Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
JMIR Hum Factors ; 9(4): e38701, 2022 Nov 04.
Article in English | MEDLINE | ID: mdl-35930561

ABSTRACT

BACKGROUND: Over the past few years, studies have increasingly focused on the development of mobile apps as complementary tools to existing traditional pharmacovigilance surveillance systems for improving and facilitating adverse drug reaction (ADR) reporting. OBJECTIVE: In this research, we evaluated the potentiality of a new mobile app (vaxEffect@UniMiB) to perform longitudinal studies, while preserving the anonymity of the respondents. We applied the app to monitor the ADRs during the COVID-19 vaccination campaign in a sample of the Italian population. METHODS: We administered vaxEffect@UniMiB to a convenience sample of academic subjects vaccinated at the Milano-Bicocca University hub for COVID-19 during the Italian national vaccination campaign. vaxEffect@UniMiB was developed for both Android and iOS devices. The mobile app asks users to send their medical history and, upon every vaccine administration, their vaccination data and the ADRs that occurred within 7 days postvaccination, making it possible to follow the ADR dynamics for each respondent. The app sends data over the web to an application server. The server, along with receiving all user data, saves the data in a SQL database server and reminds patients to submit vaccine and ADR data by push notifications sent to the mobile app through Firebase Cloud Messaging (FCM). On initial startup of the app, a unique user identifier (UUID) was generated for each respondent, so its anonymity was completely ensured, while enabling longitudinal studies. RESULTS: A total of 3712 people were vaccinated during the first vaccination wave. A total of 2733 (73.6%) respondents between the ages of 19 and 80 years, coming from the University of Milano-Bicocca (UniMiB) and the Politecnico of Milan (PoliMi), participated in the survey. Overall, we collected information about vaccination and ADRs to the first vaccine dose for 2226 subjects (60.0% of the first dose vaccinated), to the second dose for 1610 subjects (43.4% of the second dose vaccinated), and, in a nonsponsored fashion, to the third dose for 169 individuals (4.6%). CONCLUSIONS: vaxEffect@UniMiB was revealed to be the first attempt in performing longitudinal studies to monitor the same subject over time in terms of the reported ADRs after each vaccine administration, while guaranteeing complete anonymity of the subject. A series of aspects contributed to the positive involvement from people in using this app to report their ADRs to vaccination: ease of use, availability from multiple platforms, anonymity of all survey participants and protection of the submitted data, and the health care workers' support.

2.
PLoS Comput Biol ; 18(2): e1009337, 2022 02.
Article in English | MEDLINE | ID: mdl-35130273

ABSTRACT

Metabolism is directly and indirectly fine-tuned by a complex web of interacting regulatory mechanisms that fall into two major classes. On the one hand, the expression level of the catalyzing enzyme sets the maximal theoretical flux level (i.e., the net rate of the reaction) for each enzyme-controlled reaction. On the other hand, metabolic regulation controls the metabolic flux through the interactions of metabolites (substrates, cofactors, allosteric modulators) with the responsible enzyme. High-throughput data, such as metabolomics and transcriptomics data, if analyzed separately, do not accurately characterize the hierarchical regulation of metabolism outlined above. They must be integrated to disassemble the interdependence between different regulatory layers controlling metabolism. To this aim, we propose INTEGRATE, a computational pipeline that integrates metabolomics and transcriptomics data, using constraint-based stoichiometric metabolic models as a scaffold. We compute differential reaction expression from transcriptomics data and use constraint-based modeling to predict if the differential expression of metabolic enzymes directly originates differences in metabolic fluxes. In parallel, we use metabolomics to predict how differences in substrate availability translate into differences in metabolic fluxes. We discriminate fluxes regulated at the metabolic and/or gene expression level by intersecting these two output datasets. We demonstrate the pipeline using a set of immortalized normal and cancer breast cell lines. In a clinical setting, knowing the regulatory level at which a given metabolic reaction is controlled will be valuable to inform targeted, truly personalized therapies in cancer patients.


Subject(s)
Computer Simulation , Metabolic Networks and Pathways , Metabolomics , Proteomics , Transcriptome , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Proof of Concept Study
3.
PLoS Comput Biol ; 17(11): e1009550, 2021 11.
Article in English | MEDLINE | ID: mdl-34748537

ABSTRACT

Metabolic network models are increasingly being used in health care and industry. As a consequence, many tools have been released to automate their reconstruction process de novo. In order to enable gene deletion simulations and integration of gene expression data, these networks must include gene-protein-reaction (GPR) rules, which describe with a Boolean logic relationships between the gene products (e.g., enzyme isoforms or subunits) associated with the catalysis of a given reaction. Nevertheless, the reconstruction of GPRs still remains a largely manual and time consuming process. Aiming at fully automating the reconstruction process of GPRs for any organism, we propose the open-source python-based framework GPRuler. By mining text and data from 9 different biological databases, GPRuler can reconstruct GPRs starting either from just the name of the target organism or from an existing metabolic model. The performance of the developed tool is evaluated at small-scale level for a manually curated metabolic model, and at genome-scale level for three metabolic models related to Homo sapiens and Saccharomyces cerevisiae organisms. By exploiting these models as benchmarks, the proposed tool shown its ability to reproduce the original GPR rules with a high level of accuracy. In all the tested scenarios, after a manual investigation of the mismatches between the rules proposed by GPRuler and the original ones, the proposed approach revealed to be in many cases more accurate than the original models. By complementing existing tools for metabolic network reconstruction with the possibility to reconstruct GPRs quickly and with a few resources, GPRuler paves the way to the study of context-specific metabolic networks, representing the active portion of the complete network in given conditions, for organisms of industrial or biomedical interest that have not been characterized metabolically yet.


Subject(s)
Metabolic Networks and Pathways/genetics , Models, Biological , Software , Computational Biology , Computer Simulation , Databases, Genetic/statistics & numerical data , Databases, Protein/statistics & numerical data , Humans , Models, Genetic , Molecular Sequence Annotation , Protein Interaction Maps/genetics , Protein Structure, Quaternary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
4.
Comput Struct Biotechnol J ; 18: 993-999, 2020.
Article in English | MEDLINE | ID: mdl-32373287

ABSTRACT

We present MaREA4Galaxy, a user-friendly tool that allows a user to characterize and to graphically compare groups of samples with different transcriptional regulation of metabolism, as estimated from cross-sectional RNA-seq data. The tool is available as plug-in for the widely-used Galaxy platform for comparative genomics and bioinformatics analyses. MaREA4Galaxy combines three modules. The Expression2RAS module, which, for each reaction of a specified set, computes a Reaction Activity Score (RAS) as a function of the expression level of genes encoding for the associated enzyme. The MaREA (Metabolic Reaction Enrichment Analysis) module that allows to highlight significant differences in reaction activities between specified groups of samples. The Clustering module which employs the RAS computed before as a metric for unsupervised clustering of samples into distinct metabolic subgroups; the Clustering tool provides different clustering techniques and implements standard methods to evaluate the goodness of the results.

5.
PLoS Comput Biol ; 15(2): e1006733, 2019 02.
Article in English | MEDLINE | ID: mdl-30818329

ABSTRACT

Metabolic reprogramming is a general feature of cancer cells. Regrettably, the comprehensive quantification of metabolites in biological specimens does not promptly translate into knowledge on the utilization of metabolic pathways. By estimating fluxes across metabolic pathways, computational models hold the promise to bridge this gap between data and biological functionality. These models currently portray the average behavior of cell populations however, masking the inherent heterogeneity that is part and parcel of tumorigenesis as much as drug resistance. To remove this limitation, we propose single-cell Flux Balance Analysis (scFBA) as a computational framework to translate single-cell transcriptomes into single-cell fluxomes. We show that the integration of single-cell RNA-seq profiles of cells derived from lung adenocarcinoma and breast cancer patients into a multi-scale stoichiometric model of a cancer cell population: significantly 1) reduces the space of feasible single-cell fluxomes; 2) allows to identify clusters of cells with different growth rates within the population; 3) points out the possible metabolic interactions among cells via exchange of metabolites. The scFBA suite of MATLAB functions is available at https://github.com/BIMIB-DISCo/scFBA, as well as the case study datasets.


Subject(s)
Computational Biology/methods , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Adenocarcinoma of Lung/genetics , Algorithms , Breast Neoplasms/genetics , Computer Simulation , Female , Gene Expression Profiling/methods , Genetics, Population/methods , Humans , Male , Metabolic Networks and Pathways , Neoplasms/genetics , Neoplasms/metabolism , RNA/genetics , Software , Transcriptome/genetics
6.
J Biomed Inform ; 87: 37-49, 2018 11.
Article in English | MEDLINE | ID: mdl-30244122

ABSTRACT

Effective stratification of cancer patients on the basis of their molecular make-up is a key open challenge. Given the altered and heterogenous nature of cancer metabolism, we here propose to use the overall expression of central carbon metabolism as biomarker to characterize groups of patients with important characteristics, such as response to ad-hoc therapeutic strategies and survival expectancy. To this end, we here introduce the data integration framework named Metabolic Reaction Enrichment Analysis (MaREA), which strives to characterize the metabolic deregulations that distinguish cancer phenotypes, by projecting RNA-seq data onto metabolic networks, without requiring metabolic measurements. MaREA computes a score for each network reaction, based on the expression of the set of genes encoding for the associated enzyme(s). The scores are first used as features for cluster analysis and then to rank and visualize in an organized fashion the metabolic deregulations that distinguish cancer sub-types. We applied our method to recent lung and breast cancer RNA-seq datasets from The Cancer Genome Atlas and we were able to identify subgroups of patients with significant differences in survival expectancy. We show how the prognostic power of MaREA improves when an extracted and further curated core model focusing on central carbon metabolism is used rather than the genome-wide reference network. The visualization of the metabolic differences between the groups with best and worst prognosis allowed to identify and analyze key metabolic properties related to cancer aggressiveness. Some of these properties are shared across different cancer (sub) types, e.g., the up-regulation of nucleic acid and amino acid synthesis, whereas some other appear to be tumor-specific, such as the up- or down-regulation of the phosphoenolpyruvate carboxykinase reaction, which display different patterns in distinct tumor (sub)types. These results might be soon employed to deliver highly automated diagnostic and prognostic strategies for cancer patients.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Sequence Analysis, RNA/methods , Transcriptome , Adenocarcinoma/diagnosis , Adenocarcinoma/metabolism , Algorithms , Biopsy , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Carbon/metabolism , Cluster Analysis , Gene Expression Profiling , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/metabolism , Metabolic Networks and Pathways , Pattern Recognition, Automated , Prognosis
7.
Bioinformatics ; 33(14): i311-i318, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28881985

ABSTRACT

MOTIVATION: Intratumour heterogeneity poses many challenges to the treatment of cancer. Unfortunately, the transcriptional and metabolic information retrieved by currently available computational and experimental techniques portrays the average behaviour of intermixed and heterogeneous cell subpopulations within a given tumour. Emerging single-cell genomic analyses are nonetheless unable to characterize the interactions among cancer subpopulations. In this study, we propose popFBA , an extension to classic Flux Balance Analysis, to explore how metabolic heterogeneity and cooperation phenomena affect the overall growth of cancer cell populations. RESULTS: We show how clones of a metabolic network of human central carbon metabolism, sharing the same stoichiometry and capacity constraints, may follow several different metabolic paths and cooperate to maximize the growth of the total population. We also introduce a method to explore the space of possible interactions, given some constraints on plasma supply of nutrients. We illustrate how alternative nutrients in plasma supply and/or a dishomogeneous distribution of oxygen provision may affect the landscape of heterogeneous phenotypes. We finally provide a technique to identify the most proliferative cells within the heterogeneous population. AVAILABILITY AND IMPLEMENTATION: the popFBA MATLAB function and the SBML model are available at https://github.com/BIMIB-DISCo/popFBA . CONTACT: chiara.damiani@unimib.it.


Subject(s)
Computational Biology/methods , Metabolic Networks and Pathways , Neoplasms/metabolism , Software , Cell Proliferation , Computer Simulation , Humans , Models, Biological , Neoplasms/physiopathology
8.
Comput Biol Chem ; 62: 60-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27085310

ABSTRACT

The metabolic rearrangements occurring in cancer cells can be effectively investigated with a Systems Biology approach supported by metabolic network modeling. We here present tissue-specific constraint-based core models for three different types of tumors (liver, breast and lung) that serve this purpose. The core models were extracted and manually curated from the corresponding genome-scale metabolic models in the Human Metabolic Atlas database with a focus on the pathways that are known to play a key role in cancer growth and proliferation. Along similar lines, we also reconstructed a core model from the original general human metabolic network to be used as a reference model. A comparative Flux Balance Analysis between the reference and the cancer models highlighted both a clear distinction between the two conditions and a heterogeneity within the three different cancer types in terms of metabolic flux distribution. These results emphasize the need for modeling approaches able to keep up with this tumoral heterogeneity in order to identify more suitable drug targets and develop effective treatments. According to this perspective, we identified key points able to reverse the tumoral phenotype toward the reference one or vice-versa.


Subject(s)
Computational Biology , Metabolic Networks and Pathways , Models, Biological , Neoplasms/metabolism , Humans , Neoplasms/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...