Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Ophthalmol ; 16(6): 904-908, 2023.
Article in English | MEDLINE | ID: mdl-37332549

ABSTRACT

AIM: To investigate the efficacy of ripasudil, a Rho kinase inhibitor, in reducing intraocular pressure (IOP) and medication scores of anti-glaucoma drugs in patients with ocular hypertension with inflammation and corticosteroid. METHODS: The study included 11 patients diagnosed with ocular hypertension with inflammation and corticosteroid, all of whom were prescribed ripasudil eye drops and followed up for at least 2y after the initiation of treatment. IOP was measured using a non-contact tonometer before enrollment and at each follow-up visit. The medication score of glaucoma eye drops was calculated for each patient. RESULTS: The mean IOP (26.4±2.9 mm Hg before treatment) significantly decreased after ripasudil therapy (13.7±3.3 mm Hg at 3mo) and remained stable in the low-teens during the 2-year follow-up period (P<0.0001). A significant decrease in the medication score was observed at 12mo or later after the initiation of ripasudil therapy (P<0.05). Both baseline medication scores and glaucomatous optic disc change rates were significantly higher in the five eyes that required glaucoma surgery during the 2-year observation period than the 10 eyes that did not require surgery. CONCLUSION: Our results demonstrate the efficacy of ripasudil, in reducing IOP and the medication score over a 2-year treatment period in patients with ocular hypertension with inflammation and corticosteroid. Our findings also suggest that ripasudil could reduce the IOP in uveitic glaucoma patients with both lower baseline medication score and lower glaucomatous optic disc change rate.

2.
PLoS One ; 15(9): e0237728, 2020.
Article in English | MEDLINE | ID: mdl-32925945

ABSTRACT

PURPOSE: To investigate the influence of EDOF IOLs, TECNIS Symfony® (Johnson & Johnson Surgical Vision, Inc.), on visual field sensitivity and to compare the IOLs with other kinds of IOLs. METHODS: The subjects included the normal fellow eyes of patients who underwent the Humphrey Field Analyzer (HFA) 30-2 with Swedish Interactive Threshold Algorithm Fast within 6 months after cataract due to glaucoma or suspected glaucoma. Each parameter of HFA was compared among eyes implanted with TENIS Symfony® (EDOF group), diffractive bifocal IOLs (bifocal group), and monofocal IOLs (monofocal group). RESULTS: The total of 76 eyes, including 24 eyes in the EDOF group, 26 eyes in the bifocal group, and 26 eyes in the monofocal group, were included in this study. Mean deviation (MD) of HFA was -0.24±0.58 dB in the EDOF group, -1.38±0.58 dB in the bifocal group, and 0.02±0.44 dB in the monofocal group. Foveal threshold (FT) of HFA was 35.8±1.6 dB in the EDOF group, 33.6±1.7 dB in the bifocal group, and 36.6±1.4 dB in the monofocal group. In both MD and FT, there was significant difference between the bifocal group and the others (p<0.001). There was no difference between the EDOF group and the monofocal group. Moreover, there was no significant difference between the three groups about pattern standard deviation (PSD) of HFA. CONCLUSION: TECNIS Symfony® may have little influence on visual field sensitivity, whereas diffractive bifocal IOLs decrease visual field sensitivity.


Subject(s)
Contrast Sensitivity/physiology , Depth Perception/physiology , Lenses, Intraocular , Visual Fields/physiology , Aged , Female , Humans , Lens Implantation, Intraocular , Male , Middle Aged
4.
PLoS One ; 14(7): e0219405, 2019.
Article in English | MEDLINE | ID: mdl-31335861

ABSTRACT

We previously showed that dietary omega (ω)-3 long-chain polyunsaturated fatty acids (LCPUFAs) suppress inflammation in mice with experimental autoimmune uveitis (EAU). We have now investigated the role of antigen presenting cells (APCs) in this action of ω-3 LCPUFAs. C57BL/6 mice were fed a diet supplemented with ω-3 or ω-6 LCPUFAs for 2 weeks, after which splenocytes were isolated from the mice and cocultured with CD4+ T cells isolated from mice with EAU induced by injection of a human interphotoreceptor retinoid-binding protein peptide together with complete Freund's adjuvant. The proliferation of and production of interferon-γ and interleukin-17 by T cells from EAU mice in vitro were attenuated in the presence of splenocytes from ω-3 LCPUFA-fed mice as compared with those from mice fed ω-6 LCPUFAs. Splenocyte fractionation by magnetic-activated cell sorting revealed that, among APCs, dendritic cells (DCs) were the target of ω-3 LCPUFAs. Adoptive transfer of DCs from mice fed ω-3 LCPUFAs attenuated disease progression in EAU mice as well as the production of pro-inflammatory cytokines by T cells isolated from these latter animals. The proliferation of T cells from control Balb/c mice was also attenuated in the presence of DCs from ω-3 LCPUFA-fed mice as compared with those from ω-6 LCPUFA-fed mice. Furthermore, T cell proliferation in such a mixed lymphocyte reaction was inhibited by prior exposure of DCs from mice fed an ω-6 LCPUFA diet to ω-3 LCPUFAs in vitro. Our results thus suggest that DCs mediate the anti-inflammatory action of dietary ω-3 LCPUFAs in EAU.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Autoimmune Diseases/drug therapy , Dendritic Cells/immunology , Fatty Acids, Omega-3/therapeutic use , Uveitis/drug therapy , Adoptive Transfer , Animals , Anti-Inflammatory Agents/pharmacology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Autoimmune Diseases/complications , Autoimmune Diseases/pathology , Cell Proliferation , Cytokines/metabolism , Dendritic Cells/drug effects , Disease Models, Animal , Fatty Acids, Omega-3/pharmacology , Female , Inflammation/complications , Inflammation/drug therapy , Inflammation/pathology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , Mice, Inbred C57BL , Spleen/drug effects , Spleen/pathology , Th1 Cells/drug effects , Th17 Cells/drug effects , Uveitis/complications , Uveitis/pathology
5.
Nat Biotechnol ; 36(4): 346-351, 2018 04.
Article in English | MEDLINE | ID: mdl-29505028

ABSTRACT

Infiltration, accumulation, and survival of chimeric antigen receptor T (CAR-T) cells in solid tumors is crucial for tumor clearance. We engineered CAR-T cells to express interleukin (IL)-7 and CCL19 (7 × 19 CAR-T cells), as these factors are essential for the maintenance of T-cell zones in lymphoid organs. In mice, 7 × 19 CAR-T cells achieved complete regression of pre-established solid tumors and prolonged mouse survival, with superior anti-tumor activity compared to conventional CAR-T cells. Histopathological analyses showed increased infiltration of dendritic cells (DC) and T cells into tumor tissues following 7 × 19 CAR-T cell therapy. Depletion of recipient T cells before 7 × 19 CAR-T cell administration dampened the therapeutic effects of 7 × 19 CAR-T cell treatment, suggesting that CAR-T cells and recipient immune cells collaborated to exert anti-tumor activity. Following treatment of mice with 7 × 19 CAR-T cells, both recipient conventional T cells and administered CAR-T cells generated memory responses against tumors.


Subject(s)
Cell- and Tissue-Based Therapy , Chemokine CCL19/administration & dosage , Interleukin-7/administration & dosage , Receptors, Antigen, T-Cell/administration & dosage , Receptors, Chimeric Antigen/administration & dosage , Allografts , Animals , Cell Line, Tumor , Cell Survival/drug effects , Chemokine CCL19/genetics , Dendritic Cells/immunology , Dendritic Cells/transplantation , Gene Expression Regulation/drug effects , Interleukin-7/genetics , Mice , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology
6.
J Immunol ; 196(7): 2947-54, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26912321

ABSTRACT

Herpesvirus entry mediator (HVEM), a member of the TNFR superfamily, serves as a unique molecular switch to mediate both stimulatory and inhibitory cosignals, depending on its functions as a receptor or ligand interacting with multiple binding partners. In this study, we explored the cosignaling functions of HVEM in experimental autoimmune uveitis (EAU), a mouse model resembling human autoimmune uveitis conditions such as ocular sarcoidosis and Behcet disease. Our studies revealed that EAU severity significantly decreased in HVEM-knockout mice compared with wild-type mice, suggesting that stimulatory cosignals from the HVEM receptor are predominant in EAU. Further studies elucidated that the HVEM cosignal plays an important role in the induction of both Th1- and Th17-type pathogenic T cells in EAU, including differentiation of IL-17-producing αß(+)γδ(-) conventional CD4(+) T cells. Mice lacking lymphotoxin-like, inducible expression, competes with herpes simplex virus glycoprotein D for HVEM, a receptor expressed by T lymphocytes : LIGHT), B- and T-lymphocyte attenuator (BTLA) or both LIGHT and BTLA are also less susceptible to EAU, indicating that LIGHT-HVEM and BTLA-HVEM interactions, two major molecular pathways mediating HVEM functions, are both important in determining EAU pathogenesis. Finally, blocking HVEM cosignals by antagonistic anti-HVEM Abs ameliorated EAU. Taken together, our studies revealed a novel function of the HVEM cosignaling molecule and its ligands in EAU pathogenesis through the induction of Th1- and Th17-type T cell responses and suggested that HVEM-related molecular pathways can be therapeutic targets in autoimmune uveitis.


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Receptors, Tumor Necrosis Factor, Member 14/genetics , Th1 Cells/immunology , Th17 Cells/immunology , Uveitis/genetics , Uveitis/immunology , Animals , Antibodies, Monoclonal/pharmacology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/metabolism , Cell Differentiation/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Ligands , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Signal Transduction , Th1 Cells/cytology , Th1 Cells/metabolism , Th17 Cells/cytology , Th17 Cells/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Uveitis/drug therapy , Uveitis/metabolism
7.
PLoS One ; 10(9): e0138241, 2015.
Article in English | MEDLINE | ID: mdl-26393358

ABSTRACT

Omega (ω)-3 long-chain polyunsaturated fatty acids (LCPUFAs) inhibit the production of inflammatory mediators and thereby contribute to the regulation of inflammation. Experimental autoimmune uveitis (EAU) is a well-established animal model of autoimmune retinal inflammation. To investigate the potential effects of dietary intake of ω-3 LCPUFAs on uveitis, we examined the anti-inflammatory properties of these molecules in comparison with ω-6 LCPUFAs in a mouse EAU model. C57BL/6 mice were fed a diet containing ω-3 LCPUFAs or ω-6 LCPUFAs for 2 weeks before as well as after the induction of EAU by subcutaneous injection of a fragment of human interphotoreceptor retinoid-binding protein emulsified with complete Freund's adjuvant. Both clinical and histological scores for uveitis were smaller for mice fed ω-3 LCPUFAs than for those fed ω-6 LCPUFAs. The concentrations of the T helper 1 (Th1) cytokine interferon-γ and the Th17 cytokine interleukin-17 in intraocular fluid as well as the production of these cytokines by lymph node cells were reduced for mice fed ω-3 LCPUFAs. Furthermore, the amounts of mRNAs for the Th1- and Th17-related transcription factors T-bet and RORγt, respectively, were reduced both in the retina and in lymph node cells of mice fed ω-3 LCPUFAs. Our results thus show that a diet enriched in ω-3 LCPUFAs suppressed uveitis in mice in association with inhibition of Th1 and Th17 cell function.


Subject(s)
Autoimmune Diseases/prevention & control , Diet , Fatty Acids, Omega-3/pharmacology , Th1 Cells/drug effects , Th17 Cells/drug effects , Uveitis/prevention & control , Animals , Autoimmune Diseases/blood , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Fatty Acids, Omega-3/administration & dosage , Female , Gene Expression/drug effects , Humans , Inflammation Mediators/metabolism , Lipids/blood , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Retina/drug effects , Retina/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Box Domain Proteins/genetics , Th1 Cells/metabolism , Th17 Cells/metabolism , Uveitis/blood , Uveitis/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...