Cancer Letters

Cancer Letters

Volume 433, 1 October 2018, Pages 86-98
Cancer Letters

Original Articles
N-α-acetyltransferase 10 protein promotes metastasis by stabilizing matrix metalloproteinase-2 protein in human osteosarcomas

https://doi.org/10.1016/j.canlet.2018.06.033Get rights and content

Highlights

  • Naa10p promotes invasion and metastasis of osteosarcoma cells.

  • Naa10p promotes metastasis via increasing MMP-2 expression in osteosarcoma cells.

  • Naa10p acetylates MMP-2 and prevents its degradation via NatA activity.

Abstract

N-α-Acetyltransferase 10 protein (Naa10p) mediates N-terminal acetylation of nascent proteins. Oncogenic or tumor suppressive roles of Naa10p were reported in cancers. Here, we report an oncogenic role of Naa10p in promoting metastasis of osteosarcomas. Higher NAA10 transcripts were observed in metastatic osteosarcoma tissues compared to non-metastatic tissues and were also correlated with a worse prognosis of patients. Knockdown and overexpression of Naa10p in osteosarcoma cells respectively led to decreased and increased cell migratory/invasive abilities. Re-expression of Naa10p, but not an enzymatically inactive mutant, relieved suppression of the invasive ability in vitro and metastasis in vivo imposed by Naa10p-knockdown. According to protease array screening, we identified that matrix metalloproteinase (MMP)-2 was responsible for the Naa10p-induced invasive phenotype. Naa10p was directly associated with MMP-2 protein through its acetyltransferase domain and maintained MMP-2 protein stability via NatA complex activity. MMP-2 expression levels were also significantly correlated with Naa10p levels in osteosarcoma tissues. These results reveal a novel function of Naa10p in the regulation of cell invasiveness by preventing MMP-2 protein degradation that is crucial during osteosarcoma metastasis.

Introduction

Osteosarcomas are the most common primary malignant tumor of the bone in children and adolescents [1]. Although clinical management of osteosarcomas, including surgery and chemotherapy, has significantly improved long-term survival over the past few decades, outcomes for those patients with metastatic or recurrent osteosarcoma remain dismally poor and, therefore, novel therapeutic strategies are urgently required [2].

Matrix metalloproteinases (MMPs) play important roles in the progression of several types of cancer by increasing tumor growth, migration, invasion, and metastasis and are associated with poor disease prognosis [3]. Tumor growth of osteosarcomas is accompanied by both enhanced local bone destruction and bone formation, two processes that are dependent on proteolytic enzymes. Indeed, many MMPs were found to be expressed in primary and metastatic osteosarcoma tissues [4]. MMP-2, -9, and -14 as well as tissue inhibitor of matrix metalloproteinase (TIMP)-1 were shown to be associated with a poor prognosis of osteosarcomas [5,6]. Intense immunostaining for MMP-2 and -9 was also detected in metastatic lesions from osteosarcomas [4,7]. However, MMP-2 was recently considered to be a more-important MMP involved in osteosarcoma progression. A shift from MMP-9 to predominant MMP-2 activity was observed in zymographic screening of cryo-preserved osteosarcoma biopsies and was associated with a poor response to chemotherapy [8].

N-α-acetyltransferase 10 protein (Naa10p) is the catalytic subunit of a heterodimeric complex, NatA, which co-translationally acetylates N termini of small side-chain amino acids such as Ser, Ala, Thr, Gly, Val, and Cys, after the initiator methionine has been cleaved from nascent polypeptides [9,10]. Despite being debated, Naa10p was also found to acetylate proteins at internal lysine residues of diverse targets [11]. Through N-α or N-ε acetylation, Naa10p regulates protein stability [12], protein activity [13], and protein-protein interactions [14,15] in a variety of target proteins. Non-acetylation-dependent functions of Naa10p were also documented in regulating cell behaviors [16,17]. Thus, Naa10p seems to be a diverse protein with different capacities for executing its effects on target proteins. This diversity also reflects the identified roles of Naa10p in cancer cells. Dependent on the cell context, Naa10p is involved in regulating cell proliferation, apoptosis, autophagy, metastasis, and chemosensitivity of different cancer cells [9]. Naa10p overexpression was reported in breast [18], colorectal [19], liver [20], and lung cancers [21], while low expression in non-small cell lung cancer (NSCLC) was also documented [12]. Furthermore, loss of heterozygosity at Naa10p gene loci was also found in tumors including lung, breast, pancreatic, and ovarian cancers [12]. Considering the diverse targets Naa10p may regulate in different types of cancer cells or in different stages during cancer development, identifying cancer-type specific targets may help to understand the role of Naa10p in a particular cancer type.

Recently, Naa10p was found to participate in controlling osteoblast differentiation and bone formation as a feedback regulator of Runx2 [22]. Naa10p transgenic mice showed delayed calvarial bone development, while Naa10p-knockout mice showed facilitated calvarial bone development. Naa10p-knockdown also augmented the healing of rat calvarial defects. These results highlight a negative role of Naa10p in osteoblast differentiation and bone development. However, whether Naa10p is involved in the formation or progression of osteosarcomas is completely unknown. Herein, we showed that Naa10p is functionally involved in the invasion and metastasis of osteosarcomas. Naa10p is highly expressed in late-stage osteosarcomas and was correlated with shorter survival in patients. Mechanistic studies revealed that Naa10p directly interacts with MMP-2. Naa10p increases MMP-2 protein stability in an N-α-acetylation-dependent manner and thus increases the invasive ability of osteosarcoma cells. Our findings highlight the potential of targeting Naa10p-MMP2 regulation in therapeutic applications against osteosarcomas.

Section snippets

Materials

MG132 and cycloheximide (CHX) were purchased from Sigma-Aldrich (St Louis, MO). Cell culture materials and fetal bovine serum (FBS) were obtained from Gibco-BRL (Gaithersburg, MD). The following antibodies were used: Naa10p (GB-10511, Genesis Biotech, Taiwan) and MMP-2 (sc-13594, Santa Cruz Biotechnology, Santa Cruz, CA) for immunohistochemistry staining; Naa10p (GB-10511, Genesis Biotech) and MMP-2 (#4022, Cell Signaling Technology, Danver, MA, USA) for immunoblotting; Naa10p (sc-33820, Santa

Naa10p expression correlated with poor prognosis of osteosarcoma patients and invasiveness of osteosarcoma cell lines

Naa10p was recently reported to be expressed by osteoblasts which are located around new bone surfaces [22]. To examine the role of Naa10p in osteosarcomas, which arise from a mesenchymal origin and exhibit osteoblastic differentiation, we analyzed Naa10p expression in a tissue microarray composed of osteosarcomas at different clinical stages. Our results showed that Naa10p was barely expressed by normal cartilage; on the contrary, Naa10p expression was enriched in osteosarcoma tissues (Fig. 1

Discussion

MMPs are known for their role in promoting cancer progression and are considered ideal drug targets. However, MMPs' intervention strategies have met with limited clinical success due to severe toxicities [35]. These poor results are likely due to the fact that MMPs play complex roles in both normal and cancer tissues. Most of the early programs targeting MMPs with broad-spectrum MMP-inhibitors were terminated due to poorly tolerated musculoskeletal pain and inflammation [36]. Therefore, for

Conflicts of interest

The authors declare that no conflicts of interest exist.

Authors' contributions

M-H Chien and K-T Hua designed and conceived the study. W-J Lee, Y-C Yang, P Tan, K-F Pan, and Y-C Liu performed the experiments. H-C Tsai, C-H Hsu, and M Hsiao technically or materially supported the experiments. M-H Chien and K-T Hua wrote the manuscript.

Acknowledgements

We thank Mass Core Facility of Genomics Research Center, Academia Sinica, Taipei, Taiwan for Mass spectrometry analyses. We also thank Dr. Junn-Liang Chang for the scoring of Naa10p in tissue arrays. This research was supported by grants from the Ministry of Science and Technology, Taiwan (104-0210-01-09-02, 105-0210-01-13-01, and 106-0210-01-15-02 to M. Hsiao, 105-2320-B-002-035-MY3 to K.T. Hua and 105-2320-B-038 -058 -MY3 to M.H. Chien). This study was also supported by the “TMU Research

References (44)

  • A.F. Foukas et al.

    Stage-IIB osteosarcomas around the knee. A study of MMP-9 in surviving tumour cells

    J. Bone Joint Surg. Br.

    (2002)
  • M. Uchibori et al.

    Increased expression of membrane-type matrix metalloproteinase-1 is correlated with poor prognosis in patients with osteosarcoma

    Int. J. Oncol.

    (2006)
  • B.P. Himelstein et al.

    Matrix metalloproteinase-9 (MMP-9) expression in childhood osseous osteosarcoma

    Med. Pediatr. Oncol.

    (1998)
  • P. Kunz et al.

    Elevated ratio of MMP2/MMP9 activity is associated with poor response to chemotherapy in osteosarcoma

    BMC Canc.

    (2016)
  • T.V. Kalvik et al.

    Protein N-terminal acetyltransferases in cancer

    Oncogene

    (2013)
  • T. Arnesen et al.

    Proteomics analyses reveal the evolutionary conservation and divergence of N-terminal acetyltransferases from yeast and humans

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

    (2009)
  • H.P. Kuo et al.

    ARD1 stabilization of TSC2 suppresses tumorigenesis through the mTOR signaling pathway

    Sci. Signal.

    (2010)
  • D.H. Shin et al.

    Arrest defective-1 controls tumor cell behavior by acetylating myosin light chain kinase

    PLoS One

    (2009)
  • J.S. DePaolo et al.

    Acetylation of androgen receptor by ARD1 promotes dissociation from HSP90 complex and prostate tumorigenesis

    Oncotarget

    (2016)
  • Z. Wang et al.

    Inactivation of androgen-induced regulator ARD1 inhibits androgen receptor acetylation and prostate tumorigenesis

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

    (2012)
  • C.F. Lee et al.

    hNaa10p contributes to tumorigenesis by facilitating DNMT1-mediated tumor suppressor gene silencing

    J. Clin. Invest.

    (2010)
  • M. Yu et al.

    Correlation of expression of human arrest-defective-1 (hARD1) protein with breast cancer

    Canc. Invest.

    (2009)
  • Cited by (0)

    1

    These authors contributed equally to this work.

    View full text