Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
Food Funct ; 15(7): 3522-3538, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38465872

RESUMEN

The numerous beneficial probiotic properties of Lactobacillus reuteri (L. reuteri) include decreasing metabolic syndrome, preventing disorders linked to oxidative stress, improving gut flora imbalances, controlling immunological function, and extending life span. Exposure to ionizing radiation is closely associated with several disorders. We examined the protective and salvaging effects of L. reuteri on ionizing radiation-induced injury to the intestinal tract, reproductive system, and nervous system of Drosophila melanogaster. We also examined its effects on lifespan, antioxidant capacity, progeny development, and behavioral aspects to assess the interaction between L. reuteri and ionizing radiation-induced injury. The findings demonstrated that L. reuteri improved the median survival time following irradiation and greatly extended its lifespan. In addition, it raised SOD activity, reduced ROS levels in intestinal epithelial cells, and increased the quantity of intestinal stem cells. Furthermore, L. reuteri enhanced the adult male flies' capacity to move. It also successfully safeguarded the generations' growth and development. L. reuteri dramatically enhanced expression of the AMPKα gene and regulated expression of its pathway-related gene, mTOR, as well as the autophagy-related genes Atg1 and Atg5 in female Drosophila exposed to irradiation. Notably, no prior reports have been made on the possible effects of L. reuteri on injuries caused by irradiation. As a result, our research offers important new information regarding L. reuteri's possible role as a shield against ionizing radiation-induced injury.


Asunto(s)
Limosilactobacillus reuteri , Probióticos , Animales , Masculino , Femenino , Lactobacillus , Drosophila melanogaster , Intestinos , Radiación Ionizante , Probióticos/farmacología
3.
ACS Nano ; 17(14): 14079-14098, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37399352

RESUMEN

Ionizing radiation (IR) is associated with the occurrence of enteritis, and protecting the whole intestine from radiation-induced gut injury remains an unmet clinical need. Circulating extracellular vesicles (EVs) are proven to be vital factors in the establishment of tissue and cell microenvironments. In this study, we aimed to investigate a radioprotective strategy mediated by small EVs (exosomes) in the context of irradiation-induced intestinal injury. We found that exosomes derived from donor mice exposed to total body irradiation (TBI) could protect recipient mice against TBI-induced lethality and alleviate radiation-induced gastrointestinal (GI) tract toxicity. To enhance the protective effect of EVs, profilings of mouse and human exosomal microRNAs (miRNAs) were performed to identify the functional molecule in exosomes. We found that miRNA-142-5p was highly expressed in exosomes from both donor mice exposed to TBI and patients after radiotherapy (RT). Moreover, miR-142 protected intestinal epithelial cells from irradiation-induced apoptosis and death and mediated EV protection against radiation enteritis by ameliorating the intestinal microenvironment. Then, biomodification of EVs was accomplished via enhancing miR-142 expression and intestinal specificity of exosomes, and thus improved EV-mediated protection from radiation enteritis. Our findings provide an effective approach for protecting against GI syndrome in people exposed to irradiation.


Asunto(s)
Enteritis , Exosomas , Vesículas Extracelulares , MicroARNs , Humanos , Vesículas Extracelulares/metabolismo , MicroARNs/metabolismo , Exosomas/metabolismo , Enteritis/metabolismo
4.
Environ Pollut ; 316(Pt 2): 120424, 2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36272602

RESUMEN

The major health risks of dual exposure to two hazardous factors of plastics and radioactive contamination are obscure. In the present study, we systematically evaluated the combinational toxic effects of tetrabromobisphenol A (TBBPA), one of the most influential plastic ingredients, mainly from electronic wastes, and γ-irradiation in zebrafish for the first time. TBBPA (0.25 µg/mL for embryos and larvae, 300 µg/L for adults) contamination aggravated the radiation (6 Gy for embryos and larvae, 20 Gy for adults)-induced early dysplasia and aberrant angiogenesis of embryos, further impaired the locomotor vitality of irradiated larvae, and worsened the radioactive multiorganic histologic injury, neurobehavioural disturbances and dysgenesis of zebrafish adults as well as the inter-generational neurotoxicity in offspring. TBBPA exaggerated the radiative toxic effects not only by enhancing the inflammatory and apoptotic response but also by further unbalancing the endocrine system and disrupting the underlying gene expression profiles. In conclusion, TBBPA exacerbates radiation-induced injury in zebrafish, including embryos, larvae, adults and even the next generation. Our findings provide new insights into the toxicology of TBBPA and γ-irradiation, shedding light on the severity of cocontamination of MP components and radioactive substances and thereby inspiring novel remediation and rehabilitation strategies for radiation-injured aqueous organisms and radiotherapy patients.


Asunto(s)
Retardadores de Llama , Bifenilos Polibrominados , Animales , Pez Cebra/metabolismo , Embrión no Mamífero , Retardadores de Llama/toxicidad , Retardadores de Llama/metabolismo , Bifenilos Polibrominados/toxicidad , Bifenilos Polibrominados/metabolismo , Larva
5.
Mol Biol Rep ; 50(2): 1829-1843, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36507968

RESUMEN

Radiation therapy is a commonly used tool in cancer management due to its ability to destroy malignant tumors. Mechanically, the efficacy of radiotherapy mainly depends on the inherent radiosensitivity of cancer cells and surrounding normal tissues, which mostly accounts for molecular dynamics associated with radiation-induced DNA damage. However, the relationship between radiosensitivity and DNA damage mechanism deserves to be further probed. As the well-established RNA regulators or effectors, long noncoding RNAs (lncRNAs) dominate vital roles in modulating ionizing radiation response by targeting crucial molecular pathways, including DNA damage repair. Recently, emerging evidence has constantly confirmed that overexpression or inhibition of lncRNAs can greatly influence the sensitivity of radiotherapy for many kinds of cancers, by driving a diverse array of DNA damage-associated signaling cascades. In conclusion, this review critically summarizes the recent progress in the molecular mechanism of IR-responsive lncRNAs in the context of radiation-induced DNA damage. The different response of lncRNAs when IR exposure. IR exposure can trigger the changes in expression pattern and subcellular localization of lncRNAs that influences the different radiology processes.


Asunto(s)
Daño del ADN , Reparación del ADN , Neoplasias , ARN Largo no Codificante , Traumatismos por Radiación , Tolerancia a Radiación , Humanos , Daño del ADN/genética , Daño del ADN/efectos de la radiación , Reparación del ADN/genética , Reparación del ADN/efectos de la radiación , Neoplasias/genética , Neoplasias/radioterapia , Neoplasias/metabolismo , Tolerancia a Radiación/genética , Tolerancia a Radiación/efectos de la radiación , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Radioterapia/efectos adversos , Traumatismos por Radiación/complicaciones , Traumatismos por Radiación/genética
6.
Thorac Cancer ; 13(10): 1441-1448, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35388633

RESUMEN

BACKGROUND: To identify radio-responsive genes and explore the biological function of encoded proteins in non-small cell lung cancer (NSCLC). METHODS: Radio-responsive genes in irradiated H460 cells were screened from microarray data deposited in the Gene Expression Omnibus (GEO) database. A quantitative real time polymerase chain reaction assay was used to detect the expression of candidate radio-responsive genes in irradiated cells. CCK-8 assay, EDU assay, clone formation assay, immunofluorescence and flow cytometry were conducted to evaluate the biological function of B cell translocation gene 2 (BTG2) in NSCLC. RESULTS: Bioinformatic analysis using GES20549 showed that BTG2 was a radio-responsive gene in irradiated H460 cells. The mRNA expression level of BTG2 was lower in H460 cells compared with that in BEAS-2B normal lung epithelial cells. BTG2 expression was elevated upon IR exposure, in a dose-dependent but not a time-dependent manner. CCK-8 and EDU assays revealed that BTG2 overexpression inhibited the growth rate of irradiated cells. Clone formation showed that elevated BTG2 promoted DNA damage of irradiated H460 cells. The number of γ-H2AX foci induced by DNA damage was also markedly increased upon BTG2 overexpression. Flow cytometry showed that BTG2 increased IR-induced cell apoptosis. CONCLUSIONS: BTG2 may be a novel radio-responsive factor and a promising therapeutic target for radiotherapy of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Proteínas Inmediatas-Precoces , Neoplasias Pulmonares , Proteínas Supresoras de Tumor , Apoptosis/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/radioterapia , Tolerancia a Radiación/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
7.
Thorac Cancer ; 13(9): 1361-1368, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35429143

RESUMEN

BACKGROUND: Radiotherapy is an effective therapeutic approach widely used clinically in non-small cell lung cancer (NSCLC), but radioresistance remains a major challenge. New and effective radiosensitizing approaches are thus urgently needed. The activation of DNA-sensing cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway has become an attractive therapeutic target, but the relationship between activation of cGAS-STING pathway and radiosensitization of NSCLC cells remains unknown. METHODS: Considering low expression of cGAS-STING pathway genes in NSCLC, including STING, we used an activator (STING agonist, dimeric amidobenzimidazole [diABZI]) of cGAS-STING pathway and increased activation factor (DNA double strand breaks) of cGAS-STING pathway to respectively reinforce the activation of cGAS-STING pathway in NSCLC cells. We then investigated the effect of increased activation of cGAS-STING pathway on the proliferation of H460 and A549 cells by CCK-8 and colony formation assays, and revealed the underlying mechanism. RESULTS: We found that both diABZI and the increased DNA double strand breaks could sensitize NSCLC cells to irradiation. Mechanically, our results showed that the increased activation of cGAS-STING pathway enhanced radiosensitivity by promoting apoptosis in NSCLC cells. CONCLUSION: Taken together, we concluded that diABZI could be used as a radiosensitizer in NSCLC cells, and targeting the activation of cGAS-STING pathway has a potential to be a new approach for NSCLC radiosensitizing.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , ADN/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/radioterapia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Tolerancia a Radiación , Transducción de Señal
8.
Carcinogenesis ; 43(3): 288-296, 2022 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-34888652

RESUMEN

Radioresistance is a major problem encountered in the treatment of non-small cell lung cancer (NSCLC). Aberrant microRNA (miRNA) expression contributes to multiple cancer-associated signaling pathways and profoundly influences effects of radiotherapy (RT) in cancers. MicroRNA-145-5p (miR-145) is recognized as a tumor suppresser in NSCLC. However, the roles of miR-145 during radiotherapy of NSCLC are largely unknown. The present study aimed to investigate the function and underlying mechanism of miR-145 in modulation of radiosensitivity in NSCLC. We generated radioresistant H460 and A549 subclones, named H460R and A549R, respectively, and found that irradiation (IR) could suppress the expression levels of miR-145 in radioresistant NSCLC cells. Furthermore, overexpression of miR-145 could sensitize radioresistant NSCLC cells to IR, whereas knockdown of miR-145 in NSCLC cells acted the converse manner. Mechanically, miR-145 was able to directly target 3'UTR of tropomodulin 3 (TMOD3) mRNA and decrease the expression of TMOD3 at the levels of mRNA and protein. Additionally, we confirmed that miR-145 could enhance the radiosensitivity of radioresistant NSCLC cells by targeting TMOD3 in vitro and in vivo, and could be used as a target in clinical treatment of NSCLC. Collectively, restoration of miR-145 expression increases the radiosensitivity of radioresistant NSCLC cells by suppression of TMOD3, and miR-145 can act as a new radiosensitizer for NSCLC therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , MicroARNs , Regiones no Traducidas 3'/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/radioterapia , MicroARNs/genética , MicroARNs/metabolismo , Tolerancia a Radiación/genética
9.
Acta Pharmacol Sin ; 42(7): 1111-1123, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33637947

RESUMEN

Ionizing radiation-induced intestinal injury is a catastrophic complication in patients receiving radiotherapy. Circulating exosomes from patients undergoing radiotherapy can mediate communication between cells and facilitate a variety of pathological processes in vivo, but its effects on ionizing radiation-induced intestinal damage are undetermined. In this study we investigated the roles of exosomes during total body irradiation (TBI)-induced intestinal injury in vivo and in vitro. We isolated exosomes from serum of donor mice 24 h after lethal dose (9 Gy) TBI (Exo-IR-24h), then intravenously injected the exosomes into receipt mice, and found that Exo-IR-24h injection not only exacerbated 9 Gy TBI-induced lethality and weight loss, but also promoted crypt-villus structural and functional injury of the small intestine in receipt mice. Moreover, Exo-IR-24h injection significantly enhanced the apoptosis and DNA damage of small intestine in receipt mice following TBI exposure. In murine intestinal epithelial MODE-K cells, treatment with Exo-IR-24h significantly promoted 4 Gy ionizing radiation-induced apoptosis, resulting in decreased cell vitality. We further demonstrated that Exo-IR-24h promoted the IR-induced injury in receipt mice partially through its DNA damage-promoting effects and attenuating Nrf2 antioxidant response in irradiated MODE-K cells. In addition, TBI-related miRNAs and their targets in the exosomes of mice were enriched functionally using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Finally, injection of GW4869 (an inhibitor of exosome biogenesis and release, 1.25 mg·kg-1·d-1, ip, for 5 consecutive days starting 3 days before radiation exposure) was able to rescue mice against 9 Gy TBI-induced lethality and intestinal damage. Collectively, this study reveals that exosomes are involved in TBI-induced intestinal injury in mice and provides a new target to protect patients against irradiation-induced intestinal injury during radiotherapy.


Asunto(s)
Exosomas/metabolismo , Enfermedades Intestinales/metabolismo , Mucosa Intestinal/metabolismo , Animales , Apoptosis/fisiología , Proliferación Celular/fisiología , Daño del ADN/fisiología , Rayos gamma , Enfermedades Intestinales/patología , Mucosa Intestinal/patología , Intestino Delgado/metabolismo , Intestino Delgado/patología , Masculino , Ratones Endogámicos BALB C , MicroARNs/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Traumatismos Experimentales por Radiación , Irradiación Corporal Total
10.
Cell Commun Signal ; 18(1): 152, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32943060

RESUMEN

INTRODUCTION: Radioresistance is a major challenge in lung cancer radiotherapy, and new radiosensitizers are urgently needed. Estrogen receptor ß (ERß) is involved in the progression of non-small cell lung cancer (NSCLC), however, the role of ERß in the response to radiotherapy in lung cancer remains elusive. In the present study, we investigated the mechanism underlying ERß-mediated transcriptional activation and radioresistance of NSCLC cells. METHODS: Quantitative real-time PCR, western blot and immunohistochemistry were used to detect the expression of CLPTM1L, ERß and other target genes. The mechanism of CLPTM1L in modulation of radiosensitivity was investigated by chromatin immunoprecipitation assay, luciferase reporter gene assay, immunofluorescence staining, confocal microscopy, coimmunoprecipitation and GST pull-down assays. The functional role of CLPTM1L was detected by function assays in vitro and in vivo. RESULTS: CLPTM1L expression was negatively correlated with the radiosensitivity of NSCLC cell lines, and irradiation upregulated CLPTM1L in radioresistant (A549) but not in radiosensitive (H460) NSCLC cells. Meanwhile, IR induced the translocation of CLPTM1L from the cytoplasm into the nucleus in NSCLC cells. Moreover, CLPTM1L induced radioresistance in NSCLC cells. iTRAQ-based analysis and cDNA microarray identified irradiation-related genes commonly targeted by CLPTM1L and ERß, and CLPTM1L upregulated ERß-induced genes CDC25A, c-Jun, and BCL2. Mechanistically, CLPTM1L coactivated ERß by directly interacting with ERß through the LXXLL NR (nuclear receptor)-binding motif. Functionally, ERß silencing was sufficient to block CLPTM1L-enhanced radioresistance of NSCLC cells in vitro. CLPTM1L shRNA treatment in combination with irradiation significantly inhibited cancer cell growth in NSCLC xenograft tumors in vivo. CONCLUSIONS: The present results indicate that CLPTM1L acts as a critical coactivator of ERß to promote the transcription of its target genes and induce radioresistance of NSCLC cells, suggesting a new target for radiosensitization in NSCLC therapy. Video Abstract.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Receptor beta de Estrógeno/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/radioterapia , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Apoptosis/efectos de la radiación , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Receptor beta de Estrógeno/genética , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Neoplasias Pulmonares/genética , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Tolerancia a Radiación , Transducción de Señal/efectos de la radiación
11.
Dose Response ; 18(2): 1559325820931252, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32684870

RESUMEN

Radiotherapy is mainly a traditional treatment for breast cancer; however, the key genes and pathways in breast cancer associated with irradiation are not clear. In this study, we aimed to explore the messenger RNA expression changes between preradiation and postradiation breast cancer. The gene expression data set (GSE59733) was downloaded from Gene Expression Omnibus database. According to |log2FC (fold change) | ≥ 1 and with false discovery rate adjusted P value <.05, differentially expressed genes (DEGs) were screened and annotated by R programming software. The protein-protein interaction (PPI) network was conducted through STRING database, and subnetworks and hub genes were extracted by plug-in in Cytoscape. A total of 82 DEGs (74 upregulated and 8 downregulated genes) were identified. These DEGs mainly enriched in an intrinsic apoptotic signaling pathway and G-protein-coupled receptor binding. What's more, tumor necrosis factor signaling pathway and interleukin 17 signaling pathway abnormally activated in postradiation tumor samples. Two characteristic subnetworks and 3 hub genes (FOS, CCL2, and CXCL12) were strongly distinguished in PPI network. Moreover, the expression level of the hub genes was confirmed in irradiated MCF-7 cell and SUM-159 cell using quantitative real-time polymerase chain reaction assay. These findings imply that these hub genes may play momentous function in breast cancer to irradiation.

12.
Sheng Wu Gong Cheng Xue Bao ; 36(12): 2868-2876, 2020 Dec 25.
Artículo en Chino | MEDLINE | ID: mdl-33398979

RESUMEN

Tissue inhibitor of metalloproteinases-2 (TIMP-2) inhibits tumor migration and invasion. Obtaining TIMP-2 protein is conducive to a comprehensive and in-depth study of its function and mechanism in tumorigenesis and development. We collected human TIMP-2 protein through prokaryotic expression in vitro. We expressed, purified and characterized human TIMP-2 protein. First, the human TIMP-2 gene was cloned from the cDNA obtained by reverse transcription of total RNA of human lung cancer A549 cells, and constructed to pET28a vector. The recombinant plasmid pET28a-TIMP-2 was transformed into Escherichia coli BL21(DE3) after restriction endonuclease digestion and sequencing analysis. The expression of TIMP-2 protein was induced by isopropyl-ß-D-thiogalactoside (IPTG), and the expression conditions were optimized. After purification by nickel affinity column, the fusion protein His-TIMP-2 was identified by Western blotting method and its biological activity was detected by gelatin zymography. The fusion protein His-TIMP-2 existed in the form of inclusion body in E. coli. In a certain range, the concentration of IPTG had no significant effect on the expression amount of His-TIMP-2. But in this expression system, induction temperature and time were the key parameters, and the expression amount of His-TIMP-2 in E. coli increased with the increase of induction temperature. The purified and refolded fusion protein could effectively inhibit the activity of matrix metalloproteinases expressed by human lung cancer A549 cells. The acquisition of active fusion protein lays a foundation for further study of the function and mechanism of human TIMP-2, and is of great significance for tumor therapy.


Asunto(s)
Escherichia coli , Inhibidor Tisular de Metaloproteinasa-2 , Clonación Molecular , Escherichia coli/genética , Humanos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes , Inhibidor Tisular de Metaloproteinasa-2/genética
13.
Cancer Biol Med ; 16(2): 276-287, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31516748

RESUMEN

OBJECTIVE: The aim of this study was to investigate the underlying mechanism whereby HBx modulates the targeting of NUSAP1 by miR-18b to enhance hepatocarcinogenesis. METHODS: We employed an integrated approach of bioinformatics analysis and molecular experiments in hepatoma cells, HBV transgenic mice, and clinical liver cancer tissues to investigate the role of HBx-regulated miR-18b in the development of liver cancer. RESULTS: In this study, we report that the HBx-mediated tumor suppressor miR-18b modulates hepatocarcinogenesis during the host-HBV interaction. The expression levels of miR-18b were lower in clinical HBV-positive liver cancer tissues and liver tissues of HBV-transgenic mice. Interestingly, HBx inhibited miR-18b expression by inducing the methylation of CpG islands in its promoter. Accordingly, we tested the hypothesis that HBx enhanced hepatocarcinogenesis by increasing the expression of target genes of miR-18b. Moreover, we identified nucleolar spindle-associated protein 1 (NUSAP1) as one of the target genes of miR-18b. NUSAP1 was expressed at high levels in liver cancer tissues. Interestingly, HBx up-regulated NUSAP1 by suppressing miR-18b. Functionally, miR-18b significantly inhibited the proliferation of hepatoma cells by depressing NUSAP1 levels in vivo and in vitro. CONCLUSIONS: Thus, we conclude that the targeting of NUSAP1 mRNA by the tumor suppressor miR-18b is controlled by HBx-modulated promoter methylation during the host-virus interaction, leading to hepatocarcinogenesis. Our findings provide new insights into the mechanism by which HBx-mediated miRNAs modulate hepatocarcinogenesis.

14.
Biochem Biophys Res Commun ; 512(2): 392-398, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-30902389

RESUMEN

Radioresistance is a major challenge in lung cancer radiotherapy (RT), and consequently, new radiosensitizers are urgently needed. MicroRNAs (miRNAs) have been demonstrated to participate in many important cellular processes including radiosensitization. MiR-365 is dysregulated in non-small cell lung cancer (NSCLC) and is able to restrain the development of NSCLC. However, the relationship between miR-365 and radiosensitivities of NSCLC cells remains largely unknown. Here we reveal that overexpression of miR-365 is able to enhance the radiosensitivity of NSCLC cells through targeting CDC25A. We found that the expression level of miR-365 was positively correlated with the radiosensitivity of NSCLC cell lines. Furthermore, our results showed that overexpression of miR-365 could sensitize A549 cells to the irradiation. However, knockdown of miR-365 in H460 cells could act the converse manner. Mechanically, miR-365 was able to directly target 3'UTR of cell division cycle 25A (CDC25A) mRNA and reduce the expression of CDC25A at the levels of mRNA and protein. And we confirmed that miR-365 could increase the radiosensitivity of NSCLC cells by targeting CDC25A using in vitro and in vivo assays. Taken together, restoration of miR-365 expression enhances the radiosensitivity of NSCLC cells by suppressing CDC25A, and miR-365 could be used as a radiosensitizer for NSCLC therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/radioterapia , MicroARNs/genética , Tolerancia a Radiación/genética , Fosfatasas cdc25/antagonistas & inhibidores , Fosfatasas cdc25/genética , Regiones no Traducidas 3' , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/metabolismo , Fármacos Sensibilizantes a Radiaciones/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Fosfatasas cdc25/metabolismo
15.
Mol Carcinog ; 57(7): 926-935, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29573465

RESUMEN

Irradiation exposure positive correlates with tumor formation, such as breast cancer and lung cancer. However, whether low dose irradiation induces hepatocarcinogenesis and the underlying mechanism remain poorly defined. In the present study, we reported that low dose irradiation facilitated the proliferation of hepatocyte through up-regulating HULC in vitro and in vivo. Low dose irradiation exposure elevated HULC expression level in hepatocyte. Deletion of heightened HULC erased the cells growth accelerated following low dose irradiation exposure. CDKN1, the neighbor gene of HULC, was down-regulated by overexpression of HULC following low dose irradiation exposure via complementary base pairing, resulting in promoting cell cycle process. Thus, our findings provide new insights into the mechanism of low dose irradiation-induced hepatocarcinogenesis through HULC/CDKN1 signaling, and shed light on the potential risk of low dose irradiation for the development of hepatocellular carcinoma in pre-clinical settings.


Asunto(s)
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , ARN Largo no Codificante/genética , Animales , Carcinogénesis/genética , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica/genética , Células Hep G2 , Humanos , Ratones , Ratones Desnudos , Dosis de Radiación , Regulación hacia Arriba/genética
16.
Toxicol Lett ; 287: 23-30, 2018 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-29391279

RESUMEN

Excessive alcohol consumption remains a major public health problem that affects millions of people worldwide. Accumulative experimental evidence has suggested an important involvement of gut microbiota in the modulation of host's immunological and neurological functions. However, it is previously unknown whether enteric microbiota is implicated in the formation of alcohol withdrawal-induced anxiety. Using a murine model of chronic alcoholism and withdrawal, we examined the impact of alcohol consumption on the possible alterations of gut microbiota as well as alcohol withdrawal-induced anxiety and behavior changes. The 16S rRNA sequencing revealed that alcohol consumption did not alter the abundance of bacteria, but markedly changed the composition of gut microbiota. Moreover, the transplantation of enteric microbes from alcohol-fed mice to normal healthy controls remarkably shaped the composition of gut bacteria, and elicited behavioral signs of alcohol withdrawal-induced anxiety. Using quantitative real-time polymerase chain reaction, we further confirmed that the expression of genes implicated in alcohol addiction, BDNF, CRHR1 and OPRM1, was also altered by transplantation of gut microbes from alcohol-exposed donors. Collectively, our findings suggested a possibility that the alterations of gut microbiota composition might contribute to the development of alcohol withdrawal-induced anxiety, and reveal potentially new etiologies for treating alcohol addiction.


Asunto(s)
Alcoholismo/microbiología , Ansiedad/microbiología , Bacterias/crecimiento & desarrollo , Conducta Animal , Etanol , Microbioma Gastrointestinal , Intestinos/microbiología , Síndrome de Abstinencia a Sustancias/microbiología , Alcoholismo/genética , Alcoholismo/metabolismo , Alcoholismo/psicología , Animales , Ansiedad/genética , Ansiedad/metabolismo , Ansiedad/psicología , Bacterias/clasificación , Bacterias/genética , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Trasplante de Microbiota Fecal , Heces/microbiología , Hipocampo/metabolismo , Interacciones Huésped-Patógeno , Masculino , Ratones Endogámicos C57BL , Actividad Motora , Receptores de Hormona Liberadora de Corticotropina/genética , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Ribotipificación , Síndrome de Abstinencia a Sustancias/genética , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/psicología
17.
Biochim Biophys Acta Mol Basis Dis ; 1863(9): 2333-2341, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28668331

RESUMEN

Radiotherapy is often employed to treat abdominal and pelvic malignancies, but is frequently accompanied by diverse acute and chronic local injuries. It was previously unknown whether abdominal and pelvic radiotherapy impairs distant cognitive dysfunction. In the present study, we demonstrated that total abdominal irradiation (TAI) exposure caused cognitive deficits in mouse models. Mechanically, microarray assay analysis revealed that TAI elevated the expression level of miR-34a-5p in small intestine tissues and peripheral blood (PD), which targeted the 3'UTR of Brain-derived neurotrophic factor (Bdnf) mRNA in hippocampus to mediate cognitive dysfunction. Tail intravenous injection of miR-34a-5p antagomir immediately after TAI exposure rescued TAI-mediated cognitive impairment via blocking the up-regulation of miR-34a-5p in PD, resulting in restoring the Bdnf expression in the hippocampus. More importantly, high throughput sequencing validated that the gut bacterial composition of mice was shifted after TAI exposure, which was retained by miR-34a-5p antagomir injection. Thus, our findings provide new insights into pathogenic mechanism underlying abdominal and pelvic radiotherapy-mediated distant cognitive impairment.


Asunto(s)
Abdomen , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Cognición/efectos de la radiación , Rayos gamma/efectos adversos , MicroARNs/biosíntesis , Traumatismos Experimentales por Radiación/metabolismo , Animales , Femenino , Ratones , Traumatismos Experimentales por Radiación/patología , Radioterapia/efectos adversos
18.
Acta Pharmacol Sin ; 38(8): 1161-1170, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28603285

RESUMEN

Aberrant expression of miR-511 is involved in the development of cancer, but the role of miR-511 in hepatocellular carcinoma (HCC) is not well documented. In this study, we explored the molecular mechanisms of miR-511 in hepatocarcinogenesis. Our results of bioinformatics analysis suggested that B cell translocation gene 1 (BTG1), a member of anti-proliferative gene family, was one of the putative targets of miR-511. The expression levels of miR-511 were significantly higher in 30 clinical HCC tissues than in corresponding peritumor tissues, and were negatively correlated with those of BTG1 in the HCC tissues (r=-0.6105, P<0.01). In human hepatoma cell lines HepG2 and H7402, overexpression of miR-511 dose-dependently inhibited the expression of BTG1, whereas knockdown of miR-511 dose-dependently increased the expression of BTG1. Luciferase reporter gene assays verified that miR-511 targeted the 3'UTR of BTG1 mRNA. In the hepatoma cells, overexpression of miR-511 significantly decreased BTG1-induced G1 phase arrest, which was rescued by overexpression of BTG1. Furthermore, overexpression of miR-511 promoted the proliferation of the hepatoma cells, which was rescued by overexpression of BTG1. Conversely, knockdown of miR-511 inhibited cell proliferation, which was reversed by knockdown of BTG1. In conclusion, miR-511 promotes the proliferation of human hepatoma cells in vitro by targeting the 3'UTR of BTG1 mRNA.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , MicroARNs/metabolismo , Proteínas de Neoplasias/metabolismo , Regiones no Traducidas 3' , Western Blotting , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Citometría de Flujo , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
J Biol Chem ; 292(9): 3614-3623, 2017 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-28104805

RESUMEN

N6-Methyladenosine (m6A) is a prevalent modification present in the mRNAs of higher eukaryotes. YTH domain family 2 (YTHDF2), an m6A "reader" protein, can recognize mRNA m6A sites to mediate mRNA degradation. However, the regulatory mechanism of YTHDF2 is poorly understood. To this end, we investigated the post-transcriptional regulation of YTHDF2. Bioinformatics analysis suggested that the microRNA miR-145 might target the 3'-untranslated region (3'-UTR) of YTHDF2 mRNA. The levels of miR-145 were negatively correlated with those of YTHDF2 mRNA in clinical hepatocellular carcinoma (HCC) tissues, and immunohistochemical staining revealed that YTHDF2 was closely associated with malignancy of HCC. Interestingly, miR-145 decreased the luciferase activities of 3'-UTR of YTHDF2 mRNA. Mutation of predicted miR-145 binding sites in the 3'-UTR of YTHDF2 mRNA abolished the miR-145-induced decrease in luciferase activity. Overexpression of miR-145 dose-dependently down-regulated YTHDF2 expression in HCC cells at the levels of both mRNA and protein. Conversely, inhibition of miR-145 resulted in the up-regulation of YTHDF2 in the cells. Dot blot analysis and immunofluorescence staining revealed that the overexpression of miR-145 strongly increased m6A levels relative to those in control HCC cells, and this increase could be blocked by YTHDF2 overexpression. Moreover, miR-145 inhibition strongly decreased m6A levels, which were rescued by treatment with a small interfering RNA-based YTHDF2 knockdown. Thus, we conclude that miR-145 modulates m6A levels by targeting the 3'-UTR of YTHDF2 mRNA in HCC cells.


Asunto(s)
Regiones no Traducidas 3' , Adenosina/análogos & derivados , MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo , Adenosina/química , Carcinoma Hepatocelular/metabolismo , Proliferación Celular , Regulación hacia Abajo , Células Hep G2 , Humanos , Inmunohistoquímica , Neoplasias Hepáticas/metabolismo , MicroARNs/genética , Microscopía Fluorescente , Mutación , Plásmidos/metabolismo , Unión Proteica , Dominios Proteicos , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/genética
20.
Biochem Biophys Res Commun ; 482(4): 1246-1251, 2017 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-27939887

RESUMEN

B7-H3 is a glycoprotein overexpressed in cancer, but its functional contribution in this setting remains poorly understood. In the present study, we identified that the overexpression of B7-H3 in lung cancer resulted in aberrant lipid metabolism via SREBP-1/FASN signaling pathway. Immunohistochemical analysis of tissue microarrays revealed that approximately 80.4% (37/46) of lung cancer tissues were positive for B7-H3 accompanying poor prognosis. Notably, Oil red O staining and total triglyceride assay exhibited that down-regulation of B7-H3 decreased lipid synthesis in lung cancer A549 and H446 cell lines. Mechanistic investigations showed that B7-H3 modulated the expression of FASN, a fatty acid synthase, specifically. Furthermore, deletion of B7-H3 down-regulated the mRNA and protein levels of SREBP-1, a transcription factor governing the expression of FASN. Finally, correlation analysis between expression levels of B7-H3 and FASN exhibited a positive correlation in clinical lung cancer tissues. Overall, we conclude that B7-H3 hijacks SREBP-1/FASN signaling mediating abnormal lipid metabolism in lung cancer. Our finding provides new insights into the function and mechanism of B7-H3 in the development of lung cancer.


Asunto(s)
Antígenos B7/metabolismo , Acido Graso Sintasa Tipo I/metabolismo , Metabolismo de los Lípidos , Neoplasias Pulmonares/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Células A549 , Línea Celular Tumoral , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Inmunohistoquímica , Pronóstico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Matrices Tisulares , Triglicéridos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA