1
|
Panigrahi T, Khamar P, Shetty R, Kannan R, Ashok N, Nishtala K, Ghosh A, Deshpande V. Longitudinal analysis of wound healing response post SMILE and LASIK surgery using proteomic profiling of tears. Exp Eye Res 2024; 246:109987. [PMID: 38964497 DOI: 10.1016/j.exer.2024.109987] [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: 02/26/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
Different types of refractive surgeries often exhibit differences in wound healing responses. The current study investigated post-operative tear protein profiles in subjects who underwent LASIK and SMILE to elucidate global changes to the proteomic profile during the period the patient cornea undergoes healing. In this study, 10 patients underwent LASIK and SMILE surgery with a contralateral paired eye design. Tear samples were collected using Schirmer's strips preoperatively, at 1 month, 3 months and 6 months postoperatively. Quantitative ITRAQ labeled proteomics was performed and the tear protein ratios were normalized to pre-operative protein levels for each subject. Whole proteomics identified 1345 proteins in tears from LASIK and 1584 proteins in SMILE across time points. About 67 proteins were common in LASIK and SMILE tears across all the time points. Wound healing responses were differentially regulated between two refractive surgeries (SMILE and LASIK). The proteins Ceruloplasmin, Clusterin, Serotransferrin were upregulated at 1 month and 3 months and downregulated at 6 months post operatively in LASIK surgery where as in SMILE these were downregulated. Galectin 3 binding protein showed upregulation at 1 month and the levels decreased at 3 months and 6 months postop in LASIK tears whereas the levels increased at 3 months and 6 months post-op in SMILE tears. The levels of proteins that protect from oxidative stress were higher in SMILE as compared to LASIK postoperatively. The extracellular matrix proteins showed an increase in expression at 6 months in SMILE tears and was stabilized at 6 months in LASIK tears post operatively. Different refractive surgeries induce distinct wound healing responses as identified in tears. This study has implications in targeting key proteins for improving the clinical outcome postrefractive surgery.
Collapse
Affiliation(s)
| | - Pooja Khamar
- Cornea and Refractive Services, Narayana Nethralaya, Bangalore, India
| | - Rohit Shetty
- Cornea and Refractive Services, Narayana Nethralaya, Bangalore, India
| | - Ramaraj Kannan
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| | - Nikhil Ashok
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| | | | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India.
| | - Vrushali Deshpande
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India.
| |
Collapse
|
2
|
Jin L, Zhang L, Yan C, Liu M, Dean DC, Liu Y. Corneal injury repair and the potential involvement of ZEB1. EYE AND VISION (LONDON, ENGLAND) 2024; 11:20. [PMID: 38822380 PMCID: PMC11143703 DOI: 10.1186/s40662-024-00387-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 05/07/2024] [Indexed: 06/03/2024]
Abstract
The cornea, consisting of three cellular and two non-cellular layers, is the outermost part of the eyeball and frequently injured by external physical, chemical, and microbial insults. The epithelial-to-mesenchymal transition (EMT) plays a crucial role in the repair of corneal injuries. Zinc finger E-box binding homeobox 1 (ZEB1), an important transcription factor involved in EMT, is expressed in the corneal tissues. It regulates cell activities like migration, transformation, and proliferation, and thereby affects tissue inflammation, fibrosis, tumor metastasis, and necrosis by mediating various major signaling pathways, including transforming growth factor (TGF)-β. Dysfunction of ZEB1 would impair corneal tissue repair leading to epithelial healing delay, interstitial fibrosis, neovascularization, and squamous cell metaplasia. Understanding the mechanism underlying ZEB1 regulation of corneal injury repair will help us to formulate a therapeutic approach to enhance corneal injury repair.
Collapse
Affiliation(s)
- Lin Jin
- Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, 116033, China
| | - Lijun Zhang
- Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, 116033, China
| | - Chunxiao Yan
- Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, 116033, China
| | - Mengxin Liu
- Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, 116033, China
| | - Douglas C Dean
- James Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| | - Yongqing Liu
- James Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| |
Collapse
|
3
|
Pan J, Pany S, Martinez-Carrasco R, Fini ME. Differential Efficacy of Small Molecules Dynasore and Mdivi-1 for the Treatment of Dry Eye Epitheliopathy or as a Countermeasure for Nitrogen Mustard Exposure of the Ocular Surface. J Pharmacol Exp Ther 2024; 388:506-517. [PMID: 37442618 PMCID: PMC10801785 DOI: 10.1124/jpet.123.001697] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/19/2023] [Accepted: 06/05/2023] [Indexed: 07/15/2023] Open
Abstract
The ocular surface comprises the wet mucosal epithelia of the cornea and conjunctiva, the associated glands, and the overlying tear film. Epitheliopathy is the common pathologic outcome when the ocular surface is subjected to oxidative stress. Whether different stresses act via the same or different mechanisms is not known. Dynasore and dyngo-4a, small molecules developed to inhibit the GTPase activity of classic dynamins DNM1, DNM2, and DNM3, but not mdivi-1, a specific inhibitor of DNM1L, protect corneal epithelial cells exposed to the oxidant tert-butyl hydroperoxide (tBHP). Here we report that, while dyngo-4a is the more potent inhibitor of endocytosis, dynasore is the better cytoprotectant. Dynasore also protects corneal epithelial cells against exposure to high salt in an in vitro model of dysfunctional tears in dry eye. We now validate this finding in vivo, demonstrating that dynasore protects against epitheliopathy in a mouse model of dry eye. Knockdown of classic dynamin DNM2 was also cytoprotective against tBHP exposure, suggesting that dynasore's effect is at least partially on target. Like tBHP and high salt, exposure of corneal epithelial cells to nitrogen mustard upregulated the unfolded protein response and inflammatory markers, but dynasore did not protect against nitrogen mustard exposure. In contrast, mdivi-1 was cytoprotective. Interestingly, mdivi-1 did not inhibit the nitrogen mustard-induced expression of inflammatory cytokines. We conclude that exposure to tBHP or nitrogen mustard, two different oxidative stress agents, cause corneal epitheliopathy via different pathologic pathways. SIGNIFICANCE STATEMENT: Results presented in this paper, for the first time, implicate the dynamin DNM2 in ocular surface epitheliopathy. The findings suggest that dynasore could serve as a new topical treatment for dry eye epitheliopathy and that mdivi-1 could serve as a medical countermeasure for epitheliopathy due to nitrogen mustard exposure, with potentially increased efficacy when combined with anti-inflammatory agents and/or UPR modulators.
Collapse
Affiliation(s)
- Jinhong Pan
- New England Eye Center, Tufts Medical Center and Department of Ophthalmology, Tufts University School of Medicine (J.P., S.P., R.M.-C., M.E.F.) and Program in Pharmacology and Drug Development, Tufts Graduate School of Biomedical Sciences (M.E.F.), Tufts University, Boston, Massachusetts
| | - Satyabrata Pany
- New England Eye Center, Tufts Medical Center and Department of Ophthalmology, Tufts University School of Medicine (J.P., S.P., R.M.-C., M.E.F.) and Program in Pharmacology and Drug Development, Tufts Graduate School of Biomedical Sciences (M.E.F.), Tufts University, Boston, Massachusetts
| | - Rafael Martinez-Carrasco
- New England Eye Center, Tufts Medical Center and Department of Ophthalmology, Tufts University School of Medicine (J.P., S.P., R.M.-C., M.E.F.) and Program in Pharmacology and Drug Development, Tufts Graduate School of Biomedical Sciences (M.E.F.), Tufts University, Boston, Massachusetts
| | - M Elizabeth Fini
- New England Eye Center, Tufts Medical Center and Department of Ophthalmology, Tufts University School of Medicine (J.P., S.P., R.M.-C., M.E.F.) and Program in Pharmacology and Drug Development, Tufts Graduate School of Biomedical Sciences (M.E.F.), Tufts University, Boston, Massachusetts
| |
Collapse
|
4
|
Schicht M, Farger J, Wedel S, Sisignano M, Scholich K, Geisslinger G, Perumal N, Grus FH, Singh S, Sahin A, Paulsen F, Lütjen-Drecoll E. Ocular surface changes in mice with streptozotocin-induced diabetes and diabetic polyneuropathy. Ocul Surf 2024; 31:43-55. [PMID: 38141818 DOI: 10.1016/j.jtos.2023.12.006] [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: 09/09/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 12/25/2023]
Abstract
PURPOSE Diabetes mellitus (DM) is a leading risk factor for corneal neuropathy and dry eye disease (DED). Another common consequence of DM is diabetic peripheral polyneuropathy (DPN). Both complications affect around 50 % of the DM patients but the relationship between DM, DED and DPN remains unclear. METHODS In this study, we examined mice with early onset of DM and PN after streptozotocin (STZ)-induced diabetes (DPN). We compared the early morphological changes of the sciatic nerve, dorsal root and trigeminal ganglia with the changes in the ocular surface, including tear proteomic and we also investigated respective changes in the gene expressions and morphological alterations in the eye tissues involved in tear production. RESULTS The lacrimal gland, conjunctival goblet cells and cornea showed morphological changes along with alterations in tear proteins without any obvious signs of ocular surface inflammation. The gene expression for respectively altered tear proteins i.e., of Clusterin in cornea, Car6, Adh3a1, and Eef1a1 in eyelids, and Pigr in the lacrimal gland also showed significant changes compared to control mice. In the trigeminal ganglia like in the dorsal root ganglia neuronal cells showed swollen mitochondria and, in the latter, there was a significant increase of NADPH oxidases and MMP9 suggestive of oxidative and neuronal stress. In the dorsal root ganglia and the sciatic nerve, there was an upregulation of a number of pro-inflammatory cytokines and pain-mediating chemokines. CONCLUSION The early ocular changes in DM Mice only affect the lacrimal gland. Which, is reflected in the tear film composition of DPN mice. Due to the high protein concentration in tear fluid in humans, proteomic analysis in addition to noninvasive investigation of goblet cells and cornea can serve as a tools for the early diagnosis of DPN, DED in clinical practice. Early treatment could delay or even prevent the ocular complications of DM such as DED and PN.
Collapse
Affiliation(s)
- Martin Schicht
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| | - Jessica Farger
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Saskia Wedel
- Institute of Clinical Pharmacology, Goethe University, Frankfurt am Main, Germany
| | - Marco Sisignano
- Institute of Clinical Pharmacology, Goethe University, Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Department of Experimental and Translational Ophthalmology, Germany
| | - Klaus Scholich
- Institute of Clinical Pharmacology, Goethe University, Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Department of Experimental and Translational Ophthalmology, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Goethe University, Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Department of Experimental and Translational Ophthalmology, Germany
| | - Natarajan Perumal
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Franz H Grus
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Swati Singh
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Afsun Sahin
- Department of Ophthalmology, Koc University Medical School, Istanbul, Turkey
| | - Friedrich Paulsen
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Elke Lütjen-Drecoll
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
5
|
Loiseau A, Raîche-Marcoux G, Maranda C, Bertrand N, Boisselier E. Animal Models in Eye Research: Focus on Corneal Pathologies. Int J Mol Sci 2023; 24:16661. [PMID: 38068983 PMCID: PMC10706114 DOI: 10.3390/ijms242316661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 09/27/2023] [Accepted: 11/19/2023] [Indexed: 12/18/2023] Open
Abstract
The eye is a complex sensory organ that enables visual perception of the world. The dysfunction of any of these tissues can impair vision. Conduction studies on laboratory animals are essential to ensure the safety of therapeutic products directly applied or injected into the eye to treat ocular diseases before eventually proceeding to clinical trials. Among these tissues, the cornea has unique homeostatic and regenerative mechanisms for maintaining transparency and refraction of external light, which are essential for vision. However, being the outermost tissue of the eye and directly exposed to the external environment, the cornea is particularly susceptible to injury and diseases. This review highlights the evidence for selecting appropriate animals to better understand and treat corneal diseases, which rank as the fifth leading cause of blindness worldwide. The development of reliable and human-relevant animal models is, therefore, a valuable research tool for understanding and translating fundamental mechanistic findings, as well as for assessing therapeutic potential in humans. First, this review emphasizes the unique characteristics of animal models used in ocular research. Subsequently, it discusses current animal models associated with human corneal pathologies, their utility in understanding ocular disease mechanisms, and their role as translational models for patients.
Collapse
Affiliation(s)
- Alexis Loiseau
- Faculty of Medicine, Department of Ophthalmology and Otolaryngology—Head and Neck Surgery, CHU de Québec Research Center, Université Laval, Québec, QC G1S 4L8, Canada; (G.R.-M.); (C.M.)
| | - Gabrielle Raîche-Marcoux
- Faculty of Medicine, Department of Ophthalmology and Otolaryngology—Head and Neck Surgery, CHU de Québec Research Center, Université Laval, Québec, QC G1S 4L8, Canada; (G.R.-M.); (C.M.)
| | - Cloé Maranda
- Faculty of Medicine, Department of Ophthalmology and Otolaryngology—Head and Neck Surgery, CHU de Québec Research Center, Université Laval, Québec, QC G1S 4L8, Canada; (G.R.-M.); (C.M.)
| | - Nicolas Bertrand
- Faculty of Pharmacy, CHU de Quebec Research Center, Université Laval, Québec, QC G1V 4G2, Canada;
| | - Elodie Boisselier
- Faculty of Medicine, Department of Ophthalmology and Otolaryngology—Head and Neck Surgery, CHU de Québec Research Center, Université Laval, Québec, QC G1S 4L8, Canada; (G.R.-M.); (C.M.)
| |
Collapse
|
6
|
Gross C, Guérin LP, Socol BG, Germain L, Guérin SL. The Ins and Outs of Clusterin: Its Role in Cancer, Eye Diseases and Wound Healing. Int J Mol Sci 2023; 24:13182. [PMID: 37685987 PMCID: PMC10488069 DOI: 10.3390/ijms241713182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Clusterin (CLU) is a glycoprotein originally discovered in 1983 in ram testis fluid. Rapidly observed in other tissues, it was initially given various names based on its function in different tissues. In 1992, it was finally named CLU by consensus. Nearly omnipresent in human tissues, CLU is strongly expressed at fluid-tissue interfaces, including in the eye and in particular the cornea. Recent research has identified different forms of CLU, with the most prominent being a 75-80 kDa heterodimeric protein that is secreted. Another truncated version of CLU (55 kDa) is localized to the nucleus and exerts pro-apoptotic activities. CLU has been reported to be involved in various physiological processes such as sperm maturation, lipid transportation, complement inhibition and chaperone activity. CLU was also reported to exert important functions in tissue remodeling, cell-cell adhesion, cell-substratum interaction, cytoprotection, apoptotic cell death, cell proliferation and migration. Hence, this protein is sparking interest in tissue wound healing. Moreover, CLU gene expression is finely regulated by cytokines, growth factors and stress-inducing agents, leading to abnormally elevated levels of CLU in many states of cellular disturbance, including cancer and neurodegenerative conditions. In the eye, CLU expression has been reported as being severely increased in several pathologies, such as age-related macular degeneration and Fuch's corneal dystrophy, while it is depleted in others, such as pathologic keratinization. Nevertheless, the precise role of CLU in the development of ocular pathologies has yet to be deciphered. The question of whether CLU expression is influenced by these disorders or contributes to them remains open. In this article, we review the actual knowledge about CLU at both the protein and gene expression level in wound healing, and explore the possibility that CLU is a key factor in cancer and eye diseases. Understanding the expression and regulation of CLU could lead to the development of novel therapeutics for promoting wound healing.
Collapse
Affiliation(s)
- Christelle Gross
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec City, QC G1V 0A6, Canada; (C.G.); (B.G.S.); (L.G.)
- Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Québec City, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | | | - Bianca G. Socol
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec City, QC G1V 0A6, Canada; (C.G.); (B.G.S.); (L.G.)
| | - Lucie Germain
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec City, QC G1V 0A6, Canada; (C.G.); (B.G.S.); (L.G.)
- Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Québec City, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
- Département de Chirurgie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Sylvain L. Guérin
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec City, QC G1V 0A6, Canada; (C.G.); (B.G.S.); (L.G.)
- Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Québec City, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| |
Collapse
|
7
|
Chintala SK, Pan J, Satapathy S, Condruti R, Hao Z, Liu PW, O’Conner CF, Barr JT, Wilson MR, Jeong S, Fini ME. Recombinant Human Clusterin Seals Damage to the Ocular Surface Barrier in a Mouse Model of Ophthalmic Preservative-Induced Epitheliopathy. Int J Mol Sci 2023; 24:981. [PMID: 36674497 PMCID: PMC9861099 DOI: 10.3390/ijms24020981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/23/2022] [Accepted: 12/31/2022] [Indexed: 01/07/2023] Open
Abstract
There is a significant unmet need for therapeutics to treat ocular surface barrier damage, also called epitheliopathy, due to dry eye and related diseases. We recently reported that the natural tear glycoprotein CLU (clusterin), a molecular chaperone and matrix metalloproteinase inhibitor, seals and heals epitheliopathy in mice subjected to desiccating stress in a model of aqueous-deficient/evaporative dry eye. Here we investigated CLU sealing using a second model with features of ophthalmic preservative-induced dry eye. The ocular surface was stressed by topical application of the ophthalmic preservative benzalkonium chloride (BAC). Then eyes were treated with CLU and sealing was evaluated immediately by quantification of clinical dye uptake. A commercial recombinant form of human CLU (rhCLU), as well as an rhCLU form produced in our laboratory, designed to be compatible with U.S. Food and Drug Administration guidelines on current Good Manufacturing Practices (cGMP), were as effective as natural plasma-derived human CLU (pCLU) in sealing the damaged ocular surface barrier. In contrast, two other proteins found in tears: TIMP1 and LCN1 (tear lipocalin), exhibited no sealing activity. The efficacy and selectivity of rhCLU for sealing of the damaged ocular surface epithelial barrier suggests that it could be of therapeutic value in treating BAC-induced epitheliopathy and related diseases.
Collapse
Affiliation(s)
- Shravan K. Chintala
- USC Institute for Genetic Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| | - Jinhong Pan
- New England Eye Center, Tufts Medical Center, Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Sandeep Satapathy
- School of Chemistry and Molecular Bioscience, Molecular Horizons Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Rebecca Condruti
- Training Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Zixuan Hao
- Training Program in Pharmacology and Drug Development, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Pei-wen Liu
- Training Program in Pharmacology and Drug Development, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Christian F. O’Conner
- Doctor of Medicine Training Program, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Joseph T. Barr
- The Ohio State University College of Optometry, Columbus, OH 43210, USA
| | - Mark R. Wilson
- School of Chemistry and Molecular Bioscience, Molecular Horizons Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Shinwu Jeong
- USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| | - M. Elizabeth Fini
- New England Eye Center, Tufts Medical Center, Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
8
|
Wilson MR, Satapathy S, Jeong S, Fini ME. Clusterin, other extracellular chaperones, and eye disease. Prog Retin Eye Res 2022; 89:101032. [PMID: 34896599 PMCID: PMC9184305 DOI: 10.1016/j.preteyeres.2021.101032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022]
Abstract
Proteostasis refers to all the processes that maintain the correct expression level, location, folding and turnover of proteins, essential to organismal survival. Both inside cells and in body fluids, molecular chaperones play key roles in maintaining proteostasis. In this article, we focus on clusterin, the first-recognized extracellular mammalian chaperone, and its role in diseases of the eye. Clusterin binds to and inhibits the aggregation of proteins that are misfolded due to mutations or stresses, clears these aggregating proteins from extracellular spaces, and facilitates their degradation. Clusterin exhibits three main homeostatic activities: proteostasis, cytoprotection, and anti-inflammation. The so-called "protein misfolding diseases" are caused by aggregation of misfolded proteins that accumulate pathologically as deposits in tissues; we discuss several such diseases that occur in the eye. Clusterin is typically found in these deposits, which is interpreted to mean that its capacity as a molecular chaperone to maintain proteostasis is overwhelmed in the disease state. Nevertheless, the role of clusterin in diseases involving such deposits needs to be better defined before therapeutic approaches can be entertained. A more straightforward case can be made for therapeutic use of clusterin based on its proteostatic role as a proteinase inhibitor, as well as its cytoprotective and anti-inflammatory properties. It is likely that clusterin works together in this way with other extracellular chaperones to protect the eye from disease, and we discuss several examples. We end this article by predicting future steps that may lead to development of clusterin as a biological drug.
Collapse
Affiliation(s)
- Mark R Wilson
- Molecular Horizons and the School of Chemistry and Molecular Bioscience, University of Wollongong; Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, New South Wales, 2522, Australia.
| | - Sandeep Satapathy
- Molecular Horizons and the School of Chemistry and Molecular Bioscience, University of Wollongong; Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, New South Wales, 2522, Australia.
| | - Shinwu Jeong
- USC Roski Eye Institute and Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, 1333 San Pablo Street., Los Angeles, CA, 90033, USA.
| | - M Elizabeth Fini
- New England Eye Center, Tufts Medical Center and Department of Ophthalmology, Tufts University School of Medicine; Program in Pharmacology & Drug Development, Graduate School of Biomedical Sciences, Tufts University, 800 Washington St, Boston, MA, 02111, USA.
| |
Collapse
|
9
|
López-Cano JJ, González-Cela-Casamayor MA, Andrés-Guerrero V, Vicario-de-la-Torre M, Benítez Del Castillo JM, Herrero-Vanrell R, Molina-Martínez IT. Development of an osmoprotective microemulsion as a therapeutic platform for ocular surface protection. Int J Pharm 2022; 623:121948. [PMID: 35752388 DOI: 10.1016/j.ijpharm.2022.121948] [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: 03/27/2022] [Revised: 06/03/2022] [Accepted: 06/19/2022] [Indexed: 10/17/2022]
Abstract
Self-emulsified osmoprotective ophthalmic microemulsions (O/A) were prepared by combining betaine/leucine, clusterin/oleanolic acid, and hyaluronic acid or Dextran. The microemulsions contained an internal oily phase (1.2%), an external aqueous phase (96.3%), cosolvents (1%), and surfactants (1.5%). Physicochemical characterization and in vivo and in vitro tolerance were analyzed. The formulations' osmoprotective in vitro activity was assayed in a hyperosmolar model in human corneal cells. Average internal phase sizes were 16-26 nm for the microemulsions including Dextran. Addition of hyaluronic acid increased the size range (25-39 nm). Addition of osmoprotectants did not change nanodroplet size. The formulations were isotonic (280-290 mOsm/L) with neutral pH (≈7) and zeta potential (-10 to 0 mV), low surface tension (≈35-40mN·m-1), and low viscosity (≈1 mPa·s), except for the microemulsions containing hyaluronic acid (≈4-5 mPa·s). SEM and cryo-TEM showed that all formulations exhibited sphere-shaped morphology with good cell tolerance (≈100%) and were stable at 8 °C for 9 months. Osmoprotective formulations were well tolerated in vitro and in vivo, protecting cells from hypertonic stress. We therefore developed stable microemulsions compatible with the ocular surface that could constitute a novel tool for treatment of ophthalmic diseases.
Collapse
Affiliation(s)
- J J López-Cano
- Ophthalmology Innovation, Therapy and Pharmaceutical Development (InnOftal) Research Group, UCM 920415, Department of Pharmaceutics and Food Technology, Faculty of Pharmacy (UCM), Plaza Ramón y Cajal s/n, Madrid 28040, Spain; National Ocular Pathology Network (OFTARED), Carlos III Health Institute, San Carlos Clinical Hospital Health Research Institute (IdISSC), Madrid 28040, Spain
| | - M A González-Cela-Casamayor
- Ophthalmology Innovation, Therapy and Pharmaceutical Development (InnOftal) Research Group, UCM 920415, Department of Pharmaceutics and Food Technology, Faculty of Pharmacy (UCM), Plaza Ramón y Cajal s/n, Madrid 28040, Spain; National Ocular Pathology Network (OFTARED), Carlos III Health Institute, San Carlos Clinical Hospital Health Research Institute (IdISSC), Madrid 28040, Spain
| | - V Andrés-Guerrero
- Ophthalmology Innovation, Therapy and Pharmaceutical Development (InnOftal) Research Group, UCM 920415, Department of Pharmaceutics and Food Technology, Faculty of Pharmacy (UCM), Plaza Ramón y Cajal s/n, Madrid 28040, Spain; National Ocular Pathology Network (OFTARED), Carlos III Health Institute, San Carlos Clinical Hospital Health Research Institute (IdISSC), Madrid 28040, Spain
| | - M Vicario-de-la-Torre
- Ophthalmology Innovation, Therapy and Pharmaceutical Development (InnOftal) Research Group, UCM 920415, Department of Pharmaceutics and Food Technology, Faculty of Pharmacy (UCM), Plaza Ramón y Cajal s/n, Madrid 28040, Spain; National Ocular Pathology Network (OFTARED), Carlos III Health Institute, San Carlos Clinical Hospital Health Research Institute (IdISSC), Madrid 28040, Spain
| | - J M Benítez Del Castillo
- National Ocular Pathology Network (OFTARED), Carlos III Health Institute, San Carlos Clinical Hospital Health Research Institute (IdISSC), Madrid 28040, Spain; Ocular Surface and Inflammation Unit, Ophthalmology Department, San Carlos Clinical Hospital Health Research Institute (IdISSC), Madrid 28040, Spain
| | - R Herrero-Vanrell
- Ophthalmology Innovation, Therapy and Pharmaceutical Development (InnOftal) Research Group, UCM 920415, Department of Pharmaceutics and Food Technology, Faculty of Pharmacy (UCM), Plaza Ramón y Cajal s/n, Madrid 28040, Spain; National Ocular Pathology Network (OFTARED), Carlos III Health Institute, San Carlos Clinical Hospital Health Research Institute (IdISSC), Madrid 28040, Spain.
| | - I T Molina-Martínez
- Ophthalmology Innovation, Therapy and Pharmaceutical Development (InnOftal) Research Group, UCM 920415, Department of Pharmaceutics and Food Technology, Faculty of Pharmacy (UCM), Plaza Ramón y Cajal s/n, Madrid 28040, Spain; National Ocular Pathology Network (OFTARED), Carlos III Health Institute, San Carlos Clinical Hospital Health Research Institute (IdISSC), Madrid 28040, Spain.
| |
Collapse
|
10
|
Marozzi M, Parnigoni A, Negri A, Viola M, Vigetti D, Passi A, Karousou E, Rizzi F. Inflammation, Extracellular Matrix Remodeling, and Proteostasis in Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms22158102. [PMID: 34360868 PMCID: PMC8346982 DOI: 10.3390/ijms22158102] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/11/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer is a multifaceted and complex pathology characterized by uncontrolled cell proliferation and decreased apoptosis. Most cancers are recognized by an inflammatory environment rich in a myriad of factors produced by immune infiltrate cells that induce host cells to differentiate and to produce a matrix that is more favorable to tumor cells’ survival and metastasis. As a result, the extracellular matrix (ECM) is changed in terms of macromolecules content, degrading enzymes, and proteins. Altered ECM components, derived from remodeling processes, interact with a variety of surface receptors triggering intracellular signaling that, in turn, cancer cells exploit to their own benefit. This review aims to present the role of different aspects of ECM components in the tumor microenvironment. Particularly, we highlight the effect of pro- and inflammatory factors on ECM degrading enzymes, such as metalloproteases, and in a more detailed manner on hyaluronan metabolism and the signaling pathways triggered by the binding of hyaluronan with its receptors. In addition, we sought to explore the role of extracellular chaperones, especially of clusterin which is one of the most prominent in the extracellular space, in proteostasis and signaling transduction in the tumor microenvironment. Although the described tumor microenvironment components have different biological roles, they may engage common signaling pathways that favor tumor growth and metastasis.
Collapse
Affiliation(s)
- Marina Marozzi
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43125 Parma, Italy; (M.M.); (A.N.); (F.R.)
| | - Arianna Parnigoni
- Department of Medicine and Surgery, University of Insubria, Via J.H. Dunant 5, 21100 Varese, Italy; (A.P.); (M.V.); (D.V.); (A.P.)
| | - Aide Negri
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43125 Parma, Italy; (M.M.); (A.N.); (F.R.)
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, Via J.H. Dunant 5, 21100 Varese, Italy; (A.P.); (M.V.); (D.V.); (A.P.)
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, Via J.H. Dunant 5, 21100 Varese, Italy; (A.P.); (M.V.); (D.V.); (A.P.)
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Via J.H. Dunant 5, 21100 Varese, Italy; (A.P.); (M.V.); (D.V.); (A.P.)
| | - Evgenia Karousou
- Department of Medicine and Surgery, University of Insubria, Via J.H. Dunant 5, 21100 Varese, Italy; (A.P.); (M.V.); (D.V.); (A.P.)
- Correspondence:
| | - Federica Rizzi
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43125 Parma, Italy; (M.M.); (A.N.); (F.R.)
| |
Collapse
|
11
|
de Campos TDP, da Cruz Rodrigues KC, Pereira RM, Anaruma CP, Dos Santos Canciglieri R, de Melo DG, da Silva ASR, Cintra DE, Ropelle ER, Pauli JR, de Moura LP. The protective roles of clusterin in ocular diseases caused by obesity and diabetes mellitus type 2. Mol Biol Rep 2021; 48:4637-4645. [PMID: 34036481 DOI: 10.1007/s11033-021-06419-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/17/2021] [Indexed: 11/26/2022]
Abstract
Obesity is a chronic, non-transmissible and multifactorial disease commonly associated with systemic inflammation and damage to health. This disorder has been pointed out as leading to the development of a diversity of eye diseases and, consequently, damage to visual acuity. More specifically, cardiometabolic risk is associated with lacrimal gland dysfunctions, since it changes the inflammatory profile favoring the development and worsening of dry eye disease. In more severe and extreme cases, obesity, inflammation, and diabetes mellitus type 2 can trigger the total loss of vision. In this scenario, besides its numerous metabolic functions, clusterin, an apolipoprotein, has been described as protective to the ocular surface through the seal mechanism. Thus, the current review aimed to explain the role of clusterin in dry eye disease that can be triggered by obesity and diabetes.
Collapse
Affiliation(s)
- Thaís Dantis Pereira de Campos
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas, 1300 Pedro Zaccaria Street, Limeira, SP, Brazil
| | - Kellen Cristina da Cruz Rodrigues
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas, 1300 Pedro Zaccaria Street, Limeira, SP, Brazil
| | - Rodrigo Martins Pereira
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas, 1300 Pedro Zaccaria Street, Limeira, SP, Brazil
| | - Chadi Pellegrini Anaruma
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas, 1300 Pedro Zaccaria Street, Limeira, SP, Brazil
| | - Raphael Dos Santos Canciglieri
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas, 1300 Pedro Zaccaria Street, Limeira, SP, Brazil
| | - Diego Gomes de Melo
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas, 1300 Pedro Zaccaria Street, Limeira, SP, Brazil
| | | | - Dennys Esper Cintra
- Laboratory of Nutritional Genomics (LABGeN), School of Applied Sciences, University of Campinas, Limeira, SP, Brazil
- CEPECE - Center of Research in Sport Sciences. School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas, 1300 Pedro Zaccaria Street, Limeira, SP, Brazil
- CEPECE - Center of Research in Sport Sciences. School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas, 1300 Pedro Zaccaria Street, Limeira, SP, Brazil
- CEPECE - Center of Research in Sport Sciences. School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, Brazil
| | - Leandro Pereira de Moura
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas, 1300 Pedro Zaccaria Street, Limeira, SP, Brazil.
- CEPECE - Center of Research in Sport Sciences. School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, Brazil.
- Postgraduate Program in Motricity Sciences, São Paulo State University (UNESP), São Paulo, Brazil.
| |
Collapse
|
12
|
Shoari A, Kanavi MR, Rasaee MJ. Inhibition of matrix metalloproteinase-9 for the treatment of dry eye syndrome; a review study. Exp Eye Res 2021; 205:108523. [PMID: 33662353 DOI: 10.1016/j.exer.2021.108523] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 02/06/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022]
Abstract
Dry eye syndrome (DES) and tear dysfunction are multifactorial conditions affecting meibomian glands, lacrimal glands, and ocular surface. This ocular disorder can cause eye irritation, irregular cornea, corneal barrier disruption, and blurred vision. Uncontrolled increase in matrix metalloproteinase-9 (MMP-9) level and activity has been detected in the tears and ocular surface in the patients with DES, which has been proved to be related to disruption of tight junctions in apical corneal epithelium associated with severe signs of DES. These uncontrolled activities of MMP-9 lead to desquamation of ocular surface epithelia. Therefore, this review study was conducted to summarize the evidence regarding MMP-9 contribution in DES, and inhibition of MMP-9, as a therapeutic target for treatment of DES. For this purpose, herein, the related studies designed novel pharmaceutical compounds for direct and indirect inhibition of MMP-9 as treatment approaches for DES were reviewed. These compounds were designed to improve corneal barrier function, reduce inflammation on ocular surface, and restore tear production.
Collapse
Affiliation(s)
- Alireza Shoari
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mozhgan Rezaei Kanavi
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Javad Rasaee
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
13
|
Satapathy S, Wilson MR. The Dual Roles of Clusterin in Extracellular and Intracellular Proteostasis. Trends Biochem Sci 2021; 46:652-660. [PMID: 33573881 DOI: 10.1016/j.tibs.2021.01.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/04/2021] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Abstract
Clusterin (CLU) was the first reported secreted mammalian chaperone and impacts on serious diseases associated with inappropriate extracellular protein aggregation. Many studies have described intracellular CLU in locations outside the secretory system and recent work has shown that CLU can be released into the cytosol during cell stress. In this article, we critically evaluate evidence relevant to the proposed origins of cellular CLU found outside the secretory system, and advance the hypothesis that the cytosolic release of CLU induced by stress serves to facilitate the trafficking of misfolded proteins to the proteasome and autophagy for degradation. We also propose future research directions that could help establish CLU as a unique chaperone performing critical and synergic roles in both intracellular and extracellular proteostasis.
Collapse
Affiliation(s)
- Sandeep Satapathy
- School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia; Molecular Horizons Research Institute, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia
| | - Mark R Wilson
- School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia; Molecular Horizons Research Institute, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia.
| |
Collapse
|
14
|
Fini ME, Jeong S, Wilson MR. Therapeutic Potential of the Molecular Chaperone and Matrix Metalloproteinase Inhibitor Clusterin for Dry Eye. Int J Mol Sci 2020; 22:E116. [PMID: 33374364 PMCID: PMC7794831 DOI: 10.3390/ijms22010116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 12/29/2022] Open
Abstract
Evidence is presented herein supporting the potential of the natural homeostatic glycoprotein CLU (clusterin) as a novel therapeutic for the treatment of dry eye. This idea began with the demonstration that matrix metalloproteinase MMP9 is required for damage to the ocular surface in mouse dry eye. Damage was characterized by degradation of OCLN (occludin), a known substrate of MMP9 and a key component of the paracellular barrier. Following up on this finding, a yeast two-hybrid screen was conducted using MMP9 as the bait to identify other proteins involved. CLU emerged as a strong interacting protein that inhibits the enzymatic activity of MMP9. Previously characterized as a molecular chaperone, CLU is expressed prominently by epithelia at fluid-tissue interfaces and secreted into bodily fluids, where it protects cells and tissues against damaging stress. It was demonstrated that CLU also protects the ocular surface in mouse dry eye when applied topically to replace the natural protein depleted from the dysfunctional tears. CLU is similarly depleted from tears in human dry eye. The most novel and interesting finding was that CLU binds selectively to the damaged ocular surface. In this position, CLU protects against epithelial cell death and barrier proteolysis, and dampens the autoimmune response, while the apical epithelial cell layer is renewed. When present at high enough concentration, CLU also blocks staining by vital dyes used clinically to diagnose dry eye. None of the current therapeutics have this combination of properties to "protect, seal, and heal". Future work will be directed towards human clinical trials to investigate the therapeutic promise of CLU.
Collapse
Affiliation(s)
- M. Elizabeth Fini
- New England Eye Center, Tufts Medical Center and Department of Ophthalmology, Tufts University School of Medicine, Program in Pharmacology & Drug Development, Graduate School of Biomedical Sciences Tufts University, Boston, MA 02111, USA
| | - Shinwu Jeong
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90089, USA;
| | - Mark R. Wilson
- The Illawarra Health and Medical Research Institute, Molecular Horizons and the School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia;
| |
Collapse
|
15
|
Abstract
The cornea is a transparent avascular tissue on the anterior segment of the eye responsible for providing refractive power and forming a protective barrier against the external environment. Infectious and inflammatory conditions can compromise the structure of the cornea, leading to visual impairment and blindness. Galectins are a group of β-galactoside-binding proteins expressed by immune and non-immune cells that play pivotal roles in innate and adaptive immunity. In this brief review, we discuss how different members of this family of proteins affect both pro-inflammatory and anti-inflammatory responses in the cornea, particularly in the context of infection, transplantation and wound healing. We further describe recent research showing beneficial effects of galectin-targeted therapy in corneal diseases.
Collapse
|
16
|
Fini ME, Jeong S, Gong H, Martinez-Carrasco R, Laver NMV, Hijikata M, Keicho N, Argüeso P. Membrane-associated mucins of the ocular surface: New genes, new protein functions and new biological roles in human and mouse. Prog Retin Eye Res 2019; 75:100777. [PMID: 31493487 DOI: 10.1016/j.preteyeres.2019.100777] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/26/2019] [Accepted: 08/31/2019] [Indexed: 01/17/2023]
Abstract
The mucosal glycocalyx of the ocular surface constitutes the point of interaction between the tear film and the apical epithelial cells. Membrane-associated mucins (MAMs) are the defining molecules of the glycocalyx in all mucosal epithelia. Long recognized for their biophysical properties of hydration, lubrication, anti-adhesion and repulsion, MAMs maintain the wet ocular surface, lubricate the blink, stabilize the tear film and create a physical barrier to the outside world. However, it is increasingly appreciated that MAMs also function as cell surface receptors that transduce information from the outside to the inside of the cell. A number of excellent review articles have provided perspective on the field as it has progressed since 1987, when molecular cloning of the first MAM was reported. The current article provides an update for the ocular surface, placing it into the broad context of findings made in other organ systems, and including new genes, new protein functions and new biological roles. We discuss the epithelial tissue-equivalent with mucosal differentiation, the key model system making these advances possible. In addition, we make the first systematic comparison of MAMs in human and mouse, establishing the basis for using knockout mice for investigations with the complexity of an in vivo system. Lastly, we discuss findings from human genetics/genomics, which are providing clues to new MAM roles previously unimagined. Taken together, this information allows us to generate hypotheses for the next stage of investigation to expand our knowledge of MAM function in intracellular signaling and roles unique to the ocular surface.
Collapse
Affiliation(s)
- M Elizabeth Fini
- Department of Ophthalmology, Tufts University School of Medicine, at New England Eye Center, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA.
| | - Shinwu Jeong
- USC Roski Eye Institute and Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, 1975 Zonal Ave, Los Angeles, CA, 90033, USA.
| | - Haiyan Gong
- Department of Ophthalmology, Boston University School of Medicine, 72 E Concord St, Boston, MA, 02118, USA.
| | - Rafael Martinez-Carrasco
- Department of Ophthalmology, Tufts University School of Medicine, at New England Eye Center, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA.
| | - Nora M V Laver
- Department of Ophthalmology, Tufts University School of Medicine, at New England Eye Center, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA.
| | - Minako Hijikata
- Department of Pathophysiology and Host Defense, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, 3-1-24 Matsuyama, Kiyose-shi, Tokyo, 204-8533, Japan.
| | - Naoto Keicho
- Department of Pathophysiology and Host Defense, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, 3-1-24 Matsuyama, Kiyose-shi, Tokyo, 204-8533, Japan.
| | - Pablo Argüeso
- Department of Ophthalmology, Harvard Medical School, at Schepens Eye Research Institute of Mass. Eye and Ear, 20 Staniford St, Boston, MA, 02114, USA.
| |
Collapse
|
17
|
Abstract
Purpose Dry eye is a major ocular pathology worldwide. Although dry eye is a multifactorial disease, recent studies have shown that chronic immunologic processes have a pivotal role in its pathogenesis, characterized by the infiltration of immune cells in the lacrimal glands, elevated levels of tear inflammatory cytokines, and increased density of immune cells in the cornea and conjunctiva. This review describes the recent advances in understanding the relationship between dry eye and inflammation. Methods This narrative review is based on searches of recent international literature using terms related to the immune response in dry eye, and includes clinical trials, animal experiments, and expert reviews. Results Although dry eye presents clinically as tear film instability associated with corneal/conjunctival epithelial disorders, Meibomian gland dysfunction, and decreased visual function, recent laboratory and clinical studies have indicated inflammation in the lacrimal glands, Meibomian glands, conjunctiva, cornea, and aqueous tears. Furthermore, inflammation at these locations leads to conjunctival goblet cell apoptosis, corneal epithelial barrier disruption, and corneal nerve damage. These inflammatory outcomes can be exacerbated by intrinsic and extrinsic factors, such as aging, sex steroid hormone, autoimmune diseases, contact lens use, visual display terminals, and dry environment. Conclusions Recent advances in dry eye research have revealed the inflammatory process and its pathogenesis, which has been proposed as an "inflammatory vicious cycle" of dry eye. Comprehensive assessment of dry eye based on inflammation will improve the selection of treatments and help break the inflammatory cycle in clinical settings.
Collapse
Affiliation(s)
- Takefumi Yamaguchi
- Department of Ophthalmology, Ichikawa General Hospital, Tokyo Dental College, Chiba, Japan
| |
Collapse
|
18
|
Roby DA, Ruiz F, Kermath BA, Voorhees JR, Niehoff M, Zhang J, Morley JE, Musiek ES, Farr SA, Burris TP. Pharmacological activation of the nuclear receptor REV-ERB reverses cognitive deficits and reduces amyloid-β burden in a mouse model of Alzheimer's disease. PLoS One 2019; 14:e0215004. [PMID: 30973894 PMCID: PMC6459530 DOI: 10.1371/journal.pone.0215004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 03/26/2019] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease currently lacks treatment options that effectively reverse the biological/anatomical pathology and cognitive deficits associated with the disease. Loss of function of the nuclear receptor REV-ERB is associated with reduced cognitive function in mouse models. The effect of enhanced REV-ERB activity on cognitive function has not been examined. In this study, we tested the hypothesis that enhanced REV-ERB function may enhance cognitive function in a model of Alzheimer's disease. We utilized the REV-ERB agonist SR9009 to pharmacologically activate the activity of REV-ERB in the SAMP8 mouse model of Alzheimer's disease. SR9009 reversed cognitive dysfunction of an aged SAMP8 mouse in several behavioral assays including novel object recognition, T-maze foot shock avoidance, and lever press operant conditioning task assessments. SR9009 treatment reduced amyloid-β 1-40 and 1-42 levels in the cortex, which is consistent with improved cognitive function. Furthermore, SR9009 treatment led to increased hippocampal PSD-95, cortical synaptophysin expression and the number of synapses suggesting improvement in synaptic function. We conclude that REV-ERB is a potential target for treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Deborah A. Roby
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - Fernanda Ruiz
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - Bailey A. Kermath
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - Jaymie R. Voorhees
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, MO, United States of America
| | - Michael Niehoff
- Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - Jinsong Zhang
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - John E. Morley
- Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - Erik S. Musiek
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Susan A. Farr
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States of America
- Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - Thomas P. Burris
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, MO, United States of America
| |
Collapse
|
19
|
Webster A, Chintala SK, Kim J, Ngan M, Itakura T, Panjwani N, Argüeso P, Barr JT, Jeong S, Fini ME. Dynasore protects the ocular surface against damaging oxidative stress. PLoS One 2018; 13:e0204288. [PMID: 30303976 PMCID: PMC6179211 DOI: 10.1371/journal.pone.0204288] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/04/2018] [Indexed: 02/04/2023] Open
Abstract
Water soluble "vital" dyes are commonly used clinically to evaluate health of the ocular surface; however, staining mechanisms remain poorly understood. Recent evidence suggests that sublethal damage stimulates vital dye uptake by individual living cells. Since cell damage can also stimulate reparative plasma membrane remodeling, we hypothesized that dye uptake occurs via endocytic vesicles. In support of this idea, we show here that application of oxidative stress to relatively undifferentiated monolayer cultures of human corneal epithelial cells stimulates both dye uptake and endocytosis, and that dye uptake is blocked by co-treatment with three different endocytosis inhibitors. Stress application to stratified and differentiated corneal epithelial cell cultures, which are a better model of the ocular surface, also stimulated dye uptake; however, endocytosis was not stimulated, and two of the endocytosis inhibitors did not block dye uptake. The exception was Dynasore and its more potent analogue Dyngo-4a, both small molecules developed to target dynamin family GTPases, but also having off-target effects on the plasma membrane. Significantly, while Dynasore blocked stress-stimulated dye uptake at the ocular surface of ex vivo mouse eyes when treatment was performed at the same time as eyes were stressed, it had no effect when used after stress was applied and the ocular surface was already damaged. Thus, Dynasore could not be working by inhibiting endocytosis. Employing cytotoxicity and western blotting assays, we went on to demonstrate an alternative mechanism. We show that Dynasore is remarkably protective of cells and their surface glycocalyx, preventing damage due to stress, and thus precluding dye entry. These unexpected and novel findings provide greater insight into the mechanisms of vital dye uptake and point the direction for future study. Significantly, they also suggest that Dynasore and its analogues might be used therapeutically to protect the ocular surface and to treat ocular surface disease.
Collapse
Affiliation(s)
- Andrew Webster
- USC Institute for Genetic Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| | - Shravan K. Chintala
- USC Institute for Genetic Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| | - Jasmine Kim
- Program in Biological Sciences, USC Dana and David Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, United States of America
| | - Michelle Ngan
- Program in Preventive Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| | - Tatsuo Itakura
- USC Institute for Genetic Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| | - Noorjahan Panjwani
- New England Eye Center/Department of Ophthalmology and Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, United States of America
| | - Pablo Argüeso
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, United States of America
| | - Joseph T. Barr
- The Ohio State University College of Optometry, Columbus, OH, United States of America
| | - Shinwu Jeong
- USC Institute for Genetic Medicine and USC Roski Eye Institute/Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| | - M. Elizabeth Fini
- USC Institute for Genetic Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| |
Collapse
|
20
|
AbuSamra DB, Argüeso P. Lectin-Glycan Interactions in Corneal Infection and Inflammation. Front Immunol 2018; 9:2338. [PMID: 30349544 PMCID: PMC6186829 DOI: 10.3389/fimmu.2018.02338] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 09/20/2018] [Indexed: 11/13/2022] Open
Abstract
The cornea is an extraordinary component of vision that functions as the principal barrier to pathogens in the eye while allowing light transmission into the retina. Understanding the cellular and molecular mechanisms that maintain homeostasis in this tissue is the subject of intense scientific study given the high prevalence of corneal disease. Over the past decade, the interactions between lectins and glycans on plasma membranes have emerged as important regulatory factors in corneal biology. In particular, members of the galectin family have been shown to bind multiple β-galactoside-containing receptors to regulate immunopathological processes associated with viral and bacterial infection, transplantation, wound healing, dry eye, angiogenesis, and lymphangiogenesis. In this review, we describe the current understanding of how these surface interactions intersect with different pathways to activate unique cellular responses in cornea as well as their potential therapeutic implications.
Collapse
Affiliation(s)
- Dina B AbuSamra
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Pablo Argüeso
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
21
|
Yu V, Bhattacharya D, Webster A, Bauskar A, Flowers C, Heur M, Chintala SK, Itakura T, Wilson MR, Barr JT, Jeong S, Wang M, Fini ME. Clusterin from human clinical tear samples: Positive correlation between tear concentration and Schirmer strip test results. Ocul Surf 2018; 16:478-486. [PMID: 30077709 PMCID: PMC6175631 DOI: 10.1016/j.jtos.2018.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 07/24/2018] [Accepted: 08/01/2018] [Indexed: 01/02/2023]
Abstract
PURPOSE To investigate the relationship between tear concentration of the homeostatic protein clusterin (CLU) and dry eye signs and symptoms, and to characterize tear CLU protein. METHODS Two independent studies were conducted, one in Tucson (44 subjects), the other in Los Angeles (52 subjects). A cohort study design was employed to enroll patients without regard to dry eye diagnosis. Dry eye signs and symptoms were assessed using clinical tests. Tear samples were collected by Schirmer strip, and also by micropipette at slit lamp when possible. CLU from both sample types was quantified by immunoassay. The relationship between CLU concentration and clinical test scores was determined by Pearson's correlation coefficient (for individual eyes) and multiple linear regression analysis (including both eyes). CLU was also evaluated biochemically by western blotting. RESULTS In the Tucson cohort, a positive correlation was observed between tear CLU concentration and results of the Schirmer strip test, a measure of tear flow (p = 0.021 includes both eyes). This result was corroborated in the Los Angeles cohort (p = 0.013). The mean tear CLU concentration was 31 ± 14 μg/mL (n = 18 subjects, 33 eyes; range = 7-48 μg/mL). CLU from clinical tear samples appeared biochemically similar to CLU from a non-clinical tear sample and from blood plasma. CONCLUSIONS Results support the hypothesis that an optimal concentration of tear CLU is important for ocular surface health, and that this drops below the effective threshold in dry eye. Tear CLU measurement might identify patients that could benefit from supplementation. Information about concentration will aid development of therapeutic dosage parameters.
Collapse
Affiliation(s)
- Valerie Yu
- MD Program, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Dhruva Bhattacharya
- Department of Ophthalmology & Vision Science, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Andrew Webster
- USC Institute for Genetic Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Aditi Bauskar
- PhD Program in Medical Biology, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Charles Flowers
- USC Roski Eye Institute and Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Martin Heur
- USC Roski Eye Institute and Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Shravan K Chintala
- USC Institute for Genetic Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Tatsuo Itakura
- USC Institute for Genetic Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Mark R Wilson
- Illawarra Health and Medical Research Institute, School of Biological Sciences, University of Wollongong, Wollongong, New South Wales, Australia
| | - Joseph T Barr
- The Ohio State University College of Optometry, Columbus, OH, USA
| | - Shinwu Jeong
- USC Institute for Genetic Medicine, USC Roski Eye Institute and Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Mingwu Wang
- Department of Ophthalmology & Vision Science, University of Arizona College of Medicine, Tucson, AZ, USA
| | - M Elizabeth Fini
- USC Institute for Genetic Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
22
|
Wiggs JL, Kang JH, Fan B, Levkovitch-Verbin H, Pasquale LR. A Role for Clusterin in Exfoliation Syndrome and Exfoliation Glaucoma? J Glaucoma 2018; 27 Suppl 1:S61-S66. [PMID: 29965900 PMCID: PMC8035929 DOI: 10.1097/ijg.0000000000000916] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The multifunctional protein clusterin (CLU) is a secreted glycoprotein ubiquitously expressed throughout the body, including in the eye. Its primary function is to act as an extracellular molecular chaperone, preventing the precipitation and aggregation of misfolded extracellular proteins. Clusterin is commonly identified at fluid-tissue interfaces, and has been identified in most body fluids. It is a component of exfoliation material, and CLU mRNA is reduced in eyes with exfoliation syndrome compared with controls. SNPs located in the CLU genomic region have been associated with Alzheimer disease (AD) at the genome-wide level and several CLU SNPs located in an apparent regulatory region have been nominally associated with XFS/XFG in Caucasians with European ancestry and in south Indians. Interestingly, clusterin associates with altered elastic fibers in human photoaged skin and prevents UV-induced elastin aggregation in vitro. In light of the known geographic risk factors for XFS/XFG, which could include UV light, investigations of CLU-geographic interactions could be of interest. Future studies investigating rare CLU variation and other complex interactions including gene-gene interactions in XFS/XFG cases and controls may also be fruitful. Although CLU has been considered as a therapeutic target in AD, cancer and dry eye, a role for clusterin in XFS/XFG needs to be better defined before therapeutic approaches involving CLU can be entertained.
Collapse
Affiliation(s)
- Janey L. Wiggs
- Department of Ophthalmology, Mass Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Jae Hee Kang
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - BaoJian Fan
- Department of Ophthalmology, Mass Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Hani Levkovitch-Verbin
- Goldschleger Eye Institute, Tel Hashomer, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Louis R. Pasquale
- Department of Ophthalmology, Mass Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Rohne P, Wolf S, Dörr C, Ringen J, Holtz A, Gollan R, Renner B, Prochnow H, Baiersdörfer M, Koch-Brandt C. Exposure of vital cells to necrotic cell lysates induce the IRE1α branch of the unfolded protein response and cell proliferation. Cell Stress Chaperones 2018; 23:77-88. [PMID: 28687980 PMCID: PMC5741583 DOI: 10.1007/s12192-017-0825-6] [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: 03/29/2017] [Revised: 05/15/2017] [Accepted: 06/16/2017] [Indexed: 10/19/2022] Open
Abstract
Necrosis is a form of cell death that is detrimental to the affected tissue because the cell ruptures and releases its content (reactive oxygen species among others) into the extracellular space. Clusterin (CLU), a cytoprotective extracellular chaperone has been shown to be upregulated in the face of necrosis. We here show that in addition to CLU upregulation, necrotic cell lysates induce JNK/SAPK signaling, the IRE1α branch of the unfolded protein response (UPR), the MAPK/ERK1/2, and the mTOR signaling pathways and results in an enhanced proliferation of the vital surrounding cells. We name this novel response mechanism: Necrosis-induced Proliferation (NiP).
Collapse
Affiliation(s)
- Philipp Rohne
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - Steven Wolf
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
- Department of Pathology, The University of Chicago, Chicago, IL USA
| | - Carolin Dörr
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - Julia Ringen
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - Andrew Holtz
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - René Gollan
- Department of Neurology, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Benjamin Renner
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - Hans Prochnow
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
- Department of Chemical Biology, Helmholtz Centre for Infection Research GmbH, Braunschweig, Germany
| | - Markus Baiersdörfer
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - Claudia Koch-Brandt
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| |
Collapse
|
24
|
Na YJ, Choi KJ, Park SB, Sung HR, Jung WH, Kim HY, Rhee SD, Kim KY. Protective effects of carbenoxolone, an 11β-HSD1 inhibitor, against chemical induced dry eye syndrome. Apoptosis 2017; 22:1441-1453. [DOI: 10.1007/s10495-017-1419-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
25
|
Abstract
The members of the Tear Film Subcommittee reviewed the role of the tear film in dry eye disease (DED). The Subcommittee reviewed biophysical and biochemical aspects of tears and how these change in DED. Clinically, DED is characterized by loss of tear volume, more rapid breakup of the tear film and increased evaporation of tears from the ocular surface. The tear film is composed of many substances including lipids, proteins, mucins and electrolytes. All of these contribute to the integrity of the tear film but exactly how they interact is still an area of active research. Tear film osmolarity increases in DED. Changes to other components such as proteins and mucins can be used as biomarkers for DED. The Subcommittee recommended areas for future research to advance our understanding of the tear film and how this changes with DED. The final report was written after review by all Subcommittee members and the entire TFOS DEWS II membership.
Collapse
|
26
|
Galletti JG, Guzmán M, Giordano MN. Mucosal immune tolerance at the ocular surface in health and disease. Immunology 2017; 150:397-407. [PMID: 28108991 DOI: 10.1111/imm.12716] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/28/2016] [Accepted: 01/12/2017] [Indexed: 12/15/2022] Open
Abstract
The ocular surface is constantly exposed to environmental irritants, allergens and pathogens, against which it can mount a prompt immune response to preserve its integrity. But to avoid unnecessary inflammation, the ocular surface's mucosal immune system must also discriminate between harmless and potentially dangerous antigens, a seemingly complicated task. Despite its unique features, the ocular surface is a mucosal lining, and as such, it shares some homeostatic and pathophysiological mechanisms with other mucosal surfaces. The purpose of this review is to explore the mucosal homeostatic immune function of the ocular surface in both the healthy and diseased states, with a special focus on mucosal immunology concepts. The information discussed in this review has been retrieved by PubMed searches for literature published from January 1981 to October 2016.
Collapse
Affiliation(s)
- Jeremías G Galletti
- Immunology Laboratory, Institute of Experimental Medicine, National Academy of Medicine/CONICET, Buenos Aires, Argentina
| | - Mauricio Guzmán
- Immunology Laboratory, Institute of Experimental Medicine, National Academy of Medicine/CONICET, Buenos Aires, Argentina
| | - Mirta N Giordano
- Immunology Laboratory, Institute of Experimental Medicine, National Academy of Medicine/CONICET, Buenos Aires, Argentina
| |
Collapse
|
27
|
Abstract
INTRODUCTION Clusterin (CLU) is a stress-activated, ATP-independent molecular chaperone, normally secreted from cells, that is up-regulated in Alzheimer disease and in many cancers. It plays important roles in protein homeostasis/proteostasis, inhibition of cell death pathways, and modulation of pro-survival signalling and transcriptional networks. Changes in the CLU gene locus are highly associated with Alzheimer disease, and many therapy-resistant cancers over-express CLU. The extensive post-translational processing and heterogeneous oligomerization of CLU have so far prevented any definitive structure determination. This in turn has meant that targeting CLU with small molecule inhibitors is challenging. Therefore, inhibiting CLU at the gene-expression level using siRNA or antisense is a valid approach to inhibit its function. Areas covered: This article reviews recent advances regarding the role of CLU in proteostasis, cellular trafficking, human diseases, and signalling pathways involved in oncogenesis. It addresses the rationale for CLU as a therapeutic target in cancer, and the current status of pre-clinical and clinical studies using CLU antisense inhibitor OGX011. Expert opinion: Discusses challenges facing the therapeutic targeting of CLU including rapid changes in the treatment landscape for prostate cancer with multiple new FDA approved drugs, selection of windows of intervention, and potential side effects when silencing CLU expression.
Collapse
Affiliation(s)
- Mark R Wilson
- a School of Biological Sciences , University of Wollongong , Wollongong , Australia
| | - Amina Zoubeidi
- b Department of Urologic Sciences, Vancouver Prostate Centre , University of British Columbia and Vancouver General Hospital , Vancouver , Canada
| |
Collapse
|
28
|
Fini ME, Bauskar A, Jeong S, Wilson MR. Clusterin in the eye: An old dog with new tricks at the ocular surface. Exp Eye Res 2016; 147:57-71. [PMID: 27131907 DOI: 10.1016/j.exer.2016.04.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/22/2016] [Accepted: 04/23/2016] [Indexed: 12/30/2022]
Abstract
The multifunctional protein clusterin (CLU) was first described in 1983 as a secreted glycoprotein present in ram rete testis fluid that enhanced aggregation ('clustering') of a variety of cells in vitro. It was also independently discovered in a number of other systems. By the early 1990s, CLU was known under many names and its expression had been demonstrated throughout the body, including in the eye. Its homeostatic activities in proteostasis, cytoprotection, and anti-inflammation have been well documented, however its roles in health and disease are still not well understood. CLU is prominent at fluid-tissue interfaces, and in 1996 it was demonstrated to be the most highly expressed transcript in the human cornea, the protein product being localized to the apical layers of the mucosal epithelia of the cornea and conjunctiva. CLU protein is also present in human tears. Using a preclinical mouse model for desiccating stress that mimics human dry eye disease, the authors recently demonstrated that CLU prevents and ameliorates ocular surface barrier disruption by a remarkable sealing mechanism dependent on attainment of a critical all-or-none concentration in the tears. When the CLU level drops below the critical all-or-none threshold, the barrier becomes vulnerable to desiccating stress. CLU binds selectively to the ocular surface subjected to desiccating stress in vivo, and in vitro to LGALS3 (galectin-3), a key barrier component. Positioned in this way, CLU not only physically seals the ocular surface barrier, but it also protects the barrier cells and prevents further damage to barrier structure. CLU depletion from the ocular surface epithelia is seen in a variety of inflammatory conditions in humans and mice that lead to squamous metaplasia and a keratinized epithelium. This suggests that CLU might have a specific role in maintaining mucosal epithelial differentiation, an idea that can now be tested using the mouse model for desiccating stress. Most excitingly, the new findings suggest that CLU could serve as a novel biotherapeutic for dry eye disease.
Collapse
Affiliation(s)
- M Elizabeth Fini
- USC Institute for Genetic Medicine and Departments of Cell & Neurobiology and Ophthalmology, Keck School of Medicine of USC, University of Southern California, 2250 Alcatraz St., Suite 240, Los Angeles, CA 90089-9037, USA.
| | - Aditi Bauskar
- USC Institute for Genetic Medicine and Graduate Program in Medical Biology, Keck School of Medicine of USC, University of Southern California, 2250 Alcatraz St., Suite 240, Los Angeles, CA 90089-9037, USA.
| | - Shinwu Jeong
- USC Institute for Genetic Medicine and Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, 2250 Alcatraz St., Suite 240, Los Angeles, CA 90089-9037, USA.
| | - Mark R Wilson
- Illawarra Health and Medical Research Institute, School of Biological Sciences, University of Wollongong, Northfields Avenue, Wollongong, New South Wales, 2522 Australia.
| |
Collapse
|