Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cornea ; 41(12): 1545-1552, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-35965398

RESUMO

PURPOSE: Circular RNAs (circRNAs) are a novel class of endogenous noncoding RNAs that regulate gene expression through the competitive endogenous RNA (ceRNA) mechanism. CircRNA-associated-ceRNA networks are closely related to oxidative stress-related diseases. Oxidative stress-induced dysfunction of the corneal endothelium (CE) is a major pathological feature in many corneal diseases. This study was aimed to analyze circRNA-associated-ceRNA networks in oxidative stress-induced CE dysfunction. METHODS: A CE dysfunction model was established using human corneal endothelial cells (HCECs) treated with H 2 O 2 at a concentration of 250 µM for 4 hours at 37°C. High-throughput sequencing was conducted to determine the expression profiles of circRNA, miRNA, and mRNA. Bioinformatic analyses, including Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes analysis, were conducted to identify the potential biological modules and pathologic pathways of dysregulated circRNAs. CircRNA-associated-ceRNA networks were established based on the data of sequencing and bioinformatic analyses. RESULTS: We obtained 108 differentially expressed circRNAs, including 77 upregulated and 31 downregulated circRNAs. GO analysis suggested that dysregulated circRNAs were mainly targeted to protein quality control for misfolded or incompletely synthesized proteins (biologic process), nuclear chromatin (cellular component), and ubiquitin protein ligase binding (molecular function). GO terms related to CE functions responding to oxidative stress were also identified. Kyoto Encyclopedia of Genes and Genomes pathway analysis indicated that dysregulated circRNAs were mostly enriched in the adherens junction pathway. Network analysis identified several potential therapeutic targets for CE dysfunction. CONCLUSIONS: CircRNAs are significantly dysregulated in HCECs under oxidative stress. The circRNA-associated-ceRNA networks are closely related to HCEC functions. Targeting these networks might provide novel therapies for CE dysfunction.


Assuntos
MicroRNAs , RNA Circular , Humanos , RNA Circular/genética , Células Endoteliais/metabolismo , Redes Reguladoras de Genes , Perfilação da Expressão Gênica , MicroRNAs/genética
2.
Front Genet ; 13: 823517, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35368656

RESUMO

Macrophage polarization is a process that macrophages exert different functions according to surrounding micro-environment. Macrophages commonly exist in two distinct subsets: classically activated M1 macrophages and alternatively activated M2 macrophages. Circular RNAs (circRNAs) are a novel class of non-coding RNAs generated by back-splicing. Thousands of circRNAs were identified in different cells and tissues. Recent studies have revealed that circRNAs play a crucial role in regulating transcriptional and post-transcriptional gene expression. However, the effects and roles of circRNAs in macrophage polarization have not been well elucidated. Here, circRNAs expression profiles were determined in human THP-1 macrophages incubated in conditions causing activation toward M1 (interferon-γ + LPS) or M2 (interleukin-4) phenotypes. Overall, 9,720 circular RNA were detected from RNA sequencing data. Compared with M2 macrophages, a total of 140 circRNAs were aberrantly expressed in M1 macrophages, including 71 up-regulated circRNAs and 69 down-regulated circRNAs. Quantitative real-time PCR (qRT-PCR) results were generally consistent with the selected differentially expressed circRNAs. Gene Ontology (GO) and KEGG pathway analyses were used to predict biological functions and potential mechanisms of the host linear transcripts of these up-regulated and down-regulated circRNAs. Furthermore, we found that the expression level of circRNA-RNF19B (circRNF19B) in M1 macrophages was significantly higher than that in THP-1 macrophages and M2 macrophages. circRNF19B expression was increased when M2 converted to M1 whereas decreased when M1 converted to M2. Knockdown of circRNF19B following the activation of THP-1 cells using interferon-γ + LPS diminished the expression of M1 macrophages markers and elevated the expression of M2 macrophages markers. In conclusion, these data suggest the involvement of altered circRNAs expression patterns in macrophages exposure to different activating conditions. Circular RNAs may play important roles in regulating macrophage polarization.

3.
Exp Eye Res ; 197: 108107, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32531187

RESUMO

Pathological ocular angiogenesis commonly results in visual impairment or even blindness. Unveiling the mechanisms of pathological angiogenesis is critical to identify the regulators and develop effective targeted therapies. Here, we used corneal neovascularization (CNV) model to investigate the mechanism of pathological ocular angiogenesis. We show that N6-methyladenosine (m6A) mRNA demethylation mediated by fat mass- and obesity-associated protein (FTO) could regulate endothelial cell (EC) function and pathological angiogenesis during CNV. FTO levels are increased in neovascularized corneas and ECs under pathological conditions. In vitro silencing of FTO in ECs results in reduced cellular proliferation, migration, and tube formation under both basal and pathological conditions. Furthermore, FTO silencing attenuates suture-induced CNV in vivo. Mechanically, FTO silencing in ECs could increase m6A methylation levels in critical pro-angiogenic genes, such as FAK, leading to decreased RNA stability and increased RNA decay through m6A reader YTHDF2. Our study demonstrates that FTO regulates pathological ocular angiogenesis by controlling EC function in an m6A-YTHDF2-dependent manner.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Neovascularização da Córnea/genética , Regulação da Expressão Gênica , Glicoproteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Neovascularização da Córnea/metabolismo , Neovascularização da Córnea/patologia , Modelos Animais de Doenças , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Estabilidade de RNA , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo
4.
Theranostics ; 10(7): 3293-3307, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194869

RESUMO

Rationale: Choroidal neovascularization (CNV) is a major cause of severe vision loss and occurs in many ocular diseases, especially neovascular age-related macular degeneration (nAMD). Circular RNAs (circRNAs) are emerging as a new class of endogenous noncoding RNAs, which have been implicated in the regulation of endothelial cell dysfunction in diabetes mellitus and cancer. In this study, we aimed to determine the role of circRNA-ZBTB44 (cZBTB44) in the pathogenesis of CNV. Methods: Quantitative polymerase chain reaction was conducted to detect cZBTB44 expression pattern during CNV development. Isolectin B4 staining, hematoxylin and eosin (HE) staining, and choroidal sprouting assay ex vivo were conducted to evaluate the role of cZBTB44 in the development of CNV. Endothelial cell proliferation, migration and tube formation assays were conducted to determine the role of cZBTB44 in angiogenic effect in vitro. Bioinformatics analysis, RNA immunoprecipitation assay, luciferase assay, and in vitro studies were conducted to investigate the mechanism of cZBTB44-mediated CNV development. Results: cZBTB44 expression was significantly up-regulated in a laser-induced CNV mouse model in vivo and in endothelial cells upon hypoxia stress in vitro. cZBTB44 silencing retarded CNV development, while overexpression of cZBTB44 showed the opposite effects. The role of cZBTB44 in CNV development was confirmed in choroidal sprouting assay ex vivo. cZBTB44 silencing reduced endothelial cell viability, proliferation, migration and tube formation in vitro. cZBTB44 acted as miR-578 sponge to sequester and inhibit miR-578 activity, which led to increased expression of vascular endothelial growth factor A (VEGFA) and vascular cell adhesion molecule-1 (VCAM1). Overexpression of miR-578 mimicked cZBTB44 silencing-mediated anti-angiogenic effects in vivo and in vitro. Furthermore, dysregulated cZBTB44 expression was detected in the clinical samples of nAMD patients. Conclusions: This study provided novel insights into the molecular pathogenesis of CNV. The cZBTB44-miR-578-VEGFA/VCAM1 axis might be a potential source of novel therapeutic targets for neovascularization-related diseases.


Assuntos
Neovascularização de Coroide/genética , RNA Circular/metabolismo , Regiões 3' não Traduzidas , Animais , Hipóxia Celular , Corioide/citologia , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Vetores Genéticos , Lasers , Macaca mulatta , Camundongos , Camundongos Endogâmicos C57BL , RNA Circular/biossíntese , RNA Circular/genética , RNA Interferente Pequeno/genética , Retina/citologia , Coloração e Rotulagem , Regulação para Cima , Molécula 1 de Adesão de Célula Vascular/genética , Fator A de Crescimento do Endotélio Vascular/genética
5.
Proc Natl Acad Sci U S A ; 116(15): 7455-7464, 2019 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-30914462

RESUMO

The crosstalk between vascular pericytes and endothelial cells (ECs) is critical for microvascular stabilization and remodeling; however, the crosstalk is often disrupted by diabetes, leading to severe and even lethal vascular damage. Circular RNAs are a class of endogenous RNAs that regulate several important physiological and pathological processes. Here we show that diabetes-related stress up-regulates cPWWP2A expression in pericytes but not in ECs. In vitro studies show that cPWWP2A directly regulates pericyte biology but indirectly regulates EC biology via exosomes carrying cPWWP2A. cPWWP2A acts as an endogenous miR-579 sponge to sequester and inhibit miR-579 activity, leading to increased expression of angiopoietin 1, occludin, and SIRT1. In vivo studies show that cPWWP2A overexpression or miR-579 inhibition alleviates diabetes mellitus-induced retinal vascular dysfunction. By contrast, inhibition of cPWWP2A-mediated signaling by silencing cPWWP2A or overexpressing miR-579 aggravates retinal vascular dysfunction. Collectively, this study unveils a mechanism by which pericytes and ECs communicate. Intervention of cPWWP2A or miR-579 expression may offer opportunities for treating diabetic microvascular complications.


Assuntos
Comunicação Celular , Retinopatia Diabética/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , MicroRNAs/biossíntese , Pericitos/metabolismo , Transdução de Sinais , Regulação para Cima , Animais , Retinopatia Diabética/patologia , Exossomos/metabolismo , Exossomos/patologia , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Masculino , Camundongos , MicroRNAs/genética , Pericitos/patologia , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia
6.
Biomed Pharmacother ; 111: 548-554, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30597308

RESUMO

Proliferative vitreoretinopathy (PVR) is one of the major challenges in retinal surgery, which occurs in the patient with complex retinal surgery or penetrating eye injury. Circular RNAs (circRNAs) have emerged as important regulators in many biological processes and disease development. However, the characterization and function of circRNAs in PVR remains elusive. In this study, we identified 91 dysregulated circRNAs in the epiretinal membranes (ERMs) of PVR patients. We further investigated the expression pattern of circ_0043144. circ_0043144 was significantly up-regulated in the vitreous samples and the corresponding serum samples of the patients with PVR. circ_0043144 expression was significantly down-regulated after PVR operation. In vitro studies revealed that circ_0043144 was involved in the regulation of the proliferation, migration and secretion ability of ARPE-19 cells, which is critical for ERM formation. Collectively, this study indicates that circRNAs are potential regulators of the pathogenesis of PVR. circ_0043144 is a promising prognostic and diagnostic indicator for PVR diseases.


Assuntos
Perfilação da Expressão Gênica/métodos , RNA/genética , RNA/metabolismo , Vitreorretinopatia Proliferativa/genética , Vitreorretinopatia Proliferativa/metabolismo , Células Cultivadas , Membrana Epirretiniana/genética , Membrana Epirretiniana/metabolismo , Membrana Epirretiniana/patologia , Humanos , RNA Circular , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Vitreorretinopatia Proliferativa/patologia
7.
Theranostics ; 8(12): 3408-3415, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29930739

RESUMO

Glaucoma is a major cause of visual impairment characterized by progressive retinal neurodegeneration. Circular RNAs are a class of endogenous noncoding RNAs that regulate gene expression in eukaryotes. In this study, we investigated the role of cZNF609 in retinal neurodegeneration induced by glaucoma. Methods: qRT-PCR and Sanger sequencing were conducted to detect cZNF609 expression pattern during retinal neurodegeneration. Immunofluorescence staining was conducted to detect the effect of cZNF609 silencing on retinal neurodegeneration in vivo. MTT assay, Ki67 staining, and PI staining were conducted to detect the effect of cZNF609 silencing on retinal glial cells and RGC function in vitro. Bioinformatics analysis, RNA pull-down assays, and in vitro studies were conducted to reveal the mechanism of cZNF609-mediated retinal neurodegeneration. Results: cZNF609 expression was significantly up-regulated during retinal neurodegeneration. cZNF609 silencing reduced retinal reactive gliosis and glial cell activation, and facilitated RGC survival in vivo. cZNF609 silencing directly regulated Müller cell function but indirectly regulated RGC function in vitro. cZNF609 acted as an endogenous miR-615 sponge to sequester and inhibit miR-615 activity, which led to increased METRN. METRN overexpression could partially rescue cZNF609 silencing-mediated inhibitory effects on retinal glial cell proliferation. Conclusion: Intervention of cZNF609 expression is a promising therapeutic strategy for retinal neurodegeneration.


Assuntos
Glaucoma/patologia , Gliose/patologia , MicroRNAs/metabolismo , Doenças Neurodegenerativas/patologia , Retina/patologia , Transativadores/genética , Animais , Proliferação de Células , Células Ependimogliais/fisiologia , Imunofluorescência , Perfilação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/fisiologia , RNA Circular , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de DNA
8.
Cell Death Dis ; 9(5): 540, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29748605

RESUMO

Glaucoma is a neurodegenerative disease characterized by retinal ganglion cell (RGC) loss, optic disc excavation, and progressive visual field loss. Direct or indirect ameliorating retinal neurodegeneration is a promising therapeutic therapy for glaucoma. Circular RNAs (circRNAs) are a class of covalently closed circular RNA transcripts and have emerged as potential regulators in several neurodegenerative diseases. In this study, we show that cZRANB1 expression is significantly upregulated in retinal neurodegeneration induced by glaucoma. cZRANB1 knockdown decreases retinal reactive gliosis, glial cell activation, and facilitates RGC survival in vivo. cZRANB1 knockdown directly regulates Müller cell function and indirectly regulates RGC function in vitro. cZRANB1 acts as miRNA sponge to regulate Müller cell function through cZRANB1/miR-217/RUNX2 network. Intervention of cZRANB1 expression would become an effective strategy for treating retinal neurodegeneration.


Assuntos
Glaucoma/metabolismo , Doenças Neurodegenerativas/metabolismo , RNA/biossíntese , Retina/metabolismo , Regulação para Cima , Animais , Linhagem Celular , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Células Ependimogliais/metabolismo , Células Ependimogliais/patologia , Glaucoma/genética , Glaucoma/patologia , Glaucoma/terapia , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/terapia , RNA/genética , RNA Circular , Ratos , Ratos Sprague-Dawley , Retina/patologia
9.
Oncotarget ; 7(31): 49688-49698, 2016 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-27391072

RESUMO

Although nervous and vascular systems are functionally different, they usually share similar mechanisms for function maintenance. Neurovascular dysfunction has became the pathogenesis of several vascular and nervous disorders. Here we show that long non-coding RNA-MIAT is aberrantly expressed under neurovascular dysfunction condition. MIAT is shown as a regulator of vascular dysfunction, including retinal angiogenesis, corneal angiogenesis, and vascular permeability. MIAT is also shown as a regulator of retinal neurodegeneration under diabetic condition. Mechanistically, MIAT regulates neural and vascular cell function via MIAT/miR-150-5p/VEGF network. The eye is a valuable model to study central nervous system (CNS) disorders. We show that MIAT knockdown leads to cerebral microvascular degeneration, progressive neuronal loss and neurodegeneration, and behavioral deficits in a CNS neurovascular disorder, Alzheimer's disease. MIAT may represent a pharmacological target for treating neurovascular-related disorders.


Assuntos
Neovascularização Patológica , RNA Longo não Codificante/genética , Retina/patologia , Remodelação Vascular , Doença de Alzheimer/patologia , Animais , Encéfalo/patologia , Doenças do Sistema Nervoso Central/patologia , Diabetes Mellitus Experimental/patologia , Modelos Animais de Doenças , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Microcirculação , Doenças Neurodegenerativas/patologia , Neuroglia/patologia , Navegação Espacial
10.
Oncotarget ; 7(28): 44630-44643, 2016 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-27329839

RESUMO

Angiogenesis is important for invasive tumor growth and metastasis. Its inhibition is a promising tactic for limiting tumor progression. Here, we showed that Piezo2 knockdown led to decreased glioma angiogenesis and reduced vascular hyperpermeability. Piezo2 was highly expressed in tumor endothelial cells, and its knockdown suppressed vascular leakage and tumor angiogenesis. In a retinal vasculature development assay, corneal angiogenesis assay and a modified Miles assay, Piezo2 knockdown obviously decreased angiogenesis and vascular hyperpermeability. In vitro assays revealed that Piezo2 knockdown inhibited endothelial cell proliferation, migration, and tube formation. Moreover, In vitro co-culture system assay showed that Piezo2 knockdown in endothelial cells suppressed cell proliferation, migration, and invasion of glioma tumor cells. Piezo2 could regulate glioma angiogenesis via Ca2+/Wnt11/ß-catenin signaling in endothelial cells. Taken together, these studies provide the evidence for Piezo2 as a critical regulator of tumor angiogenesis and vascular permeability.


Assuntos
Permeabilidade Capilar/genética , Glioma/genética , Canais Iônicos/genética , Neovascularização Patológica/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Células Cultivadas , Feminino , Glioma/irrigação sanguínea , Glioma/terapia , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Canais Iônicos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Nus , Neovascularização Patológica/metabolismo , Interferência de RNA , Terapêutica com RNAi
11.
Cell Physiol Biochem ; 38(6): 2207-18, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27184051

RESUMO

BACKGROUND/AIMS: Retinal neurodegeneration is an early event in the pathological process of diabetic retinopathy (DR). Retinal ganglion cell (RGC) injury is an important pathological feature during neurodegenerative process. Protecting RGCs from high glucose-induced injury is a promising strategy for delaying or hindering diabetes mellitus-related retinal neuropathy. This study aims to investigate the role of Nmnat1, an enzyme which catalyzes a key step in the biosynthesis of nicotinamide adenine dinucleotide (NAD), in high glucose-induced RGC injury. METHODS: Western blot and immunofluorescence analysis was conducted to detect Nmnat1 expression pattern in the retina and RGC-5 cell. MTT assay, Hoechst staining, trypan blue staining, and calcein-AM/ propidium iodide (PI) staining was conducted to determine the effect of Nmnat1 knockdown on RGC-5 cell function. Microarray and bioinformatics analysis was conducted to identify potential signaling pathways affected by Nmnat1 knockdown. Pharmacological intervention, molecular intervention, and in vitro experiments were conducted to reveal molecular mechanism of Nmnat1-mediated protective effect on RGC-5 cell function. RESULTS: Nmnat1 is constitutively expressed in retina and RGC-5 cells. Nmnat1 knockdown aggravates RGC injury, and accelerates the development of RGC-5 cell apoptosis upon high glucose stress. MAPK signaling is the primary signaling pathway affected by Nmnat1 knockdown. Under high glucose stress, Nmnat1 knockdown leads to p38-MAPK signaling inactivation. p38-MAPK pathway inhibitor strongly blocks Nmnat1-mediated protective effect on RGC-5 cell function. CONCLUSION: Nmnat1 protects RGC against high glucose-induced injury via p38-MAPK signaling pathway. Nmnat1 may serve as a neuroprotective target for diabetes mellitus-related retinal neuropathy.


Assuntos
Retinopatia Diabética/metabolismo , Glucose/metabolismo , Sistema de Sinalização das MAP Quinases , Nicotinamida-Nucleotídeo Adenililtransferase/metabolismo , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Animais , Linhagem Celular , Sobrevivência Celular , Retinopatia Diabética/genética , Retinopatia Diabética/patologia , Nicotinamida-Nucleotídeo Adenililtransferase/análise , Nicotinamida-Nucleotídeo Adenililtransferase/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Células Ganglionares da Retina/citologia
12.
J Cell Mol Med ; 20(3): 537-48, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26818536

RESUMO

Age-related cataract is among the most common chronic disorders of ageing and is the world's leading blinding disorder. Long non-coding RNAs play important roles in several biological processes and complicated diseases. However, the role of lncRNAs in the setting of cataract is still unknown. Here, we extracted total RNAs from the transparent and age-matched cataractous human lenses, and determined lncRNA expression profiles using microarray analysis. We found that 38 lncRNAs were differentially expressed between transparent and cataractous lenses. 17 of 20 differentially expressed lncRNAs were further verified by quantitative RT-PCRs. One top abundant lncRNA, MIAT, was specifically up-regulated both in the plasma fraction of whole blood and aqueous humor of cataract patients. MIAT knockdown could affect the proliferation, apoptosis and migration of Human lens epithelial cells (HLECs) upon oxidative stress. Posterior capsule opacification (PCO) is a common complication of cataract surgery, which is associated with abnormal production of inflammatory factors. MIAT knockdown could repress tumour necrosis factor-α-induced abnormal proliferation and migration of HLECs, suggesting a potential role of MIAT in PCO-related pathological process. Moreover, we found that MIAT acted as a ceRNA, and formed a feedback loop with Akt and miR-150-5p to regulate HLEC function. Collectively, this study provides a novel insight into the pathogenesis of age-related cataract.


Assuntos
Catarata/metabolismo , Células Epiteliais/fisiologia , Cristalino/patologia , RNA Longo não Codificante/fisiologia , Idoso , Apoptose , Biomarcadores/metabolismo , Catarata/patologia , Linhagem Celular , Movimento Celular , Proliferação de Células , Feminino , Expressão Gênica , Humanos , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas c-akt/metabolismo , Regulação para Cima
13.
Biochem Biophys Res Commun ; 465(3): 324-30, 2015 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-26241674

RESUMO

Proliferative vitreoretinopathy (PVR) is a serious complication of retinal detachment and vitreoretinal surgery, which can lead to severe vision reduction. Long non-coding RNAs (lncRNAs) play critical roles in many biological processes and disease development. We attempted to determine the role of lncRNAs in the setting of PVR. Microarray analysis revealed that 78 lncRNAs were abnormally expressed in the epiretinal membranes (ERMs) of PVR patients, including 48 up-regulated and 30 down-regulated lncRNA transcripts. We subsequently focus on one lncRNA, MALAT1, and investigated its expression pattern in the biofluid of PVR patients. MALAT1 was significantly up-regulated in the cellular and plasma fraction of peripheral blood in PVR patients. MALAT1 expression was obviously reduced after PVR operation. In vitro experiments revealed the role of MALAT1 in regulating RPE proliferation and migration, which is critical for ERMs formation. This study suggests that lncRNAs are the potential regulators of PVR pathology. MALAT1 is a potential prognostic indicator and a target for the diagnosis and gene therapy for PVR diseases.


Assuntos
Predisposição Genética para Doença/genética , RNA Longo não Codificante/sangue , RNA Longo não Codificante/genética , Vitreorretinopatia Proliferativa/sangue , Vitreorretinopatia Proliferativa/genética , Sequência de Bases , Biomarcadores Tumorais/sangue , Movimento Celular/genética , Proliferação de Células/genética , Marcadores Genéticos/genética , Humanos , Dados de Sequência Molecular , Vitreorretinopatia Proliferativa/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA