1
|
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
|
2
|
He Y, Feng J, Shi W, Ren Y, Liu Y, Kang H, Tian J, Jie Y. Correlation among ocular surface changes and systemic hematologic indexes and disease activity in primary Sjögren's syndrome: a cross-sectional study. BMC Ophthalmol 2025; 25:270. [PMID: 40329232 PMCID: PMC12054185 DOI: 10.1186/s12886-025-04050-7] [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: 01/09/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND To explore the relationship among ocular surface changes, systemic hematologic indexes, and disease activity in primary Sjögren's syndrome patients. METHODS Thirty-three primary Sjögren's syndrome patients and 36 healthy controls were recruited in this cross-sectional study. All participants underwent complete ocular surface testing, including dry eye symptoms and signs, tear multi-cytokine analysis, and conjunctival impression cytology (CIC). Multiple systemic hematologic indexes and disease activity were also evaluated, including autoantibodies, immune cells, the EULAR Sjögren's Syndrome Patient Reported Index (ESSPRI), and the EULAR Sjögren's Syndrome Disease Activity Index (ESSDAI). RESULTS Primary Sjögren's syndrome patients exhibited significant dry eye, severe conjunctivochalasis, decreased goblet cell density, and severe squamous epithelial on the ocular surface. Interferon-inducible T cell alpha chemoattractant (I-TAC), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin (IL)-1β, IL-5, IL-8, IL-10, IL-13, IL-21, C-C motif chemokine ligand (CCL)4, interferon-gamma (IFN-γ), CCL20, and tumor necrosis factor-gamma (TNF-α) in the tear fluid of pSS patients changed significantly. Correlation analysis showed that anti-SSA was relevant to ocular surface disease index (OSDI) score, tear break-up time (TBUT), and meibomian gland secretion (MGS). CD8+ T cell percentages were relevant to TBUT and corneal fluorescein staining score (CFS). IL-8, IL-13, CCL4, and TNF-α were correlated with RF-IgA. IL-1β, CCL4, and TNF-α were correlated with CD8+ T cell counts. IL-5 and CCL20 were correlated with the ratio of helper T cells and suppressor T cells. Tear I-TAC, IL-8, CCL20, and TNF-α were significantly correlated with the ESSDAI of different domains. CONCLUSIONS Our results revealed that the ocular surface changes in pSS patients were significantly correlated with systemic hematologic indexes and disease activity.
Collapse
Affiliation(s)
- Yan He
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmic and Visual Science Key Laboratory, Beijing, China
| | - Jianing Feng
- Xi'an People's Hospital (Xi'an Fourth Hospital), Shaanxi Eye Hospital, Northwest University Affiliated People's Hospital, Xi'an, Shaanxi, China
| | - Wen Shi
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Yuerong Ren
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Yingyi Liu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmic and Visual Science Key Laboratory, Beijing, China
| | - Huanmin Kang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jing Tian
- Department of Rheumatism and Immunology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Ying Jie
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmic and Visual Science Key Laboratory, Beijing, China.
| |
Collapse
|
3
|
Zhou X, Wu Y, Zhang Y, Chu B, Yang K, Hong J, He Y. Targeting α7 Nicotinic Acetylcholine Receptor for Modulating the Neuroinflammation of Dry Eye Disease Via Macrophages. Invest Ophthalmol Vis Sci 2025; 66:13. [PMID: 40327011 PMCID: PMC12061059 DOI: 10.1167/iovs.66.5.13] [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: 01/25/2025] [Accepted: 04/10/2025] [Indexed: 05/07/2025] Open
Abstract
Purpose Patients with dry eye disease (DED) often exhibit neurological abnormalities and may even suffer from neuropathic pain and pain-related anxiety or depression. The α-7 nicotinic acetylcholine receptor (α7nAChR) is a pivotal regulator in the anti-inflammatory pathway connecting the nervous and immune systems, Here, we investigate the potential of α7nAChR agonist as a novel treatment for DED. Methods We induced DED model by unilateral excision of extraorbital lachrymal gland in C57/BL6 mice. After seven days of treatment, RNA sequencing was performed to identify differentially expressed genes in the cornea of lacrimal gland excision (LGE) mice. Quantitative polymerase chain reaction, Western blotting, and flow cytometry tests were carried out to elucidate neuroinflammation changes after α7nAChR activation. Corneal nerve abnormalities were assessed by corneal esthesiometry and immunofluorescence staining. Results The activation of α7nAChR stimulates genes involved in immune-mediated inflammatory progression and neuroregulation, inhibits the expression of transient receptor potential vanilloid-1, reinstates corneal nerve density, and alleviates anxiety-like behaviors associated with severe DED. Furthermore, we demonstrated that α7nAChR agonist restored corneal nerve abnormality and alleviated inflammation response by down-regulating the proportion of CD86+ M1 macrophages (proinflammatory phenotypes). Conclusions Our findings underscore the activation of α7nAChR as a pioneering therapeutic approach for preserving corneal nerves balance and controlling inflammation in DED.
Collapse
Affiliation(s)
- Xujiao Zhou
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Gene Editing and Cell Therapy for Rare Diseases, Fudan University, Shanghai, China
| | - Yuqing Wu
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Yirou Zhang
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Binbin Chu
- Institute of Functional Nano and Soft Materials Soochow University, Suzhou, China
| | - Kan Yang
- Department of Ophthalmology, The First People's Hospital of Lanzhou City, Gansu, China
| | - Jiaxu Hong
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Gene Editing and Cell Therapy for Rare Diseases, Fudan University, Shanghai, China
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- NHC Key laboratory of Myopia and Related Eye Diseases Shanghai, China
- Shanghai Key Laboratory of Rare Disease Gene Editing and Cell Therapy; Shanghai Engineering Research Center of Synthetic Immunology, Shanghai, China
- Department of Ophthalmology, Children's Hospital of Fudan University, National Pediatric Medical Center of China, Shanghai, China
| | - Yao He
- Macao Translational Medicine Center, Macau University of Science and Technology, Taipa, China
- Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, China
| |
Collapse
|
4
|
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
|
5
|
Harrell CR, Volarevic V. Ion Channels as Potential Drug Targets in Dry Eye Disease and Their Clinical Relevance: A Review. Cells 2024; 13:2017. [PMID: 39682765 PMCID: PMC11639998 DOI: 10.3390/cells13232017] [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: 07/18/2024] [Revised: 10/17/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Dry eye disease (DED) is a common multifactorial disorder characterized by a deficiency in the quality and/or quantity of tear fluid. Tear hyperosmolarity, the dysfunction of ion channel proteins, and eye inflammation are primarily responsible for the development and progression of DED. Alterations in the structure and/or function of ion channel receptors (transient receptor potential ankyrin 1 (TRPA1), transient receptor potential melastatin 8 (TRPM8), transient receptor potential vanilloid 1 and 4 (TRPV1 and TRPV4)), and consequent hyperosmolarity of the tears represent the initial step in the development and progression of DED. Hyperosmolarity triggers the activation of ion channel-dependent signaling pathways in corneal epithelial cells and eye-infiltrated immune cells, leading to the activation of transcriptional factors that enhance the expression of genes regulating inflammatory cytokine production, resulting in a potent inflammatory response in the eyes of DED patients. A persistent and untreated detrimental immune response further modifies the structure and function of ion channel proteins, perpetuating tear hyperosmolarity and exacerbating DED symptoms. Accordingly, suppressing immune cell-driven eye inflammation and alleviating tear hyperosmolarity through the modulation of ion channels in DED patients holds promise for developing new therapeutic strategies. Here, we summarize current knowledge about the molecular mechanisms responsible for the inflammation-induced modification of ion channels leading to tear hyperosmolarity and immune cell dysfunction in DED patients. We also emphasize the therapeutic potential of the newly designed immunomodulatory and hypo-osmotic solution d-MAPPS™ Hypo-Osmotic Ophthalmic Solution, which can activate TRPV4 in corneal epithelial cells, stabilize the tear film, enhance natural cytokine communication, and suppress detrimental immune responses, an important novel approach for DED treatment.
Collapse
Affiliation(s)
| | - Vladislav Volarevic
- Center for Research on Harmful Effects of Biological and Chemical Hazards, Departments of Genetics, Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, 34000 Kragujevac, Serbia;
- Faculty of Pharmacy Novi Sad, Heroja Pinkija 4, 21000 Novi Sad, Serbia
| |
Collapse
|
6
|
Vereertbrugghen A, Pizzano M, Cernutto A, Sabbione F, Keitelman IA, Aguilar DV, Podhorzer A, Fuentes F, Corral-Vázquez C, Guzmán M, Giordano MN, Trevani A, de Paiva CS, Galletti JG. CD4 + T cells drive corneal nerve damage but not epitheliopathy in an acute aqueous-deficient dry eye model. Proc Natl Acad Sci U S A 2024; 121:e2407648121. [PMID: 39560641 PMCID: PMC11621630 DOI: 10.1073/pnas.2407648121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024] Open
Abstract
Dry eye disease (DED) is characterized by a dysfunctional tear film in which the corneal epithelium and its abundant nerves are affected by ocular desiccation and inflammation. Although adaptive immunity and specifically CD4+ T cells play a role in DED pathogenesis, the exact contribution of these cells to corneal epithelial and neural damage remains undetermined. To address this, we explored the progression of a surgical DED model in wild-type (WT) and T cell-deficient mice. We observed that adaptive immune-deficient mice developed all aspects of DED comparably to WT mice except for the absence of functional and morphological corneal nerve changes, nerve damage-associated transcriptomic signature in the trigeminal ganglia, and sustained tear cytokine levels. Adoptive transfer of CD4+ T cells from WT DED mice to T cell-deficient mice reproduced corneal nerve damage but not epitheliopathy. Conversely, T cell-deficient mice reconstituted solely with naïve CD4+ T cells developed corneal nerve impairment and epitheliopathy upon DED induction, thus replicating the WT DED phenotype. Collectively, our data show that while corneal neuropathy is driven by CD4+ T cells in DED, corneal epithelial damage develops independently of the adaptive immune response. These findings have implications for T cell-targeting therapies currently in use for DED.
Collapse
Affiliation(s)
- Alexia Vereertbrugghen
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Manuela Pizzano
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Agostina Cernutto
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Florencia Sabbione
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Irene A. Keitelman
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Douglas Vera Aguilar
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Ariel Podhorzer
- Flow Cytometry Unit, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Federico Fuentes
- Confocal Microscopy Unit, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Celia Corral-Vázquez
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
| | - Mauricio Guzmán
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
| | - Mirta N. Giordano
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Analía Trevani
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | | | - Jeremías G. Galletti
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| |
Collapse
|
7
|
Cong L, Qi B, Ma W, Ren Z, Liang Q, Zhou Q, Zhang BN, Xie L. Preventing and treating neurotrophic keratopathy by a single intrastromal injection of AAV-mediated gene therapy. Ocul Surf 2024; 34:406-414. [PMID: 39362525 DOI: 10.1016/j.jtos.2024.09.010] [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: 06/19/2024] [Revised: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
PURPOSE Neurotrophic keratopathy (NK) is a degenerative corneal condition resulting from corneal nerve injury. Current therapies, including the recombinant human nerve growth factor (rhNGF) therapy, requires continuous administration. This study aims to develop a novel and highly effective gene therapy strategy for the prevention and treatment of NK. METHODS Adeno-associated virus (AAV) was transduced into corneal stromal cells by intrastromal injection. Three dimensional corneal wholemount imaging with co-immunostaining of ZO-1 and tubulin was utilized to assess the transduction of AAV.rh10. The efficacy of prevention and treatment of NK by a single intrastromal injection of AAV-Ngf was tested using capsaicin mouse model, herpes simplex keratitis (HSK) model, type Ⅱ diabetes model and alkali burn model. rhNGF eye drops served as the positive control. RESULTS Intrastromal injection of AAV.rh10 efficiently transduced the subepithelial nerve plexus and retrogradely transported to the trigeminal ganglion (TG). A single injection of AAV.rh10-Ngf can significantly promote corneal nerve repair, accelerate corneal epithelial repair, reduce corneal stromal edema, and improve corneal sensitivity across the four NK models. The therapeutic effects were consistent with those achieved by continuous administration of rhNGF drops by 6 times daily. CONCLUSIONS This proof-of-concept study demonstrates that AAV.rh10-Ngf gene therapy is a promising method for preventing and treating of NK. Our results underline the potential for developing clinical trials to further explore the safety and efficacy of such gene therapy.
Collapse
Affiliation(s)
- Lin Cong
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China; School of Ophthalmology, Shandong First Medical University, Qingdao, China
| | - Benxiang Qi
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China; School of Ophthalmology, Shandong First Medical University, Qingdao, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, China
| | - Wenhui Ma
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China; School of Ophthalmology, Shandong First Medical University, Qingdao, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, China
| | - Zhongmei Ren
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China; School of Ophthalmology, Shandong First Medical University, Qingdao, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, China
| | - Qian Liang
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Qingjun Zhou
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China; School of Ophthalmology, Shandong First Medical University, Qingdao, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, China
| | - Bi Ning Zhang
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China; School of Ophthalmology, Shandong First Medical University, Qingdao, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, China.
| | - Lixin Xie
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China; School of Ophthalmology, Shandong First Medical University, Qingdao, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, China.
| |
Collapse
|
8
|
Vereertbrugghen A, Pizzano M, Sabbione F, del Papa MS, Rodríguez G, Passerini MS, Galletti JG. Hyaluronate Protects From Benzalkonium Chloride-Induced Ocular Surface Toxicity. Transl Vis Sci Technol 2024; 13:31. [PMID: 39432403 PMCID: PMC11498636 DOI: 10.1167/tvst.13.10.31] [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: 05/15/2024] [Accepted: 09/16/2024] [Indexed: 10/23/2024] Open
Abstract
Purpose The purpose of this study was to investigate the effect of sodium hyaluronate (SH) on benzalkonium chloride (BAK)-induced toxicity in the ocular surface epithelium and corneal nerves. Methods Ocular surface epithelial cells from Balb/c mice were cultured with 0.1% to 0.4% SH and 0.001% to 0.01% BAK and their metabolic activity, viability, and wound repair capacity were assessed in vitro. Following a controlled corneal wound, re-epithelialization and recovery of epithelial barrier function and mechanosensitivity were measured in Balb/c mice treated with 0.4% SH 3 times/day and 0.01% BAK twice daily for 3 weeks. Nerve morphology was assessed by confocal microscopy of corneal whole mounts. Results Whereas BAK exposure reduced metabolic activity, viability, and wound repair ability of ocular epithelial cells in vitro, pretreatment with SH ameliorated BAK toxicity in a concentration-dependent manner. The highest SH concentration partially reversed the effects of 0.01% BAK in vitro and increased the corneal healing rate of BAK-exposed mice. Although all corneal wounds closed after 4 days, continuous SH treatment improved corneal barrier dysfunction 18 days after wounding and accelerated the recovery of corneal mechanical sensitivity to baseline levels in BAK-exposed mice. SH treatment also increased corneal nerve density in the wounded area after 3 weeks. Conclusions SH mitigates BAK-associated ocular epithelial and neurotoxicity in a concentration-dependent manner. Translational Relevance Commercially available, high-concentration SH formulations may have added benefits in treating BAK-associated ocular surface toxicity.
Collapse
Affiliation(s)
- Alexia Vereertbrugghen
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Manuela Pizzano
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Florencia Sabbione
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | | | | | | | - Jeremías G. Galletti
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| |
Collapse
|