Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cell Mol Biol Lett ; 28(1): 77, 2023 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-37805473

RESUMEN

BACKGROUND: Hepatic fibrosis is a common consequence of chronic liver diseases without approved antifibrotic therapies. Long noncoding RNAs (lncRNAs) play an important role in various pathophysiological processes. However, the functions of certain lncRNAs involved in mediating the antifibrotic role remain largely unclear. METHODS: The RNA level of lnc-High Expressed in Liver Fibrosis (Helf) was detected in both mouse and human fibrotic livers. Furthermore, lnc-Helf-silenced mice were treated with carbon tetrachloride (CCl4) or bile duct ligation (BDL) to investigate the function of lnc-Helf in liver fibrosis. RESULTS: We found that lnc-Helf has significantly higher expression in human and mouse fibrotic livers as well as M1 polarized hepatic macrophages (HMs) and activated hepatic stellate cells (HSCs). In vivo studies showed that silencing lnc-Helf by AAV8 vector alleviates CCl4- and BDL-induced hepatic inflammation and fibrosis. Furthermore, in vitro experiments revealed that lnc-Helf promotes HSCs activation and proliferation, as well as HMs M1 polarization and proliferation in the absence or presence of cytokine stimulation. Mechanistically, our data illustrated that lnc-Helf interacts with RNA binding protein PTBP1 to promote its interaction with PIK3R5 mRNA, resulting in increased stability and activating the AKT pathway, thus promoting HSCs and HMs activation and proliferation, which augments hepatic inflammation and fibrosis. CONCLUSION: Our results unveil a lnc-Helf/PTBP1/PIK3R5/AKT feedforward, amplifying signaling that exacerbates the process of hepatic inflammation and fibrosis, thus providing a possible therapeutic strategy for hepatic fibrosis.


Asunto(s)
Fosfatidilinositol 3-Quinasa , ARN Largo no Codificante , Animales , Humanos , Ratones , Células Cultivadas , Ribonucleoproteínas Nucleares Heterogéneas/genética , Inflamación , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Proteína de Unión al Tracto de Polipirimidina/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética , Factores de Transcripción/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo
2.
BMC Med ; 20(1): 335, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36171606

RESUMEN

BACKGROUND: Long noncoding RNAs (lncRNAs) have emerged as important regulators in a variety of human diseases. The dysregulation of liver sinusoidal endothelial cell (LSEC) phenotype is a critical early event in the fibrotic process. However, the biological function of lncRNAs in LSEC still remains unclear. METHODS: The expression level of lncRNA Airn was evaluated in both human fibrotic livers and serums, as well as mouse fibrotic livers. Gain- and loss-of-function experiments were performed to detect the effect of Airn on LSEC differentiation and hepatic stellate cell (HSC) activation in liver fibrosis. Furthermore, RIP, RNA pull-down-immunoblotting, and ChIP experiments were performed to explore the underlying mechanisms of Airn. RESULTS: We have identified Airn was significantly upregulated in liver tissues and LSEC of carbon tetrachloride (CCl4)-induced liver fibrosis mouse model. Moreover, the expression of AIRN in fibrotic human liver tissues and serums was remarkably increased compared with healthy controls. In vivo studies showed that Airn deficiency aggravated CCl4- and bile duct ligation (BDL)-induced liver fibrosis, while Airn over-expression by AAV8 alleviated CCl4-induced liver fibrosis. Furthermore, we revealed that Airn maintained LSEC differentiation in vivo and in vitro. Additionally, Airn inhibited HSC activation indirectly by regulating LSEC differentiation and promoted hepatocyte (HC) proliferation by increasing paracrine secretion of Wnt2a and HGF from LSEC. Mechanistically, Airn interacted with EZH2 to maintain LSEC differentiation through KLF2-eNOS-sGC pathway, thereby maintaining HSC quiescence and promoting HC proliferation. CONCLUSIONS: Our work identified that Airn is beneficial to liver fibrosis by maintaining LSEC differentiation and might be a serum biomarker for liver fibrogenesis.


