We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Long-term genome-wide blood RNA expression profiles yield novel molecular response candidates for IFN-β-1b treatment in relapsing remitting MS

    Robert H Goertsches

    † Author for correspondence

    Department of Neurology, University of Rostock, Gehlsheimer Str. 20, 18047 Rostock, Germany.

    Leibniz Institute for Natural Product Research & Infection Biology – Hans Knöll Institute, Jena, Germany

    ,
    Michael Hecker

    Leibniz Institute for Natural Product Research & Infection Biology – Hans Knöll Institute, Jena, Germany

    ,
    Dirk Koczan

    Institute of Immunology, University of Rostock, Schillingallee 70, 18055 Rostock, Germany

    ,
    Pablo Serrano-Fernandez

    Institute of Immunology, University of Rostock, Schillingallee 70, 18055 Rostock, Germany

    ,
    Steffen Moeller

    Institute of Immunology, University of Rostock, Schillingallee 70, 18055 Rostock, Germany

    ,
    Hans-Juergen Thiesen

    Institute of Immunology, University of Rostock, Schillingallee 70, 18055 Rostock, Germany

    &
    Uwe K Zettl

    Department of Neurology, University of Rostock, Gehlsheimer Str. 20, 18047 Rostock, Germany.

    Published Online:https://doi.org/10.2217/pgs.09.152

    Aims: In multiple sclerosis patients, treatment with recombinant IFN-β (rIFN-β) is partially efficient in reducing clinical exacerbations. However, its molecular mechanism of action is still under scrutiny. Materials & methods: We used DNA microarrays (Affymetrix, CA, USA) and peripheral mononuclear blood cells from 25 relapsing remitting multiple sclerosis patients to analyze the longitudinal transcriptional profile within 2 years of rIFN-β administration. Sets of differentially expressed genes were attained by applying a combination of independent criteria, thereby providing efficient data curation and gene filtering that accounted for technical and biological noise. Gene ontology term-association analysis and scientific literature text mining were used to explore evidence of gene interaction. Results: Post-therapy initiation, we identified 42 (day 2), 175 (month 1), 103 (month 12) and 108 (month 24) differentially expressed genes. Increased expression of established IFN-β marker genes, as well as differential expression of circulating IFN-β-responsive candidate genes, were observed. MS4A1 (CD20), a known target of B-cell depletion therapy, was significantly downregulated after one month. CMPK2, FCER1A, and FFAR2 appeared as hitherto unrecognized multiple sclerosis treatment-related differentially expressed genes that were consistently modulated over time. Overall, 84 interactions between 54 genes were attained, of which two major gene networks were identified at an earlier stage of therapy: the first (n = 15 genes) consisted of mostly known IFN-β-activated genes, whereas the second (n = 12) mainly contained downregulated genes that to date have not been associated with IFN-β effects in multiple sclerosis array research. Conclusion: We achieved both a broadening of the knowledge of IFN-β mechanism-of-action-related constituents and the identification of time-dependent interactions between IFN-β regulated genes.

    Papers of special note have been highlighted as: ▪▪ of considerable interest

    Bibliography

    • Hauser SL, Oksenberg JR: The neurobiology of multiple sclerosis: genes, inflammation, and neurodegeneration. Neuron52(1),61–76 (2006).
    • Hemmer B, Hartung HP: Toward the development of rational therapies in multiple sclerosis: what is on the horizon? Ann. Neurol.62(4),314–326 (2007).
    • Wiendl H, Toyka KV, Rieckmann P, Gold R, Hartung HP, Hohlfeld R; Multiple Sclerosis Therapy Consensus Group (MSTCG): Basic and escalating immunomodulatory treatments in multiple sclerosis: current therapeutic recommendations. J. Neurol.255(10),1449–1463 (2008).
    • Villoslada P, Steinman L, Baranzini SE: Systems biology and its application to the understanding of neurological diseases. Ann. Neurol.65(2),124–139 (2009).▪▪ Overview of accessible systems biology approaches in the field of neurological diseases, and describes the driving forces to complement classic reductionist approaches in the biomedical sciences.
    • Quintana FJ, Farez MF, Weiner HL: Systems biology approaches for the study of multiple sclerosis. J. Cell. Mol. Med.12(4),1087–1093 (2008).
    • Stark GR, Kerr IM, Williams BR, Silverman RH, Schreiber RD: How cells respond to interferons. Annu. Rev. Biochem.67,227–264 (1998).
    • Der SD, Zhou A, Williams BR, Silverman RH: Identification of genes differentially regulated by interferon α, β, or γ using oligonucleotide arrays. Proc. Natl Acad. Sci. USA95(26),15623–15628 (1998).
    • de Veer MJ, Holko M, Frevel M et al.: Functional classification of interferon-stimulated genes identified using microarrays. J. Leukocyte Biol.69(6),912–920 (2001).
    • Rani MR, Shrock J, Appachi S, Rudick RA, Williams BR, Ransohoff RM: Novel interferon-β-induced gene expression in peripheral blood cells. J. Leukoc. Biol.82(5),1353–1360 (2007).
    • 10  Wandinger KP, Sturzebecher CS, Bielekova B et al.: Complex immunomodulatory effects of interferon-β in multiple sclerosis include the upregulation of T helper 1-associated marker genes. Ann. Neurol.50(3),349–357 (2001).
    • 11  Koike F, Satoh J, Miyake S et al.: Microarray analysis identifies interferon β-regulated genes in multiple sclerosis. J. Neuroimmunol.139(1-2),109–118 (2003).
    • 12  Sturzebecher S, Wandinger KP, Rosenwald A et al.: Expression profiling identifies responder and non-responder phenotypes to interferon-β in multiple sclerosis. Brain126(6),1419–1429 (2003).
    • 13  Weinstock-Guttman B, Badgett D, Patrick K et al.: Genomic effects of IFN-β in multiple sclerosis patients. J. Immunol.171(5),2694–2702 (2003).
    • 14  Achiron A, Gurevich M, Magalashvili D, Kishner I, Dolev M, Mandel M: Understanding autoimmune mechanisms in multiple sclerosis using gene expression microarrays: treatment effect and cytokine-related pathways. Clin. Dev. Immunol.11(3-4),299–305 (2004).
    • 15  Iglesias AH, Camelo S, Hwang D, Villanueva R, Stephanopoulos G, Dangond F: Microarray detection of E2F pathway activation and other targets in multiple sclerosis peripheral blood mononuclear cells. J. Neuroimmunol.150(1-2),163–177 (2004).
    • 16  Hong J, Zang YC, Hutton G, Rivera VM, Zhang JZ: Gene expression profiling of relevant biomarkers for treatment evaluation in multiple sclerosis. J. Neuroimmunol.152(1–2),126–139 (2004).
    • 17  Baranzini SE, Mousavi P, Rio J et al.: Transcription-based prediction of response to IFNβ using supervised computational methods. PLoS Biol.3(1),E2(2005).
    • 18  Satoh J, Nakanishi M, Koike F et al.: T cell gene expression profiling identifies distinct subgroups of Japanese multiple sclerosis patients. J. Neuroimmunol.174(1–2),108–118 (2006).
    • 19  Satoh J, Nanri Y, Tabunoki H, Yamamura T: Microarray analysis identifies a set of CXCR3 and CCR2 ligand chemokines as early IFNβ-responsive genes in peripheral blood lymphocytes in vitro: an implication for IFNβ-related adverse effects in multiple sclerosis. BMC Neurol.19,6–18 (2006).
    • 20  Palacios R, Goni J, Martinez-Forero I et al.: A network analysis of the human T-cell activation gene network identifies JAGGED1 as a therapeutic target for autoimmune diseases. PLoS ONE2(11),E1222 (2007).
    • 21  Annibali V, Di Giavanni S, Cannoni S et al.: Gene expression profiles reveal homeostatic dynamics during interferon-β therapy in multiple sclerosis. Autoimmunity40(1),16–22 (2007).
    • 22  Singh MK, Scott TF, La Framboise WA, Hu FZ, Post JC, Ehrlich GD: Gene expression changes in peripheral blood mononuclear cells from multiple sclerosis patients undergoing β-interferon therapy. J. Neurol. Sci.258(1–2),52–59 (2007).
    • 23  Fernald GH, Knott S, Pachner A et al.: Genome-wide network analysis reveals the global properties of IFN-β immediate transcriptional effects in humans. J. Immunol.178(8),5076–5085 (2007).▪▪ First study in the field that incorporated network analysis to explore gene regulation in response to recombinant IFN-β. Effective demonstration of the combination of data- and knowledge-driven analysis.
    • 24  Reder AT, Velichko S, Yamaguchi KD et al.: IFN-β1b induces transient and variable gene expression in relapsing-remitting multiple sclerosis patients independent of neutralizing antibodies or changes in IFN receptor RNA expression. J. Interferon Cytokine Res.28(5),317–331 (2008).
    • 25  Hilpert J, Beekman JM, Schwenke S et al.: Biological response genes after single dose administration of interferon β-1b to healthy male volunteers. J. Neuroimmunol.199(1–2),115–125 (2008).
    • 26  Weinstock-Guttman B, Bhasi K, Badgett D et al.: Genomic effects of once-weekly, intramuscular interferon-β1a treatment after the first dose and on chronic dosing: Relationships to 5-year clinical outcomes in multiple sclerosis patients. J. Neuroimmunol.205(1-2),113–125 (2008).
    • 27  Goertsches RH, Hecker M, Zettl UK: Monitoring of multiple sclerosis immunotherapy: from single candidates to biomarker networks. J. Neurol.225 (Suppl. 6),48–57 (2008).
    • 28  Borden EC, Sen GC, Uze G et al.: Interferons at age 50: past, current and future impact on biomedicine. Nat. Rev. Drug Discov.6(12),975–990 (2007).▪▪ Accessible and highly exhaustive description of interferon modes of action, and discussion of its relevance.
    • 29  van Baarsen LGM, Vosslamber S, Tijssen M et al.: Pharmacogenomics of interferon-β therapy in multiple sclerosis: baseline IFN signature determines pharmacological differences between patients. PLoS One3(4),E1927 (2008).
    • 30  Satoh J, Illes Z, Peterfalvi A, Tabunoki H, Rozsa C, Yamamura T: Aberrant transcriptional regulatory network in T cells of multiple sclerosis. Neurosci. Lett.422(1),30–33 (2007).
    • 31  Gneiss C, Tripp P, Reichartseder F et al.: Differing immunogenic potentials of interferon b preparations in multiple sclerosis patients. Mult. Scler.12(6),731–737 (2006).
    • 32  McDonald WI, Compston A, Edan G et al.: Recommended diagnostic criteria for multiple sclerosis: guidelines from the International Panel on the Diagnosis of Multiple Sclerosis. Ann. Neurol.50(1),121–127 (2001).
    • 33  Yang IV, Chen E, Hasseman JP et al.: Within the fold: assessing differential expression measures and reproducibility in microarray assays. Genome Biol.3(11), research0062 (2002).
    • 34  Hecker M, Goertsches RH, Engelmann R, Thiesen HJ, Guthke R: Integrative modelling of transcriptional regulation in response to antirheumatic therapy. BMC Bioinformatics10(1),262 (2009).▪▪ Bioinformatics study that presents an analytical tool that incorporates and adjusts for the variability in gene expression intensities. Respective MA-plot-based signal intensity-dependent fold-change criterion (MAID) filtering was applied in the present study.
    • 35  Pepper SD, Saunders EK, Edwards LE, Wilson CL, Miller CJ: The utility of MAS5 expression summary and detection call algorithms. BMC Bioinformatics30(8),273 (2007).
    • 36  Ferrari F, Bortoluzzi S, Coppe A et al.: Novel definition files for human GeneChips based on GeneAnnot. BMC Bioinformatics15(8),446 (2007).
    • 37  Chalifa-Caspi V, Yanai I, Ophir R et al.: GeneAnnot: comprehensive two-way linking between oligonucleotide array probesets and GeneCards genes. Bioinformatics20(9),1457–1458 (2004).
    • 38  Ashburner M, Ball CA, Blake JA et al.; The Gene Ontology Consortium: Gene Ontology: tool for the unification of biology. Nat. Genet.25(1),25–29 (2000).
    • 39  Falcon S, Gentleman R: Using GOstats to test gene lists for GO term association. Bioinformatics23(2),257–258 (2007).
    • 40  Shannon P, Markiel A, Ozier O et al.: Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res.13(11),2498–2504 (2003).
    • 41  Gilli F, Marnetto F, Caldano M et al.: Biological responsiveness to first injections of interferon-β in patients with multiple sclerosis. J. Neuroimmunol.158(1–2),195–203 (2005).
    • 42  Brown AJ, Jupe S, Briscoe CP: A family of fatty acid binding receptors. DNA Cell Biol.24(1),54–61 (2005).
    • 43  Hirasawa A, Hara T, Katsuma S, Adachi T, Tsujimoto G: Free fatty acid receptors and drug discovery. Biol. Pharm. Bull.31(10),1847–1851 (2008).
    • 44  Hemmi H, Kaisho T, Takeuchi O et al.: Small anti-viral compounds activate immune cells via the TLR7 MyD88-dependent signaling pathway. Nat. Immunol.3(2),196–200 (2002).
    • 45  O’Neill LA: Targeting signal transduction as a strategy to treat inflammatory diseases. Nat. Rev. Drug Discov.5(7),549–563 (2006).
    • 46  Schulthess FT, Paroni F, Sauter NS et al.: CXCL10 impairs b cell function and viability in diabetes through TLR4 signaling. Cell Metab.9(2),125–139 (2009).
    • 47  Krumbholz M, Faber H, Steinmeyer F et al.: interferon-β increases BAFF levels in multiple sclerosis: implications for B cell autoimmunity. Brain131(6),1455–1463 (2008).
    • 48  Gilli F, Marnetto F, Caldano M et al.: Biological markers of interferon-β therapy: comparison among interferon-stimulated genes MxA, TRAIL and XAF-1. Mult. Scler.12(1),47–57 (2006)
    • 49  Hesse D, Sellebjerg F, Sorensen PS: Absence of MxA induction by interferon b in patients with MS reflects complete loss of bioactivity. Neurology73(5),372–377 (2009).
    • 50  Sellebjerg F, Krakauer M, Hesse D et al.: Identification of new sensitive biomarkers for the in vivo response to interferon-β treatment in multiple sclerosis using DNA-array evaluation. Eur. J. Neurol. (2009) (Epub ahead of print).
    • 51  Comabella M, Lünemann JD, Río J et al.: A type I interferon signature in monocytes is associated with poor response to interferon-β in multiple sclerosis. Brain2(Pt 12),3353–3365 (2009).
    • 52  O’Doherty C, Villoslada P, Vandenbroeck K: Pharmacogenomics of Type I interferon therapy: a survey of response-modifying genes. Cytokine Growth Factor Rev.18(3-4),211–222 (2007).
    • 53  Vandenbroeck K, Matute C: Pharmacogenomics of the response to IFN-β in multiple sclerosis: ramifications from the first genome-wide screen. Pharmacogenomics9(5),639–645 (2008).
    • 54  Hwang D, Rust AG, Ramsey S et al.: A data integration methodology for systems biology. Proc. Natl Acad. Sci. USA102(48),17296–17301 (2005).
    • 55  Bauch A, Superti-Furga G: Charting protein complexes, signaling pathways, and networks in the immune system. Immunol. Rev.210,187–207 (2006).
    • 56  Lamb J, Crawford ED, Peck D et al.: The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science313(5795),1929–1935 (2006).
    • 101  Pathway Architect 2.0.1 (Iobion) www.iobion.com
    • 102  National Center for Biotechnology Information Gene Expression Omnibus www.ncbi.nlm.nih.gov/geo