Pub Date : 2024-12-19DOI: 10.1016/j.exer.2024.110217
Lingli Kong, Jiali Sun, Razzagh Abedi-Firouzjah
Dry eye disease (DED) is a common ocular condition characterized by chronic inflammation and tear film disruption. It affects millions of people worldwide, causing significant eye discomfort and vision disturbances. Despite its prevalence, DED remains a complex condition that is not yet fully understood. It can arise from various ocular pathologies, including endocrinological disorders such as diabetes and Graves' orbitopathy. Natural products, including plant- and blood-based therapies, have shown promise in alleviating DED symptoms and may represent effective therapeutic approaches. In this study, we review recent research on natural product treatments for DED, focusing on blood-derived therapies (e.g., autologous serum, albumin serum, and allogeneic serum) and plant-based compounds such as omega-3 fatty acids (O3FA), omega-6 fatty acids (O6FA), antioxidants, polyphenols, and flavonoids. Additionally, we examine the efficacy, mechanisms of action, and delivery systems of these treatments, highlighting the potential of blood-derived therapies, polyphenols, and flavonoids to improve or treat DED through multiple mechanisms. However, the use of these natural products as instilled drugs is limited by challenges such as solubility, stability, and biological barriers. Finally, we discuss drug delivery systems and structural modifications designed to enhance the therapeutic effects of these treatments, emphasizing their potential in managing DED.
{"title":"Emerging treatment strategies in dry eye disease: Potential of blood-derived approaches and natural plant-based products.","authors":"Lingli Kong, Jiali Sun, Razzagh Abedi-Firouzjah","doi":"10.1016/j.exer.2024.110217","DOIUrl":"10.1016/j.exer.2024.110217","url":null,"abstract":"<p><p>Dry eye disease (DED) is a common ocular condition characterized by chronic inflammation and tear film disruption. It affects millions of people worldwide, causing significant eye discomfort and vision disturbances. Despite its prevalence, DED remains a complex condition that is not yet fully understood. It can arise from various ocular pathologies, including endocrinological disorders such as diabetes and Graves' orbitopathy. Natural products, including plant- and blood-based therapies, have shown promise in alleviating DED symptoms and may represent effective therapeutic approaches. In this study, we review recent research on natural product treatments for DED, focusing on blood-derived therapies (e.g., autologous serum, albumin serum, and allogeneic serum) and plant-based compounds such as omega-3 fatty acids (O3FA), omega-6 fatty acids (O6FA), antioxidants, polyphenols, and flavonoids. Additionally, we examine the efficacy, mechanisms of action, and delivery systems of these treatments, highlighting the potential of blood-derived therapies, polyphenols, and flavonoids to improve or treat DED through multiple mechanisms. However, the use of these natural products as instilled drugs is limited by challenges such as solubility, stability, and biological barriers. Finally, we discuss drug delivery systems and structural modifications designed to enhance the therapeutic effects of these treatments, emphasizing their potential in managing DED.</p>","PeriodicalId":12177,"journal":{"name":"Experimental eye research","volume":" ","pages":"110217"},"PeriodicalIF":3.0,"publicationDate":"2024-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142871862","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-18DOI: 10.1016/j.exer.2024.110213
Nishant R Sinha, Alexandria C Hofmann, Laila A Suleiman, Riley Laub, Ratnakar Tripathi, Shyam S Chaurasia, Rajiv R Mohan
Sulfur mustard gas (SM), an alkylating and vesicating agent, has been used frequently in many wars and conflicts. SM exposure to the eye results in several corneal abnormalities including scar/fibrosis formation. However, molecular mechanism for SM induced corneal fibrosis development is poorly understood. After SM insult to the eye, excessive synthesis/secretion of extracellular matrix components (ECM) including collagen (COL) I, COL III, and lysyl oxidase (LOX) by corneal myofibroblasts causes corneal fibrosis, however, precise mechanism remains elusive. This study tested the hypothesis that Phosphoinositide 3-kinase (PI3K) signaling alterations post SM in cornea enhances stromal ECM synthesis and corneal fibrosis. New Zealand White Rabbits were used. The right eyes were exposed to SM (200 mg-min/m3) and left eye to the air for 8min at MRI Global. Rabbit corneas were collected on day-3, day-7, and day-14 for molecular analysis. SM exposure caused a significant increase in mRNA expression of PI3K, AKT, COL I, COL III, and LOX and protein levels of LOX in a time-dependent manner in rabbit corneas. The in vitro studies were performed with human corneal stromal fibroblasts (hCSFs) by growing cultures in -/+ nitrogen mustard (NM) and LY294002, a PI3K specific inhibitor, for 30min, 8h, 24h, 48h, and 72h. NM significantly increased mRNA and protein levels of PI3K, AKT, COL I, COL III, and LOX. On the contrary, LY294002 in NM hCSFs significantly reduced PI3K, AKT, COL I, COL III, and LOX protein expression. We concluded that PI3K signaling mediates stromal collagen synthesis and LOX production following SM injury.
{"title":"PI3K signaling and lysyl oxidase is critical to corneal stroma fibrosis following mustard gas injury.","authors":"Nishant R Sinha, Alexandria C Hofmann, Laila A Suleiman, Riley Laub, Ratnakar Tripathi, Shyam S Chaurasia, Rajiv R Mohan","doi":"10.1016/j.exer.2024.110213","DOIUrl":"10.1016/j.exer.2024.110213","url":null,"abstract":"<p><p>Sulfur mustard gas (SM), an alkylating and vesicating agent, has been used frequently in many wars and conflicts. SM exposure to the eye results in several corneal abnormalities including scar/fibrosis formation. However, molecular mechanism for SM induced corneal fibrosis development is poorly understood. After SM insult to the eye, excessive synthesis/secretion of extracellular matrix components (ECM) including collagen (COL) I, COL III, and lysyl oxidase (LOX) by corneal myofibroblasts causes corneal fibrosis, however, precise mechanism remains elusive. This study tested the hypothesis that Phosphoinositide 3-kinase (PI3K) signaling alterations post SM in cornea enhances stromal ECM synthesis and corneal fibrosis. New Zealand White Rabbits were used. The right eyes were exposed to SM (200 mg-min/m<sup>3</sup>) and left eye to the air for 8min at MRI Global. Rabbit corneas were collected on day-3, day-7, and day-14 for molecular analysis. SM exposure caused a significant increase in mRNA expression of PI3K, AKT, COL I, COL III, and LOX and protein levels of LOX in a time-dependent manner in rabbit corneas. The in vitro studies were performed with human corneal stromal fibroblasts (hCSFs) by growing cultures in -/+ nitrogen mustard (NM) and LY294002, a PI3K specific inhibitor, for 30min, 8h, 24h, 48h, and 72h. NM significantly increased mRNA and protein levels of PI3K, AKT, COL I, COL III, and LOX. On the contrary, LY294002 in NM hCSFs significantly reduced PI3K, AKT, COL I, COL III, and LOX protein expression. We concluded that PI3K signaling mediates stromal collagen synthesis and LOX production following SM injury.</p>","PeriodicalId":12177,"journal":{"name":"Experimental eye research","volume":" ","pages":"110213"},"PeriodicalIF":3.0,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142871933","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-18DOI: 10.1016/j.exer.2024.110214
Danyang Che, Danlei Qiao, Lingfeng Lv, Yingjie Zhang, Yiting Cao, Fang Li, Shanbao Tong, Peng Miao, Jibo Zhou
We investigated the mechanism of action of atropine in myopia control by examining its effect on choroidal hemodynamics. Blood flow was evaluated using indocyanine green (ICG) fluorescence and molecular variation during the development of form-deprivation myopia (FDM) and atropine treatment in guinea pigs. Guinea pigs were divided randomly into the normal control (NC), FDM, and FDM + 1% atropine (ATR) groups, and evaluated by spherical equivalent refractive error (SE) and axial length (AL). Choroidal hemodynamic parameters were measured via ICG fluorescence imaging including the maximal ICG fluorescence intensity (Imax), rising time (Trising), blood flow index (BFI), and mean transit time (MTT). Additionally, the expression in the choroid-RPE complex of choroidal vascular endothelial growth factor A (VEGFA) and HIF-1 α were assessed via Western blotting. Atropine inhibited the development of FDM, with effects of FD on both SE and AL being reduced. ICG fluorescence hemodynamic wide-field maps and time-series curves revealed that the atropine significantly accelerated choroidal blood flow, with reduced Trising and MTT, while increasing Imax, BFI and the number of lobulated choriocapillaris structures compared with the FDM group. In terms of molecular markers, atropine inhibited the effect of FDM, increasing VEGFA levels and reducing HIF-1α expression. These findings suggest that atropine improved choroidal hemodynamics and changed vascular markers, potentially contributing to its role in inhibiting the progression of myopia in the FDM model.
{"title":"Effects of atropine on choroidal hemodynamics and VEGFA and HIF-1α expression in form-deprivation myopia guinea pigs.","authors":"Danyang Che, Danlei Qiao, Lingfeng Lv, Yingjie Zhang, Yiting Cao, Fang Li, Shanbao Tong, Peng Miao, Jibo Zhou","doi":"10.1016/j.exer.2024.110214","DOIUrl":"10.1016/j.exer.2024.110214","url":null,"abstract":"<p><p>We investigated the mechanism of action of atropine in myopia control by examining its effect on choroidal hemodynamics. Blood flow was evaluated using indocyanine green (ICG) fluorescence and molecular variation during the development of form-deprivation myopia (FDM) and atropine treatment in guinea pigs. Guinea pigs were divided randomly into the normal control (NC), FDM, and FDM + 1% atropine (ATR) groups, and evaluated by spherical equivalent refractive error (SE) and axial length (AL). Choroidal hemodynamic parameters were measured via ICG fluorescence imaging including the maximal ICG fluorescence intensity (I<sub>max</sub>), rising time (T<sub>rising</sub>), blood flow index (BFI), and mean transit time (MTT). Additionally, the expression in the choroid-RPE complex of choroidal vascular endothelial growth factor A (VEGFA) and HIF-1 α were assessed via Western blotting. Atropine inhibited the development of FDM, with effects of FD on both SE and AL being reduced. ICG fluorescence hemodynamic wide-field maps and time-series curves revealed that the atropine significantly accelerated choroidal blood flow, with reduced T<sub>rising</sub> and MTT, while increasing I<sub>max</sub>, BFI and the number of lobulated choriocapillaris structures compared with the FDM group. In terms of molecular markers, atropine inhibited the effect of FDM, increasing VEGFA levels and reducing HIF-1α expression. These findings suggest that atropine improved choroidal hemodynamics and changed vascular markers, potentially contributing to its role in inhibiting the progression of myopia in the FDM model.</p>","PeriodicalId":12177,"journal":{"name":"Experimental eye research","volume":" ","pages":"110214"},"PeriodicalIF":3.0,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142871860","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-17DOI: 10.1016/j.exer.2024.110212
Yongzhen Yu, Mengyi Zhang, Wenjie Zhou, Yangyang Yu, Liulian Jian, Yuping Zou, Long Pang, Xiulan Zou
Diabetes mellitus (DM) is always accompanied by various complications, where diabetic retinopathy was a serious microvascular complications threatening the visual function of patients. This study evaluated the significance of miR-151a-5p and its effect on DR progression aiming to explore a novel biomarker for disease screening and monitoring. Study enrolled 137 patients with DM and 103 diabetes patients with DR. Serum miR-151a-5p was compared with PCR, and its clinical significance was evaluated from the perspectives of diagnosis and severity prediction. High-glucose-treated human retinal cell model was established, the effect of miR-151a-5p on high-glucose-induced cell injury was assessed based on cell growth, inflammation, oxidative stress, and endoplasmic reticulum stress. In mechanism, the downstream targets of miR-151a-5p were predicted, based on the function enrichment, the involvement of DKK3 and the MAPK signaling was estimated. Increasing miR-151a-5p was identified as a risk factor for DR in DM patients diagnosing DR patients and was positively correlated with disease severity predicting severe development of DR. Silencing miR-151a-5p alleviated high-glucose-induced reducing proliferation, activated inflammation, oxidative stress, and endoplasmic reticulum stress in human retinal cells. Negative regulation of DKK3 by miR-151a-5p was observed, and the knockdown of DKK3 could reversed the protective effect of miR-151a-5p. High-glucose activate the MAPK signaling, which was suppressed by the miR-151a-5p/DKK3 axis, and MAPK signaling was demonstrated to mediate the functional role of the miR-151a-5p/DKK3 axis. miR-151a-5p can be considered as a biomarker for the onset and progression of DR. miR-151a-5p potentially modulates the progression of DR through regulating inflammation, oxidative stress, and endoplasmic reticulum stress via the MAPK signaling.
{"title":"miR-151a-5p predicts severity of diabetic retinopathy and protects from retinal cell injury by inactivating MAPK signaling via DKK3.","authors":"Yongzhen Yu, Mengyi Zhang, Wenjie Zhou, Yangyang Yu, Liulian Jian, Yuping Zou, Long Pang, Xiulan Zou","doi":"10.1016/j.exer.2024.110212","DOIUrl":"10.1016/j.exer.2024.110212","url":null,"abstract":"<p><p>Diabetes mellitus (DM) is always accompanied by various complications, where diabetic retinopathy was a serious microvascular complications threatening the visual function of patients. This study evaluated the significance of miR-151a-5p and its effect on DR progression aiming to explore a novel biomarker for disease screening and monitoring. Study enrolled 137 patients with DM and 103 diabetes patients with DR. Serum miR-151a-5p was compared with PCR, and its clinical significance was evaluated from the perspectives of diagnosis and severity prediction. High-glucose-treated human retinal cell model was established, the effect of miR-151a-5p on high-glucose-induced cell injury was assessed based on cell growth, inflammation, oxidative stress, and endoplasmic reticulum stress. In mechanism, the downstream targets of miR-151a-5p were predicted, based on the function enrichment, the involvement of DKK3 and the MAPK signaling was estimated. Increasing miR-151a-5p was identified as a risk factor for DR in DM patients diagnosing DR patients and was positively correlated with disease severity predicting severe development of DR. Silencing miR-151a-5p alleviated high-glucose-induced reducing proliferation, activated inflammation, oxidative stress, and endoplasmic reticulum stress in human retinal cells. Negative regulation of DKK3 by miR-151a-5p was observed, and the knockdown of DKK3 could reversed the protective effect of miR-151a-5p. High-glucose activate the MAPK signaling, which was suppressed by the miR-151a-5p/DKK3 axis, and MAPK signaling was demonstrated to mediate the functional role of the miR-151a-5p/DKK3 axis. miR-151a-5p can be considered as a biomarker for the onset and progression of DR. miR-151a-5p potentially modulates the progression of DR through regulating inflammation, oxidative stress, and endoplasmic reticulum stress via the MAPK signaling.</p>","PeriodicalId":12177,"journal":{"name":"Experimental eye research","volume":" ","pages":"110212"},"PeriodicalIF":3.0,"publicationDate":"2024-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142863868","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-16DOI: 10.1016/j.exer.2024.110209
Anne Nathalie Longakit, Hannah Bourget, Catherine D Van Raamsdonk
The Mitf transcription factor is a critical regulator of the melanocyte lineage and eye development. Mitf activity in different cell types is controlled in part by ten alternative promoters and their resulting isoforms. A useful tool for melanocyte-based research, Mitf-cre was designed to express Cre from the Mitf-M promoter, which is melanocyte specific. However, Mitf-cre mice are also microphthalmic, perhaps because of insertional mutagenesis or disrupted gene expression. Here, we investigated these possibilities and described the eye phenotype. Targeted locus amplification indicated that the transgene integrated on chromosome 2, in between Spred1 and Meis2. The BAC transgene used to make Mitf-cre was larger than expected, carrying three upstream alternative promoters, Mitf-H, Mitf-D, and Mitf-B, which could express their isoforms intact off the transgene. RT-qPCR using eye tissue demonstrated a 5-fold increase in Mitf transcripts containing exon 1B1b, which is shared by Mitf-H, Mitf-D, and Mitf-B, while Spred1 and Meis2 did not differ in their expression. These findings clarify and support the usage of Mitf-cre in conditional mutagenesis in melanocytes. The specific over-expression of these isoforms, which are preferentially expressed in the RPE, presents a unique resource for those interested in eye development and coloboma.
{"title":"Mitf over-expression leads to microphthalmia and coloboma in Mitf-cre mice.","authors":"Anne Nathalie Longakit, Hannah Bourget, Catherine D Van Raamsdonk","doi":"10.1016/j.exer.2024.110209","DOIUrl":"10.1016/j.exer.2024.110209","url":null,"abstract":"<p><p>The Mitf transcription factor is a critical regulator of the melanocyte lineage and eye development. Mitf activity in different cell types is controlled in part by ten alternative promoters and their resulting isoforms. A useful tool for melanocyte-based research, Mitf-cre was designed to express Cre from the Mitf-M promoter, which is melanocyte specific. However, Mitf-cre mice are also microphthalmic, perhaps because of insertional mutagenesis or disrupted gene expression. Here, we investigated these possibilities and described the eye phenotype. Targeted locus amplification indicated that the transgene integrated on chromosome 2, in between Spred1 and Meis2. The BAC transgene used to make Mitf-cre was larger than expected, carrying three upstream alternative promoters, Mitf-H, Mitf-D, and Mitf-B, which could express their isoforms intact off the transgene. RT-qPCR using eye tissue demonstrated a 5-fold increase in Mitf transcripts containing exon 1B1b, which is shared by Mitf-H, Mitf-D, and Mitf-B, while Spred1 and Meis2 did not differ in their expression. These findings clarify and support the usage of Mitf-cre in conditional mutagenesis in melanocytes. The specific over-expression of these isoforms, which are preferentially expressed in the RPE, presents a unique resource for those interested in eye development and coloboma.</p>","PeriodicalId":12177,"journal":{"name":"Experimental eye research","volume":" ","pages":"110209"},"PeriodicalIF":3.0,"publicationDate":"2024-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142853412","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-16DOI: 10.1016/j.exer.2024.110211
Ehsan Misaghi, Peter Kannu, Ian M MacDonald, Matthew D Benson
Many studies have identified disease-causing variants of PIKFYVE in ocular tissues; however, a comprehensive review of these variants and their ocular phenotypes is lacking. The phosphoinositide kinase PIKFYVE plays crucial roles in the endolysosomal pathway in autophagy and phagocytosis, both essential for cellular homeostasis. In this review, we evaluate the reported disease-causing PIKFYVE variants and their associated phenotypes in humans to identify potential genotype-phenotype correlations. Variants in PIKFYVE have been associated with corneal fleck dystrophy, congenital cataracts and possibly keratoconus. There are unvalidated associations of variants in PIKFYVE with autism spectrum disorder and congenital heart disease. We show that variants causing corneal fleck dystrophy exist in the chaperonin-like domain of PIKFYVE as well as the region between the chaperonin-like and the kinase domains. Similarly, congenital cataract variants appear to be specific to the kinase domain of the protein. This review consolidates existing knowledge on PIKFYVE variants in ocular disease and bridges fundamental science and clinical manifestations, potentially informing future diagnostic and treatment strategies for PIKFYVE-associated ocular disorders.
{"title":"Genetic variants in PIKFYVE: A review of ocular phenotypes.","authors":"Ehsan Misaghi, Peter Kannu, Ian M MacDonald, Matthew D Benson","doi":"10.1016/j.exer.2024.110211","DOIUrl":"10.1016/j.exer.2024.110211","url":null,"abstract":"<p><p>Many studies have identified disease-causing variants of PIKFYVE in ocular tissues; however, a comprehensive review of these variants and their ocular phenotypes is lacking. The phosphoinositide kinase PIKFYVE plays crucial roles in the endolysosomal pathway in autophagy and phagocytosis, both essential for cellular homeostasis. In this review, we evaluate the reported disease-causing PIKFYVE variants and their associated phenotypes in humans to identify potential genotype-phenotype correlations. Variants in PIKFYVE have been associated with corneal fleck dystrophy, congenital cataracts and possibly keratoconus. There are unvalidated associations of variants in PIKFYVE with autism spectrum disorder and congenital heart disease. We show that variants causing corneal fleck dystrophy exist in the chaperonin-like domain of PIKFYVE as well as the region between the chaperonin-like and the kinase domains. Similarly, congenital cataract variants appear to be specific to the kinase domain of the protein. This review consolidates existing knowledge on PIKFYVE variants in ocular disease and bridges fundamental science and clinical manifestations, potentially informing future diagnostic and treatment strategies for PIKFYVE-associated ocular disorders.</p>","PeriodicalId":12177,"journal":{"name":"Experimental eye research","volume":" ","pages":"110211"},"PeriodicalIF":3.0,"publicationDate":"2024-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142853408","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-15DOI: 10.1016/j.exer.2024.110210
Zhihao Wang, Long Guo, Pengfei Dong, Xinyi Zhu, Jianji Li, Luying Cui, Junsheng Dong, Kangjun Liu, Xia Meng, Heng Wang
The resistance of pathogenic bacteria to various clinical antibiotics is the major problem in treating bacterial keratitis. Dimethyl fumarate (DMF) has good anti-fungal and anti-inflammatory effects in fungal keratitis, but its effect on bacterial keratitis is unclear. This study aims to investigate DMF's anti-inflammatory and antibacterial effects. The pyroptosis model was constructed by intracellular infection of canine corneal epithelial cells (CCECs) with Staphylococcus pseudintermedius (S. pseudintermedius), and 200 μM DMF was added to explore its function. Western blot, ELISA, immunostaining, flow cytometry, qRT-PCR, and bacterial counts were used to examine the expression of the NLRP3-GSDMD signaling pathway, virulence genes, and oxidant mediators. 111 clinical keratitis isolates or S. pseudintermedius were treated with different concentrations of DMF to detect bacterial growth and biofilm formation. Adding DMF resulted in the inhibition of the NLRP3-GSDMD pathway while activating the NRF2 pathway. This led to a decrease in pyroptosis rate, intracellular bacteria count, and ROS content. Additionally, DMF blocked the mRNA expression of virulence genes ebpS, hlgB, siet, lukS-I, PVL, icaA, icaD, spsD, and spsL associated with S. pseudintermedius infection. Furthermore, DMF demonstrated concentration-dependent inhibition of the growth of clinical isolates and the formation of S. pseudintermedius biofilm. In conclusion, our results indicate that DMF can inhibit pyroptosis and the growth of various clinical isolates, making it a novel ophthalmic drug with anti-inflammatory and antibacterial properties.
{"title":"Dimethyl fumarate alleviates Staphylococcus pseudintermedius-induced cell damage by inhibiting pyroptosis and bacterial virulence.","authors":"Zhihao Wang, Long Guo, Pengfei Dong, Xinyi Zhu, Jianji Li, Luying Cui, Junsheng Dong, Kangjun Liu, Xia Meng, Heng Wang","doi":"10.1016/j.exer.2024.110210","DOIUrl":"10.1016/j.exer.2024.110210","url":null,"abstract":"<p><p>The resistance of pathogenic bacteria to various clinical antibiotics is the major problem in treating bacterial keratitis. Dimethyl fumarate (DMF) has good anti-fungal and anti-inflammatory effects in fungal keratitis, but its effect on bacterial keratitis is unclear. This study aims to investigate DMF's anti-inflammatory and antibacterial effects. The pyroptosis model was constructed by intracellular infection of canine corneal epithelial cells (CCECs) with Staphylococcus pseudintermedius (S. pseudintermedius), and 200 μM DMF was added to explore its function. Western blot, ELISA, immunostaining, flow cytometry, qRT-PCR, and bacterial counts were used to examine the expression of the NLRP3-GSDMD signaling pathway, virulence genes, and oxidant mediators. 111 clinical keratitis isolates or S. pseudintermedius were treated with different concentrations of DMF to detect bacterial growth and biofilm formation. Adding DMF resulted in the inhibition of the NLRP3-GSDMD pathway while activating the NRF2 pathway. This led to a decrease in pyroptosis rate, intracellular bacteria count, and ROS content. Additionally, DMF blocked the mRNA expression of virulence genes ebpS, hlgB, siet, lukS-I, PVL, icaA, icaD, spsD, and spsL associated with S. pseudintermedius infection. Furthermore, DMF demonstrated concentration-dependent inhibition of the growth of clinical isolates and the formation of S. pseudintermedius biofilm. In conclusion, our results indicate that DMF can inhibit pyroptosis and the growth of various clinical isolates, making it a novel ophthalmic drug with anti-inflammatory and antibacterial properties.</p>","PeriodicalId":12177,"journal":{"name":"Experimental eye research","volume":" ","pages":"110210"},"PeriodicalIF":3.0,"publicationDate":"2024-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142834844","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Keratoconus (KC) is the most common ectatic corneal disease with unknown pathogenesis. This study aimed to investigate the role of methyltransferase-like enzyme 3 (METTL3) in KC pathogenesis. In the present study, we examined the levels of METTL3 and other N6-methyladenosine (m6A) modification-related proteins in KC samples and human stromal keratocyte (HTK) cells stimulated by mechanical stretch (MS) using Western blotting and immunohistochemistry. The level of m6A RNA methylation was quantified using the m6A RNA methylation assay kit. Genetic (Mettl3 knockdown mice) and pharmacological (STM2457) approaches were employed to investigate the effect of METTL3 on the expression of metalloproteinases (MMPs) in MS-treated corneal stromal cells (CSCs) via Western blotting and real-time polymerase chain reaction. Moreover, YAP signaling activity was assessed to explore the relationship between METTL3 and MMPs in MS-treated CSCs. Increased expression of METTL3 and decreased expression of METTL14, WTAP, and YTHDF2 were detected in KC samples and MS-stimulated HTK cells. Correspondingly, the m6A levels in KC specimens and MS-stimulated CSCs were significantly higher than those in controls but were significantly reduced when METTL3 activity was genetically and pharmacologically blocked. Inhibition of METTL3 significantly reduced the expression of MMP1 and MMP3 in mechanically stretched CSCs and reduced YAP activity. Furthermore, pharmacologically inhibiting YAP signaling in MS-stimulated HTK cells significantly reduced MMP1 and MMP3 expression. Our findings highlight the pathogenic role of METTL3 in KC. Further investigation is required to investigate the underlying mechanism.
{"title":"Role of m<sup>6</sup>A methyltransferase METTL3 in keratoconus pathogenesis.","authors":"Huimin Yu, Shengqian Dou, Huijin Wang, Yaru Sun, Junpeng Qu, Ting Liu, Xiaoxue Liu, Chao Wei, Hua Gao","doi":"10.1016/j.exer.2024.110207","DOIUrl":"10.1016/j.exer.2024.110207","url":null,"abstract":"<p><p>Keratoconus (KC) is the most common ectatic corneal disease with unknown pathogenesis. This study aimed to investigate the role of methyltransferase-like enzyme 3 (METTL3) in KC pathogenesis. In the present study, we examined the levels of METTL3 and other N6-methyladenosine (m<sup>6</sup>A) modification-related proteins in KC samples and human stromal keratocyte (HTK) cells stimulated by mechanical stretch (MS) using Western blotting and immunohistochemistry. The level of m<sup>6</sup>A RNA methylation was quantified using the m<sup>6</sup>A RNA methylation assay kit. Genetic (Mettl3 knockdown mice) and pharmacological (STM2457) approaches were employed to investigate the effect of METTL3 on the expression of metalloproteinases (MMPs) in MS-treated corneal stromal cells (CSCs) via Western blotting and real-time polymerase chain reaction. Moreover, YAP signaling activity was assessed to explore the relationship between METTL3 and MMPs in MS-treated CSCs. Increased expression of METTL3 and decreased expression of METTL14, WTAP, and YTHDF2 were detected in KC samples and MS-stimulated HTK cells. Correspondingly, the m<sup>6</sup>A levels in KC specimens and MS-stimulated CSCs were significantly higher than those in controls but were significantly reduced when METTL3 activity was genetically and pharmacologically blocked. Inhibition of METTL3 significantly reduced the expression of MMP1 and MMP3 in mechanically stretched CSCs and reduced YAP activity. Furthermore, pharmacologically inhibiting YAP signaling in MS-stimulated HTK cells significantly reduced MMP1 and MMP3 expression. Our findings highlight the pathogenic role of METTL3 in KC. Further investigation is required to investigate the underlying mechanism.</p>","PeriodicalId":12177,"journal":{"name":"Experimental eye research","volume":" ","pages":"110207"},"PeriodicalIF":3.0,"publicationDate":"2024-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142834846","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The need for better and simpler alternative crosslinking strategies to treat keratoconus (KC) is becoming essential as there is only a single approved way to treat it. Recently, conventional UV-A Riboflavin crosslinking is proven to have some disadvantages such as causing damage to the corneal endothelium and inducing keratocyte apoptosis. A chemical cross-linker (CXL) using carbodiimide chemistry and an octanedioic acid spacer is found effective in stiffening the cornea and has the potential to be developed as an alternative therapy to halt KC progression. In order to investigate the molecular changes induced by the cross-linker, we have analyzed the effect of the cross-linker on the activity of matrix metalloproteases (MMPs) in epithelial and stromal layers of KC corneas and in vitro cellular systems to determine its role in stiffening the KC cornea. At well-optimized concentration, KC corneal buttons were treated with the CXL and the stiffening of the cornea was measured. The collagen fibril assembly in the stroma was analyzed using transmission electron microscopy and the activity of MMPs 2 and 9 were visualized using gelatin zymography. KC corneal fibroblasts in culture and tumor necrosis factor-α (TNF-α) induced human corneal epithelial (HCE) cell line were treated with CXL and secretion of MMPs 1, 2, 3 and 9 were analyzed by enzyme-linked immunosorbent assay (ELISA). We found that the CXL stiffened the KC corneas comparable to the normal corneas, with very less cytotoxicity. The collagen fiber assembly was reorganized in an orderly fashion and fibril density and diameter increased after CXL treatment. The activity of MMPs and cathepsin G in the epithelial and stromal layers of KC tissues decreased post-treatment. Secretion and activity of MMPs from the corneal epithelial and stromal cells after CXL treatment were significantly reduced while the epithelial lysyl oxidase activity increased. The CXL, intended to stop the KC progression, modified the extracellular matrix collagen assembly in the stroma and decreased the secretion of a group of metalloproteases and their activity. We have demonstrated a set of molecular changes effected by the CXL, which might aid in the stiffening of the KC cornea.
{"title":"Inhibition of matrix metalloproteases by a chemical cross-linker to halt the corneal degradation in keratoconus.","authors":"Adhithya Subramanian Gopalakrishnan, Sumaiya Sirajudeen, Nasrin Banu, Jessica Nunes, Divya T Rajendran, Seema Yadav, Namperumalsamy Venkatesh Prajna, Rachel Williams, Dharmalingam Kuppamuthu, Ramprasad Obula Giridhara Gopalan","doi":"10.1016/j.exer.2024.110208","DOIUrl":"10.1016/j.exer.2024.110208","url":null,"abstract":"<p><p>The need for better and simpler alternative crosslinking strategies to treat keratoconus (KC) is becoming essential as there is only a single approved way to treat it. Recently, conventional UV-A Riboflavin crosslinking is proven to have some disadvantages such as causing damage to the corneal endothelium and inducing keratocyte apoptosis. A chemical cross-linker (CXL) using carbodiimide chemistry and an octanedioic acid spacer is found effective in stiffening the cornea and has the potential to be developed as an alternative therapy to halt KC progression. In order to investigate the molecular changes induced by the cross-linker, we have analyzed the effect of the cross-linker on the activity of matrix metalloproteases (MMPs) in epithelial and stromal layers of KC corneas and in vitro cellular systems to determine its role in stiffening the KC cornea. At well-optimized concentration, KC corneal buttons were treated with the CXL and the stiffening of the cornea was measured. The collagen fibril assembly in the stroma was analyzed using transmission electron microscopy and the activity of MMPs 2 and 9 were visualized using gelatin zymography. KC corneal fibroblasts in culture and tumor necrosis factor-α (TNF-α) induced human corneal epithelial (HCE) cell line were treated with CXL and secretion of MMPs 1, 2, 3 and 9 were analyzed by enzyme-linked immunosorbent assay (ELISA). We found that the CXL stiffened the KC corneas comparable to the normal corneas, with very less cytotoxicity. The collagen fiber assembly was reorganized in an orderly fashion and fibril density and diameter increased after CXL treatment. The activity of MMPs and cathepsin G in the epithelial and stromal layers of KC tissues decreased post-treatment. Secretion and activity of MMPs from the corneal epithelial and stromal cells after CXL treatment were significantly reduced while the epithelial lysyl oxidase activity increased. The CXL, intended to stop the KC progression, modified the extracellular matrix collagen assembly in the stroma and decreased the secretion of a group of metalloproteases and their activity. We have demonstrated a set of molecular changes effected by the CXL, which might aid in the stiffening of the KC cornea.</p>","PeriodicalId":12177,"journal":{"name":"Experimental eye research","volume":" ","pages":"110208"},"PeriodicalIF":3.0,"publicationDate":"2024-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142834845","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-11DOI: 10.1016/j.exer.2024.110206
Guodong Liu, Hui Li, Le Feng, Min Li, Peng Gao, Fang Wang
This study aimed to investigate the effects of O-linked N-acetylglucosamine modification (O-GlcNAcylation) on astroglial-mesenchymal transition through connexin43 (Cx43) pathway under high-glucose conditions. The primary rat astrocytes were cultured under normal and high-glucose conditions, and level of GFAP, α-SMA and Cx43 was investigated. To explore the influence of O-GlcNAcylation on astroglial-mesenchymal transition, Thiamet G treatment was employed to enhance O-GlcNAcylation, while Alloxan was used to decrease it. Cx43 knockdown was acquired through lentivirus constructs to explore its role in astrocyte transition. The levels of GFAP and α-SMA expressions were examined, while astrocyte proliferation was evaluated using the CCK-8 assay, and migration was assessed through wound healing assays. The results showed that primary rat astrocytes were identified by GFAP antibody staining. Under high-glucose conditions, the levels of GFAP, α-SMA, and Cx43 increased, as confirmed by Western blot and immunofluorescence. O-GlcNAcylation augmentation induced by Thiamet G treatment significantly increased the expression of GFAP, α-SMA, and Cx43 compared to both normal and high-glucose conditions. Conversely, the inhibition of O-GlcNAcylation reversed the high-glucose-induced increase in GFAP and α-SMA. Cx43 knockout led to the downregulation of GFAP and α-SMA compared to high-glucose and O-GlcNAcylation-augmented conditions. Additionally, levels of O-GlcNAcylation and VEGF were reduced in Cx43 knockout group. Consistently, CCK8 and wound healing assays demonstrated that Cx43 knockout could inhibit astrocyte proliferation and migration compared to the high-glucose and O-GlcNAcylation augmented groups. These findings demonstrate that astroglial-mesenchymal transition occurs under high-glucose conditions, and can be promoted by O-GlcNAcylation augmentation, but suppressed by Cx43 knockout. The study underscores the significant role of Cx43 in this transition and its potential involvement in diabetic complications.
{"title":"O-GlcNAcylation promotes astroglial-mesenchymal transition via the connexin43 pathway under high-glucose conditions.","authors":"Guodong Liu, Hui Li, Le Feng, Min Li, Peng Gao, Fang Wang","doi":"10.1016/j.exer.2024.110206","DOIUrl":"10.1016/j.exer.2024.110206","url":null,"abstract":"<p><p>This study aimed to investigate the effects of O-linked N-acetylglucosamine modification (O-GlcNAcylation) on astroglial-mesenchymal transition through connexin43 (Cx43) pathway under high-glucose conditions. The primary rat astrocytes were cultured under normal and high-glucose conditions, and level of GFAP, α-SMA and Cx43 was investigated. To explore the influence of O-GlcNAcylation on astroglial-mesenchymal transition, Thiamet G treatment was employed to enhance O-GlcNAcylation, while Alloxan was used to decrease it. Cx43 knockdown was acquired through lentivirus constructs to explore its role in astrocyte transition. The levels of GFAP and α-SMA expressions were examined, while astrocyte proliferation was evaluated using the CCK-8 assay, and migration was assessed through wound healing assays. The results showed that primary rat astrocytes were identified by GFAP antibody staining. Under high-glucose conditions, the levels of GFAP, α-SMA, and Cx43 increased, as confirmed by Western blot and immunofluorescence. O-GlcNAcylation augmentation induced by Thiamet G treatment significantly increased the expression of GFAP, α-SMA, and Cx43 compared to both normal and high-glucose conditions. Conversely, the inhibition of O-GlcNAcylation reversed the high-glucose-induced increase in GFAP and α-SMA. Cx43 knockout led to the downregulation of GFAP and α-SMA compared to high-glucose and O-GlcNAcylation-augmented conditions. Additionally, levels of O-GlcNAcylation and VEGF were reduced in Cx43 knockout group. Consistently, CCK8 and wound healing assays demonstrated that Cx43 knockout could inhibit astrocyte proliferation and migration compared to the high-glucose and O-GlcNAcylation augmented groups. These findings demonstrate that astroglial-mesenchymal transition occurs under high-glucose conditions, and can be promoted by O-GlcNAcylation augmentation, but suppressed by Cx43 knockout. The study underscores the significant role of Cx43 in this transition and its potential involvement in diabetic complications.</p>","PeriodicalId":12177,"journal":{"name":"Experimental eye research","volume":" ","pages":"110206"},"PeriodicalIF":3.0,"publicationDate":"2024-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142822030","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}