD4, which can impair its nuclear localization. Overexpression of αB-crystallin enhanced nuclear translocation and accumulation of SMAD4 and SMAD5. Thus, αB-crystallin is an important regulator of EMT, acting as a molecular chaperone for SMAD4 and as its potential therapeutic target for preventing subretinal fibrosis development in neovascular age-related macular degeneration.
Collapse
Affiliation(s)
- Keijiro Ishikawa
- Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, California; Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | | | - Christine Spee
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Hossein Nazari
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Danhong Zhu
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Ram Kannan
- Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, California
| | - David R Hinton
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California; Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California.
| |
Collapse
|
19
|
Iannaccone A, Giorgianni F, New DD, Hollingsworth TJ, Umfress A, Alhatem AH, Neeli I, Lenchik NI, Jennings BJ, Calzada JI, Satterfield S, Mathews D, Diaz RI, Harris T, Johnson KC, Charles S, Kritchevsky SB, Gerling IC, Beranova-Giorgianni S, Radic MZ, Health ABC study. Circulating Autoantibodies in Age-Related Macular Degeneration Recognize Human Macular Tissue Antigens Implicated in Autophagy, Immunomodulation, and Protection from Oxidative Stress and Apoptosis. PLoS One 2015; 10:e0145323. [PMID: 26717306 PMCID: PMC4696815 DOI: 10.1371/journal.pone.0145323] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 12/01/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND We investigated sera from elderly subjects with and without age-related macular degeneration (AMD) for presence of autoantibodies (AAbs) against human macular antigens and characterized their identity. METHODS Sera were collected from participants in the Age-Related Maculopathy Ancillary (ARMA) Study, a cross-sectional investigation ancillary to the Health ABC Study, enriched with participants from the general population. The resulting sample (mean age: 79.2±3.9 years old) included subjects with early to advanced AMD (n = 131) and controls (n = 231). Sera were tested by Western blots for immunoreactive bands against human donor macular tissue homogenates. Immunoreactive bands were identified and graded, and odds ratios (OR) calculated. Based on these findings, sera were immunoprecipitated, and subjected to 2D gel electrophoresis (GE). Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to identify the targets recognized by circulating AAbs seen on 2D-GE, followed by ELISAs with recombinant proteins to confirm LC-MS/MS results, and quantify autoreactivities. RESULTS In AMD, 11 immunoreactive bands were significantly more frequent and 13 were significantly stronger than in controls. Nine of the more frequent bands also showed stronger reactivity. OR estimates ranged between 4.06 and 1.93, and all clearly excluded the null value. Following immunoprecipitation, 2D-GE and LC-MS/MS, five of the possible autoreactivity targets were conclusively identified: two members of the heat shock protein 70 (HSP70) family, HSPA8 and HSPA9; another member of the HSP family, HSPB4, also known as alpha-crystallin A chain (CRYAA); Annexin A5 (ANXA5); and Protein S100-A9, also known as calgranulin B that, when complexed with S100A8, forms calprotectin. ELISA testing with recombinant proteins confirmed, on average, significantly higher reactivities against all targets in AMD samples compared to controls. CONCLUSIONS Consistent with other evidence supporting the role of inflammation and the immune system in AMD pathogenesis, AAbs were identified in AMD sera, including early-stage disease. Identified targets may be mechanistically linked to AMD pathogenesis because the identified proteins are implicated in autophagy, immunomodulation, and protection from oxidative stress and apoptosis. In particular, a role in autophagy activation is shared by all five autoantigens, raising the possibility that the detected AAbs may play a role in AMD via autophagy compromise and downstream activation of the inflammasome. Thus, we propose that the detected AAbs provide further insight into AMD pathogenesis and have the potential to contribute to disease biogenesis and progression.
Collapse
Affiliation(s)
- Alessandro Iannaccone
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
- * E-mail:
| | - Francesco Giorgianni
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - David D. New
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - T. J. Hollingsworth
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Allison Umfress
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Albert H. Alhatem
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Indira Neeli
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Department of Microbiology, Immunology & Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Nataliya I. Lenchik
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Department of Internal Medicine/Endocrinology, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Barbara J. Jennings
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Jorge I. Calzada
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Charles Retina Institute, Memphis, TN, United States of America
| | - Suzanne Satterfield
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Dennis Mathews
- Eye Specialty Group, Memphis, TN, United States of America
- Southern College of Optometry, Memphis, TN, United States of America
| | - Rocio I. Diaz
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Charles Retina Institute, Memphis, TN, United States of America
| | - Tamara Harris
- National Institute on Aging, NIH, Bethesda, MD, United States of America
| | - Karen C. Johnson
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Steve Charles
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Charles Retina Institute, Memphis, TN, United States of America
| | - Stephen B. Kritchevsky
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Sticht Center on Aging, Wake Forest University, Winston-Salem, NC, United States of America
| | - Ivan C. Gerling
- Department of Internal Medicine/Endocrinology, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Sarka Beranova-Giorgianni
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Marko Z. Radic
- Department of Microbiology, Immunology & Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | | |
Collapse
|
20
|
Alpha crystallins in the retinal pigment epithelium and implications for the pathogenesis and treatment of age-related macular degeneration. Biochim Biophys Acta Gen Subj 2015; 1860:258-68. [PMID: 26026469 DOI: 10.1016/j.bbagen.2015.05.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/13/2015] [Accepted: 05/17/2015] [Indexed: 01/18/2023]
Abstract
BACKGROUND αA- and αB crystallins are principal members of the small heat shock protein family and elicit both a cell protective function and a chaperone function. α-Crystallins have been found to be prominent proteins in normal and pathological retina emphasizing the importance for in-depth understanding of their function and significance. SCOPE OF REVIEW Retinal pigment epithelial cells (RPE) play a vital role in the pathogenesis of age-related macular degeneration (AMD). This review addresses a number of cellular functions mediated by α-crystallins in the retina. Prominent expression of αB crystallin in mitochondria may serve to protect cells from oxidative injury. αB crystallin as secretory protein via exosomes can offer neuroprotection to adjacent RPE cells and photoreceptors. The availability of chaperone-containing minipeptides of αB crystallin could prove to be a valuable new tool for therapeutic treatment of retinal disorders. MAJOR CONCLUSIONS α-Crystallins are expressed in cytosol and mitochondria of RPE cells and are regulated during oxygen-induced retinopathy and during development. α-Crystallins protect RPE from oxidative-and ER stress-induced injury and autophagy. αB-Crystallin is a modulator of angiogenesis and vascular endothelial growth factor. αB Crystallin is secreted via exosomal pathway. Minichaperone peptides derived from αB Crystallin prevent oxidant induced cell death and have therapeutic potential. GENERAL SIGNIFICANCE Overall, this review summarizes several novel properties of α-crystallins and their relevance to maintaining normal retinal function. In particular, the use of α-crystallin derived peptides is a promising therapeutic strategy to combat retinal diseases such as AMD. This article is part of a Special Issue entitled Crystallin biochemistry in health and disease.
Collapse
|
21
|
Baba T, Oshitari T, Yamamoto S. Level of vitreous alpha-B crystallin in eyes with rhegmatogenous retinal detachment. Graefes Arch Clin Exp Ophthalmol 2014; 253:1251-4. [PMID: 25311653 DOI: 10.1007/s00417-014-2815-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/06/2014] [Accepted: 09/22/2014] [Indexed: 11/30/2022] Open
Abstract
PURPOSE Alpha-B crystallin (αB-crystallin) is a molecular chaperone, and one of the small heat shock proteins (sHSPs). αB-crystallin has been shown to have neuroprotective properties. The purpose of this study was to determine the level of αB-crystallin in eyes with a rhegmatogenous retinal detachment (RRD). METHODS Vitreous samples were collected before vitrectomy from 32 eyes of 32 consecutive patients with RRD. The concentration of αB-crystallin was measured by enzyme-linked immunosorbent assay (ELISA). Vitreous samples were also collected before vitrectomy from 16 patients with an idiopathic macular hole and epiretinal membrane, and used as controls. RESULTS The mean age was 56 years (range, 31-67 years) in the RRD cases and 66 years (range, 49-77 years) in the controls. The mean level of αB-crystallin was significantly higher in the RRD cases (36.7 ± 37.4 ng/ml) than that in controls (8.5 ± 12.2 ng/ml; P = 0.0028). CONCLUSIONS The increased level of αB-crystallin suggests that αB-crystallin may play a neuroprotective role by lessening retinal cell death after a retinal detachment.
Collapse
Affiliation(s)
- Takayuki Baba
- Department of Ophthalmology and Visual Science, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-0856, Japan,
| | | | | |
Collapse
|
22
|
Zhou P, Kannan R, Spee C, Sreekumar PG, Dou G, Hinton DR. Protection of retina by αB crystallin in sodium iodate induced retinal degeneration. PLoS One 2014; 9:e98275. [PMID: 24874187 PMCID: PMC4038555 DOI: 10.1371/journal.pone.0098275] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 04/29/2014] [Indexed: 12/21/2022] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness in the developed world. The retinal pigment epithelium (RPE) is a critical site of pathology in AMD and αB crystallin expression is increased in RPE and associated drusen in AMD. The purpose of this study was to investigate the role of αB crystallin in sodium iodate (NaIO3)-induced retinal degeneration, a model of AMD in which the primary site of pathology is the RPE. Dose dependent effects of intravenous NaIO3 (20-70 mg/kg) on development of retinal degeneration (fundus photography) and RPE and retinal neuronal loss (histology) were determined in wild type and αB crystallin knockout mice. Absence of αB crystallin augmented retinal degeneration in low dose (20 mg/kg) NaIO3-treated mice and increased retinal cell apoptosis which was mainly localized to the RPE layer. Generation of reactive oxygen species (ROS) was observed with NaIO3 in mouse and human RPE which increased further after αB crystallin knockout or siRNA knockdown, respectively. NaIO3 upregulated AKT phosphorylation and peroxisome proliferator–activator receptor–γ (PPARγ) which was suppressed after αB crystallin siRNA knockdown. Further, PPARγ ligand inhibited NaIO3-induced ROS generation. Our data suggest that αB crystallin plays a critical role in protection of NaIO3-induced oxidative stress and retinal degeneration in part through upregulation of AKT phosphorylation and PPARγ expression.
Collapse
Affiliation(s)
- Peng Zhou
- Doheny Eye Institute, Los Angeles, California, United States of America
| | - Ram Kannan
- Doheny Eye Institute, Los Angeles, California, United States of America
| | - Christine Spee
- Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | | | - Guorui Dou
- Doheny Eye Institute, Los Angeles, California, United States of America
| | - David R. Hinton
- Departments of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
- Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
23
|
Abstract
The scope of the current paper is to review existing and potential applications of proteomic analysis to aging research. The focus will lie on the unique opportunities of high-throughput studies for uncovering specific alterations in protein expression, protein complexes or protein modifications caused by biological aging. The result of such studies will outline aging phenotypes and potentially indicate pathways involved in the pathogenesis of age-associated disfunctions. Specific attention is paid to the illustrations of successful applications of proteomic technologies and potential applications of new proteomic concepts to biogerontological studies.
Collapse
Affiliation(s)
- Victor S Sharov
- University of Kansas, Pharmaceutical Chemistry Department, Lawrence, KS 66047, USA.
| | | |
Collapse
|
24
|
Renganathan K, Gu J, Rayborn ME, Crabb JS, Salomon RG, Collier RJ, Kapin MA, Romano C, Hollyfield JG, Crabb JW. CEP biomarkers as potential tools for monitoring therapeutics. PLoS One 2013; 8:e76325. [PMID: 24098476 PMCID: PMC3788138 DOI: 10.1371/journal.pone.0076325] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 08/18/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Carboxyethylpyrrole (CEP) adducts are oxidative modifications derived from docosahexaenoate-containing lipids that are elevated in ocular tissues and plasma in age-related macular degeneration (AMD) and in rodents exposed to intense light. The goal of this study was to determine whether light-induced CEP adducts and autoantibodies are modulated by pretreatment with AL-8309A under conditions that prevent photo-oxidative damage of rat retina. AL-8309A is a serotonin 5-HT1A receptor agonist. METHODS Albino rats were dark adapted prior to blue light exposure. Control rats were maintained in normal cyclic light. Rats were injected subcutaneously 3x with 10 mg/kg AL-8309A (2 days, 1 day and 0 hours) before light exposure for 6 h (3.1 mW/cm(2), λ=450 nm). Animals were sacrificed immediately following light exposure and eyes, retinas and plasma were collected. CEP adducts and autoantibodies were quantified by Western analysis or ELISA. RESULTS ANOVA supported significant differences in mean amounts of CEP adducts and autoantibodies among the light + vehicle, light + drug and dark control groups from both retina and plasma. Light-induced CEP adducts in retina were reduced ~20% following pretreatment with AL-8309A (n = 62 rats, p = 0.006) and retinal CEP immunoreactivity was less intense by immunohistochemistry. Plasma levels of light-induced CEP adducts were reduced at least 30% (n = 15 rats, p = 0.004) by drug pretreatment. Following drug treatment, average CEP autoantibody titer in light exposed rats (n = 22) was unchanged from dark control levels, and ~20% (p = 0.046) lower than in vehicle-treated rats. CONCLUSIONS Light-induced CEP adducts in rat retina and plasma were significantly decreased by pretreatment with AL-8309A. These results are consistent with and extend previous studies showing AL-8309A reduces light-induced retinal lesions in rats and support CEP biomarkers as possible tools for monitoring the efficacy of select therapeutics.
Collapse
Affiliation(s)
- Kutralanathan Renganathan
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
- Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Jiayin Gu
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
- Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Mary E. Rayborn
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
- Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
| | - John S. Crabb
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
- Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
| | - Robert G. Salomon
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Robert J. Collier
- Retina Research, Novartis Institutes for Biomedical Research, Fort Worth, Texas, United States of America
| | - Michael A. Kapin
- Retina Research, Novartis Institutes for Biomedical Research, Fort Worth, Texas, United States of America
| | - Carmelo Romano
- Retina Research, Novartis Institutes for Biomedical Research, Fort Worth, Texas, United States of America
| | - Joe G. Hollyfield
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
- Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
- Departments of Ophthalmology and Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, United States of America
| | - John W. Crabb
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
- Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
- Retina Research, Novartis Institutes for Biomedical Research, Fort Worth, Texas, United States of America
- Departments of Ophthalmology and Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
25
|
Murakami Y, Notomi S, Hisatomi T, Nakazawa T, Ishibashi T, Miller JW, Vavvas DG. Photoreceptor cell death and rescue in retinal detachment and degenerations. Prog Retin Eye Res 2013; 37:114-40. [PMID: 23994436 DOI: 10.1016/j.preteyeres.2013.08.001] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 08/08/2013] [Accepted: 08/10/2013] [Indexed: 02/08/2023]
Abstract
Photoreceptor cell death is the ultimate cause of vision loss in various retinal disorders, including retinal detachment (RD). Photoreceptor cell death has been thought to occur mainly through apoptosis, which is the most characterized form of programmed cell death. The caspase family of cysteine proteases plays a central role for inducing apoptosis, and in experimental models of RD, dying photoreceptor cells exhibit caspase activation; however, there is a paradox that caspase inhibition alone does not provide a sufficient protection against photoreceptor cell loss, suggesting that other mechanisms of cell death are involved. Recent accumulating evidence demonstrates that non-apoptotic forms of cell death, such as autophagy and necrosis, are also regulated by specific molecular machinery, such as those mediated by autophagy-related proteins and receptor-interacting protein kinases, respectively. Here we summarize the current knowledge of cell death signaling and its roles in photoreceptor cell death after RD and other retinal degenerative diseases. A body of studies indicate that not only apoptotic but also autophagic and necrotic signaling are involved in photoreceptor cell death, and that combined targeting of these pathways may be an effective neuroprotective strategy for retinal diseases associated with photoreceptor cell loss.
Collapse
Affiliation(s)
- Yusuke Murakami
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA; Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
26
|
Athanasiou D, Aguilà M, Bevilacqua D, Novoselov SS, Parfitt DA, Cheetham ME. The cell stress machinery and retinal degeneration. FEBS Lett 2013; 587:2008-17. [PMID: 23684651 PMCID: PMC4471140 DOI: 10.1016/j.febslet.2013.05.020] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 05/01/2013] [Accepted: 05/02/2013] [Indexed: 12/12/2022]
Abstract
Retinal degenerations are a group of clinically and genetically heterogeneous disorders characterised by progressive loss of vision due to neurodegeneration. The retina is a highly specialised tissue with a unique architecture and maintaining homeostasis in all the different retinal cell types is crucial for healthy vision. The retina can be exposed to a variety of environmental insults and stress, including light-induced damage, oxidative stress and inherited mutations that can lead to protein misfolding. Within retinal cells there are different mechanisms to cope with disturbances in proteostasis, such as the heat shock response, the unfolded protein response and autophagy. In this review, we discuss the multiple responses of the retina to different types of stress involved in retinal degenerations, such as retinitis pigmentosa, age-related macular degeneration and glaucoma. Understanding the mechanisms that maintain and re-establish proteostasis in the retina is important for developing new therapeutic approaches to fight blindness.
Collapse
Affiliation(s)
| | - Monica Aguilà
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Dalila Bevilacqua
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK
| | | | - David A. Parfitt
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK
| | | |
Collapse
|
27
|
Yao J, Liu X, Yang Q, Zhuang M, Wang F, Chen X, Hang H, Zhang W, Liu Q. Proteomic analysis of the aqueous humor in patients with wet age-related macular degeneration. Proteomics Clin Appl 2013; 7:550-60. [PMID: 23418058 DOI: 10.1002/prca.201200012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 12/20/2012] [Accepted: 01/07/2013] [Indexed: 12/12/2022]
Abstract
PURPOSE A number of studies have shown that the levels of some proteins in the aqueous humor (AH) are altered and correlate with the mechanisms or prognosis of many eye diseases. To identify the possible mechanisms that lead to the development of wet age-related macular degeneration (AMD), a proteomic analysis of the AH composition from wet AMD patients was performed and compared with that from non-AMD cataract patients. EXPERIMENTAL DESIGN Six wet AMD and six non-AMD cataract patients were enrolled. A proteomic approach which included two-dimensional electrophoresis coupled with MS and bioinformatics methods were used to identify AH proteins with altered expression in wet AMD compared with non-AMD patients. An ELISA was used to validate the proteomic results. RESULTS We separated 78 protein spots and identified 68 that were differently expressed in the wet AMD group and controls. Numerous proteins identified in this study are implicated in inflammation, apoptosis, angiogenesis, and oxidative stress. CONCLUSIONS AND CLINICAL RELEVANCE The AH protein composition was significantly different between wet AMD and non-AMD patients. The proteins identified in this study may be potential biomarkers of wet AMD development and might play a role in the mechanisms of wet AMD.
Collapse
Affiliation(s)
- Jiaqi Yao
- Department of Ophthalmology, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Sreekumar PG, Chothe P, Sharma KK, Baid R, Kompella U, Spee C, Kannan N, Manh C, Ryan SJ, Ganapathy V, Kannan R, Hinton DR. Antiapoptotic properties of α-crystallin-derived peptide chaperones and characterization of their uptake transporters in human RPE cells. Invest Ophthalmol Vis Sci 2013; 54:2787-98. [PMID: 23532520 PMCID: PMC3632268 DOI: 10.1167/iovs.12-11571] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 03/19/2013] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The chaperone proteins, α-crystallins, also possess antiapoptotic properties. The purpose of the present study was to investigate whether 19 to 20-mer α-crystallin-derived mini-chaperone peptides (α-crystallin mini-chaperone) are antiapoptotic, and to identify their putative transporters in human fetal RPE (hfRPE) cells. METHODS Cell death and caspase-3 activation induced by oxidative stress were quantified in early passage hfRPE cells in the presence of 19 to 20-mer αA- or αB-crystallin-derived or scrambled peptides. Cellular uptake of fluorescein-labeled, α-crystallin-derived mini-peptides and recombinant full-length αB-crystallin was determined in confluent hfRPE. The entry mechanism in hfRPE cells for α-crystallin mini-peptides was investigated. The protective role of polycaprolactone (PCL) nanoparticle encapsulated αB-crystallin mini-chaperone peptides from H2O2-induced cell death was studied. RESULTS Primary hfRPE cells exposed to oxidative stress and either αA- or αB-crystallin mini-chaperones remained viable and showed marked inhibition of both cell death and activation of caspase-3. Uptake of full-length αB-crystallin was minimal while a time-dependent uptake of αB-crystallin-derived peptide was observed. The mini-peptides entered the hfRPE cells via the sodium-coupled oligopeptide transporters 1 and 2 (SOPT1, SOPT2). PCL nanoparticles containing αB-crystallin mini-chaperone were also taken up and protected hfRPE from H2O2-induced cell death at significantly lower concentrations than free αB-crystallin mini-chaperone peptide. CONCLUSIONS αA- and αB-crystallin mini-chaperones offer protection to hfRPE cells and inhibit caspase-3 activation. The oligopeptide transporters SOPT1 and SOPT2 mediate the uptake of these peptides in RPE cells. Nanodelivery of αB-crystallin-derived mini-chaperone peptide offers an alternative approach for protection of hfRPE cells from oxidant injury.
