Joo Youn Oh’s research while affiliated with Seoul National University Hospital and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (208)


Figure 2. Temporary TLR2 inhibition, but not TLR4 blocking, exacerbates corneal NV after sterile suturing injury. A. Experimental design. WT (C57BL/6) mice were treated intraperitoneally (i.p.) with anti-TLR2 Ab, control IgG (control Ab for anti-TLR2 Ab), LPS-RS, or PBS (vehicle for LPS-RS) immediately after corneal suture placement. The corneas were evaluated 7 d after injury. B. Clinical scores of corneal NV graded under slit-lamp biomicroscopy. C, D. Representative microphotographs of CD31 and LYVE1 co-immunostaining of corneal whole mounts (C) and quantitation of CD31-and LYVE1-stained areas (D). Scale bar: 500 μm (upper panel), 200 μm (lower panel). E, F. qRT-PCR assays for angiogenesis-related markers and inflammatory cytokines in the cornea. mRNA levels are presented as fold changes relative to those in normal corneas without injury or treatment. Mean values ± SD are shown, where each dot depicts the data from an individual mouse. Data are pooled from 2-3 independent experiments. *p < 0.05, ** p < 0.01, ***p < 0.001, ****p < 0.0001, ns: not significant, as analyzed by one-way ANOVA with Tukey's test
Figure 3. Aberrant activation is not observed in response to inflammatory stimuli in TLR2 KO keratocytes compared to WT keratocytes.A. Experimental protocol. Keratocytes were isolated from the corneas of WT (C57BL/6) mice and TLR2 KO mice and cultured for 18 h in the presence or absence of TNF-α and IL-1α. B, C. qRT-PCR assays of WT keratocytes and TLR2 KO keratocytes for angiogenesis-related factors and inflammatory cytokines/chemokines. mRNA levels are shown relative to those in WT keratocytes not treated with TNF-α or IL-1α. D. ELISA for secreted levels of inflammatory cytokines and chemokines in culture supernatants of WT keratocytes and TLR2 KO keratocytes. Mean values ± SD are presented. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, ns: not significant, as analyzed by one-way ANOVA with Tukey's test or by Kruskal-Wallis test with Dunn's multiple-comparisons test (Mrc1 and Il1b in C).
Figure 4. Foxp3 + Tregs are not induced in TLR2 KO mice after injury, whereas monocytes are. A, B. Representative (A) and quantitative flow cytometry results (B) for CD11b + Ly6G -Ly6C + monocytes in the blood and spleen of WT (C57BL/6) mice and TLR2 KO mice without injury and 7 d after corneal suturing injury. C, D. Representative flow cytometry cytograms (C) and quantitation (D) of CD4 + CD25 + Foxp3 + Tregs in ocular draining cervical lymph nodes (CLN), blood, and spleen of WT mice and TLR2 KO mice without injury and 7 d post-injury. E, F. Representative and quantitative flow cytometry results for CD11b + Ly6G + granulocytes in the blood and spleen of WT mice and TLR2 KO mice without injury and 7 d post-injury. Mean values ± SD are shown where each dot depicts the data from an individual mouse. Data are pooled from three independent experiments. **p < 0.01, ***p < 0.001, ****p < 0.0001, ns: not significant, as analyzed by one-way ANOVA with Tukey's test or by Kruskal-Wallis test with Dunn's multiple-comparisons test (CD4 + CD25 + Foxp3 + cells in CLN in D).
Figure 5. Increased CD11b+Ly6C+ monocytes in TLR2 KO mice following injury promote corneal NV and inflammation.A. Experimental design. CD11b + Ly6C + monocytes were flow-sorted in the spleens of TLR2 KO mice 7 d after corneal suturing injury. Either the freshly-sorted CD11b + Ly6C + monocytes or HBSS (Hank's balanced salt solution, vehicle for cells) were intravenously (i.v.) transferred into WT (C57BL/6) mice immediately after corneal suturing injury. The corneas and spleens were evaluated 7 d after the injury. B. Representative FACS plot schema showing the isolation of CD11b + Ly6C + cells from the spleen of TLR2 KO mice 7 d post-injury. C, D. Slit-lamp biomicroscopic images of the cornea and clinical scores of corneal NV 7 d after injury and adoptive transfer of CD11b + Ly6C + cells. E. CD31-stained area in corneal whole mounts. F, G. qRT-PCR assays for angiogenesis-and inflammation-related markers in the cornea. Shown are mRNA levels relative to the levels in normal corneas without injury or treatment. Mean values ± SD are shown, where each dot depicts the data from an individual mouse. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, as analyzed by one-way ANOVA with Tukey's test, by Kruskal-Wallis test with Dunn's multiple-comparisons test (Mrc1 in G) or by Student's t-test (Il6 in G).
Figure 7. TLR2 KO Tregs exhibit reduced cell proliferation and IL-2 receptor expression, lower IL-10 secretion, and higher IFN-γ secretion. A. Experimental scheme for comparative analysis of WT (C57BL/6) Tregs and TLR2 KO Tregs. CD4 + CD25 + Foxp3 + Tregs were isolated from the spleens of WT mice and TLR2 KO mice and cultured for 5 d in the presence or absence of anti-CD3 Ab and IL-2. Then, assays were performed for evaluation of cell proliferation, marker expression, and cytokine secretion. B, C. Representative flow cytometry histograms of CFSE dilution assay (B) and quantitation of cell proliferation (C). D, E. Representative (D) and quantitative flow cytometric analysis (E) for expression of intrinsic Treg markers CD25 and Foxp3 in WT Tregs and TLR2 KO Tregs over time in culture. The percentage of CD4 + CD25 + Foxp3 + cells out of CD4 + cells is presented. F. qRT-PCR for transcript levels of Il2ra, Il2rb, and Il2rg. G. qRT-PCR and ELISA for transcript and secreted levels of IL-10 and IFN-γ. Mean values ± SD are presented. *p < 0.05, ***p < 0.001, ****p < 0.0001, ns: not significant, as analyzed by one-way ANOVA with Tukey's test or Student's t-test (CD4 + CD25 + Foxp3 + (Day 0) in E, F and G).

