RORα suppresses breast tumor invasion by inducing SEMA3F expression.

Inactivation of tumor suppressors and inhibitory microenvironmental factors is necessary for breast cancer invasion; therefore, identifying those suppressors and factors is crucial not only to advancing our knowledge of breast cancer, but also to discovering potential therapeutic targets. By analyzing gene expression profiles of polarized and disorganized human mammary epithelial cells in a physiologically relevant three-dimensional (3D) culture system, we identified retinoid orphan nuclear receptor alpha (RORα) as a transcription regulator of semaphorin 3F (SEMA3F), a suppressive microenvironmental factor. We showed that expression of RORα was downregulated in human breast cancer tissue and cell lines, and that reduced mRNA levels of RORα and SEMA3F correlated with poor prognosis. Restoring RORα expression reprogrammed breast cancer cells to form noninvasiveness structures in 3D culture and inhibited tumor growth in nude mice, accompanied by enhanced SEMA3F expression. Inactivation of RORα in nonmalignant human mammary epithelial cells inhibited SEMA3F transcription and impaired polarized acinar morphogenesis. Using chromatin immunoprecipitation and luciferase reporter assays, we showed that transcription of SEMA3F is directly regulated by RORα. Knockdown of SEMA3F in RORα-expressing cancer cells rescued the aggressive 3D phenotypes and tumor invasion. These findings indicate that RORα is a potential tumor suppressor and inhibits tumor invasion by inducing suppressive cell microenvironment.

[1]  A. Mercurio,et al.  Neuropilin-2 promotes branching morphogenesis in the mouse mammary gland , 2011, Development.

[2]  M. Bissell,et al.  IL-25 Causes Apoptosis of IL-25R–Expressing Breast Cancer Cells Without Toxicity to Nonmalignant Cells , 2011, Science Translational Medicine.

[3]  R. Xu,et al.  Gene transcriptional networks integrate microenvironmental signals in human breast cancer. , 2011, Integrative biology : quantitative biosciences from nano to macro.

[4]  William C Hines,et al.  Why don't we get more cancer? A proposed role of the microenvironment in restraining cancer progression , 2011, Nature Medicine.

[5]  Dihua Yu,et al.  Microenvironment Determinants of Brain Metastasis , 2011, Cell & Bioscience.

[6]  Tim Hui-Ming Huang,et al.  Inference of hierarchical regulatory network of estrogen-dependent breast cancer through ChIP-based data , 2010, BMC Systems Biology.

[7]  M. Bissell,et al.  Raf-induced MMP9 disrupts tissue architecture of human breast cells in three-dimensional culture and is necessary for tumor growth in vivo. , 2010, Genes & development.

[8]  A. Balmain,et al.  Deletion of the PER3 gene on chromosome 1p36 in recurrent ER-positive breast cancer. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[9]  Dhara N. Amin,et al.  Id2 promotes tumor cell migration and invasion through transcriptional repression of semaphorin 3F. , 2010, Cancer research.

[10]  M. Rosenfeld,et al.  RORalpha attenuates Wnt/beta-catenin signaling by PKCalpha-dependent phosphorylation in colon cancer. , 2010, Molecular cell.

[11]  D. Sgroi Preinvasive breast cancer. , 2010, Annual review of pathology.

[12]  T. Sanke,et al.  Neuropilin-2 expression in breast cancer: correlation with lymph node metastasis, poor prognosis, and regulation of CXCR4 expression , 2009, BMC Cancer.

[13]  W. Miyazaki,et al.  Activation of Aromatase Expression by Retinoic Acid Receptor-related Orphan Receptor (ROR) α in Breast Cancer Cells , 2009, The Journal of Biological Chemistry.

[14]  R. Gemmill,et al.  ZEB-1, a repressor of the semaphorin 3F tumor suppressor gene in lung cancer cells. , 2009, Neoplasia.

[15]  Mina J. Bissell,et al.  Sustained activation of STAT5 is essential for chromatin remodeling and maintenance of mammary-specific function , 2009, The Journal of cell biology.

[16]  A. Jetten Retinoid-related orphan receptors (RORs): critical roles in development, immunity, circadian rhythm, and cellular metabolism , 2009, Nuclear receptor signaling.

[17]  D. Ingber,et al.  ABL2/ARG Tyrosine Kinase Mediates SEMA3F-induced RhoA Inactivation and Cytoskeleton Collapse in Human Glioma Cells* , 2008, Journal of Biological Chemistry.

[18]  G. Neufeld,et al.  Successful Inhibition of Tumor Development by Specific Class-3 Semaphorins Is Associated with Expression of Appropriate Semaphorin Receptors by Tumor Cells , 2008, PloS one.

[19]  Il-Jin Kim,et al.  FBXW7 Targets mTOR for Degradation and Cooperates with PTEN in Tumor Suppression , 2008, Science.

[20]  Roy A Jensen,et al.  A human breast cell model of preinvasive to invasive transition. , 2008, Cancer research.

[21]  G. Vodjdani,et al.  Phosphorylation and transcriptional activity regulation of retinoid‐related orphan receptor alpha 1 by protein kinases C , 2008, Journal of neurochemistry.

[22]  Pengyuan Liu,et al.  Common Human Cancer Genes Discovered by Integrated Gene-Expression Analysis , 2007, PloS one.

[23]  S. Reed,et al.  FBXW7/hCDC4 is a general tumor suppressor in human cancer. , 2007, Cancer research.

[24]  H. Augustin,et al.  Semaphorin SEMA3F affects multiple signaling pathways in lung cancer cells. , 2007, Cancer research.

[25]  O. Werz,et al.  Extracellular signal-regulated kinase-2 phosphorylates RORα4 in vitro , 2007 .

[26]  Genee Y. Lee,et al.  Three-dimensional culture models of normal and malignant breast epithelial cells , 2007, Nature Methods.

[27]  H. Arakawa,et al.  Possible role of semaphorin 3F, a candidate tumor suppressor gene at 3p21.3, in p53-regulated tumor angiogenesis suppression. , 2007, Cancer research.

[28]  Ajay N. Jain,et al.  Genomic and transcriptional aberrations linked to breast cancer pathophysiologies. , 2006, Cancer cell.

[29]  Mina J Bissell,et al.  Extracellular Matrix-regulated Gene Expression Requires Cooperation of SWI/SNF and Transcription Factors* , 2006, Journal of Biological Chemistry.

[30]  R. Kuhn,et al.  RORA, a large common fragile site gene, is involved in cellular stress response , 2006, Oncogene.

[31]  Hong Liu,et al.  Mechanism of Akt1 inhibition of breast cancer cell invasion reveals a protumorigenic role for TSC2. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[32]  M. Bronner‐Fraser,et al.  Guidance of trunk neural crest migration requires neuropilin 2/semaphorin 3F signaling , 2006, Development.

[33]  A. Moon,et al.  H-Ras-specific Activation of Rac-MKK3/6-p38 Pathway , 2005, Journal of Biological Chemistry.

[34]  J. Foekens,et al.  Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer , 2005, The Lancet.

[35]  M. Kreuter,et al.  Semaphorin 3F, a chemorepulsant for endothelial cells, induces a poorly vascularized, encapsulated, nonmetastatic tumor phenotype. , 2004, The Journal of clinical investigation.

[36]  V. Giguère,et al.  Lymphocyte Development and Function in the Absence of Retinoic Acid-Related Orphan Receptor α1 , 2004, The Journal of Immunology.

[37]  G. Muscat,et al.  RORalpha regulates the expression of genes involved in lipid homeostasis in skeletal muscle cells: caveolin-3 and CPT-1 are direct targets of ROR. , 2004, The Journal of biological chemistry.

[38]  L. Miraglia,et al.  A Functional Genomics Strategy Reveals Rora as a Component of the Mammalian Circadian Clock , 2004, Neuron.

[39]  D. Radisky,et al.  Polarity and proliferation are controlled by distinct signaling pathways downstream of PI3-kinase in breast epithelial tumor cells , 2004, The Journal of cell biology.

[40]  V. Giguère,et al.  The Co-repressor Hairless Protects RORα Orphan Nuclear Receptor from Proteasome-mediated Degradation* , 2003, Journal of Biological Chemistry.

[41]  L. Ellis,et al.  Angiopoietin-1 inhibits vascular permeability, angiogenesis, and growth of hepatic colon cancer tumors. , 2003, Cancer research.

[42]  P. Leder,et al.  MEK1 signaling mediates transformation and metastasis of EpH4 mammary epithelial cells independent of an epithelial to mesenchymal transition. , 2002, Cancer research.

[43]  Mina J. Bissell,et al.  Putting tumours in context , 2001, Nature Reviews Cancer.

[44]  J. Mariani,et al.  The orphan nuclear receptor RORα is a negative regulator of the inflammatory response , 2001 .

[45]  V. Weaver,et al.  Tissue structure, nuclear organization, and gene expression in normal and malignant breast. , 1999, Cancer research.

[46]  C. Larabell,et al.  Reciprocal interactions between beta1-integrin and epidermal growth factor receptor in three-dimensional basement membrane breast cultures: a different perspective in epithelial biology. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[47]  F. Crépel,et al.  staggerer phenotype in retinoid-related orphan receptor α-deficient mice , 1998 .

[48]  V. Giguère,et al.  Orphan nuclear receptor RORα-deficient mice display the cerebellar defects of staggerer , 1998, Mechanisms of Development.

[49]  M. Nehls,et al.  The Structural Integrity of RORα Isoforms Is Mutated instaggererMice: Cerebellar Coexpression of RORα1 and RORα4 , 1997 .

[50]  C. Larabell,et al.  Reversion of the Malignant Phenotype of Human Breast Cells in Three-Dimensional Culture and In Vivo by Integrin Blocking Antibodies , 1997, The Journal of cell biology.

[51]  M. Downes,et al.  Transcriptional repression by the orphan steroid receptor RVR/Rev-erb beta is dependent on the signature motif and helix 5 in the E region: functional evidence for a biological role of RVR in myogenesis. , 1996, Nucleic acids research.

[52]  S. Naylor,et al.  Isolation of the human semaphorin III/F gene (SEMA3F) at chromosome 3p21, a region deleted in lung cancer. , 1996, Genomics.

[53]  M. Becker‐André,et al.  RZRs, a new family of retinoid-related orphan receptors that function as both monomers and homodimers. , 1994, Molecular endocrinology.

[54]  M. Bissell,et al.  Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells. , 1992, Proceedings of the National Academy of Sciences of the United States of America.

[55]  M J Bissell,et al.  Functional differentiation and alveolar morphogenesis of primary mammary cultures on reconstituted basement membrane. , 1989, Development.

[56]  山内 健太 FGF8 signaling regulates growth of midbrain dopaminergic axons by inducing semaphorin 3F , 2008 .

[57]  O. Werz,et al.  Extracellular signal-regulated kinase-2 phosphorylates RORalpha4 in vitro. , 2007, Biochemical and biophysical research communications.

[58]  S. Kurebayashi,et al.  The ROR nuclear orphan receptor subfamily: critical regulators of multiple biological processes. , 2001, Progress in nucleic acid research and molecular biology.

[59]  J. Mariani,et al.  The orphan nuclear receptor ROR alpha is a negative regulator of the inflammatory response. , 2001, EMBO reports.

[60]  F. Crépel,et al.  staggerer phenotype in retinoid-related orphan receptor alpha-deficient mice. , 1998, Proceedings of the National Academy of Sciences of the United States of America.