Conserved and Divergent Modulation of Calcification in Atherosclerosis and Aortic Valve Disease by Tissue Extracellular Vesicles

Background Fewer than 50% of patients develop calcification of both atherosclerotic plaques and aortic valves, implying differential pathogenesis. While circulating extracellular vesicles (EVs) act as biomarkers of cardiovascular diseases, tissue-entrapped EVs associate with early mineralization, but their contents, function, and contributions to disease remain unknown. Results Global proteomics of human carotid artery endarterectomies and calcified aortic valves from a total of 27 donors/patients revealed significant over-representation of proteins with vesicle-associated pathways/ontologies common to both diseases. We exploited enzymatic digestion, serial (ultra)centrifugation and OptiPrep density-gradient separation to isolate EV populations from diseased arteries and valves. Mass spectrometry found 22 EV marker proteins to be highly enriched in the four least-dense OptiPrep fractions while extracellular matrix proteins predominated in denser fractions, as confirmed by CD63 immunogold electron microscopy and nanoparticle tracking analysis. Proteomics and miRNA-sequencing of OptiPrep-enriched tissue EVs quantified 1,104 proteins and 123 miR cargoes linked to 5,182 target genes. Pathway networks of proteins and miR targets common to artery and valve tissue EVs revealed a shared regulation of Rho GTPase and MAPK intracellular signaling cascades. 179 proteins and 5 miRs were significantly altered between artery and valve EVs; multi-omics integration determined that EVs differentially modulated cellular contraction and p53-mediated transcriptional regulation in diseased vascular vs. valvular tissue. Conclusions Our findings delineate a strategy to isolate, purify, and study protein and RNA cargoes from EVs entrapped in fibrocalcific tissues. Multi-omics and network approaches implicated tissue-resident EVs in human cardiovascular disease.

[1]  S. Body,et al.  Standardization of Human Calcific Aortic Valve Disease in vitro Modeling Reveals Passage-Dependent Calcification , 2019, Front. Cardiovasc. Med..

[2]  Yong Guo,et al.  miR-29b-3p regulated osteoblast differentiation via regulating IGF-1 secretion of mechanically stimulated osteocytes , 2019, Cellular & Molecular Biology Letters.

[3]  J. Michel,et al.  Relationship of Iron Deposition to Calcium Deposition in Human Aortic Valve Leaflets. , 2019, Journal of the American College of Cardiology.

[4]  E. Aikawa,et al.  Differential miRNA Loading Underpins Dual Harmful and Protective Roles for Extracellular Vesicles in Atherogenesis. , 2019, Circulation research.

[5]  J. Olcese,et al.  Extraction of Extracellular Vesicles from Whole Tissue. , 2019, Journal of visualized experiments : JoVE.

[6]  Damian Szklarczyk,et al.  STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets , 2018, Nucleic Acids Res..

[7]  Ana Kozomara,et al.  miRBase: from microRNA sequences to function , 2018, Nucleic Acids Res..

[8]  Hiroshi Iwata,et al.  XINA: A Workflow for the Integration of Multiplexed Proteomics Kinetics Data with Network Analysis. , 2019, Journal of proteome research.

[9]  Jiafeng Wang,et al.  MicroRNA let‐7c‐5p promotes osteogenic differentiation of dental pulp stem cells by inhibiting lipopolysaccharide‐induced inflammation via HMGA2/PI3K/Akt signal blockade , 2019, Clinical and experimental pharmacology & physiology.

[10]  Jing Xu,et al.  Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines , 2018, Journal of Extracellular Vesicles.

[11]  Joshua D. Hutcheson,et al.  Spatiotemporal Multi-Omics Mapping Generates a Molecular Atlas of the Aortic Valve and Reveals Networks Driving Disease , 2018, Circulation.

[12]  Sarah J. Parker,et al.  Proteomic Architecture of Human Coronary and Aortic Atherosclerosis , 2018, Circulation.

[13]  Mark W. Tibbitt,et al.  Engineering a 3D-Bioprinted Model of Human Heart Valve Disease Using Nanoindentation-Based Biomechanics , 2018, Nanomaterials.

[14]  E. Aikawa,et al.  Dimerization of sortilin regulates its trafficking to extracellular vesicles , 2018, The Journal of Biological Chemistry.

[15]  Y. Shao,et al.  Low-level overexpression of p53 promotes warfarin-induced calcification of porcine aortic valve interstitial cells by activating Slug gene transcription , 2018, The Journal of Biological Chemistry.

[16]  Graça Raposo,et al.  Shedding light on the cell biology of extracellular vesicles , 2018, Nature Reviews Molecular Cell Biology.

[17]  Thawfeek M. Varusai,et al.  The Reactome Pathway Knowledgebase , 2017, Nucleic acids research.

[18]  Y. Hannun,et al.  Sphingosine 1-phosphate activation of ERM contributes to vascular calcification[S] , 2017, Journal of Lipid Research.

[19]  C. Masters,et al.  A rigorous method to enrich for exosomes from brain tissue , 2017, Journal of extracellular vesicles.

[20]  Anna C. Porter,et al.  Coronary Artery Calcification and Risk of Cardiovascular Disease and Death Among Patients With Chronic Kidney Disease , 2017, JAMA cardiology.

[21]  M. Dweck,et al.  End stage renal disease‐induced hypercalcemia may promote aortic valve calcification via Annexin VI enrichment of valve interstitial cell derived‐matrix vesicles , 2017, Journal of cellular physiology.

[22]  X. Loyer,et al.  Extracellular vesicles in coronary artery disease , 2017, Nature Reviews Cardiology.

[23]  D. Tang,et al.  The roles and regulation of the actin cytoskeleton, intermediate filaments and microtubules in smooth muscle cell migration , 2017, Respiratory Research.

[24]  A. Didangelos,et al.  Extracellular matrix proteomics identifies molecular signature of symptomatic carotid plaques , 2017, The Journal of clinical investigation.

[25]  Y. Bossé,et al.  Altered DNA Methylation of Long Noncoding RNA H19 in Calcific Aortic Valve Disease Promotes Mineralization by Silencing NOTCH1 , 2016, Circulation.

[26]  Zhiping Weng,et al.  DNApi: A De Novo Adapter Prediction Algorithm for Small RNA Sequencing Data , 2016, PloS one.

[27]  P. Libby,et al.  Sortilin mediates vascular calcification via its recruitment into extracellular vesicles. , 2016, The Journal of clinical investigation.

[28]  Joshua D. Hutcheson,et al.  Genesis and growth of extracellular vesicle-derived microcalcification in atherosclerotic plaques , 2015, Nature materials.

[29]  Douglas E. Vaughan,et al.  MiR-125b Is Critical for Fibroblast-to-Myofibroblast Transition and Cardiac Fibrosis , 2016, Circulation.

[30]  M. Mayr,et al.  Vascular smooth muscle cell calcification is mediated by regulated exosome secretion. , 2015, Circulation research.

[31]  Ram Rup Sarkar,et al.  Comparison of human cell signaling pathway databases—evolution, drawbacks and challenges , 2015, Database J. Biol. Databases Curation.

[32]  A. Brisson,et al.  High-speed centrifugation induces aggregation of extracellular vesicles , 2015, Journal of extracellular vesicles.

[33]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[34]  Jason L. Johnson Emerging regulators of vascular smooth muscle cell function in the development and progression of atherosclerosis. , 2014, Cardiovascular research.

[35]  Joshua D. Hutcheson,et al.  Enrichment of calcifying extracellular vesicles using density-based ultracentrifugation protocol , 2014, Journal of extracellular vesicles.

[36]  MasanoriAikawa,et al.  Cystathionine γ-lyase Accelerates Osteoclast Differentiation , 2014 .

[37]  Elena Aikawa,et al.  Macrophage-Derived Matrix Vesicles: An Alternative Novel Mechanism for Microcalcification in Atherosclerotic Plaques , 2013, Circulation research.

[38]  Lynne T. Bemis,et al.  Standardization of sample collection, isolation and analysis methods in extracellular vesicle research , 2013, Journal of extracellular vesicles.

[39]  Ralf Herwig,et al.  The ConsensusPathDB interaction database: 2013 update , 2012, Nucleic Acids Res..

[40]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[41]  F. Cipollone,et al.  MicroRNAs and atherosclerosis: new actors for an old movie. , 2012, Nutrition, metabolism, and cardiovascular diseases : NMCD.

[42]  J. Zhan,et al.  p53 negatively regulates the osteogenic differentiation of vascular smooth muscle cells in mice with chronic kidney disease , 2012, Cardiovascular journal of Africa.

[43]  B. Cookson,et al.  Membrane Vesicle Release in Bacteria, Eukaryotes, and Archaea: a Conserved yet Underappreciated Aspect of Microbial Life , 2012, Infection and Immunity.

[44]  Xavier Estivill,et al.  A non-biased framework for the annotation and classification of the non-miRNA small RNA transcriptome , 2011, Bioinform..

[45]  Heng Li,et al.  A statistical framework for SNP calling, mutation discovery, association mapping and population genetical parameter estimation from sequencing data , 2011, Bioinform..

[46]  L. Hofbauer,et al.  miR-125b regulates calcification of vascular smooth muscle cells. , 2011, The American journal of pathology.

[47]  G. Nickenig,et al.  Circulating CD31+/Annexin V+ microparticles correlate with cardiovascular outcomes. , 2011, European heart journal.

[48]  M. Mayr,et al.  Calcium Regulates Key Components of Vascular Smooth Muscle Cell–Derived Matrix Vesicles to Enhance Mineralization , 2011, Circulation research.

[49]  Marcel Martin Cutadapt removes adapter sequences from high-throughput sequencing reads , 2011 .

[50]  Xavier Estivill,et al.  SeqBuster, a bioinformatic tool for the processing and analysis of small RNAs datasets, reveals ubiquitous miRNA modifications in human embryonic cells , 2009, Nucleic acids research.

[51]  Manuel Mayr,et al.  Proteomics, Metabolomics, and Immunomics on Microparticles Derived From Human Atherosclerotic Plaques , 2009, Circulation. Cardiovascular genetics.

[52]  Deepak Srivastava,et al.  miR-145 and miR-143 Regulate Smooth Muscle Cell Fate Decisions , 2009, Nature.

[53]  Shelly R. Peyton,et al.  ECM Compliance Regulates Osteogenesis by Influencing MAPK Signaling Downstream of RhoA and ROCK , 2009, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[54]  N. Toma Intensive Lipid Lowering with Simvastatin and Ezetimibe in Aortic Stenosis , 2009 .

[55]  J. Chambers,et al.  Intensive lipid lowering with simvastatin and ezetimibe in aortic stenosis. , 2008, The New England journal of medicine.

[56]  Jean-Loup Guillaume,et al.  Fast unfolding of communities in large networks , 2008, 0803.0476.

[57]  J. Lötvall,et al.  Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells , 2007, Nature Cell Biology.

[58]  Y. Castier,et al.  Cellular origins and thrombogenic activity of microparticles isolated from human atherosclerotic plaques. , 2007, Journal of the American College of Cardiology.

[59]  J. Cigarroa,et al.  Prevalence of coronary artery disease in patients with aortic stenosis with and without angina pectoris. , 2001, The American journal of cardiology.

[60]  Susumu Goto,et al.  KEGG: Kyoto Encyclopedia of Genes and Genomes , 2000, Nucleic Acids Res..

[61]  Y. Benjamini,et al.  Controlling the false discovery rate: a practical and powerful approach to multiple testing , 1995 .

[62]  A. Gown,et al.  Characterization of the Early Lesion of ‘Degenerative’ Valvular Aortic Stenosis: Histological and Immunohistochemical Studies , 1994, Circulation.