Figure 3 - uploaded by Lixin Ma
Content may be subject to copyright.
Appearance of mucinous adenocarcinoma in situ ; outlined (A) and MRI axial images (B) and consecutive micro-MRI coronal images (C) of a male mouse at 16 weeks PI, demonstrating the corresponding lesion (arrows, B, C) and background signal mimicking lesions (arrowhead, B). b indicates bladder. 

Appearance of mucinous adenocarcinoma in situ ; outlined (A) and MRI axial images (B) and consecutive micro-MRI coronal images (C) of a male mouse at 16 weeks PI, demonstrating the corresponding lesion (arrows, B, C) and background signal mimicking lesions (arrowhead, B). b indicates bladder. 

Source publication
Article
Full-text available
Helicobacter bilis-infected Smad3(-/-) mice represent an attractive model of inflammation-associated colon cancer. Most infected mice develop mucinous adenocarcinoma (MUC) by 6 weeks post inoculation (PI); however, approximately one third do not progress to MUC. The ability to predict the development of MUC in mice used in therapeutic studies would...

Contexts in source publication

Context 1
... of differences in normalized expression levels between H. bilis – infected ( n = 7) and sham-infected ( n = 9) Smad3 − / − mice at 9 weeks PI was determined using the Mann-Whitney rank sum test. The significance of differences between H. bilis – infected MUC+ ( n = 11), H. bilis – infected MUC − ( n = 3), and sham-infected mice ( n = 9) at 1 to 7 weeks PI was determined using the Kruskal-Wallis one-way analysis of variance on ranks and Dunn ’ s method of multiple pairwise comparisons. Coli were evaluated histologically and ranked according to lesion severity. Rank order of lesion severity in H. bilis – infected Smad3 − / − mice was correlated to the rank order of normalized expression of each biomarker using Spearman rank order correlation (SROC). On the basis of the presence or absence of CRC on histologic interpretation, receiver-operator characteristic (ROC) curves were generated from H. bilis – infected ( n = 14) Smad3 − / − mice using the statistical software package SigmaPlot (SPSS, Chicago, IL). R software [22] was used to compute the con- fidence intervals (CIs) for the area under the curve (AUC) based on DeLong ’ s method [26] as well as to compare ROC curves to each other based on the Hanley and McNeil method [27]. For CIs for the AUC, upper limits were truncated at unity. Smad3 mice on a 129/Sv background develop colonic neoplasia, but this phenotype is dependent on infection with either H. bilis or H. hepaticus , with tumors developing most often in the proximal colon [11,25]. In the present study, the tumors were typically single masses, although less than 10% developed a second mass at other sites in the colon. Grossly, tumors appeared either as a thickened pale area of the proximal colon or as pearlescent, lobulated, exophytic masses reflecting the abundant mucin production seen in most masses (Figure 1 A ). Histologically, these tumors were best classified as MUC, with an appearance similar to that seen in humans. Tumors were characterized by marked goblet and epithelial cell hyperplasia with extensive production of mucus, often seen sequestered in large dilated “ mucin lakes, ” spilling into the lumen of the gastrointestinal (GI) tract or penetrating the serosal surface and spilling into the peritoneal space. In the latter case, peritonitis was not uncommon. Many of the neoplastic epithelial cells retained a simple tall columnar morphology with centrally located, oblong nuclei containing a vesicular chromatin pattern. There were also abundant goblet cells, often approaching a 1:1 ratio with columnar epithelial cells. Surrounding the accumulations of mucinous material, the epithelium was variably attenuated, and sloughed epithelial and inflammatory cells were seen in the mucinous material, which was characterized by a lightly basophilic, homogenous to lacy appearance (Figure 1, B-D ). There were also mild to moderate mixed inflammatory infiltrates in the areas around the tumor. Mitotic figures ranged from 0 to 4, with an average of 1 per high-power field (×400). In the absence of Helicobacter infection, Smad3 − / − mice did not develop MUC or any detectable intestinal inflammation. To determine whether the incidence of MUC would increase with increasing duration of infection, Smad3 − / − mice orally infected with H. bilis were killed at multiple time points after infection, and the GI tract was collected for histologic examination. Tissues were nominally classified as being neoplastic (possessing a characteristic MUC lesion), hyperplastic (showing evidence of focal or diffuse epithelial hyperplasia but not neoplasia), or lesion-free. Whereas 100% of mice (5/5) examined at 9 weeks PI had developed characteristic MUC lesions, only 40% of mice (2/5) at 11 weeks PI and 88% of mice (7/8) at 13 weeks PI showed histologic evidence of neoplasia (Figure 2). This was not a function of the early time point because similar results were obtained with mice at 20 weeks PI (data not shown). Thus, whereas most mice developed identifiable MUC by as early as 6 weeks PI, not all mice progressed to MUC regard- less of the duration of infection. H. bilis – infected Smad3 mice ( n = 12, 6 males and 6 females) and naive wild-type mice of the same background strain (1 male and 1 female) were imaged using MRI without contrast at 3, 5, 8, 10, 12, 14, and 16 weeks PI. Immediately after the final imaging, mice were killed and carefully dissected and photographed without disturbing the po- sition of abdominal contents in situ . Gross findings were then compared with the 16-week PI images to assess the capacity of MRI to detect intestinal lesions and to evaluate the level of background signal in control mice. The earliest time point at which Helicobacter -infected mice were interpreted as possessing a neoplastic lesion was 8 weeks PI. Although we were able to identify a reasonable agreement between the final (16 weeks PI) images and gross necropsy findings in 7 of 12 H. bilis – infected mice (sensitivity = 58.33%; Figure 3), 5 of 12 mice with histologically identifiable MUC were interpreted as MUC-negative at 16 weeks PI. At 9 weeks PI, H. bilis – infected Smad3 mice expressed significantly higher levels of IL-1 β ( P = .001, Mann-Whitney rank sum test), MIP-1 α ( P = .004), and RANTES ( P = .003; Figure 4, A-C ). However, no difference was detected in the expression of MCP-2 despite a trend toward an elevated expression in H. bilis – infected mice (Figure 4 D ). Two sham- inoculated mice expressed levels of MCP-2 comparable to even the highest-expressing H. bilis – infected mice; thus, MCP-2 was eliminated from further experiments. In this cohort of mice, all seven Helicobacter infected mice developed histologically identifiable MUC. SROC was then performed to correlate the expression of IL-1 β , MIP-1 α , and RANTES with lesion severity in the H. bilis – infected mice. Correlation coefficients for IL-1 β , MIP-1 α , and RANTES were 0.929 ( P < .001), 0.536 ( P = .18), and 0.750 ( P = .04), respectively, indicating a significant correlation between lesion scores and expression of both IL-1 β and RANTES. On the basis of the significant overall difference in expression between infected and control mice in MIP-1 α expression, along with the fact that the two mice with the lowest MIP-1 α mRNA levels also demonstrated the lowest lesion scores, we opted to retain MIP-1 α in subsequent studies. In addition, the kinetics of chemokine and cytokine expression vary, and we reasoned that, at earlier time points, MIP-1 α might still prove to be a useful biomarker, despite poor overall correlation at 9 weeks PI. To determine whether fecal mRNA levels of IL-1 β , MIP-1 α , and RANTES at time points earlier than 9 weeks PI could predict subsequent disease occurrence or severity, mice were inoculated as before with H. bilis ( n = 14) or sterile broth ( n = 9), and fecal samples were collected every 2 weeks starting at 1 week PI and continuing until 7 weeks PI. Mice were killed at 9 weeks PI, and tissues were collected for histologic examination. The normalized expression of each biomarker was determined at each time point, revealing similar kinetics for all three biomarkers (Figure 5). The expression of all three biomarkers peaked at 1 week PI and then steadily declined thereafter in Helicobacter infected mice. Nonetheless, even at 7 weeks PI, there was a significant difference (Mann-Whitney rank sum test, P < .05) between H. bilis – infected MUC+ and sham-infected mice for all three biomarkers. To assess the value of each biomarker and determine the optimal screening paradigm, the expression at each time point was correlated to lesion rank at 9 weeks PI using SROC. In addition, ROC curves were generated to establish sensitivity, specificity, and appropriate cutoff values for each biomarker. SROC analysis of samples from 1 to 7 weeks PI revealed a significant correlation ( P < .05) between lesion severity at 9 weeks PI and expression of IL-1 β at 1, 3, and 5 weeks PI, of MIP-1 α at 3 and 5 weeks PI, and of RANTES at 3 weeks PI, indicating that 3 weeks PI may be the optimal time for testing mice. Surprisingly, the correlation between lesion rank and the expression of all three biomarkers was not statistically significant at 7 weeks PI. However, SROC indicates the overall agreement between lesions and the selected biomarkers across the entire range of lesion severity. Because our goal was to identify “ poor responders ” and eliminate mice at the low end of the disease spectrum, ROC curves at each time point were compared with determine whether very high specificity (>0.98) and acceptable sensitivity (>0.80) could be achieved simultaneously. Considering the poor correlation for all markers at 7 weeks PI, empirical ROC curves were produced for only 1, 3, and 5 weeks PI (Figure 6). CIs were established using the method of DeLong et al. [26], and the upper limit was truncated at 1 because AUC values, by definition, cannot exceed unity. All three biomarkers examined provided an estimated AUC of 0.97 at 3 weeks PI (Figure 6, D - F ; 95% CI = 0.89-1). At 5 weeks PI, however, fecal mRNA levels of IL-1 β yielded an estimated AUC of 1, thus providing, in this sample, perfect sensitivity and specificity in predicting the presence or absence of MUC in mice at 9 weeks PI (Figure 6 G ). However, ranking the relative performance of IL-1 β at 5 weeks PI and IL-1 β , MIP-1 α , or RANTES at 3 weeks PI is difficult because the small sample size gives limited power to discern differences in AUC; not surprisingly, none of the markers shown in Figure 6 were statistically different from each other (smallest P = .1), using the method of Hanley and McNeil [27]. Because the goal of this study was to establish a screening method with the ability to predict which mice would not develop CRC as a means of conserving resources, it should be noted that by as early as 1 week PI, IL-1 β provided an AUC of 0.97 (95% CI = 0.89-1), the same as all three biomarkers at ...
Context 2
... lack of robust sensitivity and specificity, and although colonoscopy is possible in mice [8,9], it is both time- and cost- intensive and requires anesthesia and substantial expertise. Helicobacter -infected Smad3 knockout mice represent an attractive animal model for the study of CRC. Mice deficient in Smad3, a transcription factor downstream of the anti-inflammatory and pro- apoptotic cytokine transforming growth factor β , develop only a few mild phenotypic abnormalities including megaesophagus and, at a very low incidence, angular limb deformities [10] when raised in specific pathogen-free conditions. However, when infected with enterohepatic species of Helicobacter , e.g., Helicobacter bilis or Helicobacter hepaticus [11], approximately two-thirds of these mice develop mucinous adenocarcinoma (MUC) of the proximal colon by as early as 6 weeks post inoculation (PI). H. bilis and H. hepaticus induce intestinal inflammation in susceptible strains of mice [12,13], and the neoplasia seen in Helicobacter infected Smad3 − / − mice is considered a postinflammatory phenomenon [11]. In addition, loss of normal transforming growth factor β signaling is widely recognized as an indicator of malignancy in human CRC [14,15]. Thus, the targeted deletion of Smad3 in mice is a biologically relevant model of the human condition. This model is ideal for therapeutic studies of CRC in that most mice develop disease on a predicted time course. Moreover, cancer develops in its natural setting as opposed to the more commonly used models that use flank injections of immunocompromised mice with human cancer cell lines. To enhance the usefulness of this model, it would be ideal to be able to identify CRC at early stages of disease or even before disease onset. This would allow for both the elimination of mice from expensive therapeutic studies as well as the assessment of therapeutics at various stages of disease. For the purposes of evaluating therapeutic compounds for the treatment of CRC, we endeavored to establish a method of screening H. bilis – infected Smad3 − / − mice before the time at which disease typically occurs. Our goal was to refine this mouse model of CRC in an effort to reduce animal numbers and the associated costs and to increase the power of the data generated in these trials. Toward these ends, we evaluated both magnetic resonance imaging (MRI) and several fecal RNA biomarkers as means of detecting disease and predicting disease severity in H. bilis – and sham-inoculated Smad3 − / − mice in two separate, sequential longitudinal studies using different cohorts of mice. MRI was selected, as opposed to CT, based on the greater soft tissue definition afforded by this modality. Non – contrast-enhanced MRI was able to detect MUC lesions beginning at 8 weeks PI, and 58% of mice with histologically confirmed lesions were correctly identified at 16 weeks PI. However, serial images produced inconsistent results. In addition, MRI requires considerable resources to perform, and image interpretation is inherently subjective and requires expertise. Alternatively, fecal expression of messenger RNA (mRNA) specific for interleukin 1 β (IL-1 β ), macrophage inflammatory protein 1 α (MIP-1 α ), and regulated on activation, normal T-cell expressed, and secreted (RANTES) at 3 weeks PI correlated significantly with MUC lesion severity at 9 weeks PI in H. bilis – infected Smad3 − / − mice, allowing the identification of which mice have a high probability of developing MUC. A H. bilis isolate was obtained from an endemically infected mouse colony using a previously described culture technique [16]. The isolate was identified as H. bilis based on ultrastructural morphology, bio- chemical characteristics, and sequence analysis of the 16S ribosomal RNA gene [17]. For inoculation, H. bilis cultures were grown in 5 ml of Brucella broth (Becton Dickinson, Franklin Lakes, NJ) sup- plemented with 5% fetal calf serum (Sigma-Aldrich Co, St Louis, MO) and overlaid on blood agar plates and incubated for 24 to 48 hours at 37°C in a microaerobic environment containing 90% N 2 , 5% H , and 5% CO . All studies were performed in accordance with the Guide for the Care and Use of Laboratory Animals and were approved by the University of Missouri Institutional Animal Care and Use Committee. 129- Smad 3tm/Par /J (referred to as Smad3 − / − ) mice were bred on site for these studies. Mice were confirmed to be free of adventitious viruses, parasites, and pathogenic enteric and respiratory bacteria, including all known murine Helicobacter spp. Three- to four-week-old Smad3 − / − mice were inoculated twice, 24 hours apart, with 10 8 H. bilis organisms in 0.5 ml of Brucella broth, or an equivalent volume of sterile broth, through gastric gavage. Separate cohorts of mice were used for each study including incidence of CRC ( n = 5-9 mice per time point; Figures 1 and 2), MRI ( n = 6 of each sex infected with H. bilis and n = 1 of each sex sham-infected; Figure 3), 9 weeks PI fecal mRNA levels ( n = 7 H. bilis – infected and 9 sham-infected; Figure 4), and 1 to 7 weeks PI fecal mRNA levels ( n = 14 of H. bilis – infected and n = 9 of sham-infected; Figures 5 and 6). Mice were group-housed according to infection status in autoclaved microisolator cages and were provided autoclaved food and water. All manipulations and sample collections were performed in a biosafety hood except for MRI. Mice were killed at 16 or 9 weeks PI for the MRI and fecal biomarker studies, respectively, through inhaled over- dose of CO . MRI was performed at 3, 5, 8, 10, 12, 14, and 16 weeks PI using a 7 T/210 mm Varian Unity Inova MRI system equipped with a quadrature-driven birdcage coil (38-mm ID; Varian, Inc, Palo Alto, CA). Mice were anesthetized with 1% to 2% isoflurane in oxygen through a nose cone. A respiratory sensor was placed on the abdomen for monitoring of vital signs; body temperature was supported with warm air circulating in the magnet bore (SA Instruments, Inc, Stony Brook, NY). Coronal and axial planes were collected using a spin-echo T 1 -weighted imaging sequence with a fat saturation pulse applied to suppress the strong signals from fatty tissues. Typically, images were collected with 21 slices, 0.8-mm slice thickness, pixel resolution of 59 mm × 127 mm (coronal planes) and 59 mm × 59 mm (axial planes), and four scans to average the motion artifacts. Images were processed using VnmrJ (Varian, Inc, CA). For collection of fecal samples, mice were individually placed in autoclaved cages containing no bedding within a biosafety hood. Fecal pellets were collected at 1, 3, 5, 7, and 9 weeks PI with a sterile tuber- culin syringe and placed in 200 μ l of RNAlater (Ambion, Austin, TX) for isolation of RNA. Pellets in RNAlater were homogenized with a TissueLyser (Qiagen, Inc, Valencia, CA), centrifuged briefly (Marathon 16 km; Fisher Scientific, Pittsburgh, PA), and then vortexed to re- suspend fecal material. After euthanasia, the intestinal tract from ileum to rectum was collected and fixed in formalin. Formalin-fixed tissues from H. bilis – and sham-infected mice were embedded in paraffin, cut in 5- μ m-thick sections, and processed for staining with hematoxylin and eosin. CRC lesions in H. bilis – infected mice were ranked in a blinded fashion by two laboratory animal pathologists (A.E. and C.F.) according to lesion severity, based on the number of lesions, the longitudinal and vertical extent of neoplastic lesions, and the degree of associated inflammation. Rankings were compared, and in the case of discrepancies in ranking, pathologists con- ferred and agreed on a rank order. Total RNA was extracted using the RNeasy Mini Kit respectively, according to the manufacturer ’ s protocols (Qiagen). RNA was quantified by measuring the absorbance at 260 and 280 nm using a Nanodrop- 1000 spectrophotometer (Nanodrop, Wilmington, DE). Five micrograms of total RNA was reverse-transcribed using reverse transcriptase and oligo(dT) primers according to the manufacturer ’ s protocol (SuperScript First-Strand; Invitrogen, Carlsbad, CA). cDNA was diluted 1:1 with diethylpyrocarbonate-treated water. Semiquantitative real-time reverse transcription – polymerase chain reaction (RT-PCR) was used to measure mRNA levels in feces (LightCycler 1.5; Roche Diagnostic, Nutley, NJ). PCRs and melting curves were performed in a 20- μ l volume in glass capillaries containing 0.5 μ M of each primer, 3 mM MgCl 2 , QuantiTect SYBR Green PCR Master Mix (Qiagen), and cDNA. To quantify the number of copies of specific cDNA, a standard curve was created using known concentrations (10 1 to 10 6 copies) of the pCR4-TOPO (Invitrogen) plasmid containing the transcript of interest. PCRs were incubated at 95°C for 15 minutes to activate the polymerase followed by 40 cycles consisting of a 15-second denaturing at 94°C, 20-second annealing (see Table 1 for primer-specific annealing temperature), and a 30-second extension at 72°C. The ramp rate was 3°C/sec for annealing and 20°C/sec for all other steps. Fluorescence was monitored at the end of each extension phase. After amplification, melting curves were generated to con- firm PCR product identity. The sequences for hypoxanthine guanine phosphoribosyltransferase (HPRT) [18], IL-1 β [19], monocyte chemotactic protein 2 (MCP-2) [20], and MIP-1 α [21] have been previously reported in the literature. The primer sequence for RANTES was designed from published mRNA sequences using DS Gene software (Accelrys, San Diego, CA). Standards were generated using linearized plasmids containing cloned amplicons of selected targets using the Topo TA PCR cloning kit (Invitrogen). Transcripts were quantified by comparing fluorescence of experimental samples to that of plasmid standards containing known concentrations of the cloned product. Semiquantitative real-time RT-PCR was used to measure mRNA levels in feces. The expressions of IL-1 β , MIP-1 α , MCP-2, and RANTES were ...
Context 3
... recapitulate the human condition faithfully, with extensive mucin production seen multifocally in neoplastic foci, forming dilated pockets of mucinous material ex- tending into the lumen and frequently through the tunica muscularis to the serosal surface of the GI tract. There are several reasons to believe that the mechanisms leading to MUC in humans and Helicobacter infected Smad3 − / − mice are similar. In humans, as in Helicobacter -infected Smad3 − / − mice, MUC occurs more frequently in the proximal colon than elsewhere in the GI tract [30 – 32]. Although not all studies agree, possibly due to geographical differences [33] or the presence of two subtypes of colorectal MUC [30], MUC in humans seems to be more prevalent as a sequela to IBD than as a spontaneous CRC not associated with previous IBD [32,34 – 37]. Supporting this concept, MUC occurs primarily in areas of chronic inflammation, and the risk of MUC increases with duration of IBD [38,39]. Similarly, the findings detailed herein support the notion that the severity of MUC in Helicobacter infected Smad3 − / − mice is largely dependent on the robustness of the inflammatory response to a member of the gut flora, as measured by the expression of certain fecal cytokines and chemokines. In addition, MUC in humans is frequently associated with fistula formation [32,35,40 – 43], a phenomenon attributed to adenomatous transformation of the epithelium lining the fistula tract [44]. Many H. bilis – infected Smad3 − / − mice showed histologic evidence of perforation of the bowel wall and areas in which dysplastic epithelial cells are seen invading and penetrating the serosal surface (Figure 1, B-D ), and it is tempting to speculate that a similar mechanism is at work in the formation of MUC lesions in Smad3 − / − mice. Regardless of the pathogenesis, not all Smad3 − / − mice will develop MUC despite persistent colonization with H. bilis . As a consequence, many of these MUC-resistant mice will be used in expensive therapeutic trials lasting up to 8 months in duration. Along with the time and money spent maintaining mice that will not progress to MUC, one must also consider that these mice are potentially confounding the research by making therapeutic compounds seem falsely efficacious. The ability to noninvasively identify which mice will not progress to cancer would both conserve resources and increase the power of data generated by using only mice with a high probability of developing MUC. We first evaluated MRI as a means of detecting early inflammatory or precancerous lesions in Helicobacter -infected Smad3 − / − mice. Mice were imaged every 2 to 3 weeks until 16 weeks PI, a time by which previous studies (Figure 2) had demonstrated most mice would develop MUC. Although MRI provided some diagnostic information, that is, the presence or absence of a lesion, it afforded little prognostic information regarding the severity or extent of disease at necropsy when images were analyzed retrospectively. This is partially due to the vari- ability between images from week to week. Frequently, MRI would indicate a possible lesion at one time point, followed by images at subsequent time points interpreted as negative. In only 1 of 12 mice did images consistently contain suspect lesions after the initial appearance. In addition, in those mice in which a correlation between 16-week PI MRI images and gross lesions was detected, it was difficult to reliably track the course of intestinal neoplasms retrospectively. The background noise, seen in both experimental and control mice, was considerable and was most problematic in highly glandular tissue such as the repro- ductive tract. In addition, the severity of the lesions could not be predicted based on the size and intensity of suspect lesions on MRI. Mice with hyperintense signal on the final imaging, indicating a large or se- vere lesion, often revealed mild or moderate MUC lesions at necropsy. Conversely, mice with borderline “ positive ” final images often revealed extensive marked MUC or even multiple lesions. MRI has been applied to the human population as a screening method for MUC; however, the method requires insufflation of the colon with air to enhance imaging [1]. The lack of insufflation in our study may partially explain the lack of adequate definition with MRI. Also, the imaging in this study was performed without the use of contrast. MRI studies of the gastrointestinal tract using a fecal-tagging based MRI contrast agent may enhance visualization of MUC lesions [45]. The primary goal of these studies was to evaluate two distinct methods of detecting CRC in a mouse model, noninvasively and as early in the disease process as possible. Because our motivation was to conserve resources, we opted to omit insufflation and contrast in an effort to keep the procedure as simple and cost-effective as possible. In our study, a mass showing hyperintense and heterogeneous signal contents would indicate a MUC lesion (Figure 3, B and C ). However, abdominal motion artifacts and significant signals from feces often cause ambiguities or missed detections. A respiratory-gated T 2 -weighted MRI protocol may be applied to increase the detection accuracy and specificity for MUC because of the brighter signal nature of mucin contents in MUC lesions on T 2 -weighted images, however, with the expense of prolonged imaging time. Lastly, considering the expense of the initial purchase, maintenance, and opera- tion, MRI is a costly technique for screening large numbers of animals. Imaging in both coronal and axial planes resulted in 21 and 42 images, respectively, per mouse at each time point, which, along with the user- dependent nature of image interpretation, made this a time-consuming and subjective technique. Because our impetus for screening animals is to eliminate mice that are not going to develop MUC as a means of saving costs, MRI is problematic. Thus, the inability of MRI to detect disease in a reproducible manner, the associated costs, and the labor- intensive and subjective nature of this method make it less than ideal for our purposes. Next, we elected to determine whether CRC in our model could be predicted through the analysis of fecal cytokine and chemokine mes- sage levels. This concept, although not new in humans [46,47], has not been applied to murine models to the authors ’ knowledge. Because humans and mice both constitutively slough colonic epithelial cells in feces, the RNA isolated from these cells should reflect the state of health or inflammation present in the gut. Inflammation is now recognized as a risk factor and negative prognostic indicator for certain types of neoplasia in humans [48]. Adaptive antitumoral immune responses, particularly those mediated by CD8 + T cells, are considered protective and beneficial in destroying tumor cells, whereas inflammation due to innate immune responses is often associated with a poor prognosis [49]. This concept can be extrapolated to the chemokines responsible for attracting T cells or macrophages; accumulation of lymphocytes due to increased expression of CXCL16 correlates with a favorable outcome in human CRC [50], whereas accumulation of tumor-associated macrophages due to increased levels of CCL2 correlates with poor outcome [51]. Thus, as a means of noninvasively assessing the degree of colonic inflammation in Smad3 − / − mice, we measured the fecal levels of several factors involved in macrophage recruitment, and which have been shown to be elevated in human CRC [49,52,53], including the chemokines MIP-1 α , RANTES, and MCP-2 and IL-1 β , a highly pleiotropic proinflammatory cytokine with effects on virtually all cell types [54]. Our initial fecal biomarker study was performed at 9 weeks PI, when most mice were expected to have developed MUC. It was established that a significant difference in the fecal expression of IL-1 β , MIP-1 α , and RANTES existed between Helicobacter - and sham-infected mice (Figure 4); however, 100% (7/7) of the infected mice in this group developed MUC, making a com- parison of MUC+ and MUC − mice within the infected group impos- sible. Pursuing earlier time points PI (Figure 5) with a second group of mice provided evidence that there is also a significant difference in the expression of these biomarkers between H. bilis – infected MUC+ and MUC − mice, supporting their use as predictors of MUC in this model. It is notable that the expression of IL-1 β , MIP-1 α , and RANTES showed an acute increase after infection with Helicobacter , which waned steadily thereafter. Although these biomarkers are primarily associated with innate immune responses, infection with H. bilis eventually induces an adaptive immune response [55], allowing the innate response to wane accordingly. Like the selected chemokines, IL-1 β is produced by the intestinal epithelium. Because intestinal epithelial cells also express the activating receptor IL-1RI [54], IL-1 β functions in an autocrine and paracrine manner to amplify chemokine expression. Similarly, RANTES has been shown to stimulate production of MIP-1 α by human monocytes [56], suggesting that activation of tumor-associated macrophages may amplify recruitment of additional monocytes and other leukocytes, thought to be the source of harmful reactive oxidative intermediaries. Interestingly, several studies indicate that IL-1 β may have a more direct role in colorectal tumorigenesis. In 2003, Liu et al. [57] demonstrated that IL-1 β upregulates the expression of cyclooxygenase-2 (COX-2), which is overexpressed in 80% to 90% of human CRC [58] and is also found to be elevated in Helicobacter -infected Smad3 sup> − / − mice relative to naive mice [11]. Similarly, Maihofner et al. [59] showed that, in both human CRC-associated neoplastic epithelium and tumor-associated macrophages, COX-2 expression was markedly increased and that increase correlated with increases in IL-1 β . COX-2 functions to facilitate ...

Citations

... Intestinal carcinogenesis is driven by microbial enrichment, chronic inflammation, and intestinal immunomodulation [78,79]. In 2010, Ericsson et al. demonstrated that H. bilis infection in Smad3(-/-) mice leads to the development of colitisassociated mucinous colonic adenocarcinoma, which is strongly correlated with the expression of Il-1b, Mip-1a, and Cd5 (RANTES) [80]. In 2013, Nguyen et al. pointed out that H. bilis is not only an important risk factor for the development of chronic intestinal inflammation in WASPdeficient mice but also for dysplasia and CRC [81]. ...
Article
Full-text available
Gut microbiota coevolve with humans to achieve a symbiotic relationship, which ultimately leads to physiological homeostasis. A variety of diseases can occur once this balance is disrupted. Helicobacter bilis (H. bilis) is an opportunistic pathogen in humans, triggering multiple diseases, including inflammatory bowel disease (IBD). IBD is a chronic immunologically mediated inflammation of the human gastrointestinal tract, and its occurrence is closely related to the gut microbiota. Several studies have demonstrated that H. bilis colonization is associated with IBD, and its mechanism is related to host immunity. However, few studies have investigated these mechanisms of action. Therefore, this article is aimed at reviewing these studies and summarizing the mechanisms of H. bilis-induced IBD from two perspectives: adaptive immunity and innate immunity. Furthermore, this study provides a preliminary discussion on treating H. bilis-related IBD. In addition, we also demonstrated that H. bilis played an important role in promoting the carcinogenesis of IBD and discussed its mechanism.
... Abbreviations: CRC, colorectal cancer; CAFs, cancer-associated fibroblasts; Th, T helper cells; ROS, reactive oxygen species in patients with CRC [65]. Moreover, IL-1β and C-C motif chemokine ligand 3 (CCL3) levels are increased in the feces and associated with lesion severity in Helicobacter bilisinfected mothers against decapentaplegic homolog 3 -/-(SMAD3 -/-) mice, an inflammation-associated colon cancer model [66]. Cytokines, such as IFN-γ, IL-15, and IL-18, inhibit CD8 + cytotoxic T lymphocyte (CTL)-mediated antitumor immune responses [34]. ...
Article
Full-text available
Colorectal cancer (CRC) is a predominant life‐threatening cancer, with liver and peritoneal metastases as the primary causes of death. Intestinal inflammation, a known CRC risk factor, nurtures a local inflammatory environment enriched with tumor cells, endothelial cells, immune cells, cancer‐associated fibroblasts, immunosuppressive cells, and secretory growth factors. The complex interactions of aberrantly expressed cytokines, chemokines, growth factors, and matrix‐remodeling enzymes promote CRC pathogenesis and evoke systemic responses that affect disease outcomes. Mounting evidence suggests that these cytokines and chemokines play a role in the progression of CRC through immunosuppression and modulation of the tumor microenvironment, which is partly achieved by the recruitment of immunosuppressive cells. These cells impart features such as cancer stem cell‐like properties, drug resistance, invasion, and formation of the premetastatic niche in distant organs, promoting metastasis and aggressive CRC growth. A deeper understanding of the cytokine‐ and chemokine‐mediated signaling networks that link tumor progression and metastasis will provide insights into the mechanistic details of disease aggressiveness and facilitate the development of novel therapeutics for CRC. Here, we summarized the current knowledge of cytokine‐ and chemokine‐mediated crosstalk in the inflammatory tumor microenvironment, which drives immunosuppression, resistance to therapeutics, and metastasis during CRC progression. We also outlined the potential of this crosstalk as a novel therapeutic target for CRC. The major cytokine/chemokine pathways involved in cancer immunotherapy are also discussed in this review.
... In 2017 alone, there were an estimated 50,260 deaths [1]. Colitis-associated CRC (CAC) is a devastating sequela of inflammatory bowel diseases (IBD) such as Ulcerative Colitis (UC) and Crohn's Disease (CD), recurrent, chronic inflammatory diseases affecting the colon or any region of the gastrointestinal tract (GIT) respectively [2]. The risk of developing CAC increases with the duration of IBD, reaching twenty percent after thirty years duration [3]. ...
... Although CAC represents less than two percent of cases of CRC, the challenges posed by difficulty of detection and treatment of this subset negatively affect prognosis [4][5][6]. Inflammation, among other risk factors of CRC, such as diet, smoking, and obesity, have a complex relationship with the gut microbiota (GM) [1][2][3]. The gut microbiota is the ecosystem of microorganisms inhabiting the GIT, with a profound influence on immune development and function, nutrient absorption and metabolite generation, and susceptibility to diseases of the gut and peripheral systems [7]. ...
... Mutations affecting the TGF-β pathway, an anti-inflammatory and pro-apoptotic cytokine, are commonly cited as an inciting incident for CRCs [9]. For this reason, Smad3 -/mice, harboring a deletion for the Smad3 signaling molecule downstream of the TGF-β receptor, are often used in combination with pathobiont bacterial species such as Helicobacter bilis and H. hepaticus to study colitis-associated CRC [2,10]. These Helicobacter spp. ...
Article
Full-text available
Colorectal cancer (CRC) risk is influenced by host genetics, sex, and the gut microbiota. Using a genetically susceptible mouse model of CRC induced via inoculation with pathobiont Helicobacter spp. and demonstrating variable tumor incidence, we tested the ability of the Th17-enhancing commensal Candidatus Savagella, more commonly denoted as Segmented Filamentous Bacteria (SFB), to influence the incidence and severity of colitis-associated CRC in male and female mice. To document the composition of the gut microbiota during CRC development and identify taxa associated with disease, fecal samples were collected before and throughout disease development and characterized via 16S rRNA sequencing. While there were no significant SFB-dependent effects on disease incidence or severity, SFB was found to exert a sex-dependent protective effect in male mice. Furthermore, SFB stabilized the GM against Helicobacter-induced changes post-inoculation, resulting in a shift in disease association from Helicobacter spp. to Escherichia coli. These data support sex-dependent SFB-mediated effects on CRC risk, and highlight the complex community dynamics within the GM during exposure to inflammatory pathobionts.
... Quantitative PCR was used to determine MNV copy number present in the MLN, liver, and spleen of infected mice, as already described above. Previously published primer sequences were used for Il6, 23 Il1b, 34 Hprt, 34 Il10, 35 Tnfa, 23 Ifnb, 36 Vegfc, 37 and Vegfd. 38 Il7 primer sequences were from Primer Bank. ...
Article
Full-text available
Murine norovirus (MNV) is an RNA virus that can prove lethal in mice with impaired innate immunity. We found that MNV-4 infection of Stat1-/- mice was not lethal, but produced a 100% penetrant, previously undescribed lymphatic phenotype characterized by chronic-active lymphangitis with hepatitis, splenitis, and chronic cecal and colonic inflammation. Lesion pathogenesis progressed from early ileal enteritis and regional dilated lymphatics to lymphangitis, granulomatous changes in the liver and spleen, and ultimately, typhlocolitis. Lesion development was neither affected by antibiotics nor reproduced by infection with another enteric RNA virus, rotavirus. MNV-4 infection in Stat1-/- mice decreased expression of Vegfr3, Vegf-c, and Vegf-d and increased Ifnγ, Tnfα, and iNos. However, anti-IFNγ and anti-TNFα antibody treatment did not attenuate the histologic lesions. Studies in Ifnαβγr-/- mice suggested that canonical signaling via interferon receptors did not cause MNV-4-induced disease. Infected Stat1-/- mice had increased STAT3 phosphorylation and expressed many STAT3-regulated genes, consistent with our findings of increased myeloid cell subsets and serum G-CSF, which are also associated with increased STAT3 activity. In conclusion, in Stat1-/- mice, MNV-4 induces lymphatic lesions similar to those seen in Crohn's disease as well as hepatitis, splenitis, and typhlocolitis. MNV-4-infected Stat1-/- mice may be a useful model to study mechanistic associations between viral infections, lymphatic dysfunction, and intestinal inflammation in a genetically susceptible host.
... However, neither C. jejuni nor its CDT proteins are associated with malignancies in the gastrointestinal tract (Brauner et al., 2010). Although rare studies reported CDT capable of inducing tumorigenesis, CDT-producing Helicobacter bilis-and H. hepaticus-infected mice developed colon and hepatic cancers, respectively (Ericsson et al., 2010;Fox et al., 2011). These reports revealed a potential correlation between CDT and carcinogenesis (Guerra et al., 2011). ...
Article
Full-text available
Cytolethal distending toxin (CDT), a genotoxin produced by Campylobacter jejuni, is composed of three subunits: CdtA, CdtB, and CdtC. CdtB is a DNase that causes DNA double-strand breaks (DSB) in the nucleus resulting in cell cycle arrest at the G2/M stage and apoptosis. CdtA and CdtC bind to cholesterol-rich microdomains on the cytoplasmic membrane, a process required for the delivery of CdtB to cells. Although a unique motif associated with cholesterol-binding activity has been identified in other pathogens, the mechanism underlying the interaction between the CdtA and CdtC subunits and membrane cholesterol remains unclear. Also, the processes of cell uptake and delivery of CdtB in host cells and the translocation of CdtB into the nucleus are only partially understood. In this review, we focus on the underlying relationship among CDT, membrane cholesterol, and the intracellular trafficking pathway as a unique mechanism for C. jejuni-induced pathogenesis. Moreover, we discuss the clinical aspects of a possible therapeutic application of CDT in cancer therapy. Understanding the molecular mechanism of CDT-host interactions may provide insights into novel strategies to control C. jejuni infection and the development of potential clinical applications of CDT.
... In such individuals, increased levels of Foxp3 and reduced intestinal inflammation have often been observed [256,257]. Experimental colitis leading to cancer has also been induced using H. bilis and H. hepaticus in mice 4-6 weeks post-infection, and can contribute to the development of chronic intestinal inflammation in mice [258,259]. Helicobacter hepaticus is often isolated in the livers and colons of infected mice [260], and induces hepatitis, enteritis, typhlocolitis, and IBD-like tissue injury in many genetically modified mouse models [64,261]. Helicobacter hepaticus-induced enteric inflammation has been observed in A/JCr, BALB/cAnNCr, SJL/NCr, C3H/HeNCr, Rag −/− , IL10 −/− , and SCID mice reconstituted with CD45RB high T-cells [262]. ...
Article
Full-text available
Acute and chronic inflammatory diseases of the intestine impart a significant and negative impact on the health and well-being of human and non-human mammalian animals. Understanding the underlying mechanisms of inflammatory disease is mandatory to develop effective treatment and prevention strategies. As inflammatory disease etiologies are multifactorial, the use of appropriate animal models and associated metrics of disease are essential. In this regard, animal models used alone or in combination to study acute and chronic inflammatory disease of the mammalian intestine paired with commonly used inflammation-inducing agents are reviewed. This includes both chemical and biological incitants of inflammation, and both non-mammalian (i.e. nematodes, insects, and fish) and mammalian (i.e. rodents, rabbits, pigs, ruminants, dogs, and non-human primates) models of intestinal inflammation including germ-free, gnotobiotic, as well as surgical, and genetically modified animals. Importantly, chemical and biological incitants induce inflammation via a multitude of mechanisms, and intestinal inflammation and injury can vary greatly according to the incitant and animal model used, allowing studies to ascertain both long-term and short-term effects of inflammation. Thus, researchers and clinicians should be aware of the relative strengths and limitations of the various animal models used to study acute and chronic inflammatory diseases of the mammalian intestine, and the scope and relevance of outcomes achievable based on this knowledge. The ability to induce inflammation to mimic common human diseases is an important factor of a successful animal model, however other mechanisms of disease such as the amount of infective agent to induce disease, invasion mechanisms, and the effect various physiologic changes can have on inducing damage are also important features. In many cases, the use of multiple animal models in combination with both chemical and biological incitants is necessary to answer the specific question being addressed regarding intestinal disease. Some incitants can induce acute responses in certain animal models while others can be used to induce chronic responses; this review aims to illustrate the strengths and weaknesses in each animal model and to guide the choice of an appropriate acute or chronic incitant to facilitate intestinal disease.
... After incubation for 24 h, RNA extraction and gene expression analysis by real-time RT-PCR was performed as previously described. 42 Target gene expression was normalized to the housekeeping gene hypoxanthine-guanine phosphoribosyltransferase 18 and then calculated as the fold change relative to the average value obtained from the no-treatment control. Primers for PCR were: TNFα forward, 5′ CTG AAC TTC GGG GTG ATC GG 3′; TNFα reverse, 5′ GGC TTG TCA CTC GAA TTT TGA GA 3′; and those previously described for IL6, 50 IL1β, 18 inducible nitric oxide synthase (iNOS), 46 MCP1, 28 IFNβ, 79 CD36, 63 and ABCA1. ...
... 42 Target gene expression was normalized to the housekeeping gene hypoxanthine-guanine phosphoribosyltransferase 18 and then calculated as the fold change relative to the average value obtained from the no-treatment control. Primers for PCR were: TNFα forward, 5′ CTG AAC TTC GGG GTG ATC GG 3′; TNFα reverse, 5′ GGC TTG TCA CTC GAA TTT TGA GA 3′; and those previously described for IL6, 50 IL1β, 18 inducible nitric oxide synthase (iNOS), 46 MCP1, 28 IFNβ, 79 CD36, 63 and ABCA1. 63 For western blot analysis, protein was extracted by using MPER Protein Extraction Kit (Thermo Scientific). ...
Article
Full-text available
Macrophages play a key role in the development of atherosclerosis. Murine noroviruses (MNV) are highly prevalent in research mouse colonies and infect macrophages and dendritic cells. Our laboratory found that MNV4 infection in mice lacking the LDL receptor alters the development of atherosclerosis, potentially confounding research outcomes. Therefore, we investigated whether MNV4 likewise altered atherosclerosis in ApoE(-/-) mice. In the presence of oxidized LDL, MNV4 infection of ApoE(-/-) bone marrow-derived macrophages increased the gene expression of the inflammatory markers inducible nitric oxide synthase, monocyte chemoattractant protein 1, and IL6. In addition, proteins involved in cholesterol transport were altered in MNV4-infected ApoE -/- bone marrow-derived macrophages and consisted of increased CD36 and decreased ATP-binding cassette transporter A1. MNV4 infection of ApoE(-/-) mice at 12 wk of age (during the development of atherosclerosis) had a variable effect on atherosclerotic lesion size. In one study, MNV4 significantly increased atherosclerotic plaque area whereas in a second study, no effect was observed. Compared with controls, MNV4-infected mice had higher circulating Ly6C-positive monocytes, and viral RNA was detected in the aortas of some mice, suggesting potential mechanisms by which MNV4 alters disease progression. Plaque size did not differ when ApoE -/- mice were infected at 4 wk of age (early during disease development) or in ApoE -/- mice maintained on a high-fat, high-cholesterol diet. Therefore, these data show that MNV4 has the potential to exert a variable and unpredictable effect on atherosclerosis in ApoE(-/-) mice. We therefore propose that performing experiments in MNV-free mouse colonies is warranted.
... TGFβ is generally accepted to have tumour-suppressive roles in early-stage cancer [34][35][36] , an effect that is consistent with a high frequency of mutations in TGFβ signalling components in CRC 37 . Rapid onset of CRC was recently reported in Helicobacter bilis-infected Smad3 −/− mice, two-thirds of which develop inflammation-associated mucinous carcinomas as early as 6 weeks post-infection 38 . Consistent with a tumour-suppressive role, human CRC cells with high TGFβ sensitivity are weakly metastatic in orthotopic xenograft models 39 . ...
Article
Full-text available
Cytokine networks are crucial aspects of tumour immunology, particularly for colorectal cancer (CRC), in which inflammation and antitumour immunity are key determinants of disease progression. In this Review, we highlight new insights into the functions of well-known cytokines in CRC, describe recently discovered roles for a growing number of novel players, and emphasize the complexity and therapeutic implications of the cytokine milieu. We also discuss how cancer mutations and epigenetic adaptations influence the oncogenic potential of cytokines, a relatively unexplored area that could yield crucial insights into tumour immunology and facilitate the effective application of cytokine-modulatory therapies for CRC.
... Further experimental work, including characterization of cdtB mutants in animal models, is clearly needed though to more completely define the contributions of CdtB and the other proteins encoded in the CdtB-islet, as well as the artAB operon, to pathogenesis of a growing list of non-typhoidal Salmonella serovars recognized as encoding these virulence factors; our mutants are freely available to other researchers for these types of experiments. Given the association of several CdtB-producing bacterial pathogens with cancer [23,24] and the ability of CdtB to cause a DDR in a wide range of eukaryotic cells, it would also be important to explore whether CdtB-producing bacteria can promote cancer development in their respective hosts. As an analysis of previously reported data on distribution of invasive and non-invasive disease among patients infected with different Salmonella serovars provided preliminary support that cdtB positive isolates may be more likely to cause invasive disease, future epidemiological studies are also needed to determine whether cdtB-positive Salmonella isolates are associated with different or more severe disease outcomes in human hosts, in addition to animal studies on the role of CdtB in invasive infection by non-typhoidal Salmonella. ...
Article
Full-text available
For many putative Salmonella enterica subsp. enterica virulence genes, functional characterization across serovars has been limited. Cytolethal distending toxin B (CdtB) is an incompletely characterized virulence factor that is found not only in Salmonella enterica subsp. enterica serovar Typhi (Salmonella Typhi) and dozens of Gram negative bacterial pathogens, but also in non-typhoidal Salmonella (NTS) serovars. A comparative genomics approach was performed to characterize sequence conservation of the typhoid toxin (TT), encoded in the CdtB-islet, between Salmonella Typhi and NTS serovars. The cytotoxic activity of representative Salmonella enterica subsp. enterica serovars Javiana, Montevideo and Schwarzengrund strains and their respective isogenic cdtB mutants was determined in human intestinal epithelial Henle-407 cells by assessment of cell cycle progression of infected cells using fluorescence-activated cell sorting (FACS). Two-way analysis of variance (ANOVA) was used to determine whether cdtB deletion had a significant (p < 0.05) effect on the percentage of Henle-407 cells at each stage of the cell cycle. Here we show that a CdtB-islet encoding the cytolethal distending toxin B (CdtB), pertussis-like toxin A (PltA), and pertussis-like toxin B (PltB) is present in a dozen NTS serovars and that these proteins have a high level of sequence conservation and each form monophyletic clades with corresponding Salmonella Typhi genes. Human epithelial Henle-407 cells infected with three representative CdtB-encoding NTS serovars displayed G2/M phase cell cycle arrest that was absent in cells infected with corresponding isogenic cdtB null mutants (p < 0.0001 for the factor ∆cdtB deletion). Our results show that CdtB encoded by NTS serovars has a genomic organization, amino acid sequence conservation and biological activity similar to the TT, and thus, may contribute to disease pathogenesis.
... Mucosal inflammation may be an independent risk factor for colon cancer [50]. The connection between inflammation and tumorigenesis is well-established and has been reported in genetic, pharmacological, and epidemiological studies in the last decade [51][52][53][54]. Moreover, inflammation is likely involved in other forms of sporadic and heritable colon cancer [52]. ...
Article
Full-text available
Purpose: We evaluated the chemopreventive effect of statins on colon cancer in patients with chronic obstructive pulmonary disease (COPD) and identified the statin exerting the strongest chemopreventive effect. Methods: Using the National Health Insurance Research Database, we identified patients who received a COPD diagnosis in Taiwan between January 1, 2001, and December 31, 2012, and included them in the study cohort. Each patient was followed to assess the colon cancer risk and protective factors. A propensity score was derived using a logistic regression model to estimate the effect of statins by accounting for covariates predicted during the intervention (statins). To examine the dose-response relationship, we categorized statin doses into four groups in each cohort [<28, 28-90, 91-365, and >365 cumulative defined daily dose]. Results: Compared with the statin nonusers, the adjusted hazard ratio (aHR) for colon cancer decreased in the statin users (aHR = 0.52, 95% confidence interval = 0.44, 0.62). Hydrophilic statins exerted a stronger preventive effect against colon cancer. Regarding the statin type, lovastatin, pravastatin, and fluvastatin nonsignificantly reduced the colon cancer risk in the patients with COPD. Compared with the statin nonusers, the aHRs for colon cancer decreased in the individual statin users (rosuvastatin, simvastatin, and atorvastatin: aHRs = 0.28, 0.64, and 0.65, respectively). In the sensitivity analysis, statins dose-dependently reduced the colon cancer risk. Conclusions: Statins dose-dependently exert significant chemopreventive effects on colon cancer in patients with COPD, with rosuvastatin exerting the largest chemopreventive effect.