Elsevier

Biochemical Pharmacology

Volume 86, Issue 9, 1 November 2013, Pages 1285-1300
Biochemical Pharmacology

Adenosine dialdehyde suppresses MMP-9-mediated invasion of cancer cells by blocking the Ras/Raf-1/ERK/AP-1 signaling pathway

https://doi.org/10.1016/j.bcp.2013.08.022Get rights and content

Abstract

Adenosine dialdehyde (AdOx) inhibits transmethylation by the accumulation of S-adenosylhomocysteine (SAH), a negative feedback inhibitor of methylation, through the suppression of SAH hydrolase (SAHH). In this study, we aimed to determine the regulatory effect of AdOx on cancer invasion by using three different cell lines: MDA-MB-231, MCF-7, and U87. The invasive capacity of these cells in the presence (MCF-7) or absence (MDA-MB-231 and U87) of phorbal 12-myristate 13-acetate (PMA) was strongly decreased by AdOx treatment. Furthermore, the expression, secretion, and activation of matrix metalloproteinase (MMP)-9, a critical enzyme regulating cell invasion, in these cells were diminished by AdOx treatment. AdOx strongly suppressed AP-1-mediated luciferase activity and, in parallel, reduced the translocation of c-Fos and c-Jun into the nucleus. AdOx was shown to block a series of upstream AP-1 activation signaling complexes composed of extracellular signal-related kinase (ERK), mitogen-activated protein ERK kinase (MEK)1/2, Raf-1, and Ras, as assessed by measuring the levels of the phosphorylated and membrane-translocated forms. Furthermore, we found that suppression of SAHH by siRNA and 3-deazaadenosine, knock down of isoprenylcysteine carboxyl methyltransferase (ICMT), and treatment with SAH showed inhibitory patterns similar to those of AdOx. Therefore, our data suggest that AdOx is capable of targeting the methylation reaction regulated by SAHH and ICMT and subsequently downregulating MMP-9 expression and decreasing invasion of cancer cells through inhibition of the Ras/Raf-1/ERK/AP-1 pathway.

Introduction

It is well-known that when a tumor metastasizes [1], the cancer becomes very difficult to cure. Six steps lead to metastasis: (1) aggressive proliferation of transformed cells; (2) breakdown of the basal lamina; (3) intravasation and circulation through the bloodstream or lymphatic system; (4) attachment to the inner wall of the blood vessel; (5) extravasation; and (6) abnormal proliferation at the metastasized site [2], [3]. Among these steps, acquisition of invasive capacity is a key step toward cancer malignancy.

Tumor cell invasion, i.e., the expansion of tumor cells into surrounding tissues, is a complex process that leads to proteolysis and destruction of biological barriers [4]. Type I collagen is the most abundant component of the extracellular matrix, and the invasive capacity of tumor cells involves proteolytic enzymes such as matrix metalloproteinases (MMPs) [5]. Among the extracellular proteases responsible for the degradation of the extracellular matrix, MMP-2 and MMP-9 play critical roles in the invasiveness of carcinoma [6], [7]. Therefore, targeting of the enzyme activity of MMP has been regarded as an important strategy in the development of anti-tumor agents with anti-invasive properties.

Methylation is a representative biochemical reaction in which a methyl group from S-adenosylmethionine (SAM) is transferred to numerous methyl-accepting donors such as proteins, DNA, and RNA [8], [9]. This reaction is catalyzed by various protein/DNA/RNA methyltransferases (N-, O-, and S-methyltransferases) [10], [11]. Although methylation is regarded as a critical pathway in many different human diseases including aging, obesity, and Parkinson's disease, it is unclear how methylation modulates tumorigenic responses [12]. In particular, because epigenetic processes such as DNA methylation and histone modifications, including methylation and acetylation, at lysine and arginine residues are tightly associated with the proliferation, apoptosis, survival, migration, and invasion of tumor cells [13], it is expected that elucidation of the methylation reaction could lead to therapeutic solutions.

Adenosine dialdehyde (AdOx) is a broadly used indirect methylation inhibitor [10], [14], [15]. AdOx inhibits adenosylhomocysteine hydrolase (SAHH), which is an important enzyme for SAM metabolism [16], [17]. SAHH converts S-adenosyl-l-homocysteine (SAH), a negative feedback inhibitor of methylatransferase that utilizes S-adenosyl-l-methionine (SAM) as its methyl donor, into adenosine and l-homocysteine, which are necessary for producing SAM [18]. AdOx treatment induces a hypomethylated state by suppressing methyltransferase activity in cells in two ways: (1) accumulation of SAH and (2) deficiency of SAM [19], [20]. Whereas the functional role of AdOx in the methylation of tumorigenic responses is known, the inhibitory activity of AdOx in various cancer stages has not been fully elucidated. Indeed, this compound has been shown to block the proliferation of some cancer cells such as prostate cancer cells, P19 embryonic carcinoma cells, MCF breast cancer cells, and neuroblastoma cells [10], [21], [22]. In particular, AdOx is able to arrest the cell cycle at the G2 phase and induce p53-dependent apoptosis [11], [23]. These results strongly suggest the possibility that AdOx can be used as a methylation-inhibitory anti-cancer drug. However, no studies have demonstrated the suppressive role of AdOx in tumor cell invasion and migration. The aim of this study, therefore, was to evaluate the inhibitory effect of AdOx on tumor invasion in view of its molecular suppressive mechanism.

Section snippets

Materials

AdOx, gelatin, Coomassie brilliant blue, and phorbal 12-myristate 13-acetate (PMA) were purchased from Sigma Chemical Co. (St. Louis, MO). SB203580, a p38 inhibitor, U0126, a MEK inhibitor, and SP600125, a JNK inhibitor were obtained from Calbiochem (La Jolla, CA). Antibodies against phospho- and total forms of extracellular signal-related kinase (ERK), p38, c-Jun N-terminal kinase (JNK), mitogen-activated protein kinase/ERK (extracellular-signal-regulated kinase) kinase 1/2 (MEK1/2), Ras,

AdOx inhibits the invasion and migration capabilities of cancer cells

MDA-MB-231 cells, a human adenocarcinoma cell line of the mammary gland, and U87 cells, a human glioblastoma cell line, are known to be highly aggressive and invasive by increased expression of MMPs catalyzing proteolytic degradation of the extracellular matrix [37], [38]. In contrast, since MCF-7 cells, derived from human breast adenocarcinoma, do barely possess metastatic and invasive capacities, it is reported that PMA-stimulated conditions are required for triggering the migration and

Discussion

Because methylation has many functions in tumor development and aggressiveness, methyltransferases have been suggested as targets of anti-cancer drugs [47]. Indeed, it has already been reported that DNA and protein methylation reactions are involved in tumorigenic responses through the regulation of the proliferation, differentiation, and migration of cancer cells [48]. It was previously reported that AdOx, a general methylation inhibitor that is broadly used for blocking methylation [19], [20]

Conflicts of interest statement

The authors declare that there are no conflicts of interest.

Acknowledgements

This study was supported by a Grant (HI12C0050) of the Korean Health Technology R&D Project, Ministry of Health & Welfare, Republic of Korea.

References (71)

  • S.E. Byeon et al.

    Hydroquinone regulates hemeoxygenase-1 expression via modulation of Src kinase activity through thiolation of cysteine residues

    Free Radic Biol Med

    (2013 April)
  • L. Ge et al.

    Constitutive protease-activated receptor-2-mediated migration of MDA MB-231 breast cancer cells requires both beta-arrestin-1 and -2

    J Biol Chem

    (2004)
  • S.O. Lee et al.

    Silibinin suppresses PMA-induced MMP-9 expression by blocking the AP-1 activation via MAPK signaling pathways in MCF-7 human breast carcinoma cells

    Biochem Biophys Res Commun

    (2007)
  • M. Karin

    The regulation of AP-1 activity by mitogen-activated protein kinases

    J Biol Chem

    (1995)
  • E. Chicoine et al.

    Evidence for the role of promoter methylation in the regulation of MMP-9 gene expression

    Biochem Biophys Res Commun

    (2002)
  • K. Hata et al.

    Role for Btg1 and Btg2 in growth arrest of WEHI-231 cells through arginine methylation following membrane immunoglobulin engagement

    Exp Cell Res

    (2007)
  • C. Torricelli et al.

    Role of PTHrp and PTHrp-engaged pathways in MCF-7 cells migration/invasion

    Matrix Biol

    (2006)
  • T. Yu et al.

    Anti-inflammatory activity of ethanol extract derived from Phaseolus angularis beans

    J Ethnopharmacol

    (2011)
  • T. Curran et al.

    Fos and Jun: the AP-1 connection

    Cell

    (1988)
  • R. Roskoski

    ERK1/2 MAP kinases: structure, function, and regulation

    Pharmacol Res

    (2012)
  • C.J. Chalmers et al.

    The duration of ERK1/2 activity determines the activation of c-Fos and Fra-1 and the composition and quantitative transcriptional output of AP-1

    Cell Signal

    (2007)
  • K.A. Creedon et al.

    Plasmodium falciparum S-adenosylhomocysteine hydrolase. cDNA identification, predicted protein sequence, and expression in Escherichia coli

    J Biol Chem

    (1994)
  • C.A. Klein

    Cancer. The metastasis cascade

    Science

    (2008)
  • M. Bacac et al.

    Metastatic cancer cell

    Annu Rev Pathol

    (2008)
  • K.H. Jung et al.

    Progress in cancer therapy targeting c-Met signaling pathway

    Arch Pharm Res

    (2012)
  • W.G. Stetler-Stevenson et al.

    Tumor cell interactions with the extracellular matrix during invasion and metastasis

    Annu Rev Cell Biol

    (1993)
  • M. Sillem et al.

    Invasiveness corresponds to differentiation rather than to proteinase secretion in endometrial cancer cell lines

    Eur J Gynaecol Oncol

    (1999)
  • Y.M. Kim et al.

    Anti-metastatic effect of cantharidin in A549 human lung cancer cells

    Arch Pharm Res

    (2013)
  • W.K. Paik et al.

    Protein methylation: chemical, enzymological, and biological significance

    Adv Enzymol Relat Areas Mol Biol

    (1975)
  • C. Schwerk et al.

    Methyltransferase inhibition induces p53-dependent apoptosis and a novel form of cell death

    Oncogene

    (2005)
  • Y. Bergman et al.

    DNA methylation dynamics in health and disease

    Nat Struct Mol Biol

    (2013)
  • S.V. Liu et al.

    Epigenetic therapy in lung cancer

    Front Oncol

    (2013)
  • N. Bae et al.

    Knock-down of protein l-isoaspartyl O-methyltransferase increases beta-amyloid production by decreasing ADAM10 and ADAM17 levels

    Acta Pharmacol Sin

    (2011)
  • R.L. Bartel et al.

    Effects of adenosine dialdehyde on S-adenosylhomocysteine hydrolase and S-adenosylmethionine-dependent transmethylations in mouse L929 cells

    Mol Pharmacol

    (1984)
  • U. Patel-Thombre et al.

    Adenine nucleoside dialdehydes: potent inhibitors of bovine liver S-adenosylhomocysteine hydrolase

    Biochemistry

    (1985)
  • Cited by (0)

    1

    These authors equally contributed to this work.

    View full text