+1

Toll-like receptor 2 deficiency exacerbates corneal angiogenesis in injury by impairing regulatory T cells
  • Article
  • Full-text available

May 2025

·

9 Reads

Theranostics

Jung Hwa Ko

·

Hyun Ju Lee

·

Hyeon Ji Kim

·

[...]

·

Joo Youn Oh

Background: Toll-like receptor (TLR) 2 is a primary sensor of injury, and regulatory T cells (Tregs) are crucial mediators of tissue homeostasis. In this study, we aimed to investigate whether TLR2 is necessary for Treg-mediated restoration of corneal homeostasis following injury. Methods: We evaluated inflammatory corneal neovascularization and the proportions of Tregs, along with pro-angiogenic, pro-inflammatory monocytes, using a suture-induced corneal angiogenesis model in mice that either lacked TLR2 or were subjected to temporary TLR2 inhibition. The roles of injury-induced Tregs in corneal angiogenesis were further verified in vivo through adoptive transfer and in vitro using cultures of vascular endothelial cells. Results: Inflammatory corneal neovascularization was significantly more pronounced in TLR2 knockout mice compared to wild-type mice, while no differences were observed in TLR4 knockout mice. Temporary TLR2 inhibition also exacerbated corneal neovascularization, whereas TLR4 inhibition did not. Mechanistically, corneal injury induced an increase in Tregs in wild-type mice, which was absent in TLR2 knockout mice. Conversely, pro-angiogenic, pro-inflammatory monocytes were elevated in TLR2 knockout mice. Adoptive transfer of injury-induced Tregs from wild-type to TLR2 knockout mice reduced corneal neovascularization and decreased the number of monocytes. Functional assays demonstrated that Tregs from TLR2 knockout mice exhibited lower cell proliferation and IL-10 secretion, but increased IFN-γ secretion compared to Tregs from wild-type mice. Furthermore, TLR2 knockout Tregs were less effective at inducing apoptosis and suppressing pro-inflammatory activation and tube formation of vascular endothelial cells than their wild-type counterparts. Conclusion: Our findings suggest an expanded role for TLR2 in promoting corneal angiogenic and immunologic homeostasis during injury by regulating Treg numbers and functions.

Download

Double angle plots of anterior, posterior, and total astigmatism from (A) initial measurement, (B) final measurement, and (C) average of each. (A, B, C) Anterior corneal astigmatism showed with-the-rule (WTR) astigmatism at initial measurements, which remained WTR with decreased magnitude at final measurements. Posterior corneal astigmatism showed against-the-rule (ATR) astigmatism at initial measurements, which remained ATR with minimal change in the magnitude at final measurements. Total corneal astigmatism showed WTR astigmatism at initial measurement, which remained WTR with increased magnitude at final measurements
Longitudinal changes in ocular biometry and their effect on intraocular lens power calculation accuracy in cataract patients

February 2025

·

26 Reads

Graefe's Archive for Clinical and Experimental Ophthalmology

To investigate the changes in ocular biometry over time and their impact on intraocular lens (IOL) calculation in adult Korean patients with cataracts. Inclusion criteria were patients who underwent two consecutive ocular biometric measurements spaced more than one year apart using the IOLMaster 700 between November 2019 and February 2024 at a tertiary hospital in Seoul, Korea. Longitudinal changes in ocular biometry were evaluated. Predictive errors were compared among patients who underwent cataract surgery using the SRK/T, Kane, Barrett Universal II, Cook K6, EVO, Hill-RBF, Hoffer QST, and Pearl DGS formulas. A total of 448 eyes from 448 patients were included. Ocular biometry measured over an average interval of 23.4 months showed that with increasing age, axial length elongated (0.04 ± 0.10 mm, p < 0.001), and the magnitude of total corneal astigmatism increased (0.04 ± 0.39 D, p = 0.018). The mean absolute predictive errors of the final measurements were significantly smaller compared to the initial measurements in the Barrett Universal II, EVO, Kane, and Pearl DGS formulas (difference of -0.05 D, -0.05 D, -0.06 D, and − 0.05 D, respectively). In the subgroup of eyes with an axial length of 25 mm or longer, the final measurements showed even greater reduction in mean absolute predictive errors across multiple formulas, including Barrett Universal II, Cook K6, EVO, Hill-RBF, Hoffer QST, Kane, and Pearl DGS, with reductions of -0.11 D, -0.11 D, -0.10 D, -0.08 D, -0.10 D, -0.09 D and − 0.10 D, respectively. Axial length increases and corneal curvature changes with aging. IOLMaster 700 ocular biometry results measured closer to the date of surgery were more accurate in IOL power calculation than those measured more than one year earlier, with the greatest improvement observed in myopic eyes. Therefore, it is recommended to repeat IOLMaster 700 biometry before surgery if the previous measurements were taken more than a year ago.


Schematic design of how to select the eyes with presbyopia-correcting IOL implantation for this study
Various patterns of dissatisfaction are depicted based on the types of IOLs. A The proportion of dissatisfaction for each IOL type regardless of the nature of dissatisfaction. B The proportion of dissatisfaction of photic phenomena according to IOL types, with the ReSTOR® group having the highest rate of 16.4%, which was significantly different from the Symfony® group. C The proportion of dissatisfaction of distance visual acuity according to IOL types, with the ReSTOR® group having the highest rate of 9.8%, which was significantly different from the Symfony® group. D The proportion of dissatisfaction of near visual acuity according to IOL types, with no significant differences observed. E The proportion of dissatisfaction of intermediate visual acuity according to IOL types, with the PanOptix® group having the rate of 3.5%, while the Symfony® group had a rate of 0%
The proportion of dissatisfaction according to the laterality of IOL implanted eyes
Risk factors in self-reported dissatisfied patients implanted with various presbyopia-correcting intraocular lenses after cataract surgery

February 2025

·

24 Reads

BMC Ophthalmology

Background This study aimed to investigate the self-reported dissatisfaction rates and associated risk factors among patients who underwent cataract surgery using different types of presbyopia-correcting intraocular lenses (IOLs). Methods This retrospective case–control study analyzed the medical records in 340 eyes from 211 cataract surgery patients with presbyopia-correcting IOLs. The analyzed IOL types included bifocal (ReSTOR®), trifocal (PanOptix®), and extended depth-of-focus (EDOF; Symfony®) IOLs. The rates of self-reported dissatisfaction related to vision or photic disturbances were compared between these IOLs. Various factors, including sex, age, preoperative visual acuity and refractive status, and biometric indices, were analyzed to identify potential risk factors for dissatisfaction. Results The overall dissatisfaction rate was 18.5% (63/340). Among the IOL types, Symfony®-implanted eyes had the highest rate of near-vision dissatisfaction, while PanOptix®-implanted eyes showed similar proportions of photic disturbances and near-vision discomfort. The major risk factor identified for overall dissatisfaction, regardless of IOL type, was preoperative myopia, which aligns with the risk factor for near discomfort. Meanwhile, the risk factors for photic phenomena were revealed to be thinner corneal thickness and greater corneal astigmatism. By IOL types, preoperative myopia caused near-vision discomfort in Symfony® eyes, whereas greater corneal astigmatism and thinner corneas were linked to photic disturbances in PanOptix® eyes. Conclusions It suggests that near-vision discomfort is related to myopic factors, whereas photic disturbances are associated with ocular aberrations. The types of dissatisfaction vary depending on the designs of presbyopia-correcting IOLs. Trial Registration This retrospective study adhered to the principles of the Declaration of Helsinki and was approved by the Institutional Review Board of the Seoul National University Hospital on March 13, 2023 (IRB No: 2303–025-1409).


Shapley Additive Explanations (SHAP) values and mean absolute SHAP values for the IOL formulas. ACD, anterior chamber depth measured from corneal epithelium to lens; AL, axial length; BUII, Barrett Universal II; CCT, central corneal thickness; CD, horizontal corneal diameter; IOL, intraocular lens; K, Keratometry; LT, lens thickness. The Shapley additive explanation (SHAP) values and mean absolute SHAP values for each intraocular lens (IOL) formula. The SHAP value for each participant is visually presented as a colored dot. The x-axis represents the SHAP values, wherein negative values correspond to the eyes contributing to a negative prediction error (PE) and positive values correspond to a positive PE. Colors ranging from blue to red denote the value of the ocular biometric variables, with red dots representing eyes with greater values and blue dots representing eyes with smaller values. The average absolute SHAP values for each variable indicate the contribution of the variable variation within the formula. The height of the horizontal bar indicates the contribution of the variable, with larger bars signifying higher contributions and smaller bars indicating lower contributions
Partial dependence plots (PDPs) between the Shapley Additive Explanations (SHAP) values and ocular biometric variables. ACD, anterior chamber depth measured from corneal epithelium to lens; AL, axial length; BUII, Barrett Universal II; CCT, central corneal thickness; CD, horizontal corneal diameter; IOL, intraocular lens; K, Keratometry; LT, lens thickness. The partial dependence plots (PDPs) between SHAP values and ocular biometric variables present the marginal impact of each variable on each intraocular lens (IOL) formula. The x- and y-axes represent the value of the variable and the SHAP value, respectively. The blue dots represent the eyes. SHAP values of < 0.0 and > 0.0 result in myopic and hyperopic shifts, respectively
Evaluation of prediction errors in nine intraocular lens calculation formulas using an explainable machine learning model

December 2024

·

29 Reads

BMC Ophthalmology

Background The purpose of the study was to evaluate the relationship between prediction errors (PEs) and ocular biometric variables in cataract surgery using nine intraocular lens (IOL) formulas with an explainable machine learning model. Methods We retrospectively analyzed the medical records of consecutive patients who underwent standard cataract surgery with a Tecnis 1-piece IOL (ZCB00) at a single center. We calculated predicted refraction using the following IOL formulas: Barrett Universal II (BUII), Cooke K6, EVO V2.0, Haigis, Hoffer QST, Holladay 1, Kane, SRK/T, and PEARL-DGS. We used a LightGBM-based machine learning model to evaluate the explanatory power of ocular biometric variables for PEs and assessed the relationship between PEs and ocular biometric variables using Shapley additive explanation (SHAP) values. Results We included 1,430 eyes of 1,430 patients in the analysis. The SRK/T formula exhibited the highest R² value (0.231) in the test set among the machine-learning models. In contrast, the Kane formula exhibited the lowest R² value (0.021) in the test set, indicating that the model could explain only 2.1% of the PEs using ocular biometric variables. BUII, Cooke K6, EVO V2.0, Haigis, Hoffer QST, Holladay 1, PEARL-DGS formulas exhibited R² values of 0.046, 0.025, 0.037, 0.194, 0.106, 0.191, and 0.058, respectively. Lower R² values for the IOL formulas corresponded to smaller SHAP values. Conclusion The explanatory power of currently used ocular biometric variables for PEs in new-generation formulas such as BUII, Cooke K6, EVO V2.0 and Kane is low, implying that these formulas are already optimized. Therefore, the introduction of new ocular biometric variables into IOL calculation formulas could potentially reduce PEs, enhancing the accuracy of surgical outcomes.


Recurrence of pterygium and symblepharon after treatment with conjunctival autograft. (A, B) Anterior segment photographs of a patient at presentation. A fleshy pterygial mass and symblepharon extending to the medial end of the lower lid were noted. (C) Anterior OCT at presentation. Up to 0.68 mm thick pterygial mass invaded the corneal surface. (D) One month after the first surgery using pterygium excision, symblepharolysis, intraoperative MMC application, and conjunctival autograft. (E, F) Six months after the first surgery. Fibrovascular ingrowth into the cornea causing 10.0 D of astigmatism and symblepharon recurrence was observed. (G, H) One month after a second surgery using pterygium excision, symblepharolysis, intraoperative MMC application, limbal allograft, and AM transplantation.
Recurrences of pterygium and symblepharon after repeated surgeries using limbal allografts and AM transplantation. (A, B) Anterior segment photographs of the patient 6 (A) and 30 months (B) after a second surgery. Pterygial mass and symblepharon started to reform and overgrew onto the previous limbal graft and corneal surface progressively. (C) Anterior segment photographs of the patient 6 months after a fourth surgery.
Resolution of pterygium and symblepharon after treatment with labial mucosal autograft. (A–C) Anterior segment photographs of the patient 12 months after a fourth surgery. A recalcitrant recurrence of pterygium and symblepharon was observed. (D–F) Anterior segment photographs of the patient at 1 week (D), 3 weeks (E), and 3 months (F) after a fifth surgery using labial mucosal autograft. (G–I) Anterior segment photographs without and with fluorescein staining at 30 months after labial mucosal autograft. Pterygium and symblepharon did not recur, and a smooth and stable ocular surface was achieved.
Corneal topography before and after labial mucosal autograft. (A) Corneal topographic image at the time of recurrence 3 years after a third surgery. (B) Corneal topographic image 2 years after a fifth surgery using labial mucosal autograft, indicating a significant reduction in corneal astigmatism and irregularities with the resolution of pterygium.
Superior outcome of labial mucosal autograft over limbal allograft in the management of recurrent pterygium with symblepharon: a case report

October 2024

·

36 Reads

A 40-year-old woman visited our clinic for recurred pterygium and symblepharon in the right eye. She had a history of pterygium excision 8 years before. Over the course of 7 years, we performed pterygium excision combined initially with mitomycin C (MMC) application and conjunctival autograft. This was followed by three procedures using limbal allografts, MMC application, and amniotic membrane transplantation. All procedures were unsuccessful, resulting in aggressive recurrences of pterygial mass and symblepharon, extraocular movement limitation, corneal astigmatism, and decreased visual acuity. Ultimately, we applied a labial mucosal autograft after the recession of pterygial tissue. No complications were observed. Two and a half years postoperatively, the labial mucosal autograft was well-integrated into the conjunctival surface without symblepharon recurrence or abduction limitation. Corneal clarity was restored, and astigmatism was reduced, with no recurrence of pterygium. In conclusion, a labial mucosal autograft is a viable treatment option in complex cases of recalcitrantly recurrent pterygium with symblepharon.


The Bland-Altman plots of the (a) anterior Ks, (b) anterior Kf, (c) ACA, (d) posterior Ks, (e) posterior Kf, (f) PCA, (g) total Ks, (h), total Kf, (i) TCA, (j) CCT, (k) TCT, and (l) ACD. The solid line represents the mean difference, while the dotted lines on each side show upper and lower 95% LoA. K = keratometry; s = steep; f = flat; ACA = anterior corneal astigmatism; PCA = posterior corneal astigmatism; TCA = total corneal astigmatism; CCT = central corneal thickness; TCT = thinnest corneal thickness; ACD = anterior chamber depth; D = diopters; LoA = limits of agreement.
Agreement in anterior segment measurements between swept-source optical coherence and dual Scheimpflug tomography devices in keratoconus eyes

October 2024

·

19 Reads

Purpose To evaluate the agreement of ocular biometry measured using a swept-source optical coherence (Casia 2) and a dual Scheimpflug (Galilei G6) tomography in keratoconus. Methods This retrospective study included 102 eyes from 102 keratoconus patient examined using both devices. Parameters compared included flat (Kf) and steep (Ks) keratometry, astigmatism of anterior, posterior, and total keratometry, central (CCT) and thinnest (TCT) corneal thickness. To assess the agreement, intraclass coefficient (ICC) and Bland-Altman analysis with 95% limits of agreement (LoA) were used. Results Anterior and total Ks and Kf showed moderate or good agreement with 95% limits of agreement (LoA) range over 9.75 D. Posterior Ks and Kf were lower in the Galilei G6 (all p < 0.001). Astigmatism showed moderate, poor, and moderate agreement for anterior, posterior, and total keratometry, respectively. CCT and TCT showed excellent agreement; however, the 95% LoA range was over 60 μm. Conclusion The agreement between the two devices was not excellent for most parameters used to diagnose keratoconus and assess disease progression, and the differences were clinically significant. Therefore, the measurements from these two devices are not interchangeable for patients with keratoconus.


Biopotency and surrogate assays to validate the immunomodulatory potency of extracellular vesicles derived from mesenchymal stem/stromal cells for the treatment of experimental autoimmune uveitis

August 2024

·

70 Reads

·

4 Citations

Extracellular vesicles (EVs) derived from mesenchymal stem/stromal cells (MSCs) have been recognized as promising cytotherapeutics due to their demonstrated immunomodulatory effects in various preclinical models. The immunomodulatory capabilities of EVs stem from the proteins and genetic materials they carry from parent cells, but the cargo contents of EVs are significantly influenced by MSC tissues and donors, cellular age and culture conditions, resulting in functional variations. However, there are no surrogate assays available to validate the immunomodulatory potency of MSC‐EVs before in vivo administration. In previous work, we discovered that microcarrier culture conditions enhance the immunomodulatory function of MSC‐EVs, as well as the levels of immunosuppressive molecules such as TGF‐β1 and let‐7b in MSC‐EVs. Building on these findings, we investigated whether TGF‐β1 levels in MSC‐EVs could serve as a surrogate biomarker for predicting their potency in vivo. Our studies revealed a strong correlation between TGF‐β1 and let‐7b levels in MSC‐EVs, as well as their capacity to suppress IFN‐γ secretion in stimulated splenocytes, establishing biopotency and surrogate assays for MSC‐EVs. Subsequently, we validated MSC‐EVs generated from monolayer cultures (ML‐EVs) or microcarrier cultures (MC‐EVs) using murine models of experimental autoimmune uveoretinitis (EAU) and additional in vitro assays reflecting the Mode of Action of MSC‐EVs in vivo. Our findings demonstrated that MC‐EVs carrying high levels of TGF‐β1 exhibited greater efficacy than ML‐EVs in halting disease progression in mice with EAU as well as inducing apoptosis and inhibiting the chemotaxis of retina‐reactive T cells. Additionally, MSC‐EVs suppressed the MAPK/ERK pathway in activated T cells, with treatment using TGF‐β1 or let‐7b showing similar effects on the MAPK/ERK pathway. Collectively, our data suggest that MSC‐EVs directly inhibit the infiltration of retina‐reactive T cells toward the eyes, thereby halting the disease progression in EAU mice, and their immunomodulatory potency in vivo can be predicted by their TGF‐β1 levels.


Figure 3. TLR2 signaling in MSCs is essential for suppression of inflammatory angiogenesis in the cornea following sterile injury. A. Experimental protocol. Immediately after corneal suturing injuries, MSCs, Pam2CSK4-pretreated MSCs, TLR2 siRNA-transfected MSCs, SCR siRNA-transfected MSCs or HBSS (vehicle) were administered via tail vein injection in BALB/c mice. Seven days later, the corneas were subjected to assays. B, C. Representative corneal photographs and microphotographs of CD31/LYVE1-stained corneal whole-mounts. Scale bar: 500 μm for the first and second rows and 200 μm for the third row (magnified images of yellow-outlined insets from the first row). D, E. Clinical scoring of corneal NV and quantification of CD31-stained area in corneal whole-mounts. F, G. qRT-PCR for pro-angiogenic factors and pro-inflammatory cytokines in cornea. The mRNA levels are presented relative to those in BALB/c control corneas that had not received injury or treatment. H. Experimental protocol. Corneal sutures were applied to TLR2 KO mice, and either MSCs or HBSS were intravenously injected. After 7 d, the corneas were subjected to assays. I, J. Representative corneal photographs and microphotographs after CD31/LYVE1 immunostaining. Scale bar: 500 μm for the upper row and 200 μm for the lower row (magnified images of yellow-outlined insets from the upper row). K, L. Clinical scoring of corneal NV and measurement of CD31-and LYVE1-stained areas. Data represent means ± SD, where a circle indicates the data from an individual animal. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, as analyzed by one-way ANOVA and Tukey's test or by Student's t-test (Injury + MSC vs. Injury + MSC/Pam2CSK4 in F, G).
Figure 4. Single-cell transcriptional profiling of Pam2CSK4-treated and untreated MSCs. A. Experimental scheme. MSCs treated with Pam2CSK4 100 ng/mL for 24 h and untreated MSCs were subjected to scRNA-seq on the 10x Genomics Chromium platform. B. Combined UMAP and tSNE plots of Pam2CSK4-treated MSCs (blue) and untreated MSCs (orange) showing overlapping. C. tSNE plots of Pam2CSK4-treated MSCs and untreated MSCs depicting 7 clusters (C0-C6) in both cell libraries. D, E. Feature and dot plots displaying positive and negative markers for MSCs. F. Heatmap of top 10 DEGs in Pam2CSK4-treated MSCs vs. untreated MSCs as determined by RNA-Seq analysis. AKR1C1 was identified as the top DEG upregulated in Pam2CSK4-treated MSCs relative to untreated MSCs. G. ELISA for secreted levels of AKR1C1 in cell-free supernatants of MSC cultures treated with Pam2CSK4 (0-100 ng/mL). H. qRT-PCR and ELISA for mRNA and protein levels of AKR1C1 in MSCs transfected with TLR2 siRNA or control SCR siRNA. Means ± SD are presented. **p < 0.01 as analyzed by Kruskal-Wallis test and Dunn's multiple-comparison test (G) or Student's t-test (H).
Figure 5. AKR1C1 inhibition promotes MSC ferroptosis. A. Cell viability assay in MSCs transfected with AKR1C1 siRNA or control SCR siRNA or in MSCs treated with AKR1C1 inhibitor, 5-PBSA (10 or 100 ng/mL). B, C. Quantification of intracellular Fe 2+ fluorescence intensity in MSCs and representative fluorescence images. Red indicates FerroOrange staining, while blue represents DAPI-stained nuclei. Scale bar: 50 μm for the upper row and 200 μm for the lower row. Data are means ± SD from 3-4 independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, ns: not significant, as analyzed by one-way ANOVA and Tukey's test.
Figure 7. AKR1C1 downregulates pro-inflammatory cytokines in monocytes and macrophages. A. Experimental scheme. BM cells were cultured under GM-CSF stimulation for 5 d in the presence or absence of rhAKR1C1 (1 or 5 ng/mL) or 5-PBSA (10 or 100 ng/mL). Then, the cells were assessed for MDSC marker Arg1 and pro-inflammatory cytokines. B. qRT-PCR for Arg1, Tnfa and Il1b in BM monocytes. The mRNA levels are presented as fold changes relative to GM-CSF-unstimulated BM cells. C. ELISA for TNF-α and IL-1β secretion in culture supernatants of BM monocytes. D. Experimental scheme. THP-1 macrophages were stimulated with LPS followed by ATP, and were treated with rhAKR1C1 (1 or 5 ng/mL) or 5-PBSA (10 or 100 ng/mL). 18 h later, the cells were analyzed for pro-inflammatory cytokine expression. E, F. qRT-PCR and ELISA for transcript and protein levels of TNF-α, IL-1β and IL-12B in LPS/ATP-stimulated THP-1 macrophages. Shown are the mRNA levels relative to LPS/ATP-unstimulated THP-1 macrophages. Means ± SD are presented. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, as analyzed by one-way ANOVA and Tukey's test.
Figure 8. AKR1C1 in MSCs is required for amelioration of inflammatory angiogenesis in the cornea following sterile injury. A. Experimental protocol. Immediately after corneal suturing injury, AKR1C1 siRNA-transfected MSCs, SCR siRNA-transfected MSCs or HBSS (vehicle) were intravenously administered into BALB/c mice. Seven days later, the corneas were assayed. B, C. Representative corneal photographs and microphotographs of corneal flat mounts with CD31/LYVE1 immunostaining. Scale bar: 500 μm or 200 μm (magnified views of yellow-outlined insets). D, E. Clinical scoring of corneal NV and quantification of LYVE1-stained area in corneal whole-mounts. F, G. Representative flow cytometry cytograms of CD11b hi Ly6C hi cells in the spleen (F) and quantitation of CD11b hi Ly6C hi cells in spleen and blood (G). H, I. qRT-PCR assays for pro-inflammatory cytokines in ocular draining cervical lymph nodes (DLNs) and blood cells. The mRNA levels are presented as fold changes relative to naïve BALB/c mice without injury or treatment. Data represent means ± SD, where a circle indicates the data from an individual animal. *p < 0.05, **p < 0.01, ***p < 0.001, ns: not significant, as analyzed by one-way ANOVA and Tukey's test.
Activation of Toll-like receptor 2 promotes mesenchymal stem/stromal cell-mediated immunoregulation and angiostasis through AKR1C1

August 2024

·

29 Reads

·

2 Citations

Theranostics

Background: Mesenchymal stem/stromal cells (MSCs) maintain tissue homeostasis in response to microenvironmental perturbations. Toll-like receptors (TLRs) are key sensors for exogenous and endogenous signals produced during injury. In this study, we aimed to investigate whether TLRs affect the homeostatic functions of MSCs after injury. Methods: We examined the expression of TLR2, TLR3 and TLR4 in MSCs, and analyzed the functional significance of TLR2 activation using single-cell RNA sequencing. Additionally, we investigated the effects and mechanisms of TLR2 and its downstream activation in MSCs on the MSCs themselves, on monocytes/macrophages, and in a mouse model of sterile injury-induced inflammatory corneal angiogenesis. Results: MSCs expressed TLR2, which was upregulated by monocytes/macrophages. Activation of TLR2 in MSCs promoted their immunoregulatory and angiostatic functions in monocytes/macrophages and in mice with inflammatory corneal angiogenesis, whereas TLR2 inhibition attenuated these functions. Single-cell RNA sequencing revealed AKR1C1, a gene encoding aldo-keto reductase family 1 member C1, as the most significantly inducible gene in MSCs upon TLR2 stimulation, though its stimulation did not affect cell compositions. AKR1C1 protected MSCs against ferroptosis, increased secretion of anti-inflammatory cytokines, and enhanced their ability to drive monocytes/macrophages towards immunoregulatory phenotypes, leading to the amelioration of inflammatory corneal neovascularization in mice. Conclusion: Our findings suggest that activation of TLR2-AKR1C1 signaling in MSCs serves as an important pathway for the survival and homeostatic activities of MSCs during injury.


Subconjunctival aflibercept attenuates corneal neovascularization. a Experimental scheme. Corneal sutures were applied in BALB/c mice for induction of corneal neovascularization, and aflibercept (200 µg in 5 µL) (Eylea®, Regeneron Pharmaceuticals, Inc., Tarrytown, NY) or PBS (5 µL) was subconjunctivally injected. Seven days later, the corneas were clinically observed, and the corneas, blood and DLNs were collected for assays. b, c Representative corneal photographs (b) and quantification of corneal new vessels as graded by the standardized scoring system (c). d, e Representative microphotographs of whole-corneal flat mounts with CD31 immunostaining (d) and quantification of CD31-stained area (e). Mean values ± SD are shown from three independent experiments. Each circle depicts the data from an individual eye. **p < 0.01, ****p < 0.0001, as analyzed by Mann-Whitney test (c) or by unpaired t-test (e)
Subconjunctival aflibercept suppresses the expression of pro-angiogenic and inflammatory molecules in the cornea. a, b, c, d RT-qPCR for Cd31 (a), vascular growth factors (Vegfc and Angpt1) (b), pro-angiogenic monocyte/macrophage markers (Tek/Tie2, Mrc1, and Mrc2) (c), and pro-inflammatory cytokine (Il6) (d) in the cornea. The mRNA levels are presented as fold changes relative to the levels in control eyes which had not received injury or treatment. Mean values ± SD are shown from two independent experiments. Each circle depicts the data from an individual mouse. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, as analyzed by one-way ANOVA with Tukey’s test or by Kruskal–Wallis test with Dunn’s multiple-comparisons test (Tek in c)
Subconjunctival aflibercept reduces the percentage of circulating and cornea-infiltrating VEGFR-3⁺CD11b⁺myeloid cells. a, b Representative and quantitative flow cytometry results for VEGFR-3⁺CD11b⁺ cells (a) or VEGFR-2⁺CD11b⁺ cells (b) in the blood and DLN. (c) Enumeration of VEGFR-3⁺CD11b⁺ cells or VEGFR-2⁺CD11b⁺ cells in the corneal flat mounts immunostained with CD11b, VEGFR-2, or VEGFR-3. Mean values ± SD are shown from two independent experiments. Each circle depicts the data from an individual mouse. **p < 0.01, ***p < 0.001, ****p < 0.0001, ns: not significant, as analyzed by one-way ANOVA with Tukey’s test (a, b) or by unpaired t-test (c)
Subconjunctival aflibercept inhibits corneal angiogenesis and VEGFR-3CD11b cells

July 2024

·

20 Reads

Graefe's Archive for Clinical and Experimental Ophthalmology

Purpose This study aimed to investigate the effects of subconjunctival injection of aflibercept, a soluble protein decoy for VEGFR-1 and VEGFR-2, on corneal angiogenesis and VEGFR-expressing CD11b⁺ cells in a mouse model of suture-induced corneal neovascularization. Methods Corneal neovascularization was induced in BALB/c mice by placing three sutures on the cornea. Immediately after surgery, either 200 µg aflibercept (5 µL) or an equal volume of phosphate-buffered saline (PBS) was administered into the subconjunctival space. Seven days after later, corneal new vessels were quantified through clinical examination and measurement of the CD31-stained area in corneal flat mounts. The levels of pro-angiogenic and inflammatory markers in the cornea were evaluated using RT-qPCR. The percentages of VEGFR-2⁺CD11b⁺ cells and VEGFR-3⁺CD11b⁺ cells were analyzed in the cornea, blood, and draining cervical lymph nodes (DLNs) using flow cytometry. Results Subconjunctival injection of aflibercept significantly reduced the growth of corneal new vessels compared to subconjunctival PBS injection. The mRNA levels of Cd31, vascular growth factors (Vegfc and Angpt1), and pro-angiogenic/inflammatory markers (Tek/Tie2, Mrc1, Mrc2, and Il6) in the cornea were downregulated by subconjunctival aflibercept. Also, the percentage of VEGFR-3⁺CD11b⁺ cells in the cornea, blood, and DLNs was decreased by aflibercept, whereas that of VEGFR-2⁺CD11b⁺ cells was unaffected. Conclusion Subconjunctival aflibercept administration inhibits inflammatory angiogenesis in the cornea and reduces the numbers of cornea-infiltrating and circulating VEGFR-3⁺CD11b⁺ cells.



Citations (72)


... However, little is known about how and what determines the miRNA content of extracellular vesicles (EVs) from MSCs, which is a critical issue for further therapeutic applications [26].Furthermore, MSCs-derived exosomes provide considerable stability to the miRNAs they carry, protecting them from enzymatic degradation and facilitating delivery to recipient cells through various pathways [27]. Additionally, while intracellular miRNAs are produced through their own transcription and processing [28], miRNAs in exosomes primarily originate from the secretion and release of intracellular miRNAs [29]. ...

Reference:

Advances in the roles and mechanisms of mesenchymal stem cell derived microRNAs on periodontal tissue regeneration
Biopotency and surrogate assays to validate the immunomodulatory potency of extracellular vesicles derived from mesenchymal stem/stromal cells for the treatment of experimental autoimmune uveitis

... Primary keratocytes were isolated from the corneas of C57BL/6 WT or TLR2 KO mice using a previously-established protocol [26,27]. Briefly, the corneal epithelium and endothelium-Descemet membrane were removed, and the corneal stroma was treated with 10 mg/mL dispase (Roche, Indianapolis, IN) at 37 ˚C for 90 min, followed by 200 U collagenase I (Worthington Biochemical Corp., Lakewood, NJ) for 30 min. ...

Toxicity and efficacy of type I interferons on the ocular surface: in vitro, animal, and clinical studies
  • Citing Article
  • July 2024

The Ocular Surface

... In a parallel experiment, murine BM-derived monocytes were cocultured in a Transwell system with MSCs under GM-CSF stimulation ( Figure 2D). In our previous studies, MSCs directed the differentiation of GM-CSF-stimulated BM cells from pro-inflammatory CD11b hi Ly6C hi Ly6G lo monocytes into immunosuppressive CD11b mid Ly6C mid Ly6G lo monocytic MDSCs [41,42]. Consistent with these findings, the present results revealed that MSCs generated a distinct population of CD11b mid Ly6C mid Ly6G lo cells in BM monocytes, while reducing CD11b hi Ly6C hi Ly6G lo cells ( Figure 2E). ...

Defining Mesenchymal Stem/Stromal Cell-Induced Myeloid-Derived Suppressor Cells Using Single-Cell Transcriptomics
  • Citing Article
  • April 2024

Molecular Therapy

... In addition to these study design issues, the fact that we did not detect any new leukocyte or T-and B-cell subpopulation signatures of rejection risk in PBMC analyzed at 6 and 12 mo posttransplant, could either indicate a true lack of systemic donor antigen-driven immune response in those who developed rejection or that alloantigen-responsive cells had already migrated into the CT or to secondary lymphoid organs by the time the samples were obtained. Of interest in this regard, Lee et al 28 recently reported in a mouse model of corneal allotransplantation in which 50% of animals undergo graft rejection within 28 d that the level of a serum extracellular vesicle-derived major histocompatibility complex protein at day 7 posttransplant was predictive of rejection. ...

Serum Extracellular Vesicle Protein Profiling for Prediction of Corneal Transplant Rejection
  • Citing Article
  • February 2024

Transplantation

... Treating these cells with the MEK inhibitor PD98059 led to a reduction in Jun phosphorylation after PAUF treatment and a down-regulation of immunosuppressive factors at the transcript level [54]. In line with these findings, Lee et al. reported that co-culture of MDSCs with mesenchymal stromal cells (MSC) induced the MAPK signalling pathway, resulting in robust phosphorylation of JNK and a highly induced expression of immunosuppressive cytokines [55]. JNK is responsible for the phosphorylation of Jun as well as other transcription factors such as c-Myc, Elk-1 and ATF2 [56]. ...

Mesenchymal Stem/Stromal Cells Induce Myeloid-Derived Suppressor Cells in the Bone Marrow via the Activation of the c-Jun N-Terminal Kinase Signaling Pathway

... FGF2 is also a central mediator in angiogenic pathways essential for wound healing 60,61 . Conversely, FUT1 is recognized for its involvement in ABO self-antigen recognition 62 whereas recent studies have unveiled its immunomodulatory properties, particularly its role in inhibiting CD4 T cell proliferation 63 . Additionally, MCAM (also known as CD146) was found to be upregulated after TIC + OXT priming which has unique implication to MSC functionality relating to immunomodulation and bone regeneration. ...

Splenocytes with fucosylation deficiency promote T cell proliferation and differentiation through thrombospondin‐1 downregulation

... Previous studies have evaluated the accuracy of IOL power formulas for specific subgroups of patients according to the ocular biometric variables [6,7,[20][21][22][23][24][25][26][27][28] and provided valuable insights into the strengths and weaknesses of various formulas. The Kane formula exhibits a significantly lower MAE value and higher accuracy in eyes with long AL. ...

Intraocular lens power calculation in eyes with a shallow anterior chamber depth and normal axial length

... Dendranthema morifolium is a medicinal and edible cognate plant [26]. Modern studies showed that the medicinal ingredients of D. morifolium have many biological and pharmacological characteristics including antibacterial, anti-inflammatory, antioxidant, vasodilator, hypolipidemic, and anti-tumor characteristics, such as chlorogenic acid and 1, 5-dicaffeoylquinic acid [27][28][29]. The quality of the medication produced was also impacted by the accumulation of heavy metals in the soil that occurred during the planting of D. morifolium [30][31][32]. ...

1,5-Dicaffeoylquinic acid from Pseudognaphalium affine ameliorates dry eye disease via suppression of inflammation and protection of the ocular surface
  • Citing Article
  • June 2023

... It induces apoptosis of the corneal, conjunctival (including GCs), and lacrimal gland acinar epithelium [37,38]. At the cellular and molecular levels, the detrimental effects of IFN-γ on ocular surface are manifested through its impact on corneal stromal fibroblasts and epithelial cells, promoting inflammation, opacification, and barrier disruption [39]. In line with these concepts, we found the level of IFN-γ was related to OSDI and CFS, illustrating the damage to cornea epithelium may be associated with IFN-γ as well. ...

Interferon-γ elicits the ocular surface pathology mimicking dry eye through direct modulation of resident corneal cells

Cell Death Discovery

... In some clinical scenarios of corneal endothelial injury, we did observe that the corneal endothelial cell loss progresses even if the causative incident did not proceed. The onset of bullous keratopathy following cataract surgery and Argon laser iridotomy was reported at mean 148.4 and 107 months respectively [12]. The exact mechanism for the progression of endothelial cell loss beyond normal aging loss following primary insult remains uncertain. ...

Etiology and outcome of penetrating keratoplasty in bullous keratopathy post-cataract surgery vs post-glaucoma surgery