1
|
Yu M, Ning FTE, Liu C, Liu YC. Interconnections between diabetic corneal neuropathy and diabetic retinopathy: diagnostic and therapeutic implications. Neural Regen Res 2025; 20:2169-2180. [PMID: 39359077 PMCID: PMC11759029 DOI: 10.4103/nrr.nrr-d-24-00509] [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: 05/06/2024] [Revised: 06/28/2024] [Accepted: 07/24/2024] [Indexed: 10/04/2024] Open
Abstract
Diabetic corneal neuropathy and diabetic retinopathy are ocular complications occurring in the context of diabetes mellitus. Diabetic corneal neuropathy refers to the progressive damage of corneal nerves. Diabetic retinopathy has traditionally been considered as damage to the retinal microvasculature. However, growing evidence suggests that diabetic retinopathy is a complex neurovascular disorder resulting from dysfunction of the neurovascular unit, which includes both the retinal vascular structures and neural tissues. Diabetic retinopathy is one of the leading causes of blindness and is frequently screened for as part of diabetic ocular screening. However, diabetic corneal neuropathy is commonly overlooked and underdiagnosed, leading to severe ocular surface impairment. Several studies have found that these two conditions tend to occur together, and they share similarities in their pathogenesis pathways, being triggered by a status of chronic hyperglycemia. This review aims to discuss the interconnection between diabetic corneal neuropathy and diabetic retinopathy, whether diabetic corneal neuropathy precedes diabetic retinopathy, as well as the relation between the stage of diabetic retinopathy and the severity of corneal neuropathy. We also endeavor to explore the relevance of a corneal screening in diabetic eyes and the possibility of using corneal nerve measurements to monitor the progression of diabetic retinopathy.
Collapse
Affiliation(s)
- Mingyi Yu
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore
| | - Faith Teo En Ning
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chang Liu
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore
| | - Yu-Chi Liu
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore
- Department of Cornea and External Eye Disease, Singapore National Eye Center, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
| |
Collapse
|
2
|
Galor A, Gallar J, Acosta MC, Meseguer V, Benítez‐Del‐Castillo JM, Stachs O, Szentmáry N, Versura P, Müller‐Lierheim WGK, Belmonte C, Pujol‐Martí J. CORONIS symposium 2023: Scientific and clinical frontiers in ocular surface innervation. Acta Ophthalmol 2025; 103:e240-e255. [PMID: 39891368 PMCID: PMC12069973 DOI: 10.1111/aos.17450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/19/2025] [Indexed: 02/03/2025]
Abstract
The 5th CORONIS Symposium, held during the 2023 Congress of the European Association for Vision and Eye Research (EVER), highlighted the growing importance of ocular surface innervation in eye surface disorders. This article summarises the insights and perspectives shared during the symposium, which focused on the clinical relevance of ocular surface innervation, as well as on the development of innovative diagnostic and therapeutic approaches for ocular surface pathologies linked to disturbed sensory innervation. Through robust interdisciplinary collaborations, these developments hold great potential to improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Anat Galor
- Department of OphthalmologyMiami Veterans Affairs Medical CenterMiamiFloridaUSA
- Bascom Palmer Eye InstituteUniversity of MiamiMiamiFloridaUSA
| | - Juana Gallar
- Instituto de NeurocienciasUniversidad Miguel Hernández‐CSICSan Juan de AlicanteSpain
- Instituto de Investigación Sanitaria y Biomédica de AlicanteAlicanteSpain
| | - M. Carmen Acosta
- Instituto de NeurocienciasUniversidad Miguel Hernández‐CSICSan Juan de AlicanteSpain
| | - Víctor Meseguer
- Instituto de NeurocienciasUniversidad Miguel Hernández‐CSICSan Juan de AlicanteSpain
| | | | - Oliver Stachs
- Klinik und Poliklinik für AugenheilkundeUniversitätsmedizin RostockRostockGermany
- Interdisziplinäre Fakultät Leben, Licht & MaterieUniversität RostockRostockGermany
| | - Nóra Szentmáry
- Dr Rolf M. Schwiete Center for Limbal Stem Cell and Congenital Aniridia ResearchSaarland UniversityHomburgGermany
- Department of OphthalmologySemmelweis UniversityBudapestHungary
| | - Piera Versura
- Ophthalmology Unit, Dipartimento di Scienze Mediche e Chirurgiche (DIMEC)Alma Mater Studiorum Università di BolognaBolognaItaly
- IRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | | | - Carlos Belmonte
- Instituto de NeurocienciasUniversidad Miguel Hernández‐CSICSan Juan de AlicanteSpain
| | | |
Collapse
|
3
|
Mobeen R, Stapleton F, Chao C, Swarbrick H, Naduvilath T, Golebiowski B. Does orthokeratology contact lens wear suppress the immune cell response in the human corneal epithelium, while soft contact lens wear enhances it? Cont Lens Anterior Eye 2025; 48:102380. [PMID: 39884954 DOI: 10.1016/j.clae.2025.102380] [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: 08/26/2024] [Revised: 01/17/2025] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Orthokeratology (OK) contact lenses are increasingly prescribed for myopia control but their impact on corneal epithelial immune cells (CEIC) is unclear. This study compares CEIC in OK wearers to soft contact lens (SCL) wearers and non-wearers. METHODS In vivo confocal microscope images at the corneal central and mid-peripheral subbasal level were evaluated in 18 OK wearers, 18 SCL wearers and 18 non-wearers (mean age 27.6±8.0 years; 65% female). Corneal epithelial immune cell density was manually quantified and morphology was graded using a published system. Statistical significances (p<0.05) were examined using generalised estimating equations. RESULTS Corneal epithelial immune cell density in OK wearers was lower than in SCL wearers (p=0.03) at the central cornea, but not at the mid-periphery. Compared to SCL wearers, OK wearers had smaller cell bodies, with fewer participants displaying CEIC with dendrites, long and thick dendrites at both corneal locations (p<0.001). Orthokeratology wearers also had smaller CEIC bodies (p=0.01) and fewer participants had CEIC with dendrites (p=0.01) than non-wearers at both locations. Contrarily, SCL wearers had larger CEIC bodies, with a greater proportion of SCL wearers displaying CEIC with dendrites, long and thick dendrites compared to non-wearers (p≤0.04) at both locations. Corneal epithelial immune cell density was higher at the central cornea than at mid-periphery in SCL wearers (p<0.001) and non-wearers (p=0.01), but not in OK wearers (p=0.26). CONCLUSIONS In long-term OK lens wear, immune cells observed in the corneal epithelium are fewer in number and are less likely to present with dendrites, suggesting a suppressed CEIC response in OK lens wear which should be investigated further.
Collapse
Affiliation(s)
- Rabia Mobeen
- School of Optometry and Vision Science UNSW Sydney Australia.
| | - Fiona Stapleton
- School of Optometry and Vision Science UNSW Sydney Australia
| | - Cecilia Chao
- School of Optometry and Vision Science UNSW Sydney Australia
| | - Helen Swarbrick
- School of Optometry and Vision Science UNSW Sydney Australia
| | - Thomas Naduvilath
- School of Optometry and Vision Science UNSW Sydney Australia; Brien Holden Vision Institute UNSW Sydney Australia
| | | |
Collapse
|
4
|
Chen L, Li S, Fu Y. MicroRNAs in Corneal Diseases: Emerging Roles as Biomarkers, Regulators, and Therapeutics. Ocul Surf 2025:S1542-0124(25)00068-0. [PMID: 40412549 DOI: 10.1016/j.jtos.2025.05.007] [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: 11/07/2024] [Revised: 03/29/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
MicroRNAs (miRNAs) are conserved, short, non-coding RNAs that play a crucial role in regulating gene expression. Emerging evidence suggests that miRNAs are closely involved in the pathophysiology of various corneal diseases, particularly in regulating corneal wound healing, inflammation and neovascularization. In this review, we summarized the recent progress of miRNAs in corneal diseases, especially focused on their application as diagnostic biomarkers, regulators of cell biology, and therapeutic targets. Recent advances in miRNA detection technology have made it possible to analyze minimal miRNAs in samples such as tears or exosomes, further enhancing the ability to identify disease-specific miRNA profiles and providing potential objective indicators for the early diagnosis of disease. Meanwhile, we summarized the mechanisms and pathways of multiple miRNAs in regulating various biological processes of corneal cells, as well as the advantages of studying miRNA compared to proteins or genes. Furthermore, we explore the potential of miRNAs-based therapies, especially introduce various miRNA delivery systems and challenges associated with clinical translation. This review highlights the need for further research to harness the full potential of miRNAs in treating various corneal diseases.
Collapse
Affiliation(s)
- Liangbo Chen
- Department of Ophthalmology, Shanghai Ninth People's Hospill, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, 200011 Shanghai, China
| | - Shiding Li
- Department of Ophthalmology, Shanghai Ninth People's Hospill, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, 200011 Shanghai, China
| | - Yao Fu
- Department of Ophthalmology, Shanghai Ninth People's Hospill, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, 200011 Shanghai, China.
| |
Collapse
|
5
|
Hopkinson A, Figueiredo FC. A Narrative Review of Amniotic Membrane Transplantation in Ocular Surface Repair: Unveiling the Immunoregulatory Pathways for Timely Intervention. Ophthalmol Ther 2025:10.1007/s40123-025-01143-w. [PMID: 40360962 DOI: 10.1007/s40123-025-01143-w] [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: 12/14/2024] [Accepted: 04/04/2025] [Indexed: 05/15/2025] Open
Abstract
This narrative review explores the pathophysiology of ocular surface inflammation and highlights the therapeutic potential of patch amniotic membrane transplantation (patch-AMT) in ocular surface repair. Disruptions in ocular surface homeostasis caused by trauma, disease, or immune dysregulation trigger an inflammatory cascade that, if unresolved, can impair epithelial healing, lead to fibrosis, corneal haze, and vision loss. Patch-AMT provides a biological intervention with epitheliotropic, anti-inflammatory, anti-fibrotic, anti-angiogenic, and neuroprotective effects that support wound healing, regulate inflammation, and reduce pain. The review examines patch-AMT's role in acute conditions (chemical burns, Stevens-Johnson Syndrome) and chronic disease (persistent epithelial defects, dry eye disease), focusing on its ability to entrap immune cells, regulate cytokine signaling, and prevent fibrotic remodeling while releasing trophic proteins. Additionally, this review explores how preservation methods, application orientation, and intervention timing influences patch-AMT's efficacy. Recent advancements in non-surgical application methods have expanded accessibility, enabling earlier intervention and outpatient use. However, variability in clinical protocols emphasize the need for standardized guidelines. The review concludes by highlighting the need for further research to refine treatment timing, optimize repeat application strategies, and evaluate cost-effectiveness. While patch-AMT remains underutilized, growing evidence underscores its potential to improve clinical outcomes, particularly when applied early in disease progression.
Collapse
Affiliation(s)
- Andrew Hopkinson
- Academic Ophthalmology, Division of Clinical Neuroscience, Queen's Medical Centre (QMC), University of Nottingham, Queen's Medical Centre Campus, Nottingham, NG7 2UH, UK.
- NuVision Biotherapies, MediCity Nottingham, Nottingham, NG90 6BH, UK.
| | - Francisco C Figueiredo
- Department of Ophthalmology, Royal Victoria Infirmary, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
6
|
Park R, Spritz S, Zeng AY, Erukulla R, Zavala D, Merchant T, Gascon A, Jung R, Bigit B, Azar DT, Chang JH, Jalilian E, Djalilian AR, Guaiquil VH, Rosenblatt MI. Corneal Sensory Receptors and Pharmacological Therapies to Modulate Ocular Pain. Int J Mol Sci 2025; 26:4663. [PMID: 40429806 PMCID: PMC12111238 DOI: 10.3390/ijms26104663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 05/06/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Nociceptors respond to noxious stimuli and transmit pain signals to the central nervous system. In the cornea, the nociceptors located in the most external layer provide a myriad of sensation modalities. Damage to these corneal nerve fibers can induce neuropathic pain. In response, corneal nerves become sensitized to previously non-noxious stimuli. Assessing corneal pain origin is a complex ophthalmic challenge due to variations in its causes and manifestations. Current FDA-approved therapies for corneal nociceptive pain, such as acetaminophen and NSAIDs, provide only broad-acting relief with unwanted side effects, highlighting the need for precision medicine for corneal nociceptive pain. A few targeted treatments, including perfluorohexyloctane (F6H8) eye drops and Optive Plus (TRPV1 antagonist), are FDA-approved, while others are in preclinical development. Treatments that target signaling pathways related to neurotrophic factors, such as nerve growth factors and ion channels, such as the transient receptor potential (TRP) family or tropomyosin receptor kinase A, may provide a potential combinatory therapeutic approach. This review describes the roles of nociceptors in corneal pain. In addition, it evaluates molecules within nociceptor signaling pathways for their potential to serve as targets for efficient therapeutic strategies for corneal nociceptive pain aimed at modulating neurotrophic factors and nociceptive channel sensitivity.
Collapse
Affiliation(s)
- Ryan Park
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (R.P.); (S.S.); (D.Z.); (B.B.); (E.J.)
| | - Samantha Spritz
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (R.P.); (S.S.); (D.Z.); (B.B.); (E.J.)
| | - Anne Y. Zeng
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (R.P.); (S.S.); (D.Z.); (B.B.); (E.J.)
| | - Rohith Erukulla
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (R.P.); (S.S.); (D.Z.); (B.B.); (E.J.)
| | - Deneb Zavala
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (R.P.); (S.S.); (D.Z.); (B.B.); (E.J.)
| | - Tasha Merchant
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (R.P.); (S.S.); (D.Z.); (B.B.); (E.J.)
| | - Andres Gascon
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (R.P.); (S.S.); (D.Z.); (B.B.); (E.J.)
| | - Rebecca Jung
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (R.P.); (S.S.); (D.Z.); (B.B.); (E.J.)
| | - Bianca Bigit
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (R.P.); (S.S.); (D.Z.); (B.B.); (E.J.)
| | - Dimitri T. Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (R.P.); (S.S.); (D.Z.); (B.B.); (E.J.)
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (R.P.); (S.S.); (D.Z.); (B.B.); (E.J.)
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| | - Elmira Jalilian
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (R.P.); (S.S.); (D.Z.); (B.B.); (E.J.)
- Richard and Loan Hill Department of Bioengineering, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Ali R. Djalilian
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (R.P.); (S.S.); (D.Z.); (B.B.); (E.J.)
| | - Victor H. Guaiquil
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (R.P.); (S.S.); (D.Z.); (B.B.); (E.J.)
| | - Mark I. Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (R.P.); (S.S.); (D.Z.); (B.B.); (E.J.)
| |
Collapse
|
7
|
Chandler HL, Moradi S, Green SW, Chen P, Madden C, Zhang L, Zhang Z, Park KH, Ma J, Zhu H, Swindle-Reilly KE. Development of an Ophthalmic Hydrogel to Deliver MG53 and Promote Corneal Wound Healing. Pharmaceutics 2025; 17:526. [PMID: 40284520 PMCID: PMC12030682 DOI: 10.3390/pharmaceutics17040526] [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: 02/27/2025] [Revised: 03/28/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objective: A clinical need exists for more effective therapeutics and sustained drug delivery systems to promote ocular surface healing. This study tested the hypothesis that a novel biodegradable, thermoresponsive hydrogel loaded with the human recombinant (rh)MG53 protein, which we have demonstrated to promote corneal healing without fibrosis, would exhibit safety and biocompatibility in vitro and in vivo. Methods: Hydrogel optimization was performed based on varying concentrations of poloxamer 407, poloxamer 188, and hydroxypropyl methylcellulose. Hydrogels were characterized and potential toxicity was evaluated in vitro in cultured corneal epithelium, fibroblasts, and endothelium. In vivo safety and tolerability were assessed in mice and hydrogels were used to evaluate corneal healing following alkali injury. Results: The optimized hydrogel formulation did not result in any detrimental changes to the corneal cells and released functional rhMG53 protein for at least 24 h. In vivo rhMG53-loaded hydrogels improved re-epithelialization, reduced stromal opacification and vascularization, and promoted corneal nerve density. Mechanistically, rhMG53 reduced vascular endothelial cell migration and tube formation by inhibiting pSTAT3 signaling. Conclusions: Taken together, our poloxamer-based thermoresponsive hydrogel effectively released rhMG53 protein and enhanced multiple corneal healing outcomes.
Collapse
Affiliation(s)
- Heather L. Chandler
- College of Optometry, The Ohio State University, Columbus, OH 43210, USA; (C.M.); (L.Z.)
| | - Sara Moradi
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; (S.M.); (S.W.G.)
| | - Spencer W. Green
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; (S.M.); (S.W.G.)
| | - Peng Chen
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (P.C.); (Z.Z.); (H.Z.)
| | - Christopher Madden
- College of Optometry, The Ohio State University, Columbus, OH 43210, USA; (C.M.); (L.Z.)
| | - Luxi Zhang
- College of Optometry, The Ohio State University, Columbus, OH 43210, USA; (C.M.); (L.Z.)
| | - Zhentao Zhang
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (P.C.); (Z.Z.); (H.Z.)
| | - Ki Ho Park
- Division of Surgical Sciences, Department of Surgery, University of Virginia Medical School, Charlottesville, VA 22903, USA; (K.H.P.); (J.M.)
| | - Jianjie Ma
- Division of Surgical Sciences, Department of Surgery, University of Virginia Medical School, Charlottesville, VA 22903, USA; (K.H.P.); (J.M.)
| | - Hua Zhu
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (P.C.); (Z.Z.); (H.Z.)
| | - Katelyn E. Swindle-Reilly
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; (S.M.); (S.W.G.)
| |
Collapse
|
8
|
Lin Y, Yang L, Li Y, Dou S, Zhang Z, Zhou Q. CD4+CD25- T-Cell-Secreted IFN-γ Promotes Corneal Nerve Degeneration in Diabetic Mice. Invest Ophthalmol Vis Sci 2025; 66:15. [PMID: 40192636 PMCID: PMC11980951 DOI: 10.1167/iovs.66.4.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
Purpose This study aimed to explore the relationship between corneal nerve degeneration and elevated dendritic cells (DCs) in diabetic keratopathy. Methods Corneas from diabetic and healthy mice were analyzed using single-cell RNA sequencing. Corneal nerve density and DC and T-cell infiltration were quantified through whole-mount corneal staining. Freshly isolated mouse trigeminal ganglion (TG) neurons were co-cultured with immature DCs, mature DCs, activated CD8+ T cells, and CD4+CD25- T cells. TG neurite outgrowth was assessed to identify potential effector cells driving corneal nerve degeneration. In addition, interferon-gamma (IFN-γ) and blocking antibodies were used to evaluate their effects on TG neurite outgrowth and corneal nerve degeneration in mice. Results Compared with age-matched healthy mice, diabetic mice exhibited a significant reduction in corneal nerve density and sensitivity, along with increased infiltration of DCs, CD4+ T cells, and CD8+ T cells. In vitro co-culture experiments revealed that CD4+CD25- T cells, rather than DCs and CD8+ T cells, significantly inhibited TG neurite outgrowth. Among cytokines, elevated IFN-γ in diabetic corneas impaired TG neurite outgrowth and induced corneal nerve degeneration, whereas IL-4 and IL-17 had no such effect. Blocking IFN-γ alleviated CD4+CD25- T-cell-induced inhibition of TG neurite outgrowth and corneal nerve degeneration in diabetic mice. Conclusions CD4+CD25- T cells, but not DCs or CD8+ T cells, contribute to corneal nerve degeneration in diabetic mice, a process partially mediated by IFN-γ.
Collapse
Affiliation(s)
- Yujing Lin
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Lingling Yang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Ya Li
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Shengqian Dou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Zhenzhen Zhang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| |
Collapse
|
9
|
Liu Y, Ma B, Zhao L, Li H, Li W, Sun Z, Duan H, Zhao Y, Qi H. Influence of dendritic cells on corneal nerve morphological analysis and clinical relevance in chronic dry eye disease after femtosecond laser-assisted laser in situ keratomileusis. Front Med (Lausanne) 2025; 12:1568787. [PMID: 40231078 PMCID: PMC11994687 DOI: 10.3389/fmed.2025.1568787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/03/2025] [Indexed: 04/16/2025] Open
Abstract
Purpose This study aims to investigate the influence of dendritic cells (DCs) on corneal nerve morphology and the clinical significance in chronic Femtosecond Laser-Assisted Laser in Situ Keratomileusis (FS-LASIK) related dry eye disease (DED). Methods The cross-sectional study was conducted involving healthy control, DED without FS-LASIK group, and DED after FS-LASIK group. Clinical parameters such as ocular surface disease index (OSDI), fluorescein tear breakup time (FBUT), corneal fluorescein staining (CFS) scores, Schirmer I test (SIt), Cochet-Bonnet esthesiometer (C-BE) were recorded. DCs of in vivo confocal microscopy images were included or excluded during corneal nerve segmentation. Key morphological parameters, including corneal nerve fiber density (CNFD), corneal nerve branch density (CNBD), tortuosity, and box-count fractal dimension (Boxdim), were measured. The impact of DCs on nerve metrics and clinical parameters and the correlations between each other were assessed. Results The significant reduce in key morphological parameters was observed after eliminating DCs. Significant differences of morphological parameters were observed in DED after FS-LASIK group compared with other two groups. With the increased presence of DCs density in DED especially in DED after FS-LASIK group, the presence of DCs introduced false positives in the correlation analysis of DCs density with corneal morphology in DED after FS-LASIK and in the correlation analysis of corneal morphology with clinical characteristics in DED without FS-LASIK. Conclusion The presence of DCs introduces significant biases in the assessments of corneal nerve morphology, primarily false-positive results in DED especially chronic FS-LASIK related DED. Their exclusion improves the precision of nerve measurements, which may enhance the clinical evaluation of corneal nerve morphology. These findings highlight the importance of precise segmentation techniques to minimize DCs related interference in clinical practice.
Collapse
Affiliation(s)
- Yilin Liu
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Baikai Ma
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Lu Zhao
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmic and Visual Science Key Laboratory, Beijing, China
| | - Hongshuo Li
- Cixi Institute of BioMedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Wenlong Li
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Zhengze Sun
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Hongyu Duan
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Yitian Zhao
- Cixi Institute of BioMedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Hong Qi
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| |
Collapse
|
10
|
Özkan G, Turhan SA, Toker E. Effect of high and low molecular weight sodium hyaluronic acid eye drops on corneal recovery after crosslinking in keratoconus patients. BMJ Open Ophthalmol 2025; 10:e001890. [PMID: 40157723 PMCID: PMC11956357 DOI: 10.1136/bmjophth-2024-001890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 03/19/2025] [Indexed: 04/01/2025] Open
Abstract
OBJECTIVE To assess the impact of eye drops containing high molecular weight hyaluronic acid (HMW-HA) and low molecular weight hyaluronic acid (LMW-HA) on corneal nerve regeneration, dendritic cell (DC) density, corneal sensitivity (CS) and ocular surface parameters in patients with keratoconus following corneal crosslinking (CXL). METHODS AND ANALYSIS 63 eyes of 55 patients with keratoconus were randomised to receive eye drops containing HMW-HA (n: 20) for 12 months, LMW-HA (n: 23) for 12 months and polyvinyl alcohol (n: 20) until closure of the epithelial defect in the control group after CXL. Subbasal nerve plexus (SNP) was imaged with corneal confocal microscopy and quantified with ACCMetrics. DC density was calculated with Image J. Ocular Surface Disease Index (OSDI) questionnaire and non-invasive break-up time (NI-TBUT) were evaluated. All measurements were performed before CXL and 1, 3, 6 and 12 months postsurgery. RESULTS At 6 months post-CXL, SNP reached to its preoperative. CS was higher in the HMW-HA groups compared with the other two groups in the 3rd and 6th month post-CXL. DC density was higher in the LMW-HA group compared with the HMW-HA group in the postoperative 3rd month. OSDI were higher in the control group compared with both the LMW-HA and HMW-HA groups at postoperative 3rd and 6th months. NI-TBUT was lower in the control group in the 6th and 12th months compared with the other groups. CONCLUSIONS The use of artificial tear drops containing HMW-HA may have a therapeutic effect to promote corneal nerve regrowth and support faster functional recovery after CXL. TRIAL REGISTRATION NUMBER NCT06243991.
Collapse
Affiliation(s)
- Gamze Özkan
- Ophthalmology, Marmara University School of Medicine, Istanbul, Turkey
| | | | - Ebru Toker
- Ophthalmology, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
11
|
Xu K, Wu K, Chen L, Zhao Y, Li H, Lin N, Ye Z, Xu J, Huang D, Huang X. Selective promotion of sensory innervation-mediated immunoregulation for tissue repair. SCIENCE ADVANCES 2025; 11:eads9581. [PMID: 40117376 PMCID: PMC11927663 DOI: 10.1126/sciadv.ads9581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/14/2025] [Indexed: 03/23/2025]
Abstract
Sensory innervation triggers the regenerative response after injury. However, dysfunction and impairment of sensory nerves, accompanied by excessive inflammation impede tissue regeneration. Consequently, specific induction of sensory innervation to mediate immunoregulation becomes a promising therapeutic approach. Herein, we developed a cell/drug-free strategy to selectively boost endogenous sensory innervation to harness immune responses for promoting tissue rehabilitation. Specifically, a dual-functional phage was constructed with a sensory nerve-homing peptide and a β-subunit of nerve growth factor (β-NGF)-binding peptide. These double-displayed phages captured endogenic β-NGF and localized to sensory nerves to promote sensory innervation. Furthermore, regarding bone regeneration, phage-loaded hydrogels achieved rapid sensory nerve ingrowth in bone defect areas. Mechanistically, sensory neurotization facilitated M2 polarization of macrophages through the Sema3A/XIAP/PAX6 pathway, thus decreasing the M1/M2 ratio to induce the dissipation of local inflammation. Collectively, these findings highlight the essential role of sensory innervation in manipulating inflammation and provide a conceptual framework based on neuroimmune interactions for promoting tissue regeneration.
Collapse
Affiliation(s)
- Kaicheng Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Kaile Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Liang Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yubin Zhao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hengyuan Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Nong Lin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhaoming Ye
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jianbin Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Donghua Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xin Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Anam A, Yu M, Liu C, Lee IXY, Yang J, Shanmathi AV, Cheng CY, Liu YC. Smoking negatively impacts ocular surface health and corneal nerve metrics. Ocul Surf 2025; 37:105-114. [PMID: 40088969 DOI: 10.1016/j.jtos.2025.03.002] [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: 11/10/2024] [Revised: 02/19/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
PURPOSE To evaluate the effects of smoking on ocular surface through comprehensive analysis of corneal nerves, corneal epithelium, dendritic cells (DCs), and clinical assessments. METHODS This cross-sectional study included 250 healthy smokers and 272 healthy non-smokers. Patients' smoking status and duration were recorded. In vivo confocal microscopy was performed to assess 7 quantitative corneal nerves parameters, 3 corneal neuroma parameters, 3 DCs parameters, and 3 epithelial parameters. Ocular surface evaluations included tear break-up time (TBUT), ocular surface and corneal staining, corneal sensitivity, and Schirmer test. Ocular Surface Disease Index questionnaire was used for symptom assessment. RESULTS Compared to non-smokers, smokers exhibited significantly lower corneal nerve fiber density (CNFD), nerve branch density, nerve fiber length, nerve total branch density, corneal nerve fiber area (CNFA), and corneal nerve fractal dimension (CFracDim; all p < 0.001). Smokers also presented with a significantly swollen corneal nerve fiber (p < 0.001). Longer smoking duration was significantly associated with lower CNFD (β = -0.04, P = 0.010), lower CNFA (β = -0.00002, P = 0.033), and lower CFracDim (β = -0.0008, P = 0.016). Additionally, a significantly larger neuroma total area (p = 0.040), size (p < 0.001) and perimeter (p < 0.001), as well as a significantly higher DCs density (p < 0.001), DCs count (p = 0.003), and lower DCs elongation which suggested higher DCs maturity (p < 0.001), were observed in the smoking group. Smokers demonstrated significantly higher ocular surface staining scores (p < 0.001) and reduced TBUT (p = 0.001). Corneal epithelial circularity was borderline higher in the smoking subjects (p = 0.059). CONCLUSIONS Smoking is associated with significant alterations in corneal nerve morphology and quantity, increased immunological cells, and compromised ocular surface integrity.
Collapse
Affiliation(s)
- Ansa Anam
- Department of Ophthalmology, MTI Khyber Teaching Hospital, Peshawar, Pakistan
| | - Mingyi Yu
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore
| | - Chang Liu
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore
| | - Isabelle Xin Yu Lee
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore
| | - Juanita Yang
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore
| | - A V Shanmathi
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore
| | - Ching-Yu Cheng
- Epidemiology Group, Singapore Eye Research Institute, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore
| | - Yu-Chi Liu
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore; Department of Cornea and External Eye Disease, Singapore National Eye Centre, Singapore; Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
13
|
Cui Z, Li X, Ou Y, Sun X, Gu J, Ding C, Yu Z, Guo Y, Liang Y, Mao S, Ma JH, Chan HF, Tang S, Chen J. Novel full-thickness biomimetic corneal model for studying pathogenesis and treatment of diabetic keratopathy. Mater Today Bio 2025; 30:101409. [PMID: 39807180 PMCID: PMC11729032 DOI: 10.1016/j.mtbio.2024.101409] [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: 10/08/2024] [Revised: 12/08/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025] Open
Abstract
Diabetic keratopathy (DK), a significant complication of diabetes, often leads to corneal damage and vision impairment. Effective models are essential for studying DK pathogenesis and evaluating potential therapeutic interventions. This study developed a novel biomimetic full-thickness corneal model for the first time, incorporating corneal epithelial cells, stromal cells, endothelial cells, and nerves to simulate DK conditions in vitro. By exposing the model to a high-glucose (HG) environment, the pathological characteristics of DK, including nerve bundle disintegration, compromised barrier integrity, increased inflammation, and oxidative stress, were successfully replicated. Transcriptomic analysis revealed that HG downregulated genes associated with axon and synapse formation while upregulating immune response and oxidative stress pathways, with C-C Motif Chemokine Ligand 5 (CCL5) identified as a key hub gene in DK pathogenesis. The therapeutic effects of Lycium barbarum glycopeptide (LBGP) were evaluated using this model and validated in db/db diabetic mice. LBGP promoted nerve regeneration, alleviated inflammation and oxidative stress in both in vitro and in vivo models. Notably, LBGP suppressed the expression of CCL5, highlighting its potential mechanism of action. This study establishes a robust biomimetic platform for investigating DK and other corneal diseases, and identifies LBGP as a promising therapeutic candidate for DK. These findings provide valuable insights into corneal disease mechanisms and pave the way for future translational research and clinical applications.
Collapse
Affiliation(s)
- Zekai Cui
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan, China
- Changsha Aier Eye Hospital, Changsha, Hunan, China
| | - Xiaoxue Li
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan, China
| | - Yiwen Ou
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan, China
| | - Xihao Sun
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan, China
- Changsha Aier Eye Hospital, Changsha, Hunan, China
| | - Jianing Gu
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan, China
| | - Chengcheng Ding
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan, China
| | - Zhexiong Yu
- Tianren Goji Biotechnology Co., Ltd, Ningxia, China
| | - Yonglong Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yuqin Liang
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan, China
| | - Shengru Mao
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan, China
| | - Jacey Hongjie Ma
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan, China
- Changsha Aier Eye Hospital, Changsha, Hunan, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Shibo Tang
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan, China
- Changsha Aier Eye Hospital, Changsha, Hunan, China
| | - Jiansu Chen
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan, China
- Changsha Aier Eye Hospital, Changsha, Hunan, China
| |
Collapse
|
14
|
Surico PL, Naderi A, Singh RB, Kahale F, Farsi Y, Lee S, Musayeva A, Chen Y, Dana R. Antagonizing NK-1R modulates pain perception following corneal injury. Exp Eye Res 2025; 251:110230. [PMID: 39761841 DOI: 10.1016/j.exer.2025.110230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 02/07/2025]
Abstract
Substance P (SP) expressed by corneal nerves, is an 11-amino acid long neuropeptide from the tachykinin family, encoded by the Tac1 gene, and binds to neurokinin receptors. SP overexpression is associated with various pathological responses in the cornea including vasodilation, pain, inflammation, and angiogenesis in the normally avascular tissue. This study investigates the role of neurokinin-1 receptor (NK-1R) mediated signaling in nociception, nerve regeneration, and neuronal activation following mechanical corneal injury in mice. Corneal injuries were induced in age- and sex-matched C57BL/6 mice by removing corneal epithelium and partial anterior stroma. Following injury, mice were treated with either L-733,060, an NK-1R antagonist, or vehicle, administered topically twice daily for 21 days. Corneal SP levels were measured using ELISA, and nerve regeneration was assessed by quantifying corneal nerve fiber density (CNFD) via β-Tubulin III staining. Gene expression of neuronal markers (ATF3, GFAP, cFos, TRPV1, and TRPM8) in the trigeminal ganglia was measured using qPCR. Pain responses were evaluated using the eye-wiping test (EWT) and palpebral ratio (PR). Results indicated a persistent increase in corneal SP post-injury, significantly reduced by NK-1R antagonism. At 21 days, NK-1R antagonist-treated mice showed higher CNFD, reduced expression of neuronal activation markers, and lower pain perception compared to controls. These findings suggest that SP/NK-1R signaling is critical in corneal nociception post-injury, and its inhibition reduces pain, prevents neuronal hyperactivation, and supports nerve regeneration.
Collapse
Affiliation(s)
- Pier Luigi Surico
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Amirreza Naderi
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Rohan Bir Singh
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Francesca Kahale
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Yeganeh Farsi
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Seokjoo Lee
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Aytan Musayeva
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Yihe Chen
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Reza Dana
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
Buonanno M, Hashmi R, Petersen CE, Tang Z, Welch D, Shuryak I, Brenner DJ. Wavelength-dependent DNA damage induced by single wavelengths of UV-C radiation (215 to 255 nm) in a human cornea model. Sci Rep 2025; 15:252. [PMID: 39747969 PMCID: PMC11696903 DOI: 10.1038/s41598-024-84196-4] [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: 09/21/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
Scientific bodies overseeing UV radiation protection recommend safety limits for exposure to ultraviolet radiation in the workplace based on published peer-reviewed data. To support this goal, a 3D model of the human cornea was used to assess the wavelength dependence of corneal damage induced by UV-C radiation. In the first set of experiments the models were exposed with or without simulated tears; at each wavelength (215-255 nm) cells with DNA dimers and their distribution within the epithelium were measured. Simulated tears reduced the fraction of damaged cells to an extent dependent on the wavelength and tissue layer. Subsequent experiments were performed with models exposed without simulated tears; yields of DNA-damaged cells and their distribution within the corneal epithelium were evaluated at each wavelength, together with other markers of cell and tissue integrity. Unlike relatively longer wavelengths, the range of wavelengths commonly referred to as far-UV-C (215-235 nm) only induced dimers in the uppermost layers of the epithelium and did not result in lasting damage or halt proliferation of the germinative cells. These results provide evidence for the recommended exposure limits for far-UV-C wavelengths, which have been proposed as a practical technology to reduce the risk of transmission of airborne diseases in occupied locations.
Collapse
Affiliation(s)
- Manuela Buonanno
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th Street, New York, 10032, USA.
| | - Raabia Hashmi
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th Street, New York, 10032, USA
| | - Camryn E Petersen
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th Street, New York, 10032, USA
| | - Zheng Tang
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th Street, New York, 10032, USA
| | - David Welch
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th Street, New York, 10032, USA
| | - Igor Shuryak
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th Street, New York, 10032, USA
| | - David J Brenner
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th Street, New York, 10032, USA
| |
Collapse
|
16
|
Wu M, Fletcher EL, Chinnery HR, Downie LE, Mueller SN. Redefining our vision: an updated guide to the ocular immune system. Nat Rev Immunol 2024; 24:896-911. [PMID: 39215057 DOI: 10.1038/s41577-024-01064-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2024] [Indexed: 09/04/2024]
Abstract
Balanced immune responses in the eyes are crucial to preserve vision. The ocular immune system has long been considered distinct, owing to the so-called 'immune privilege' of its component tissues. More recently, intravital imaging and transcriptomic techniques have reshaped scientific understanding of the ocular immune landscape, such as revealing the specialization of immune cell populations in the various tissues of the eye. As knowledge of the phenotypes of corneal and retinal immune cells has evolved, links to both the systemic immune system, and the central and peripheral nervous systems, have been identified. Using intravital imaging, T cells have recently been found to reside in, and actively patrol, the healthy human cornea. Disease-associated retinal microglia with links to retinal degeneration have also been identified. This Review provides an updated guide to the ocular immune system, highlighting current knowledge of the immune cells that are present in steady-state and specific diseased ocular tissues, as well as evidence for their relationship to systemic disease. In addition, we discuss emerging intravital imaging techniques that can be used to visualize immune cell morphology and dynamics in living human eyes and how these could be applied to advance understanding of the human immune system.
Collapse
Affiliation(s)
- Mengliang Wu
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Erica L Fletcher
- Department of Anatomy and Physiology, The University of Melbourne, Carlton, Victoria, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia.
- Lions Eye Institute, Nedlands, Western Australia, Australia.
- Optometry, The University of Western Australia, Crawley, Western Australia, Australia.
| | - Laura E Downie
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia.
| | - Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
17
|
Idiaquez JF, Barnett‐Tapia C, Perkins BA, Bril V. Assessing corneal dendritic cells in glucose dysregulation small-fibre neuropathy. J Peripher Nerv Syst 2024; 29:400-405. [PMID: 39532698 PMCID: PMC11625976 DOI: 10.1111/jns.12671] [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: 09/16/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND AIMS Small-fibre neuropathy (SFN) is associated with glucose dysregulation, including impaired glucose tolerance (IGT) and type 2 diabetes (T2D). Corneal confocal microscopy (CCM) offers a non-invasive tool to assess corneal nerve damage and dendritic cell density (DCD). In this study, we investigated corneal DCD in patients with SFN and glucose dysregulation, defined as IGT or T2D. METHODS We enrolled 38 patients with SFN + glucose dysregulation, 51 with SFN + non-glucose dysregulation and 20 healthy controls. All participants underwent neurological examination, neurophysiology and CCM. RESULTS Individuals with SFN and glucose dysregulation had higher DCD compared with healthy controls (p = .01), and mature DCD was higher in IGT SFN patients than in T2D patients. INTERPRETATION Higher DCD in IGT compared with controls and patients with established T2D may suggest that DCD is a biomarker of early neuropathy.
Collapse
Affiliation(s)
- Juan Francisco Idiaquez
- Ellen and Martin Prosserman Centre for Neuromuscular Diseases, Division of Neurology, Department of MedicineUniversity Health Network, University of TorontoTorontoCanada
| | - Carolina Barnett‐Tapia
- Ellen and Martin Prosserman Centre for Neuromuscular Diseases, Division of Neurology, Department of MedicineUniversity Health Network, University of TorontoTorontoCanada
| | - Bruce A. Perkins
- Division of Endocrinology and MetabolismUniversity of Toronto, and the Leadership Sinai Centre for Diabetes, Sinai HealthTorontoCanada
| | - Vera Bril
- Ellen and Martin Prosserman Centre for Neuromuscular Diseases, Division of Neurology, Department of MedicineUniversity Health Network, University of TorontoTorontoCanada
| |
Collapse
|
18
|
Huang D, Li Z. Multidimensional immunotherapy for dry eye disease: current status and future directions. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1449283. [PMID: 39554604 PMCID: PMC11564177 DOI: 10.3389/fopht.2024.1449283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/10/2024] [Indexed: 11/19/2024]
Abstract
Dry Eye Disease (DED) is a multifactorial condition driven by tear film hyperosmolarity, immune dysregulation, and neuro-immune interactions. The immune system plays a central role in its pathogenesis, influencing both inflammation and ocular surface damage. While traditional immunotherapies like anti-inflammatory agents and immunosuppressants offer symptom relief, their long-term use is limited by side effects. This review focuses on emerging immunotherapies, including biologics, stem cell therapy, gene therapy, nanotechnology, and exosome-based treatments, all of which hold promise in modulating immune responses and promoting tissue repair. The relationship between the ocular microbiome and DED is also explored, with an emphasis on personalized immunotherapy. Key challenges for future research include identifying novel therapeutic targets, optimizing clinical translation, and evaluating the long-term efficacy of these innovative treatments.
Collapse
Affiliation(s)
- Duliurui Huang
- Department of Ophthalmology, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Henan University, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhijie Li
- Department of Ophthalmology, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Henan University, People’s Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
19
|
Blanco T, Nakagawa H, Musayeva A, Krauthammer M, Singh RB, Narimatsu A, Ge H, Shoushtari SI, Dana R. Acquired immunostimulatory phenotype of migratory CD103+ DCs promotes alloimmunity following corneal transplantation. JCI Insight 2024; 9:e182469. [PMID: 39235864 PMCID: PMC11530131 DOI: 10.1172/jci.insight.182469] [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: 05/01/2024] [Accepted: 08/15/2024] [Indexed: 09/07/2024] Open
Abstract
After transplantation, Th1-mediated immune rejection is the predominant cause of graft failure. Th1 cell sensitization occurs through complex and context-dependent interaction among antigen-presenting cell subsets, particularly CD11b+ DCs (DC2) and CD103+ DCs (DC1). This interaction necessitates further investigation in the context of transplant immunity. We used well-established preclinical models of corneal transplantation and identified distinct roles of migratory CD103+ DC1 in influencing the outcomes of the grafted tissue. In recipients with uninflamed corneal beds, migratory CD103+ DC1 demonstrate a tolerogenic phenotype that modulates the immunogenic capacity of CD11b+ DC2 primarily mediated by IL-10, suppressing alloreactive CD4+ Th1 cells via the PD-L1/PD-1 pathway and enhancing Treg-mediated tolerance via αvβ8 integrin-activated TGF-β1, thus facilitating graft survival. Conversely, in recipients with inflamed and vascularized corneal beds, IFN-γ produced by CD4+ Th1 cells induced migratory CD103+ DC1 to adopt an immunostimulatory phenotype, characterized by the downregulation of regulatory markers, including αvβ8 integrin and IL-10, and the upregulation of IL-12 and costimulatory molecules CD80/86, resulting in graft failure. The adoptive transfer of ex vivo induced tolerogenic CD103+ DC1 (iDC1) effectively inhibited Th1 polarization and preserved the tolerogenic phenotype of their physiological counterparts. Collectively, our findings underscore the essential role played by CD103+ DC1 in modulating host alloimmune responses.
Collapse
|
20
|
Barone V, Surico PL, Cutrupi F, Mori T, Gallo Afflitto G, Di Zazzo A, Coassin M. The Role of Immune Cells and Signaling Pathways in Diabetic Eye Disease: A Comprehensive Review. Biomedicines 2024; 12:2346. [PMID: 39457658 PMCID: PMC11505591 DOI: 10.3390/biomedicines12102346] [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: 09/24/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Diabetic eye disease (DED) encompasses a range of ocular complications arising from diabetes mellitus, including diabetic retinopathy, diabetic macular edema, diabetic keratopathy, diabetic cataract, and glaucoma. These conditions are leading causes of visual impairments and blindness, especially among working-age adults. Despite advancements in our understanding of DED, its underlying pathophysiological mechanisms remain incompletely understood. Chronic hyperglycemia, oxidative stress, inflammation, and neurodegeneration play central roles in the development and progression of DED, with immune-mediated processes increasingly recognized as key contributors. This review provides a comprehensive examination of the complex interactions between immune cells, inflammatory mediators, and signaling pathways implicated in the pathogenesis of DED. By delving in current research, this review aims to identify potential therapeutic targets, suggesting directions of research for future studies to address the immunopathological aspects of DED.
Collapse
Affiliation(s)
- Vincenzo Barone
- Department of Ophthalmology, Campus Bio-Medico University, 00128 Rome, Italy; (V.B.); (F.C.); (T.M.); (A.D.Z.); (M.C.)
- Ophthalmology Operative Complex Unit, Campus Bio-Medico University Hospital Foundation, 00128 Rome, Italy
| | - Pier Luigi Surico
- Department of Ophthalmology, Campus Bio-Medico University, 00128 Rome, Italy; (V.B.); (F.C.); (T.M.); (A.D.Z.); (M.C.)
- Ophthalmology Operative Complex Unit, Campus Bio-Medico University Hospital Foundation, 00128 Rome, Italy
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Francesco Cutrupi
- Department of Ophthalmology, Campus Bio-Medico University, 00128 Rome, Italy; (V.B.); (F.C.); (T.M.); (A.D.Z.); (M.C.)
- Ophthalmology Operative Complex Unit, Campus Bio-Medico University Hospital Foundation, 00128 Rome, Italy
| | - Tommaso Mori
- Department of Ophthalmology, Campus Bio-Medico University, 00128 Rome, Italy; (V.B.); (F.C.); (T.M.); (A.D.Z.); (M.C.)
- Ophthalmology Operative Complex Unit, Campus Bio-Medico University Hospital Foundation, 00128 Rome, Italy
- Department of Ophthalmology, University of California San Diego, La Jolla, CA 92122, USA
| | - Gabriele Gallo Afflitto
- Ophthalmology Unit, Department of Experimental Medicine, University of Rome “Tor Vergata”, 00128 Rome, Italy;
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
| | - Antonio Di Zazzo
- Department of Ophthalmology, Campus Bio-Medico University, 00128 Rome, Italy; (V.B.); (F.C.); (T.M.); (A.D.Z.); (M.C.)
- Ophthalmology Operative Complex Unit, Campus Bio-Medico University Hospital Foundation, 00128 Rome, Italy
| | - Marco Coassin
- Department of Ophthalmology, Campus Bio-Medico University, 00128 Rome, Italy; (V.B.); (F.C.); (T.M.); (A.D.Z.); (M.C.)
- Ophthalmology Operative Complex Unit, Campus Bio-Medico University Hospital Foundation, 00128 Rome, Italy
| |
Collapse
|
21
|
Yaman E, Heyer N, de Paiva CS, Stepp MA, Pflugfelder SC, Alam J. Mouse Corneal Immune Cell Heterogeneity Revealed by Single-Cell RNA Sequencing. Invest Ophthalmol Vis Sci 2024; 65:29. [PMID: 39432400 PMCID: PMC11500044 DOI: 10.1167/iovs.65.12.29] [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: 05/01/2024] [Accepted: 10/03/2024] [Indexed: 10/23/2024] Open
Abstract
Purpose This study aimed to define the heterogeneity, spatial localization, and functional roles of immune cells in the mouse cornea using single-cell RNA sequencing (scRNA-seq) and immunofluorescent staining. Methods Enriched mouse corneal immune cells (C57BL/6 strain, age 16-20 weeks) underwent single-cell RNA sequencing library preparation, sequencing, and analysis with Seurat, Monocle 3, and CellChat packages in R. Pathway analysis used Qiagen Ingenuity Pathway Analysis software. Immunostaining confirmed cell distribution. Results We identified 14 distinct immune cell clusters (56% myeloid and 44% lymphoid). Myeloid populations included resident macrophages, conventional dendritic cells (cDC2s), Langerhans cells, neutrophils, monocytes, and mast cells. Additionally, lymphocyte subsets (B, CD8, CD4, γδT, natural killer, natural killer T, and group 2 innate lymphoid cells) were found. We also found three new subtypes of resident macrophages in the cornea. Trajectory analysis suggested a differentiation pathway from monocytes to conventional dendritic cells, resident macrophages, and LCs. Intercellular communication network analysis using cord diagram identified amyloid precursor protein, chemokine (C-C motif) ligands (2, 3, 4, 6, 7, 9, and 12), Cxcl2, Mif, Tnf, Tgfb1, Igf1, and Il10 as prominent ligands involved in these interactions. Sexually dimorphic gene expression patterns were observed, with male myeloid cells expressing genes linked to immune regulation (Egr1, Foxp1, Mrc1, and Il1rn) and females showing higher expression of antigen presentation genes (Clic1, Psmb8, and Psmb9). Finally, immunostaining confirmed the spatial distribution of these cell populations within the cornea. Conclusions This study unveils a diverse immune landscape in the mouse cornea, with evidence for cell differentiation and sex-based differences. Immunostaining validates the spatial distribution of these populations, furthering our knowledge of corneal immune function.