Collapse
Affiliation(s)
| | - Paresh Chothe
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Health Sciences University, Augusta, Georgia
| | - Krishna K. Sharma
- Department of Ophthalmology, University of Missouri-Columbia School of Medicine, Columbia, Missouri
| | - Rinku Baid
- Departments of Pharmaceutical Sciences and Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Uday Kompella
- Departments of Pharmaceutical Sciences and Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Christine Spee
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Nandini Kannan
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Christina Manh
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Stephen J. Ryan
- Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, California
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Vadivel Ganapathy
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Health Sciences University, Augusta, Georgia
| | - Ram Kannan
- Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, California
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - David R. Hinton
- Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, California
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| |
Collapse
|
29
|
Bonilha VL, Shadrach KG, Rayborn ME, Li Y, Pauer GJT, Hagstrom SA, Bhattacharya SK, Hollyfield JG. Retinal deimination and PAD2 levels in retinas from donors with age-related macular degeneration (AMD). Exp Eye Res 2013; 111:71-8. [PMID: 23562679 DOI: 10.1016/j.exer.2013.03.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 03/21/2013] [Accepted: 03/22/2013] [Indexed: 12/17/2022]
Abstract
Deimination is a form of protein posttranslational modification carried out by the peptidyl arginine deiminases (PADs) enzymes. PAD2 is the principal deiminase expressed in the retina. Elevated levels of PAD2 and protein deimination are present in a number of human neurological diseases, with or without ocular manifestation. To define the association of deimination with the pathogenesis of age-related macular degeneration (AMD), we studied protein deimination and PAD2 levels in retinas of AMD donor eyes compared to age-matched non-AMD retinas. Eyes from non-AMD and AMD donors were fixed in 4% paraformaldehyde and 0.5% glutaraldehyde in phosphate buffer. Retina and retinal pigment epithelium (RPE) from donor eyes were processed for immunohistochemical detection and western blotting using antibodies to PAD2 and citrulline residues. The ganglion cell, inner plexiform, inner nuclear and outer nuclear layers were labeled by both PAD2 and citrulline antibodies. Changes in the localization of deiminated residues and PAD2 were evident as the retinal layers were remodeled coincident with photoreceptor degeneration in AMD retinas. Immunodetection of either PAD2 or citrulline residues could not be evaluated in the RPE layer due to the high autofluorescence levels in this layer. Interestingly, higher deimination immunoreactivity was detected in AMD retinal lysates. However, no significant changes in PAD2 were detected in the AMD and non-AMD retinas and RPE lysates. Our observations show increased levels of protein deimination but not PAD2 in AMD retinas and RPE, suggesting a reduced rate of turnover of deiminated proteins in these AMD retinas.
Collapse
Affiliation(s)
- Vera L Bonilha
- Department of Ophthalmology, The Cole Eye Institute(i31), Cleveland Clinic Lerner College of Medicine, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
McGreal RS, Brennan LA, Kantorow WL, Wilcox JD, Wei J, Chauss D, Kantorow M. Chaperone-independent mitochondrial translocation and protection by αB-crystallin in RPE cells. Exp Eye Res 2013; 110:10-7. [PMID: 23466869 DOI: 10.1016/j.exer.2013.02.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 02/12/2013] [Accepted: 02/19/2013] [Indexed: 11/16/2022]
Abstract
αB-crystallin is a small heat shock protein that exhibits chaperone activity and can protect multiple cell types against oxidative stress damage. Altered levels and specific mutations of αB-crystallin are associated with multiple degenerative diseases. We previously found that αB-crystallin translocates to lens and retinal cell mitochondria upon oxidative stress exposure where it provides protection against oxidative stress damage. To date, the role of the chaperone function of αB-crystallin in mitochondrial translocation and protection has not been established. Here, we sought to determine the relationship between the chaperone activity of αB-crystallin and its ability to translocate to and protect retinal cell mitochondria against oxidative stress damage. Our data provide evidence that three forms of αB-crystallin exhibiting different chaperone activity levels including wild-type, R120G (decreased chaperone activity) and M68A (increased chaperone activity) provide comparable levels of mitochondrial translocation and protection to retinal cells exposed to oxidative stress. The results provide evidence that mitochondrial translocation and protection by αB-crystallin is independent of its chaperone activity and that other functions of αB-crystallin may also be independent of its chaperone activity.
Collapse
Affiliation(s)
- Rebecca S McGreal
- Biomedical Sciences Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL 33431, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Hamann S, Métrailler S, Schorderet DF, Cottet S. Analysis of the cytoprotective role of α-crystallins in cell survival and implication of the αA-crystallin C-terminal extension domain in preventing Bax-induced apoptosis. PLoS One 2013; 8:e55372. [PMID: 23383327 PMCID: PMC3562314 DOI: 10.1371/journal.pone.0055372] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 12/21/2012] [Indexed: 12/17/2022] Open
Abstract
α-Crystallins, initially described as the major structural proteins of the lens, belong to the small heat shock protein family. Apart from their function as chaperones, α-crystallins are involved in the regulation of intracellular apoptotic signals. αA- and αB-crystallins have been shown to interfere with the mitochondrial apoptotic pathway triggering Bax pro-apoptotic activity and downstream activation of effector caspases. Differential regulation of α-crystallins has been observed in several eye diseases such as age-related macular degeneration and stress-induced and inherited retinal degenerations. Although the function of α-crystallins in healthy and diseased retina remains poorly understood, their altered expression in pathological conditions argue in favor of a role in cellular defensive response. In the Rpe65⁻/⁻ mouse model of Leber's congenital amaurosis, we previously observed decreased expression of αA- and αB-crystallins during disease progression, which was correlated with Bax pro-death activity and photoreceptor apoptosis. In the present study, we demonstrated that α-crystallins interacted with pro-apoptotic Bax and displayed cytoprotective action against Bax-triggered apoptosis, as assessed by TUNEL and caspase assays. We further observed in staurosporine-treated photoreceptor-like 661W cells stably overexpressing αA- or αB-crystallin that Bax-dependent apoptosis and caspase activation were inhibited. Finally, we reported that the C-terminal extension domain of αA-crystallin was sufficient to provide protection against Bax-triggered apoptosis. Altogether, these data suggest that α-crystallins interfere with Bax-induced apoptosis in several cell types, including the cone-derived 661W cells. They further suggest that αA-crystallin-derived peptides might be sufficient to promote cytoprotective action in response to apoptotic cell death.
Collapse
Affiliation(s)
- Séverine Hamann
- IRO, Institute for Research in Ophthalmology, Sion, Switzerland
- School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | | | - Daniel F. Schorderet
- IRO, Institute for Research in Ophthalmology, Sion, Switzerland
- School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Department of Ophthalmology, University of Lausanne, Lausanne, Switzerland
| | - Sandra Cottet
- IRO, Institute for Research in Ophthalmology, Sion, Switzerland
- Department of Ophthalmology, University of Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
32
|
|
33
|
Chan CC, Smith WM. Inflammatory Response and Mediators in Retinal Injury. Retina 2013. [DOI: 10.1016/b978-1-4557-0737-9.00025-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
34
|
Abstract
PURPOSE To examine the expression of alphaB-crystallin and its colocalization with vascular endothelial growth factor in the epiretinal membrane of human proliferative diabetic retinopathy. METHODS Ten epiretinal membranes of proliferative diabetic retinopathy and three normal retinas surgically excised were included in this study. Paraformaldehyde-fixed, paraffin-embedded tissue sections were processed for immunohistochemistry with alphaB-crystallin, vascular endothelial growth factor, and CD31 antibodies. RESULTS AlphaB-crystallin was expressed in all epiretinal membranes examined. The immunolocalization of alphaB-crystallin was detected in the cytoplasm of CD31-positive endothelial cells, but not in normal retinal blood vessels. Furthermore, alphaB-crystallin immunoreactivity was colocalized in vascular endothelial growth factor-positive endothelial cells in proliferative diabetic retinopathy membranes. CONCLUSION AlphaB-crystallin was expressed in proliferative diabetic retinopathy membranes, and colocalized with vascular endothelial growth factor-positive neovessels. AlphaB-crystallin may play a potential role in the pathogenesis of epiretinal membranes in proliferative diabetic retinopathy, together with vascular endothelial growth factor.
Collapse
|
35
|
Kannan R, Sreekumar PG, Hinton DR. Novel roles for α-crystallins in retinal function and disease. Prog Retin Eye Res 2012; 31:576-604. [PMID: 22721717 DOI: 10.1016/j.preteyeres.2012.06.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 05/31/2012] [Accepted: 06/04/2012] [Indexed: 01/18/2023]
Abstract
α-Crystallins are key members of the superfamily of small heat shock proteins that have been studied in detail in the ocular lens. Recently, novel functions for α-crystallins have been identified in the retina and in the retinal pigmented epithelium (RPE). αB-Crystallin has been localized to multiple compartments and organelles including mitochondria, golgi apparatus, endoplasmic reticulum and nucleus. α-Crystallins are regulated by oxidative and endoplasmic reticulum stress, and inhibit apoptosis-induced cell death. α-Crystallins interact with a large number of proteins that include other crystallins, and apoptotic, cytoskeletal, inflammatory, signaling, angiogenic, and growth factor molecules. Studies with RPE from αB-crystallin deficient mice have shown that αB-crystallin supports retinal and choroidal angiogenesis through its interaction with vascular endothelial growth factor. αB-Crystallin has also been shown to have novel functions in the extracellular space. In RPE, αB-crystallin is released from the apical surface in exosomes where it accumulates in the interphotoreceptor matrix and may function to protect neighboring cells. In other systems administration of exogenous recombinant αB-crystallin has been shown to be anti-inflammatory. Another newly described function of αB-crystallin is its ability to inhibit β-amyloid fibril formation. α-Crystallin minichaperone peptides have been identified that elicit anti-apoptotic function in addition to being efficient chaperones. Generation of liposomal particles and other modes of nanoencapsulation of these minipeptides could offer great therapeutic advantage in ocular delivery for a wide variety of retinal degenerative, inflammatory and vascular diseases including age-related macular degeneration and diabetic retinopathy.
Collapse
Affiliation(s)
- Ram Kannan
- Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, CA 90033, United States
| | | | | |
Collapse
|
36
|
Li S, Wu J, Ding H, Liao A, He H, Stell WK, Zhong X. Flicker downregulates the content of crystallin proteins in form-deprived C57BL/6 mouse retina. Exp Eye Res 2012; 101:1-8. [PMID: 22659691 DOI: 10.1016/j.exer.2012.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 04/16/2012] [Accepted: 05/16/2012] [Indexed: 10/28/2022]
Abstract
Image degradation by loss of higher spatial frequencies causes form-deprivation myopia (FDM) in humans and animals, and cyclical illumination (flicker) at certain frequencies may prevent FDM. The molecular mechanisms underlying FDM and its prevention by flicker are poorly known. To understand them better, we have identified proteins that differ in amount in form-deprived (FD) mouse retinas, under steady versus flickering light. Male C57BL/6 mice (age 27-29 days) were randomly divided into three groups: Experimental - monocularly form-deprived, and kept under either normal room light ("FD-Only") or 20 Hz flickering light ("FD-Flicker"), throughout the 12-hour light phase; and Control ("Open-Control") - kept under normal illumination, without form deprivation. After two weeks of treatment, retinal proteins were extracted and separated by two-dimensional gel electrophoresis (2D-GE); proteins that differ in content in FD-only versus FD-flicker retinas were identified by mass spectroscopy ("MS"), and their identities were verified by western blotting. The contents of three identified proteins differed statistically in FD-only compared to FD-flicker retinas. These proteins were identified by MS as α-A-crystallin, crystallin β A2 and crystallin β A1. Quantitative western blotting showed that the relative amount of α-A-crystallin in FD-only retinas was significantly higher than that in FD-Flicker and control retinas. In conclusion, form deprivation induced significant increases in the amounts of crystallins in mouse retinas. These increases were significantly reduced by exposure to 20 Hz flicker. Since form deprivation is known to induce myopia development, and flicker to prevent it, our data suggest that FD- and flicker-responsive changes in the content of crystallin proteins may be involved causally or protectively in myopia development.
Collapse
Affiliation(s)
- Saiqun Li
- Zhongshan Ophthalmic Center and State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou 510060, China
| | | | | | | | | | | | | |
Collapse
|
37
|
McGreal RS, Kantorow WL, Chauss DC, Wei J, Brennan LA, Kantorow M. αB-crystallin/sHSP protects cytochrome c and mitochondrial function against oxidative stress in lens and retinal cells. Biochim Biophys Acta Gen Subj 2012; 1820:921-30. [PMID: 22521365 DOI: 10.1016/j.bbagen.2012.04.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 03/20/2012] [Accepted: 04/05/2012] [Indexed: 12/12/2022]
Abstract
BACKGROUND αB-crystallin/sHSP protects cells against oxidative stress damage. Here, we mechanistically examined its ability to preserve mitochondrial function in lens and retinal cells and protect cytochrome c under oxidative stress conditions. METHODS αB-crystallin/sHSP was localized in human lens (HLE-B3) and retinal (ARPE-19) cells. αB-crystallin/sHSP was stably over-expressed and its ability to preserve mitochondrial membrane potential under oxidative stress conditions was monitored. Interactions between αB-crystallin/sHSP and cytochrome c were examined by fluorescent resonance energy transfer (FRET) and by co-immune precipitation. The ability of αB-crystallin/sHSP to protect cytochrome c against methionine-80 oxidation was monitored. RESULTS αB-crystallin/sHSP is present in the mitochondria of lens and retinal cells and is translocated to the mitochondria under oxidative conditions. αB-crystallin/sHSP specifically interacts with cytochrome c in vitro and in vivo and its overexpression preserves mitochondrial membrane potential under oxidative stress conditions. αB-crystallin/sHSP directly protects cytochrome c against oxidation. GENERAL SIGNIFICANCE These data demonstrate that αB-crystallin/sHSP maintains lens and retinal cells under oxidative stress conditions at least in part by preserving mitochondrial function and by protecting cytochrome c against oxidation. Since oxidative stress and loss of mitochondrial function are associated with eye lens cataract and age-related macular degeneration, loss of these αB-crystallin/sHSP functions likely plays a key role in the development of these diseases. αB-crystallin/sHSP is expressed throughout the body and its ability to maintain mitochondrial function is likely important for the prevention of multiple degenerative diseases.
Collapse
Affiliation(s)
- Rebecca S McGreal
- Biomedical Sciences Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | | | | | | | | | | |
Collapse
|
38
|
Organisciak D, Wong P, Rapp C, Darrow R, Ziesel A, Rangarajan R, Lang J. Light-induced retinal degeneration is prevented by zinc, a component in the age-related eye disease study formulation. Photochem Photobiol 2012; 88:1396-407. [PMID: 22385127 DOI: 10.1111/j.1751-1097.2012.01134.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Mineral supplements are often included in multivitamin preparations because of their beneficial effects on metabolism. In this study, we used an animal model of light-induced retinal degeneration to test for photoreceptor cell protection by the essential trace element zinc. Rats were treated with various doses of zinc oxide and then exposed to intense visible light for as long as 8 h. Zinc treatment effectively prevented retinal light damage as determined by rhodopsin and retinal DNA recovery, histology and electrophoretic analysis of DNA damage and oxidized retinal proteins. Zinc oxide was particularly effective when given before light exposure and at doses two- to four-fold higher than recommended by the age-related eye disease study group. Treated rats exhibited higher serum and retinal pigment epithelial zinc levels and an altered retinal gene expression profile. Using an Ingenuity database, 512 genes with known functional annotations were found to be responsive to zinc supplementation, with 45% of these falling into a network related to cellular growth, proliferation, cell cycle and death. Although these data suggest an integrated and extensive regulatory response, zinc induced changes in gene expression also appear to enhance antioxidative capacity in retina and reduce oxidative damage arising from intense light exposure.
Collapse
Affiliation(s)
- Daniel Organisciak
- Petticrew Research Laboratory, Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA.
| | | | | | | | | | | | | |
Collapse
|
39
|
Ng TK, Liang XY, Pang CP. HTRA1 in Age-Related Macular Degeneration. ASIA-PACIFIC JOURNAL OF OPHTHALMOLOGY (PHILADELPHIA, PA.) 2012; 1:51-63. [PMID: 26107018 DOI: 10.1097/apo.0b013e31823e57fe] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Age-related macular degeneration (AMD) is a leading cause of severe visual impairment and irreversible blindness in most developed countries, affecting more than 50 million of elderly people worldwide. Current treatments, such as intravitreal injection of antiangiogenic agents, mitigate the effect of advanced AMD but cannot completely cure the disease. Comprehensive understanding of the AMD pathological mechanisms is important for the development of new therapies. Previously, we identified a single-nucleotide polymorphism (rs11200638) in the promoter region of the high temperature requirement factor A1 (HTRA1) gene on chromosome 10q26 to be associated with exudative AMD. In further biological studies, we have provided evidence that HTRA1 could be a potential disease-causing gene within the 10q26 locus. In this review, we summarize the pathology of AMD and the molecular function of the HtrA1 protein. Also evaluated are the genetic effects of HTRA1 polymorphism on AMD in different populations and interactions with other AMD-associated genes, especially with the complement factor H (CFH) gene, which was identified for nonexudative AMD. The biological roles of HtrA1 are exhaustively examined on its contribution to the multifactorial pathogenic mechanism of AMD. Although HtrA1 should play a part in AMD pathogenesis, a host of other genetic and environmental factors, known and unknown, is involved and warrants intensive future research.
Collapse
Affiliation(s)
- Tsz Kin Ng
- From the Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, China
| | | | | |
Collapse
|
40
|
Sreekumar PG, Hinton DR, Kannan R. Methionine sulfoxide reductase A: Structure, function and role in ocular pathology. World J Biol Chem 2011; 2:184-92. [PMID: 21909460 PMCID: PMC3163237 DOI: 10.4331/wjbc.v2.i8.184] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 07/27/2011] [Accepted: 08/03/2011] [Indexed: 02/05/2023] Open
Abstract
Methionine is a highly susceptible amino acid that can be oxidized to S and R diastereomeric forms of methionine sulfoxide by many of the reactive oxygen species generated in biological systems. Methionine sulfoxide reductases (Msrs) are thioredoxin-linked enzymes involved in the enzymatic conversion of methionine sulfoxide to methionine. Although MsrA and MsrB have the same function of methionine reduction, they differ in substrate specificity, active site composition, subcellular localization, and evolution. MsrA has been localized in different ocular regions and is abundantly expressed in the retina and in retinal pigment epithelial (RPE) cells. MsrA protects cells from oxidative stress. Overexpression of MsrA increases resistance to cell death, while silencing or knocking down MsrA decreases cell survival; events that are mediated by mitochondria. MsrA participates in protein-protein interaction with several other cellular proteins. The interaction of MsrA with α-crystallins is of utmost importance given the known functions of the latter in protein folding, neuroprotection, and cell survival. Oxidation of methionine residues in α-crystallins results in loss of chaperone function and possibly its antiapoptotic properties. Recent work from our laboratory has shown that MsrA is co-localized with αA and αB crystallins in the retinal samples of patients with age-related macular degeneration. We have also found that chemically induced hypoxia regulates the expression of MsrA and MsrB2 in human RPE cells. Thus, MsrA is a critical enzyme that participates in cell and tissue protection, and its interaction with other proteins/growth factors may provide a target for therapeutic strategies to prevent degenerative diseases.
Collapse
Affiliation(s)
- Parameswaran G Sreekumar
- Parameswaran G Sreekumar, David R Hinton, Ram Kannan, Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, CA 90033, United States
| | | | | |
Collapse
|
41
|
Organisciak D, Darrow R, Barsalou L, Rapp C, McDonald B, Wong P. Light induced and circadian effects on retinal photoreceptor cell crystallins. Photochem Photobiol 2010; 87:151-9. [PMID: 21091955 DOI: 10.1111/j.1751-1097.2010.00844.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Crystallins in the retina may serve a chaperone-like protective function. In this study we measured mRNA levels for alpha-, beta- and gamma-crystallins in rat retinas following treatment with potentially damaging levels of light. We also determined crystallin protein patterns in photoreceptor cell rod outer segments (ROSs) isolated from rats exposed to intense light. Weanling albino rats were maintained in a dim cyclic light environment or in darkness for 40days. At P60 animals were treated with intense visible light, for as long as 8h, beginning at various times of the day or night. Retinas were excised immediately after light treatment and used for quantitative RT-PCR, or to prepare ROSs for western analysis. Some eyes were frozen in OCT for crystallin immunohistochemistry. Intense light exposure led to increases in mRNA expression for all retinal crystallins and to changes in ROS crystallin immunoreactivity. These light-induced changes were found to depend on the time of day that exposure started, duration of light treatment and previous light rearing history. We suggest that crystallin synthesis in retina exhibits a dependence on both light stress and circadian rhythm and that within photoreceptor cells crystallins appear to migrate in a light-independent, circadian fashion.
Collapse
Affiliation(s)
- Daniel Organisciak
- Petticrew Research Laboratory, Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine Wright State University, Dayton, OH, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Sturgill GM, Bala E, Yaniglos SS, Peachey NS, Hagstrom SA. Mutation screen of beta-crystallin genes in 274 patients with age-related macular degeneration. Ophthalmic Genet 2010; 31:129-34. [PMID: 20565250 DOI: 10.3109/13816810.2010.486774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE The crystallin family of proteins comprise the main structural proteins of the vertebrate lens and have been classified into alpha-, beta-, and gamma- families. Several of the beta-crystallin proteins have been detected in the retina where they are each localized to different compartments of rod and cone photoreceptors. Functionally, beta-crystallins have been implicated in the protection of the retina from intense light exposure. Two members of the beta-crystallins, CRYBB1 and CRYBB2, have been identified in drusen preparations isolated from the retina of donor eyes of patients with age-related macular degeneration (AMD), the leading cause of blindness in the elderly population of developed countries. We therefore investigated CRYBB1 and CRYBB2 as candidate genes for AMD in 274 unrelated patients. RESULTS A mutation screen of the entire coding region of the CRYBB1gene uncovered eight sequence variations, including three missense changes, two intronic changes and three isocoding changes. A mutation screen of the entire coding region of the CRYBB2 gene uncovered three sequence variations, one isocoding change and two intronic changes. CONCLUSIONS Although variant alleles of the CRYBB1 and CRYBB2 genes were found, none are considered pathogenic.
Collapse
Affiliation(s)
- Gwen M Sturgill
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, Ohio, USA
| | | | | | | | | |
Collapse
|
43
|
αB crystallin is apically secreted within exosomes by polarized human retinal pigment epithelium and provides neuroprotection to adjacent cells. PLoS One 2010; 5:e12578. [PMID: 20949024 PMCID: PMC2951891 DOI: 10.1371/journal.pone.0012578] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Accepted: 08/08/2010] [Indexed: 12/20/2022] Open
Abstract
αB Crystallin is a chaperone protein with anti-apoptotic and anti-inflammatory functions and has been identified as a biomarker in age-related macular degeneration. The purpose of this study was to determine whether αB crystallin is secreted from retinal pigment epithelial (RPE) cells, the mechanism of this secretory pathway and to determine whether extracellular αB crystallin can be taken up by adjacent retinal cells and provide protection from oxidant stress. We used human RPE cells to establish that αB crystallin is secreted by a non-classical pathway that involves exosomes. Evidence for the release of exosomes by RPE and localization of αB crystallin within the exosomes was achieved by immunoblot, immunofluorescence, and electron microscopic analyses. Inhibition of lipid rafts or exosomes significantly reduced αB crystallin secretion, while inhibitors of classic secretory pathways had no effect. In highly polarized RPE monolayers, αB crystallin was selectively secreted towards the apical, photoreceptor-facing side. In support, confocal microscopy established that αB crystallin was localized predominantly in the apical compartment of RPE monolayers, where it co-localized in part with exosomal marker CD63. Severe oxidative stress resulted in barrier breakdown and release of αB crystallin to the basolateral side. In normal mouse retinal sections, αB crystallin was identified in the interphotoreceptor matrix. An increased uptake of exogenous αB crystallin and protection from apoptosis by inhibition of caspase 3 and PARP activation were observed in stressed RPE cultures. αB Crystallin was taken up by photoreceptors in mouse retinal explants exposed to oxidative stress. These results demonstrate an important role for αB crystallin in maintaining and facilitating a neuroprotective outer retinal environment and may also explain the accumulation of αB crystallin in extracellular sub-RPE deposits in the stressed microenvironment in age-related macular degeneration. Thus evidence from our studies supports a neuroprotective role for αB crystallin in ocular diseases.
Collapse
|
44
|
Yuan X, Gu X, Crabb JS, Yue X, Shadrach K, Hollyfield JG, Crabb JW. Quantitative proteomics: comparison of the macular Bruch membrane/choroid complex from age-related macular degeneration and normal eyes. Mol Cell Proteomics 2010; 9:1031-46. [PMID: 20177130 PMCID: PMC2877969 DOI: 10.1074/mcp.m900523-mcp200] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 01/19/2010] [Indexed: 11/29/2022] Open
Abstract
A quantitative proteomics analysis of the macular Bruch membrane/choroid complex was pursued for insights into the molecular mechanisms of age-related macular degeneration (AMD). Protein in trephine samples from the macular region of 10 early/mid-stage dry AMD, six advanced dry AMD, eight wet AMD, and 25 normal control post-mortem eyes was analyzed by LC MS/MS iTRAQ (isobaric tags for relative and absolute quantitation) technology. A total of 901 proteins was quantified, including 556 proteins from > or =3 AMD samples. Most proteins differed little in amount between AMD and control samples and therefore reflect the proteome of normal macular tissues of average age 81. A total of 56 proteins were found to be elevated and 43 were found to be reduced in AMD tissues relative to controls. Analysis by category of disease progression revealed up to 16 proteins elevated or decreased in each category. About 60% of the elevated proteins are involved in immune response and host defense, including many complement proteins and damage-associated molecular pattern proteins such as alpha-defensins 1-3, protein S100s, crystallins, histones, and galectin-3. Four retinoid processing proteins were elevated only in early/mid-stage AMD, supporting a role for retinoids in AMD initiation. Proteins uniquely decreased in early/mid-stage AMD implicate hematologic malfunctions and weakened extracellular matrix integrity and cellular interactions. Galectin-3, a receptor for advanced glycation end products, was the most significantly elevated protein in advanced dry AMD, supporting a role for advanced glycation end products in dry AMD progression. The results endorse inflammatory processes in both early and advanced AMD pathology, implicate different pathways of progression to advanced dry and wet AMD, and provide a new database for hypothesis-driven and discovery-based studies of AMD.
Collapse
Affiliation(s)
- Xianglin Yuan
- From the ‡Cole Eye Institute and
- §Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195
| | - Xiaorong Gu
- From the ‡Cole Eye Institute and
- §Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195
| | - John S. Crabb
- From the ‡Cole Eye Institute and
- §Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195
| | - Xiuzhen Yue
- From the ‡Cole Eye Institute and
- §Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195
| | - Karen Shadrach
- From the ‡Cole Eye Institute and
- §Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195
| | - Joe G. Hollyfield
- From the ‡Cole Eye Institute and
- §Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195
- ¶Departments of Ophthalmology and Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio 44106
| | - John W. Crabb
- From the ‡Cole Eye Institute and
- §Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195
- ‖Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, and
- ¶Departments of Ophthalmology and Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio 44106
| |
Collapse
|
45
|
Hollyfield JG, Perez VL, Salomon RG. A hapten generated from an oxidation fragment of docosahexaenoic acid is sufficient to initiate age-related macular degeneration. Mol Neurobiol 2010; 41:290-8. [PMID: 20221855 DOI: 10.1007/s12035-010-8110-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Accepted: 02/15/2010] [Indexed: 01/16/2023]
Abstract
The protein adduct carboxyethylpyrrole (CEP) is present in age-related macular degeneration (AMD) eye tissue and in the blood of AMD patients at higher levels than found in age-matched non-AMD tissues. Autoantibodies to CEP are also higher in AMD blood samples than in controls. To test the hypothesis that this hapten is causally involved in initiating an inflammatory response in AMD, we immunized C57BL/6J mice with mouse serum albumin (MSA) adducted with CEP. Immunized mice develop antibodies to CEP, fix complement component-3 in Bruch's membrane, accumulate drusen below the retinal pigment epithelium during aging, show decreased a- and b-wave amplitudes in response to light, and develop lesions in the retinal pigment epithelium mimicking geographic atrophy, the blinding end-stage condition characteristic of the dry form of AMD. Inflammatory cells are present in the region of lesions and may be actively involved in the pathology observed. We conclude that early immunization of mice with CEP-adducted MSA sensitizes these animals to the ongoing production of CEP adducts in the outer retina where DHA is abundant and the conditions for oxidative damage are permissive. In response to this early sensitization, the immune system mounts a complement-mediated attack on the cells of the outer retina where CEP adducts are formed. This animal model for AMD is the first that was developed from an inflammatory signal discovered in eye tissue and blood from AMD patients. It provides a novel opportunity for dissecting the early pathology of AMD and the immune response contributing to this disorder. The availability of a mouse with a mechanistically based AMD-like disease that progresses rapidly is highly desirable. Such a model will allow for the efficient preclinical testing of the much-needed therapeutics quickly and inexpensively.
Collapse
Affiliation(s)
- Joe G Hollyfield
- Cole Eye Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA.
| | | | | |
Collapse
|
46
|
Brennan LA, Lee W, Giblin FJ, David LL, Kantorow M. Methionine sulfoxide reductase A (MsrA) restores alpha-crystallin chaperone activity lost upon methionine oxidation. Biochim Biophys Acta Gen Subj 2009; 1790:1665-72. [PMID: 19733220 DOI: 10.1016/j.bbagen.2009.08.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Revised: 08/05/2009] [Accepted: 08/26/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND Lens cataract is associated with protein oxidation and aggregation. Two proteins that cause cataract when deleted from the lens are methionine sulfoxide reductase A (MsrA) that repairs protein methionine sulfoxide (PMSO) oxidized proteins and alpha-crystallin which is a two-subunit (alphaA and alphaB) chaperone. Here, we tested whether PMSO formation damages alpha-crystallin chaperone function and whether MsrA could repair PMSO-alpha-crystallin. METHODS Total alpha-crystallin was oxidized to PMSO and evaluated by CNBr-cleavage and mass spectrometry. Chaperone activity was measured by light scattering using lysozyme as target. PMSO-alpha-crystallin was treated with MsrA, and repair was assessed by CNBr cleavage, mass spectrometry and recovery of chaperone function. The levels of alpha-crystallin-PMSO in the lenses of MsrA-knockout relative to wild-type mice were determined. RESULTS PMSO oxidation of total alpha-crystallin (met 138 of alphaA and met 68 of alphaB) resulted in loss of alpha-crystallin chaperone activity. MsrA treatment of PMSO-alpha-crystallin repaired its chaperone activity through reduction of PMSO. Deletion of MsrA in mice resulted in increased levels of PMSO-alpha-crystallin. CONCLUSIONS Methionine oxidation damages alpha-crystallin chaperone function and MsrA can repair PMSO-alpha-crystallin restoring its chaperone function. MsrA is required for maintaining the reduced state of alpha-crystallin methionines in the lens. SIGNIFICANCE Methionine oxidation of alpha-crystallin in combination with loss of MsrA repair causes loss of alpha-crystallin chaperone function. Since increased PMSO levels and loss of alpha-crystallin function are hallmarks of cataract, these results provide insight into the mechanisms of cataract development and likely those of other age-related diseases.
Collapse
Affiliation(s)
- Lisa A Brennan
- Biomedical Sciences Department, Charles E. Schmidt College of Biomedical Science, Florida Atlantic University, Boca Raton, FL, USA
| | | | | | | | | |
Collapse
|
47
|
Chen L, Holland GN, Yu F, Levinson RD, Lampi KJ, Horwitz J, Gordon LK. Associations of seroreactivity against crystallin proteins with disease activity and cataract in patients with uveitis. Invest Ophthalmol Vis Sci 2008; 49:4476-81. [PMID: 18539933 DOI: 10.1167/iovs.08-1971] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE betaB1-crystallin is a putative target of an autoantibody observed in a subset of patients with uveitis. The purpose of this study was to determine whether seroreactivity against betaB1 or other specific purified crystallin proteins is observed in patients with uveitis and whether this reactivity is associated with either cataract or active intraocular inflammation. METHODS Sera from patients with uveitis were tested for IgG antibodies with reactivity against alphaA-, alphaB-, betaB1-, or betaB2-crystallin proteins using a modified slot-blot protocol. Ophthalmic evaluations included analysis of the degree of intraocular inflammation and assessment of lens opacity by the Lens Opacities Classification System (LOCS) III. Positive anti-crystallin reactivity was defined as greater than the mean + 2 SD of the reactivity of a commercially available control serum panel. Statistical analysis was performed with the Fisher exact test, Kruskal-Wallis test, and Student's t-test. RESULTS IgG antibodies against alphaA-, alphaB-, or betaB1-crystallin were identified in 70% of 39 subjects; in contrast, only 30% of the control sera exhibited reactivity against one or more of these crystallin proteins (P <or= 0.01). Seroreactivity against alphaA-, alphaB-, or betaB1-, but not betaB2-crystallin was related to active anterior segment inflammation. Seroreactivity against alphaB and betaB1 was significantly related to cortical cataract (P <or= 0.05). CONCLUSIONS Serum antibodies against specific crystallin proteins are present in most patients with uveitis. The relationship between the presence of specific anti-crystallin antibodies and active inflammation may indicate a role for these autoantibodies in uveitis pathogenesis.
Collapse
Affiliation(s)
- Ling Chen
- Departments of 2Ophthalmology, Ocular Inflammatory Disease Center, Jules Stein Eye Institute, University of California, Los Angeles, California 90095-7000, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
As the retinal pigment epithelium (RPE) ages, a number of structural changes occur, including loss of melanin granules, increase in the density of residual bodies, accumulation of lipofuscin, accumulation of basal deposits on or within Bruch's membrane, formation of drusen (between the basal lamina of the RPE and the inner collagenous layer of Bruch's membrane), thickening of Bruch's membrane, microvilli atrophy and disorganization of the basal infoldings. Although these changes are well known, the basic mechanisms involved in them are frequently poorly understood. These age-related changes progress slowly and vary in severity in different individuals. These changes are also found in age-related macular degeneration (AMD), a late onset disease that severely impacts the RPE, but they are much more pronounced than during normal aging. However, the changes in AMD lead to severe loss of vision. Given the many supporting functions which the RPE serves for the retina, it is important to decipher the age-related changes in this epithelium in order to understand age-related changes in vision.
Collapse
Affiliation(s)
- Vera L Bonilha
- Cole Eye Institute, The Cleveland Clinic, Cleveland, 9500 Euclid Avenue, OH, USA.
| |
Collapse
|
49
|
Penn JS, Madan A, Caldwell RB, Bartoli M, Caldwell RW, Hartnett ME. Vascular endothelial growth factor in eye disease. Prog Retin Eye Res 2008; 27:331-71. [PMID: 18653375 DOI: 10.1016/j.preteyeres.2008.05.001] [Citation(s) in RCA: 563] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Collectively, angiogenic ocular conditions represent the leading cause of irreversible vision loss in developed countries. In the US, for example, retinopathy of prematurity, diabetic retinopathy and age-related macular degeneration are the principal causes of blindness in the infant, working age and elderly populations, respectively. Evidence suggests that vascular endothelial growth factor (VEGF), a 40kDa dimeric glycoprotein, promotes angiogenesis in each of these conditions, making it a highly significant therapeutic target. However, VEGF is pleiotropic, affecting a broad spectrum of endothelial, neuronal and glial behaviors, and confounding the validity of anti-VEGF strategies, particularly under chronic disease conditions. In fact, among other functions VEGF can influence cell proliferation, cell migration, proteolysis, cell survival and vessel permeability in a wide variety of biological contexts. This article will describe the roles played by VEGF in the pathogenesis of retinopathy of prematurity, diabetic retinopathy and age-related macular degeneration. The potential disadvantages of inhibiting VEGF will be discussed, as will the rationales for targeting other VEGF-related modulators of angiogenesis.
Collapse
Affiliation(s)
- J S Penn
- Vanderbilt University School of Medicine, Nashville, TN, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Proteomics as a research tool in clinical and experimental ophthalmology. Proteomics Clin Appl 2008; 2:762-75. [DOI: 10.1002/prca.200780094] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|