Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Brief Bioinform ; 23(5)2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36063560

RESUMO

Biological pathways are a broadly used formalism for representing and interpreting the cascade of biochemical reactions underlying cellular and biological mechanisms. Pathway representation provides an ontological link among biomolecules such as RNA, DNA, small molecules, proteins, protein complexes, hormones and genes. Frequently, pathway annotations are used to identify mechanisms linked to genes within affected biological contexts. This important role and the simplicity and elegance in representing complex interactions led to an explosion of pathway representations and databases. Unfortunately, the lack of overlap across databases results in inconsistent enrichment analysis results, unless databases are integrated. However, due to absence of consensus, guidelines or gold standards in pathway definition and representation, integration of data across pathway databases is not straightforward. Despite multiple attempts to provide consolidated pathways, highly related, redundant, poorly overlapping or ambiguous pathways continue to render pathways analysis inconsistent and hard to interpret. Ontology-based integration will promote unbiased, comprehensive yet streamlined analysis of experiments, and will reduce the number of enriched pathways when performing pathway enrichment analysis. Moreover, appropriate and consolidated pathways provide better training data for pathway prediction algorithms. In this manuscript, we describe the current methods for pathway consolidation, their strengths and pitfalls, and highlight directions for future improvements to this research area.


Assuntos
Algoritmos , Proteínas , Bases de Dados Factuais , Hormônios , Anotação de Sequência Molecular , RNA
2.
Int J Mol Sci ; 25(4)2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-38396873

RESUMO

The identification of biomarkers for predicting inter-individual sorafenib response variability could allow hepatocellular carcinoma (HCC) patient stratification. SNPs in angiogenesis- and drug absorption, distribution, metabolism, and excretion (ADME)-related genes were evaluated to identify new potential predictive biomarkers of sorafenib response in HCC patients. Five known SNPs in angiogenesis-related genes, including VEGF-A, VEGF-C, HIF-1a, ANGPT2, and NOS3, were investigated in 34 HCC patients (9 sorafenib responders and 25 non-responders). A subgroup of 23 patients was genotyped for SNPs in ADME genes. A machine learning classifier method was used to discover classification rules for our dataset. We found that only the VEGF-A (rs2010963) C allele and CC genotype were significantly associated with sorafenib response. ADME-related gene analysis identified 10 polymorphic variants in ADH1A (rs6811453), ADH6 (rs10008281), SULT1A2/CCDC101 (rs11401), CYP26A1 (rs7905939), DPYD (rs2297595 and rs1801265), FMO2 (rs2020863), and SLC22A14 (rs149738, rs171248, and rs183574) significantly associated with sorafenib response. We have identified a genetic signature of predictive response that could permit non-responder/responder patient stratification. Angiogenesis- and ADME-related genes correlation was confirmed by cumulative genetic risk score and network and pathway enrichment analysis. Our findings provide a proof of concept that needs further validation in follow-up studies for HCC patient stratification for sorafenib prescription.


Assuntos
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Sorafenibe/farmacologia , Sorafenibe/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Antineoplásicos/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Niacinamida/uso terapêutico , Compostos de Fenilureia/uso terapêutico , Marcadores Genéticos
3.
PLoS Comput Biol ; 18(8): e1010348, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35951505

RESUMO

Pathway enrichment analysis (PEA) is a computational biology method that identifies biological functions that are overrepresented in a group of genes more than would be expected by chance and ranks these functions by relevance. The relative abundance of genes pertinent to specific pathways is measured through statistical methods, and associated functional pathways are retrieved from online bioinformatics databases. In the last decade, along with the spread of the internet, higher availability of computational resources made PEA software tools easy to access and to use for bioinformatics practitioners worldwide. Although it became easier to use these tools, it also became easier to make mistakes that could generate inflated or misleading results, especially for beginners and inexperienced computational biologists. With this article, we propose nine quick tips to avoid common mistakes and to out a complete, sound, thorough PEA, which can produce relevant and robust results. We describe our nine guidelines in a simple way, so that they can be understood and used by anyone, including students and beginners. Some tips explain what to do before starting a PEA, others are suggestions of how to correctly generate meaningful results, and some final guidelines indicate some useful steps to properly interpret PEA results. Our nine tips can help users perform better pathway enrichment analyses and eventually contribute to a better understanding of current biology.


Assuntos
Biologia Computacional , Software , Biologia Computacional/métodos , Bases de Dados Factuais , Humanos
4.
BMC Bioinformatics ; 23(Suppl 6): 393, 2022 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-36167506

RESUMO

BACKGROUND: Microarrays can perform large scale studies of differential expressed gene (DEGs) and even single nucleotide polymorphisms (SNPs), thereby screening thousands of genes for single experiment simultaneously. However, DEGs and SNPs are still just as enigmatic as the first sequence of the genome. Because they are independent from the affected biological context. Pathway enrichment analysis (PEA) can overcome this obstacle by linking both DEGs and SNPs to the affected biological pathways and consequently to the underlying biological functions and processes. RESULTS: To improve the enrichment analysis results, we present a new statistical network pre-processing method by mapping DEGs and SNPs on a biological network that can improve the relevance and significance of the DEGs or SNPs of interest to incorporate pathway topology information into the PEA. The proposed methodology improves the statistical significance of the PEA analysis in terms of computed p value for each enriched pathways and limit the number of enriched pathways. This helps reduce the number of relevant biological pathways with respect to a non-specific list of genes. CONCLUSION: The proposed method provides two-fold enhancements. Network analysis reveals fewer DEGs, by selecting only relevant DEGs and the detected DEGs improve the enriched pathways' statistical significance, rather than simply using a general list of genes.


Assuntos
Fenômenos Biológicos , Perfilação da Expressão Gênica , Expressão Gênica , Perfilação da Expressão Gênica/métodos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Polimorfismo de Nucleotídeo Único
5.
BMC Bioinformatics ; 22(Suppl 13): 376, 2021 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-34592927

RESUMO

BACKGROUND: Pathway enrichment analysis (PEA) is a well-established methodology for interpreting a list of genes and proteins of interest related to a condition under investigation. This paper aims to extend our previous work in which we introduced a preliminary comparative analysis of pathway enrichment analysis tools. We extended the earlier work by providing more case studies, comparing BiP enrichment performance with other well-known PEA software tools. METHODS: PEA uses pathway information to discover connections between a list of genes and proteins as well as biological mechanisms, helping researchers to overcome the problem of explaining biological entity lists of interest disconnected from the biological context. RESULTS: We compared the results of BiP with some existing pathway enrichment analysis tools comprising Centrality-based Pathway Enrichment, pathDIP, and Signaling Pathway Impact Analysis, considering three cancer types (colorectal, endometrial, and thyroid), for a total of six datasets (that is, two datasets per cancer type) obtained from the The Cancer Genome Atlas and Gene Expression Omnibus databases. We measured the similarities between the overlap of the enrichment results obtained using each couple of cancer datasets related to the same cancer. CONCLUSION: As a result, BiP identified some well-known pathways related to the investigated cancer type, validated by the available literature. We also used the Jaccard and meet-min indices to evaluate the stability and the similarity between the enrichment results obtained from each couple of cancer datasets. The obtained results show that BiP provides more stable enrichment results than other tools.


Assuntos
Neoplasias , Software , Biologia Computacional , Bases de Dados Factuais , Perfilação da Expressão Gênica , Humanos , Neoplasias/genética , Proteínas/genética , Transdução de Sinais
6.
Bioinformatics ; 36(15): 4377-4378, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32437515

RESUMO

SUMMARY: Biological pathways are fundamental for learning about healthy and disease states. Many existing formats support automatic software analysis of biological pathways, e.g. BioPAX (Biological Pathway Exchange). Although some algorithms are available as web application or stand-alone tools, no general graphical application for the parsing of BioPAX pathway data exists. Also, very few tools can perform pathway enrichment analysis (PEA) using pathway encoded in the BioPAX format. To fill this gap, we introduce BiP (BioPAX-Parser), an automatic and graphical software tool aimed at performing the parsing and accessing of BioPAX pathway data, along with PEA by using information coming from pathways encoded in BioPAX. AVAILABILITY AND IMPLEMENTATION: BiP is freely available for academic and non-profit organizations at https://gitlab.com/giuseppeagapito/bip under the LGPL 2.1, the GNU Lesser General Public License. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Transdução de Sinais , Software , Algoritmos
7.
Brief Bioinform ; 17(4): 553-61, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26351205

RESUMO

Predictive, preventive, personalized and participatory (P4) medicine is an emerging medical model that is based on the customization of all medical aspects (i.e. practices, drugs, decisions) of the individual patient. P4 medicine presupposes the elucidation of the so-called omic world, under the assumption that this knowledge may explain differences of patients with respect to disease prevention, diagnosis and therapies. Here, we elucidate the role of some selected omics sciences for different aspects of disease management, such as early diagnosis of diseases, prevention of diseases, selection of personalized appropriate and optimal therapies based on molecular profiling of patients. After introducing basic concepts of P4 medicine and omics sciences, we review some computational tools and approaches for analysing selected omics data, with a special focus on microarray and mass spectrometry data, which may be used to support P4 medicine. Some applications of biomarker discovery and pharmacogenomics and some experiences on the study of drug reactions are also described.


Assuntos
Análise em Microsséries , Humanos , Espectrometria de Massas , Medicina de Precisão
8.
J Biomed Inform ; 56: 273-83, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26092773

RESUMO

Microarray platforms enable the investigation of allelic variants that may be correlated to phenotypes. Among those, the Affymetrix DMET (Drug Metabolism Enzymes and Transporters) platform enables the simultaneous investigation of all the genes that are related to drug absorption, distribution, metabolism and excretion (ADME). Although recent studies demonstrated the effectiveness of the use of DMET data for studying drug response or toxicity in clinical studies, there is a lack of tools for the automatic analysis of DMET data. In a previous work we developed DMET-Analyzer, a methodology and a supporting platform able to automatize the statistical study of allelic variants, that has been validated in several clinical studies. Although DMET-Analyzer is able to correlate a single variant for each probe (related to a portion of a gene) through the use of the Fisher test, it is unable to discover multiple associations among allelic variants, due to its underlying statistic analysis strategy that focuses on a single variant for each time. To overcome those limitations, here we propose a new analysis methodology for DMET data based on Association Rules mining, and an efficient implementation of this methodology, named DMET-Miner. DMET-Miner extends the DMET-Analyzer tool with data mining capabilities and correlates the presence of a set of allelic variants with the conditions of patient's samples by exploiting association rules. To face the high number of frequent itemsets generated when considering large clinical studies based on DMET data, DMET-Miner uses an efficient data structure and implements an optimized search strategy that reduces the search space and the execution time. Preliminary experiments on synthetic DMET datasets, show how DMET-Miner outperforms off-the-shelf data mining suites such as the FP-Growth algorithms available in Weka and RapidMiner. To demonstrate the biological relevance of the extracted association rules and the effectiveness of the proposed approach from a medical point of view, some preliminary studies on a real clinical dataset are currently under medical investigation.


Assuntos
Coleta de Dados/métodos , Mineração de Dados/métodos , Farmacogenética/instrumentação , Algoritmos , Alelos , Automação , Variação Genética , Genótipo , Informática Médica/métodos , Análise de Sequência com Séries de Oligonucleotídeos , Preparações Farmacêuticas , Farmacogenética/métodos , Polimorfismo de Nucleotídeo Único , Medicina de Precisão/instrumentação , Medicina de Precisão/métodos , Software
9.
PLoS Comput Biol ; 9(1): e1002833, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23341759

RESUMO

High-throughput technologies produce massive amounts of data. However, individual methods yield data specific to the technique used and biological setup. The integration of such diverse data is necessary for the qualitative analysis of information relevant to hypotheses or discoveries. It is often useful to integrate these datasets using pathways and protein interaction networks to get a broader view of the experiment. The resulting network needs to be able to focus on either the large-scale picture or on the more detailed small-scale subsets, depending on the research question and goals. In this tutorial, we illustrate a workflow useful to integrate, analyze, and visualize data from different sources, and highlight important features of tools to support such analyses.


Assuntos
Biologia Computacional , Armazenamento e Recuperação da Informação , Visão Ocular , Envelhecimento/genética , Humanos , Neoplasias/genética , Neoplasias/patologia
10.
Biomed Pharmacother ; 174: 116478, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38547766

RESUMO

BACKGROUND: Long-term survival induced by anticancer treatments discloses emerging frailty among breast cancer (BC) survivors. Trastuzumab-induced cardiotoxicity (TIC) is reported in at least 5% of HER2+BC patients. However, TIC mechanism remains unclear and predictive genetic biomarkers are still lacking. Interaction between systemic inflammation, cytokine release and ADME genes in cancer patients might contribute to explain mechanisms underlying individual susceptibility to TIC and drug response variability. We present a single institution case series to investigate the potential role of genetic variants in ADME genes in HER2+BC patients TIC experienced. METHODS: We selected data related to 40 HER2+ BC patients undergone to DMET genotyping of ADME constitutive variant profiling, with the aim to prospectively explore their potential role in developing TIC. Only 3 patients ("case series"), who experienced TIC, were compared to 37 "control group" matched patients cardiotoxicity-sparing. All patients underwent to left ventricular ejection fraction (LVEF) evaluation at diagnosis and during anti-HER2 therapy. Each single probe was clustered to detect SNPs related to cardiotoxicity. RESULTS: In this retrospective analysis, our 3 cases were homogeneous in terms of clinical-pathological characteristics, trastuzumab-based treatment and LVEF decline. We identified 9 polymorphic variants in 8 ADME genes (UGT1A1, UGT1A6, UGT1A7, UGT2B15, SLC22A1, CYP3A5, ABCC4, CYP2D6) potentially associated with TIC. CONCLUSION: Real-world TIC incidence is higher compared to randomized clinical trials and biomarkers with potential predictive value aren't available. Our preliminary data, as proof of concept, could suggest a predictive role of pharmacogenomic approach in the identification of cardiotoxicity risk biomarkers for anti-HER2 treatment.


Assuntos
Neoplasias da Mama , Cardiotoxicidade , Polimorfismo de Nucleotídeo Único , Trastuzumab , Humanos , Feminino , Trastuzumab/efeitos adversos , Trastuzumab/farmacocinética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Cardiotoxicidade/genética , Pessoa de Meia-Idade , Estudos Retrospectivos , Antineoplásicos Imunológicos/efeitos adversos , Antineoplásicos Imunológicos/farmacocinética , Idoso , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Adulto
11.
BMC Bioinformatics ; 14 Suppl 1: S1, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23368786

RESUMO

BACKGROUND: Visualization concerns the representation of data visually and is an important task in scientific research. Protein-protein interactions (PPI) are discovered using either wet lab techniques, such mass spectrometry, or in silico predictions tools, resulting in large collections of interactions stored in specialized databases. The set of all interactions of an organism forms a protein-protein interaction network (PIN) and is an important tool for studying the behaviour of the cell machinery. Since graphic representation of PINs may highlight important substructures, e.g. protein complexes, visualization is more and more used to study the underlying graph structure of PINs. Although graphs are well known data structures, there are different open problems regarding PINs visualization: the high number of nodes and connections, the heterogeneity of nodes (proteins) and edges (interactions), the possibility to annotate proteins and interactions with biological information extracted by ontologies (e.g. Gene Ontology) that enriches the PINs with semantic information, but complicates their visualization. METHODS: In these last years many software tools for the visualization of PINs have been developed. Initially thought for visualization only, some of them have been successively enriched with new functions for PPI data management and PIN analysis. The paper analyzes the main software tools for PINs visualization considering four main criteria: (i) technology, i.e. availability/license of the software and supported OS (Operating System) platforms; (ii) interoperability, i.e. ability to import/export networks in various formats, ability to export data in a graphic format, extensibility of the system, e.g. through plug-ins; (iii) visualization, i.e. supported layout and rendering algorithms and availability of parallel implementation; (iv) analysis, i.e. availability of network analysis functions, such as clustering or mining of the graph, and the possibility to interact with external databases. RESULTS: Currently, many tools are available and it is not easy for the users choosing one of them. Some tools offer sophisticated 2D and 3D network visualization making available many layout algorithms, others tools are more data-oriented and support integration of interaction data coming from different sources and data annotation. Finally, some specialistic tools are dedicated to the analysis of pathways and cellular processes and are oriented toward systems biology studies, where the dynamic aspects of the processes being studied are central. CONCLUSION: A current trend is the deployment of open, extensible visualization tools (e.g. Cytoscape), that may be incrementally enriched by the interactomics community with novel and more powerful functions for PIN analysis, through the development of plug-ins. On the other hand, another emerging trend regards the efficient and parallel implementation of the visualization engine that may provide high interactivity and near real-time response time, as in NAViGaTOR. From a technological point of view, open-source, free and extensible tools, like Cytoscape, guarantee a long term sustainability due to the largeness of the developers and users communities, and provide a great flexibility since new functions are continuously added by the developer community through new plug-ins, but the emerging parallel, often closed-source tools like NAViGaTOR, can offer near real-time response time also in the analysis of very huge PINs.


Assuntos
Mapas de Interação de Proteínas , Software , Algoritmos , Análise por Conglomerados , Simulação por Computador , Bases de Dados de Proteínas , Mapeamento de Interação de Proteínas , Biologia de Sistemas
12.
Genes (Basel) ; 14(10)2023 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-37895264

RESUMO

Over the years, network analysis has become a promising strategy for analysing complex system, i.e., systems composed of a large number of interacting elements. In particular, multilayer networks have emerged as a powerful framework for modelling and analysing complex systems with multiple types of interactions. Network analysis can be applied to pharmacogenomics to gain insights into the interactions between genes, drugs, and diseases. By integrating network analysis techniques with pharmacogenomic data, the goal consists of uncovering complex relationships and identifying key genes to use in pathway enrichment analysis to figure out biological pathways involved in drug response and adverse reactions. In this study, we modelled omics, disease, and drug data together through multilayer network representation. Then, we mined the multilayer network with a community detection algorithm to obtain the top communities. After that, we used the identified list of genes from the communities to perform pathway enrichment analysis (PEA) to figure out the biological function affected by the selected genes. The results show that the genes forming the top community have multiple roles through different pathways.


Assuntos
Redes Reguladoras de Genes , Farmacogenética , Algoritmos
13.
BMC Bioinformatics ; 13: 258, 2012 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-23035929

RESUMO

BACKGROUND: Clinical Bioinformatics is currently growing and is based on the integration of clinical and omics data aiming at the development of personalized medicine. Thus the introduction of novel technologies able to investigate the relationship among clinical states and biological machineries may help the development of this field. For instance the Affymetrix DMET platform (drug metabolism enzymes and transporters) is able to study the relationship among the variation of the genome of patients and drug metabolism, detecting SNPs (Single Nucleotide Polymorphism) on genes related to drug metabolism. This may allow for instance to find genetic variants in patients which present different drug responses, in pharmacogenomics and clinical studies. Despite this, there is currently a lack in the development of open-source algorithms and tools for the analysis of DMET data. Existing software tools for DMET data generally allow only the preprocessing of binary data (e.g. the DMET-Console provided by Affymetrix) and simple data analysis operations, but do not allow to test the association of the presence of SNPs with the response to drugs. RESULTS: We developed DMET-Analyzer a tool for the automatic association analysis among the variation of the patient genomes and the clinical conditions of patients, i.e. the different response to drugs. The proposed system allows: (i) to automatize the workflow of analysis of DMET-SNP data avoiding the use of multiple tools; (ii) the automatic annotation of DMET-SNP data and the search in existing databases of SNPs (e.g. dbSNP), (iii) the association of SNP with pathway through the search in PharmaGKB, a major knowledge base for pharmacogenomic studies. DMET-Analyzer has a simple graphical user interface that allows users (doctors/biologists) to upload and analyse DMET files produced by Affymetrix DMET-Console in an interactive way. The effectiveness and easy use of DMET Analyzer is demonstrated through different case studies regarding the analysis of clinical datasets produced in the University Hospital of Catanzaro, Italy. CONCLUSION: DMET Analyzer is a novel tool able to automatically analyse data produced by the DMET-platform in case-control association studies. Using such tool user may avoid wasting time in the manual execution of multiple statistical tests avoiding possible errors and reducing the amount of time needed for a whole experiment. Moreover annotations and the direct link to external databases may increase the biological knowledge extracted. The system is freely available for academic purposes at: https://sourceforge.net/projects/dmetanalyzer/files/


Assuntos
Biologia Computacional/métodos , Genoma Humano/genética , Preparações Farmacêuticas/metabolismo , Farmacogenética/métodos , Polimorfismo de Nucleotídeo Único , Software , Algoritmos , Humanos
14.
Methods Mol Biol ; 2401: 249-261, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34902133

RESUMO

Microarrays are experimental methods that can provide information about gene expression and SNP data that hold great potential for new understanding, driving advances in functional genomics and clinical and molecular biology. Cluster analysis is used to analyze data that are not a priori to contain any specific subgroup. The goal is to use the data itself to recognize meaningful and informative subgroups. Also, cluster analysis helps data reduction purposes, exposes hidden patterns, and generates hypotheses regarding the relationship between genes and phenotypes. This chapter outlines a collection of cluster methods suitable for the analysis of microarray data sets.


Assuntos
Análise em Microsséries , Algoritmos , Análise por Conglomerados , Expressão Gênica , Perfilação da Expressão Gênica , Genômica , Análise de Sequência com Séries de Oligonucleotídeos
15.
Methods Mol Biol ; 2401: 263-271, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34902134

RESUMO

Microarrays are broadly used in the omic investigation and have several areas of applications in biology and medicine, providing a significant amount of data for a single experiment. Different kinds of microarrays are available, identifiable by characteristics such as the type of probes, the surface used as support, and the method used for the target detection. To better deal with microarray datasets, the development of microarray data analysis protocols simple to use as well as able to produce accurate reports, and comprehensible results arise. The object of this paper is to provide a general protocol showing how to choose the best software tool to analyze microarray data, allowing to efficiently figure out genomic/pharmacogenomic biomarkers.


Assuntos
Análise de Dados , Análise em Microsséries , Perfilação da Expressão Gênica , Genômica , Análise de Sequência com Séries de Oligonucleotídeos , Software
16.
BioTech (Basel) ; 11(4)2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-36278559

RESUMO

The COVID-19 disease (Coronavirus Disease 19), caused by the SARS-CoV-2 virus (Severe Acute Respiratory Syndrome Coronavirus 2), has posed many challenges worldwide at various levels, with special focus to the biological, medical, and epidemiological ones [...].

17.
BioTech (Basel) ; 11(3)2022 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-35892929

RESUMO

High-Throughput technologies are producing an increasing volume of data that needs large amounts of data storage, effective data models and efficient, possibly parallel analysis algorithms. Pathway and interactomics data are represented as graphs and add a new dimension of analysis, allowing, among other features, graph-based comparison of organisms' properties. For instance, in biological pathway representation, the nodes can represent proteins, RNA and fat molecules, while the edges represent the interaction between molecules. Otherwise, biological networks such as Protein-Protein Interaction (PPI) Networks, represent the biochemical interactions among proteins by using nodes that model the proteins from a given organism, and edges that model the protein-protein interactions, whereas pathway networks enable the representation of biochemical-reaction cascades that happen within the cells or tissues. In this paper, we discuss the main models for standard representation of pathways and PPI networks, the data models for the representation and exchange of pathway and protein interaction data, the main databases in which they are stored and the alignment algorithms for the comparison of pathways and PPI networks of different organisms. Finally, we discuss the challenges and the limitations of pathways and PPI network representation and analysis. We have identified that network alignment presents a lot of open problems worthy of further investigation, especially concerning pathway alignment.

18.
BioTech (Basel) ; 11(3)2022 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-35997341

RESUMO

Italy was one of the European countries most afflicted by the COVID-19 pandemic. From 2020 to 2022, Italy adopted strong containment measures against the COVID-19 epidemic and then started an important vaccination campaign. Here, we extended previous work by applying the COVID-19 Community Temporal Visualizer (CCTV) methodology to Italian COVID-19 data related to 2020, 2021, and five months of 2022. The aim of this work was to evaluate how Italy reacted to the pandemic in the first two waves of COVID-19, in which only containment measures such as the lockdown had been adopted, in the months following the start of the vaccination campaign, the months with the mildest weather, and the months affected by the new COVID-19 variants. This assessment was conducted by observing the behavior of single regions. CCTV methodology allows us to map the similarities in the behavior of Italian regions on a graph and use a community detection algorithm to visualize and analyze the spatio-temporal evolution of data. The results depict that the communities formed by Italian regions change with respect to the ten data measures and time.

19.
Genes (Basel) ; 13(10)2022 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-36292724

RESUMO

Gene expression and SNPs data hold great potential for a new understanding of disease prognosis, drug sensitivity, and toxicity evaluations. Cluster analysis is used to analyze data that do not contain any specific subgroups. The goal is to use the data itself to recognize meaningful and informative subgroups. In addition, cluster investigation helps data reduction purposes, exposes hidden patterns, and generates hypotheses regarding the relationship between genes and phenotypes. Cluster analysis could also be used to identify bio-markers and yield computational predictive models. The methods used to analyze microarrays data can profoundly influence the interpretation of the results. Therefore, a basic understanding of these computational tools is necessary for optimal experimental design and meaningful data analysis. This manuscript provides an analysis protocol to effectively analyze gene expression data sets through the K-means and DBSCAN algorithms. The general protocol enables analyzing omics data to identify subsets of features with low redundancy and high robustness, speeding up the identification of new bio-markers through pathway enrichment analysis. In addition, to demonstrate the effectiveness of our clustering analysis protocol, we analyze a real data set from the GEO database. Finally, the manuscript provides some best practice and tips to overcome some issues in the analysis of omics data sets through unsupervised learning.


Assuntos
Boidae , Animais , Análise por Conglomerados , Algoritmos , Análise em Microsséries , Análise de Dados
20.
Cells ; 11(2)2022 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-35053305

RESUMO

The cause of multiple myeloma (MM) remains largely unknown. Several pieces of evidence support the involvement of genetic and multiple environmental factors (i.e., chemical agents) in MM onset. The inter-individual variability in the bioactivation, detoxification, and clearance of chemical carcinogens such as asbestos, benzene, and pesticides might increase the MM risk. This inter-individual variability can be explained by the presence of polymorphic variants in absorption, distribution, metabolism, and excretion (ADME) genes. Despite the high relevance of this issue, few studies have focused on the inter-individual variability in ADME genes in MM risk. To identify new MM susceptibility loci, we performed an extended candidate gene approach by comparing high-throughput genotyping data of 1936 markers in 231 ADME genes on 64 MM patients and 59 controls from the CEU population. Differences in genotype and allele frequencies were validated using an internal control group of 35 non-cancer samples from the same geographic area as the patient group. We detected an association between MM risk and ADH1B rs1229984 (OR = 3.78; 95% CI, 1.18-12.13; p = 0.0282), PPARD rs6937483 (OR = 3.27; 95% CI, 1.01-10.56; p = 0.0479), SLC28A1 rs8187737 (OR = 11.33; 95% CI, 1.43-89.59; p = 0.005), SLC28A2 rs1060896 (OR = 6.58; 95% CI, 1.42-30.43; p = 0.0072), SLC29A1 rs8187630 (OR = 3.27; 95% CI, 1.01-10.56; p = 0.0479), and ALDH3A2 rs72547554 (OR = 2.46; 95% CI, 0.64-9.40; p = 0.0293). The prognostic value of these genes in MM was investigated in two public datasets showing that shorter overall survival was associated with low expression of ADH1B and SLC28A1. In conclusion, our proof-of-concept findings provide novel insights into the genetic bases of MM susceptibility.


Assuntos
Alelos , Predisposição Genética para Doença , Mieloma Múltiplo/genética , Regulação da Expressão Gênica , Humanos , Redes e Vias Metabólicas/genética , Polimorfismo de Nucleotídeo Único/genética , Fatores de Risco , Análise de Sobrevida
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa