Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.654
Filtrar
1.
Arch Endocrinol Metab ; 68: e230097, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38739522

RESUMEN

Objective: This study sought to investigate the regulation of long noncoding RNA (lncRNA) XIST on the microRNA (miR)-101-3p/vascular endothelial growth factor A (VEGFA) axis in neovascularization in diabetic retinopathy (DR). Materials and methods: Serum of patients with DR was extracted for the analysis of XIST, miR-101-3p, and VEGFA expression levels. High glucose (HG)-insulted HRMECs and DR model rats were treated with lentiviral vectors. MTT, transwell, and tube formation assays were performed to evaluate cell viability, migration, and angiogenesis, and ELISA was conducted to detect the levels of inflammatory cytokines. Dual-luciferase reporter, RIP, and RNA pull-down experiments were used to validate the relationships among XIST, miR-101-3p, and VEGFA. Results: XIST and VEGFA were upregulated and miR-101-3p was downregulated in serum from patients with DR. XIST knockdown inhibited proliferation, migration, vessel tube formation, and inflammatory responsein HG-treated HRMECs, whereas the above effects were nullified by miR-101-3p inhibition or VEGFA overexpression. miR-101-3p could bind to XIST and VEGFA. XIST promoted DR development in rats by regulating the miR-101-3p/VEGFA axis. Conclusion: LncRNA XIST promotes VEGFA expression by downregulating miR-101-3p, thereby stimulating angiogenesis and inflammatory response in DR.


Asunto(s)
Retinopatía Diabética , MicroARNs , Neovascularización Patológica , ARN Largo no Codificante , Factor A de Crecimiento Endotelial Vascular , ARN Largo no Codificante/genética , Retinopatía Diabética/genética , Retinopatía Diabética/sangre , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Ratas , Humanos , Masculino , Neovascularización Patológica/genética , Ratas Sprague-Dawley , Femenino , Movimiento Celular/genética , Proliferación Celular/genética , Persona de Mediana Edad , Diabetes Mellitus Experimental
2.
PLoS One ; 19(5): e0303094, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38768222

RESUMEN

In response to the growing number of diabetes cases worldwide, Our study addresses the escalating issue of diabetic eye disease (DED), a significant contributor to vision loss globally, through a pioneering approach. We propose a novel integration of a Genetic Grey Wolf Optimization (G-GWO) algorithm with a Fully Convolutional Encoder-Decoder Network (FCEDN), further enhanced by a Kernel Extreme Learning Machine (KELM) for refined image segmentation and disease classification. This innovative combination leverages the genetic algorithm and grey wolf optimization to boost the FCEDN's efficiency, enabling precise detection of DED stages and differentiation among disease types. Tested across diverse datasets, including IDRiD, DR-HAGIS, and ODIR, our model showcased superior performance, achieving classification accuracies between 98.5% to 98.8%, surpassing existing methods. This advancement sets a new standard in DED detection and offers significant potential for automating fundus image analysis, reducing reliance on manual examination, and improving patient care efficiency. Our findings are crucial to enhancing diagnostic accuracy and patient outcomes in DED management.


Asunto(s)
Algoritmos , Retinopatía Diabética , Aprendizaje Automático , Humanos , Retinopatía Diabética/genética , Retinopatía Diabética/diagnóstico , Procesamiento de Imagen Asistido por Computador/métodos , Redes Neurales de la Computación
3.
Front Endocrinol (Lausanne) ; 15: 1383035, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38752182

RESUMEN

Background: Diabetic retinopathy (DR) is a microvascular complication of diabetes, severely affecting patients' vision and even leading to blindness. The development of DR is influenced by metabolic disturbance and genetic factors, including gene polymorphisms. The research aimed to uncover the causal relationships between blood metabolites and DR. Methods: The two-sample mendelian randomization (MR) analysis was employed to estimate the causality of blood metabolites on DR. The genetic variables for exposure were obtained from the genome-wide association study (GWAS) dataset of 486 blood metabolites, while the genetic predictors for outcomes including all-stage DR (All DR), non-proliferative DR (NPDR) and proliferative DR (PDR) were derived from the FinnGen database. The primary analysis employed inverse variance weighted (IVW) method, and supplementary analyses were performed using MR-Egger, weighted median (WM), simple mode and weighted mode methods. Additionally, MR-Egger intercept test, Cochran's Q test, and leave-one-out analysis were also conducted to guarantee the accuracy and robustness of the results. Subsequently, we replicated the MR analysis using three additional datasets from the FinnGen database and conducted a meta-analysis to determine blood metabolites associated with DR. Finally, reverse MR analysis and metabolic pathway analysis were performed. Results: The study identified 13 blood metabolites associated with All DR, 9 blood metabolites associated with NPDR and 12 blood metabolites associated with PDR. In summary, a total of 21 blood metabolites were identified as having potential causal relationships with DR. Additionally, we identified 4 metabolic pathways that are related to DR. Conclusion: The research revealed a number of blood metabolites and metabolic pathways that are causally associated with DR, which holds significant importance for screening and prevention of DR. However, it is noteworthy that these causal relationships should be validated in larger cohorts and experiments.


Asunto(s)
Retinopatía Diabética , Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Humanos , Retinopatía Diabética/sangre , Retinopatía Diabética/genética , Polimorfismo de Nucleótido Simple
4.
PLoS One ; 19(5): e0303010, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38748682

RESUMEN

Diabetic Retinopathy (DR) is the leading cause of vision loss in working-age adults. The hallmark features of DR include vascular leakage, capillary loss, retinal ischemia, and aberrant neovascularization. Although the pathophysiology is not fully understood, accumulating evidence supports elevated reactive oxygen species associated with increased activity of NADPH oxidase 4 (Nox4) as major drivers of disease progression. Previously, we have shown that Nox4 upregulation in retinal endothelial cells by diabetes leads to increased vascular leakage by an unknown mechanism. Platelet endothelial cell adhesion molecule 1 (PECAM-1) is a cell surface molecule that is highly expressed in endothelial cells and regulates endothelial barrier function. In the present study, using endothelial cell-specific human Nox4 transgenic (TG) mice and endothelial cell-specific Nox4 conditional knockout (cKO) mice, we investigated the impact of Nox4 upregulation on PECAM-1 expression in mouse retinas and brain microvascular endothelial cells (BMECs). Additionally, cultured human retinal endothelial cells (HRECs) transduced with adenovirus overexpressing human Nox4 were used in the study. We found that overexpression of Nox4 increases PECAM-1 mRNA but has no effect on its protein expression in the mouse retina, BMECs, or HRECs. Furthermore, PECAM-1 mRNA and protein expression was unchanged in BMECs isolated from cKO mice compared to wild type (WT) mice with or without 2 months of diabetes. Together, these findings do not support a significant role of Nox4 in the regulation of PECAM-1 expression in the diabetic retina and endothelial cells. Further studies are warranted to elucidate the mechanism of Nox4-induced vascular leakage by investigating other intercellular junctional proteins in endothelial cells and their implications in the pathophysiology of diabetic retinopathy.


Asunto(s)
Retinopatía Diabética , Células Endoteliales , NADPH Oxidasa 4 , Molécula-1 de Adhesión Celular Endotelial de Plaqueta , Regulación hacia Arriba , Animales , NADPH Oxidasa 4/metabolismo , NADPH Oxidasa 4/genética , Retinopatía Diabética/metabolismo , Retinopatía Diabética/genética , Retinopatía Diabética/patología , Ratones , Humanos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Células Endoteliales/metabolismo , Ratones Noqueados , NADPH Oxidasas/metabolismo , NADPH Oxidasas/genética , Retina/metabolismo , Retina/patología , Modelos Animales de Enfermedad , Ratones Transgénicos
5.
Eur J Med Res ; 29(1): 265, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38698486

RESUMEN

Diabetic retinopathy (DR), a leading cause of visual impairment, demands a profound comprehension of its cellular mechanisms to formulate effective therapeutic strategies. Our study presentes a comprehensive single-cell analysis elucidating the intricate landscape of Müller cells within DR, emphasizing their nuanced involvement. Utilizing scRNA-seq data from both Sprague-Dawley rat models and human patients, we delineated distinct Müller cell clusters and their corresponding gene expression profiles. These findings were further validated through differential gene expression analysis utilizing human transcriptomic data. Notably, certain Müller cell clusters displayed upregulation of the Rho gene, implying a phagocytic response to damaged photoreceptors within the DR microenvironment. This phenomenon was consistently observed across species. Additionally, the co-expression patterns of RHO and PDE6G within Müller cell clusters provided compelling evidence supporting their potential role in maintaining retinal integrity during DR. Our results offer novel insights into the cellular dynamics of DR and underscore Müller cells as promising therapeutic targets for preserving vision in retinal disorders induced by diabetes.


Asunto(s)
Retinopatía Diabética , Células Ependimogliales , Ratas Sprague-Dawley , Análisis de la Célula Individual , Retinopatía Diabética/patología , Retinopatía Diabética/genética , Células Ependimogliales/patología , Células Ependimogliales/metabolismo , Análisis de la Célula Individual/métodos , Animales , Humanos , Ratas , Transcriptoma
6.
Front Endocrinol (Lausanne) ; 15: 1340608, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38699385

RESUMEN

Objectives: This study aims to investigate the causal relationship between Alzheimer's Disease (AD) and Diabetic Retinopathy (DR). Methods: Employing Mendelian Randomization (MR), Generalized Summary-data-based Mendelian Randomization (GSMR), and the MR-Steiger test, this study scrutinizes the genetic underpinnings of the hypothesized causal association between AD and DR, as well as its Proliferative DR (PDR) and Non-Proliferative DR (NPDR) subtypes. Comprehensive data from Genome-Wide Association Studies (GWAS) were analyzed, specifically AD data from the Psychiatric Genomics Consortium (71,880 cases/383,378 controls), and DR, PDR, and NPDR data from both the FinnGen consortium (FinnGen release R8, DR: 5,988 cases/314,042 controls; PDR: 8,383 cases/329,756 controls; NPDR: 3,446 cases/314,042 controls) and the IEU OpenGWAS (DR: 14,584 cases/176,010 controls; PDR: 8,681 cases/204,208 controls; NPDR: 2,026 cases/204,208 controls). The study also incorporated Functional Mapping and Annotation (FUMA) for an in-depth analysis of the GWAS results. Results: The MR analyses revealed that genetic susceptibility to AD significantly increases the risk of DR, as evidenced by GWAS data from the FinnGen consortium (OR: 2.5090; 95% confidence interval (CI):1.2102-5.2018, false discovery rate P-value (PFDR)=0.0201; GSMR: bxy=0.8936, bxy_se=0.3759, P=0.0174), NPDR (OR: 2.7455; 95% CI: 1.3178-5.7197, PFDR=0.0166; GSMR: bxy=0.9682, bxy_se=0.3802, P=0.0126), and PDR (OR: 2.3098; 95% CI: 1.2411-4.2986, PFDR=0.0164; GSMR: bxy=0.7962, bxy_se=0.3205, P=0.0129) using DR GWAS from FinnGen consortium. These results were corroborated by DR GWAS datasets from IEU OpenGWAS. The MR-Steiger test confirmed a significant association of all identified instrumental variables (IVs) with AD. While a potential causal effect of DR and its subtypes on AD was identified, the robustness of these results was constrained by a low power value. FUMA analysis identified OARD1, NFYA, TREM1 as shared risk genes between DR and AD, suggesting a potential genetic overlap between these complex diseases. Discussion: This study underscores the contribution of AD to an increased risk of DR, as well as NPDR and PDR subtypes, underscoring the necessity of a holistic approach in the management of patients affected by these conditions.


Asunto(s)
Enfermedad de Alzheimer , Retinopatía Diabética , Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Humanos , Enfermedad de Alzheimer/genética , Factores de Riesgo , Retinopatía Diabética/genética , Retinopatía Diabética/epidemiología , Retinopatía Diabética/etiología , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple
7.
Cell Commun Signal ; 22(1): 275, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755602

RESUMEN

BACKGROUND: Diabetic retinopathy (DR) is a major cause of blindness and is characterized by dysfunction of the retinal microvasculature. Neutrophil stasis, resulting in retinal inflammation and the occlusion of retinal microvessels, is a key mechanism driving DR. These plugging neutrophils subsequently release neutrophil extracellular traps (NETs), which further disrupts the retinal vasculature. Nevertheless, the primary catalyst for NETs extrusion in the retinal microenvironment under diabetic conditions remains unidentified. In recent studies, cellular communication network factor 1 (CCN1) has emerged as a central molecule modulating inflammation in pathological settings. Additionally, our previous research has shed light on the pathogenic role of CCN1 in maintaining endothelial integrity. However, the precise role of CCN1 in microvascular occlusion and its potential interaction with neutrophils in diabetic retinopathy have not yet been investigated. METHODS: We first examined the circulating level of CCN1 and NETs in our study cohort and analyzed related clinical parameters. To further evaluate the effects of CCN1 in vivo, we used recombinant CCN1 protein and CCN1 overexpression for gain-of-function, and CCN1 knockdown for loss-of-function by intravitreal injection in diabetic mice. The underlying mechanisms were further validated on human and mouse primary neutrophils and dHL60 cells. RESULTS: We detected increases in CCN1 and neutrophil elastase in the plasma of DR patients and the retinas of diabetic mice. CCN1 gain-of-function in the retina resulted in neutrophil stasis, NETs extrusion, capillary degeneration, and retinal leakage. Pre-treatment with DNase I to reduce NETs effectively eliminated CCN1-induced retinal leakage. Notably, both CCN1 knockdown and DNase I treatment rescued the retinal leakage in the context of diabetes. In vitro, CCN1 promoted adherence, migration, and NETs extrusion of neutrophils. CONCLUSION: In this study, we uncover that CCN1 contributed to retinal inflammation, vessel occlusion and leakage by recruiting neutrophils and triggering NETs extrusion under diabetic conditions. Notably, manipulating CCN1 was able to hold therapeutic promise for the treatment of diabetic retinopathy.


Asunto(s)
Proteína 61 Rica en Cisteína , Retinopatía Diabética , Trampas Extracelulares , Ratones Endogámicos C57BL , Neutrófilos , Retinopatía Diabética/patología , Retinopatía Diabética/metabolismo , Retinopatía Diabética/genética , Trampas Extracelulares/metabolismo , Animales , Neutrófilos/metabolismo , Humanos , Proteína 61 Rica en Cisteína/metabolismo , Proteína 61 Rica en Cisteína/genética , Ratones , Masculino , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/complicaciones , Retina/patología , Retina/metabolismo , Femenino , Persona de Mediana Edad
8.
J Transl Med ; 22(1): 476, 2024 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-38764010

RESUMEN

BACKGROUND: Diabetic retinopathy (DR) is the leading cause of blinding eye disease among working adults and is primarily attributed to the excessive proliferation of microvessels, which leads to vitreous hemorrhage and retinal traction, thereby significantly impairing patient vision. NSUN2-mediated RNA m5C methylation is implicated in various diseases, and in this investigation, we focused on elucidating the impact of NSUN2 on the regulation of the expression of the downstream gene MUC1, specifically through RNA m5C methylation, on the progression of DR. METHOD: Utilizing Microarray analysis, we examined patient vitreous fluid to pinpoint potential therapeutic targets for DR. Differential expression of NSUN2 was validated through qRT-PCR, Western blot, and immunofluorescence in human tissue, animal tissue, and cell model of DR. The relationship between NSUN2 and DR was explored in vitro and in vivo through gene knockdown and overexpression. Various techniques, such as MeRIP-qPCR and dot blot, were applied to reveal the downstream targets and mechanism of action of NSUN2. RESULTS: The levels of both NSUN2 and RNA m5C methylation were significantly elevated in the DR model. Knockdown of NSUN2 mitigated DR lesion formation both in vitro and in vivo. Mechanistically, NSUN2 promoted MUC1 expression by binding to the RNA m5C reader ALYREF. Knockdown of ALYREF resulted in DR lesion alterations similar to those observed with NSUN2 knockdown. Moreover, MUC1 overexpression successfully reversed a series of DR alterations induced by NSUN2 silencing. CONCLUSIONS: NSUN2 regulates the expression of MUC1 through ALYREF-mediated RNA m5C methylation, thereby regulating the progression of DR and providing a new option for the treatment of DR in the future.


Asunto(s)
Retinopatía Diabética , Progresión de la Enfermedad , Mucina-1 , Retinopatía Diabética/metabolismo , Retinopatía Diabética/genética , Retinopatía Diabética/patología , Humanos , Mucina-1/metabolismo , Mucina-1/genética , Metilación , Animales , Regulación de la Expresión Génica , ARN/metabolismo , Masculino , Metiltransferasas/metabolismo , Metiltransferasas/genética , Técnicas de Silenciamiento del Gen , Ratones Endogámicos C57BL
9.
Mol Biol Rep ; 51(1): 477, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38573426

RESUMEN

BACKGROUND: The retinal pigment epithelium (RPE) is essential for retinal homeostasis. Comprehensively exploring the transcriptional patterns of diabetic human RPE promotes the understanding of diabetic retinopathy (DR). METHODS AND RESULTS: A total of 4125 differentially expressed genes (DEGs) were screened out from the human primary RPE cells subjected to prolonged high glucose (HG). The subsequent bioinformatics analysis is divided into 3 steps. In Step 1, 21 genes were revealed by intersecting the enriched genes from the KEGG, WIKI, and Reactome databases. In Step 2, WGCNA was applied and intersected with the DEGs. Further intersection based on the enrichments with the GO biological processes, GO cellular components, and GO molecular functions databases screened out 12 candidate genes. In Step 3, 13 genes were found to be simultaneously up-regulated in the DEGs and a GEO dataset involving human diabetic retinal tissues. VEGFA and ERN1 were the 2 starred genes finally screened out by overlapping the 3 Steps. CONCLUSION: In this study, multiple genes were identified as crucial in the pathological process of RPE under protracted HG, providing potential candidates for future researches on DR. The current study highlights the importance of RPE in DR pathogenesis.


Asunto(s)
Retinopatía Diabética , Retina , Humanos , Retinopatía Diabética/genética , Células Epiteliales , Pigmentos Retinianos , Glucosa
10.
Transl Vis Sci Technol ; 13(4): 12, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38587436

RESUMEN

Purpose: Circular RNAs (circRNAs) have been verified to participate in multiple biological processes and disease progression. Yet, the role of circRNAs in the pathogenesis of diabetic retinopathy (DR) is still poorly understood and deserves further study. This study aimed to investigate the role of circRNAs in the regulation of high glucose (HG)-induced apoptosis of retinal microvascular endothelial cells (RMECs). Methods: Epiretinal membranes from patients with DR and nondiabetic patients with idiopathic macular epiretinal membrane were collected for this study. The circRNA microarrays were performed using high-throughput sequencing. Hierarchical clustering, functional enrichment, and network regulation analyses were used to analyze the data generated by high-throughput sequencing. Next, RMECs were subjected to HG (25 mM) conditions to induce RMECs apoptosis in vitro. A series of experiments, such as Transwell, the Scratch wound, and tube formation, were conducted to explore the regulatory effect of circRNA on RMECs. Fluorescence in situ hybridization (FISH), immunofluorescence staining, and Western blot were used to study the mechanism underlying circRNA-mediated regulation. Results: A total of 53 differentially expressed circRNAs were found in patients with DR. Among these, hsa_circ_0000880 was significantly upregulated in both the diabetic epiretinal membranes and in an in vitro DR model of HG-treated RMECs. Hsa_circ_0000880 knockout facilitated RMECs vitality and decreased the paracellular permeability of RMECs under hyperglycemia. More importantly, silencing of hsa_circ_0000880 significantly inhibited HG-induced ROS production and RMECs apoptosis. Hsa_circ_0000880 acted as an endogenous sponge for eukaryotic initiation factor 4A-III (EIF4A3). Knockout of hsa_circ_0000880 reversed HG-induced decrease in EIF4A3 protein level. Conclusions: Our findings suggest that hsa_circ_0000880 is a novel circRNA can induce RMECs apoptosis in response to HG conditions by sponging EIF4A3, offering an innovative treatment approach against DR. Translational Relevance: The circRNAs participate in the dysregulation of microvascular endothelial function induced by HG conditions, indicating a promising therapeutic target for DR.


Asunto(s)
Retinopatía Diabética , Membrana Epirretinal , Humanos , Células Endoteliales , ARN Circular/genética , Hibridación Fluorescente in Situ , Retinopatía Diabética/genética , Apoptosis/genética , Glucosa/toxicidad , Factor 4A Eucariótico de Iniciación , ARN Helicasas DEAD-box
11.
Mol Vis ; 30: 167-174, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38601015

RESUMEN

Purpose: To examine whether increased ephrin type-B receptor 1 (EphB1) leads to inflammatory mediators in retinal Müller cells. Methods: Diabetic human and mouse retinal samples were examined for EphB1 protein levels. Rat Müller cells (rMC-1) were grown in culture and treated with EphB1 siRNA or ephrin B1-Fc to explore inflammatory mediators in cells grown in high glucose. An EphB1 overexpression adeno-associated virus (AAV) was used to increase EphB1 in Müller cells in vivo. Ischemia/reperfusion (I/R) was performed on mice treated with the EphB1 overexpression AAV to explore the actions of EphB1 on retinal neuronal changes in vivo. Results: EphB1 protein levels were increased in diabetic human and mouse retinal samples. Knockdown of EphB1 reduced inflammatory mediator levels in Müller cells grown in high glucose. Ephrin B1-Fc increased inflammatory proteins in rMC-1 cells grown in normal and high glucose. Treatment of mice with I/R caused retinal thinning and loss of cell numbers in the ganglion cell layer. This was increased in mice exposed to I/R and treated with the EphB1 overexpressing AAVs. Conclusions: EphB1 is increased in the retinas of diabetic humans and mice and in high glucose-treated Müller cells. This increase leads to inflammatory proteins. EphB1 also enhanced retinal damage in response to I/R. Taken together, inhibition of EphB1 may offer a new therapeutic option for diabetic retinopathy.


Asunto(s)
Retinopatía Diabética , Efrina-B1 , Enfermedades de la Retina , Animales , Humanos , Ratones , Ratas , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Células Ependimogliales/metabolismo , Efrina-B1/genética , Efrina-B1/metabolismo , Glucosa/metabolismo , Mediadores de Inflamación/metabolismo , Retina/metabolismo , Enfermedades de la Retina/metabolismo
12.
Diabetes Res Clin Pract ; 211: 111683, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38663549

RESUMEN

BACKGROUND AND AIMS: Because FTO gene is connected with the risk of obesity, cardiovascular disease and hypertension, as well as type 2 diabetes, we hypothesize that the rs9939609 FTO polymorphism may affect type 1 diabetes (T1D) complications and comorbidities. METHODS: We have investigated the associations of the FTO gene variant with the T1D and its complications and comorbidities, as well as the serum levels of pro- and anti-inflammatory markers and lipid profiles. RESULTS: The key results of our study are as follows: (1) the rs9939609 FTO polymorphism does not predispose individuals to T1D; (2) AA genotype is associated with an increased risk of overweight and obesity, retinopathy, hypertension, dyslipidemia and celiac disease; (3) AT genotype is associated with a decreased risk of retinopathy and celiac disease, whereas TT genotype is connected with decreased risk of dyslipidemia; (4) the FTO rs9939609 polymorphism affects the inflammatory status as well as lipid profile in T1D patients. CONCLUSIONS: Our results, for the first time, comprehensively indicate that the rs9939609 FTO polymorphism could be considered a genetic marker for increased susceptibility to T1D complications and comorbidities as well as suggests importance of FTO-mediated pathways in their etiology.


Asunto(s)
Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Diabetes Mellitus Tipo 1 , Obesidad , Humanos , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Diabetes Mellitus Tipo 1/genética , Femenino , Masculino , Adulto , Obesidad/genética , Proteínas/genética , Dislipidemias/genética , Dislipidemias/epidemiología , Comorbilidad , Persona de Mediana Edad , Predisposición Genética a la Enfermedad , Genotipo , Enfermedad Celíaca/genética , Enfermedad Celíaca/epidemiología , Hipertensión/genética , Hipertensión/epidemiología , Retinopatía Diabética/genética , Retinopatía Diabética/epidemiología , Polimorfismo de Nucleótido Simple , Adulto Joven
13.
Free Radic Biol Med ; 219: 76-87, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38604315

RESUMEN

Diabetic retinopathy (DR) is a highly hazardous and widespread complication of diabetes mellitus (DM). The accumulated reactive oxygen species (ROS) play a central role in DR development. The aim of this research was to examine the impact and mechanisms of mesenchymal stem cell (MSC)-derived small extracellular vesicles (sEV) on regulating ROS and retinal damage in DR. Intravitreal injection of sEV inhibited Cullin3 neddylation, stabilized Nrf2, decreased ROS, reduced retinal inflammation, suppressed Müller gliosis, and mitigated DR. Based on MSC-sEV miRNA sequencing, bioinformatics software, and dual-luciferase reporter assay, miR-143-3p was identified to be the key effector for MSC-sEV's role in regulating neural precursor cell expressed developmentally down-regulated 8 (NEDD8)-mediated neddylation. sEV were able to be internalized by Müller cells. Compared to advanced glycation end-products (AGEs)-induced Müller cells, sEV coculture decreased Cullin3 neddylation, activated Nrf2 signal pathway to combat ROS-induced inflammation. The barrier function of endothelial cells was impaired when endothelial cells were treated with the supernatant of AGEs-induced Müller cells, but was restored when treated with supernatant of AGEs-induced Müller cells cocultured with sEV. The protective effect of sEV was, however, compromised when miR-143-3p was inhibited in sEV. Moreover, the protective efficacy of sEV was diminished when NEDD8 was overexpressed in Müller cells. These findings showed MSC-sEV delivered miR-143-3p to inhibit Cullin3 neddylation, stabilizing Nrf2 to counteract ROS-induced inflammation and reducing vascular leakage. Our findings suggest that MSC-sEV may be a potential nanotherapeutic agent for DR, and that Cullin3 neddylation could be a new target for DR therapy.


Asunto(s)
Proteínas Cullin , Retinopatía Diabética , Vesículas Extracelulares , Células Madre Mesenquimatosas , MicroARNs , Proteína NEDD8 , Factor 2 Relacionado con NF-E2 , Especies Reactivas de Oxígeno , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Retinopatía Diabética/patología , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/genética , MicroARNs/genética , MicroARNs/metabolismo , Animales , Células Madre Mesenquimatosas/metabolismo , Ratones , Proteínas Cullin/metabolismo , Proteínas Cullin/genética , Humanos , Especies Reactivas de Oxígeno/metabolismo , Proteína NEDD8/metabolismo , Proteína NEDD8/genética , Transducción de Señal , Masculino , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/genética , Productos Finales de Glicación Avanzada/metabolismo , Ratones Endogámicos C57BL
14.
Exp Eye Res ; 243: 109912, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38670210

RESUMEN

Diabetic retinopathy (DR), a most common microangiopathy of diabetes, causes vision loss and even blindness. The mechanisms of exosomal lncRNA remain unclear in the development of DR. Here, we first identifed the pro-angiogenic effect of exosomes derived from vitreous humor of proliferative diabetic retinopathy patients, where lncRNA-MIAT was enriched inside. Secondly, lncRNA-MIAT was demonstrated significantly increased in exosomes from high glucose induced human retinal vascular endothelial cell, and can regulate tube formation, migration and proliferation ability to promote angiogenesis in vitro and in vivo. Mechanistically, the pro-angiogenic effect of lncRNA-MIAT was via the lncRNA-MIAT/miR-133a-3p/MMP-X1 axis. The reduced level of lncRNA-MIAT in this axis mitigated the generation of retinal neovascular in mouse model of oxygen-induced retinopathy (OIR), providing crucial evidence for lncRNA-MIAT as a potential clinical target. These findings enhance our understanding of the role of exosomal lncRNA-MIAT in retinal angiogenesis, and propose a promising therapeutic strategy against diabetic retinopathy.


Asunto(s)
Retinopatía Diabética , Exosomas , MicroARNs , ARN Largo no Codificante , Neovascularización Retiniana , Retinopatía Diabética/metabolismo , Retinopatía Diabética/genética , Retinopatía Diabética/patología , ARN Largo no Codificante/genética , Animales , Exosomas/metabolismo , Exosomas/genética , Humanos , MicroARNs/genética , Ratones , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/genética , Neovascularización Retiniana/patología , Ratones Endogámicos C57BL , Proliferación Celular , Masculino , Diabetes Mellitus Experimental , Movimiento Celular , Células Cultivadas , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica
15.
Mol Biol Rep ; 51(1): 583, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38683407

RESUMEN

PURPOSE: Oxidative stress in chronic hyperglycemia could injure the tissues and onset of diabetes-related complications like retinopathy and neuropathy. This study investigates the association between methylenetetrahydrofolate reductase (MTHFR) and glutathione peroxidase (GPx) genetic variants with these complications. METHODS: In this case-control study, 400 individuals, including 100 healthy subjects and 300 patients with type 2 diabetes mellitus (T2DM) in three subgroups: with retinopathy(n = 100), with neuropathy(n = 100), and without complication (n = 100) from West Iran, were studied. MTHFR (rs1801133) and GPx-1 (rs1050450) variants were identified by the PCR-RFLP method. The plasma levels of GPx activity, glutathione, malondialdehyde (MDA), total antioxidant capacity (TAC), and total oxidative stress (TOS) were measured by chemical methods. RESULTS: Higher BMI, TOS and MDA levels were observed in patients with neuropathy compared to other patients and controls. Diabetic patients with neuropathy had lower levels of glutathione (7.8 ± 4.5; P < 0.001), GPx activity (39.5 ± 8.5; P < 0.001), and TAC (703.1 ± 129.1; P = 0.0001) in comparison with other groups. The patients without complication and retinopathic patients had higher plasma levels of glutathione (12.2 ± 2.4; p = 0.02) and TAC (793.4 ± 124.6; P < 0.001), respectively. MTHFR TT genotype significantly correlated with lower levels of TOS (3.5 ± 1.1; P < 0.001) and OSI (0.0050 ± 0.001; P < 0.001). Subjects with the GPx-1 TT genotype had higher levels of MDA (6.8 ± 2.5; P = 0.02) and lower levels of TOS (3.7 ± 1.6; P < 0.001), which is statistically significant. TT genotype of MTHFR was associated with 3.9 fold (95% CI 1.04-4.76; P = 0.0436) increased risk of neuropathy. Also, GPx-1 CT genotype increased the risk of retinopathy [OR = 2.7 (95% CI = 1.38-5.44; P = 0.0039)]. CONCLUSION: The MTHFR TT genotype increased the risk of neuropathy in diabetic patients significantly. The GPx-1 CT genotype is related to increased retinopathy risk among diabetic patients. Both MTHFR and Gpx-1 TT genotypes were associated with higher BMI levels.


Asunto(s)
Diabetes Mellitus Tipo 2 , Neuropatías Diabéticas , Retinopatía Diabética , Predisposición Genética a la Enfermedad , Glutatión Peroxidasa GPX1 , Glutatión Peroxidasa , Metilenotetrahidrofolato Reductasa (NADPH2) , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/complicaciones , Neuropatías Diabéticas/genética , Retinopatía Diabética/genética , Estudios de Asociación Genética , Genotipo , Glutatión Peroxidasa/genética , Irán , Malondialdehído/sangre , Malondialdehído/metabolismo , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Estrés Oxidativo/genética , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo
16.
Sci Rep ; 14(1): 9585, 2024 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-38671284

RESUMEN

This study aimed to investigate the probable existence of a causal relationship between sleep phenotypes and proliferative diabetic retinopathy (PDR). Single nucleotide polymorphisms associated with sleep phenotypes were selected as instrumental variables at the genome-wide significance threshold (P < 5 × 10-8). Inverse-variance weighted was applied as the primary Mendelian randomization (MR) analysis method, and MR Egger regression, weighted median, simple mode, and weighted mode methods were used as complementary analysis methods to estimate the causal association between sleep phenotypes and PDR. Results indicated that genetically predicted sleep phenotypes had no causal effects on PDR risk after Bonferroni correction (P = 0.05/10) [Chronotype: P = 0.143; Daytime napping: P = 0.691; Daytime sleepiness: P = 0.473; Insomnia: P = 0.181; Long sleep duration: P = 0.671; Morning person:P = 0.113; Short sleep duration: P = 0.517; Obstructive sleep apnea: P = 0.091; Sleep duration: P = 0.216; and snoring: P = 0.014]. Meanwhile, there are no reverse causality for genetically predicted PDR on sleep phenotypes [Chronotype: P = 0.100; Daytime napping: P = 0.146; Daytime sleepiness: P = 0.469; Insomnia: P = 0.571; Long sleep duration: P = 0.779; Morning person: P = 0.040; Short sleep duration: P = 0.875; Obstructive sleep apnea: P = 0.628; Sleep duration: P = 0.896; and snoring: P = 0.047]. This study's findings did not support the causal effect of between sleep phenotypes and PDR. Whereas, longitudinal studies can further verify results validation.


Asunto(s)
Retinopatía Diabética , Análisis de la Aleatorización Mendeliana , Fenotipo , Polimorfismo de Nucleótido Simple , Sueño , Humanos , Retinopatía Diabética/genética , Sueño/genética , Factores de Riesgo , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo
17.
Food Funct ; 15(9): 5147-5157, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38682722

RESUMEN

Age-related eye diseases (AREDs), including age-related cataracts (ARCs), age-related macular degeneration (AMD), diabetic retinopathy (DR), and glaucoma, are a leading cause of visual loss globally. This study aimed to explore the effects of dietary water intake on AREDs using Mendelian randomization. In the European population, genome-wide association study (GWAS) summary statistics of water intake and AREDs were obtained from the UK Biobank database and the FinnGen Consortium, respectively. The causal associations between water intake and ARED risks were explored by univariable and multivariable MR analyses, followed by sensitivity analyses to test the robustness of the results and detect potential pleiotropy bias. Water intake was associated with reduced risks of ARCs (odds ratio [OR]: 0.61; 95% confidence interval [CI]: 0.46-0.83; P = 1.44 × 10-3) and DR (OR: 0.52; 95% CI: 0.36-0.76; P = 5.47 × 10-4), and a suggestive reduced risk of AMD (OR: 0.42; 95% CI: 0.20-0.88; P = 2.18 × 10-2). Water intake had no effect on glaucoma (OR: 1.16; 95% CI: 0.72-1.88; P = 0.549). After adjusting confounders, the causal effects of water intake on ARCs and DR persisted. Our study provides evidence of the preventive role of water intake in ARCs and DR from a genetic perspective.


Asunto(s)
Ingestión de Líquidos , Estudio de Asociación del Genoma Completo , Degeneración Macular , Análisis de la Aleatorización Mendeliana , Humanos , Degeneración Macular/genética , Degeneración Macular/epidemiología , Masculino , Femenino , Anciano , Oftalmopatías/genética , Oftalmopatías/epidemiología , Catarata/genética , Catarata/prevención & control , Catarata/epidemiología , Glaucoma/genética , Glaucoma/epidemiología , Persona de Mediana Edad , Retinopatía Diabética/genética , Retinopatía Diabética/epidemiología , Retinopatía Diabética/prevención & control , Polimorfismo de Nucleótido Simple
19.
Nutr Metab Cardiovasc Dis ; 34(5): 1295-1304, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38508994

RESUMEN

BACKGROUND AND AIM: Diabetes retinopathy (DR) is a common microvascular complication of diabetes, and it is the main cause of global vision loss. The current observational research results show that the causal relationship between Vitamin D and DR is still controversial. Therefore, we conducted a Mendelian randomization study to determine the potential causal relationship between serum 25-hydroxyvitamin D 25(OH)D and DR. METHODS AND RESULTS: In this study, we selected aggregated data on serum 25(OH)D levels (GWAS ID: ebi-a-GCST90000615) and DR (GWAS ID: finn-b-DM_RETINOPATHY) from a large-scale GWAS database. Then use MR analysis to evaluate the possible causal relationship between them. We mainly use inverse variance weighted (IVW), supplemented by MR Egger and weighted median methods. Sensitivity analysis is also used to ensure the stability of the results, such as Cochran's Q-test, MR-PRESSO, MR-Egger interception test, and retention method. The MR analysis results showed that there was no significant causal relationship between 25(OH)D and DR (OR = 1.0128, 95%CI=(0.9593,1.0693), P = 0.6447); Similarly, there was no significant causal relationship between DR and serum 25 (OH) D levels (OR = 0.9900, 95% CI=(0.9758,1.0045), P = 0.1771). CONCLUSION: Our study found no significant causal relationship between serum 25(OH)D levels and DR, and vice versa. A larger sample size randomized controlled trial is needed to further reveal its potential causal relationship.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Enfermedades de la Retina , Humanos , Análisis de la Aleatorización Mendeliana , Retinopatía Diabética/diagnóstico , Retinopatía Diabética/epidemiología , Retinopatía Diabética/genética , Vitamina D , Bases de Datos Factuales , Estudio de Asociación del Genoma Completo
20.
Biochem Biophys Res Commun ; 709: 149760, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38554602

RESUMEN

BACKGROUND: Diabetic retinopathy (DR) is a retinal microvascular complication caused by hyperglycemia, which can lead to visual impairment or blindness. Pyroptosis is a type of inflammation-related programmed cell death, activated by caspase-1, resulting in the maturation of IL-1ß and IL-18 and the rupture of the cell membrane. RNA sequencing (RNA-seq) is a high-throughput sequencing technique that reveals the presence and quantity of RNA in the genome at a specific time point, i.e., the transcriptome. RNA-seq can analyze gene expression levels, splicing variants, mutations, fusions, editing and other post-transcriptional modifications, as well as gene expression differences between different samples or conditions. It has been widely used in biological and medical research, clinical diagnosis and new drug development. This study aimed to establish an in vitro model of diabetic retinopathy by culturing human retinal endothelial cells (HREC) with high glucose (30 mmol/L), and to detect their transcriptome expression by RNA-seq, screen for key genes related to pyroptosis, and validate the sequencing results by subsequent experiments. METHODS: We used RNA-seq to detect the transcriptome expression differences between HREC cells cultured with high glucose and control group, and identified differentially expressed genes by GO/KEGG analysis. We constructed a PPI network and determined the key genes by Cytoscape software and CytoHubba plugin. We validated the expression of related factors by Western Blot, qPCR and ELISA. RESULTS: We performed GO and KEGG analysis on the RNA-seq data and found differentially expressed genes. We used Cytoscape and CytoHubba plugin to screen out IRF1 as the key gene, and then detected the expression of IRF1 in HREC under high glucose and control group by Western Blot and qPCR. We found that the expression of Caspase-1, GSDMD and IL-1ß proteins in HREC under high glucose increased, while the expression of these proteins decreased after the inhibition of IRF1 by siRNA. ELISA showed that the secretion of IL-1ß in HREC under high glucose increased, while the inhibition of IRF1 reduced the secretion of IL-1ß. These results indicate that IRF1 plays an important role in DR, and provides a new target and strategy for the prevention and treatment of this disease.


Asunto(s)
Retinopatía Diabética , Factor 1 Regulador del Interferón , Piroptosis , Humanos , Caspasas/metabolismo , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica/métodos , Glucosa/metabolismo , Factor 1 Regulador del Interferón/genética , Piroptosis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA