Review
Smurf E3 ubiquitin ligases at the cross roads of oncogenesis and tumor suppression

https://doi.org/10.1016/j.bbcan.2012.11.003Get rights and content

Abstract

Smad ubiquitin regulatory factors (Smurfs) belong to the HECT- family of E3 ubiquitin ligases and comprise mainly of two members, Smurf1 and Smurf2. Initially, Smurfs have been implicated in determining the competence of cells to respond to TGF-β/BMP signaling pathway. Nevertheless, the intrinsic catalytic activity has extended the repertoire of Smurf substrates beyond the TGF-β/BMP super family expanding its realm further to epigenetic modifications of histones governing the chromatin landscape. Through regulation of a large number of proteins in multiple cellular compartments, Smurfs regulate diverse cellular processes, including cell-cycle progression, cell proliferation, differentiation, DNA damage response, maintenance of genomic stability, and metastasis. As the genomic ablation of Smurfs leads to global changes in histone modifications and predisposition to a wide spectrum of tumors, Smurfs are also considered to have a novel tumor suppressor function. This review focuses on regulation network and biological functions of Smurfs in connection with its role in cancer progression. By providing a portrait of their protein targets, we intend to link the substrate specificity of Smurfs with their contribution to tumorigenesis. Since the regulation and biological functions of Smurfs are quite complex, understanding the oncogenic potential of these E3 ubiquitin ligases may facilitate the development of mechanism-based drugs in cancer treatment.

Introduction

Protein ubiquitination, a post translational modification, mediated by the ubiquitin–proteasome system (UPS) plays an essential regulatory role in critical cellular processes including cell cycle, cell proliferation, differentiation, angiogenesis, stem cell quiescence, DNA damage repair, and apoptosis. Clearly, a process that is so critical for cellular homeostasis is itself tightly regulated by multiple enzymatic steps catalyzed by ubiquitin-activating enzyme E1, ubiquitin-conjugating enzyme E2, and ubiquitin ligase E3, and many accessory proteins that provide specificity to the system. Specifically, E3 ubiquitin ligases (E3s) play an indispensable role in this cascade by recruiting ubiquitin-loaded E2s, recognizing specific substrates, and facilitating or directly catalyzing ubiquitin transfer to the respective molecular targets. The relevance of the E3s in several biological processes is emphasized in vivo by the observation that their genetic alteration, abnormal expression, or dysfunction is often accompanied by the occurrence of pathological disorders, including cancer. Recently various HECT E3 ubiquitin ligases have emerged as crucial regulators of cancer development. Indeed, in view of their substrate specificity, the E3s represent potentially attractive targets for anticancer treatment [1], [2], [3].

Smurfs are popularly known as little blue comic creatures and are versatile. Similarly in molecular biology the two Smurfs, Smurf1 (Smad ubiquitination-related factor 1) and Smurf2 constitute the Smurf village which are closely related C2-WW-HECT-domain E3 ubiquitin ligases that play important roles in determining the competence of cells to respond to TGF- β/BMP signaling pathway [4], [5]. However, the pleiotropic effect of Smurfs has expanded the repertoire of its substrates beyond the TGF-β/BMP super family ranging from planar cell polarity during embryonic development to cell proliferation, migration, differentiation and senescence [6], [7], [8], [9], [10], [11]. Notably, aberrant expression of Smurfs occurs in several types of cancers, including breast, esophageal, pancreatic and renal cell carcinomas [12], [13], [14], [15], [16]. In contrast, Smurfs are found to induce senescence [10] and recent mouse model studies by Blank et al. [17] showed that Smurf2 is a bona fide tumor suppressor, as the Smurf2-deficient mice are prone to a variety of cancers, including lymphoma, hepatocellular, lung and mammary carcinoma.

Since Smurfs are considered to play a contradictory role as tumor promoter and suppressor, understanding the biological functions of Smurfs and its associated regulatory networks would be crucial for providing insights into the mechanisms of Smurf-mediated cancer progression and also in developing therapeutic strategies that target the Smurf pathway in human cancers. In this review, we focus on the major functions and regulatory network of Smurfs, as well as their role in human cancer.

Section snippets

Smurfs structure, isoforms and localization

Smurfs were initially identified in Xenopus laevis by yeast two- hybrid screening using Smad1 as a bait, regulating the bone morphogenetic protein (BMP) signaling pathway. This gene, XSmurf1, and a closely related human homologue, hSmurf1 (for Xenopus and human Smurf-1), encoded proteins comprising 731 amino acids, and share 91% sequence identity [4]. Similar to Smurf1, Smurf2- which contains 748 amino acids and 83% identical to Smurf1 was found to physically interact with activated Smad2 and

Regulation of Smurfs activity

Although much is known about the Smurf mediated protein degradation pathway and the substrates they target, the regulation and upstream regulators of Smurfs themselves are poorly understood. Here, we summarize several Smurf regulators detailed in literature.

The autoregulatory mechanisms and the WW domain interaction with HECT domain could probably be disrupted by phosphorylation, aiding E3s in ubiquitiating target proteins. Recently, it was observed that protein kinase A (PKA)-dependent

Role of Smurfs in carcinogenesis

The mechanism of cancer initiation and development is not fully understood. However, gain of function of oncogenes and loss-of-function of tumor suppressors are possible basis of this disease. Recently, a flurry of papers reported that Smurfs perform a dual role in cancer by functioning as both tumor promoter and suppressor by controlling the stability of several important proteins with central role in cell-cycle progression, proliferation, differentiation, metastasis, genomic stability and

Concluding remarks and future perspectives

The UPS has been previously shown to be an important mechanism in regulating cellular homeostasis. Here we summarized the conspicuous role of Smurfs, Smurf1 and Smurf2 in the maintenance of this homeostasis, given that Smurfs are frequently deregulated in human cancers and are critical regulators of multiple oncogenic factors [12], [13], [14], [15], [16]. However, the specific role of Smurfs in tumorigenesis has been highly obscure, with numerous studies showing that it is either

Conflict of interest statement

The authors confirm that there are no conflicts of interest.

References (91)

  • S. Wiesner et al.

    Autoinhibition of the HECT-type ubiquitin ligase Smurf2 through its C2 domain

    Cell

    (2007)
  • J. Nie et al.

    Smad ubiquitylation regulatory factor 1/2 (Smurf1/2) promotes p53 degradation by stabilizing the E3 ligase MDM2

    J. Biol. Chem.

    (2010)
  • Y. Zhang et al.

    Regulation of Smad degradation and activity by Smurf2, an E3 ubiquitin ligase

    Proc. Natl. Acad. Sci.

    (2001)
  • S.X. Ying et al.

    Smurf1 facilitates myogenic differentiation and antagonizes the bone morphogenetic protein-2-induced osteoblast conversion by targeting Smad5 for degradation

    J. Biol. Chem.

    (2003)
  • A. Morén et al.

    Degradation of the tumor suppressor Smad4 by WW and HECT domain ubiquitin ligases

    J. Biol. Chem.

    (2005)
  • I. Ito et al.

    Estrogen inhibits transforming growth factor β signaling by promoting Smad2/3 degradation

    J. Biol. Chem.

    (2010)
  • T.R. Pray et al.

    Cell cycle regulatory E3 ubiquitin ligases as anticancer targets

    Drug Resist. Updat.

    (2002)
  • P. Xie et al.

    Smurf1 ubiquitin ligase targets Kruppel-like factor KLF2 for ubiquitination and degradation in human lung cancer H1299 cells

    Biochem. Biophys. Res. Commun.

    (2011)
  • J.X. Du et al.

    The E3 ubiquitin ligase SMAD ubiquitination regulatory factor 2 negatively regulates Krüppel-like factor 5 protein

    J. Biol. Chem.

    (2011)
  • R. Sotillo et al.

    Mad2 overexpression promotes aneuploidy and tumorigenesis in mice

    Cancer Cell

    (2007)
  • M. Kim et al.

    Comparative oncogenomics identifies NEDD9 as a melanoma metastasis gene

    Cell

    (2006)
  • R. Sun et al.

    Intestinal-enriched Krüppel-like factor (Krüppel-like factor 5) is a positive regulator of cellular proliferation

    J. Biol. Chem.

    (2001)
  • L.E.S. Crose et al.

    Cerebral cavernous malformation 2 protein promotes smad ubiquitin regulatory factor 1-mediated RhoA degradation in endothelial cells

    J. Biol. Chem.

    (2009)
  • E. Fukunaga et al.

    Smurf2 induces ubiquitin-dependent degradation of Smurf1 to prevent migration of breast cancer cells

    J. Biol. Chem.

    (2008)
  • J. Nie et al.

    HECT ubiquitin ligase Smurf1 targets the tumor suppressor ING2 for ubiquitination and degradation

    FEBS Lett.

    (2010)
  • X. Guo et al.

    The E3 ligase Smurf1 regulates Wolfram syndrome protein stability at the endoplasmic reticulum

    J. Biol. Chem.

    (2011)
  • R.P. Leng et al.

    Pirh2, a p53-induced ubiquitin-protein ligase, promotes p53 degradation

    Cell

    (2003)
  • D. Chen et al.

    ARF-BP1/Mule is a critical mediator of the ARF tumor suppressor

    Cell

    (2005)
  • C. Esser et al.

    The chaperone-associated ubiquitin ligase CHIP is able to target p53 for proteasomal degradation

    J. Biol. Chem.

    (2005)
  • A.A. Khalil et al.

    Heat shock proteins in oncology: diagnostic biomarkers or therapeutic targets?

    Biochim. Biophys. Acta, Rev. Cancer

    (2011)
  • T. Bellido et al.

    Proteasomal degradation of Runx2 shortens parathyroid hormone-induced anti-apoptotic signaling in osteoblasts: a putative explanation for why intermittent administration is needed for bone anabolism

    Sci. STKE

    (2003)
  • G. Han et al.

    Smad7-induced [beta]-catenin degradation alters epidermal appendage development

    Dev. Cell

    (2006)
  • I. Carpentier et al.

    Smurf2 is a TRAF2 binding protein that triggers TNF-R2 ubiquitination and TNF-R2-induced JNK activation

    Biochem. Biophys. Res. Commun.

    (2008)
  • R. Benezra et al.

    The protein Id: a negative regulator of helix-loop-helix DNA binding proteins

    Cell

    (1990)
  • J. Bond et al.

    Direct evidence from siRNA-directed

    Exp. Cell Res.

    (2004)
  • S. Brookes et al.

    Contribution of p16INK4a to replicative senescence of human fibroblasts

    Exp. Cell Res.

    (2004)
  • B. Zhu et al.

    Monoubiquitination of human histone H2B: the factors involved and their roles in < i > HOX </i > gene regulation

    Mol. Cell

    (2005)
  • L. Moyal et al.

    Requirement of ATM-dependent monoubiquitylation of histone H2B for timely repair of DNA double-strand breaks

    Mol. Cell

    (2011)
  • K. Nakamura et al.

    Regulation of homologous recombination by RNF20-dependent H2B ubiquitination

    Mol. Cell

    (2011)
  • Y. Sun

    E3 ubiquitin ligases as cancer targets and biomarkers

    Neoplasia

    (2006)
  • D. Chen et al.

    The ubiquitin-proteasome system as a prospective molecular target for cancer treatment and prevention

    Curr. Protein Pept. Sci.

    (2010)
  • H. Zhu et al.

    A SMAD ubiquitin ligase targets the BMP pathway and affects embryonic pattern formation

    Nature

    (1999)
  • E.C. Osmundson et al.

    The HECT E3 ligase Smurf2 is required for Mad2-dependent spindle assembly checkpoint

    J. Cell Biol.

    (2008)
  • C. Huang

    Roles of E3 ubiquitin ligases in cell adhesion and migration

    Cell Adh. Migr.

    (2010)
  • H. Zhang et al.

    Smurf2 up-regulation activates telomere-dependent senescence

    Genes Dev.

    (2004)
  • Cited by (72)

    View all citing articles on Scopus
    View full text