PhenoWin – An R Shiny application for visualization and extraction of phenological windows in Germany

2020 ◽  
Vol 175 ◽  
pp. 105534
Author(s):  
Markus Möller ◽  
Lucas Boutarfa ◽  
Jörn Strassemeyer
Keyword(s):  
R Shiny ◽  
2019 ◽  
Author(s):  
Shuoguo Wang ◽  
Constance Brett ◽  
Mohan Bolisetty ◽  
Ryan Golhar ◽  
Isaac Neuhaus ◽  
...  

AbstractMotivationThanks to technological advances made in the last few years, we are now able to study transcriptomes from thousands of single cells. These have been applied widely to study various aspects of Biology. Nevertheless, comprehending and inferring meaningful biological insights from these large datasets is still a challenge. Although tools are being developed to deal with the data complexity and data volume, we do not have yet an effective visualizations and comparative analysis tools to realize the full value of these datasets.ResultsIn order to address this gap, we implemented a single cell data visualization portal called Single Cell Viewer (SCV). SCV is an R shiny application that offers users rich visualization and exploratory data analysis options for single cell datasets.AvailabilitySource code for the application is available online at GitHub (http://www.github.com/neuhausi/single-cell-viewer) and there is a hosted exploration application using the same example dataset as this publication at http://periscopeapps.org/[email protected]; [email protected]


2021 ◽  
Author(s):  
Theodoros Evrenoglou ◽  
Isabelle Boutron ◽  
Anna Chaimani

Abstract“Living” evidence synthesis is of primary interest for decision-makers to overcome the COVID-19 pandemic. The COVID-NMA provides open-access living meta-analyses assessing different therapeutic and preventive interventions. Data are posted on a platform (https://covid-nma.com/) and analyses are updated every week. However, guideline developers and other stakeholders also need to investigate the data and perform their own analyses. This requires resources, time, statistical expertise, and software knowledge. To assist them, we created the “metaCOVID” application which, based on automation processes, facilitates the fast exploration of the data and the conduct of analyses tailored to end-users needs. metaCOVID has been created in R and is freely available as an R-Shiny application. The application conducts living meta-analyses for every outcome. Several options are available for subgroup and sensitivity analyses. The results are presented in downloadable forest plots. metaCOVID is freely available from https://covid-nma.com/metacovid/ and the source code from https://github.com/TEvrenoglou/metaCovid.


2020 ◽  
Vol 36 (9) ◽  
pp. 2932-2933 ◽  
Author(s):  
Angela Serra ◽  
Laura Aliisa Saarimäki ◽  
Michele Fratello ◽  
Veer Singh Marwah ◽  
Dario Greco

Abstract 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.


2020 ◽  
Vol 36 (11) ◽  
pp. 3582-3584
Author(s):  
Nathan Lawlor ◽  
Eladio J Marquez ◽  
Donghyung Lee ◽  
Duygu Ucar

Abstract Summary Single-cell RNA-sequencing (scRNA-seq) technology enables studying gene expression programs from individual cells. However, these data are subject to diverse sources of variation, including ‘unwanted’ variation that needs to be removed in downstream analyses (e.g. batch effects) and ‘wanted’ or biological sources of variation (e.g. variation associated with a cell type) that needs to be precisely described. Surrogate variable analysis (SVA)-based algorithms, are commonly used for batch correction and more recently for studying ‘wanted’ variation in scRNA-seq data. However, interpreting whether these variables are biologically meaningful or stemming from technical reasons remains a challenge. To facilitate the interpretation of surrogate variables detected by algorithms including IA-SVA, SVA or ZINB-WaVE, we developed an R Shiny application [Visual Surrogate Variable Analysis (V-SVA)] that provides a web-browser interface for the identification and annotation of hidden sources of variation in scRNA-seq data. This interactive framework includes tools for discovery of genes associated with detected sources of variation, gene annotation using publicly available databases and gene sets, and data visualization using dimension reduction methods. Availability and implementation The V-SVA Shiny application is publicly hosted at https://vsva.jax.org/ and the source code is freely available at https://github.com/nlawlor/V-SVA. Contact [email protected] or [email protected] Supplementary information Supplementary data are available at Bioinformatics online.


2018 ◽  
Vol 18 (2) ◽  
pp. 241-243 ◽  
Author(s):  
Filipe Inácio Matias ◽  
Italo Granato ◽  
Roberto Fritsche-Neto

2019 ◽  
Author(s):  
Jorge Cimentada ◽  
Sebastian Klüsener ◽  
Tim Riffe

Lexis surfaces are widely used to analyze demographic trends across periods, ages, and birth cohorts. When used to visualize rates or similar, these plots usually do not convey information about population size. The failure to communicate population size in Lexis surfaces can lead to misinterpretations of the mortality conditions populations face because, for example, high mortality rates at very high ages have historically been experienced by only a small proportion of a population or cohort. We propose enhanced Lexis surfaces that include a visual representation of population size. The examples we present demonstrate how such plots can give readers a more intuitive understanding of the demographic development of a population over time. Visualizations are implemented using an R-Shiny application, building upon perception theories. We present example plots for enhanced Lexis surfaces that show trends in cohort mortality and first-order differences in cohort mortality developments. These plots illustrate how adding the cohort size dimension allows us to extend the analytical potential of standard Lexis surfaces. Our enhanced Lexis surfaces improve conventional depictions of period, age, and cohort trends in demographic developments of populations and cohorts. An online interactive visualization tool based on Human Mortality Database data allows users to generate and export enhanced Lexis surfaces for their research. The R code to generate the application (and a link to the deployed application) can be accessed at https://github.com/cimentadaj/lexis_plot.


F1000Research ◽  
2017 ◽  
Vol 6 ◽  
pp. 2046 ◽  
Author(s):  
Lauren Kemperman ◽  
Matthew N. McCall

The miRcomp-Shiny web application allows interactive performance assessments and comparisons of qPCR-based microRNA expression and quality estimation methods using a benchmark data set. This work is motivated by two distinct use cases: (1) selection of methodology and quality thresholds for use analyzing one's own data, and (2) comparison of novel expression estimation algorithms with currently-available methodology. The miRcomp-Shiny application is implemented in the R/Shiny language and can be installed on any operating system on which R can be installed. It is made freely available as part of the miRcomp package (version 1.3.3 and later) available through the Bioconductor project at: http://bioconductor.org/packages/miRcomp. The web application is hosted at https://laurenkemperman.shinyapps.io/mircomp/. A detailed description of how to use the web application is available at: http://lkemperm.github.io/miRcomp_shiny_app


2021 ◽  
Author(s):  
Hamzah Syed ◽  
Georg W Otto ◽  
Daniel Kelberman ◽  
Chiara Bacchelli ◽  
Philip L Beales

Background: Multi-omics studies are increasingly used to help understand the underlying mechanisms of clinical phenotypes, integrating information from the genome, transcriptome, epigenome, metabolome, proteome and microbiome. This integration of data is of particular use in rare disease studies where the sample sizes are often relatively small. Methods development for multi-omics studies is in its early stages due to the complexity of the different individual data types. There is a need for software to perform data simulation and power calculation for multi-omics studies to test these different methodologies and help calculate sample size before the initiation of a study. This software, in turn, will optimise the success of a study. Results: The interactive R shiny application MOPower described below simulates data based on three different omics using statistical distributions. It calculates the power to detect an association with the phenotype through analysis of n number of replicates using a variety of the latest multi-omics analysis models and packages. The simulation study confirms the efficiency of the software when handling thousands of simulations over ten different sample sizes. The average time elapsed for a power calculation run between integration models was approximately 500 seconds. Additionally, for the given study design model, power varied with the increase in the number of features affecting each method differently. For example, using MOFA had an increase in power to detect an association when the study sample size equally matched the number of features. Conclusions: MOPower addresses the need for flexible and user-friendly software that undertakes power calculations for multi-omics studies. MOPower offers users a wide variety of integration methods to test and full customisation of omics features to cover a range of study designs.


Author(s):  
Jeremiah Rounds ◽  
Lauren Charles-Smith ◽  
Courtney D. Corley

ObjectiveTo introduce Soda Pop, an R/Shiny application designed to be adisease agnostic time-series clustering, alarming, and forecastingtool to assist in disease surveillance “triage, analysis and reporting”workflows within the Biosurveillance Ecosystem (BSVE) [1]. In thisposter, we highlight the new capabilities that are brought to the BSVEby Soda Pop with an emphasis on the impact of metholodogicaldecisions.IntroductionThe Biosurveillance Ecosystem (BSVE) is a biological andchemical threat surveillance system sponsored by the Defense ThreatReduction Agency (DTRA). BSVE is intended to be user-friendly,multi-agency, cooperative, modular and threat agnostic platformfor biosurveillance [2]. In BSVE, a web-based workbench presentsthe analyst with applications (apps) developed by various DTRAfundedresearchers, which are deployed on-demand in the cloud(e.g., Amazon Web Services). These apps aim to address emergingneeds and refine capabilities to enable early warning of chemical andbiological threats for multiple users across local, state, and federalagencies.Soda Pop is an app developed by Pacific Northwest NationalLaboratory (PNNL) to meet the current needs of the BSVE forearly warning and detection of disease outbreaks. Aimed for use bya diverse set of analysts, the application is agnostic to data sourceand spatial scale enabling it to be generalizable across many diseasesand locations. To achieve this, we placed a particular emphasis onclustering and alerting of disease signals within Soda Pop withoutstrong prior assumptions on the nature of observed diseased counts.MethodsAlthough designed to be agnostic to the data source, Soda Pop wasinitially developed and tested on data summarizing Influenza-LikeIllness in military hospitals from collaboration with the Armed ForcesHealth Surveillance Branch. Currently, the data incorporated alsoincludes the CDC’s National Notifiable Diseases Surveillance System(NNDSS) tables [3] and the WHO’s Influenza A/B Influenza Data(Flunet) [4]. These data sources are now present in BSVE’s Postgresdata storage for direct access.Soda Pop is designed to automate time-series tasks of datasummarization, exploration, clustering, alarming and forecasting.Built as an R/Shiny application, Soda Pop is founded on the powerfulstatistical tool R [5]. Where applicable, Soda Pop facilitates nonparametricseasonal decomposition of time-series; hierarchicalagglomerative clustering across reporting areas and between diseaseswithin reporting areas; and a variety of alarming techniques includingExponential Weighted Moving Average alarms and Early AberrationDetection [6].Soda Pop embeds these techniques within a user-interface designedto enhance an analyst’s understanding of emerging trends in their dataand enables the inclusion of its graphical elements into their dossierfor further tracking and reporting. The ultimate goal of this softwareis to facilitate the discovery of unknown disease signals along withincreasing the speed of detection of unusual patterns within thesesignals.ConclusionsSoda Pop organizes common statistical disease surveillance tasksin a manner integrated with BSVE data source inputs and outputs.The app analyzes time-series disease data and supports a robust set ofclustering and alarming routines that avoid strong assumptions on thenature of observed disease counts. This attribute allows for flexibilityin the data source, spatial scale, and disease types making it useful toa wide range of analystsSoda Pop within the BSVE.KeywordsBSVE; Biosurveillance; R/Shiny; Clustering; AlarmingAcknowledgmentsThis work was supported by the Defense Threat Reduction Agency undercontract CB10082 with Pacific Northwest National LaboratoryReferences1. Dasey, Timothy, et al. “Biosurveillance Ecosystem (BSVE) WorkflowAnalysis.” Online journal of public health informatics 5.1 (2013).2. http://www.defense.gov/News/Article/Article/681832/dtra-scientistsdevelop-cloud-based-biosurveillance-ecosystem. Accessed 9/6/2016.3. Centers for Disease Control and Prevention. “National NotifiableDiseases Surveillance System (NNDSS).”4. World Health Organization. “FluNet.” Global Influenza Surveillanceand Response System (GISRS).5. R Core Team (2016). R: A language and environment for statisticalcomputing. R Foundation for Statistical Computing, Vienna, Austria.6. Salmon, Maëlle, et al. “Monitoring Count Time Series in R: AberrationDetection in Public Health Surveillance.” Journal of StatisticalSoftware [Online], 70.10 (2016): 1 - 35.


Sign in / Sign up

Export Citation Format

Share Document