Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 8762, 2024 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-38627442

RESUMEN

Metastatic colorectal cancer (CRC) is still in need of effective treatments. This study applies a holistic approach to propose new targets for treatment of primary and liver metastatic CRC and investigates their therapeutic potential in-vitro. An integrative analysis of primary and metastatic CRC samples was implemented for alternative target and treatment proposals. Integrated microarray samples were grouped based on a co-expression network analysis. Significant gene modules correlated with primary CRC and metastatic phenotypes were identified. Network clustering and pathway enrichments were applied to gene modules to prioritize potential targets, which were shortlisted by independent validation. Finally, drug-target interaction search led to three agents for primary and liver metastatic CRC phenotypes. Hesperadin and BAY-1217389 suppress colony formation over a 14-day period, with Hesperadin showing additional efficacy in reducing cell viability within 48 h. As both candidates target the G2/M phase proteins NEK2 or TTK, we confirmed their anti-proliferative properties by Ki-67 staining. Hesperadinin particular arrested the cell cycle at the G2/M phase. IL-29A treatment reduced migration and invasion capacities of TGF-ß induced metastatic cell lines. In addition, this anti-metastatic treatment attenuated TGF-ß dependent mesenchymal transition. Network analysis suggests IL-29A induces the JAK/STAT pathway in a preventive manner.


Asunto(s)
Neoplasias del Colon , Neoplasias Colorrectales , Indoles , Neoplasias Hepáticas , Neoplasias del Recto , Sulfonamidas , Humanos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Transcriptoma , Quinasas Janus/metabolismo , Transducción de Señal , Factores de Transcripción STAT/metabolismo , Neoplasias del Colon/genética , Neoplasias del Recto/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundario , Factor de Crecimiento Transformador beta/metabolismo , Línea Celular Tumoral , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Quinasas Relacionadas con NIMA/genética
2.
Curr Issues Mol Biol ; 46(3): 1777-1798, 2024 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-38534733

RESUMEN

This paper aims to elucidate the differentially coexpressed genes, their potential mechanisms, and possible drug targets in low-grade invasive serous ovarian carcinoma (LGSC) in terms of the biologic continuity of normal, borderline, and malignant LGSC. We performed a bioinformatics analysis, integrating datasets generated using the GPL570 platform from different studies from the GEO database to identify changes in this transition, gene expression, drug targets, and their relationships with tumor microenvironmental characteristics. In the transition from ovarian epithelial cells to the serous borderline, the FGFR3 gene in the "Estrogen Response Late" pathway, the ITGB2 gene in the "Cell Adhesion Molecule", the CD74 gene in the "Regulation of Cell Migration", and the IGF1 gene in the "Xenobiotic Metabolism" pathway were upregulated in the transition from borderline to LGSC. The ERBB4 gene in "Proteoglycan in Cancer", the AR gene in "Pathways in Cancer" and "Estrogen Response Early" pathways, were upregulated in the transition from ovarian epithelial cells to LGSC. In addition, SPP1 and ITGB2 genes were correlated with macrophage infiltration in the LGSC group. This research provides a valuable framework for the development of personalized therapeutic approaches in the context of LGSC, with the aim of improving patient outcomes and quality of life. Furthermore, the main goal of the current study is a preliminary study designed to generate in silico inferences, and it is also important to note that subsequent in vitro and in vivo studies will be necessary to confirm the results before considering these results as fully reliable.

3.
PeerJ ; 11: e15624, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37456868

RESUMEN

Computational drug repositioning approaches are important, as they cost less compared to the traditional drug development processes. This study proposes a novel network-based drug repositioning approach, which computes similarities between disease-causing genes and drug-affected genes in a network topology to suggest candidate drugs with highest similarity scores. This new method aims to identify better treatment options by integrating systems biology approaches. It uses a protein-protein interaction network that is the main topology to compute a similarity score between candidate drugs and disease-causing genes. The disease-causing genes were mapped on this network structure. Transcriptome profiles of drug candidates were taken from the LINCS project and mapped individually on the network structure. The similarity of these two networks was calculated by different network neighborhood metrics, including Adamic-Adar, PageRank and neighborhood scoring. The proposed approach identifies the best candidates by choosing the drugs with significant similarity scores. The method was experimented on melanoma, colorectal, and prostate cancers. Several candidate drugs were predicted by applying AUC values of 0.6 or higher. Some of the predictions were approved by clinical phase trials or other in-vivo studies found in literature. The proposed drug repositioning approach would suggest better treatment options with integration of functional information between genes and transcriptome level effects of drug perturbations and diseases.


Asunto(s)
Reposicionamiento de Medicamentos , Neoplasias de la Próstata , Masculino , Humanos , Reposicionamiento de Medicamentos/métodos , Biología Computacional/métodos , Mapas de Interacción de Proteínas , Biología de Sistemas
4.
Med Sci (Basel) ; 11(3)2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37489460

RESUMEN

Combining omics data from different layers using integrative methods provides a better understanding of the biology of a complex disease such as cancer. The discovery of biomarkers related to cancer development or prognosis helps to find more effective treatment options. This study integrates multi-omics data of different cancer types with a network-based approach to explore common gene modules among different tumors by running community detection methods on the integrated network. The common modules were evaluated by several biological metrics adapted to cancer. Then, a new prognostic scoring method was developed by weighting mRNA expression, methylation, and mutation status of genes. The survival analysis pointed out statistically significant results for GNG11, CBX2, CDKN3, ARHGEF10, CLN8, SEC61G and PTDSS1 genes. The literature search reveals that the identified biomarkers are associated with the same or different types of cancers. Our method does not only identify known cancer-specific biomarker genes, but also proposes new potential biomarkers. Thus, this study provides a rationale for identifying new gene targets and expanding treatment options across cancer types.


Asunto(s)
Multiómica , Neoplasias , Humanos , Pronóstico , Neoplasias/diagnóstico , Neoplasias/genética , Biomarcadores de Tumor/genética , Análisis de Datos , Canales de Translocación SEC
5.
Mol Inform ; 42(3): e2200077, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36411244

RESUMEN

Computational drug repurposing aims to discover new treatment regimens by analyzing approved drugs on the market. This study proposes previously approved compounds that can change the expression profile of disease-causing proteins by developing a network theory-based drug repurposing approach. The novelty of the proposed approach is an exploration of module similarity between a disease-causing network and a compound-specific interaction network; thus, such an association leads to more realistic modeling of molecular cell responses at a system biology level. The overlap of the disease network and each compound-specific network is calculated based on a shortest-path similarity of networks by accounting for all protein pairs between networks. A higher similarity score indicates a significant potential of a compound. The approach was validated for breast and lung cancers. When all compounds are sorted by their normalized-similarity scores, 36 and 16 drugs are proposed as new candidates for breast and lung cancer treatment, respectively. A literature survey on candidate compounds revealed that some of our predictions have been clinically investigated in phase II/III trials for the treatment of two cancer types. As a summary, the proposed approach has provided promising initial results by modeling biochemical cell responses in a network-level data representation.


Asunto(s)
Neoplasias , Transcriptoma , Humanos , Biología Computacional/métodos , Reposicionamiento de Medicamentos/métodos , Neoplasias/tratamiento farmacológico , Proteínas
6.
PLoS One ; 17(4): e0267973, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35486660

RESUMEN

Adenomatous polyps of the colon are the most common neoplastic polyps. Although most of adenomatous polyps do not show malign transformation, majority of colorectal carcinomas originate from neoplastic polyps. Therefore, understanding of this transformation process would help in both preventive therapies and evaluation of malignancy risks. This study uncovers alterations in gene expressions as potential biomarkers that are revealed by integration of several network-based approaches. In silico analysis performed on a unified microarray cohort, which is covering 150 normal colon and adenomatous polyp samples. Significant gene modules were obtained by a weighted gene co-expression network analysis. Gene modules with similar profiles were mapped to a colon tissue specific functional interaction network. Several clustering algorithms run on the colon-specific network and the most significant sub-modules between the clusters were identified. The biomarkers were selected by filtering differentially expressed genes which also involve in significant biological processes and pathways. Biomarkers were also validated on two independent datasets based on their differential gene expressions. To the best of our knowledge, such a cascaded network analysis pipeline was implemented for the first time on a large collection of normal colon and polyp samples. We identified significant increases in TLR4 and MSX1 expressions as well as decrease in chemokine profiles with mostly pro-tumoral activities. These biomarkers might appear as both preventive targets and biomarkers for risk evaluation. As a result, this research proposes novel molecular markers that might be alternative to endoscopic approaches for diagnosis of adenomatous polyps.


Asunto(s)
Pólipos Adenomatosos , Neoplasias Colorrectales , Pólipos Adenomatosos/genética , Pólipos Adenomatosos/patología , Biomarcadores , Estudios de Cohortes , Humanos
7.
Artículo en Inglés | MEDLINE | ID: mdl-32396100

RESUMEN

Identification of common molecular mechanisms in interrelated diseases is essential for better prognoses and targeted therapies. However, complexity of metabolic pathways makes it difficult to discover common disease genes underlying metabolic disorders; and it requires more sophisticated bioinformatics models that combine different types of biological data and computational methods. Accordingly, we built an integrative network analysis model to identify shared disease genes in metabolic syndrome (MS), type 2 diabetes (T2D), and coronary artery disease (CAD). We constructed weighted gene co-expression networks by combining gene expression, protein-protein interaction, and gene ontology data from multiple sources. For 90 different configurations of disease networks, we detected the significant modules by using MCL, SPICi, and Linkcomm graph clustering algorithms. We also performed a comparative evaluation on disease modules to determine the best method providing the highest biological validity. By overlapping the disease modules, we identified 22 shared genes for MS-CAD and T2D-CAD. Moreover, 19 out of these genes were directly or indirectly associated with relevant diseases in the previous medical studies. This study does not only demonstrate the performance of different biological data sources and computational methods in disease-gene discovery, but also offers potential insights into common genetic mechanisms of the metabolic disorders.


Asunto(s)
Diabetes Mellitus Tipo 2 , Análisis por Conglomerados , Biología Computacional , Diabetes Mellitus Tipo 2/genética , Perfilación de la Expresión Génica , Ontología de Genes , Redes Reguladoras de Genes/genética , Humanos
8.
Diagnostics (Basel) ; 9(3)2019 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-31450720

RESUMEN

Leukemia is a fatal cancer and has two main types: Acute and chronic. Each type has two more subtypes: Lymphoid and myeloid. Hence, in total, there are four subtypes of leukemia. This study proposes a new approach for diagnosis of all subtypes of leukemia from microscopic blood cell images using convolutional neural networks (CNN), which requires a large training data set. Therefore, we also investigated the effects of data augmentation for an increasing number of training samples synthetically. We used two publicly available leukemia data sources: ALL-IDB and ASH Image Bank. Next, we applied seven different image transformation techniques as data augmentation. We designed a CNN architecture capable of recognizing all subtypes of leukemia. Besides, we also explored other well-known machine learning algorithms such as naive Bayes, support vector machine, k-nearest neighbor, and decision tree. To evaluate our approach, we set up a set of experiments and used 5-fold cross-validation. The results we obtained from experiments showed that our CNN model performance has 88.25% and 81.74% accuracy, in leukemia versus healthy and multiclass classification of all subtypes, respectively. Finally, we also showed that the CNN model has a better performance than other wellknown machine learning algorithms.

9.
J Bioinform Comput Biol ; 17(2): 1950012, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31057072

RESUMEN

Identification of effective drug combinations for patients is an expensive and time-consuming procedure, especially for in vitro experiments. To accelerate the synergistic drug discovery process, we present a new classification model to identify more effective anti-cancer drug pairs using in silico network biology approach. Based on the hypotheses that the drug synergy comes from the collective effects on the biological network, therefore, we developed six network biology features, including overlap and distance of drug perturbation network, that were derived by using individual drug-perturbed transcriptome profiles and the relevant biological network analysis. Using publicly available drug synergy databases and three machine-learning (ML) methods, the model was trained to discriminate the positive (synergistic) and negative (nonsynergistic) drug combinations. The proposed models were evaluated on the test cases to predict the most promising network biology feature, which is the network degree activity, i.e. the synergistic effect between drug pairs is mainly accounted by the complementary signaling pathways or molecular networks from two drugs.


Asunto(s)
Biología Computacional/métodos , Sinergismo Farmacológico , Aprendizaje Automático , Bases de Datos Farmacéuticas , Ontología de Genes , Humanos , Medicina de Precisión , Mapas de Interacción de Proteínas/efectos de los fármacos , Reproducibilidad de los Resultados , Transcriptoma
10.
Reprod Sci ; 26(6): 794-805, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30198418

RESUMEN

OBJECTIVES: To investigate gene expression differences and related functions between primary tumor, malignant cells in ascites, and metastatic peritoneal implant in high-grade serous ovarian cancer. METHODS: Biopsies from primary tumor, peritoneal implant, and ascites were collected from 10 patients operated primarily for high-grade, advanced-staged serous ovarian cancer. Total RNA isolation was performed from collected tissue biopsy and fluid samples, and RNA expression profile was measured. Messenger RNA expression profiles of 3 different groups were compared. Functional analyses of candidate genes were carried out by gene ontology and pathway analysis. RESULTS: There were significant differences in the expression of 5 genes between primary tumor and peritoneal implant, 979 genes between primary tumor and malignant cells in ascites, and 649 genes between peritoneal implant and malignant cells in ascites. Three commonly enriched gene ontology functions between "primary tumor and malignant cells in the ascites" and "peritoneal implant and malignant cells in the ascites" were protein deubiquitination, ubiquitin-dependent protein catabolism, and apoptotic processes. All genes related to these functions belonged to USP17 gene family. CONCLUSION: Gene expression difference between primary tumor and the peritoneal implant is not as much as the difference between primary tumor and free cells in the ascites. These results show that malignant cells in the ascites return into its genetic origin after they invade on the peritoneum. Significantly increased expression of DUB-enzyme genes, SNAR gene family, and ribosomal pathway genes in epithelial-mesenchymal transition suggests that this regulation is ubiquitin-proteasome dependent. Especially, this is the first study that offers USP17 as a potential target for epithelial-mesenchymal transition.


Asunto(s)
Cistadenocarcinoma Seroso/genética , Endopeptidasas/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Ováricas/genética , Proteasas Ubiquitina-Específicas/genética , Adulto , Ascitis/genética , Cistadenocarcinoma Seroso/patología , Cistadenocarcinoma Seroso/fisiopatología , Femenino , Humanos , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Ováricas/patología , Neoplasias Ováricas/fisiopatología , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/secundario , Dominios y Motivos de Interacción de Proteínas/genética , ARN Mensajero/análisis , Ubiquitina/metabolismo
11.
Comput Biol Med ; 89: 397-404, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28869900

RESUMEN

Integration of several types of patient data in a computational framework can accelerate the identification of more reliable biomarkers, especially for prognostic purposes. This study aims to identify biomarkers that can successfully predict the potential survival time of a cancer patient by integrating the transcriptomic (RNA-Seq), proteomic (RPPA), and protein-protein interaction (PPI) data. The proposed method -RPBioNet- employs a random walk-based algorithm that works on a PPI network to identify a limited number of protein biomarkers. Later, the method uses gene expression measurements of the selected biomarkers to train a classifier for the survival time prediction of patients. RPBioNet was applied to classify kidney renal clear cell carcinoma (KIRC), glioblastoma multiforme (GBM), and lung squamous cell carcinoma (LUSC) patients based on their survival time classes (long- or short-term). The RPBioNet method correctly identified the survival time classes of patients with between 66% and 78% average accuracy for three data sets. RPBioNet operates with only 20 to 50 biomarkers and can achieve on average 6% higher accuracy compared to the closest alternative method, which uses only RNA-Seq data in the biomarker selection. Further analysis of the most predictive biomarkers highlighted genes that are common for both cancer types, as they may be driver proteins responsible for cancer progression. The novelty of this study is the integration of a PPI network with mRNA and protein expression data to identify more accurate prognostic biomarkers that can be used for clinical purposes in the future.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Modelos Biológicos , Proteínas de Neoplasias/biosíntesis , Neoplasias/metabolismo , Neoplasias/mortalidad , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , Bases de Datos de Proteínas , Supervivencia sin Enfermedad , Perfilación de la Expresión Génica , Humanos , Proteínas de Neoplasias/genética , Neoplasias/genética , Valor Predictivo de las Pruebas , Proteómica , ARN Mensajero/genética , ARN Neoplásico/genética , Análisis de Secuencia de ARN , Tasa de Supervivencia
12.
Oncotarget ; 7(7): 7415-25, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26871731

RESUMEN

During a cell state transition, cells travel along trajectories in a gene expression state space. This dynamical systems framework complements the traditional concept of molecular pathways that drive cell phenotype switching. To expose the structure that hinders cancer cells from exiting robust proliferative state, we assessed the perturbation capacity of a drug library and identified 16 non-cytotoxic compounds that stimulate MCF7 breast cancer cells to exit from proliferative state to differentiated state. The transcriptome trajectories triggered by these drugs diverged, then converged. Chemical structures and drug targets of these compounds overlapped minimally. However, a network analysis of targeted pathways identified a core signaling pathway--indicating common stress-response and down-regulation of STAT1 before differentiation. This multi-trajectory analysis explores the cells' state transition with a multitude of perturbations in combination with traditional pathway analysis, leading to an encompassing picture of the dynamics of a therapeutically desired cell-state switching.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Biología Computacional/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de la Mama/genética , Femenino , Perfilación de la Expresión Génica , Redes Reguladoras de Genes/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Humanos , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas
13.
Sci Rep ; 5: 17417, 2015 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-26615774

RESUMEN

Drugs bind to their target proteins, which interact with downstream effectors and ultimately perturb the transcriptome of a cancer cell. These perturbations reveal information about their source, i.e., drugs' targets. Here, we investigate whether these perturbations and protein interaction networks can uncover drug targets and key pathways. We performed the first systematic analysis of over 500 drugs from the Connectivity Map. First, we show that the gene expression of drug targets is usually not significantly affected by the drug perturbation. Hence, expression changes after drug treatment on their own are not sufficient to identify drug targets. However, ranking of candidate drug targets by network topological measures prioritizes the targets. We introduce a novel measure, local radiality, which combines perturbed genes and functional interaction network information. The new measure outperforms other methods in target prioritization and proposes cancer-specific pathways from drugs to affected genes for the first time. Local radiality identifies more diverse targets with fewer neighbors and possibly less side effects.


Asunto(s)
Biología Computacional/métodos , Descubrimiento de Drogas , Regulación de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Algoritmos , Línea Celular , Perfilación de la Expresión Génica , Humanos , Mapas de Interacción de Proteínas , Curva ROC , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos
14.
Brief Bioinform ; 15(4): 612-25, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23255167

RESUMEN

Disease progression in cancer can vary substantially between patients. Yet, patients often receive the same treatment. Recently, there has been much work on predicting disease progression and patient outcome variables from gene expression in order to personalize treatment options. Despite first diagnostic kits in the market, there are open problems such as the choice of random gene signatures or noisy expression data. One approach to deal with these two problems employs protein-protein interaction networks and ranks genes using the random surfer model of Google's PageRank algorithm. In this work, we created a benchmark dataset collection comprising 25 cancer outcome prediction datasets from literature and systematically evaluated the use of networks and a PageRank derivative, NetRank, for signature identification. We show that the NetRank performs significantly better than classical methods such as fold change or t-test. Despite an order of magnitude difference in network size, a regulatory and protein-protein interaction network perform equally well. Experimental evaluation on cancer outcome prediction in all of the 25 underlying datasets suggests that the network-based methodology identifies highly overlapping signatures over all cancer types, in contrast to classical methods that fail to identify highly common gene sets across the same cancer types. Integration of network information into gene expression analysis allows the identification of more reliable and accurate biomarkers and provides a deeper understanding of processes occurring in cancer development and progression.


Asunto(s)
Neoplasias/fisiopatología , Algoritmos , Biomarcadores de Tumor , Progresión de la Enfermedad , Humanos
15.
Mol Biosyst ; 8(12): 3224-31, 2012 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-23042589

RESUMEN

Determination of cell signalling behaviour is crucial for understanding the physiological response to a specific stimulus or drug treatment. Current approaches for large-scale data analysis do not effectively incorporate critical topological information provided by the signalling network. We herein describe a novel model- and data-driven hybrid approach, or signal transduction score flow algorithm, which allows quantitative visualization of cyclic cell signalling pathways that lead to ultimate cell responses such as survival, migration or death. This score flow algorithm translates signalling pathways as a directed graph and maps experimental data, including negative and positive feedbacks, onto gene nodes as scores, which then computationally traverse the signalling pathway until a pre-defined biological target response is attained. Initially, experimental data-driven enrichment scores of the genes were computed in a pathway, then a heuristic approach was applied using the gene score partition as a solution for protein node stoichiometry during dynamic scoring of the pathway of interest. Incorporation of a score partition during the signal flow and cyclic feedback loops in the signalling pathway significantly improves the usefulness of this model, as compared to other approaches. Evaluation of the score flow algorithm using both transcriptome and ChIP-seq data-generated signalling pathways showed good correlation with expected cellular behaviour on both KEGG and manually generated pathways. Implementation of the algorithm as a Cytoscape plug-in allows interactive visualization and analysis of KEGG pathways as well as user-generated and curated Cytoscape pathways. Moreover, the algorithm accurately predicts gene-level and global impacts of single or multiple in silico gene knockouts.


Asunto(s)
Algoritmos , Biología Computacional , Perfilación de la Expresión Génica , Análisis por Matrices de Proteínas , Transducción de Señal , Modelos Biológicos , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA