Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Hum Mutat ; 43(2): 200-214, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34859522

RESUMEN

Rare germline variations contribute to the missing heritability of human complex diseases including cancers. Given their very low frequency, discovering and testing disease-causing rare germline variations remains challenging. The tag-single nucleotide polymorphism rs17728461 in 22q12.2 is highly associated with lung cancer risk. Here, we identified a functional rare germline variation rs548071605 (A>G) in a p65-responsive enhancer located within 22q12.2. The enhancer significantly promoted lung cancer cell proliferation in vitro and in a xenograft mouse model by upregulating the leukemia inhibitory factor (LIF) gene via the formation of a chromatin loop. Differential expression of LIF and its significant correlation with first progression survival time of patients further supported the lung cancer-driving effects of the 22q-Enh enhancer. Importantly, the rare variation was harbored in the p65 binding sequence and dramatically increased the enhancer activity by increasing responsiveness of the enhancer to p65 and B-cell lymphoma 3 protein, an oncoprotein that assisted the p65 binding. Our study revealed a regulatory rare germline variation with a potential lung cancer-driving role in the 22q12.2 risk region, providing intriguing clues for investigating the "missing heritability" of cancers, and also offered a useful experimental model for identifying causal rare variations.


Asunto(s)
Elementos de Facilitación Genéticos , Neoplasias Pulmonares , Animales , Células Germinativas , Humanos , Neoplasias Pulmonares/genética , Ratones , Polimorfismo de Nucleótido Simple
2.
Entropy (Basel) ; 24(2)2022 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-35205464

RESUMEN

As a data augmentation method, masking word is commonly used in many natural language processing tasks. However, most mask methods are based on rules and are not related to downstream tasks. In this paper, we propose a novel masking word generator, named Actor-Critic Mask Model (ACMM), which can adaptively adjust the mask strategy according to the performance of downstream tasks. In order to demonstrate the effectiveness of the method, we conducted experiments on two causal event extraction datasets. Experiment results show that, compared with various rule-based masking methods, the masked sentences generated by our proposed method can significantly enhance the generalization of the model and improve the model performance.

3.
Entropy (Basel) ; 24(12)2022 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-36554126

RESUMEN

The aim of explainable recommendation is not only to provide recommended items to users, but also to make users aware of why these items are recommended. Traditional recommendation methods infer user preferences for items using user-item rating information. However, the expressive power of latent representations of users and items is relatively limited due to the sparseness of the user-item rating matrix. Heterogeneous information networks (HIN) provide contextual information for improving recommendation performance and interpreting the interactions between users and items. However, due to the heterogeneity and complexity of context information in HIN, it is still a challenge to integrate this contextual information into explainable recommendation systems effectively. In this paper, we propose a novel framework-the dual-attention networks for explainable recommendation (DANER) in HINs. We first used multiple meta-paths to capture high-order semantic relations between users and items in HIN for generating similarity matrices, and then utilized matrix decomposition on similarity matrices to obtain low-dimensional sparse representations of users and items. Secondly, we introduced two-level attention networks, namely a local attention network and a global attention network, to integrate the representations of users and items from different meta-paths for obtaining high-quality representations. Finally, we use a standard multi-layer perceptron to model the interactions between users and items, which predict users' ratings of items. Furthermore, the dual-attention mechanism also contributes to identifying critical meta-paths to generate relevant explanations for users. Comprehensive experiments on two real-world datasets demonstrate the effectiveness of DANER on recommendation performance as compared with the state-of-the-art methods. A case study illustrates the interpretability of DANER.

4.
Mol Cancer Res ; 22(3): 227-239, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38047807

RESUMEN

Cancer risk loci provide special clues for uncovering pathogenesis of cancers. The TNFRSF19 gene located within the 13q12.12 lung cancer risk locus encodes TNF receptor superfamily member 19 (TNFRSF19) protein and has been proved to be a key target gene of a lung tissue-specific tumor suppressive enhancer, but its functional role in lung cancer pathogenesis remains to be elucidated. Here we showed that the TNFRSF19 gene could protect human bronchial epithelial Beas-2B cells from pulmonary carcinogen nicotine-derived nitrosamine ketone (NNK)-induced malignant transformation. Knockout of the TNFRSF19 significantly increased NNK-induced colony formation rate on soft agar. Moreover, TNFRSF19 expression was significantly reduced in lung cancer tissues and cell lines. Restoration of TNFRSF19 expression in A549 lung cancer cell line dramatically suppressed the tumor formation in xenograft mouse model. Interestingly, the TNFRSF19 protein that is an orphan membrane receptor could compete with LRP6 to bind Wnt3a, thereby inhibiting the Wnt/ß-catenin signaling pathway that is required for NNK-induced malignant transformation as indicated by protein pulldown, site mutation, and fluorescence energy resonance transfer experiments. Knockout of the TNFRSF19 enhanced LRP6-Wnt3a interaction, promoting ß-catenin nucleus translocation and the downstream target gene expression, and thus sensitized the cells to NNK carcinogen. In conclusion, our study demonstrated that the TNFRSF19 inhibited lung cancer carcinogenesis by competing with LRP6 to combine with Wnt3a to inhibit the Wnt/ß-catenin signaling pathway. IMPLICATIONS: These findings revealed a novel anti-lung cancer mechanism, highlighting the special significance of TNFRSF19 gene within the 13q12.12 risk locus in lung cancer pathogenesis.


Asunto(s)
Neoplasias Pulmonares , Animales , Humanos , Ratones , beta Catenina/genética , Carcinógenos , Modelos Animales de Enfermedad , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/genética , Ratones Noqueados , Receptores del Factor de Necrosis Tumoral , Vía de Señalización Wnt
5.
Cell Rep Med ; 5(4): 101488, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38565146

RESUMEN

Most recurrences of lung cancer (LC) occur within 3 years after surgery, but the underlying mechanism remains unclear. Here, we collect LC tissues with shorter (<3 years, recurrence group) and longer (>3 years, non-recurrence group) recurrence-free survival. By using 16S sequencing, we find that intratumor microbiome diversity is lower in the recurrence group and butyrate-producing bacteria are enriched in the recurrence group. The intratumor microbiome signature and circulating microbiome DNA can accurately predict LC recurrence. We prove that intratumor injection of butyrate-producing bacteria Roseburia can promote subcutaneous tumor growth. Mechanistically, bacteria-derived butyrate promotes LC metastasis by increasing expression of H19 in tumor cells through inhibiting HDAC2 and increasing H3K27 acetylation at the H19 promoter and inducing M2 macrophage polarization. Depletion of macrophages partially abolishes the metastasis-promoting effect of butyrate. Our results provide evidence for the cross-talk between the intratumor microbiome and LC metastasis and suggest the potential prognostic and therapeutic value of the intratumor microbiome.


Asunto(s)
Neoplasias Pulmonares , Microbiota , Humanos , Neoplasias Pulmonares/patología , Butiratos/metabolismo , Recurrencia Local de Neoplasia/metabolismo , Macrófagos
6.
Cancer Res Commun ; 3(5): 933-942, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37377889

RESUMEN

Currently, approximately 30%-55% of the patients with non-small cell lung cancer (NSCLC) develop recurrence due to minimal residual disease (MRD) after receiving surgical resection of the tumor. This study aims to develop an ultrasensitive and affordable fragmentomic assay for MRD detection in patients with NSCLC. A total of 87 patients with NSCLC, who received curative surgical resections (23 patients relapsed during follow-up), enrolled in this study. A total of 163 plasma samples, collected at 7 days and 6 months postsurgical, were used for both whole-genome sequencing (WGS) and targeted sequencing. WGS-based cell-free DNA (cfDNA) fragment profile was used to fit regularized Cox regression models, and leave-one-out cross-validation was further used to evaluate models' performance. The models showed excellent performances in detecting patients with a high risk of recurrence. At 7 days postsurgical, the high-risk patients detected by our model showed an increased risk of 4.6 times, while the risk increased to 8.3 times at 6 months postsurgical. These fragmentomics determined higher risk compared with the targeted sequencing-based circulating mutations both at 7 days and 6 months postsurgical. The overall sensitivity for detecting patients with recurrence reached 78.3% while using both fragmentomics and mutation results from 7 days and 6 months postsurgical, which increased from the 43.5% sensitivity by using only the circulating mutations. The fragmentomics showed great sensitivity in predicting patient recurrence compared with the traditional circulating mutation, especially after the surgery for early-stage NSCLC, therefore exhibiting great potential to guide adjuvant therapeutics. Significance: The circulating tumor DNA mutation-based approach shows limited performance in MRD detection, especially for landmark MRD detection at an early-stage cancer after surgery. Here, we describe a cfDNA fragmentomics-based method in MRD detection of resectable NSCLC using WGS, and the cfDNA fragmentomics showed a great sensitivity in predicting prognosis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Ácidos Nucleicos Libres de Células , ADN Tumoral Circulante , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Ácidos Nucleicos Libres de Células/genética , Neoplasia Residual/diagnóstico , ADN Tumoral Circulante/genética
7.
J Hematol Oncol ; 15(1): 141, 2022 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-36209111

RESUMEN

BACKGROUND: Besides featured glucose consumption, recent studies reveal that cancer cells might prefer "addicting" specific energy substrates from the tumor microenvironment (TME); however, the underlying mechanisms remain unclear. METHODS: Fibroblast-specific long noncoding RNAs were screened using RNA-seq data of our NJLCC cohort, TCGA, and CCLE datasets. The expression and package of LINC01614 into exosomes were identified using flow cytometric sorting, fluorescence in situ hybridization (FISH), and quantitative reverse transcription polymerase chain reaction (RT-PCR). The transfer and functional role of LINC01614 in lung adenocarcinoma (LUAD) and CAFs were investigated using 4-thiouracil-labeled RNA transfer and gain- and loss-of-function approaches. RNA pull-down, RNA immunoprecipitation, dual-luciferase assay, gene expression microarray, and bioinformatics analysis were performed to investigate the underlying mechanisms involved. RESULTS: We demonstrate that cancer-associated fibroblasts (CAFs) in LUAD primarily enhance the glutamine metabolism of cancer cells. A CAF-specific long noncoding RNA, LINC01614, packaged by CAF-derived exosomes, mediates the enhancement of glutamine uptake in LUAD cells. Mechanistically, LINC01614 directly interacts with ANXA2 and p65 to facilitate the activation of NF-κB, which leads to the upregulation of the glutamine transporters SLC38A2 and SLC7A5 and eventually enhances the glutamine influx of cancer cells. Reciprocally, tumor-derived proinflammatory cytokines upregulate LINC01614 in CAFs, constituting a feedforward loop between CAFs and cancer cells. Blocking exosome-transmitted LINC01614 inhibits glutamine addiction and LUAD growth in vivo. Clinically, LINC01614 expression in CAFs is associated with the glutamine influx and poor prognosis of patients with LUAD. CONCLUSION: Our study highlights the therapeutic potential of targeting a CAF-specific lncRNA to inhibit glutamine utilization and cancer progression in LUAD.


Asunto(s)
Adenocarcinoma , Fibroblastos Asociados al Cáncer , Neoplasias Pulmonares , ARN Largo no Codificante , Adenocarcinoma/genética , Fibroblastos Asociados al Cáncer/patología , Citocinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Glucosa/metabolismo , Glutamina/metabolismo , Humanos , Hibridación Fluorescente in Situ , Transportador de Aminoácidos Neutros Grandes 1/genética , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Pulmón/patología , Neoplasias Pulmonares/patología , FN-kappa B/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Microambiente Tumoral
8.
Cancer Biomark ; 32(4): 531-539, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34275895

RESUMEN

BACKGROUND: The incidence of lung cancer in Yunnan area ranks firstly in the world and underlying molecular mechanisms of lung cancer in Yunnan region are still unclear. We screened a novel potential oncogene CYP2S1 used mRNA microassay and bioinformation database. The function of CYP2S1 in lung cancer has not been reported. OBJECTIVE: To investigate the functions of CYP2S1 in lung cancer. METHODS: Immunohistochemistry and Real-time PCR were used to verify the expression of CYP2S1. Colony formation and Transwell assays were used to determine cell proliferation, invasion and migration. Xenograft assays were used to detected cell growth in vivo. RESULTS: CYP2S1 is significantly up-regulated in lung cancer tissues and cells. Knockdown CYP2S1 in lung cancer cells resulted in decrease cell proliferation, invasion and migration in vitro. Animal experiments showed downregulation of CYP2S1 inhibited lung cancer cell growth in vivo. GSEA analysis suggested that CYP2S1 played functions by regulating E2F targets and G2M checkpoint pathway which involved in cell cycle. Kaplan-Meier analysis indicated that patients with high CYP2S1 had markedly shorter event overall survival (OS) time. CONCLUSIONS: Our data demonstrate that CYP2S1 exerts tumor suppressor function in lung cancer. The high expression of CYP2S1 is an unfavorable prognostic marker for patient survival.


Asunto(s)
Adenocarcinoma del Pulmón/enzimología , Sistema Enzimático del Citocromo P-450/metabolismo , Neoplasias Pulmonares/genética , Células A549 , Adenocarcinoma del Pulmón/patología , Animales , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Femenino , Xenoinjertos , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Regulación hacia Arriba
9.
Mol Cancer Res ; 19(12): 1992-2002, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34535561

RESUMEN

Genome-wide association studies (GWAS) have implicated the 1q22 gastric cancer risk locus in disease, but little is known about its underlying oncogenic functions. This study represents a systematic investigation of the biological significance and potential mechanism associated with the gastric cancer risk of SNP rs2075570(C>T) in 1q22. We identified two functional germline variations (rs2049805-C and rs2974931-G) in an active enhancer in a 64.8 kb high-linkage disequilibrium block of rs2075570. The enhancer upregulated ubiquitin associated protein 2 like (UBAP2L) gene expression over a 960 kb distance by chromatin looping. Gastric cancer tissues expressed significantly higher levels of UBAP2L than was observed in the matched noncancerous tissues, and the UBAP2L expression was negatively correlated with patient survival. Downregulation of UBAP2L inhibited the proliferation and invasion of human gastric cancer cells in vitro and in a xenograft mouse model. Notably, the two mutant variations significantly enforced the enhancer activity and UBAP2L expression. In conclusion, this study revealed two causal variations in the 1q22 region using tag-SNP rs2075570 as a genetic marker. These variations may affect the occurrence and progression of gastric cancer by reinforcing the expression of the 1q22-Enh enhancer-regulated UBAP2L target gene. IMPLICATIONS: Our study provides an important clue of how noncoding germline variations contribute to gastric cancer, which gives a novel insight into understanding the genetic mechanism of gastric cancer.


Asunto(s)
Proteínas Portadoras/metabolismo , Cromatina/metabolismo , Sitios Genéticos/genética , Neoplasias Gástricas/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Ratones , Ratones Desnudos , Factores de Riesgo , Neoplasias Gástricas/patología , Transfección
10.
Genome Biol ; 20(1): 103, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-31126313

RESUMEN

BACKGROUND: Inherited factors contribute to lung cancer risk, but the mechanism is not well understood. Defining the biological consequence of GWAS hits in cancers is a promising strategy to elucidate the inherited mechanisms of cancers. The tag-SNP rs753955 (A>G) in 13q12.12 is highly associated with lung cancer risk in the Chinese population. Here, we systematically investigate the biological significance and the underlying mechanism behind 13q12.12 risk locus in vitro and in vivo. RESULTS: We characterize a novel p53-responsive enhancer with lung tissue cell specificity in a 49-kb high linkage disequilibrium block of rs753955. This enhancer harbors 3 highly linked common inherited variations (rs17336602, rs4770489, and rs34354770) and six p53 binding sequences either close to or located between the variations. The enhancer effectively protects normal lung cell lines against pulmonary carcinogen NNK-induced DNA damages and malignant transformation by upregulating TNFRSF19 through chromatin looping. These variations significantly weaken the enhancer activity by affecting its p53 response, especially when cells are exposed to NNK. The effect of the mutant enhancer alleles on TNFRSF19 target gene in vivo is supported by expression quantitative trait loci analysis of 117 Chinese NSCLC samples and GTEx data. Differentiated expression of TNFRSF19 and its statistical significant correlation with tumor TNM staging and patient survival indicate a suppressor role of TNFRSF19 in lung cancer. CONCLUSION: This study provides evidence of how the inherited variations in 13q12.12 contribute to lung cancer risk, highlighting the protective roles of the p53-responsive enhancer-mediated TNFRSF19 activation in lung cells under carcinogen stress.


Asunto(s)
Cromosomas Humanos Par 13 , Elementos de Facilitación Genéticos , Neoplasias Pulmonares/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis , Línea Celular Tumoral , Reparación del ADN , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Desequilibrio de Ligamiento , Neoplasias Pulmonares/metabolismo , Polimorfismo de Nucleótido Simple
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA