Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
Exp Eye Res ; 244: 109939, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38789021

RESUMO

Transforming growth factor-ß2 (TGF-ß2) induced fibrogenic changes in human trabecular meshwork (HTM) cells have been implicated in trabecular meshwork (TM) damage and intraocular pressure (IOP) elevation in primary open-angle glaucoma (POAG) patients. Silibinin (SIL) exhibited anti-fibrotic properties in various organs and tissues. This study aimed to assess the effects of SIL on the TGF-ß2-treated HTM cells and to elucidate the underlying mechanisms. Our study found that SIL effectively inhibited HTM cell proliferation, attenuated TGF-ß2-induced cell migration, and mitigated TGF-ß2-induced reorganization of both actin and vimentin filaments. Moreover, SIL suppressed the expressions of fibronectin (FN), collagen type I alpha 1 chain (COL1A1), and alpha-smooth muscle actin (α-SMA) in the TGF-ß2-treated HTM cells. RNA sequencing indicated that SIL interfered with the phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB, also known as AKT) signaling pathway, extracellular matrix (ECM)-receptor interaction, and focal adhesion in the TGF-ß2-treated HTM cells. Western blotting demonstrated SIL inhibited the activation of Janus kinase 2 (JAK2)/signal transducers and activators of transcription 3 (STAT3) and the downstream PI3K/AKT signaling pathways induced by TGF-ß2, potentially contributing to its inhibitory effects on ECM protein production in the TGF-ß2-treated HTM cells. Our study demonstrated the ability of SIL to inhibit TGF-ß2-induced fibrogenic changes in HTM cells. SIL could be a potential IOP-lowering agent by reducing the fibrotic changes in the TM tissue of POAG patients, which warrants further investigation through additional animal and clinical studies.


Assuntos
Movimento Celular , Proliferação de Células , Janus Quinase 2 , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Fator de Transcrição STAT3 , Transdução de Sinais , Silibina , Malha Trabecular , Fator de Crescimento Transformador beta2 , Humanos , Malha Trabecular/efeitos dos fármacos , Malha Trabecular/metabolismo , Malha Trabecular/patologia , Fator de Crescimento Transformador beta2/farmacologia , Fator de Crescimento Transformador beta2/metabolismo , Fator de Transcrição STAT3/metabolismo , Janus Quinase 2/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Cultivadas , Silibina/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proliferação de Células/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Western Blotting , Fibrose , Silimarina/farmacologia , Antioxidantes/farmacologia , Glaucoma de Ângulo Aberto/metabolismo , Glaucoma de Ângulo Aberto/tratamento farmacológico , Glaucoma de Ângulo Aberto/patologia
2.
Tissue Eng Regen Med ; 21(5): 695-710, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38642251

RESUMO

BACKGROUND: Various cell culture platforms that could display native environmental cue-mimicking stimuli were developed, and effects of environmental cues on cell behaviors were studied with the cell culture platforms. Likewise, various cell culture platforms mimicking native trabecular meshwork (TM) composed of juxtacanalicular, corneoscleral and uveal meshwork located in internal scleral sulcus were used to study effects of environmental cues and/or drug treatments on TM cells and glaucoma development. Glaucoma is a disease that could cause blindness, and cause of glaucoma is not clearly identified yet. It appears that aqueous humor (AH) outflow resistance increased by damages on pathway of AH outflow can elevate intraocular pressure (IOP). These overall possibly contribute to development of glaucoma. METHODS: For the study of glaucoma, static and dynamic cell culture platforms were developed. Particularly, the dynamic platforms exploiting AH outflow-mimicking perfusion or increased IOP-mimicking increased pressure were used to study how perfusion or increased pressure could affect TM cells. Overall, potential mechanisms of glaucoma development, TM structures and compositions, TM cell culture platform types and researches on TM cells and glaucoma development with the platforms were described in this review. RESULTS AND CONCLUSION: This will be useful to improve researches on TM cells and develop enhanced therapies targeting glaucoma.


Assuntos
Técnicas de Cultura de Células , Glaucoma , Malha Trabecular , Malha Trabecular/citologia , Humanos , Técnicas de Cultura de Células/métodos , Pressão Intraocular , Humor Aquoso , Animais
3.
Front Biosci (Landmark Ed) ; 29(3): 91, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38538254

RESUMO

OBJECTIVE: The morphology and functions of the human trabecular meshwork (HTM) are dysregulated in glaucoma, and the molecular mechanisms of this dysregulation remain unknown. According to an established in vitro model, whose function was to study the regulatory networks sustaining the response of HTM cells to the increased substrate stiffness, we systematically analyzed the expression pattern of long noncoding RNAs (lncRNAs), the important regulatory RNAs in cells. METHODS: Bioinformatics analysis was performed to identify the dysregulated lncRNAs in response to increased substrate stiffness using transcriptome sequencing data (RNA-seq). Then we interfered with the expression of several dysregulated lncRNAs in HTM cells to explore their molecular targets. The cross-linking immunoprecipitation and sequencing method (CLIP-seq) was used to identify enhancer of zeste homolog 2 (EZH2)-targeted RNAs in HTM cells. The chromatin IP and sequencing method (ChIP-seq) was used to identify the targets of EZH2 and histone H3 at lysine 27 (H3K27me3). RESULTS: The response of thousands of dysregulated lncRNAs to increased substrate stiffness was identified through RNA-seq. Functional prediction of these lncRNAs revealed that they potentially regulated key biological processes, including extracellular matrix (ECM) organization. By interfering with the expression of lncRNA SHNG8, ZFHX4-AS1, and RP11-552M11.4, the results demonstrated that those lncRNAs extensively regulated the expression levels of ECM-associated genes. Moreover, we found that EZH2 expression was significantly decreased at high substrate stiffness. Using CLIP-seq to identify EZH2-targeted RNAs in HTM cells, we found that SNHG8 was bound by EZH2. According to the CLIP-seq data of EZH2, we found that EZH2 binding sites were observed in the transcripts of SNHG8-regulated genes, but not in the ChIP-seq results of EZH2 and H3K27me3. CONCLUSION: Our results suggest that SNHG8 and EZH2 may cooperate to regulate the expression of a subset of genes by influencing their RNA abundance, explaining how they support HTM cell morphology and high density. This study contributes to the understanding of the alteration of HTM during the progression of glaucoma by identifying functional lncRNAs, especially SNHG8, and suggests novel therapeutic targets to treat glaucoma.


Assuntos
Glaucoma , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Histonas/metabolismo , Transcriptoma , Malha Trabecular/metabolismo , Cromatina/metabolismo , Biologia Computacional/métodos , Glaucoma/genética , Glaucoma/metabolismo
4.
Hum Cell ; 37(2): 394-407, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38316716

RESUMO

Glaucoma is one of the leading causes of irreversible blindness in developed countries, and intraocular pressure (IOP) is primary and only treatable risk factor, suggesting that to a significant extent, glaucoma is a disease of IOP disorder and pathological mechanotransduction. IOP-lowering ways are limited to decreaseing aqueous humour (AH) production or increasing the uveoscleral outflow pathway. Still, therapeutic approaches have been lacking to control IOP by enhancing the trabecular meshwork (TM) pathway. Trabecular meshwork cells (TMCs) have endothelial and myofibroblast properties and are responsible for the renewal of the extracellular matrix (ECM). Mechanosensitive cation channels, including Piezo1 and TRPV4, are abundantly expressed in primary TMCs and trigger mechanostress-dependent ECM and cytoskeletal remodelling. However, prolonged mechanical stimulation severely affects cellular biosynthesis through TMC mechanotransduction, including signaling, gene expression, ECM remodelling, and cytoskeletal structural changes, involving outflow facilities and elevating IOP. As for the functional coupling relationship between Piezo1 and TRPV4 channels, inspired by VECs and osteoblasts, we hypothesized that Piezo1 may also act upstream of TRPV4 in glaucomatous TM tissue, mediating the activation of TRPV4 via Ca2+ inflow or Ca2+ binding to phospholipase A2(PLA2), and thus be involved in increasing TM outflow resistance and elevated IOP. Therefore, this review aims to help identify new potential targets for IOP stabilization in ocular hypertension and primary open-angle glaucoma by understanding the mechanical transduction mechanisms associated with the development of glaucoma and may provide ideas into novel treatments for preventing the progression of glaucoma by targeting mechanotransduction.


Assuntos
Glaucoma de Ângulo Aberto , Glaucoma , Humanos , Mecanotransdução Celular , Malha Trabecular , Canais de Cátion TRPV/genética , Cátions
5.
Exp Eye Res ; 240: 109806, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38272381

RESUMO

Primary open-angle glaucoma (POAG) is the most common type of glaucoma leading to blindness. The search for ways to prevent/treat this entity is one of the main challenges of today's ophthalmology. One of such solution seems to be biologically active substances of natural origin, such as genistein (GEN), which can affect the function of isolated trabecular meshwork by the inhibition of protein tyrosine kinase. However, the role of GEN in viability as well as myofibroblastic transformation in human trabecular meshwork cells stimulated by TGF-ß is unknown. Using human trabecular meshwork cells (HTMCs) we investigated the effect of genistein on cell viability and myofibroblastic transformation stimulated by TGF-ß1 and TGF-ß2. Using Real-Time PCR, western blot and immunofluorescence we determined the effect on the expression changes of αSMA, TIMP1, collagen 1 and 3 at mRNA and protein level. We found that genistein increases the viability of HTMCs (1, 2, 3 µg/ml; P < 0.05 and 4, 5, 10, 15, 20 µg/ml; P < 0.01). Moreover, we found that addition of 10, 15 and 20 µg/ml is able to prevent myofibroblastic transformation of HTMCs by decreasing αSMA, TIMP1, collagen 1 and 3 mRNA and protein expression (P < 0.01). Based on the obtained results, we can conclude that genistein is a potential factor that can prevent the myofibroblastic transformation of HTMCs accompanying glaucoma. Describing GEN influence on myofibroblastic transformation processes in HTMC allows us to conclude that it can be considered a potential therapeutic agent or a substance supporting treatment in patients with glaucoma.


Assuntos
Glaucoma de Ângulo Aberto , Glaucoma , Humanos , Genisteína/farmacologia , Glaucoma de Ângulo Aberto/tratamento farmacológico , Glaucoma de Ângulo Aberto/prevenção & controle , Glaucoma de Ângulo Aberto/genética , Malha Trabecular/metabolismo , Células Cultivadas , Fator de Crescimento Transformador beta2/farmacologia , Fator de Crescimento Transformador beta2/metabolismo , Glaucoma/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Colágeno/metabolismo
6.
Biochim Biophys Acta Gen Subj ; 1868(2): 130538, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38072209

RESUMO

BACKGROUND: Hyperglycemia, which can lead to apoptosis, hypertrophy, fibrosis, and induces hyperinflammation in diabetic vascular complications due to oxidative stress. In order to elucidate the potential dual roles and regulatory signal transduction of TGF-ß1 and TGF-ß2 in human trabecular meshwork cells (HTMCs), we established an oxidative cell model in HTMCs using 5.5, 25, 50, and 100 mM d-glucose-supplemented media and characterized the TGF-ß-related oxidative stress pathway. METHODS: Further analysis was conducted to investigate oxidative damage and protein alterations in the HTMC caused by the signal transduction. This was done through a series of qualitative cell function studies, such as cell viability/apoptosis analysis, intracellular reactive oxygen species (ROS) detection, analysis of calcium release concentration, immunoblot analysis to detect the related protein expression alteration, and analysis of cell fibrosis to study the effect of different severities of hyperglycemia. Also, we illustrated the role of TGF-ß1/2 in oxidative stress-induced injury by shRNA-mediated knockdown or stimulation with recombinant human TGF-ß1 protein (rhTGF-ß1). RESULTS: Results from the protein expression analysis showed that p-JNK, p-p38, p-AKT, and related SMAD family members were upregulated in HTMCs under hyperglycemia. In the cell functional assays, HTMCs treated with rhTGFß-1 (1 ng/mL) under hyperglycemic conditions showed higher proliferation rates and lower ROS and calcium levels. CONCLUSIONS: To summarize, mechanistic analyses in HTMCs showed that hyperglycemia-induced oxidative stress activated TGF-ß1 along with its associated pathway. GENERAL SIGNIFICANCE: While at low concentrations, TGF-ß1 protects cells from antioxidation, whereas at high concentrations, it accumulates in the extracellular matrix, causing further HTMC dysfunction.


Assuntos
Hiperglicemia , Fator de Crescimento Transformador beta1 , Humanos , Fator de Crescimento Transformador beta1/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Malha Trabecular/metabolismo , Cálcio/metabolismo , Hiperglicemia/metabolismo , Fibrose
7.
J Fr Ophtalmol ; 47(1): 103746, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37806937

RESUMO

Glaucoma is a group of neurodegenerative diseases characterized by loss of retinal ganglion cells and visual field defects and is one of the major causes of irreversible blindness worldwide. Primary open-angle glaucoma (POAG) is one of the classifications of glaucoma. Oxidative stress in trabecular reticulated cells is one of the possible mechanisms of the development of glaucoma. At present, there is still a lack of effective methods to treat glaucoma. Ghrelin is characterized by its wide distribution and high potency and has anti-inflammatory, antioxidant, and anti-apoptotic effects, which may be beneficial in the treatment of glaucoma. In this study, we investigated whether ghrelin can protect human trabecular meshwork cells (HTMCs) from oxidative damage induced by hydrogen peroxide (H2O2), as well as the possible mechanism of action. CCK8 and flow cytometry results revealed that treatment of HTMCs with ghrelin showed a dose-dependent protective effect against H2O2-induced damage. Ghrelin significantly decreased the rate of apoptosis and levels of reactive oxygen species (ROS) and malondialdehyde (MDA) and increased the level of superoxide dismutase (SOD) and catalase (CAT) in HTMCs. The difference was statistically significant compared with the H2O2 group. Ghrelin activated Nrf2/HO-1/NQO-1 signaling pathways and decreased HIF-1α level in H2O2-injured HTMCs as shown on qPCR and Western blot. In conclusion, ghrelin can protect HTMCs from oxidative damage induced by H2O2 and reduce apoptosis in HTMCs, which can be a new approach to treating POAG. The underlying therapeutic mechanism may be related to Nrf2/HO-1/NQO-1 signaling pathways and HIF-1α.


