Epigenetic deregulation of the COX pathway in cancer

Epigenetic deregulation of the COX pathway in cancer

Progress in Lipid Research 51 (2012) 301–313 Contents lists available at SciVerse ScienceDirect Progress in Lipid Research journal homepage: www.els...

826KB Sizes 0 Downloads 42 Views

Progress in Lipid Research 51 (2012) 301–313

Contents lists available at SciVerse ScienceDirect

Progress in Lipid Research journal homepage: www.elsevier.com/locate/plipres

Review

Epigenetic deregulation of the COX pathway in cancer Inês Cebola 1, Miguel A. Peinado ⇑ Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Badalona, Catalonia, Spain

a r t i c l e

i n f o

Article history: Received 2 December 2011 Received in revised form 8 February 2012 Accepted 8 February 2012 Available online 3 May 2012 Keywords: DNA methylation Histone modification Gene regulation Gene silencing Prostaglandins Inflammation and cancer

a b s t r a c t Inflammation is a major cause of cancer and may condition its progression. The deregulation of the cyclooxygenase (COX) pathway is implicated in several pathophysiological processes, including inflammation and cancer. Although, its targeting with nonsteroidal antiinflammatory drugs (NSAIDs) and COX-2 selective inhibitors has been investigated for years with promising results at both preventive and therapeutic levels, undesirable side effects and the limited understanding of the regulation and functionalities of the COX pathway compromise a more extensive application of these drugs. Epigenetics is bringing additional levels of complexity to the understanding of basic biological and pathological processes. The deregulation of signaling and biosynthetic pathways by epigenetic mechanisms may account for new molecular targets in cancer therapeutics. Genes of the COX pathway are seldom mutated in neoplastic cells, but a large proportion of them show aberrant expression in different types of cancer. A growing body of evidence indicates that epigenetic alterations play a critical role in the deregulation of the genes of the COX pathway. This review summarizes the current knowledge on the contribution of epigenetic processes to the deregulation of the COX pathway in cancer, getting insights into how these alterations may be relevant for the clinical management of patients. Ó 2012 Elsevier Ltd. All rights reserved.

Contents 1.

2.

3.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1.1. The COX pathway and cancer. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1.2. Epigenetics and cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Epigenetic alterations of the COX pathway in cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.1. The cyclooxygenases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.2. Prostaglandin E2 biosynthesis and signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.3. Prostacyclin biosynthesis and signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.4. Prostaglandin D2 biosynthesis and signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.5. Thromboxane A2 biosynthesis and signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.6. Prostaglandin F2a biosynthesis and signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

302 302 303 303 303 304 306 306 307 307 307 308 308

Abbreviations: AKR1B1, aldo–keto reductase family 1 member B1; AOM, azoxymethane; COX, cycloxigenase; COX-1 (also known as PTGS1), cycloxigenase 1; COX-2 (also known as PTGS2), cycloxigenase 2; Coxibs, selective COX-2 inhibitors; CRTH2 (also known as DP2), chemoattractant receptor-homologous expressed on T helper type 2 cells; EP1 (also known as PTGER1), PGE2 receptor 1; EP2 (also known as PTGER2), PGE2 receptor 2; EP3 (also known as PTGER3), PGE2 receptor 3; EP4 (also known as PTGER4), PGE2 receptor 4; HDACs, histone deacetylases; HPGD, 15-hydroxyprostaglandin dehydrogenase; HPGDS (also known as H-PGDS), hematopoietic PGD2 synthase; HPV, human papillomavirus; IP (PTGIR), prostacyclin receptor; LOX, lipoxygenase; MBDs, methyl-binding domain proteins; NSAIDs, nonsteroidal antiinflammatory drugs; PLA2, phospholipase A2; PPAR, peroxisome-proliferator-activated receptor; PGD2, prostaglandin D2; PGE2, prostaglandin E2; PGF2a, prostaglandin F2a; PGH2, prostaglandin H2; PGI2, prostacyclin (also known as prostaglandin I2); PKA, protein kinase A; PTGDR (also known as DP1), prostaglandin D2 receptor; PTGDS (also known as L-PGDS), lipocalin-type PGD2 synthase; PTGES (also known as mPGES-1), microsomal membrane-bound PGE2 synthase-1; PTGES2 (also known as mPGES-2), microsomal membrane-bound PGE2 synthase-2; PTGES3 (also known as cPGES), cytosolic PGE2 synthase 3; PTGFR (also known as FP), PGF2a receptor; PTGIS (also known as PGIS), prostacyclin synthase; TBXAS1 (also known as TXAS), thromboxane synthase; TP (also known as TBXA2R), thromboxane receptor; TXA2, thromboxane A2. ⇑ Corresponding author. Address: Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain. Tel.: +34 93 554 3050; fax: +34 93 465 1472. E-mail address: [email protected] (M.A. Peinado). 1 Present address: Institut d’Investigacións Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Rosselló 153, Edifici CEK, Planta 5, 08036 Barcelona, Spain. 0163-7827/$ - see front matter Ó 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.plipres.2012.02.005

302

I. Cebola, M.A. Peinado / Progress in Lipid Research 51 (2012) 301–313

1. Introduction Several factors contribute to cancer development and maintenance, including genetic and epigenetic alterations, chronic inflammation and lifestyle factors, such as diet and tobacco smoking. In particular, a large body of evidence from epidemiological and animal model studies demonstrates an association between high-fat diets and increased risk for particular malignant diseases, such as colorectal, pancreatic, breast and prostate cancer [1]. Arachidonic acid is a polyunsaturated fatty acid present in most mammalian cell membranes and a major component of animal fats. Mostly released from the membrane by phospholipase A2 (PLA2), this fatty acid can be substrate for the generation of distinct eicosanoids, being metabolized by the cyclooxygenase (COX) pathway, the lipoxygenase (LOX) pathway or the cytochrome P450 monooxygenase pathway. Several reports show that both COX and LOX pathways are frequently overactivated during chronic inflammation and cancer (reviewed in reference [2]), which may account for the association between high-fat diets and higher incidence of chronic inflammation settings and malignant growths (reviewed in reference [3]). Cyclooxygenases catalyze the two-step conversion reaction of arachidonic acid into prostaglandin H2 (PGH2), which constitutes the precursor for the subsequent synthesis of a number of structurally related prostanoids, the prostaglandins and thromboxanes, by specialized eicosanoid synthases. Importantly, not only the prostanoid production, but the presence of specific G protein-coupled receptors at the cell surface as well, is determining for the establishment of downstream prostanoid-dependent signaling. These

surface receptors are designated according to the concomitant ligand. In addition, prostanoids such as prostacyclin are able to bind nuclear receptors from the peroxisome proliferator-activated receptors family (see below) (Fig. 1). Noteworthy, whereas biosynthesis and receptor binding account for the final outcome of the pathway, maintenance of a given metabolic state implicates influx and efflux transporters, as well as cytoplasmatic inactivation, which may be enzyme-dependent or not.

1.1. The COX pathway and cancer There are two major COX isoforms, COX-1 (also known as PTGS1) and COX-2 (also known as PTGS2). Although the expression profiles of both isoforms are variable from tissue to tissue, COX-1 is generally considered the constitutive form, being responsible for the homeostatic production of prostanoids and highly expressed in most tissues, including platelets, lung, prostate, brain, gastrointestinal tract, kidney, liver and spleen. On the contrary, COX-2, often referred as the inducible or rate-limiting isoform, is usually undetectable in most normal tissues, being responsible for most of the prostanoid production during inflammation and markedly upregulated in various types of cancer, as well as in other diseases [4–7]. However, recent data indicates that the ‘constitutive’ and ‘inducible’ terms may be too reductionist for the complex dynamics of COX-1 versus COX-2. In fact, several normal tissues show detectable COX-2 levels [8]. Moreover, high levels of both COX-1 and COX-2 have been found in mouse lung tumors [9]. A third isoform, COX-3, has been also described, however, it appears to

Fig. 1. Diagram of the COX pathway. Arachidonic acid is a polyunsaturated fatty acid that constitutes the phospholipid domain of most cell membranes and is liberated from the cellular membranes by cytoplasmic phospholipase A2 (PLA2). Free arachidonic acid can be metabolized to eicosanoids through three major pathways: the cyclooxygenase (COX), the lipoxygenase (LOX) and the cytochrome P450 monooxygenase pathways. In the COX pathway, the key step is the enzymatic conversion of arachidonic acid to the intermediate prostaglandin G2 (PGG2), which is then reduced to PGH2 by the peroxidase activity of COX. PGH2 is sequentially metabolized to prostanoids, including prostaglandins (PGs) and thromboxanes (TXs) by specific prostaglandin and thromboxane synthases. Each of the prostaglandins exerts its biological effects by binding to its cognate G protein-coupled receptor.

I. Cebola, M.A. Peinado / Progress in Lipid Research 51 (2012) 301–313

be an inactive protein, which results from the alternative splicing of COX-1 [10]. Overexpression of COX-2 in tumors is usually linked to overproduction of the pro-inflammatory prostaglandin PGE2 [11,12]. Furthermore, cancer-causing agents have been reported to induce constitutive COX-2 expression, including tobacco smoke (reviewed in Ref. [13]), UV irradiation [14], essential polyunsaturated fatty acids [15–20], mitogens [21], growth factors [22–26], pro-inflammatory cytokines [24,27] and microbial agents [28–36]. Due to its pro-inflammatory potential, COX-2 overexpression may trigger the acquisition of essential cancer-cell traits [37], including evasion of apoptosis [38–43], immunosuppresion [44], sustained proliferation [45–48], angiogenesis [49–52], invasion and metastasis [53–57]. The important participation of cyclooxygenases in cancer promotion and progression is also well illustrated by models of murine carcinogenesis, as the Min mouse carrying a mutation in the APC gene [58,59] and the azoxymethane (AOM) induced tumors [60]. Several epidemiological studies have shown that the regular intake of NSAIDs, such as aspirin, ibuprofen and sulindac, inhibitors of COX, reduces the risk of cancer (for detailed reviews see references [61–63]). In colorectal cancer, sulindac, not only was shown to present chemopreventive functions in sporadic tumors, but in familial adenomatous polyposis as well [64]. Nevertheless, NSAIDs-based cancer therapies have been associated with increased risk of gastrointestinal complications, mainly due to the inhibition of COX-1 [65], and long-term administration of selective COX-2 inhibitors (coxibs) shows a strong association with higher risk of cardiovascular outcomes (myocardial infarction or stroke) [66], making the targeting of COX-2 a less attractive therapeutic strategy to pursuit. Even though this is still a main topic of discussion, important considerations must be taken before enrolling new clinical trials, including the particular genetic backgrounds that might confer additional susceptibilities to the inhibition of COX by NSAIDs [67,68] and the NSAIDs COX-independent actions [69,70]. 1.2. Epigenetics and cancer Chromatin is a dynamic structure able to change the accessibility of regulatory regions of the genome to the transcriptional machinery. In a simplified scenario, chromatin can switch between two interconvertible states manifested at structural and functional level: (1) active regions, which are transcribed or ready to start transcription upon proper stimulation and present a relaxed structure; and (2) inactive regions, which hold a repressed transcriptional state and display a condensed structure. Several epigenetic mechanisms account for the establishment of either one state or the other, including DNA methylation, modification of histones and non coding RNAs [71–75]. In humans and other mammals, DNA methylation occurs predominantly in CpG dinucleotides and is associated with transcriptional repression [76]. CpGs are not randomly distributed in the genome, but found enriched in clusters called CpG islands [76]. It is estimated that about 76% of the human genes possess a CpG island associated with their promoter regions [77] and most of them are unmethylated [76]. On the other hand, most methylated CpG sites are located in repeat elements and other inactive regions. The repressive state involves many components and layers of complexity. In brief, methylated DNA is able to recruit methyl-binding domain proteins (MBDs), together with histone deacetylases (HDACs) and other repressor elements, to promote chromatin remodeling to a compact conformation, leading to transcription impairment and acquisition of a permanent repressed state [78,79]. The epigenomic landscape varies markedly across tissue types and between individuals [75,80,81]. Recent genome-scale studies

303

have revealed that most CpG methylation changes associated with development and cancer do not occur in CpG islands, but rather in sequences up to 2 kb distant, which have been termed CpG island shores [82,83]. During carcinogenesis, the epigenetic landscape is severely distorted, presenting global changes in the DNA methylation content among other epigenetic aberrations (reviewed in [75,84–87]). As a consequence, the alteration of the DNA methylation patterns may have profound consequences in human disease. Global loss of DNA methylation (hypomethylation) throughout the genome is a common feature of cancer cells and relates with the acquisition of genomic instability and activation of normally repressed genes, such as germline-specific genes. Furthermore, cancer cells also present punctual gain of DNA methylation (hypermethylation) in particular CpG islands, which results in the gene silencing of the associated genes, many of which are bona fide tumor-suppressor genes (reviewed in [84–86]) (Fig. 2). Histone tail post-translational modifications are also important regulatory epigenetic marks for transcriptional activation or repression (for review see references [74,88,89]). Histone methylation may actually be associated with both chromatin states, being the methylation of H3K4 and H3K36 generally associated with active genes, whereas the methylation of H3K9 predominantly occurs in repressed regions. As in the case of histone lysine acetylation, by weakening the histone–DNA interactions, this modification maintains chromatin in a low condensation state, being therefore found in transcribed genes. In accordance, HDACs, which reverse histone lysine acetylation, are usually considered elements of transcriptional repression [74,89]. In embryonic stem cells it has been reported the coexistence of active and inactive chromatin histone marks in a subset of genes with key functions in development and cell identity [90,91]. Noteworthy, these bivalent chromatin domains are also characteristic of the genes that become silenced in cancer by hypermethylation of the corresponding CpG island [92–94]. 2. Epigenetic alterations of the COX pathway in cancer The aberrant expression of several COX pathway genes has been recurrently shown as a frequent event in numerous malignant diseases, including colorectal, breast and lung cancer and has been related with functional features of cancer cells (reviewed in reference [3]). Current knowledge of the regulatory mechanisms involved in this deregulation mainly arises from fractioned studies generally focusing on a single gene or a short subset of genes. Few studies have pursuit an integrative perspective of the mechanisms underlying the deregulation of the COX pathway during carcinogenesis. Although, somatic mutations are not frequent, epigenetic alterations in the COX pathway, in particular DNA methylation, are recurrent events in several types of cancer (Table 1 and Fig. 3). Cumulated evidences underscore the participation of epigenetic mechanisms in the deregulation of cyclooxygenases in cancer, as well as the downstream biosynthesis and signaling of the main prostaglandins and thromboxanes and will be reviewed here. 2.1. The cyclooxygenases As mentioned above, COX is responsible for the conversion of arachidonic acid to a metabolic intermediate, PGH2, which constitutes the commitment step for prostanoid biosynthesis. The overexpression of COX-2 in tumors is a common feature of numerous cancer cells, but not a universal condition of all tumors. Even if the association between COX-2 expression and a certain type of cancer is very strong, as in colorectal and gastric cancer, a small subset of patients present unaltered levels or downregulation of COX-2. Pioneer studies from the laboratories of Issa and

304

I. Cebola, M.A. Peinado / Progress in Lipid Research 51 (2012) 301–313

Fig. 2. Diagram of the epigenetic profile of a genomic region including a gene and a repeat element. The gene is active in normal cells and exhibits an open chromatin conformation that allows the transcription. The open chromatin conformation is characterized by low nucleosome density, presence of histone modifications associated with activity (i.e.: histone 3 lysine 4 trimethylation) and unmethylated CpG island. Alternatively the repeat element is embedded in closed chromatin. Closed chromatin is characterized by a high compactation and nucleosome density, presence of repressing histone modifications (i.e.: histone 3 lysine 9 trimethylation) and methylation of CpG dinucleotides. In tumor cells, the chromatin conformation (compactation, presence of repressing histone modifications and DNA methylation) of the promoter region of the tumor suppressor gene denotes its silencing, while the remodeled chromatin in the repeat element is associated with reactivation.

Bang revealed that the observed COX-2 downregulation in tumors of a subset of colorectal and gastric cancer patients, respectively, was strongly correlated with CpG island DNA methylation [95,96]. Furthermore, it seems that there is a strong correlation between CpG island methylator phenotype and the gain of methylation in this region in colorectal malignant growths [97]. However, recent studies have reported inconsistent profiles between COX-2 expression and the methylation status of its promoter in colorectal cancer [98], which indicates that other factors may also contribute to the downregulation of this gene in a subset of patients. In gastric cancer, it has been shown that COX-2 downregulation is also mediated by histone deacetylation [99]. Recent analyses report COX-2 CpG island hypermethylation in subsets of patients of several other cancer types, including breast [100,101], hepatic [102–104], nasopharyngeal cancer [105], prostate [106] [107], esophageal [108,109], pancreatic [110] and cervical [111–113]. Cyclooxygenase-1 (COX-1) has only been found epigenetically silenced by promoter hypermethylation in pancreatic tumors [110]. Conversely, it has also been observed that epigenetic mechanisms also contribute to COX-2 overexpression in cancer, such as chronic myelogenous leukemia (CML), which is associated with HDAC activity via CREB transcription factor [114]. Whereas in the colorectal cancer cell line HT-29 treatment with HDAC inhibitors downregulates COX-2 expression involving suppression of RNA polymerase II elongation [115]. Overexpression of COX-2 in some types of breast cancer has been associated with copy number gains [116]. Helicobacter pylori gastritis, which is intimately related to higher gastric cancer risk, has been associated with COX-2 overexpression [96,117]. Moreover, COX-2 deficient gastric epithelial cell lines, which present COX-2 hypermethylation, are less prone to augment COX-2 levels in response to H. pylori than unmethylated cell lines [118]. Following H. pylori infection, several epigenetic alterations contribute to COX-2 reactivation, such as DNA hypomethylation, release of HDACs from COX-2 promoter, increase of H3 acetylation, H3K4 dimethylation, decrease of H3K9 dimethylation and increase of H3K27 trimethylation [119]. In this case, loss of

CpG island methylation might facilitate pathogen-associated inflammation and, as a consequence, increase the risk of gastric cancer. In a pilot study that involved 40 primary gastric cancer tissues, COX-2 methylation associated with longer recurrence times and better overall survival [120]. Regarding cervical cancer is has been observed that the human papillomavirus HPV16 oncoproteins E6 and E7 are able to activate COX-2 transcription by mechanisms that include the dissociation of nuclear receptor corepressor from its promoter [111]. Finally, the presence of COX-2 mRNA in serum and feces DNA has been evaluated as marker for lung [121] and colorectal cancer respectively [122,123] and low levels of COX-2 protein are associated with better response to chemotherapy in rectal adenocarcinoma [124]. As a whole, these data support the notion that targeting the epigenetic control of COX-2 in diagnostic and therapeutic settings may have important clinical applications. 2.2. Prostaglandin E2 biosynthesis and signaling During tumorigenesis, the upregulation of COX-2 leads to a drastic increase in the levels of prostaglandin E2 (PGE2), a bioactive lipid with pleiotropic effects, most of which are related with inflammation (reviewed in reference [2]) and the acquisition of the different hallmarks of cancer [125]. Elevated levels of PGE2 can be found in colorectal, lung, breast, head and neck, and esophageal cancer, frequently correlating with poor prognosis [3,126– 129]. In addition, it has been observed that the administration of PGE2 to AOM mice induces colorectal carcinogenesis [130] and several other studies in murine models have demonstrated the close link between COX-2 derived PGE2 and tumorigenesis in colon. In Min mice, treatments with PGE2 receptor agonists were sufficient to impair NSAIDs-induced tumor regression [131]. There are three known enzymes with PGE2 synthase activity, two microsomal membrane-bound PGE2 synthases (PTGES and PTGES2, also known as mPGES-1 and mPGES-2) and a cytosolic isoform (PTGES3, also known as cPGES). Microsomal PGE2 synthase-1 is the primary responsible for inflammatory responses, being kept at low levels in most tissues, but recurrently upregulated in a number of

305

I. Cebola, M.A. Peinado / Progress in Lipid Research 51 (2012) 301–313 Table 1 Genetic and epigenetic modifiers of the COX pathway regulation in Cancer. Lipid

Gene

Cancer type

Polymorphismsa

PGH2

COX-1 (PTGS1)

Colorectal

L15-L16del [222]; G213G [223]; C639A [224]

Epigenetic alterations

Pancreatic COX-2 (PTGS2)

Bile duct

837T > C [225]

Breast

169C > G [226]; rs5277 [227]; rs7550380 [228]; 8473T>C [229] 837T>C [230]

Bladder CML Colorectal Gallbladder Esophagus Gastric

Glioma Hepatic Lung Nasopharyngeal Non-melanoma skin Ovarian Pancreatic Prostate PGE2

PGI2

PGD2

PTGES EP2 (PTGER2)

EP3 (PTGER3) EP4 (PTGER4) HPGD

Hepatic Gastric Lung Neuroblastoma Colorectal Gastric Colorectal

PTGIS

Breast

PTGDS PTGDR

a b

Hypermethylation of TSS region (not a CpG island) [110]

Hypermethylation [100,101]

Induction by HDACs [114] Hypermethylation [95,123,233,234]

V102 V [231]; rs4648298 [232] 765G>C [235]; 1195G>A [236]

Hypermethylation [108] Hypermethylation [96,99,238] [120,239,240]; Hypomethylation associated with H. pylori infection [118,119]; histone deacetylation [99]; hypomethylation [241]

5939C [237]

rs20417 [242] Hypermethylation [102] 8473 C > C [243] 765 G > C [244] 765G > C and -1195A>G [245] rs5275 [246]

Hypermethylation [105] Hypermethylation [247] Hypermethylation [110] Hypermethylation [106] Hypermethylation Hypermethylation Hypermethylation Hypermethylation Hypermethylation Hypermethylation

[146]b [180] [152,153,248] [151] [155] [180]

rs2612656 and rs8752 [249] rs5602 and rs8183919 [175]; repeat polymorphism [174]

Colorectal Lung

Hypermethylation [177] Hypermethylation [178,179]; Histone deacetylation [179]b

Glioblastoma multiforme Neuroblastoma Osteosarcoma

Hypermethylation [193]

TXA2

TBXAS1 TBXA2R

Breast Colorectal

PGF2a

PTGFR

Breast Colorectal Prostate

Hypermethylation [151] Hypermethylation [201] rs41727 [175] Hypermethylation [208] Hypermethylation [219] Hypermethylation [219]

All the polymorphisms listed are associated with higher cancer risk. Analyzed in cancer cell lines only.

cancers, including colon, lung, stomach, pancreas, cervix, prostate, papillary thyroid carcinoma, head and neck, and brain [132–143]. The tumorigenic role of PTGES is further demonstrated in Min and AOM mice, where deletion of this PGE2 synthase results in the inhibition of tumorigenic development [144,145]. An induction of PTGES expression was found in a hepatocellular carcinoma cell line upon DNMT1 knockdown [146]. This observation could indicate that PTGES role in carcinogenesis is not universal, being its expression less required for hepatic cancer progression. Nevertheless, this result might be also due to a cross-reaction of other signaling pathways, not being a direct effect of PTGES demethylation. There are four PGE2 receptors, EP1, EP2, EP3 and EP4 (also known as PTGER1, PTGER2, PTGER3 and PTGER4, respectively), which mediate a variety of biological functions in several cell types. Among these, EP2 and EP4 have been mostly related with inflammatory and tumorigenic processes. In fact, EP2 is found overexpressed in esophageal, lung and colorectal cancer [128,147,148] and implicated in mouse skin papilloma and in Kaposi’s Sarcoma development [149,150]. However, it is recurrently silenced through promoter

hypermethylation in neuroblastomas and lung cancers [151–153], which suggests that it is not required for tumor progression in these tissues. In addition, it was recently shown in vitro that EP4 is involved in angiogenesis through activation of protein kinase A (PKA) [154]. The EP3 receptor may also undergo epigenetic silencing during carcinogenesis by promoter hypermethylation in mouse, rat and human colorectal tumors [155]. In contrast with COX-2, the responsible for the cytosolic degradation of PGE2, 15-hydroxyprostaglandin dehydrogenase (HPGD), is highly expressed in non-neoplastic tissues and often downregulated in colon, gastric, lung and breast cancer, which contributes for the accumulation of PGE2 in cancer cells [156– 161]. In colorectal cancer, HPGD silencing is mediated by HDACs and, in tumors from patients, elevated HDAC expression correlates with HPGD downregulation [162]. This mechanism of gene silencing seems to be common to colorectal and lung cancer, since the treatment of lung adenocarcinoma cells with HDAC inhibitors induces chromatin remodeling and HPGD re-expression [163].

306

I. Cebola, M.A. Peinado / Progress in Lipid Research 51 (2012) 301–313

Fig. 3. Effectors of the Cox pathway (either synthases or receptors) silenced by promoter DNA hypermethylation in different tumor types. Hypermethylated genes are shown in red boxes and linked to the cancer types in which have been reported to be epigenetically repressed.

In addition to the epigenetic mechanisms mentioned above, there is a growing body of evidence showing that microRNAs are important in the regulation of inflammatory processes and in particular for prostaglandin synthesis, especially at the level of PGE2 synthesis and signaling (reviewed in reference [164]).

2.3. Prostacyclin biosynthesis and signaling Prostacyclin (also referred as prostaglandin I2 or PGI2) is a potent vasodilator and platelet aggregation inhibitor. Mostly studied as putative target for the treatment of cardiovascular system malfunctions [165,166], its role in carcinogenesis remains uncertain. Hypoxia appears to increase the overexpression of prostacyclin synthase (PTGIS, also known as PGIS) in vascular cells, which results in high levels of prostacyclin in highly vascularized tissues [167]. It has been suggested that this mechanism may counteract the negative effects of inflammatory stimuli [167]. Overexpression PTGIS induces apoptosis through nuclear receptor PPARd-specific signaling in HEK 293T cells [168], and it has been shown to be anti-tumorigenic in lung cancer models [169–172]. Furthermore, iloprost, a prostacyclin analog, induces apoptosis in vascular smooth muscle [173]. It was also reported that polymorphisms at PTGIS promoter conferring lower expression levels are significantly associated with higher risk of colorectal cancer and lower chemopreventive action of NSAIDs [174]. In addition, Abraham et al. observed differential susceptibility and survival rates of breast cancer patients with different variants of PTGIS [175]. However, prostacyclin appears to have a complex mechanism of action, which is most likely context-dependent as Cutler et al. observed that the stromal production of prostacyclin in the colon is able to protect epithelial cells from entering apoptosis [176]. Very few studies have addressed the epigenetic mechanisms that modulate PTGIS expression in cancer. We have shown that PTGIS suffers an epigenetic deregulation in colorectal cancer by promoter hypermethylation, which is significantly associated with transcriptional silencing and aneuploidy in several cancer cell lines and human tumors [177]. Subsequent works from Stearman et al.

and Cathcart et al. showed that PTGIS also undergoes epigenetic silencing in lung cancer cell lines and tissues [178,179]. Although at the moment there are no specific reports on the epigenetic regulation of the prostacyclin concomitant surface receptor IP (also known as PTGIR), its mRNA, along with that of two prostaglandin E2 receptors, EP2 and EP4, was found upregulated in a metastatic gastric cancer cell line upon treatment with the DNA-methyl-transferase inhibitor 5-aza-2-deoxycytidine in a cDNA microarray analysis [180]. However, it is not clear whether these genes are directly regulated by epigenetic mechanisms, or if the observed upregulation is only an indirect effect of alterations in the expression of other genes. Further studies should address the potential epigenetic silencing of these genes and their functional implications in tumorigenesis. In addition to the cell-surface receptor, prostacyclin is able to bind to different members of the PPAR nuclear receptor family. Prostacyclin was reported to signal through PPARd [181] and PPARc, being able to induce apoptosis [168] or growth arrest [182] in particular cellular contexts. Yang et al. have reported that RNA interference against PPARd promoted colorectal cancer cell proliferation [183]. However, the role of PPARd in cancer, especially in colorectal and breast cancers, is controversial, and some reports show that PPARd activation may promote cell proliferation and tumor progression [184,185]. At the moment, we have no knowledge of reports on epigenetic mechanisms regulating PPARd expression. As in the case of PPARc, its expression in colorectal tumors correlates with good prognosis [186], but it may also undergo epigenetic silencing through promoter hypermethylation, being this event associated with poor prognosis [187]. 2.4. Prostaglandin D2 biosynthesis and signaling Similarly to prostacyclin, prostaglandin D2 (PGD2) is a vasodilator and inhibits platelet aggregation. Preferentially produced in the brain, it plays an important role in the central nervous system modulation, being important for the regulation of sleep, as well as allergic and pain responses, through PGD2 receptor-mediated signaling [188,189]. Noteworthy, this eicosanoid may suffer

I. Cebola, M.A. Peinado / Progress in Lipid Research 51 (2012) 301–313

further dehydration to produce PGJ2, D12-PGJ2, and 15-deoxyD(12,14)-PGJ2, being the last a natural ligand for the nuclear receptor PPARc (mentioned in the previous section). Two distinct forms of PGD2 synthase have been described: the lipocalin-type (PTGDS, also known as L-PGDS) and the hematopoietic (HPGDS, also known as H-PGDS). The first is mostly found in the central nervous system, male genital organs and heart, where it functions as a lipophilic ligand-binding protein as well. On the other hand, the hematopoietic form, HPGDS, can be found ubiquitously expressed in peripheral tissues, being preferentially expressed in antigen-presenting cells, mast cells, and megakaryocytes [189]. High levels of PTGDS were found in breast, brain and ovarian cancers [190–192]. In contrast, it was recently reported that PTGDS is recurrently downregulated in astrocytomas, by aberrant hypermethylation of the first introns [193], and in lung tumors, although in this case the silencing mechanisms have not been described yet [194]. Prostaglandin D2, may bind and signal through two G proteincoupled receptors, PTGDR (also known as DP1) and CRTH2 (Chemoattractant Receptor-Homologous expressed on T Helper type 2 cells, also referred as DP2). Activation of PTGDR is involved in the regulation of several physiological and pathological processes, such as sleep, allergic responses [195] and, importantly, cell survival and angiogenesis [196,197], whereas CRTH2 is thought to be mainly involved in the induction of chemotaxis in T helper type 2 cells, eosinophils, and basophils [198]. Apparently, only PTGDR is subjected to gene expression modulationin colorectalcancer,being significantly downregulatedin tumors [199]. Interestingly, when characterizing the PGD2 receptors expression in a panel of colorectal cancer cell lines, Hawcroft et al. could only detect expression of PTGDR in one cell line [200]. Although the mechanisms responsible of the downregulation or silencing of this gene in colorectal cancer have not been studied yet, its promoter was already found hypermethylated in osteosarcoma and neuroblastoma cell lines [151,201], being this mechanism likely to mediate PTGDR silencing in colorectal carcinogenesis as well.

307

pressure, renal filtration, induction of luteolysis, inhibition of adipocyte differentiation and hair growth stimulation [209]. In addition, PGF2a has been shown to promote carcinogen-induced malignant transformation in C3H/M2 mouse fibroblasts [210]. Moreover, colorectal cancer cells produce high levels of this prostanoid, which, in this context, is able to stimulate cell motility and invasion [211]. The biosynthesis of PGF2a may occur via three distinct pathways: from PGE2, PGD2 or PGH2, by PGE 9-ketoreductase (AKR1C5), PGD 11-ketoreductase (AKR1C3) or PGH 9-, 11-endoperoxide reductase (AKR1C3), respectively. Despite the abundance of PGF2a synthases, most of them belong to the aldo–keto reductase superfamily [209,212,213]. Of particular interest from the therapeutic point of view is AKR1B1 (aldo–keto reductase family 1 member B1), which is the major responsible for human endometrium PGF2a production [214] and may be implicated in hepatic tumorigenesis [215]. Conversely, its silencing has been reported in sporadic adrenocortical and colorectal tumors [216,217]. In a comparative study of colorectal cancer samples, AKR1B10, another member of the AKR1B family, with weaker PGF2a synthase activity [209], was found downregulated in 88% of the analyzed tumors [217]. So far, we have no knowledge of studies regarding the epigenetic regulation of the PGF2a synthases. Similarly to other prostanoids, PGF2a binds to a cognate G protein-coupled receptor (PTGFR, also known as FP). The PTGFR-mediated signaling has been related with enhanced epithelial cell proliferation in endometrial adenocarcinoma [218]. However, in a genome-wide analysis of cancer cell methylation, the PTGFR promoter was found epigenetically silenced by promoter methylation in a prostate cancer cell line and in colorectal tumors [219]. Loss of heterozygosity affecting this gene is frequent in human breast tumors, supporting its possible involvement in cancer [220]. Importantly, PGF2a is also able to bind to the PGE2 receptors EP1 and EP3 with high affinity [221], adding further complexity into the understanding of the biological role of PGF2a.

2.5. Thromboxane A2 biosynthesis and signaling In opposition to prostacyclin, thromboxane A2 (TXA2) is a powerful vasoconstrictor and platelet aggregation mediator. Thromboxane synthase (TBXAS1, also known as TXAS) overexpression has been observed in a number of cancer types, including papillary thyroid carcinoma [202], bladder [203], prostate [204] and colorectal [205], and its expression is generally negatively associated with survival. When comparing different human cell lines, Lee et al. found cell-specific signatures in the methylation pattern of the TBXAS1 promoter. Surprisingly, higher expression levels could only be observed in the cell lines that presented hypermethylation of this promoter [206]. The thromboxane receptor (TP, also known as TBXA2R) has been found commonly expressed in breast cancer tumors [207]. On the other hand, in a molecular screening designed to detect alterations in the patterns of methylation, Paz et al. identified the thromboxane receptor as a putative target of promoter hypermethylation and transcriptional silencing in colorectal cancer. In the same study, re-introduction of this receptor in the colorectal cancer cell line HCT116 resulted in colony formation inhibition [208]. According to these results, the role of TXA2 varies amongst different cancer types and epigenetic mechanisms may be involved in the regulation of both synthesis and membrane–receptor-mediated activity. 2.6. Prostaglandin F2a biosynthesis and signaling Prostaglandin F2a (PGF2a) is involved in biological processes as diverse as smooth muscle contraction, regulation of intraocular

3. Concluding remarks The COX pathway is involved in the regulation of multiple cellular functions that are critical in the tumorigenic process (Fig. 1). This implies that its study represents a prime target in all the strategies against cancer, from prevention and risk assessment to diagnosis, prognosis and treatment. With the advent of epigenetic analyses in the last decade, it has been uncovered that promoter DNA methylation, histone deacetylation and microRNA differential expression, among other modifications, participate in the abnormal expression of several members of the COX pathway during carcinogenesis (summarized in Table 1 and Fig. 3). Despite the abundant literature pointing up the deregulation of one or more of these genes in a particular type of cancer, there is a lack of studies targeting the analysis of this complex pathway as a functional unit. Comprehensive epigenetic profiling of the elements of the COX pathway is needed to delineate an integrated chart of the pathway regulation and the cellular mechanisms modulated by the different prostaglandins and their ligands in physiological but especially in pathological situations, as inflammation and cancer. Gene silencing by DNA hypermethylation of the promoter-associated CpG island of different genes of the COX pathway is the most frequently reported alteration in particular types of cancer. This is probably due to technical constrictions, since, so far, DNA methylation is the most accessible epigenetic mark to be tested in tissue samples. Technical advances are needed to analyze other epigenetic modifications in a consistent way.

308

I. Cebola, M.A. Peinado / Progress in Lipid Research 51 (2012) 301–313

Fig. 4. Deregulation of the COX pathway in cancer. It is known that lifestyle factors, such as high-fat diets and tobacco smoking, particular genetic backgrounds, and epigenetic alterations may contribute to the deregulation of the COX pathway in chronic inflammation pathologies and cancer. Most often, this deregulation results in a generalized overproduction and downstream cognate receptor-mediated activity of prostaglandin E2 over the other possible products of this metabolic pathway (other PG, other prostaglandins).

The epigenetic deregulation of some of effectors of the COX pathway in inflammation and cancer collapses the balance between prostaglandins and results in loss of compensating mechanisms (Fig. 4), which results in functional impairments. Epigenetics acts as a bidirectional control of the COX pathway that gauges and modulates the impact of exogenous and endogenous factors at multiple levels. Hence, the understanding of the contribution of environmental factors and the genetic background to inflammation and cancer cannot be achieved without a deep understanding of the epigenetic mechanisms involved in the fine regulation and integration of environmental and molecular information. The development and application of personalized and more effective therapies in cancer is based in a better knowledge of the molecular mechanisms underlying disease and normal physiology. Future studies should tackle the complete profiling of the COX pathway at functional level, and this includes the epigenetic mechanisms controlling its different components. Acknowledgements I.C. was supported by a fellowship from the Fundação para a Ciência e a Tecnologia. Research in the lab was supported by grants from the Spanish Ministry of Science and Innovation (SAF2008/ 1409, SAF2011/23638, CSD2006/49) and Generalitat de Catalunya (2009 SGR 1356). References [1] Woutersen RA, Appel MJ, van Garderen-Hoetmer A, Wijnands MV. Dietary fat and carcinogenesis. Mutat Res 1999;443:111–27. [2] Greene ER, Huang S, Serhan CN, Panigrahy D. Regulation of inflammation in cancer by eicosanoids. Prostaglandins Other Lipid Mediators 2011;96:27–36. [3] Wang D, Dubois RN. Eicosanoids and cancer. Nat Rev Cancer 2010;10:181–93. [4] Eberhart CE, Coffey RJ, Radhika A, Giardiello FM, Ferrenbach S, Dubois RN. Upregulation of cyclooxygenase 2 gene expression in human colorectal adenomas and adenocarcinomas. Gastroenterology 1994;107:1183–8. [5] Ristimäki A, Honkanen N, Jänkälä H, Sipponen P, Härkönen M. Expression of cyclooxygenase-2 in human gastric carcinoma. Cancer Res 1997;57:1276–80. [6] Wolff H, Saukkonen K, Anttila S, Karjalainen A, Vainio H, Ristimäki A. Expression of cyclooxygenase-2 in human lung carcinoma. Cancer Res 1998;58:4997–5001. [7] Liu XH, Rose DP. Differential expression and regulation of cyclooxygenase-1 and -2 in two human breast cancer cell lines. Cancer Res 1996;56:5125–7. [8] Zidar N, Odar K, Glavac D, Jerse M, Zupanc T, Stajer D. Cyclooxygenase in normal human tissues: is COX-1 really a constitutive isoform, and COX-2 an inducible isoform? J Cell Mol Med 2009;13:3753–63. [9] Bauer AK, Dwyer-Nield LD, Malkinson AM. High cyclooxygenase 1 (COX-1) and cyclooxygenase 2 (COX-2) contents in mouse lung tumors. Carcinogenesis 2000;21:543–50. [10] Nurmi JT, Puolakkainen PA, Rautonen NE. Intron 1 retaining cyclooxygenase 1 splice variant is induced by osmotic stress in human intestinal epithelial cells. Prostaglandins Leukotrienes Essent Fatty Acids 2005;73:343–50. [11] Chell S, Kaidi A, Williams AC, Paraskeva C. Mediators of PGE2 synthesis and signalling downstream of COX-2 represent potential targets for the

[12] [13] [14]

[15]

[16]

[17]

[18]

[19]

[20] [21]

[22]

[23]

[24]

[25]

[26]

[27]

[28]

[29]

[30]

prevention/treatment of colorectal cancer. Biochim Biophys Acta 2006;1766:104–19. Prescott SM, Fitzpatrick FA. Cyclooxygenase-2 and carcinogenesis. Biochim Biophys Acta 2000;1470:M69–78. Huang RY, Chen GG. Cigarette smoking, cyclooxygenase-2 pathway and cancer. Biochim Biophys Acta 2011;1815:158–69. Buckman SY, Gresham A, Hale P, Hruza G, Anast J, Masferrer J, et al. COX-2 expression is induced by UVB exposure in human skin: implications for the development of skin cancer. Carcinogenesis 1998;19:723–9. Weylandt KH, Krause LF, Gomolka B, Chiu C-Y, Bilal S, Nadolny A, et al. Suppressed liver tumorigenesis in fat-1 mice with elevated omega-3 fatty acids is associated with increased omega-3 derived lipid mediators and reduced TNF-{alpha}. Carcinogenesis 2011. Walker DR, Bond JP, Tarone RE, Harris CC, Makalowski W, Boguski MS, et al. Evolutionary conservation and somatic mutation hotspot maps of p53: correlation with p53 protein structural and functional features. Oncogene 1999;18:211–8. Funahashi H, Satake M, Hasan S, Sawai H, Newman RA, Reber HA, et al. Opposing effects of n-6 and n-3 polyunsaturated fatty acids on pancreatic cancer growth. Pancreas 2008;36:353–62. Angelucci A, Garofalo S, Speca S, Bovadilla A, Gravina GL, Muzi P, et al. Arachidonic acid modulates the crosstalk between prostate carcinoma and bone stromal cells. Endocr Relat Cancer 2008;15:91–100. Hughes-Fulford M, Chen Y, Tjandrawinata RR. Fatty acid regulates gene expression and growth of human prostate cancer PC-3 cells. Carcinogenesis 2001;22:701–7. Rose DP, Connolly JM. Omega-3 fatty acids as cancer chemopreventive agents. Pharmacol Ther 1999;83:217–44. Chang Y-WE, Putzer K, Ren L, Kaboord B, Chance TW, Qoronfleh MW, et al. Differential regulation of cyclooxygenase 2 expression by small GTPases Ras, Rac1, and RhoA. J Cell Biochem 2005;96:314–29. Coffey RJ, Hawkey CJ, Damstrup L, Graves-Deal R, Daniel VC, Dempsey PJ, et al. Epidermal growth factor receptor activation induces nuclear targeting of cyclooxygenase-2, basolateral release of prostaglandins, and mitogenesis in polarizing colon cancer cells. Proc Natl Acad Sci U S A 1997;94:657–62. Moraitis D, Du B, De Lorenzo MS, Boyle JO, Weksler BB, Cohen EG, et al. Levels of cyclooxygenase-2 are increased in the oral mucosa of smokers: evidence for the role of epidermal growth factor receptor and its ligands. Cancer Res 2005;65:664–70. Reed JR, Leon RP, Hall MK, Schwertfeger KL. Interleukin-1beta and fibroblast growth factor receptor 1 cooperate to induce cyclooxygenase-2 during early mammary tumourigenesis. Breast Cancer Res 2009;11:R21. Chang K-Y, Shen M-R, Lee M-Y, Wang W-L, Su W-C, Chang W-C, et al. Epidermal growth factor-activated aryl hydrocarbon receptor nuclear translocator/HIF-1{beta} signal pathway up-regulates cyclooxygenase-2 gene expression associated with squamous cell carcinoma. J Biol Chem 2009;284:9908–16. Saha D, Datta PK, Sheng H, Morrow JD, Wada M, Moses HL, et al. Synergistic induction of cyclooxygenase-2 by transforming growth factor-beta1 and epidermal growth factor inhibits apoptosis in epithelial cells. Neoplasia 1999;1:508–17. Liu W, Reinmuth N, Stoeltzing O, Parikh AA, Tellez C, Williams S, et al. Cyclooxygenase-2 is up-regulated by interleukin-1 beta in human colorectal cancer cells via multiple signaling pathways. Cancer Res 2003;63:3632–6. Konturek PC, Hartwich A, Zuchowicz M, Labza H, Pierzchalski P, Karczewska E, et al. Helicobacter pylori, gastrin and cyclooxygenases in gastric cancer. J Physiol Pharmacol 2000;51:737–49. Chang YJ, Wu MS, Lin JT, Sheu BS, Muta T, Inoue H, et al. Induction of cyclooxygenase-2 overexpression in human gastric epithelial cells by Helicobacter pylori involves TLR2/TLR9 and c-Src-dependent nuclear factorkappaB activation. Mol Pharmacol 2004;66:1465–77. Abdel-Latif MMM, Windle H, Terres A, Eidhin DN, Kelleher D, Reynolds JV. Helicobacter pylori extract induces nuclear factor-kappa B, activator protein-1,

I. Cebola, M.A. Peinado / Progress in Lipid Research 51 (2012) 301–313

[31]

[32]

[33]

[34]

[35]

[36]

[37] [38]

[39]

[40]

[41]

[42]

[43]

[44]

[45]

[46]

[47]

[48]

[49]

[50]

[51]

[52]

[53]

[54]

and cyclooxygenase-2 in esophageal epithelial cells. J Gastrointest Surg 2006;10:551–62. Singh A, Sharma H, Salhan S, Gupta SD, Bhatla N, Jain SK, et al. Evaluation of expression of apoptosis-related proteins and their correlation with HPV, telomerase activity, and apoptotic index in cervical cancer. Pathobiology 2004;71:314–22. Cheng AS-L, Chan HL-Y, Leung WK, To KF, Go MY-Y, Chan JY-H, et al. Expression of HBx and COX-2 in chronic hepatitis B, cirrhosis and hepatocellular carcinoma: implication of HBx in upregulation of COX-2. Mod Pathol 2004;17:1169–79. Kaul R, Verma SC, Murakami M, Lan K, Choudhuri T, Robertson ES. Epstein– Barr virus protein can upregulate cyclo-oxygenase-2 expression through association with the suppressor of metastasis Nm23-H1. J Virol 2006;80:1321–31. Lara-Pezzi E, Gómez-Gaviro MV, Gálvez BG, Mira E, Iñiguez MA, Fresno M, et al. The hepatitis B virus X protein promotes tumor cell invasion by inducing membrane-type matrix metalloproteinase-1 and cyclooxygenase-2 expression. J Clin Invest 2002;110:1831–8. Waris G, Siddiqui A. Hepatitis C virus stimulates the expression of cyclooxygenase-2 via oxidative stress: role of prostaglandin E2 in RNA replication. J Virol 2005;79:9725–34. Murono S, Inoue H, Tanabe T, Joab I, Yoshizaki T, Furukawa M, et al. Induction of cyclooxygenase-2 by Epstein–Barr virus latent membrane protein 1 is involved in vascular endothelial growth factor production in nasopharyngeal carcinoma cells. Proc Natl Acad Sci U S A 2001;98:6905–10. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646–74. Battu S, Rigaud M, Beneytout JL. Resistance to apoptosis and cyclooxygenase2 expression in a human adenocarcinoma cell line HT29 CL.19A. Anticancer Res 1998;18:3579–83. Tsujii M, DuBois RN. Alterations in cellular adhesion and apoptosis in epithelial cells overexpressing prostaglandin endoperoxide synthase 2. Cell 1995;83:493–501. Tang X, Sun YJ, Half E, Kuo MT, Sinicrope F. Cyclooxygenase-2 overexpression inhibits death receptor 5 expression and confers resistance to tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis in human colon cancer cells. Cancer Res 2002;62:4903–8. Totzke G, Schulze-Osthoff K, Jänicke RU. Cyclooxygenase-2 (COX-2) inhibitors sensitize tumor cells specifically to death receptor-induced apoptosis independently of COX-2 inhibition. Oncogene 2003;22:8021–30. Yamanaka Y, Shiraki K, Inoue T, Miyashita K, Fuke H, Yamaguchi Y, et al. COX2 inhibitors sensitize human hepatocellular carcinoma cells to TRAIL-induced apoptosis. Int J Mol Med 2006;18:41–7. Martin S, Phillips DC, Szekely-Szucs K, Elghazi L, Desmots F, Houghton JA. Cyclooxygenase-2 inhibition sensitizes human colon carcinoma cells to TRAIL-induced apoptosis through clustering of DR5 and concentrating death-inducing signaling complex components into ceramide-enriched caveolae. Cancer Res 2005;65:11447–58. Pockaj BA, Basu GD, Pathangey LB, Gray RJ, Hernandez JL, Gendler SJ, et al. Reduced T-cell and dendritic cell function is related to cyclooxygenase-2 overexpression and prostaglandin E2 secretion in patients with breast cancer. Ann Surg Oncol 2004;11:328–39. Pai R, Soreghan B, Szabo IL, Pavelka M, Baatar D, Tarnawski AS. Prostaglandin E2 transactivates EGF receptor: a novel mechanism for promoting colon cancer growth and gastrointestinal hypertrophy. Nat Med 2002;8:289–93. Kinoshita T, Takahashi Y, Sakashita T, Inoue H, Tanabe T, Yoshimoto T. Growth stimulation and induction of epidermal growth factor receptor by overexpression of cyclooxygenases 1 and 2 in human colon carcinoma cells. Biochim Biophys Acta 1999;1438:120–30. Zhao Y, Agarwal VR, Mendelson CR, Simpson ER. Estrogen biosynthesis proximal to a breast tumor is stimulated by PGE2 via cyclic AMP, leading to activation of promoter II of the CYP19 (aromatase) gene. Endocrinology 1996;137:5739–42. Castellone MD, Teramoto H, Williams BO, Druey KM, Gutkind JS. Prostaglandin E2 promotes colon cancer cell growth through a Gs-axinbeta-catenin signaling axis. Science 2005;310:1504–10. Gallo O, Franchi A, Magnelli L, Sardi I, Vannacci A, Boddi V, et al. Cyclooxygenase-2 pathway correlates with VEGF expression in head and neck cancer. implications for tumor angiogenesis and metastasis. Neoplasia 2001;3:53–61. Masferrer JL, Leahy KM, Koki AT, Zweifel BS, Settle SL, Woerner BM, et al. Antiangiogenic and antitumor activities of cyclooxygenase-2 inhibitors. Cancer Res 2000;60:1306–11. Leung WK, To KF, Go MYY, Chan K-K, Chan FKL, Ng EKW, et al. Cyclooxygenase-2 upregulates vascular endothelial growth factor expression and angiogenesis in human gastric carcinoma. Int J Oncol 2003;23:1317–22. Sun WH, Sun YL, Fang RN, Shao Y, Xu HC, Xue QP, et al. Expression of cyclooxygenase-2 and matrix metalloproteinase-9 in gastric carcinoma and its correlation with angiogenesis. Jpn J Clin Oncol 2005;35:707–13. Tsujii M, Kawano S, DuBois RN. Cyclooxygenase-2 expression in human colon cancer cells increases metastatic potential. Proc Natl Acad Sci U S A 1997;94:3336–40. Sivula A, Talvensaari-Mattila A, Lundin J, Joensuu H, Haglund C, Ristimäki A, et al. Association of cyclooxygenase-2 and matrix metalloproteinase-2

[55]

[56]

[57]

[58]

[59]

[60]

[61]

[62] [63] [64] [65]

[66] [67]

[68]

[69] [70]

[71] [72] [73] [74] [75] [76] [77] [78] [79]

[80]

[81] [82]

[83]

[84] [85] [86]

309

expression in human breast cancer. Breast Cancer Res Treat 2005;89:215–20. Byun JH, Lee MA, Roh SY, Shim BY, Hong SH, Ko YH, et al. Association between cyclooxygenase-2 and matrix metalloproteinase-2 expression in non-small cell lung cancer. Jpn J Clin Oncol 2006;36:263–8. Larkins TL, Nowell M, Singh S, Sanford GL. Inhibition of cyclooxygenase-2 decreases breast cancer cell motility, invasion and matrix metalloproteinase expression. BMC Cancer 2006;6:181. Kinugasa Y, Hatori M, Ito H, Kurihara Y, Ito D, Nagumo M. Inhibition of cyclooxygenase-2 suppresses invasiveness of oral squamous cell carcinoma cell lines via down-regulation of matrix metalloproteinase-2 and CD44. Clin Exp Metastasis 2004;21:737–45. Oshima M, Dinchuk JE, Kargman SL, Oshima H, Hancock B, Kwong E, et al. Suppression of intestinal polyposis in Apc delta716 knockout mice by inhibition of cyclooxygenase 2 (COX-2). Cell 1996;87:803–9. Chulada PC, Thompson MB, Mahler JF, Doyle CM, Gaul BW, Lee C, et al. Genetic disruption of PTGS-1, as well as PTGS-2, reduces intestinal tumorigenesis in Min mice. Cancer Res 2000;60:4705–8. Tanaka T, Kohno H, Suzuki R, Yamada Y, Sugie S, Mori H. A novel inflammation-related mouse colon carcinogenesis model induced by azoxymethane and dextran sodium sulfate. Cancer Sci 2003;94:965–73. Harris RE. Cyclooxygenase-2 (cox-2) blockade in the chemoprevention of cancers of the colon, breast, prostate, and lung. Inflammopharmacology 2009;17:55–67. Arber N, Levin B. Chemoprevention of colorectal neoplasia: the potential for personalized medicine. Gastroenterology 2008;134:1224–37. Chan AT, Giovannucci EL. Primary prevention of colorectal cancer. Gastroenterology 2010;138(2029–43):e10. Giardiello FM. Sulindac and polyp regression. Cancer Metastasis Rev 1994;13:279–83. Warner TD, Giuliano F, Vojnovic I, Bukasa A, Mitchell JA, Vane JR. Nonsteroid drug selectivities for cyclo-oxygenase-1 rather than cyclo-oxygenase-2 are associated with human gastrointestinal toxicity: a full in vitro analysis. Proc Natl Acad Sci U S A 1999;96:7563–8. Patrignani P, Capone ML, Tacconelli S. NSAIDs and cardiovascular disease. Heart 2008;94:395–7. Arehart E, Stitham J, Asselbergs FW, Douville K, MacKenzie T, Fetalvero KM, et al. Acceleration of cardiovascular disease by a dysfunctional prostacyclin receptor mutation: potential implications for cyclooxygenase-2 inhibition. Circ Res 2008;102:986–93. Vogel U, Christensen J, Wallin H, Friis S, Nexø BA, Raaschou-Nielsen O, et al. Polymorphisms in genes involved in the inflammatory response and interaction with NSAID use or smoking in relation to lung cancer risk in a prospective study. Mutat Res 2008;639:89–100. Tegeder I, Pfeilschifter J, Geisslinger G. Cyclooxygenase-independent actions of cyclooxygenase inhibitors. FASEB J 2001;15:2057–72. Din FVN, Dunlop MG, Stark LA. Evidence for colorectal cancer cell specificity of aspirin effects on NF kappa B signalling and apoptosis. Br J Cancer 2004;91:381–8. Goldberg AD, Allis CD, Bernstein E. Epigenetics: a landscape takes shape. Cell 2007;128:635–8. Esteller M. Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet 2007;8:286–98. Jones PA, Baylin SB. The epigenomics of cancer. Cell 2007;128:683–92. Felsenfeld G, Groudine M. Controlling the double helix. Nature 2003;421:448–53. Portela A, Esteller M. Epigenetic modifications and human disease. Nat Biotechnol 2010;28:1057–68. Bird A. DNA methylation patterns and epigenetic memory. Genes Dev 2002;16:6–21. Goll MG, Bestor TH. Eukaryotic cytosine methyltransferases. Annu Rev Biochem 2005;74:481–514. Jones PA, Takai D. The role of DNA methylation in mammalian epigenetics. Science 2001;293:1068–70. Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet 2003;33(Suppl):245–54. Bernstein BE, Stamatoyannopoulos JA, Costello JF, Ren B, Milosavljevic A, Meissner A, et al. The NIH roadmap epigenomics mapping consortium. Nat Biotechnol 2010;28:1045–8. Feinberg AP. Phenotypic plasticity and the epigenetics of human disease. Nature 2007;447:433–40. Irizarry RA, Ladd-Acosta C, Wen B, Wu Z, Montano C, Onyango P, et al. The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet 2009;41: 178–86. Doi A, Park IH, Wen B, Murakami P, Aryee MJ, Irizarry R, et al. Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts. Nat Genet 2009. Feinberg AP, Tycko B. The history of cancer epigenetics. Nat Rev Cancer 2004;4:143–53. Herman JG, Baylin SB. Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med 2003;349:2042–54. Esteller M. Epigenetics in cancer. N Engl J Med 2008;358:1148–59.

310

I. Cebola, M.A. Peinado / Progress in Lipid Research 51 (2012) 301–313

[87] Ehrlich M. DNA methylation in cancer: too much, but also too little. Oncogene 2002;21:5400–13. [88] Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Res 2011;21:381–95. [89] Kouzarides T. Chromatin modifications and their function. Cell 2007;128:693–705. [90] Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 2006;125:315–26. [91] Barrero MJ, Boue S, Izpisua Belmonte JC. Epigenetic mechanisms that regulate cell identity. Cell Stem Cell 2010;7:565–70. [92] Widschwendter M, Fiegl H, Egle D, Mueller-Holzner E, Spizzo G, Marth C, et al. Epigenetic stem cell signature in cancer. Nat Genet 2007;39:157–8. [93] Ohm JE, McGarvey KM, Yu X, Cheng L, Schuebel KE, Cope L, et al. A stem celllike chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing. Nat Genet 2007;39:237–42. [94] Rodriguez J, Muñoz M, Vives L, Frangou CG, Groudine M, Peinado MA. Bivalent domains enforce transcriptional memory of DNA methylated genes in cancer cells. Proc Natl Acad Sci U S A 2008;105:19809–14. [95] Toyota M, Shen L, Ohe-Toyota M, Hamilton SR, Sinicrope FA, Issa JP. Aberrant methylation of the cyclooxygenase 2 CpG island in colorectal tumors. Cancer Res 2000;60:4044–8. [96] Song SH, Jong HS, Choi HH, Inoue H, Tanabe T, Kim NK, et al. Transcriptional silencing of cyclooxygenase-2 by hyper-methylation of the 50 CpG island in human gastric carcinoma cells. Cancer Res 2001;61:4628–35. [97] Kim JC, Choi JS, Roh SA, Cho DH, Kim TW, Kim YS. Promoter methylation of specific genes is associated with the phenotype and progression of colorectal adenocarcinomas. Ann Surg Oncol 2010;17:1767–76. [98] Asting AG, Caren H, Andersson M, Lonnroth C, Lagerstedt K, Lundholm K. COX-2 gene expression in colon cancer tissue related to regulating factors and promoter methylation status. BMC Cancer 2011;11:238. [99] Kikuchi T, Itoh F, Toyota M, Suzuki H, Yamamoto H, Fujita M, et al. Aberrant methylation and histone deacetylation of cyclooxygenase 2 in gastric cancer. Int J Cancer 2002;97:272–7. [100] Ma X, Yang Q, Wilson KT, Kundu N, Meltzer SJ, Fulton AM. Promoter methylation regulates cyclooxygenase expression in breast cancer. Breast Cancer Res 2004;6:R316–21. [101] Chow LWC, Zhu L, Loo WTY, Lui ELH. Aberrant methylation of cyclooxygenase-2 in breast cancer patients. Biomed Pharmacother 2005;59(Suppl 2):S264–7. [102] Murata H, Tsuji S, Tsujii M, Sakaguchi Y, Fu HY, Kawano S, et al. Promoter hypermethylation silences cyclooxygenase-2 (Cox-2) and regulates growth of human hepatocellular carcinoma cells. Lab Invest 2004;84:1050–9. [103] Nishida N, Nagasaka T, Nishimura T, Ikai I, Boland CR, Goel A. Aberrant methylation of multiple tumor suppressor genes in aging liver, chronic hepatitis, and hepatocellular carcinoma. Hepatology 2008;47:908–18. [104] Um TH, Kim H, Oh BK, Kim MS, Kim KS, Jung G, et al. Aberrant CpG island hypermethylation in dysplastic nodules and early HCC of hepatitis B virusrelated human multistep hepatocarcinogenesis. J Hepatol 2011;54: 939–47. [105] Soo R, Putti T, Tao Q, Goh B-C, Lee K-H, Kwok-Seng L, et al. Overexpression of cyclooxygenase-2 in nasopharyngeal carcinoma and association with epidermal growth factor receptor expression. Arch Otolaryngol Head Neck Surg 2005;131:147–52. [106] Bastian PJ, Ellinger J, Heukamp LC, Kahl P, Müller SC, von Rücker A. Prognostic value of CpG island hypermethylation at PTGS2, RAR-beta, EDNRB, and other gene loci in patients undergoing radical prostatectomy. Eur Urol 2007;51:665–74. [107] Lodygin D, Epanchintsev A, Menssen A, Diebold J, Hermeking H. Functional epigenomics identifies genes frequently silenced in prostate cancer. Cancer Res 2005;65:4218–27. [108] Dawsey SP, Roth MJ, Adams L, Hu N, Wang Q-H, Taylor PR, et al. COX-2 (PTGS2) gene methylation in epithelial, subepithelial lymphocyte and stromal tissue compartments in a spectrum of esophageal squamous neoplasia. Cancer Detect Prev 2008;32:135–9. [109] Meng XY, Zhu ST, Zong Y, Wang YJ, Li P, Zhang ST. Promoter hypermethylation of cyclooxygenase-2 gene in esophageal squamous cell carcinoma. Dis Esophagus 2011;24:444–9. [110] Omura N, Griffith M, Vincent A, Li A, Hong S-M, Walter K, et al. Cyclooxygenase-deficient pancreatic cancer cells use exogenous sources of prostaglandins. Mol Cancer Res 2010;8:821–32. [111] Subbaramaiah K, Dannenberg AJ. Cyclooxygenase-2 transcription is regulated by human papillomavirus 16 E6 and E7 oncoproteins: evidence of a corepressor/coactivator exchange. Cancer Res 2007;67:3976–85. [112] Kulkarni S, Rader JS, Zhang F, Liapis H, Koki AT, Masferrer JL, et al. Cyclooxygenase-2 is overexpressed in human cervical cancer. Clin Cancer Res 2001;7:429–34. [113] Ryu HS, Chang KH, Yang HW, Kim MS, Kwon HC, Oh KS. High cyclooxygenase2 expression in stage IB cervical cancer with lymph node metastasis or parametrial invasion. Gynecol Oncol 2000;76:320–5. [114] Kalle AM, Sachchidanand S, Pallu R. Bcr-Abl-independent mechanism of resistance to imatinib in K562 cells: induction of cyclooxygenase-2 (COX-2) by histone deacetylases (HDACs). Leuk Res 2010;34:1132–8. [115] Tong X, Yin L, Joshi S, Rosenberg DW, Giardina C. Cyclooxygenase-2 regulation in colon cancer cells: modulation of RNA polymerase II elongation by histone deacetylase inhibitors. J Biol Chem 2005;280:15503–9.

[116] Stange DE, Radlwimmer B, Schubert F, Traub F, Pich A, Toedt G, et al. Highresolution genomic profiling reveals association of chromosomal aberrations on 1q and 16p with histologic and genetic subgroups of invasive breast cancer. Clin Cancer Res 2006;12:345–52. [117] Fu S, Ramanujam KS, Wong A, Fantry GT, Drachenberg CB, James SP, et al. Increased expression and cellular localization of inducible nitric oxide synthase and cyclooxygenase 2 in Helicobacter pylori gastritis. Gastroenterology 1999;116:1319–29. [118] Akhtar M, Cheng Y, Magno RM, Ashktorab H, Smoot DT, Meltzer SJ, et al. Promoter methylation regulates Helicobacter pylori-stimulated cyclooxygenase-2 expression in gastric epithelial cells. Cancer Res 2001;61:2399–403. [119] Pero R, Peluso S, Angrisano T, Tuccillo C, Sacchetti S, Keller S, et al. Chromatin and DNA methylation dynamics of Helicobacter pylori-induced COX-2 activation. Int J Med Microbiol 2011;301:140–9. [120] de Maat MFG, van de Velde CJH, Umetani N, de Heer P, Putter H, van Hoesel AQ, et al. Epigenetic silencing of cyclooxygenase-2 affects clinical outcome in gastric cancer. J Clin Oncol 2007;25:4887–94. [121] Ulivi P, Mercatali L, Zoli W, Dell’amore D, Poletti V, Casoni GL, et al. Serum free DNA and COX-2 mRNA expression in peripheral blood for lung cancer detection. Thorax 2008;63:843–4. [122] Kanaoka S, Yoshida K, Miura N, Sugimura H, Kajimura M. Potential usefulness of detecting cyclooxygenase 2 messenger RNA in feces for colorectal cancer screening. Gastroenterology 2004;127:422–7. [123] Leung WK, To K-F, Man EPS, Chan MWY, Hui AJ, Ng SSM, et al. Detection of hypermethylated DNA or cyclooxygenase-2 messenger RNA in fecal samples of patients with colorectal cancer or polyps. Am J Gastroenterol 2007;102:1070–6. [124] Edden Y, Wexner SD, Berho M. The use of molecular markers as a method to predict the response to neoadjuvant therapy for advanced stage rectal adenocarcinoma. Colorectal Dis 2011. [125] Greenhough A, Smartt HJM, Moore AE, Roberts HR, Williams AC, Paraskeva C, et al. The COX-2/PGE2 pathway: key roles in the hallmarks of cancer and adaptation to the tumour microenvironment. Carcinogenesis 2009;30:377–86. [126] Rigas B, Goldman IS, Levine L. Altered eicosanoid levels in human colon cancer. J Lab Clin Med 1993;122:518–23. [127] Pugh S, Thomas GA. Patients with adenomatous polyps and carcinomas have increased colonic mucosal prostaglandin E2. Gut 1994;35:675–8. [128] Kuo K-T, Wang H-W, Chou T-Y, Hsu W-H, Hsu H-S, Lin C-H, et al. Prognostic role of PGE2 receptor EP2 in esophageal squamous cell carcinoma. Ann Surg Oncol 2009;16:352–60. [129] Hambek M, Baghi M, Wagenblast J, Schmitt J, Baumann H, Knecht R. Inverse correlation between serum PGE2 and T classification in head and neck cancer. Head Neck 2007;29:244–8. [130] Kawamori T, Uchiya N, Sugimura T, Wakabayashi K. Enhancement of colon carcinogenesis by prostaglandin E2 administration. Carcinogenesis 2003;24:985–90. [131] Hansen-Petrik MB, McEntee MF, Jull B, Shi H, Zemel MB, Whelan J. Prostaglandin E(2) protects intestinal tumors from nonsteroidal antiinflammatory drug-induced regression in Apc(Min/+) mice. Cancer Res 2002;62:403–8. [132] Kamei D, Murakami M, Nakatani Y, Ishikawa Y, Ishii T, Kudo I. Potential role of microsomal prostaglandin E synthase-1 in tumorigenesis. J Biol Chem 2003;278:19396–405. [133] Yoshimatsu K, Altorki NK, Golijanin D, Zhang F, Jakobsson PJ, Dannenberg AJ, et al. Inducible prostaglandin E synthase is overexpressed in non-small cell lung cancer. Clin Cancer Res 2001;7:2669–74. [134] Yoshimatsu K, Golijanin D, Paty PB, Soslow RA, Jakobsson PJ, DeLellis RA, et al. Inducible microsomal prostaglandin E synthase is overexpressed in colorectal adenomas and cancer. Clin Cancer Res 2001;7:3971–6. [135] Gudis K, Tatsuguchi A, Wada K, Hiratsuka T, Futagami S, Fukuda Y, et al. Clinical significance of prostaglandin E synthase expression in gastric cancer tissue. Hum Pathol 2007;38:1826–35. [136] Hasan S, Satake M, Dawson DW, Funahashi H, Angst E, Go VLW, et al. Expression analysis of the prostaglandin E2 production pathway in human pancreatic cancers. Pancreas 2008;37:121–7. [137] Herfs M, Herman L, Hubert P, Minner F, Arafa M, Roncarati P, et al. High expression of PGE2 enzymatic pathways in cervical (pre)neoplastic lesions and functional consequences for antigen-presenting cells. Cancer Immunol Immunother 2009;58:603–14. [138] Hanaka H, Pawelzik S-C, Johnsen JI, Rakonjac M, Terawaki K, Rasmuson A, et al. Microsomal prostaglandin E synthase 1 determines tumor growth in vivo of prostate and lung cancer cells. Proc Natl Acad Sci U S A 2009;106:18757–62. [139] Omi Y, Shibata N, Okamoto T, Obara T, Kobayashi M. Immunohistochemical demonstration of membrane-bound prostaglandin E2 synthase-1 in papillary thyroid carcinoma. Acta Histochem Cytochem 2009;42: 105–9. [140] Baryawno N, Sveinbjörnsson B, Eksborg S, Orrego A, Segerström L, Oqvist CO, et al. Tumor-growth-promoting cyclooxygenase-2 prostaglandin E2 pathway provides medulloblastoma therapeutic targets. Neuro-oncol 2008;10:661–74. [141] Mattila S, Tuominen H, Koivukangas J, Stenbäck F. The terminal prostaglandin synthases mPGES-1, mPGES-2, and cPGES are all overexpressed in human gliomas. Neuropathology 2009;29:156–65.

I. Cebola, M.A. Peinado / Progress in Lipid Research 51 (2012) 301–313 [142] Kawata R, Hyo S, Araki M, Takenaka H. Expression of cyclooxygenase-2 and microsomal prostagalandin E synthase-1 in head and neck squamous cell carcinoma. Auris Nasus Larynx 2010;37:482–7. [143] Camacho M, León X, Fernández-Figueras M-T, Quer M, Vila L. Prostaglandin E(2) pathway in head and neck squamous cell carcinoma. Head Neck 2008;30:1175–81. [144] Nakanishi M, Montrose DC, Clark P, Nambiar PR, Belinsky GS, Claffey KP, et al. Genetic deletion of mPGES-1 suppresses intestinal tumorigenesis. Cancer Res 2008;68:3251–9. [145] Sasaki Y, Kamei D, Ishikawa Y, Ishii T, Uematsu S, Akira S, et al. Microsomal prostaglandin E synthase-1 is involved in multiple steps of colon carcinogenesis. Oncogene 2011. [146] Fan H, Zhao Z, Quan Y, Xu J, Zhang J, Xie W. DNA methyltransferase 1 knockdown induces silenced CDH1 gene reexpression by demethylation of methylated CpG in hepatocellular carcinoma cell line SMMC-7721. Eur J Gastroenterol Hepatol 2007;19:952–61. [147] Alaa M, Suzuki M, Yoshino M, Tian L, Suzuki H, Nagato K, et al. Prostaglandin E2 receptor 2 overexpression in squamous cell carcinoma of the lung correlates with p16INK4A methylation and an unfavorable prognosis. Int J Oncol 2009;34:805–12. [148] Baba Y, Nosho K, Shima K, Goessling W, Chan AT, Ng K, et al. PTGER2 overexpression in colorectal cancer is associated with microsatellite instability, independent of CpG island methylator phenotype. Cancer Epidemiol Biomarkers Prev 2010;19:822–31. [149] Chun K-S, Lao H-C, Trempus CS, Okada M, Langenbach R. The prostaglandin receptor EP2 activates multiple signaling pathways and beta-arrestin1 complex formation during mouse skin papilloma development. Carcinogenesis 2009;30:1620–7. [150] Paul AG, Sharma-Walia N, Kerur N, White C, Chandran B. Piracy of prostaglandin E2/EP receptor-mediated signaling by Kaposi’s sarcomaassociated herpes virus (HHV-8) for latency gene expression: strategy of a successful pathogen. Cancer Res 2010;70:3697–708. [151] Sugino Y, Misawa A, Inoue J, Kitagawa M, Hosoi H, Sugimoto T, et al. Epigenetic silencing of prostaglandin E receptor 2 (PTGER2) is associated with progression of neuroblastomas. Oncogene 2007;26:7401–13. [152] Tian L, Suzuki M, Nakajima T, Kubo R, Sekine Y, Shibuya K, et al. Clinical significance of aberrant methylation of prostaglandin E receptor 2 (PTGER2) in nonsmall cell lung cancer: association with prognosis, PTGER2 expression, and epidermal growth factor receptor mutation. Cancer 2008;113: 1396–403. [153] Gray SG, Al-Sarraf N, Baird A-M, Cathcart M-C, McGovern E, O’Byrne KJ. Regulation of EP receptors in non-small cell lung cancer by epigenetic modifications. Eur J Cancer 2009;45:3087–97. [154] Zhang Y, Daaka Y. PGE2 promotes angiogenesis through EP4 and PKA Cc pathway. Blood 2011;118:5355–64. [155] Shoji Y, Takahashi M, Kitamura T, Watanabe K, Kawamori T, Maruyama T, et al. Downregulation of prostaglandin E receptor subtype EP3 during colon cancer development. Gut 2004;53:1151–8. [156] Backlund MG, Mann JR, Holla VR, Buchanan FG, Tai HH, Musiek ES, et al. 15Hydroxyprostaglandin dehydrogenase is down-regulated in colorectal cancer. J Biol Chem 2005;280:3217–23. [157] Leslie A, Stewart A, Baty DU, Mechan D, McGreavey L, Smith G, et al. Chromosomal changes in colorectal adenomas: relationship to gene mutations and potential for clinical utility. Genes Chromosomes Cancer 2006;45:126–35. [158] Jang TJ, Ji YS, Jung KH. Decreased expression of 15-hydroxyprostaglandin dehydrogenase in gastric carcinomas. Yonsei Med J 2008;49:917–22. [159] Hughes D, Otani T, Yang P, Newman RA, Yantiss RK, Altorki NK, et al. NAD+dependent 15-hydroxyprostaglandin dehydrogenase regulates levels of bioactive lipids in non-small cell lung cancer. Cancer Prev Res (Phila) 2008;1:241–9. [160] Thiel A, Ganesan A, Mrena J, Junnila S, Nykänen A, Hemmes A, et al. 15hydroxyprostaglandin dehydrogenase is down-regulated in gastric cancer. Clin Cancer Res 2009;15:4572–80. [161] Na HK, Park JM, Lee HG, Lee HN, Myung SJ, Surh YJ. 15-Hydroxyprostaglandin dehydrogenase as a novel molecular target for cancer chemoprevention and therapy. Biochem Pharmacol 2011;82:1352–60. [162] Backlund MG, Mann JR, Holla VR, Shi Q, Daikoku T, Dey SK, et al. Repression of 15-hydroxyprostaglandin dehydrogenase involves histone deacetylase 2 and snail in colorectal cancer. Cancer Res 2008;68:9331–7. [163] Tong M, Ding Y, Tai H-H. Histone deacetylase inhibitors and transforming growth factor-beta induce 15-hydroxyprostaglandin dehydrogenase expression in human lung adenocarcinoma cells. Biochem Pharmacol 2006;72:701–9. [164] Moore AE, Young LE, Dixon DA. MicroRNA and AU-rich element regulation of prostaglandin synthesis. Cancer metastasis rev 2011. [165] Iwai N, Katsuya T, Ishikawa K, Mannami T, Ogata J, Higaki J, et al. Human prostacyclin synthase gene and hypertension: the Suita study. Circulation 1999;100:2231–6. [166] FitzGerald GA, Pedersen AK, Patrono C. Analysis of prostacyclin and thromboxane biosynthesis in cardiovascular disease. Circulation 1983;67:1174–7. [167] Camacho M, Rodriguez C, Guadall A, Alcolea S, Orriols M, Escudero JR, et al. Hypoxia upregulates PGI-synthase and increases PGI release in human vascular cells exposed to inflammatory stimuli. J Lipid Res 2011;52: 720–31.

311

[168] Hatae T, Wada M, Yokoyama C, Shimonishi M, Tanabe T. Prostacyclindependent apoptosis mediated by PPAR delta. J Biol Chem 2001;276:46260–7. [169] Keith RL, Miller YE, Hoshikawa Y, Moore MD, Gesell TL, Gao B, et al. Manipulation of pulmonary prostacyclin synthase expression prevents murine lung cancer. Cancer Res 2002;62:734–40. [170] Keith RL, Karoor V, Mozer AB, Hudish TM, Le M, Miller YE. Chemoprevention of murine lung cancer by gefitinib in combination with prostacyclin synthase overexpression. Lung Cancer 2010;70:37–42. [171] Keith RL, Miller YE, Hudish TM, Girod CE, Sotto-Santiago S, Franklin WA, et al. Pulmonary prostacyclin synthase overexpression chemoprevents tobacco smoke lung carcinogenesis in mice. Cancer Res 2004;64:5897–904. [172] Nemenoff R, Meyer AM, Hudish TM, Mozer AB, Snee A, Narumiya S, et al. Prostacyclin prevents murine lung cancer independent of the membrane receptor by activation of peroxisomal proliferator-activated receptor gamma. Cancer Prev Res (Phila) 2008;1:349–56. [173] Li R-C, Cindrova-Davies T, Skepper JN, Sellers LA. Prostacyclin induces apoptosis of vascular smooth muscle cells by a cAMP-mediated inhibition of extracellular signal-regulated kinase activity and can counteract the mitogenic activity of endothelin-1 or basic fibroblast growth factor. Circ Res 2004;94:759–67. [174] Poole EM, Bigler J, Whitton J, Sibert JG, Potter JD, Ulrich CM. Prostacyclin synthase and arachidonate 5-lipoxygenase polymorphisms and risk of colorectal polyps. Cancer Epidemiol Biomarkers Prev 2006;15:502–8. [175] Abraham JE, Harrington P, Driver KE, Tyrer J, Easton DF, Dunning AM, et al. Common polymorphisms in the prostaglandin pathway genes and their association with breast cancer susceptibility and survival. Clin Cancer Res 2009;15:2181–91. [176] Cutler NS, Graves-Deal R, LaFleur BJ, Gao Z, Boman BM, Whitehead RH, et al. Stromal production of prostacyclin confers an antiapoptotic effect to colonic epithelial cells. Cancer Res 2003;63:1748–51. [177] Frigola J, Muñoz M, Clark SJ, Moreno V, Capella G, Peinado MA. Hypermethylation of the prostacyclin synthase (PTGIS) promoter is a frequent event in colorectal cancer and associated with aneuploidy. Oncogene 2005;24:7320–6. [178] Stearman RS, Grady MC, Nana-Sinkam P, Varella-Garcia M, Geraci MW. Genetic and epigenetic regulation of the human prostacyclin synthase promoter in lung cancer cell lines. Mol Cancer Res 2007;5:295–308. [179] Cathcart MC, Gray SG, Baird AM, Boyle E, Gately K, Kay E, et al. Prostacyclin synthase expression and epigenetic regulation in nonsmall cell lung cancer. Cancer 2011. [180] Chen J, Röcken C, Klein-Hitpass L, Götze T, Leodolter A, Malfertheiner P, et al. Microarray analysis of gene expression in metastatic gastric cancer cells after incubation with the methylation inhibitor 5-aza-20 -deoxycytidine. Clin Exp Metastasis 2004;21:389–97. [181] Gupta RA, Tan J, Krause WF, Geraci MW, Willson TM, Dey SK, et al. Prostacyclin-mediated activation of peroxisome proliferator-activated receptor delta in colorectal cancer. Proc Natl Acad Sci U S A 2000;97:13275–80. [182] Falcetti E, Flavell DM, Staels B, Tinker A, Haworth SG, Clapp LH. IP receptordependent activation of PPARgamma by stable prostacyclin analogues. Biochem Biophys Res Commun 2007;360:821–7. [183] Yang L, Zhou Z-G, Zheng X-L, Wang L, Yu Y-Y, Zhou B, et al. RNA interference against peroxisome proliferator-activated receptor delta gene promotes proliferation of human colorectal cancer cells. Dis Colon Rectum 2008;51:318–26. [184] Tachibana K, Yamasaki D, Ishimoto K, Doi T. The Role of PPARs in Cancer. PPAR Res 2008;2008:102737. [185] Ghosh M, Ai Y, Narko K, Wang Z, Peters JM, Hla T. PPARdelta is protumorigenic in a mouse model of COX-2-induced mammary cancer. Prostaglandins Other Lipid Mediat 2009;88:97–100. [186] Ogino S, Shima K, Baba Y, Nosho K, Irahara N, Kure S, et al. Colorectal cancer expression of peroxisome proliferator-activated receptor gamma (PPARG, PPARgamma) is associated with good prognosis. Gastroenterology 2009;136:1242–50. [187] Pancione M, Sabatino L, Fucci A, Carafa V, Nebbioso A, Forte N, et al. Epigenetic silencing of peroxisome proliferator-activated receptor c is a biomarker for colorectal cancer progression and adverse patients’ outcome. PLoS One 2010;5:e14229. [188] Urade Y, Hayaishi O. Prostaglandin D synthase: structure and function. Vitam Horm 2000;58:89–120. [189] Urade Y, Eguchi N. Lipocalin-type and hematopoietic prostaglandin D synthases as a novel example of functional convergence. Prostaglandins Other Lipid Mediat 2002;68–69:375–82. [190] Melegos DN, Diamandis EP. Diagnostic value of molecular forms of prostatespecific antigen for female breast cancer. Clin Biochem 1996;29:193–200. [191] Saso L, Leone MG, Sorrentino C, Giacomelli S, Silvestrini B, Grima J, et al. Quantification of prostaglandin D synthetase in cerebrospinal fluid: a potential marker for brain tumor. Biochem Mol Biol Int 1998;46:643–56. [192] Su B, Guan M, Zhao R, Lu Y. Expression of prostaglandin D synthase in ovarian cancer. Clin Chem Lab Med 2001;39:1198–203. [193] Payne CA, Maleki S, Messina M, O’Sullivan MG, Stone G, Hall NR, et al. Loss of prostaglandin D2 synthase: a key molecular event in the transition of a lowgrade astrocytoma to an anaplastic astrocytoma. Mol Cancer Ther 2008;7:3420–8.

312

I. Cebola, M.A. Peinado / Progress in Lipid Research 51 (2012) 301–313

[194] Ragolia L, Palaia T, Hall CE, Klein J, Büyük A. Diminished lipocalin-type prostaglandin D(2) synthase expression in human lung tumors. Lung Cancer 2010;70:103–9. [195] Bos CL, Richel DJ, Ritsema T, Peppelenbosch MP, Versteeg HH. Prostanoids and prostanoid receptors in signal transduction. Int J Biochem Cell Biol 2004;36:1187–205. [196] Gervais FG, Cruz RP, Chateauneuf A, Gale S, Sawyer N, Nantel F, et al. Selective modulation of chemokinesis, degranulation, and apoptosis in eosinophils through the PGD2 receptors CRTH2 and DP. J Allergy Clin Immunol 2001;108:982–8. [197] Murata T, Lin MI, Aritake K, Matsumoto S, Narumiya S, Ozaki H, et al. Role of prostaglandin D2 receptor DP as a suppressor of tumor hyperpermeability and angiogenesis in vivo. Proc Natl Acad Sci U S A 2008;105:20009–14. [198] Hirai H, Tanaka K, Yoshie O, Ogawa K, Kenmotsu K, Takamori Y, et al. Prostaglandin D2 selectively induces chemotaxis in T helper type 2 cells, eosinophils, and basophils via seven-transmembrane receptor CRTH2. J Exp Med 2001;193:255–61. [199] Gustafsson A, Hansson E, Kressner U, Nordgren S, Andersson M, Lonnroth C, et al. Prostanoid receptor expression in colorectal cancer related to tumor stage, differentiation and progression. Acta Oncol 2007;46:1107–12. [200] Hawcroft G, Gardner SH, Hull MA. Expression of prostaglandin D2 receptors DP1 and DP2 by human colorectal cancer cells. Cancer Lett 2004;210:81–4. [201] Sadikovic B, Yoshimoto M, Al-Romaih K, Maire G, Zielenska M, Squire JA. In vitro analysis of integrated global high-resolution DNA methylation profiling with genomic imbalance and gene expression in osteosarcoma. PLoS One 2008;3:e2834. [202] Kajita S, Ruebel KH, Casey MB, Nakamura N, Lloyd RV. Role of COX-2, thromboxane A2 synthase, and prostaglandin I2 synthase in papillary thyroid carcinoma growth. Mod Pathol 2005;18:221–7. [203] Moussa O, Yordy JS, Abol-Enein H, Sinha D, Bissada NK, Halushka PV, et al. Prognostic and functional significance of thromboxane synthase gene overexpression in invasive bladder cancer. Cancer Res 2005;65:11581–7. [204] Dassesse T, de Leval X, de Leval L, Pirotte B, Castronovo V, Waltregny D. Activation of the thromboxane A2 pathway in human prostate cancer correlates with tumor Gleason score and pathologic stage. Eur Urol 2006;50:1021–31. [205] Sakai H, Suzuki T, Takahashi Y, Ukai M, Tauchi K, Fujii T, et al. Upregulation of thromboxane synthase in human colorectal carcinoma and the cancer cell proliferation by thromboxane A2. FEBS Lett 2006;580:3368–74. [206] Lee KD, Baek SJ, Shen RF. Multiple factors regulating the expression of human thromboxane synthase gene. Biochem J 1996;319(Pt 3):783–91. [207] Watkins G, Douglas-Jones A, Mansel RE, Jiang WG. Expression of thromboxane synthase, TBXAS1 and the thromboxane A2 receptor, TBXA2R, in human breast cancer. Int Semin Surg Oncol 2005;2:23. [208] Paz MF, Wei S, Cigudosa JC, Rodriguez-Perales S, Peinado MA, Huang TH, et al. Genetic unmasking of epigenetically silenced tumor suppressor genes in colon cancer cells deficient in DNA methyltransferases. Hum Mol Genet 2003;12:2209–19. [209] Kabututu Z, Manin M, Pointud J-C, Maruyama T, Nagata N, Lambert S, et al. Prostaglandin F2alpha synthase activities of aldo-keto reductase 1B1, 1B3 and 1B7. J Biochem (Tokyo) 2009;145:161–8. [210] Wölfle D. Enhancement of carcinogen-induced malignant cell transformation by prostaglandin F(2 alpha). Toxicology 2003;188:139–47. [211] Qualtrough D, Kaidi A, Chell S, Jabbour HN, Williams AC, Paraskeva C. Prostaglandin F(2alpha) stimulates motility and invasion in colorectal tumor cells. Int J Cancer 2007;121:734–40. [212] Watanabe K. Prostaglandin F Synthase. Prostaglandins Other Lipid Mediat 2002;68–69:401–7. [213] Nigam S, Zakrzewicz A, Eskafi S, Roscher A. Clinical significance of prostacyclin and thromboxane in cancer of the female breast and genital tract. Cancer Metastasis Rev 1992;11:411–20. [214] Bresson E, Boucher-Kovalik S, Chapdelaine P, Madore E, Harvey N, Laberge PY, et al. The human aldose reductase AKR1B1 qualifies as the primary prostaglandin F synthase in the endometrium. J Clin Endocrinol Metab 2011;96:210–9. [215] Liao C-S, Tai P-J, Huang Y-H, Chen R-N, Wu S-M, Kuo L-W, et al. Regulation of AKR1B1 by thyroid hormone and its receptors. Mol Cell Endocrinol 2009;307:109–17. [216] Lefrançois-Martinez A-M, Bertherat J, Val P, Tournaire C, Gallo-Payet N, Hyndman D, et al. Decreased expression of cyclic adenosine monophosphateregulated aldose reductase (AKR1B1) is associated with malignancy in human sporadic adrenocortical tumors. J Clin Endocrinol Metab 2004;89:3010–9. [217] Kropotova ES, Tychko RA, Zinov’eva OL, Zyrianova AF, Khankin SL, Cherkes VL, et al. Downregulation of AKR1B10 gene expression in colorectal cancer. Mol Biol (Mosk) 2010;44:243–50. [218] Sales KJ, Maldonado-Pérez D, Grant V, Catalano RD, Wilson MR, Brown P, et al. Prostaglandin F(2alpha)-F-prostanoid receptor regulates CXCL8 expression in endometrial adenocarcinoma cells via the calcium– calcineurin–NFAT pathway. Biochim Biophys Acta 2009;1793:1917–28. [219] Keshet I, Schlesinger Y, Farkash S, Rand E, Hecht M, Segal E, et al. Evidence for an instructive mechanism of de novo methylation in cancer cells. Nat Genet 2006;38:149–53. [220] Sossey-Alaoui K, Kitamura E, Cowell JK. Fine mapping of the PTGFR gene to 1p31 region and mutation analysis in human breast cancer. Int J Mol Med 2001;7:543–6.

[221] Breyer RM, Bagdassarian CK, Myers SA, Breyer MD. Prostanoid receptors: subtypes and signaling. Annu Rev Pharmacol Toxicol 2001;41:661–90. [222] Ulrich CM, Bigler J, Sparks R, Whitton J, Sibert JG, Goode EL, et al. Polymorphisms in PTGS1 (=COX-1) and risk of colorectal polyps. Cancer Epidemiol Biomarkers Prev 2004;13:889–93. [223] Frank B, Hoffmeister M, Klopp N, Illig T, Chang-Claude J, Brenner H. Polymorphisms in inflammatory pathway genes and their association with colorectal cancer risk. Int J Cancer 2010;127:2822–30. [224] Küry S, Buecher B, Robiou-du-Pont S, Scoul C, Colman H, Le Neel T, et al. Lowpenetrance alleles predisposing to sporadic colorectal cancers: a French casecontrolled genetic association study. BMC Cancer 2008;8:326. [225] Sakoda LC, Gao Y-T, Chen BE, Chen J, Rosenberg PS, Rashid A, et al. Prostaglandin-endoperoxide synthase 2 (PTGS2) gene polymorphisms and risk of biliary tract cancer and gallstones: a population-based study in Shanghai, China. Carcinogenesis 2006;27:1251–6. [226] Li F, Ren GS, Li HY, Wang XY, Chen L, Li J. A novel single nucleotide polymorphism of the cyclooxygenase-2 gene associated with breast cancer. Clin Oncol (R Coll Radiol) 2009;21:302–5. [227] Schonfeld S, Bhatti P, Brown EE, Linet MS, Simon SL, Weinstock RM, et al. Polymorphisms in oxidative stress and inflammation pathway genes, lowdose ionizing radiation, and the risk of breast cancer among US radiologic technologists. Cancer Causes Control 2010;21:1857–66. [228] Dossus L, Kaaks R, Canzian F, Albanes D, Berndt SI, Boeing H, et al. PTGS2 and IL6 genetic variation and risk of breast and prostate cancer: results from the Breast and Prostate Cancer Cohort Consortium (BPC3). Carcinogenesis 2010;31:455–61. [229] Langsenlehner U, Yazdani-Biuki B, Eder T, Renner W, Wascher TC, Paulweber B, et al. The cyclooxygenase-2 (PTGS2) 8473T>C polymorphism is associated with breast cancer risk. Clin Cancer Res 2006;12:1392–4. [230] Yang H, Gu J, Lin X, Grossman HB, Ye Y, Dinney CP, et al. Profiling of genetic variations in inflammation pathway genes in relation to bladder cancer predisposition. Clin Cancer Res 2008;14:2236–44. [231] Siezen CLE, Bueno-de-Mesquita HB, Peeters PHM, Kram NR, van Doeselaar M, van Kranen HJ. Polymorphisms in the genes involved in the arachidonic acidpathway, fish consumption and the risk of colorectal cancer. Int J Cancer 2006;119:297–303. [232] Cox DG, Pontes C, Guino E, Navarro M, Osorio A, Canzian F, et al. Polymorphisms in prostaglandin synthase 2/cyclooxygenase 2 (PTGS2/ COX2) and risk of colorectal cancer. Br J Cancer 2004;91:339–43. [233] Kawasaki T, Nosho K, Ohnishi M, Suemoto Y, Kirkner GJ, Dehari R, et al. Correlation of beta-catenin localization with cyclooxygenase-2 expression and CpG island methylator phenotype (CIMP) in colorectal cancer. Neoplasia 2007;9:569–77. [234] Ogino S, Kirkner GJ, Nosho K, Irahara N, Kure S, Shima K, et al. Cyclooxygenase-2 expression is an independent predictor of poor prognosis in colon cancer. Clin Cancer Res 2008;14:8221–7. [235] Srivastava K, Srivastava A, Kumar A, Mittal B. Gallbladder cancer predisposition: a multigenic approach to DNA-repair, apoptotic and inflammatory pathway genes. PLoS One 2011;6:e16449. [236] Srivastava K, Srivastava A, Pandey SN, Kumar A, Mittal B. Functional polymorphisms of the cyclooxygenase (PTGS2) gene and risk for gallbladder cancer in a North Indian population. J Gastroenterol 2009;44:774–80. [237] Li Y, He W, Liu T, Zhang Q. A new cyclo-oxygenase-2 gene variant in the Han Chinese population is associated with an increased risk of gastric carcinoma. Mol Diag Ther 2010;14:351–5. [238] Hur K, Song SH, Lee HS, Ho Kim W, Bang Y-J, Yang H-K. Aberrant methylation of the specific CpG island portion regulates cyclooxygenase-2 gene expression in human gastric carcinomas. Biochem Biophys Res Commun 2003;310:844–51. [239] Huang L, Zhang K-L, Li H, Chen X-Y, Kong Q-Y, Sun Y, et al. Infrequent COX-2 expression due to promoter hypermethylation in gastric cancers in Dalian. China. Hum Pathol 2006;37:1557–67. [240] Silva M, Azenha D, Pereira C, Almeida A, Balseiro S, Sampaio AM, et al. Gastric carcinoma and chronic gastritis: epigenetic regulation of CDH1 (E-Cadherin), CDKN2A (p16INK4A), PTGS2 (COX-2) and EGFR genes through methylation. Acta Med Port 2010;23:5–14. [241] Wang B-C, Guo C-Q, Sun C, Sun Q-L, Liu G-Y, Li D-G. Mechanism and clinical significance of cyclooxygenase-2 expression in gastric cancer. World J Gastroenterol 2005;11:3240–4. [242] Amirian E, Liu Y, Scheurer ME, El-Zein R, Gilbert MR, Bondy ML. Genetic variants in inflammation pathway genes and asthma in glioma susceptibility. Neuro-oncology 2010;12:444–52. [243] Campa D, Zienolddiny S, Maggini V, Skaug V, Haugen A, Canzian F. Association of a common polymorphism in the cyclooxygenase 2 gene with risk of non-small cell lung cancer. Carcinogenesis 2004;25:229–35. [244] Ben Nasr H, Chahed K, Bouaouina N, Chouchane L. PTGS2 (COX-2) -765 G > C functional promoter polymorphism and its association with risk and lymph node metastasis in nasopharyngeal carcinoma. Mol Biol Rep 2009;36:193–200. [245] Lira MG, Mazzola S, Tessari G, Malerba G, Ortombina M, Naldi L, et al. Association of functional gene variants in the regulatory regions of COX-2 gene (PTGS2) with nonmelanoma skin cancer after organ transplantation. Br J Dermatol 2007;157:49–57. [246] Lurie G, Terry KL, Wilkens LR, Thompson PJ, McDuffie KE, Carney ME, et al. Pooled analysis of the association of PTGS2 rs5275 polymorphism and NSAID use with invasive ovarian carcinoma risk. Cancer Causes Control 2010;21:1731–41.

I. Cebola, M.A. Peinado / Progress in Lipid Research 51 (2012) 301–313 [247] Huang Y-W, Jansen RA, Fabbri E, Potter D, Liyanarachchi S, Chan MWY, et al. Identification of candidate epigenetic biomarkers for ovarian cancer detection. Oncol Rep 2009;22:853–61. [248] Huang SK, Fisher AS, Scruggs AM, White ES, Hogaboam CM, Richardson BC, et al. Hypermethylation of PTGER2 confers prostaglandin E2 resistance in fibrotic fibroblasts from humans and mice. Am J Pathol 2010.

313

[249] Hoeft B, Linseisen J, Beckmann L, Müller-Decker K, Canzian F, Hüsing A, et al. Polymorphisms in fatty-acid-metabolism-related genes are associated with colorectal cancer risk. Carcinogenesis 2010;31:466–72.