Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Brief Bioinform ; 23(1)2022 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-34962256

RESUMO

The pharmacological arsenal against the COVID-19 pandemic is largely based on generic anti-inflammatory strategies or poorly scalable solutions. Moreover, as the ongoing vaccination campaign is rolling slower than wished, affordable and effective therapeutics are needed. To this end, there is increasing attention toward computational methods for drug repositioning and de novo drug design. Here, multiple data-driven computational approaches are systematically integrated to perform a virtual screening and prioritize candidate drugs for the treatment of COVID-19. From the list of prioritized drugs, a subset of representative candidates to test in human cells is selected. Two compounds, 7-hydroxystaurosporine and bafetinib, show synergistic antiviral effects in vitro and strongly inhibit viral-induced syncytia formation. Moreover, since existing drug repositioning methods provide limited usable information for de novo drug design, the relevant chemical substructures of the identified drugs are extracted to provide a chemical vocabulary that may help to design new effective drugs.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , COVID-19 , Células Gigantes , Pirimidinas/farmacologia , SARS-CoV-2/metabolismo , Estaurosporina/análogos & derivados , Células A549 , COVID-19/metabolismo , Biologia Computacional , Avaliação Pré-Clínica de Medicamentos , Reposicionamento de Medicamentos , Células Gigantes/metabolismo , Células Gigantes/virologia , Humanos , Estaurosporina/farmacologia
2.
Bioinformatics ; 39(7)2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37471593

RESUMO

MOTIVATION: De novo drug development is a long and expensive process that poses significant challenges from the design to the preclinical testing, making the introduction into the market slow and difficult. This limitation paved the way to the development of drug repurposing, which consists in the re-usage of already approved drugs, developed for other therapeutic indications. Although several efforts have been carried out in the last decade in order to achieve clinically relevant drug repurposing predictions, the amount of repurposed drugs that have been employed in actual pharmacological therapies is still limited. On one hand, mechanistic approaches, including profile-based and network-based methods, exploit the wealth of data about drug sensitivity and perturbational profiles as well as disease transcriptomics profiles. On the other hand, chemocentric approaches, including structure-based methods, take into consideration the intrinsic structural properties of the drugs and their molecular targets. The poor integration between mechanistic and chemocentric approaches is one of the main limiting factors behind the poor translatability of drug repurposing predictions into the clinics. RESULTS: In this work, we introduce DREAM, an R package aimed to integrate mechanistic and chemocentric approaches in a unified computational workflow. DREAM is devoted to the druggability evaluation of pathological conditions of interest, leveraging robust drug repurposing predictions. In addition, the user can derive optimized sets of drugs putatively suitable for combination therapy. In order to show the functionalities of the DREAM package, we report a case study on atopic dermatitis. AVAILABILITY AND IMPLEMENTATION: DREAM is freely available at https://github.com/fhaive/dream. The docker image of DREAM is available at: https://hub.docker.com/r/fhaive/dream.


Assuntos
Reposicionamento de Medicamentos , Transcriptoma , Humanos , Reposicionamento de Medicamentos/métodos
3.
Bioinformatics ; 39(6)2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37225400

RESUMO

MOTIVATION: Transcriptomic data can be used to describe the mechanism of action (MOA) of a chemical compound. However, omics data tend to be complex and prone to noise, making the comparison of different datasets challenging. Often, transcriptomic profiles are compared at the level of individual gene expression values, or sets of differentially expressed genes. Such approaches can suffer from underlying technical and biological variance, such as the biological system exposed on or the machine/method used to measure gene expression data, technical errors and further neglect the relationships between the genes. We propose a network mapping approach for knowledge-driven comparison of transcriptomic profiles (KNeMAP), which combines genes into similarity groups based on multiple levels of prior information, hence adding a higher-level view onto the individual gene view. When comparing KNeMAP with fold change (expression) based and deregulated gene set-based methods, KNeMAP was able to group compounds with higher accuracy with respect to prior information as well as is less prone to noise corrupted data. RESULT: We applied KNeMAP to analyze the Connectivity Map dataset, where the gene expression changes of three cell lines were analyzed after treatment with 676 drugs as well as the Fortino et al. dataset where two cell lines with 31 nanomaterials were analyzed. Although the expression profiles across the biological systems are highly different, KNeMAP was able to identify sets of compounds that induce similar molecular responses when exposed on the same biological system. AVAILABILITY AND IMPLEMENTATION: Relevant data and the KNeMAP function is available at: https://github.com/fhaive/KNeMAP and 10.5281/zenodo.7334711.


Assuntos
Perfilação da Expressão Gênica , Transcriptoma
4.
Bioinformatics ; 39(6)2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37354497

RESUMO

SUMMARY: Biological data repositories are an invaluable source of publicly available research evidence. Unfortunately, the lack of convergence of the scientific community on a common metadata annotation strategy has resulted in large amounts of data with low FAIRness (Findable, Accessible, Interoperable and Reusable). The possibility of generating high-quality insights from their integration relies on data curation, which is typically an error-prone process while also being expensive in terms of time and human labour. Here, we present ESPERANTO, an innovative framework that enables a standardized semi-supervised harmonization and integration of toxicogenomics metadata and increases their FAIRness in a Good Laboratory Practice-compliant fashion. The harmonization across metadata is guaranteed with the definition of an ad hoc vocabulary. The tool interface is designed to support the user in metadata harmonization in a user-friendly manner, regardless of the background and the type of expertise. AVAILABILITY AND IMPLEMENTATION: ESPERANTO and its user manual are freely available for academic purposes at https://github.com/fhaive/esperanto. The input and the results showcased in Supplementary File S1 are available at the same link.


Assuntos
Metadados , Software , Humanos , Toxicogenética , Idioma , Curadoria de Dados
5.
Bioinformatics ; 36(9): 2932-2933, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31950985

RESUMO

MOTIVATION: The analysis of dose-dependent effects on the gene expression is gaining attention in the field of toxicogenomics. Currently available computational methods are usually limited to specific omics platforms or biological annotations and are able to analyse only one experiment at a time. RESULTS: We developed the software BMDx with a graphical user interface for the Benchmark Dose (BMD) analysis of transcriptomics data. We implemented an approach based on the fitting of multiple models and the selection of the optimal model based on the Akaike Information Criterion. The BMDx tool takes as an input a gene expression matrix and a phenotype table, computes the BMD, its related values, and IC50/EC50 estimations. It reports interactive tables and plots that the user can investigate for further details of the fitting, dose effects and functional enrichment. BMDx allows a fast and convenient comparison of the BMD values of a transcriptomics experiment at different time points and an effortless way to interpret the results. Furthermore, BMDx allows to analyse and to compare multiple experiments at once. AVAILABILITY AND IMPLEMENTATION: BMDx is implemented as an R/Shiny software and is available at https://github.com/Greco-Lab/BMDx/. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Benchmarking , Biologia Computacional , Software , Toxicogenética , Transcriptoma
6.
Int J Mol Sci ; 22(4)2021 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-33562347

RESUMO

. De novo drug design is a computational approach that generates novel molecular structures from atomic building blocks with no a priori relationships. Conventional methods include structure-based and ligand-based design, which depend on the properties of the active site of a biological target or its known active binders, respectively. Artificial intelligence, including machine learning, is an emerging field that has positively impacted the drug discovery process. Deep reinforcement learning is a subdivision of machine learning that combines artificial neural networks with reinforcement-learning architectures. This method has successfully been employed to develop novel de novo drug design approaches using a variety of artificial networks including recurrent neural networks, convolutional neural networks, generative adversarial networks, and autoencoders. This review article summarizes advances in de novo drug design, from conventional growth algorithms to advanced machine-learning methodologies and highlights hot topics for further development.


Assuntos
Desenho de Fármacos , Aprendizado de Máquina , Redes Neurais de Computação , Preparações Farmacêuticas/química , Animais , Humanos
7.
Bioinformatics ; 34(4): 625-634, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29040390

RESUMO

Motivation: Microarray technology can be used to study the expression of thousands of genes across a number of different experimental conditions, usually hundreds. The underlying principle is that genes sharing similar expression patterns, across different samples, can be part of the same co-expression system, or they may share the same biological functions. Groups of genes are usually identified based on cluster analysis. Clustering methods rely on the similarity matrix between genes. A common choice to measure similarity is to compute the sample correlation matrix. Dimensionality reduction is another popular data analysis task which is also based on covariance/correlation matrix estimates. Unfortunately, covariance/correlation matrix estimation suffers from the intrinsic noise present in high-dimensional data. Sources of noise are: sampling variations, presents of outlying sample units, and the fact that in most cases the number of units is much larger than the number of genes. Results: In this paper, we propose a robust correlation matrix estimator that is regularized based on adaptive thresholding. The resulting method jointly tames the effects of the high-dimensionality, and data contamination. Computations are easy to implement and do not require hand tunings. Both simulated and real data are analyzed. A Monte Carlo experiment shows that the proposed method is capable of remarkable performances. Our correlation metric is more robust to outliers compared with the existing alternatives in two gene expression datasets. It is also shown how the regularization allows to automatically detect and filter spurious correlations. The same regularization is also extended to other less robust correlation measures. Finally, we apply the ARACNE algorithm on the SyNTreN gene expression data. Sensitivity and specificity of the reconstructed network is compared with the gold standard. We show that ARACNE performs better when it takes the proposed correlation matrix estimator as input. Availability and implementation: The R software is available at https://github.com/angy89/RobustSparseCorrelation. Contact: aserra@unisa.it or robtag@unisa.it. Supplementary information: Supplementary data are available at Bioinformatics online.


Assuntos
Análise por Conglomerados , Perfilação da Expressão Gênica/métodos , Software , Algoritmos , Humanos , Neoplasias/genética , Sensibilidade e Especificidade , Análise de Sequência de RNA/métodos
8.
Hum Brain Mapp ; 39(2): 932-940, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29143414

RESUMO

BACKGROUND: Despite its clinical relevance, the pathophysiology of pain in Parkinson's disease (PD) is still largely unknown, and both central and peripheral mechanisms have been invoked. OBJECTIVES: To investigate whether central pain processing is altered in "drug-naive" pain-free PD (dnPD) patients. METHODS: Using event-related functional MRI (fMRI), functional response to forearm heat stimulation (FHS) at two different intensities (41°C and 53°C) was investigated in 20 pain-free dnPD patients, compared with 18 healthy controls (HCs). Secondary analyses were performed to evaluate associations between BOLD signal changes and PD clinical features and behavioral responses. RESULTS: During low-innocuous FHS (41°C), no activation differences were found between dnPD patients and HCs. During high-noxious FHS (53°C) a significantly increased activation in the left somatosensory cortex, left cerebellum, and right low pons was observed in dnPD patients compared to HCs. In the latter experimental condition, fMRI BOLD signal changes in the right low pons (p < .0001; R = -0.8) and in the cerebellum (p = .004; R = -0.7) were negatively correlated with pain intensity ratings only in dnPD patients. No statistically significant difference in experimental pain perception was detected between dnPD patients and HCs. CONCLUSIONS: Our findings suggest that a functional remodulation of pain processing pathways occurs even in the absence of clinically overt pain symptoms in dnPD patients. These mechanisms may eventually become dysfunctional over time, contributing to the emergence of pain symptoms in more advanced PD stages. The comprehension of pain-related mechanisms may improve the clinical approach and therapeutic management of this disabling nonmotor symptom.


Assuntos
Encéfalo/fisiopatologia , Percepção da Dor/fisiologia , Dor/fisiopatologia , Doença de Parkinson/fisiopatologia , Encéfalo/diagnóstico por imagem , Mapeamento Encefálico , Circulação Cerebrovascular , Feminino , Temperatura Alta , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Oxigênio/sangue , Dor/diagnóstico por imagem
9.
Neuroradiology ; 60(5): 497-504, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29520641

RESUMO

PURPOSE: Advances in computational network analysis have enabled the characterization of topological properties of human brain networks (connectomics) from high angular resolution diffusion imaging (HARDI) MRI structural measurements. In this study, the effect of changing the diffusion weighting (b value) and sampling (number of gradient directions) was investigated in ten healthy volunteers, with specific focus on graph theoretical network metrics used to characterize the human connectome. METHODS: Probabilistic tractography based on the Q-ball reconstruction of HARDI MRI measurements was performed and structural connections between all pairs of regions from the automated anatomical labeling (AAL) atlas were estimated, to compare two HARDI schemes: low b value (b = 1000) and low direction number (n = 32) (LBLD); high b value (b = 3000) and high number (n = 54) of directions (HBHD). RESULTS: LBLD and HBHD data sets produced connectome images with highly overlapping hub structure. Overall, the HBHD scheme yielded significantly higher connection probabilities between cortical and subcortical sites and allowed detecting more connections. Small worldness and modularity were reduced in HBHD data. The clustering coefficient was significantly higher in HBHD data indicating a higher level of segregation in the resulting connectome for the HBHD scheme. CONCLUSION: Our results demonstrate that the HARDI scheme as an impact on structural connectome measures which is not automatically implied by the tractography outcome. As the number of gradient directions and b values applied may introduce a bias in the assessment of network properties, the choice of a given HARDI protocol must be carefully considered when comparing results across connectomic studies.


Assuntos
Conectoma , Imagem de Difusão por Ressonância Magnética/métodos , Feminino , Voluntários Saudáveis , Humanos , Processamento de Imagem Assistida por Computador , Imageamento Tridimensional , Masculino , Pessoa de Meia-Idade
10.
Cephalalgia ; 37(4): 305-314, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27084886

RESUMO

Objective A prospective clinical imaging study has been conducted to investigate pain processing functional pathways during trigeminal heat stimulation (THS) in patients with migraine without aura experiencing ictal cutaneous allodynia (CA) (MwoA CA+). Methods Using whole-brain BOLD-fMRI, functional response to THS at three different intensities (41°, 51° and 53℃) was investigated interictally in 20 adult MwoA CA+ patients compared with 20 MwoA patients without ictal CA (MwoA CA-) and 20 healthy controls (HCs). Secondary analyses evaluated associations between BOLD signal change and clinical features of migraine. Results During moderate-noxious THS (51℃), we observed a significantly greater activation in (a) the anterior cingulate cortex in MwoA CA+ patients compared to HCs and (b) the middle frontal gyrus in MwoA CA+ patients compared to both MwoA CA- patients and HCs. Furthermore, during high-noxious THS (53℃) a significantly decreased activation in the secondary somatosensory cortices was observed in (a) MwoA CA- patients compared to both MwoA CA+ patients and HCs and (b) MwoA CA+ patients compared to HCs. CA severity was positively correlated with the secondary somatosensory cortices activation. Conclusions Our findings suggest that CA may be subtended by both a dysfunctional analgesic compensatory mechanism and an abnormal internal representation of pain in migraine patients.


Assuntos
Hiperalgesia/fisiopatologia , Transtornos de Enxaqueca/fisiopatologia , Dor/fisiopatologia , Adulto , Encéfalo/fisiopatologia , Mapeamento Encefálico , Feminino , Temperatura Alta , Humanos , Imageamento por Ressonância Magnética , Masculino , Estudos Prospectivos
11.
BMC Bioinformatics ; 16: 261, 2015 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-26283178

RESUMO

BACKGROUND: Multiple high-throughput molecular profiling by omics technologies can be collected for the same individuals. Combining these data, rather than exploiting them separately, can significantly increase the power of clinically relevant patients subclassifications. RESULTS: We propose a multi-view approach in which the information from different data layers (views) is integrated at the levels of the results of each single view clustering iterations. It works by factorizing the membership matrices in a late integration manner. We evaluated the effectiveness and the performance of our method on six multi-view cancer datasets. In all the cases, we found patient sub-classes with statistical significance, identifying novel sub-groups previously not emphasized in literature. Our method performed better as compared to other multi-view clustering algorithms and, unlike other existing methods, it is able to quantify the contribution of single views on the final results. CONCLUSION: Our observations suggest that integration of prior information with genomic features in the subtyping analysis is an effective strategy in identifying disease subgroups. The methodology is implemented in R and the source code is available online at http://neuronelab.unisa.it/a-multi-view-genomic-data-integration-methodology/ .


Assuntos
Algoritmos , Genômica/métodos , Análise por Conglomerados , MicroRNAs/genética , MicroRNAs/metabolismo , Análise de Sequência de RNA
12.
BMC Bioinformatics ; 16: 151, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25962835

RESUMO

BACKGROUND: OMICs technologies allow to assay the state of a large number of different features (e.g., mRNA expression, miRNA expression, copy number variation, DNA methylation, etc.) from the same samples. The objective of these experiments is usually to find a reduced set of significant features, which can be used to differentiate the conditions assayed. In terms of development of novel feature selection computational methods, this task is challenging for the lack of fully annotated biological datasets to be used for benchmarking. A possible way to tackle this problem is generating appropriate synthetic datasets, whose composition and behaviour are fully controlled and known a priori. RESULTS: Here we propose a novel method centred on the generation of networks of interactions among different biological molecules, especially involved in regulating gene expression. Synthetic datasets are obtained from ordinary differential equations based models with known parameters. Our results show that the generated datasets are well mimicking the behaviour of real data, for popular data analysis methods are able to selectively identify existing interactions. CONCLUSIONS: The proposed method can be used in conjunction to real biological datasets in the assessment of data mining techniques. The main strength of this method consists in the full control on the simulated data while retaining coherence with the real biological processes. The R package MVBioDataSim is freely available to the scientific community at http://neuronelab.unisa.it/?p=1722.


Assuntos
Algoritmos , Biologia Computacional/métodos , Simulação por Computador , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Genômica/métodos , Variações do Número de Cópias de DNA , Metilação de DNA , Conjuntos de Dados como Assunto , Regulação da Expressão Gênica , Humanos , MicroRNAs/genética
13.
Adv Sci (Weinh) ; : e2401754, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38840452

RESUMO

The categorization of human diseases is mainly based on the affected organ system and phenotypic characteristics. This is limiting the view to the pathological manifestations, while it neglects mechanistic relationships that are crucial to develop therapeutic strategies. This work aims to advance the understanding of diseases and their relatedness beyond traditional phenotypic views. Hence, the similarity among 502 diseases is mapped using six different data dimensions encompassing molecular, clinical, and pharmacological information retrieved from public sources. Multiple distance measures and multi-view clustering are used to assess the patterns of disease relatedness. The integration of all six dimensions into a consensus map of disease relationships reveals a divergent disease view from the International Classification of Diseases (ICD), emphasizing novel insights offered by a multi-view disease map. Disease features such as genes, pathways, and chemicals that are enriched in distinct disease groups are identified. Finally, an evaluation of the top similar diseases of three candidate diseases common in the Western population shows concordance with known epidemiological associations and reveals rare features shared between Type 2 diabetes (T2D) and Alzheimer's disease. A revision of disease relationships holds promise for facilitating the reconstruction of comorbidity patterns, repurposing drugs, and advancing drug discovery in the future.

14.
Adv Sci (Weinh) ; : e2400389, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38923832

RESUMO

Hazard assessment is the first step in evaluating the potential adverse effects of chemicals. Traditionally, toxicological assessment has focused on the exposure, overlooking the impact of the exposed system on the observed toxicity. However, systems toxicology emphasizes how system properties significantly contribute to the observed response. Hence, systems theory states that interactions store more information than individual elements, leading to the adoption of network based models to represent complex systems in many fields of life sciences. Here, they develop a network-based approach to characterize toxicological responses in the context of a biological system, inferring biological system specific networks. They directly link molecular alterations to the adverse outcome pathway (AOP) framework, establishing direct connections between omics data and toxicologically relevant phenotypic events. They apply this framework to a dataset including 31 engineered nanomaterials with different physicochemical properties in two different in vitro and one in vivo models and demonstrate how the biological system is the driving force of the observed response. This work highlights the potential of network-based methods to significantly improve their understanding of toxicological mechanisms from a systems biology perspective and provides relevant considerations and future data-driven approaches for the hazard assessment of nanomaterials and other advanced materials.

15.
Sci Data ; 10(1): 409, 2023 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-37355733

RESUMO

Adverse outcome pathways (AOPs) are emerging as a central framework in modern toxicology and other fields in biomedicine. They serve as an extension of pathway-based concepts by depicting biological mechanisms as causally linked sequences of key events (KEs) from a molecular initiating event (MIE) to an adverse outcome. AOPs guide the use and development of new approach methodologies (NAMs) aimed at reducing animal experimentation. While AOPs model the systemic mechanisms at various levels of biological organisation, toxicogenomics provides the means to study the molecular mechanisms of chemical exposures. Systematic integration of these two concepts would improve the application of AOP-based knowledge while also supporting the interpretation of complex omics data. Hence, we established this link through rigorous curation of molecular annotations for the KEs of human relevant AOPs. We further expanded and consolidated the annotations of the biological context of KEs. These curated annotations pave the way to embed AOPs in molecular data interpretation, facilitating the emergence of new knowledge in biomedicine.


Assuntos
Rotas de Resultados Adversos , Humanos , Bases de Conhecimento , Toxicogenética
16.
Adv Sci (Weinh) ; 10(2): e2203984, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36479815

RESUMO

Mechanistic toxicology provides a powerful approach to inform on the safety of chemicals and the development of safe-by-design compounds. Although toxicogenomics supports mechanistic evaluation of chemical exposures, its implementation into the regulatory framework is hindered by uncertainties in the analysis and interpretation of such data. The use of mechanistic evidence through the adverse outcome pathway (AOP) concept is promoted for the development of new approach methodologies (NAMs) that can reduce animal experimentation. However, to unleash the full potential of AOPs and build confidence into toxicogenomics, robust associations between AOPs and patterns of molecular alteration need to be established. Systematic curation of molecular events to AOPs will create the much-needed link between toxicogenomics and systemic mechanisms depicted by the AOPs. This, in turn, will introduce novel ways of benefitting from the AOPs, including predictive models and targeted assays, while also reducing the need for multiple testing strategies. Hence, a multi-step strategy to annotate AOPs is developed, and the resulting associations are applied to successfully highlight relevant adverse outcomes for chemical exposures with strong in vitro and in vivo convergence, supporting chemical grouping and other data-driven approaches. Finally, a panel of AOP-derived in vitro biomarkers for pulmonary fibrosis (PF) is identified and experimentally validated.


Assuntos
Rotas de Resultados Adversos , Segurança Química , Animais , Medição de Risco/métodos , Toxicogenética
17.
Methods Mol Biol ; 2401: 101-120, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34902125

RESUMO

Biomarkers are valuable indicators of the state of a biological system. Microarray technology has been extensively used to identify biomarkers and build computational predictive models for disease prognosis, drug sensitivity and toxicity evaluations. Activation biomarkers can be used to understand the underlying signaling cascades, mechanisms of action and biological cross talk. Biomarker detection from microarray data requires several considerations both from the biological and computational points of view. In this chapter, we describe the main methodology used in biomarkers discovery and predictive modeling and we address some of the related challenges. Moreover, we discuss biomarker validation and give some insights into multiomics strategies for biomarker detection.


Assuntos
Análise em Microsséries , Biomarcadores , Pesquisa Biomédica
18.
Methods Mol Biol ; 2401: 121-146, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34902126

RESUMO

The amount of data made available by microarrays gives researchers the opportunity to delve into the complexity of biological systems. However, the noisy and extremely high-dimensional nature of this kind of data poses significant challenges. Microarrays allow for the parallel measurement of thousands of molecular objects spanning different layers of interactions. In order to be able to discover hidden patterns, the most disparate analytical techniques have been proposed. Here, we describe the basic methodologies to approach the analysis of microarray datasets that focus on the task of (sub)group discovery.


Assuntos
Algoritmos , Análise por Conglomerados , Perfilação da Expressão Gênica , Análise de Sequência com Séries de Oligonucleotídeos
19.
Cancers (Basel) ; 14(8)2022 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-35454948

RESUMO

Despite remarkable efforts of computational and predictive pharmacology to improve therapeutic strategies for complex diseases, only in a few cases have the predictions been eventually employed in the clinics. One of the reasons behind this drawback is that current predictive approaches are based only on the integration of molecular perturbation of a certain disease with drug sensitivity signatures, neglecting intrinsic properties of the drugs. Here we integrate mechanistic and chemocentric approaches to drug repositioning by developing an innovative network pharmacology strategy. We developed a multilayer network-based computational framework integrating perturbational signatures of the disease as well as intrinsic characteristics of the drugs, such as their mechanism of action and chemical structure. We present five case studies carried out on public data from The Cancer Genome Atlas, including invasive breast cancer, colon adenocarcinoma, lung squamous cell carcinoma, hepatocellular carcinoma and prostate adenocarcinoma. Our results highlight paclitaxel as a suitable drug for combination therapy for many of the considered cancer types. In addition, several non-cancer-related genes representing unusual drug targets were identified as potential candidates for pharmacological treatment of cancer.

20.
Comput Struct Biotechnol J ; 20: 1413-1426, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35386103

RESUMO

The recent advancements in toxicogenomics have led to the availability of large omics data sets, representing the starting point for studying the exposure mechanism of action and identifying candidate biomarkers for toxicity prediction. The current lack of standard methods in data generation and analysis hampers the full exploitation of toxicogenomics-based evidence in regulatory risk assessment. Moreover, the pipelines for the preprocessing and downstream analyses of toxicogenomic data sets can be quite challenging to implement. During the years, we have developed a number of software packages to address specific questions related to multiple steps of toxicogenomics data analysis and modelling. In this review we present the Nextcast software collection and discuss how its individual tools can be combined into efficient pipelines to answer specific biological questions. Nextcast components are of great support to the scientific community for analysing and interpreting large data sets for the toxicity evaluation of compounds in an unbiased, straightforward, and reliable manner. The Nextcast software suite is available at: ( https://github.com/fhaive/nextcast).

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA