The stretch responsive microRNA miR‐148a‐3p is a novel repressor of IKBKB, NF‐κB signaling, and inflammatory gene expression in human aortic valve cells

Bicuspid aortic valves calcify at a significantly higher rate than normal aortic valves, a process that involves increased inflammation. Because we have previously found that bicuspid aortic valve experience greater stretch, we investigated the potential connection between stretch and inflammation in human aortic valve interstitial cells (AVICs). Microarray, quantitative PCR (qPCR), and protein assays performed on AVICs exposed to cyclic stretch showed that stretch was sufficient to increase expression of interleukin and metalloproteinase family members by more than 1.5‐fold. Conditioned medium from stretched AVICs was sufficient to activate leukocytes. microRNA sequencing and qPCR experiments demonstrated that miR‐148a‐3p was repressed in both stretched AVICs (43% repression) and, as a clinical correlate, human bicuspid aortic valves (63% reduction). miR‐148a‐3p was found to be a novel repressor of IKBKB based on data from qPCR, luciferase, and Western blot experiments. Furthermore, increasing miR‐148a‐3p levels in AVICs was sufficient to decrease NF‐κB (nuclear factor kappa‐light‐chain‐enhancer of activated B cells) signaling and NF‐κB target gene expression. Our data demonstrate that stretch‐mediated activation of inflammatory pathways is at least partly the result of stretch‐repression of miR‐148a‐3p and a consequent failure to repress IKBKB. To our knowledge, we are the first to report that cyclic stretch of human AVICs activates inflammatory genes in a tissue‐autonomous manner via a microRNA that regulates a central inflammatory pathway.—Patel, V., Carrion, K., Hollands, A., Hinton, A., Gallegos, T., Dyo, J., Sasik, R., Leire, E., Hardiman, G., Mohamed, S. A., Nigam, S., King, C. C., Nizet, V., Nigam V. The stretch responsive microRNA miR‐148a‐3p is a novel repressor of IKBKB, NF‐κB signaling, and inflammatory gene expression in human aortic valve cells. FASEB J. 29, 1859‐1868 (2015). www.fasebj.org

[1]  M. Daniels,et al.  Architectural Trends in the Human Normal and Bicuspid Aortic Valve Leaflet and Its Relevance to Valve Disease , 2014, Annals of Biomedical Engineering.

[2]  Vishal Nigam,et al.  Bicuspid Aortic Valves Experience Increased Strain as Compared to Tricuspid Aortic Valves , 2013, World journal for pediatric & congenital heart surgery.

[3]  S. Kauppinen,et al.  Treatment of HCV infection by targeting microRNA. , 2013, The New England journal of medicine.

[4]  Libing Song,et al.  miR-486 sustains NF-κB activity by disrupting multiple NF-κB-negative feedback loops , 2012, Cell Research.

[5]  S. Verma,et al.  miRNA-141 is a novel regulator of BMP-2-mediated calcification in aortic stenosis. , 2012, The Journal of thoracic and cardiovascular surgery.

[6]  O. Bukulmez,et al.  Endometrial miR-200c is Altered During Transformation into Cancerous States and Targets the Expression of ZEBs, VEGFA, FLT1, IKKβ, KLF9, and FBLN5 , 2012, Reproductive Sciences.

[7]  Chris B Schaffer,et al.  Cyclic strain anisotropy regulates valvular interstitial cell phenotype and tissue remodeling in three-dimensional culture. , 2012, Acta biomaterialia.

[8]  Norbert Gretz,et al.  miRWalk - Database: Prediction of possible miRNA binding sites by "walking" the genes of three genomes , 2011, J. Biomed. Informatics.

[9]  Robert M Nerem,et al.  Discovery of shear- and side-specific mRNAs and miRNAs in human aortic valvular endothelial cells. , 2011, American journal of physiology. Heart and circulatory physiology.

[10]  Jun Li,et al.  miR-218 inhibits the invasive ability of glioma cells by direct downregulation of IKK-β. , 2010, Biochemical and biophysical research communications.

[11]  S. Hirota,et al.  Increased interleukin-18 expression in nonrheumatic aortic valve stenosis. , 2010, International journal of cardiology.

[12]  D. Srivastava,et al.  Altered microRNAs in bicuspid aortic valve: a comparison between stenotic and insufficient valves. , 2010, The Journal of heart valve disease.

[13]  Michael A Lopez,et al.  Mechanical Stretch Up-regulates MicroRNA-26a and Induces Human Airway Smooth Muscle Hypertrophy by Suppressing Glycogen Synthase Kinase-3β* , 2010, The Journal of Biological Chemistry.

[14]  Kathryn E. Smith,et al.  Cyclic strain inhibits acute pro-inflammatory gene expression in aortic valve interstitial cells , 2010, Biomechanics and modeling in mechanobiology.

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

[16]  K. Waters,et al.  MicroRNA 132 regulates nutritional stress-induced chemokine production through repression of SirT1. , 2009, Molecular endocrinology.

[17]  Nectarios Koziris,et al.  DIANA-microT web server: elucidating microRNA functions through target prediction , 2009, Nucleic Acids Res..

[18]  Cole Trapnell,et al.  Ultrafast and memory-efficient alignment of short DNA sequences to the human genome , 2009, Genome Biology.

[19]  Ajit P Yoganathan,et al.  Elevated cyclic stretch alters matrix remodeling in aortic valve cusps: implications for degenerative aortic valve disease. , 2009, American journal of physiology. Heart and circulatory physiology.

[20]  R. Chen,et al.  Regulation of IKKβ by miR-199a affects NF-κB activity in ovarian cancer cells , 2008, Oncogene.

[21]  Xiaowei Wang miRDB: a microRNA target prediction and functional annotation database with a wiki interface. , 2008, RNA.

[22]  G. Nickenig,et al.  Differential profile of the OPG/RANKL/RANK-system in degenerative aortic native and bioprosthetic valves. , 2008, The Journal of heart valve disease.

[23]  Michael Kertesz,et al.  The role of site accessibility in microRNA target recognition , 2007, Nature Genetics.

[24]  Younghee Lee,et al.  Enhancement of NF-kappaB expression and activity upon differentiation of human embryonic stem cell line SNUhES3. , 2007, Stem cells and development.

[25]  M. Amrani,et al.  Role of Human Valve Interstitial Cells in Valve Calcification and Their Response to Atorvastatin , 2006, Circulation.

[26]  Y. Suematsu,et al.  Age-associated aortic stenosis in apolipoprotein E-deficient mice. , 2005, Journal of the American College of Cardiology.

[27]  B. Iung,et al.  Extracellular matrix remodelling in human aortic valve disease: the role of matrix metalloproteinases and their tissue inhibitors. , 2005, European heart journal.

[28]  Rosario V. Freeman,et al.  Spectrum of Calcific Aortic Valve Disease: Pathogenesis, Disease Progression, and Treatment Strategies , 2005, Circulation.

[29]  K. Gunsalus,et al.  Combinatorial microRNA target predictions , 2005, Nature Genetics.

[30]  S. Hagl,et al.  Inflammatory regulation of extracellular matrix remodeling in calcific aortic valve stenosis. , 2005, Cardiovascular pathology : the official journal of the Society for Cardiovascular Pathology.

[31]  C. Burge,et al.  Conserved Seed Pairing, Often Flanked by Adenosines, Indicates that Thousands of Human Genes are MicroRNA Targets , 2005, Cell.

[32]  Anton J. Enright,et al.  Human MicroRNA Targets , 2004, PLoS biology.

[33]  M. Bernardo,et al.  Differential Inhibition of Membrane Type 3 (MT3)-Matrix Metalloproteinase (MMP) and MT1-MMP by Tissue Inhibitor of Metalloproteinase (TIMP)-2 and TIMP-3 Regulates Pro-MMP-2 Activation* , 2004, Journal of Biological Chemistry.

[34]  S. Hagl,et al.  Interleukin-1 beta promotes matrix metalloproteinase expression and cell proliferation in calcific aortic valve stenosis. , 2003, Atherosclerosis.

[35]  T. Salo,et al.  Evidence for an altered balance between matrix metalloproteinase-9 and its inhibitors in calcific aortic stenosis. , 2003, The Annals of thoracic surgery.

[36]  R. Levy,et al.  Progression of aortic valve stenosis: TGF-beta1 is present in calcified aortic valve cusps and promotes aortic valve interstitial cell calcification via apoptosis. , 2003, The Annals of thoracic surgery.

[37]  B. Janerot-Sjöberg,et al.  T lymphocyte infiltration in non-rheumatic aortic stenosis: a comparative descriptive study between tricuspid and bicuspid aortic valves , 2002, Heart.

[38]  S. Verma,et al.  Clinical and Pathophysiological Implications of a Bicuspid Aortic Valve , 2002, Circulation.

[39]  Y. Soini,et al.  Expression of MMP2, MMP9, MT1‐MMP, TIMP1, and TIMP2 mRNA in valvular lesions of the heart , 2001, The Journal of pathology.

[40]  J. Shirani,et al.  Matrix metalloproteinase expression in nonrheumatic aortic stenosis. , 2000, Cardiovascular pathology : the official journal of the Society for Cardiovascular Pathology.

[41]  C. Ward Clinical significance of the bicuspid aortic valve , 2000, Heart.

[42]  C. Brinckerhoff,et al.  Nuclear factor kappaB/p50 activates an element in the distal matrix metalloproteinase 1 promoter in interleukin-1beta-stimulated synovial fibroblasts. , 1998, Arthritis and rheumatism.

[43]  T. Lüscher,et al.  Pulsatile Stretch Stimulates Superoxide Production and Activates Nuclear Factor-κB in Human Coronary Smooth Muscle , 1997 .

[44]  E. Mohler,et al.  Detection of osteopontin in calcified human aortic valves. , 1997, Arteriosclerosis, thrombosis, and vascular biology.

[45]  N. Mori,et al.  Transactivation of the interleukin-1alpha promoter by human T-cell leukemia virus type I and type II Tax proteins. , 1996, Blood.

[46]  M. Davies,et al.  Demographic characteristics of patients undergoing aortic valve replacement for stenosis: relation to valve morphology. , 1996, Heart.

[47]  B. Sumpio,et al.  Cyclic strain causes heterogeneous induction of transcription factors, AP-1, CRE binding protein and NF-kB, in endothelial cells: species and vascular bed diversity. , 1995, Journal of biomechanics.

[48]  M. Ferguson,et al.  Osteopontin is expressed in human aortic valvular lesions. , 1995, Circulation.

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

[50]  M. Rosenqvist,et al.  Accumulation of T lymphocytes and expression of interleukin-2 receptors in nonrheumatic stenotic aortic valves. , 1994, Journal of the American College of Cardiology.

[51]  J. Hiscott,et al.  Characterization of a functional NF-kappa B site in the human interleukin 1 beta promoter: evidence for a positive autoregulatory loop , 1993, Molecular and cellular biology.

[52]  S. Beppu,et al.  Rapidity of progression of aortic stenosis in patients with congenital bicuspid aortic valves. , 1993, The American journal of cardiology.

[53]  A. Brasier,et al.  Optimized use of the firefly luciferase assay as a reporter gene in mammalian cell lines. , 1989, BioTechniques.

[54]  J. Cleveland,et al.  Expression of functional Toll-like receptors 2 and 4 in human aortic valve interstitial cells: potential roles in aortic valve inflammation and stenosis. , 2008, American journal of physiology. Cell physiology.

[55]  H. S. Jung,et al.  Optimized THP-1 differentiation is required for the detection of responses to weak stimuli , 2007, Inflammation Research.

[56]  M. Thubrikar,et al.  The congenitally bicuspid aortic valve: how does it function? Why does it fail? , 2004, The Annals of thoracic surgery.