Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Genom ; 4(3): 100501, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38335956

RESUMEN

The precise roles of chromatin organization at osteoporosis risk loci remain largely elusive. Here, we combined chromatin interaction conformation (Hi-C) profiling and self-transcribing active regulatory region sequencing (STARR-seq) to qualify enhancer activities of prioritized osteoporosis-associated single-nucleotide polymorphisms (SNPs). We identified 319 SNPs with biased allelic enhancer activity effect (baaSNPs) that linked to hundreds of candidate target genes through chromatin interactions across 146 loci. Functional characterizations revealed active epigenetic enrichment for baaSNPs and prevailing osteoporosis-relevant regulatory roles for their chromatin interaction genes. Further motif enrichment and network mapping prioritized several putative, key transcription factors (TFs) controlling osteoporosis binding to baaSNPs. Specifically, we selected one top-ranked TF and deciphered that an intronic baaSNP (rs11202530) could allele-preferentially bind to YY2 to augment PAPSS2 expression through chromatin interactions and promote osteoblast differentiation. Our results underline the roles of TF-mediated enhancer-promoter contacts for osteoporosis, which may help to better understand the intricate molecular regulatory mechanisms underlying osteoporosis risk loci.


Asunto(s)
Osteoporosis , Secuencias Reguladoras de Ácidos Nucleicos , Humanos , Factores de Transcripción/genética , Osteoporosis/genética , Cromatina/genética , Regiones Promotoras Genéticas/genética
2.
Biomed Mater ; 19(2)2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38194710

RESUMEN

Posterior capsule opacification (PCO) is a main complication after cataract surgery and intraocular lens (IOLs) implantation and is attributed to residual lens epithelial cells (LECs) migrating to the IOL surface and posterior capsules. IOL surface modification has been a newly-developing research filed in recent years; however, the applicability and economical acquisition of modified materials remain unsolved. In this study, we first applied a metal-polyphenolic network coating with a self-assembly technique on the IOL surface by using tannic acid (TA) combined with AlCl3, which are easily acquire and applying on the IOL surface to solve the IOL transmittance affair. Using wound healing and Transwell assay to verify AZD0364 inhibits cell migration (P< 0.05), the lipopolysaccharide-induced macrophage inflammation model to verify pterostilbene (PTE) inhibits the inflammatory reaction (P< 0.01). By optimizes its self-assembly coating parameters and calculating its drug release kinetics, we successfully loaded these two drugs on the coating, named TA (AZD0364/PTE) IOL. Its surface morphology characteristics were analyzed by scanning electron microscope, x-ray photoelectron spectrometer and water contact angle. The optical performance was carefully investigated by optical instruments and equipment (n= 3). Thein vitroresults showed that TA (AZD0364/PTE) IOL can significantly inhibit cell adhesion and acute inflammation (n= 3,P< 0.0001). Importantly, afterin vivoimplantation for 28 d with eight rabbits PCO models in two groups, the TA (AZD0364/PTE) IOL group maintained clear refracting media and decreased the inflammatory reaction compared with the original IOL group (P< 0.05). This study provides a new applicable and economical strategy for preventing PCO and offers a reference for the next generation of IOLs that benefit cataract patients.


Asunto(s)
Opacificación Capsular , Lentes Intraoculares , Polifenoles , Animales , Humanos , Conejos , Opacificación Capsular/prevención & control , Inflamación/prevención & control , Diseño de Prótesis , Complicaciones Posoperatorias/prevención & control
3.
Invest Ophthalmol Vis Sci ; 65(1): 16, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38180772

RESUMEN

Purpose: The purpose of this study was to present our findings of the distribution pattern of choroidal arteries and large veins in patients with age-related macular degeneration (AMD) using indocyanine green angiography (ICGA). Methods: A retrospective analysis was conducted on 980 patients who underwent ICGA at The Second Affiliated Hospital of Xi'an Jiaotong University from 2017 to 2023, including 240 patients with AMD. Secondary image processing was applied to the angiographic videos to obtain posterior distribution maps of choroidal arteries and large veins. Differences between different distribution patterns regarding age, gender, eye laterality, and circulation time were compared. We also conducted a comparison of choroidal vascular distribution characteristics between patients with AMD and patients with diabetic retinopathy (DR) and provided a summary of choroidal vascular distribution patterns in AMD. Results: The filling patterns of choroidal arteries can be classified into the invisible trunk arteries type, the partially masked trunk arteries type, and the exposed trunk arteries type. The vascular topography of the large choroidal vein can be classified into the watershed type, the non-watershed type, and the unknown type, further divided into six subtypes. The distribution patterns of choroidal arteries and veins were significantly correlated with age (P < 0.001). Left eye, older age, and the exposed trunk arteries type were independent risk factors for non-watershed large choroidal vein (P < 0.05). The non-watershed type was the main characteristic of the venous phase in AMD. Conclusions: The distribution characteristics of the arterial and venous patterns in AMD suggest atrophy of the small blood vessels in the choroid and insufficient perfusion pressure of the blood flow.


Asunto(s)
Verde de Indocianina , Degeneración Macular , Humanos , Estudios Retrospectivos , Angiografía , Degeneración Macular/diagnóstico , Coroides
4.
Int J Ophthalmol ; 16(12): 1935-1941, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38111943

RESUMEN

AIM: To investigate the role of reactive oxygen species (ROS) in epithelial-mesenchymal transition (EMT) and apoptosis of human lens epithelial cells (HLECs). METHODS: Flow cytometry was used to assess ROS production after transforming growth factor ß2 (TGF-ß2) induction. Apoptosis of HLECs after H2O2 and TGF-ß2 interference with or without ROS scavenger N-acetylcysteine (NAC) were assessed by flow cytometry. The corresponding protein expression levels of the EMT marker α-smooth muscle actin (α-SMA), the extracellular matrix (ECM), marker fibronectin (Fn), and apoptosis-associated proteins were detected by using Western blotting in the presence of an ROS scavenger (NAC). Wound-healing and Transwell assays were used to assess the migration capability of HLECs. RESULTS: TGF-ß2 stimulates ROS production within 8h in HLECs. Additionally, TGF-ß2 induced HLECs cell apoptosis, EMT/ECM synthesis protein markers expression, and pro-apoptotic proteins production; nonetheless, NAC treatment prevented these responses. Similarly, TGF-ß2 promoted HLECs cell migration, whereas NAC inhibited cell migration. We further determined that although ROS initiated apoptosis, it only induced the accumulation of the EMT marker α-SMA protein, but not COL-1 or Fn. CONCLUSION: ROS contribute to TGF-ß2-induced EMT/ECM synthesis and cell apoptosis of HLECs; however, ROS alone are not sufficient for EMT/ECM synthesis.

5.
Am J Hum Genet ; 110(8): 1266-1288, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37506691

RESUMEN

Most of the single-nucleotide polymorphisms (SNPs) associated with insulin resistance (IR)-relevant phenotypes by genome-wide association studies (GWASs) are located in noncoding regions, complicating their functional interpretation. Here, we utilized an adapted STARR-seq to evaluate the regulatory activities of 5,987 noncoding SNPs associated with IR-relevant phenotypes. We identified 876 SNPs with biased allelic enhancer activity effects (baaSNPs) across 133 loci in three IR-relevant cell lines (HepG2, preadipocyte, and A673), which showed pervasive cell specificity and significant enrichment for cell-specific open chromatin regions or enhancer-indicative markers (H3K4me1, H3K27ac). Further functional characterization suggested several transcription factors (TFs) with preferential allelic binding to baaSNPs. We also incorporated multi-omics data to prioritize 102 candidate regulatory target genes for baaSNPs and revealed prevalent long-range regulatory effects and cell-specific IR-relevant biological functional enrichment on them. Specifically, we experimentally verified the distal regulatory mechanism at IRS1 locus, in which rs952227-A reinforces IRS1 expression by long-range chromatin interaction and preferential binding to the transcription factor HOXC6 to augment the enhancer activity. Finally, based on our STARR-seq screening data, we predicted the enhancer activity of 227,343 noncoding SNPs associated with IR-relevant phenotypes (fasting insulin adjusted for BMI, HDL cholesterol, and triglycerides) from the largest available GWAS summary statistics. We further provided an open resource (http://www.bigc.online/fnSNP-IR) for better understanding genetic regulatory mechanisms of IR-relevant phenotypes.


Asunto(s)
Resistencia a la Insulina , Polimorfismo de Nucleótido Simple , Humanos , Polimorfismo de Nucleótido Simple/genética , Estudio de Asociación del Genoma Completo , Resistencia a la Insulina/genética , Factores de Transcripción/genética , Cromatina/genética , Fenotipo , Elementos de Facilitación Genéticos/genética
6.
Curr Eye Res ; 48(7): 627-638, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36988559

RESUMEN

PURPOSE: Posterior capsular opacification is the most common complication after cataract surgery. Abnormal proliferation, migration, epithelial-mesenchymal transition, and extracellular matrix synthesis of residual lens epithelial cells are considered to be the main pathogenic mechanisms. Hepatocyte nuclear factor 4α has been reported to regulate epithelial-mesenchymal transition in different tumors. Our objective was to investigate the role and mechanism of hepatocyte nuclear factor 4α in posterior capsular opacification. METHODS: Hepatocyte nuclear factor 4α expression was tested in posterior capsular opacification rat lens capsules and cell models. Hepatocyte nuclear factor 4α was knocked down using small hairpin RNA. Cell viability was measured by Cell Counting Kit-8 assay. Cell migration ability was evaluated by wound healing and Transwell assays. Epithelial-mesenchymal transition markers were detected by Western blotting. Transcriptome sequencing was used to screen for downstream effectors of hepatocyte nuclear factor 4α. Chromatin immunoprecipitation and a dual luciferase reporter assay were used to determine the binding of hepatocyte nuclear factor 4α to the MMP2 promoter region. RESULTS: Hepatocyte nuclear factor 4α was downregulated in posterior capsular opacification tissue and cell models. In vitro studies showed that hepatocyte nuclear factor 4α deletion facilitated cell proliferation, migration, and epithelial-mesenchymal transition protein marker expression in lens epithelial cells. Hepatocyte nuclear factor 4α knockdown promoted epithelial-mesenchymal transition and migration of lens epithelial cells via MMP2. Mechanistically, hepatocyte nuclear factor 4α decreased MMP2 expression by binding to the MMP2 promoter region. Hepatocyte nuclear factor 4α deletion also promoted epithelial-mesenchymal transition in rat lens capsules. CONCLUSIONS: We demonstrated that hepatocyte nuclear factor 4α inhibited epithelial-mesenchymal transition of lens epithelial cells by directly binding to the MMP2 promoter region and inhibiting the expression of MMP2, thus leading to retardation of posterior capsular opacification formation and development, suggesting that hepatocyte nuclear factor 4α is a potential therapeutic target for posterior capsular opacification.


Asunto(s)
Opacificación Capsular , Factor Nuclear 4 del Hepatocito , Cápsula del Cristalino , Cristalino , Metaloproteinasa 2 de la Matriz , Animales , Ratas , Opacificación Capsular/metabolismo , Cápsulas/metabolismo , Movimiento Celular , Proliferación Celular , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Cápsula del Cristalino/patología , Cristalino/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo
7.
Cell Signal ; 97: 110378, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35690292

RESUMEN

Posterior capsular opacification (PCO) can cause postoperative visual loss after cataract surgery. Residual human lens epithelial cell (HLEC) proliferation, migration, epithelial-mesenchymal transition (EMT) and synthesis of extracellular matrix (ECM) are the entitative reasons for PCO. Low expression of Ral-binding protein 1-associated Eps domain-containing 2 (REPS2) and high levels of basic fibroblast growth factor (b-FGF) were observed in the lens and postoperative aqueous humor of cataract patients. REPS2 was identified as a negative regulator in growth factor signaling; however, its function in HLECs is unknown. This was first investigated in the present study by evaluating REPS2 expression in anterior lens capsules from cataract patients, a mouse cataract model, and HLE-b3 cells. The biological function of REPS2 in HLE-B3 cells was assessed by REPS2 silencing and Cell Counting Kit 8, wound healing, Transwell migration, F-actin staining, G-protein pulldown and western blot assays. In the present study, REPS2 was significantly downregulated in human and mouse cataract capsules and H2O2-treated HLE-B3 cells. REPS2 knockdown increased fibronectin, type I collagen, and α-smooth muscle actin expression levels and stimulated HLECs proliferation and migration; these effects were enhanced by FGF treatment and accompanied with focal adhesion kinase (FAK) phosphorylation, cell division cycle 42 (Cdc42) activation, focal adhesion protein upregulation, and F-actin cytoskeleton reorganization. However, treatment with the FAK inhibitor PF573228 abolished these effects. Thus, REPS2 downregulation in cataract HLECs induces their proliferation and facilitates FGF-induced ECM synthesis, EMT, cell adhesion and migration by activating FAK/Cdc42 signaling, which may underlie PCO pathogenesis.


Asunto(s)
Proteínas de Unión al Calcio , Opacificación Capsular , Animales , Proteínas de Unión al Calcio/metabolismo , Opacificación Capsular/metabolismo , Opacificación Capsular/patología , Cápsulas/metabolismo , Cápsulas/farmacología , Adhesión Celular , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Factor 2 de Crecimiento de Fibroblastos/farmacología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Ratones , Proteína de Unión al GTP cdc42
8.
Invest Ophthalmol Vis Sci ; 63(4): 11, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35435923

RESUMEN

Purpose: Age-related cataract (ARC) is a major cause of vision impairment worldwide. The E3 ubiquitin ligase RING finger protein 157 (RNF157) is involved in regulating cell survival and downregulated in human cataractous lens samples. However, the function of RNF157 in cataracts remains unclear. This study aimed to determine the role of RNF157 in ARC. Methods: Real-time polymerase chain reaction (PCR) and Western blotting were used to analyze the expression of RNF157 in clinical lens capsules, rat cataract models, and oxidative stress cell models. Western blot analysis and flow cytometry were used to evaluate cell apoptosis. Co-IP assay, protein stability assay, and ubiquitination assay were used to detect the interaction between RNF157 and its substrate p53. Results: The expression of RNF157 was downregulated in human cataract samples, UVB-induced rat cataract model, and H2O2-treated human lens epithelial cells (LECs). Ectopic expression of RNF157 protected LECs from H2O2-induced apoptosis. In contrast, knockdown of RNF157 enhanced oxidative stress-induced apoptotic cell death. Moreover, silence of RNF157 in the rat ex vivo lens model exacerbated lens opacity. Mechanistically, RNF157 causes ubiquitination and degradation of the tumor antigen p53. Overexpression of p53 eliminated the antiapoptotic effects of RNF157, whereas p53 knockdown rescued RNF157 silencing-induced cell death. Conclusions: Our findings revealed that reduced RNF157 expression promoted LEC apoptosis by upregulating p53 in cataracts, suggesting that the regulation of RNF157 expression may serve as a potential therapeutic strategy for cataracts.


Asunto(s)
Catarata , Cristalino , Animales , Apoptosis , Catarata/metabolismo , Células Epiteliales/metabolismo , Peróxido de Hidrógeno/efectos adversos , Cristalino/metabolismo , Ratas , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
9.
Life Sci ; 286: 120061, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34666037

RESUMEN

AIMS: The epithelial-mesenchymal transition (EMT), extracellular matrix (ECM) synthesis and cell migration of residual lens cells constitute the canonical mechanisms of posterior capsular opacification (PCO). Recently, myofibroblast cell apoptosis is also observed in the rabbit PCO model. However, whether cell apoptosis is a key factor affecting PCO and regulates EMT/ECM synthesis/cell migration remains obscure. MAIN METHODS: Flow cytometry was utilized to assess cell cycle and apoptosis. EMT marker α-smooth muscle actin (α-SMA), ECM markers fibronectin (Fn), type 1 collagen (COL-1) and apoptosis-associated proteins in the presence or absence of EMT/ECM inhibitor (LY2109761), apoptosis inhibitor (ZVAD) or apoptosis activator (BTSA1) were detected by Western blotting. Downstream effector genes in apoptosis-induced lens epithelial cell lines (LECs) were analyzed by RNA-seq. Gene silencing and overexpression in LECs were performed to validate the role of effector genes. We measured cell migration capability using Wound healing and Transwell assays. KEY FINDINGS: We found that TGF-ß2 induced cell apoptosis. ZVAD inhibited α-SMA expression in the ex vivo capsule model and decreased the expression of both EMT and ECM markers in TGF-ß2-treated LECs. RNA-seq revealed that FILIP1L was significantly decreased in apoptosis-activated cells. We further validated that the knockdown of FILIP1L could enhance EMT and ECM synthesis and promote cell migration and that FILIP1L overexpression could reverse these effects. Apoptosis might contribute to TGF-ß2-induced EMT and ECM synthesis during PCO, and these contributions are mediated by FILIP1L. SIGNIFICANCE: Our findings uncover the role of apoptosis in PCO development and provide new drug targets.


Asunto(s)
Opacificación Capsular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Apoptosis/efectos de los fármacos , Opacificación Capsular/genética , Ciclo Celular , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , China , Colágeno Tipo I/metabolismo , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/fisiología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/fisiología , Fibronectinas/metabolismo , Citometría de Flujo/métodos , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Cristalino/metabolismo , Masculino , Cápsula Posterior del Cristalino/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Porcinos , Factor de Crecimiento Transformador beta2/metabolismo , Factor de Crecimiento Transformador beta2/farmacología
10.
Invest Ophthalmol Vis Sci ; 61(12): 3, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33006594

RESUMEN

Purpose: Cataract, a clouding of the intraocular lens, is the leading cause of blindness. The lens-expressed long noncoding RNA OIP5-AS1 was upregulated in lens epithelial cells from patients with cataracts, suggesting its pathogenic role in cataracts. We investigated the regulatory role of OIP5-AS1 in the development of cataracts as well as potential RNA binding proteins, downstream target genes, and upstream transcription factors. Methods: Clinical capsules and ex vivo and in vitro cataract models were used to test OIP5-AS1 expression. Cell apoptosis was detected using Western blots, JC-1 staining, and flow cytometry. Ribonucleoprotein immunoprecipitation-qPCR was performed to confirm the interaction of OIP5-AS1 and POLG. Chromatin immunoprecipitation-qPCR was used to determine the binding of TFAP2A and the OIP5-AS1 promoter region. Results: OIP5-AS1 was upregulated in cataract lenses and B3 cells under oxidative stress. OIP5-AS1 knockdown protected B3 cells from H2O2-induced apoptosis and alleviated lens opacity in the ex vivo cataract model. HuR functioned as a scaffold carrying OIP5-AS1 and POLG mRNA and mediated the decay of POLG mRNA. POLG was downregulated in the cataract lens and oxidative-stressed B3 cells, and POLG depletion decreased the mtDNA copy number and MMP, increased reactive oxygen species production, and sensitized B3 cells to oxidative stress-induced apoptosis. POLG overexpression reversed these effects. TFAP2A bound the OIP5-AS1 promoter and contributed to OIP5-AS1 expression. Conclusions: We demonstrated that OIP5-AS1, activated by TFAP2A, contributed to cataract formation by inhibiting POLG expression mediated by HuR, thus leading to increased apoptosis of lens epithelial cells and aggravated lens opacity, suggesting that OIP5-AS1 is a potential target for cataract treatment.


Asunto(s)
Apoptosis/genética , Catarata/genética , ADN Polimerasa gamma/antagonistas & inhibidores , Regulación de la Expresión Génica/fisiología , ARN Largo no Codificante/genética , Animales , Western Blotting , Catarata/metabolismo , Catarata/patología , Proliferación Celular , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Femenino , Citometría de Flujo , Humanos , Peróxido de Hidrógeno/toxicidad , Cristalino/citología , Masculino , Potenciales de la Membrana , Estrés Oxidativo , Plásmidos/genética , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Factor de Transcripción AP-2/metabolismo , Transfección
11.
Histochem Cell Biol ; 154(6): 629-638, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32997263

RESUMEN

Proliferative vitreoretinopathy (PVR) is the most common cause of surgical failure in the rhegmatogenous retinal detachment (RD) treatment. Retinal pigment epithelial (RPE) cell proliferation, migration, and the synthesis of extracellular matrix (ECM) are intrinsic to the formation of a PVR membrane. High level of interleukin-6 (IL-6) has been found in the vitreous of PVR patients, while the role of IL-6 in RPE cells remaining further characterized. In the present study, we evaluated the potential regulatory effects of IL-6 on cell migration, ECM components, and transforming growth factor ß2 (TGF-ß2) expression in RPE cells. Furthermore, cell counting kit-8 (CCK­8) assay was used to investigate cell proliferation activity. We found that IL-6 promoted fibronectin (Fn) and type I collagen (COL-1), TGF-ß2 expression in RPE cells, also stimulate RPE cell migration effectively. Moreover, the induction of IL-6 activated the Janus kinase/signal transducers and activators of transcription (JAK/STAT3) and the nuclear factor kappa-B (NF-κB) signaling pathways significantly. Simultaneously, both JAK/STAT3 and NF-κB pathways inhibitors, WP1066 and BAY11-7082, alleviated IL-6-induced biological effects, respectively. However, it was noted that IL-6 had little effect on α-smooth muscle actin (α-SMA) expression. Collectively, our results reveal that IL-6 promotes RPE cell migration and ECM synthesis via activating JAK/STAT3 and NF-κB signaling pathways, which may play a crucial role in PVR formation.


Asunto(s)
Matriz Extracelular/metabolismo , Interleucina-6/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Movimiento Celular , Células Cultivadas , Humanos , Epitelio Pigmentado de la Retina/citología
12.
Dev Growth Differ ; 61(7-8): 410-418, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31608440

RESUMEN

Macular fibrosis is a vital obstacle of vision acuity improvement of age-related macular degeneration patients. This study was to investigate the effects of interleukin 2 (IL-2) on epithelial-mesenchymal transition (EMT), extracellular matrix (ECM) synthesis and transforming growth factor ß2 (TGF-ß2) expression in retinal pigment epithelial (RPE) cells. 10 µg/L IL-2 was used to induce fibrosis in RPE cells for various times. Western blot was used to detect the EMT marker α-smooth muscle actin (α-SMA), ECM markers fibronectin (Fn) and type 1 collagen (COL-1), TGF-ß2, and the activation of the JAK/STAT3 and NF-κB signaling pathway. Furthermore, JAK/STAT3 and NF-κB signaling pathways were specifically blocked by WP1066 or BAY11-7082, respectively, and the expression of α-SMA, COL-1, Fn and TGF-ß2 protein were detected. Wound healing and Transwell assays were used to measure cell migration ability of IL-2 with or without WP1066 or BAY11-7082. After induction of IL-2, the expressions of Fn, COL-1, TGF-ß2 protein were significantly increased, and this effect was correlated with IL-2 treatment duration, while α-SMA protein expression did not change significantly. Both WP1066 and BAY11-7082 could effectively downregulate the expression of Fn, COL-1 and TGF-ß2 induced by IL-2. What's more, both NF-κB and JAK/STAT3 inhibitors could suppress the activation of the other signaling pathway. Additionally, JAK/STAT3 inhibitor WP1066 and NF-κB inhibitor BAY 11-7082 could obviously decrease RPE cells migration capability induced by IL-2. IL-2 promotes cell migration, ECM synthesis and TGF-ß2 expression in RPE cells via JAK/STAT3 and NF-κB signaling pathways, which may play an important role in proliferative vitreoretinopathy.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Interleucina-2/farmacología , Factor de Crecimiento Transformador beta2/metabolismo , Actinas/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Colágeno Tipo I/metabolismo , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Humanos , Quinasas Janus/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Nitrilos/farmacología , Piridinas/farmacología , Epitelio Pigmentado de la Retina/citología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Sulfonas/farmacología , Tirfostinos/farmacología
13.
Cell Cycle ; 18(15): 1714-1726, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31234714

RESUMEN

Objective: The present study was conducted to determine the role of gremlin during the development of posterior capsular opacification (PCO) via in vitro and in vivo experiments. Methods: The activation, roles and relationships of the BMPs/Smad1/5, MAPK, FAK and AKT signaling pathways in human lens epithelial cells (HLECs) after gremlin induction were detected by western blotting and real-time PCR. Wound-healing, transwell, capsular bag models and rat PCO models assays were used to test the effects of gremlin on HLECs' migration, proliferation, EMT-specific protein α-smooth muscle actin(α-SMA)and development of PCO in rats. Results: Our data showed that knockdown of the gremlin inhibited the development of PCO and reduced expression of α-SMA in rats. While gremlin did not alter the migration of HLECs, it increased the expression of p-ERK and p-AKT. Knockout of Smad2 or Smad3 inhibited the expression of p-ERK and p-AKT proteins induced by gremlin. Gremlin also reduced BMP4-induced expression of the p-Smad1/5 protein. Finally, knockout of Smad1/5 increased gremlin-induced expression of α-SMA, fibronectin and type I collagen (COL-1) in HLECs. Conclusion: These results suggested that gremlin contributed to the development of PCO by promoting LEC proliferation, activation of TGF-ß/Smad, ERK and AKT signaling and inhibition of BMPs/Smad1/5 signaling. Furthermore, inhibiting gremlin effectively impaired both PCO development in rats and EMT in the lens capsule. Thus, our data suggest that gremlin might be a potential target for PCO.


Asunto(s)
Opacificación Capsular/metabolismo , Células Epiteliales/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas/metabolismo , Actinas/metabolismo , Animales , Opacificación Capsular/genética , Línea Celular , Movimiento Celular/genética , Proliferación Celular/genética , Colágeno Tipo I/metabolismo , Transición Epitelial-Mesenquimal/genética , Femenino , Fibronectinas/metabolismo , Silenciador del Gen , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/farmacología , Sistema de Señalización de MAP Quinasas/genética , Masculino , Proteínas/genética , ARN Interferente Pequeño , Ratas , Proteínas Smad Reguladas por Receptores/genética , Proteínas Smad Reguladas por Receptores/metabolismo , Porcinos , Factor de Crecimiento Transformador beta2/metabolismo , Cicatrización de Heridas/genética
14.
Int J Mol Med ; 42(6): 3591-3601, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30280182

RESUMEN

The migration of lens epithelial cells towards the posterior capsule is a key event in the development of posterior capsule opacification (PCO). Accumulating evidence has described crosstalk between growth factors and adhesive signaling pathways in wound healing and cell migration. The aim of the present study was to elucidate an aberrant transforming growth factor (TGF)­ß2 signaling pathway that regulated the migration of lens epithelial cells in the pathological context of PCO. The expression of fibronectin, focal adhesion kinase (FAK) and phosphorylated (p)­FAK in HLE­B3 cells following TGF­ß2 treatment was determined by western blot analysis and the expression of integrin α5ß1 was detected by flow cytometry. Cell migration capacity was measured by wound healing and Transwell assays in the presence of 1,2,4,5­tetraaminobenzene tetrahydrochloride, a selective FAK inhibitor, fibronectin small interfering RNA interference, arginylglycylaspartic acid peptides or α5ß1­integrin neutralizing antibodies. The 1,2,4,5­tetraaminobenzene tetrahydrochloride was administered daily to 16 rabbits following cataract surgery. Fibronectin and TGF­ß expression were increased in the PCO group, demonstrated by immunofluorescence assays. PCO grading was conducted by slit­lamp biomicroscopy and evaluation of posterior capsule opacification software. It was observed that TGF­ß2 promoted HLE­B3 cell migration and upregulated fibronectin expression, which was followed by an increased phosphorylation of FAK. In addition, TGF­ß2 treatment and fibronectin surface coating significantly increased cell migration and FAK activation, which was inhibited by disrupting fibronectin­integrin α5ß1 interaction with the arginylglycylaspartic acid peptide, α5ß1­integrin neutralizing antibody or fibronectin depletion. Finally, suppression of FAK signaling by its inhibitor significantly decreased cell migration in vitro and attenuated PCO development in vivo. In summary, TGF­ß2 was indicated to promote the migration of lens epithelial cells through the TGF­ß2/fibronectin/integrin/FAK axis. Inhibition of FAK activity decreased TGF­ß2­mediated cell migration in vitro and improved the symptoms of PCO in a rabbit model.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/enzimología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Cristalino/citología , Factor de Crecimiento Transformador beta2/farmacología , Animales , Opacificación Capsular/enzimología , Opacificación Capsular/patología , Línea Celular , Células Epiteliales/efectos de los fármacos , Fibronectinas/metabolismo , Humanos , Integrina alfa5beta1/metabolismo , Masculino , Conejos , Transducción de Señal
15.
Int J Biol Sci ; 14(4): 437-448, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29725265

RESUMEN

Connective tissue growth factor (CTGF) is a crucial factor that plays a major role in the process of posterior capsule opacification (PCO). However, the effects of CTGF on the proliferation and migration of lens epithelial cells (LECs) and on the mechanism of the epithelial mesenchymal transition (EMT) and extracellular matrix (ECM) in human lens epithelial cells (HLECs) as well as the effects of shRNA-mediated CTGF knockdown on the development of PCO in rats remain unclear. In the present study, we found that CTGF promoted EMT, proliferation, migration and the expression of p-ERK1/2 protein in HLECs but exerted little effect on the expression of p-p38 and p-JNK1/2 proteins. MEK inhibitor U0126 effectively restrained the CTGF-induced expression of α-smooth muscle actin (α-SMA), fibronectin (Fn) and type I collagen (COL-1) in HLECs. CTGF knockdown effectively postponed the onset of PCO in the rats and significantly reduced the expression of α-SMA in the capsule. In conclusion, CTGF contributed to the development of PCO presumably by promoting proliferation, migration of LECs, EMT specific protein expression and ECM synthesis in HLECs, which is dependent on ERK signalling. Furthermore, blocking CTGF effectively inhibited PCO in the rats and the EMT specific protein expression in the lens capsule.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/fisiología , Cápsula Posterior del Cristalino/patología , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Células Epiteliales , Técnicas de Silenciamiento del Gen , Humanos , Cristalino/citología , Sistema de Señalización de MAP Quinasas/genética , Interferencia de ARN , Ratas , Porcinos
16.
Exp Eye Res ; 172: 94-103, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29617629

RESUMEN

The purpose of this work was to determine the effects of interleukin-6 (IL-6) on the development of posterior capsular opacification (PCO) in vitro and in vivo. Western blot and real-time PCR were used to test the IL-6-induced epithelial-mesenchymal transition (EMT) marker α-smooth muscle actin (α-SMA), the extracellular matrix (ECM) markers fibronectin (Fn) and type I collagen (COL-1), transforming growth factor ß2 (TGF-ß2), and the activation and role of the JAK/STAT3 signaling pathway in human lens epithelial cells (HLECs). Immunocytofluorescence staining was performed to detect gp130 and IL-6Rα expression in HLECs. Rat PCO models were then established to examine the impact of STAT3 knockdown by shRNA adeno-associated virus on PCO development, and immunohistochemical staining was performed to detect the expression of Fn in the anterior and posterior capsule in vivo. We found that IL-6 promotes the expression of Fn, COL-1, TGF-ß2, p-JAK2 and p-STAT3 in HLECs but exerts little effect on α-SMA. The JAK/STAT3 inhibitor WP1066 effectively suppressed the IL-6-induced expression of Fn and COL-1 in lens epithelial cells. STAT3 knockdown effectively inhibited the development of PCO in rats and significantly reduced the expression of Fn in the anterior and posterior capsule. These data suggest that IL-6 contributes to the development of PCO by promoting TGF-ß2 activation and ECM synthesis through a JAK/STAT3 signaling-dependent mechanism. Furthermore, inhibiting JAK/STAT3 signaling effectively impairs both PCO development in rats and ECM synthesis in the lens capsule.


Asunto(s)
Opacificación Capsular/etiología , Células Epiteliales/efectos de los fármacos , Interleucina-6/farmacología , Cristalino/efectos de los fármacos , Cápsula Posterior del Cristalino/efectos de los fármacos , Actinas/metabolismo , Animales , Western Blotting , Opacificación Capsular/metabolismo , Colágeno Tipo I/metabolismo , Receptor gp130 de Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/fisiología , Fibronectinas/metabolismo , Humanos , Subunidad alfa del Receptor de Interleucina-6/metabolismo , Quinasas Janus/metabolismo , Cristalino/metabolismo , Cápsula Posterior del Cristalino/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT3/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA