Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Swiss Med Wkly ; 150: w20195, 2020 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-32083704

RESUMO

With the emerging advances made in genomics and functional genomics approaches, there is a critical and growing unmet need to integrate plural datasets in order to identify driver genes in cancer. An integrative approach, with the convergence of multiple types of genetic evidence, can limit false positives through a posterior filtering strategy and reduce the need for multiple hypothesis testing to identify true cancer vulnerabilities. We performed a pooled shRNA screen against 906 human genes in the oral cancer cell line AW13516 in triplicate. The genes that were depleted in the screen were integrated with copy number alteration and gene expression data and ranked based on ROAST analysis, using an integrative scoring system, DepRanker, to compute a Rank Impact Score (RIS) for each gene. The RIS-based ranking of candidate driver genes was used to identify the putative oncogenes AURKB and TK1 as essential for oral cancer cell proliferation. We validated the findings, showing that shRNA mediated genetic knockdown of TK1 or pharmacological inhibition of AURKB by AZD-1152 HQPA in AW13516 cells could significantly impede their proliferation. Next we analysed alterations in AURKB and TK1 genes in head and neck cancer and their association with prognosis using data on 528 patients obtained from TCGA. Patients harbouring alterations in AURKB and TK1 genes were associated with poor survival. To summarise, we present DepRanker as a simple yet robust package with no third-party dependencies for the identification of potential driver genes from a pooled shRNA functional genomic screen by integrating results from RNAi screens with gene expression and copy number data. Using DepRanker, we identify AURKB and TK1 as potential therapeutic targets in oral cancer. DepRanker is in the public domain and available for download at http://www.actrec.gov.in/pi-webpages/AmitDutt/DepRanker/DepRanker.html.


Assuntos
Aurora Quinase B/genética , Tecnologia de Impulso Genético/métodos , Neoplasias de Cabeça e Pescoço/genética , RNA Interferente Pequeno/genética , Timidina Quinase/genética , Linhagem Celular , Genômica/métodos , Humanos , Oncogenes , Software , Sobrevida , Neoplasias da Língua/genética
2.
PLoS Comput Biol ; 15(11): e1007520, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31765387

RESUMO

Although existing computational models have identified many common driver genes, it remains challenging to identify the personalized driver genes by using samples of an individual patient. Recently, the methods of exploiting the structure-based control principles of complex networks provide new clues for identifying minimum number of driver nodes to drive the state transition of large-scale complex networks from an initial state to the desired state. However, the structure-based network control methods cannot be directly applied to identify the personalized driver genes due to the unknown network dynamics of the personalized system. Here we proposed the personalized network control model (PNC) to identify the personalized driver genes by employing the structure-based network control principle on genetic data of individual patients. In PNC model, we firstly presented a paired single sample network construction method to construct the personalized state transition network for capturing the phenotype transitions between healthy and disease states. Then, we designed a novel structure-based network control method from the Feedback Vertex Sets-based control perspective to identify the personalized driver genes. The wide experimental results on 13 cancer datasets from The Cancer Genome Atlas firstly showed that PNC model outperforms current state-of-the-art methods, in terms of F-measures for identifying cancer driver genes enriched in the gold-standard cancer driver gene lists. Furthermore, these results showed that personalized driver genes can be explored by their network characteristics even when they are hidden factors in transcription and mutation profiles. Our PNC gives novel insights and useful tools into understanding the tumor heterogeneity in cancer. The PNC package and data resources used in this work can be freely downloaded from https://github.com/NWPU-903PR/PNC.


Assuntos
Biologia Computacional/métodos , Neoplasias/genética , Medicina de Precisão/métodos , Algoritmos , Tecnologia de Impulso Genético/métodos , Redes Reguladoras de Genes/genética , Genômica/métodos , Humanos , Modelos Genéticos , Modelos Teóricos , Mutação/genética , Oncogenes/genética
3.
Sci Rep ; 9(1): 5959, 2019 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-30976053

RESUMO

High coverage and mutual exclusivity (HCME), which are considered two combinatorial properties of mutations in a collection of driver genes in cancers, have been used to develop mathematical programming models for distinguishing cancer driver gene sets. In this paper, we summarize a weak HCME pattern to justify the description of practical mutation datasets. We then present AWRMP, a method for identifying driver gene sets through the adaptive assignment of appropriate weights to gene candidates to tune the balance between coverage and mutual exclusivity. It embeds the genetic algorithm into the subsampling strategy to provide the optimization results robust against the uncertainty and noise in the data. Using biological datasets, we show that AWRMP can identify driver gene sets that satisfy the weak HCME pattern and outperform the state-of-arts methods in terms of robustness.


Assuntos
Biologia Computacional/métodos , Tecnologia de Impulso Genético/métodos , Modelos Teóricos , Mutação , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patologia , Algoritmos , Bases de Dados Genéticas , Humanos
4.
Sci Rep ; 9(1): 5685, 2019 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-30952905

RESUMO

Long intergenic non-coding RNAs (lincRNAs) are emerging as integral components of signaling pathways in various cancer types. In neuroblastoma, only a handful of lincRNAs are known as upstream regulators or downstream effectors of oncogenes. Here, we exploit RNA sequencing data of primary neuroblastoma tumors, neuroblast precursor cells, neuroblastoma cell lines and various cellular perturbation model systems to define the neuroblastoma lincRNome and map lincRNAs up- and downstream of neuroblastoma driver genes MYCN, ALK and PHOX2B. Each of these driver genes controls the expression of a particular subset of lincRNAs, several of which are associated with poor survival and are differentially expressed in neuroblastoma tumors compared to neuroblasts. By integrating RNA sequencing data from both primary tumor tissue and cancer cell lines, we demonstrate that several of these lincRNAs are expressed in stromal cells. Deconvolution of primary tumor gene expression data revealed a strong association between stromal cell composition and driver gene status, resulting in differential expression of these lincRNAs. We also explored lincRNAs that putatively act upstream of neuroblastoma driver genes, either as presumed modulators of driver gene activity, or as modulators of effectors regulating driver gene expression. This analysis revealed strong associations between the neuroblastoma lincRNAs MIAT and MEG3 and MYCN and PHOX2B activity or expression. Together, our results provide a comprehensive catalogue of the neuroblastoma lincRNome, highlighting lincRNAs up- and downstream of key neuroblastoma driver genes. This catalogue forms a solid basis for further functional validation of candidate neuroblastoma lincRNAs.


Assuntos
Neuroblastoma/genética , RNA Longo não Codificante/genética , Linhagem Celular Tumoral , Tecnologia de Impulso Genético/métodos , Perfilação da Expressão Gênica/métodos , Humanos , Células-Tronco Neurais/fisiologia , Análise de Sequência de RNA/métodos , Transdução de Sinais/genética , Fatores de Transcrição/genética
5.
World Neurosurg ; 107: 990-1000, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28751139

RESUMO

OBJECTIVE: This study is to identify pediatric brain tumors (PBT) driver genes and key pathways to detect the expression of the driver genes and also to clarify the relationship between patients' prognosis and expression of driver genes. METHODS: The gene expression profile of GSE50161 was analyzed to identify the differentially expressed genes (DEGs) between tumor tissue and the normal tissue. Gene ontology, Kyoto Encyclopedia of Genes and Genomes analysis, and protein-protein interaction network analysis were conducted to identify the enrichment functions, pathways, and hub genes. After hub genes were identified, quantitative reverse transcription polymerase chain reaction was used to confirm the differential expression of these hub genes. Survival data of 325 patients' were analyzed to clarify the relationship between prognosis and expression levels of the mutual hub genes. RESULTS: Gene ontology and Kyoto Encyclopedia of Genes and Genomes analysis showed that there were 13 common functions and 3 common pathways which were upregulated or downregulated among the 4 groups. Mutual hub genes were somatostatin (SST), glutamate decarboxylase 2 (GAD2), and single copy human parvalbumin gene (PVALB). The expression of SST, GAD2, and PVALB in glioma cells significantly decreased compared with normal glial cells (P < 0.05). In addition, survival analysis showed a favorable progression-free and overall survival in patients with glioma with SST, GAD2, and PVALB high expression (P < 0.05). CONCLUSIONS: SST, GAD2, and PVALB significantly decrease in glioma cells compared with normal glial cells. Survival analysis suggests that patients with high-expressed SST, GAD2, and PVALB have a longer overall and progression-free survival. The differential expressed genes identified in this study provide novel targets for diagnosis and treatment.


Assuntos
Neoplasias Encefálicas/genética , Tecnologia de Impulso Genético/métodos , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes/genética , Neoplasias Encefálicas/diagnóstico , Linhagem Celular Tumoral , Criança , Marcadores Genéticos/genética , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA