Elsevier

Seminars in Cancer Biology

Volume 59, December 2019, Pages 66-79
Seminars in Cancer Biology

Review
Mechanisms of PTEN loss in cancer: It’s all about diversity

https://doi.org/10.1016/j.semcancer.2019.02.001Get rights and content

Abstract

PTEN is a phosphatase which metabolises PIP3, the lipid product of PI 3-Kinase, directly opposing the activation of the oncogenic PI3K/AKT/mTOR signalling network. Accordingly, loss of function of the PTEN tumour suppressor is one of the most common events observed in many types of cancer. Although the mechanisms by which PTEN function is disrupted are diverse, the most frequently observed events are deletion of a single gene copy of PTEN and gene silencing, usually observed in tumours with little or no PTEN protein detectable by immunohistochemistry. Accordingly, with the exceptions of glioblastoma and endometrial cancer, mutations of the PTEN coding sequence are uncommon (<10%) in most types of cancer. Here we review the data relating to PTEN loss in seven common tumour types and discuss mechanisms of PTEN regulation, some of which appear to contribute to reduced PTEN protein levels in cancers.

Introduction

PTEN is a ubiquitously expressed tumour suppressor that is commonly inactivated in human sporadic cancers. It was firstly identified in 1997 by two independent research groups while studying the chromosomal location 10q23, which appeared to show frequent deletion in tumours of the brain, prostate and bladder [1,2]. Shortly thereafter, PTEN mutations were also found in the germline of patients with a group of autosomal dominant syndromes, termed collectively as PTEN hamartoma tumour syndromes (PHTS) that are characterized by the presence of multiple hamartomas, an increased cancer predisposition and neurological symptoms. The role of PTEN as a tumour suppressor has been extensively studied since its initial discovery [3,4] and it was identified as the locus with greatest selection pressure for deletion in an analysis of 746 human cancer genomes [5].

In many cases, PTEN appears to be a haploinsufficient tumour suppressor. In contrast to classical tumour suppressor models that need a complete inactivation to induce cancer and were based upon studies of the Retinoblastoma gene, [6] partial loss of PTEN function can have a dramatic impact on tumorigenesis and cancer progression. Studies using hypomorphic mouse models expressing reduced PTEN levels have shown that even subtle reductions in PTEN expression can significantly increase cancer susceptibility [[7], [8], [9]]. This is also in accordance with evidence that the diverse mechanisms controlling PTEN stability and function can have substantial impacts on cancer development [[10], [11], [12], [13], [14]] and with the observation that loss of one copy of PTEN is far more common than mutation or deletion of both copies (Tables 1 and S1).

PTEN is a major negative regulator of the signalling pathway defined by class I phosphoinositide 3 kinase (PI3K), AKT and the mechanistic target of rapamycin (mTOR) and which plays a key role controlling a wide range of essential cellular processes including cell proliferation, growth, survival and metabolism [[15], [16], [17]]. The PI3K-AKT-mTOR signalling pathway is evolutionarily conserved within metazoans although the linked functions of the class I PI3Ks and PTEN appear to have evolved earlier as regulators of cell polarity and membrane remodelling [18].

The activation of intracellular class I PI3Ks is caused by diverse cell surface receptors which promote cell growth and proliferation, including many growth factor-activated members of the receptor tyrosine kinases (RTK) cytokine receptors, some integrins and a subset of G-protein coupled receptors which includes several chemokine receptors [19]. These activated receptors directly or indirectly recruit and activate class I PI3K which in turn phosphorylates a small fraction of plasma membrane phosphatidylinositol-4,5-bisphosphate (PIP2) to generate phosphatidylinositol-3,4,5-trisphosphate (PIP3), a membrane-associated lipid that acts as a second messenger driving downstream signalling (Fig. 1). Increases in local PIP3 levels facilitate the binding of a large number of proteins that carry selective PIP3-binding domains which in turn promote the effects of pathway activation on cell metabolism, growth, proliferation, etc [[20], [21], [22]]. The best studied of these PIP3-binding effector proteins are the AKT protein kinases, which have a large and diverse range of substrates and are important proto-oncogenes in their own right. However, other proteins directly regulated by PIP3 binding include the BTK/TEC family of tyrosine kinases and several regulators of small GTPase of the ARF and RHO families [19,22].

PTEN’s role within the pathway is as a lipid phosphatase, directly opposing the activation of the PI3K signalling by converting the PIP3 generated by PI3K back to PIP2. Loss of PTEN results in the lack of regulation of PIP3 levels which in turn promote the hyper-activation of the pathway thus leading to cellular transformation and tumorigenesis, as observed in studies with PTEN-null tumour cell lines, immortalized fibroblasts and tumours arising in PTEN-deficient mice [3,23,24]. The PI3K/AKT/mTOR signalling axis is one of the most frequently deregulated pathways in cancer with mutations occurring in most of the major components of the network [15,[25], [26], [27]]. Therefore, targeting the pathway has become an attractive strategy for cancer therapy and this has led to the development of numerous compounds designed to counteract activated PI3K signalling, although to date clinical success has been limited to the approval of the PI3K delta inhibitor idelalisib for the treatment of B cell malignancies [15].

Even though its main biological activity relies on its ability to dephosphorylate lipid substrates, PTEN has also been reported to display phosphatase activity against tyrosine, serine and threonine residues towards protein substrates in vitro as well as on itself [28] although the biological significance of these functions is still controversial. In addition, PTEN has been proposed to exert some of its biological functions in a catalysis-independent manner through protein-protein interactions [29,30]. Both phosphatase-dependent and independent functions appear related to PTEN’s subcellular localization: interestingly, loss of the nuclear pool of PTEN seems to correlate with cancer progression and poor clinical outcome in certain types of tumours thus highlighting the importance of its nuclear localisation [13,31,32]. However, the molecular mechanisms through which PTEN exerts its tumour-suppressor functions in the nucleus and its biological relevance still remain unclear.

The PTEN gene is located on chromosome 10q23 and its 9 exons encode a predominant protein product of 403 amino acids and 48 kDa that shares sequence homology with the tyrosine phosphatase superfamily as well as with tensin and auxilin. Therefore it was named Phosphate and Tensin Homolog deleted from chromosome ten (PTEN) when first discovered [1]. The protein sequence is highly conserved within vertebrates with only one amino acid difference between the human and murine orthologs. The first analysis of a crystal structure of human PTEN revealed the existence of 2 tightly associated domains: a catalytic N-terminal phosphatase domain (amino acids 6–185) and a C2 domain required for membrane binding (amino acids 186–351) [33]. The protein also includes an extreme N-terminal PtdIns(4,5)P2 binding sequence (amino acids 6–15) that enables the interaction with substrate-containing membrane surfaces and cytoplasmic and nuclear localization signals (amino acids 19–25) that helps dictate its subcellular localization [[34], [35], [36]]. Furthermore, the C-terminal portion of PTEN has a less-structured C-terminal tail (amino acids 352–403) that contributes to the post-translational regulation of the PTEN protein, containing two clusters of phosphorylation sites and a PDZ binding sequence [33].

In addition to the most abundant 403 amino acid form of PTEN, many cells contain lower levels of N-terminally extended isoforms of the enzyme [37,38]. The first of these to be discovered, PTEN-L, has an additional 173 amino acid N-terminal region translated from an alternate upstream start codon which notably includes a signal peptide. This leads to the secretion of PTEN-L and suggested the hypothesis that active PTEN-L protein may be shared between cells to suppress PI3K/AKT signalling [39]. However, the functions of these longer forms of PTEN remains somewhat mysterious.

Section snippets

Changes in PTEN activity in health and disease

Given the biological importance of its functions and the profound pathological effects that arise as a consequence of subtle disruptions on its expression and activity, it is unsurprising that PTEN levels are tightly regulated through multiple physiological mechanisms [40,41]. These mechanisms of PTEN regulation act at transcriptional, post-transcriptional and post-translational levels. Significantly, these physiological mechanisms appear to be subverted in some cancers to suppress PTEN

Tumour type-specific patterns of PTEN loss

Stark changes in PTEN activity are seen in tumours that display changes in the PTEN gene, mutating or deleting the gene in many cases leading to complete loss of activity or of expression. PTEN shows different patterns of loss in different tumour types and these are discussed in individual sections below and illustrated in Table 1 and Table S1. It should be noted that despite work to compare antibodies and advise best practice [108], sources of uncertainty within datasets analysing PTEN loss

Conclusion

The spectrum of changes and processes which contribute to the loss of PTEN function in different types of tumours is diverse and as discussed reveals some specific patterns. In many cases, the source and consequence of these patterns are unclear. It seems likely that some may be driven by mutational processes, such as tumour cells which are deficient in mismatch repair, identifiable by microsatellite instability. However, other patterns of PTEN loss seem likely to be driven by selection,

Acknowledgements and conflict of interest statement

The authors declare they have no conflict of interest with publication of this manuscript. Work in the NRL laboratory is funded by Medical Research Scotland (Grant number: 1034-2016), Prostate Cancer UK (PG14-006), CRUK (C50604/A28050) and PTEN Research.

References (223)

  • W.S. Park et al.

    Comprehensive identification of PIP3-regulated PH domains from C. elegans to H. sapiens by model prediction and live imaging

    Mol. Cell

    (2008)
  • L. Mahimainathan et al.

    Inactivation of platelet-derived growth factor receptor by the tumor suppressor PTEN provides a novel mechanism of action of the phosphatase

    J. Biol. Chem.

    (2004)
  • O. Gimm et al.

    Differential nuclear and cytoplasmic expression of PTEN in normal thyroid tissue, and benign and malignant epithelial thyroid tumors

    Am. J. Pathol.

    (2000)
  • A. Perren et al.

    Mutation and expression analyses reveal differential subcellular compartmentalization of PTEN in endocrine pancreatic tumors compared to normal islet cells

    Am. J. Pathol.

    (2000)
  • J.-O. Lee et al.

    Crystal structure of the PTEN tumor suppressor: implications for its phosphoinositide phosphatase activity and membrane association

    Cell

    (1999)
  • T.L. Mighell et al.

    A saturation mutagenesis approach to understanding PTEN lipid phosphatase activity and genotype-phenotype relationships

    Am. J. Hum. Genet.

    (2018)
  • L. Patel et al.

    Tumor suppressor and anti-inflammatory actions of PPARgamma agonists are mediated via upregulation of PTEN

    Curr. Biol.

    (2001)
  • V. Stambolic et al.

    Regulation of PTEN transcription by p53

    Mol. Cell

    (2001)
  • Y.-H. Kang et al.

    Promoter methylation and silencing of PTEN in gastric carcinoma

    Lab. Investig.

    (2002)
  • G. Piras et al.

    Methylation of the PTEN promoter leads to PTEN inactivation in multiple myeloma

    Blood

    (2008)
  • M.A. Zysman et al.

    Considerations when analyzing the methylation status of PTEN tumor suppressor gene

    Am. J. Pathol.

    (2002)
  • F. Meng et al.

    MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer

    Gastroenterology

    (2007)
  • J. Zhang et al.

    MicroRNA-21 (miR-21) represses tumor suppressor PTEN and promotes growth and invasion in non-small cell lung cancer (NSCLC)

    Clin. Chim. Acta

    (2010)
  • J. Torres et al.

    The tumor suppressor PTEN is phosphorylated by the protein kinase CK2 at its C terminus implications for pten stability to proteasome-mediated degradation

    J. Biol. Chem.

    (2001)
  • A.M. Al-Khouri et al.

    Cooperative phosphorylation of the tumor suppressor phosphatase and tensin homologue (PTEN) by casein kinases and glycogen synthase kinase 3beta

    J. Biol. Chem.

    (2005)
  • R. Fragoso et al.

    Kinases, tails and more: regulation of PTEN function by phosphorylation

    Methods

    (2015)
  • H. Maccario et al.

    Ubiquitination of PTEN (phosphatase and tensin homolog) inhibits phosphatase activity and is enhanced by membrane targeting and hyperosmotic stress

    J. Biol. Chem.

    (2010)
  • F. Vazquez et al.

    Phosphorylation of the PTEN tail acts as an inhibitory switch by preventing its recruitment into a protein complex

    J. Biol. Chem.

    (2001)
  • S.F. Ahmed et al.

    The chaperone-assisted E3 ligase C terminus of Hsc70-interacting protein (CHIP) targets PTEN for proteasomal degradation

    J. Biol. Chem.

    (2012)
  • C. Van Themsche et al.

    X-linked inhibitor of apoptosis protein (XIAP) regulates PTEN ubiquitination, content, and compartmentalization

    J. Biol. Chem.

    (2009)
  • S.-R. Lee et al.

    Reversible inactivation of the tumor suppressor PTEN by H2O2

    J. Biol. Chem.

    (2002)
  • J. Li et al.

    PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer

    Science

    (1997)
  • P.A. Steck et al.

    Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers

    Nat. Genet.

    (1997)
  • C.A. Worby et al.

    PTEN

    Annu. Rev. Biochem.

    (2014)
  • G.R. Bignell et al.

    Signatures of mutation and selection in the cancer genome

    Nature

    (2010)
  • A.G. Knudson

    Mutation and cancer: statistical study of retinoblastoma

    Proc. Natl. Acad. Sci. U. S. A.

    (1971)
  • A. Alimonti et al.

    Subtle variations in Pten dose determine cancer susceptibility

    Nat. Genet.

    (2010)
  • A. Carracedo et al.

    PTEN level in tumor suppression: how much is too little?

    Cancer Res.

    (2011)
  • L.C. Trotman et al.

    Pten dose dictates Cancer progression in the prostate

    PLoS Biol.

    (2003)
  • A. Silva et al.

    PTEN posttranslational inactivation and hyperactivation of the PI3K/Akt pathway sustain primary T cell leukemia viability

    J. Clin. Invest.

    (2008)
  • D.A. Fruman et al.

    PI3K and cancer: lessons, challenges and opportunities

    Nat. Rev. Drug Discov.

    (2014)
  • B. Vanhaesebroeck et al.

    PI3K signalling: the path to discovery and understanding

    Nat. Rev. Mol. Cell Biol.

    (2012)
  • L.C. Cantley et al.

    New insights into tumor suppression: PTEN suppresses tumor formation by restraining the phosphoinositide 3-kinase/AKT pathway

    Proc. Natl. Acad. Sci. U. S. A.

    (1999)
  • D. Freeman et al.

    Genetic background controls tumor development in PTEN-deficient mice

    Cancer Res.

    (2006)
  • V.W.Y. Lui et al.

    Frequent mutation of the PI3K pathway in head and neck cancer defines predictive biomarkers

    Cancer Discov.

    (2013)
  • T.L. Yuan et al.

    PI3K pathway alterations in cancer: variations on a theme

    Oncogene

    (2008)
  • Y. Samuels et al.

    Oncogenic PI3K and its role in cancer

    Curr. Opin. Oncol.

    (2006)
  • E. Lima-Fernandes et al.

    Distinct functional outputs of PTEN signalling are controlled by dynamic association with β-arrestins

    EMBO J.

    (2011)
  • H. Serra et al.

    PTEN mediates Notch-dependent stalk cell arrest in angiogenesis

    Nat. Commun.

    (2015)
  • G. Denning et al.

    A short N-terminal sequence of PTEN controls cytoplasmic localization and is required for suppression of cell growth

    Oncogene

    (2007)
  • Cited by (228)

    View all citing articles on Scopus
    1

    Present address: Cancer Research UK Edinburgh Centre, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XR, UK.

    View full text