Skip to main content

Suppression of colitis-related mouse colon carcinogenesis by a COX-2 inhibitor and PPAR ligands

Abstract

Background

It is generally assumed that inflammatory bowel disease (IBD)-related carcinogenesis occurs as a result of chronic inflammation. We previously developed a novel colitis-related mouse colon carcinogenesis model initiated with azoxymethane (AOM) and followed by dextran sodium sulfate (DSS). In the present study we investigated whether a cyclooxygenase (COX)-2 inhibitor nimesulide and ligands for peroxisome proliferator-activated receptors (PPARs), troglitazone (a PPARγ ligand) and bezafibrate (a PPARα ligand) inhibit colitis-related colon carcinogenesis using our model to evaluate the efficacy of these drugs in prevention of IBD-related colon carcinogenesis.

Methods

Female CD-1 (ICR) mice were given a single intraperitoneal administration of AOM (10 mg/kg body weight) and followed by one-week oral exposure of 2% (w/v) DSS in drinking water, and then maintained on the basal diets mixed with or without nimesulide (0.04%, w/w), troglitazone (0.05%, w/w), and bezafibrate (0.05%, w/w) for 14 weeks. The inhibitory effects of dietary administration of these compounds were determined by histopathological and immunohistochemical analyses.

Results

Feeding with nimesulide and troglitazone significantly inhibited both the incidence and multiplicity of colonic adenocarcinoma induced by AOM/DSS in mice. Bezafibrate feeding significantly reduced the incidence of colonic adenocarcinoma, but did not significantly lower the multiplicity. Feeding with nimesulide and troglitazone decreased the proliferating cell nuclear antigen (PCNA)-labeling index and expression of β-catenin, COX-2, inducible nitric oxide synthase (iNOS) and nitrotyrosine. The treatments increased the apoptosis index in the colonic adenocarcinoma. Feeding with bezafibrate also affected these parameters except for β-catenin expression in the colonic malignancy.

Conclusion

Dietary administration of nimesulide, troglitazone and bezafibrate effectively suppressed the development of colonic epithelial malignancy induced by AOM/DSS in female ICR mice. The results suggest that COX-2 inhibitor and PPAR ligands could serve as an effective agent against colitis-related colon cancer development.

Peer Review reports

Background

Colorectal cancer (CRC) is one of the leading causes of death in the world. This malignancy is also one of the most serious complications of inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease [1]. Long-term UC patients have an increased risk of developing CRC compared with the general population [2]. The precise mechanisms of the IBD-related carcinogenesis process are largely unclear, although it is generally assumed that IBD-related carcinogenesis occurs as a result of chronic inflammation [3].

Several agents, such as folic acid, short chain fatty acid (butyrate), ursodeoxycholic acid, and 5-aminosalicylic acid, have been suggested to be useful for prevention of CRC in UC [4]. Epidemiological studies have shown that prolonged use of aspirin is associated with a reduced risk of CRC [5]. Consistent with these data, several non-steroidal anti-inflammatory drugs (NSAIDs), including cyclooxygenase (COX)-2 inhibitors, suppressed the development of chemically-induced colon carcinomas in rats [6] and intestinal polyps in Min mice with a nonsense mutation of the Apc gene [7]. In addition, clinical trials have demonstrated that a NSAID sulindac causes regression of adenomas in patients with familial adenomatous polyposis [8]. Nimesulide (4-nitro-2-phenoxymethanesulfonanilide), a selective inhibitor of COX-2, belonging to the sulfonamide class [9], is less ulcerogenic than other NSAIDs [10], and suppresses the formation of aberrant crypt foci (ACF), being putative precancerous lesions of the colon cancer, induced by a colon carcinogen, azoxymethane (AOM) in rats [11]. Moreover, this COX-2 inhibitor could effectively reduce the development of intestinal polyps in Min mice [12].

Peroxisome proliferator-activated receptors (PPARs?) are ligand-activated transcription factors, belonging to the nuclear hormone receptor superfamily [13]. Three PPAR isotypes, PPARα, PPARδ(β), and PPARγ, have been identified. PPARγ is highly expressed in fat tissue, and play important roles in adipocyte differentiation and lipid storage [14]. PPARγ is also expressed in a number of epithelial neoplasms, such as cancers in colon, breast, and prostate [13]. PPARγ ligands, including thiazolidinediones (troglitazone and rosiglitazone) and tyrosine analogue (GW7845), can induce apoptosis and adipogenic differentiation, and inhibit tumor growth both in vitro and in vivo studies [15–17]. We previously reported that pioglitazone, bezafibrate or troglitazone in diet are able to suppress ACF formation induced by dextran sodium sulfate (DSS)/AOM in the rat colon [18]. Osawa et al. [19] confirmed our findings by demonstrating that ligands for PPARγ (troglitazone, rosiglitazone, and pioglitazone) suppress the occurrence of colonic tumors in mice initiated with AOM. Niho et al. [20] also demonstrated that ligands for PPARα (bezafibrate) and PPARγ (pioglitazone) suppress intestinal polyp formation in Apc-deficient mice. Moreover, PPARγ could suppress β-catenin levels and colon carcinogenesis during the early steps of tumor formation [21]. On the other hand, high doses of troglitazone and rosiglitazone can promote polyp formation in the Min mouse colon [22, 23].

For understanding the pathogenesis of IBD and IBD-related CRC, several animal models have been established. Most used is a mouse model with DSS [24]. Modifying effects of several xenobiotics on IBD-related colon carcinogenesis were reported [25] in animal models of IBD. However, the colitis model using DSS with or without carcinogen needs to a long period and repeated administration of DSS to induce colitis and colitis-related CRC. Recently, an endogenous anti-inflammatory PPARγ pathway was suggested in the intestine, which was found in PPARγ-deficient mice [26, 27]. To search novel and effective chemopreventive agents against IBD-related CRC, we recently have developed a novel colitis-related CRC mouse model, in which large bowel adenocarcinomas occur within 20 weeks and their histology and biological characteristics are resemble to those found in human cased [28]. As a part of our search for a safer chemopreventive agent for colitis-related colon cancer, in the present study we examined the chemopreventive ability of nimesulide, troglitazone, and bezafibrate using our mouse colon carcinogenesis model for colitis-related colon carcinogenesis [28].

Methods

Animals, chemicals and diets

Female Crj: CD-1 (ICR) (Charles River Japan Inc., Tokyo, Japan) aged 5 weeks were used in this study. They were maintained at Kanazawa Medical University Animal Facility according to the Institutional Animal Care Guidelines. The mice were quarantined for the first 7 days then randomized by body weight into experimental and control groups. All animals were housed in plastic cages (five or six mice/cage) with free access to drinking water and a pelleted basal diet, CRF-1 (Oriental Yeast Co., Ltd., Tokyo, Japan), under controlled conditions of humidity (50 ± 10%), light (12/12 hour light/dark cycle) and temperature (23 ± 2°C). A colonic carcinogen AOM was purchased from Sigma Chemical Co. (St. Louis, MO, USA). DSS with a molecular weight of 40,000 was purchased from ICN Biochemicals, Inc. (Aurora, OH, USA). DSS for induction of colitis was dissolved in distilled water at a concentration of 2% (w/v). Nimesulide was purchased from Sigma Chemical Co. (St. Louis, MO, USA). Troglitazone and bezafibrate were kindly supplied by Sankyo Co. (Tokyo, Japan) and Kissei Pharmaceutical Co. (Matsumoto, Japan), respectively. Experimental diet containing nimesulide (0.04%, w/w), troglitazone (0.05%, w/w) or bezafibrate (0.05%, w/w) was prepared every week by mixing the respective compound in powdered basal diet CRF-1. The dose levels were determined on the basis of previous studies [18, 29].

Experimental procedure

A total of 71 female ICR mice were divided into 7 experimental and control groups (Figure 1). Mice in groups 1 through 4 were given a single intraperitoneal injection of AOM (10 mg/kg body weight). Starting one week after the AOM injection, animals in group 1 were administered to 2%DSS in drinking water for 7 days, and then followed without any further treatment for 15 weeks. Mice in groups 2, 3, and 4 were fed the diets containing 0.04% nimesulide, 0.05% troglitazone, and 0.05% bezafibrate, respectively, for 14 weeks, starting 1 week after the stop of DSS administration. Animals in groups 5, 6, and 7 were respectively given the diets containing 0.04% nimesulide, 0.05% troglitazone, and 0.05% bezafibrate alone for 14 weeks. Group 8 was given a single dose of AOM. Group 9 was given 2% DSS for 7 days. Group 10 consisted of untreated mice. All animals were sacrificed at the end of the study (Week 17) by ether overdose. Their large bowels were flushed with saline, excised, measured their length (from ileocecal junction to the anal verge), cut open longitudinally along the main axis, and then washed with saline. The large bowels were macroscopically inspected, cut, and fixed in 10% buffered formalin for at least 24 hours. Histopathological examination was performed on paraffin-embedded sections after hematoxylin and eosin (H & E) staining. Colonic mucosal dysplasia (mild- and severe-graded) was diagnosed according to the criteria described by Ridell et al. [30] and Pascal [31]. Colonic neoplasms were diagnosed according to the description by Ward [32].

Figure 1
figure 1

Experimental protocol. Arrows, AOM 10 mg/kg body weight, i.p. injection; densely cross-hatched bars, 2% dextran sodium sulfate (DSS) in drinking water; open bars, basal diet and tap water; crosses, death.

Immunohistochemistry

Immunohistochemistry for the proliferating cell nuclear antigen (PCNA), apoptotic nuclei, β-catenin, COX-2, inducible nitric oxide synthase (iNOS), and nitrotyrosine was performed on 4-μm-thick paraffin-embedded sections from colons of mice in each group by the labeled streptavidin biotin method using a LSAB KIT (DAKO Japan, Kyoto, Japan) with microwave accentuation. The paraffin-embedded sections were heated for 30 min at 65°C, deparaffinized in xylene, and rehydrated through graded ethanol at room temperature. A 0.05 M Tris HCl buffer (pH 7.6) was used to prepare solutions and for washes between various steps. Incubations were performed in a humidified chamber. For the determination of PCNA-incorporated nuclei, the PCNA-immunohistochemistry was performed according to the method described by Watanabe et al. [33]. Apoptotic index was also evaluated by immunohistochemistry for single stranded DNA (ssDNA) [33]. Sections were treated for 40 min at room temperature with 2% BSA and incubated overnight at 4°C with primary antibodies. Primary antibodies included anti-PCNA mouse monoclonal antibody (diluted 1:50; PC10, DAKO Japan), anti-ssDNA rabbit polyclonal antibody (diluted 1:300, DAKO Japan), anti-β-catenin mouse monoclonal antibody (diluted 1:1000, Transduction Laboratories, Lexington, KY, USA), anti-COX-2 rabbit polyclonal antibody (diluted 1:50, IBL Co., Ltd., Gunma, Japan), anti-iNOS rabbit polyclonal antibody (diluted 1:1000, Wako Pure Chemical Industries, Ltd., Osaka, Japan), and anti-nitrotyrosine rabbit polyclonal antibody (diluted 1:500, Upstate Biotechnology, Lake Placid, NY, USA). To reduce the non-specific staining of mouse tissue by the mouse antibodies, a Mouse On Mouse IgG blocking reagent (Vector Laboratories, Inc., Burlingame, CA, USA) was applied for 1 h. Horseradish peroxidase activity was visualized by treatment with H2O2 and 3,3'-diaminobenzidine for 5 min. At the last step, the sections were weakly counterstained with Mayer's hematoxylin (Merck Ltd., Tokyo, Japan). For each case, negative controls were performed on serial sections. On the control sections, incubation with the primary antibodies was omitted.

Intensity and localization of immunoreactivities against all primary antibodies used were examined on all sections using a microscope (Olympus BX41, Olympus Optical Co., Ltd., Tokyo, Japan). The PCNA and apoptotic indices were determined by counting the number of positive cells among at least 200 cells in the lesion, and were indicated as percentages. Each slide for β-catenin, COX-2, iNOS, and nitrotyrosine immunohistochemistry was evaluated for intensity of immunoreactivity on a 0 to 4+ scale. The overall intensity of the staining reaction was scored with 0 indicating no immunoreactivity and no positive cells, 1+ weak immunoreactivity and < 10% of positive cells, 2+ mild immunoreactivity and 10–30% of positive cells, 3+ moderate immunoreactivity and 31–60% of positive cells, and 4+ strong immunoreactivity and 61–100% of positive cells.

Statistical analysis

All measurements were compared by Student's t-test, Welch's t-test or Fisher's exact probability test for paired samples.

Results

General observation

Bloody stool was noted in a few mice received 2% DSS and their body weight gains were slightly decreased during the period of the treatment. However, thereafter no such clinical symptoms were observed. After Week 12, anal prolapsus due to the tumor development in the distal colon was found in a few mice treated with AOM and 2% DSS (group 1). The body weights and lengths of large bowel of mice in all groups at the end of the study are shown in Table 1. The mean body weights of groups 2 (AOM/DSS/0.04% nimesulide, P < 0.005), 3 (AOM/DSS/0.04% troglitazone, P < 0.02), and 4 (AOM/DSS/0.05% bezafibrate, P < 0.05), were significantly higher than that of group 1 (AOM/DSS). The mean body weight of group 9 (DSS alone, P < 0.05) was significantly lower than that of group 10 (untreated). The mean liver weights of mice in groups 3 (P < 0.05) and 4 (P < 0.01) were significantly greater than that of group 1. However, there were no pathological alterations suggesting toxicity of test compounds in the liver, kidneys, lung, and heart of mice (data not shown).

Table 1 Body, liver, relative liver weights, and length of colon.

Pathological findings

Macroscopically, nodular, polypoid or caterpillar-like tumors were observed in the middle and distal colon of mice in groups 1 through 4. They were histologically tubular adenoma (Figure 2a) or well-/moderately-differentiated tubular adenocarcinoma (Figure 2b). Dysplasia (Figure 2c–e) was also developed in mice of these groups. Animals of groups 5–10 did not have large bowel neoplasms and dysplasia. The incidences and multiplicity of colon neoplasma are shown in Table 2. Group 1 (AOM/DSS) induced 100% incidence of colon adenocarcinomas with a multiplicity of 3.0 ± 1.8. The incidences of colorectal adenocarcinomas in groups 2 (AOM/DSS/0.04% nimesulide), 3 (AOM/DSS/0.05% troglitazone), and 4 (AOM/DSS/0.05% bezafibrate) were significantly smaller than that of group 1 (P < 0.01, P < 0.01 and P < 0.05, respectively). The multiplicities of colon adenocarcinomas in groups 2 and 3 were also significantly lower than that of group 1 (P < 0.005 and P < 0.05, respectively). While the multiplicity of colon adenocarcinoma of group 4 (AOM/DSS/0.05% bezafibrate) was smaller than group 1, the difference was insignificant. In this study, mucosal ulcer with or without focal dysplasia (Figure 2c–e) were also found in the distal colon of mice in groups 1 through 4. The incidences and multiplicity of colonic ulceration and dysplasia are shown in Table 3. The incidences and multiplicities of colorectal mucosal ulcer and dysplasia of groups 2, 3, and 4 were smaller than group 1, but the differences did not reach to statistical significance.

Table 2 Incidence and multiplicity of colonic neoplasia.
Table 3 Incidence of multiplicity colonic mucosal ulcer and dysplasia.
Figure 2
figure 2

Histopathology of colonic lesions in mice of group 1. (a) adenoma, (b) adenocarcinoma, (c) mild dysplasia, and (d) severe dysplasia. Hematoxylin and eosin stain. Original magnifications, × 10.

Immunohistochemistry for PCNA, ssDNA, β-catenin, COX-2, iNOS and nitrotyrosine in colonic adenocarcinoma

As summarized in Table 4, PCNA-labeling index (Figure 3a) of colonic adenocarcinomas developed in groups 2, 3, and 4 was significantly smaller than group 1 (P < 0.001). Apoptotic index measured by ssDNA immunohistochemistry (Figure 3b) in groups 2, 3, and 4 was significantly greater than group 1 (P < 0.001).

Table 4 PCNA and apoptosis indices and scores of β-catenin, COX-2, iNOS and nitrotyrosine expression in colonic adenocarcinomas.
Figure 3
figure 3

Immunohistochemistry of PCNA, ssDNA, β-catenin COX-2, iNOS, and nitrotyrosine in mice of group 1. (a) PCNA immunohistochemistry, (b) ssDNA immunohistochemistry, (c) β-catenin immunohistochemistry, (d) COX-2 immunohistochemistry, (e) iNOS immunohistochemistry, and (f) nitrotyrosine immunohistochemistry. Original magnifications: a, d, e, f, × 10; c, × 20; b, × 40. Insets, immunohistochemistry staining for each antibody in the mouse colon from group 10 (Original magnifications, × 10).

Strong β-catenin expression was seen in the nucleus and cytoplasm of adenocarcinoma cells (Figure 3c). Although the intensity was relatively weaker than carcinoma cells, adenoma cells showed positivity for β-catenin in their cytoplasm and cell membrane. β-catenin immunoreactivity was also found in the cell membrane and cytoplasm of dysplastic cells. Non-lesional cryptal cells showed weak positivity of β-catenin in their cell membrane. In the positive cases of COX-2 (Figure 3d), and iNOS (Figure 3e) expression in the dysplasia and adenocarcinoma, the staining pattern was granular and localized to cytoplasm and/or nuclei. Slight immunoreactivity for COX-2 and iNOS was observed in the superficial layers of the non-lesional colonic mucosa and in parts of basal layer in all groups. The expression pattern between COX-2 and iNOS of colorectal adenocarcinomas was well correlated. Furthermore, a positive staining for nitrotyrosine, a marker of nitrosative injury, was mainly observed in mononuclear cells infiltrated in the colonic mucosa (Figure 3f). Neoplastic cells also showed negative or very weakly positive immnoreactivity of nitrotyrosine. Scores for β-catenin, COX-2 and iNOS expression in colonic adenocarcinomas are given in Table 4. β-Catenin expression scores of colorectal adenocarcinomas in groups 2 and 3 were significantly decreased when compared with that in group 1 (P < 0.05 and P < 0.05, respectively). Scores for COX-2 and iNOS expression of colorectal adenocarcinomas in groups 2, 3, and 4, were significantly smaller than those in group 1 (P < 0.001). The scores of nitrotyrosine positivity in groups 2, 3, and 4 were significantly lower than group 1 (P < 0.001).

Discussion

The results of the present work clearly indicated that a COX-2 inhibitor nimesulide and a PPARγ ligand troglitazone effectively inhibited AOM/DSS-induced colitis-related colonic carcinogenesis in mice. Inhibitory effect of nimesulide was superior to that of troglitazone. Bezafibrate also reduced the occurrence of colonic adenocarcinoma, but the ability was relatively weaker than that of nimesulide and troglitazone. The suppressive effects of nimesulide, troglitazone and bezafibrate on the development of colonic adenocarcinoma was well correlated with the inhibition of cell proliferation activity, induction of apoptosis, and lowered immunoreactivity of β-catenin, COX-2, iNOS, and nitrotyrosine in the colonic malignancies. However, no differences on the frequency of dysplastic lesions could be observed among the groups. These data may suggest that the pharmacological classes tested under the present investigation slow down the time course of tumor development rather than completely preventing it.

The pathogenesis of IBD-associated colorectal carcinogenesis is widely believed to involve a step-wise progression from inflamed and hyperplastic epithelia through flat dysplasia to finally adenocarcinoma [30]. IBD-associated colorectal carcinogenesis is probably promoted by chronic inflammation, but the mechanism is still unclear. However, mucosal inflammation may result in colonic carcinogenesis through several proposed mechanisms such as induction of genetic mutations, increased-cryptal cell proliferation, changes in crypt cell metabolism, changes in bile acid enterohepatic circulation, and alterations in bacteria flora [4, 34]. These events are considered to promote IBD-associated CRC development. Given the correlation between increased COX-2 expression and colonic carcinoma and/or inflammation, the chemopreventive effects of NSAIDs seem to be mediated, at least in part, by COX inhibition [35]. We [36] and others [29, 37] demonstrated that NSAIDs including nimesulide inhibited both colon tumorigenesis and colitis. In the current study, powerful chemopreventive ability of nimesulide was observed in our colitis-related mouse colon carcinogenesis model, suggesting that nimesulide can be applied as an effective chemopreventor of both sporadic and IBD-associated cololectral carcinogenesis.

We previously demonstrated that dietary administration of PPARα and PPARγ ligands inhibits AOM and/or DSS-induced ACF in rodents [38]. In the present study, cancer chemopreventive ability of the PPARγ ligand, troglitazone, or the PPARα ligand, bezafibrate, was found in AOM/DSS-induced mouse colon carcinogenesis model, although their ability was lower than nimesulide. Inhibition of colonic inflammation and decrease in cell proliferation activity by these PPARs ligands might be responsible for their chemopreventive effects on colitis-associated colon carcinogenesis [38]. DNA damage caused by reactive oxygen and nitrogen species may contribute to colitis-related colon tumorigenesis [39]. Several NSAIDs can bind to PPARα and PPARγ [40]. Their anti-inflammatory activities might be mediated through inhibition of COX-1 and/or COX-2. PPARα could suppress COX-2 induction [41]. In addition, immunomodulation by the PPARs ligands might contribute to inhibition of colitis and colon carcinogenesis [42].

Expression and activity of iNOS is increased in colonic mucosa in patients with IBD [43] and colonic adenomas [44]. Several studies using experimental colon carcinogenesis models indicate that chemically induced colon tumors have higher expression and/or activity of iNOS compared with those in their adjacent non-tumorous tissues [12, 25]. Numerous iNOS-positive and nitrotyrosine-positive inflammatory cells are observed in non-cancerous colonic mucosa of mice treated with DSS [25]. PPARα [45] and PPARγ [46] involve in inflammation control, and can inhibit iNOS expression [47]. In addition, Rao et al. [48] showed that an iNOS-selective inhibitor suppresses AOM-induced colonic ACF development and iNOS activity. Furthermore, nitrotyrosine may originate from the reaction of iNOS generated NO with reactive oxygen species [49] or the myeloperoxidase-dependent pathway [50]. In the present study, we found a positive immunoreactivity for iNOS and nitrotyrosine in the inflamed colon, suggesting the formation of peroxynitrite and other NO-derived oxidants. These results may suggest that one of the mechanisms by which tested agents exert chemopreventive ability might be related to suppression of iNOS activity and/or expression.

Cell proliferation plays an important role in multi-step carcinogenesis [51]. In the colon, the number of cryptal cells is strictly regulated by a balance between cell proliferation and cell death that maintains homeostasis [52]. Changes in cell proliferation and apoptosis are regarded as a common denominator in the pathogenesis of tumor formation [53]. Reduced tumor incidence is generally associated with decreases in cellular proliferation and/or increases in apoptosis [54]. An increased COX-2 expression in CRC [55, 56] may confer a survival advantage on cells by inhibition of apoptosis and a change in cellular adhesion to the extracellular matrix [57]. Cancer cells treated with PPARγ ligands induce cell differentiation and apoptosis [14, 17]. Recently, Tardieu et al. [58] demonstrated that nimesulide increases apoptosis in colonic mucosa of DSS-treated rats. Our findings that nimesulide and troglitazone inhibited cell proliferation activity and induced apoptosis in colorectal mucosa are in accordance with these findings. Thus, cellular responses like cell growth and/or apoptosis to nimesulide and troglitazone may contribute to chemopreventive effects against colon carcinogenesis processes.

β-Catenin is a key regulator of the cadherin-mediated cell-cell adhesion system and an important element in the Wnt signal transduction pathway [59]. Accumulated β-catenin interacts with T-cell factor (Tcf) or lymphoid-enhancer factor (Lef) and translocates to the nucleus, in which it transactivates target genes including c-myc and cyclin D1 that are the potentially oncogenic [47, 60] in the cytoplasm or nucleus as a consequence of mutant Apc or β-catenin genesis frequently observed in early stages of colorectal carcinogenesis [61, 62]. Recently, Girnun et al. [21] indicated that a ligand of PPARγ suppresses β-catenin levels and colon carcinogenesis in Pparγ+/- mice treated with AOM. Furthermore, COX-2 is regulated by nuclear β-catenin accumulation [63]. In the current study, treatment with nimesulide, troglitazone, and bezafibrate significantly suppressed β-catenin expression in colorectal adenocarcinomas. Thus, it seems likely that the preventive efficacies of the COX-2 inhibitor (nimesulide) or the PPAR ligands (troglitazone and bezafibrate) against AOM/DSS induced mouse colon carcinogenesis might be mediated, at least in part, by β-catenin down-regulation.

Chemoprevention of cancer might be defined as the deliberate introduction of these selected non-toxic substances into the diet for the purpose of reducing cancer development. In the present study, the mean liver weights of mice in dietary feeding of troglitazone and bezafibrate were significantly increased. However, there were no pathological alterations suggesting toxicity of the drugs examined. It is known that administration of PPARs ligands to rodent exhibit hepatomegaly due to both cellular hypertrophy and hyperplasia [64]. Increase in liver weight by exposure of troglitazone and bezafibrate in this study may be caused by these pathological changes. The estimated daily intakes of nimesulide, troglitazone, and bezafibrate in mice given the diets containing 400 ppm and 500 ppm were approximately 160 mg/kg and 200 mg/kg in the present study. In a direct extrapolation to a 60 kg person, these doses are slightly lower than those of clinical trial [65–67]. These findings may suggest that the efficacy of these agents at dietary dose-levels has a direct practical and translational relevance to human.

Conclusion

In conclusion, dietary administration of nimesulide, troglitazone, and bezafibrate could effectively suppress colon carcinogenesis induced by AOM and DSS in female ICR mice. Our on-going study on molecular profiles in colonic samples from the current experiment will provide precise molecular mechanisms involved in their inhibitory action in AOM/DSS-induced mouse colon carcinogenesis.

Abbreviations

AOM:

azoxymethane

ACF:

aberrant crypt foci

CRC:

Colorectal cancer

COX-2:

cyclooxygenase-2

DSS:

dextran sodium sulfate

FAP:

familial adenomatous polyposis

H&E:

hematoxylin and eosin

IBD:

inflammatory bowel disease

iNOS:

inducible nitric oxide synthase

NSAID:

non-steroidal anti-inflammatory drug

PPAR:

peroxisome proliferator-activated receptor

PCNA:

proliferating cell nuclear antigen

ssDNA:

single stranded DNA

UC:

ulcerative colitis.

References

  1. Eaden JA, Abrams KR, Mayberry JF: The risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut. 2001, 48: 526-535. 10.1136/gut.48.4.526.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ekbom A, Helmick C, Zack M, Adami HO: Ulcerative colitis and colorectal cancer. A population-based study. N Engl J Med. 1990, 323: 1228-1233.

    Article  CAS  PubMed  Google Scholar 

  3. Weitzman SA, Gordon LI: Inflammation and cancer: role of phagocyte-generated oxidants in carcinogenesis. Blood. 1990, 76: 655-663.

    CAS  PubMed  Google Scholar 

  4. Seril DN, Liao J, Yang GY, Yang CS: Oxidative stress and ulcerative colitis-associated carcinogenesis: stuides in human and animal models. Carcinogenesis. 2003, 24: 353-362. 10.1093/carcin/24.3.353.

    Article  CAS  PubMed  Google Scholar 

  5. Thun MJ, Namboodiri MM, Heath CWJ: Aspirin use and reduced risk of fatal colon cancer. N Engl J Med. 1991, 325: 1593-1596.

    Article  CAS  PubMed  Google Scholar 

  6. Reddy BS, Tokumo K, Kulkarni N, Aligia C, Kelloff G: Inhibition of colon carcinogenesis by prostaglandin synthesis inhibitors and related compounds. Carcinogenesis. 1992, 13: 1019-1023.

    Article  CAS  PubMed  Google Scholar 

  7. Jacoby RF, Marshall DJ, Newton MA, Novakovic K, Tutsch K, Cole CE, Lubet RA, Kelloff GJ, Verma A, Moser AR, Dove WF: Chemoprevention of spontaneous intestinal adenomas in the Apc Min mouse model by the nonsteroidal anti-inflammatory drug piroxicam. Cancer Res. 1996, 56: 710-714.

    CAS  PubMed  Google Scholar 

  8. Giardiello FM, Hamilton SR, Krush AJ, Piantadosi S, Hylind LM, Celano P, Booker SV, Robinson CR, Offerhaus GJ: Treatment of colonic and rectal adenomas with sulindac in familial adenomatous polyposis. N Engl J Med. 1993, 328: 1313-1316. 10.1056/NEJM199305063281805.

    Article  CAS  PubMed  Google Scholar 

  9. Taniguchi Y, Ikesue A, Yokoyama K, Noda K, Debuchi H, Nakamura T, Toda A: Selective inhibition by nimesulide, a novel non-steroidal anti-inflammatory drug, with prostaglandin endoperoxide synthase-2 activity in-vitro. Pharm Sci. 1995, 1: 173-175.

    Google Scholar 

  10. Cipollini F, Mecozzi V, Altilia F: Endoscopic assessment of the effects on nimesulide on the gastric mucosa: comparison with indomethacin. Curr Ther Res. 1989, 45: 1042-1048.

    Google Scholar 

  11. Takahashi M, Fukutake M, Yokota S, Ishida K, Wakabayashi K, Sugimura T: Suppression of azoxymethane-induced aberrant crypt foci in rat colon by nimesulide, a selective inhibitor of cyclooxygenase 2. J Cancer Res Clin Oncol. 1996, 122: 219-222. 10.1007/BF01209649.

    Article  CAS  PubMed  Google Scholar 

  12. Nakatsugi S, Fukutake M, Takahashi M, Fukuda K, Isoi T, Taniguchi Y, Sugimura T, Wakabayashi K: Suppression of intestinal polyp development by nimesulide, a selective cyclooxygenase-2 inhibitor, in Min mice. Jpn J Cancer Res. 1997, 88: 1117-1120.

    Article  CAS  PubMed  Google Scholar 

  13. Rosen ED, Spiegelman BM: PPARg : a nuclear regulator of metabolism, differentiation, and cell growth. J Biol Chem. 2001, 276: 37731-37734. 10.1074/jbc.M106424200.

    Article  CAS  PubMed  Google Scholar 

  14. Tontonoz P, Singer S, Forman BM, Sarraf P, Fletcher JA, Fletcher CD, Brun RP, Mueller E, Altiok S, Oppenheim H, Evans RM, Spiegelman BM: Terminal differentiation of human liposarcoma cells induced by ligands for peroxisome proliferator-activated receptor gamma and the retinoid X receptor. Proc Natl Acad Sci USA. 1997, 94: 237-241. 10.1073/pnas.94.1.237.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Sarraf P, Mueller E, Jones D, King FJ, DeAngelo DJ, Partridge JB, Holden SA, Chen LB, Singer S, Fletcher C, Spiegelman BM: Differentiation and reversal of malignant changes in colon cancer through PPARg. Nat Med. 1998, 4: 1046-1052. 10.1038/2030.

    Article  CAS  PubMed  Google Scholar 

  16. Suh N, Wang Y, Williams CR, Risingsong R, Gilmer T, Willson TM, Sporn MB: A new ligand for the peroxisome proliferator-activated receptor-g (PPAR-g), GW7845, inhibits rat mammary carcinogenesis. Cancer Res. 1999, 59: 5671-5673.

    CAS  PubMed  Google Scholar 

  17. Takahashi N, Okumura T, Motomura W, Fujimoto Y, Kawabata I, Kohgo Y: Activation of PPARgamma inhibits cell growth and induces apoptosis in human gastric cancer cells. FEBS Lett. 1999, 455: 135-139. 10.1016/S0014-5793(99)00871-6.

    Article  CAS  PubMed  Google Scholar 

  18. Tanaka T, Kohno H, Yoshitani S, Takashima S, Okumura A, Murakami A, Hosokawa M: Ligands for peroxisome proliferator-activated receptors a and g inhibit chemically induced colitis and formation of aberrant crypt foci in rats. Cancer Res. 2001, 61: 2424-2428.

    CAS  PubMed  Google Scholar 

  19. Osawa E, Nakajima A, Wada K, Ishimine S, Fujisawa N, Kawamori T, Matsuhashi N, Kadowaki T, Ochiai M, Sekihara H, Nakagama H: Peroxisome proliferator-activated receptor g ligands suppress colon carcinogenesis induced by azoxymethane in mice. Gastroenterology. 2003, 124: 361-367. 10.1053/gast.2003.50067.

    Article  CAS  PubMed  Google Scholar 

  20. Niho N, Takahashi M, Kitamura T, Shoji Y, Itoh M, Noda T, Sugimura T, Wakabayashi K: Concomitant suppression of hyperlipidemia and intestinal polyp formation in Apc-deficient mice by peroxisome proliferator-activated receptor ligands. Cancer Res. 2003, 63: 6090-6095.

    CAS  PubMed  Google Scholar 

  21. Girnun GD, Smith WM, Drori S, Sarraf P, Mueller E, Eng C, Nambiar P, Rosenberg DW, Bronson RT, Edelmann W, Kucherlapati R, Gonzalez FJ, Spiegelman BM: APC-dependent suppression of colon carcinogenesis by PPARg. Proc Natl Acad Sci USA. 2002, 99: 13771-13776. 10.1073/pnas.162480299.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lefebvre AM, Chen I, Desreumaux P, Najib J, Fruchart JC, Geboes K, Briggs M, Heyman R, Auwerx J: Activation of the peroxisome proliferator-activated receptor g promotes the development of colon tumors in C57BL/6J-APCMin/+ mice. Nat Med. 1998, 4: 1053-1057. 10.1038/2036.

    Article  CAS  PubMed  Google Scholar 

  23. Saez E, Tontonoz P, Nelson MC, Alvarez JGA, Ming UT, Baird SM, Thomazy VA, Evans RM: Activators of the nuclear receptor PPARg enhance colon polyp formation. Nat Med. 1998, 4: 1058-1061. 10.1038/2042.

    Article  CAS  PubMed  Google Scholar 

  24. Okayasu I, Hatakeyama S, Yamada M, Ohkusa T, Inagaki Y, Nakaya R: Novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mice. Gastroenterology. 1990, 98: 694-702.

    CAS  PubMed  Google Scholar 

  25. Seril DN, Liao J, Ho KLK, Warsi A, Yang CY, Yang GY: Dietary iron supplementation enhances DSS-induced colitis and associated colorectal carcinoma development in mice. Dig Dis Sci. 2002, 47: 1266-1278. 10.1023/A:1015362228659.

    Article  CAS  PubMed  Google Scholar 

  26. Su CG, Wen X, Bailey ST, Jiang W, Rangwala SM, Keilbaugh SA, Flanigan A, Murthy S, Lazar MA, Wu GD: A novel therapy for colitis utilizing PPAR-g ligands to inhibit the epithelial inflammation response. J Clin Invest. 1999, 104: 383-389.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Nakajima A, Wada K, Miki H, Kubota N, Nakajima N, Terauchi Y, Ohnishi S, Saubermann LJ, Kadowaki T, Blumberg RS, Nagai R, Matsuhashi N: Endogenous PPAR g mediates anti-inflammatory activity in murine ischemia-reperfusion injury. Gastroenterology. 2001, 120: 460-469.

    Article  CAS  PubMed  Google Scholar 

  28. 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: 965-973.

    Article  CAS  PubMed  Google Scholar 

  29. Fukutake M, Nakatsugi S, Isoi T, Takahashi M, Ohta T, Mamiya S, Taniguchi Y, Sato H, Fukuda K, Sugimura T, Wakabayashi K: Suppressive effects of nimesulide, a selective inhibitor of cyclooxygenase-2, on azoxymethane-induced colon carcinogenesis in mice. Carcinogenesis. 1998, 19: 1939-1942. 10.1093/carcin/19.11.1939.

    Article  CAS  PubMed  Google Scholar 

  30. Riddell RH, Goldman H, Ransohof DF, Appleman HD, Fenoglio CM, Haggitt RC, Ahren C, Correa P, Hamilton SR, Morson BC, Sommers SC, Yardley JH: Dysplasia in inflammatory bowel disease: standardized classification with provisional clinical application. Human Pathol. 1983, 14: 931-968.

    Article  CAS  Google Scholar 

  31. Pascal RR: Dysplasia and early carcinoma in inflammatory bowel disease and colorectal carcinomas. Human Pathol. 1994, 25: 1160-1171. 10.1016/0046-8177(94)90032-9.

    Article  CAS  Google Scholar 

  32. Ward JM: Morphogenesis of chemically induced neoplasms of the colon and small intestine in rats. Lab Invest. 1974, 30: 505-513.

    CAS  PubMed  Google Scholar 

  33. Watanabe I, Toyoda M, Okuda J, Tenjo T, Tanaka K, Yamamoto T, Kawasaki H, Sugiyama T, Kawarada Y, Tanigawa N: Detection of apoptotic cells in human colorectal cancer by two different in situ methods; antibody against single-stranded DNA and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick and end-labeling (TUNEL) methods. Jpn J Cancer Res. 1999, 90: 188-193.

    Article  CAS  PubMed  Google Scholar 

  34. Tanaka T, Kohno H, Murakami M, Shimada R, Kagami S: Colitis-related rat colon carcinogenesis induced by 1-hydroxyanthraquinone and methylazoxymethanol acetate (Review). Oncol Rep. 2000, 7: 501-508.

    CAS  PubMed  Google Scholar 

  35. Wakabayashi K: NSAIDs as cancer preventive agents. Asian Pacific J Cancer Prev. 2000, 1: 97-113.

    Google Scholar 

  36. Tanaka T, Kojima T, Yoshimi N, Sugie S, Mori H: Inhibitory effect of the non-steroidal anti-inflammatory drug, indomethacin on the naturally occurring carcinogen, 1-hydroxyanthraquinone in male ACI/N rats. Carcinogenesis. 1991, 12: 1949-1952.

    Article  CAS  PubMed  Google Scholar 

  37. Kawamori T, Rao CV, Seibert K, Reddy BS: Chemopreventive activity of celecoxib, a specific cyclooxygenase-2 inhibitor, against colon carcinogenesis. Cancer Res. 1998, 58: 409-412.

    CAS  PubMed  Google Scholar 

  38. Kohno H, Yoshitani S, Takashima S, Okumura A, Hosokawa M, Yamaguchi N, Tanaka T: Troglitazone, a ligands for peroxisome proliferator-activated receptor g inhibits chemically-induced aberrant crypt foci in rats. Jpn J Cancer Res. 2001, 92: 396-403.

    Article  CAS  PubMed  Google Scholar 

  39. Maeda H, Akaike T: Review: Nitric oxide and oxygen radicals in infection, inflammation, and cancer. Biochemistry (Mosc). 1998, 63: 854-865.

    CAS  Google Scholar 

  40. Lehmann JM, Lenhard JM, Oliver BB, Ringold GM, Kliewer SA: Peroxisome proliferator-activated receptors a and g are activated by indomethacin and other non-steroidal anti-inflammatory drugs. J Biol Chem. 1997, 272: 3406-3410. 10.1074/jbc.272.6.3406.

    Article  CAS  PubMed  Google Scholar 

  41. Vamecq J, Latruffe N: Medical significance of peroxisome proliferator-activated receptor. Lancet. 1999, 354: 141-148. 10.1016/S0140-6736(98)10364-1.

    Article  CAS  PubMed  Google Scholar 

  42. Gelman L, Fruchart JC, Auwerx J: An update on the mechanisms of action of the peroxisome proliferator-activated receptors (PPARs) and their roles in inflammation and cancer. Cell Mol Life Sci. 1999, 55: 932-943. 10.1007/s000180050345.

    Article  CAS  PubMed  Google Scholar 

  43. Singer II, Kawka DW, Scott S, Weidner JR, Mumford RA, Riehl TE, Stenson WF: Expression of inducible nitric oxide synthase and nitrotyrosine in colonic epithelium in inflammatory bowel disease. Gastroenterology. 1996, 111: 871-885.

    Article  CAS  PubMed  Google Scholar 

  44. Ambs S, Merriam WG, Bennett WP, Felley-Bosco E, Ogunfusika MO, Oser SM, Klein S, Shields PG, Billiar TR, Harris CC: Frequent nitric oxide synthase-2 expression in human colon adenomas: implication for tumor angiogenesis and colon cancer progression. Cancer Res. 1998, 58: 334-341.

    CAS  PubMed  Google Scholar 

  45. Devchand PR, Keller H, Peters JM, Vazquez M, Gonzalez FJ, Wahli W: The PPARa-leukotriene B4 pathway to inflammatory control. Nature. 1996, 384: 39-43. 10.1038/384039a0.

    Article  CAS  PubMed  Google Scholar 

  46. Jiang C, Ting AT, Seed B: PPAR-g agonists inhibit production of monocyte inflammatory cytokines. Nature. 1998, 391: 82-86. 10.1038/35154.

    Article  CAS  PubMed  Google Scholar 

  47. Colville-Nash PR, Qureshi SS, Willis D, Willoughby DA: Inhibition of inducible nitric oxide synthase by peroxisome proliferator-activated receptor agonists: correlation with induction of heme oxygenase 1. J Immunol. 1998, 161: 978-984.

    CAS  PubMed  Google Scholar 

  48. Rao CV, Kawamori T, Hamid R, Reddy BS: Chemoprevention of colonic aberrant crypt foci by an inducible nitric oxide synthase-selective inhibitor. Carcinogenesis. 1999, 20: 641-644. 10.1093/carcin/20.4.641.

    Article  CAS  PubMed  Google Scholar 

  49. Zingarelli B, Szabo C, Salzman AL: Reduced oxidative and nitrosative damage in murine experimental colitis in the absence of inducible nitric oxide synthase. Gut. 1999, 45: 199-209.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Eiserich JP, Hristova M, Cross CE, Jones AD, Freeman BA, Halliwell B, van der Vliet A: Formation of nitric oxide-derived inflammatory oxidants by myeloperoxidase in neutrophils. Nature. 1998, 391: 393-397. 10.1038/34923.

    Article  CAS  PubMed  Google Scholar 

  51. Cohen SM, Ellwein LB: Cell proliferation in carcinogenesis. Science. 1990, 249: 1007-1011.

    Article  CAS  PubMed  Google Scholar 

  52. Kellett M, Potten CS, Rew DA: A comparison of in vivo cell proliferation measurements in the intestine of mouse and man. Epithelial Cell Biol. 1992, 1: 147-155.

    CAS  PubMed  Google Scholar 

  53. McGarrity TJ, Peiffer LP, Colony PC: Cellular proliferation in proximal and distal rat colon during 1,2-dimethylhydrazine-induced carcinogenesis. Gastroenterology. 1988, 95: 343-348.

    Article  CAS  PubMed  Google Scholar 

  54. Barnes CJ, Cameron IL, Hardman WE, Lee M: Non-steroidol anti-inflammatory drug effect on crypt cell proliferation and apoptosis during initiation of rat colon carcinogenesis. Br J Cancer. 1998, 77: 573-580.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. DuBois RN, Radhika A, Reddy BS, Entingh AJ: Increased cycloooxygenase-2 levels in carcinogen-induced rat colonic tumors. Gastroenterology. 1996, 110: 1259-1262.

    Article  CAS  PubMed  Google Scholar 

  56. 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.

    Article  CAS  PubMed  Google Scholar 

  57. Tsujii M, DuBois RN: Alterations in cellular adhesion and apoptosis in epithelial cells overexpressing prostaglandin endoperoxide synthase 2. Cell. 1995, 83: 493-501. 10.1016/0092-8674(95)90127-2.

    Article  CAS  PubMed  Google Scholar 

  58. Tardieu D, Jaeg JP, Deloly A, Corpet DE, Cadet J, Petit CR: The COX-2 inhibitor nimesulide suppresses superoxide and 8-hydroxy-deoxyguanosine formation, and stimulates apoptosis in mucosa during early colonic inflammation in rats. Carcinogenesis. 2000, 21: 973-976. 10.1093/carcin/21.5.973.

    Article  CAS  PubMed  Google Scholar 

  59. Miller JR, Moon RT: Signal transduction through b-catenin and specification of cell fate during embryogenesis. Genes Dev. 1996, 10: 2527-2539.

    Article  CAS  PubMed  Google Scholar 

  60. Tetsu O, McCormick F: b-Catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature. 1999, 398: 422-426. 10.1038/18884.

    Article  CAS  PubMed  Google Scholar 

  61. Iwao K, Nakamori S, Kameyama M, Imaoka S, Kinoshita M, Fukui T, Ishiguro S, Nakamura Y, Miyoshi Y: Activation of the b-catenin gene by interstitial deletions involving exon 3 in primary colorectal carcinomas without adenomatous polyposis coli mutations. Cancer Res. 1998, 58: 1021-1026.

    CAS  PubMed  Google Scholar 

  62. He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, Morin PJ, Vogelstein B, Kinzler KW: Identification of c-MYC as a target of the APC pathway. Science. 1998, 281: 1509-1512. 10.1126/science.281.5382.1509.

    Article  CAS  PubMed  Google Scholar 

  63. Araki Y, Okamura S, Hussain SP, Nagashima M, He P, Shiseki M, Miura K, Harris CC: Regulation of cyclooxygenase-2 expression by the Wnt and ras pathways. Cancer Res. 2003, 63: 728-734.

    CAS  PubMed  Google Scholar 

  64. Fahimi HD, Maumgart E, Beier K, Pill J, Hartig F, Volkl A: Ultrastructural and biochemical aspects of peroxisome proliferation and biogenesis in different mammalian species. Peroxisomes: biology and importance in toxicology and medicine. Edited by: Gibson G and Lake B. 1993, London, Taylor and Francis Ltd., 395-424.

    Google Scholar 

  65. Reiner M, Massera E, Magni E: Nimesulide in the treatment of fever: a double-blind, crossover clinical trial. J Int Med Res. 1984, 12: 102-107.

    CAS  PubMed  Google Scholar 

  66. Kumar S, Prange A, Schulze J, Lettis S, Barnett AH: Troglitazone, an insulin action enhancer, improves glycaemic control and insulin sensitivity in elderly type 2 diabetic patients. Diabet Med. 1998, 15: 772-779. 10.1002/(SICI)1096-9136(199809)15:9<772::AID-DIA677>3.0.CO;2-X.

    Article  CAS  PubMed  Google Scholar 

  67. Wheeler KA, West RJ, Lloyd JK, Barley J: Double blind trial of bezafibrate in familial hypercholesterolaemia. Arch Dis Child. 1985, 60: 34-37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Pre-publication history

Download references

Acknowledgements

Dr. Rikako Suzuki is a Research Fellow of the Japan Society for the Promotion of Science. This research was supported in part by the Grant-in-Aid (13–15) for Cancer Research from the Ministry of Health, Labour and Welfare of Japan; the Grant-in-Aid for the 3rd Term Comprehensive Control Research from the Ministry of Health, Labour and Welfare of Japan; the Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan; and the grant (C2004-4) for the Collaborative Research from Kanazawa Medical University; and the grant (H2004-6) for the Project Research from the High-Technology Center of Kanazawa Medical University.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Hiroyuki Kohno.

Additional information

Competing interests

The author(s) declare that they have no competing interests.

Authors' contributions

HK participated in study design, performed the animal studies, and drafted the manuscript. RS carried out tissue collection and data analysis. SS participated in the histopathological and immunohistological analysis. TT participated in study design, coordination, and manuscript preparation. All authors read and approved the final manuscript.

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Kohno, H., Suzuki, R., Sugie, S. et al. Suppression of colitis-related mouse colon carcinogenesis by a COX-2 inhibitor and PPAR ligands. BMC Cancer 5, 46 (2005). https://doi.org/10.1186/1471-2407-5-46

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1471-2407-5-46

Keywords