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

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(11): 6067-6074, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32123064

RESUMEN

Ocular inflammation is a major cause of visual impairment attributed to dysregulation of the immune system. Previously, we have shown that the receptor for growth-hormone-releasing hormone (GHRH-R) affects multiple inflammatory processes. To clarify the pathological roles of GHRH-R in acute ocular inflammation, we investigated the inflammatory cascades mediated by this receptor. In human ciliary epithelial cells, the NF-κB subunit p65 was phosphorylated in response to stimulation with lipopolysaccharide (LPS), resulting in transcriptional up-regulation of GHRH-R. Bioinformatics analysis and coimmunoprecipitation showed that GHRH-R had a direct interaction with JAK2. JAK2, but not JAK1, JAK3, and TYK2, was elevated in ciliary body and iris after treatment with LPS in a rat model of endotoxin-induced uveitis. This elevation augmented the phosphorylation of STAT3 and production of proinflammatory factors, including IL-6, IL-17A, COX2, and iNOS. In explants of iris and ciliary body, the GHRH-R antagonist, MIA-602, suppressed phosphorylation of STAT3 and attenuated expression of downstream proinflammatory factors after LPS treatment. A similar suppression of STAT3 phosphorylation was observed in human ciliary epithelial cells. In vivo studies showed that blocking of the GHRH-R/JAK2/STAT3 axis with the JAK inhibitor Ruxolitinib alleviated partially the LPS-induced acute ocular inflammation by reducing inflammatory cells and protein leakage in the aqueous humor and by repressing expression of STAT3 target genes in rat ciliary body and iris and in human ciliary epithelial cells. Our findings indicate a functional role of the GHRH-R/JAK2/STAT3-signaling axis in acute anterior uveitis and suggest a therapeutic strategy based on treatment with antagonists targeting this signaling pathway.


Asunto(s)
Células Epiteliales/patología , Receptores de Neuropéptido/metabolismo , Receptores de Hormona Reguladora de Hormona Hipofisaria/metabolismo , Transducción de Señal/inmunología , Uveítis/patología , Animales , Línea Celular , Cuerpo Ciliar/citología , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Humanos , Janus Quinasa 2/metabolismo , Lipopolisacáridos/inmunología , Masculino , Nitrilos , Pirazoles/farmacología , Pirazoles/uso terapéutico , Pirimidinas , Ratas , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/inmunología , Receptores de Hormona Reguladora de Hormona Hipofisaria/antagonistas & inhibidores , Receptores de Hormona Reguladora de Hormona Hipofisaria/inmunología , Factor de Transcripción STAT3/metabolismo , Sermorelina/análogos & derivados , Sermorelina/farmacología , Sermorelina/uso terapéutico , Transducción de Señal/efectos de los fármacos , Uveítis/tratamiento farmacológico , Uveítis/inmunología
2.
Exp Eye Res ; 181: 277-284, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30831084

RESUMEN

The receptor for growth hormone-releasing hormone (GHRH-R) has been shown to upregulate specifically in the ciliary and iris epithelial cells and infiltrating cells in the aqueous humor in a rat model of acute anterior uveitis. Treatment with GHRHR-R antagonist alleviates significantly these inflammatory responses. Herein we investigated whether the ciliary and iris epithelial cells can respond directly to lipopolysaccharide (LPS) without the influences of circulating leukocytes to produce inflammatory mediators through a GHRH-R mediated mechanism. In explant cultures of rat ciliary body and iris, LPS caused a substantial increase of GHRH-R in 24 h. Immunohistochemistry showed a localization of TLR4, the receptor for LPS, and an elevated expression of IL-6 and IL-1ß in ciliary and iris epithelial cells after LPS treatment. LPS also elevated the level of IL-1ß, IL-6, and iNOS and increased secretion of IL-1ß and IL-6 from the explants. The GHRH-R antagonist, MIA-602, suppressed the elevated expression of IL-1ß and IL-6, and reduced the release of IL-6. Such effects were not seen for the GHRHR agonist, MR-409. When co-cultured with leukocytes, expression of GHRH-R in the ocular explants was further enhanced during LPS treatment. Our results demonstrate a direct action of LPS on ciliary and iris epithelial cells to produce pro-inflammatory factors through a GHRH-R mediated mechanism, and suggest a role of these epithelial cells, in addition to the resident antigen presenting cells, in immune surveillance of the eye. Infiltrating leukocytes may enhance these inflammatory responses by regulating GHRH-R in ciliary and iris epithelial cells, in addition to their functions of synthesizing proinflammatory cytokines.


Asunto(s)
Humor Acuoso/metabolismo , Cuerpo Ciliar/metabolismo , Citocinas/biosíntesis , Infecciones Bacterianas del Ojo/genética , Regulación de la Expresión Génica , Receptores de Neuropéptido/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Uveítis Anterior/genética , Animales , Cuerpo Ciliar/patología , Modelos Animales de Enfermedad , Infecciones Bacterianas del Ojo/metabolismo , Infecciones Bacterianas del Ojo/patología , Inmunohistoquímica , Iris/metabolismo , Masculino , ARN/genética , Ratas , Ratas Sprague-Dawley , Receptores de Neuropéptido/biosíntesis , Receptores de Hormona Reguladora de Hormona Hipofisaria/biosíntesis , Uveítis Anterior/metabolismo , Uveítis Anterior/patología
3.
Exp Cell Res ; 350(2): 312-317, 2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27919747

RESUMEN

Colorectal cancer (CRC) is a common malignancy, most of which remain unresponsive to chemotherapy. As one of the earliest cytotoxic drugs, methotrexate (MTX) serves as an anti-metabolite and anti-folate chemotherapy for various cancers. Unfortunately, MTX resistance prevents its clinical application in cancer therapy. Thereby, overcoming the drug resistance is an alternative strategy to maximize the therapeutic efficacy of MTX in clinics. Long noncoding RNAs (lncRNAs) have gained widespread attention in recent years. More and more emerging evidences have demonstrated that they play important regulatory roles in various biological activities and disease progression including drug resistance. In the present study, a MTX-resistant colorectal cell line HT-29 (HT-29-R) was developed, which displayed the active proliferation and shortened cell cycle. LncRNA H19 was found to be significantly upregulated in this resistant cell line. Further investigation showed that H19 knockdown sensitized the MTX resistance in HT-29-R cells while its overexpression improved the MTX resistance in the parental cells, suggesting that H19 mediate MTX resistance. The Wnt/ß-catenin signaling was activated in HT-29-R cells, and H19 knockdown suppressed this signaling in the parental cells. In conclusion, H19 mediated MTX resistance via activating Wnt/ß-catenin signaling, which help to develop H19 as a promising therapeutic target for MTX resistant CRC.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Resistencia a Antineoplásicos/genética , ARN Largo no Codificante/genética , Vía de Señalización Wnt , Neoplasias Colorrectales/genética , Células HT29 , Humanos , Metotrexato/farmacología
4.
J Cell Biochem ; 118(3): 530-536, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27585307

RESUMEN

Fulminant hepatic failure is a life-threatening disease which occurs in patients without preexisting liver disease. Nowadays, there is no ideal therapeutic tool in the treatment of fulminant hepatic failure. Recent studies suggested that a novel technology termed CRISPR/Cas9 may be a promising approach for the treatment of fulminant hepatic failure. In this project, we have designed single chimeric guide RNAs specifically targeting the genomic regions of mouse Fas gene. The in vitro and in vivo effects of sgRNAs on the production of Fas protein were examined in cultured mouse cells and in a hydrodynamic injection-based mouse model, respectively. The in vivo delivery of CRISPR/Cas9 could maintain liver homeostasis and protect hepatocytes from Fas-mediated cell apoptosis in the fulminant hepatic failure model. Our study indicates the clinical potential of developing the CRISPR/Cas9 system as a novel therapeutic strategy to rescue Concanavalin-A-induced fulminant hepatic failure in the mouse model. J. Cell. Biochem. 118: 530-536, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Concanavalina A/toxicidad , Marcación de Gen , Fallo Hepático Agudo , Receptor fas/genética , Animales , Apoptosis/genética , Línea Celular , Modelos Animales de Enfermedad , Hepatocitos/metabolismo , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/genética , Fallo Hepático Agudo/metabolismo , Fallo Hepático Agudo/prevención & control , Ratones , Ratones Endogámicos ICR , Receptor fas/metabolismo
5.
J Cell Biochem ; 116(11): 2658-66, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25923408

RESUMEN

In the past decade, miRNA emerges as a vital player in orchestrating gene regulation and maintaining cellular homeostasis. It is well documented that miRNA influences a variety of biological events, including embryogenesis, cell fate decision, and cellular differentiation. Adipogenesis is an organized process of cellular differentiation by which pre-adipocytes differentiate towards mature adipocytes. It has been shown that adipogenesis is tightly modulated by a number of transcription factors such as PPARγ, KLF4, and C/EBPα. However, the molecular mechanisms underlying the missing link between miRNA and adipogenesis-related transcription factors remain elusive. In this study, we unveiled that miR-25, a member of miR-106b-25 cluster, was remarkably downregulated during 3T3-L1 adipogenesis. Restored expression of miR-25 significantly impaired 3T3-L1 adipogenesis and downregulated the expression of serial adipogenesis-related genes. Further experiments presented that ectopic expression of miR-25 did not affect cell proliferation and cell cycle progression. Finally, KLF4 and C/EBPα, two key regulators of adipocyte differentiation, were experimentally identified as bona fide targets for miR-25. These data indicate that miR-25 is a novel negative regulator of adipocyte differentiation and it suppressed 3T3-L1 adipogenesis by targeting KLF4 and C/EBPα, which provides novel insights into the molecular mechanism of miRNA-mediated cellular differentiation.


Asunto(s)
Adipogénesis , Proteínas Potenciadoras de Unión a CCAAT/genética , Factores de Transcripción de Tipo Kruppel/genética , MicroARNs/metabolismo , Células 3T3-L1 , Animales , Diferenciación Celular , Regulación de la Expresión Génica , Factor 4 Similar a Kruppel , Ratones , Regiones Promotoras Genéticas
6.
J Hepatol ; 63(4): 886-95, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26024833

RESUMEN

BACKGROUND & AIMS: Long non-coding RNA Hotair has been considered as a pro-oncogene in multiple cancers. Although there is emerging evidence that reveals its biological function and the association with clinical prognosis, the precise mechanism remains largely elusive. METHODS: We investigated the function and mechanism of Hotair in hepatocellular carcinoma (HCC) cell models and a xenograft mouse model. The regulatory network between miR-218 and Hotair was elucidated by RNA immunoprecipitation and luciferase reporter assays. Finally, the correlation between Hotair, miR-218 and the target gene Bmi-1 were evaluated in 52 paired HCC specimens. RESULTS: In this study, we reported that Hotair negatively regulated miR-218 expression in HCC, which might be mediated through an EZH2-targeting-miR-218-2 promoter regulatory axis. Further investigation revealed that Hotair knockdown dramatically inhibited cell viability and induced G1-phase arrest in vitro and suppressed tumorigenicity in vivo by promoting miR-218 expression. Oncogene Bmi-1 was shown to be a functional target of miR-218, and the main downstream targets signaling, P16(Ink4a) and P14(ARF), were activated in Hotair-suppressed tumorigenesis. In primary human HCC specimens, Hotair and Bmi-1 were concordantly upregulated whereas miR-218 was downregulated in these tissues. Furthermore, Hotair was inversely associated with miR-218 expression and positively correlated with Bmi-1 expression in these clinical tissues. CONCLUSION: Hotair silence activates P16(Ink4a) and P14(ARF) signaling by enhancing miR-218 expression and suppressing Bmi-1 expression, resulting in the suppression of tumorigenesis in HCC.


Asunto(s)
Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas Experimentales/genética , MicroARNs/genética , Proteínas de Neoplasias/genética , Proteínas/genética , ARN Largo no Codificante/genética , Animales , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Humanos , Inmunoprecipitación , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Ratones , MicroARNs/biosíntesis , Proteínas de Neoplasias/biosíntesis , Regiones Promotoras Genéticas , Proteínas/metabolismo , ARN Largo no Codificante/biosíntesis , ARN Neoplásico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética
7.
Apoptosis ; 20(1): 75-82, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25416134

RESUMEN

Traditional Chinese medicine is recently emerged as anti-cancer therapy or adjuvant with reduced side-effects and improved quality of life. In the present study, an active ingredient, 1,6,7-trihydroxyxanthone (THA), derived from Goodyera oblongifolia was found to strongly suppress cell growth and induce apoptosis in liver cancer cells. MicroRNAs are a group of small non-coding RNAs that regulate gene expression at post-transcriptional levels. Our results demonstrated that miR-218 was up-regulated and oncogene Bmi-1 was down-regulated by THA treatment. Further investigation showed that THA-induced-miR-218 up-regulation could lead to activation of tumor suppressor P16(Ink4a) and P14(ARF), the main down-stream targets of Bmi-1. In conclusion, THA might be a potential anti-cancer drug candidate, at least in part, through the activation of miR-218 and suppression of Bmi-1 expression.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Hepáticas , MicroARNs/metabolismo , Orchidaceae/química , Complejo Represivo Polycomb 1/metabolismo , Xantonas/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , MicroARNs/genética , Extractos Vegetales/farmacología , Xantonas/química
8.
RNA Biol ; 11(7): 845-54, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24921656

RESUMEN

The Krüppel like factor 6 (KLF6) gene encodes multiple protein isoforms derived from alternative mRNA splicing, most of which are intimately involved in hepatocarcinogenesis and tumor progression. Recent bioinformatics analysis shows that alternative mRNA splicing of the KLF6 gene produces around 16 alternatively spliced variants with divergent or even opposing functions. Intriguingly, the full-length KLF6 (KLF6-FL) is a tumor suppressor gene frequently inactivated in liver cancer, whereas KLF6 splice variant 1 (KLF6-SV1) is an oncogenic isoform with antagonistic function against KLF6-FL. Compelling evidence indicates that miRNA, the small endogenous non-coding RNA (ncRNA), acts as a vital player in modulating a variety of cellular biological processes through targeting different mRNA regions of protein-coding genes. To identify the potential miRNAs specifically targeting KLF6-FL, we utilized bioinformatics analysis in combination with the luciferase reporter assays and screened out two miRNAs, namely miR-210 and miR-1301, specifically targeted the tumor suppressive KLF6-FL rather than the oncogenic KLF6-SV1. Our in vitro experiments demonstrated that stable expression of KLF6-FL inhibited cell proliferation, migration and angiogenesis while overexpression of miR-1301 promoted cell migration and angiogenesis. Further experiments demonstrated that miR-1301 was highly expressed in liver cancer cell lines as well as clinical specimens and we also identified the potential methylation and histone acetylation for miR-1301 gene. To sum up, our findings unveiled a novel molecular mechanism that specific miRNAs promoted tumorigenesis by targeting the tumor suppressive isoform KLF6-FL rather than its oncogenic isoform KLF6-SV1.


Asunto(s)
Carcinoma Hepatocelular/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Neoplasias Hepáticas/genética , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Células Hep G2 , Humanos , Factor 6 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Neoplasias Hepáticas/metabolismo , Metilación , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas/genética
9.
J Cell Biochem ; 114(12): 2699-707, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23798503

RESUMEN

Although accumulating evidences indicate that miRNA emerge as a vital player in cell growth, development, and differentiation, how they contribute to the process of adipocyte differentiation remains elusive. In the present study, we revealed that the expression level of miR-210 was dramatically upregulated during 3T3-L1 adipogenesis. Ectopic introduction of miR-210 into 3T3-L1 cells promoted terminal differentiation as well as the expression of adipogenic markers. MTT assay showed that miR-210 significantly inhibited cell proliferation whereas the BrdU incorporation assay and flow cytometry analysis showed that miR-210 did not impair G1/S phase transition. Further experiments demonstrated that enhanced expression of miR-210 in 3T3-L1 cells provoked adipocyte differentiation via activation of PI3K/Akt pathway by targeting SHIP1, a negative regulator of PI3K/Akt pathway. Moreover, blockade of endogenous miR-210 during adipogenesis significantly repressed adipocyte differentiation. In summary, we have identified miR-210 as an important positive regulator in adipocyte differentiation through the activation of PI3K/Akt pathway.


Asunto(s)
Adipogénesis/genética , Diferenciación Celular/genética , MicroARNs/genética , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Proliferación Celular , Ratones , Proteína Oncogénica v-akt/genética , Proteína Oncogénica v-akt/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación
10.
Genome Biol ; 20(1): 84, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-31027518

RESUMEN

BACKGROUND: Circular RNAs are a class of regulatory RNA transcripts, which are ubiquitously expressed in eukaryotes. In the current study, we evaluate the function of a novel circRNA derived from the ß-catenin gene locus, circß-catenin. RESULTS: Circß-catenin is predominantly localized in the cytoplasm and displays resistance to RNase-R treatment. We find that circß-catenin is highly expressed in liver cancer tissues when compared to adjacent normal tissues. Silencing of circß-catenin significantly suppresses malignant phenotypes in vitro and in vivo, and knockdown of this circRNA reduces the protein level of ß-catenin without affecting its mRNA level. We show that circß-catenin affects a wide spectrum of Wnt pathway-related genes, and furthermore, circß-catenin produces a novel 370-amino acid ß-catenin isoform that uses the start codon as the linear ß-catenin mRNA transcript and translation is terminated at a new stop codon created by circularization. We find that this novel isoform can stabilize full-length ß-catenin by antagonizing GSK3ß-induced ß-catenin phosphorylation and degradation, leading to activation of the Wnt pathway. CONCLUSIONS: Our findings illustrate a non-canonical function of circRNA in modulating liver cancer cell growth through the Wnt pathway, which can provide novel mechanistic insights into the underlying mechanisms of hepatocellular carcinoma.


Asunto(s)
Carcinoma Hepatocelular/etiología , Neoplasias Hepáticas/etiología , ARN/metabolismo , Vía de Señalización Wnt , beta Catenina/genética , Animales , Carcinogénesis , Línea Celular Tumoral , Movimiento Celular , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Ratones Desnudos , Metástasis de la Neoplasia , ARN Circular
11.
Sci Rep ; 8(1): 429, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29323215

RESUMEN

Inflammation is in a wide spectrum of retinal diseases, causing irreversible blindness and visual impairment. We have previously demonstrated that Green Tea Extract (GTE) is a potent anti-inflammatory agent for anterior uveitis. Here we investigated the anti-inflammatory effect of GTE on lipopolysaccharides (LPS)-induced retinal inflammation in rats and explored the underlying mechanism. Adult rats were injected with LPS and GTE was administered intra-gastrically at 2, 8, 26 and 32 hours post-injection. Staining of whole-mount retina showed that the number of activated microglia cells was significantly increased at 48 hours post-injection, which was suppressed after GTE treatment in a dose-dependent manner. Activation of astrocytes and Müller glia in the retina was also suppressed after GTE treatment. Meanwhile, GTE reduced the expression of pro-inflammatory cytokines including IL-1ß, TNF-α and IL-6 in retina and vitreous humor. These anti-inflammatory effects were associated with a reduced phosphorylation of STAT3 and NF-κB in the retina. Furthermore, the surface receptor of EGCG, 67LR, was localized on the neurons and glia in the retina. These findings demonstrate that GTE is an effective agent in suppressing LPS-induced retinal inflammation, probably through its potent anti-oxidative property and a receptor-mediated action on transcription factors that regulate production of pro-inflammatory cytokines.


Asunto(s)
Antiinflamatorios/administración & dosificación , Lipopolisacáridos/efectos adversos , Extractos Vegetales/administración & dosificación , Retinitis/tratamiento farmacológico , Té/química , Animales , Antiinflamatorios/farmacología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , FN-kappa B/metabolismo , Fosforilación , Extractos Vegetales/farmacología , Ratas , Retinitis/inducido químicamente , Retinitis/inmunología , Factor de Transcripción STAT3/metabolismo
12.
Oncogene ; 37(11): 1445-1456, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29311643

RESUMEN

Emerging evidence indicates that the long noncoding RNAs extensively participate in cancer progression. Nevertheless, the molecular pathogenesis of how these lncRNAs regulate tumorigenesis has not been fully elucidated especially in hepatocellular carcinoma (HCC). Here, we sought to define the role of a novel lncRNA named lncRNA-NEF in modulating epithelial to mesenchymal transition (EMT) in HCC. It was found that the lncRNA-NEF was transcriptionally activated by EMT suppressor FOXA2 and frequently downregulated in HCC cell lines as well as clinical specimens. Although enhanced expression of lncRNA-NEF did not affect tumor cell growth, ectopic expression of lncRNA-NEF significantly suppressed EMT program and cell migration. Animal studies validated that lncRNA-NEF alleviated in vivo tumor metastasis and protected mice from tumor-induced mortality. Interestingly, we verified that lncRNA-NEF acted as a novel activator of its neighbor gene FOXA2, which formed a positive feedback loop. Subsequent studies revealed that lncRNA-NEF physically interacted with ß-catenin to increase the binding of GSK3ß with ß-catenin and therefore promoted the inhibitory phosphorylation of ß-catenin, leading to the suppression on Wnt/ß-catenin signaling and activation of FOXA2 expression. Hence, our findings illustrated a novel feedback loop including FOXA2 and its neighboring gene lncRNA-NEF, which might provide mechanistic insights into the metastatic progress of HCC.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Factor Nuclear 3-beta del Hepatocito/genética , Metástasis de la Neoplasia/genética , ARN Largo no Codificante/fisiología , Vía de Señalización Wnt/genética , Animales , Línea Celular Tumoral , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , beta Catenina/genética , beta Catenina/metabolismo
13.
Oncotarget ; 7(4): 4712-23, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26717040

RESUMEN

Nasopharyngeal carcinoma (NPC), as a unique head and neck cancer type, is particularly prevalent in certain geographic areas such as eastern Asia. Until now, the therapeutic options have been restricted mainly to radiotherapy or chemotherapy. However, the clinical treatment effect remains unsatisfactory even if the combined radio-chemotherapies. Therefore, it is urgently needed to develop effective novel therapies against NPC. In this study, we discovered that lncRNA Hotair was extremely abundant in NPC cells and clinical NPC samples. Further studies showed that Hotair knockdown significantly attenuated both in vitro and in vivo tumor cell growth and angiogenesis. Our study also demonstrated that Hotair promoted angiogenesis through directly activating the transcription of angiogenic factor VEGFA as well as through GRP78-mediated upregulation of VEGFA and Ang2 expression. Therefore, Hotair may serve as a promising diagnostic marker and therapeutic target for NPC patients.


Asunto(s)
Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas Mitocondriales/metabolismo , Neoplasias Nasofaríngeas/irrigación sanguínea , Neovascularización Patológica/genética , ARN Largo no Codificante/genética , Transducción de Señal , Animales , Apoptosis , Western Blotting , Carcinoma , Proliferación Celular , Células Cultivadas , Chaperón BiP del Retículo Endoplásmico , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Desnudos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Estadificación de Neoplasias , Pronóstico , Regiones Promotoras Genéticas/genética , ARN Interferente Pequeño/genética , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Sci Rep ; 6: 20121, 2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26853553

RESUMEN

Bone homeostasis is tightly orchestrated and maintained by the balance between osteoblasts and osteoclasts. Recent studies have greatly expanded our understanding of the molecular mechanisms of cellular differentiation. However, the functional roles of non-coding RNAs particularly lncRNAs in remodeling bone architecture remain elusive. In our study, lncRNA H19 was found to be upregulated during osteogenesis in hMSCs. Stable expression of H19 significantly accelerated in vivo and in vitro osteoblast differentiation. Meanwhile, by using bioinformatic investigations and RIP assays combined with luciferase reporter assays, we demonstrated that H19 functioned as an miRNA sponge for miR-141 and miR-22, both of which were negative regulators of osteogenesis and Wnt/ß-catenin pathway. Further investigations revealed that H19 antagonized the functions of these two miRNAs and led to de-repression of their shared target gene ß-catenin, which eventually activated Wnt/ß-catenin pathway and hence potentiated osteogenesis. In addition, we also identified a novel regulatory feedback loop between H19 and its encoded miR-675-5p. And miR-675-5p was found to directly target H19 and counteracted osteoblast differentiation. To sum up, these observations indicate that the lncRNA H19 modulates Wnt/ß-catenin pathway by acting as a competing endogenous RNA, which may shed light on the functional role of lncRNAs in coordinating osteogenesis.


Asunto(s)
MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Regiones no Traducidas 3' , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Secuencia de Bases , Western Blotting , Diferenciación Celular , Línea Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Genes Reporteros , Humanos , Inmunoprecipitación , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis , ARN Largo no Codificante/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Alineación de Secuencia , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/genética , beta Catenina/antagonistas & inhibidores , beta Catenina/genética , beta Catenina/metabolismo
15.
ACS Chem Neurosci ; 6(2): 211-8, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25437245

RESUMEN

Pimozide is a conventional antipsychotic of the diphenylbutylpiperidine class that has been clinically used for over 30 years. The obvious side effect of this drug is weight gain. However, the mechanism of pimozide-induced weight gain is still unknown. In the present study, we identified pimozide as a novel fatty acid binding protein 4 (FABP4) inhibitor using molecular docking simulation as well as biochemical characterizations. BMS309403, a well-known FABP4 inhibitor, elevated the basal protein levels of PPARγ, therefore stimulating adipogenesis in adipocytes. The present study showed that the inhibitory effect of pimozide on FABP4 promoted adipocyte differentiation with the potency proportional to their propensities for weight gain. These effects in adipogenesis by pimozide may help to explain the weight gain that is frequently observed in patients treated with pimozide.


Asunto(s)
Adipogénesis/efectos de los fármacos , Fármacos del Sistema Nervioso Central/farmacología , Proteínas de Unión a Ácidos Grasos/antagonistas & inhibidores , PPAR gamma/metabolismo , Pimozida/farmacología , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/fisiología , Adipogénesis/fisiología , Anilidas/farmacología , Animales , Compuestos de Bifenilo/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Relación Dosis-Respuesta a Droga , Proteínas de Unión a Ácidos Grasos/metabolismo , Humanos , Ratones , Simulación del Acoplamiento Molecular , PPAR gamma/antagonistas & inhibidores , Pirazoles/farmacología
16.
Oncotarget ; 6(26): 22513-25, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26068968

RESUMEN

Recently, the long non-coding RNA (lncRNA) H19 has been identified as an oncogenic gene in multiple cancer types and elevated expression of H19 was tightly linked to tumorigenesis and cancer progression. However, the molecular basis for this observation has not been characterized in colorectal cancer (CRC) especially during epithelial to mesenchymal transition (EMT) progression. In our studies, H19 was characterized as a novel regulator of EMT in CRC. We found that H19 was highly expressed in mesenchymal-like cancer cells and primary CRC tissues. Stable expression of H19 significantly promotes EMT progression and accelerates in vivo and in vitro tumor growth. Furthermore, by using bioinformatics study and RNA immunoprecipitation combined with luciferase reporter assays, we demonstrated that H19 functioned as a competing endogenous RNA (ceRNA) for miR-138 and miR-200a, antagonized their functions and led to the de-repression of their endogenous targets Vimentin, ZEB1, and ZEB2, all of which were core marker genes for mesenchymal cells. Taken together, these observations imply that the lncRNA H19 modulated the expression of multiple genes involved in EMT by acting as a competing endogenous RNA, which may build up the missing link between the regulatory miRNA network and EMT progression.


Asunto(s)
Neoplasias Colorrectales/genética , Transición Epitelial-Mesenquimal/genética , MicroARNs/genética , ARN Largo no Codificante/genética , Animales , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Células HCT116 , Células HEK293 , Células HT29 , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Regulación hacia Arriba
17.
World J Gastroenterol ; 21(36): 10375-84, 2015 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-26420964

RESUMEN

AIM: To investigate the effects of Recql5 deficiency on liver injury induced by lipopolysaccharide/D-galactosamine (LPS/D-Gal). METHODS: Liver injury was induced in wild type (WT) or Recql5-deficient mice using LPS/D-Gal, and assessed by histological, serum transaminases, and mortality analyses. Hepatocellular apoptosis was quantified by transferase dUTP nick end labeling assay and Western blot analysis of cleaved caspase-3. Liver inflammatory chemokine and cytochrome P450 expression was analyzed by quantitative reverse transcription-PCR. Neutrophil infiltration was evaluated by myeloperoxidase activity. Expression and phosphorylation of ERK, JNK, p65, and H2A.X was determined by Western blot. Oxidative stress was evaluated by measuring malondialdehyde production and nitric oxide synthase, superoxide dismutase, glutathione peroxidase, catalase, and glutathione reductase activity. RESULTS: Following LPS/D-Gal exposure, Recql5-deficient mice exhibited enhanced liver injury, as evidenced by more severe hepatic hemorrhage, higher serum aspartate transaminase and alanine transaminase levels, and lower survival rate. As compared to WT mice, Recql5-deficient mice showed an increased number of apoptotic hepatocytes and higher cleaved caspase-3 levels. Recql5-deficient mice exhibited increased DNA damage, as evidenced by increased γ-H2A.X levels. Inflammatory cytokine levels, neutrophil infiltration, and ERK phosphorylation were also significantly increased in the knockout mice. Additionally, Recql5-deficient mice exhibited increased malondialdehyde production and elevated inducible nitric oxide synthase, superoxide dismutase, glutathione peroxidase, catalase, and glutathione reductase activity, indicative of enhanced oxidative stress. Moreover, CYP450 expression was significantly downregulated in Recql5-deficient mice after LPS/D-Gal treatment. CONCLUSION: Recql5 protects the liver against LPS/D-Gal-induced injury through suppression of hepatocyte apoptosis and oxidative stress and modulation of CYP450 expression.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Galactosamina , Hepatocitos/enzimología , Lipopolisacáridos , Hígado/enzimología , RecQ Helicasas/metabolismo , Animales , Apoptosis , Caspasa 3/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Citocinas/genética , Citocinas/metabolismo , Daño del ADN , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hepatocitos/patología , Mediadores de Inflamación/metabolismo , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Estrés Oxidativo , Fosforilación , RecQ Helicasas/deficiencia , RecQ Helicasas/genética , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
18.
Phytomedicine ; 21(11): 1310-7, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25172795

RESUMEN

PURPOSE: C-X-C chemokine receptor type 4 (CXCR4) signaling has been demonstrated to be involved in cancer invasion and migration; therefore, CXCR4 antagonist can serve as an anti-cancer drug by preventing tumor metastasis. This study aimed to identify the CXCR4 antagonists that could reduce and/or inhibit tumor metastasis from natural products. METHODS AND RESULTS: According to the molecular docking screening, we reported here silibinin as a novel CXCR4 antagonist. Biochemical characterization showed that silibinin blocked chemokine ligand 12 (CXCL12)-induced CXCR4 internalization by competitive binding to CXCR4, therefore inhibiting downstream intracellular signaling. In human breast cancer cells MDA-MB-231, which expresses high levels of CXCR4, inhibition of CXCL12-induced chemomigration can be found under silibinin treatment. Overexpression of CXCL12 sensitized MDA-MB-231 cells to the inhibition of silibinin, which was abolished by CXCR4 knockdown. The inhibition of silibinin was also observed in MCF-7/CXCR4 cells rather than MCF-7 cells that express low level of CXCR4. CONCLUSIONS: Our work demonstrated that silibinin is a novel CXCR4 antagonist that may have potential therapeutic use for prevention of tumor metastasis.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Silimarina/farmacología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Simulación del Acoplamiento Molecular , Transducción de Señal/efectos de los fármacos , Silibina
19.
Artículo en Inglés | MEDLINE | ID: mdl-24302971

RESUMEN

Acetylcholinesterase inhibitors are prominent alternative in current clinical treatment for AD patients. Therefore, there is a continued need to search for novel AChEIs with good clinical efficacy and less side effects. By using our in-house natural product database and AutoDock Vina as a tool in docking study, we have identified twelve phytochemicals (emodin, aloe-emodin, chrysophanol, and rhein in Rhei Radix Et Rhizoma; xanthotoxin, phellopterin, alloisoimperatorin, and imperatorin in Angelicae dahuricae Radix; shikonin, acetylshikonin, isovalerylshikonin, and ß,ß-dimethylacrylshikonin in Arnebiae Radix) as candidates of AChEIs that were not previously reported in the literature. In addition to AChEI activity, a series of cell-based experiments were conducted for the investigation of their neuroprotective activities. We found that acetylshikonin and its derivatives prevented apoptotic cell death induced by hydrogen peroxide in human and rat neuronal SH-SY5Y and PC12 cells at 10 µM. We showed that acetylshikonin exhibited the most potent antiapoptosis activity through the inhibition of the generation of reactive oxygen species as well as protection of the loss of mitochondria membrane potential. Furthermore, we identified for the first time that the upregulation of heme oxygenase 1 by acetylshikonin is a key step mediating its antiapoptotic activity from oxidative stress in SH-SY5Y cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA