Whole-Blood RNA Profiles Associated with Pulmonary Arterial Hypertension and Clinical Outcome

Rationale: Idiopathic and heritable pulmonary arterial hypertension (PAH) are rare but comprise a genetically heterogeneous patient group. RNA sequencing linked to the underlying genetic architecture can be used to better understand the underlying pathology by identifying key signaling pathways and stratify patients more robustly according to clinical risk. Objectives: To use a three-stage design of RNA discovery, RNA validation and model construction, and model validation to define a set of PAH-associated RNAs and a single summarizing RNA model score. To define genes most likely to be involved in disease development, we performed Mendelian randomization (MR) analysis. Methods: RNA sequencing was performed on whole-blood samples from 359 patients with idiopathic, heritable, and drug-induced PAH and 72 age- and sex-matched healthy volunteers. The score was evaluated against disease severity markers including survival analysis using all-cause mortality from diagnosis. MR used known expression quantitative trait loci and summary statistics from a PAH genome-wide association study. Measurements and Main Results: We identified 507 genes with differential RNA expression in patients with PAH compared with control subjects. A model of 25 RNAs distinguished PAH with 87% accuracy (area under the curve 95% confidence interval: 0.791–0.945) in model validation. The RNA model score was associated with disease severity and long-term survival (P = 4.66 × 10−6) in PAH. MR detected an association between SMAD5 levels and PAH disease susceptibility (odds ratio, 0.317; 95% confidence interval, 0.129–0.776; P = 0.012). Conclusions: A whole-blood RNA signature of PAH, which includes RNAs relevant to disease pathogenesis, associates with disease severity and identifies patients with poor clinical outcomes. Genetic variants associated with lower SMAD5 expression may increase susceptibility to PAH.

[1]  Yuan Guo,et al.  The Role of Regulatory T Cells in Pulmonary Arterial Hypertension , 2019, Journal of the American Heart Association.

[2]  E. Mickler,et al.  Systems Analysis of the Human Pulmonary Arterial Hypertension Lung Transcriptome. , 2019, American journal of respiratory cell and molecular biology.

[3]  Lisa J. Martin,et al.  Genetic determinants of risk in pulmonary arterial hypertension: international genome-wide association studies and meta-analysis , 2018, bioRxiv.

[4]  R. Danner,et al.  Meta-analysis of Blood Genome-Wide Expression Profiling Studies in Pulmonary Arterial Hypertension. , 2019, American journal of physiology. Lung cellular and molecular physiology.

[5]  A. Pegg,et al.  Polyamine metabolism and cancer: treatments, challenges and opportunities , 2018, Nature Reviews Cancer.

[6]  Yu Wang,et al.  Molecular and pharmacological modulators of the tumor immune contexture revealed by deconvolution of RNA-seq data , 2017, Genome Medicine.

[7]  Henning Gall,et al.  Identification of rare sequence variation underlying heritable pulmonary arterial hypertension , 2018, Nature Communications.

[8]  M. Humbert,et al.  Plasma proteome analysis in patients with pulmonary arterial hypertension: an observational cohort study , 2017, The Lancet. Respiratory medicine.

[9]  H. Hedlin,et al.  Randomised placebo-controlled safety and tolerability trial of FK506 (tacrolimus) for pulmonary arterial hypertension , 2017, European Respiratory Journal.

[10]  Jodie L Babitt,et al.  Smad1/5 is required for erythropoietin-mediated suppression of hepcidin in mice. , 2017, Blood.

[11]  Cizhong Jiang,et al.  Smad5 acts as an intracellular pH messenger and maintains bioenergetic homeostasis , 2017, Cell Research.

[12]  Geet Duggal,et al.  Salmon: fast and bias-aware quantification of transcript expression using dual-phase inference , 2017, Nature Methods.

[13]  P. Corris,et al.  Plasma Metabolomics Implicates Modified Transfer RNAs and Altered Bioenergetics in the Outcomes of Pulmonary Arterial Hypertension , 2017, Circulation.

[14]  Roby Joehanes,et al.  Integrated genome-wide analysis of expression quantitative trait loci aids interpretation of genomic association studies , 2017, Genome Biology.

[15]  M. Humbert,et al.  Regulatory T Cell Dysfunction in Idiopathic, Heritable and Connective Tissue-Associated Pulmonary Arterial Hypertension. , 2016, Chest.

[16]  F. Saudek,et al.  Unstable Expression of Commonly Used Reference Genes in Rat Pancreatic Islets Early after Isolation Affects Results of Gene Expression Studies , 2016, PloS one.

[17]  Stephen Burgess,et al.  Combining information on multiple instrumental variables in Mendelian randomization: comparison of allele score and summarized data methods , 2015, Statistics in medicine.

[18]  D. Geerts,et al.  ATP13A3 and caveolin-1 as potential biomarkers for difluoromethylornithine-based therapies in pancreatic cancers. , 2016, American journal of cancer research.

[19]  Simon Gibbs,et al.  2015 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension , 2015, European Respiratory Journal.

[20]  S. Gräf,et al.  Selective enhancement of endothelial BMPR-II with BMP9 reverses pulmonary arterial hypertension , 2015, Nature Medicine.

[21]  Ash A. Alizadeh,et al.  Robust enumeration of cell subsets from tissue expression profiles , 2015, Nature Methods.

[22]  S. Rich,et al.  Peripheral Blood Signature of Vasodilator-Responsive Pulmonary Arterial Hypertension , 2015, Circulation.

[23]  M. Humbert,et al.  Inflammation and immunity in the pathogenesis of pulmonary arterial hypertension. , 2014, Circulation research.

[24]  W. Seeger,et al.  Impact of S-Adenosylmethionine Decarboxylase 1 on Pulmonary Vascular Remodeling , 2014, Circulation.

[25]  M. Humbert,et al.  Upfront triple combination therapy in pulmonary arterial hypertension: a pilot study , 2014, European Respiratory Journal.

[26]  R Core Team,et al.  R: A language and environment for statistical computing. , 2014 .

[27]  T. Gudermann,et al.  Classical transient receptor potential channel 1 in hypoxia-induced pulmonary hypertension. , 2013, American Journal of Respiratory and Critical Care Medicine.

[28]  M. Peters,et al.  Systematic identification of trans eQTLs as putative drivers of known disease associations , 2013, Nature Genetics.

[29]  N. Morrell,et al.  Correction of nonsense BMPR2 and SMAD9 mutations by ataluren in pulmonary arterial hypertension. , 2013, American journal of respiratory cell and molecular biology.

[30]  J. Wharton,et al.  Iron deficiency and raised hepcidin in idiopathic pulmonary arterial hypertension: clinical prevalence, outcomes, and mechanistic insights. , 2011, Journal of the American College of Cardiology.

[31]  Xavier Robin,et al.  pROC: an open-source package for R and S+ to analyze and compare ROC curves , 2011, BMC Bioinformatics.

[32]  Mark D. Robinson,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[33]  R. Speich,et al.  Increased Regulatory and Decreased CD8+ Cytotoxic T Cells in the Blood of Patients with Idiopathic Pulmonary Arterial Hypertension , 2007, Respiration.