Module Network Inference from a Cancer Gene Expression Data Set Identifies MicroRNA Regulated Modules

Background MicroRNAs (miRNAs) are small RNAs that recognize and regulate mRNA target genes. Multiple lines of evidence indicate that they are key regulators of numerous critical functions in development and disease, including cancer. However, defining the place and function of miRNAs in complex regulatory networks is not straightforward. Systems approaches, like the inference of a module network from expression data, can help to achieve this goal. Methodology/Principal Findings During the last decade, much progress has been made in the development of robust and powerful module network inference algorithms. In this study, we analyze and assess experimentally a module network inferred from both miRNA and mRNA expression data, using our recently developed module network inference algorithm based on probabilistic optimization techniques. We show that several miRNAs are predicted as statistically significant regulators for various modules of tightly co-expressed genes. A detailed analysis of three of those modules demonstrates that the specific assignment of miRNAs is functionally coherent and supported by literature. We further designed a set of experiments to test the assignment of miR-200a as the top regulator of a small module of nine genes. The results strongly suggest that miR-200a is regulating the module genes via the transcription factor ZEB1. Interestingly, this module is most likely involved in epithelial homeostasis and its dysregulation might contribute to the malignant process in cancer cells. Conclusions/Significance Our results show that a robust module network analysis of expression data can provide novel insights of miRNA function in important cellular processes. Such a computational approach, starting from expression data alone, can be helpful in the process of identifying the function of miRNAs by suggesting modules of co-expressed genes in which they play a regulatory role. As shown in this study, those modules can then be tested experimentally to further investigate and refine the function of the miRNA in the regulatory network.

[1]  A. Nordheim,et al.  Repression of c‐fos transcription is mediated through p67SRF bound to the SRE. , 1989, The EMBO journal.

[2]  M. Sheng,et al.  The inner core of the serum response element mediates both the rapid induction and subsequent repression of c-fos transcription following serum stimulation. , 1990, Genes & development.

[3]  M. Welsh,et al.  Cloning, expression, and tissue distribution of a human amiloride-sensitive Na+ channel. , 1994, The American journal of physiology.

[4]  R. Koski,et al.  The Transforming Receptor Tyrosine Kinase, Axl, Is Post-translationally Regulated by Proteolytic Cleavage (*) , 1995, The Journal of Biological Chemistry.

[5]  H. Weintraub,et al.  B-lymphocyte development is regulated by the combined dosage of three basic helix-loop-helix genes, E2A, E2-2, and HEB , 1996, Molecular and cellular biology.

[6]  T. Schall,et al.  CC chemokines induce the generation of killer cells from CD56+ cells , 1996, European journal of immunology.

[7]  Jaime Prilusky,et al.  GeneCards: a novel functional genomics compendium with automated data mining and query reformulation support , 1998, Bioinform..

[8]  J J Gómez-Reino Carnota,et al.  [Genetics of rheumatoid arthritis]. , 2000, Medicina clinica.

[9]  A. Sahin,et al.  In vivo expression of the novel CXC chemokine BRAK in normal and cancerous human tissue. , 2000, The American journal of pathology.

[10]  I. Kurth,et al.  Monocyte Selectivity and Tissue Localization Suggests a Role for Breast and Kidney–Expressed Chemokine (Brak) in Macrophage Development , 2001, The Journal of experimental medicine.

[11]  J. Stull,et al.  Dedicated Myosin Light Chain Kinases with Diverse Cellular Functions* , 2001, The Journal of Biological Chemistry.

[12]  E. Filardo,et al.  Epidermal growth factor receptor (EGFR) transactivation by estrogen via the G-protein-coupled receptor, GPR30: a novel signaling pathway with potential significance for breast cancer , 2002, The Journal of Steroid Biochemistry and Molecular Biology.

[13]  Markus J. Herrgård,et al.  Reconciling gene expression data with known genome-scale regulatory network structures. , 2003, Genome research.

[14]  C. Moskaluk,et al.  Overexpression of the 32‐kilodalton dopamine and cyclic adenosine 3′,5′‐monophosphate‐regulated phosphoprotein in common adenocarcinomas , 2003, Cancer.

[15]  D. Pe’er,et al.  Module networks: identifying regulatory modules and their condition-specific regulators from gene expression data , 2003, Nature Genetics.

[16]  Cloning and characterisation of a 1.1 kb fragment of the carcinoma-associated epithelial cell adhesion molecule promoter. , 2003, Anticancer research.

[17]  Da-Zhi Wang,et al.  Myocardin is a master regulator of smooth muscle gene expression , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[18]  R. Reeves,et al.  HMGA1 Co-activates Transcription in B Cells through Indirect Association with DNA* , 2003, Journal of Biological Chemistry.

[19]  D. Bartel MicroRNAs Genomics, Biogenesis, Mechanism, and Function , 2004, Cell.

[20]  C. Perou,et al.  A custom microarray platform for analysis of microRNA gene expression , 2004, Nature Methods.

[21]  M. Grzmil,et al.  Up-regulated expression of the MAT-8 gene in prostate cancer and its siRNA-mediated inhibition of expression induces a decrease in proliferation of human prostate carcinoma cells. , 2004, International journal of oncology.

[22]  G. Mills,et al.  The RAB25 small GTPase determines aggressiveness of ovarian and breast cancers , 2004, Nature Medicine.

[23]  D. Bartel,et al.  MicroRNAs Modulate Hematopoietic Lineage Differentiation , 2004, Science.

[24]  Adam A. Margolin,et al.  Reverse engineering of regulatory networks in human B cells , 2005, Nature Genetics.

[25]  Giovanni De Micheli,et al.  Prediction of regulatory modules comprising microRNAs and target genes , 2005, ECCB/JBI.

[26]  Eugene Berezikov,et al.  Phylogenetic Shadowing and Computational Identification of Human microRNA Genes , 2005, Cell.

[27]  J. Castle,et al.  Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs , 2005, Nature.

[28]  Martin Kuiper,et al.  BiNGO: a Cytoscape plugin to assess overrepresentation of Gene Ontology categories in Biological Networks , 2005, Bioinform..

[29]  D. Bartel,et al.  Microarray profiling of microRNAs reveals frequent coexpression with neighboring miRNAs and host genes. , 2005, RNA.

[30]  H. Horvitz,et al.  MicroRNA expression profiles classify human cancers , 2005, Nature.

[31]  K. Geering,et al.  FXYD3 (Mat-8), a new regulator of Na,K-ATPase. , 2005, Molecular biology of the cell.

[32]  Jian-Fu Chen,et al.  The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation , 2006, Nature Genetics.

[33]  Albertha J. M. Walhout,et al.  Unraveling transcription regulatory networks by protein-DNA and protein-protein interaction mapping. , 2006, Genome research.

[34]  J. Collins,et al.  Large-Scale Mapping and Validation of Escherichia coli Transcriptional Regulation from a Compendium of Expression Profiles , 2007, PLoS biology.

[35]  C. Croce,et al.  MicroRNA-133 controls cardiac hypertrophy , 2007, Nature Medicine.

[36]  Byoung-Tak Zhang,et al.  BIOINFORMATICS ORIGINAL PAPER doi:10.1093/bioinformatics/btm045 Data and text mining Discovery of microRNA–mRNA modules via population-based probabilistic learning , 2007 .

[37]  K. Fujiwara,et al.  Serum response factor: master regulator of the actin cytoskeleton and contractile apparatus. , 2007, American journal of physiology. Cell physiology.

[38]  L. Lim,et al.  MicroRNA targeting specificity in mammals: determinants beyond seed pairing. , 2007, Molecular cell.

[39]  Chen-Yong Lin,et al.  Prostasin induces protease-dependent and independent molecular changes in the human prostate carcinoma cell line PC-3. , 2007, Biochimica et biophysica acta.

[40]  T. Brabletz,et al.  A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells , 2008, EMBO reports.

[41]  G. Berx,et al.  The role of the ZEB family of transcription factors in development and disease , 2009, Cellular and Molecular Life Sciences.

[42]  Stijn van Dongen,et al.  miRBase: tools for microRNA genomics , 2007, Nucleic Acids Res..

[43]  Yves Van de Peer,et al.  Analysis of a Gibbs sampler method for model-based clustering of gene expression data , 2008, Bioinform..

[44]  Pieter J. De Bleser,et al.  ConTra: a promoter alignment analysis tool for identification of transcription factor binding sites across species , 2008, Nucleic Acids Res..

[45]  Kathleen Marchal,et al.  Comparative analysis of module-based versus direct methods for reverse-engineering transcriptional regulatory networks , 2009, BMC Systems Biology.

[46]  Sun-Mi Park,et al.  The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. , 2008, Genes & development.

[47]  G. Goodall,et al.  The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1 , 2008, Nature Cell Biology.

[48]  Frank J. Slack,et al.  MicroRNAs and cancer: An overview , 2008, Cell cycle.

[49]  Shaoxiang Zhang,et al.  MicroRNAs play a role in the development of human hematopoietic stem cells , 2008, Journal of cellular biochemistry.

[50]  M. Vingron,et al.  Paracrine control of oligodendrocyte differentiation by SRF-directed neuronal gene expression , 2009, Nature Neuroscience.

[51]  C. Burge,et al.  Most mammalian mRNAs are conserved targets of microRNAs. , 2008, Genome research.

[52]  Jiuyong Li,et al.  Exploring complex miRNA-mRNA interactions with Bayesian networks by splitting-averaging strategy , 2009, BMC Bioinformatics.

[53]  Woonyoung Choi,et al.  miR-200 Expression Regulates Epithelial-to-Mesenchymal Transition in Bladder Cancer Cells and Reverses Resistance to Epidermal Growth Factor Receptor Therapy , 2009, Clinical Cancer Research.

[54]  Zhiwei Wang,et al.  miR‐200 Regulates PDGF‐D‐Mediated Epithelial–Mesenchymal Transition, Adhesion, and Invasion of Prostate Cancer Cells , 2009, Stem cells.

[55]  P. Tam Faculty Opinions recommendation of miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. , 2009 .

[56]  Kathleen Marchal,et al.  Module networks revisited: computational assessment and prioritization of model predictions , 2009, Bioinform..