Collapse
Affiliation(s)
- Ebru Yaman
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Nicole Heyer
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Cintia S. de Paiva
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Mary Ann Stepp
- Departments of Anatomy, Regenerative Biology and Ophthalmology, The George Washington University Medical School and Health Sciences, Washington, DC, United States
| | - Stephen C. Pflugfelder
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Jehan Alam
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
22
|
Wu M, Zhang X, Karunaratne S, Lee JH, Lampugnani ER, Selva KJ, Chung AW, Mueller SN, Chinnery HR, Downie LE. Intravital Imaging of the Human Cornea Reveals the Differential Effects of Season on Innate and Adaptive Immune Cell Morphodynamics. Ophthalmology 2024; 131:1185-1195. [PMID: 38703795 DOI: 10.1016/j.ophtha.2024.04.020] [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: 01/22/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024] Open
Abstract
PURPOSE Defining how the in vivo immune status of peripheral tissues is shaped by the external environment has remained a technical challenge. We recently developed Functional in vivo confocal microscopy (Fun-IVCM) for dynamic, longitudinal imaging of corneal immune cells in living humans. This study investigated the effect of seasonal-driven environmental factors on the morphodynamic features of human corneal immune cell subsets. DESIGN Longitudinal, observational clinical study. PARTICIPANTS Sixteen healthy participants (aged 18-40 years) attended 2 visits in distinct seasons in Melbourne, Australia (Visit 1, November-December 2021 [spring-summer]; Visit 2, April-June 2022 [autumn-winter]). METHODS Environmental data were collected over each period. Participants underwent ocular surface examinations and corneal Fun-IVCM (Heidelberg Engineering). Corneal scans were acquired at 5.5 ± 1.5-minute intervals for up to 5 time points. Time-lapse Fun-IVCM videos were created to analyze corneal immune cells, comprising epithelial T cells and dendritic cells (DCs), and stromal macrophages. Tear cytokines were analyzed using a multiplex bead-based immunoassay. MAIN OUTCOME MEASURES Difference in the density, morphology, and dynamic parameters of corneal immune cell subsets over the study periods. RESULTS Visit 1 was characterized by higher temperature, lower humidity, and higher air particulate and pollen levels compared with Visit 2. Clinical ocular surface parameters and the density of immune cell subsets were similar across visits. At Visit 1 , corneal epithelial DCs were larger, with a lower dendrite probing speed (0.38 ± 0.21 vs. 0.68 ± 0.33 μm/min; P < 0.001) relative to Visit 2; stromal macrophages were more circular and had less dynamic activity (Visit 1, 7.2 ± 1.9 vs. Visit 2, 10.3 ± 3.7 dancing index; P < 0.001). Corneal T cell morphodynamics were unchanged across periods. Basal tear levels of interleukin 2 and CXCL10 were relatively lower during spring-summer. CONCLUSIONS This study identifies that the in vivo morphodynamics of innate corneal immune cells (DCs, macrophages) are modified by environmental factors, but such effects are not evident for adaptive immune cells (T cells). The cornea is a potential in vivo window to investigate season-dependent environmental influences on the human immune system. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Mengliang Wu
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia; Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Xinyuan Zhang
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia
| | - Senuri Karunaratne
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia
| | - Ji-Hyun Lee
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia
| | - Edwin R Lampugnani
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia; Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kevin J Selva
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia
| | - Laura E Downie
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia.
| |
Collapse
|
23
|
Suanno G, Genna VG, Maurizi E, Dieh AA, Griffith M, Ferrari G. Cell therapy in the cornea: The emerging role of microenvironment. Prog Retin Eye Res 2024; 102:101275. [PMID: 38797320 DOI: 10.1016/j.preteyeres.2024.101275] [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: 10/11/2023] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
The cornea is an ideal testing field for cell therapies. Its highly ordered structure, where specific cell populations are sequestered in different layers, together with its accessibility, has allowed the development of the first stem cell-based therapy approved by the European Medicine Agency. Today, different techniques have been proposed for autologous and allogeneic limbal and non-limbal cell transplantation. Cell replacement has also been attempted in cases of endothelial cell decompensation as it occurs in Fuchs dystrophy: injection of cultivated allogeneic endothelial cells is now in advanced phases of clinical development. Recently, stromal substitutes have been developed with excellent integration capability and transparency. Finally, cell-derived products, such as exosomes obtained from different sources, have been investigated for the treatment of severe corneal diseases with encouraging results. Optimization of the success rate of cell therapies obviously requires high-quality cultured cells/products, but the role of the surrounding microenvironment is equally important to allow engraftment of transplanted cells, to preserve their functions and, ultimately, lead to restoration of tissue integrity and transparency of the cornea.
Collapse
Affiliation(s)
- Giuseppe Suanno
- Vita-Salute San Raffaele University, Milan, Italy; Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Eleonora Maurizi
- Centre for Regenerative Medicine ''S. Ferrari'', University of Modena and Reggio Emilia, Modena, Italy
| | - Anas Abu Dieh
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada
| | - May Griffith
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada.
| | - Giulio Ferrari
- Vita-Salute San Raffaele University, Milan, Italy; Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy; Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
24
|
Meng T, Zheng J, Shin CS, Gao N, Bande D, Sudarjat H, Chow W, Halquist MS, Yu FS, Acharya G, Xu Q. Combination Nanomedicine Strategy for Preventing High-Risk Corneal Transplantation Rejection. ACS NANO 2024; 18:20679-20693. [PMID: 39074146 PMCID: PMC11308920 DOI: 10.1021/acsnano.4c06595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 07/31/2024]
Abstract
High-risk (HR) corneal transplantation presents a formidable challenge, with over 50% of grafts experiencing rejection despite intensive postoperative care involving frequent topical eyedrop administration up to every 2 h, gradually tapering over 6-12 months, and ongoing maintenance dosing. While clinical evidence underscores the potential benefits of inhibiting postoperative angiogenesis, effective antiangiogenesis therapy remains elusive in this context. Here, we engineered controlled-release nanomedicine formulations comprising immunosuppressants (nanoparticles) and antiangiogenesis drugs (nanowafer) and demonstrated that these formulations can prevent HR corneal transplantation rejection for at least 6 months in a clinically relevant rat model. Unlike untreated corneal grafts, which universally faced rejection within 2 weeks postsurgery, a single subconjunctival injection of the long-acting immunosuppressant nanoparticle alone effectively averted graft rejection for 6 months, achieving a graft survival rate of ∼70%. Notably, the combination of an immunosuppressant nanoparticle and an anti-VEGF nanowafer yielded significantly better efficacy with a graft survival rate of >85%. The significantly enhanced efficacy demonstrated that a combination nanomedicine strategy incorporating immunosuppressants and antiangiogenesis drugs can greatly enhance the ocular drug delivery and benefit the outcome of HR corneal transplantation with increased survival rate, ensuring patient compliance and mitigating dosing frequency and toxicity concerns.
Collapse
Affiliation(s)
- Tuo Meng
- Department
of Pharmaceutics, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
| | - Jinhua Zheng
- Department
of Pharmaceutics, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
- Department
of Ophthalmology, Affiliated Hospital of
Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Crystal S. Shin
- Michale
E. DeBakey Department of Surgery, Baylor
College of Medicine, Houston, Texas 77030, United States
| | - Nan Gao
- Departments
of Ophthalmology, Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Divya Bande
- Department
of Pharmaceutics, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
| | - Hadi Sudarjat
- Department
of Pharmaceutics, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
| | - Woon Chow
- Department
of Ophthalmology, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
- Department
of Pathology, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Matthew Sean Halquist
- Department
of Pharmaceutics, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
| | - Fu-Shin Yu
- Departments
of Ophthalmology, Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Ghanashyam Acharya
- Michale
E. DeBakey Department of Surgery, Baylor
College of Medicine, Houston, Texas 77030, United States
- Department
of Materials Science and Nanoengineering, Rice University, Houston, Texas 77005, United States
| | - Qingguo Xu
- Department
of Pharmaceutics, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
- Department
of Ophthalmology, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
- Center
for Pharmaceutical Engineering; Institute for Structural Biology,
Drug Discovery & Development (ISB3D); and Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
25
|
Tang Y, Qu S, Ning Z, Wu H. Immunopeptides: immunomodulatory strategies and prospects for ocular immunity applications. Front Immunol 2024; 15:1406762. [PMID: 39076973 PMCID: PMC11284077 DOI: 10.3389/fimmu.2024.1406762] [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: 03/25/2024] [Accepted: 07/01/2024] [Indexed: 07/31/2024] Open
Abstract
Immunopeptides have low toxicity, low immunogenicity and targeting, and broad application prospects in drug delivery and assembly, which are diverse in application strategies and drug combinations. Immunopeptides are particularly important for regulating ocular immune homeostasis, as the eye is an immune-privileged organ. Immunopeptides have advantages in adaptive immunity and innate immunity, treating eye immune-related diseases by regulating T cells, B cells, immune checkpoints, and cytokines. This article summarizes the application strategies of immunopeptides in innate immunity and adaptive immunity, including autoimmunity, infection, vaccine strategies, and tumors. Furthermore, it focuses on the mechanisms of immunopeptides in mediating ocular immunity (autoimmune diseases, inflammatory storms, and tumors). Moreover, it reviews immunopeptides' application strategies and the therapeutic potential of immunopeptides in the eye. We expect the immune peptide to get attention in treating eye diseases and to provide a direction for eye disease immune peptide research.
Collapse
Affiliation(s)
| | | | | | - Hong Wu
- Eye Center of Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
26
|
Crabtree JR, Tannir S, Tran K, Boente CS, Ali A, Borschel GH. Corneal Nerve Assessment by Aesthesiometry: History, Advancements, and Future Directions. Vision (Basel) 2024; 8:34. [PMID: 38804355 PMCID: PMC11130793 DOI: 10.3390/vision8020034] [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: 03/01/2024] [Revised: 04/28/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024] Open
Abstract
The measurement of corneal sensation allows clinicians to assess the status of corneal innervation and serves as a crucial indicator of corneal disease and eye health. Many devices are available to assess corneal sensation, including the Cochet-Bonnet aesthesiometer, the Belmonte Aesthesiometer, the Swiss Liquid Jet Aesthesiometer, and the newly introduced Corneal Esthesiometer Brill. Increasing the clinical use of in vivo confocal microscopy and optical coherence tomography will allow for greater insight into the diagnosis, classification, and monitoring of ocular surface diseases such as neurotrophic keratopathy; however, formal esthesiometric measurement remains necessary to assess the functional status of corneal nerves. These aesthesiometers vary widely in their mode of corneal stimulus generation and their relative accessibility, precision, and ease of clinical use. The development of future devices to optimize these characteristics, as well as further comparative studies between device types should enable more accurate and precise diagnosis and treatment of corneal innervation deficits. The purpose of this narrative review is to describe the advancements in the use of aesthesiometers since their introduction to clinical practice, compare currently available devices for assessing corneal innervation and their relative limitations, and discuss how the assessment of corneal innervation is crucial to understanding and treating pathologies of the ocular surface.
Collapse
Affiliation(s)
- Jordan R. Crabtree
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.R.C.)
| | - Shadia Tannir
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.R.C.)
| | - Khoa Tran
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.R.C.)
| | - Charline S. Boente
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Asim Ali
- Department of Ophthalmology and Vision Sciences, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Gregory H. Borschel
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.R.C.)
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
27
|
Surico PL, Narimatsu A, Forouzanfar K, Singh RB, Shoushtari S, Dana R, Blanco T. Effects of Diabetes Mellitus on Corneal Immune Cell Activation and the Development of Keratopathy. Cells 2024; 13:532. [PMID: 38534376 PMCID: PMC10969384 DOI: 10.3390/cells13060532] [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: 01/16/2024] [Revised: 03/12/2024] [Accepted: 03/16/2024] [Indexed: 03/28/2024] Open
Abstract
Diabetes mellitus (DM) is one of the most prevalent diseases globally, and its prevalence is rapidly increasing. Most patients with a long-term history of DM present with some degree of keratopathy (DK). Despite its high incidence, the underlying inflammatory mechanism of DK has not been elucidated yet. For further insights into the underlying immunopathologic processes, we utilized streptozotocin-induced mice to model type 1 DM (T1D) and B6.Cg-Lepob/J mice to model type 2 DM (T2D). We evaluated the animals for the development of clinical manifestations of DK. Four weeks post-induction, the total frequencies of corneal CD45+CD11b+Ly-6G- myeloid cells, with enhanced gene and protein expression levels for the proinflammatory cytokines TNF-α and IL-1β, were higher in both T1D and T2D animals. Additionally, the frequencies of myeloid cells/mm2 in the sub-basal neural plexus (SBNP) were significantly higher in T1D and T2D compared to non-diabetic mice. DK clinical manifestations were observed four weeks post-induction, including significantly lower tear production, corneal sensitivity, and epitheliopathy. Nerve density in the SBNP and intraepithelial terminal endings per 40x field were lower in both models compared to the normal controls. The findings of this study indicate that DM alters the immune quiescent state of the cornea during disease onset, which may be associated with the progressive development of the clinical manifestations of DK.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tomas Blanco
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA; (P.L.S.); (A.N.); (K.F.); (R.B.S.); (S.S.); (R.D.)
| |
Collapse
|
28
|
Zidan AA, Zhu S, Elbasiony E, Najafi S, Lin Z, Singh RB, Naderi A, Yin J. Topical application of calcitonin gene-related peptide as a regenerative, antifibrotic, and immunomodulatory therapy for corneal injury. Commun Biol 2024; 7:264. [PMID: 38438549 PMCID: PMC10912681 DOI: 10.1038/s42003-024-05934-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 02/19/2024] [Indexed: 03/06/2024] Open
Abstract
Calcitonin gene-related peptide (CGRP) is a multifunctional neuropeptide abundantly expressed by corneal nerves. Using a murine model of corneal mechanical injury, we found CGRP levels in the cornea significantly reduced after injury. Topical application of CGRP as an eye drop accelerates corneal epithelial wound closure, reduces corneal opacification, and prevents corneal edema after injury in vivo. CGRP promotes corneal epithelial cell migration, proliferation, and the secretion of laminin. It reduces TGF-β1 signaling and prevents TGF-β1-mediated stromal fibroblast activation and tissue fibrosis. CGRP preserves corneal endothelial cell density, morphology, and pump function, thus reducing corneal edema. Lastly, CGRP reduces neutrophil infiltration, macrophage maturation, and the production of inflammatory cytokines in the cornea. Taken together, our results show that corneal nerve-derived CGRP plays a cytoprotective, pro-regenerative, anti-fibrotic, and anti-inflammatory role in corneal wound healing. In addition, our results highlight the critical role of sensory nerves in ocular surface homeostasis and injury repair.
Collapse
Affiliation(s)
- Asmaa A Zidan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Shuyan Zhu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Elsayed Elbasiony
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Sheyda Najafi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Zhirong Lin
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Rohan Bir Singh
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Amirreza Naderi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Jia Yin
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Buonfiglio F, Wasielica-Poslednik J, Pfeiffer N, Gericke A. Diabetic Keratopathy: Redox Signaling Pathways and Therapeutic Prospects. Antioxidants (Basel) 2024; 13:120. [PMID: 38247544 PMCID: PMC10812573 DOI: 10.3390/antiox13010120] [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: 12/19/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
Diabetes mellitus, the most prevalent endocrine disorder, not only impacts the retina but also significantly involves the ocular surface. Diabetes contributes to the development of dry eye disease and induces morphological and functional corneal alterations, particularly affecting nerves and epithelial cells. These changes manifest as epithelial defects, reduced sensitivity, and delayed wound healing, collectively encapsulated in the context of diabetic keratopathy. In advanced stages of this condition, the progression to corneal ulcers and scarring further unfolds, eventually leading to corneal opacities. This critical complication hampers vision and carries the potential for irreversible visual loss. The primary objective of this review article is to offer a comprehensive overview of the pathomechanisms underlying diabetic keratopathy. Emphasis is placed on exploring the redox molecular pathways responsible for the aberrant structural changes observed in the cornea and tear film during diabetes. Additionally, we provide insights into the latest experimental findings concerning potential treatments targeting oxidative stress. This endeavor aims to enhance our understanding of the intricate interplay between diabetes and ocular complications, offering valuable perspectives for future therapeutic interventions.
Collapse
Affiliation(s)
- Francesco Buonfiglio
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (J.W.-P.); (N.P.)
| | | | | | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (J.W.-P.); (N.P.)
| |
Collapse
|
30
|
Wang C, Liu X, Zhou J, Zhang Q. The Role of Sensory Nerves in Dental Pulp Homeostasis: Histological Changes and Cellular Consequences after Sensory Denervation. Int J Mol Sci 2024; 25:1126. [PMID: 38256202 PMCID: PMC10815945 DOI: 10.3390/ijms25021126] [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: 12/20/2023] [Revised: 01/10/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
Homeostatic maintenance is essential for pulp function. Disrupting pulp homeostasis may lead to pulp degeneration, such as fibrosis and calcifications. Sensory nerves constitute a crucial component of the dental pulp. However, the precise involvement of sensory nerves in pulp homeostasis remains uncertain. In this study, we observed the short-term and long-term histological changes in the dental pulp after inferior alveolar nerve transection. Additionally, we cultured primary dental pulp cells (DPCs) from the innervated and denervated groups and compared indicators of cellular senescence and cellular function. The results revealed that pulp fibrosis occurred at 2 w after the operation. Furthermore, the pulp area, as well as the height and width of the pulp cavity, showed accelerated reductions after sensory denervation. Notably, the pulp area at 16 w after the operation was comparable to that of 56 w old rats. Sensory denervation induced excessive extracellular matrix (ECM) deposition and increased predisposition to mineralization. Furthermore, sensory denervation promoted the senescence of DPCs. Denervated DPCs exhibited decelerated cell proliferation, arrest in the G2/M phase of the cell cycle, imbalance in the synthesis and degradation of ECM, and enhanced mineralization. These findings indicate that sensory nerves play an essential role in pulp homeostasis maintenance and dental pulp cell fate decisions, which may provide novel insights into the prevention of pulp degeneration.
Collapse
Affiliation(s)
| | | | | | - Qi Zhang
- Department of Endodontics, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Yanchang Middle Road, Jing’an District, Shanghai 200072, China
| |
Collapse
|
31
|
Volatier T, Cursiefen C, Notara M. Current Advances in Corneal Stromal Stem Cell Biology and Therapeutic Applications. Cells 2024; 13:163. [PMID: 38247854 PMCID: PMC10814767 DOI: 10.3390/cells13020163] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
Corneal stromal stem cells (CSSCs) are of particular interest in regenerative ophthalmology, offering a new therapeutic target for corneal injuries and diseases. This review provides a comprehensive examination of CSSCs, exploring their anatomy, functions, and role in maintaining corneal integrity. Molecular markers, wound healing mechanisms, and potential therapeutic applications are discussed. Global corneal blindness, especially in more resource-limited regions, underscores the need for innovative solutions. Challenges posed by corneal defects, emphasizing the urgent need for advanced therapeutic interventions, are discussed. The review places a spotlight on exosome therapy as a potential therapy. CSSC-derived exosomes exhibit significant potential for modulating inflammation, promoting tissue repair, and addressing corneal transparency. Additionally, the rejuvenation potential of CSSCs through epigenetic reprogramming adds to the evolving regenerative landscape. The imperative for clinical trials and human studies to seamlessly integrate these strategies into practice is emphasized. This points towards a future where CSSC-based therapies, particularly leveraging exosomes, play a central role in diversifying ophthalmic regenerative medicine.
Collapse
Affiliation(s)
- Thomas Volatier
- Department of Ophthalmology, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Claus Cursiefen
- Department of Ophthalmology, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
- Cologne Excellence Cluster for Cellular Stress Responses Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine, University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Maria Notara
- Department of Ophthalmology, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine, University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
32
|
Huang R, Su C, Zhang N, Shi C, Pu G, Ding Y, Wei W, Chen J. Cord blood-derived biologics lead to robust axonal regeneration in benzalkonium chloride-injured mouse corneas by modulating the Il-17 pathway and neuropeptide Y. Mol Med 2024; 30:2. [PMID: 38172658 PMCID: PMC10763178 DOI: 10.1186/s10020-023-00772-w] [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: 09/25/2023] [Accepted: 12/17/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Umbilical cord blood-derived therapeutics, such as serum (UCS) and platelet-rich plasma (UCPRP), are popular treatment options in clinical trials and can potentially be utilized to address a clinically unmet need caused by preservatives, specifically benzalkonium chloride (BAK), present in ophthalmic formulations. As current clinical interventions for secondary injuries caused by BAK are suboptimal, this study will explore the feasibility of utilizing UCS and UCPRP for cornea treatment and investigate the underlying mechanisms associated with this approach. METHODS Mice's corneas were administered BAK to induce damage. UCS and UCPRP were then utilized to attempt to treat the injuries. Ocular tests were performed on the animals to evaluate recovery, while immunostaining, RNA-seq, and subsequent bioinformatics analysis were conducted to investigate the treatment mechanism. RESULTS BAK administration led to widespread inflammatory responses in the cornea. Subsequent treatment with UCS and UCPRP led to the downregulation of immune-related 'interactions between cytokine receptors' and 'IL-17 signaling' pathways. Although axonal enhancers such as Ngf, Rac2, Robo2, Srgap1, and Rock2 were found to be present in the injured group, robust axonal regeneration was observed only in the UCS and UCPRP treatment groups. Further analysis revealed that, as compared to normal corneas, inflammation was not restored to pre-injury levels post-treatment. Importantly, Neuropeptide Y (Npy) was also involved in regulating immune responses, indicating neuroimmune axis interactions. CONCLUSIONS Cord blood-derived therapeutics are feasible options for overcoming the sustained injuries induced by BAK in the cornea. They also have potential applications in areas where axonal regeneration is required.
Collapse
Affiliation(s)
- Ruojing Huang
- The First Affiliated Hospital of Jinan University, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Caiying Su
- The First Affiliated Hospital of Jinan University, Tianhe District, Guangzhou, 510630, Guangdong Province, China
- Peking University Shenzhen Hospital, Futian District, Shenzhen, 518036, Guangdong Province, China
| | - Na Zhang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Dr, Singapore, 637459, Singapore
| | - Congying Shi
- Institution of Guangdong Cord Blood Bank, Guangdong Women and Children Hospital, Guangzhou, 510705, Guangdong Province, China
- Department of Experimental Center, Guangzhou Municipality Tianhe Nuoya Bio-Engineering Co., Ltd, Guangzhou, 510705, Guangdong Province, China
| | - Guangming Pu
- Jinan University Affiliated Heyuan Hospital, Guangzhou, 517000, Guangdong Province, China
| | - Yong Ding
- The First Affiliated Hospital of Jinan University, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Wei Wei
- Institution of Guangdong Cord Blood Bank, Guangdong Women and Children Hospital, Guangzhou, 510705, Guangdong Province, China.
- Department of Experimental Center, Guangzhou Municipality Tianhe Nuoya Bio-Engineering Co., Ltd, Guangzhou, 510705, Guangdong Province, China.
| | - Jiansu Chen
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, 510630, Guangdong Province, China.
| |
Collapse
|
33
|
Li L, Yu Y, Zhuang Z, Wu Q, Lin S, Hu J. Dopamine Receptor 1 Treatment Promotes Epithelial Repair of Corneal Injury by Inhibiting NOD-Like Receptor Protein 3-Associated Inflammation. Invest Ophthalmol Vis Sci 2024; 65:49. [PMID: 38294802 PMCID: PMC10839817 DOI: 10.1167/iovs.65.1.49] [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: 08/23/2023] [Accepted: 01/10/2024] [Indexed: 02/01/2024] Open
Abstract
Purpose To elucidate the influence of dopamine receptor 1 (DRD1) on the proliferation of mouse corneal epithelial cells (MCECs) under inflammatory conditions. Methods In vitro, immortalized MCECs (iMCECs) were treated with IL-1β, with and without pcDNA3.1_DRD1. Primary MCECs (pMCECs) were exposed to IL-1β, with and without DRD1 agonist (A68930). Cell proliferation was quantified using the Cell Counting Kit-8 (CCK-8) assay and immunofluorescence staining for Ki-67 and p63. Expression levels of NOD-like receptor protein 3 (NLRP3), IL-1β, and IL-6 were assessed. To establish a corneal injury model in mice, a 2-mm superficial keratectomy was performed. Either 0.1% A68930 or PBS was topically administered three times daily to the injured eyes for up to 5 days post-injury. Immunofluorescence analysis was employed to evaluate the expression of Ki-67, p63, and CD45 in mouse corneas. Western blotting and real-time quantitative PCR were utilized for quantitative analysis of DRD1, NLRP3, IL-1β, and IL-6 in mouse corneas. Corneal epithelial regeneration was monitored through fluorescein sodium staining for a duration of up to 5 days following the injury. Results Overexpression of DRD1 and A68930 promoted MCEC proliferation and suppressed the expression of NLRP3, IL-1β, and IL-6 in vitro. Topical application of the 0.1% A68930 following mechanical corneal injury in mice led to increased Ki-67 and p63 expression compared to PBS treatment. Furthermore, topical administration of the 0.1% A68930 reduced the expression of CD45, NLRP3, IL-1β, and IL-6. Analysis with fluorescein sodium indicated accelerated corneal epithelial regeneration in the 0.1% A68930 treatment group. Conclusions DRD1 treatment counteracts NLRP3-associated inflammation and facilitates epithelial repair of corneal injury.
Collapse
Affiliation(s)
- Licheng Li
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Centre of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, Fujian Province, China
| | - Yang Yu
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Centre of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, Fujian Province, China
| | - Zihao Zhuang
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Centre of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, Fujian Province, China
| | - Qi Wu
- Group of Neuroendocrinology, Garvan Institute of Medical Research, Sydney, Australia
| | - Shu Lin
- Group of Neuroendocrinology, Garvan Institute of Medical Research, Sydney, Australia
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Jianmin Hu
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Centre of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, Fujian Province, China
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
34
|
Lu X, Chen Z, Lu J, Watsky MA. Effects of 1,25-Vitamin D3 and 24,25-Vitamin D3 on Corneal Nerve Regeneration in Diabetic Mice. Biomolecules 2023; 13:1754. [PMID: 38136625 PMCID: PMC10742127 DOI: 10.3390/biom13121754] [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: 11/07/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Corneal nerve homeostasis is essential for the functional integrity of the ocular surface. Vitamin D deficiency (VDD) and vitamin D receptor knockout (VDR KO) have been found to reduce corneal nerve density in diabetic mice. This is the first study to comprehensively examine the influence of vitamin D on nerve regeneration following corneal epithelial injury in diabetic mice. Corneal nerve regeneration was significantly retarded by diabetes, VDR KO, and VDD, and it was accelerated following topical 1,25 Vit D and 24,25 Vit D administration. Furthermore, topical 1,25 Vit D and 24,25 Vit D increased nerve growth factor, glial cell line-derived neurotropic factor, and neurotropin-3 protein expression, and it increased secretion of GDNF protein from human corneal epithelial cells. CD45+ cells and macrophage numbers were significantly decreased, and vitamin D increased CD45+ cell and macrophage recruitment in these wounded diabetic mouse corneas. The accelerated nerve regeneration observed in these corneas following topical 1,25 Vit D and 24,25 Vit D administration may be related to the vitamin D-stimulated expression, secretion of neurotrophic factors, and recruitment of immune cells.
Collapse
Affiliation(s)
- Xiaowen Lu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15th Street, CB-2901, Augusta, GA 30912, USA
| | | | | | - Mitchell A. Watsky
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15th Street, CB-2901, Augusta, GA 30912, USA
| |
Collapse
|
35
|
Rodrigues-Braz D, Zhu L, Gélizé E, Clarin JP, Chatagnon X, Benzine Y, Rampignon P, Thouvenin A, Bourges JL, Behar-Cohen F, Zhao M. Spironolactone Eyedrop Favors Restoration of Corneal Integrity after Wound Healing in the Rat. Pharmaceuticals (Basel) 2023; 16:1446. [PMID: 37895917 PMCID: PMC10609951 DOI: 10.3390/ph16101446] [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: 07/20/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Abnormal corneal wound healing can compromise corneal transparency and lead to visual impairment. Mineralocorticoid receptor antagonists (MRA) are promising candidates to promote corneal remodeling with anti-inflammatory properties and lack gluococorticoids-associated side effects. In this preclinical study, a new polymer-free hydroxypropyl-gamma-cyclodextrin-based eyedrop containing 0.1% spironolactone (SPL), a potent but non-water-soluble MRA, was investigated for its ocular surface tolerance and efficacy in a rat model of corneal wound healing. SPL eyedrops were stable for up to 9 months at 4 °C. The formulation was well-tolerated since no morphological changes or inflammatory reactions were observed in the rat cornea after multiple daily instillations over 7 days. SPL eyedrops accelerated rat corneal wound healing, reduced corneal edema and inflammation, enhanced epithelial integrity, and improved nerve regeneration, suggesting restoration of corneal homeostasis, while potassium canrenoate, an active and soluble metabolite of SPL, had no effect. SPL eyedrops could benefit patients with impaired corneal wound healing, including that secondary to glucocorticoid therapy. Repurposing known drugs with known excipients will expedite translation to the clinic.
Collapse
Affiliation(s)
- Daniela Rodrigues-Braz
- Centre de Recherche des Cordeliers, Inserm, Université Paris Cité, Sorbonne Université, 75006 Paris, France; (D.R.-B.); (L.Z.); (E.G.); (J.-L.B.); (M.Z.)
| | - Linxin Zhu
- Centre de Recherche des Cordeliers, Inserm, Université Paris Cité, Sorbonne Université, 75006 Paris, France; (D.R.-B.); (L.Z.); (E.G.); (J.-L.B.); (M.Z.)
| | - Emmanuelle Gélizé
- Centre de Recherche des Cordeliers, Inserm, Université Paris Cité, Sorbonne Université, 75006 Paris, France; (D.R.-B.); (L.Z.); (E.G.); (J.-L.B.); (M.Z.)
| | | | | | | | | | - Agathe Thouvenin
- CNRS, Inserm, UTCBS, Université Paris Cité, 75006 Paris, France;
- Département Recherche et Développement Pharmaceutique, Agence Générale des Equipements et Produits de Santé (AGEPS), AP-HP, 75005 Paris, France
| | - Jean-Louis Bourges
- Centre de Recherche des Cordeliers, Inserm, Université Paris Cité, Sorbonne Université, 75006 Paris, France; (D.R.-B.); (L.Z.); (E.G.); (J.-L.B.); (M.Z.)
- Ophtalmopole, AP-HP, Cochin Hospital, 75014 Paris, France
| | - Francine Behar-Cohen
- Centre de Recherche des Cordeliers, Inserm, Université Paris Cité, Sorbonne Université, 75006 Paris, France; (D.R.-B.); (L.Z.); (E.G.); (J.-L.B.); (M.Z.)
- Ophtalmopole, AP-HP, Cochin Hospital, 75014 Paris, France
- Hôpital Foch, Service D’ophtalmologie, 92150 Suresnes, France
| | - Min Zhao
- Centre de Recherche des Cordeliers, Inserm, Université Paris Cité, Sorbonne Université, 75006 Paris, France; (D.R.-B.); (L.Z.); (E.G.); (J.-L.B.); (M.Z.)
| |
Collapse
|
36
|
Su D, Zhang J, Wu Y, Wang W, Wang W, Shao C, Li J. Evaluation of Corneal Nerve Regeneration After Minimally Invasive Corneal Neurotization. Asia Pac J Ophthalmol (Phila) 2023; 12:427-436. [PMID: 37527446 DOI: 10.1097/apo.0000000000000626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/31/2023] [Indexed: 08/03/2023] Open
Abstract
PURPOSE To evaluate the corneal nerve regeneration after minimally invasive corneal neurotization (MICN) and to further clarify the recovery patterns of sensory and trophic functions of the corneal nerves. DESIGN A retrospective cohort study based in the Shanghai Ninth People's Hospital. METHODS Eighteen patients (18 eyes) who underwent MICN for neurotrophic keratopathy due to intracranial surgery was conducted to analyze their follow-up data at 6, 12, 18, and 24 months after surgery. RESULTS At 12 months postoperatively, the growth of the central and peripheral corneal nerve fiber density (CNFD) was 11.47±8.56 and 14.73±8.08 n/mm 2 with subsequent improvement slowing down, and the patient's corneal epithelium defect was healed ahead of the accomplishment of corneal nerve regeneration. The number of dendritic cells also reached its peak. At 18 months postoperatively, the recovery of central and peripheral corneal sensation was 37.22±23.06 mm and 39.38±18.08 mm with no subsequent improvement, and the growth of the central and peripheral corneal nerve branch density (CNBD) was 29.69±11.05 and 43.75±1.41 n/mm 2 , with a positive and significant correlation between corneal sensation and CNBD (at central r =0.632, P <0.005; at peripheral r =0.645, P <0.005). At 24 months postoperatively, mean CNFD, CNBD, and corneal sensation recovered significantly compared with preoperative, but a few patients' corneal sensation recovered insignificantly with good CNFD recovery and poor CNBD recovery. CONCLUSIONS After MICN, the trophic function of the corneal nerve recovers before the sensory function, and in particular, the recovery of sensation is based on the coexistence of the corneal nerve trunk and branches.
Collapse
Affiliation(s)
- Dai Su
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jiaying Zhang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yue Wu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Wenjin Wang
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wang
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunyi Shao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jin Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
37
|
Datta A, Lee JH, Flandrin O, Horneman H, Lee J, Metruccio MME, Bautista D, Evans DJ, Fleiszig SMJ. TRPA1 and TPRV1 Ion Channels Are Required for Contact Lens-Induced Corneal Parainflammation and Can Modulate Levels of Resident Corneal Immune Cells. Invest Ophthalmol Vis Sci 2023; 64:21. [PMID: 37585189 PMCID: PMC10434714 DOI: 10.1167/iovs.64.11.21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/17/2023] [Indexed: 08/17/2023] Open
Abstract
Purpose Contact lens wear can induce corneal parainflammation involving CD11c+ cell responses (24 hours), γδ T cell responses (24 hours and 6 days), and IL-17-dependent Ly6G+ cell responses (6 days). Topical antibiotics blocked these CD11c+ responses. Because corneal CD11c+ responses to bacteria require transient receptor potential (TRP) ion-channels (TRPA1/TRPV1), we determined if these channels mediate lens-induced corneal parainflammation. Methods Wild-type mice were fitted with contact lenses for 24 hours or 6 days and compared to lens wearing TRPA1 (-/-) or TRPV1 (-/-) mice or resiniferatoxin (RTX)-treated mice. Contralateral eyes were not fitted with lenses. Corneas were examined for major histocompatibility complex (MHC) class II+, CD45+, γδ T, or TNF-α+ cell responses (24 hours) or Ly6G+ responses (6 days) by quantitative imaging. The quantitative PCR (qPCR) determined cytokine gene expression. Results Lens-induced increases in MHC class II+ cells after 24 hours were abrogated in TRPV1 (-/-) but not TRPA1 (-/-) mice. Increases in CD45+ cells were unaffected. Increases in γδ T cells after 24 hours of wear were abrogated in TRPA1 (-/-) and TRPV1 (-/-) mice, as were 6 day Ly6G+ cell responses. Contralateral corneas of TRPA1 (-/-) and TRPV1 (-/-) mice showed reduced MHC class II+ and γδ T cells at 24 hours. RTX inhibited lens-induced parainflammatory phenotypes (24 hours and 6 days), blocked lens-induced TNF-α and IL-18 gene expression, TNF-α+ cell infiltration (24 hours), and reduced baseline MHC class II+ cells. Conclusions TRPA1 and TRPV1 mediate contact lens-induced corneal parainflammation after 24 hours and 6 days of wear and can modulate baseline levels of resident corneal immune cells.
Collapse
Affiliation(s)
- Ananya Datta
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
| | - Ji Hyun Lee
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
| | - Orneika Flandrin
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
| | - Hart Horneman
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
| | - Justin Lee
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
| | - Matteo M E Metruccio
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
| | - Diana Bautista
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States
| | - David J Evans
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
- College of Pharmacy, Touro University California, Vallejo, California, United States
| | - Suzanne M J Fleiszig
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, California, United States
- Graduate groups in Vision Science, Microbiology, and Infectious Diseases & Immunity, University of California, Berkeley, California, United States
| |
Collapse
|
38
|
Lu X, Chen Z, Lu J, Watsky M. Effects of Topical 1,25 and 24,25 Vitamin D on Diabetic, Vitamin D Deficient and Vitamin D Receptor Knockout Mouse Corneal Wound Healing. Biomolecules 2023; 13:1065. [PMID: 37509101 PMCID: PMC10377579 DOI: 10.3390/biom13071065] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
Delayed or prolonged corneal wound healing and non-healing corneas put patients at risk for ocular surface infections and subsequent stromal opacification, resulting in discomfort or visual loss. It is important to enhance corneal wound healing efficiency and quality. Vitamin D (Vit D) is both a hormone and a vitamin, and its insufficiency has been linked to immune disorders and diabetes. For this study, wound healing and recruitment of CD45+ cells into the wound area of normoglycemic and diabetic mice were examined following corneal epithelial debridement and treatment with 1,25-dihyroxyvitamin D (1,25 Vit D) or 24,25-dihydroxyvitamin D (24,25 Vit D). Treatment with topical 1,25-dihyroxyvitamin D (1,25 Vit D) resulted in significantly increased corneal wound healing rates of normoglycemic, diabetic and diabetic Vit D deficient mice. Furthermore, 24,25-dihydroxyvitamin D (24,25 Vit D) significantly increased corneal wound healing of diabetic Vit D deficient and Vit D receptor knockout (VDR KO) mice. In addition, CD45+ cell numbers were reduced in diabetic and VDR KO mouse corneas compared to normoglycemic mice, and 24,25 Vit D increased the recruitment of CD45+ cells to diabetic mouse corneas after epithelial debridement. CD45+ cells were found to infiltrate into the corneal basal epithelial layer after corneal epithelial debridement. Our data indicate that topical Vit D promotes corneal wound healing and further supports previous work that the Vit D corneal wound healing effect is not totally VDR-dependent.
Collapse
Affiliation(s)
| | | | | | - Mitchell Watsky
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
39
|
Wareham LK, Holden JM, Bossardet OL, Baratta RO, Del Buono BJ, Schlumpf E, Calkins DJ. Collagen mimetic peptide repair of the corneal nerve bed in a mouse model of dry eye disease. Front Neurosci 2023; 17:1148950. [PMID: 37260844 PMCID: PMC10228686 DOI: 10.3389/fnins.2023.1148950] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/21/2023] [Indexed: 06/02/2023] Open
Abstract
The intraepithelial sub-basal nerve plexus of the cornea is characterized by a central swirl of nerve processes that terminate between the apical cells of the epithelium. This plexus is a critical component of maintaining homeostatic function of the ocular surface. The cornea contains a high concentration of collagen, which is susceptible to damage in conditions such as neuropathic pain, neurotrophic keratitis, and dry eye disease. Here we tested whether topical application of a collagen mimetic peptide (CMP) is efficacious in repairing the corneal sub-basal nerve plexus in a mouse model of ocular surface desiccation. We induced corneal tear film reduction, epithelial damage, and nerve bed degradation through a combination of environmental and pharmaceutical (atropine) desiccation. Mice were subjected to desiccating air flow and bilateral topical application of 1% atropine solution (4× daily) for 2 weeks. During the latter half of this exposure, mice received topical vehicle [phosphate buffered saline (PBS)] or CMP [200 μm (Pro-Pro-Gly)7, 10 μl] once daily, 2 h prior to the first atropine treatment for that day. After euthanasia, cornea were labeled with antibodies against βIII tubulin to visualize and quantify changes to the nerve bed. For mice receiving vehicle only, the two-week desiccation regimen reduced neuronal coverage of the central sub-basal plexus and epithelial terminals compared to naïve, with some corneas demonstrating complete degeneration of nerve beds. Accordingly, both sub-basal and epithelial βIII tubulin-labeled processes demonstrated increased fragmentation, indicative of nerve disassembly. Treatment with CMP significantly reduced nerve fragmentation, expanded both sub-basal and epithelial neuronal coverage compared to vehicle controls, and improved corneal epithelium integrity, tear film production, and corneal sensitivity. Together, these results indicate that topical CMP significantly counters neurodegeneration characteristic of corneal surface desiccation. Repairing underlying collagen in conditions that damage the ocular surface could represent a novel therapeutic avenue in treating a broad spectrum of diseases or injury.
Collapse
Affiliation(s)
- Lauren K. Wareham
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Joseph M. Holden
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Olivia L. Bossardet
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| | | | | | - Eric Schlumpf
- Stuart Therapeutics, Inc., Stuart, FL, United States
| | - David J. Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
40
|
Shu DY, Chaudhary S, Cho KS, Lennikov A, Miller WP, Thorn DC, Yang M, McKay TB. Role of Oxidative Stress in Ocular Diseases: A Balancing Act. Metabolites 2023; 13:187. [PMID: 36837806 PMCID: PMC9960073 DOI: 10.3390/metabo13020187] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Redox homeostasis is a delicate balancing act of maintaining appropriate levels of antioxidant defense mechanisms and reactive oxidizing oxygen and nitrogen species. Any disruption of this balance leads to oxidative stress, which is a key pathogenic factor in several ocular diseases. In this review, we present the current evidence for oxidative stress and mitochondrial dysfunction in conditions affecting both the anterior segment (e.g., dry eye disease, keratoconus, cataract) and posterior segment (age-related macular degeneration, proliferative vitreoretinopathy, diabetic retinopathy, glaucoma) of the human eye. We posit that further development of therapeutic interventions to promote pro-regenerative responses and maintenance of the redox balance may delay or prevent the progression of these major ocular pathologies. Continued efforts in this field will not only yield a better understanding of the molecular mechanisms underlying the pathogenesis of ocular diseases but also enable the identification of novel druggable redox targets and antioxidant therapies.
Collapse
Affiliation(s)
- Daisy Y. Shu
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Suman Chaudhary
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Kin-Sang Cho
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Anton Lennikov
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - William P. Miller
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - David C. Thorn
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Menglu Yang
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Tina B. McKay
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
41
|
Liu F, Liu C, Lee IXY, Lin MTY, Liu YC. Corneal dendritic cells in diabetes mellitus: A narrative review. Front Endocrinol (Lausanne) 2023; 14:1078660. [PMID: 36777336 PMCID: PMC9911453 DOI: 10.3389/fendo.2023.1078660] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/12/2023] [Indexed: 01/28/2023] Open
Abstract
Diabetes mellitus is a global public health problem with both macrovascular and microvascular complications, such as diabetic corneal neuropathy (DCN). Using in-vivo confocal microscopy, corneal nerve changes in DCN patients can be examined. Additionally, changes in the morphology and quantity of corneal dendritic cells (DCs) in diabetic corneas have also been observed. DCs are bone marrow-derived antigen-presenting cells that serve both immunological and non-immunological roles in human corneas. However, the role and pathogenesis of corneal DC in diabetic corneas have not been well understood. In this article, we provide a comprehensive review of both animal and clinical studies that report changes in DCs, including the DC density, maturation stages, as well as relationships between the corneal DCs, corneal nerves, and corneal epithelium, in diabetic corneas. We have also discussed the associations between the changes in corneal DCs and various clinical or imaging parameters, including age, corneal nerve status, and blood metabolic parameters. Such information would provide valuable insight into the development of diagnostic, preventive, and therapeutic strategies for DM-associated ocular surface complications.
Collapse
Affiliation(s)
- Fengyi Liu
- University of Cambridge, Girton College, Cambridgeshire, United Kingdom
| | - Chang Liu
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Isabelle Xin Yu Lee
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Molly Tzu Yu Lin
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Yu-Chi Liu
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore, Singapore
- Cornea and Refractive Surgery Group, Singapore Eye Research Institute, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
- Department of Ophthalmology, National Taiwan University, Taipei, Taiwan
- *Correspondence: Yu-Chi Liu,
| |
Collapse
|
42
|
Puri S, Kenyon BM, Hamrah P. Immunomodulatory Role of Neuropeptides in the Cornea. Biomedicines 2022; 10:1985. [PMID: 36009532 PMCID: PMC9406019 DOI: 10.3390/biomedicines10081985] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/21/2022] Open
Abstract
The transparency of the cornea along with its dense sensory innervation and resident leukocyte populations make it an ideal tissue to study interactions between the nervous and immune systems. The cornea is the most densely innervated tissue of the body and possesses both immune and vascular privilege, in part due to its unique repertoire of resident immune cells. Corneal nerves produce various neuropeptides that have a wide range of functions on immune cells. As research in this area expands, further insights are made into the role of neuropeptides and their immunomodulatory functions in the healthy and diseased cornea. Much remains to be known regarding the details of neuropeptide signaling and how it contributes to pathophysiology, which is likely due to complex interactions among neuropeptides, receptor isoform-specific signaling events, and the inflammatory microenvironment in disease. However, progress in this area has led to an increase in studies that have begun modulating neuropeptide activity for the treatment of corneal diseases with promising results, necessitating the need for a comprehensive review of the literature. This review focuses on the role of neuropeptides in maintaining the homeostasis of the ocular surface, alterations in disease settings, and the possible therapeutic potential of targeting these systems.
Collapse
Affiliation(s)
- Sudan Puri
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Brendan M. Kenyon
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Pedram Hamrah
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
- Departments of Immunology and Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
- Cornea Service, Tufts New England Eye Center, Boston, MA 02111, USA
| |
Collapse
|
43
|
Zhou Q, Yang L, Wang Q, Li Y, Wei C, Xie L. Mechanistic investigations of diabetic ocular surface diseases. Front Endocrinol (Lausanne) 2022; 13:1079541. [PMID: 36589805 PMCID: PMC9800783 DOI: 10.3389/fendo.2022.1079541] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
With the global prevalence of diabetes mellitus over recent decades, more patients suffered from various diabetic complications, including diabetic ocular surface diseases that may seriously affect the quality of life and even vision sight. The major diabetic ocular surface diseases include diabetic keratopathy and dry eye. Diabetic keratopathy is characterized with the delayed corneal epithelial wound healing, reduced corneal nerve density, decreased corneal sensation and feeling of burning or dryness. Diabetic dry eye is manifested as the reduction of tear secretion accompanied with the ocular discomfort. The early clinical symptoms include dry eye and corneal nerve degeneration, suggesting the early diagnosis should be focused on the examination of confocal microscopy and dry eye symptoms. The pathogenesis of diabetic keratopathy involves the accumulation of advanced glycation end-products, impaired neurotrophic innervations and limbal stem cell function, and dysregulated growth factor signaling, and inflammation alterations. Diabetic dry eye may be associated with the abnormal mitochondrial metabolism of lacrimal gland caused by the overactivation of sympathetic nervous system. Considering the important roles of the dense innervations in the homeostatic maintenance of cornea and lacrimal gland, further studies on the neuroepithelial and neuroimmune interactions will reveal the predominant pathogenic mechanisms and develop the targeting intervention strategies of diabetic ocular surface complications.
Collapse
Affiliation(s)
- Qingjun Zhou
- State Key Laboratory Cultivation Base, Eye Institute of Shandong First Medical University, Qingdao, China
- Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Lingling Yang
- State Key Laboratory Cultivation Base, Eye Institute of Shandong First Medical University, Qingdao, China
- Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Qun Wang
- State Key Laboratory Cultivation Base, Eye Institute of Shandong First Medical University, Qingdao, China
- Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Ya Li
- State Key Laboratory Cultivation Base, Eye Institute of Shandong First Medical University, Qingdao, China
- Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Chao Wei
- State Key Laboratory Cultivation Base, Eye Institute of Shandong First Medical University, Qingdao, China
- Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Lixin Xie
- State Key Laboratory Cultivation Base, Eye Institute of Shandong First Medical University, Qingdao, China
- Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
- *Correspondence: Lixin Xie,
| |
Collapse
|