Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
1.
Heliyon ; 10(8): e29775, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38699726

ABSTRACT

Objective: To develop an algorithm using deep learning methods to calculate the volume of intraretinal and subretinal fluid in optical coherence tomography (OCT) images for assessing diabetic macular edema (DME) patients' condition changes. Design: Cross-sectional study. Participants: Treatment-naive patients diagnosed with DME recruited from April 2020 to November 2021. Methods: The deep learning network, which was built for autonomous segmentation utilizing an encoder-decoder network based on the U-Net architecture, was used to calculate the volume of intraretinal fluid (IRF) and subretinal fluid (SRF). The alterations of retinal vessel density and thickness, and the correlation between best-corrected visual acuity (BCVA) and OCT parameters were analyzed. Results: 2,955 OCT images of fourteen eyes from DME patients with IRF and SRF who received anti-vascular endothelial growth factor (VEGF) agents were obtained. The area under the curve (AUC) of the receiver operating characteristic (ROC) curve of the algorithm was 0.993 for IRF and 0.998 for SRF. The volumes of IRF and SRF were significantly decreased from 1.93 ± 0.58 /1.14 ± 0.25 mm3 (baseline) to 0.26 ± 0.13 /0.26 ± 0.18 mm3 (post-injection), respectively (p = 0.0170 for IRF, and p = 0.0004 for SRF). The Spearman correlation demonstrated that the reduction of IRF volume was negatively correlated with age (coefficient = -0.698, p = 0.006). Conclusion: We developed a deep learning assisted fluid volume calculation algorithm with high sensitivity and specificity for assessing the volume of IRF and SRF in DME patients. Key words: deep learning; diabetic macular edema; optical coherence tomography.

2.
Int J Biol Macromol ; 268(Pt 2): 131678, 2024 May.
Article in English | MEDLINE | ID: mdl-38657921

ABSTRACT

BACKGROUND: Glia maturation factor beta (GMFB) is a growth and differentiation factor that acts as an intracellular regulator of signal transduction pathways. The small ubiquitin-related modifier (SUMO) modification, SUMOylation, is a posttranslational modification (PTM) that plays a key role in protein subcellular localization, stability, transcription, and enzymatic activity. Recent studies have highlighted the importance of SUMOylation in the inflammation and progression of numerous diseases. However, the relationship between GMFB and SUMOylation is unclear. RESULTS: Here, we report for the first time that GMFB and SUMO1 are markedly increased in retinal pigment epithelial (RPE) cells at the early stage of diabetes mellitus (DM) under hyperglycemia. The GMFΒ protein could be mono-SUMOylated by SUMO1 at the K20, K35, K58 or K97 sites. SUMOylation of GMFB led to its increased protein stability and subcellular translocation. Furthermore, deSUMOylation of GMFΒ downregulates multiple signaling pathways, including the Jak-STAT signaling pathway, p38 pathway and NF-kappa B signaling pathway. CONCLUSIONS: This work provides novel insight into the role of SUMOylated GMFB in RPE cells and provides a novel therapeutic target for diabetic retinopathy (DR).


Subject(s)
Hyperglycemia , Protein Stability , Retinal Pigment Epithelium , Signal Transduction , Sumoylation , Retinal Pigment Epithelium/metabolism , Hyperglycemia/metabolism , Humans , Epithelial Cells/metabolism , Cell Line , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , NF-kappa B/metabolism , SUMO-1 Protein/metabolism
3.
J Neuroinflammation ; 21(1): 75, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38532410

ABSTRACT

BACKGROUND: Neovascular age-related macular degeneration (nAMD), accounts for up to 90% of AMD-associated vision loss, ultimately resulting in the formation of fibrotic scar in the macular region. The pathogenesis of subretinal fibrosis in nAMD involves the process of epithelial-mesenchymal transition (EMT) occurring in retinal pigment epithelium (RPE). Here, we aim to investigate the underlying mechanisms involved in the Wnt signaling during the EMT of RPE cells and in the pathological process of subretinal fibrosis secondary to nAMD. METHODS: In vivo, the induction of subretinal fibrosis was performed in male C57BL/6J mice through laser photocoagulation. Either FH535 (a ß-catenin inhibitor) or Box5 (a Wnt5a inhibitor) was intravitreally administered on the same day or 14 days following laser induction. The RPE-Bruch's membrane-choriocapillaris complex (RBCC) tissues were collected and subjected to Western blot analysis and immunofluorescence to examine fibrovascular and Wnt-related markers. In vitro, transforming growth factor beta 1 (TGFß1)-treated ARPE-19 cells were co-incubated with or without FH535, Foxy-5 (a Wnt5a-mimicking peptide), Box5, or Wnt5a shRNA, respectively. The changes in EMT- and Wnt-related signaling molecules, as well as cell functions were assessed using qRT-PCR, nuclear-cytoplasmic fractionation assay, Western blot, immunofluorescence, scratch assay or transwell migration assay. The cell viability of ARPE-19 cells was determined using Cell Counting Kit (CCK)-8. RESULTS: The in vivo analysis demonstrated Wnt5a/ROR1, but not Wnt3a, was upregulated in the RBCCs of the laser-induced CNV mice compared to the normal control group. Intravitreal injection of FH535 effectively reduced Wnt5a protein expression. Both FH535 and Box5 effectively attenuated subretinal fibrosis and EMT, as well as the activation of ß-catenin in laser-induced CNV mice, as evidenced by the significant reduction in areas positive for fibronectin, alpha-smooth muscle actin (α-SMA), collagen I, and active ß-catenin labeling. In vitro, Wnt5a/ROR1, active ß-catenin, and some other Wnt signaling molecules were upregulated in the TGFß1-induced EMT cell model using ARPE-19 cells. Co-treatment with FH535, Box5, or Wnt5a shRNA markedly suppressed the activation of Wnt5a, nuclear translocation of active ß-catenin, as well as the EMT in TGFß1-treated ARPE-19 cells. Conversely, treatment with Foxy-5 independently resulted in the activation of abovementioned molecules and subsequent induction of EMT in ARPE-19 cells. CONCLUSIONS: Our study reveals a reciprocal activation between Wnt5a and ß-catenin to mediate EMT as a pivotal driver of subretinal fibrosis in nAMD. This positive feedback loop provides valuable insights into potential therapeutic strategies to treat subretinal fibrosis in nAMD patients.


Subject(s)
Macular Degeneration , Sulfonamides , beta Catenin , Humans , Male , Animals , Mice , beta Catenin/metabolism , Wnt-5a Protein , Mice, Inbred C57BL , Retinal Pigment Epithelium/metabolism , Epithelial-Mesenchymal Transition , Macular Degeneration/metabolism , Fibrosis , RNA, Small Interfering/metabolism
4.
J Proteome Res ; 23(3): 916-928, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38367214

ABSTRACT

Myopia accounts for a significant proportion of visual lesions worldwide and has the potential to progress toward pathological myopia. This study aims to reveal the difference in protein content in aqueous humor between high myopic and nonhigh myopic patients, as well as better understand the dysregulation of proteins in myopic eyes. Aqueous humor was collected for liquid chromatograph mass spectrometer (LC/MS) analysis from 30 individual eyes that underwent phacoemulsification and intraocular lens (IOL) implantation. Results showed that a total of 190 differentially expressed proteins were identified, which revealed their involvement in cell metabolism, immune and inflammatory response, and system and anatomical structure. Further analysis focused on 15 intensively interacted hub proteins, encompassing functions related to complement cascades, lipoprotein metabolism, and fibrin biological function. Subsequent validations demonstrated elevated levels of APOE (apolipoprotein E), C3 (complement 3), and AHSG (α-2-HS-glycoprotein) in the high myopia group (31 eyes of cataracts and 45 eyes of high myopia with cataracts). AHSG had a significant positive correlation with axial length in high myopic patients, with good efficacy in distinguishing between myopic and nonmyopic groups. AHSG may be a potential indicator of the pathological severity and participator in the pathological progress of high myopia. This study depicted differential expression characteristics of aqueous humor in patients with high myopia and provided optional information for further experimental research on exploring the molecular mechanisms and potential therapeutic targets for high myopia. Data are available via ProteomeXchange with the identifier PXD047584.


Subject(s)
Cataract Extraction , Cataract , Myopia , Humans , Aqueous Humor , Proteomics
5.
Clin Epigenetics ; 16(1): 6, 2024 01 03.
Article in English | MEDLINE | ID: mdl-38172938

ABSTRACT

BACKGROUND: Studies have shown that tet methylcytosine dioxygenase 2 (TET2) is highly expressed in diabetic retinopathy (DR), which reduces the DNA methylation of downstream gene promoters and activates the transcription. Abnormally expressed TET2 and downstream genes in a high-glucose environment are associated with retinal capillary leakage and neovascularization. Here, we investigated the downstream genes of TET2 and its potential association with neovascularization in proliferative diabetic retinopathy (PDR). METHODS: GSE60436, GSE57362, and GSE158333 datasets were analyzed to identify TET2-related hypomethylated and upregulated genes in PDR. Gene expression and promoter methylation of these genes under high glucose treatment were verified. Moreover, TET2 knockdown was used to assess its impact on tube formation and migration in human retinal microvascular endothelial cells (HRMECs), as well as its influence on downstream genes. RESULTS: Our analysis identified three key genes (PARVB, PTPRE, ECM1) that were closely associated with TET2 regulation. High glucose-treated HRMECs exhibited increased expression of TET2 and ECM1 while decreasing the promoter methylation level of ECM1. Subsequently, TET2 knockdown led to decreased migration ability and tube formation function of HRMECs. We further found a decreased expression of PARVB, PTPRE, and ECM1, accompanied by an increase in the promoter methylation of ECM1. CONCLUSIONS: Our findings indicate the involvement of dysregulated TET2 expression in neovascularization by regulating the promoter methylation and transcription of downstream genes (notably ECM1), eventually leading to PDR. The TET2-induced hypomethylation of downstream gene promoters represents a potential therapeutic target and offers a novel perspective on the mechanism underlying neovascularization in PDR.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Dioxygenases , Humans , Diabetic Retinopathy/genetics , DNA Methylation , Endothelial Cells/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Glucose/pharmacology , Glucose/metabolism , Diabetes Mellitus/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases/genetics , Dioxygenases/metabolism
6.
Glia ; 72(3): 504-528, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37904673

ABSTRACT

Retinal degeneration, characterized by Müller cell gliosis and photoreceptor apoptosis, is considered an early event in diabetic retinopathy (DR). Our previous study proposed that GMFB may mediate diabetic retinal degeneration. This study identified GMFB as a sensitive and functional gliosis marker for DR. Compared to the wild type (WT) group, Gmfb knockout (KO) significantly improved visual function, attenuated gliosis, reduced the apoptosis of neurons, and decreased the mRNA levels of tumor necrosis factor α (Tnf-α) and interleukin-1ß (Il-1ß) in diabetic retinas. Tgf-ß3 was enriched by hub genes using RNA sequencing in primary WT and KO Müller cells. Gmfb KO significantly upregulated the transforming growth factor (TGF)-ß3 protein level via the AKT pathway. The protective effect of TGF-ß3 in the vitreous resulted in significantly improved visual function and decreased the number of apoptotic cells in the diabetic retina. The protection of Gmfb KO in primary Müller cells against high glucose (HG)-induced photoreceptor apoptosis was partially counteracted by TGF-ß3 antibody and administration of TGFBR1/2 inhibitors. Nuclear receptor subfamily 3 group C member 1 (NR3C1) binds to the promoter region of Gmfb and regulates Gmfb mRNA at the transcriptional level. NR3C1 was increased in the retinas of early diabetic rats but decreased in the retinas of late diabetic rats. N'-[(1E)-(3-Methoxyphenyl)Methylene]-3-Methyl-1H-Pyrazole-5-Carbohydrazide (DS-5) was identified as an inhibitor of GMFB, having a protective role in DR. We demonstrated that GMFB/AKT/TGF-ß3 mediated early diabetic retinal degeneration in diabetic rats. This study provides a novel therapeutic strategy for treating retinal degeneration in patients with DR.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Retinopathy , Retinal Degeneration , Humans , Rats , Animals , Retinal Degeneration/pathology , Ependymoglial Cells/metabolism , Streptozocin/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Transforming Growth Factor beta3/adverse effects , Transforming Growth Factor beta3/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Gliosis/pathology , Retina/metabolism , Diabetic Retinopathy/pathology , RNA, Messenger/metabolism
7.
Retina ; 44(4): 680-688, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38011844

ABSTRACT

PURPOSE: To investigate the effectiveness of two regimens of ranibizumab-assisted pars plana vitrectomy in the treatment of patients with proliferative diabetic retinopathy. METHODS: This is a prospective, 6-month, randomized controlled trial. Eighty patients with 87 eyes requiring pars plana vitrectomy treatment for proliferative diabetic retinopathy were included and randomly divided into a 1.0-mg injection group and a 0.5-mg injection group. The ranibizumab was delivered intraoperatively, at the close of surgery. The vitreous hemorrhage grade, best-corrected visual acuity, central macular thickness, and safety data were assessed to Month 6. RESULTS: The 1.0-mg injection group had a milder grade and a lower reoccurrence rate of early postoperatively vitreous hemorrhage than the 0.5-mg injection group (35.0% and 63.4%, respectively, P = 0.0195). The mean best-corrected visual acuity of two groups was significantly improved from baseline to 6 months after surgery, 1.60 ± 0.72 Logarithm of the Minimum Angle of Resolution (LogMAR) (<20/200) to 0.47 ± 0.49 LogMAR (20/59) for the 1.0-mg injection group and 1.51 ± 0.69 LogMAR (<20/200) to 0.50 ± 0.31 LogMAR (20/63) for the 0.5-mg injection group, but there was no significant difference between the two groups ( P = 0.74). There was no significant difference in the mean decrease in central macular thickness and probability of postoperative adverse events between the two groups. CONCLUSION: Intravitreal injection of 1.0 mg of ranibizumab after pars plana vitrectomy compared with the recommended dose of 0.5 mg significantly reduced the recurrence and severity of early postoperative vitreous hemorrhage in patients with proliferative diabetic retinopathy. It also contributed to the early recovery of visual acuity after surgery and did not increase postoperative adverse events.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Humans , Diabetic Retinopathy/drug therapy , Intravitreal Injections , Prospective Studies , Ranibizumab/adverse effects , Ranibizumab/therapeutic use , Treatment Outcome , Vitrectomy/adverse effects , Vitreous Hemorrhage/surgery
8.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 166995, 2024 03.
Article in English | MEDLINE | ID: mdl-38142757

ABSTRACT

Diabetic retinopathy (DR) is the most common reason for blindness in working-age individuals globally. Prolonged high blood glucose is a main causative factor for DR development, and glucose transport is prerequisite for the disturbances in DR caused by hyperglycemia. Glucose transport is mediated by its transporters, including the facilitated transporters (glucose transporter, GLUTs), the "active" glucose transporters (sodium-dependent glucose transporters, SGLTs), and the SLC50 family of uniporters (sugars will eventually be exported transporters, SWEETs). Glucose transport across the blood-retinal barrier (BRB) is crucial for nourishing the neuronal retina in the context of retinal physiology. This physiological process primarily relies on GLUTs and SGLTs, which mediate the glucose transportation across both the cell membrane of retinal capillary endothelial cells and the retinal pigment epithelium (RPE). Under diabetic conditions, increased accumulation of extracellular glucose enhances the retinal cellular glucose uptake and metabolism via both glycolysis and glycolytic side branches, which activates several biochemical pathways, including the protein kinase C (PKC), advanced glycation end-products (AGEs), polyol pathway and hexosamine biosynthetic pathway (HBP). These activated biochemical pathways further increase the production of reactive oxygen species (ROS), leading to oxidative stress and activation of Poly (ADP-ribose) polymerase (PARP). The activated PARP further affects all the cellular components in the retina, and finally resulting in microangiopathy, neurodegeneration and low-to-moderate grade inflammation in DR. This review aims to discuss the changes of glucose transport, glucose transporters, as well as its metabolism in DR, which influences the retinal neurovascular unit (NVU) and implies the possible therapeutic strategies for treating DR.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Hyperglycemia , Humans , Diabetic Retinopathy/metabolism , Endothelial Cells/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Retina/metabolism , Glucose/metabolism , Hyperglycemia/metabolism , Glycation End Products, Advanced/metabolism , Diabetes Mellitus/metabolism
9.
Stem Cell Res Ther ; 14(1): 281, 2023 10 02.
Article in English | MEDLINE | ID: mdl-37784129

ABSTRACT

BACKGROUND: T helper 2 (Th2) cells are thought to play critical roles in allergic conjunctivitis (AC). They release inflammatory cytokines to promote an allergic response in AC. Due to individual heterogeneity and long-term chronic management, current therapies do not always effectively control AC. Mesenchymal stem cells (MSCs) have been shown to be effective in treating allergy-related disorders, but it is unclear how exactly the Th2-mediated allergic response is attenuated. This study aims to elucidate the therapeutic effect and mechanism of the human umbilical cord MSCs (hUCMSCs) in a mouse model of experimental AC (EAC). METHODS: A mouse EAC model was established by inoculating short ragweed (SRW) pollen. After the SRW pollen challenge, the mice received a single subconjunctival or tail vein injection of 2 × 106 hUCMSCs, or subconjunctival injection of hUCMSCs conditioned medium (hUCMSC-CM), and dexamethasone eye drops was used as positive control; subsequent scratching behavior and clinical symptoms were assessed. Immunostaining and flow cytometry were carried out to show allergic reactions and the activation of CD4 + T cell subsets in the conjunctiva and cervical lymph nodes (CLNs). Gene expression was determined by RNA-seq and further verified by qRT-PCR and Western blot. Co-culture assays were performed to explore the regulatory role of hUCMSCs in the differentiation of CD4 + naive T cells (Th0) into Th2 cells. RESULTS: Subconjunctival administration of hUCMSCs resulted in fewer instances of scratching and lower inflammation scores in EAC mice compared to the tail vein delivery, hUCMSC-CM and control groups. Subconjunctival administration of hUCMSCs reduced the number of activated mast cells and infiltrated eosinophils in the conjunctiva, as well as decreased the number of Th2 cells in CLNs. After pretreatment with EAC mouse serum in vitro to mimic the in vivo milieu, hUCMSCs were able to inhibit the differentiation of Th0 into Th2 cells. Further evidence demonstrated that repression of Th2 cell differentiation by hUCMSCs is mediated by CRISPLD2 through downregulation of STAT6 phosphorylation. Additionally, hUMCSCs were able to promote the differentiation of Th0 cells into regulatory T cells in CLNs of EAC mice. CONCLUSIONS: Subconjunctival injection of hUCMSCs suppressed the Th2-allergic response and alleviated clinical symptoms. This study provides not only a potential therapeutic target for the treatment of AC but also other T cell-mediated diseases.


Subject(s)
Conjunctivitis, Allergic , Mesenchymal Stem Cells , Humans , Animals , Mice , Conjunctivitis, Allergic/drug therapy , Conjunctivitis, Allergic/pathology , Conjunctiva/metabolism , Conjunctiva/pathology , Cytokines/metabolism , Disease Models, Animal , Mesenchymal Stem Cells/metabolism , Umbilical Cord
10.
Front Med (Lausanne) ; 10: 1202691, 2023.
Article in English | MEDLINE | ID: mdl-37601785

ABSTRACT

Background: Spontaneous lens absorption is rare and usually occurs in eyes with certain syndromes, hyper-mature cataracts, and ocular trauma. The application of an implantable collamer lens (ICL) is widely performed in patients with high myopia for refractive correction. This study reports a case of spontaneous lens absorption after ICL implantation. Case summary: A 23-year-old man was referred with complaints of poor vision in his left eye. The patient had undergone binocular ICL implantation for refractive correction of high myopia 1.5 years prior. Approximately 10 months later, he experienced a sudden loss of vision and pain in his left eye, which resolved spontaneously the next day without any treatment. Since then, the visual acuity in his left eye gradually decreased. At presentation, slit-lamp examination revealed an ICL in the posterior chamber of both eyes, with anterior capsular fibrotic proliferation and posterior capsular opacity, and the residual lens cortex sandwiched between the anterior fibrotic membrane and opacified posterior lens capsule in his left eye. The number of corneal endothelial cells in his left eye was 1,337, which was lower than before ICL implantation (2,902). The patient then underwent ICL extraction, anterior capsular capsulotomy, residual cortex aspiration, posterior capsular polishing, and intraocular lens implantation. Conclusions: Spontaneous lens absorption may occur in patients with ICL implantation. Patients should undergo routine follow-ups after ICL implantation.

11.
Neurobiol Dis ; 185: 106250, 2023 09.
Article in English | MEDLINE | ID: mdl-37536385

ABSTRACT

Age-related macular degeneration (AMD) is a leading cause of vision loss among elderly people in developed countries. Neovascular AMD (nAMD) accounts for more than 90% of AMD-related vision loss. At present, intravitreal injection of anti-vascular endothelial growth factor (anti-VEGF) is widely used as the first-line therapy to decrease the choroidal and retinal neovascularizations, and thus to improve or maintain the visual acuity of the patients with nAMD. However, about 1/3 patients still progress to irreversible visual impairment due to subretinal fibrosis even with adequate anti-VEGF treatment. Extensive literatures support the critical role of epithelial-mesenchymal transformation (EMT) of retinal pigment epithelium (RPE) in the pathogenesis of subretinal fibrosis in nAMD, but the underlying mechanisms still remain largely unknown. This review summarized the molecular pathogenesis of subretinal fibrosis in nAMD, especially focusing on the transforming growth factor-ß (TGF-ß)-induced EMT pathways. It was also discussed how these pathways crosstalk and respond to signals from the microenvironment to mediate EMT and contribute to the progression of nAMD-related subretinal fibrosis. Targeting EMT signaling pathways might provide a promising and effective therapeutic strategy to treat subretinal fibrosis secondary to nAMD.


Subject(s)
Retinal Pigment Epithelium , Wet Macular Degeneration , Humans , Aged , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/therapeutic use , Epithelial-Mesenchymal Transition , Vascular Endothelial Growth Factor A/metabolism , Visual Acuity , Wet Macular Degeneration/drug therapy , Wet Macular Degeneration/metabolism , Wet Macular Degeneration/pathology , Fibrosis
12.
Phytomedicine ; 116: 154865, 2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37201365

ABSTRACT

BACKGROUND: Subretinal fibrosis (SF) accounts for vision loss in patients with neovascular age-related macular degeneration (nAMD) even treated with adequate intravitreal injection of anti-vascular endothelial growth factor (anti-VEGF) drugs. Currently, there is no treatment available to prevent or treat SF caused by nAMD. PURPOSE: This study aims to investigate the potential effects of luteolin on SF and epithelial-mesenchymal transition (EMT) as well as the underlying molecular pathways both in vivo and in vitro. METHODS: Seven-week-old male C57BL/6J mice were employed to establish laser-induced choroidal neovascularization (CNV) and SF. One day after the laser induction, luteolin was administered intravitreally. SF and CNV were assessed with the immunolabeling of collagen type I (collagen I) and isolectin B4 (IB4), respectively. RPE65 and α-SMA colocalization in the lesions was used to evaluate the extent of EMT in retinal pigment epithelial (RPE) cells by using immunofluorescence. In vitro, luteolin was administered to TGFß1-treated primary human RPE (phRPE) cells. RT-qPCR, Western blot and immunofluorescence were employed to evaluate the change of EMT-related molecules, epithelial markers, and relevant signaling pathways. The functional changes associated with EMT were investigated using the scratch assay, Transwell migration assay, and collagen gel contraction assay. CCK-8 was used to determine the cell viability of phRPE cells. RESULTS: On day 7 and 14 after laser induction in mice, intravitreal injection of luteolin dramatically decreased the immunolabeled sizes of both collagen I and IB4, as well as the amount of colocalized double immunostaining of α-SMA and RPE65 in laser-induced SF lesions. In vitro, TGFß1-treated phRPE cells demonstrated increased cell migration and contraction capacity, accompanied with considerable overexpression of fibronectin, α-SMA, N-cadherin and vimentin, as well as downregulation of E-cadherin and ZO-1. The above changes were largely inhibited by luteolin co-incubation. Mechanistically, luteolin could evidently decrease the phosphorylation of Smad2/3, whereas increase the phosphorylation of YAP in TGFß1-treated phRPE cells. CONCLUSION: This study demonstrates that luteolin exhibits the anti-fibrotic effect in a laser-induced mouse model by inhibiting EMT of RPE cells via deactivating Smad2/3 and YAP signaling, which provides a potential natural compound for the prevention and treatment of SF and fibrosis-related diseases.


Subject(s)
Epithelial-Mesenchymal Transition , Retinal Pigment Epithelium , Humans , Male , Animals , Mice , Retinal Pigment Epithelium/pathology , Luteolin/pharmacology , Mice, Inbred C57BL , Fibrosis , Collagen/metabolism , Collagen Type I/metabolism , Lasers
13.
Exp Cell Res ; 429(1): 113649, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37225012

ABSTRACT

BACKGROUND: Fibrotic scar is a severe side effect of trabeculectomy, resulting in unsatisfactory outcomes for glaucoma surgery. Accumulating evidence showed human Tenon's fibroblasts (HTFs) play an important role in fibrosis formation. We previously reported that the aqueous level of secreted protein acidic and rich in cysteine (SPARC) was higher in the patients with primary angle closure glaucoma, which was associated with the failure of trabeculectomy. In this study, the potential effect and mechanism of SPARC in promoting fibrosis were explored by using HTFs. METHODS: HTFs were employed in this study and examined under a phase-contrast microscope. Cell viability was determined by CCK-8. The expressions of SPARC-YAP/TAZ signaling and the fibrosis-related markers were examined with reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), Western blot, and immunofluorescence, subcellular fractionation was conducted to further determined the variation of YAP and phosphorylated YAP. The differential gene expressions were analyzed with RNA sequencing (RNAseq), followed by Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. RESULTS: Exogenous SPARC induced HTFs-myofibroblast transformation, as evidenced by the increased expression of α-SMA, collagen I and fibronectin in both protein and mRNA levels. SPARC knockdown decreased the expressions of the above genes in TGF-ß2-treated HTFs. KEGG analysis showed that the Hippo signaling pathway was mostly enriched. SPARC treatment increased the expressions of YAP, TAZ, CTGF and CYR61 as well as enhanced YAP translocation from cytoplasm to nucleus, and decreased the phosphorylation of YAP and LAST1/2, which was reversed by SPARC knockdown. Knockdown of YAP1 decreased the fibrosis-related markers, such as α-SMA, collagen I and Fibronectin, in SPARC-treated HTFs. CONCLUSIONS: SPARC induced HTFs-myofibroblast transformation via activating YAP/TAZ signaling. Targeting SPARC-YAP/TAZ axis in HTFs might provide a novel strategy for inhibiting fibrosis formation after trabeculectomy.


Subject(s)
Fibronectins , Myofibroblasts , Humans , Myofibroblasts/metabolism , Fibronectins/metabolism , Osteonectin/genetics , Osteonectin/metabolism , Fibroblasts/metabolism , Collagen Type I/metabolism , Fibrosis , Cells, Cultured
14.
J Fungi (Basel) ; 9(4)2023 Apr 13.
Article in English | MEDLINE | ID: mdl-37108923

ABSTRACT

Bioremediation is an effective way to remove heavy metals from pollutants. This study investigated the effects of Yarrowia lipolytica (Y. lipolytica) on the bioremediation of chromated copper arsenate (CCA)-treated wood wastes. Copper ions stressed the yeast strains to improve their bioremediation efficiency. A comparison of changes in morphology, chemical composition, and metal content of CCA wood before and after bioremediation was conducted. The amount of arsenic (As), chromium (Cr), and copper (Cu) was quantified by microwave plasma atomic emission spectrometer. The results showed that yeast strains remained on the surface of CCA-treated wood after bioremediation. The morphologies of the strains changed from net to spherical because of the Cu2+ stress. Fourier-transform infrared spectroscopy showed that carboxylic acid groups of wood were released after removing heavy metals. A large amount of oxalic acid was observed when the optical density (OD600nm) was 0.05 on the 21st day. Meanwhile, the highest removal rate of Cu, As, and Cr was 82.8%, 68.3%, and 43.1%, respectively. Furthermore, the Cu removal from CCA-treated wood increased by about 20% after Cu2+ stress. This study showed that it is feasible to remove heavy metals from CCA-treated wood by Y. lipolytica without destroying the wood structure, especially by copper-induced Y. lipolytica.

15.
Heliyon ; 9(3): e13897, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36915480

ABSTRACT

Deprivation of one sense can be followed by enhanced development of other senses via cross-modal plasticity mechanisms. To study the effect of whisker tactile deprivation on vision during the early stages of development, we clipped the bilateral whiskers of young mice and found that their vision was impaired but later recovered to normal levels. Our results demonstrate that inhibition of the PI3K/AKT/ERK signaling pathway caused short-term visual impairment during early development, while high expression levels of Crystallin Alpha A (CRYAA) and Gap Junction Protein Alpha 8 (GJA8) in the retina led to the recovery of developmental visual acuity. Interestingly, analysis of single-cell sequencing results from human embryonic retinas at 9-19 gestational weeks (GW) revealed that CRYAA and GJA8 display stage-specific peak expression during human embryonic retinal development, suggesting potential functions in visual development. Our data show that high expression levels of CRYAA and GJA8 in the retina after whisker deprivation rescue impaired visual development, which may provide a foundation for further research on the mechanisms of cross-modal plasticity and in particular, offer new insights into the mechanisms underlying tactile-visual cross-modal development.

16.
Pharmacol Res ; 187: 106559, 2023 01.
Article in English | MEDLINE | ID: mdl-36403720

ABSTRACT

Retinal Müller glial dysfunction and intracellular edema are important mechanisms leading to diabetic macular edema (DME). Aquaporin 11 (AQP11) is primarily expressed in Müller glia with unclear functions. This study aims to explore the role of AQP11 in the pathogenesis of intracellular edema of Müller glia in diabetic retinopathy (DR). Here, we found that AQP11 expression, primarily located at the endfeet of Müller glia, was down-regulated with diabetes progression, accompanied by intracellular edema, which was alleviated by intravitreal injection of lentivirus-mediated AQP11 overexpression. Similarly, intracellular edema of hypoxia-treated rat Müller cell line (rMC-1) was aggravated by AQP11 inhibition, while attenuated by AQP11 overexpression, accompanied by enhanced function in glutamate metabolism and reduced cell death. The down-regulation of AQP11 was also verified in the Müller glia from the epiretinal membranes (ERMs) of proliferative DR (PDR) patients. Mechanistically, down-regulation of AQP11 in DR was mediated by the HIF-1α-dependent and independent miRNA-AQP11 axis. Overall, we deciphered the AQP11 down-regulation, mediated by miRNA-AQP11 axis, resulted in Müller drainage dysfunction and subsequent intracellular edema in DR, which was partially reversed by AQP11 overexpression. Our findings propose a novel mechanism for the pathogenesis of DME, thus targeting AQP11 regulation provides a new therapeutic strategy for DME.


Subject(s)
Aquaporins , Diabetes Mellitus , Diabetic Retinopathy , Macular Edema , MicroRNAs , Rats , Animals , Diabetic Retinopathy/pathology , MicroRNAs/genetics , Down-Regulation , Aquaporins/metabolism
17.
Neural Regen Res ; 18(7): 1441-1449, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36571340

ABSTRACT

Epigenetics focuses on DNA methylation, histone modification, chromatin remodeling, noncoding RNAs, and other gene regulation mechanisms beyond the DNA sequence. In the past decade, epigenetic modifications have drawn more attention as they participate in the development and progression of diabetic retinopathy despite tight control of glucose levels. The underlying mechanisms of epigenetic modifications in diabetic retinopathy still urgently need to be elucidated. The diabetic condition facilitates epigenetic changes and influences target gene expression. In this review, we summarize the involvement of epigenetic modifications and metabolic memory in the development and progression of diabetic retinopathy and propose novel insights into the treatment of diabetic retinopathy.

18.
Neural Regen Res ; 18(5): 976-982, 2023 May.
Article in English | MEDLINE | ID: mdl-36254977

ABSTRACT

Diabetic retinopathy, characterized as a microangiopathy and neurodegenerative disease, is the leading cause of visual impairment in diabetic patients. Many clinical features observed in diabetic retinopathy, such as capillary occlusion, acellular capillaries and retinal non-perfusion, aggregate retinal ischemia and represent relatively late events in diabetic retinopathy. In fact, retinal microvascular injury is an early event in diabetic retinopathy involving multiple biochemical alterations, and is manifested by changes to the retinal neurovascular unit and its cellular components. Currently, intravitreal anti-vascular endothelial growth factor therapy is the first-line treatment for diabetic macular edema, and benefits the patient by decreasing the edema and improving visual acuity. However, a significant proportion of patients respond poorly to anti-vascular endothelial growth factor treatments, indicating that factors other than vascular endothelial growth factor are involved in the pathogenesis of diabetic macular edema. Accumulating evidence confirms that low-grade inflammation plays a critical role in the pathogenesis and development of diabetic retinopathy as multiple inflammatory factors, such as interleukin-1ß, monocyte chemotactic protein-1 and tumor necrosis factor -α, are increased in the vitreous and retina of diabetic retinopathy patients. These inflammatory factors, together with growth factors such as vascular endothelial growth factor, contribute to blood-retinal barrier breakdown, vascular damage and neuroinflammation, as well as pathological angiogenesis in diabetic retinopathy, complicated by diabetic macular edema and proliferative diabetic retinopathy. In addition, retinal cell types including microglia, Müller glia, astrocytes, retinal pigment epithelial cells, and others are activated, to secrete inflammatory mediators, aggravating cell apoptosis and subsequent vascular leakage. New therapies, targeting these inflammatory molecules or related signaling pathways, have the potential to inhibit retinal inflammation and prevent diabetic retinopathy progression. Here, we review the relevant literature to date, summarize the inflammatory mechanisms underlying the pathogenesis of diabetic retinopathy, and propose inflammation-based treatments for diabetic retinopathy and diabetic macular edema.

20.
Cells ; 11(21)2022 10 25.
Article in English | MEDLINE | ID: mdl-36359761

ABSTRACT

Diabetic retinopathy (DR), with increasing incidence, is the major cause of vision loss and blindness worldwide in working-age adults. Diabetic macular edema (DME) remains the main cause of vision impairment in diabetic patients, with its pathogenesis still not completely elucidated. Vascular endothelial growth factor (VEGF) plays a pivotal role in the pathogenesis of DR and DME. Currently, intravitreal injection of anti-VEGF agents remains as the first-line therapy in DME treatment due to the superior anatomic and functional outcomes. However, some patients do not respond satisfactorily to anti-VEGF injections. More than 30% patients still exist with persistent DME even after regular intravitreal injection for at least 4 injections within 24 weeks, suggesting other pathogenic factors, beyond VEGF, might contribute to the pathogenesis of DME. Recent advances showed nearly all the retinal cells are involved in DR and DME, including breakdown of blood-retinal barrier (BRB), drainage dysfunction of Müller glia and retinal pigment epithelium (RPE), involvement of inflammation, oxidative stress, and neurodegeneration, all complicating the pathogenesis of DME. The profound understanding of the changes in proteomics and metabolomics helps improve the elucidation of the pathogenesis of DR and DME and leads to the identification of novel targets, biomarkers and potential therapeutic strategies for DME treatment. The present review aimed to summarize the current understanding of DME, the involved molecular mechanisms, and the changes in proteomics and metabolomics, thus to propose the potential therapeutic recommendations for personalized treatment of DME.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Macular Edema , Humans , Macular Edema/drug therapy , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/therapeutic use , Intravitreal Injections , Diabetes Mellitus/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...