Review Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Feb 7, 2015; 21(5): 1414-1423
Published online Feb 7, 2015. doi: 10.3748/wjg.v21.i5.1414
Association between Helicobacter spp. infections and hepatobiliary malignancies: A review
Fany Karina Segura-López, Hospital de Especialidades UMAE No 71, Instituto Mexicano del Seguro Social, Torreón, Coahuila 27000, México
Alfredo Güitrón-Cantú, Departamento de Endoscopia Gastrointestinal, Hospital de Especialidades UMAE No 71, Instituto Mexicano del Seguro Social, Torreón, Coahuila 2700, México
Javier Torres, Unidad de Investigación en Enfermedades Infecciosas y Parasitarias, Hospital de Pediatría UMAE, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, México CP 06720, DF, México
Author contributions: All authors contributed to the manuscript.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Javier Torres, PhD, Unidad de Investigación en Enfermedades Infecciosas y Parasitarias, Hospital de Pediatría UMAE, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Cuauhtémoc 330, Delegacion Cuauhtémoc, México CP 06720, DF, México. jtorresl57@yahoo.com.mx
Telephone: +52-55-56276940 Fax: +52-55-56276949
Received: September 9, 2014
Peer-review started: September 10, 2014
First decision: October 14, 2014
Revised: October 29, 2014
Accepted: November 19, 2014
Article in press: November 19, 2014
Published online: February 7, 2015

Abstract

Hepatobiliary cancers are highly lethal cancers that comprise a spectrum of invasive carcinomas originating in the liver hepatocellular carcinoma, the bile ducts intrahepatic cholangiocarcinoma and extrahepatic cholangiocarcinoma, the gallbladder and the ampulla of Vater (collectively known as biliary tract cancers). These tumors account for approximately 13% of all annual cancer-related deaths worldwide and for 10%-20% of deaths from hepatobiliary malignancies. Cholangiocarcinoma (CCA) is a devastating disease that displays a poor survival rate for which few therapeutic options are available. Population genetics, geographical and environmental factors, cholelithiasis, obesity, parity, and endemic infection with liver flukes have been identified as risk factors that influence the development of biliary tract tumors. Other important factors affecting the carcinogenesis of these tumors include chronic inflammation, obstruction of the bile ducts, and impaired bile flow. It has been suggested that CCA is caused by infection with Helicobacter species, such as Helicobacter bilis and Helicobacter hepaticus, in a manner that is similar to the reported role of Helicobacter pylori in distal gastric cancer. Due to the difficulty in culturing these Helicobacter species, molecular methods, such as polymerase chain reaction and sequencing, or immunologic assays have become the methods of choice for diagnosis. However, clinical studies of benign or malignant biliary tract diseases revealed remarkable variability in the methods and the findings, and the use of uniform and validated techniques is needed.

Key Words: Helicobacter bilis, Helicobacter hepaticus, Helicobacter species, Extrahepatic cholangiocarcinoma, Cholangiocarcinoma, Biliary tract cancer

Core tip: Hepatobiliary cancers are highly lethal cancers which are difficult to diagnose early and for which few therapeutic options are available. Its etiological factors have been suggested to include chronic inflammation, obstruction of the bile ducts, and impaired bile flow, as well as infection with Helicobacter species, such as Helicobacter bilis and Helicobacter hepaticus, in a manner that is similar to the reported role of Helicobacter pylori in distal gastric cancer. Population genetics, geographical and environmental factors, cholelithiasis, obesity, parity, and endemic infection with liver flukes have been identified as risk factors that influence the development of biliary tract tumors.



INTRODUCTION

Hepatobiliary cancers are highly lethal cancers that comprise a spectrum of invasive carcinomas originating in the liver [hepatocellular carcinoma (HCC)], the bile ducts intrahepatic cholangiocarcinoma (ICCA) and extrahepatic cholangiocarcinoma (ECCA), the gallbladder and the ampulla of Vater (collectively known as biliary tract cancers)[1]. These tumors account for approximately 13% of all annual cancer-related deaths worldwide and for 10%-20% of deaths from hepatobiliary malignancies[2]. The incidence and mortality of CCA has increased dramatically in North America and Europe in recent decades[3,4]. The CCA prevalence in North America is heterogeneously distributed among different racial and ethnic groups, with the highest age-adjusted prevalence in Hispanics (1.22/100000) and the lowest in African Americans (0.17-0.50/100000)[2]. In 2012, the worldwide incidence of hepatobiliary cancer was 960552 cases, resulting in 888330 deaths, whereas in the United States, an estimated 39880 cases of liver or other biliary tract cancer were diagnosed, including approximately 24312 deaths from liver cancer or CCA and 3845 deaths from gallbladder cancer[5].

In 2012, the worldwide mortality of gallbladder and biliary tract cancers was estimated to be 42813 cases, which represents 1.7% of deaths due to all cancers[5]. The regional prevalence of these tumors is highly variable; they are rare in most regions of Europe and North America but are exceptionally high in regions such as Chile, Thailand, Japan, central Europe, Northeastern India and Pakistan, with a 5-year survival rate of less than 10% in most cases. These contrasting rates are partially explained by differences in the prevalence of risk factors of this disease, such as population genetics, geographical and environmental factors, cholelithiasis, obesity, and parity, as well as endemic infection with liver flukes such as Opisthorchis viverrini or Clonorchis sinensis, which occurs in some regions of Asia[6-14]. Other important factors affecting hepatobiliary carcinogenesis include chronic inflammation, obstruction of the bile ducts, and impaired bile flow[15,16].

CCA is a devastating disease that displays a poor survival rate for which few therapeutic options are available. Although surgical resection has been considered to be the best option for localized CCA, local recurrence is very common because of persistent micro-metastatic disease in the regional lymph nodes or at the surgical margins[17]. Multiple studies have reported a survival rate of 20%-40% after 10 years for patients with resectable tumors localized to the hilar or distal third of the ductus choledochus, although the survival rate is decreased in advances stages of this disease. Previous reports have concluded that tumor extension directly correlates to the mortality rate[2,18-24]. Another therapeutic option is liver transplantation, for which the reported 5-year survival rate is 38%-50%[17,25]. For patients with unresectable disease, the placement of a self-expandable biliary stent is a widely accepted palliative procedure because it improves the quality of life by improving biliary drainage and by reducing the frequency of cholangitis and its complications[26,27].

The etiology of most of these tumors remains unknown, although the role of an infectious agent has been suggested, and studies of patients and animal models have indicated that Helicobacter species might play this role. Because clinical and experimental evidence for a role of Helicobacter spp. infection in the carcinogenesis of CCA has been accumulating in recent years, we considered it to be appropriate to review and summarize the relevant studies addressing the probable participation of Helicobacter species infection in the development of these tumors.

EVIDENCE SUGGESTING A ROLE OF HELICOBACTER SPP. INFECTION IN CCA

CCA displays the histological characteristic of adenocarcinoma of the bile-duct epithelial cells, and it has been suggested that the progression to cancer is similar to that which is observed in the intestinal type of gastric cancer: from hyperplasia to metaplasia, dysplasia, and, ultimately, the development of adenocarcinoma. In agreement with this proposal, it has been suggested that CCA may be caused by infection with Helicobacter species, such as Helicobacter bilis (H. bilis) and Helicobacter hepaticus (H. hepaticus), in a manner that is similar to the reported role of Helicobacter pylori (H. pylori) in distal gastric cancer; in fact, H. pylori was classified as a type I carcinogen by the International Agency for Research on Cancer (IARC)[9-11,28-30].

The enterohepatic Helicobacter spp., such as H. bilis, H. hepaticus, and H. cholecystus, have been isolated from the human gallbladder, liver tissue and bile juice and are suggested to express genes that facilitate their colonization in this hostile environment, which is highly osmotic and filled with biliary salts. The pathogenicity of H. bilis and H. hepaticus has been studied using various animals infected experimentally or naturally, and Helicobacter infection has been found to induce chronic active hepatitis, hepatocellular and biliary tract carcinomas, typhlocolitis, and lower bowel cancer in susceptible strains of inbred and genetically engineered mice[29-39].

H. hepaticus may establish a chronic infection in A/JCr mice by colonizing the lower bowel and, sporadically, the liver, particularly in the bile canaliculi[40]. This persistent infection caused chronic inflammation and necrosis in the hepatic parenchyma and portal triads, and infected animals exhibited oval cell, Kupffer cell, and Ito cell hyperplasia, hepatocytomegaly, and bile duct proliferation. Notably, hepatic adenomas were observed only in male animals. Thus, persistent H. hepaticus infection was associated with chronic proliferative hepatitis and hepatomas in exclusively male mice, suggesting that H. hepaticus may cause an increased risk for hepatic cancer. The constitutive androstane receptor (CAR) regulates the metabolic detoxification of endobiotics, and CAR knockout (KO) mice were used to examine tumor progression after H. hepaticus infection[41]. Although H. hepaticus infection induced chronic hepatitis in both wild type and CAR KO mice, the CAR KO mice exhibited increased numbers of liver lobes displaying dysplasia and neoplasia. The authors found that this enhanced tumor promotion was associated with decreased hepatic expression of the P450 enzymes Cyp2b10 and Cyp3a11, increased expression of Camp, and increased serum concentrations of chenodeoxycholic acid. Thus, liver tumor promotion in mice infected with H. hepaticus might be enhanced by impaired metabolic detoxification.

A study of aged hamsters detected liver infection with Helicobacter of the H. Bilis cluster[42], and the infected animals exhibited predominant portocentric and, to a lesser extent, perivenular fibrosis and nodular dysplasia. Lesion formation was associated with chronic active portal/interface and lobular inflammation, along with significant portal hepatitis. These results suggest the participation of H. bilis in hepatobiliary disease in both animals and humans. Enterohepatic Helicobacter infection might also contribute to the development of cholesterol gallstones and intrahepatic cholelithiasis, as suggested by recent studies of animal models[12,13,39,43-52].

The geographical distribution of H. bilis and other enteric Helicobacter species among the human population is unknown, although it is presumed that their prevalence is low in populations which display low risk for biliary duct malignancies and nonviral hepatitis. Similarly, its increased prevalence may contribute to the unexplained increase in biliary cancer incidence in specific regions of East and South Asia[53]. Because of the difficulty in culturing these Helicobacter spp., molecular techniques, such polymerase chain reaction (PCR) and sequencing, have become the methods of choice to identify these species in clinical specimens[54,55].

H. hepaticus

In 1994, spiral bacilli were isolated from the liver, the colon and the cecum of A/JCr mice and were identified as a novel species of Helicobacter termed H. hepaticus[40]. H. hepaticus is a microaerophilic, 1.5- to 5-µm-long, gram-negative, spiral-shaped bacterium that displays positive urease, catalase, and oxidase activities. It reduces nitrate, grows at 37 °C but not at 25 °C or 42 °C, is resistant to cephalothin and nalidixic acid but is sensitive to metronidazole, and as expected, exhibits resistance to bile. H. hepaticus must express genes that enable it to colonize the hostile, highly osmotic environment of the biliary duct. It lacks the genes expressed by H. pylori which are required for the colonization of the stomach, but it shares genes with Campylobacter spp. that may assist in the colonization of other enteric regions. The current availability of its genomic sequence provides investigators with the opportunity to examine the mechanisms underlying its tissue tropism and carcinogenesis[56]. H. hepaticus contains two flagella that are localized in a characteristic bipolar manner and possesses a flagellar sheath. These flagellar genes are closely related to those of H. pylori, and as in H. pylori, these genes are scattered throughout the chromosome, with few small clusters of functionally linked genes[57]. The flaA and flaB genes likely encode the major and minor flagellin subunits, which form the multimeric flagellar filament. One particular characteristic that is common to all of the H. hepaticus genomes analyzed is the presence of two identical copies of the flaA gene, including its promoter region (flaA_1 and flaA_2), which are present at two separate locations in the genome[58]. The expression of flagella and their regulatory genes are essential for the motility required for the intestinal colonization of H. hepaticus. Motility and chemotaxis could play a predominant role in enabling these bacteria to locate their appropriate intestinal habitat, particularly during the initial steps of colonization[59]. Cytolethal-distending toxin (CDT) is a multimeric cytotoxin that displays nuclease activity which may contribute to the exposure of endogenous antigens to the immune system following infection with H. hepaticus. CDTs are a family of proteins that cause cell cycle arrest and cellular distention, leading to the enlargement of both cells and nuclei followed by chromatin fragmentation[46,57,58]. These CDTs are composed of three subunits: a catalytic subunit, CdtB, which displays DNase I-like activity, and CdtA and CdtC, which bind to the cell membrane and deliver CdtB to the target cells[39,59]. The translocation of CdtB to the nucleus exerts genotoxic effects on the host DNA, triggering DNA repair cascades that induce cell cycle arrest and, ultimately, cell death. It was recently shown that H. hepaticus CDT plays a crucial role in promoting the progression of hepatitis to pre-malignant, dysplastic lesions via the activation of pro-inflammatory NF-κB and the increased proliferation of hepatocytes[60-63]. These results represent the first evidence that CDT displays carcinogenic potential in vivo. The urease enzyme is an important virulence factor involved in gastric colonization by H. pylori and H. mustale, although the contribution of urease to the development of H. hepaticus-associated disorders has yet to be demonstrated. Although urease expression by H. hepaticus is lower than that by H. pylori, its expression remains relatively high compared to that in other urease-positive pathogens. Active urease converts urea into ammonia and bicarbonate, and the produced ammonia mediates protection against acidic microenvironments and may also serve as a nitrogen source[64]. In contrast to H. pylori, the urease activity of H. hepaticus is not induced at acidic pH, and H. hepaticus does not grow or survive at pH 3.0. This finding suggests that the urease system is not a universal acid-resistance factor throughout ureolytic Helicobacter species and that its functions may differ between gastric and enterohepatic Helicobacter species; in fact, urease expression in H. hepaticus may respond to distinct environmental stimuli[64,65].

The complete genomic sequence of H. hepaticus strain ATCC51449 was published in 2003[56]. This sequence consists of 1779146 bp and shares certain characteristics with those from H. pylori, C. jejuni, and other enteric bacteria, such as V. cholerae and E. coli. Thus, approximately 50% of H. hepaticus open reading frames (ORFs) have orthologs in the sequences of the H. pylori 26695 and J99 strains, and 50.8% have orthologs in the sequence of C. jejuni NCTC 11168. It was also found that 821 H. hepaticus proteins have the same ortholog in both H. pylori and C. jejuni and that 109 H. hepaticus proteins have orthologs in both H. pylori genomes but not in C. jejuni. In addition, 130 H. hepaticus ORFs have an ortholog in C. jejuni but not in H. pylori[40,57,58,66,67]. The H. hepaticus genome contains one large region and many small regions that differ from the rest of the chromosome with respect to their G + C content, suggesting that these regions may have been generated via horizontal gene transfer. The largest region is a 71 Kb region termed genomic island 1 (HHGI1), which displays a G+C content of 33.2% and contains 70 ORFs (HH0233-HH0302). Most genes in HHGI1 encode hypothetical proteins, although it also encodes three proteins that are homologous to the structural components of the type IV secretion system, which is present in several H. pylori strains. HHGI1 also contains a gene that displays homology to V. cholerae hcp, which encodes a secreted protein that is co-regulated with V. cholerae hemolysin, and a gene cluster (HH244 to HH251) that displays significant homology to a cluster of genes of unknown function on the small chromosome of V. cholerae (VCA0107 to VCA0115) and on the Yersinia pestis genome. Additionally, the HH252 gene displays high similarity to the IcmF gene of V. cholerae and Legionella pneumophila, which encodes a protein that is involved in macrophage killing and bacterial conjugation. These data suggest that HHGI1 might be involved in the virulence of H. Hepaticus[49,68]. The genome also contains a katA gene (HH0043), which encodes a catalase that is homologous to that of Bordetella pertussis (65.95%) and H. pylori (62.5%)[49]. katA may confer protection against hydrogen peroxide and other ROS produced as a result of the inflammatory reaction induced by H. hepaticus infection. In fact, there is evidence that the levels of 8-oxo-guanine and lipid peroxidation are significantly increased in H. hepaticus-infected mice and that these oxidized macromolecules increase with the duration of infection[60,69].

The epidemiology of H. hepaticus infection is unknown; although the persistent infection of the lower intestine suggests that the fecal-oral route, particularly in mice considering their coprophagic habits, may transmit infection[40,48,70-72]. H. hepaticus is routinely cultured from the colon and the liver of mice suffering from various hepatobiliary disorders[31,40,67]. In contrast, attempts to culture these bacteria from specimens of patients suffering from various hepatobiliary disorders have largely been unsuccessful, although H. hepaticus infection has been detected via PCR or immunologic assays[6,13,14,28,29]. This difference suggests that characteristics such as the host hepatobiliary environment play a role in determining the adaptability and cultivability of this bacterium in vitro[72].

Chronic inflammation as the mediator of tumor promotion by H. hepaticus

Similar to H. pylori, H. hepaticus can establish a persistent infection in its host, inducing chronic inflammation that may represent the primary risk factor of the progression to carcinogenesis[58]. Nod2-deficient mice challenged with H. hepaticus secrete these bacteria in feces for longer periods than wild type mice, and an increased number of H. hepaticus was detected in the terminal ilea, suggesting that Nod2 protects the host from intestinal colonization. However, intestinal pathology was not detected following infection of either WT or Nod2-deficient mice. In contrast, in mice exhibiting altered immune function, the typhlocolitis/colitis lesion can be severe, leading to rectal prolapse, weight loss, and death[44,73]. Mice lacking IL-10 and infected with H. hepaticus developed spontaneous colitis, which was less severe when the mice were housed under specific-pathogen-free (SPF) conditions, suggesting enhanced susceptibility to pathogen-induced inflammation[69]. Inflammation was associated with a Th-1-related cytokine profile, including increased mRNA expression of IFN-γ and IL-17 in the colon. These results indicate that H. hepaticus infection induces Th1 and Th17 responses in mice exhibiting an impaired anti-inflammatory response or IL-10 deficiency[74]. Early studies demonstrated that in regulatory T-lymphocyte-deficient mice, H. hepaticus induced colitis and the progression to colonic carcinoma, including neoplastic peritoneal invasion[75,76]. Recombinase-activating gene-2-deficient (Rag2-/-) mice, which lack functional lymphocytes, represent a useful model of chronic inflammatory bowel disease. Infection of Rag2-/- mice with H. hepaticus led to the accumulation of macrophages and neutrophils and the up-regulation of inducible nitric oxide synthase (iNOS) expression in tissue, along with increased nitric oxide (NO) production in the colonic wall. Increasingly severe inflammation led to metaplasia, hyperplasia, dysplasia, and cancer. Concurrent administration of an iNOS inhibitor prevented NO production and abrogated this epithelial pathology and the onset of cancer. The presence of Gr-1+ cells and elevated TNF-α expression in the colon were also required for increased iNOS expression and for cancer development; alternatively, anti-inflammatory CD4+ Treg lymphocytes and IL-10 down-regulated the expression of TNF-α and iNOS and suppressed cancer. Collectively, these results confirm the essential roles of elevated NO production and TNF-α expression in carcinogenesis in this experimental model[77,78]. Chronic H. hepaticus infection is characterized by the infiltration of neutrophils and macrophages in the mouse cecum and liver, where they secrete ROS that may contribute to tumor promotion by damaging DNA in hepatocytes or intestinal epithelial cells, as suggested by the increased levels of oxidized nucleoside 8-hydroxydeoxyguanosine (8-oxo-dG) in liver DNA after H. hepaticus infection[79]. Orogastric infection of C57BL/6 ApcMin/+ (Min) mice with H. hepaticus rapidly promoted extraintestinal tumors in mammary tissue[45], and breast cancer has been suggested to arise from intestinal bacteria-induced inflammation. The pro-inflammatory cytokine TNF-α plays a prominent role in this tumor model, as it participates in cancer progression in the bowel, the liver, and other sites in mice[49,80,81].

H. bilis

H. bilis is a gram-negative, opportunistic, nonsporulating, microaerophilic, fusiform bacteria that measures 0.5 nm in width and 4 to 5 nm in length. This bacterium produces 3 to 14 bipolar-sheathed flagella and periplasmic fibers that surround the cell. Comparative analysis of its 16S rRNA sequences revealed that it displays a similarity of 98% to H. cinaedi, 97.4% to H. hepaticus and 93.4% to H. pylori[30,42,82,83]. It was originally isolated from the colonic crypts, the cecum, the bile and the liver of mice with hepatitis[30], and because it has not been found in the stomach, it is also referred to as non-gastric Helicobacter. H. bilis is an enterohepatic Helicobacter species which is endemic in most mouse facilities that may induce disease in susceptible animals[84]. H. bilis infection has been associated with an increased incidence of typhlocolitis[85,86] inflammatory bowel disease[33], hepatitis[30], and cholecystitis[85].

In humans, H. bilis has been associated with chronic liver disease[55], biliary tract and gallbladder cancer[87,88], chronic diarrhea[89], and pyoderma gangrenosum-like ulcers[90]. H. bilis has not been cultured from human specimens, and it was first detected via PCR in the gallbladder and the bile of Chilean patients with chronic cholecystitis[91]. H. bilis antibodies have been identified in sera from patients with chronic liver disease and cirrhosis[88], and H. bilis infection has also been demonstrated via PCR in patients with benign and malignant liver neoplasm-like hepatoma and HCC[36,37].

In mice, H. bilis is known to be responsible for chronic hepatitis and hepatocellular tumors and may play a major role in cholesterol gallstone formation[30,54,87,88,92,93]. However, a direct association between H. bilis and bile duct cancer has yet to be established.

Due to the presence of enterohepatic Helicobacter in gallbladder carcinoma, this species has been considered as a cofactor that contributes to biliary carcinogenesis, although the mechanisms responsible for the processes that lead to cancer remain unclear[94]. Enterohepatic Helicobacter infection has been suggested to participate in the development of cholesterol gallstones and intrahepatic cholelithiasis, which may subsequently lead to carcinogenesis[54,95]. Its promotion of stone formation has been suggested to be due to its serving as a foreign body nest around which the stone develops, likely by producing hydrolyzing enzymes or nucleating proteins, such as immunoglobulins[54,96]. The risk for lithogenicity might also be associated with the modulation of enterohepatic cycling of conjugated bile acid, including the transit time through the gut[54,82,87,97-101]. It was recently confirmed that H. bilis infection activates NF-κB in bile duct carcinoma cells and increases the expression of the angiogenic factor VEGF, elevating the angiogenic potential of these cells via CRE-binding protein[83,97]. Recently, gamma-glutamyl transpeptidase (gGT) has been reported as a novel H. bilis virulence factor (HBgGT). The H. bilis genome encodes two putative gGT sequences, one of which was found to be functionally active, inhibiting the activity of T cells and suppressing the proliferation of epithelial AGS cells, in a manner that is similar to H. pylori gGT (HPgGT)[102]. HBgGT has been implicated as an important regulator of inflammation in H. bilis-infected intestinal epithelial cells that may be responsible for the inflammatory disorders observed in experimental animal models[103].

Clinical evidence for the role of Helicobacter spp. in biliary diseases

Currently, multiple human clinical studies have associated Helicobacter spp. infection with biliary diseases. H. pylori has been suggested to infect the biliary tract and cause cancer. Although H. pylori DNA has been detected in human bile, whether this organism rarely colonizes the bile duct epithelium was not clearly documented. In this infection, carcinogenesis includes the activity of the virulence factor CagA, an oncoprotein that interferes with signal transduction pathways, and the host response to Helicobacter antigens in the form of cytokines and other inflammatory mediators[101,104,105]. However, based on the currently available evidence, inflammation followed by epithelial cell proliferation and interference with cell cycle via alterations in signal transduction appear to be the most plausible explanations of carcinogenesis[42,57,97,98]. As different Helicobacter spp. have been successfully identified in the biliary system, these organisms have been suggested to participate in the development of malignant and benign biliary diseases. A meta-analysis based on a systematic review of 18 studies published between 1998 and 2011 revealed significantly higher frequencies of H. pylori and H. hepaticus infection in patients with lithiasis[99]. Other studies have focused on patients with chronic liver disease, particularly cholelithiasis, or cholecystitis and demonstrated a significantly higher prevalence of H. hepaticus in samples from these patients than in samples from patients with other diseases[105,106]. Increased concentrations of anti-H. Hepaticus antibodies were found in sera from patients with liver disease compared to those suffering from other diseases, especially HBV- and/or HCV-infected patients[13].

Based on a meta-analyses of 10 case-control studies that contained a cumulative sample size of 205 cases, 115 cases (56%) were positive for Helicobacter spp. infection, whereas among the 263 controls, 53 (20%) were positive for Helicobacter spp. infection. The authors concluded that there is sufficient evidence to suggest a role of Helicobacter spp. in hepatobiliary tract cancers, although there was some variation in the results from different regions of the world and in the methods used to detect Helicobacter[96]. Another meta-analysis explored the association between the Helicobacter species H. pylori, H. bilis, H. hepaticus, and H. ganmani and biliary tract cancer. The presence of Helicobacter spp. was determined via PCR or immunohistochemical analysis of specimens from bile and biliary tissues. A significantly higher pooled infection rate of Helicobacter spp. (predominantly H. pylori and H. bilis) was observed in the biliary tract of the cancer group (P = 0.0001) and in the benign biliary disease group (P = 0.0001) than in the asymptomatic group[53]. In another case-control study of patients with HCC, the authors searched for Helicobacter spp. in liver tissue. PCR detected Helicobacter spp. in 60.7% of the cases, whereas the controls displayed no infection (P < 0.01), suggesting that Helicobacter spp. colonized the liver tissues of HCC patients[107].

CONCLUSION

The high mortality rates observed in biliary tract cancer demands further investigation to elucidate the etiology and carcinogenesis of these tumors. Population genetics, geographical and environmental factors, cholelithiasis, obesity, parity, and endemic infection with liver flukes (Opisthorchis viverrini or Clonorchis sinensis), particularly in certain regions of Asia, have been identified as risk factors that influence the development of biliary tract tumors. Other important factors in the carcinogenesis of these tumors include chronic inflammation, obstruction of the bile ducts, and impaired bile flow. CCA has been suggested to be caused by infection with Helicobacter spp., such as H. bilis and H. hepaticus, in a manner that is similar to the reported role of H. pylori in distal gastric cancer. The pathogenicity of H. bilis and H. hepaticus has been studied in various animals infected experimentally or naturally, in which Helicobacter infection has been found to cause chronic active hepatitis, hepatocellular and biliary tract carcinoma, typhlocolitis, and lower bowel cancer. These infections can also promote the development of cholesterol gallstones and intrahepatic cholelithiasis, which is another risk factor for CCA. The availability of the entire genomic sequence of H. hepaticus has facilitated the identification of potential virulence factors and the elucidation of their mechanisms of action, including their participation in inflammation, cell proliferation and interference with the cell cycle.

Multiple clinical studies have identified different Helicobacter spp. in the biliary tract, including H. pylori, and have associated these infections with the development of benign and malignant biliary diseases. The geographical distribution of enteric Helicobacter species has been suggested to be low in populations that display a low incidence of biliary duct malignancies. Because of the difficulty in culturing these Helicobacter species, molecular methods, such as PCR and sequencing, and immunologic assays have become the methods of choice for diagnosis. However, clinical studies of benign or malignant biliary tract diseases revealed remarkable variability in the methods and the findings, and the use of uniform and validated techniques is needed.

Because the biliary tract is only accessible via invasive procedures or surgery, it is necessary to develop PCR assay protocols, more suitable antigens for immunohistochemistry, and easy and effective serological methods for the early detection of Helicobacter spp. to help reduce the incidence of biliary diseases, as well as the morbidity and mortality of this group of patients.

Footnotes

P- Reviewer: Christodoulidis G, Koch TR, Ozen H S- Editor: Qi Y L- Editor: A E- Editor: Wang CH

References
1.  Guitron-Cantu A. Endoscopic treatment in malignant obstructive jaundice: In Endoscopic procedures in Gastroenterology. 2nd ed. Endoscopic Procedures in Gastroenterology. Mexico: Ed Panamericana 2009; 489-504.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Shaib Y, El-Serag HB. The epidemiology of cholangiocarcinoma. Semin Liver Dis. 2004;24:115-125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 839]  [Cited by in F6Publishing: 815]  [Article Influence: 40.8]  [Reference Citation Analysis (0)]
3.  Patel T. Increasing incidence and mortality of primary intrahepatic cholangiocarcinoma in the United States. Hepatology. 2001;33:1353-1357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 773]  [Cited by in F6Publishing: 756]  [Article Influence: 32.9]  [Reference Citation Analysis (0)]
4.  Shaib YH, Davila JA, McGlynn K, El-Serag HB. Rising incidence of intrahepatic cholangiocarcinoma in the United States: a true increase? J Hepatol. 2004;40:472-477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 514]  [Cited by in F6Publishing: 513]  [Article Influence: 25.7]  [Reference Citation Analysis (0)]
5.  International Agency For Research On Cancer. 2012.  Available from: http://globocan.IARC.fr.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Murakami K, Takahashi R, Ono M, Watanabe K, Okimoto T, Kodama M, Abe D, Kimura M, Fujioka T. Serodiagnosis of Helicobacter hepaticus infection in patients with liver and gastrointestinal diseases: western blot analysis and ELISA using a highly specific monoclonal antibody for H. hepaticus antigen. J Gastroenterol. 2011;46:1120-1126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
7.  Andia ME, Hsing AW, Andreotti G, Ferreccio C. Geographic variation of gallbladder cancer mortality and risk factors in Chile: a population-based ecologic study. Int J Cancer. 2008;123:1411-1416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 53]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
8.  Jongsuksuntigul P, Imsomboon T. Opisthorchiasis control in Thailand. Acta Trop. 2003;88:229-232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 183]  [Cited by in F6Publishing: 190]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
9.  Marshall BJ, Warren JR. Unidentified curved bacilli in the stomach of patients with gastritis and peptic ulceration. Lancet. 1984;1:1311-1315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3302]  [Cited by in F6Publishing: 3116]  [Article Influence: 77.9]  [Reference Citation Analysis (0)]
10.  Hattori T, Sugihara H. The pathological sequence in the development of gastric cancer: I. Scand J Gastroenterol Suppl. 1996;214:34-35; discussion 40-43.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Schistosomes , liver flukes and Helicobacter pylori. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Lyon, 7-14 June 1994. IARC Monogr Eval Carcinog Risks Hum. 1994;61:1-241.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Ward JM, Anver DC, Haines LM. Chronic active hepatitis of unknown origin in mice from a large research facility. Vet Pathol. 1993;30:A4769.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Fox JG. The non-H pylori helicobacters: their expanding role in gastrointestinal and systemic diseases. Gut. 2002;50:273-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 220]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
14.  Rossi M, Hänninen ML. Helicobacter spp. other than H. pylori. Helicobacter. 2012;17 Suppl 1:56-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
15.  Holzinger F, Z‘graggen K, Büchler MW. Mechanisms of biliary carcinogenesis: a pathogenetic multi-stage cascade towards cholangiocarcinoma. Ann Oncol. 1999;10 Suppl 4:122-126.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Gores GJ. Cholangiocarcinoma: current concepts and insights. Hepatology. 2003;37:961-969.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 195]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
17.  Meza-Junco J, Montano-Loza AJ, Ma M, Wong W, Sawyer MB, Bain VG. Cholangiocarcinoma: has there been any progress? Can J Gastroenterol. 2010;24:52-57.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Ojima H, Kanai Y, Iwasaki M, Hiraoka N, Shimada K, Sano T, Sakamoto Y, Esaki M, Kosuge T, Sakamoto M. Intraductal carcinoma component as a favorable prognostic factor in biliary tract carcinoma. Cancer Sci. 2009;100:62-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
19.  Borghero Y, Crane CH, Szklaruk J, Oyarzo M, Curley S, Pisters PW, Evans D, Abdalla EK, Thomas MB, Das P. Extrahepatic bile duct adenocarcinoma: patients at high-risk for local recurrence treated with surgery and adjuvant chemoradiation have an equivalent overall survival to patients with standard-risk treated with surgery alone. Ann Surg Oncol. 2008;15:3147-3156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 78]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
20.  Lim KH, Oh DY, Chie EK, Jang JY, Im SA, Kim TY, Kim SW, Ha SW, Bang YJ. Adjuvant concurrent chemoradiation therapy (CCRT) alone versus CCRT followed by adjuvant chemotherapy: which is better in patients with radically resected extrahepatic biliary tract cancer?: a non-randomized, single center study. BMC Cancer. 2009;9:345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 36]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
21.  Zaydfudim VM, Clark CJ, Kendrick ML, Que FG, Reid-Lombardo KM, Donohue JH, Farnell MB, Nagorney DM. Correlation of staging systems to survival in patients with resected hilar cholangiocarcinoma. Am J Surg. 2013;206:159-165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
22.  Nagahashi M, Shirai Y, Wakai T, Sakata J, Ajioka Y, Nomura T, Tsuchiya Y, Hatakeyama K. Depth of invasion determines the postresectional prognosis for patients with T1 extrahepatic cholangiocarcinoma. Cancer. 2010;116:400-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 17]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
23.  Shindoh J, Vauthey JN. Staging of biliary tract and primary liver tumors. Surg Oncol Clin N Am. 2014;23:313-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 8]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
24.  Ebata T, Kosuge T, Hirano S, Unno M, Yamamoto M, Miyazaki M, Kokudo N, Miyagawa S, Takada T, Nagino M. Proposal to modify the International Union Against Cancer staging system for perihilar cholangiocarcinomas. Br J Surg. 2014;101:79-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 145]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
25.  Robles R, Sánchez-Bueno F, Ramírez P, Brusadin R, Parrilla P. Liver transplantation for hilar cholangiocarcinoma. World J Gastroenterol. 2013;19:9209-9215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 19]  [Cited by in F6Publishing: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
26.  Isayama H, Nakai Y, Kogure H, Yamamoto N, Koike K. Biliary self-expandable metallic stent for unresectable malignant distal biliary obstruction: which is better: covered or uncovered? Dig Endosc. 2013;25 Suppl 2:71-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 31]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
27.  Güitrón-Cantú A, Adalid-Martínez R, Gutiérrez-Bermúdez JA. [Self-expandable metallic biliary endoprosthesis in malignant obstructive jaundice]. Rev Gastroenterol Mex. 2005;70:247-252.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Roe IH, Kim JT, Lee HS, Lee JH. Detection of Helicobacter DNA in bile from bile duct diseases. J Korean Med Sci. 1999;14:182-186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
29.  Abu Al-Soud W, Stenram U, Ljungh A, Tranberg KG, Nilsson HO, Wadström T. DNA of Helicobacter spp. and common gut bacteria in primary liver carcinoma. Dig Liver Dis. 2008;40:126-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
30.  Fox JG, Dewhirst FE, Tully JG, Paster BJ, Yan L, Taylor NS, Collins MJ, Gorelick PL, Ward JM. Helicobacter hepaticus sp. nov., a microaerophilic bacterium isolated from livers and intestinal mucosal scrapings from mice. J Clin Microbiol. 1994;32:1238-1245.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Fox JG, Lee A. The role of Helicobacter species in newly recognized gastrointestinal tract diseases of animals. Lab Anim Sci. 1997;47:222-255.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Fox JG, Rogers AB, Whary MT, Taylor NS, Xu S, Feng Y, Keys S. Helicobacter bilis-associated hepatitis in outbred mice. Comp Med. 2004;54:571-577.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Shomer NH, Dangler CA, Schrenzel MD, Fox JG. Helicobacter bilis-induced inflammatory bowel disease in scid mice with defined flora. Infect Immun. 1997;65:4858-4864.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Casswall TH, Németh A, Nilsson I, Wadström T, Nilsson HO. Helicobacter species DNA in liver and gastric tissues in children and adolescents with chronic liver disease. Scand J Gastroenterol. 2010;45:160-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
35.  Kobayashi T, Harada K, Miwa K, Nakanuma Y. Helicobacter genus DNA fragments are commonly detectable in bile from patients with extrahepatic biliary diseases and associated with their pathogenesis. Dig Dis Sci. 2005;50:862-867.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 38]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
36.  Kosaka T, Tajima Y, Kuroki T, Mishima T, Adachi T, Tsuneoka N, Fukuda K, Kanematsu T. Helicobacter bilis colonization of the biliary system in patients with pancreaticobiliary maljunction. Br J Surg. 2010;97:544-549.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
37.  Fukuda K, Kuroki T, Tajima Y, Tsuneoka N, Kitajima T, Matsuzaki S, Furui J, Kanematsu T. Comparative analysis of Helicobacter DNAs and biliary pathology in patients with and without hepatobiliary cancer. Carcinogenesis. 2002;23:1927-1931.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 60]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
38.  Fowsantear W, Argo E, Pattinson C, Cash P. Comparative proteomics of Helicobacter species: the discrimination of gastric and enterohepatic Helicobacter species. J Proteomics. 2014;97:245-255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
39.  Young VB, Knox KA, Schauer DB. Cytolethal distending toxin sequence and activity in the enterohepatic pathogen Helicobacter hepaticus. Infect Immun. 2000;68:184-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 119]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
40.  Fox JG, Li X, Yan L, Cahill RJ, Hurley R, Lewis R, Murphy JC. Chronic proliferative hepatitis in A/JCr mice associated with persistent Helicobacter hepaticus infection: a model of helicobacter-induced carcinogenesis. Infect Immun. 1996;64:1548-1558.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  García A, Zeng Y, Muthupalani S, Ge Z, Potter A, Mobley MW, Boussahmain C, Feng Y, Wishnok JS, Fox JG. Helicobacter hepaticus--induced liver tumor promotion is associated with increased serum bile acid and a persistent microbial-induced immune response. Cancer Res. 2011;71:2529-2540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
42.  Fox JG, Shen Z, Muthupalani S, Rogers AR, Kirchain SM, Dewhirst FE. Chronic hepatitis, hepatic dysplasia, fibrosis, and biliary hyperplasia in hamsters naturally infected with a novel Helicobacter classified in the H. bilis cluster. J Clin Microbiol. 2009;47:3673-3681.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 38]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
43.  Petnicki-Ocwieja T, Hrncir T, Liu YJ, Biswas A, Hudcovic T, Tlaskalova-Hogenova H, Kobayashi KS. Nod2 is required for the regulation of commensal microbiota in the intestine. Proc Natl Acad Sci USA. 2009;106:15813-15818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 421]  [Cited by in F6Publishing: 444]  [Article Influence: 29.6]  [Reference Citation Analysis (0)]
44.  Mandell L, Moran AP, Cocchiarella A, Houghton J, Taylor N, Fox JG, Wang TC, Kurt-Jones EA. Intact gram-negative Helicobacter pylori, Helicobacter felis, and Helicobacter hepaticus bacteria activate innate immunity via toll-like receptor 2 but not toll-like receptor 4. Infect Immun. 2004;72:6446-6454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 171]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
45.  Rao VP, Poutahidis T, Ge Z, Nambiar PR, Boussahmain C, Wang YY, Horwitz BH, Fox JG, Erdman SE. Innate immune inflammatory response against enteric bacteria Helicobacter hepaticus induces mammary adenocarcinoma in mice. Cancer Res. 2006;66:7395-7400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 145]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
46.  Rudi J, Rudy A, Maiwald M, Stremmel W. Helicobacter sp. are not detectable in bile from German patients with biliary disease. Gastroenterology. 1999;116:1016-1017.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
47.  Rice JM. Helicobacter hepaticus, a recently recognized bacterial pathogen, associated with chronic hepatitis and hepatocellular neoplasia in laboratory mice. Emerg Infect Dis. 1995;1:129-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
48.  Erdman SE, Poutahidis T, Tomczak M, Rogers AB, Cormier K, Plank B, Horwitz BH, Fox JG. CD4+ CD25+ regulatory T lymphocytes inhibit microbially induced colon cancer in Rag2-deficient mice. Am J Pathol. 2003;162:691-702.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in F6Publishing: 239]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
49.  Rao VP, Poutahidis T, Fox JG, Erdman SE. Breast cancer: should gastrointestinal bacteria be on our radar screen? Cancer Res. 2007;67:847-850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 55]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
50.  Veijola L, Nilsson I, Halme L, Al-Soud WA, Mäkinen J, Ljungh A, Rautelin H. Detection of Helicobacter species in chronic liver disease and chronic inflammatory bowel disease. Ann Med. 2007;39:554-560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
51.  Chen W, Li D, Cannan RJ, Stubbs RS. Common presence of Helicobacter DNA in the gallbladder of patients with gallstone diseases and controls. Dig Liver Dis. 2003;35:237-243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 42]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
52.  Nilsson HO, Taneera J, Castedal M, Glatz E, Olsson R, Wadström T. 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. 2000;38:1072-1076.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Zhou D, Wang JD, Weng MZ, Zhang Y, Wang XF, Gong W, Quan ZW. Infections of Helicobacter spp. in the biliary system are associated with biliary tract cancer: a meta-analysis. Eur J Gastroenterol Hepatol. 2013;25:447-454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 37]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
54.  Maurer KJ, Ihrig MM, Rogers AB, Ng V, Bouchard G, Leonard MR, Carey MC, Fox JG. Identification of cholelithogenic enterohepatic helicobacter species and their role in murine cholesterol gallstone formation. Gastroenterology. 2005;128:1023-1033.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 109]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
55.  Vorobjova T, Nilsson I, Terjajev S, Granholm M, Lyyra M, Porkka T, Prükk T, Salupere R, Maaroos HI, Wadström T. 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. 2006;38:171-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 22]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
56.  Suerbaum S, Josenhans C, Sterzenbach T, Drescher B, Brandt P, Bell M, Droge M, Fartmann B, Fischer HP, Ge Z. The complete genome sequence of the carcinogenic bacterium Helicobacter hepaticus. Proc Natl Acad Sci USA. 2003;100:7901-7906.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 192]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
57.  Chien CC, Taylor NS, Ge Z, Schauer DB, Young VB, Fox JG. Identification of cdtB homologues and cytolethal distending toxin activity in enterohepatic Helicobacter spp. J Med Microbiol. 2000;49:525-534.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Alyamani EJ, Brandt P, Pena JA, Major AM, Fox JG, Suerbaum S, Versalovic J. Helicobacter hepaticus catalase shares surface-predicted epitopes with mammalian catalases. Microbiology. 2007;153:1006-1016.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
59.  Sterzenbach T, Bartonickova L, Behrens W, Brenneke B, Schulze J, Kops F, Chin EY, Katzowitsch E, Schauer DB, Fox JG. Role of the Helicobacter hepaticus flagellar sigma factor FliA in gene regulation and murine colonization. J Bacteriol. 2008;190:6398-6408.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
60.  Ge Z, Feng Y, Whary MT, Nambiar PR, Xu S, Ng V, Taylor NS, Fox JG. Cytolethal distending toxin is essential for Helicobacter hepaticus colonization in outbred Swiss Webster mice. Infect Immun. 2005;73:3559-3567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 92]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
61.  Smith JL, Bayles DO. The contribution of cytolethal distending toxin to bacterial pathogenesis. Crit Rev Microbiol. 2006;32:227-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 140]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
62.  Ge Z, Schauer DB, Fox JG. In vivo virulence properties of bacterial cytolethal-distending toxin. Cell Microbiol. 2008;10:1599-1607.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 78]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
63.  Liyanage NP, Manthey KC, Dassanayake RP, Kuszynski CA, Oakley GG, Duhamel GE. Helicobacter hepaticus cytolethal distending toxin causes cell death in intestinal epithelial cells via mitochondrial apoptotic pathway. Helicobacter. 2010;15:98-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
64.  Belzer C, van Schendel BA, Kuipers EJ, Kusters JG, van Vliet AH. Iron-responsive repression of urease expression in Helicobacter hepaticus is mediated by the transcriptional regulator Fur. Infect Immun. 2007;75:745-752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
65.  Williams CL, Preston T, Hossack M, Slater C, McColl KE. Helicobacter pylori utilises urea for amino acid synthesis. FEMS Immunol Med Microbiol. 1996;13:87-94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 77]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
66.  Ward JM, Fox JG, Anver MR, Haines DC, George CV, Collins MJ, Gorelick PL, Nagashima K, Gonda MA, Gilden RV. Chronic active hepatitis and associated liver tumors in mice caused by a persistent bacterial infection with a novel Helicobacter species. J Natl Cancer Inst. 1994;86:1222-1227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 299]  [Cited by in F6Publishing: 272]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
67.  Boutin SR, Shen Z, Rogers AB, Feng Y, Ge Z, Xu S, Sterzenbach T, Josenhans C, Schauer DB, Suerbaum S. Different Helicobacter hepaticus strains with variable genomic content induce various degrees of hepatitis. Infect Immun. 2005;73:8449-8452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 28]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
68.  Hong Y, Wang G, Maier RJ. A Helicobacter hepaticus catalase mutant is hypersensitive to oxidative stress and suffers increased DNA damage. J Med Microbiol. 2007;56:557-562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
69.  Kullberg MC, Ward JM, Gorelick PL, Caspar P, Hieny S, Cheever A, Jankovic D, Sher A. Helicobacter hepaticus triggers colitis in specific-pathogen-free interleukin-10 (IL-10)-deficient mice through an IL-12- and gamma interferon-dependent mechanism. Infect Immun. 1998;66:5157-5166.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Whary MT, Morgan TJ, Dangler CA, Gaudes KJ, Taylor NS, Fox JG. Chronic active hepatitis induced by Helicobacter hepaticus in the A/JCr mouse is associated with a Th1 cell-mediated immune response. Infect Immun. 1998;66:3142-3148.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Cahill RJ, Foltz CJ, Fox JG, Dangler CA, Powrie F, Schauer DB. Inflammatory bowel disease: an immunity-mediated condition triggered by bacterial infection with Helicobacter hepaticus. Infect Immun. 1997;65:3126-3131.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Okoli AS, Raftery MJ, Mendz GL. Effects of human and porcine bile on the proteome of Helicobacter hepaticus. Proteome Sci. 2012;10:27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
73.  Fritz JH, Ferrero RL, Philpott DJ, Girardin SE. Nod-like proteins in immunity, inflammation and disease. Nat Immunol. 2006;7:1250-1257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 648]  [Cited by in F6Publishing: 635]  [Article Influence: 35.3]  [Reference Citation Analysis (0)]
