Multifaceted Regulation of PTEN Subcellular Distributions and Biological Functions
Abstract
:1. Introduction
2. Domains of PTEN
2.1. N-Terminal Domain
2.2. Catalytic Domain
2.3. C2 Domain
2.4. Tail Region
2.5. PDZ-BM
3. Subcellular Distribution of PTEN
3.1. Cytoplasmic PTEN
3.2. Nuclear PTEN
3.3. PTEN in Cell Organelles
3.4. Secreted PTEN
4. PTEN and Cancer Hallmarks
4.1. PTEN and Oncogenic Signaling
4.2. PTEN and Cell Cycle
4.3. PTEN and Cancer Genome Stability
4.4. PTEN and Cellular Energetics
4.5. PTEN and Metastasis
4.6. PTEN and Angiogenesis
5. Regulation of PTEN in Physiological and Pathological States
5.1. miRNAs and LncRNAs
5.2. Catalytic Activity
5.2.1. PIP2
5.2.2. Phosphorylation
5.2.3. Redox
5.2.4. Ubiquitination
5.3. Membrane Targeting
5.4. Stability
5.5. Nuclear Targeting
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
4EBP1 | 4E-binding protein 1 |
AMD1 | S-adenosylmethionine decarboxylase 1 |
ATM | Ataxia telangiectasia mutated |
ATP | Adenosine triphosphate |
BCR-ABL | Breakpoint cluster region-Abelson |
C | Carboxyl |
Ca2+ | Calcium |
CBR1 | Ca2+-binding region 1 |
Cdh1 | Cadherin-1 |
CDK1 | Cyclin D1 |
CHIP | C-terminus of Hsc70-interacting protein |
CKII | Casein kinase II |
COSMIC | Catalogue of Somatic Mutations In Cancer |
cPLA2 | Phospholipase A2 |
DSP | Dual-specificity phosphatase |
ER | Endoplasmic reticulum |
FOXO3a | Forkhead box protein O3a |
FOXO1 | Forkhead box protein O1 |
GLUT4 | Glucose transporter type 4 |
Grb2 | Growth factor receptor-bound protein 2 |
GSK-3β | Glycogen synthase kinase 3 beta |
HAUSP | Herpesvirus-associated ubiquitin-specific protease |
LKB-1 | Liver kinase B-1 |
LncRNAs | Long-noncoding RNAs |
LRR | Leucine rich repeat |
MAPK | Mitogen-activated protein kinase |
miRNAs | MicroRNAs |
ms | Millisecond |
N | Amino |
ncRNAs | Noncoding RNAs |
Ndfip1 | NEDD4 family-interacting protein 1 |
Ndfip1 | NEDD4 family-interacting protein 1 |
NEDD4-1 | Developmentally down-regulated protein 4-1 |
NPM-1 | Nucleophosmin-1 |
OTUD3 | Ovarian tumor domain-containing protein 3 |
PBM | PIP2-binding motif |
PDZ | Postsynaptic density protein-Drosophila disc large tumor suppressor-Zonula occludens-1 protein |
PDZ-BM | PDZ-binding motif |
PERK | Protein kinase R-like endoplasmic reticulum kinase |
PEST | Proline, glutamate, serine, threonine |
PI(5)P | Phosphatidylinositol 5-monophosphate |
PI3K | Phosphoinositide 3-kinase |
PIASX | Protein Inhibitor of Activated STAT2 |
PIP2/PI(4,5)P2 | Phosphatidylinositol 4,5-bisphosphate |
PIP3 | 3′ phosphate of phosphatidylinositol 3,4,5-triphosphate |
PKCδ | Protein kinase Cδ |
PLCδ1 | Phospholipase C δ1 |
PNUTS | Protein phosphatase nuclear targeting subunit |
POPC | 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine |
POPCS | 1-palmitoyl-2-oleoyl-glycero-3-phosphoserine |
PPARγ | Proliferator-activated receptor-γ |
PRKN | Parkin RBR E3 ubiquitin protein ligase |
PTEN | Phosphatase and tensin homolog deleted on chromosome 10 |
PTEN-L | PTEN-long |
PTENP1 | PTEN pseudogene 1 |
PTP | Protein tyrosine phosphatase |
SDHD | Succinate dehydrogenase complex subunit D |
SREBP | Sterol regulatory element-binding proteins |
TCGA | The Cancer Genome Atlas |
TOP2A | DNA topoisomerase-2 alpha |
USP11 | Ubiquitin specific peptidase 11 |
VEGF | Vascular endothelial growth factor |
WHSC1 | Wolf–Hirschhorn syndrome candidate 1 |
WWP2 | WW domain-containing protein 2 |
XIAP | X-linked inhibitor of apoptosis |
References
- Li, D.M.; Sun, H. TEP1, encoded by a candidate tumor suppressor locus, is a novel protein tyrosine phosphatase regulated by transforming growth factor beta. Cancer Res. 1997, 57, 2124–2129. [Google Scholar] [PubMed]
- Li, J.; Yen, C.; Liaw, D.; Podsypanina, K.; Bose, S.; Wang, S.I.; Puc, J.; Miliaresis, C.; Rodgers, L.; McCombie, R.; et al. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science 1997, 275, 1943–1947. [Google Scholar] [CrossRef] [PubMed]
- Steck, P.A.; Pershouse, M.A.; Jasser, S.A.; Yung, W.K.; Lin, H.; Ligon, A.H.; Langford, L.A.; Baumgard, M.L.; Hattier, T.; Davis, T.; et al. Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat. Genet. 1997, 15, 356–362. [Google Scholar] [CrossRef] [PubMed]
- Ali, I.U.; Schriml, L.M.; Dean, M. Mutational spectra of PTEN/MMAC1 gene: A tumor suppressor with lipid phosphatase activity. J. Natl. Cancer Inst. 1999, 91, 1922–1932. [Google Scholar] [CrossRef] [PubMed]
- Ali, I.U. Gatekeeper for endometrium: The PTEN tumor suppressor gene. J. Natl. Cancer Inst. 2000, 92, 861–863. [Google Scholar] [CrossRef]
- Kwabi-Addo, B.; Giri, D.; Schmidt, K.; Podsypanina, K.; Parsons, R.; Greenberg, N.; Ittmann, M. Haploinsufficiency of the Pten tumor suppressor gene promotes prostate cancer progression. Proc. Natl. Acad. Sci. USA 2001, 98, 11563–11568. [Google Scholar] [CrossRef]
- Trotman, L.C.; Niki, M.; Dotan, Z.A.; Koutcher, J.A.; Di Cristofano, A.; Xiao, A.; Khoo, A.S.; Roy-Burman, P.; Greenberg, N.M.; Van Dyke, T.; et al. Pten dose dictates cancer progression in the prostate. PLoS Biol. 2003, 1, E59. [Google Scholar] [CrossRef]
- Li, W.; Zhang, T.; Guo, L.; Huang, L. Regulation of PTEN expression by noncoding RNAs. J. Exp. Clin. Cancer Res. 2018, 37, 223. [Google Scholar] [CrossRef]
- Poliseno, L.; Pandolfi, P.P. PTEN ceRNA networks in human cancer. Methods 2015, 77–78, 41–50. [Google Scholar] [CrossRef]
- Campbell, R.B.; Liu, F.; Ross, A.H. Allosteric activation of PTEN phosphatase by phosphatidylinositol 4,5-bisphosphate. J. Biol. Chem. 2003, 278, 33617–33620. [Google Scholar] [CrossRef]
- Tibarewal, P.; Zilidis, G.; Spinelli, L.; Schurch, N.; Maccario, H.; Gray, A.; Perera, N.M.; Davidson, L.; Barton, G.J.; Leslie, N.R. PTEN protein phosphatase activity correlates with control of gene expression and invasion, a tumor-suppressing phenotype, but not with AKT activity. Sci. Signal. 2012, 5, ra18. [Google Scholar] [CrossRef]
- Lee, Y.R.; Chen, M.; Pandolfi, P.P. The functions and regulation of the PTEN tumour suppressor: New modes and prospects. Nat. Rev. Mol. Cell Biol. 2018, 19, 547–562. [Google Scholar] [CrossRef]
- Fruman, D.A.; Chiu, H.; Hopkins, B.D.; Bagrodia, S.; Cantley, L.C.; Abraham, R.T. The PI3K Pathway in Human Disease. Cell 2017, 170, 605–635. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.O.; Yang, H.; Georgescu, M.M.; Di Cristofano, A.; Maehama, T.; Shi, Y.; Dixon, J.E.; Pandolfi, P.; Pavletich, N.P. Crystal structure of the PTEN tumor suppressor: Implications for its phosphoinositide phosphatase activity and membrane association. Cell 1999, 99, 323–334. [Google Scholar] [CrossRef]
- Das, S.; Dixon, J.E.; Cho, W. Membrane-binding and activation mechanism of PTEN. Proc. Natl. Acad. Sci. USA 2003, 100, 7491–7496. [Google Scholar] [CrossRef] [Green Version]
- Fragoso, R.; Barata, J.T. Kinases, tails and more: Regulation of PTEN function by phosphorylation. Methods 2015, 77–78, 75–81. [Google Scholar] [CrossRef]
- Sotelo, N.S.; Schepens, J.T.; Valiente, M.; Hendriks, W.J.; Pulido, R. PTEN-PDZ domain interactions: Binding of PTEN to PDZ domains of PTPN13. Methods 2015, 77–78, 147–156. [Google Scholar] [CrossRef]
- Vaquero, J.; Nguyen Ho-Bouldoires, T.H.; Claperon, A.; Fouassier, L. Role of the PDZ-scaffold protein NHERF1/EBP50 in cancer biology: From signaling regulation to clinical relevance. Oncogene 2017, 36, 3067–3079. [Google Scholar] [CrossRef]
- Haynie, D.T.; Xue, B. Superdomains in the protein structure hierarchy: The case of PTP-C2. Protein Sci. 2015, 24, 874–882. [Google Scholar] [CrossRef] [Green Version]
- Gil, A.; Rodriguez-Escudero, I.; Stumpf, M.; Molina, M.; Cid, V.J.; Pulido, R. A functional dissection of PTEN N-terminus: Implications in PTEN subcellular targeting and tumor suppressor activity. PLoS ONE 2015, 10, e0119287. [Google Scholar] [CrossRef]
- Duerr, E.M.; Rollbrocker, B.; Hayashi, Y.; Peters, N.; Meyer-Puttlitz, B.; Louis, D.N.; Schramm, J.; Wiestler, O.D.; Parsons, R.; Eng, C.; et al. PTEN mutations in gliomas and glioneuronal tumors. Oncogene 1998, 16, 2259–2264. [Google Scholar] [CrossRef] [Green Version]
- Trotman, L.C.; Wang, X.; Alimonti, A.; Chen, Z.; Teruya-Feldstein, J.; Yang, H.; Pavletich, N.P.; Carver, B.S.; Cordon-Cardo, C.; Erdjument-Bromage, H.; et al. Ubiquitination regulates PTEN nuclear import and tumor suppression. Cell 2007, 128, 141–156. [Google Scholar] [CrossRef]
- Radivojac, P.; Vacic, V.; Haynes, C.; Cocklin, R.R.; Mohan, A.; Heyen, J.W.; Goebl, M.G.; Iakoucheva, L.M. Identification, analysis, and prediction of protein ubiquitination sites. Proteins 2010, 78, 365–380. [Google Scholar] [CrossRef]
- Denning, G.; Jean-Joseph, B.; Prince, C.; Durden, D.L.; Vogt, P.K. A short N-terminal sequence of PTEN controls cytoplasmic localization and is required for suppression of cell growth. Oncogene 2007, 26, 3930–3940. [Google Scholar] [CrossRef] [Green Version]
- Walker, S.M.; Downes, C.P.; Leslie, N.R. TPIP: A novel phosphoinositide 3-phosphatase. Biochem. J. 2001, 360, 277–283. [Google Scholar] [CrossRef]
- Furnari, F.B.; Huang, H.J.; Cavenee, W.K. The phosphoinositol phosphatase activity of PTEN mediates a serum-sensitive G1 growth arrest in glioma cells. Cancer Res. 1998, 58, 5002–5008. [Google Scholar]
- Davidson, L.; Maccario, H.; Perera, N.M.; Yang, X.; Spinelli, L.; Tibarewal, P.; Glancy, B.; Gray, A.; Weijer, C.J.; Downes, C.P.; et al. Suppression of cellular proliferation and invasion by the concerted lipid and protein phosphatase activities of PTEN. Oncogene 2010, 29, 687–697. [Google Scholar] [CrossRef]
- Rizo, J.; Sudhof, T.C. Mechanics of membrane fusion. Nat. Struct. Biol. 1998, 5, 839–842. [Google Scholar] [CrossRef]
- Corbalan-Garcia, S.; Gomez-Fernandez, J.C. Signaling through C2 domains: More than one lipid target. Biochim. Biophys. Acta 2014, 1838, 1536–1547. [Google Scholar] [CrossRef] [Green Version]
- Singh, G.; Chan, A.M. Post-translational modifications of PTEN and their potential therapeutic implications. Curr. Cancer Drug Targets 2011, 11, 536–547. [Google Scholar] [CrossRef]
- Kalyoncu, S.; Keskin, O.; Gursoy, A. Interaction prediction and classification of PDZ domains. BMC Bioinform. 2010, 11, 357. [Google Scholar] [CrossRef]
- Luck, K.; Charbonnier, S.; Trave, G. The emerging contribution of sequence context to the specificity of protein interactions mediated by PDZ domains. FEBS Lett. 2012, 586, 2648–2661. [Google Scholar] [CrossRef] [Green Version]
- Feng, W.; Zhang, M. Organization and dynamics of PDZ-domain-related supramodules in the postsynaptic density. Nat. Rev. Neurosci. 2009, 10, 87–99. [Google Scholar] [CrossRef]
- Terrien, E.; Chaffotte, A.; Lafage, M.; Khan, Z.; Prehaud, C.; Cordier, F.; Simenel, C.; Delepierre, M.; Buc, H.; Lafon, M.; et al. Interference with the PTEN-MAST2 interaction by a viral protein leads to cellular relocalization of PTEN. Sci. Signal. 2012, 5, ra58. [Google Scholar] [CrossRef]
- Vazquez, F.; Matsuoka, S.; Sellers, W.R.; Yanagida, T.; Ueda, M.; Devreotes, P.N. Tumor suppressor PTEN acts through dynamic interaction with the plasma membrane. Proc. Natl. Acad. Sci. USA 2006, 103, 3633–3638. [Google Scholar] [CrossRef] [Green Version]
- Franke, T.F.; Hornik, C.P.; Segev, L.; Shostak, G.A.; Sugimoto, C. PI3K/Akt and apoptosis: Size matters. Oncogene 2003, 22, 8983–8998. [Google Scholar] [CrossRef]
- Christie, K.J.; Zochodne, D. Peripheral Axon Regrowth: New Molecular Approaches. Neuroscience 2013, 240, 310–324. [Google Scholar] [CrossRef]
- Chung, J.H.; Eng, C. Nuclear-cytoplasmic partitioning of phosphatase and tensin homologue deleted on chromosome 10 (PTEN) differentially regulates the cell cycle and apoptosis. Cancer Res. 2005, 65, 8096–8100. [Google Scholar] [CrossRef]
- Manning, B.D.; Cantley, L.C. AKT/PKB signaling: Navigating downstream. Cell 2007, 129, 1261–1274. [Google Scholar] [CrossRef]
- Song, M.S.; Salmena, L.; Pandolfi, P.P. The functions and regulation of the PTEN tumour suppressor. Nat. Rev. Mol. Cell Biol. 2012, 13, 283–296. [Google Scholar] [CrossRef]
- Gimm, O.; Perren, A.; Weng, L.P.; Marsh, D.J.; Yeh, J.J.; Ziebold, U.; Gil, E.; Hinze, R.; Delbridge, L.; Lees, J.A.; et al. Differential nuclear and cytoplasmic expression of PTEN in normal thyroid tissue, and benign and malignant epithelial thyroid tumors. Am. J. Pathol. 2000, 156, 1693–1700. [Google Scholar] [CrossRef]
- Sano, T.; Lin, H.; Chen, X.S.; Langford, L.A.; Koul, D.; Bondy, M.L.; Hess, K.R.; Myers, J.N.; Hong, Y.K.; Yung, W.K.A.; et al. Differential expression of MMAC/PTEN in glioblastoma multiforme: Relationship to localization and prognosis. Cancer Res. 1999, 59, 1820–1824. [Google Scholar]
- Whiteman, D.C.; Zhou, X.P.; Cummings, M.C.; Pavey, S.; Hayward, N.K.; Eng, C. Nuclear PTEN expression and clinicopathologic features in a population-based series of primary cutaneous melanoma. Int. J. Cancer 2002, 99, 63–67. [Google Scholar] [CrossRef]
- McCall, P.; Witton, C.J.; Grimsley, S.; Nielsen, K.V.; Edwards, J. Is PTEN loss associated with clinical outcome measures in human prostate cancer? Brit. J. Cancer 2008, 99, 1296–1301. [Google Scholar] [CrossRef] [Green Version]
- Zhu, X.H.; Qin, X.; Fei, M.G.; Hou, W.M.; Greshock, J.; Bachman, K.E.; Kang, J.H.; Qin, C.Y. Loss and reduced expression of PTEN correlate with advanced-stage gastric carcinoma. Exp. Ther. Med. 2013, 5, 57–64. [Google Scholar] [CrossRef]
- Puc, J.; Keniry, M.; Li, H.S.; Pandita, T.K.; Choudhury, A.D.; Memeo, L.; Mansukhani, M.; Murty, V.V.V.S.; Gaciong, Z.; Meek, S.E.M.; et al. Lack of PTEN sequesters CHK1 and initiates genetic instability. Cancer Cell 2005, 7, 193–204. [Google Scholar] [CrossRef] [Green Version]
- Shen, W.H.; Balajee, A.S.; Wang, J.L.; Wu, H.; Eng, C.; Pandolfi, P.P.; Yin, Y.X. Essential role for nuclear PTEN in maintaining chromosomal integrity. Cell 2007, 128, 157–170. [Google Scholar] [CrossRef]
- Zhang, Z.; Hou, S.Q.; He, J.X.; Gu, T.T.; Yin, Y.X.; Shen, W.H. PTEN regulates PLK1 and controls chromosomal stability during cell division. Cell Cycle 2016, 15, 2476–2485. [Google Scholar] [CrossRef] [Green Version]
- Bassi, C.; Ho, J.; Srikumar, T.; Dowling, R.J.O.; Gorrini, C.; Miller, S.J.; Mak, T.W.; Neel, B.G.; Raught, B.; Stambolic, V. Nuclear PTEN Controls DNA Repair and Sensitivity to Genotoxic Stress. Science 2013, 341, 395–399. [Google Scholar] [CrossRef] [Green Version]
- Brandmaier, A.; Hou, S.Q.; Shen, W.H. Cell Cycle Control by PTEN. J. Mol. Biol. 2017, 429, 2265–2277. [Google Scholar] [CrossRef]
- Li, A.G.; Piluso, L.G.; Cai, X.; Wei, G.; Sellers, W.R.; Liu, X. Mechanistic insights into maintenance of high p53 acetylation by PTEN. Mol. Cell 2006, 23, 575–587. [Google Scholar] [CrossRef]
- Kim, S.J.; Lee, H.W.; Baek, J.H.; Cho, Y.H.; Kang, H.G.; Jeong, J.S.; Song, J.; Park, H.S.; Chun, K.H. Activation of nuclear PTEN by inhibition of Notch signaling induces G2/M cell cycle arrest in gastric cancer. Oncogene 2016, 35, 251–260. [Google Scholar] [CrossRef]
- Kang, X.; Song, C.; Du, X.; Zhang, C.; Liu, Y.; Liang, L.; He, J.X.; Lamb, K.; Shen, W.H.; Yin, Y.X. PTEN stabilizes TOP2A and regulates the DNA decatenation. Sci. Rep. 2015, 5, 17873. [Google Scholar] [CrossRef] [Green Version]
- Bononi, A.; Bonora, M.; Marchi, S.; Missiroli, S.; Poletti, F.; Giorgi, C.; Pandolfi, P.P.; Pinton, P. Identification of PTEN at the ER and MAMs and its regulation of Ca2+ signaling and apoptosis in a protein phosphatase-dependent manner. Cell Death Differ. 2013, 20, 1631–1643. [Google Scholar] [CrossRef]
- Kuchay, S.; Giorgi, C.; Simoneschi, D.; Pagan, J.; Missiroli, S.; Saraf, A.; Florens, L.; Washburn, M.P.; Collazo-Lorduy, A.; Castillo-Martin, M.; et al. PTEN counteracts FBXL2 to promote IP3R3- and Ca(2+)-mediated apoptosis limiting tumour growth. Nature 2017, 546, 554–558. [Google Scholar] [CrossRef]
- Zhu, Y.; Hoell, P.; Ahlemeyer, B.; Krieglstein, J. PTEN: A crucial mediator of mitochondria-dependent apoptosis. Apoptosis 2006, 11, 197–207. [Google Scholar] [CrossRef]
- Comelli, M.; Pretis, I.; Buso, A.; Mavelli, I. Mitochondrial energy metabolism and signalling in human glioblastoma cell lines with different PTEN gene status. J. Bioenerg. Biomembr. 2018, 50, 33–52. [Google Scholar] [CrossRef]
- Li, P.D.; Wang, D.N.; Li, H.Y.; Yu, Z.K.; Chen, X.H.; Fang, J.G. Identification of nucleolus-localized PTEN and its function in regulating ribosome biogenesis. Mol. Biol. Rep. 2014, 41, 6383–6390. [Google Scholar] [CrossRef]
- Derenzini, M.; Montanaro, L.; Trere, D. Ribosome biogenesis and cancer. Acta Histochem. 2017, 119, 190–197. [Google Scholar] [CrossRef]
- Liang, H.; Chen, X.; Yin, Q.; Ruan, D.H.; Zhao, X.Y.; Zhang, C.; McNutt, M.A.; Yin, Y.X. PTEN beta is an alternatively translated isoform of PTEN that regulates rDNA transcription. Nat. Commun. 2017, 8, 14771. [Google Scholar] [CrossRef]
- Van Ree, J.H.; Nam, H.J.; Jeganathan, K.B.; Kanakkanthara, A.; van Deursen, J.M. Pten regulates spindle pole movement through Dlg1-mediated recruitment of Eg5 to centrosomes. Nat. Cell Biol. 2016, 18, 814–821. [Google Scholar] [CrossRef]
- Shnitsar, I.; Bashkurov, M.; Masson, G.R.; Ogunjimi, A.A.; Mosessian, S.; Cabeza, E.A.; Hirsch, C.L.; Trcka, D.; Gish, G.; Jiao, J.; et al. PTEN regulates cilia through Dishevelled. Nat. Commun. 2015, 6, 8388. [Google Scholar] [CrossRef] [Green Version]
- Hopkins, B.D.; Fine, B.; Steinbach, N.; Dendy, M.; Rapp, Z.; Shaw, J.; Pappas, K.; Yu, J.S.; Hodakoski, C.; Mense, S.; et al. A Secreted PTEN Phosphatase That Enters Cells to Alter Signaling and Survival. Science 2013, 341, 399–402. [Google Scholar] [CrossRef] [Green Version]
- Liang, H.; He, S.; Yang, J.; Jia, X.; Wang, P.; Chen, X.; Zhang, Z.; Zou, X.; McNutt, M.A.; Shen, W.H.; et al. PTENalpha, a PTEN isoform translated through alternative initiation, regulates mitochondrial function and energy metabolism. Cell Metab. 2014, 19, 836–848. [Google Scholar] [CrossRef]
- Putz, U.; Howitt, J.; Doan, A.; Goh, C.P.; Low, L.H.; Silke, J.; Tan, S.S. The Tumor Suppressor PTEN Is Exported in Exosomes and Has Phosphatase Activity in Recipient Cells. Sci. Signal. 2012, 5, ra70. [Google Scholar] [CrossRef]
- Cao, Y.Y.; Wang, H.Y.; Yang, L.; Zhang, Z.; Li, C.L.; Yuan, X.; Bu, L.; Chen, L.; Chen, Y.; Li, C.M.; et al. PTEN-L promotes type I interferon responses and antiviral immunity. Cell. Mol. Immunol. 2018, 15, 48–57. [Google Scholar] [CrossRef]
- Goncalves, M.B.; Malmqvist, T.; Clarke, E.; Hubens, C.J.; Grist, J.; Hobbs, C.; Trigo, D.; Risling, M.; Angeria, M.; Damberg, P.; et al. Neuronal RAR beta Signaling Modulates PTEN Activity Directly in Neurons and via Exosome Transfer in Astrocytes to Prevent Glial Scar Formation and Induce Spinal Cord Regeneration. J. Neurosci. 2015, 35, 15731–15745. [Google Scholar] [CrossRef]
- Lavon, I.; Refael, M.; Zelikovitch, B.; Shalom, E.; Siegal, T. Serum DNA can define tumor-specific genetic and epigenetic markers in gliomas of various grades. Neuro Oncol. 2010, 12, 173–180. [Google Scholar] [CrossRef] [Green Version]
- De Mattos-Arruda, L.; Mayor, R.; Ng, C.K.Y.; Weigelt, B.; Martinez-Ricarte, F.; Torrejon, D.; Oliveira, M.; Arias, A.; Raventos, C.; Tang, J.B.; et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat. Commun. 2015, 6, 8839. [Google Scholar] [CrossRef]
- Papa, A.; Wan, L.X.; Bonora, M.; Salmena, L.; Song, M.S.; Hobbs, R.M.; Lunardi, A.; Webster, K.; Ng, C.; Newton, R.H.; et al. Cancer-Associated PTEN Mutants Act in a Dominant-Negative Manner to Suppress PTEN Protein Function. Cell 2014, 157, 595–610. [Google Scholar] [CrossRef] [Green Version]
- Jamaspishvili, T.; Berman, D.M.; Ross, A.E.; Scher, H.I.; De Marzo, A.M.; Squire, J.A.; Lotan, T.L. Clinical implications of PTEN loss in prostate cancer. Nat. Rev. Urol. 2018, 15, 222–234. [Google Scholar] [CrossRef]
- McConechy, M.K.; Ding, J.; Cheang, M.C.; Wiegand, K.; Senz, J.; Tone, A.; Yang, W.; Prentice, L.; Tse, K.; Zeng, T.; et al. Use of mutation profiles to refine the classification of endometrial carcinomas. J. Pathol. 2012, 228, 20–30. [Google Scholar] [CrossRef] [Green Version]
- Álvarez-Garcia, V.; Tawil, Y.; Wise, H.M.; Leslie, N.R. Mechanisms of PTEN loss in cancer: It’s all about diversity. Semin. Cancer Biol. 2019. [Google Scholar] [CrossRef]
- Wiencke, J.K.; Zheng, S.; Jelluma, N.; Tihan, T.; Vandenberg, S.; Tamguney, T.; Baumber, R.; Parsons, R.; Lamborn, K.R.; Berger, M.S.; et al. Methylation of the PTEN promoter defines low-grade gliomas and secondary glioblastoma. Neuro Oncol. 2007, 9, 271–279. [Google Scholar] [CrossRef] [Green Version]
- Roh, M.R.; Gupta, S.; Park, K.H.; Chung, K.Y.; Lauss, M.; Flaherty, K.T.; Jonsson, G.; Rha, S.Y.; Tsao, H. Promoter Methylation of PTEN Is a Significant Prognostic Factor in Melanoma Survival. J. Investig. Dermatol. 2016, 136, 1002–1011. [Google Scholar] [CrossRef]
- Berger, A.H.; Knudson, A.G.; Pandolfi, P.P. A continuum model for tumour suppression. Nature 2011, 476, 163–169. [Google Scholar] [CrossRef] [Green Version]
- Alimonti, A.; Carracedo, A.; Clohessy, J.G.; Trotman, L.C.; Nardella, C.; Egia, A.; Salmena, L.; Sampieri, K.; Haveman, W.J.; Brogi, E.; et al. Subtle variations in Pten dose determine cancer susceptibility. Nat. Genet. 2010, 42, 454–458. [Google Scholar] [CrossRef]
- Carracedo, A.; Alimonti, A.; Pandolfi, P.P. PTEN level in tumor suppression: How much is too little? Cancer Res. 2011, 71, 629–633. [Google Scholar] [CrossRef]
- Salvesen, H.B.; MacDonald, N.; Ryan, A.; Jacobs, I.J.; Lynch, E.D.; Akslen, L.A.; Das, S. PTEN methylation is associated with advanced stage and microsatellite instability in endometrial carcinoma. Int. J. Cancer 2001, 91, 22–26. [Google Scholar] [CrossRef]
- Koul, D. PTEN signaling pathways in glioblastoma. Cancer Biol. Ther. 2008, 7, 1321–1325. [Google Scholar] [CrossRef]
- Soria, J.C.; Lee, H.Y.; Lee, J.I.; Wang, L.; Issa, J.P.; Kemp, B.L.; Liu, D.D.; Kurie, J.M.; Mao, L.; Khuri, F.R. Lack of PTEN expression in non-small cell lung cancer could be related to promoter methylation. Clin. Cancer Res. 2002, 8, 1178–1184. [Google Scholar]
- Wang, X.; Jiang, X. Post-translational regulation of PTEN. Oncogene 2008, 27, 5454–5463. [Google Scholar] [CrossRef] [Green Version]
- Carver, B.S.; Tran, J.; Gopalan, A.; Chen, Z.; Shaikh, S.; Carracedo, A.; Alimonti, A.; Nardella, C.; Varmeh, S.; Scardino, P.T.; et al. Aberrant ERG expression cooperates with loss of PTEN to promote cancer progression in the prostate. Nat. Genet. 2009, 41, 619–624. [Google Scholar] [CrossRef] [Green Version]
- Yoshimoto, M.; Joshua, A.M.; Cunha, I.W.; Coudry, R.A.; Fonseca, F.P.; Ludkovski, O.; Zielenska, M.; Soares, F.A.; Squire, J.A. Absence of TMPRSS2:ERG fusions and PTEN losses in prostate cancer is associated with a favorable outcome. Mod. Pathol. 2008, 21, 1451–1460. [Google Scholar] [CrossRef] [Green Version]
- Krohn, A.; Freudenthaler, F.; Harasimowicz, S.; Kluth, M.; Fuchs, S.; Burkhardt, L.; Stahl, P.; Tsourlakis, M.T.; Bauer, M.; Tennstedt, P.; et al. Heterogeneity and chronology of PTEN deletion and ERG fusion in prostate cancer. Mod. Pathol. 2014, 27, 1612–1620. [Google Scholar] [CrossRef] [Green Version]
- King, J.C.; Xu, J.; Wongvipat, J.; Hieronymus, H.; Carver, B.S.; Leung, D.H.; Taylor, B.S.; Sander, C.; Cardiff, R.D.; Couto, S.S.; et al. Cooperativity of TMPRSS2-ERG with PI3-kinase pathway activation in prostate oncogenesis. Nat. Genet. 2009, 41, 524–526. [Google Scholar] [CrossRef] [Green Version]
- Chen, Z.B.; Trotman, L.C.; Shaffer, D.; Lin, H.K.; Dotan, Z.A.; Niki, M.; Koutcher, J.A.; Scher, H.I.; Ludwig, T.; Gerald, W.; et al. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature 2005, 436, 725–730. [Google Scholar] [CrossRef] [Green Version]
- Mayo, L.D.; Donner, D.B. The PTEN, Mdm2, p53 tumor suppressor-oncoprotein network. Trends Biochem. Sci. 2002, 27, 462–467. [Google Scholar] [CrossRef]
- Freeman, D.J.; Li, A.G.; Wei, G.; Li, H.H.; Kertesz, N.; Lesche, R.; Whale, A.D.; Martinez-Diaz, H.; Rozengurt, N.; Cardiff, R.D.; et al. PTEN tumor suppressor regulates p53 protein levels and activity through phosphatase-dependent and -independent mechanisms. Cancer Cell 2003, 3, 117–130. [Google Scholar] [CrossRef] [Green Version]
- Stambolic, V.; MacPherson, D.; Sas, D.; Lin, Y.; Snow, B.; Jang, Y.; Benchimol, S.; Mak, T.W. Regulation of PTEN transcription by p53. Mol. Cell 2001, 8, 317–325. [Google Scholar] [CrossRef]
- Puszynski, K.; Hat, B.; Lipniacki, T. Oscillations and bistability in the stochastic model of p53 regulation. J. Theor. Biol. 2008, 254, 452–465. [Google Scholar] [CrossRef]
- Gallia, G.L.; Rand, V.; Siu, I.M.; Eberhart, C.G.; James, C.D.; Marie, S.K.N.; Oba-Shinjo, S.M.; Carlotti, C.G.; Caballero, O.L.; Simpson, A.J.G.; et al. PIK3CA gene mutations in pediatric and adult glioblastoma multiforme. Mol. Cancer Res. 2006, 4, 709–714. [Google Scholar] [CrossRef]
- Ito, C.; Nishizuka, S.S.; Ishida, K.; Uesugi, N.; Sugai, T.; Tamura, G.; Koeda, K.; Sasaki, A. Analysis of PIK3CA mutations and PI3K pathway proteins in advanced gastric cancer. J. Surg. Res. 2017, 212, 195–204. [Google Scholar] [CrossRef]
- VanderLaan, P.A.; Rangachari, D.; Mockus, S.M.; Spotlow, V.; Reddi, H.V.; Malcolm, J.; Huberman, M.S.; Joseph, L.J.; Kobayashi, S.S.; Costa, D.B. Mutations in TP53, PIK3CA, PTEN and other genes in EGFR mutated lung cancers: Correlation with clinical outcomes. Lung Cancer 2017, 106, 17–21. [Google Scholar] [CrossRef]
- Manning, B.D.; Toker, A. AKT/PKB Signaling: Navigating the Network. Cell 2017, 169, 381–405. [Google Scholar] [CrossRef] [Green Version]
- Wu, X.Y.; Senechal, K.; Neshat, M.S.; Whang, Y.E.; Sawyers, C.L. The PTEN/MMAC1 tumor suppressor phosphatase functions as a negative regulator of the phosphoinositide 3-kinase/Akt pathway. Proc. Natl. Acad. Sci. USA 1998, 95, 15587–15591. [Google Scholar] [CrossRef] [Green Version]
- Datta, S.R.; Brunet, A.; Greenberg, M.E. Cellular survival: A play in three Akts. Genes Dev. 1999, 13, 2905–2927. [Google Scholar] [CrossRef]
- Maroulakou, I.G.; Oemler, W.; Naber, S.P.; Tsichlis, P.N. Akt1 ablation inhibits, whereas Akt2 ablation accelerates, the development of mammary adenocarcinomas in mouse mammary tumor virus (MMTV)-ErbB2/Neu and MMTV-polyoma middle T transgenic mice. Cancer Res. 2007, 67, 167–177. [Google Scholar] [CrossRef]
- Fraser, M.M.; Zhu, X.Y.; Kwon, C.H.; Uhlmann, E.J.; Gutmann, D.H.; Baker, S.J. Pten loss causes hypertrophy and increased proliferation of astrocytes in vivo. Cancer Res. 2004, 64, 7773–7779. [Google Scholar] [CrossRef]
- Gottschalk, A.R.; Basila, D.; Wong, M.; Dean, N.M.; Brandts, C.H.; Stokoe, D.; Haas-Kogan, D.A. p27Kip1 is required for PTEN-induced G1 growth arrest. Cancer Res. 2001, 61, 2105–2111. [Google Scholar]
- Seminario, M.C.; Precht, P.; Wersto, R.P.; Gorospe, M.; Wange, R.L. PTEN expression in PTEN-null leukaemic T cell lines leads to reduced proliferation via slowed cell cycle progression. Oncogene 2003, 22, 8195–8204. [Google Scholar] [CrossRef] [Green Version]
- Di Cristofano, A.; De Acetis, M.; Koff, A.; Cordon-Cardo, C.; Pandolfi, P.P. Pten and p27KIP1 cooperate in prostate cancer tumor suppression in the mouse. Nat. Genet. 2001, 27, 222–224. [Google Scholar] [CrossRef]
- Tomkiel, J.; Cooke, C.A.; Saitoh, H.; Bernat, R.L.; Earnshaw, W.C. Cenp-C Is Required for Maintaining Proper Kinetochore Size and for a Timely Transition to Anaphase. J. Cell Biol. 1994, 125, 531–545. [Google Scholar] [CrossRef]
- Yan, M.; Wang, Y.; Wong, C.W.; Or, P.M.; Wong, K.L.; Li, L.; Many, A.M.; Guan, H.; Khoo, U.S.; Chan, A.M. PTEN PDZ-binding domain suppresses mammary carcinogenesis in the MMTV-PyMT breast cancer model. Cancer Lett. 2018, 430, 67–78. [Google Scholar] [CrossRef]
- Warburg, O. On the origin of cancer cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef]
- Ortega-Molina, A.; Efeyan, A.; Lopez-Guadamillas, E.; Munoz-Martin, M.; Gomez-Lopez, G.; Canamero, M.; Mulero, F.; Pastor, J.; Martinez, S.; Romanos, E.; et al. Pten positively regulates brown adipose function, energy expenditure, and longevity. Cell Metab. 2012, 15, 382–394. [Google Scholar] [CrossRef]
- Shimobayashi, M.; Hall, M.N. Making new contacts: The mTOR network in metabolism and signalling crosstalk. Nat. Rev. Mol. Cell Biol. 2014, 15, 155–162. [Google Scholar] [CrossRef]
- Wong, J.T.; Kim, P.T.; Peacock, J.W.; Yau, T.Y.; Mui, A.L.; Chung, S.W.; Sossi, V.; Doudet, D.; Green, D.; Ruth, T.J.; et al. Pten (phosphatase and tensin homologue gene) haploinsufficiency promotes insulin hypersensitivity. Diabetologia 2007, 50, 395–403. [Google Scholar] [CrossRef]
- Miinea, C.P.; Sano, H.; Kane, S.; Sano, E.; Fukuda, M.; Peranen, J.; Lane, W.S.; Lienhard, G.E. AS160, the Akt substrate regulating GLUT4 translocation, has a functional Rab GTPase-activating protein domain. Biochem. J. 2005, 391, 87–93. [Google Scholar] [CrossRef]
- Liu, F.; Dallas-Yang, Q.; Castriota, G.; Fischer, P.; Santini, F.; Ferrer, M.; Li, J.; Akiyama, T.E.; Berger, J.P.; Zhang, B.B.; et al. Development of a novel GLUT4 translocation assay for identifying potential novel therapeutic targets for insulin sensitization. Biochem. J. 2009, 418, 413–420. [Google Scholar] [CrossRef] [Green Version]
- Puigserver, P.; Rhee, J.; Donovan, J.; Walkey, C.J.; Yoon, J.C.; Oriente, F.; Kitamura, Y.; Altomonte, J.; Dong, H.J.; Accili, D.; et al. Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1 alpha interaction. Nature 2003, 423, 550–555. [Google Scholar] [CrossRef]
- Li, X.H.; Monks, B.; Ge, Q.Y.; Birnbaum, M.J. Akt/PKB regulates hepatic metabolism by directly inhibiting PGC-1 alpha transcription coactivator. Nature 2007, 447, 1012-U1018. [Google Scholar] [CrossRef]
- Fang, M.; Shen, Z.R.; Huang, S.; Zhao, L.P.; Chen, S.; Mak, T.W.; Wang, X.D. The ER UDPase ENTPD5 Promotes Protein N-Glycosylation, the Warburg Effect, and Proliferation in the PTEN Pathway. Cell 2010, 143, 711–724. [Google Scholar] [CrossRef] [Green Version]
- Chen, M.; Zhang, J.W.; Sampieri, K.; Clohessy, J.G.; Mendez, L.; Gonzalez-Billalabeitia, E.; Liu, X.S.; Lee, Y.R.; Fung, J.; Katon, J.M.; et al. An aberrant SREBP-dependent lipogenic program promotes metastatic prostate cancer. Nat. Genet. 2018, 50, 206–218. [Google Scholar] [CrossRef] [Green Version]
- Zoncu, R.; Efeyan, A.; Sabatini, D.M. mTOR: From growth signal integration to cancer, diabetes and ageing. Nat. Rev. Mol. Cell Biol. 2011, 12, 21–35. [Google Scholar] [CrossRef]
- Zabala-Letona, A.; Arruabarrena-Aristorena, A.; Martin-Martin, N.; Fernandez-Ruiz, S.; Sutherland, J.D.; Clasquin, M.; Tomas-Cortazar, J.; Jimenez, J.; Torres, I.; Quang, P.; et al. mTORC1-dependent AMD1 regulation sustains polyamine metabolism in prostate cancer. Nature 2017, 547, 109–113. [Google Scholar] [CrossRef]
- Garcia-Cao, I.; Song, M.S.; Hobbs, R.M.; Laurent, G.; Giorgi, C.; de Boer, V.C.J.; Anastasiou, D.; Ito, K.; Sasaki, A.T.; Rameh, L.; et al. Systemic Elevation of PTEN Induces a Tumor-Suppressive Metabolic State. Cell 2012, 149, 49–62. [Google Scholar] [CrossRef] [Green Version]
- Tosoian, J.J.; Almutairi, F.; Morais, C.L.; Glavaris, S.; Hicks, J.; Sundi, D.; Humphreys, E.; Han, M.; De Marzo, A.M.; Ross, A.E.; et al. Prevalence and Prognostic Significance of PTEN Loss in African-American and European-American Men Undergoing Radical Prostatectomy. Eur. Urol. 2017, 71, 697–700. [Google Scholar] [CrossRef]
- Li, Y.; Cui, J.; Zhang, C.H.; Yang, D.J.; Chen, J.H.; Zan, W.H.; Li, B.; Li, Z.; He, Y.L. High-expression of DJ-1 and Loss of PTEN Associated with Tumor Metastasis and Correlated with Poor Prognosis of Gastric Carcinoma. Int. J. Med. Sci. 2013, 10, 1689–1697. [Google Scholar] [CrossRef] [Green Version]
- Robinson, D.R.; Wu, Y.M.; Lonigro, R.J.; Vats, P.; Cobain, E.; Everett, J.; Cao, X.H.; Rabban, E.; Kumar-Sinha, C.; Raymond, V.; et al. Integrative clinical genomics of metastatic cancer. Nature 2017, 548, 297–303. [Google Scholar] [CrossRef]
- Wang, S.Y.; Gao, J.; Lei, Q.Y.; Rozengurt, N.; Pritchard, C.; Jiao, J.; Thomas, G.V.; Li, G.; Roy-Burman, P.; Nelson, P.S.; et al. Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer. Cancer Cell 2003, 4, 209–221. [Google Scholar] [CrossRef] [Green Version]
- Mulholland, D.J.; Kobayashi, N.; Ruscetti, M.; Zhi, A.; Tran, L.M.; Huang, J.T.; Gleave, M.; Wu, H. Pten Loss and RAS/MAPK Activation Cooperate to Promote EMT and Metastasis Initiated from Prostate Cancer Stem/Progenitor Cells. Cancer Res. 2012, 72, 1878–1889. [Google Scholar] [CrossRef]
- Nogueira, C.; Kim, K.H.; Sung, H.; Paraiso, K.H.T.; Dannenberg, J.H.; Bosenberg, M.; Chin, L.; Kim, M. Cooperative interactions of PTEN deficiency and RAS activation in melanoma metastasis. Oncogene 2010, 29, 6222–6232. [Google Scholar] [CrossRef] [Green Version]
- Li, N.; Xue, W.; Yuan, H.R.; Dong, B.J.; Ding, Y.F.; Liu, Y.F.; Jiang, M.; Kan, S.; Sun, T.Y.; Ren, J.L.; et al. AKT-mediated stabilization of histone methyltransferase WHSC1 promotes prostate cancer metastasis. J. Clin. Investig. 2017, 127, 1284–1302. [Google Scholar] [CrossRef] [Green Version]
- Won, O.J.; Zhang, L.; Wang, J.H.; Su, Q.T.; Feng, Q.; Zhang, X.H.F.; Mani, S.A.; Paulter, R.; Creighton, C.J.; Ittmann, M.M.; et al. Notch promotes tumor metastasis in a prostate-specific Pten-null mouse model. J. Clin. Investig. 2016, 126, 2626–2641. [Google Scholar] [CrossRef]
- Dankort, D.; Curley, D.P.; Cartlidge, R.A.; Nelson, B.; Karnezis, A.N.; Damsky, W.E.; You, M.J.; DePinho, R.A.; McMahon, M.; Bosenberg, M. Braf(V600E) cooperates with Pten loss to induce metastatic melanoma. Nat. Genet. 2009, 41, 544–552. [Google Scholar] [CrossRef]
- Ding, Z.H.; Wu, C.J.; Chu, G.C.; Xiao, Y.H.; Ho, D.; Zhang, J.F.; Perry, S.R.; Labrot, E.S.; Wu, X.Q.; Lis, R.; et al. SMAD4-dependent barrier constrains prostate cancer growth and metastatic progression. Nature 2011, 470, 269–273. [Google Scholar] [CrossRef] [Green Version]
- Liliental, J.; Moon, S.Y.; Lesche, R.; Mamillapalli, R.; Li, D.M.; Zheng, Y.; Sun, H.; Wu, H. Genetic deletion of the Pten tumor suppressor gene promotes cell motility by activation of Rac1 and Cdc42 GTPases. Curr. Biol. 2000, 10, 401–404. [Google Scholar] [CrossRef] [Green Version]
- Raftopoulou, M.; Etienne-Manneville, S.; Self, A.; Nicholls, S.; Hall, A. Regulation of cell migration by the C2 domain of the tumor suppressor PTEN. Science 2004, 303, 1179–1181. [Google Scholar] [CrossRef]
- Leslie, N.R.; Yang, X.S.; Downes, C.P.; Weijer, C.J. PtdIns(3,4,5)P-3-dependent and -independent roles for PTEN in the control of cell migration. Curr. Biol. 2007, 17, 115–125. [Google Scholar] [CrossRef]
- Tian, K.; Di, R.; Wang, L. MicroRNA-23a enhances migration and invasion through PTEN in osteosarcoma. Cancer Gene Ther. 2015, 22, 351–359. [Google Scholar] [CrossRef]
- Carmeliet, P.; Jain, R.K. Angiogenesis in cancer and other diseases. Nature 2000, 407, 249–257. [Google Scholar] [CrossRef]
- Wen, S.; Stolarov, J.; Myers, M.P.; Su, J.D.; Wigler, M.H.; Tonks, N.K.; Durden, D.L. PTEN controls tumor-induced angiogenesis. Proc. Natl. Acad. Sci. USA 2001, 98, 4622–4627. [Google Scholar] [CrossRef] [Green Version]
- Choorapoikayil, S.; Weijts, B.; Kers, R.; de Bruin, A.; den Hertog, J. Loss of Pten promotes angiogenesis and enhanced vegfaa expression in zebrafish. Dis. Models Mech. 2013, 6, 1159–1166. [Google Scholar] [CrossRef]
- Tian, T.; Nan, K.J.; Wang, S.H.; Liang, X.A.; Lu, C.X.; Guo, H.; Wang, W.J.; Ruan, Z.P. PTEN regulates angiogenesis and VEGF expression through phosphatase-dependent and -independent mechanisms in HepG2 cells. Carcinogenesis 2010, 31, 1211–1219. [Google Scholar] [CrossRef] [Green Version]
- Serra, H.; Chivite, I.; Angulo-Urarte, A.; Soler, A.; Sutherland, J.D.; Arruabarrena-Aristorena, A.; Ragab, A.; Lim, R.; Malumbres, M.; Fruttiger, M.; et al. PTEN mediates Notch-dependent stalk cell arrest in angiogenesis. Nat. Commun. 2015, 6, 7935. [Google Scholar] [CrossRef] [Green Version]
- Meng, F.; Henson, R.; Wehbe-Janek, H.; Ghoshal, K.; Jacob, S.T.; Patel, T. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 2007, 133, 647–658. [Google Scholar] [CrossRef]
- Huse, J.T.; Brennan, C.; Hambardzumyan, D.; Wee, B.; Pena, J.; Rouhanifard, S.H.; Sohn-Lee, C.; le Sage, C.; Agami, R.; Tuschl, T.; et al. The PTEN-regulating microRNA miR-26a is amplified in high-grade glioma and facilitates gliomagenesis in vivo. Genes Dev. 2009, 23, 1327–1337. [Google Scholar] [CrossRef] [Green Version]
- Zhang, H.; Cao, H.; Xu, D.; Zhu, K. MicroRNA-92a promotes metastasis of nasopharyngeal carcinoma by targeting the PTEN/AKT pathway. Onco Targets Ther. 2016, 9, 3579–3588. [Google Scholar] [CrossRef] [Green Version]
- Wei, H.; Cui, R.; Bahr, J.; Zanesi, N.; Luo, Z.; Meng, W.; Liang, G.; Croce, C.M. miR-130a Deregulates PTEN and Stimulates Tumor Growth. Cancer Res. 2017, 77, 6168–6178. [Google Scholar] [CrossRef] [Green Version]
- Zhang, P.; Lu, X.; Shi, Z.; Li, X.; Zhang, Y.; Zhao, S.; Liu, H. miR-205-5p regulates epithelial-mesenchymal transition by targeting PTEN via PI3K/AKT signaling pathway in cisplatin-resistant nasopharyngeal carcinoma cells. Gene 2019, 710, 103–113. [Google Scholar] [CrossRef]
- Ma, J.; Liu, J.; Wang, Z.; Gu, X.; Fan, Y.; Zhang, W.; Xu, L.; Zhang, J.; Cai, D. NF-kappaB-dependent microRNA-425 upregulation promotes gastric cancer cell growth by targeting PTEN upon IL-1beta induction. Mol. Cancer 2014, 13, 40. [Google Scholar] [CrossRef]
- Poliseno, L.; Salmena, L.; Zhang, J.; Carver, B.; Haveman, W.J.; Pandolfi, P.P. A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature 2010, 465, 1033–1038. [Google Scholar] [CrossRef] [Green Version]
- Hu, S.; Xu, L.; Li, L.; Luo, D.; Zhao, H.; Li, D.; Peng, B. Overexpression of lncRNA PTENP1 suppresses glioma cell proliferation and metastasis in vitro. Onco Targets Ther. 2019, 12, 147–156. [Google Scholar] [CrossRef]
- Shi, X.; Tang, X.; Su, L. Overexpression of Long Noncoding RNA PTENP1 Inhibits Cell Proliferation and Migration via Suppression of miR-19b in Breast Cancer Cells. Oncol. Res. 2018, 26, 869–878. [Google Scholar] [CrossRef]
- Zheng, R.; Du, M.; Wang, X.; Xu, W.; Liang, J.; Wang, W.; Lv, Q.; Qin, C.; Chu, H.; Wang, M.; et al. Exosome-transmitted long non-coding RNA PTENP1 suppresses bladder cancer progression. Mol. Cancer 2018, 17, 143. [Google Scholar] [CrossRef]
- McConnachie, G.; Pass, I.; Walker, S.M.; Downes, C.P. Interfacial kinetic analysis of the tumour suppressor phosphatase, PTEN: Evidence for activation by anionic phospholipids. Biochem. J. 2003, 371, 947–955. [Google Scholar] [CrossRef]
- Redfern, R.E.; Redfern, D.; Furgason, M.L.; Munson, M.; Ross, A.H.; Gericke, A. PTEN phosphatase selectively binds phosphoinositides and undergoes structural changes. Biochemistry 2008, 47, 2162–2171. [Google Scholar] [CrossRef]
- Morotti, A.; Panuzzo, C.; Crivellaro, S.; Carra, G.; Fava, C.; Guerrasio, A.; Pandolfi, P.P.; Saglio, G. BCR-ABL inactivates cytosolic PTEN through Casein Kinase II mediated tail phosphorylation. Cell Cycle 2015, 14, 973–979. [Google Scholar] [CrossRef]
- Cordier, F.; Chaffotte, A.; Terrien, E.; Prehaud, C.; Theillet, F.X.; Delepierre, M.; Lafon, M.; Buc, H.; Wolff, N. Ordered phosphorylation events in two independent cascades of the PTEN C-tail revealed by NMR. J. Am. Chem. Soc. 2012, 134, 20533–20543. [Google Scholar] [CrossRef]
- Odriozola, L.; Singh, G.; Hoang, T.; Chan, A.M. Regulation of PTEN activity by its carboxyl-terminal autoinhibitory domain. J. Biol. Chem. 2007, 282, 23306–23315. [Google Scholar] [CrossRef]
- Rahdar, M.; Inoue, T.; Meyer, T.; Zhang, J.; Vazquez, F.; Devreotes, P.N. A phosphorylation-dependent intramolecular interaction regulates the membrane association and activity of the tumor suppressor PTEN. Proc. Natl. Acad. Sci. USA 2009, 106, 480–485. [Google Scholar] [CrossRef] [Green Version]
- Leslie, N.R.; Kriplani, N.; Hermida, M.A.; Alvarez-Garcia, V.; Wise, H.M. The PTEN protein: Cellular localization and post-translational regulation. Biochem. Soc. Trans. 2016, 44, 273–278. [Google Scholar] [CrossRef]
- Nguyen, H.N.; Afkari, Y.; Senoo, H.; Sesaki, H.; Devreotes, P.N.; Iijima, M. Mechanism of human PTEN localization revealed by heterologous expression in Dictyostelium. Oncogene 2014, 33, 5688–5696. [Google Scholar] [CrossRef]
- Nguyen, H.N.; Yang, J.M.; Afkari, Y.; Park, B.H.; Sesaki, H.; Devreotes, P.N.; Iijima, M. Engineering ePTEN, an enhanced PTEN with increased tumor suppressor activities. Proc. Natl. Acad. Sci. USA 2014, 111, E2684–E2693. [Google Scholar] [CrossRef] [Green Version]
- Lee, S.R.; Yang, K.S.; Kwon, J.; Lee, C.; Jeong, W.; Rhee, S.G. Reversible inactivation of the tumor suppressor PTEN by H2O2. J. Biol. Chem. 2002, 277, 20336–20342. [Google Scholar] [CrossRef]
- Kwon, J.; Lee, S.R.; Yang, K.S.; Ahn, Y.; Kim, Y.J.; Stadtman, E.R.; Rhee, S.G. Reversible oxidation and inactivation of the tumor suppressor PTEN in cells stimulated with peptide growth factors. Proc. Natl. Acad. Sci. USA 2004, 101, 16419–16424. [Google Scholar] [CrossRef] [Green Version]
- Schwertassek, U.; Haque, A.; Krishnan, N.; Greiner, R.; Weingarten, L.; Dick, T.P.; Tonks, N.K. Reactivation of oxidized PTP1B and PTEN by thioredoxin 1. FEBS J. 2014, 281, 3545–3558. [Google Scholar] [CrossRef] [Green Version]
- Han, S.J.; Zhang, Y.; Kim, I.; Chay, K.O.; Yoon, H.J.; Jang, D.I.; Yang, S.Y.; Park, J.; Woo, H.A.; Park, I.; et al. Redox regulation of the tumor suppressor PTEN by the thioredoxin system and cumene hydroperoxide. Free Radic. Biol. Med. 2017, 112, 277–286. [Google Scholar] [CrossRef]
- Kim, Y.; Song, Y.B.; Kim, T.Y.; Kim, I.; Han, S.J.; Ahn, Y.; Cho, S.H.; Choi, C.Y.; Chay, K.O.; Yang, S.Y.; et al. Redox regulation of the tumor suppressor PTEN by glutathione. FEBS Lett. 2010, 584, 3550–3556. [Google Scholar] [CrossRef]
- Zhang, Y.; Han, S.J.; Park, I.; Kim, I.; Chay, K.O.; Kim, S.M.; Jang, D.I.; Lee, T.H.; Lee, S.R. Redox Regulation of the Tumor Suppressor PTEN by Hydrogen Peroxide and Tert-Butyl Hydroperoxide. Int. J. Mol. Sci. 2017, 18. [Google Scholar] [CrossRef]
- Lee, J.T.; Shan, J.; Zhong, J.; Li, M.; Zhou, B.; Zhou, A.; Parsons, R.; Gu, W. RFP-mediated ubiquitination of PTEN modulates its effect on AKT activation. Cell Res. 2013, 23, 552–564. [Google Scholar] [CrossRef]
- Naguib, A.; Bencze, G.; Cho, H.; Zheng, W.; Tocilj, A.; Elkayam, E.; Faehnle, C.R.; Jaber, N.; Pratt, C.P.; Chen, M.; et al. PTEN functions by recruitment to cytoplasmic vesicles. Mol. Cell 2015, 58, 255–268. [Google Scholar] [CrossRef]
- Fotin, A.; Cheng, Y.; Grigorieff, N.; Walz, T.; Harrison, S.C.; Kirchhausen, T. Structure of an auxilin-bound clathrin coat and its implications for the mechanism of uncoating. Nature 2004, 432, 649–653. [Google Scholar] [CrossRef]
- Shenoy, S.S.; Nanda, H.; Losche, M. Membrane association of the PTEN tumor suppressor: Electrostatic interaction with phosphatidylserine-containing bilayers and regulatory role of the C-terminal tail. J. Struct. Biol. 2012, 180, 394–408. [Google Scholar] [CrossRef] [Green Version]
- Chen, Z.; Dempsey, D.R.; Thomas, S.N.; Hayward, D.; Bolduc, D.M.; Cole, P.A. Molecular Features of Phosphatase and Tensin Homolog (PTEN) Regulation by C-terminal Phosphorylation. J. Biol. Chem. 2016, 291, 14160–14169. [Google Scholar] [CrossRef] [Green Version]
- Yasui, M.; Matsuoka, S.; Ueda, M. PTEN hopping on the cell membrane is regulated via a positively-charged C2 domain. PLoS Comput. Biol. 2014, 10, e1003817. [Google Scholar] [CrossRef]
- Masson, G.R.; Perisic, O.; Burke, J.E.; Williams, R.L. The intrinsically disordered tails of PTEN and PTEN-L have distinct roles in regulating substrate specificity and membrane activity. Biochem. J. 2016, 473, 135–144. [Google Scholar] [CrossRef] [Green Version]
- Spinelli, L.; Black, F.M.; Berg, J.N.; Eickholt, B.J.; Leslie, N.R. Functionally distinct groups of inherited PTEN mutations in autism and tumour syndromes. J. Med. Genet. 2015, 52, 128–134. [Google Scholar] [CrossRef]
- Sun, Z.; Huang, C.; He, J.; Lamb, K.L.; Kang, X.; Gu, T.; Shen, W.H.; Yin, Y. PTEN C-terminal deletion causes genomic instability and tumor development. Cell Rep. 2014, 6, 844–854. [Google Scholar] [CrossRef]
- Wang, X.; Trotman, L.C.; Koppie, T.; Alimonti, A.; Chen, Z.; Gao, Z.; Wang, J.; Erdjument-Bromage, H.; Tempst, P.; Cordon-Cardo, C.; et al. NEDD4-1 is a proto-oncogenic ubiquitin ligase for PTEN. Cell 2007, 128, 129–139. [Google Scholar] [CrossRef]
- Correia, N.C.; Girio, A.; Antunes, I.; Martins, L.R.; Barata, J.T. The multiple layers of non-genetic regulation of PTEN tumour suppressor activity. Eur. J. Cancer 2014, 50, 216–225. [Google Scholar] [CrossRef]
- Hong, S.W.; Moon, J.H.; Kim, J.S.; Shin, J.S.; Jung, K.A.; Lee, W.K.; Jeong, S.Y.; Hwang, J.J.; Lee, S.J.; Suh, Y.A.; et al. p34 is a novel regulator of the oncogenic behavior of NEDD4-1 and PTEN. Cell Death Differ. 2014, 21, 146–160. [Google Scholar] [CrossRef]
- Zou, X.; Levy-Cohen, G.; Blank, M. Molecular functions of NEDD4 E3 ubiquitin ligases in cancer. Biochim. Biophys. Acta 2015, 1856, 91–106. [Google Scholar] [CrossRef]
- Cai, J.; Li, R.; Xu, X.; Zhang, L.; Lian, R.; Fang, L.; Huang, Y.; Feng, X.; Liu, X.; Li, X.; et al. CK1alpha suppresses lung tumour growth by stabilizing PTEN and inducing autophagy. Nat. Cell Biol. 2018, 20, 465–478. [Google Scholar] [CrossRef]
- Ahmed, D.; Eide, P.W.; Eilertsen, I.A.; Danielsen, S.A.; Eknaes, M.; Hektoen, M.; Lind, G.E.; Lothe, R.A. Epigenetic and genetic features of 24 colon cancer cell lines. Oncogenesis 2013, 2, e71. [Google Scholar] [CrossRef]
- Maddika, S.; Kavela, S.; Rani, N.; Palicharla, V.R.; Pokorny, J.L.; Sarkaria, J.N.; Chen, J. WWP2 is an E3 ubiquitin ligase for PTEN. Nat. Cell Biol. 2011, 13, 728–733. [Google Scholar] [CrossRef] [Green Version]
- Cao, J.; Wan, L.; Hacker, E.; Dai, X.; Lenna, S.; Jimenez-Cervantes, C.; Wang, Y.; Leslie, N.R.; Xu, G.X.; Widlund, H.R.; et al. MC1R is a potent regulator of PTEN after UV exposure in melanocytes. Mol. Cell 2013, 51, 409–422. [Google Scholar] [CrossRef]
- Clements, A.E.; Bravo, V.; Koivisto, C.; Cohn, D.E.; Leone, G. WWP2 and its association with PTEN in endometrial cancer. Gynecol. Oncol. Rep. 2015, 13, 26–29. [Google Scholar] [CrossRef]
- Liu, J.; Wan, L.; Liu, J.; Yuan, Z.; Zhang, J.; Guo, J.; Malumbres, M.; Liu, J.; Zou, W.; Wei, W. Cdh1 inhibits WWP2-mediated ubiquitination of PTEN to suppress tumorigenesis in an APC-independent manner. Cell Discov. 2016, 2, 15044. [Google Scholar] [CrossRef]
- Li, H.; Zhang, P.; Zhang, Q.; Li, C.; Zou, W.; Chang, Z.; Cui, C.P.; Zhang, L. WWP2 is a physiological ubiquitin ligase for phosphatase and tensin homolog (PTEN) in mice. J. Biol. Chem. 2018, 293, 8886–8899. [Google Scholar] [CrossRef] [Green Version]
- Yuan, L.L.; Green, A.S.; Bertoli, S.; Grimal, F.; Mansat-De Mas, V.; Dozier, C.; Tamburini, J.; Recher, C.; Didier, C.; Manenti, S. Pim kinases phosphorylate Chk1 and regulate its functions in acute myeloid leukemia. Leukemia 2014, 28, 293–301. [Google Scholar] [CrossRef]
- Park, M.K.; Yao, Y.; Xia, W.; Setijono, S.R.; Kim, J.H.; Vila, I.K.; Chiu, H.H.; Wu, Y.; Billalabeitia, E.G.; Lee, M.G.; et al. PTEN self-regulates through USP11 via the PI3K-FOXO pathway to stabilize tumor suppression. Nat. Commun. 2019, 10, 636. [Google Scholar] [CrossRef]
- Watt, J.E.; Hughes, G.R.; Walpole, S.; Monaco, S.; Stephenson, G.R.; Bulman Page, P.C.; Hemmings, A.M.; Angulo, J.; Chantry, A. Discovery of Small Molecule WWP2 Ubiquitin Ligase Inhibitors. Chemistry 2018, 24, 17677–17680. [Google Scholar] [CrossRef]
- Zhang, J.; Xie, J.J.; Zhou, S.J.; Chen, J.; Hu, Q.; Pu, J.X.; Lu, J.L. Diosgenin inhibits the expression of NEDD4 in prostate cancer cells. Am. J. Transl. Res. 2019, 11, 3461–3471. [Google Scholar]
- Lee, Y.R.; Chen, M.; Lee, J.D.; Zhang, J.; Lin, S.Y.; Fu, T.M.; Chen, H.; Ishikawa, T.; Chiang, S.Y.; Katon, J.; et al. Reactivation of PTEN tumor suppressor for cancer treatment through inhibition of a MYC-WWP1 inhibitory pathway. Science 2019, 364. [Google Scholar] [CrossRef]
- Leslie, N.R.; Longy, M. Inherited PTEN mutations and the prediction of phenotype. Semin. Cell Dev. Biol. 2016, 52, 30–38. [Google Scholar] [CrossRef] [Green Version]
- Gil, A.; Andres-Pons, A.; Fernandez, E.; Valiente, M.; Torres, J.; Cervera, J.; Pulido, R. Nuclear localization of PTEN by a Ran-dependent mechanism enhances apoptosis: Involvement of an N-terminal nuclear localization domain and multiple nuclear exclusion motifs. Mol. Biol. Cell 2006, 17, 4002–4013. [Google Scholar] [CrossRef]
- Van Themsche, C.; Leblanc, V.; Parent, S.; Asselin, E. X-linked inhibitor of apoptosis protein (XIAP) regulates PTEN ubiquitination, content, and compartmentalization. J. Biol. Chem. 2009, 284, 20462–20466. [Google Scholar] [CrossRef]
- Wang, W.; Chen, Y.; Wang, S.; Hu, N.; Cao, Z.; Wang, W.; Tong, T.; Zhang, X. PIASxalpha ligase enhances SUMO1 modification of PTEN protein as a SUMO E3 ligase. J. Biol. Chem. 2014, 289, 3217–3230. [Google Scholar] [CrossRef]
- Song, M.S.; Salmena, L.; Carracedo, A.; Egia, A.; Lo-Coco, F.; Teruya-Feldstein, J.; Pandolfi, P.P. The deubiquitinylation and localization of PTEN are regulated by a HAUSP-PML network. Nature 2008, 455, 813–817. [Google Scholar] [CrossRef]
- Minaguchi, T.; Waite, K.A.; Eng, C. Nuclear localization of PTEN is regulated by Ca(2+) through a tyrosil phosphorylation-independent conformational modification in major vault protein. Cancer Res. 2006, 66, 11677–11682. [Google Scholar] [CrossRef]
- Zhang, W.; Neo, S.P.; Gunaratne, J.; Poulsen, A.; Boping, L.; Ong, E.H.; Sangthongpitag, K.; Pendharkar, V.; Hill, J.; Cohen, S.M. Feedback regulation on PTEN/AKT pathway by the ER stress kinase PERK mediated by interaction with the Vault complex. Cell Signal. 2015, 27, 436–442. [Google Scholar] [CrossRef] [Green Version]
- Chen, M.; Nowak, D.G.; Narula, N.; Robinson, B.; Watrud, K.; Ambrico, A.; Herzka, T.M.; Zeeman, M.E.; Minderer, M.; Zheng, W.; et al. The nuclear transport receptor Importin-11 is a tumor suppressor that maintains PTEN protein. J. Cell Biol. 2017, 216, 641–656. [Google Scholar] [CrossRef] [Green Version]
- Howitt, J.; Lackovic, J.; Low, L.H.; Naguib, A.; Macintyre, A.; Goh, C.P.; Callaway, J.K.; Hammond, V.; Thomas, T.; Dixon, M.; et al. Ndfip1 regulates nuclear Pten import in vivo to promote neuronal survival following cerebral ischemia. J. Cell Biol. 2012, 196, 29–36. [Google Scholar] [CrossRef] [Green Version]
- Hou, B.; Xu, S.; Xu, Y.; Gao, Q.; Zhang, C.; Liu, L.; Yang, H.; Jiang, X.; Che, Y. Grb2 binds to PTEN and regulates its nuclear translocation to maintain the genomic stability in DNA damage response. Cell Death Dis. 2019, 10, 546. [Google Scholar] [CrossRef]
- Chen, J.H.; Zhang, P.; Chen, W.D.; Li, D.D.; Wu, X.Q.; Deng, R.; Jiao, L.; Li, X.; Ji, J.; Feng, G.K.; et al. ATM-mediated PTEN phosphorylation promotes PTEN nuclear translocation and autophagy in response to DNA-damaging agents in cancer cells. Autophagy 2015, 11, 239–252. [Google Scholar] [CrossRef] [Green Version]
- Yu, W.; He, X.; Ni, Y.; Ngeow, J.; Eng, C. Cowden syndrome-associated germline SDHD variants alter PTEN nuclear translocation through SRC-induced PTEN oxidation. Hum. Mol. Genet. 2015, 24, 142–153. [Google Scholar] [CrossRef]
- Li, Y.; Low, L.H.; Putz, U.; Goh, C.P.; Tan, S.S.; Howitt, J. Rab5 and Ndfip1 are involved in Pten ubiquitination and nuclear trafficking. Traffic 2014, 15, 749–761. [Google Scholar] [CrossRef]
- Kavela, S.; Shinde, S.R.; Ratheesh, R.; Viswakalyan, K.; Bashyam, M.D.; Gowrishankar, S.; Vamsy, M.; Pattnaik, S.; Rao, S.; Sastry, R.A.; et al. PNUTS functions as a proto-oncogene by sequestering PTEN. Cancer Res. 2013, 73, 205–214. [Google Scholar] [CrossRef]
- Liu, J.L.; Mao, Z.; Gallick, G.E.; Yung, W.K. AMPK/TSC2/mTOR-signaling intermediates are not necessary for LKB1-mediated nuclear retention of PTEN tumor suppressor. Neuro Oncol. 2011, 13, 184–194. [Google Scholar] [CrossRef]
- Chang, C.J.; Mulholland, D.J.; Valamehr, B.; Mosessian, S.; Sellers, W.R.; Wu, H. PTEN nuclear localization is regulated by oxidative stress and mediates p53-dependent tumor suppression. Mol. Cell Biol. 2008, 28, 3281–3289. [Google Scholar] [CrossRef]
- Beckham, T.H.; Cheng, J.C.; Lu, P.; Marrison, S.T.; Norris, J.S.; Liu, X. Acid ceramidase promotes nuclear export of PTEN through sphingosine 1-phosphate mediated Akt signaling. PLoS ONE 2013, 8, e76593. [Google Scholar] [CrossRef]
- Wu, Y.; Zhou, H.; Wu, K.; Lee, S.; Li, R.; Liu, X. PTEN phosphorylation and nuclear export mediate free fatty acid-induced oxidative stress. Antioxid. Redox Signal. 2014, 20, 1382–1395. [Google Scholar] [CrossRef]
- Leonard, M.K.; Hill, N.T.; Grant, E.D.; Kadakia, M.P. DeltaNp63alpha represses nuclear translocation of PTEN by inhibition of NEDD4-1 in keratinocytes. Arch. Derm. Res. 2013, 305, 733–739. [Google Scholar] [CrossRef]
- Leonard, M.K.; Kommagani, R.; Payal, V.; Mayo, L.D.; Shamma, H.N.; Kadakia, M.P. DeltaNp63alpha regulates keratinocyte proliferation by controlling PTEN expression and localization. Cell Death Differ. 2011, 18, 1924–1933. [Google Scholar] [CrossRef]
- Noguera, N.I.; Song, M.S.; Divona, M.; Catalano, G.; Calvo, K.L.; Garcia, F.; Ottone, T.; Florenzano, F.; Faraoni, I.; Battistini, L.; et al. Nucleophosmin/B26 regulates PTEN through interaction with HAUSP in acute myeloid leukemia. Leukemia 2013, 27, 1037–1043. [Google Scholar] [CrossRef]
- Lobo, G.P.; Waite, K.A.; Planchon, S.M.; Romigh, T.; Houghton, J.A.; Eng, C. ATP modulates PTEN subcellular localization in multiple cancer cell lines. Hum. Mol. Genet. 2008, 17, 2877–2885. [Google Scholar] [CrossRef] [Green Version]
- Yang, J.M.; Schiapparelli, P.; Nguyen, H.N.; Igarashi, A.; Zhang, Q.; Abbadi, S.; Amzel, L.M.; Sesaki, H.; Quinones-Hinojosa, A.; Iijima, M. Characterization of PTEN mutations in brain cancer reveals that pten mono-ubiquitination promotes protein stability and nuclear localization. Oncogene 2017, 36, 3673–3685. [Google Scholar] [CrossRef]
- Morotti, A.; Panuzzo, C.; Crivellaro, S.; Pergolizzi, B.; Familiari, U.; Berger, A.H.; Saglio, G.; Pandolfi, P.P. BCR-ABL disrupts PTEN nuclear-cytoplasmic shuttling through phosphorylation-dependent activation of HAUSP. Leukemia 2014, 28, 1326–1333. [Google Scholar] [CrossRef]
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Liu, T.; Wang, Y.; Wang, Y.; Chan, A.M. Multifaceted Regulation of PTEN Subcellular Distributions and Biological Functions. Cancers 2019, 11, 1247. https://doi.org/10.3390/cancers11091247
Liu T, Wang Y, Wang Y, Chan AM. Multifaceted Regulation of PTEN Subcellular Distributions and Biological Functions. Cancers. 2019; 11(9):1247. https://doi.org/10.3390/cancers11091247
Chicago/Turabian StyleLiu, Tian, Yiwei Wang, Yubing Wang, and Andrew M. Chan. 2019. "Multifaceted Regulation of PTEN Subcellular Distributions and Biological Functions" Cancers 11, no. 9: 1247. https://doi.org/10.3390/cancers11091247
APA StyleLiu, T., Wang, Y., Wang, Y., & Chan, A. M. (2019). Multifaceted Regulation of PTEN Subcellular Distributions and Biological Functions. Cancers, 11(9), 1247. https://doi.org/10.3390/cancers11091247