Assuntos
Glaucoma de Ângulo Aberto , Glaucoma , Humanos , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Malha Trabecular , Glaucoma de Ângulo Aberto/tratamento farmacológico , Peróxido de Hidrogênio/farmacologia , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/uso terapêutico , Grelina/farmacologia , Grelina/metabolismo , Grelina/uso terapêutico , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/farmacologia , Fator 2 Relacionado a NF-E2/uso terapêutico
8.
J Cell Physiol ; 238(10): 2451-2467, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37584382

RESUMO

The trabecular meshwork (TM) cells of the eye are important for controlling intraocular pressure (IOP) and regulating outflow resistance in the aqueous humor. TM cells can remove particles and cellular debris by phagocytosis, decreasing both outflow resistance and IOP. However, the underlying mechanisms remain unclear. Here, we investigate whether apoptosis inhibitor of macrophages (AIM), which mediates the removal of dead cells and debris in renal tubular epithelial cells, regulates the phagocytic capacity of TM cells. In vitro experiments revealed that CD36, the main receptor for AIM, colocalized with AIM in human TM cells; additionally, phagocytosis was stimulated when AIM was provided. Furthermore, in a mouse model with transient IOP elevation induced by laser iridotomy (LI), removal of accumulated iris pigment epithelial cells or debris in the TM and recovery of IOP to baseline levels were delayed in AIM-/- mice, compared with control mice. However, treatment with AIM eyedrops rescued AIM-/- mice from the elevated IOP after LI. Since AIM is a protein known to inhibit macrophage apoptosis, we additionally verified its involvement in macrophage removal of cellular debris and IOP. There were no statistically significant differences in the number of macrophages between control mice and AIM-/- mice in the TM. Additionally, we confirmed the rescue effect of the rAIM eyedrops after macrophages had been removed by clodronate liposomes. Therefore, AIM plays an important role in regulating the phagocytic capacity of TM cells, thereby affecting outflow resistance. Our results suggest that drugs targeting the phagocytic capacity of TM cells via the AIM-CD36 pathway may be used to treat glaucoma.

9.
Curr Eye Res ; 48(11): 1014-1025, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37466387

RESUMO

PURPOSE: We investigated whether hydrogen peroxide (H2O2)-induced oxidative stress causes human trabecular meshwork (HTM) cell dysfunction observed in open angle glaucoma (OAG) in vitro, and the effects of topical glaucoma medications on oxidative stress in HTM cells. METHODS: We used commercially available ophthalmic solutions of brimonidine, omidenepag isopropyl, and ripasudil in the study. HTM cells were exposed to H2O2 for 1 h, with or without glaucoma medications. We assessed cell viability and senescence via WST-1 and senescence-associated-ß-galactosidase (SA-ß-Gal) activity assays. After exposure to H2O2 and glaucoma medications, we evaluated changes in markers of fibrosis and stress by using real-time quantitative polymerase chain reaction (qPCR) to measure the mRNA levels of collagen type I alpha 1 chain (COL1A1), fibronectin, alpha-smooth muscle actin (α-SMA), matrix metalloproteinase-2 (MMP-2), endoplasmic reticulum stress markers of C/EBP homologous protein (CHOP), 78-kDa glucose-regulated protein (GRP78), and splicing X-box binding protein-1 (sXBP-1). RESULTS: HTM cell viability decreased and SA-ß-Gal activity increased significantly after exposure to H2O2. Treatment with three ophthalmic solutions attenuated these changes. Real-time qPCR revealed that H2O2 upregulated the mRNA levels of COL1A1, fibronectin, α-SMA, CHOP, GRP78, and sXBP-1, whereas it downregulated MMP-2 mRNA expression significantly. Brimonidine suppressed the upregulation of stress markers CHOP and GRP78. Additionally, omidenepag isopropyl and ripasudil decreased the upregulation of COL1A1 and sXBP-1. Furthermore, ripasudil significantly suppressed fibrotic markers fibronectin and α-SMA, compared with the other two medications. CONCLUSION: In vitro, H2O2 treatment of HTM cells induced characteristic changes of OAG, such as fibrosis changes and the upregulation of stress markers. These glaucomatous changes were attenuated by additional treatments with brimonidine, omidenepag isopropyl, and ripasudil ophthalmic solutions.

10.
J Ocul Pharmacol Ther ; 39(7): 430-438, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37307020

RESUMO

Purpose: To investigate the effects of Sonic hedgehog (Shh) signaling on primary human trabecular meshwork (HTM) cells. Methods: Primary HTM cells were isolated from healthy donors and cultured. Recombinant Shh (rShh) protein and cyclopamine were used to activate and inhibit the Shh signaling pathway, respectively. A cell viability assay was performed to assess the effects of rShh on the activity of primary HTM cells. Functional assessment of cell adhesion and phagocytosis was also performed. The proportion of apoptotic cells was examined using flow cytometry. Fibronectin (FN) and transforming growth factor beta2 (TGF-ß2) protein were detected to assess the influence of rShh on the metabolism of the extracellular matrix (ECM). Real-time polymerase chain reaction (RT-PCR) and western blot analyses were used to examine mRNA and protein expression of Shh signaling pathway-associated factors GLI Family Zinc Finger 1 (GLI1) and Suppressor of Fused (SUFU). Results: rShh significantly enhanced primary HTM cell viability at a concentration of 0.5 µg/mL. rShh increased the adhesion and phagocytic abilities of primary HTM cells, and decreased cell apoptosis. FN and TGF-ß2 protein expression increased in primary HTM cells treated with rShh. rShh upregulated the transcriptional activity and protein levels of GLI1, and downregulated those of SUFU. Correspondingly, the rShh-induced GLI1 upexpression was partially blocked by pretreatment with the Shh pathway inhibitor cyclopamine at a concentration of 10 µM. Conclusions: Activation of Shh signaling can regulate the function of primary HTM cells through GLI1. Regulation of Shh signaling may be a potential target for attenuating cell damage in glaucoma.


Assuntos
Proteínas Hedgehog , Fator de Crescimento Transformador beta2 , Humanos , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Transdução de Sinais , Malha Trabecular/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Proteína GLI1 em Dedos de Zinco/genética , Proteína GLI1 em Dedos de Zinco/metabolismo , Proteína GLI1 em Dedos de Zinco/farmacologia
11.
Biomolecules ; 12(9)2022 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-36139071

RESUMO

This study examines the potential role of transforming growth factor-beta 3 (TGF-ß3) on the fibrotic response of cultured human trabecular meshwork (HTM) cells. The relationships and trans-signaling interactions between TGF-ß3 and autotaxin (ATX) in HTM cells were also examined. The levels of TGF-ß and ATX in the aqueous humor (AH) of patients were measured by an immunoenzymetric assay. The TGF-ß3-induced expression of the fibrogenic markers, fibronectin, collagen type I alpha 1 chain, and alpha-smooth muscle actin, and ATX were examined by quantitative real-time PCR, Western blotting, and immunocytochemistry, and the trans-signaling regulatory effect of TGF-ß3 on ATX expression was also evaluated. In HTM cells, the significant upregulation of ATX was induced by TGF-ß3 at a concentration of 0.1 ng/mL, corresponding to the physiological concentration in the AH of patients with exfoliative glaucoma (XFG). However, higher concentrations of TGF-ß3 significantly suppressed ATX expression. TGF-ß3 regulated ATX transcription and signaling in HTM cells, inducing the upregulation of fibrogenic proteins in a dose-dependent manner. Trans-signaling of TGF-ß3 regulated ATX transcription, protein expression, and signaling, and was thereby suggested to induce fibrosis of the trabecular meshwork. Modulation of trans-signaling between TGF-ß3 and ATX may be key to elucidate the pathology of XFG, and for the development of novel treatment modalities.


Assuntos
Humor Aquoso , Diester Fosfórico Hidrolases , Malha Trabecular , Fator de Crescimento Transformador beta3 , Actinas/metabolismo , Humor Aquoso/metabolismo , Células Cultivadas , Colágeno Tipo I/metabolismo , Fibronectinas/metabolismo , Fibrose , Humanos , Diester Fosfórico Hidrolases/metabolismo , Malha Trabecular/metabolismo , Malha Trabecular/patologia , Fator de Crescimento Transformador beta3/metabolismo
12.
Int J Ophthalmol ; 15(8): 1240-1248, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36017046

RESUMO

AIM: To investigate potential gene changes in trabecular meshwork (TM) induced by dexamethasone (DEX) in steroid-induced glaucoma (SIG). METHODS: The expression data of 24 cases from a public functional genomics data were sorted to identify the mechanisms of action of DEX on the TM. The relationships of the differentially expressed genes (DEGs) were enriched using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. In addition, the hub genes were screened by the Search Tool for the Retrieval of Interacting Genes Database (STRING) and Cytoscape tools. Finally, human TM cells (HTMCs) were treated with DEX to preliminarily explore the function of hub genes. RESULTS: Totally 47 DEGs, including 21 downregulated and 26 upregulated genes were identified. The primary enriched results of the DEGs consisted of inflammatory response, extracellular matrix (ECM), negative regulation of cell proliferation, TNF signalling pathway and the regulation of tryptophan channels by inflammatory mediators. Subsequently, pro-melanin-enriched hormone (PMCH) and Bradykinin B1 receptor (BDKRB1) were screened as hub genes. It is verified in GSE37474 data set. Western blot and quantitative real-time polymerase chain reaction (qPCR) results showed that protein and RNA expression levels of BDKRB1 were significantly decreased after DEX treatment, while PMCH was not significantly changed. CONCLUSION: BDKRB1 may be a key gene involved in SIG onset, providing a suitable therapeutic target for improving the prognosis of SIG patients.

13.
Genes (Basel) ; 13(5)2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35627267

RESUMO

Aim: To investigate genes and pathways involved in differential glucocorticoid (GC) responsiveness in human trabecular meshwork (HTM) cells using RNA sequencing. Methods: Using paired human donor eyes, human organ-cultured anterior segment (HOCAS) was established in one eye to characterize GC responsiveness based on intra ocular pressure (IOP) change and, in the other eye, primary HTM cell culture was established. For RNA sequencing, total RNA extracted from GC-responder (GC-R) and non-responder (GC-NR) cells after dexamethasone (DEX) or ethanol (ETH) treatment for 7d was used. Differentially expressed genes (DEGs) were compared among five groups and validated. Results: In total, 616 and 216 genes were identified as significantly dysregulated in Group #1 and #2 (#1: ETH vs. DEX-treated GC-R; #2: ETH vs. DEX-treated GC-NR), respectively. Around 80 genes were commonly dysregulated in Group #3 (overlapping DEGs between #1 and #2), whereas 536 and 136 genes were uniquely expressed in GC-R (#4) and GC-NR HTM (#5) cells, respectively. Pathway analysis revealed that WNT signaling, drug metabolism cytochrome p450, cell adhesion, TGF-ß signaling, and MAPK signaling were associated with GC responsiveness. Conclusion: This is the first study reporting distinct gene signatures and their associated pathways for GC-R and GC-NR HTM cells. WNT and MAPK signaling are potential therapeutic targets for the management of GC-induced glaucoma.


Assuntos
Glucocorticoides , Malha Trabecular , Perfilação da Expressão Gênica , Glucocorticoides/metabolismo , Glucocorticoides/farmacologia , Humanos , Pressão Intraocular , Malha Trabecular/metabolismo , Transcriptoma/genética
14.
Clin Proteomics ; 19(1): 12, 2022 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-35562675

RESUMO

BACKGROUND: Treatment for glaucoma has traditionally been limited to reducing intraocular pressure (IOP). Inhibiting oxidative stress in the trabecular meshwork (TM) is regarded as a new treatment for glaucoma; however, the effects do not meet expectations. Exploring the mechanism by which oxidative stress and antioxidant stress occur in TM cells will offer clues to aid the development of new treatments. METHODS AND RESULTS: In our study, we cultured TM cells and used H2O2 and SOD to induce and inhibit oxidative stress, respectively. Label-free LC-MS/MS quantitative proteomic analysis was conducted to analyze the differentially expressed proteins and relevant signaling pathways. A total of 24 upregulated proteins and 18 downregulated proteins were identified under oxidative stress. PTGS2, TGFßr2 and ICAM-1 are the key proteins. The PTGS2/NF-ĸb pathway, TGF-ß/Smad signaling pathway and AGE-RAGE signaling pathway in diabetic complications may be the major signaling pathways under conditions of ROS-induced damage in TM cells. Seventy-eight proteins were upregulated and 73 proteins were downregulated under antioxidant stress in TM cells. The key protein was ICAM-1, which participates in the African trypanosomiasis pathway, one of the most important pathways under antioxidant stress. Combining the results of the Venn diagram with protein-protein interactions (PPIs), ICAM-1 was identified as the major protein. Cell Counting Kit-8 (CCK-8) and western blotting (WB) were used to reveal that suppressing the expression of ICAM-1 would improve the survival of TM cells. CONCLUSIONS: Key proteins and signaling pathways play important roles in the mechanisms of oxidative stress and antioxidant strategies in TM cells. ICAM-1 knockdown can suppress the apoptosis of TM cells induced by H2O2, which may reveal new therapeutic targets and biomarkers for glaucoma.

15.
Int J Mol Med ; 49(6)2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35417030

RESUMO

To explore the role of atorvastatin in regulating intraocular pressure (IOP) in glaucoma in vivo, and to investigate its related molecular pathway in vitro, an ocular hypertension model was generated by intravitreal injection of an adenoviral vector encoding transforming growth factor (TGF)­ß2 in the right eye of BALB/cJ mice, while the left was treated with an empty control adenovirus. To determine its anti­intraocular hypertension role, these induced hyper­IOP mice were gavaged with atorvastatin (20 mg/kg/day). Furthermore, extracellular matrix (ECM) factors were examined in the primary human trabecular meshwork (HTM) cells followed atorvastatin (0~200 µM) treatment in vitro. Whole genome microarray was employed to identify potential therapeutic target molecules associated with ECM regulation. Unilateral murine ocular hypertension was induced, via intravitreal injection of the adenoviral vector carrying the human TGF­ß2 gene (Ad.hTGF­ß2226/228), raising IOP from 12±1.6 to 32.3±0.7 mmHg (n=6, P<0.05) at day 15, which plateaued from day 15 to 30. Atorvastatin administration from day 15 to 30 decreased IOP from 32.3±0.7 to 15.4±1.1 mmHg (n=6, P<0.05) at day 30. Additionally, atorvastatin administration changed the morphology of cultured HTM cells from an elongated and adherent morphology into rounded, less elongated and less adherent cells, accompanied with suppressed expression of ECM. Gene Ontology and Genome analysis revealed that FGD4 (FYVE, RhoGEF and PH domain containing 4) might be a key factor contributing to these changes. Our data demonstrated that atorvastatin reduced TGF­ß2­induced ocular hypertension in vivo, perhaps via modifying cellular structure and decreasing ECM, using the FGD4 signaling pathway, as demonstrated in HTM cells. Our findings provide some useful information for the management of glaucoma, with statin therapy revealing a potential novel therapeutic pathway for glaucoma treatment.


Assuntos
Atorvastatina , Glaucoma , Proteínas dos Microfilamentos , Hipertensão Ocular , Animais , Atorvastatina/farmacologia , Células Cultivadas , Matriz Extracelular/metabolismo , Glaucoma/metabolismo , Pressão Intraocular , Camundongos , Proteínas dos Microfilamentos/genética , Hipertensão Ocular/induzido quimicamente , Hipertensão Ocular/tratamento farmacológico , Hipertensão Ocular/metabolismo , Malha Trabecular/metabolismo , Fator de Crescimento Transformador beta2/farmacologia
16.
Front Mol Biosci ; 8: 728932, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34552960

RESUMO

High intraocular pressure (IOP) is a major risk factor for glaucoma, a leading cause of irreversible blindness. Abnormal fibrotic activity in the human trabecular meshwork (HTM) cells is considered to be partly responsible for the increased resistance of aqueous humor outflow and IOP. This study aimed to identify the fibrotic pathways using integrated bioinformatics and further elucidate their mechanism of regulating fibrotic activity in dexamethasone (DEX)-treated HTM cells. Microarray datasets from the GEO database were obtained and analyzed by GEO2R. Bioinformatics analyses, including GO and KEGG analyses, were performed to explore biological functions and signaling pathways of differentially expressed genes (DEGs). The fibrotic pathways and targets were determined by western blot, RT-qPCR, or immunofluorescence staining. The cellular elastic modulus was measured using an atomic force microscope. A total of 204 DEGs, partly enriched in fibrotic activity (collagen-containing ECM, fibroblast activation) and Rap1, Ras, TGF-ß, and Hippo pathways, were identified. Experimental results showed that DEX induced fibrotic activity and regulated the expression of RhoA/ROCK in HTM cells. Similarly, the constitutively active RhoA (RhoAG14V) also promoted the fibrotic activity of HTM cells. Mechanistically, RhoAG14V induced the expression and nuclear translocation of YAP/TAZ to produce CTGF. Moreover, inhibition of ROCK or YAP decreased the expression of Collagen I and α-SMA proteins induced by DEX or RhoAG14V in HTM cells. In conclusion, these results indicate that RhoA/ROCK-YAP/TAZ axis plays a crucial role in regulating the fibrotic activity of DEX-treated HTM cells.

17.
Environ Toxicol ; 36(6): 1070-1079, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33522089

RESUMO

Glaucoma is the main reason for irreversible blindness, and pathological increased intraocular pressure is the leading risk factor for glaucoma. It is reported that trabecular meshwork cell injury is closely associated with the elevated intraocular pressure. The current study aimed to investigate the role of small nucleolar RNA host gene 3 (SNHG3) in human trabecular meshwork (HTM) cells under oxidative stress. A series of experiments including real-time quantitative polymerase chain reaction, subcellular fractionation assay, western blot analysis, cell counting kit-8 assay, RNA pull down, flow cytometry analysis, and RNA immunoprecipitation assay were used to explore the biological function and regulatory mechanism of SNHG3 in HTM cells under oxidative stress. First, we observed that H2 O2 induced SNHG3 upregulation in HTM cells. Then, we found that SNHG3 silencing alleviated H2 O2 -induced oxidative damage in HTM cells. Moreover, snail family transcriptional repressor 2 (SNAI2) knockdown alleviated the oxidative damage induced by H2 O2 in HTM cells. Mechanistically, SNHG3 bound with ELAV like RNA binding protein 2 (ELAVL2) to stabilize SNAI2. Finally, SNAI2 overexpression counteracted the effect of SNHG3 silencing on H2 O2 -treated HTM cells. In conclusion, our results demonstrated that SNHG3 cooperated with ELAVL2 to modulate cell apoptosis and extracellular matrix accumulation by stabilizing SNAI2 in HTM cells under oxidative stress.


Assuntos
Estresse Oxidativo , Malha Trabecular , Apoptose , Células Cultivadas , Matriz Extracelular/metabolismo , Humanos , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Regulação para Cima
18.
Antioxidants (Basel) ; 10(1)2021 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-33451157

RESUMO

Oxidative stress generated by reactive oxygen species (ROS) plays a critical role in the pathomechanism of glaucoma, which is a multifactorial blinding disease that may cause irreversible damage within human trabecular meshwork cells (HTMCs). It is known that the transforming growth factor-ß (TGF-ß) signaling pathway is an important component of oxidative stress-induced damage related to extracellular matrix (ECM) fibrosis and activates cell antioxidative mechanisms. To elucidate the dual potential roles and regulatory mechanisms of TGF-ß in effects on HTMCs, we established an in vitro oxidative model using hydrogen peroxide (H2O2) and further focused on TGF-ß-related oxidative stress pathways and the related signal transduction. Via a series of cell functional qualitative analyses to detect related protein level alterations and cell fibrosis status, we illustrated the role of TGF-ß1 and TGF-ß2 in oxidative stress-induced injury by shTGF-ß1 and shTGF-ß2 knockdown or added recombinant human TGF-ß1 protein (rhTGF-ß1). The results of protein level showed that p38 MAPK, TGF-ß, and its related SMAD family were activated after H2O2 stimulation. Cell functional assays showed that HTMCs with H2O2 exposure duration had a more irregular actin architecture compared to normal TM cells. Data with rhTGF-ß1 (1 ng/mL) pretreatment reduced the cell apoptosis rate and amount of reactive oxygen species (ROS), while it also enhanced survival. Furthermore, TGF-ß1 and TGF-ß2 in terms of antioxidant signaling were related to the activation of collagen I and laminin, which are fibrosis-response proteins. Succinctly, our study demonstrated that low concentrations of TGF-ß1 (1 ng/mL) preserves HTMCs from free radical-mediated injury by p-p38 MAPK level and p-AKT signaling balance, presenting a signaling transduction mechanism of TGF-ß1 in HTMC oxidative stress-related therapies.

19.
Curr Eye Res ; 46(4): 496-503, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32847411

RESUMO

PURPOSE: RhoA signaling is important for the regulation of intraocular pressure through the trabecular meshwork (TM). However, the relationship between RhoA signaling and phagocytosis in TM cells is unclear. The purpose of this study was to investigate the effects of RhoA signaling on the phagocytosis of TM cells. MATERIALS AND METHODS: TM cells were isolated from enucleated porcine eyes and treated with lysophosphatidic acid (LPA) or calpeptin to activate RhoA to determine phagocytic activity. To assess phagocytic activity, TM cells were incubated with pHrodo® Red S. aureus bioparticles, and the fluorescence intensity was measured using a cell sorter. The phagocytic activity of RhoA knockdown TM cells was also assessed using small interfering RNA (siRNA). To resolve the effects of dexamethasone on phagocytosis, TM cells were treated with dexamethasone for 72 h. The immunocytochemistry of vinculin and F-actin were evaluated in LPA- and dexamethasone-treated TM cells. RESULTS: RhoA activities after treatment with 10 µM LPA and 100 µM calpeptin were 1.38 ± 0.026-fold and 1.47 ± 0.070-fold higher, respectively, compared with the control. The phagocytic activity was reduced by LPA (0.67 ± 0.099) and calpeptin (0.57 ± 0.016), compared with the control. C3 transferase (Rho inhibitor) and Y-27632 (Rho-associated kinase inhibitor) prevented the effects of LPA on phagocytosis, and C3 partially inhibited the effects of calpeptin on phagocytosis. Knockdown of RhoA prevented the effect of LPA on phagocytosis. By immunostaining, LPA-induced stress fiber and focal adhesion formation was prevented by C3 and Y-27632 treatment. Moreover, RhoA knockdown prevented the effects of LPA on F-actin and focal adhesion. Dexamethasone treatment decreased phagocytic activity and increased stress fiber and focal adhesion. Y-27632 prevented the effects of dexamethasone on phagocytosis, and on stress fiber and focal adhesion fomation. CONCLUSIONS: These results suggest that the RhoA signal pathway regulates the phagocytic activity of TM cells.Abbreviations: TM: trabecular meshwork; LPA: lysophosphatidic acid; C3: C3 transferase; ROCK: Rho-associated kinase; siRNA: small interfering RNA.


Assuntos
Fagocitose/fisiologia , Transdução de Sinais/fisiologia , Malha Trabecular/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/metabolismo , Actinas/metabolismo , Animais , Western Blotting , Sobrevivência Celular , Células Cultivadas , Dexametasona/farmacologia , Dipeptídeos/farmacologia , Glucocorticoides/farmacologia , Imuno-Histoquímica , Lisofosfolipídeos/farmacologia , Staphylococcus aureus , Suínos , Malha Trabecular/metabolismo , Vinculina/metabolismo
20.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-908610

RESUMO

Glaucoma, a neurodegenerative disease characterized by progressive death of retinal ganglion cells (RGCs) and chronic axonal degeneration, is often associated with elevated intraocular pressure.Autophagy, which means self-eating, is a mechanism of cell degradation and recycling.Excessive autophagy or impaired autophagy may lead to cell dysfunction and even cell death.Recent studies have shown that autophagy is closely related to trabecular meshwork cell dysfunction, RGCs apoptosis and optic nerve degeneration caused by different factors, including oxidative stress, mechanical stimulation, high intraocular pressure, axon injury, genetic factors and so on.Regulating autophagy to protect RGCs may provide new ideas for glaucoma treatments.In this article, the definition and classification of autophagy, the regulation of autophagy, the role of autophagy in the process of oxidative stress and mechanical stimulation on the function of trabecular meshwork cells, and the impact of autophagy on RGCs under high intraocular pressure and RGCs axonal injury, the relationship between autophagy and apoptosis in RGCs, as well as the latest research results on autophagy and hereditary optic nerve degeneration, regulation of autophagy via gene and drug for the treatment of glaucoma were reviewed.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...