Cadmium Induces Migration of Colon Cancer Cells: Roles of Reactive Oxygen Species, P38 and Cyclooxygenase-2

 

Sara Najia    Khodr Issaa    Assaad Eidb    Rabah Iratnic    Ali H. Eida,d

 

aDepartment of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon, bDepartment of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon, cDepartment of Biology, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates, dDepartment of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar

 

 

 

 

Key Words

Cadmium • Colorectal cancer • COX-2 • Environmental pollution

 

Abstract

Background/Aims: Cadmium (Cd) is a heavy metal contaminant whose toxicity is associated with colorectal cancer (CRC). However, the underlying molecular mechanisms of Cd-induced CRC malignancy remain obscure. Methods: A monolayer scratch assay was employed to assess the migration of HT-29 human adenocarcinoma cells.Luciferase reporter assay was used to determine cyclooxygenase-2 (COX-2) transcriptional activity, and Western blotting was used to detect p38 Mitogen Activated Protein Kinase (MAPK) and Akt phosphorylation as well as COX-2 expression. Prostaglandin E2 (PGE2) levels were measured using Enzyme Linked Immunosorbent Assay (ELISA) and reactive oxygen species (ROS) formation was assessed using dihydroethidium (DHE) stain. Results: Here, we show that Cd potentiates the migratory capacity of HT-29 CRC cells. Cd caused a time-dependent increase in COX-2 expression. Celecoxib, a COX-2 selective inhibitor, significantly reduced Cd-induced migration. Cd also increased levels of ROS and phosphorylated p38. Importantly, Cd-induced COX-2 expression and migration were significantly abolished by N-Acetyl-Cysteine (NAC), a ROS scavenger, or SB202190, a specific p38 inhibitor. Furthermore, Cd-induced p38 phosphorylation was inhibited by NAC. Cd (100 nM) also increased PGE2 levels, which was abrogated by NAC, SB202190, or celecoxib. Exogenous PGE2 significantly potentiated cell migration. Cd caused a significant increase in Akt phosphorylation in a ROS-mediated pathway. Moreover, Cd-induced migration was significantly attenuated by LY294 002, a phosphatidylinositol-3-kinase inhibitor.Conclusion: Taken together, our results suggest that exposure to low levels of Cd promotes a more migratory cancer phenotype in a ROS-p38-COX-2-PGE2 pathway as well as ROS-Akt pathway. Therefore, COX-2, PGE2 receptors or Akt represent potential targets in the treatment of CRC, particularly in patients exposed to Cd.

 

 

Introduction

 

Colorectal cancer (CRC) is currently the second leading cause of cancer related deaths in the world [1]. Risk factors associated with CRC are mainly divided into the modifiable (obesity, physical inactivity, diet, smoking) and the non-modifiable ones (age, gender, family history) [2]. Recently, the exposure to environmental pollutants such as heavy metals has gained support as possible contributors to increased CRC risk. These metal contaminants generally include: Arsenic, lead, mercury, chromium and cadmium [3, 4].

Cadmium (Cd) is a toxic metal present in the environment and used in industrial activities [4, 5]. Accumulating evidence suggests that Cd is associated with a wide range of adverse health effects such as cardiovascular diseases [6], bone-related disorders [7], reproductive toxicity [8], nephrotoxicity [9], inflammatory diseases, and cancer [10]. In fact, Cd is classified as human carcinogen (Group 1) by the International Agency for Research on Cancer (IARC). The exact molecular mechanisms by which Cd promotes malignancy are poorly understood [11]. Nevertheless, it has been postulated that Cd-induced reactive oxygen species (ROS) formation, by activating multiple signaling players such as the Mitogen activated protein kinases (MAPKs) [12], play a key role in Cd-mediated carcinogenicity [11, 13].

Different cancer types have been linked to Cd exposure. Being a metalloestrogen, Cd was reported to mimic the actions of estrogen in biological systems. It is not surprising then that Cd has been positively associated with hormone-dependent cancers such as endometrial [14] and breast cancers [15]. It has been suggested that Cd targets the prostate gland as well [16]. In vitro studies indicate that Cd promotes the transformation of normal human prostate epithelial cells to a malignant phenotype [17, 18]. Recent evidence shows that dietary Cd exposure contributes to the development of prostate cancer [16]. Contextually, CRC is among the different types of cancers associated with Cd. A significantly higher Cd plasma level was reported in metastatic CRC patients when compared to healthy group [19]. Experimental evidence further demonstrates that by suppressing DNA mismatch repair mechanism, Cd acts a mutagen [20] and might therefore contribute to CRC. Moreover, Cd was shown to induce transformative and carcinogenic effects in human colorectal cells CRL-1807 and a xenograft animal model [21].

Inflammation has been also linked to Cd exposure [22]. In fact, inflammatory bowel diseases (IBD) are among those that have been positively correlated with Cd. Oral Cd exposure in rats was reported to increase the severity of colonic inflammation and to delay the healing of acetic acid-induced colitis [23]. Furthermore, evidence indicates that Cd initiates a pro-inflammatory response by inducingcyclooxygenase-2 (COX-2) upregulation in mouse neuronal cells [24].

Substantial evidence indicates that inflammation is strongly correlated with the initiation and development of cancer [25, 26]. In fact, persistent inflammation as a result of infection or injury could predispose to cancer development. For instance, chronic infections as in hepatitis B or C are associated with elevated hepatocellular carcinoma risk [25]. Moreover, chronic inflammation of intestine as in patients with IBD is strongly linked to increased CRC risk [27]. The cyclooxygenase (COX) pathway is among those inflammatory pathways that have been suggested to play a central role in colorectal malignancy. The COX enzyme exists in two major isoforms: COX-1, the constitutive enzyme and COX-2 the inducible one [28]. It is important to note that both enzymes serve as molecular targets for the Non-steroidal Anti-inflammatory drugs (NSAIDs), a class of medications used in the management of pain, fever and inflammation.

Overwhelming data suggests that COX-2 is positively correlated with colorectal malignancy. COX-2, but not COX-1, gene expression was reported to be highly elevated in human CRC mucosa when compared to the normal one [29]. Moreover, COX-2 overexpression was found to be associated with worse survival among CRC patients [30]. The regular use of aspirin (an NSAID) after diagnosis with CRC is linked to a lower CRC specific and overall mortality [31]. Experimental evidence demonstrates that selective COX-2 inhibitors negatively regulate CRC cells growth, migration and invasion [32, 33].

The tumorigenic effects of COX-2 are thought to be largely attributed to its major metabolite, Prostaglandin E2 (PGE2). In fact, elevated PGE2 levels was reported among patients with colonic adenomas as well as carcinoma [34]. The biological functions of PGE2 are meditated by binding to its target receptors, namely the E type prostanoid (EP) receptors [35]. These receptors comprise the following EP1, EP2, EP3 and EP4 subtypes. Recent evidence suggests that the EP receptors, namely EP4, play a central role in colorectal neoplasia [36]. PGE2 exerts a plethora of effects in colorectal cancer promoting proliferation [37], survival, angiogenesis [38], invasion and migration [39, 40] . Multiple players were shown to be implicated in COX-2/PGE2 mediated carcinogenicity including the Phosphatidylinositol 3-kinase (PI3K)/Akt pathway. For instance, PGE2 via activating EP4 receptors, increases the growth, migration and invasion of LS-174T colorectal carcinoma cells in a PI3K/Akt mediated pathway [41].

