Activation of PKA leads to mesenchymal-to-epithelial transition and loss of tumor-initiating ability

Have cancer stem cells MET their match? Solid tumors have been hypothesized to contain a subset of highly aggressive cells that fuel tumor growth and metastasis. The search is on for drugs that selectively kill or diminish the malignant properties of these tumor-initiating cells (TICs; previously called “cancer stem cells”). Pattabiraman et al. hypothesized that compounds that induce TICs to undergo a phenotypic change called the mesenchymal-to-epithelial transition (MET) would therefore cause TICs to lose their tumor-initiating ability. Indeed, drugs activating the protein kinase A signaling pathway triggered an epigenetic reprogramming of TICs that resulted in the cells acquiring a more benign epithelial-like phenotype. Science, this issue p. 10.1126/science.aad3680 Tumor-initiating cells differentiate to a more benign state when treated with drugs that activate protein kinase A. INTRODUCTION Tumor-initiating cells (TICs) have emerged in recent years as important targets for cancer therapy owing to their elevated resistance to conventional chemotherapy and their tumor-initiating ability. Although their mode of generation and biological properties have been explored in a diverse array of cancer types, our understanding of the biology of TICs remains superficial. The epithelial-to-mesenchymal transition (EMT) is a cell-biological program that confers mesenchymal traits on both normal and neoplastic epithelial cells, which enables both to acquire stemlike properties. In the case of carcinoma cells, entrance into a more mesenchymal state is associated with elevated resistance to a variety of conventional chemotherapeutics. This association between the EMT program and the TIC state has presented an attractive opportunity for drug development using agents that preferentially target more mesenchymal carcinoma cells, rather than their epithelial counterparts, in an effort to eliminate TICs. Adenosine 3′,5′-monophosphate (cAMP) is a second messenger that transmits intracellular signals through multiple downstream effectors; the most well studied of these is protein kinase A (PKA). In this study, we explore the role of PKA in determining the epithelial versus mesenchymal properties of mammary epithelial cells and how this signaling pathway affects the tumor-initiating ability of transformed cells. RATIONALE At least two approaches might be taken to target mesenchymal TICs. One strategy that has been used previously is the development of agents that show specific or preferential cytotoxicity toward TICs. In the current study, we have embraced an alternative strategy that is designed to induce TICs to undergo a mesenchymal-to-epithelial transition (MET). This “induced differentiation” approach would trigger cells to exit the more mesenchymal tumor-initiating state and enter into an epithelial non-stemlike state. In principle, this transition would make the cells more vulnerable to conventional cytotoxic treatments and thereby reduce the likelihood of metastasis and clinical relapse. RESULTS To identify agents that might induce an MET in mesenchymal mammary epithelial cells, we performed a screen for compounds that stimulate transcription of CDH1, which encodes E-cadherin, a key epithelial protein. Through this screen, compounds that activate adenylate cyclase (cholera toxin, CTx; and forskolin, Fsk) were identified as key inducers of the epithelial state. We found that mesenchymal cells treated with either CTx or Fsk differentiated into benign epithelial derivatives that had lost their ability to effectively initiate tumors and that were more susceptible to conventional chemotherapeutic agents in vitro. Further interrogation revealed that these agents elevated the intracellular levels of cAMP, which in turn activates PKA. PHF2, a histone H3 with acetylated lysine 9 (H3K9) histone demethylase and PKA substrate, was found to be essential for the cAMP-induced MET. By studying the genome occupancy of PHF2 and the epigenomic state of the cells before and after PKA activation, we determined that PHF2 promotes the demethylation and derepression of epithelial genes that ultimately contribute to acquisition of an epithelial state. CONCLUSION We conclude that PKA participates in the differentiation of TICs by enforcing residence in the epithelial state and preventing or reversing the EMT program. Our study reveals a new direction for targeting the TIC population. We propose that pharmacological induction of epigenetic reprogramming of these cells could promote their differentiation to a more epithelial state and increase their susceptibility to conventional chemotherapeutic drugs. Induction of the MET as a potential cancer therapy. TICs have mesenchymal attributes that contribute to their ability to seed new tumors. Treatment of TICs with compounds that increase cAMP levels (e.g., CTx and Fsk) activates PKA. This leads to epigenetic reprogramming through subsequent activation of the histone demethylase PHF2, a PKA substrate, which in turn promotes differentiation of the cells into a more epithelial state, accompanied by a loss of their tumor-initiating ability. Drugs targeting various steps of this signaling pathway might be developed into a differentiation-based cancer therapy for certain breast cancers. Cite this article as D. R. Pattabiraman et al., Science 351, aad3680 (2016). DOI: 10.1126/science.aad3680 The epithelial-to-mesenchymal transition enables carcinoma cells to acquire malignancy-associated traits and the properties of tumor-initiating cells (TICs). TICs have emerged in recent years as important targets for cancer therapy, owing to their ability to drive clinical relapse and enable metastasis. Here, we propose a strategy to eliminate mesenchymal TICs by inducing their conversion to more epithelial counterparts that have lost tumor-initiating ability. We report that increases in intracellular levels of the second messenger, adenosine 3′,5′-monophosphate, and the subsequent activation of protein kinase A (PKA) induce a mesenchymal-to-epithelial transition (MET) in mesenchymal human mammary epithelial cells. PKA activation triggers epigenetic reprogramming of TICs by the histone demethylase PHF2, which promotes their differentiation and loss of tumor-initiating ability. This study provides proof-of-principle for inducing an MET as differentiation therapy for TICs and uncovers a role for PKA in enforcing and maintaining the epithelial state.

[1]  A. Wittinghofer,et al.  Epac is a Rap1 guanine-nucleotide-exchange factor directly activated by cyclic AMP , 1998, Nature.

[2]  Ping Zhang,et al.  Assembly of allosteric macromolecular switches: lessons from PKA , 2012, Nature Reviews Molecular Cell Biology.

[3]  E. Lander,et al.  Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. , 2008, Cancer research.

[4]  J. Shabb Physiological substrates of cAMP-dependent protein kinase. , 2001, Chemical reviews.

[5]  R. Weinberg,et al.  Tackling the cancer stem cells — what challenges do they pose? , 2014, Nature Reviews Drug Discovery.

[6]  Susan G Hilsenbeck,et al.  Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. , 2008, Journal of the National Cancer Institute.

[7]  F. Bertucci,et al.  Gene expression profiling of breast cell lines identifies potential new basal markers , 2006, Oncogene.

[8]  Jun Song,et al.  CEAS: cis-regulatory element annotation system , 2006, Nucleic Acids Res..

[9]  U. Moens,et al.  What turns CREB on? , 2004, Cellular signalling.

[10]  M. Montminy,et al.  Cyclic AMP stimulates somatostatin gene transcription by phosphorylation of CREB at serine 133 , 1989, Cell.

[11]  J. Miller,et al.  Analogs of cyclic AMP and cyclic GMP: general methods of synthesis and the relationship of structure to enzymic activity. , 1974, Life sciences.

[12]  M. Hummel,et al.  Evidence for Epithelial-Mesenchymal Transition in Cancer Stem Cells of Head and Neck Squamous Cell Carcinoma , 2011, PloS one.

[13]  David C. Lee,et al.  Increased Basal cAMP-dependent Protein Kinase Activity Inhibits the Formation of Mesoderm-derived Structures in the Developing Mouse Embryo* , 2002, The Journal of Biological Chemistry.

[14]  Tzong-Shiue Yu,et al.  A restricted cell population propagates glioblastoma growth following chemotherapy , 2012, Nature.

[15]  G. Dontu,et al.  In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. , 2003, Genes & development.

[16]  Yonghong Xiao,et al.  Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. , 2011, Cancer cell.

[17]  E. Gelmann,et al.  Growth properties and tumorigenesis of MCF-7 cells transfected with isogenic mutants of rasH. , 1990, Cancer research.

[18]  Clifford A. Meyer,et al.  Model-based Analysis of ChIP-Seq (MACS) , 2008, Genome Biology.

[19]  F. Marshall,et al.  cAMP-responsive element-binding protein regulates vascular endothelial growth factor expression: implication in human prostate cancer bone metastasis , 2007, Oncogene.

[20]  H. Green,et al.  Cyclic AMP in relation to proliferation of the Epidermal cell: a new view , 1978, Cell.

[21]  S. Firestein,et al.  Cyclic Nucleotide‐Gated Channels: Molecular Mechanisms of Activation , 1999, Annals of the New York Academy of Sciences.

[22]  Cole Trapnell,et al.  TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions , 2013, Genome Biology.

[23]  Elgene Lim,et al.  Protein kinase C α is a central signaling node and therapeutic target for breast cancer stem cells. , 2013, Cancer cell.

[24]  W. Hahn,et al.  Human breast cancer cells generated by oncogenic transformation of primary mammary epithelial cells. , 2001, Genes & development.

[25]  Helga Thorvaldsdóttir,et al.  Integrative Genomics Viewer , 2011, Nature Biotechnology.

[26]  Susan S. Taylor,et al.  Inactivation of a Gαs-PKA tumor suppressor pathway in skin stem cells initiates basal-cell carcinogenesis , 2015, Nature Cell Biology.

[27]  Steven L Salzberg,et al.  Fast gapped-read alignment with Bowtie 2 , 2012, Nature Methods.

[28]  M. Quinn,et al.  Epithelial mesenchymal transition and cancer stem cell-like phenotypes facilitate chemoresistance in recurrent ovarian cancer. , 2010, Current cancer drug targets.

[29]  G. McKnight,et al.  Mutations in the catalytic subunit of cAMP-dependent protein kinase result in unregulated biological activity. , 1992, Proceedings of the National Academy of Sciences of the United States of America.

[30]  S. Morrison,et al.  Prospective identification of tumorigenic breast cancer cells , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[31]  Hans Clevers,et al.  Distinct gene mutation profiles among luminal-type and basal-type breast cancer cell lines , 2010, Breast Cancer Research and Treatment.

[32]  A. Peters,et al.  Mechanisms of transcriptional repression by histone lysine methylation. , 2009, The International journal of developmental biology.

[33]  J. Beavo,et al.  Cyclic nucleotide research — still expanding after half a century , 2002, Nature Reviews Molecular Cell Biology.

[34]  A. Puisieux,et al.  Generation of Breast Cancer Stem Cells through Epithelial-Mesenchymal Transition , 2008, PloS one.

[35]  Fabian J Theis,et al.  Quantification of regenerative potential in primary human mammary epithelial cells , 2015, Development.

[36]  A. Krešo,et al.  Evolution of the cancer stem cell model. , 2014, Cell stem cell.

[37]  Wenjun Guo,et al.  Slug and Sox9 Cooperatively Determine the Mammary Stem Cell State , 2012, Cell.

[38]  G. Berx,et al.  Regulatory networks defining EMT during cancer initiation and progression , 2013, Nature Reviews Cancer.

[39]  C. Wernstedt,et al.  Phosphorylation of Serine 11 and Serine 92 as New Positive Regulators of Human Snail1 Function: Potential Involvement of Casein Kinase-2 and the cAMP-activated Kinase Protein Kinase A , 2010, Molecular biology of the cell.

[40]  C. Stratakis,et al.  Targeted deletion of Prkar1a reveals a role for protein kinase A in mesenchymal-to-epithelial transition. , 2008, Cancer research.

[41]  Jingwu Xie,et al.  Regulation of Gli1 Localization by the cAMP/Protein Kinase A Signaling Axis through a Site Near the Nuclear Localization Signal* , 2006, Journal of Biological Chemistry.

[42]  T. Seufferlein,et al.  Transforming growth factor beta1 treatment leads to an epithelial-mesenchymal transdifferentiation of pancreatic cancer cells requiring extracellular signal-regulated kinase 2 activation. , 2001, Cancer research.

[43]  Aaron R. Quinlan,et al.  BIOINFORMATICS APPLICATIONS NOTE , 2022 .

[44]  R. Cardiff,et al.  Induction of mammary tumors by expression of polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease , 1992, Molecular and cellular biology.

[45]  Wenjun Guo,et al.  The Epithelial-Mesenchymal Transition Generates Cells with Properties of Stem Cells , 2008, Cell.

[46]  Guofei Zhou,et al.  cAMP-dependent protein kinase is essential for hypoxia-mediated epithelial-mesenchymal transition, migration, and invasion in lung cancer cells. , 2012, Cellular signalling.

[47]  S. Døskeland,et al.  A novel Epac-specific cAMP analogue demonstrates independent regulation of Rap1 and ERK , 2002, Nature Cell Biology.

[48]  Jun Yao,et al.  G9a interacts with Snail and is critical for Snail-mediated E-cadherin repression in human breast cancer. , 2012, The Journal of clinical investigation.

[49]  Eric Nestler,et al.  ngs.plot: Quick mining and visualization of next-generation sequencing data by integrating genomic databases , 2014, BMC Genomics.

[50]  B. Shankar,et al.  TGF-β1-ROS-ATM-CREB signaling axis in macrophage mediated migration of human breast cancer MCF7 cells. , 2014, Cellular signalling.

[51]  Dimitris Anastassiou,et al.  Human cancer cells express Slug-based epithelial-mesenchymal transition gene expression signature obtained in vivo , 2011, BMC Cancer.

[52]  Tao Liu,et al.  CEAS: cis-regulatory element annotation system , 2009, Bioinform..

[53]  Tzong-Shiue Yu,et al.  A restricted cell population propagates glioblastoma growth after chemotherapy , 2012 .

[54]  Eric S. Lander,et al.  Identification of Selective Inhibitors of Cancer Stem Cells by High-Throughput Screening , 2009, Cell.

[55]  Clifford A. Meyer,et al.  PKA-dependent regulation of the histone lysine demethylase complex PHF2–ARID5B , 2011, Nature Cell Biology.

[56]  E. Aandahl,et al.  Localized effects of cAMP mediated by distinct routes of protein kinase A. , 2004, Physiological reviews.

[57]  Paul Theodor Pyl,et al.  HTSeq—a Python framework to work with high-throughput sequencing data , 2014, bioRxiv.

[58]  I. Fentiman,et al.  Cholera toxin and analogues of cyclic AMP stimulate the growth of cultured human mammary epithelial cells , 1980, Journal of cellular physiology.

[59]  W. Huber,et al.  which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. MAnorm: a robust model for quantitative comparison of ChIP-Seq data sets , 2011 .

[60]  M. Dowsett,et al.  Phase I study of the novel cyclic AMP (cAMP) analogue 8-chloro-cAMP in patients with cancer: toxicity, hormonal, and immunological effects. , 1999, Clinical cancer research : an official journal of the American Association for Cancer Research.