MicroRNAs profiling in murine models of acute and chronic asthma: a relationship with mRNAs targets

Nancy Garbacki, Emmanuel Di Valentin, Vân Anh Huynh-Thu, Pierre Geurts, Alexandre Irrthum, Céline Crahay, Thierry Arnould, Christophe Deroanne, Jacques Piette, Didier Cataldo, Alain Colige, Nancy Garbacki, Emmanuel Di Valentin, Vân Anh Huynh-Thu, Pierre Geurts, Alexandre Irrthum, Céline Crahay, Thierry Arnould, Christophe Deroanne, Jacques Piette, Didier Cataldo, Alain Colige

Abstract

Background: miRNAs are now recognized as key regulator elements in gene expression. Although they have been associated with a number of human diseases, their implication in acute and chronic asthma and their association with lung remodelling have never been thoroughly investigated.

Methodology/principal findings: In order to establish a miRNAs expression profile in lung tissue, mice were sensitized and challenged with ovalbumin mimicking acute, intermediate and chronic human asthma. Levels of lung miRNAs were profiled by microarray and in silico analyses were performed to identify potential mRNA targets and to point out signalling pathways and biological processes regulated by miRNA-dependent mechanisms. Fifty-eight, 66 and 75 miRNAs were found to be significantly modulated at short-, intermediate- and long-term challenge, respectively. Inverse correlation with the expression of potential mRNA targets identified mmu-miR-146b, -223, -29b, -29c, -483, -574-5p, -672 and -690 as the best candidates for an active implication in asthma pathogenesis. A functional validation assay was performed by cotransfecting in human lung fibroblasts (WI26) synthetic miRNAs and engineered expression constructs containing the coding sequence of luciferase upstream of the 3'UTR of various potential mRNA targets. The bioinformatics analysis identified miRNA-linked regulation of several signalling pathways, as matrix metalloproteinases, inflammatory response and TGF-β signalling, and biological processes, including apoptosis and inflammation.

Conclusions/significance: This study highlights that specific miRNAs are likely to be involved in asthma disease and could represent a valuable resource both for biological makers identification and for unveiling mechanisms underlying the pathogenesis of asthma.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Assessment of airway inflammation, sensitization…
Figure 1. Assessment of airway inflammation, sensitization and hyperresponsiveness.
Assessment of airway responsiveness to metacholine (Panel A), of glandular hyperplasia as percentage of goblet cells per total epithelial cells (Panel B), of peribronchial collagen deposition (Panel C) and of eosinophils accumulation (Panel D) in randomly selected bronchi in PBS and OVA-treated groups of mice at short-term (ST), intermediate-term (IT) and long-term (LT) sensitization and exposure protocols. Mean scores were measured as described in . Results are expressed as means ± SE and the comparison between groups was performed using Mann-Whitney U test (* p-value <0.05; ** p-value <0.005; *** p-value <0.001; N.S.: not significant).
Figure 2. Validation of microarray data by…
Figure 2. Validation of microarray data by real-time RT-qPCR.
Comparison of miRNA level regulation as determined by microarray hybridization (several probes per target) performed on pooled total RNA and by RT-qPCR performed on total RNA of each individual mice. Results are expressed as means ± SD. The value “1” is arbitrarily given when no change is observed. ST, IT, LT: short, intermediate and long-term treatments, respectively. FI: fold induction.
Figure 3. Potential regulation performed by miRNAs…
Figure 3. Potential regulation performed by miRNAs during the development of allergen-induced asthma.
This in silico prediction is based on significant inverse correlation between mRNA and miRNA modulation of expression as detailed in tables 4 and 5. ST, IT, LT: short, intermediate and long-term treatments, respectively. The observed up- (<$>\raster="rg1"<$>) and down- (<$>\raster="rg2"<$>) regulations of the expression of the selected miRNAs are reported.
Figure 4. Dose-response analysis of the effect…
Figure 4. Dose-response analysis of the effect of miR-29b, -29c and -146b on their predicted target in lung cells.
Transient transfection analysis for luciferase reporter expression with mouse Mmp-15 3′UTR in the presence of miR-29b and -29c (Panel A); mouse Mmp-24 3′UTR in the presence and absence of miR-29b and -29c (Panel B); human Mmp-15 3′UTR in the presence of miR-29b and -29c (Panel C); human Mmp-24 3′UTR in the presence of miR-29b and -29c (Panel D); mouse Col6a2 3′UTR in the presence of miR-29c (Panel E); mouse Ctsk 3′UTR in the presence of miR-29c (Panel F); mouse Scube2 3′UTR in the presence of miR-146b (Panel G); mouse Card10 3′UTR in the presence of miR-146b (Panel H). Universal negative siRNA was used at 20 nM as non-functional small RNA control. For each expression vector, the specific effect of the miRNA on luciferase activity was expressed as compared to the activity measured in the control condition, arbitrarily set at “100”. Results are expressed as mean ± SD. (* p-value <0.05; ** p-value <0.001; # p-value <0.005).
Figure 5. Analysis of 13 miRNAs-predicted target…
Figure 5. Analysis of 13 miRNAs-predicted target murine genes in vitro.
Transient transfection analysis for luciferase reporter expression with Arid4b, Il-6 or Lpin2 3′UTR in the presence of miR-223; with Gmnn, Nola2 or Ube2c 3′UTR in the presence of miR-483; with Dera or Nusap1 3′UTR in the presence of miR-574-5p; with Cd3g or Phb2 3′UTR in the presence of miR-672; and with Fst, Ctse or Cdca8 3′UTR in the presence of miR-690. Universal negative siRNA were used at 20 nM as non-functional small RNA control. For each expression vector, the specific effect of the miRNA on luciferase activity was expressed as compared to the activity measured in the control condition, arbitrarily set at “100”. Results are expressed as mean ± SD. Each p-value is indicated in the graph.
Figure 6. Potential influence of miRNAs on…
Figure 6. Potential influence of miRNAs on immune response induced by OVA.
In antigen presenting cells, MHC complexes are maturated in endosomes by lysosomal reductases while CTSE processes antigen (Ag), i.e. OVA. Finally, the MHC/antigenic peptide complex translocates to the plasma membrane and is presented to the TCR/CD3 complex on CD4+ T helper cell surface. Activation of TCR/CD3 induces the activation of NF-κB through PKC activation and CARD11/BCL10/MALT1 complex recruitment. MiRNA modulation could occur through mmu-miR-690, -672 and -146b. Cross-linking of costimulatory receptors on the T helper cell with corresponding ligands, such as TNFRSF9 with TNFSF9, also induces NF-κB and regulators (MAPKs, JNK) of the activity of multiple transcription factors. Production of TNFSF9 could be under the control of mmu-miR-146b, thus regulating the T helper cells properties. ST, IT, LT: short, intermediate and long-term treatments, respectively. The observed up- (\raster="rg1") and down- (\raster="rg2") regulations of the expression of some specific miRNAs are reported.
Figure 7. Regulatory pathways regulated by miRNAs…
Figure 7. Regulatory pathways regulated by miRNAs as determined by the MicroCosm Targets algorithm.
Fifty-one pathways were identified at one time-point at least. While 28 pathways appeared to be modulated at only one stage of the disease (ST, IT or LT), 17 were regulated at 2 different time-points and 6 during the entire course of the disease. Stouffer's method was used to identify significant enrichment for pathways annotations among predicted targets of modulated miRNA in the model. ST, IT, LT: short, intermediate and long-term treatments, respectively.
Figure 8. Regulatory pathways regulated by miRNAs…
Figure 8. Regulatory pathways regulated by miRNAs as determined by the TargetScan algorithm.
Fifty-three pathways were identified at one time-point at least. While 30 pathways appeared to be modulated at only one stage of the disease (ST, IT or LT), 18 were regulated at 2 different time-points and 5 during the entire study. Stouffer's method was used to identify significant enrichment for pathways annotations among predicted targets of modulated miRNA in the model. ST, IT, LT: short, intermediate and long-term treatments, respectively.
Figure 9. miRNAs and MMP-2 activation.
Figure 9. miRNAs and MMP-2 activation.
Panel A represents MMP-14 (MT1-MMP)-dependent activation pathway for MMP-2. TIMP-2 activates pro-MMP-2 by forming a complex that interacts with the MMP-14/TIMP-2 complex at the cell membrane. Activation of pro-MMP-2 occurs in a two step process: cleavage within the MMP-2 prodomain followed by an autocatalytic cleavage which results in the active 62 kD form. Panel B represents the presumed MMP-15 (MT2-MMP) and MMP-24 (MT5-MMP)-dependent activation pathway for MMP-2. Activation of pro-MMP-2 occurs in a two step process: cleavage within the MMP-2 prodomain in the absence of TIMP-2 followed by a second cleavage, enhanced by an unidentified secreted soluble protein which results in the active 62 kD form. The mechanism by which MMP-24 releases active MMP-2 is currently unknown. The intensive activation of MMP-2 contributes to collagen deposition and interstitial fibrosis. An excess of TIMP-2 and the extracellular matrix-anchored TIMP-3 contribute, respectively, to the degradation of pro-MMP-2 and to the inhibition of MMP-2. ST, IT, LT: short, intermediate and long-term treatments, respectively. The observed up- (\raster="rg1") and down- (\raster="rg2") regulations of the expression of some specific miRNA are reported.
Figure 10. Experimental protocol.
Figure 10. Experimental protocol.
Sensitization and short-term (ST), intermediate-term (IT) and long-term (LT) PBS/ovalbumin (OVA) exposure protocols. BALB/c male mice were sensitized on days 1 and 7 (ST) or 11 (IT and LT) by intraperitoneal injection of 10 µg OVA. At day 22, mice were subsequently exposed to PBS or OVA 1% aerosol for 30 min per day. For ST, aerosol challenge was performed for 7 consecutive days (grey box). For IT or LT, aerosol challenges were performed three or five times (black boxes) according to a pattern of 5-day inhalation (black boxes) followed by a 9-day time off (white boxes). Mice were sacrificed the day after last aerosol challenge.

References

    1. Lange P, Parner J, Vestbo J, Schnohr P, Jensen G. A 15-year follow-up study of ventilatory function in adults with asthma. N Engl J Med. 1998;339:1194–1200.
    1. Pascual RM, Peters SP. Airway remodeling contributes to the progressive loss of lung function in asthma: an overview. J Allergy Clin Immunol. 2005;116:477–486; quiz 487.
    1. Hirst SJ, Martin JG, Bonacci JV, Chan V, Fixman ED, et al. Proliferative aspects of airway smooth muscle. J Allergy Clin Immunol. 2004;114:S2–17.
    1. Di Valentin E, Crahay C, Garbacki N, Hennuy B, Gueders M, et al. New asthma biomarkers: lessons from murine models of acute and chronic asthma. Am J Physiol Lung Cell Mol Physiol. 2009;296:L185–197.
    1. Pennings JL, Kimman TG, Janssen R. Identification of a common gene expression response in different lung inflammatory diseases in rodents and macaques. PLoS ONE. 2008;3:e2596.
    1. Novershtern N, Itzhaki Z, Manor O, Friedman N, Kaminski N. A functional and regulatory map of asthma. Am J Respir Cell Mol Biol. 2008;38:324–336.
    1. Rolph MS, Sisavanh M, Liu SM, Mackay CR. Clues to asthma pathogenesis from microarray expression studies. Pharmacol Ther. 2006;109:284–294.
    1. Izuhara K, Saito H. Microarray-based identification of novel biomarkers in asthma. Allergol Int. 2006;55:361–367.
    1. Park SG, Choi JW, Kim H, Roh GS, Bok J, et al. Genome-wide profiling of antigen-induced time course expression using murine models for acute and chronic asthma. Int Arch Allergy Immunol. 2008;146:44–56.
    1. Zimmermann N, King NE, Laporte J, Yang M, Mishra A, et al. Dissection of experimental asthma with DNA microarray analysis identifies arginase in asthma pathogenesis. J Clin Invest. 2003;111:1863–1874.
    1. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–297.
    1. Griffiths-Jones S, Saini HK, van Dongen S, Enright AJ. miRBase: tools for microRNA genomics. Nucleic Acids Res. 2008;36:D154–158.
    1. Erson AE, Petty EM. MicroRNAs in development and disease. Clin Genet. 2008;74:296–306.
    1. Zhang B, Farwell MA. microRNAs: a new emerging class of players for disease diagnostics and gene therapy. J Cell Mol Med. 2008;12:3–21.
    1. Wang Y, Liang Y, Lu Q. MicroRNA epigenetic alterations: predicting biomarkers and therapeutic targets in human diseases. Clin Genet. 2008;74:307–315.
    1. Zhang B, Pan X, Cobb GP, Anderson TA. microRNAs as oncogenes and tumor suppressors. Dev Biol. 2007;302:1–12.
    1. Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer. 2006;6:857–866.
    1. Esquela-Kerscher A, Slack FJ. Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 2006;6:259–269.
    1. Wiemer EA. The role of microRNAs in cancer: no small matter. Eur J Cancer. 2007;43:1529–1544.
    1. Ma L, Teruya-Feldstein J, Weinberg RA. Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature. 2007;449:682–688.
    1. Thum T, Catalucci D, Bauersachs J, Thum T, Catalucci D, et al. MicroRNAs: novel regulators in cardiac development and disease. [see comment]. Cardiovascular Research. 2008;79:562–570.
    1. Catalucci D, Latronico MV, Condorelli G, Catalucci D, Latronico MVG, et al. MicroRNAs control gene expression: importance for cardiac development and pathophysiology. Annals of the New York Academy of Sciences. 2008;1123:20–29.
    1. Callis TE, Chen JF, Wang DZ, Callis TE, Chen J-F, et al. MicroRNAs in skeletal and cardiac muscle development. DNA & Cell Biology. 2007;26:219–225.
    1. Poy MN, Eliasson L, Krutzfeldt J, Kuwajima S, Ma X, et al. A pancreatic islet-specific microRNA regulates insulin secretion. Nature. 2004;432:226–230.
    1. Hennessy E, O'Driscoll L, Hennessy E, O'Driscoll L. Molecular medicine of microRNAs: structure, function and implications for diabetes. Expert Reviews in Molecular Medicine. 2008;10:e24.
    1. Esau C, Davis S, Murray SF, Yu XX, Pandey SK, et al. miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell Metab. 2006;3:87–98.
    1. Poy MN, Spranger M, Stoffel M. microRNAs and the regulation of glucose and lipid metabolism. Diabetes Obes Metab. 2007;9(Suppl 2):67–73.
    1. Gottwein E, Cullen BR, Gottwein E, Cullen BR. Viral and cellular microRNAs as determinants of viral pathogenesis and immunity. Cell Host & Microbe. 2008;3:375–387.
    1. Lecellier CH, Dunoyer P, Arar K, Lehmann-Che J, Eyquem S, et al. A cellular microRNA mediates antiviral defense in human cells. Science. 2005;308:557–560.
    1. Cullen BR. Viruses and microRNAs. Nat Genet. 2006;38(Suppl):S25–30.
    1. Izzotti A, Calin GA, Arrigo P, Steele VE, Croce CM, et al. Downregulation of microRNA expression in the lungs of rats exposed to cigarette smoke. Faseb J. 2009;23:806–812.
    1. Nana-Sinkam SP, Hunter MG, Nuovo GJ, Schmittgen TD, Gelinas R, et al. Integrating the MicroRNome into the study of lung disease. Am J Respir Crit Care Med. 2009;179:4–10.
    1. Moschos SA, Williams AE, Perry MM, Birrell MA, Belvisi MG, et al. Expression profiling in vivo demonstrates rapid changes in lung microRNA levels following lipopolysaccharide-induced inflammation but not in the anti-inflammatory action of glucocorticoids. BMC Genomics. 2007;8:240.
    1. Williams AE, Larner-Svensson H, Perry MM, Campbell GA, Herrick SE, et al. MicroRNA expression profiling in mild asthmatic human airways and effect of corticosteroid therapy. PLoS ONE. 2009;4:e5889.
    1. Lu TX, Munitz A, Rothenberg ME. MicroRNA-21 is up-regulated in allergic airway inflammation and regulates IL-12p35 expression. J Immunol. 2009;182:4994–5002.
    1. Mattes J, Collison A, Plank M, Phipps S, Foster PS. Antagonism of microRNA-126 suppresses the effector function of TH2 cells and the development of allergic airways disease. Proc Natl Acad Sci U S A. 2009;106:18704–18709.
    1. Chiba Y, Tanabe M, Goto K, Sakai H, Misawa M. Down-Regulation of miR-133a Contributes to Up-Regulation of RhoA in Bronchial Smooth Muscle Cells. Am J Respir Crit Care Med. 2009;180:713–719.
    1. Polikepahad S, Knight JM, Naghavi AO, Oplt T, Creighton CJ, et al. Proinflammatory role for let-7 microRNAS in experimental asthma. J Biol Chem. 2010;285:30139–30149.
    1. Southam DS, Ellis R, Wattie J, Glass W, Inman MD. Goblet cell rebound and airway dysfunction with corticosteroid withdrawal in a mouse model of asthma. Am J Respir Crit Care Med. 2008;178:1115–1122.
    1. Gueders MM, Paulissen G, Crahay C, Quesada-Calvo F, Hacha J, et al. Mouse models of asthma: a comparison between C57BL/6 and BALB/c strains regarding bronchial responsiveness, inflammation, and cytokine production. Inflamm Res. 2009;58:845–854.
    1. Van Hove CL, Maes T, Cataldo DD, Gueders MM, Palmans E, et al. Comparison of acute inflammatory and chronic structural asthma-like responses between C57BL/6 and BALB/c mice. Int Arch Allergy Immunol. 2009;149:195–207.
    1. Gueders MM, Bertholet P, Perin F, Rocks N, Maree R, et al. A novel formulation of inhaled doxycycline reduces allergen-induced inflammation, hyperresponsiveness and remodeling by matrix metalloproteinases and cytokines modulation in a mouse model of asthma. Biochemical Pharmacology. 2008;75:514–526.
    1. Tang X, Muniappan L, Tang G, Ozcan S. Identification of glucose-regulated miRNAs from pancreatic {beta} cells reveals a role for miR-30d in insulin transcription. Rna. 2009;15:287–293.
    1. Malumbres R, Sarosiek KA, Cubedo E, Ruiz JW, Jiang X, et al. Differentiation stage-specific expression of microRNAs in B lymphocytes and diffuse large B-cell lymphomas. Blood. 2009;113:3754–3764.
    1. Baltimore D, Boldin MP, O'Connell RM, Rao DS, Taganov KD. MicroRNAs: new regulators of immune cell development and function. Nat Immunol. 2008;9:839–845.
    1. Bellon M, Lepelletier Y, Hermine O, Nicot C. Deregulation of microRNA involved in hematopoiesis and the immune response in HTLV-I adult T-cell leukemia. Blood. 2009;113:4914–4917.
    1. Johnnidis JB, Harris MH, Wheeler RT, Stehling-Sun S, Lam MH, et al. Regulation of progenitor cell proliferation and granulocyte function by microRNA-223. Nature. 2008;451:1125–1129.
    1. Anglicheau D, Sharma VK, Ding R, Hummel A, Snopkowski C, et al. MicroRNA expression profiles predictive of human renal allograft status. Proc Natl Acad Sci U S A. 2009;106:5330–5335.
    1. Laios A, O'Toole S, Flavin R, Martin C, Kelly L, et al. Potential role of miR-9 and miR-223 in recurrent ovarian cancer. Mol Cancer. 2008;7:35.
    1. Wong QW, Lung RW, Law PT, Lai PB, Chan KY, et al. MicroRNA-223 is commonly repressed in hepatocellular carcinoma and potentiates expression of Stathmin1. Gastroenterology. 2008;135:257–269.
    1. Gottardo F, Liu CG, Ferracin M, Calin GA, Fassan M, et al. Micro-RNA profiling in kidney and bladder cancers. Urol Oncol. 2007;25:387–392.
    1. Wang J, Xu R, Lin F, Zhang S, Zhang G, et al. MicroRNA: novel regulators involved in the remodeling and reverse remodeling of the heart. Cardiology. 2009;113:81–88.
    1. Walden TB, Timmons JA, Keller P, Nedergaard J, Cannon B. Distinct expression of muscle-specific microRNAs (myomirs) in brown adipocytes. J Cell Physiol. 2009;218:444–449.
    1. Pekarsky Y, Santanam U, Cimmino A, Palamarchuk A, Efanov A, et al. Tcl1 expression in chronic lymphocytic leukemia is regulated by miR-29 and miR-181. Cancer Res. 2006;66:11590–11593.
    1. Yanaihara N, Caplen N, Bowman E, Seike M, Kumamoto K, et al. Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell. 2006;9:189–198.
    1. Porkka KP, Pfeiffer MJ, Waltering KK, Vessella RL, Tammela TL, et al. MicroRNA expression profiling in prostate cancer. Cancer Res. 2007;67:6130–6135.
    1. Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, et al. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005;65:7065–7070.
    1. Fabbri M, Garzon R, Cimmino A, Liu Z, Zanesi N, et al. MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B. Proc Natl Acad Sci U S A. 2007;104:15805–15810.
    1. Park SY, Lee JH, Ha M, Nam JW, Kim VN. miR-29 miRNAs activate p53 by targeting p85 alpha and CDC42. Nat Struct Mol Biol. 2009;16:23–29.
    1. Sengupta S, den Boon JA, Chen IH, Newton MA, Stanhope SA, et al. MicroRNA 29c is down-regulated in nasopharyngeal carcinomas, up-regulating mRNAs encoding extracellular matrix proteins. Proc Natl Acad Sci U S A. 2008;105:5874–5878.
    1. Wang H, Garzon R, Sun H, Ladner KJ, Singh R, et al. NF-kappaB-YY1-miR-29 regulatory circuitry in skeletal myogenesis and rhabdomyosarcoma. Cancer Cell. 2008;14:369–381.
    1. Schmidt WM, Spiel AO, Jilma B, Wolzt M, Muller M. In vivo profile of the human leukocyte microRNA response to endotoxemia. Biochem Biophys Res Commun. 2009;380:437–441.
    1. Taganov KD, Boldin MP, Chang KJ, Baltimore D. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci USA. 2006;103:12481–12486.
    1. Bhaumik D, Scott GK, Schokrpur S, Patil CK, Campisi J, et al. Expression of microRNA-146 suppresses NF-[kappa]B activity with reduction of metastatic potential in breast cancer cells. Oncogene. 2008;27:5643–5647.
    1. Park SE, Xu J, Frolova A, Liao L, O'Malley BW, et al. Genetic deletion of the repressor of estrogen receptor activity (REA) enhances the response to estrogen in target tissues in vivo. Mol Cell Biol. 2005;25:1989–1999.
    1. Luo L, Yang X, Takihara Y, Knoetgen H, Kessel M. The cell-cycle regulator geminin inhibits Hox function through direct and polycomb-mediated interactions. Nature. 2004;427:749–753.
    1. Wolgemuth DJ. Function of cyclins in regulating the mitotic and meiotic cell cycles in male germ cells. Cell Cycle. 2008;7:3509–3513.
    1. Yamanaka Y, Heike T, Kumada T, Shibata M, Takaoka Y, et al. Loss of Borealin/DasraB leads to defective cell proliferation, p53 accumulation and early embryonic lethality. Mech Dev. 2008;125:441–450.
    1. Raemaekers T, Ribbeck K, Beaudouin J, Annaert W, Van Camp M, et al. NuSAP, a novel microtubule-associated protein involved in mitotic spindle organization. J Cell Biol. 2003;162:1017–1029.
    1. Buhling F, Rocken C, Brasch F, Hartig R, Yasuda Y, et al. Pivotal role of cathepsin K in lung fibrosis. Am J Pathol. 2004;164:2203–2216.
    1. Ohta K, Yamashita N, Tajima M, Miyasaka T, Kawashima R, et al. In vivo effects of apoptosis in asthma examined by a murine model. Int Arch Allergy Immunol. 2001;124:259–261.
    1. Noma T, Sugawara Y, Aoki K, Kawano Y, Ishikawa Y, et al. Induction of peripheral mononuclear cell apoptosis in asthmatic patients in remission. J Asthma. 2002;39:591–601.
    1. Wang L, Guo Y, Huang WJ, Ke X, Poyet JL, et al. Card10 is a novel caspase recruitment domain/membrane-associated guanylate kinase family member that interacts with BCL10 and activates NF-kappa B. J Biol Chem. 2001;276:21405–21409.
    1. Yang RB, Ng CK, Wasserman SM, Colman SD, Shenoy S, et al. Identification of a novel family of cell-surface proteins expressed in human vascular endothelium. J Biol Chem. 2002;277:46364–46373.
    1. Akira S, Kishimoto T. IL-6 and NF-IL6 in acute-phase response and viral infection. Immunol Rev. 1992;127:25–50.
    1. Nishioku T, Hashimoto K, Yamashita K, Liou SY, Kagamiishi Y, et al. Involvement of cathepsin E in exogenous antigen processing in primary cultured murine microglia. J Biol Chem. 2002;277:4816–4822.
    1. Shin SM, Kim YH, Choi BK, Kwon PM, Lee HW, et al. 4-1BB triggers IL-13 production from T cells to limit the polarized, Th1-mediated inflammation. J Leukoc Biol. 2007;81:1455–1465.
    1. Polte T, Foell J, Werner C, Hoymann HG, Braun A, et al. CD137-mediated immunotherapy for allergic asthma. J Clin Invest. 2006;116:1025–1036.
    1. Seo SK, Choi JH, Kim YH, Kang WJ, Park HY, et al. 4-1BB-mediated immunotherapy of rheumatoid arthritis. Nat Med. 2004;10:1088–1094.
    1. Foell J, Strahotin S, O'Neil SP, McCausland MM, Suwyn C, et al. CD137 costimulatory T cell receptor engagement reverses acute disease in lupus-prone NZB x NZW F1 mice. J Clin Invest. 2003;111:1505–1518.
    1. Kuang W, Tan J, Duan Y, Duan J, Wang W, et al. Cyclic stretch induced miR-146a upregulation delays C2C12 myogenic differentiation through inhibition of Numb. Biochem Biophys Res Commun. 2009;378:259–263.
    1. Pece S, Serresi M, Santolini E, Capra M, Hulleman E, et al. Loss of negative regulation by Numb over Notch is relevant to human breast carcinogenesis. J Cell Biol. 2004;167:215–221.
    1. Nal B, Mohr E, Silva MI, Tagett R, Navarro C, et al. Wdr12, a mouse gene encoding a novel WD-Repeat Protein with a notchless-like amino-terminal domain. Genomics. 2002;79:77–86.
    1. Mestdagh P, Van Vlierberghe P, De Weer A, Muth D, Westermann F, et al. A novel and universal method for microRNA RT-qPCR data normalization. Genome Biol. 2009;10:R64.
    1. Tzur G, Levy A, Meiri E, Barad O, Spector Y, et al. MicroRNA expression patterns and function in endodermal differentiation of human embryonic stem cells. PLoS ONE. 2008;3:e3726.
    1. Chang TC, Yu D, Lee YS, Wentzel EA, Arking DE, et al. Widespread microRNA repression by Myc contributes to tumorigenesis. Nat Genet. 2008;40:43–50.
    1. Betel D, Wilson M, Gabow A, Marks DS, Sander C. The resource: targets and expression. Nucleic Acids Res. 2008;36:D149–153.
    1. Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB. Prediction of mammalian microRNA targets. Cell. 2003;115:787–798.
    1. Gueders MM, Foidart JM, Noel A, Cataldo DD. Matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs in the respiratory tract: potential implications in asthma and other lung diseases. Eur J Pharmacol. 2006;533:133–144.
    1. Henderson N, Markwick LJ, Elshaw SR, Freyer AM, Knox AJ, et al. Collagen I and thrombin activate MMP-2 by MMP-14-dependent and -independent pathways: implications for airway smooth muscle migration. Am J Physiol Lung Cell Mol Physiol. 2007;292:L1030–1038.
    1. Zucker S, Drews M, Conner C, Foda HD, DeClerck YA, et al. Tissue inhibitor of metalloproteinase-2 (TIMP-2) binds to the catalytic domain of the cell surface receptor, membrane type 1-matrix metalloproteinase 1 (MT1-MMP). J Biol Chem. 1998;273:1216–1222.
    1. Murphy G, Nagase H. Progress in matrix metalloproteinase research. Mol Aspects Med. 2008;29:290–308.
    1. Barnes PJ, Drazen JM, Rennard S, Thomson NC, editors. Asthma and COPD, Basic Mechanisms and Clinical Management Academic Press. 2002. 780
    1. Zhu ML, Kyprianou N. Androgen receptor and growth factor signaling cross-talk in prostate cancer cells. Endocr Relat Cancer. 2008;15:841–849.
    1. Recchia AG, Musti AM, Lanzino M, Panno ML, Turano E, et al. A cross-talk between the androgen receptor and the epidermal growth factor receptor leads to p38MAPK-dependent activation of mTOR and cyclinD1 expression in prostate and lung cancer cells. Int J Biochem Cell Biol. 2009;41:603–614.
    1. Yamashita N, Tashimo H, Ishida H, Matsuo Y, Arai H, et al. Role of insulin-like growth factor-I in allergen-induced airway inflammation and remodeling. Cell Immunol. 2005;235:85–91.
    1. Gosens R, Schaafsma D, Grootte Bromhaar MM, Vrugt B, Zaagsma J, et al. Growth factor-induced contraction of human bronchial smooth muscle is Rho-kinase-dependent. Eur J Pharmacol. 2004;494:73–76.
    1. Delafontaine P, Song YH, Li Y. Expression, regulation, and function of IGF-1, IGF-1R, and IGF-1 binding proteins in blood vessels. Arterioscler Thromb Vasc Biol. 2004;24:435–444.
    1. Asirvatham AJ, Magner WJ, Tomasi TB. miRNA regulation of cytokine genes. Cytokine. 2009;45:58–69.
    1. Sheedy FJ, O'Neill LA. Adding fuel to fire: microRNAs as a new class of mediators of inflammation. Ann Rheum Dis. 2008;67(Suppl 3):iii50–55.
    1. Brightling C, Berry M, Amrani Y. Targeting TNF-alpha: a novel therapeutic approach for asthma. J Allergy Clin Immunol. 2008;121:5–10; quiz 11-12.
    1. Thomas PS. Tumour necrosis factor-alpha: the role of this multifunctional cytokine in asthma. Immunol Cell Biol. 2001;79:132–140.
    1. van Rooij E, Sutherland LB, Thatcher JE, DiMaio JM, Naseem RH, et al. Dysregulation of microRNAs after myocardial infarction reveals a role of miR-29 in cardiac fibrosis. Proc Natl Acad Sci U S A. 2008;105:13027–13032.
    1. Li Z, Hassan MQ, Jafferji M, Aqeilan RI, Garzon R, et al. Biological functions of miR-29b contribute to positive regulation of osteoblast differentiation. J Biol Chem. 2009;284:15676–15684.
    1. Kuhn C, 3rd, Boldt J, King TE, Jr, Crouch E, Vartio T, et al. An immunohistochemical study of architectural remodeling and connective tissue synthesis in pulmonary fibrosis. Am Rev Respir Dis. 1989;140:1693–1703.
    1. Muro AF, Moretti FA, Moore BB, Yan M, Atrasz RG, et al. An essential role for fibronectin extra type III domain A in pulmonary fibrosis. Am J Respir Crit Care Med. 2008;177:638–645.
    1. Shan SW, Lee DY, Deng Z, Shatseva T, Jeyapalan Z, et al. MicroRNA MiR-17 retards tissue growth and represses fibronectin expression. Nat Cell Biol. 2009;11:1031–1038.
    1. Zhang X, Liu S, Hu T, Liu S, He Y, et al. Up-regulated microRNA-143 transcribed by nuclear factor kappa B enhances hepatocarcinoma metastasis by repressing fibronectin expression. Hepatology. 2009;50:490–499.
    1. Wang Q, Wang Y, Minto AW, Wang J, Shi Q, et al. MicroRNA-377 is up-regulated and can lead to increased fibronectin production in diabetic nephropathy. Faseb J. 2008;22:4126–4135.
    1. Saotome A, Kanai N, Nagai T, Yashiro T, Tokudome S. Immunohistochemical classification of the localization of laminin in the thickened bronchial epithelial basement membrane of deceased bronchial asthma patients. Respir Med. 2003;97:688–694.
    1. Meeker TC, Hardy D, Willman C, Hogan T, Abrams J. Activation of the interleukin-3 gene by chromosome translocation in acute lymphocytic leukemia with eosinophilia. Blood. 1990;76:285–289.
    1. Asquith KL, Ramshaw HS, Hansbro PM, Beagley KW, Lopez AF, et al. The IL-3/IL-5/GM-CSF common receptor plays a pivotal role in the regulation of Th2 immunity and allergic airway inflammation. J Immunol. 2008;180:1199–1206.
    1. Fabry B, Fredberg JJ. Mechanotransduction, asthma, and airway smooth muscle. Drug Discov Today Dis Models. 2007;4:131–137.
    1. Turner CE. Molecules in focus Paxillin. The International Journal of Biochemistry & Cell Biology. 1998;30:955–959.
    1. Opazo Saez A, Zhang W, Wu Y, Turner CE, Tang DD, et al. Tension development during contractile stimulation of smooth muscle requires recruitment of paxillin and vinculin to the membrane. Am J Physiol Cell Physiol. 2004;286:C433–447.
    1. Ebner S, Hofer S, Nguyen VA, Furhapter C, Herold M, et al. A novel role for IL-3: human monocytes cultured in the presence of IL-3 and IL-4 differentiate into dendritic cells that produce less IL-12 and shift Th cell responses toward a Th2 cytokine pattern. J Immunol. 2002;168:6199–6207.
    1. Moser M, Murphy KM. Dendritic cell regulation of TH1-TH2 development. Nat Immunol. 2000;1:199–205.
    1. Gardino AK, Smerdon SJ, Yaffe MB. Structural determinants of 14-3-3 binding specificities and regulation of subcellular localization of 14-3-3-ligand complexes: a comparison of the X-ray crystal structures of all human 14-3-3 isoforms. Semin Cancer Biol. 2006;16:173–182.
    1. Agarwal-Mawal A, Qureshi HY, Cafferty PW, Yuan Z, Han D, et al. 14-3-3 connects glycogen synthase kinase-3 beta to tau within a brain microtubule-associated tau phosphorylation complex. J Biol Chem. 2003;278:12722–12728.
    1. Zhu Q, Hong A, Sheng N, Zhang X, Matejko A, et al. microParaflo biochip for nucleic acid and protein analysis. Methods Mol Biol. 2007;382:287–312.
    1. Bolstad BM, Irizarry RA, Astrand M, Speed TP. A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics. 2003;19:185–193.
    1. Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 2003;4:249–264.
    1. John B, Enright AJ, Aravin A, Tuschl T, Sander C, et al. Human MicroRNA targets. PLoS Biol. 2004;2:e363.
    1. Rehmsmeier M, Steffen P, Hochsmann M, Giegerich R. Fast and effective prediction of microRNA/target duplexes. Rna. 2004;10:1507–1517.
    1. Pico AR, Kelder T, van Iersel MP, Hanspers K, Conklin BR, et al. WikiPathways: pathway editing for the people. PLoS Biol. 2008;6:e184.
    1. Leroy Folks J, Krishnaiah PR, Sen PK. Handbook of Statistics Elsevier; 1984. 6 Combination of independent tests. pp. 113–121.

Source: PubMed

3
Abonnieren