Dysregulation of YAP by ARF Stimulated with Tea-derived Carbon Nanodots

YAP is a downstream nuclear transcription factor of Hippo pathway which plays an essential role in development, cell growth, organ size and homeostasis. It was previously identified that elevation of YAP in genomics of genetic engineered mouse (GEM) model of prostate cancer is associated with Pten/Trp53 inactivation and ARF elevation hypothesizing the essential crosstalk of AKT/mTOR/YAP with ARF in prostate cancer. However, the detailed function and trafficking of YAP in cancer cells remains unclear. Using GEM microarray model, we found ARF dysregulates Hippo and Wnt pathways. In particular, ARF knockdown reduced non-nuclear localization of YAP which led to an increase in F-actin. Mechanistically, ARF knockdown suppressed protein turnover of β-catenin/YAP, and therefore enhanced the activity of AKT and phosphorylation of YAP. Moreover, we found tea-derived carbon dots can interact with ARF in nucleus that may further lead to the non-nuclear localization of YAP. Thus, we reported a novel crosstalk of ARF/β-catenin dysregulated YAP in Hippo pathway and a new approach to stimulate ARF-mediated signaling to inhibit nuclear YAP using nanomaterials implicating an innovative avenue for treatment of cancer.

[1]  Li Li,et al.  Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. , 2007, Genes & development.

[2]  F. Camargo,et al.  YAP mediates crosstalk between the Hippo and PI(3)K–TOR pathways by suppressing PTEN via miR-29 , 2012, Nature Cell Biology.

[3]  R. Matusik,et al.  ARF represses androgen receptor transactivation in prostate cancer. , 2013, Molecular endocrinology.

[4]  Feng Li,et al.  Resonantly enhanced Raman scattering and high-order Raman spectra of single-walled carbon nanotubes , 1999 .

[5]  Yuguo Tang,et al.  Recent advances in carbon nanodots: synthesis, properties and biomedical applications. , 2015, Nanoscale.

[6]  Jindan Yu,et al.  Cell detachment activates the Hippo pathway via cytoskeleton reorganization to induce anoikis. , 2012, Genes & development.

[7]  M. Loda,et al.  Differential p53-Independent Outcomes of p19Arf Loss in Oncogenesis , 2009, Science Signaling.

[8]  Y. Shaul,et al.  The Hippo pathway kinase Lats2 prevents DNA damage-induced apoptosis through inhibition of the tyrosine kinase c-Abl , 2013, Cell Death and Differentiation.

[9]  Hiroyuki Ogata,et al.  KEGG: Kyoto Encyclopedia of Genes and Genomes , 1999, Nucleic Acids Res..

[10]  L. Dai,et al.  Highly luminescent carbon nanodots by microwave-assisted pyrolysis. , 2012, Chemical communications.

[11]  K. Harvey,et al.  The Hippo pathway effector YAP is a critical regulator of skeletal muscle fibre size , 2015, Nature Communications.

[12]  C. Sander,et al.  Identifying Actionable Targets through Integrative Analyses of GEM Model and Human Prostate Cancer Genomic Profiling , 2014, Molecular Cancer Therapeutics.

[13]  Juanxia Wu,et al.  Raman spectroscopy of graphene , 2014 .

[14]  Brad T. Sherman,et al.  Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources , 2008, Nature Protocols.

[15]  Jason A. Koutcher,et al.  Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis , 2005, Nature.

[16]  Choon-Sik Park,et al.  Inhibiting DX2-p14/ARF Interaction Exerts Antitumor Effects in Lung Cancer and Delays Tumor Progression. , 2016, Cancer research.

[17]  Zhi-hua Chen,et al.  Kyoto Encyclopedia of Genes and Genomes were used for functional enrichment analysis of differentially expressed genes (DEGs). A protein‐protein interaction network was constructed, and the hub genes were subjected to module analysis and identification using Search Tool for the Retrieval , 2019 .

[18]  W. Gu,et al.  p53-Dependent and p53-independent activation of autophagy by ARF. , 2008, Cancer research.

[19]  X. Qu,et al.  Programmed Bacteria Death Induced by Carbon Dots with Different Surface Charge. , 2016, Small.

[20]  Y. Qiu,et al.  The 44 kDa Pim-1 kinase directly interacts with tyrosine kinase Etk/BMX and protects human prostate cancer cells from apoptosis induced by chemotherapeutic drugs , 2006, Oncogene.

[21]  Minoru Kanehisa,et al.  KEGG: new perspectives on genomes, pathways, diseases and drugs , 2016, Nucleic Acids Res..

[22]  C. Sherr Divorcing ARF and p53: an unsettled case , 2006, Nature Reviews Cancer.

[23]  Y. Dong,et al.  The preparation of ethylenediamine-modified fluorescent carbon dots and their use in imaging of cells. , 2015, Luminescence : the journal of biological and chemical luminescence.

[24]  Sarah E Seton-Rogers,et al.  Oncogenes: All eyes on YAP1 , 2014, Nature Reviews. Cancer.

[25]  R. Kefford,et al.  p14ARF Interacts with the SUMO-Conjugating Enzyme Ubc9 and Promotes the Sumoylation of Its Binding Partners , 2005, Cell cycle.

[26]  E. Olson,et al.  Regulation of YAP by mTOR and autophagy reveals a therapeutic target of tuberous sclerosis complex , 2014, The Journal of experimental medicine.

[27]  Huan‐Tsung Chang,et al.  Carbon dots prepared from ginger exhibiting efficient inhibition of human hepatocellular carcinoma cells. , 2014, Journal of materials chemistry. B.

[28]  Minoru Kanehisa,et al.  KEGG as a reference resource for gene and protein annotation , 2015, Nucleic Acids Res..

[29]  Huan‐Tsung Chang,et al.  Extremely high inhibition activity of photoluminescent carbon nanodots toward cancer cells. , 2013, Journal of materials chemistry. B.

[30]  N. Sharpless,et al.  The Regulation of INK4/ARF in Cancer and Aging , 2006, Cell.

[31]  X. Qu,et al.  Recent advances in bioapplications of C-dots , 2015 .

[32]  A. Ferrari,et al.  Raman scattering efficiency of graphene , 2013 .

[33]  A. Valette,et al.  p14ARF interacts with the SUMO-conjugating enzyme Ubc9 and promotes the sumoylation of its binding partners. , 2005 .

[34]  X. Qu,et al.  Non-Enzymatic-Browning-Reaction: A Versatile Route for Production of Nitrogen-Doped Carbon Dots with Tunable Multicolor Luminescent Display , 2014, Scientific Reports.

[35]  Johannes T. Margraf,et al.  Carbon nanodots: toward a comprehensive understanding of their photoluminescence. , 2014, Journal of the American Chemical Society.

[36]  I. Garraway,et al.  YAP1 and AR interactions contribute to the switch from androgen-dependent to castration-resistant growth in prostate cancer , 2015, Nature Communications.

[37]  J. Downward,et al.  Akt phosphorylates the Yes-associated protein, YAP, to induce interaction with 14-3-3 and attenuation of p73-mediated apoptosis. , 2003, Molecular cell.

[38]  Effects of layer stacking on the combination Raman modes in graphene. , 2011, ACS nano.

[39]  Wei Gu,et al.  Hydrothermal synthesis of nitrogen-doped carbon dots with real-time live-cell imaging and blood–brain barrier penetration capabilities , 2016, International journal of nanomedicine.

[40]  Yan Li,et al.  Wnt-YAP interactions in the neural fate of human pluripotent stem cells and the implications for neural organoid formation , 2016, Organogenesis.

[41]  Kun-Liang Guan,et al.  The emerging roles of YAP and TAZ in cancer , 2015, Nature Reviews Cancer.

[42]  S. Pratap,et al.  MMP7 interacts with ARF in nucleus to potentiate tumor microenvironments for prostate cancer progression in vivo , 2016, Oncotarget.

[43]  Sheila N. Baker,et al.  Luminescent carbon nanodots: emergent nanolights. , 2010, Angewandte Chemie.

[44]  R. Matusik,et al.  Slug regulates E‐cadherin repression via p19Arf in prostate tumorigenesis , 2014, Molecular oncology.

[45]  C. Liang,et al.  In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro , 2007, Nature Protocols.