Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Invest Ophthalmol Vis Sci ; 65(2): 25, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38345552

ABSTRACT

Purpose: To evaluate the expression of sry-box transcription factor 9 (SOX9) in orbital fibroblasts (OFs) of thyroid eye disease (TED) and to find its potential role and underlying mechanism in orbital fibrosis. Methods: OFs were cultured from orbital connective tissues obtained from patients with TED (n = 10) and healthy controls (n = 6). SOX9 was depleted by small interfering RNA or overexpressed through lentivirus transduction in OFs. Fibroblast contractile activity was measured by collagen gel contraction assay and proliferation was examined by EdU assay. Transcriptomic changes were assessed by RNA sequencing. Results: The mRNA and protein levels of SOX9 were significantly higher in OFs cultured from patients with TED than those from healthy controls. Extracellular matrix-related genes were down-regulated by SOX9 knockdown and up-regulated by SOX9 overexpression in TED-OFs. SOX9 knockdown significantly decrease the contraction and the antiapoptotic ability of OFs, whereas the overexpression of SOX9 increased the ability of transformation, migration, and proliferation of OFs. SOX9 knockdown suppressed the expression of phosphorylated ERK1/2, whereas its overexpression showed the opposite effect. Epidermal growth factor receptor (EGFR) is one of the notably down-regulated genes screened out by RNA sequencing. Chromatin immunoprecipitation-qPCR demonstrated SOX9 binding to the EGFR promoter. Conclusions: A high expression of SOX9 was found in TED-OFs. SOX9 can activate OFs via MAPK/ERK1/2 signaling pathway, which in turn promotes proliferation and differentiation of OFs. EGFR was a downstream target gene of SOX9. SOX9/EGFR can be considered as therapeutic targets for the treatment of orbital fibrosis in TED.


Subject(s)
Graves Ophthalmopathy , Humans , Graves Ophthalmopathy/genetics , Graves Ophthalmopathy/metabolism , Orbit/metabolism , MAP Kinase Signaling System , ErbB Receptors/metabolism , Fibroblasts/metabolism , Fibrosis , Cells, Cultured , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism
2.
Antioxidants (Basel) ; 12(6)2023 May 29.
Article in English | MEDLINE | ID: mdl-37371903

ABSTRACT

Retinal ischemia-reperfusion (RIR) injury caused by high intraocular pressure (IOP) is an important risk factor contributing to retinal ganglion cell (RGC) death, eventually causing blindness. A key progressive pathological process in the development of RIR is the death of RGCs. However, the detailed mechanisms underlying RGC death caused by RIR have not yet been clearly elucidated, and effective treatments are lacking. Ferroptosis is a recently defined form of programmed cell death that is closely related to organ injury. Melatonin (MT) is a promising neuroprotective agent, but its effects on RIR injury remain unclear. In this study, murine models of acute ocular hypertension and oxygen and glucose deprivation/reoxygenation (OGD/R) model were adopted to simulate retinal ischemia. MT alleviated retinal damage and RGC death in RIR mice, significantly attenuating RIR-induced ferroptosis. Furthermore, MT reduced the expression of p53, a master regulator of ferroptosis pathways, and the upregulation of p53 promoted ferroptosis and largely abolished the neuroprotective effects of MT. Mechanistically, the overexpression (OE) of p53 suppressed the expression of the solute carrier family 7 member 11 (Slc7a11), which was accompanied by increased 12-lipoxygenase (Alox12) expression, triggering retinal ferroptosis. Moreover, MT-ameliorated apoptosis, neuroinflammation and microglial activation were observed. In summary, MT conferred neuroprotection against RIR injury by inhibiting p53-mediated ferroptosis. These findings indicate that MT is a retina-specific ferroptosis inhibitor and a promising therapeutic agent for retinal neuroprotection.

3.
Exp Eye Res ; 232: 109515, 2023 07.
Article in English | MEDLINE | ID: mdl-37207866

ABSTRACT

Lutein (LU) is a carotenoid that has recently been implicated in multiple roles in fibrosis, inflammation, and oxidative stress. Thyroid-associated ophthalmopathy (TAO) is particularly relevant to these pathological changes. We thus aim to probe the potential therapeutic effects of TAO in an in vitro model. We used LU pre-treating OFs derived from patients with TAO or not, then treated with TGF-ß1(or IL-1ß)to induce fibrosis (or inflammation). We analyzed the different expressions of related genes and proteins, and the molecular mechanism pathway on TAO OFs was screened by RNA sequencing, which is identified in vitro. We found that LU attenuates fibrotic and inflammatory effects in TAO. LU inhibited ACTA2, COL1A1, FN1, and CTGF mRNA expression and suppressed α-SMA, and FN1 protein expression induced by TGF-ß1. Besides, LU suppressed OFs migration. Besides, it is shown that LU suppressed inflammation-related genes, such as IL-6, IL-8, CXCL1, and MCP-1. Moreover, LU inhibited oxidative stress induced by IL-1ß, which is analyzed by DHE fluorescent probe staining. RNA sequencing suggested ERK/AP-1 pathway may be the molecular mechanism of LU protective effect on TAO, which is identified by RT-qPCR and western-blot. In summary, this study provides the first evidence that LU significantly attenuates the pathogenic manifestations of TAO by inhibiting the expression of fibrotic and inflammation-related genes and ROS produced by OFs. These data suggested that LU may be a potential medicine for TAO.


Subject(s)
Graves Ophthalmopathy , Humans , Graves Ophthalmopathy/metabolism , Lutein/pharmacology , Transforming Growth Factor beta1/pharmacology , Orbit/metabolism , Inflammation/metabolism , Fibroblasts/metabolism , Fibrosis , Cells, Cultured
4.
Medicina (Kaunas) ; 59(3)2023 Mar 22.
Article in English | MEDLINE | ID: mdl-36984631

ABSTRACT

Background and objectives: Previous studies on ptosis recurrence after correction surgery have tended to focus on postoperative complications, surgical methods and suspension materials, few have mentioned refractive error. This research is to investigate the potential relation between refractive error and recurrence after correction surgery in pediatric patients with simple congenital ptosis. Materials and Methods: We conducted a retrospective analysis of data from patients with simple congenital ptosis who were treated at Zhongshan Ophthalmic Center (ZOC) between 2017 and 2020. In total, 111 eyelids of 85 patients without surgery-related complications who underwent frontalis muscle flap suspension (FMFS) for simple congenital ptosis were included. Postoperative changes in eyelid height were assessed. Cycloplegic refraction was assessed before surgery and during the follow-up period (every 3 months after surgery). Recurrence in the postoperative period was defined as a marginal reflex distance 1 (MRD1) of <1 mm. Results: There were 16 recurrence and 69 non-recurrence cases, with no statistically significant differences, in terms of patient age at the time of surgery, patient sex, or preoperative MRD1, between the recurrence and non-recurrence groups. The postoperative cylindrical diopter (adjusted odds ratio [OR] = 0.432, p = 0.005), laterality (adjusted OR = 0.202, p = 0.006), and preoperative MRD1 (adjusted OR = 0.617, p = 0.019) were associated with ptosis recurrence after surgery. Differences between the recurrence and non-recurrence groups in spherical diopter and spherical equivalent (SE) before and after surgery were not statistically significant. In addition, preoperative refractive error and postoperative spherical diopter were not significantly associated with ptosis recurrence after correction surgery. Conclusions: Ptosis recurrence after FMFS in pediatric cases of congenital ptosis is associated with refractive error. Timely refractive correction and amblyopia treatment may help to reduce ptosis recurrence.


Subject(s)
Blepharoptosis , Refractive Errors , Child , Humans , Retrospective Studies , Oculomotor Muscles/surgery , Blepharoptosis/surgery , Blepharoptosis/congenital , Eyelids/surgery , Refractive Errors/etiology , Treatment Outcome
5.
Ann Transl Med ; 10(8): 432, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35571431

ABSTRACT

Background: Corneal neovascularization (CNV) caused by alkali burn injury is tightly associated with an inflammatory reaction and can lead to vision loss. Melatonin is involved in anti-inflammation and anti-angiogenesis, but its role in CNV has not yet been investigated. Methods: We induced CNV using sodium hydroxide (NaOH) and compared the reactions of vehicle control and melatonin-treated male C57BL/6 mice at 7 and 14 days following the corneal burn. The infiltration of inflammatory cells and the expression of proangiogenic factors, chemokines, and inflammation-related molecules were quantified via immunohistochemical (IHC) analysis and quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR), respectively. Murine peritoneal macrophages were used in vitro to further verify the effect of melatonin in inflammatory CNV. Results: Compared with the vehicle control mice, the melatonin-treated mice showed significant inhibition of angiogenesis and reduction of corneal epithelial defects in alkali-burned corneas. Concomitantly, the infiltration of inflammatory cells and F4/80+ cells were dramatically reduced after melatonin treatment. The messenger RNA (mRNA) expression of proangiogenic factors [vascular endothelial growth factors (VEGF) and matrix metalloproteinase-9 (MMP-9)], monocyte chemotactic protein-1 (MCP-1), nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3), and pro-inflammatory cytokines [tumor necrosis factor-alpha (TNF-α) and interleukin-1ß (IL-1ß) and IL-6] were down-regulated in the melatonin-treated mice. Moreover, melatonin inhibited the expression of these factors in murine peritoneal macrophages. Conclusions: Melatonin inhibits the neovascular and inflammatory responses in corneal alkali burn injury, suggesting that it may be a potential therapy for CNV.

6.
Front Endocrinol (Lausanne) ; 13: 846106, 2022.
Article in English | MEDLINE | ID: mdl-35273577

ABSTRACT

Orbital fibrosis is a hallmark of tissue remodeling in thyroid-associated ophthalmopathy (TAO). Previous studies have shown that interleukin (IL)-11 plays a pivotal profibrotic role in various inflammatory and autoimmune diseases. However, the expression pattern of IL-11 in patients with TAO and whether IL-11 is mechanistically linked with pathological fibrosis remains unknown. In this study, we investigated IL-11 levels in the serum and orbital connective tissue of patients with TAO, and evaluated the correlation of these levels with the patient's clinical activity score. We also evaluated the expression pattern of IL-11Rα in orbital connective tissue. Furthermore, we elucidated the regulatory factors, profibrotic function, and downstream signaling pathways for IL-11 in TAO using in vitro studies. IL-11 levels in serum and orbital connective tissues were increased in patients with TAO, as compared with healthy controls. In addition, both levels were positively correlated with disease activity. Single-cell RNA sequencing of orbital connective tissue indicated that IL-11Rα was dominantly expressed in orbital fibroblasts (OFs). RNA sequencing of paired unstimulated and transforming growth factor (TGF)-ß1-stimulated samples demonstrated that upregulation of IL-11 expression defined the dominant transcriptional response. IL-11 signaling was also confirmed to be downstream of TGF-ß1 and IL-1ß. Therefore, we deduced that IL-11 protein is secreted in an autocrine loop in TAO. We also indicated that IL-11 mediated the profibrotic phenotype switch by inducing the expression of myofibroblast differentiation markers, including α-smooth muscle actin and collagen type I α1, which could be abrogated by an anti-IL-11 neutralizing antibody. Furthermore, we revealed that extracellular regulated protein kinase may be a crucial factor in the pro-fibrotic, translationally specific signaling activity of IL-11. These data demonstrate that IL-11 plays a crucial role in orbital fibroblast phenotype switching and may be a potential therapeutic target candidate for the treatment of TAO.


Subject(s)
Graves Ophthalmopathy , Interleukin-11/metabolism , Fibroblasts/metabolism , Fibrosis , Graves Ophthalmopathy/metabolism , Humans , Interleukin-11/genetics , Orbit/pathology , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL