Elsevier

Cellular Signalling

Volume 26, Issue 7, July 2014, Pages 1532-1538
Cellular Signalling

Rlim, an E3 ubiquitin ligase, influences the stability of Stathmin protein in human osteosarcoma cells

https://doi.org/10.1016/j.cellsig.2014.03.018Get rights and content

Highlights

  • We identify that Rlim could interact with Stathmin.

  • Rlim induces ubiquitination of Stathmin and promotes degradation of Stathmin.

  • Rlim is involved in the negative regulation of Stathmin protein level.

  • Rlim-Stathmin interaction can turn over malignant phenotype in osteosarcomas cells.

Abstract

Stathmin is an oncoprotein and is expressed at high levels in a wide variety of human malignancies, which plays important roles in maintenance of malignant phenotypes. The regulation of Stathmin gene overexpression has been wildly explored, but the exact mechanism still needs to be elucidated. It is believed that regulation of an oncogene protein abundance through post-translational modifications is essential for maintenance of malignant phenotypes. Here we identified the Rlim, a Ring H2 zinc finger protein with intrinsic ubiquitin ligase activity, as a Stathmin-interacting protein that could increase Stathmin turnover through binding with this targeted protein and then induce its degradation by proteasome in a ubiquitin-dependent manner. Inhibition of endogenous Rlim expression by siRNA could increase the level of Stathmin protein, which further led to cell proliferation and cell cycle changes in human osteosarcoma cell lines. On the other hand, forced overexpression of Rlim could decrease the level of Stathmin protein. These results demonstrate that Rlim is involved in the negative regulation of Stathmin protein level through physical interaction and ubiquitin-mediated proteolysis. Hence, Rlim is a novel regulator of Stathmin protein in a ubiquitin-dependent manner, and represents a new pathway for malignant phenotype turnover by modulating the level of Stathmin protein in human osteosarcomas.

Introduction

Stathmin (STMN1), a 19 kDa cytosolic protein and also known as Op18, is a major phosphoprotein that plays crucial roles in the control of cell division and proliferation by regulating the dynamics of the microtubules [1], [2], [3]. Stathmin is also an oncoprotein and is expressed at high levels in a wide variety of human malignancies including osteosarcoma, lung cancer, uterine cervix cancer, bladder cancer, and breast cancer, which demonstrates that the overexpression of Stathmin may play an important role in maintenance of malignant phenotypes in malignant tumors [4], [5], [6], [7], [8]. Our previous studies on Stathmin have shown that Stathmin overexpression is associated with cellular proliferation, and inhibition of Stathmin expression can abolish the transformed phenotypes in malignant cells [9], [10], [11]. In addition, inhibition of Stathmin expression in malignant tumor cells interferes with their proliferation through the cell cycle arrest and abrogates their transformed phenotypes [12], [13], [14]. Thus, Stathmin provides an attractive molecular target for the treatment of malignant tumors.

In a search for proteins capable of interacting with Stathmin, we identified Rlim as a candidate of such proteins. Rlim (RING finger LIM domain-binding protein), encoded by the Rnf12 gene and located around 500 kb telomeric to the Xist gene on the X chromosome [15], [16], is originally identified as an E3 ubiquitin ligase. Rlim acts as a negative coregulator for LIM homeodomain transcription factors by mediating the ubiquitination and subsequent degradation of LIM cofactors LDB1 and LDB2 [17], [18], [19]. It has been reported that Rlim also plays a role in telomere length-mediated growth suppression by mediating the ubiquitination and degradation of TRF1, and acts as an activator of random inactivation of X chromosome in the embryo by targeting ZFP42 for degradation [20], [21]. Furthermore, study has shown that Rlim also enhances transcriptional activation of endogenous estrogen receptor alpha (ERalpha) target genes [22], [23]. Since signaling pathways controlled by ERalpha are profoundly implicated in mammary oncogenesis, Rlim may play an important role during the development of human cancers.

However, whether and how Rlim affects Stathmin protein levels and further influences the Stathmin functional roles during carcinogenesis have not been determined so far. In this study, we found that Rlim increased Stathmin turnover by binding it for degradation through the proteasome in a ubiquitin-dependent manner. Depletion of endogenous Rlim expression by siRNA stabilized Stathmin protein, whereas overexpression of Rlim promoted degradation of Stathmin and further impaired cell growth. In sum, these results demonstrated that Rlim was a critical negative regulator of Stathmin protein abundance and represented a new target for cancer therapy.

Section snippets

Chemicals and antibodies

MG132 (CAS Number: 133407-82-6) and cycloheximide (CAS Number: 66-81-9) were purchased from Sigma (MO, USA). MG132 was dissolved in DMSO and the dosage of MG132 treatment was 10 μM for 4 h unless otherwise mentioned. Cycloheximide was dissolved in water and the concentration for cell treatment was 25 μM at indicated time points. Antibodies against Stathmin (ab52630), Rlim (ab22813), Ubiquitin (ab7780) and Flag tags (ab122902) were purchased from Abcam (UK). The antibodies against His (sc-804), GST

Identification of Rlim as a partner of Stathmin in osteosarcoma cells

To identify the natural protein partners of Stathmin in osteosarcoma cells, we expressed a Flag–Stathmin in the human osteosarcoma cell line MG-63. We analyzed proteins that co-purified with Flag–Stathmin by LC/MS/MS after sequential Flag immunoprecipitation and peptide elution [28] and discovered that Stathmin complexes contain Rlim (Fig. 1A). Rlim (Ring finger LIM domain-binding protein, also known as RNF12), is originally identified as an E3 ubiquitin ligase and capable of targeting

Discussion

Stathmin, a kind of small molecular weight cytosolic phosphoprotein, regulates microtubule dynamics and influences cell cycle through integrating and transducing various transduction signals within cells. Expression studies have shown that Stathmin is aberrantly overexpressed in the majority of human malignancies, and plays an important role in maintaining malignant phenotype of tumors [33]. Stathmin expression and its function are controlled by multiple steps and different regulating levels

Acknowledgment

Thanks to everyone from the Department of Clinical Laboratory for their sincere help and excellent technical assistance. This study was supported by a grant from the National Natural Science Foundation of China (No. 81001195).

References (39)

  • L. Cassimeris

    Cell Biol.

    (2002)
  • G. Chen et al.

    Mol. Cell. Proteomics

    (2003)
  • M.O. Steinmetz

    J. Struct. Biol.

    (2007)
  • I. Jonkers et al.

    Cell

    (2009)
  • H.P. Ostendorff et al.

    Genomics

    (2000)
  • Y.R. Her et al.

    J. Biol. Chem.

    (2009)
  • R. Nikolay et al.

    J. Biol. Chem.

    (2004)
  • S.F. Ahmed et al.

    J. Biol. Chem.

    (2012)
  • X. Li et al.

    J. Biol. Chem.

    (2011)
  • R.G. Polzin et al.

    Gene

    (2004)
  • H. Benlhabib et al.

    Gene

    (2006)
  • L. Kashima et al.

    J. Biol. Chem.

    (2012)
  • P. Niethammer et al.

    Science

    (2004)
  • C.I. Rubin et al.

    J. Cell. Biochem.

    (2004)
  • X. Wang et al.

    Cancer Biol. Ther.

    (2010)
  • S. Rana et al.

    Expert. Rev. Anticancer. Ther.

    (2008)
  • S.J. Mistry et al.

    Mt Sinai J. Med.

    (2002)
  • H.Z. Zhang et al.

    Ai Zheng

    (2004)
  • H.Z. Zhang et al.

    Cancer Biol. Ther.

    (2006)
  • Cited by (19)

    • RNF12 promotes p53-dependent cell growth suppression and apoptosis by targeting MDM2 for destruction

      2016, Cancer Letters
      Citation Excerpt :

      RNF12 modulates the telomere length homeostasis through degradation of TRF1 [9]. RNF12 influences the stability of Stathmin protein in human osteosarcoma cells [10]. Recently studies identified X-encoded RNF12 as a dose-dependent activator of X-chromosome inactivation (XCI) in mouse embryonic stem cells (ESCs) [11].

    View all citing articles on Scopus

    Grant support: This study was supported by a grant from the National Natural Science Foundation of China (No. 81001195).

    1

    These authors contributed equally to this work.

    View full text