Elsevier

Drug Discovery Today

Volume 26, Issue 11, November 2021, Pages 2680-2698
Drug Discovery Today

Keynote (green)
Modulation of serine/threonine-protein phosphatase 1 (PP1) complexes: A promising approach in cancer treatment

https://doi.org/10.1016/j.drudis.2021.08.001Get rights and content

Highlights

  • The development of new and optimized anticancer therapies is an urgent requirement.

  • Serine/threonine-protein phosphatase 1 (PP1) has been recognized as a potential drug target in cancer.

  • Several PP1 complexes have been characterized in cancer models.

  • The disruption or stabilization of PP1 complexes offer advantages in cancer treatment.

Abstract

Cancer is the second leading cause of death worldwide. Despite the availability of numerous therapeutic options, tumor heterogeneity and chemoresistance have limited the success of these treatments, and the development of effective anticancer therapies remains a major focus in oncology research. The serine/threonine-protein phosphatase 1 (PP1) and its complexes have been recognized as potential drug targets. Research on the modulation of PP1 complexes is currently at an early stage, but has immense potential. Chemically diverse compounds have been developed to disrupt or stabilize different PP1 complexes in various cancer types, with the objective of inhibiting disease progression. Beneficial results obtained in vitro now require further pre-clinical and clinical validation. In conclusion, the modulation of PP1 complexes seems to be a promising, albeit challenging, therapeutic strategy for cancer.

Introduction

According to the World Health Organization (WHO), cancer is considered a major public health concern, estimated to be the second leading cause of death worldwide. The incidence of cancer has increased latterly, with a total of 18.1 million new cases and 9.6 million deaths reported globally in 2018.1 At present, therapy decisions are dictated by cancer type and clinical staging. Options include both localized therapies, including surgery or radiation therapy, and systemic therapies that encompass chemotherapy and hormonal or immune interventions. The success of conventional therapies is essentially limited by tumor heterogeneity and their acquired resistance to therapy.2 In the past decade, important advances have been provided by increasingly detailed knowledge of the molecular biology of tumors, coupled with the emergence of new approaches for cancer treatment such as more personalized cancer medicine. Nevertheless, serious challenges remain and the establishment of improved therapies is urgently needed.2

Interventions that target the post-translational phosphorylation of intracellular proteins have been considered as viable anticancer therapies. Transient phosphorylation events control most cellular signaling processes, and abnormal phosphorylation profiles have been associated with several pathological conditions, including cancers.3 The phosphoproteome is formed by the activities of both protein kinases and phosphatases, which add or remove phosphate groups, respectively. The balance between the activities of these two types of enzyme is essential to maintain cellular homeostasis.4 Thus, the targeting of both protein kinases and protein phosphatases has been proposed for cancer treatment.5., 6.

Serine/threonine-protein phosphatase 1 (PP1) is a major protein phosphatase, which catalyzes a wide range of protein dephosphorylation reactions in human cells.7 It regulates critical cellular processes including cell cycle progression, apoptosis and metabolism.8 The involvement of PP1 in several oncogenic pathways has become evident, and its expression level seems to be altered in the presence of a tumor.9 Nevertheless, the direction in which PP1 expression levels are altered is not clear as contradictory results have been published. Importantly, PP1 deregulation seems to depend on the type of cancer, on the interacting proteins and on the PP1 isoform.10., 11., 12., 13. Indeed, the catalytic subunit of PP1 (PP1c) is encoded by three genes (PPP1CA, PPP1CB and PPP1CC), which give rise to three different isoforms (PP1-alpha catalytic subunit (PP1α), PP1-beta catalytic subunit (PP1β) and PP1-gamma catalytic subunit (PP1ϒ)) that are ubiquitously expressed and differ mainly in their extremities.14 The roles of PP1 depend on the interaction of PP1c with different regulatory interactors of PP1 (RIPPOs)15 (previously called PP1-interacting proteins (PIPs)), which can act as targeting subunits, substrates, activity regulators, or through a combination of these roles. A determined effort over several decades has identified the PP1c interactome in different tissues and specific biological contexts, including pathological conditions.16., 17., 18., 19. Despite the relatively high number of PP1 complexes identified in human tissues, the highly dynamic nature of these complexes has clearly hampered their functional characterization.20

Targeting of PP1 has been considered for the treatment of several other diseases, including heart failure21 and neurological conditions.22 Compared with conventional chemotherapies, interventions that modulate discrete PP1 complexes could provide a more specific option with reduced cytotoxicity. In fact, this novel approach has been proposed for the treatment of various pathologies,23., 24., 25. including cancer.

In this context, we rigorously reviewed the potential of modulating PP1 complexes in cancer treatment. Herein, we summarize the PP1 complexes characterized in different types of cancer, highlighting their roles as tumor promoters or suppressors. The PP1 complexes that are modulated by either small molecules or peptides in cancer are also described. Finally, we define the main conclusions that can be drawn from the studies, and the principal challenges to be addressed by future work in this topic.

Section snippets

PP1 complexes in cancer: tumor promoters or suppressors?

The interaction of PP1c with its regulatory interactors plays important roles in key oncogenic pathways. Furthermore, the dysregulation of some PP1 complexes has been associated with cancer initiation and/or progression.26 Contradictory roles have been attributed to different PP1 complexes in cancer. Indeed, some are considered tumor promoters, whereas others are associated with a tumor suppressor activity. The tumor promoter/suppressor activity of a PP1 holoenzyme seems to depend principally

Targeting PP1 complexes

Targeted interference with protein phosphorylation mechanisms has long been considered a potential approach in the treatment of several diseases, including cancer. Of the various enzymes that are intimately involved in such events, protein kinases emerged as the first generic target for anticancer therapies.88 Despite the treatment resistance associated with various kinase inhibitors, and the widely variable therapeutic responses observed across patients, several kinase inhibitors have been

Conclusions and future challenges

Although it is not yet a well-studied and established topic, the modulation of PP1 complexes has recently emerged as a promising strategy for cancer treatment. Limited knowledge of the specific roles of PP1 complexes in tumorigenesis, and the molecular characterization of only a small proportion of all PP1 complexes, have necessarily limited this approach. Indeed, to the best of our knowledge, only 38 different PP1 complexes have been functionally characterized in cancer models, and even then,

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

We thank the Portuguese Foundation for Science and Technology (FCT) and the European Union (QREN, FEDER and COMPETE frameworks) for funding both iBiMED (UID/BIM/04501/2020, POCI-01-0145-FEDER-007628 and UID/BIM/04501/2019, respectively) and an individual scholarship for BM (SFRH/BD/146032/2019).

Bárbara Matos received a BSc in Biotechnology in 2016 and an MSc in Clinical Biochemistry in 2018 from the University of Aveiro, Portugal. At present, she is a PhD student in the doctoral program of Biomedicine, University of Aveiro. The main goal of her project is to establish an efficient strategy to disrupt key PP1 complexes in prostate carcinogenesis, with the ultimate objective of impairing the progression of prostate cancer.

References (196)

  • D. Paul et al.

    Protein Phosphatase 1 regulatory subunit SDS22 inhibits breast cancer cell tumorigenesis by functioning as a negative regulator

    Neoplasia

    (2019)
  • Christian Hansen et al.

    Wnt-5a-induced phosphorylation of DARPP-32 inhibits breast cancer cell migration in a CREB-dependent manner

    J Biol Chem

    (2009)
  • E.H. Hall et al.

    Tensin1 requires protein phosphatase-1alpha in addition to RhoGAP DLC-1 to control cell polarization, migration, and invasion

    J Biol Chem

    (2009)
  • Emily H. Hall et al.

    Comprehensive analysis of phosphorylation sites in Tensin1 reveals regulation by p38MAPK

    Mol Cell Proteomics

    (2010)
  • Chao Li et al.

    Essential phosphatases and a phospho-degron are critical for regulation of SRC-3/AIB1 coactivator function and turnover

    Mol Cell

    (2008)
  • Nae-Fang Twu et al.

    Expression of Aurora kinase A and B in normal and malignant cervical tissue: high Aurora A kinase expression in squamous cervical cancer

    Eur J Obstet Gynecol Reprod Biol

    (2009)
  • Hiroshi Katayama et al.

    Interaction and feedback regulation between STK15/BTAK/Aurora-A kinase and protein phosphatase 1 through mitotic cell division cycle

    J Biol Chem

    (2001)
  • Qiyan Zeng et al.

    IPP5, a novel inhibitor of protein phosphatase 1, suppresses tumor growth and progression of cervical carcinoma cells by inducing G2/M arrest

    Cancer Genet

    (2012)
  • Huihui Li et al.

    Antagonistic effects of p53 and HIF1A on microRNA-34a regulation of PPP1R11 and STAT3 and hypoxia-induced epithelial to mesenchymal transition in colorectal cancer cells

    Gastroenterology

    (2017)
  • Rebecca S. Henkhaus et al.

    Caveolin-1-mediated expression and secretion of kallikrein 6 in colon cancer cells

    Neoplasia

    (2008)
  • L. He et al.

    Protease-activated receptor 2 signaling modulates susceptibility of colonic epithelium to injury through stabilization of YAP in vivo

    Cell Death Dis

    (2018)
  • James A. McCubrey et al.

    Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance

    Biochim Biophys Acta

    (2007)
  • M. Chen et al.

    Deregulated PP1α phosphatase activity towards suppressive failsafe mechanism

    Nat Commun

    (2018)
  • Shaoyong Chen et al.

    Androgen receptor phosphorylation and activity are regulated by an association with protein phosphatase 1

    J Biol Chem

    (2009)
  • Pablo Rodriguez-Viciana et al.

    A phosphatase holoenzyme comprised of Shoc2/Sur8 and the catalytic subunit of PP1 functions as an M-Ras effector to modulate Raf activity

    Mol Cell

    (2006)
  • Masumi Eto et al.

    Nuclear localization of CPI-17, a protein phosphatase-1 inhibitor protein, affects histone H3 phosphorylation and corresponds to proliferation of cancer and smooth muscle cells

    Biochem Biophys Res Commun

    (2013)
  • Matthew S. Kelker et al.

    Crystal structures of protein phosphatase-1 bound to nodularin-R and tautomycin: a novel scaffold for structure-based drug design of serine/threonine phosphatase inhibitors

    J Mol Biol

    (2009)
  • Paola Vagnarelli et al.

    Previews PP1 phosphatase complexes: undruggable no longer

    Cell

    (2018)
  • Mathieu Bollen

    Combinatorial control of protein phosphatase-1

    Trends Biochem Sci

    (2001)
  • Annick Hendrickx et al.

    Docking motif-guided mapping of the interactome of protein phosphatase-1

    Chem Biol

    (2009)
  • Thomas D. Hurley et al.

    Structural basis for regulation of protein phosphatase 1 by inhibitor-2

    J Biol Chem

    (2007)
  • Heike Meiselbach et al.

    Structural analysis of the protein phosphatase 1 docking motif: molecular description of binding specificities identifies interacting proteins

    Chem Biol

    (2006)
  • J. Ferlay et al.

    Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods

    Int J Cancer

    (2019)
  • V. Singh et al.

    Phosphorylation: implications in cancer

    Protein J

    (2017)
  • A. Bononi et al.

    Protein kinases and phosphatases in the control of cell fate

    Enzyme Res

    (2011)
  • K.S. Bhullar et al.

    Kinase-targeted cancer therapies: progress, challenges and future directions

    Mol Cancer

    (2018)
  • L. Scott et al.

    Targeting protein tyrosine phosphatases for anticancer drug discovery

    Curr Pharm Des

    (2010)
  • Hugo Ceulemans et al.

    Functional diversity of protein phosphatase-1, a cellular economizer and reset button

    Physiol Rev

    (2004)
  • L.C. Hsu et al.

    Gene amplification and overexpression of protein phosphatase 1alpha in oral squamous cell carcinoma cell lines

    Oncogene

    (2006)
  • K. Sasaki et al.

    Identification of members of the protein phosphatase 1 gene family in the rat enhanced expression of protein phosphatase 1 alpha gene in rat hepatocellular carcinoma

    Jpn J Cancer Res

    (1990)
  • S.L. Winter et al.

    The interaction of PP1 with BRCA1 and analysis of their expression in breast tumors

    BMC Cancer

    (2007)
  • I. Prowatke et al.

    Expression analysis of imbalanced genes in prostate carcinoma using tissue microarrays

    Br J Cancer

    (2007)
  • M. Köhn

    Turn and face the strange: a new view on phosphatases

    ACS Cent Sci

    (2020)
  • S.L.C. Esteves et al.

    Protein phosphatase 1α interacting proteins in the human brain

    OMICS

    (2012)
  • L. Trinkle-Mulcahy et al.

    Dynamic targeting of protein phosphatase 1 within the nuclei of living mammalian cells

    J Cell Sci

    (2001)
  • Y. Miyazaki et al.

    Heart failure-inducible gene therapy targeting protein phosphatase 1 prevents progressive left ventricular remodeling

    PLoS One

    (2012)
  • Branco-Santos J, Herrera F, Poças G, Pires-Afonso Y, Giorgini F, Domingos PM, Outeiro TF. Protein phosphatase 1...
  • M. Fardilha et al.

    Protein phosphatase 1 complexes modulate sperm motility and present novel targets for male infertility

    Mol Hum Reprod

    (2011)
  • M. Jerebtsova et al.

    Inhibition of HIV-1 transcription through disruption of TAT-PP1 interaction as a novel target for functional HIV cure

    Nov Appro Drug Des Dev

    (2019)
  • X. Lin et al.

    Targeting the non-catalytic RVxF site of protein phosphatase-1 with small molecules for Ebola virus inhibition

    Front Microbiol

    (2019)
  • Cited by (10)

    View all citing articles on Scopus

    Bárbara Matos received a BSc in Biotechnology in 2016 and an MSc in Clinical Biochemistry in 2018 from the University of Aveiro, Portugal. At present, she is a PhD student in the doctoral program of Biomedicine, University of Aveiro. The main goal of her project is to establish an efficient strategy to disrupt key PP1 complexes in prostate carcinogenesis, with the ultimate objective of impairing the progression of prostate cancer.

    John Howl obtained his BSc in Biological Sciences in 1984 and his PhD in Molecular Pathology in 1988 from the University of Birmingham, UK. At present, he is Professor of Molecular Pharmacology at the University of Wolverhampton and the coordinating member of the Molecular Pharmacology research group. His research focuses on exploring the design, microwave-enhanced synthesis and biomedical applications of cell-penetrating peptides and bioportides.

    Carmen Jeronimo obtained her BSc in Biology (1994), MSc in Oncology (1998) and PhD in Biomedical Sciences (2001) from the University of Porto, Portugal. She is Invited Full Professor in the Department of Pathology and Molecular Genetics in the University of Porto and the group leader of the Cancer Biology and Epigenetics group at the Portuguese Oncology Institute of Porto (IPO-Porto), Portugal. Her research interests are focused on characterizing the epigenome of tumor cells, and on identifying functional changes that are involved in the breakdown of epigenetic homeostasis in these cells.

    Margarida Fardilha received her BSc in Biochemistry in 1996 from the University of Porto and her PhD in Biology in 2004 from the University of Aveiro, Portugal. She is currently an Assistant Professor with the habilitation qualification at the Department of Medical Sciences, University of Aveiro. She is also coordinator of the Signal Transduction Laboratory at the Institute for Biomedicine (iBiMED), University of Aveiro. Her main topics of research are related to the role of protein phosphatases in male-related disorders.

    View full text