Skip to main content
Experimental Biology and Medicine logoLink to Experimental Biology and Medicine
. 2017 Nov 15;243(2):107–117. doi: 10.1177/1535370217740860

Methionine adenosyltransferases in cancers: Mechanisms of dysregulation and implications for therapy

Lauren Y Maldonado 1,*, Diana Arsene 2,*, José M Mato 3, Shelly C Lu 4,
PMCID: PMC5788143  PMID: 29141455

Short abstract

Methionine adenosyltransferase genes encode enzymes responsible for the biosynthesis of S-adenosylmethionine, the principal biological methyl donor and precursor of polyamines and glutathione. Mammalian cells express three genes – MAT1A, MAT2A, and MAT2B – with distinct expression and functions. MAT1A is mainly expressed in the liver and maintains the differentiated states of both hepatocytes and bile duct epithelial cells. Conversely, MAT2A and MAT2B are widely distributed in non-parenchymal cells of the liver and extrahepatic tissues. Increasing evidence suggests that methionine adenosyltransferases play significant roles in the development of cancers. Liver cancers, namely hepatocellular carcinoma and cholangiocarcinoma, involve dysregulation of all three methionine adenosyltransferase genes. MAT1A reduction is associated with increased oxidative stress, progenitor cell expansion, genomic instability, and other mechanisms implicated in tumorigenesis. MAT2A/MAT2B induction confers growth and survival advantage to cancerous cells, enhancing tumor migration. Highlighted examples from colon, gastric, breast, pancreas and prostate cancer studies further underscore methionine adenosyltransferase genes’ role beyond the liver in cancer development. In this subset of extra-hepatic cancers, MAT2A and MAT2B are induced via different regulatory mechanisms. Understanding the role of methionine adenosyltransferase genes in tumorigenesis helps identify attributes of these genes that may serve as valuable targets for therapy. While S-adenosylmethionine, and its metabolite, methylthioadenosine, have been largely explored as therapeutic interventions, targets aimed at regulation of MAT gene expression and methionine adenosyltransferase protein–protein interactions are now surfacing as potential effective strategies for treatment and chemoprevention of cancers.

Impact statement

This review examines the role of methionine adenosyltransferases (MATs) in human cancer development, with a particular focus on liver cancers in which all three MAT genes are implicated in tumorigenesis. An overview of MAT genes, isoenzymes and their regulation provide context for understanding consequences of dysregulation. Highlighting examples from liver, colon, gastric, breast, pancreas and prostate cancers underscore the importance of understanding MAT’s tumorigenic role in identifying future targets for cancer therapy.

Keywords: Chemoprevention, cholangiocarcinoma, colon cancer, hepatocellular carcinoma, methionine adenosyltransferase, S-adenoylmethionine

MAT genes and isoenzymes

MATs are an essential enzyme family that synthesize S-adenosylmethionine (SAMe), the main methyl donor and precursor in polyamine and glutathione synthesis.1 SAMe is vital to the methylation of nucleic acids, phospholipids, histones, biologic amines, and proteins; thus, changes in its biosynthesis can profoundly affect cellular growth, differentiation, and function. Recent data also suggest that MATs play significant roles apart from SAMe synthesis, most notably, serving as transcription factors and co-factors, as well as part of a scaffold complex implicated in cancer development (see below).

Mammalian cells express three genes – MAT1A, MAT2A, and MAT2B – with distinct expression and functions. MAT1A is mainly expressed in the liver and maintains the differentiated states of both hepatocytes and bile duct epithelial cells.1,2 MAT1A encodes the catalytic subunit α1, which forms a homo-dimer (MATIII) and tetramer (MATI).3 Conversely, MAT2A and MAT2B are widely distributed in non-parenchymal cells of the liver and extrahepatic tissues. MAT2A encodes the catalytic α2 subunit found in MATII, predominates in fetal liver and is replaced by MAT1A shortly after birth.4,5 MAT2B encodes the regulatory subunit MATβ, which regulates MATII activity by lowering its Km for methionine and its Ki for SAMe.6,7 MAT2B additionally encodes two major splicing variants, V1 (same as MATβ) and V2, which differs from V1 in the first 20 amino acids at the N-terminus.8 Both MAT2B variants regulate important processes apart from MATII activity, as they interact with many proteins that affect cell growth and signaling pathways (see below). MATI and MATIII have higher Kms than MATII and are not feedback inhibited by SAMe. These differences in kinetic and regulatory properties between the MAT isoenzymes allow a marked increase in hepatic SAMe level after a high methionine load.1 This is important, as the liver is the main site of methionine catabolism, where half of daily intake is converted to SAMe.1 Table 1 summarizes the three MAT genes, isoenzymes, kinetic and regulatory properties of the isoenzymes.

Table 1.

Mammalian MAT genes and isoenzymes.

MAT gene Protein product MAT isoenzyme Regulatory subunit Km for methionine Ki for SAMe
MAT1A α1

MATI (tetramer)

MATIII (dimer)

None

MATI: 23 µM–1 mM

MATIII: 210 µM–7 mM

MATI: 400 µM

MATIII: none

MAT2A α2 Catalytic subunit of MATII Yes, both V1 and V2 regulate MATII by lowering the Km for methionine, V1 also lowers the Ki for SAMe (V2 has not been examined) 4–10 µM 60 µM
MAT2B V1 (same as β), V2 Regulatory subunit of MATII

MAT: methionine adenosyltransferase.

MAT gene regulation and dysregulation

All three MAT genes are regulated at transcriptional, post-transcriptional and post-translational levels. This section describes these regulatory mechanisms for each of the human MAT genes in the normal setting and in cancers. The following section describes the consequences of their dysregulation.

MAT1A

Epigenetic mechanisms and transcription factors are involved in MAT1A transcriptional regulation. In normal liver, MAT1A is epigenetically upregulated by hyperacetylation and cytosine hypomethylation upstream of the MAT1A transcription start site.9 Hypermethylation of the MAT1A promoter is observed in hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA).2,9,10 Methylation of the coding region also reduces MAT1A transcription.11 Importantly, hypermethylation of the MAT1A promoter and coding region was reported in patients with advanced non-alcoholic fatty liver disease (NAFLD) with fibrosis score 3–4, but not mild NAFLD.12

The MAT1A promoter contains consensus binding sites for multiple transcription factors/co-factors that include: glucocorticoid, interleukin-6 (IL-6), hepatocyte nuclear factor (HNF), activator protein 1 (AP-1), CCAAT enhancer binding protein (C/EBP), cyclic AMP response element binding protein (CREBP), E2F, signal transducers and activators of transcription (STAT), c-MYC and v-MYB.13 Among these transcription factors, glucocorticoid and C/EBP demonstrate positive regulation of human MAT1A transcription.13,14 Recently, c-MYC, MAF BZIP transcription factor G (MAFG), and c-MAF were shown to negatively regulation the human MAT1A promoter.2 All three proteins are upregulated in both HCC and CCA, and repress MAT1A transcription by interacting at the MAT1A promoter’s E-box region.2

Multiple factors downregulate MAT1A post-transcriptionally. Binding of AU-rich RNA binding factor (AUF1) to its 3′- untranslated region negatively regulates MAT1A mRNA.15 AUF1 expression is high in HCC and fetal liver and falls during liver development, coinciding with increased MAT1A expression.15 Furthermore, MAT1A mRNA level is downregulated by microRNAs (miRs) in HCC.16,17 Preneoplastic liver lesions induced by 2-acetylaminofluorene injections in rats caused induction of miR-22 and miR-29b that inhibited Mat1a mRNA expression.16 MicroRNAs miR-485–3p, miR-495 and miR-664 are induced in human HCC and all three negatively regulate MAT1A mRNA directly.17 Reduced MAT1A expression resulted in a lower nuclear SAMe level, hypomethylation of the LIN28B promoter region and increased LIN28B expression.17 Importantly, LIN28B is an oncogene that exerts reciprocal regulation with Let-7, a tumor suppressor. Blocking expression of these miRNAs recovers MAT1A expression, functionally inhibits growth, induces apoptosis in HCC cell lines, and inhibits HCC growth in vivo.17

Post-translationally, MATI/III is inactivated by covalent modification of cysteine 121 either by oxidation or nitrosylation.1,18 This is observed in multiple liver diseases and explains the fall in hepatic MAT activity. High GSH level can reverse cysteine 121 covalent modification; however, GSH is often reduced in chronic liver disease leading to a vicious feed-forward cycle. MATI/III can be phosphorylated by protein kinase C,19 but this does not alter enzymatic activity.

MAT2A

MAT2A is expressed at low levels in normal adult liver, but is induced during rapid liver growth and dedifferentiation.1 Like MAT1A, MAT2A expression is regulated both transcriptionally and post-transcriptionally. We identified Sp1, c-MYB, nuclear factor-κB (NF-κB), and AP-1 as participants in MAT2A transcriptional up-regulation in HCC.2022 Hypoxia-inducible factor factor-1α (HIF-1α), activated in HCC, binds to a consensus binding site in the MAT2A promoter and activates transcription in hepatoma cells.23 Hepatitis B X protein also activates MAT2A gene transcription by facilitating NF-κB and CREB binding to the MAT2A promoter, contributing to induction of MAT2A in HBV-associated HCC.24 Promoter methylation and histone acetylation also regulate human MAT2A transcription.25,26

Post-transcriptionally, MAT2A mRNA level is regulated by HuR and methylated-HuR.15 HuR is a ubiquitously expressed mRNA binding protein known to stabilize its target mRNAs, whereas methylated-HuR exerts the opposite effect.15 Interestingly, during hepatocyte de-differentiation and in HCC, there is a switch from methylated-HuR to HuR binding of the 3′-UTR of MAT2A, resulting in increased MAT2A mRNA level.15 Several miRNAs also regulate MAT2A post-transcriptionally. MiR-21–3p, induced by the anticancer drug berberine, was shown to suppress MAT2A mRNA level in HepG2 cells leading to growth arrest and apoptosis.27 Recently, both miR-34a and miR-34b were shown to directly target the MAT2A 3′UTR, downregulating MAT2A expression in colon, prostate, and pancreas cancer cells.28 Both miR-34a and miR-34b are tumor suppressor miRNAs and the former is downregulated in HCC,29 which may contribute to MAT2A induction in HCC.

MATα2, the protein encoded by MAT2A, is also regulated via multiple post-translational modifications (PTMs). MATα2 is stabilized by sumoylation at several lysine residues, K340, K372, and K394.30 Sumoylation also enhances MATα2’s interaction with an important pro-survival protein, B-cell lymphoma 2 (BCL-2), leading to BCL-2 stabilization.30 Other HCC-relevant PTMs of MATα2 are acetylation and ubiquitylation. MATα2 can be acetylated at K81 by P300 (E1A binding protein), which promotes subsequent ubiquitin protein ligase E3 component n-recognin 4-mediated ubiquitylation/degradation.31 Deacetylation by histone deacetylase 3 (HDAC3) stabilizes MATα2. Interestingly, folate deprivation upregulated MATα2 K81 acetylation, destabilized MATα2 and reduced cell growth.31 In HCC, a decrease in MATα2 K81 acetylation is associated with increased expression of HDAC3 compared to normal tissue, underscoring relevance of this PTM to liver cancer.31

MAT2B

Less is known about MAT2B regulation. At the transcriptional level, tumor necrosis factor α (TNF-α) induces V1 or MATβ expression (but not V2) by mechanisms involving AP-1 and NF-κB.8 In liver cancer cells, leptin increases, while SAMe inhibits MAT2B promoter activity and expression via ERK and AKT signaling mechanisms.32 We also demonstrated that sirtuin 1 (SIRT1) can activate MAT2B transcriptionally.33 Post-transcriptionally, HuR stabilizes MAT2B mRNA,33 whereas miR-21–3p directly targets MAT2B 3′UTR to destabilize its mRNA.27 At the protein level, MATβ stability is affected by several protein interactions. MATβ-MATα2 interaction is suspected to stabilize these two proteins as overexpression of MAT2B raised MATα2 levels.34 We confirmed this, as knockdown of endogenous MAT2A or MAT2B lowered protein level of the other in multiple human cancer cell lines.28 This finding helps explain why these two proteins are often simultaneously induced. MATβ also interacts with HuR and SIRT1, and resveratrol enhances these interactions by stabilizing them.33 Finally, both variants of MAT2B also interact with G-protein-coupled receptor kinase-interacting protein 1 (GIT1) to form a scaffold complex. This interaction stabilizes all participants and activates RAS-RAF-MEK-ERK signaling in liver and colon cancer cells.35,36 Table 2 summarizes regulation and dysregulation of the three MAT genes.

Table 2.

Regulatory mechanisms of human MAT genes and proteins.

MAT gene Expression pattern in cancers Transcriptional regulation Post-transcriptional regulation Post-translational regulation
MAT1A Downregulated in hepatocellular carcinoma and cholangiocarcinoma Silenced by promoter and coding region hypermethylation; increased by glucocorticoids and C/EBP; downregulated by c-MYC, MAFG and c-MAF Inhibited by AUF1, miR-485-3p, miR-495, miR-664 by binding to MAT1A 3′-UTR Inactivated by covalent modification of cysteine 121, which can be reversed by GSH
MAT2A Upregulated in liver, colon, gastric, breast, pancreas, prostate cancers Induced by promoter hypomethylation; increased by Sp1, c-MYB, NF-κB, AP-1, HIF-1α, and HBx; suppressed by SAMe Increased by HuR; inhibited by methylated-HuR, miR-21-3p, miR-34a, miR-34b binding to MAT2A 3′-UTR Stabilized by sumoylation at K340, 373, 394, and interaction with MATβ; destabilized by acetylation at K81
MAT2B Upregulated in liver, colon, pancreas and prostate cancers V1 (but not V2) is induced by AP-1, NF-κB, and SIRT1; inhibited by SAMe Increased by HuR; inhibited by miR-21-3p binding to MAT2B 3′-UTR Stabilized by interaction with MATα2 and GIT1

MAT: methionine adenosyltransferase.

Consequences of MAT gene dysregulation

MAT1A

MAT1A expression and MATI/III activity falls in chronic liver disease.1 Epigenetic hypermethylation of promoter and coding regions is attributed to this decline in gene expression and activity. This explains observed impairment of methionine clearance and reduced hepatic GSH level in liver disease patients, since MATI/III are largely responsible for methionine catabolism and SAMe is a key precursor for liver GSH, respectively.1 In HCC, MAT1A is often silenced and low MAT1A expression correlates with poor prognosis.10,37 The Mat1a-knockout (KO) mouse model (Mat1a-KO) has revealed consequences of chronic hepatic SAMe deficiency and provided insight on how loss of MAT1A can lead to HCC development. Mat1a-KO mice have reduced hepatic SAMe and GSH levels, consistent with what’s known about the kinetic and regulatory differences between the MAT isoenzymes and the importance of SAMe as a precursor of GSH in the liver.38 Although KO livers appear normal at three months, liver weights are greater and KO mice are sensitized to develop fatty liver when challenged with a choline-deficient diet for six days.38 KO mice have increased activity of the cytochrome P450 2E1 (CYP2E1) enzyme, which facilitates release of reactive oxygen species (ROS) during hepatotoxin metabolism.39 On a normal diet, KO mice spontaneously develop steatohepatitis by 8 months and HCC by 18 months.38,40 Mitochondrial dysfunction is also evident in Mat1a-KO mice; MAT1A deficiency depletes the mitochondrial chaperone, prohibitin 1 (PHB1), leading to increased propensity for injury, mitochondrial damage, and oxidative stress.41

While increased ROS sensitizes KO mice to liver injury, multiple dysregulated pathways are implicated in HCC development. These pathways include expansion of progenitor cells, some of which are cancer stem cells in aging KO mice,42 increased genomic instability due to decreased protein level of apurinic/apyrimidinic endonuclease 1,43 and uncontrolled extracellular signal-regulated kinases (ERK) activation due to decreased stability of dual-specificity mitogen-activated protein kinases (MAPK) phosphatase.44 Other pathways include: abnormal LKB1/AMPK signaling, (which behaves as an oncogenic pathway in liver),45,46 reduced protein level of PHB1 (a mitochondrial chaperone protein that may also function as a tumor suppressor in hepatocytes and bile duct epithelial cells),41,47 and increased expression of ubiquitin-conjugating enzyme 9 (UBC9) (the sole E2 enzyme in protein sumoylation that is often induced in cancer).48 Among these dysregulated pathways, restoration of hepatic SAMe level normalized ERK activity and reduced UBC9 expression.44,48

Though SAMe deficiency may contribute to dysregulation of many signaling pathways, we recently reported that MATα1 can act as a transcription co-factor that interacts with other E-box binding regulatory proteins, such as c-MYC.2,47 We identified c-MYC, MAFG, c-MAF, MAX network transcriptional repressor (MNT), PHB1, and MYC associated factor X (MAX) as MATα1 interacting proteins that differentially regulate gene expression at the E-box (5′-CACGTG-3′), with c-MYC/MAFG/c-MAF serving as E-box activators, and PHB1 and MATα1 as repressors.2,47 It is well known that c-MYC and MNT compete for heterodimerization with MAX for E-box binding in opposite ways.49 Our recent studies demonstrated that MATα1 can also heterodimerize with MAX, and bind to the E-box to repress E-box-driven promoter activity.47 Interestingly, while c-MYC possesses an E-box that acts as an enhancer element, MAT1A has a repressor E-box. Thus, MAT1A and c-MYC exert reciprocal regulation against each other in part through this mechanism.2 MAT1A can regulate gene expression through epigenetics (i.e. LIN28B promoter methylation mentioned above), but its repressive effect on c-MYC expression occurs despite inhibition of DNA methylation or histone 3 lysine 27 trimethylation (H3K27me3, a transcriptional repressor).2 This supports the notion that MATα1 can also regulate gene expression as a transcription co-factor (it is unable to bind to the E-box by itself), independent of epigenetics. MAT1A is expressed at high levels in normal hepatocytes and bile duct epithelial cells and is repressed in CCA, where c-MYC, MAFG, and c-MAF are induced. These changes favor c-MYC induction as well as CCA proliferation because both effects are suppressed by MAT1A overexpression.2 Together, loss of MAT1A can result in altered liver signaling pathways due to chronically low SAMe levels and/or loss of MATα1-regulated gene expression, leading to development of liver pathology including HCC and CCA.

MAT2A

In HCC, the fall in MAT1A expression is often accompanied by MAT2A induction.10,50 Increased MAT2A expression provides cancer cells a dual growth/survival benefit. Due to kinetic differences and regulatory properties discussed above, a switch from MAT1A to MAT2A expression results in lower steady state SAMe level and favors proliferative signaling in the liver.50,51 Indeed, quiescent hepatocytes have high SAMe levels and a fall in SAMe level is needed for hepatocyte proliferation in conditions such as post-partial hepatectomy.1 SAMe’s inhibitory effect on hepatocyte growth factor (HGF),52 required for liver regeneration, is one proposed mechanism that explains this observation. However, in cancer cells where MAT1A is not expressed, an increase in MAT2A crucially supplies SAMe for polyamine biosynthesis.22 Similarly, silencing MAT2A prevents leptin’s pro-survival signaling by decreasing intracellular SAMe and limiting polyamine biosynthesis.32 Cross-talk between MAT2A and polyamine biosynthesis also occurs in both liver and colon cancer cells.22 Induction of MAT2A enhances polyamine biosynthesis and growth, while increase in polyamines promotes MAT2A transcription by a feed-forward mechanism involving AP-1.22

Increased MAT2A expression can bestow a survival benefit by enhancing BCL-2 expression.30 At least two mechanisms are involved. First, MATα2 can act as a transcription factor that directly binds to the BCL-2 promoter and activates promoter activity.30 Second, MATα2 can physically interact with BCL-2 protein to enhance BCL-2 stability.30 These MATα2 mechanisms require sumoylation at critical lysine residues, which stabilize MATα230 and may enhance nuclear targeting and DNA trans-activating activity (topics for future investigation). SUMO-stabilized MATα2 confers chemo-resistance to liver and colon cancer cells by preventing apoptosis mediated by 5-fluorouracil (5-FU).30

Recently, we reported that increased MAT2A expression in cancer cells enhances tumor migration and invasion.28 This is largely because increased MATα2 stabilizes MATβ, which we believe is the critical mediator in these processes.

MAT2B

As mentioned above, MAT2B and MAT2A expression often simultaneously increase, which is unsurprising given their frequent interaction and co-stabilizing effect.28 However, other mechanisms may also be involved as their mRNA levels are often altered in parallel (such as in HCC).8 Some of these shared mechanisms include AP-1 and NF-κB, both transcription activators, and HuR, which stabilizes both mRNAs.

Similar to MAT2A, increased MAT2B confers a growth advantage; silencing either gene can lead to cancer cell apoptosis.8,24 One of MAT2B’s effects on growth is attributed to its ability to lower SAMe level to steady-state concentration when overexpressed.53 More recent studies found MAT2B regulates much more than MATII enzyme activity. To better understand the functions of MAT2B variants, we identified their interactomes54 and functionally verified the importance of two of them in growth. First is the interaction between MAT2B variants and HuR.54 We found that when either of the MAT2B variants is overexpressed, cytosolic HuR content increased, leading to increased mRNA levels of cyclin D1 and cyclin A (known targets of HuR) and increased growth.54 The opposite occurred when MAT2B variants were silenced.54 Second, the interaction between MAT2B variants and GIT1 form a scaffold complex that efficiently binds and activates MAPK components, mitogen-activated protein kinase (MEK), and ERK.35,36 Silencing either MAT2BV1, V2, or GIT1 blocks ERK activation.35 In the classical MAPK activation pathway, signal transduction from RAF kinases to MEK causes phosphorylation and activation.55 MAT2B variants-GIT1 complex activates and recruits C-RAF and B-RAF to MEK in liver cancer cell lines, promoting MEK activation.36 B-RAF and C-RAF also form homodimers and heterodimers that enhance activity compared to monomeric states.56 The MAT2B variants-GIT1 complex also induces heterodimerization between B-RAF and C-RAF,36 thereby amplifying the MEK signal. Finally, MAT2B variants interact, stabilize, and activate RAS, located upstream of RAF kinases.36 Thus, the MAT2B variants-GIT1 complex interacts and activates all components of RAS-RAF-MEK-ERK signaling – a pathway often dysregulated in cancers. Of note, the effects of MAT2B variants on this signaling pathway occurred independent of MATα2 in these studies, as the overexpression vectors used included a tag that prevents MATα2 interaction. While MAT2B variants can exert pro-tumorigenic effect without MATα2, we suspect MATα2 requires MATβ to induce ERK (this warrants further investigation).

MAT genes in other cancers

Although most studies reporting MAT gene involvement in cancer focus on liver, there are increasing reports that MAT2A and MAT2B are also dysregulated in other cancers, namely: colon, gastric, breast, pancreatic, and prostate cancer.

In colon cancer, MAT2A mRNA levels and MATα2 protein levels are elevated, mirroring expression in liver cancer.57 In contrast to liver cancer, however, MAT2B mRNA levels remain unchanged but protein levels are increased.35,57 Four signaling pathways are recognized in colorectal carcinogenesis: Wnt, K-RAS, transforming growth factor β, and p53,58 with mutations in these pathways leading to inactivation of tumor suppressor function or activation of proto-oncogenes. Additionally, epidermal growth factor (EGF), insulin-like growth factor-1 (IGF-1), and leptin are three well-known growth factors implicated in colon cancer growth and invasion.59,60 All three growth factors induce MAT2A gene expression through transcriptional up-regulation, and promote growth by via endogenous production of SAMe and polyamines; exogenous SAMe and its metabolite methylthioadenosine (MTA) prevent MAT2A induction by these growth factors.57 Mitogens such as IGF-1 are thought to up-regulate MAT2A expression at the transcriptional level, through increased binding of transcription factors to enhancer elements including AP-1 and NF-κB.61

There are shared pathways in which MAT2A is implicated in both colon and liver carcinogenesis. Crosstalk between MAT2A and polyamines, as well as BCL-2 regulation occurs in both colon and liver cancer cells. Similarly, there are significant parallels between MAT2B’s role in liver and colon cancer. MAT2B and GIT1 are over-expressed in both cancers, and regulate RAS-RAF-MEK-ERK signaling similarly in both cell types.35,36 Mutations in this signaling pathway are well known in many cancers, and targeting RAS-RAF-MEK-ERK has been the subject of intense research.62

MAT2A upregulation also occurs in gastric cancer. MAT2A mRNA levels are significantly higher in gastric cancer patients than in corresponding non-tumor tissues and are correlated with high tumor classification indicating lymph node metastasis and poor tumor differentiation.63 Studies also describe the effect of SAMe on c-MYC, H-RAS and tumor-suppressor gene p16 (INK4a) promoter methylation, as well as gastric cancer cell inhibition. SAMe treatment led to heavy promoter methylation, which consequently downregulated mRNA and protein levels, and prevented gastric cancer growth63 pointing to SAMe supplementation as an attractive therapeutic approach.

Like gastric cancer, tamoxifen-resistant breast cancer tissues (TAMR-MCF-7 cells) exhibited increased basal expression of MAT2A compared to MCF-7 control cells. This was also observed in tamoxifen-resistant human breast cancer tissues compared to tamoxifen-responsive cells. Increased transcription factors NF-κB, AP-1, and NRF2 in cell lines may be attributed to these observations.64 Furthermore, the effect of SAMe appears to depend on whether it is increased endogenously (i.e. via increased MAT2A expression) or given in pharmacologic amounts. One study showed that a 30% increase in endogenous SAMe level found in TAMR-MCF-7 cells correlated with hypermethylation of the tumor suppressor phosphatase and tensin homolog (PTEN) and reduced PTEN expression that facilitated growth65; however, exogenous SAMe (200–500µM, more than 10 times physiological level) induces autophagy and apoptosis in MCF-7 cell lines, respectively.66 This seemingly contradictory finding can be explained by whether SAMe is varied physiologically or pharmacologically (see below).

The role of MAT genes and SAMe in pancreatic cancer is not well-studied. Interestingly, outside the liver, MAT1A is highly expressed in pancreatic acinar cells67; however, Mat1a KO mice do not develop pancreatic cancer (Lu SC, unpublished observation). In severe necrotizing pancreatitis models, SAMe levels are 50% decreased and MAT1A protein levels are post-translationally diminished, while MAT2A expression is induced.67 Recently, we found MAT2A and MAT2B proteins are overexpressed in pancreatic cancer and overexpressing MAT2A induced migration of pancreatic cancer cells, likely due to enhancing MAT2B protein stability and activating ERK.28 Like colon cancer, miR-34a expression is often downregulated in pancreatic cancer.68 MAT2A is directly targeted by miR-34a (and miR-34b) to suppress expression and indirectly lower MAT2B protein level by stability reduction.28

In prostate cancer, miR-34a expression is also downregulated, facilitating cancer metastasis via miR-34a suppression of cancer stemness.69 We confirmed that miR-34a and miR-34b also negatively regulate MAT2A and MAT2B at the protein level in prostate cancer cells and overexpressing MAT2A enhanced cancer migration.28 Consistently, both MAT2A and MAT2B are induced in prostate cancer specimens.28

MAT2A and MAT2B are key targets of miR-34a and miR-34b. This is supported by evidence that miR-34a and miR-34b overexpression has minimal to no effect on cancer cell growth, migration, or invasion when either MAT2A or MAT2B is overexpressed.28 This strengthens support for targeting MAT proteins in cancer treatment development. Table 3 summarizes consequences of MAT genes dysregulation.

Table 3.

Consequences of MAT genes dysregulation.

MAT genes Consequences of dysregulation
MAT1A

• Mat1a KO mice have hepatic SAMe and GSH deficiency, increased oxidative stress, and spontaneous development of NASH and HCC

• Dysregulated pathways include cancer stem cells, genomic instability, sustained ERK activation, increased LKB1/AMPK, and sumoylation

Effects on gene expression:

MAT1A can regulate gene expression via epigenetics (i.e. promoter methylation) and as a transcription co-factor (i.e. heterodimerize with MAX, binds to the E-box and represses E-box dependent genes, such as c-MYC)

MAT2A

• Increased MAT2A expression confers cancer cell growth advantage partly by enhancing polyamine biosynthesis, which further induces MAT2A expression

• MAT2A overexpression confers cancer cell survival advantage due to MATα2’s role as a transcription factor that binds and activates the BCL-2 promoter

• MATα2 physically stabilizes BCL-2 which increases chemoresistance to 5-FU in both liver and colon cancer cell

• MAT2A enhances cancer cell migration through stabilizing MAT2B protein

MAT2B

• Increased MAT2B expression:

Promotes tumor growth and metastasis

Promotes HuR cytosolic content, stabilizing many of its targets that include cyclins to promote growth

MAT2B variants and GIT1 form a scaffold complex that interacts and activates RAS-RAF-MEK-ERK signaling in liver and colon cancer cells, promoting growth and metastasis

MAT: methionine adenosyltransferase.

Targeting MAT genes in cancer therapeutics

As our understanding of MAT genes and SAMe’s role in cancer pathogenesis evolves, increasing ideas for potential therapeutic interventions surface. Over two decades of research supports SAMe’s role as a potential chemopreventive agent for HCC, and emerging evidence now supports its role in treatment of extra-hepatic malignancies. Additionally, work over the last few years has elucidated the role of MAT1A, MAT2A and MAT2B in tumorigenesis and identifying these genes as valuable targets for cancer therapeutic research and development. This section summarizes the role of SAMe in this context and emerging approaches that target MAT genes in cancer therapeutics.

SAMe as a chemopreventive and/or therapeutic agent

As previously mentioned, in human HCC, the MAT1A to MAT2A switch is accompanied by increased MAT2B expression, associated with lower SAMe levels and faster growth.1 Interestingly, SAMe maintains MAT1A expression and suppresses MAT2A and MAT2B expression.32,70 The Mat1a-KO mouse model has clearly illustrated the multitude of dysregulated pathways that favor HCC development in the setting of chronic hepatic SAMe depletion. Indeed, SAMe has demonstrated efficacy as a chemopreventive agent against HCC rodent models in multiple studies.7173 SAMe’s chemopreventative ability is attributed to preventing proto-oncogene promoter hypomethylation in these models, which results in gene induction. Using an orthotopic HCC model where hepatic SAMe level is not altered, however, SAMe dramatically inhibited tumor establishment and growth suggesting other mechanisms also participate, such as inhibiting angiogenesis and inducing apoptosis of liver cancer cells.74 Interestingly, SAMe treatment post-HCC development was ineffective in tumor growth reduction.74 This phenomenon was attributed to compensatory induction of glycine N-methyltransferase (GNMT), the liver’s dominant methyltransferase, from chronic SAMe administration, preventing hepatic SAMe accumulation.74 Although hepatic SAMe level consistently increased 10-fold following 24 h of intravenous administration, it was only 30% higher after 24 days of SAMe administration, which is not high enough to induce apoptosis (see below).74 However, GNMT expression is often downregulated in human HCC,75 so the verdict is still out whether SAMe should be used therapeutically. One attractive feature of SAMe is that while it is pro-apoptotic in liver cancer cells, it is anti-apoptotic in normal hepatocytes.76,77 The minimum concentration required for SAMe to induce apoptosis in liver cancer cells after 18 h is 0.5 mM (nearly 10-fold higher than normal hepatic SAMe concentration),77,78 which explains why a 30% increase in hepatic SAMe level did not result in tumor shrinkage.

SAMe versus MTA

Physiological variation and implications for pharmacologic treatment are two important contexts in which we can better understand differences between SAMe and MTA. SAMe is unstable and spontaneously converts to MTA (1.3% per hour at 37˚C, pH 7.0); thus, MTA may mediate SAMe’s pharmacologic effect.57,79 Unlike SAMe, MTA is highly stable, readily traverses the plasma membrane, and inhibits both methylation and polyamine biosynthesis.1 However, MTA can regenerate SAMe via the methionine salvage pathway, which can be blocked with a MAT inhibitor like cycloleucine.1 Thus, if the biological effect is very rapid (so conversion to MTA is minimal), or associated with an increase in methylation, and/or MTA’s effect is blocked by cycloleucine, then SAMe is the active molecule. On the other hand, if the outcome is not blocked by cycloleucine or is associated with a fall in methylation, then MTA is the active molecule. This is illustrated by SAMe and MTA’s inhibitory effect on lipopolysaccharide (LPS)-induced expression of proinflammatory cytokines where MTA was the active compound that blocked the LPS-mediated increase in methylated histone 3 lysine 4 (H3K4) associated with these promoters.79 In contrast, MTA’s ability to prevent the fall in Apurinic/Apyrimidinic Endonuclease 1 (a critical protein involved in DNA repair) protein level during culture of hepatocytes was blocked by cycloleucine, which means MTA’s effect was mediated by SAMe.43 In Mat1a-KO mice, abnormalities corrected after normalization of hepatic SAMe level with supplemental administration suggest these are due to SAMe deficiency as MTA levels are not affected in the KO mice.38

Similar to SAMe, MTA also inhibits HCC cell growth and prevents development of HCC in rats.7173 In hepatoma cells, SAMe and MTA prevented leptin’s ability to induce MAT2A and MAT2B, which are required for growth.32

Both SAMe and MTA affect apoptosis. Interestingly, both are anti-apoptotic in normal hepatocytes but pro-apoptotic in liver cancer cells.76 They selectively induce Bcl-xs, a pro-apoptotic protein, in liver cancer cells by increasing alternative splicing of Bcl-x.77 SAMe and MTA also induce apoptosis in colon cancer cells, but not in NCM460 cell line – an immortalized cell line derived from normal human colonocytes.80 The mechanism of apoptosis is completely different from liver cancer cells.81 SAMe and MTA lowered expression of cellular FLICE inhibitory protein, an anti-apoptotic protein that targets procaspase 8, and had no influence on Bcl-xS expression.81

There is also substantial evidence to support SAMe’s role as a potential chemopreventive agent in colon cancer. Chronic inflammation is an underlying risk factor for colon cancer, and mouse models demonstrate that TNF-α plays a critical role in development of inflammation-induced colon cancer.82,83 Like others, we have shown that SAMe and MTA can reduce the LPS-mediated increase in TNF-α and iNOS expression in mouse macrophages in vitro and mouse livers in vivo.79,84 In addition to altering inflammation, both SAMe and MTA reduce the mitogenic effects of IGF-1, EGF, and leptin in colon cancer cells, as previously mentioned.57 In the colitis-associated colon cancer models, treatment with SAMe and MTA lowered total tumor load by suppressing multiple oncogenic pathways that include NF-κB activation, IL-6 signaling, and β-catenin.85 The mechanism for lowering β-catenin was further examined in both colon and liver cancer cells. SAMe and MTA excluded β-catenin from the nuclear compartment in cells with constitutively active β-catenin, but accelerated β-catenin degradation by a glycogen synthase kinase 3-β-dependent mechanism in cells with wild type β-catenin.86 This ability to suppress β-catenin regardless of whether this pathway is aberrantly induced further supports their use as chemopreventive agents in cancers where aberrant β-catenin pathway is a prominent feature. Recently, we showed that SAMe and MTA can raise the expression of miR-34a and miR-34b and prevent colon cancer liver metastasis in a mouse model of metastatic colon cancer.28

SAMe in other cancers

In gastric cancer, SAMe treatment inhibits the growth of gastric cancer cell lines, SGC-7901 and MKN-45, and significantly decreased mRNA expression of c-MYC and urokinase type plasminogen activator (uPA).63 These studies were confirmed using in vivo xenograft experiments.63 In breast cancer cell lines, the role of SAMe as an adjunct to existing chemotherapeutic therapies is currently being explored. When SAMe is combined with Doxorubicin, the anti-proliferative effect is strongly synergistic in the hormone-dependent CG5 and MCF-7 human breast cancer cell lines. This synergy is attributed to apoptosis induction with observed activation of caspase 3 and 8; this effect was also observed in hormone-independent MDA-MB 231 cells, but less so, suggesting an additive rather than synergistic effect.87 Additionally, SAMe may play a role in cancer epigenetic therapy. 5-Azacytidine, a chemotherapeutic agent used in myelodysplastic disorders, and its deoxy-analog, 5-aza-2′-deoxycytidine are known to induce cancer cell invasiveness and methylated prometastatic genes by DNA demethylation.88 As a methyl donor, SAMe may block the adverse demethylating activity of 5-aza-2′-deoxycytidine while maintaining growth suppression effects. Additionally, SAMe demonstrated reversal of global- and gene-specific demethylation induced by 5-aza-2′-deoxycytidine preventing cell invasiveness in breast cancer cell lines, while still augmenting growth inhibitory effects of 5-aza-2′-deoxycytidine.88 Thus, SAMe is a potential candidate for use in combination therapy aimed at targeting DNA methylation machinery as a new strategy for epigenetic cancer therapy.

Targeting miRNAs that regulate MATs

Several miRNAs regulate MAT1A, MAT2A, and MAT2B. We have demonstrated that targeting the three miRNAs induced in HCC, miR-485–3p, miR-495, and miR-664, is a viable strategy for HCC treatment using the orthotopic HCC model.17 Indeed, when these miRNAs are knocked down, we observe a recovery in MAT1A expression, growth inhibition, and increased apoptosis in HCC in vivo.17 MiR-34a and miR-34b, however, negatively regulate MAT2A/MAT2B expression; thus, one may strategically increase expression of these miRNAs to achieve gene suppression.28 Similarly, berberine was shown to increase the expression of miR-21–3p, which target MAT2A and MAT2B in a human hepatoma cell line.27 As technology evolves, targeting miRNAs that control gene expression will become one of the best approaches to cancer treatment.89

Other approaches that target MATs

Recently, a novel allosteric inhibitor of MAT2A protein that overlaps with the MAT2B binding site demonstrated promise in cancer treatment.90 Interrupting interaction between MAT2B variants and GIT1 is likely to inhibit RAS-RAF-MEK-ERK signaling, and thus may be another promising therapeutic approach.

Conclusions and future directions

In conclusion, increasing evidence suggests that MAT genes are dysregulated in many cancers and these pathways play critical roles in tumorigenesis. Targeting MAT gene dysregulation with chemicals, small molecules that block interactions or miRNAs that control expression may prove effective strategies in future cancer therapy research and development. While much advance has been made on understanding the role of MAT genes in various cancers, many questions remain for future investigation. For instance, MATα1 and MATα2 share high homology yet one is a tumor suppressor and the other an oncoprotein in the liver. The underlying molecular mechanisms for this distinct behavior are unclear. In addition, we have only begun to unravel how these MAT proteins regulate gene expression transcriptionally and proteins functionally. Future investigations should include identifying genes that are regulated by these MAT proteins, either epigenetically or as transcription factors or co-factors. Finally, a better characterization of the MAT proteins interactomes is likely to reveal unknown functions of these proteins that may provide more targets for designing therapy.

Authors’ contributions

LM, DA and SL wrote the paper, JM provided intellectual input.

Declaration of Conflicting Interests

JMM is a consultant for Abbott, Galmed and OWL; the other authors have nothing to declare

Funding

This work was supported by NIH grants R01AT001576 and R01 CA172086 (to JM Mato and SC Lu), R01DK107288 and R01DK092407 (to SC Lu), Plan Nacional of I + D SAF 2014–52097R and Severo Ochoa Excellence Accreditation SEV-2016–0644 (to JM Mato). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Lu SC, Mato JM. S-adenosylmethionine in liver health, injury and cancer. Physiol Rev 2012; 92:1515–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Yang HP, Liu T, Wang J, Li TW, Fan W, Peng H, Krishnan A, Gores GJ, Mato JM, Lu SC. Deregulated methionine adenosyltransferase α1, c-Myc and Maf proteins interplay promotes cholangiocarcinoma growth in mice and humans. Hepatology 2016; 64:439–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kotb M, Mudd SH, Mato JM, Geller AM, Kredich NM, Chou JY, Cantoni GL. Consensus nomenclature for the mammalian methionine adenosyltransferase genes and gene products. Trends Genet 1997; 13:51–2 [DOI] [PubMed] [Google Scholar]
  • 4.Horikawa S, Sasuga J, Shimizu K, Ozasa H, Tsukada K. Molecular cloning and nucleotide sequence of cDNA encoding the rat kidney Methionine adenosyltransferase. J Biol Chem 1990; 265:13683–6 [PubMed] [Google Scholar]
  • 5.Gil B, Casado M, Pajares M, Boscá L, Mato JM, Martín-Sanz P, Alvarez L. Differential expression pattern of methionine adenosyltransferase isoenzymes during rat liver development. Hepatology 1996; 24:876–81 [DOI] [PubMed] [Google Scholar]
  • 6.Halim A, LeGros L, Geller A, Kotb M. Expression and functional interaction of the catalytic and regulatory subunits of human methionine adenosyltransferase in mammalian cells. J Biol Chem 1999; 274:29720–5 [DOI] [PubMed] [Google Scholar]
  • 7.LeGros HL, Jr., Halim AB, Geller AM, Kotb M. Cloning, expression, and functional characterization of the b regulatory subunit of human methionine adenosyltransferase (MAT II). J Biol Chem 2000; 75:2359–66 [DOI] [PubMed] [Google Scholar]
  • 8.Yang HP, Iglesias Ara A, Magilnick N, Xia M, Ramani K, Chen H, Lee TD, Mato JM, Lu SC. Expression pattern, regulation and function of methionine adenosyltransferase 2β alternative splicing variants in hepatoma cells. Gastroenterology 2008; 134:281–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Torres L, Avila MA, Carretero MV, Latasa MU, Caballeria J, Lopez-Rodas G, Boukaba A, Lu SC, Franco L, Mato JM. Liver-specific methionine adenosyltransferase MAT1A gene expression is associated with a specific pattern of promoter methylation and histone acetylation: implications for MAT1A silencing during transformation. FASEB J 2000; 14:95–02 [DOI] [PubMed] [Google Scholar]
  • 10.Frau M, Tomasi ML, Simile MM, Demartis MI, Salis F, Latte G, Calvisi DF, Seddaiu MA, Daino L, Feo CF, Brozzetti S, Solinas G, Yamashita S, Ushijima T, Feo F, Pascale RM. Role of transcriptional and posttranscriptional regulation of methionine adenosyltransferases in liver cancer progression. Hepatology 2012; 56:165–75 [DOI] [PubMed] [Google Scholar]
  • 11.Tomasi ML, Li TWH, Li M, Mato JM, Lu SC. Inhibition of methionine adenosyltransferase 1A transcription by coding region methylation. J Cell Physiol 2012; 227:1583–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Murphy SK, Yang H, Moylan CA, Pang H, Dellinger A, Abdelmalek MF, Garrett ME, Ashley-Koch A, Suzuki A, Tillmann HL, Hauser MA, Diehl AM. Relationship between methylome and transcriptome in patients with nonalcoholic fatty liver disease. Gastroenterology 2013; 145:1076–87 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Zeng Z, Huang ZZ, Chen C, Yang H, Mao Z, Lu SC. Cloning and functional characterization of the 5'-flanking region of human methionine adenosyltransferase 1A gene. Biochem J 2000; 346:475–82 [PMC free article] [PubMed] [Google Scholar]
  • 14.Wu SM, Huang YH, Lu YH, Chien LF, Yeh CT, Tsai MM, Liao CH, Chen WJ, Liao CJ, Cheng WL, Lin KH. Thyroid hormone receptor-mediated regulation of the methionine adenosyltransferase 1 gene is associated with cell invasion in hepatoma cell lines. Cell Mol Life Sci 2010; 67:1831–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Vázquez-Chantada M, Fernández D, Embade N, Woodhoo A, Martinez N, Varela-Rey M, Luka Z, Wagner C, Caballería J, Gorospe M, Lu SC, Mato JM, Martínez-Chantar ML. HuR/Methylated-HuR and AUF1 regulate the expression of methionine adenosyltransferase during liver proliferation, differentiation and carcinogenesis. Gastroenterology 2010; 138:1943–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Koturbash I, Melnyk S, James SJ, Beland FA, Pogribny IP. Role of epigenetic and miR-22 and miR-29b alterations in the downregulation of Mat1a and Mthfr genes in early preneoplastic livers in rats induced by 2-acetylaminofluorene. Mol Carcinog 2013; 52:318–27 [DOI] [PubMed] [Google Scholar]
  • 17.Yang HP, Cho ME, Li TWH, Peng H, Ko KS, Mato JM, Lu SC. MiRNAs regulate methionine adenosyltransferase 1A expression in hepatocellular carcinoma. J Clin Invest 2013; 123:285–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Perez-Mato I, Castro C, Ruiz FA, Corrales FJ, Mato JM. Methionine adenosyltransferase S-nitrosylation is regulated by the basic and acidic amino acids surrounding the target thiol. J Biol Chem 1999; 274:17075–9 [DOI] [PubMed] [Google Scholar]
  • 19.Pajares MA, Duran C, Corrales F, Mato JM. Protein kinase C phosphorylation of rat liver Methionine adenosyltransferase: dissociation and product of an active monomer. Biochem J 1994; 303:949–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Yang HP, Huang ZZ, Wang JH, Lu SC. The role of c-Myb and Sp1 in the up-regulation of methionine adenosyltransferase 2A gene expression in human hepatocellular carcinoma. FASEB J 2001; 15:1507–16 [DOI] [PubMed] [Google Scholar]
  • 21.Yang HP, Sadda MR, Yu V, Zeng Y, Lee TD, Ou XP, Chen LX, Lu SC. Induction of human methionine adenosyltransferase 2A expression by tumor necrosis factor alpha: role of NF-κB and AP-1. J Biol Chem 2003; 278:50887–96 [DOI] [PubMed] [Google Scholar]
  • 22.Tomasi ML, Ryoo M, Skay A, Tomasi I, Giordano P, Mato JM, Lu SC. Polyamine and methionine adenosyltransferase 2A crosstalk in human colon and liver cancer. Exp Cell Res. 2013; 319:1902–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Liu Q, Liu L, Zhao Y, Zhang J, Wang D, Chen J, He Y, Wu J, Liu Z. Hypoxia induces genomic DNA demethylation through the activation of HIF-1α and transcriptional upregulation of MAT2A in hepatoma cells. Mol Cancer Ther 2011; 10:1113–23 [DOI] [PubMed] [Google Scholar]
  • 24.Liu Q, Chen J, Liu L, Zhang J, Wang D, Ma L, He Y, Liu Y, Liu Z, Wu J. The X protein of hepatitis B virus inhibits apoptosis in hepatoma cells through enhancing the methionine adenosyltransferase 2A gene expression and reducing S-adenosylmethionine production. J Biol Chem 2011; 286:17168–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Yang HP, Huang ZZ, Zeng ZH, Chen CJ, Selby RR, Lu SC. Role of promoter methylation in increased methionine adenosyltransferase 2A expression in human liver cancer. Am J Physiol 2001; 280:G184–90 [DOI] [PubMed] [Google Scholar]
  • 26.Latasa MU, Boukaba A, García-Trevijano ER, Torres L, Rodríguez JR, Caballería J, Lu SC, López-Rodas G, Franco L, Mato JM, Avila MA. Hepatocyte growth factor induces MAT2A expression and histone acetylation in rat hepatocytes. Role in liver regeneration. FASEB J 2001; 10:1096. [DOI] [PubMed] [Google Scholar]
  • 27.Lo TF, Tsai WC, Chen ST. MicroRNA-21-3p, a berberine-induced miRNA, directly down-regulates human methionine adenosyltransferases 2A and 2B and inhibits hepatoma cell growth. PLoS One 2013; 8:e75628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Tomasi ML, Cossu C, Spissu Y, Floris A, Ryoo M, Iglesias-Ara A, Wang Q, Pandol SJ, Bhowmick NA, Seki E, Posadas EM, Lu SC. S-adenosylmethionine and methylthioadenosine inhibit cancer metastasis by targeting microRNA 34a/b-methionine adenosyltransferase 2A/2B axis. Oncotarget 2017;8:78851-69 [DOI] [PMC free article] [PubMed]
  • 29.Xiao Z, Li CH, Chan SL, Xu F, Feng L, Wang Y, Jiang JD, Sung JJY, Cheng CHK, Chen YA. small-molecule modulator of the tumor-suppressor miR34a inhibits the growth of hepatocellular carcinoma. Cancer Res 2014; 74:6236–47 [DOI] [PubMed] [Google Scholar]
  • 30.Tomasi ML, Ryoo M, Tomasi I, Giordano P, Mato JM, Lu SC. Methionine adenosyltransferase α2 positively regulates B-cell CLL/lymphoma 2 expression in a sumoylation-dependent manner. Oncotarget 2015; 6:37706–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Yang HB, Xu YY, Zhao XN, Zou SW, Zhang Y, Zhang M, Li JT, Ren F, Wang LY, Lei QY. Acetylation of MAT IIα represses tumour cell growth and is decreased in human hepatocellular cancer. Nat Comms 2015; 6:6973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ramani K, Yang HP, Xia M, Iglesias Ara A, Mato JM, Lu SC. Leptin’s mitogenic effect in human liver cancer cells requires induction of both methionine adenosyltransferase 2A and 2B. Hepatology 2008; 47:521–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yang HP, Zheng Y, Li TWH, Peng H, Ramos DF, Martínez-Chantar ML, Rojas AL, Mato JM, Lu SC. Methionine adenosyltransferase 2B, HuR and Sirtuin 1 crosstalk impacts on resveratrol’s effect on apoptosis and growth in liver cancer cells. J Biol Chem 2013; 288:23161–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Nordgren KKS, Peng Y, Pelleymounter LL, Moon I, Abo R, Feng Q, Eckloff B, Yee YC, Wieben E, Weinshilboum RM. Methionine adenosyltransferase 2A/2B and methylation: gene sequence variation and functional genomics. Drug Metab Disp 2011; 39:2135–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Peng H, Dara L, Li TW, Zheng Y, Yang H, Tomasi LM, Tomasi I, Giordano P, Mato JM, Lu SC. Methionine adenosyltransferase 2B-GIT1 interplay activates MEK1-ERK1/2 to induce growth in human liver and colon cancer. Hepatology 2013; 57:2299–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Peng H, Li TWH, Yang HP, Mato JM, Lu SC. Methionine Adenosyltransferase 2B-GIT1 complex serves as a scaffold to regulate Ras/Raf/MEK1/2 activity in human liver and colon cancer cells. Am J Pathol 2015; 185:1135–44 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Cai J, Sun W, Hwang JJ, Stain S, Lu SC. Changes in S-adenosylmethionine synthetase in human liver cancer: molecular characterization and significance. Hepatology 1996; 24:1090–7 [DOI] [PubMed] [Google Scholar]
  • 38.Lu SC, Alvarez L, Huang ZZ, Chen L, An W, Corrales FJ, Avila MA, Kanel G, Mato JM. Methionine adenosyltransferase 1A knockout mice are predisposed to liver injury and exhibit increased expression of genes involved in proliferation. Proc Natl Acad Sci U S A 2001; 98:5560–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Wu D, Cederbaum AI. Opposite action of S-adenosyl methionine and its metabolites on CYP2E1-mediated toxicity in pyrazole-induced rat hepatocytes and HepG2 E47 cells. Am J Physiol Gastrointest Liver Physiol 2006; 290:G674–84 [DOI] [PubMed] [Google Scholar]
  • 40.Martínez-Chantar ML, Corrales FJ, Martínez-Cruz LA, García-Trevijano ER, Huang ZZ, Chen L, Kanel G, Avila MA, Mato JM, Lu SC. Spontaneous oxidative stress and liver tumors in mice lacking methionine adenosyltransferase 1A. FASEB J 2002; 16:1292–4 [DOI] [PubMed] [Google Scholar]
  • 41.Ko K, Tomasi ML, Iglesias-Ara A, French BA, French SW, Ramani K, Lozano JJ, Oh P, He L, Stiles BL, Li TWH, Yang HP, Martínez-Chantar ML, Mato JM, Lu SC. Liver-specific deletion of prohibitin 1 results in spontaneous liver injury, fibrosis and hepatocellular carcinoma in mice. Hepatology 2010; 52:2096–08 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Rountree CB, Senadheera S, Mato JM, Crooks GM, Lu SC. Expansion of liver cancer stem cells during aging in methionine adenosyltransferase 1A-deficient mice. Hepatology 2008; 47:1288–97 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Tomasi ML, Iglesias Ara A, Yang H, Ramani K, Feo F, Pascale MR, Martínez-Chantar ML, Mato JM, Lu SC. S-adenosylmethionine regulates apurinic/apyrimidinic endonuclease 1 stability: implication in hepatocarcinogenesis. Gastroenterology 2009; 136:1025–36 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Tomasi ML, Ramani K, Lopitz-Otsoa F, Rodríguez MS, Li TWH, Ko K, Yang HP, Bardag-Gorce F, Iglesias-Ara A, Feo F, Pascale MR, Mato JM, Lu SC. S-adenosylemthionine regulates dual-specificity MAPK phosphatase expression in mouse and human hepatocytes. Hepatology 2010; 51:2152–61 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Vázquez-Chantada M, Ariz U, Varela-Rey M, Embade N, Martínez-Lopez N, Fernández-Ramos D, Gómez-Santos L, Lamas S, Lu SC, Martínez-Chantar ML, Mato JM. Evidence for an LKB1/AMPK/eNOS cascade regulated by HGF, S-Adenosylmethionine and NO in hepatocyte proliferation. Hepatology 2009; 49:608–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Martínez-López N, Varela-Rey M, Fernández-Ramos D, Woodhoo A, Vázquez-Chantada M, Embade N, Espinosa-Hevia L, Bustamante FJ, Parada LA, Rodriguez MS, Lu SC, Mato JM, Martínez Chantar ML. Activation of LKB1-Akt pathway independent of phosphoinositide 3-kinase plays a critical role in the proliferation of hepatocellular carcinoma from nonalcoholic steatohepatitis. Hepatology 2010; 52:1621–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Fan W, Yang H, Liu T, Wang J, Li TWH, Mavila N, Tang Y, Yang JW, Peng H, Annamalai A, Noureddin M, Krishnan A, Gores GJ, Martínez-Chantar ML, Mato JM, Lu SC. Prohibitin 1 suppresses liver cancers tumorigenesis in mice and humans. Hepatology 2017; 65:1249–66 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Tomasi ML, Tomasi I, Ramani K, Pascale RM, Xu J, Giordano P, Mato JM, Lu SC. S-adenosyl methionine regulates ubiquitin-conjugating enzyme 9 protein expression and sumoylation in human cancers. Hepatology 2012; 56:982–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lüscher B, Vervoorts J. Regulation of gene transcription by the oncoprotein MYC. Gene 2012; 494:145–60 [DOI] [PubMed] [Google Scholar]
  • 50.Cai J, Mao Z, Hwang JJ, Lu SC. Differential expression of methionine adenosyltransferase genes influences the rate of growth of human hepatocellular carcinoma cells. Cancer Res 1998; 58:1444–50 [PubMed] [Google Scholar]
  • 51.Ramani K, Lu SC. Methionine adenosyltransferase genes in liver health and disease. In: Tirosh O (ed) Liver metabolism and fatty liver disease. Boca Raton, Florida: Taylor & Francis Group/CRC Press 2014, pp.103–24
  • 52.Garcia-Trevijano ER, Martínez-Chantar ML, Latasa MU, Mato JM, Avila MA. NO sensitizes rat hepatocytes to hepatocyte growth factor-induced proliferation through the modulation of S-adenosylmethionine levels. Gastroenterology 2002; 122:1355–63 [DOI] [PubMed] [Google Scholar]
  • 53.Martínez-Chantar ML, Garcia-Trevijano ER, Latasa MU, Martin-Duce A, Fortes P, Caballeria J, Avila MA, Mato JM. Methionine adenosyltransferase II β subunit gene expression provides a proliferative advantage in human hepatoma. Gastroenterology 2003; 124:940–8 [DOI] [PubMed] [Google Scholar]
  • 54.Xia M, Chen Y, Wang LC, Zandi E, Yang HP, Bemanian S, Martínez-Chantar ML, Mato JM, Lu SC. Novel function and intracellular localization of methionine adenosyltransferase 2β splicing variants. J Biol Chem 2010; 285:20015–21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Marais R, Light Y, Paterson HF, Mason CS, Marshall CJ. Differential regulation of Raf-1, A-Raf, and B-Raf by oncogenic ras and tyrosine kinases. J Biol Chem 1997; 272:4378–83 [DOI] [PubMed] [Google Scholar]
  • 56.Matallanas D, Birtwistle M, Romano D, Zebisch A, Rauch J, von Kriegsheim A, Kolch W. Raf family kinases: old dogs have learned new tricks. Genes Cancer 2011; 2:232–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Chen H, Xia M, Lin M, Yang H, Kuhlenkamp J, Li T, Sodir NM, Chen YH, Josef-Lenz H, Laird PW, Clarke S, Mato JM, Lu SC. Role of methionine adenosyltransferase 2A and S-adenosylmethionine in mitogen-induced growth of human colon cancer cells. Gastroenterology 2007; 133:207–18 [DOI] [PubMed] [Google Scholar]
  • 58.Piard F, Martin L, Chapusot C, Chapusot C, Ponnelle T, Faivre J. Genetic pathways in colorectal cancer: interest for the pathologist. Ann Pathol 2002; 22:277–88 [PubMed] [Google Scholar]
  • 59.Reinmuth N, Fan F, Liu W, Parikh AA, Stoeltzing O, Jung YD, Bucana CD, Radinsky R, Gallick GE, Ellis LM. Impact of insulin-like growth factor receptor-1 function on angiogenesis, growth, and metastasis of colon cancer. Lab Invest 2002; 82:1377–89 [DOI] [PubMed] [Google Scholar]
  • 60.LeRoith D, Roberts CT., Jr., The insulin-like growth factor system and cancer. Cancer Lett 2003; 195:127–37 [DOI] [PubMed] [Google Scholar]
  • 61.Yang HP, Li TW, Peng J, Mato JM, Lu SC. Insulin-like growth factor 1 activates methionine adenosyltransferase 2A transcription by multiple pathways in human colon cancer cells. Biochem J 2011; 436:507–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Sacco E, Spinelli M, Vanoni M. Approaches to Ras signaling modulation and treatment of Ras-dependent disorders: a patent review. Expert Opin Ther Pat 2012; 22:1263–87 [DOI] [PubMed] [Google Scholar]
  • 63.Zhang T, Zheng Z, Liu Y, Zhang J, Zhao Y, Liu Y, Zhu H, Zhao G, Liang J, Li Q, Xu H. Overexpression of methionine adenosyltransferase II alpha (MAT2A) in gastric cancer and induction of cell cycle arrest and apoptosis in SGC-7901 cells by shRNA-mediated silencing of MAT2A gene. Acta Histochem 2013; 115:48–55 [DOI] [PubMed] [Google Scholar]
  • 64.Phuong NT, Kim SK, Im JH, Yang JW, Choi MC, Lim SC, Lee KY, Kim YM, Yoon JH, Kang KW. Induction of methionine adenosyltransferase 2A in tamoxifen-resistant breast cancer cells. Oncotarget 2016; 7:13902–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Phuong NT, Kim SK, Lim SC, Kim HS, Kim TH, Lee KY, Ahn SG, Yoon JH, Kang KW. Role of PTEN promoter methylation in tamoxifen-resistant breast cancer cells. Breast Cancer Res Treat 2011; 130:73–83 [DOI] [PubMed] [Google Scholar]
  • 66.Cave DD, Desiderio V, Mosca L, Llisso CP, Mele L, Caraglia M, Cacciapuoti G, Porcelli M. S-Adenosylmethionine-mediated apoptosis is potentiated by autophagy inhibition induced by chloroquine in human breast cancer cells. J Cell Physiol. Epub ahead of print 2017. DOI: 10.1002/jcp.26015 [DOI] [PubMed]
  • 67.Lu SC, Gukovsky I, Lugea A, Reyes CN, Huang ZZ, Chen L, Mato JM, Bottiglieri T, Pandol SJ. Role of S-adenosylmethionine in two experimental models of pancreatitis. FASEB J 2003; 17:56–8 [DOI] [PubMed] [Google Scholar]
  • 68.Tang Y, Tang Y, Cheng YS. miR-34a inhibits pancreatic cancer progression through Snail1- mediated epithelial–mesenchymal transition and the Notch signaling pathway. Sci Rep 2017; 7:38232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Liu C, Kelner K, Liu B, Chen X, Calhoun-Davis T, Li H, Patrawala L, Yan H, Jeter C, Honorio S, Wiggins JF, Bader AG, Fagin R, Brown D, Tang DG. The microRNA miR-34a inhibits protstate cancer stem cells and metastasis by directly repressing CD44. Nature Med 2011; 17:211–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Garcia-Trevijano ER, Latasa MU, Carretero MV, Berasain C, Mato JM, Avila MAS-A. regulates MAT1A and MAT2A gene expression in cultured rat hepatocytes: a new role for S-adenosylmethionine in the maintenance of the differentiated status of the liver. FASEB J 2000; 14:2511–8 [DOI] [PubMed] [Google Scholar]
  • 71.Pascale R, Simile MM, Ruggiu ME, Seddaiu MA, Satta G, Sequenza MJ, Daino L, Vannini MG, Lai P, Feo F. Reversal by 5-azacytidine of the S-adenosyl-L-methionine-induced inhibition of the development of putative preneoplastic foci in rat liver carcinogenesis. Cancer Lett 1991; 56:259–65 [DOI] [PubMed] [Google Scholar]
  • 72.Pascale RM, Simile MM, Satta G, Seddaiu MA, Daino L, Pinna G, Vinci MA, Gaspa L, Feo F. Comparative effects of L-methionine, S-adenosyl-L-methionine and 5'-methylthioadenosine on the growth of preneoplastic lesions and DNA methylation in rat liver during the early stages of hepatocarcinogenesis. Anticancer Res 1991; 11:1617–24 [PubMed] [Google Scholar]
  • 73.Pascale RM, Simile MM, De Miglio MR, Nufris A, Daino L, Seddaiu MA, Rao PM, Rajalakshmi S, Sarma DS, Feo F. Chemoprevention by S-adenosyl-L-methionine of rat liver carcinogenesis initiated by 1,2-dimethylhydrazine and promoted by orotic acid. Carcinogenesis 1995; 16:427–30 [DOI] [PubMed] [Google Scholar]
  • 74.Lu SC, Ramani K, Ou XP, Lin M, Yu V, Ko K, Park R, Bottiglieri T, Tsukamoto H, Kanel G, French S, Mato JM, Moat R, Grant E. S-adenosylmethionine in the chemoprevention and treatment of hepatocellular carcinoma in a rat model. Hepatology 2009; 50:462–71 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Liu HH, Chen KH, Shih YP, Lui WY, Wong FH, Chen YM. Characterization of reduced expression of glycine N-methyltransferase in cancerous hepatic tissues using two newly developed monoclonal antibodies. J Biomed Sci 2003; 10:87–97 [DOI] [PubMed] [Google Scholar]
  • 76.Ansorena E, García-Trevijano ER, Martínez-Chantar ML, Huang ZZ, Chen LX, Mato JM, Iraburu M, Lu SC, Avila MA. S-adenosylmethionine and methylthioadenosine are anti-apoptotic in cultured rat hepatocytes but pro-apoptotic in human hepatoma cells. Hepatology 2002; 35:274–80 [DOI] [PubMed] [Google Scholar]
  • 77.Yang HP, Sadda MR, Li M, Zeng Y, Chen LX, Bae WJ, Ou XP, Runnegar MT, Mato JM, Lu SC. S-Adenosylmethionine and its metabolite induce apoptosis in HepG2 cells: role of protein phosphatase 1 and Bcl-xS. Hepatology 2004; 40:221–31 [DOI] [PubMed] [Google Scholar]
  • 78.Lee TD, Sadda ME, Mendler MH, Bottiglieri T, Kanel G, Mato JM, Lu SC. Abnormal hepatic methionine and GSH metabolism in patients with alcoholic hepatitis. Alcoholism 2004; 28:173–81 [DOI] [PubMed] [Google Scholar]
  • 79.Ara AI, Xia M, Ramani K, Mato JM, Lu SC. S-adenosylmethionine inhibits lipopolysaccharide-induced gene expression via modulation of histone methylation. Hepatology 2008; 47:1655–66 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Moyer MP, Manzano LA, Merriman RL, Stauffer JS, Tanzer LR. Letter to the Editor: NCM460, a normal human colon mucosal epithelial cell line. In Vitro Cell Dev Biol-Anim 1996; 32:315–7 [DOI] [PubMed] [Google Scholar]
  • 81.Li T, Zhang Q, Oh P, Xia M, Chen H, Bemanian S, Circ M, Moyer MP, Mato JM, Aw TY, Lu SC. S-Adenosylmethionine and methylthioadenosine inhibit cellular FLICE inhibitory protein expression and induce apoptosis in colon cancer cells. Mol Pharmacol 2009; 76:192–200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Terzic J, Grivennikov S, Karin E, Karin M. Inflammation and colon cancer. Gastroenterology 2010; 138:2101–14 [DOI] [PubMed] [Google Scholar]
  • 83.Karin M. Nuclear factor-kappaB in cancer development and progression. Nature 2006; 441:431–6 [DOI] [PubMed] [Google Scholar]
  • 84.Hevia H, Varela-Rey M, Corrales FJ, Berasain C, Martínez-Chantar ML, Latasa MU, Lu SC, Mato JM, García-Trevijano ER, Avila MA. 5'-methylthioadenosine modulates the inflammatory response to endotoxin in mice and in rat hepatocytes. Hepatology 2004; 39:1088–98 [DOI] [PubMed] [Google Scholar]
  • 85.Li TWH, Yang HP, Peng H, Xia M, Mato JM, Lu SC. Effects of S-adenosylmethionine and methylthioadenosine on inflammation-induced colon cancer in mice. Carcinogenesis 2012; 33:427–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Li TWH, Peng H, Yang HP, Kuriniawidjaja S, Panthaki P, Zheng Y, Mato JM, Lu SC. S-adenosylmethionine and methylthioadenosine inhibit beta catenin signaling by multiple mechanisms in liver and colon cancer. Mol Pharmacol 2015; 87:77–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ilisso CP, Castellano M, Zappavigna S, Lombardi A, Vitale G, Dicitore A, Cacciapuoti G, Caraglia M, Porcelli M. The methyl donor S-adenosylmethionine potentiates doxorubicin effects on apoptosis of hormone-dependent breast cancer cell lines. Endocrine 2015; 50:212–22 [DOI] [PubMed] [Google Scholar]
  • 88.Chik F, Machnes Z, Szyf M. Synergistic anti-breast cancer effect of a combined treatment with the methyl donor S-adenosyl methionine and the DNA methylation inhibitor 5-aza-2'-deoxycytidine. Carcinogenesis 2014; 35:138–44 [DOI] [PubMed] [Google Scholar]
  • 89.Cheng CJ, Bahar B, Babar IA, Pincus Z, Barrera F, Liu C, Svoronos A, Braddock DT, Glazer PM, Engelman DM, Saltzman WM, Slack FJ. MicroRNA silencing for cancer therapy targeted to the tumor microenvironment. Nature 107;518:107–12 [DOI] [PMC free article] [PubMed]
  • 90.Quinlan CL, Kaiser SE, Bolaños B, Nowlin D, Grantner R, Karlicek-Bryant S, Feng JL, Jenkinson S, Freeman-Cook K, Dann SG, Wang X, Wells PA, Fantin VR, Stewart AI, Grant SK. Targeting S-adenosylmethionine biosynthesis with a novel allosteric inhibitor of Mat2A. Nature Chem Biol 2017; 13:785–92 [DOI] [PubMed] [Google Scholar]

Articles from Experimental Biology and Medicine are provided here courtesy of Frontiers Media SA

RESOURCES