Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Appl Stat ; 49(9): 2349-2369, 2022.
Article in English | MEDLINE | ID: mdl-35755089

ABSTRACT

We develop and study a quantitative, interdisciplinary strategy for conducting statistical risk analyses within the 'benchmark risk' paradigm of contemporary risk assessment when potential autocorrelation exists among sample units. We use the methodology to explore information on vulnerability to natural hazards across 3108 counties in the conterminous 48 US states, applying a place-based resilience index to an existing knowledgebase of hazardous incidents and related human casualties. An extension of a centered autologistic regression model is applied to relate local, county-level vulnerability to hazardous outcomes. Adjustments for autocorrelation embedded in the resiliency information are applied via a novel, non-spatial neighborhood structure. Statistical risk-benchmarking techniques are then incorporated into the modeling framework, wherein levels of high and low vulnerability to hazards are identified.

2.
Fire (Basel) ; 5(1)2022 Feb.
Article in English | MEDLINE | ID: mdl-35295881

ABSTRACT

Wildland fires produce smoke plumes that impact air quality and human health. To understand the effects of wildland fire smoke on humans, the amount and composition of the smoke plume must be quantified. Using a fire emissions inventory is one way to determine the emissions rate and composition of smoke plumes from individual fires. There are multiple fire emissions inventories, and each uses a different method to estimate emissions. This paper presents a comparison of four emissions inventories and their products: Fire INventory from NCAR (FINN version 1.5), Global Fire Emissions Database (GFED version 4s), Missoula Fire Labs Emissions Inventory (MFLEI (250 m) and MFLEI (10 km) products), and Wildland Fire Emissions Inventory System (WFEIS (MODIS) and WFEIS (MTBS) products). The outputs from these inventories are compared directly. Because there are no validation datasets for fire emissions, the outlying points from the Bayesian models developed for each inventory were compared with visible images and fire radiative power (FRP) data from satellite remote sensing. This comparison provides a framework to check fire emissions inventory data against additional data by providing a set of days to investigate closely. Results indicate that FINN and GFED likely underestimate emissions, while the MFLEI products likely overestimate emissions. No fire emissions inventory matched the temporal distribution of emissions from an external FRP dataset. A discussion of the differences impacting the emissions estimates from the four fire emissions inventories is provided, including a qualitative comparison of the methods and inputs used by each inventory and the associated strengths and limitations.

3.
Sci Rep ; 10(1): 4188, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32144346

ABSTRACT

With the explosion of high-throughput data, effective integrative analyses are needed to decipher the knowledge accumulated in biological databases. Existing meta-analysis approaches in systems biology often focus on hypothesis testing and neglect real expression changes, i.e. effect sizes, across independent studies. In addition, most integrative tools completely ignore the topological order of gene regulatory networks that hold key characteristics in understanding biological processes. Here we introduce a novel meta-analysis framework, Network-Based Integrative Analysis (NBIA), that transforms the challenging meta-analysis problem into a set of standard pathway analysis problems that have been solved efficiently. NBIA utilizes techniques from classical and modern meta-analysis, as well as a network-based analysis, in order to identify patterns of genes and networks that are consistently impacted across multiple studies. We assess the performance of NBIA by comparing it with nine meta-analysis approaches: Impact Analysis, GSA, and GSEA combined with classical meta-analysis methods (Fisher's and the additive method), plus the three MetaPath approaches that employ multiple datasets. The 10 approaches have been tested on 1,737 samples from 27 expression datasets related to Alzheimer's disease, acute myeloid leukemia (AML), and influenza. For all of the three diseases, NBIA consistently identifies biological pathways relevant to the underlying diseases while the other 9 methods fail to capture the key phenomena. The identified AML signature is also validated on a completely independent cohort of 167 AML patients. In this independent cohort, the proposed signature identifies two groups of patients that have significantly different survival profiles (Cox p-value 2 × 10-6). The NBIA framework will be included in the next release of BLMA Bioconductor package (http://bioconductor.org/packages/release/bioc/html/BLMA.html).

4.
Brief Bioinform ; 20(3): 789-805, 2019 05 21.
Article in English | MEDLINE | ID: mdl-29272327

ABSTRACT

The development of computational methods capable of analyzing -omics data at the individual level is critical for the success of precision medicine. Although unprecedented opportunities now exist to gather data on an individual's -omics profile ('personalome'), interpreting and extracting meaningful information from single-subject -omics remain underdeveloped, particularly for quantitative non-sequence measurements, including complete transcriptome or proteome expression and metabolite abundance. Conventional bioinformatics approaches have largely been designed for making population-level inferences about 'average' disease processes; thus, they may not adequately capture and describe individual variability. Novel approaches intended to exploit a variety of -omics data are required for identifying individualized signals for meaningful interpretation. In this review-intended for biomedical researchers, computational biologists and bioinformaticians-we survey emerging computational and translational informatics methods capable of constructing a single subject's 'personalome' for predicting clinical outcomes or therapeutic responses, with an emphasis on methods that provide interpretable readouts. Key points: (i) the single-subject analytics of the transcriptome shows the greatest development to date and, (ii) the methods were all validated in simulations, cross-validations or independent retrospective data sets. This survey uncovers a growing field that offers numerous opportunities for the development of novel validation methods and opens the door for future studies focusing on the interpretation of comprehensive 'personalomes' through the integration of multiple -omics, providing valuable insights into individual patient outcomes and treatments.


Subject(s)
Precision Medicine , Transcriptome , Humans
5.
J R Stat Soc Ser A Stat Soc ; 181(3): 803-823, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29904240

ABSTRACT

We develop a quantitative methodology to characterize vulnerability among 132 U.S. urban centers ('cities') to terrorist events, applying a place-based vulnerability index to a database of terrorist incidents and related human casualties. A centered autologistic regression model is employed to relate urban vulnerability to terrorist outcomes and also to adjust for autocorrelation in the geospatial data. Risk-analytic 'benchmark' techniques are then incorporated into the modeling framework, wherein levels of high and low urban vulnerability to terrorism are identified. This new, translational adaptation of the risk-benchmark approach, including its ability to account for geospatial autocorrelation, is seen to operate quite flexibly in this socio-geographic setting.

6.
Pac Symp Biocomput ; 23: 484-495, 2018.
Article in English | MEDLINE | ID: mdl-29218907

ABSTRACT

Recent precision medicine initiatives have led to the expectation of improved clinical decisionmaking anchored in genomic data science. However, over the last decade, only a handful of new single-gene product biomarkers have been translated to clinical practice (FDA approved) in spite of considerable discovery efforts deployed and a plethora of transcriptomes available in the Gene Expression Omnibus. With this modest outcome of current approaches in mind, we developed a pilot simulation study to demonstrate the untapped benefits of developing disease detection methods for cases where the true signal lies at the pathway level, even if the pathway's gene expression alterations may be heterogeneous across patients. In other words, we relaxed the crosspatient homogeneity assumption from the transcript level (cohort assumptions of deregulated gene expression) to the pathway level (assumptions of deregulated pathway expression). Furthermore, we have expanded previous single-subject (SS) methods into cohort analyses to illustrate the benefit of accounting for an individual's variability in cohort scenarios. We compare SS and cohort-based (CB) techniques under 54 distinct scenarios, each with 1,000 simulations, to demonstrate that the emergence of a pathway-level signal occurs through the summative effect of its altered gene expression, heterogeneous across patients. Studied variables include pathway gene set size, fraction of expressed gene responsive within gene set, fraction of expressed gene responsive up- vs down-regulated, and cohort size. We demonstrated that our SS approach was uniquely suited to detect signals in heterogeneous populations in which individuals have varying levels of baseline risks that are simultaneously confounded by patient-specific "genome -by-environment" interactions (G×E). Area under the precision-recall curve of the SS approach far surpassed that of the CB (1st quartile, median, 3rd quartile: SS = 0.94, 0.96, 0.99; CB= 0.50, 0.52, 0.65). We conclude that single-subject pathway detection methods are uniquely suited for consistently detecting pathway dysregulation by the inclusion of a patient's individual variability. http://www.lussiergroup.org/publications/PathwayMarker/.


Subject(s)
Gene Expression Profiling/statistics & numerical data , Genetic Markers , Transcriptome , Cohort Studies , Computational Biology/methods , Computer Simulation , Databases, Nucleic Acid/statistics & numerical data , Gene Regulatory Networks , Gene-Environment Interaction , Humans , Models, Genetic , Pilot Projects , Precision Medicine , Systems Biology , Translational Research, Biomedical
7.
Stat Methods Med Res ; 27(12): 3797-3813, 2018 12.
Article in English | MEDLINE | ID: mdl-28552011

ABSTRACT

Modern precision medicine increasingly relies on molecular data analytics, wherein development of interpretable single-subject ("N-of-1") signals is a challenging goal. A previously developed global framework, N-of-1- pathways, employs single-subject gene expression data to identify differentially expressed gene set pathways in an individual patient. Unfortunately, the limited amount of data within the single-subject, N-of-1 setting makes construction of suitable statistical inferences for identifying differentially expressed gene set pathways difficult, especially when non-trivial inter-gene correlation is present. We propose a method that exploits external information on gene expression correlations to cluster positively co-expressed genes within pathways, then assesses differential expression across the clusters within a pathway. A simulation study illustrates that the cluster-based approach exhibits satisfactory false-positive error control and reasonable power to detect differentially expressed gene set pathways. An example with a single N-of-1 patient's triple negative breast cancer data illustrates use of the methodology.


Subject(s)
Gene Expression Profiling/statistics & numerical data , Models, Statistical , Triple Negative Breast Neoplasms/genetics , Algorithms , Computer Simulation , Female , Humans , Monte Carlo Method , Precision Medicine , Sequence Analysis, RNA
8.
J Am Med Inform Assoc ; 24(6): 1116-1126, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29016970

ABSTRACT

OBJECTIVE: To introduce a disease prognosis framework enabled by a robust classification scheme derived from patient-specific transcriptomic response to stimulation. MATERIALS AND METHODS: Within an illustrative case study to predict asthma exacerbation, we designed a stimulation assay that reveals individualized transcriptomic response to human rhinovirus. Gene expression from peripheral blood mononuclear cells was quantified from 23 pediatric asthmatic patients and stimulated in vitro with human rhinovirus. Responses were obtained via the single-subject gene set testing methodology "N-of-1-pathways." The classifier was trained on a related independent training dataset (n = 19). Novel visualizations of personal transcriptomic responses are provided. RESULTS: Of the 23 pediatric asthmatic patients, 12 experienced recurrent exacerbations. Our classifier, using individualized responses and trained on an independent dataset, obtained 74% accuracy (area under the receiver operating curve of 71%; 2-sided P = .039). Conventional classifiers using messenger RNA (mRNA) expression within the viral-exposed samples were unsuccessful (all patients predicted to have recurrent exacerbations; accuracy of 52%). DISCUSSION: Prognosis based on single time point, static mRNA expression alone neglects the importance of dynamic genome-by-environment interplay in phenotypic presentation. Individualized transcriptomic response quantified at the pathway (gene sets) level reveals interpretable signals related to clinical outcomes. CONCLUSION: The proposed framework provides an innovative approach to precision medicine. We show that quantifying personal pathway-level transcriptomic response to a disease-relevant environmental challenge predicts disease progression. This genome-by-environment interaction assay offers a noninvasive opportunity to translate omics data to clinical practice by improving the ability to predict disease exacerbation and increasing the potential to produce more effective treatment decisions.


Subject(s)
Asthma/genetics , Gene-Environment Interaction , Precision Medicine , Transcriptome , Asthma/classification , Bayes Theorem , Child , Datasets as Topic , Decision Trees , Disease Progression , Female , Humans , Leukocytes, Mononuclear/metabolism , Male , Models, Statistical , Patient-Specific Modeling , Prognosis , RNA, Messenger/metabolism , ROC Curve , Rhinovirus , Support Vector Machine , Transcriptome/immunology , Transcriptome/physiology
9.
BMC Med Genomics ; 10(Suppl 1): 27, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28589853

ABSTRACT

BACKGROUND: Transcriptome analytic tools are commonly used across patient cohorts to develop drugs and predict clinical outcomes. However, as precision medicine pursues more accurate and individualized treatment decisions, these methods are not designed to address single-patient transcriptome analyses. We previously developed and validated the N-of-1-pathways framework using two methods, Wilcoxon and Mahalanobis Distance (MD), for personal transcriptome analysis derived from a pair of samples of a single patient. Although, both methods uncover concordantly dysregulated pathways, they are not designed to detect dysregulated pathways with up- and down-regulated genes (bidirectional dysregulation) that are ubiquitous in biological systems. RESULTS: We developed N-of-1-pathways MixEnrich, a mixture model followed by a gene set enrichment test, to uncover bidirectional and concordantly dysregulated pathways one patient at a time. We assess its accuracy in a comprehensive simulation study and in a RNA-Seq data analysis of head and neck squamous cell carcinomas (HNSCCs). In presence of bidirectionally dysregulated genes in the pathway or in presence of high background noise, MixEnrich substantially outperforms previous single-subject transcriptome analysis methods, both in the simulation study and the HNSCCs data analysis (ROC Curves; higher true positive rates; lower false positive rates). Bidirectional and concordant dysregulated pathways uncovered by MixEnrich in each patient largely overlapped with the quasi-gold standard compared to other single-subject and cohort-based transcriptome analyses. CONCLUSION: The greater performance of MixEnrich presents an advantage over previous methods to meet the promise of providing accurate personal transcriptome analysis to support precision medicine at point of care.


Subject(s)
Gene Expression Profiling/methods , Head and Neck Neoplasms/genetics , Humans , Neoplasms, Squamous Cell/genetics , Precision Medicine , ROC Curve
10.
J Biomed Inform ; 66: 32-41, 2017 02.
Article in English | MEDLINE | ID: mdl-28007582

ABSTRACT

MOTIVATION: Understanding dynamic, patient-level transcriptomic response to therapy is an important step forward for precision medicine. However, conventional transcriptome analysis aims to discover cohort-level change, lacking the capacity to unveil patient-specific response to therapy. To address this gap, we previously developed two N-of-1-pathways methods, Wilcoxon and Mahalanobis distance, to detect unidirectionally responsive transcripts within a pathway using a pair of samples from a single subject. Yet, these methods cannot recognize bidirectionally (up and down) responsive pathways. Further, our previous approaches have not been assessed in presence of background noise and are not designed to identify differentially expressed mRNAs between two samples of a patient taken in different contexts (e.g. cancer vs non cancer), which we termed responsive transcripts (RTs). METHODS: We propose a new N-of-1-pathways method, k-Means Enrichment (kMEn), that detects bidirectionally responsive pathways, despite background noise, using a pair of transcriptomes from a single patient. kMEn identifies transcripts responsive to the stimulus through k-means clustering and then tests for an over-representation of the responsive genes within each pathway. The pathways identified by kMEn are mechanistically interpretable pathways significantly responding to a stimulus. RESULTS: In ∼9000 simulations varying six parameters, superior performance of kMEn over previous single-subject methods is evident by: (i) improved precision-recall at various levels of bidirectional response and (ii) lower rates of false positives (1-specificity) when more than 10% of genes in the genome are differentially expressed (background noise). In a clinical proof-of-concept, personal treatment-specific pathways identified by kMEn correlate with therapeutic response (p-value<0.01). CONCLUSION: Through improved single-subject transcriptome dynamics of bidirectionally-regulated signals, kMEn provides a novel approach to identify mechanism-level biomarkers.


Subject(s)
Gene Expression Profiling , Precision Medicine , Transcriptome , Cluster Analysis , Data Interpretation, Statistical , Humans , RNA, Messenger
11.
Bioinformatics ; 32(12): i80-i89, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27307648

ABSTRACT

MOTIVATION: As 'omics' biotechnologies accelerate the capability to contrast a myriad of molecular measurements from a single cell, they also exacerbate current analytical limitations for detecting meaningful single-cell dysregulations. Moreover, mRNA expression alone lacks functional interpretation, limiting opportunities for translation of single-cell transcriptomic insights to precision medicine. Lastly, most single-cell RNA-sequencing analytic approaches are not designed to investigate small populations of cells such as circulating tumor cells shed from solid tumors and isolated from patient blood samples. RESULTS: In response to these characteristics and limitations in current single-cell RNA-sequencing methodology, we introduce an analytic framework that models transcriptome dynamics through the analysis of aggregated cell-cell statistical distances within biomolecular pathways. Cell-cell statistical distances are calculated from pathway mRNA fold changes between two cells. Within an elaborate case study of circulating tumor cells derived from prostate cancer patients, we develop analytic methods of aggregated distances to identify five differentially expressed pathways associated to therapeutic resistance. Our aggregation analyses perform comparably with Gene Set Enrichment Analysis and better than differentially expressed genes followed by gene set enrichment. However, these methods were not designed to inform on differential pathway expression for a single cell. As such, our framework culminates with the novel aggregation method, cell-centric statistics (CCS). CCS quantifies the effect size and significance of differentially expressed pathways for a single cell of interest. Improved rose plots of differentially expressed pathways in each cell highlight the utility of CCS for therapeutic decision-making. AVAILABILITY AND IMPLEMENTATION: http://www.lussierlab.org/publications/CCS/ CONTACT: yves@email.arizona.edu or piegorsch@math.arizona.edu SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Drug Resistance, Neoplasm , Neoplastic Cells, Circulating/drug effects , Sequence Analysis, RNA , Transcriptome , Gene Expression Profiling , Humans , Male , Prostatic Neoplasms/drug therapy , RNA
12.
Bioinformatics ; 31(12): i293-302, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26072495

ABSTRACT

MOTIVATION: The conventional approach to personalized medicine relies on molecular data analytics across multiple patients. The path to precision medicine lies with molecular data analytics that can discover interpretable single-subject signals (N-of-1). We developed a global framework, N-of-1-pathways, for a mechanistic-anchored approach to single-subject gene expression data analysis. We previously employed a metric that could prioritize the statistical significance of a deregulated pathway in single subjects, however, it lacked in quantitative interpretability (e.g. the equivalent to a gene expression fold-change). RESULTS: In this study, we extend our previous approach with the application of statistical Mahalanobis distance (MD) to quantify personal pathway-level deregulation. We demonstrate that this approach, N-of-1-pathways Paired Samples MD (N-OF-1-PATHWAYS-MD), detects deregulated pathways (empirical simulations), while not inflating false-positive rate using a study with biological replicates. Finally, we establish that N-OF-1-PATHWAYS-MD scores are, biologically significant, clinically relevant and are predictive of breast cancer survival (P < 0.05, n = 80 invasive carcinoma; TCGA RNA-sequences). CONCLUSION: N-of-1-pathways MD provides a practical approach towards precision medicine. The method generates the magnitude and the biological significance of personal deregulated pathways results derived solely from the patient's transcriptome. These pathways offer the opportunities for deriving clinically actionable decisions that have the potential to complement the clinical interpretability of personal polymorphisms obtained from DNA acquired or inherited polymorphisms and mutations. In addition, it offers an opportunity for applicability to diseases in which DNA changes may not be relevant, and thus expand the 'interpretable 'omics' of single subjects (e.g. personalome). AVAILABILITY AND IMPLEMENTATION: http://www.lussierlab.net/publications/N-of-1-pathways.


Subject(s)
Breast Neoplasms/mortality , Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Data Interpretation, Statistical , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Precision Medicine
SELECTION OF CITATIONS
SEARCH DETAIL
...