Asunto(s)
ARN Largo no Codificante , Animales , Biomarcadores/metabolismo , Tetracloruro de Carbono/metabolismo , Tetracloruro de Carbono/farmacología , Células Endoteliales/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/farmacología , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/patología , Ratones , ARN Largo no Codificante/genética
3.
Med Oncol ; 39(10): 156, 2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35852638

RESUMEN

To reveal whether STARD5 is a potential biomarker for diagnosis and prognosis of HCC. Using gene expression omnibus and the cancer genome atlas (TCGA) to screen differentially expressed genes in HCC and STARD5 was selected by LASSO algorithm. Then, we analyzed the association between STARD5 and clinical characteristics of HCC patients in TCGA and International Cancer Genome Consortium. Meanwhile, the mRNA and protein level of STARD5 was also verified by collecting 87 cases of HCC patients' liver tissues using qRT-PCR and WB. Next, we applied gene set enrichment analysis (GSEA) for pathways analysis of STARD5. Finally, TIMER1.0 and TISIDB were used to explore the correlation of STARD5 with immune cell infiltration. The expression of STARD5 was lower in HCC and negatively correlated with tumor grade (p < 0.05), while high expression of STARD5 suggested a better prognosis for HCC patients (p < 0.01) and it could be an independent prognostic predictor (p < 0.001). Meanwhile, STARD5 also had strong diagnostic accuracy for HCC patients. GSEA revealed that STARD5-related genes were mainly enriched in E2F targets, G2M checkpoint and KRAS signaling. The TIMER1.0 and TISIDB databases found a negative correlation between STARD5 and tumor immune infiltrating cells. STARD5 could be used as a potential target for HCC diagnosis and prognosis.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/patología , Humanos , Neoplasias Hepáticas/patología , Pronóstico , ARN Mensajero/genética
4.
Theranostics ; 9(25): 7566-7582, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31695787

RESUMEN

Liver fibrosis leading to cirrhosis is one of the major health burdens worldwide with currently limited therapeutic options available. Long noncoding RNAs (lncRNAs) play important roles in various biological and pathological processes in a cell- or tissue-specific manner. However, there is still an important gap in the understanding of the role of hepatocyte-specific lncRNAs in liver fibrosis. Methods: The expressions of lnc-Hser in human and mice fibrotic livers as well as primary hepatocytes (HCs) of mice developing liver fibrosis were determined by real-time RT-PCR. The roles and mechanisms of lnc-Hser in HCs and liver fibrosis were determined in vitro and in vivo. Results: In this study, we have identified a hepatocyte-specifically expressed lnc-Hser, which was reduced in human and mice fibrotic livers as well as primary HCs of mice developing liver fibrosis. We have shown that silencing lnc-Hser aggravated liver fibrosis both in vitro and in vivo through inducing the epithelial-mesenchymal transition (EMT) and the apoptosis of HCs. In addition, knockdown of lnc-Hser promoted hepatic stellate cells (HSCs) activation through the signals derived from injured HCs. Mechanistically, we have revealed that lnc-Hser inhibited HCs apoptosis via the C5AR1-Hippo-YAP pathway and suppressed HCs EMT via the Notch signaling. Conclusions: Our work has identified a hepatocyte-specific lnc-HSER that regulates liver fibrosis, providing a proof that this molecule is a novel biomarker for damaged HCs and a potential target for anti-fibrotic therapy.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Hepatocitos/metabolismo , Cirrosis Hepática/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Apoptosis/fisiología , Células Cultivadas , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/fisiología , Humanos , Hígado/metabolismo , Hígado/fisiología , Ratones , Ratones Endogámicos BALB C , Transducción de Señal/fisiología
5.
Theranostics ; 9(12): 3622-3638, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31281502

RESUMEN

Long non-coding RNAs (lncRNAs) are involved in numerous biological functions and pathological processes. However, the clinical significance of lncRNAs and their functions in liver fibrosis remain largely unclear. Methods: The transcript of lncRNA SCARNA10 in serum and liver samples from patients with advanced hepatic fibrosis, liver tissues from two fibrosis mouse models, and cultured hepatic stellate cells (HSCs) was determined by real-time RT-PCR. The effects of lentivirus-mediated knockdown or over-expression of SCARNA10 in liver fibrosis were examined in vitro and in vivo. Moreover, the effects and mechanisms of down-regulation or over-expression of SCARNA10 on the expression of the genes involved in TGFß pathway were determined. Results: It was found lncRNA ENSMUST00000158992, named as Scarna10, was remarkably up-regulated in mouse fibrotic livers according to the microarray data. We observed that the transcript of SCARNA10 was increased in the serum and liver from patients with advanced hepatic fibrosis. Furthermore, we found that SCARNA10 promoted liver fibrosis both in vitro and in vivo through inducing hepatocytes (HCs) apoptosis and HSCs activation. Mechanistically, RNA immunoprecipitation (RIP) assays demonstrated that SCARNA10 physically associated with polycomb repressive complex 2 (PRC2). Additionally, our results demonstrated that SCARNA10 functioned as a novel positive regulator of TGFß signaling in hepatic fibrogenesis by inhibiting the binding of PRC2 to the promoters of the genes associated with ECM and TGFß pathway, thus promoting the transcription of these genes. Conclusions: Our study identified a crucial role of SCARNA10 in liver fibrosis, providing a proof of this molecule as a potential diagnostic marker and a possible therapeutic target against liver fibrosis.


Asunto(s)
Biomarcadores/análisis , Células Estrelladas Hepáticas/química , Cirrosis Hepática/fisiopatología , Hígado/patología , ARN Largo no Codificante/análisis , ARN Largo no Codificante/metabolismo , Suero/química , Animales , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Cirrosis Hepática/patología , Ratones , Modelos Biológicos , ARN Largo no Codificante/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Biochem Biophys Res Commun ; 516(1): 75-81, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31196627

RESUMEN

Evidence indicates that 1α, 25-dihydroxy vitamin D3 (1, 25-(OH)2D3) markedly reduces intraocular pressure (IOP) in nonhuman primates, while the biochemical mechanisms are unclear. To investigate the influence of oxidative stress on human trabecular meshwork cells (HTMCs) and the effect and regulatory mechanism of 1, 25-(OH)2D3 in HTMCs under oxidative stress, we established an oxidative stress model in HTMCs using hydrogen peroxide (H2O2) and showed that 1, 25-(OH)2D3 could inhibit oxidative stress-induced apoptosis and reduce extracellular matrix (ECM) composition of HTMCs. Moreover, 1, 25-(OH)2D3 could attenuate H2O2-induced inflammation in HTMCs. Mechanistically, our findings revealed that H2O2-induced damage was mediated by the transforming growth factor-ß (TGF-ß)-SMAD3 pathway in HTMCs, and 1, 25-(OH)2D3 could protect HTMCs against oxidative stress through vitamin D receptor (VDR), which antagonises the effects of SMAD3. Overall, these findings define a mechanism by which 1, 25-(OH)2D3 reduces ECM accumulation and suppresses the TGF-ß-SMAD3-VDR pathway in HTMCs, thus protecting the cells from oxidative stress, suggesting 1, 25-(OH)2D3 might be a potential therapeutic for glaucoma.


Asunto(s)
Calcitriol/farmacología , Estrés Oxidativo/efectos de los fármacos , Malla Trabecular/efectos de los fármacos , Vitaminas/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Humanos , Receptores de Calcitriol/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína smad3/metabolismo , Malla Trabecular/citología , Malla Trabecular/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Vitamina D/análogos & derivados , Vitamina D/farmacología
7.
Biochem Biophys Res Commun ; 493(4): 1396-1401, 2017 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-28965947

RESUMEN

Thymosin ß4 (Tß4) has been shown to have beneficial effects in a number of pathological processes. Although there was research reporting the endogenous expression of Tß4 influences hepatic stellate cells (HSCs) activation, the effect of exogenous administration of Tß4 in hepatic fibrosis remains unclear. In the current study, we used CCl4-induced liver fibrosis model mice to investigate the effect of Tß4 administration on fibrosis in vivo and the underlying mechanism. Our study indicates that Tß4 attenuates hepatic fibrosis and down-regulates the expression of fibrogenic genes in hepatic liver. In addition, Tß4 inhibits the expression of pro fibrogenic and proliferation genes in activated HSCs. Further study revealed that Tß4 attenuates liver fibrosis through inhibition of the Notch signaling, as Tß4 significantly reduces the expression of Notch2 and Notch 3 that were increased in hepatic liver. Our data indicate that Tß4 might be an effective anti-fibrotic drug for the treatment of liver fibrosis.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática Experimental/metabolismo , Cirrosis Hepática Experimental/prevención & control , Receptores Notch/metabolismo , Timosina/metabolismo , Animales , Tetracloruro de Carbono/toxicidad , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Expresión Génica , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/patología , Humanos , Cirrosis Hepática Experimental/genética , Masculino , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores Notch/genética , Transducción de Señal/efectos de los fármacos , Timosina/farmacología
8.
Nat Commun ; 8(1): 144, 2017 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-28747678

RESUMEN

Long noncoding RNAs (lncRNAs) play important roles in various biological processes such as proliferation, cell death and differentiation. Here, we show that a liver-enriched lncRNA, named liver fibrosis-associated lncRNA1 (lnc-LFAR1), promotes liver fibrosis. We demonstrate that lnc-LFAR1 silencing impairs hepatic stellate cells (HSCs) activation, reduces TGFß-induced hepatocytes apoptosis in vitro and attenuates both CCl4- and bile duct ligation-induced liver fibrosis in mice. Lnc-LFAR1 promotes the binding of Smad2/3 to TGFßR1 and its phosphorylation in the cytoplasm. Lnc-LFAR1 binds directly to Smad2/3 and promotes transcription of TGFß, Smad2, Smad3, Notch2 and Notch3 which, in turn, results in TGFß and Notch pathway activation. We show that the TGFß1/Smad2/3/lnc-LFAR1 pathway provides a positive feedback loop to increase Smad2/3 response and a novel link connecting TGFß with Notch pathway. Our work identifies a liver-enriched lncRNA that regulates liver fibrogenesis and suggests it as a potential target for fibrosis treatment.Activated hepatic stellate cells are the principal contributors to liver fibrosis by secreting a variety of pro-fibrogenic cytokines . Here Zhang et al. demonstrate that a liver-enriched lncRNA, lnc-LFAR1, promotes liver fibrosis and HSC activation by activating TGFß and Notch signaling.


Asunto(s)
Cirrosis Hepática/genética , Hígado/metabolismo , ARN Largo no Codificante/genética , Receptores Notch/genética , Factor de Crecimiento Transformador beta1/genética , Animales , Apoptosis/genética , Línea Celular , Células Cultivadas , Perfilación de la Expresión Génica/métodos , Células HEK293 , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/metabolismo , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores Notch/metabolismo , Transducción de Señal/genética , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
9.
Biochim Biophys Acta Mol Basis Dis ; 1863(9): 2398-2407, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28502705

RESUMEN

Accumulation of amyloid ß protein (Aß)-containing neuritic plaques in the brain is a neuropathological feature of Alzheimer's disease (AD). The ß-site APP-cleaving enzyme 1 (BACE1) is essential for Aß generation and dysregulation of BACE1 expression may lead to AD pathogenesis. Bcl-2-associated athanogen 1M (BAG-1M), initially identified as an anti-apoptotic protein, has also been found to be highly expressed in the same neurons that contain intracellular amyloid in the hippocampus of AD patient. In this report, we found that over-expression of BAG-1M enhances BACE1-mediated cleavage of amyloid precursor protein (APP) and Aß production by up-regulating BACE1 gene transcription. The regulation of BACE1 transcription by BAG-1M was dependent on NF-κB, as BAG-1M complexes NF-κB at the promoter of BACE1 gene and co-activates NF-κB-facilitated BACE1 transcription. Moreover, expression of BAG-1M by lentiviral vector in the hippocampus of AD transgenic model mice promotes Aß generation and formation of neuritic plaque, and subsequently accelerates memory deficits of the mice. These results provide evidence for an emerging role of BAG-1M in the regulation of BACE1 expression and AD pathogenesis and that targeting the BAG-1M-NF-κB complex may provide a mechanism for inhibiting Aß production and plaque formation.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/biosíntesis , Precursor de Proteína beta-Amiloide/biosíntesis , Ácido Aspártico Endopeptidasas/biosíntesis , Proteínas de Unión al ADN/metabolismo , Trastornos de la Memoria/metabolismo , FN-kappa B/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/genética , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Humanos , Trastornos de la Memoria/genética , Trastornos de la Memoria/patología , Ratones , Ratones Transgénicos , Factores de Transcripción/genética
10.
FEBS J ; 284(7): 1096-1109, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28296235

RESUMEN

The aberrant accumulation of ß-amyloid peptide (Aß) in the brain is a key feature of Alzheimer's disease (AD), and enhanced cleavage of ß-amyloid precursor protein (APP) by ß-site APP-cleaving enzyme 1 (BACE1) has a major causative role in AD. Despite their prominence in AD pathogenesis, the regulation of BACE1 and APP is incompletely understood. In this study, we report that the circular RNA circular RNA sponge for miR-7 (ciRS-7) has an important role in regulating BACE1 and APP protein levels. Previous studies have shown that ciRS-7, which is highly expressed in the human brain, is down-regulated in the brain of people with AD but the relevance of this finding was not clear. We have found that ciRS-7 is not involved in the regulation of APP and BACE1 gene expression, but instead reduces the protein levels of APP and BACE1 by promoting their degradation via the proteasome and lysosome. Consequently, overexpression of ciRS-7 reduces the generation of Aß, indicating a potential neuroprotective role of ciRS-7. Our data also suggest that ciRS-7 modulates APP and BACE1 levels in a nuclear factor-κB (NF-κB)-dependent manner: ciRS-7 expression inhibits translation of NF-κB and induces its cytoplasmic localization, thus derepressing expression of UCHL1, which promotes APP and BACE1 degradation. Additionally, we demonstrated that APP reduces the level of ciRS-7, revealing a mutual regulation of ciRS-7 and APP. Taken together, our data provide a molecular mechanism implicating reduced ciRS-7 expression in AD, suggesting that ciRS-7 may represent a useful target in the development of therapeutic strategies for AD.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , MicroARNs/metabolismo , FN-kappa B/metabolismo , Neuronas/metabolismo , ARN/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Precursor de Proteína beta-Amiloide/genética , Ácido Aspártico Endopeptidasas/genética , Línea Celular Tumoral , Regulación de la Expresión Génica , Genes Reporteros , Células HEK293 , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Lisosomas/metabolismo , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , FN-kappa B/genética , Neuronas/citología , Complejo de la Endopetidasa Proteasomal/metabolismo , Biosíntesis de Proteínas , Proteolisis , ARN/genética , ARN Circular , Transducción de Señal , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo
11.
Oncotarget ; 7(36): 58315-58330, 2016 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-27506947

RESUMEN

Colorectal cancer (CRC) is one of the most common cancers leading to high mortality. However, long-term administration of anti-tumor therapy for CRC is not feasible due to the side effects. Omega-3 polyunsaturated fatty acids (ω-3 PUFAs), particularly DHA and EPA, exert protection against CRC, but the mechanisms are unclear. Here, we show that ω-3 PUFAs inhibit proliferation and induce apoptosis of CRC cells in vitro and alleviate AOM/DSS-induced mice colorectal cancer in vivo. Moreover, ω-3 PUFAs promote phosphorylation and cytoplasmic retention of YAP and this effect was mediated by MST1/2 and LATS1, suggesting that the canonical Hippo Pathway is involved in ω-3 PUFAs function. We further confirmed that increase of pYAP by ω-3 PUFAs was mediated by GPRs, including GPR40 and GPR120, which subsequently activate PKA via Gαs, thus inducing the Hippo pathway activation. These data provide a novel DHA/EPA-GPR40/120-Gαs-PKA-MST1/2-LATS1-YAP signaling pathway which is linked to ω-3 PUFAs-induced inhibition of cell proliferation and promotion of apoptosis in CRC cells, indicating a mechanism that could explain the anti-cancer action of ω-3 PUFAs.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Ácidos Grasos Omega-3/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis , Azoximetano/química , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/terapia , Citoplasma/metabolismo , Sulfato de Dextran/química , Células HT29 , Vía de Señalización Hippo , Humanos , Ratones , Ratones Endogámicos BALB C , Fosfoproteínas/metabolismo , Fosforilación , Transporte de Proteínas , Proteínas Señalizadoras YAP
12.
BMC Ophthalmol ; 16: 116, 2016 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-27435453

RESUMEN

BACKGROUND: Vitamin D deficiency and vitamin D receptor gene polymorphisms are known to be significantly associated with high myopia. Whether this genetic variant may impact primary open-angle glaucoma is largely unknown. This study investigated whether vitamin D receptor gene polymorphisms are altered in primary open-angle glaucoma subjects carrying the risk allele, and whether vitamin D deficiency is an important factor in the development of glaucoma. METHODS: Seventy-three POAG patients and 71 age-matched controls from the Han population were enrolled. Serum levels of 1a, 25-Dihydroxyvitamin D3 were measured by enzyme-linked immunoabsorbent assay. Vitamin D receptor polymorphisms (Cdx-2, Fok I, Bsm I and Taq I) were analyzed using real-time polymerase-chain reaction high resolution melting analysis. RESULTS: Serum levels of 1a, 25-Dihydroxyvitamin in primary open-angle glaucoma patients were lower than in age-matched controls. Statistical analysis revealed a significant difference in the allelic frequencies of the BsmI and TaqI genotypes between primary open-angle glaucoma patients and age-matched controls, while other polymorphisms did not show any significant differences. CONCLUSIONS: Vitamin D deficiency and the presence of the BsmI 'B' allele and the TaqI 't' allele are relevant risk factors in the development of glaucoma. TRIAL REGISTRATION: Clinical Trials.gov: NCT02539745 . The study was registered retrospectively on August 3rd, 2015. The first participant was enrolled on July 4th, 2013.


Asunto(s)
Glaucoma de Ángulo Abierto/genética , Polimorfismo de Nucleótido Simple , Receptores de Calcitriol/genética , Deficiencia de Vitamina D/genética , Anciano , Alelos , Factor de Transcripción CDX2/genética , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes , Genotipo , Glaucoma de Ángulo Abierto/sangre , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Factores de Riesgo , Vitamina D/análogos & derivados , Vitamina D/sangre , Deficiencia de Vitamina D/complicaciones
13.
Sci Rep ; 6: 30029, 2016 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-27435808

RESUMEN

Elevated levels of the transcriptional regulators Yes-associated protein (YAP) and transcriptional coactivators with PDZ-binding motif (TAZ), key effectors of the Hippo pathway, have been shown to play essential roles in controlling liver cell fate and the activation of hepatic stellate cells (HSCs). The dietary intake of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) has been positively associated with a number of health benefits including prevention and reduction of cardiovascular diseases, inflammation and cancers. However, little is known about the impact of ω-3 PUFAs on liver fibrosis. In this study, we used CCl4-induced liver fibrosis mouse model and found that YAP/TAZ is over-expressed in the fibrotic liver and activated HSCs. Fish oil administration to the model mouse attenuates CCl4-induced liver fibrosis. Further study revealed that ω-3 PUFAs down-regulate the expression of pro-fibrogenic genes in activated HSCs and fibrotic liver, and the down-regulation is mediated via YAP, thus identifying YAP as a target of ω-3 PUFAs. Moreover, ω-3 PUFAs promote YAP/TAZ degradation in a proteasome-dependent manner. Our data have identified a mechanism of ω-3 PUFAs in ameliorating liver fibrosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ácidos Grasos Omega-3/administración & dosificación , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/fisiología , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/patología , Fosfoproteínas/metabolismo , Factores de Transcripción/metabolismo , Aciltransferasas , Animales , Proteínas de Ciclo Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Hígado/patología , Masculino , Ratones Endogámicos BALB C , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Resultado del Tratamiento , Proteínas Señalizadoras YAP
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA