Skip to main content
Comparative Medicine logoLink to Comparative Medicine
. 2009 Feb;59(1):10–17.

Effects of Helicobacter Infection on Research: The Case for Eradication of Helicobacter from Rodent Research Colonies

Maciej Chichlowski 1, Laura P Hale 1,*
PMCID: PMC2703140  PMID: 19295050

Abstract

Infection of mouse colonies with Helicobacter spp. has become an increasing concern for the research community. Although Helicobacter infection may cause clinical disease, investigators may be unaware that their laboratory mice are infected because the pathology of Helicobacter species is host-dependent and may not be recognized clinically. The effects of Helicobacter infections are not limited to the gastrointestinal system and can affect reproduction, the development of cancers in gastrointestinal organs and remote organs such as the breast, responses to vaccines, and other areas of research. The data we present in this review show clearly that unintentional Helicobacter infection has the potential to significantly interfere with the reliability of research studies based on murine models. Therefore, frequent screening of rodent research colonies for Helicobacter spp. and the eradication of these pathogens should be key goals of the research community.


The reliability of an experiment that uses an in vivo model system depends on understanding and controlling all variables that can influence the experimental outcome. Infections of mouse colonies are important to the scientific community because they can introduce such harmful variables. Therefore, the ultimate goal of laboratory animal facilities is to maintain disease-free animals, to eliminate those unwanted variables.

Numerous pathogenic microbes can interfere with animal research (reviewed in reference 57), and colonization of mouse colonies with members of the family Helicobacteriaceae is an increasing concern for the research community. Naturally acquired Helicobacter infections have been reported in all commonly used laboratory rodent species.3,10,36,44,45,49,82,124 A study of mice derived from 34 commercial and academic institutions in Canada, Europe, Asia, Australia, and the United States showed that 88% of these institutions had mouse colonies infected with 1 or more Helicobacter spp.109 Approximately 59% of these mice were infected with Helicobacter hepaticus ; however monoinfections with other species also were encountered. In another study, at least 1 of 5 Helicobacter spp. was detected in 88% of the 40 mouse strains tested.4

Surveys such as these have established that a broad range of Helicobacter spp. may be present in mouse research colonies. Several of those Helicobacter species cause disease in laboratory mice. H. hepaticus first was identified as a pathogen when it was discovered to be the cause of chronic hepatitis and hepatocellular carcinoma in mice,26,31,116 either alone or in combination with other Helicobacter spp.78 In addition, H. typhlonius causes intestinal inflammation in mice with immunodeficiency or defects in immune regulation;28,37 H. muridarum has been associated with gastritis,86 and H. bilis has been associated with hepatitis35,38 and colitis.60,61 Although, H. rodentium appears to be relatively nonpathogenic in wild-type and SCID mice,78 combined infection with H. rodentium and H. typhlonius results in a high incidence of inflammation-associated neoplasia in IL10−/− mice.9,46 Further, it is becoming increasingly clear that the effects of Helicobacter infections are not limited to the gastrointestinal system. Helicobacter infections have been documented to directly or indirectly affect responses as diverse as reproduction, development of breast cancer, and altered immune responses to vaccines.65,95,99 In addition to effects on rodents, Helicobacter spp. can infect other laboratory animals2,5,27,29,33,36,107 and can colonize different anatomic regions of the gastrointestinal system.35 This review focuses on the potential effect of these organisms on in vivo experiments and biomedical research. The results summarized here emphasize the importance of knowledge of colony infection status and prevention of unintentional infections to achieve the goal of providing a consistent and reliable environment for research studies.

Biologic characteristics of Helicobacter organisms

Helicobacter spp. are gram-negative bacteria that vary in their morphology, growth requirements, biochemical profiles, antibiotic susceptibility, and sequence of conserved 16S rRNA genes.121 Most Helicobacter organisms are long, narrow, slightly curved rods with bipolar sheathed flagella. Detailed data on genus characteristics and methods of detection have been published.3,57,63,88,121 The species that typically infect rodents and their sites of infection are listed in Table 1. Although it does not naturally infect rodents, H. pylori is also included in this list due to its common use in research mouse models. Several Helicobacter spp. (H. pylori, H. hepaticus, H. bilis, H. muridarum) are urease-positive, that is, capable of producing ammonia to neutralize gastric acid, whereas others (for example H. ganmani, H. rodentium, H. trogontum, H. typhlonius, and others) are urease-negative.39,100 Production of urease allows microorganisms to survive in the very acidic gastric environment.96 Interestingly, recent study has shown that H. hepaticus urease is not required for intestinal colonization but promotes hepatic inflammation in male A/JCr mice.42 Most rodent Helicobacter species are urease-negative and thus preferentially colonize the intestine, although in some cases they may translocate to the liver and biliary system, stomach,95 or other tissues.95,99

Table 1.

Rodent host species and sites of Helicobacter infection.

Helicobacter spp. Infected speciesa Site of infection References
H. aurati Hamster Stomach, intestine 82, 83
H. bilis Mouse, rat Intestine 35
H. cholecystus Hamster Gallbladder 36
H. cinaedi Hamster Intestine 108
H. ganmani Mouse, rat Intestine 89
H. hepaticus Mouse, rat, gerbil Intestine 25, 44, 97
H. mesocricetorum Hamster Intestine 104
H. mastomyrinus Mastomys natalensis Liver 101
H. muridarium Mouse, rat Intestine, stomach 54
H. pylori (experimental infections only) Mouse Stomach 93
H. rodentium Mouse, rat Intestine 100
H. trogontum Rat, mouse Intestine 73, 75
H. typhlonius Mouse, rat Intestine 39
a

Unless noted, all mice listed are Mus musculus.

PCR-based techniques, mainly genus-specific PCR and quantitative PCR, typically are used for identification and detection of most Helicobacter spp. Molecular detection of Helicobacter DNA by using PCR is rapid and sensitive to the early phases of infection. Further enhanced sensitivity is achieved by using nested primers.63 One of the most important features of the PCR assay is that it can be performed noninvasively on fecal pellets.3,25,57 Thus analysis for the presence of Helicobacter spp. does not require euthanasia of the animals being tested, an especially important aspect when monitoring valuable rodents, including transgenic and knockout mice.

Transmission

Data regarding the transmission of Helicobacter spp. are not consistent. For example, H. hepaticus—which is morphologically similar to 3 other intestinal Helicobacter spp.: H. fennelliae, H. cinaedi, and H. canis26—was transmitted from colony to sentinel mice quickly by means of contaminated bedding.58 Furthermore, H. hepaticus was established as a member of the cecal mucosal-associated microbiota rapidly when administered by gavage to adult mice and eventually grow to become a dominant member of the bacterial community by 14 d.52 Other researchers reported delayed and inconsistent transmission of H. bilis to sentinel mice.120 Clearly, transmission of Helicobacter spp. depends heavily on the species involved and on environmental factors, including husbandry practices. Fecal–oral spread is the primary route of natural acquisition of infection in rodents. Once acquired, Helicobacter infections are persistent, with long-term shedding of the organisms in feces. Endemic infections therefore are very common in research facilities, unless specific steps are taken to prevent and eradicate these microorganisms. Several studies21,22,51,95 have reported successful eradication of Helicobacter infections. Methods included depopulation followed by restocking with Helicobacter-free mice, embryo transfer, cross-fostering, and antibiotic therapy,21,22,51,95 but the efficiency of these methods may vary depending on species and strain of rodent or Helicobacter organism and experimental conditions.

In many cases, investigators can be unaware of the infections, because the pathology of most Helicobacter species is host-dependent, and infection may be subclinical.119 A recent study on prevalence and spread of enterohepatic Helicobacter species (H. ganmani, H. hepaticus, H. typhlonius, and others)4 showed that infections can be prevented effectively by harboring mice in individually ventilated cages even if infected mice are reared in neighboring cages. The same study4 showed that mice reared in open-air cages were very likely to become infected. However, the use of filter tops on conventional cages was sufficient to prevent Helicobacter transmission between cages,4 probably by preventing spread of contaminated stool and bedding.

Helicobacter infections in humans and mice

Presence of Helicobacter spp. profoundly affects the host organism. A recent study77 profiled the changes in gene expression in cecal tissue from disease-susceptible A/JCr and disease-resistant C57BL/6 mice longitudinally, from the acute to the chronic phases of H. hepaticus colonization. The results demonstrated that disease-susceptible A/JCr mice exhibit a sustained or biphasic Th1-mediated inflammatory response, whereas disease-resistant C57BL/6 mice exhibit a transient regulated response that was predominated by immunoglobulin-related gene expression. The immunologic mechanisms that regulate this balance may be related to disruption of cytokine signaling and control of the cell cycle.77 In addition to dysregulated inflammation, disruption of the epithelial barrier may play a role. Overall, Helicobacter organisms in mice14-16,46,99 have been linked to inflammatory bowel disease6,48 and breast,87 liver,105 gastric1 and colon cancers.

Colonic inflammation and neoplasia.

The first reports of pathogenic intestinal Helicobacter infections appeared in 1994.26,116 Most scientists consider that the pathogenic potential of intestinal Helicobacter spp. varies. Helicobacter hepaticus or H. bilis have been used most frequently to model microbial triggers of colon inflammation, because these species were the first to be linked to the development of inflammatory bowel disease and inflammation-associated neoplasia.6,37,60,62 A study20 investigating the association between colonization of the lower intestinal tract with H. hepaticus of multiple genetically altered lines, including Rag−/− and p53+/- mice, showed rectal prolapse and histologic evidence of proliferative typhlitis. Colitis or proctitis was present in 65% of the animals examined, 89% of which were positive for H. hepaticus as detected by species-specific PCR.20 However, H. typhlonius, H. rodentium, H. muridarum, H. ganmani, H. trogontum, and other species have also been shown to trigger intestinal inflammation in susceptible mice28,37,89,103,121,125 and are commonly endemic within research animal facilities. Table 2 lists mouse models of Helicobacter infection associated with gastrointestinal and liver cancer.

Table 2.

Rodent models of Helicobacter-associated gastrointestinal and liver cancer

Mouse strain Helicobacter spp. involved Tumor References
INS-GAS H. pylori, H. felis Gastric adenocarcinoma 50,55
BALB/c-IL10−/− H. hepaticus Colon carcinoma 79
IL10−/− (C57BL/6) H. typhlonius, H. rodentium, H. hepaticus Colon carcinoma 9,46,53
Mdr1a−/− H. hepaticus, H. bilis Colon carcinoma 60,111
Rag2−/− H. hepaticus Colon carcinoma 15
A/JCr H. hepaticus Hepatocellular carcinoma 92,110
Smad3 H. hepaticus, H. bilis Colon carcinoma 62

Infections with H. hepaticus and H. bilis have been established firmly as a trigger of the colonic inflammation in IL10−/− adult mice susceptible to inflammatory bowel disease6,60 and will not be considered in this section of the review. Genetically, H. typhlonius is very closely related to H. hepaticus, having only 2.36% difference in the 16S rRNA gene sequence, but H. typhlonius has a unique intervening sequence in this gene that makes it easily recognizable by PCR.18,39 H. typhlonius previously was shown to cause an enteric disease characterized by mucosal hyperplasia and associated inflammation in the cecum and colon in immunodeficient mice39 and IL10−/− mice.28 In those studies, colitis was relatively mild, with no development of inflammation-associated neoplasia. Another organism from the Helicobacter genus, H. rodentium, was the first urease-negative, murine Helicobacter species isolated from intestines.100 H. rodentium has been described to be nonpathogenic in adult wild-type mice, but this species did enhance cytokine production in mice also infected with H. hepaticus.78 Coinfection with H. hepaticus and H. rodentium was associated with augmented cecal gene expression and with clinical diarrheal disease in immunodeficient mice.78,102

H. rodentium and H. typhlonius caused rapid onset of severe colon inflammation and multiple neoplastic lesions in the colons of IL10−/− mice neonatally coinfected with those 2 strains.46 Those mice rapidly developed severe colitis with a high rate of rectal prolapse (this condition was the indication for euthanasia in 50% of the mice and occurred as early as 5 wk of age, with a mean of 21 wk). Colonic neoplasia was present in 13 of 14 Helicobacter-infected IL10−/− mice (compared with 0 of 8 with spontaneous colitis), with a mean of 4 neoplastic lesions per colon and invasive adenocarcinoma in 57%. In addition, rapid onset of severe inflammatory bowel disease and a high incidence of inflammation-associated neoplasia occurred in IL10−/− mice that were monoinfected with either H. typhlonius or H. rodentium.9 In that study, Helicobacter organisms were detected in the stomach and all portions of the colon, and H. rodentium DNA was detected in the small intestine and in 1 mesenteric lymph node sample. Other researchers have reported the presence of H. typhlonius in sex organs and gastric tissue of C57BL/6 mice.95 Overall, both mixed and monoinfections with H. rodentium and H. typhlonius suggest a synergistic role of those 2 in their inflammatory mechanism, but the exact mechanism is still unknown.

After infection with H. hepaticus, 129/SvEv Rag2−/− mice lacking mature lymphocytes developed several different types of colon cancer associated with colitis,15,16 whereas sham-dosed mice did not show any signs of the disease. Inflammatory bowel disease and carcinoma that developed in H. hepaticus-infected Rag2−/− mice were abrogated by treatment with IL10-competent regulatory T cells.16 Interestingly, sites of H. hepaticus-induced cancer in that study coincided with the primary sites of colonization of H. hepaticus in other inbred strains of mice.43,119 Other studies using immunodeficient mice have revealed similar protective and therapeutic effects mediated by regulatory T cells in mice with colitis.64,85 Those authors proposed that a signaling pathway involving IL10 and IL6 is essential in maintaining epithelial homeostasis and modulating epithelial invasion during bacterially driven inflammatory diseases.

Recent studies demonstrated that mice lacking Smad3, a signaling molecule in the transforming growth factor β pathway, do not manifest the colon cancer phenotype when maintained in a Helicobacter-free environment for as long as 9 mo.62 Infection of the same mouse strain with H. hepaticus, H. bilis, and a novel Helicobacter species triggered colon cancer in 50% to 66% of the animals.62 These results add to the numerous reports suggesting that bacterial triggers may be important in colorectal cancer, especially in the context of genetic loss of antiinflammatory and antiproliferative signals, such as those provided by transforming growth factor β. In addition, the same research group demonstrated that H. bilis causes inflammatory bowel disease in Mdr1−/− mice that are deficient in P-glycoprotein transporter61 and that coinfection with H. hepaticus and H. bilis results in colitis and eventually progresses to dysplasia.60

Gastric inflammation and inflammation-associated cancer.

Helicobacter spp. isolated from the stomachs of humans and animals have become a subject of intense research because of their association with gastric disease.23,66 Infection with H. pylori in humans has been associated with progression from gastritis to gastric adenocarcinoma12 and is recognized as a key risk factor for the development of gastric mucosa-associated lymphoid tissue (MALT) lymphoma.17 Although H. pylori does not naturally infect mice, this species is the best studied member of the Helicobacter genus and frequently is used in mice to generate experimental models of gastric inflammation and cancer. In a recent study, INS-GAS transgenic mice overexpressing amidated gastrin were used to elucidate the effect of H. pylori eradication therapy conducted at different stages of H. pylori-associated gastric pathology.55 In that study, all H. pylori-infected INS-GAS mice had developed gastrointestinal intraepithelial neoplasia or gastric cancer at 28 wk after infection, accompanied by inflammation, loss of parietal and chief cells, and hypertrophy of foveolar glands. When H. pylori antimicrobial eradication therapy was instituted at 8 wk after infection, the risk of gastrointestinal intraepithelial neoplasia was reduced to a level comparable to that of uninfected mice. Several clinical trials have examined the efficacy of H. pylori eradication in preventing the development of preneoplastic gastric lesions and gastric cancer in humans74,122,123 and have proven that such eradication is a successful prevention therapy against gastric cancer.

Lee and colleagues54 were the first to describe H. muridarum, a spiral bacterium morphologically and biochemically associated with an inflammatory response in both antral and oxyntic gastric mucosa of mice. The histologic changes in animals infected with H. muridarum, characterized by mononuclear and polymorphonuclear leukocytes, closely resembled those in human antral mucosa colonized by H. pylori. H. muridarum shares various characteristics (morphologically and biochemically) with other microorganisms that colonize gastric mucosa in the normal noninfected host; however, H. muridarum provoked a gastric inflammatory reaction. Another research group independently isolated the same bacterium86 and confirmed that H. muridarum has a potential to colonize both the stomach and intestinal tract of rodents. The authors concluded that this bacterium naturally colonizes the ileum and cecum but can also elicit gastritis after colonizing the gastric mucosa of older animals. Currently, no publications report the occurrence of carcinoma associated with H. muridarum infection.

Hepatocarcinoma and hepatic inflammation

H. hepaticus is an enterohepatic species that persistently colonizes the colon and cecum.98 This bacterium originally was discovered as the causative agent for the development of chronic hepatitis and hepatocellular cancer in A/JCr mice and other mouse strains.26,34,56,92,116 In addition, H. hepaticus reportedly produces hepatocarcinoma with a male bias.30,91 Tumors usually arise after 18 mo, whereas after 1 y, susceptible mice exhibit chronic hepatitis.90 Other studies41,77 have shown that H. hepaticus efficiently colonizes the colon of C57BL/6 mice; however substrains vary in their propensity to develop chronic hepatitis after infection. The isolation of H. hepaticus from the spleen of 1 mouse 3 wk after infection suggested hematogenous spread of H. hepaticus early in the course of disease, but H. hepaticus was not isolated from the spleen at later time points. In another carcinogenesis study,45 B6C3F1 mice that were infected with H. hepaticus developed hepatitis, hepatocellular carcinoma, and hemangiosarcoma of the liver. In addition, increases in cell proliferation rates and apoptosis were observed in the livers of male mice with H. hepaticus-associated hepatitis.

In an experiment using F1 hybrid mice derived from A/J and C57BL/6 matings,41 mice from both groups were infected with H. hepaticus. Significant hepatitis was noticed in all H. hepaticus-infected parental strains 18 mo after infection. Overall, hepatocellular carcinomas or dysplastic liver lesions were present in 69% of H. hepaticus-infected F1 male mice, and H. hepaticus was isolated from hepatic tissues of all F1 mice with liver tumors. Chronic active hepatitis was prevalent in other strains of mice as well, including C3H/HeNCr, SJL/NCr, BALB/cAnNCr, and SCID/NCr.115,116 Other Helicobacter spp. associated with the rodent models of liver cancer and hepatobiliary system are H. bilis,32,35 H. mastomyrinus,101 and H. cholecystus.36,84

In humans, hepatic Helicobacter species DNA has been identified in liver tissue, bile and gallbladder tissue from patients with primary sclerosing cholangitis, primary biliary cirrhosis, chronic cholecystitis, and biliary tract malignancies.24,67,81 Increased serum antibodies to enterohepatic Helicobacter species have also been detected in humans with chronic liver diseases.114 In another study,71 coinfection with both H. hepaticus and H. rodentium and monoinfection with H. bilis promoted cholesterol gallstone formation in C57BL mice. Those researchers suggested that this process was mediated by the species-specific bacterial products or a reaction of the host to these products, possibly through cytokines and other proinflammatory mediators. Interestingly, mice infected with both H. rodentium and H. hepaticus developed cholesterol gallstones at the higher rate (78%) than did those infected with either H. rodentium or H. hepaticus alone (30 and 40%, respectively).

Breast cancer.

Inflammation induced in the gut by proinflammatory microbial infection possibly could have systemic effects, which would then influence carcinogenic events in distant organs. Adenomatous polyposis coli (Apc) is a gene responsible for multiple intestinal neoplasia in human and mice.80,112 Infecting Rag2-deficient C57BL/6 ApcMin/+ (multiple intestinal neoplasia) mice with H. hepaticus significantly promotes mammary carcinoma in female mice and enhances intestinal adenoma multiplicity by a TNF-dependent mechanism.87 In that study, mammary tumors from H. hepaticus-infected Rag2−/−ApcMin/+ mice showed an 18-fold increase in TNF expression when compared with mammary tissue of uninfected mice. In addition, neutralization of TNF by antibody significantly suppressed both intestinal and mammary tumors in H. hepaticus-infected mice, suggesting that TNF or its downstream signaling mediators are required to sustain tumors. The authors of that study were the first to report that an enteric microbial infection promotes cancer in the mammary gland. However, whether this outcome is a direct or indirect effect of infection is unknown, given that the presence of Helicobacter organisms in the mammary tissue was not analyzed in the study.

Effects on reproduction.

Thus far, the information regarding effects of Helicobacter spp. on reproduction is scarce. In one study, H. typhlonius was detected by PCR in the sex organs of 3 mouse strains: immunodeficient athymic nude-nu (nu-nu), Helicobacter-sensitive C3H/HeJ and Helicobacter-resistant C57BL/6 wild type mice.95 Sentinel mice of these 3 strains became infected at different times after first exposure to an infected cagemate. H. typhlonius DNA was documented to be present in testes, epididymis, uterus, and ovaries in all 3 mouse strains tested for a period of 1 to 2 wk at different times after exposure. Most tissues from C3H/HeJ mice were positive during the first round of PCR, whereas most of the samples derived from the C57BL/6J mice needed a second round of PCR with nested primers to detect the positivity. These results suggest that mice experience a transient bacteremia after Helicobacter infection, with clearing of microorganisms rapidly from nontarget tissues followed by preferential long-term colonization of the gastrointestinal tract. In addition, H. hepaticus was cultured from fetal viscera of 2 of 11 pups sampled late in gestation from infected SCID/NCr females, suggesting transplacental infection of H. hepaticus.56 Apparent differences among different mouse strains and animal facilities in the detection of the Helicobacter in reproductive tissues might reflect quantification issues and not by the presence (or absence) of infection itself.

Experimental infection with H. pylori influenced murine pregnancy by increasing the number of fetal resorptions and producing decreased fetal weights when compared with those of noninfected CD1 mice.93 Induction of Th1-type responses at the endometrial level was 1 mechanism suggested for these phenomena, but this notion was not investigated further. We recently showed that the reproductive success of C57BL/6 IL10−/− female mice intentionally infected with H. typhlonius or H. rodentium (or both organisms) was decreased compared with that of noninfected mice.99 Pregnancy rates and the number of pups surviving to weaning were decreased in infected dams. Quantitative PCR detected Helicobacter DNA in the reproductive organs of 1 infected mouse on day 82 after infection and in 4 of 5 mice on day 7 after infection. Treatment with a 4-drug antiHelicobacter therapy eliminated PCR-detectable excretion of Helicobacter DNA, improved fecundity, and enhanced survival of pups born to previously infected dams. Clearly, additional studies are required to investigate the influence of Helicobacter spp. infection on reproductive success in other mouse strains. However, numerous published studies performed with mice that were infected unintentionally or unknown to be infected might have to be reevaluated, because the breeding results in those strains could have been due to Helicobacter infection and not to the genetic manipulations or treatments used.

Helicobacter and immunity.

Oral tolerance is a systemic unresponsiveness (that is, lack of specific antibody production or cell-mediated response) to the same antigens subsequently delivered systemically.7 In some conditions, oral tolerance can be inhibited, and this effect can be considered equivalent to promotion of sensitization. Such suppression involves signaling by an array of facultative antigen-presenting cells, dendritic cells, and regulatory T cells, as well as lymphocyte anergy or deletion. The inhibitory effect of Helicobacter infection on oral tolerance potentially could contribute to the persistent, chronic gastric inflammation and development of allergic response in the laboratory animal models. Indeed, the association between H. pylori infection and food allergy11,19 as well as with other allergic diseases like chronic urticaria,8,118 atopic dermatitis,76 and hereditary angioneurotic edema113 has been suggested. Infection with H. pylori or H. felis has been shown to increase the absorption of antigens across the digestive epithelium in vitro69 and across the gastric mucosa in vivo in mice.68 In 1 study,117 scientists studied gastric inflammation and T cell response in H. pylori-challenged mice after intraperitoneal immunization using H. pylori whole-cell lysates in the absence of adjuvants. H. pylori-challenged mice without immunization developed moderate to severe gastric inflammation, whereas mice inoculated with H. pylori after immunization had little or no gastric inflammation despite persistent colonization with this Helicobacter species. The mechanism described by the authors might lead to a state of controlled inflammation in subjects harboring H. pylori.

Bacterial flagellin, the primary structural component of flagella, is a dominant target of humoral immunity in response to infection with flagellated pathogenic bacteria94 including many Helicobacter spp. Briefly, flagellin monomers released by bacteria are recognized by receptor molecules and activate innate immunity, in particular triggering a rapid induction of proinflammatory gene expression.47 Flagellin monomers are a target of the heightened adaptive immune response associated with Crohn disease,59,106 and purified flagellin functions as a T cell adjuvant.13,72 Generation of flagellin-specific immunoglobulins in response to intraperitoneal injection with flagellin requires activation of innate immunity, is T-cell–dependent, and can originate from Helicobacter flagellin present in the intestinal tract during inflammatory conditions.94

C3H/He mice have been used as an experimental model for oral tolerance to ovalbumin.40 These mice are colonized readily by H. felis and develop gastric inflammation in response. Colonization with H. felis can inhibit the development of oral tolerance to ovalbumin in mice;70 development of this tolerance was preserved by treatment rebamipide, a gastroprotective agent used in the treatment of gastritis and ulcerative colitis. The mechanisms may involve the increase in antigenic absorption across the epithelium and the presence of bacterial adjuvants in H. felis-infected mice.68

Conclusions

The potential influence of Helicobacter spp. on in vivo experiments continues to be an important issue regarding rodent models used for biomedical research. The data presented here show clearly that infection with Helicobacter spp. can affect numerous areas of research. This review focuses on effects of Helicobacter infection on the development of cancer (including those of the gastrointestinal tract and breast), immune response, and reproduction of mouse colonies, as well as on the progress of chronic and acute inflammation. Many investigators may be unaware of the infections present in their colonies because the pathology of the infection is host-dependent and can be subclinical. Therefore, frequent screening and the eradication of Helicobacter spp. from laboratory animal facilities should be key goals of the research community. The findings we have presented stress the importance of appropriate husbandry practices and the prevention of infection to provide a reliable environment for future research studies and results.

Acknowledgment

This work was supported by grant 5R01CA115480 from the National Institutes of Health.

References

  • 1.Abad A. 2008. New drugs in the treatment of gastric tumors. Clin Transl Oncol 10:256–261 [DOI] [PubMed] [Google Scholar]
  • 2.Baele M, Decostere A, Vandamme P, Ceelen L, Hellemans A, Mast J, Chiers K, Ducatelle R, Haesebrouck F. 2008. Isolation and characterization of Helicobacter suis sp. nov. from pig stomachs. Int J Syst Evol Microbiol 58:1350–1358 [DOI] [PubMed] [Google Scholar]
  • 3.Beckwith CS, Franklin CL, Hook RR, Jr, Besch-Williford CL, Riley LK. 1997. Fecal pcr assay for diagnosis of Helicobacter infection in laboratory rodents. J Clin Microbiol 35:1620–1623 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bohr UR, Selgrad M, Ochmann C, Backert S, Konig W, Fenske A, Wex T, Malfertheiner P. 2006. Prevalence and spread of enterohepatic Helicobacter species in mice reared in a specific-pathogen-free animal facility. J Clin Microbiol 44:738–742 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Bridgeford EC, Marini RP, Feng Y, Parry NM, Rickman B, Fox JG. 2008. Gastric Helicobacter species as a cause of feline gastric lymphoma: a viable hypothesis. Vet Immunol Immunopathol 123:106–113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Burich A, Hershberg R, Waggie K, Zeng W, Brabb T, Westrich G, Viney JL, Maggio-Price L. 2001. Helicobacter-induced inflammatory bowel disease in IL10- and T cell-deficient mice. Am J Physiol Gastrointest Liver Physiol 281:G764–G778 [DOI] [PubMed] [Google Scholar]
  • 7.Burks AW, Laubach S, Jones SM. 2008. Oral tolerance, food allergy, and immunotherapy: implications for future treatment. J Allergy Clin Immunol 121:1344–1350 [DOI] [PubMed] [Google Scholar]
  • 8.Campli CD, Gasbarrini A, Nucera E, Franceschi F, Ojetti V, Torre ES, Schiavino D, Pola P, Patriarca G, Gasbarrini G. 1998. Beneficial effects of Helicobacter pylori eradication on idiopathic chronic urticaria. Dig Dis Sci 43:1226–1229 [DOI] [PubMed] [Google Scholar]
  • 9.Chichlowski M, Sharp JM, Vanderford DA, Myles MH, Hale LP. 2008. Helicobacter typhlonius and H. Rodentium differentially affect the severity of colon inflammation and inflammation-associated neoplasia in IL0-deficient mice. Comp Med 58:534–541 [PMC free article] [PubMed] [Google Scholar]
  • 10.Compton SR, Ball-Goodrich LJ, Zeiss CJ, Johnson LK, Johnson EA, Macy JD. 2003. Pathogenesis of mouse hepatitis virus infection in gamma interferon-deficient mice is modulated by coinfection with Helicobacter hepaticus. Comp Med 53:197–206 [PubMed] [Google Scholar]
  • 11.Corrado G, Luzzi I, Lucarelli S, Frediani T, Pacchiarotti C, Cavaliere M, Rea P, Cardi E. 1998. Positive association between Helicobacter pylori infection and food allergy in children. Scand J Gastroenterol 33:1135–1139 [DOI] [PubMed] [Google Scholar]
  • 12.Correa P, Houghton J. 2007. Carcinogenesis of Helicobacter pylori. Gastroenterology 133:659–672 [DOI] [PubMed] [Google Scholar]
  • 13.Didierlaurent A, Ferrero I, Otten LA, Dubois B, Reinhardt M, Carlsen H, Blomhoff R, Akira S, Kraehenbuhl J-P, Sirard J-C. 2004. Flagellin promotes myeloid differentiation factor 88-dependent development of Th2-type response. J Immunol 172:6922–6930 [DOI] [PubMed] [Google Scholar]
  • 14.Engle SJ, Ormsby I, Pawlowski S, Boivin GP, Croft J, Balish E, Doetschman T. 2002. Elimination of colon cancer in germ-free transforming growth factor β1-deficient mice. Cancer Res 62:6362–6366 [PubMed] [Google Scholar]
  • 15.Erdman SE, Poutahidis T, Tomczak M, Rogers AB, Cormier K, Plank B, Horwitz BH, Fox JG. 2003. CD4+ CD25+ regulatory T lymphocytes inhibit microbially induced colon cancer in Rag2-deficient mice. Am J Pathol 162:691–702 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Erdman SE, Rao VP, Poutahidis T, Ihrig MM, Ge Z, Feng Y, Tomczak M, Rogers AB, Horwitz BH, Fox JG. 2003. CD4+CD25+ regulatory lymphocytes require interleukin 10 to interrupt colon carcinogenesis in mice. Cancer Res 63:6042–6050 [PubMed] [Google Scholar]
  • 17.Farinha P, Gascoyne RD. 2005. Helicobacter pylori and MALT lymphoma. Gastroenterology 128:1579–1605 [DOI] [PubMed] [Google Scholar]
  • 18.Feng S, Ku K, Hodzic E, Lorenzana E, Freet K, Barthold SW. 2005. Differential detection of five mouse-infecting Helicobacter species by multiplex PCR. Clin Diagn Lab Immunol 12:531–536 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Figura N, Perrone A, Gennari C, Orlandini G, Bianciardi L, Giannace R, Vaira D, Vagliasinti M, Rottoli P. 1999. Food allergy and Helicobacter pylori infection. Ital J Gastroenterol Hepatol 31:186–191 [PubMed] [Google Scholar]
  • 20.Foltz CJ, Fox JG, Cahill R, Murphy JC, Yan L, Shames B, Schauer DB. 1998. Spontaneous inflammatory bowel disease in multiple mutant mouse lines: association with colonization by Helicobacter hepaticus. Helicobacter 3:69–78 [DOI] [PubMed] [Google Scholar]
  • 21.Foltz CJ, Fox JG, Yan L, Shames B. 1995. Evaluation of antibiotic therapies for eradication of Helicobacter hepaticus. Antimicrob Agents Chemother 39:1292–1294 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Foltz CJ, Fox JG, Yan L, Shames B. 1996. Evaluation of various oral antimicrobial formulations for eradication of Helicobacter hepaticus. Lab Anim Sci 46:193–197 [PubMed] [Google Scholar]
  • 23.Fox JG, Blanco M, Murphy JC, Taylor NS, Lee A, Kabok Z, Pappo J. 1993. Local and systemic immune responses in murine Helicobacter felis active chronic gastritis. Infect Immun 61:2309–2315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Fox JG, Dewhirst FE, Shen Z, Feng Y, Taylor NS, Paster BJ, Ericson RL, Lau CN, Correa P, Araya JC, Roa I. 1998. Hepatic Helicobacter species identified in bile and gallbladder tissue from Chileans with chronic cholecystitis. Gastroenterology 114:755–763 [DOI] [PubMed] [Google Scholar]
  • 25.Fox JG, Dewhirst FE, Fraser GJ, Paster BJ, Shames B, Murphy JC. 1994. Intracellular Campylobacter-like organism from ferrets and hamsters with proliferative bowel disease is a Desulfovibrio sp. J Clin Microbiol 32:1229–1237 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Fox JG, Dewhirst FE, Tully JG, Paster BJ, Yan L, Taylor NS, Collins MJ, Jr, Gorelick PL, Ward JM. 1994. Helicobacter hepaticus sp. nov., a microaerophilic bacterium isolated from livers and intestinal mucosal scrapings from mice. J Clin Microbiol 32:1238–1245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Fox JG, Drolet R, Higgins R, Messier S, Yan L, Coleman BE, Paster BJ, Dewhirst FE. 1996. Helicobacter canis isolated from a dog liver with multifocal necrotizing hepatitis. J Clin Microbiol 34:2479–2482 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Fox JG, Gorelick PL, Kullberg MC, Ge Z, Dewhirst FE, Ward JM. 1999. A novel urease-negative Helicobacter species associated with colitis and typhlitis in IL10-deficient mice. Infect Immun 67:1757–1762 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Fox JG, Handt L, Sheppard BJ, Xu S, Dewhirst FE, Motzel S, Klein H. 2001. Isolation of Helicobacter cinaedi from the colon, liver, and mesenteric lymph node of a rhesus monkey with chronic colitis and hepatitis. J Clin Microbiol 39:1580–1585 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Fox JG, Li X, Yan L, Cahill RJ, Hurley R, Lewis R, Murphy JC. 1996. Chronic proliferative hepatitis in A/JCr mice associated with persistent Helicobacter hepaticus infection: a model of Helicobacter-induced carcinogenesis. Infect Immun 64:1548–1558 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fox JG, MacGregor JA, Shen Z, Li X, Lewis R, Dangler CA. 1998. Comparison of methods of identifying Helicobacter hepaticus in B6C3F1 mice used in a carcinogenesis bioassay. J Clin Microbiol 36:1382–1387 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Fox JG, Rogers AB, Whary MT, Taylor NS, Xu S, Feng Y, Keys S. 2004. Helicobacter bilis-associated hepatitis in outbred mice. Comp Med 54:571–577 [PubMed] [Google Scholar]
  • 33.Fox JG, Shen Z, Xu S, Feng Y, Dangler CA, Dewhirst FE, Paster BJ, Cullen JM. 2002. Helicobacter marmotae sp. nov. isolated from livers of woodchucks and intestines of cats. J Clin Microbiol 40:2513–2519 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Fox JG, Yan L, Shames B, Campbell J, Murphy JC, Li X. 1996. Persistent hepatitis and enterocolitis in germfree mice infected with Helicobacter hepaticus. Infect Immun 64:3673–3681 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Fox JG, Yan LL, Dewhirst FE, Paster BJ, Shames B, Murphy JC, Hayward A, Belcher JC, Mendes EN. 1995. Helicobacter bilis sp. nov., a novel Helicobacter species isolated from bile, livers, and intestines of aged, inbred mice. J Clin Microbiol 33:445–454 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Franklin CL, Beckwith CS, Livingston RS, Riley LK, Gibson SV, Besch-Williford CL, Hook RR., Jr 1996. Isolation of a novel Helicobacter species, Helicobacter cholecystus sp. nov., from the gallbladders of Syrian hamsters with cholangiofibrosis and centrilobular pancreatitis. J Clin Microbiol 34:2952–2958 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Franklin CL, Gorelick PL, Riley LK, Dewhirst FE, Livingston RS, Ward JM, Beckwith CS, Fox JG. 2001. Helicobacter typhlonius sp. nov., a novel murine urease-negative Helicobacter species. J Clin Microbiol 39:3920–3926 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Franklin CL, Riley LK, Livingston RS, Beckwith CS, Besch-Williford CL, Hook RRJ. 1998. Enterohepatic lesions in SCID mice infected with Helicobacter bilis. Lab Anim Sci 48:334–339 [PubMed] [Google Scholar]
  • 39.Franklin CL, Riley LK, Livingston RS, Beckwith CS, Hook RR, Jr, Besch-Williford CL, Hunziker R, Gorelick PL. 1999. Enteric lesions in SCID mice infected with ‘Helicobacter typhlonicus,’ a novel urease-negative Helicobacter species. Lab Anim Sci 49:496–505 [PubMed] [Google Scholar]
  • 40.Gaboriau-Routhiau V, Moreau MC. 1996. Gut flora allows recovery of oral tolerance to ovalbumin in mice after transient breakdown mediated by cholera toxin or Escherichia coli heat-labile enterotoxin. Pediatr Res 39:625–629 [DOI] [PubMed] [Google Scholar]
  • 41.Garcia A, Ihrig MM, Fry RC, Feng Y, Xu S, Boutin SR, Rogers AB, Muthupalani S, Samson LD, Fox JG. 2008. Genetic susceptibility to chronic hepatitis is inherited codominantly in Helicobacter hepaticus-infected AB6F1 and B6AF1 hybrid male mice, and progression to hepatocellular carcinoma is linked to hepatic expression of lipogenic genes and immune function-associated networks. Infect Immun 76:1866–1876 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ge Z, Lee A, Whary MT, Rogers AB, Maurer KJ, Taylor NS, Schauer DB, Fox JG. 2008. Helicobacter hepaticus urease is not required for intestinal colonization but promotes hepatic inflammation in male A/JCr mice. Microb Pathog 45:18–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ge Z, White DA, Whary MT, Fox JG. 2001. Fluorogenic PCR-based quantitative detection of a murine pathogen, Helicobacter hepaticus. J Clin Microbiol 39:2598–2602 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Goto K, Jiang W, Zheng Q, Oku Y, Kamiya H, Itoh T, Ito M. 2004. Epidemiology of Helicobacter infection in wild rodents in the Xinjiang–Uygur autonomous region of China. Curr Microbiol 49:221–223 [DOI] [PubMed] [Google Scholar]
  • 45.Hailey JR, Haseman JK, Bucher JR, Radovsky AE, Malarkey DE, Miller RT, Nyska A, Maronpot RR. 1998. Impact of Helicobacter hepaticus infection in B6C3F1 mice from 12 national toxicology program 2-year carcinogenesis studies. Toxicol Pathol 26:602–611 [DOI] [PubMed] [Google Scholar]
  • 46.Hale LP, Perera D, Gottfried MR, Maggio-Price L, Srinivasan S, Marchuk D. 2007. Neonatal coinfection with Helicobacter species markedly accelerates the development of inflammation-associated colonic neoplasia in IL10 −/− mice. Helicobacter 12:598–604 [DOI] [PubMed] [Google Scholar]
  • 47.Hayashi F, Smith KD, Ozinsky A, Hawn TR, Yi EC, Goodlett DR, Eng JK, Akira S, Underhill DM, Aderem A. 2001. The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature 410:1099–1103 [DOI] [PubMed] [Google Scholar]
  • 48.Jiang H-Q, Kushnir N, Christine Thurnheer M, Bos NA, Cebra JJ. 2002. Monoassociation of SCID mice with Helicobacter muridarum, but not 4 other enterics, provokes IBD upon receipt of T cells. Gastroenterology 122:1346–1354 [DOI] [PubMed] [Google Scholar]
  • 49.Johansson SK, Feinstein RE, Johansson KE, Lindberg AV. 2006. Occurrence of Helicobacter species other than H. hepaticus in laboratory mice and rats in Sweden. Comp Med 56:110–113 [PubMed] [Google Scholar]
  • 50.Kobayashi M, Lee H, Schaffer L, Gilmartin TJ, Head SR, Takaishi S, Wang TC, Nakayama J, Fukuda M. 2007. A distinctive set of genes is upregulated during the inflammation-carcinoma sequence in mouse stomach infected by Helicobacter felis. J Histochem Cytochem 55:263–274 [DOI] [PubMed] [Google Scholar]
  • 51.Kostomitsopoulos N, Donnelly H, Kostavasili I, Paronis E, Alexakos P, Karayannacos P. 2007. Eradication of Helicobacter bilis and H. hepaticus from infected mice by using a medicated diet. Lab Anim (NY) 36:37–40 [DOI] [PubMed] [Google Scholar]
  • 52.Kuehl CJ, Wood HD, Marsh TL, Schmidt TM, Young VB. 2005. Colonization of the cecal mucosa by Helicobacter hepaticus impacts the diversity of the indigenous microbiota. Infect Immun 73:6952–6961 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Laharie D, Ménard S, Asencio C, Vidal-Martinez T, Rullier A, Zerbib F, Candalh C, Mégraud F, Heyman M, Matysiak-Budnik T. 2007. Effect of rebamipide on the colonic barrier in IL10-deficient mice. Dig Dis Sci 52:84–92 [DOI] [PubMed] [Google Scholar]
  • 54.Lee A, Phillips MW, O'Rourke JL, Paster BJ, Dewhirst FE, Fraser GJ, Fox JG, Sly LI, Romaniuk PJ, Trust TJ. 1992. Helicobacter muridarum sp. nov., a microaerophilic helical bacterium with a novel ultrastructure isolated from the intestinal mucosa of rodents. Int J Syst Bacteriol 42:27–36 [DOI] [PubMed] [Google Scholar]
  • 55.Lee CW, Rickman B, Rogers AB, Ge Z, Wang TC, Fox JG. 2008. Helicobacter pylori eradication prevents progression of gastric cancer in hypergastrinemic INS-GAS mice. Cancer Res 68:3540–3548 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Li X, Fox JG, Whary MT, Yan L, Shames B, Zhao Z. 1998. SCID/NCR mice naturally infected with Helicobacter hepaticus develop progressive hepatitis, proliferative typhlitis, and colitis. Infect Immun 66:5477–5484 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Livingston RS, Riley LK. 2003. Diagnostic testing of mouse and rat colonies for infectious agents. Lab Anim 32:44–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Livingston RS, Riley LK, Besch-Williford CL, Hook RR, Franklin CL. 1998. Transmission of Helicobacter hepaticus infection to sentinel mice by contaminated bedding. Lab Anim Sci 48:291–293 [PubMed] [Google Scholar]
  • 59.Lodes MJ, Cong Y, Elson CO, Mohamath R, Landers CJ, Targan SR, Fort M, Hershberg RM. 2004. Bacterial flagellin is a dominant antigen in Crohn disease. J Clin Invest 113:1296–1306 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Maggio-Price L, Bielefeldt-Ohmann H, Treuting P, Iritani BM, Zeng W, Nicks A, Tsang M, Shows D, Morrissey P, Viney JL. 2005. Dual infection with Helicobacter bilis and Helicobacter hepaticus in p-glycoprotein-deficient Mdr1a−/− mice results in colitis that progresses to dysplasia. Am J Pathol 166:1793–1806 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Maggio-Price L, Shows D, Waggie K, Burich A, Zeng W, Escobar S, Morrissey P, Viney JL. 2002. Helicobacter bilis infection accelerates and H. hepaticus infection delays the development of colitis in multiple drug resistance-deficient (Mdr1a−/−) mice. Am J Pathol 160:739–751 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Maggio-Price L, Treuting P, Zeng W, Tsang M, Bielefeldt-Ohmann H, Iritani BM. 2006. Helicobacter infection is required for inflammation and colon cancer in SMAD3-deficient mice. Cancer Res 66:828–838 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Mahler M, Bedigian HG, Burgett BL, Bates RJ, Hogan ME, Sundberg JP. 1998. Comparison of 4 diagnostic methods for detection of Helicobacter species in laboratory mice. Lab Anim Sci 48:85–91 [PubMed] [Google Scholar]
  • 64.Maloy KJ, Salaun L, Cahill R, Dougan G, Saunders NJ, Powrie F. 2003. CD4+CD25+ Tr cells suppress innate immune pathology through cytokine-dependent mechanisms. J Exp Med 197:111–119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Marshall B, Schoep T. 2007. Helicobacter pylori as a vaccine delivery system. Helicobacter 12 Suppl 2:75–79 [DOI] [PubMed] [Google Scholar]
  • 66.Marshall BJ, Warren JR. 1984. Unidentified curved bacilli in the stomach of patients with gastritis and peptic ulceration. Lancet 323:1311–1315 [DOI] [PubMed] [Google Scholar]
  • 67.Matsukura N, Yokomuro S, Yamada S, Tajiri T, Sundo T, Hadama T, Kamiya S, Naito Z, Fox JG. 2002. Association between Helicobacter bilis in bile and biliary tract malignancies: H. bilis in bile from Japanese and Thai patients with benign and malignant diseases in the biliary tract. Jpn J Cancer Res 93:842–847 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Matysiak-Budnik T, Hashimoto K, Heyman M, de Mascarel A, Desjeux JF, Mégraud F. 1999. Antral gastric permeability to antigens in mice is altered by infection with Helicobacter felis. Eur J Gastroenterol Hepatol 11:1371–1377 [DOI] [PubMed] [Google Scholar]
  • 69.Matysiak-Budnik T, Terpend K, Alain S, Sanson le Pors MJ, Desjeux JF, Megraud F, Heyman M. 1998. Helicobacter pylori alters exogenous antigen absorption and processing in a digestive tract epithelial cell line model. Infect Immun 66:5785–5791 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Matysiak-Budnik T, van Niel G, Megraud F, Mayo K, Bevilacqua C, Gaboriau-Routhiau V, Moreau M-C, Heyman M. 2003. Gastric Helicobacter infection inhibits development of oral tolerance to food antigens in mice. Infect Immun 71:5219–5224 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Maurer KJ, Ihrig MM, Rogers AB, Ng V, Bouchard G, Leonard MR, Carey MC, Fox JG. 2005. Identification of cholelithogenic enterohepatic Helicobacter species and their role in murine cholesterol gallstone formation. Gastroenterology 128:1023–1033 [DOI] [PubMed] [Google Scholar]
  • 72.McSorley SJ, Ehst BD, Yu Y, Gewirtz AT. 2002. Bacterial flagellin is an effective adjuvant for CD4+ T cells in vivo. J Immunol 169:3914–3919 [DOI] [PubMed] [Google Scholar]
  • 73.Mendes EN, Queiroz DM, Dewhirst FE, Paster BJ, Moura SB, Fox JG. 1996. Helicobacter trogontum sp. nov. isolated from the rat intestine. Int J Syst Bacteriol 46:916–921 [DOI] [PubMed] [Google Scholar]
  • 74.Mera R, Fontham ETH, Bravo LE, Bravo JC, Piazuelo MB, Camargo MC, Correa P. 2005. Long-term follow-up of patients treated for Helicobacter pylori infection. Gut 54:1536–1540 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Moura SB, Mendes EN, Queiroz DM, Camargos ER, Evangelina M, Fonseca F, Rocha GA, Nicoli JR. 1998. Ultrastructure of Helicobacter trogontum in culture and in the gastrointestinal tract of gnotobiotic mice. J Med Microbiol 47:513–520 [DOI] [PubMed] [Google Scholar]
  • 76.Murakami K, Fujioka T, Nishizono A, Nagai J, Tokieda M, Kodama R, Kubota T, Nasu M. 1996. Atopic dermatitis successfully treated by eradication of Helicobacter pylori. J Gastroenterol 31:77–82 [PubMed] [Google Scholar]
  • 77.Myles MH, Dieckgraefe BK, Criley JM, Franklin CL. 2007. Characterization of cecal gene expression in a differentially susceptible mouse model of bacterial-induced inflammatory bowel disease. Inflamm Bowel Dis 13:822–836 [DOI] [PubMed] [Google Scholar]
  • 78.Myles MH, Livingston RS, Franklin CL. 2004. Pathogenicity of Helicobacter rodentium in A/JCr and SCID mice. Comp Med 54:549–557 [PubMed] [Google Scholar]
  • 79.Nagamine CM, Rogers AB, Fox JG, Schauer DB. 2008. Helicobacter hepaticus promotes azoxymethane-initiated colon tumorigenesis in BALB/c IL10-deficient mice. Int J Cancer 122:832–838 [DOI] [PubMed] [Google Scholar]
  • 80.Nagamine CM, Sohn JJ, Rickman BH, Rogers AB, Fox JG, Schauer DB. 2008. Helicobacter hepaticus infection promotes colon tumorigenesis in the BALB/c Rag2−/−Apcmin/+ mouse. Infect Immun 76:2758–2766 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Nilsson H-O, Taneera J, Castedal M, Glatz E, Olsson R, Wadstrom T. 2000. Identification of Helicobacter pylori and other Helicobacter species by PCR, hybridization, and partial DNA sequencing in human liver samples from patients with primary sclerosing cholangitis or primary biliary cirrhosis. J Clin Microbiol 38:1072–1076 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Patterson MM, Schrenzel MD, Feng Y, Fox JG. 2000. Gastritis and intestinal metaplasia in Syrian hamsters infected with Helicobacter aurati and 2 other microaerobes. Vet Pathol 37:589–596 [DOI] [PubMed] [Google Scholar]
  • 83.Patterson MM, Schrenzel MD, Feng Y, Xu S, Dewhirst FE, Paster BJ, Thibodeau SA, Versalovic J, Fox JG. 2000. Helicobacter aurati sp. nov., a urease-positive Helicobacter species cultured from gastrointestinal tissues of Syrian hamsters. J Clin Microbiol 38:3722–3728 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Pellicano R, Ménard A, Rizzetto M, Mégraud F. 2008. Helicobacter species and liver diseases: association or causation? Lancet Infect Dis 8:254–260 [DOI] [PubMed] [Google Scholar]
  • 85.Powrie F. 1995. T cells in inflammatory bowel disease: protective and pathogenic roles. Immunity 3:171–174 [DOI] [PubMed] [Google Scholar]
  • 86.Queiroz DM, Contigli C, Coimbra RS, Nogueira AM, Mendes EN, Rocha GA, Moura SB. 1992. Spiral bacterium associated with gastric, ileal, and caecal mucosa of mice. Lab Anim 26:288–294 [DOI] [PubMed] [Google Scholar]
  • 87.Rao VP, Poutahidis T, Ge Z, Nambiar PR, Boussahmain C, Wang YY, Horwitz BH, Fox JG, Erdman SE. 2006. Innate immune inflammatory response against enteric bacteria Helicobacter hepaticus induces mammary adenocarcinoma in mice. Cancer Res 66:7395–7400 [DOI] [PubMed] [Google Scholar]
  • 88.Riley LK, Franklin CL, Hook RR, Jr, Besch-Williford C. 1996. Identification of murine Helicobacters by PCR and restriction enzyme analyses. J Clin Microbiol 34:942–946 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Robertson BR, O'Rourke JL, Vandamme P, On SLW, Lee A. 2001. Helicobacter ganmani sp. nov., a urease-negative anaerobe isolated from the intestines of laboratory mice. Int J Syst Evol Microbiol 51:1881–1889 [DOI] [PubMed] [Google Scholar]
  • 90.Rogers AB, Boutin SR, Whary MT, Sundina N, Ge Z, Cormier K, Fox JG. 2004. Progression of chronic hepatitis and preneoplasia in Helicobacter hepaticus-infected A/JCr mice. Toxicol Pathol 32:668–677 [DOI] [PubMed] [Google Scholar]
  • 91.Rogers AB, Fox JG. 2004. Inflammation and cancer. I. Rodent models of infectious gastrointestinal and liver cancer. Am J Physiol Gastrointest Liver Physiol 286:G361–G366 [DOI] [PubMed] [Google Scholar]
  • 92.Rogers AB, Theve EJ, Feng Y, Fry RC, Taghizadeh K, Clapp KM, Boussahmain C, Cormier KS, Fox JG. 2007. Hepatocellular carcinoma associated with liver–gender disruption in male mice. Cancer Res 67:11536–11546 [DOI] [PubMed] [Google Scholar]
  • 93.Rossi G, Romagnoli S, Lauretti L, Pancotto L, Taccini E, Rappuoli R, Del Giudice G, Ruggiero P. 2004. Helicobacter pylori infection negatively influences pregnancy outcome in a mouse model. Helicobacter 9:152–157 [DOI] [PubMed] [Google Scholar]
  • 94.Sanders CJ, Yu Y, Moore DA, 3rd, Williams IR, Gewirtz AT. 2006. Humoral immune response to flagellin requires T cells and activation of innate immunity. J Immunol 177:2810–2818 [DOI] [PubMed] [Google Scholar]
  • 95.Scavizzi F, Raspa M. 2006. Helicobacter typhlonius was detected in the sex organs of 3mouse strains but did not transmit vertically. Lab Anim 40:70–79 [DOI] [PubMed] [Google Scholar]
  • 96.Schubert ML, Peura DA. 2008. Control of gastric acid secretion in health and disease. Gastroenterology 134:1842–1860 [DOI] [PubMed] [Google Scholar]
  • 97.Seok S, Park J, Cho S, Baek M, Lee H, Kim D, Yang K, Jang D, Han B, Nam K, Park J. 2005. Health surveillance of specific pathogen-free and conventionally housed mice and rats in Korea. Exp Anim 54:85–92 [DOI] [PubMed] [Google Scholar]
  • 98.Shames B, Fox JG, Dewhirst F, Yan L, Shen Z, Taylor NS. 1995. Identification of widespread Helicobacter hepaticus infection in feces in commercial mouse colonies by culture and PCR assay. J Clin Microbiol 33:2968–2972 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Sharp JM, Vanderford DA, Chichlowski M, MM H., Hale LP. 2008. Helicobacter infection decreases reproductive success of IL10-deficient mice. Comp Med 58:447–453 [PMC free article] [PubMed] [Google Scholar]
  • 100.Shen Z, Fox JG, Dewhirst FE, Paster BJ, Foltz CJ, Yan L, Shames B, Perry L. 1997. Helicobacter rodentium sp. nov., a urease-negative Helicobacter species isolated from laboratory mice. Int J Syst Bacteriol 47:627–634 [DOI] [PubMed] [Google Scholar]
  • 101.Shen Z, Xu S, Dewhirst FE, Paster BJ, Pena JA, Modlin IM, Kidd M, Fox JG. 2005. A novel enterohepatic Helicobacter species Helicobacter mastomyrinus isolated from the liver and intestine of rodents. Helicobacter 10:59–70 [DOI] [PubMed] [Google Scholar]
  • 102.Shomer NH, Dangler CA, Marini RP, Fox JG. 1998. Helicobacter bilis–Helicobacter rodentium coinfection associated with diarrhea in a colony of SCID mice. Lab Anim Sci 48:455–459 [PubMed] [Google Scholar]
  • 103.Shomer NH, Dangler CA, Schrenzel MD, Whary MT, Xu S, Feng Y, Paster BJ, Dewhirst FE, Fox JG. 2001. Cholangiohepatitis and inflammatory bowel disease induced by a novel urease-negative Helicobacter species in A/J and tac:Icr:Hascidf RF mice. Exp Biol Med 226:420–428 [DOI] [PubMed] [Google Scholar]
  • 104.Simmons JH, Riley LK, Besch-Williford CL, Franklin CL. 2000. Helicobacter mesocricetorum sp. nov., a novel Helicobacter isolated from the feces of Syrian hamsters. J Clin Microbiol 38:1811–1817 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Sipowicz MA, Weghorst CM, Shiao YH, Buzard GS, Calvert RJ, Anver MR, Anderson LM, Rice JM. 1997. Lack of p53 and Ras mutations in Helicobacter hepaticus-induced liver tumors in A/JCr mice. Carcinogenesis 18:233–236 [DOI] [PubMed] [Google Scholar]
  • 106.Sitaraman SV, Klapproth J-M, Moore DA, 3rd, Landers C, Targan S, Williams IR, Gewirtz AT. 2005. Elevated flagellin-specific immunoglobulins in Crohn disease. Am J Physiol Gastrointest Liver Physiol 288:G403–G406 [DOI] [PubMed] [Google Scholar]
  • 107.Stanley J, Linton D, Burnens AP, Dewhirst FE, On SL, Porter A, Owen RJ, Costas M. 1994. Helicobacter pullorum sp. nov.-genotype and phenotype of a new species isolated from poultry and from human patients with gastroenteritis. Microbiology 140:3441–3449 [DOI] [PubMed] [Google Scholar]
  • 108.Stills HF, Jr, Hook RR, Jr, Kinden DA. 1989. Isolation of a Campylobacter-like organism from healthy Syrian hamsters (Mesocricetus auratus). J Clin Microbiol 27:2497–2501 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Taylor NS, Xu S, Nambiar P, Dewhirst FE, Fox JG. 2007. Enterohepatic Helicobacter species are prevalent in mice from commercial and academic institutions in Asia, Europe, and North America. J Clin Microbiol 45:2166–2172 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Theve EJ, Feng Y, Taghizadeh K, Cormier KS, Bell DR, Fox JG, Rogers AB. 2008. Sex hormone influence on hepatitis in young male A/JCr mice infected with Helicobacter hepaticus. Infect Immun 76:4071–4078 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Torrence AE, Brabb T, Viney JL, Bielefeldt-Ohmann H, Treuting P, Seamons A, Drivdahl R, Zeng W, Maggio-Price L. 2008. Serum biomarkers in a mouse model of bacterial-induced inflammatory bowel disease. Inflamm Bowel Dis 14:480–490 [DOI] [PubMed] [Google Scholar]
  • 112.van Es JH, Giles RH, Clevers HC. 2001. The many faces of the tumor suppressor gene APC. Exp Cell Res 264:126–134 [DOI] [PubMed] [Google Scholar]
  • 113.Visy B, Füst G, Bygum A, Bork K, Longhurst H, Bucher C, Bouillet L, Cicardi M, Farkas H. 2007. Helicobacter pylori infection as a triggering factor of attacks in patients with hereditary angioedema. Helicobacter 12:251–257 [DOI] [PubMed] [Google Scholar]
  • 114.Vorobjova T, Nilsson I, Terjajev S, Granholm M, Lyyra M, Porkka T, Prükk T, Salupere R, Maaroos HI, Wadström T, Uibo R. 2006. Serum antibodies to enterohepatic Helicobacter spp. in patients with chronic liver diseases and in a population with high prevalence of H. pylori infection. Dig Liver Dis 38:171–176 [DOI] [PubMed] [Google Scholar]
  • 115.Ward JM, Anver MR, Haines DC, Benveniste RE. 1994. Chronic active hepatitis in mice caused by Helicobacter hepaticus. Am J Pathol 145:959–968 [PMC free article] [PubMed] [Google Scholar]
  • 116.Ward JM, Fox JG, Anver MR, Haines DC, George CV, Collins MJ, Jr, Gorelick PL, Nagashima K, Gonda MA, Gilden RV, Tully JG, Russell RJ, Benvensite RE, Paster BJ, Dewhirst FE, Donovan JC, Anderson LM, Rice JM. 1994. Chronic active hepatitis and associated liver tumors in mice caused by a presistent bacterial infection with a novel Helicobacter species. J Natl Cancer Inst 86:1222–1227 [DOI] [PubMed] [Google Scholar]
  • 117.Watanabe K, Murakami K, Maeda K, Fujioka T, Nasu M, Nishizono A. 2002. Intraperitoneal immunization led to T cell hyporesponsiveness to Helicobacter pylori infection in mice. Microbiol Immunol 46:441–447 [DOI] [PubMed] [Google Scholar]
  • 118.Wedi B, Wagner S, Werfel T, Manns MP, Kapp A. 1998. Prevalence of Helicobacter pylori-associated gastritis in chronic urticaria. Int Arch Allergy Immunol 116:288–294 [DOI] [PubMed] [Google Scholar]
  • 119.Whary MT, Cline J, King A, Ge Z, Shen Z, Sheppard B, Fox JG. 2001. Long-term colonization levels of Helicobacter hepaticus in the cecum of hepatitis-prone A/JCr mice are significantly lower than those in hepatitis-resistant C57BL/6 mice. Comp Med 51:413–417 [PubMed] [Google Scholar]
  • 120.Whary MT, Cline JH, King AE, Hewes KM, Chojnacky D, Salvarrey A, Fox JG. 2000. Monitoring sentinel mice for Helicobacter hepaticus, H. rodentium, and H. bilis infection by use of polymerase chain reaction analysis and serologic testing. Comp Med 50:436–443 [PubMed] [Google Scholar]
  • 121.Whary MT, Fox JG. 2004. Natural and experimental Helicobacter infections. Comp Med 54:128–158 [PubMed] [Google Scholar]
  • 122.Wong BC-Y, Lam SK, Wong WM, Chen JS, Zheng TT, Feng RE, Lai KC, Hu WHC, Yuen ST, Leung SY, Fong DYT, Ho J, Ching CK, Chen JS. 2004. Helicobacter pylori eradication to prevent gastric cancer in a high-risk region of China: a randomized controlled trial. JAMA 291:187–194 [DOI] [PubMed] [Google Scholar]
  • 123.You WC, Brown LM, Zhang L, Li JY, Jin ML, Chang YS, Ma JL, Pan KF, Liu WD, Hu Y, Crystal-Mansour S, Pee D, Blot WJ, Fraumeni JF, Jr, Xu GW, Gail MH. 2006. Randomized double-blind factorial trial of 3 treatments to reduce the prevalence of precancerous gastric lesions. J Natl Cancer Inst 98:974–983 [DOI] [PubMed] [Google Scholar]
  • 124.Zhang L, Danon SJ, Grehan M, Chan V, Lee A, Mitchell H. 2005. Natural colonization with Helicobacter species and the development of inflammatory bowel disease in IL10-deficient mice. Helicobacter 10:223–230 [DOI] [PubMed] [Google Scholar]
  • 125.Zhang L, Mitchell H. 2006. The roles of mucus-associated bacteria in inflammatory bowel disease. Drugs Today (Barc) 42:605–616 [DOI] [PubMed] [Google Scholar]

Articles from Comparative Medicine are provided here courtesy of American Association for Laboratory Animal Science

RESOURCES