Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
bioRxiv ; 2024 May 01.
Article in English | MEDLINE | ID: mdl-38585820

ABSTRACT

The OmicsFootPrint framework addresses the need for advanced multi-omics data analysis methodologies by transforming data into intuitive two-dimensional circular images and facilitating the interpretation of complex diseases. Utilizing Deep Neural Networks and incorporating the SHapley Additive exPlanations (SHAP) algorithm, the framework enhances model interpretability. Tested with The Cancer Genome Atlas (TCGA) data, OmicsFootPrint effectively classified lung and breast cancer subtypes, achieving high Area Under Curve (AUC) scores - 0.98±0.02 for lung cancer subtype differentiation, 0.83±0.07 for breast cancer PAM50 subtypes, and successfully distinguishe between invasive lobular and ductal carcinomas in breast cancer, showcasing its robustness. It also demonstrated notable performance in predicting drug responses in cancer cell lines, with a median AUC of 0.74, surpassing existing algorithms. Furthermore, its effectiveness persists even with reduced training sample sizes. OmicsFootPrint marks an enhancement in multi-omics research, offering a novel, efficient, and interpretable approach that contributes to a deeper understanding of disease mechanisms.

2.
J Vasc Surg ; 80(1): 251-259.e3, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38417709

ABSTRACT

OBJECTIVE: Patients with diabetes mellitus (DM) are at increased risk for peripheral artery disease (PAD) and its complications. Arterial calcification and non-compressibility may limit test interpretation in this population. Developing tools capable of identifying PAD and predicting major adverse cardiac event (MACE) and limb event (MALE) outcomes among patients with DM would be clinically useful. Deep neural network analysis of resting Doppler arterial waveforms was used to detect PAD among patients with DM and to identify those at greatest risk for major adverse outcome events. METHODS: Consecutive patients with DM undergoing lower limb arterial testing (April 1, 2015-December 30, 2020) were randomly allocated to training, validation, and testing subsets (60%, 20%, and 20%). Deep neural networks were trained on resting posterior tibial arterial Doppler waveforms to predict all-cause mortality, MACE, and MALE at 5 years using quartiles based on the distribution of the prediction score. RESULTS: Among 11,384 total patients, 4211 patients with DM met study criteria (mean age, 68.6 ± 11.9 years; 32.0% female). After allocating the training and validation subsets, the final test subset included 856 patients. During follow-up, there were 262 deaths, 319 MACE, and 99 MALE. Patients in the upper quartile of prediction based on deep neural network analysis of the posterior tibial artery waveform provided independent prediction of death (hazard ratio [HR], 3.58; 95% confidence interval [CI], 2.31-5.56), MACE (HR, 2.06; 95% CI, 1.49-2.91), and MALE (HR, 13.50; 95% CI, 5.83-31.27). CONCLUSIONS: An artificial intelligence enabled analysis of a resting Doppler arterial waveform permits identification of major adverse outcomes including all-cause mortality, MACE, and MALE among patients with DM.


Subject(s)
Peripheral Arterial Disease , Predictive Value of Tests , Ultrasonography, Doppler , Humans , Male , Female , Aged , Peripheral Arterial Disease/physiopathology , Peripheral Arterial Disease/diagnostic imaging , Peripheral Arterial Disease/mortality , Peripheral Arterial Disease/complications , Risk Assessment , Middle Aged , Risk Factors , Deep Learning , Reproducibility of Results , Prognosis , Aged, 80 and over , Time Factors , Tibial Arteries/diagnostic imaging , Tibial Arteries/physiopathology , Diabetic Angiopathies/physiopathology , Diabetic Angiopathies/diagnostic imaging , Diabetic Angiopathies/mortality , Diabetic Angiopathies/diagnosis
3.
J Am Heart Assoc ; 13(3): e031880, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38240202

ABSTRACT

BACKGROUND: Patients with peripheral artery disease are at increased risk for major adverse cardiac events, major adverse limb events, and all-cause death. Developing tools capable of identifying those patients with peripheral artery disease at greatest risk for major adverse events is the first step for outcome prevention. This study aimed to determine whether computer-assisted analysis of a resting Doppler waveform using deep neural networks can accurately identify patients with peripheral artery disease at greatest risk for adverse outcome events. METHODS AND RESULTS: Consecutive patients (April 1, 2015, to December 31, 2020) undergoing ankle-brachial index testing were included. Patients were randomly allocated to training, validation, and testing subsets (60%/20%/20%). Deep neural networks were trained on resting posterior tibial arterial Doppler waveforms to predict major adverse cardiac events, major adverse limb events, and all-cause death at 5 years. Patients were then analyzed in groups based on the quartiles of each prediction score in the training set. Among 11 384 total patients, 10 437 patients met study inclusion criteria (mean age, 65.8±14.8 years; 40.6% women). The test subset included 2084 patients. During 5 years of follow-up, there were 447 deaths, 585 major adverse cardiac events, and 161 MALE events. After adjusting for age, sex, and Charlson comorbidity index, deep neural network analysis of the posterior tibial artery waveform provided independent prediction of death (hazard ratio [HR], 2.44 [95% CI, 1.78-3.34]), major adverse cardiac events (HR, 1.97 [95% CI, 1.49-2.61]), and major adverse limb events (HR, 11.03 [95% CI, 5.43-22.39]) at 5 years. CONCLUSIONS: An artificial intelligence-enabled analysis of Doppler arterial waveforms enables identification of major adverse outcomes among patients with peripheral artery disease, which may promote early adoption and adherence of risk factor modification.


Subject(s)
Artificial Intelligence , Peripheral Arterial Disease , Humans , Female , Middle Aged , Aged , Aged, 80 and over , Male , Peripheral Arterial Disease/diagnostic imaging , Risk Factors
4.
Vasc Med ; 27(4): 333-342, 2022 08.
Article in English | MEDLINE | ID: mdl-35535982

ABSTRACT

BACKGROUND: Patients with peripheral artery disease (PAD) are at increased risk for major adverse limb and cardiac events including mortality. Developing screening tools capable of accurate PAD identification is a necessary first step for strategies of adverse outcome prevention. This study aimed to determine whether machine analysis of a resting Doppler waveform using deep neural networks can accurately identify patients with PAD. METHODS: Consecutive patients (4/8/2015 - 12/31/2020) undergoing rest and postexercise ankle-brachial index (ABI) testing were included. Patients were randomly allocated to training, validation, and testing subsets (70%/15%/15%). Deep neural networks were trained on resting posterior tibial arterial Doppler waveforms to predict normal (> 0.9) or PAD (⩽ 0.9) using rest and postexercise ABI. A separate dataset of 151 patients who underwent testing during a period after the model had been created and validated (1/1/2021 - 3/31/2021) was used for secondary validation. Area under the receiver operating characteristic curves (AUC) were constructed to evaluate test performance. RESULTS: Among 11,748 total patients, 3432 patients met study criteria: 1941 with PAD (mean age 69 ± 12 years) and 1491 without PAD (64 ± 14 years). The predictive model with highest performance identified PAD with an AUC 0.94 (CI = 0.92-0.96), sensitivity 0.83, specificity 0.88, accuracy 0.85, and positive predictive value (PPV) 0.90. Results were similar for the validation dataset: AUC 0.94 (CI = 0.91-0.98), sensitivity 0.91, specificity 0.85, accuracy 0.89, and PPV 0.89 (postexercise ABI comparison). CONCLUSION: An artificial intelligence-enabled analysis of a resting Doppler arterial waveform permits identification of PAD at a clinically relevant performance level.


Subject(s)
Ankle Brachial Index , Peripheral Arterial Disease , Aged , Aged, 80 and over , Ankle Brachial Index/methods , Arteries , Artificial Intelligence , Humans , Middle Aged , Peripheral Arterial Disease/diagnostic imaging , Predictive Value of Tests , Ultrasonography, Doppler
5.
J Pathol Inform ; 12: 21, 2021.
Article in English | MEDLINE | ID: mdl-34267986

ABSTRACT

BACKGROUND: Adoption of the Digital Imaging and Communications in Medicine (DICOM) standard for whole slide images (WSIs) has been slow, despite significant time and effort by standards curators. One reason for the lack of adoption is that there are few tools which exist that can meet the requirements of WSIs, given an evolving ecosystem of best practices for implementation. Eventually, vendors will conform to the specification to ensure enterprise interoperability, but what about archived slides? Millions of slides have been scanned in various proprietary formats, many with examples of rare histologies. Our hypothesis is that if users and developers had access to easy to use tools for migrating proprietary formats to the open DICOM standard, then more tools would be developed as DICOM first implementations. METHODS: The technology we present here is dicom_wsi, a Python based toolkit for converting any slide capable of being read by the OpenSlide library into DICOM conformant and validated implementations. Moreover, additional postprocessing such as background removal, digital transformations (e.g., ink removal), and annotation storage are also described. dicom_wsi is a free and open source implementation that anyone can use or modify to meet their specific purposes. RESULTS: We compare the output of dicom_wsi to two other existing implementations of WSI to DICOM converters and also validate the images using DICOM capable image viewers. CONCLUSION: dicom_wsi represents the first step in a long process of DICOM adoption for WSI. It is the first open source implementation released in the developer friendly Python programming language and can be freely downloaded at .

6.
PLoS One ; 16(5): e0250518, 2021.
Article in English | MEDLINE | ID: mdl-34033669

ABSTRACT

Gestational trophoblastic disease (GTD) is a heterogeneous group of lesions arising from placental tissue. Epithelioid trophoblastic tumor (ETT), derived from chorionic-type trophoblast, is the rarest form of GTD with only approximately 130 cases described in the literature. Due to its morphologic mimicry of epithelioid smooth muscle tumors and carcinoma, ETT can be misdiagnosed. To date, molecular characterization of ETTs is lacking. Furthermore, ETT is difficult to treat when disease spreads beyond the uterus. Here using RNA-Seq analysis in a cohort of ETTs and other gestational trophoblastic lesions we describe the discovery of LPCAT1-TERT fusion transcripts that occur in ETTs and coincide with underlying genomic deletions. Through cell-growth assays we demonstrate that LPCAT1-TERT fusion proteins can positively modulate cell proliferation and therefore may represent future treatment targets. Furthermore, we demonstrate that TERT upregulation appears to be a characteristic of ETTs, even in the absence of LPCAT1-TERT fusions, and that it appears linked to copy number gains of chromosome 5. No evidence of TERT upregulation was identified in other trophoblastic lesions tested, including placental site trophoblastic tumors and placental site nodules, which are thought to be the benign chorionic-type trophoblast counterpart to ETT. These findings indicate that LPCAT1-TERT fusions and copy-number driven TERT activation may represent novel markers for ETT, with the potential to improve the diagnosis, treatment, and outcome for women with this rare form of GTD.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/genetics , Epithelioid Cells/pathology , Gestational Trophoblastic Disease/etiology , Oncogene Proteins, Fusion/genetics , Telomerase/genetics , Trophoblastic Neoplasms/pathology , Uterine Neoplasms/pathology , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Adult , Biomarkers, Tumor/genetics , Cell Proliferation , Epithelioid Cells/metabolism , Female , Gestational Trophoblastic Disease/pathology , Humans , Middle Aged , Oncogene Proteins, Fusion/metabolism , Pregnancy , Telomerase/metabolism , Trophoblastic Neoplasms/genetics , Trophoblastic Neoplasms/metabolism , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism
7.
J Med Imaging (Bellingham) ; 7(5): 057502, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33102624

ABSTRACT

Purpose: Deep learning models are showing promise in digital pathology to aid diagnoses. Training complex models requires a significant amount and diversity of well-annotated data, typically housed in institutional archives. These slides often contain clinically meaningful markings to indicate regions of interest. If slides are scanned with the ink present, then the downstream model may end up looking for regions with ink before making a classification. If scanned without the markings, the information regarding where the relevant regions are located is lost. A compromise solution is to scan the slide with the annotations present but digitally remove them. Approach: We proposed a straightforward framework to digitally remove ink markings from whole slide images using a conditional generative adversarial network based on Pix2Pix. Results: The peak signal-to-noise ratio increased 30%, structural similarity index increased 20%, and visual information fidelity increased 200% relative to previous methods. Conclusions: When comparing our digital removal of marked images with rescans of clean slides, our method qualitatively and quantitatively exceeds current benchmarks, opening the possibility of using archived clinical samples as resources to fuel the next generation of deep learning models for digital pathology.

8.
PLoS One ; 14(7): e0220074, 2019.
Article in English | MEDLINE | ID: mdl-31339943

ABSTRACT

For many disease conditions, tissue samples are colored with multiple dyes and stains to add contrast and location information for specific proteins to accurately identify and diagnose disease. This presents a computational challenge for digital pathology, as whole-slide images (WSIs) need to be properly overlaid (i.e. registered) to identify co-localized features. Traditional image registration methods sometimes fail due to the high variation of cell density and insufficient texture information in WSIs-particularly at high magnifications. In this paper, we proposed a robust image registration strategy to align re-stained WSIs precisely and efficiently. This method is applied to 30 pairs of immunohistochemical (IHC) stains and their hematoxylin and eosin (H&E) counterparts. Our approach advances the existing methods in three key ways. First, we introduce refinements to existing image registration methods. Second, we present an effective weighting strategy using kernel density estimation to mitigate registration errors. Third, we account for the linear relationship across WSI levels to improve accuracy. Our experiments show significant decreases in registration errors when matching IHC and H&E pairs, enabling subcellular-level analysis on stained and re-stained histological images. We also provide a tool to allow users to develop their own registration benchmarking experiments.


Subject(s)
Image Processing, Computer-Assisted/methods , Staining and Labeling/methods , Fluoresceins , Fluorescent Antibody Technique/methods , Hematoxylin
9.
BMC Med Genomics ; 11(Suppl 3): 67, 2018 Sep 14.
Article in English | MEDLINE | ID: mdl-30255803

ABSTRACT

BACKGROUND: RNA-seq is the most commonly used sequencing application. Not only does it measure gene expression but it is also an excellent media to detect important structural variants such as single nucleotide variants (SNVs), insertion/deletion (Indels) or fusion transcripts. However, detection of these variants is challenging and complex from RNA-seq. Here we describe a sensitive and accurate analytical pipeline which detects various mutations at once for translational precision medicine. METHODS: The pipeline incorporates most sensitive aligners for Indels in RNA-Seq, the best practice for data preprocessing and variant calling, and STAR-fusion is for chimeric transcripts. Variants/mutations are annotated, and key genes can be extracted for further investigation and clinical actions. Three datasets were used to evaluate the performance of the pipeline for SNVs, indels and fusion transcripts. RESULTS: For the well-defined variants from NA12878 by GIAB project, about 95% and 80% of sensitivities were obtained for SNVs and indels, respectively, in matching RNA-seq. Comparison with other variant specific tools showed good performance of the pipeline. For the lung cancer dataset with 41 known and oncogenic mutations, 39 were detected by the pipeline with STAR aligner and all by the GSNAP aligner. An actionable EML4 and ALK fusion was also detected in one of the tumors, which also demonstrated outlier ALK expression. For 9 fusions spiked-into RNA-seq libraries with different concentrations, the pipeline was able to detect all in unfiltered results although some at very low concentrations may be missed when filtering was applied. CONCLUSIONS: The new RNA-seq workflow is an accurate and comprehensive mutation profiler from RNA-seq. Key or actionable mutations are reliably detected from RNA-seq, which makes it a practical alternative source for personalized medicine.


Subject(s)
Biomarkers, Tumor/genetics , High-Throughput Nucleotide Sequencing/methods , INDEL Mutation , Lung Neoplasms/genetics , Polymorphism, Single Nucleotide , Precision Medicine , Sequence Analysis, RNA/methods , Adenocarcinoma/genetics , Humans , Software
10.
Sci Rep ; 8(1): 5124, 2018 Mar 20.
Article in English | MEDLINE | ID: mdl-29559662

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

11.
Sci Rep ; 7(1): 14196, 2017 10 27.
Article in English | MEDLINE | ID: mdl-29079769

ABSTRACT

Long non-coding RNA (lncRNA) is a large class of gene transcripts with regulatory functions discovered in recent years. Many more are expected to be revealed with accumulation of RNA-seq data from diverse types of normal and diseased tissues. However, discovering novel lncRNAs and accurately quantifying known lncRNAs is not trivial from massive RNA-seq data. Herein we describe UClncR, an Ultrafast and Comprehensive lncRNA detection pipeline to tackle the challenge. UClncR takes standard RNA-seq alignment file, performs transcript assembly, predicts lncRNA candidates, quantifies and annotates both known and novel lncRNA candidates, and generates a convenient report for downstream analysis. The pipeline accommodates both un-stranded and stranded RNA-seq so that lncRNAs overlapping with other genes can be predicted and quantified. UClncR is fully parallelized in a cluster environment yet allows users to run samples sequentially without a cluster. The pipeline can process a typical RNA-seq sample in a matter of minutes and complete hundreds of samples in a matter of hours. Analysis of predicted lncRNAs from two test datasets demonstrated UClncR's accuracy and their relevance to sample clinical phenotypes. UClncR would facilitate researchers' novel lncRNA discovery significantly and is publically available at http://bioinformaticstools.mayo.edu/research/UClncR .


Subject(s)
RNA, Long Noncoding/genetics , Sequence Analysis, RNA/methods , Adenocarcinoma of Lung/genetics , Computational Biology , Humans , Time Factors
12.
JCO Precis Oncol ; 20172017.
Article in English | MEDLINE | ID: mdl-30761385

ABSTRACT

PURPOSE: Genomic testing has increased the quantity of information available to oncologists. Unfortunately, many identified sequence alterations are variants of unknown significance (VUSs), which thus limit the clinician's ability to use these findings to inform treatment. We applied a combination of in silico prediction and molecular modeling tools and laboratory techniques to rapidly define actionable VUSs. MATERIALS AND METHODS: Exome sequencing was conducted on 308 tumors from various origins. Most single nucleotide alterations within gene coding regions were VUSs. These VUSs were filtered to identify a subset of therapeutically targetable genes that were predicted with in silico tools to be altered in function by their variant sequence. A subset of receptor tyrosine kinase VUSs was characterized by laboratory comparison of each VUS versus its wild-type counterpart in terms of expression and signaling activity. RESULTS: The study identified 4,327 point mutations of which 3,833 were VUSs. Filtering for mutations in genes that were therapeutically targetable and predicted to affect protein function reduced these to 522VUSs of interest, including a large number of kinases. Ten receptortyrosine kinase VUSs were selected to explore in the laboratory. Of these, seven were found to be functionally altered. Three VUSs (FGFR2 F276C, FGFR4 R78H, and KDR G539R) showed increased basal or ligand-stimulated ERK phosphorylation compared with their wild-type counterparts, which suggests that they support transformation. Treatment of a patient who carried FGFR2 F276C with an FGFR inhibitor resulted in significant and sustained tumor response with clinical benefit. CONCLUSION: The findings demonstrate the feasibility of rapid identification of the biologic relevance of somatic mutations, which thus advances clinicians' ability to make informed treatment decisions.

13.
Stud Health Technol Inform ; 245: 868-872, 2017.
Article in English | MEDLINE | ID: mdl-29295223

ABSTRACT

In this study, we describe our efforts in building a clinical statistics and analysis application platform using an emerging clinical data standard, HL7 FHIR, and an open source web application framework, Shiny. We designed two primary workflows that integrate a series of R packages to enable both patient-centered and cohort-based interactive analyses. We leveraged Shiny with R to develop interactive interfaces on FHIR-based data and used ovarian cancer study datasets as a use case to implement a prototype. Specifically, we implemented patient index, patient-centered data report and analysis, and cohort analysis. The evaluation of our study was performed by testing the adaptability of the framework on two public FHIR servers. We identify common research requirements and current outstanding issues, and discuss future enhancement work of the current studies. Overall, our study demonstrated that it is feasible to use Shiny for implementing interactive analysis on FHIR-based standardized clinical data.


Subject(s)
Electronic Health Records , Health Level Seven , Humans , Medical Informatics
14.
Brief Bioinform ; 18(6): 973-983, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-27473065

ABSTRACT

Driver somatic mutations are a hallmark of a tumor that can be used for diagnosis and targeted therapy. Mutations are primarily detected from tumor DNA. As dynamic molecules of gene activities, transcriptome profiling by RNA sequence (RNA-seq) is becoming increasingly popular, which not only measures gene expression but also structural variations such as mutations and fusion transcripts. Although single-nucleotide variants (SNVs) can be easily identified from RNA-seq, intermediate long insertions/deletions (indels > 2 bases and less than sequence reads) cause significant challenges and are ignored by most RNA-seq analysis tools. This study evaluates commonly used RNA-seq analysis programs along with variant and somatic mutation callers in a series of data sets with simulated and known indels. The aim is to develop strategies for accurate indel detection. Our results show that the RNA-seq alignment is the most important step for indel identification and the evaluated programs have a wide range of sensitivity to map sequence reads with indels, from not at all to decently sensitive. The sensitivity is impacted by sequence read lengths. Most variant calling programs rely on hard evidence indels marked in the alignment and the programs with realignment may use soft-clipped reads for indel inferencing. Based on the observations, we have provided practical recommendations for indel detection when different RNA-seq aligners are used and demonstrated the best option with highly reliable results. With careful customization of bioinformatics algorithms, RNA-seq can be reliably used for both SNV and indel mutation detection that can be used for clinical decision-making.


Subject(s)
Computational Biology/methods , ErbB Receptors/genetics , High-Throughput Nucleotide Sequencing/methods , INDEL Mutation , Lung Neoplasms/genetics , Software , Algorithms , Case-Control Studies , Humans , Exome Sequencing
15.
Epigenomics ; 7(7): 1099-110, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26039248

ABSTRACT

AIM: Abnormal inactivation or loss of inactivated X chromosome (Xi) is implicated in women's cancer. However, the underlying mechanisms and clinical relevance are little known. MATERIALS & METHODS: High-throughput sequencing was conducted on breast cancer cell lines for copy number, RNA expression and 5'-methylcytosine in ChrX. The results were examined in primary breast tumors. RESULTS & CONCLUSION: Breast cancer cells demonstrated reduced or total loss of hemimethylation. Most cell lines lost part or one of X chromosomes. Cell lines without ChrX loss were more active in gene expression. DNA methylation was corroborated with Xi control lincRNA XIST. Similar transcriptome and DNA methylation changes were observed in primary breast cancer datasets with clinical phenotype associations. Dramatic genomic and epigenomic changes in ChrX may be used for potential diagnostic or prognostic markers in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Chromosomes, Human, X/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , X Chromosome Inactivation , 5-Methylcytosine/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Chromosomes, Human, X/chemistry , DNA Copy Number Variations , DNA Methylation , Databases, Factual , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Survival Analysis , Transcriptome
16.
Circ Cardiovasc Genet ; 8(1): 141-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25452597

ABSTRACT

BACKGROUND: The goal of this study was to identify genetic determinants of plasma N-terminal proatrial natriuretic peptide (NT-proANP) in the general community by performing a large-scale genetic association study and to assess its functional significance in in vitro cell studies and on disease susceptibility. METHODS AND RESULTS: Genotyping was performed across 16 000 genes in 893 randomly selected individuals, with replication in 891 subjects from the community. Plasma NT-proANP1-98 concentrations were determined using a radioimmunoassay. Thirty-three genome-wide significant single-nucleotide polymorphisms were identified in the MTHFR-CLCN6-NPPA-NPPB locus and were all replicated. To assess the significance, in vitro functional genomic studies and clinical outcomes for carriers of a single-nucleotide polymorphism rs5063 (V32M) located in NPPA that represented the most significant variation in this genetic locus were assessed. The rs5063 variant allozyme in transfected HEK293 cells was decreased to 55±8% of wild-type protein (P=0.01) as assessed by quantitative western blots. Carriers of rs5063 had lower NT-proANP levels (1427 versus 2291 pmol/L; P<0.001) and higher diastolic blood pressures (75 versus 73 mm Hg; P=0.009) and were at an increased risk of stroke when compared with wild-type subjects independent of age, sex, diabetes mellitus, hypertension, atrial fibrillation, and cholesterol levels (hazard ratio, 1.6; P=0.004). CONCLUSIONS: This is the first large-scale genetic association study of circulating NT-proANP levels performed with replication and functional assessment that identified genetic variants in the MTHFR-CLCN6-NPPA-NPPB cluster to be significantly associated with NT-proANP levels. The clinical significance of this variation is related to lower NT-proANP levels, higher blood pressures, and an increased risk of stroke in the general community.


Subject(s)
Atrial Natriuretic Factor , Genetic Loci , Multigene Family , Stroke , White People/genetics , Atrial Natriuretic Factor/blood , Atrial Natriuretic Factor/genetics , Chloride Channels/metabolism , Female , Genome-Wide Association Study , HEK293 Cells , Humans , Male , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Stroke/blood , Stroke/genetics
17.
Cancer Res ; 74(23): 6947-57, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25320007

ABSTRACT

Metastatic recurrence is the leading cause of cancer-related deaths in patients with colorectal carcinoma. To capture the molecular underpinnings for metastasis and tumor progression, we performed integrative network analysis on 11 independent human colorectal cancer gene expression datasets and applied expression data from an immunocompetent mouse model of metastasis as an additional filter for this biologic process. In silico analysis of one metastasis-related coexpression module predicted nuclear factor of activated T-cell (NFAT) transcription factors as potential regulators for the module. Cells selected for invasiveness and metastatic capability expressed higher levels of NFATc1 as compared with poorly metastatic and less invasive parental cells. We found that inhibition of NFATc1 in human and mouse colon cancer cells resulted in decreased invasiveness in culture and downregulation of metastasis-related network genes. Overexpression of NFATc1 significantly increased the metastatic potential of colon cancer cells, whereas inhibition of NFATc1 reduced metastasis growth in an immunocompetent mouse model. Finally, we found that an 8-gene signature comprising genes upregulated by NFATc1 significantly correlated with worse clinical outcomes in stage II and III colorectal cancer patients. Thus, NFATc1 regulates colon cancer cell behavior and its transcriptional targets constitute a novel, biologically anchored gene expression signature for the identification of colon cancers with high risk of metastatic recurrence.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , NFATC Transcription Factors/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Transcription Factors/genetics
18.
Bioinformatics ; 30(18): 2678-80, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24876377

ABSTRACT

MOTIVATION: Exome sequencing (exome-seq) data, which are typically used for calling exonic mutations, have also been utilized in detecting DNA copy number variations (CNVs). Despite the existence of several CNV detection tools, there is still a great need for a sensitive and an accurate CNV-calling algorithm with built-in QC steps, and does not require a paired reference for each sample. RESULTS: We developed a novel method named PatternCNV, which (i) accounts for the read coverage variations between exons while leveraging the consistencies of this variability across different samples; (ii) reduces alignment BAM files to WIG format and therefore greatly accelerates computation; (iii) incorporates multiple QC measures designed to identify outlier samples and batch effects; and (iv) provides a variety of visualization options including chromosome, gene and exon-level views of CNVs, along with a tabular summarization of the exon-level CNVs. Compared with other CNV-calling algorithms using data from a lymphoma exome-seq study, PatternCNV has higher sensitivity and specificity. AVAILABILITY AND IMPLEMENTATION: The software for PatternCNV is implemented using Perl and R, and can be used in Mac or Linux environments. Software and user manual are available at http://bioinformaticstools.mayo.edu/research/patterncnv/, and R package at https://github.com/topsoil/patternCNV/.


Subject(s)
Algorithms , DNA Copy Number Variations , Exome/genetics , Genomics/methods , Sequence Analysis, DNA , Exons/genetics , Software
19.
AMIA Annu Symp Proc ; 2014: 1160-9, 2014.
Article in English | MEDLINE | ID: mdl-25954427

ABSTRACT

Adverse drug events (ADEs) are a critical factor for selecting cancer therapy options. The underlying molecular mechanisms of ADEs associated with cancer therapy drugs may overlap with their antineoplastic mechanisms; an aspect of toxicity. In the present study, we develop a novel knowledge-driven approach that provides an ADE-based stratification (ADEStrata) of tumor mutations. We demonstrate clinical utility of the ADEStrata approach through performing a case study of breast invasive carcinoma (BRCA) patients receiving aromatase inhibitors (AI) from The Cancer Genome Atlas (TCGA) (n=212), focusing on the musculoskeletal adverse events (MS-AEs). We prioritized somatic variants in a manner that is guided by MS-AEs codified as 6 Human Phenotype Ontology (HPO) terms. Pathway enrichment and hierarchical clustering of prioritized variants reveals clusters associated with overall survival. We demonstrated that the prediction of per-patient ADE propensity simultaneously identifies high-risk patients experiencing poor outcomes. In conclusion, the ADEStrata approach could produce clinically and biologically meaningful tumor subtypes that are potentially predictive of the drug response to the cancer therapy drugs.


Subject(s)
Antineoplastic Agents/adverse effects , Aromatase Inhibitors/adverse effects , Breast Neoplasms/genetics , Mutation , Antineoplastic Agents/therapeutic use , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Drug-Related Side Effects and Adverse Reactions , Female , Humans , Propensity Score , Unified Medical Language System
20.
Mol Biosyst ; 7(7): 2118-27, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21594272

ABSTRACT

Modification of proteins by reactive electrophiles such as the 4-hydroxy-2-nonenal (HNE) plays a critical role in oxidant-associated human diseases. However, little is known about protein adduction and the mechanism by which protein damage elicits adaptive effects and toxicity. We developed a systems approach for relating protein adduction to gene expression changes through the integration of protein adduction, gene expression, protein-DNA interaction, and protein-protein interaction data. Using a random walk strategy, we expanded a list of responsive transcription factors inferred from gene expression studies to upstream signaling networks, which in turn allowed overlaying protein adduction data on the network for the prediction of stress sensors and their associated regulatory mechanisms. We demonstrated the general applicability of transcription factor-based signaling network inference using 103 known pathways. Applying our workflow on gene expression and protein adduction data from HNE-treatment not only rediscovered known mechanisms of electrophile stress but also generated novel hypotheses regarding protein damage sensors. Although developed for analyzing protein adduction data, the framework can be easily adapted for phosphoproteomics and other types of protein modification data.


Subject(s)
Gene Expression Regulation , Proteins/metabolism , Systems Biology/methods , Aldehydes/pharmacology , DNA Damage , Gene Expression Regulation/drug effects , Heat-Shock Proteins/metabolism , Humans , Signal Transduction/drug effects , Transcription Factors/metabolism , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...