Despite being a known human carcinogen, the underlying molecular mechanisms of Cd-induced colorectal malignancy remain vague. Thus, in the present study we hypothesize that Cd promotes the migration of CRC cells by increasing PGE2 production via a ROS-p38 MAPK-COX-2 dependent mechanism, concomitant with activation of the ROS-Akt pathway.

 

 

Materials and Methods

 

Reagents

Cadmium Chloride dihydrate (CdCl2) was a kind gift from the chemistry department at American University of Beirut (AUB). Rabbit monoclonal anti-COX-2 (ab15191), anti-β actin (ab119716), anti-p38 (phospho T180+Y182 ab38238) anti-total p38 (ab7952), anti-pan AKT (ab8805), anti-pan AKT (phospho T308 ab38449), SB202190 (ab20638) and Celecoxib (ab141988) were purchased from abcam. Goat anti-rabbit secondary antibody IgG (H+L), HRP conjugate was obtained from Invitrogen. N-Acetyl-Cysteine (#1761C311) was obtained from Amresco (Solon, Ohio, USA). LY294 002, Dulbecco's Modified Eagle's medium (DMEM) and Fetal Bovine Serum (FBS) were purchased from Sigma-Aldrich (Schnelldorf, Germany) and PGE2   was obtained from Cayman Chemical (Ann Arbor, Michigan, USA). Penicillin/streptomycin was obtained from Lonza (Basel, Switzerland).

 

Cell culture

HT-29 human colorectal adenocarcinoma cells were cultured in Dulbecco's Modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin. Cells were grown in 10 mm dishes and maintained in a humidified incubator at 37°C with 5% CO2 atmosphere. Culture medium was changed every 48 hours and cells were split in a ratio of 1 to 4 once reaching 90% confluence.

 

Migration Assay

Cell migratory capacity was assessed using the scratch or wound healing assay. Cells were seeded in 12-wells plate (6x105 cells/well) in 1 ml complete DMEM. Once reaching 90-95% confluence, a scratch was created manually through the confluent monolayer using a white tip (2–20 μl). The culture medium was then aspirated, and wells were washed with PBS to remove cellular debris. Complete fresh medium was then added along with the indicated treatments.Photomicrographs were taken at baseline (0 hour) and at the indicated time points. ZEN lite from Zeiss Microscope software was used to measure the width of the scratch which reflected the extent of cell migration. The distance migrated was measured in µm.

 

Western blotting

Cells were lysed using a lysis buffer comprising 10 mM Tris pH 6.8, 2% SDS, as we previously described [42]. Proteins were quantified by DC protein assay kit (Bio-Rad, USA). Protein samples were then loaded (30-50 µg) on 5-11% SDS-PAGE along with protein ladder (Abcam), electrophoresed at 70 V (Bio-Rad, USA) and then transferred to PVDF membranes (Bio-Rad, USA). After blocking with 5% non-fat dry milk in TBS-T (TBS and 0.05% Tween 20) for 1 hour at room temperature, the blots were probed with primary antibody at 4 oC overnight. Blots were then washed three times with TBS-T and incubated with Horseradish peroxidase (HRP)-conjugated secondary antibody (diluted 1:2000) for 1 hour at room temperature. Immunoreactive bands were finally detected by ECL chemiluminescent substrate (Bio-Rad, USA). Blot images were taken by using Chemidoc MP Imaging system (Bio-Rad, USA).

 

Dihydroethidium staining (DHE)

ROS production was assessed using DHE stain. HT-29 cells were seeded on a cover slip in 12-well plates (3x105 cells/well) in 1 ml complete DMEM and incubated until reaching 50% confluency. The cells were treated as indicated. Medium was then removed, cells were washed twice with cold PBS. DHE stain (5 µM final concentration) was added to the wells (1ml/well) in the dark and cells were incubated for 1 hour. DHE stain was removed and cells were washed once with cold PBS. Stained cells on the cover slips were imaged using Microscope Zeiss Axio.

 

Transient Transfections

COX-2 transcriptional activity was measured using the luciferase reporter assay. Cells were transiently transfected with COX-2-promoter driven luciferase expression reporter plasmid using Lipofectamine 2000, to achieve an approximate 80% transfection efficiency. Cells were then allowed to recover overnight in complete growth medium. The next day, cells were treated as indicated. For the luciferase analysis, cells were washed, lysed in luciferase lysis buffer (Promega; Wisconsin, USA), snap-frozen, and then thawed at room temperature.  This was followed by the centrifugation of the cell lysates at 9, 300 g for 10 minutes. The luciferase activity was finally determined in the supernatant.

 

Enzyme linked immunosorbent assay (ELISA)

HT-29 cells were subjected to the specified treatment conditions for the indicated time points and PGE2 production was evaluated using the PGE2 Enzyme linked immunosorbent assay (ELISA) kit (R&D Systems; Minnesota, USA) according to the manufacturer’s protocol.

 

Statistical Analysis

Statistical analysis was performed by a student's t-test for either paired or unpaired observations using GraphPad Prism version 5.0 and InStat3 Software (GraphPad software, Inc. San Diego, CA). Data was presented as mean ± SEM, where n is equal to the number of times an experiment is repeated (n will be = 3). When more than two means are used for comparison, ANOVA was used: either one-way ANOVA (with Dunnett’s post hoc test) or two-way ANOVA (with Tukey-Kramer’s post hoc test). p < 0.05 is considered significant.

 

 

Results

 

Cd induces the migration of HT-29 cells

Cd, at a concentration of 100 nM, significantly (p < 0.05) promoted wound healing at 9 and 12 hours (Fig. 1A and B). The 1000 nM concentration, on the other hand, significantly (p < 0.05) increased the migration only at 9 hours (Fig. 1B), and no further increase was observed for prolonged time.

 

Fig. 1. Cd increases the migration of HT-29 cells. Cell migration was examined by the wound-healing assay. HT-29 cells were treated with Cd (100 and 1000 nM). Images were taken at the indicated time points. Values are represented as mean ± SEM of n=3. * denotes p<0.05.

 

Cd increases colon cancer migration through production of reactive oxygen species

Because Cd was previously shown to induce ROS accumulation in various cells, we therefore investigated its effect on ROS production in HT-29 colon cancer cells.  As shown in Fig. 2C and 2D treatment significantly increased ROS production as early as 5 minutes.

We next examined the effect of ROS production on the Cd-induced migration.  We found that pre-treatment of HT-29 cells with the ROS scavenger N-Acetyl-Cysteine, NAC (10 nM), for 30 minutes significantly (p < 0.05) attenuated Cd-induced migration (Fig. 3A and B).

 

Fig. 2. Cd increases the production of ROS. HT-29 cells were treated with Cd (100 nM) for 5, 10, and 30 minutes and DHE staining assay was performed.

Fig. 3. Cd-induced ROS production increases HT-29 migration. HT-29 cells were treated with Cd (100 nM) in the presence or absence of the ROS scavenger NAC (10 mM) and migration was assessed by scratch assay. Values are represented as mean ± SEM of n=3. * denotes p<0.05.

 

Cd promotes colon cancer cell migration through ROS-dependent activation of p38 MAPK

Next, we have investigated whether the p38 MAPK pathway is implicated in the Cd-mediated effect on cellular migration, by measuring the level of p38 phosphorylation.  We found that Cd caused more than 2-fold increase in p38 phosphorylation (p < 0.05) following 10 and 30 minutes of treatment (Fig. 4A and B). Furthermore, pretreating the cells with SB202190 (10 µM), a specific p38 MAPK inhibitor, significantly (p < 0.05) abrogated Cd-induced migration (Fig. 4C and D).

Next, we examined whether the Cd-induced activation of p38 MAPK is mediated by ROS production. Indeed, we found that pre-treatment with NAC significantly (p < 0.05) abolished Cd-induced p38 activation (Fig. 5A and B).

 

Fig. 4. Cd activates p38 MAPK which is implicated in migration. (A) and (B): HT-29 cells were treated with Cd (100 nM) for 0, 5, 10, 30 and 60 minutes and p38 MAPK phosphorylation levels were determined by western blotting. (C) and (D): Cells were pretreated with SB202190 (10 µM) for 30 minutes followed by Cd (100 nM) and migration was determined by wound healing assay. Values are the mean ± SEM of three replicates. * denotes p<0.05.

Fig. 5. Cd-induced ROS is upstream p38 MAPK. HT-29 cells were treated with Cd (100 nM) for 30 minutes in the presence or absence of NAC and p38 MAPK phosphorylation levels were determined by western blotting. Values are the mean ± SEM of three replicates. * denotes p<0.05.

 

Cd increases COX-2 expression at the transcriptional and protein levels

Cd significantly (p < 0.05) increases COX-2 protein levels as early as 6 hours (Fig. 6A and B) and maximally at 24 hours (Fig. 6C and D). This result was further validated by assessing the effect of 100 nM Cd on COX-2 expression at the transcription level. Consistent with the protein results, Cd caused a significant and time-dependent increase (p < 0.05) in transcriptional activity of COX-2 promoter, with this increase being evident as early as 6 hours after treatment (Fig. 6 E).

 

Fig. 6. COX-2 protein expression and transcription are enhanced in response to Cd. (A), (B), (C) and (D): HT-29 cells were treated with Cd (100 nM) for 6, 12, 24, 48 and 72 hours. COX-2 protein expression was assessed by western blotting. (E): Cells were treated with Cd (100 nM) for 3,6,12 and 24 hours. COX-2 transcription level was assessed by measuring the level of COX-2 promoter-driven luciferase using reporter assay. Values are the mean ± SEM of three independent experiments. * denotes p<0.05.

 

Cd-induced migration is mediated by ROS-dependent p38 MAPK through upregulation of COX-2

Next, we wanted to examine whether COX-2 induction is involved in Cd-induced migration of colon cancer cells.  As shown in Fig. 7A and 7B, selective inhibition of COX-2 with celecoxib (CLX; 10 µM) significantly (p <0.05) abolished Cd-induced migration, hence suggesting that Cd induced migration requires COX-2 function.

Next, we sought to investigate the mechanism through which Cd treatment upregulates COX-2 expression. As shown in Fig. 8, Inhibition of ROS production by NAC  (Fig. 8A and B) and inhibition of p38 (Fig. 8C and D), by SB202190, significantly attenuated Cd-induced upregulation of COX-2 protein. Moreover, pretreatment with SB202190 also resulted in a significant (p < 0.05) reduction in Cd-induced increase in the transcription level of COX-2 at 12 hours (Fig. 8E).

 

Fig. 7. Cd-induced migration is COX-2 dependent. HT-29 cells were pre-treated with CLX (10 µM) for 30 minutes followed by Cd (100 nM). Migration was determined by wound healing assay. Values are represented as mean ± SEM of n=3. * denotes p<0.05.

Fig. 8. Cd acts through ROS and p38 to upregulate COX-2. (A), (B), (C) and (D): HT-29 cells were treated with Cd (100 nM) for 24 hours in the presence or absence of NAC and/or SB202190. COX-2 protein levels were assessed by western blotting. (E): Cells were pre-treated with SB202190 for 30 minutes followed by Cd (100 nM) for 12 hours. COX-2 transcription was evaluated using the COX-2 promoter-driven luciferase reporter activity. Values are represented as mean ± SEM of n=3. * denotes p<0.05, ** denotes p<0.01.

 

PGE2 potentiates migration and its production is enhanced in response to Cd through ROS mediated activation of p38MAPK and subsequent COX-2 upregulation

We have shown that COX-2 is required for Cd-induced migration of colon cancer cells (Fig. 7A and B). We therefore investigated whether Prostaglandin E2 (PGE2), a COX-2 major metabolite, is involved in the increased migration of Cd-treated HT-29 cells.  We found that exogenous PGE2 significantly (p < 0.05) increased HT-29 migration (Fig. 9A). Moreover, Cd caused a concentration-dependent increase in endogenous PGE2 production (data not shown). Time-course experiment showed that Cd (100 nM) caused a time-dependent increase (p <0.05) in PGE2 production starting as early as 1 hour (Fig. 9B).

Interstingly, we found that reducing cellular levels of ROS and blocking p38 or COX-2 activity was associated with a significant reduction (p < 0.05) in Cd-induced PGE2 production at 6 hours (Fig. 10). Altogether, our data suggest that Cd increases cellular migration of colon cancer cells, through induction of ROS-mediated activation of p38 MAPK which subsequently upregulates the expression of COX-2 leading to accumulation of PGE2.

 

Fig. 9. PGE2 enhances migration and its levels are increased in response to Cd. (A): Cells were treated with 1 µM PGE2 for 12 hours and migration was assessed by scratch assay. (B): Cells were treated with Cd (100 nM) for 1, 3, 6 and 12 hours and PGE2 production was assessed using ELISA kit. Values are represented as the mean of fold increase ± SEM of three independent experiments. * denotes p<0.05.

Fig. 10. Cd increases PGE2 levels in a ROS, p38 and COX-2 dependent pathway. HT-29 cells were pre-treated with NAC, SB202190 or CLX for 30 minutes followed by Cd (100 nM) for 6 hours. PGE2 levels were measured using ELISA kit. Values are represented as the mean of fold change ± SEM of n=3. * denotes p<0.05.

 

Cd activates Akt in a ROS-dependent pathway

The effect of Cd on the Akt, another signaling pathway actively involved in the migration of cancer cells was investigated. We found that Cd caused a significant increase in the level of phosphorylated Akt detectable as early as 30 minutes post-treatment with Cd (p <0.05 and 0.01 respectively) (Fig. 11A and B). Inhibition of ROS generation by NAC significantly attenuated Cd-induced Akt phosphorylation at 60 minutes (Fig. 11C and D).

 

Fig. 11. Cd activates Akt in a ROS mediated pathway. (A) and (B). HT-29 cells were treated with Cd (100 nM) for 0, 10, 30 and 60 minutes and Akt phosphorylation levels were assessed by western blotting. (C) and (D): cells were pre-treated with NAC (10 mM) for 30 minutes followed by Cd (100 nM) for 60 minutes and Akt phosphorylation levels were determined by western blotting. Values are represented as mean ± SEM of n=3. * denotes p<0.05, ** denotes p<0.01.

 

Akt is a crucial mediator of Cd-induced migration

Next, we examined whether Cd-mediated activation of Akt signaling pathway is involved in the Cd-induced increase in colon cancer cell migration. We show that the inhibition of PI3K and thereby Akt, by the PI3K inhibitor LY294002, significantly (p < 0.05) abrogated Cd-increased migration. Pre-treatment with both CLX and LY294002 did not provide additional decrease in migration when compared to LY294002 or CLX alone (Fig. 12A and B).

 

Fig. 12. Cd-induced Akt activation is implicated in migration. HT-29 cells were treated with Cd (100 nM) in the presence or absence of LY294 002 (10 µM), a PI3K inhibitor and/or CLX. Migration was evaluated using scratch wound healing assay. Values are represented as mean ± SEM of n=3.* denotes p<0.05.

 

 

Discussion

 

Although Cd is a known human carcinogen, the underlying signaling pathways implicating it in colorectal malignancy are not well characterized. In this report, the effect of Cd on CRC cells migration was investigated. Herein, we show that Cd increases the migratory capacity of CRC cells via activating two inter-connected pathways: ROS-p38-COX-2-PGE2 and the ROS-Akt pathway.

It is widely accepted that inflammation plays an important role at different stages of tumor development [25, 26]. Overwhelming evidence shows that the cyclooxygenase-mediated pathway is a primary pro-inflammatory signaling cascade implicated in colorectal malignancy [43]. In fact, expression of COX-2 was reported to be increased in human colorectal cancer mucosa when compared to the normal tissues [29]. Epidemiological evidence demonstrates that chronic aspirin use is associated with 40- 50% reduction in CRC relative risk [44]. Indeed, selective inhibition of COX-2 is associated with reduced CRC cell proliferation [45], migration and invasion [32]. Here, we report that Cd causes a significant increase in cellular migratory capacity. To investigate the underlying mechanisms of Cd-induced cell migration, we looked at COX-2 expression. Interestingly, Cd caused a time dependent increase in the transcription and protein expression of COX-2. The fact that Cd upregulated COX-2 as early as 6 hours most likely indicates a post-transcriptional regulation, which could be explained by either increased COX-2 translational efficiency or mRNA stability. To assess whether Cd-induced COX-2 upregulation is positively implicated in migration, cells were pre-treated with CLX. Indeed, blocking COX-2 activity abolished Cd-induced migration, clearly indicating that Cd, via increasing COX-2 expression, is enhancing migration. This is consistent with several lines of evidence which established that COX-2 plays a crucial role in promoting migration of cancer cells [46, 47].

It is well known that CLX and other NSAIDs reduce carcinogenesis via inhibiting COX-2 activity. Nevertheless, COX-2 independent anti-tumor effects of NSAIDs have been also reported [48]. For instance, CLX was reported to reduce membrane fluidity and hence metastatic potential of CRC cells in a COX-2 independent mechanism [49]. Conversely, COX-2 positive but not COX-2 negative CRC cells treated with CLX showed a significant reduction in Epithelial-mesenchymal transition (EMT), strongly implying a COX-2 dependent anti-tumor mechanism [50]. Moreover, it has been reported that CLX tends to accumulate in the hydrophobic interior of plasma membrane of different cell types including B lymphocytes, vascular endothelial cells, leukemic monocytes and human colorectal cancer cells, and thus a higher intracellular concentration is needed to observe its anti-carcinogenic effects [51]. Accordingly, the concentration of CLX used in the present study is 10 µM, which is higher than the maximal plasma concentration achieved (3-5 µM) following the administration of 800 mg/day CLX [52].

A huge body of evidence indicates that ROS plays an important role in different types of cancer including CRC [53]. Several signaling pathways have been implicated in ROS-mediated carcinogenicity. In fact, ROS mediates the activation of Src kinases, PI3K/Akt, and MAPKs, all of which promote malignancy [54]. Previous findings support the notion that ROS could promote EMT [55] as well as tumor cell migration and invasion [56]. It is not surprising then, that reducing cellular levels of ROS using NAC is associated with the inhibition of migration of several cancer cells [57, 58]. The present study established that Cd induces ROS production as early as 5 minutes. This is in agreement with literature which indicates that Cd is an oxidative stressor [12]. Moreover, it has been suggested that ROS plays a crucial role in Cd-induced carcinogenicity [13]. To explore whether oxidative stress is involved in Cd-induced migration, cells were pretreated with the ROS scavenger NAC. Indeed, decreasing cellular ROS levels attenuated Cd- increased cell migration. Interestingly, ROS was reported to upregulate COX-2 in CRC [53]. In line with this, our results show that reducing oxidative stress led to a significant decrease in Cd-induced COX-2 expression, clearly implicating ROS as a COX-2 positive modulator.

It is important to note that in addition to its tumor promoting role, a tumor suppressive role of ROS has been also reported [59]. Contrary to our findings, Piskounova et al., demonstrated that NAC promoted metastasis in melanoma human xenografts [60]. Similarly, NAC markedly increased migration and invasion of human malignant melanoma cells [61]. Moreover, dietary supplementation with antioxidants such as NAC or Vitamin E was shown to accelerate the progression of lung cancer in mice [62]. It is now accepted that low levels of ROS support cells proliferation, migration and survival whereas high levels are generally cytotoxic [53, 63].

Aberrant MAPK signaling plays a crucial role in the development and progression of different malignancies [64]. Three main groups of MAPKs have been identified in mammals: ERK1/2, JNK 1/2/3 and p38 MAPK [65]. The MAPKs are activated by variety of stimuli such as growth factors, pro-inflammatory cytokines, hormones and oxidative stress [66]. Indeed, being an oxidative stressor, Cd was reported to activate MAPKs [67]. Contextually, herein we report that Cd significantly increases p38 MAPK phosphorylation in a ROS mediated pathway. Previous studies indicate that p38 is implicated in several human diseases including cancer [68]. In fact, elevated levels of phosphorylated p38 have been associated with various malignancies [69]. A recently published study shows that p38 MAPK is involved in the migration of human breast cancer cells [70]. A positive association has been reported between high endogenous p38 MAPK activity and cancer cells invasiveness [71]. Additionally, pro-proliferative [72] and anti-apoptotic effects were correlated with p38 activation [73, 74]. The fact that SB202190 abolished Cd-induced HT-29 migration is consistent with reports implicating active p38 as a mediator of cancer cells migration [72]. Moreover, we have shown that Cd-induced increase in COX-2 transcriptional activity and protein expression was significantly attenuated by SB202190. This is in agreement to previous findings suggesting that the activation of p38 could contribute to cancer progression via upregulating COX-2 in different tumors [69]. It is noteworthy that besides its tumor promoting effect, a tumor suppressive role of p38 has been also described [75]. In fact, increasing evidence highlights the pro-apoptotic roles of p38 [75]. Remarkably, p38 activity appears to be essential for some chemotherapeutic drugs induced apoptosis. The alkylating agent Cisplatin for example, was shown to induce a p38 mediated tumor cell death [76]. Importantly, the function of p38 MAPK in cancer appears to depend on the cell type, the stimuli and/or the isoform that is activated [74].

Because PGE2 is thought to be the major pro-tumorigenic metabolite of COX-2, we next explored its effect on HT-29 migration. Our findings indicate that the addition of PGE2 significantly potentiated migration. These results support the notion that PGE2 is an important mediator of colon cancer cells migration [39, 40]. Previous studies yielded conflicting results regarding the catalytic activity of COX-2 in HT-29 cell line. Parker et al. demonstrated that the addition of arachidonic acid (AA), a COX substrate, simulates the production of PGE2 in HT-29 [77]. On the contrary, other investigators have shown that although COX-2 is expressed in HT-29 cells, it is catalytically inactive and is therefore unable to convert AA to prostaglandins [78]. Our results indicate that Cd causes a significant rise in PGE2 production, clearly suggesting that the Cd-induced de novo synthesized COX-2 is enzymatically active. The present study further established Cd-induced PGE2 production was significantly attenuated by NAC, SB202190 and CLX pre-treatment. These findings support the fact that PGE2 is major COX-2 product and that ROS and p38 are critical mediators of COX-2 expression.

Substantial evidence indicates that the PI3K/Akt pathway is implicated in cancer [79]. In fact, this pathway plays an important role in promoting cell proliferation, survival, invasion, metastasis and angiogenesis [79]. The selective PI3K inhibitor LY294002 was shown to significantly decrease ovarian cancer cells proliferation [80], invasion and metastasis [81]. As a matter of fact, aberrant PI3K/Akt pathway occurs in several malignancies including CRC [79]. Interestingly, when compared to normal colonic mucosa, colorectal adenomas and carcinomas frequently overexpress Akt, the major downstream player of PI3K [82]. Importantly, Turečkovį et al. established that Akt strongly stimulates the migration of colorectal cancer cells via phosphorylating focal adhesion kinase (FAK) in a Src mediated pathway [83]. In line with this, our results indicate that Cd significantly increases the phosphorylation of Akt. Furthermore, inhibiting Akt significantly abolished Cd-increased migration, clearly implicating Akt as a mediator of migration. Our findings further demonstrate that inhibiting both COX-2 and Akt was not associated with any additional decrease in migration. The exact explanation remains to be fully elucidated; however, the following could be postulated: Cd-induced PGE2 production before 1 hour could actually be activating the PI3K/Akt pathway. Previous reports indicated that PI3K/Akt pathway is an important downstream mediator of the COX-2/PGE2 pathway. In fact, PGE2 was shown to enhance the growth, migration and invasion of colorectal cancer cells in PI3K/Akt dependent pathway [41]. Since the PI3K/Akt pathway is known to be activated by ROS [84], we determined if indeed Cd-induced Akt phosphorylation is ROS-mediated. Herein, we showed that pre-treatment with NAC significantly attenuated Cd-induced Akt phosphorylation, strongly suggesting that ROS is upstream of Akt.

Increasing evidence highlights the role of EP receptors in colorectal neoplasia. Elevated EP4 receptor expression was reported in colorectal cancers (100%) as well as adenomas (36%) when compared with normal colon epithelium [36]. Enhanced tumorigenic behavior and suppression of apoptosis was observed in HT-29 human adenocarcinoma cells overexpressing EP4 receptors [85]. Moreover, the genetic deletion or the pharmacological inhibition of EP4 receptor [86] and EP1 receptor [87] caused a significant inhibition of tumor growth, suggesting that these receptors play a key role in colon tumorigenesis.

 

 

Conclusion

 

The findings of the present study argue that exposure to low Cd levels enhances migration of CRC cells via induction of the ROS-p38-COX-2-PGE2 and the ROS-Akt pathway. Therefore, COX-2, PGE2 receptors, or Akt may represent potential targets in the treatment of CRC, specifically in patients exposed to Cd.

 

 

Acknowledgements

 

This publication was made possible by an MPP fund (#320133) from the American University of Beirut-Faculty of Medicine to Ali Eid.

 

 

Disclosure Statement

 

The authors declare that they have no competing interests.

 

 

References

 

1 WHO: Cancer Fact sheet, 2018. URL: www.who.int/en/news-room/fact-sheets/detail/cancer.

 

2 Haggar FA, Boushey RP: Colorectal cancer epidemiology: incidence, mortality, survival, and risk factors. Clin Colon Rectal Surg 2009;22:191-197.
https://doi.org/10.1055/s-0029-1242458
PMID: 21037809 PMCid:PMC2796096

 

3 Jaishankar M, Tseten T, Anbalagan N, Mathew BB, Beeregowda KN: Toxicity, mechanism and health effects of some heavy metals. Interdiscip Toxicol 2014;7:60-72.
https://doi.org/10.2478/intox-2014-0009
PMID: 26109881 PMCid:PMC4427717

 

4 Tchounwou PB, Yedjou CG, Patlolla AK, Sutton DJ: Heavy Metals Toxicity and the Environment. Exp Suppl 2012;101:133-164.
https://doi.org/10.1007/978-3-7643-8340-4_6
PMID: 22945569 PMCid:PMC4144270

 

5 Godt J, Scheidig F, Grosse-Siestrup C, Esche V, Brandenburg P, Reich A, Groneberg DA: The toxicity of cadmium and resulting hazards for human health. J Occup Med Toxicol 2006;1:22-22.
https://doi.org/10.1186/1745-6673-1-22
PMID: 16961932 PMCid:PMC1578573

 

6 Tellez-Plaza M, Navas-Acien A, Crainiceanu CM, Guallar E: Cadmium exposure and hypertension in the 1999-2004 National Health and Nutrition Examination Survey (NHANES). Environ Health Perspect 2008;116:51-56.
https://doi.org/10.1289/ehp.10764
PMID: 18197299 PMCid:PMC2199293

 

7 Bhattacharyya MH: Cadmium Osteotoxicity in Experimental Animals: Mechanisms and Relationship to Human Exposures. Toxicol Appl Pharmacol 2009;238:258-265.
https://doi.org/10.1016/j.taap.2009.05.015
PMID: 19463839 PMCid:PMC2826165

 

8 Wang HF, Chang M, Peng TT, Yang Y, Li N, Luo T, Cheng YM, Zhou MZ, Zeng XH, Zheng LP: Exposure to Cadmium Impairs Sperm Functions by Reducing CatSper in Mice. Cell Physiol Biochem 2017;42:44-54.
https://doi.org/10.1159/000477113
PMID: 28554186

 

9 Johri N, Jacquillet G, Unwin R: Heavy metal poisoning: the effects of cadmium on the kidney. Biometals 2010;23:783-792.
https://doi.org/10.1007/s10534-010-9328-y
PMID: 20354761

 

10 Huff J, Lunn RM, Waalkes MP, Tomatis L, Infante PF: Cadmium-induced Cancers in Animals and in Humans. Int J Occup Environ Health 2007;13:202-212.
https://doi.org/10.1179/oeh.2007.13.2.202
PMID: 17718178 PMCid:PMC3399253

 

11 Luevano J, Damodaran C: A Review of Molecular Events of Cadmium-Induced Carcinogenesis. J Environ Pathol Toxicol Oncol 2014;33:183-194.
https://doi.org/10.1615/JEnvironPatholToxicolOncol.2014011075

 

12 Nair AR, DeGheselle O, Smeets K, Van Kerkhove E, Cuypers A: Cadmium-Induced Pathologies: Where Is the Oxidative Balance Lost (or Not)? Int J Mol Sci 2013;14:6116-6143.
https://doi.org/10.3390/ijms14036116
PMID: 23507750 PMCid:PMC3634456

 

13 Liu J, Qu W, Kadiiska MB: Role of oxidative stress in cadmium toxicity and carcinogenesis. Toxicol Appl Pharmacol 2009;238:209-214.
https://doi.org/10.1016/j.taap.2009.01.029
PMID: 19236887 PMCid:PMC4287357

 

14 McElroy JA, Kruse RL, Guthrie J, Gangnon RE, Robertson JD: Cadmium exposure and endometrial cancer risk: A large midwestern U.S. population-based case-control study. PloS One 2017;12:e0179360.
https://doi.org/10.1371/journal.pone.0179360
PMID: 28742092 PMCid:PMC5524364

 

15 Siewit CL, Gengler B, Vegas E, Puckett R, Louie MC: Cadmium promotes breast cancer cell proliferation by potentiating the interaction between ERalpha and c-Jun. Mol Endocrinol 2010;24:981-992.
https://doi.org/10.1210/me.2009-0410
PMID: 20219890 PMCid:PMC2870938

 

16 Julin B, Wolk A, Johansson JE, Andersson SO, Andrén O, Åkesson A: Dietary cadmium exposure and prostate cancer incidence: a population-based prospective cohort study. Br J Cancer 2012;107:895.
https://doi.org/10.1038/bjc.2012.311
PMID: 22850555 PMCid:PMC3425979

 

17 Achanzar WE, Diwan BA, Liu J, Quader ST, Webber MM, Waalkes MP: Cadmium-induced Malignant Transformation of Human Prostate Epithelial Cells. Cancer Res 2001;61:455.
PMID: 11212230

 

18 Benbrahim-Tallaa L, Waterland RA, Dill AL, Webber MM, Waalkes MP: Tumor suppressor gene inactivation during cadmium-induced malignant transformation of human prostate cells correlates with overexpression of de novo DNA methyltransferase. Environ Health Perspect 2007;115:1454-1459.
https://doi.org/10.1289/ehp.10207
PMID: 17938735 PMCid:PMC2022656

 

19 Emre O, Demir H, Dogan E, Esen R, Gur T, Demir C, Gonullu E, Turan N, Özbay MF: Plasma Concentrations of Some Trace Element and Heavy Metals in Patients with Metastatic Colon Cancer. J Cancer Ther 2013;4:1085-1090.
https://doi.org/10.4236/jct.2013.46124

 

20 Jin YH, Clark AB, Slebos RJC, Al-Refai H, Taylor JA, Kunkel TA, Resnick MA, Gordenin DA: Cadmium is a mutagen that acts by inhibiting mismatch repair. Nat Genet 2003;34:326-329.
https://doi.org/10.1038/ng1172
PMID: 12796780 PMCid:PMC2662193

 

21 Lu J, Zhou Z, Zheng J, Zhang Z, Lu R, Liu H, Shi H, Tu Z: 2D-DIGE and MALDI TOF/TOF MS analysis reveal that small GTPase signaling pathways may play an important role in cadmium-induced colon cell malignant transformation. Toxicol Appl Pharmacol 2015;288:106-113.
https://doi.org/10.1016/j.taap.2015.07.020
PMID: 26220685

 

22 Olszowski T, Baranowska-Bosiacka I, Gutowska I, Chlubek D: Pro-inflammatory properties of cadmium. Acta Biochim Pol 2012;59:475-482.
https://doi.org/10.18388/abp.2012_2080
PMID: 23240106

 

23 Adegoke AG, Salami AT, Olaleye SB: Cadmium Exacerbates Aetic Acid Induced Experimental Colitis in Rats. Eur J Exp Biol 2017;7:27.
https://doi.org/10.21767/2248-9215.100027

 

24 Figueiredo-Pereira ME, Li Z, Jansen M, Rockwell P: N-Acetylcysteine and Celecoxib Lessen Cadmium Cytotoxicity Which Is Associated with Cyclooxygenase-2 Up-regulation in Mouse Neuronal Cells. J Biol Chem 2002;277:25283-25289.
https://doi.org/10.1074/jbc.M109145200
PMID: 11997384

 

25 Rakoff-Nahoum S: Why Cancer and Inflammation? Yale J Biol Med 2006;79:123-130.
PMID: 17940622

 

26 Coussens LM, Werb Z: Inflammation and cancer. Nature 2002;420:860-867.
https://doi.org/10.1038/nature01322
PMID: 12490959 PMCid:PMC2803035

 

27 Amersi F, Agustin M, Ko CY: Colorectal cancer: epidemiology, risk factors, and health services. Clin Colon Rectal Surg 2005;18:133-140.
https://doi.org/10.1055/s-2005-916274
PMID: 20011296 PMCid:PMC2780097

 

28 Dubois RN, Abramson SB, Crofford L, Gupta RA, Simon LS, Van De Putte LB, Lipsky PE: Cyclooxygenase in biology and disease. FASEB J 1998;12:1063-1073.
https://doi.org/10.1096/fasebj.12.12.1063
PMID: 9737710

 

29 Eberhart CE, Coffey RJ, Radhika A, Giardiello FM, Ferrenbach S, DuBois RN: Up-regulation of cyclooxygenase 2 gene expression in human colorectal adenomas and adenocarcinomas. Gastroenterology 1994;107:1183-1188.
https://doi.org/10.1016/0016-5085(94)90246-1

 

30 Ogino S, Kirkner GJ, Nosho K, Irahara N, Kure S, Shima K, Hazra A, Chan AT, Dehari R, Giovannucci EL, Fuchs CS: Cyclooxygenase-2 expression is an independent predictor of poor prognosis in colon cancer. Clin Cancer Res 2008;14:8221-8227.
https://doi.org/10.1158/1078-0432.CCR-08-1841
PMID: 19088039 PMCid:PMC2679582

 

31 Chan AT, Ogino S, Fuchs CS: Aspirin use and survival after diagnosis of colorectal cancer. JAMA 2009;302:649-658.
https://doi.org/10.1001/jama.2009.1112
PMID: 19671906 PMCid:PMC2848289

 

32 Yao M, Lam EC, Kelly CR, Zhou W, Wolfe MM: Cyclooxygenase-2 selective inhibition with NS-398 suppresses proliferation and invasiveness and delays liver metastasis in colorectal cancer. Br J Cancer 2004;90:712-719.
https://doi.org/10.1038/sj.bjc.6601489
PMID: 14760389 PMCid:PMC2409599

 

33 Yao M, Kargman S, Lam EC, Kelly CR, Zheng Y, Luk P, Kwong E, Evans JF, Wolfe MM: Inhibition of Cyclooxygenase-2 by Rofecoxib Attenuates the Growth and Metastatic Potential of Colorectal Carcinoma in Mice. Cancer Res 2003;63:586-592.
PMID: 12566300

 

34 Pugh S, Thomas GA: Patients with adenomatous polyps and carcinomas have increased colonic mucosal prostaglandin E2. Gut 1994;35:675-678.
https://doi.org/10.1136/gut.35.5.675
PMID: 8200564 PMCid:PMC1374755

 

35 Breyer RM, Bagdassarian CK, Myers SA, Breyer MD: Prostanoid Receptors: Subtypes and Signaling. Annu Rev Pharmacol Toxicol 2001;41:661-690.
https://doi.org/10.1146/annurev.pharmtox.41.1.661
PMID: 11264472

 

36 Chell SD, Witherden IR, Dobson RR, Moorghen M, Herman AA, Qualtrough D, Williams AC, Paraskeva C: Increased EP4 Receptor Expression in Colorectal Cancer Progression Promotes Cell Growth and Anchorage Independence. Cancer Res 2006;66:3106.
https://doi.org/10.1158/0008-5472.CAN-05-3702
PMID: 16540660

 

37 Qiao L, Kozoni V, Tsioulias GJ, Koutsos MI, Hanif R, Shiff SJ, Rigas B: Selected eicosanoids increase the proliferation rate of human colon carcinoma cell lines and mouse colonocytes in vivo. Biochim Biophys Acta 1995;1258:215-223.
https://doi.org/10.1016/0005-2760(95)00100-Q

 

38 Wang D, Wang H, Brown J, Daikoku T, Ning W, Shi Q, Richmond A, Strieter R, Dey SK, DuBois RN: CXCL1 induced by prostaglandin E2 promotes angiogenesis in colorectal cancer. J Exp Med 2006;203:941-951.
https://doi.org/10.1084/jem.20052124
PMID: 16567391 PMCid:PMC2118273

 

39 Buchanan FG, Wang D, Bargiacchi F, DuBois RN: Prostaglandin E2 Regulates Cell Migration via the Intracellular Activation of the Epidermal Growth Factor Receptor. J Biol Chem 2003;278:35451-35457.
https://doi.org/10.1074/jbc.M302474200
PMID: 12824187

 

40 Hsu HH, Lin YM, Shen CY, Shibu MA, Li SY, Chang SH, Lin CC, Chen RJ, Viswanadha VP, Shih HN, Huang CY: Prostaglandin E2-Induced COX-2 Expressions via EP2 and EP4 Signaling Pathways in Human LoVo Colon Cancer Cells. Int J Mol Sci 2017;18:pii:E1132.
https://doi.org/10.3390/ijms18061132
PMID: 28587064 PMCid:PMC5485956

 

41 Sheng H, Shao J, Washington MK, DuBois RN: Prostaglandin E2 Increases Growth and Motility of Colorectal Carcinoma Cells. J Biol Chem 2001;276:18075-18081.
https://doi.org/10.1074/jbc.M009689200
PMID: 11278548

 

42 Eid AH, Chotani MA, Mitra S, Miller TJ, Flavahan NA: Cyclic AMP acts through Rap1 and JNK signaling to increase expression of cutaneous smooth muscle alpha2C-adrenoceptors. Am J Physiol Heart Circ Physiol 2008;295:H266-H272.
https://doi.org/10.1152/ajpheart.00084.2008
PMID: 18487435 PMCid:PMC2494767

 

43 Liu Y, Sun H, Hu M, Zhang Y, Chen S, Tighe S, Zhu Y: The Role of Cyclooxygenase-2 in Colorectal Carcinogenesis. Clin Colorectal Cancer 2017;16:165-172.
https://doi.org/10.1016/j.clcc.2016.09.012
PMID: 27810226

 

44 DuBois RN, Smalley WE: Cyclooxygenase, NSAIDs, and colorectal cancer. J Gastroenterol 1996;31:898-906.
https://doi.org/10.1007/BF02358623
PMID: 9027660

 

45 Sheng H, Shao J, Kirkland SC, Isakson P, Coffey RJ, Morrow J, Beauchamp RD, DuBois RN: Inhibition of human colon cancer cell growth by selective inhibition of cyclooxygenase-2. J Clin Invest 1997;99:2254-2259.
https://doi.org/10.1172/JCI119400
PMID: 9151799 PMCid:PMC508057

 

46 Guo Z, Jiang JH, Zhang J, Yang HJ, Yang FQ, Qi YP, Zhong YP, Su J, Yang RR, Li LQ, Xiang BD: COX-2 Promotes Migration and Invasion by the Side Population of Cancer Stem Cell-Like Hepatocellular Carcinoma Cells. Medicine 2015;94:e1806.
https://doi.org/10.1097/MD.0000000000001806
PMID: 26554780 PMCid:PMC4915881

 

47 Singh B, Berry JA, Shoher A, Ramakrishnan V, Lucci A: COX-2 overexpression increases motility and invasion of breast cancer cells. Int J Oncol 2005;26:1393-1399.
https://doi.org/10.3892/ijo.26.5.1393
PMID: 15809733

 

48 Grösch S, Maier TJ, Schiffmann S, Geisslinger G: Cyclooxygenase-2 (COX-2)-Independent Anticarcinogenic Effects of Selective COX-2 Inhibitors. J Natl Cancer Inst 2006;98:736-747.
https://doi.org/10.1093/jnci/djj206
PMID: 16757698

 

49 Sade A, Tunēay S, Ēimen İ, Severcan F, Banerjee S: Celecoxib reduces fluidity and decreases metastatic potential of colon cancer cell lines irrespective of COX-2 expression. Biosci Rep 2012;32:35.
https://doi.org/10.1042/BSR20100149
PMID: 21401528

 

50 Bocca C, Bozzo F, Cannito S, Parola M, Miglietta A: Celecoxib inactivates epithelial-mesenchymal transition stimulated by hypoxia and/or epidermal growth factor in colon cancer cells. Mol Carcinog 2012;51:783-795.
https://doi.org/10.1002/mc.20846
PMID: 21882253

 

51 Maier TJ, Schiffmann S, Wobst I, Birod K, Angioni C, Hoffmann M, Lopez JJ, Glaubitz C, Steinhilber D, Geisslinger G, Grosch S: Cellular membranes function as a storage compartment for celecoxib. J Mol Med 2009;87:981-993
https://doi.org/10.1007/s00109-009-0506-8
PMID: 19641861

 

52 Niederberger E, Tegeder I, Vetter G, Schmidtko A, Schmidt H, Euchenhofer C, Bräutigam L, Grösch S, Geisslinger G: Celecoxib loses its anti-inflammatory efficacy at high doses through activation of NF-κB. FASEB J 2001;15:1622-1624.
https://doi.org/10.1096/fj.00-0716fje
PMID: 11427506

 

53 Liu H, Liu X, Zhang C, Zhu H, Xu Q, Bu Y, Lei Y: Redox Imbalance in the Development of Colorectal Cancer. J Cancer 2017;8:1586-1597.
https://doi.org/10.7150/jca.18735
PMID: 28775778 PMCid:PMC5535714

 

54 Storz P: Reactive oxygen species in tumor progression. Front Biosci 2005;10:1881-1896.
https://doi.org/10.2741/1667
PMID: 15769673

 

55 Wang Z, Li Y, Sarkar FH: Signaling mechanism(s) of reactive oxygen species in Epithelial-Mesenchymal Transition reminiscent of cancer stem cells in tumor progression. Curr Stem Cell Res Ther 2010;5:74-80.
https://doi.org/10.2174/157488810790442813

 

56 Hurd TR, DeGennaro M, Lehmann R: Redox regulation of cell migration and adhesion. Trends Cell Biol 2012;22:107-115.
https://doi.org/10.1016/j.tcb.2011.11.002
PMID: 22209517 PMCid:PMC4515034

 

57 Supabphol A, Supabphol R: Antimetastatic potential of N-acetylcysteine on human prostate cancer cells. J Med Assoc Thai 2012;95:S56-62.
PMID: 23513466

 

58 Supabphol A, Muangman V, Chavasiri W, Supabphol R, Gritsanapan W: N-acetylcysteine inhibits proliferation, adhesion, migration and invasion of human bladder cancer cells. J Med Assoc Thai 2009;92:1171-1177.
PMID: 19772176

 

59 Galadari S, Rahman A, Pallichankandy S, Thayyullathil F: Reactive oxygen species and cancer paradox: To promote or to suppress? Free Radic Biol Med 2017;104:144-164.
https://doi.org/10.1016/j.freeradbiomed.2017.01.004
PMID: 28088622

 

60 Piskounova E, Agathocleous M, Murphy MM, Hu Z, Huddlestun SE, Zhao Z, Leitch AM, Johnson TM, DeBerardinis RJ, Morrison SJ: Oxidative stress inhibits distant metastasis by human melanoma cells. Nature 2015;527:186-191.
https://doi.org/10.1038/nature15726
PMID: 26466563 PMCid:PMC4644103

 

61 Le Gal K, Ibrahim MX, Wiel C, Sayin VI, Akula MK, Karlsson C, Dalin MG, Akyurek LM, Lindahl P, Nilsson J, Bergo MO: Antioxidants can increase melanoma metastasis in mice. Sci Transl Med 2015;7:308re308.
https://doi.org/10.1126/scitranslmed.aad3740
PMID: 26446958

 

62 Sayin VI, Ibrahim MX, Larsson E, Nilsson JA, Lindahl P, Bergo MO: Antioxidants accelerate lung cancer progression in mice. Sci Transl Med 2014;6:221ra215.
https://doi.org/10.1126/scitranslmed.3007653
PMID: 24477002

 

63 Gorrini C, Harris IS, Mak TW: Modulation of oxidative stress as an anticancer strategy. Nat Rev Drug Discov 2013;12:931-947.
https://doi.org/10.1038/nrd4002
PMID: 24287781

 

64 Dhillon AS, Hagan S, Rath O, Kolch W: MAP kinase signalling pathways in cancer. Oncogene 2007;26:3279.
https://doi.org/10.1038/sj.onc.1210421
PMID: 17496922

 

65 Chen Z, Gibson TB, Robinson F, Silvestro L, Pearson G, Xu B, Wright A, Vanderbilt C, Cobb MH: MAP kinases. Chem Rev 2001;101:2449-2476.
https://doi.org/10.1021/cr000241p
PMID: 11749383

 

66 Kim EK, Choi EJ: Pathological roles of MAPK signaling pathways in human diseases. Biochim Biophys Acta 2010;1802:396-405.
https://doi.org/10.1016/j.bbadis.2009.12.009
PMID: 20079433

 

67 Chen L, Liu L, Huang S: Cadmium activates the mitogen-activated protein kinase (MAPK) pathway via induction of reactive oxygen species and inhibition of protein phosphatases 2A and 5. Free Radic Biol Med 2008;45:1035-1044.
https://doi.org/10.1016/j.freeradbiomed.2008.07.011
PMID: 18703135

 

68 Cuenda A, Rousseau S: p38 MAP-Kinases pathway regulation, function and role in human diseases. Biochimica et Biophysica Acta 2007;1773:1358-1375.
https://doi.org/10.1016/j.bbamcr.2007.03.010
PMID: 17481747

 

69 Wagner EF, Nebreda AR: Signal integration by JNK and p38 MAPK pathways in cancer development. Nat Rev Cancer 2009;9:537-549.
https://doi.org/10.1038/nrc2694
PMID: 19629069

 

70 Düzgün ŞA, Yerlikaya A, Zeren S, Bayhan Z, Okur E, Boyacı İ: Differential effects of p38 MAP kinase inhibitors SB203580 and SB202190 on growth and migration of human MDA-MB-231 cancer cell line. Cytotechnology 2017;69:711-724.
https://doi.org/10.1007/s10616-017-0079-2
PMID: 28393288 PMCid:PMC5507849

 

71 Huang S, New L, Pan Z, Han J, Nemerow GR: Urokinase Plasminogen Activator/Urokinase-specific Surface Receptor Expression and Matrix Invasion by Breast Cancer Cells Requires Constitutive p38α Mitogen-activated Protein Kinase Activity. J Biol Chem 2000;275:12266-12272.
https://doi.org/10.1074/jbc.275.16.12266
PMID: 10766865

 

72 Huth HW, Santos DM, Gravina HD, Resende JM, Goes AM, de Lima ME, Ropert C: Upregulation of p38 pathway accelerates proliferation and migration of MDA-MB-231 breast cancer cells. Oncol Rep 2017;37:2497-2505.
https://doi.org/10.3892/or.2017.5452
PMID: 28260101

 

73 Wada T, Penninger JM: Mitogen-activated protein kinases in apoptosis regulation. Oncogene 2004;23:2838-2849.
https://doi.org/10.1038/sj.onc.1207556
PMID: 15077147

 

74 Koul HK, Pal M, Koul S: Role of p38 MAP Kinase Signal Transduction in Solid Tumors. Genes Cancer 2013;4:342-359.
https://doi.org/10.1177/1947601913507951
PMID: 24349632 PMCid:PMC3863344

 

75 Han J, Sun P: The pathways to tumor suppression via route p38. Trends Biochem Sci 2007;32:364-371.
https://doi.org/10.1016/j.tibs.2007.06.007
PMID: 17624785

 

76 Hernandez Losa J, Parada Cobo C, Guinea Viniegra J, Sanchez-Arevalo Lobo VJ, Ramon y Cajal S, Sanchez-Prieto R: Role of the p38 MAPK pathway in cisplatin-based therapy. Oncogene 2003;22:3998-4006.
https://doi.org/10.1038/sj.onc.1206608
PMID: 12821934

 

77 Parker J, Kaplon MK, Alvarez CJ, Krishnaswamy G: Prostaglandin H synthase expression is variable in human colorectal adenocarcinoma cell lines. Exp Cell Res 1997;236:321-329.
https://doi.org/10.1006/excr.1997.3741
PMID: 9344613

 

78 Hsi LC, Joon Baek S, Eling TE: Lack of Cyclooxygenase-2 Activity in HT-29 Human Colorectal Carcinoma Cells. Exp Cell Res 2000;256:563-570.
https://doi.org/10.1006/excr.2000.4863
PMID: 10772828

 

79 Sadeghi N, Gerber DE: Targeting the PI3K pathway for cancer therapy. Future Med Chem 2012;4:1153-1169.
https://doi.org/10.4155/fmc.12.56
PMID: 22709255 PMCid:PMC4276042

 

80 Hu L, Zaloudek C, Mills GB, Gray J, Jaffe RB: Ovarian Carcinoma Growth Inhibition by a Phosphatidylinositol 3-Kinase Inhibitor (LY294002). Clin Cancer Res 2000;6:880.
PMID: 10741711

 

81 Thant AA, Nawa A, Kikkawa F, Ichigotani Y, Zhang Y, Sein TT, Amin AR, Hamaguchi M: Fibronectin activates matrix metalloproteinase-9 secretion via the MEK1-MAPK and the PI3K-Akt pathways in ovarian cancer cells. Clin Exp Metastasis 2000;18:423-428.
https://doi.org/10.1023/A:1010921730952

 

82 Roy HK, Olusola BF, Clemens DL, Karolski WJ, Ratashak A, Lynch HT, Smyrk TC: AKT proto-oncogene overexpression is an early event during sporadic colon carcinogenesis. Carcinogenesis 2002;23:201-205.
https://doi.org/10.1093/carcin/23.1.201
PMID: 11756242

 

83 Turečkovį J, Vojtěchovį M, Krausovį M, Šloncovį E, Korķnek V: Focal Adhesion Kinase Functions as an Akt Downstream Target in Migration of Colorectal Cancer Cells. Transl Oncol 2009;2:281-290.
https://doi.org/10.1593/tlo.09160
PMID: 19956390 PMCid:PMC2781073

 

84 Leslie NR, Downes CP: PTEN: The down side of PI 3-kinase signalling. Cell Signal 2002;14:285-295.
https://doi.org/10.1016/S0898-6568(01)00234-0

 

85 Hawcroft G, Ko CW, Hull MA: Prostaglandin E2-EP4 receptor signalling promotes tumorigenic behaviour of HT-29 human colorectal cancer cells. Oncogene 2007;26:3006-3019.
https://doi.org/10.1038/sj.onc.1210113
PMID: 17130837

 

86 Mutoh M, Watanabe K, Kitamura T, Shoji Y, Takahashi M, Kawamori T, Tani K, Kobayashi M, Maruyama T, Kobayashi K, Ohuchida S, Sugimoto Y, Narumiya S, Sugimura T, Wakabayashi K: Involvement of prostaglandin E receptor subtype EP(4) in colon carcinogenesis. Cancer Res 2002;62:28-32.
PMID: 11782353

 

87 Watanabe K, Kawamori T, Nakatsugi S, Ohta T, Ohuchida S, Yamamoto H, Maruyama T, Kondo K, Ushikubi F, Narumiya S, Sugimura T, Wakabayashi K: Role of the prostaglandin E receptor subtype EP1 in colon carcinogenesis. Cancer Res 1999;59:5093-5096.
PMID: 10537280