74.  Kullberg MC, Rothfuchs AG, Jankovic D, Caspar P, Wynn TA, Gorelick PL, Cheever AW, Sher A. Helicobacter hepaticus-induced colitis in interleukin-10-deficient mice: cytokine requirements for the induction and maintenance of intestinal inflammation. Infect Immun. 2001;69:4232-4241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 120]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
75.  Erdman SE, Poutahidis T. Cancer inflammation and regulatory T cells. Int J Cancer. 2010;127:768-779.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 41]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
76.  Erdman SE, Poutahidis T, Rogers AB, Tomszaki M, Horwitz B, Fox JG. Susceptibility of three strains of Rag2-deficient mice to Helicobacter hepaticus induced colitis and colorectal cancer. Gastroenterology. 2003;124 Suppl 1:A340.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Erdman SE, Rao VP, Poutahidis T, Rogers AB, Taylor CL, Jackson EA, Ge Z, Lee CW, Schauer DB, Wogan GN. Nitric oxide and TNF-alpha trigger colonic inflammation and carcinogenesis in Helicobacter hepaticus-infected, Rag2-deficient mice. Proc Natl Acad Sci USA. 2009;106:1027-1032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 151]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
78.  Poutahidis T, Haigis KM, Rao VP, Nambiar PR, Taylor CL, Ge Z, Watanabe K, Davidson A, Horwitz BH, Fox JG. Rapid reversal of interleukin-6-dependent epithelial invasion in a mouse model of microbially induced colon carcinoma. Carcinogenesis. 2007;28:2614-2623.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 52]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
79.  Sipowicz MA, Chomarat P, Diwan BA, Anver MA, Awasthi YC, Ward JM, Rice JM, Kasprzak KS, Wild CP, Anderson LM. Increased oxidative DNA damage and hepatocyte overexpression of specific cytochrome P450 isoforms in hepatitis of mice infected with Helicobacter hepaticus. Am J Pathol. 1997;151:933-941.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Rao VP, Poutahidis T, Ge Z, Nambiar PR, Horwitz BH, Fox JG, Erdman SE. Proinflammatory CD4+ CD45RB(hi) lymphocytes promote mammary and intestinal carcinogenesis in Apc(Min/+) mice. Cancer Res. 2006;66:57-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 73]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
81.  Erdman SE, Rao VP, Olipitz W, Taylor CL, Jackson EA, Levkovich T, Lee CW, Horwitz BH, Fox JG, Ge Z. Unifying roles for regulatory T cells and inflammation in cancer. Int J Cancer. 2010;126:1651-1665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 50]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
82.  Taniguchi T, Sekiya A, Higa M, Saeki Y, Umeki K, Okayama A, Hayashi T, Misawa N. Rapid identification and subtyping of Helicobacter cinaedi strains by intact-cell mass spectrometry profiling with the use of matrix-assisted laser desorption ionization-time of flight mass spectrometry. J Clin Microbiol. 2014;52:95-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 28]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
83.  Takayama S, Takahashi H, Matsuo Y, Okada Y, Takeyama H. Effect of Helicobacter bilis infection on human bile duct cancer cells. Dig Dis Sci. 2010;55:1905-1910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
84.  Fox JG. Helicobacter bilis: bacterial provocateur orchestrates host immune responses to commensal flora in a model of inflammatory bowel disease. Gut. 2007;56:898-900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
85.  Jergens AE, Wilson-Welder JH, Dorn A, Henderson A, Liu Z, Evans RB, Hostetter J, Wannemuehler MJ. Helicobacter bilis triggers persistent immune reactivity to antigens derived from the commensal bacteria in gnotobiotic C3H/HeN mice. Gut. 2007;56:934-940.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 90]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
86.  Liu Z, Ramer-Tait AE, Henderson AL, Demirkale CY, Nettleton D, Wang C, Hostetter JM, Jergens AE, Wannemuehler MJ. Helicobacter bilis colonization enhances susceptibility to Typhlocolitis following an inflammatory trigger. Dig Dis Sci. 2011;56:2838-2848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
87.  Murata H, Tsuji S, Tsujii M, Fu HY, Tanimura H, Tsujimoto M, Matsuura N, Kawano S, Hori M. Helicobacter bilis infection in biliary tract cancer. Aliment Pharmacol Ther. 2004;20 Suppl 1:90-94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 59]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
88.  Matsukura N, Yokomuro S, Yamada S, Tajiri T, Sundo T, Hadama T, Kamiya S, Naito Z, Fox JG. 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. 2002;93:842-847.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 120]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
89.  Romero S, Archer JR, Hamacher ME, Bologna SM, Schell RF. Case report of an unclassified microaerophilic bacterium associated with gastroenteritis. J Clin Microbiol. 1988;26:142-143.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Murray PR, Jain A, Uzel G, Ranken R, Ivy C, Blyn LB, Ecker DJ, Sampath R, Lee CC, Turner ML. Pyoderma gangrenosum-like ulcer in a patient with X-linked agammaglobulinemia: identification of Helicobacter bilis by mass spectrometry analysis. Arch Dermatol. 2010;146:523-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
91.  Fox JG, Dewhirst FE, Shen Z, Feng Y, Taylor NS, Paster BJ, Ericson RL, Lau CN, Correa P, Araya JC. Hepatic Helicobacter species identified in bile and gallbladder tissue from Chileans with chronic cholecystitis. Gastroenterology. 1998;114:755-763.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 355]  [Cited by in F6Publishing: 334]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
92.  Pellicano R, Mazzaferro V, Grigioni WF, Cutufia MA, Fagoonee S, Silengo L, Rizzetto M, Ponzetto A. Helicobacter species sequences in liver samples from patients with and without hepatocellular carcinoma. World J Gastroenterol. 2004;10:598-601.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Silva CP, Pereira-Lima JC, Oliveira AG, Guerra JB, Marques DL, Sarmanho L, Cabral MM, Queiroz DM. Association of the presence of Helicobacter in gallbladder tissue with cholelithiasis and cholecystitis. J Clin Microbiol. 2003;41:5615-5618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 62]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
94.  Fox JG. Other helicobacters involved in human diseases. Acta Gastroenterol Belg. 2002;65:24-32.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Maurer KJ, Rao VP, Ge Z, Rogers AB, Oura TJ, Carey MC, Fox JG. T-cell function is critical for murine cholesterol gallstone formation. Gastroenterology. 2007;133:1304-1315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 49]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
96.  Pandey M, Shukla M. Helicobacter species are associated with possible increase in risk of hepatobiliary tract cancers. Surg Oncol. 2009;18:51-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
97.  Tak PP, Firestein GS. NF-kappaB: a key role in inflammatory diseases. J Clin Invest. 2001;107:7-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3011]  [Cited by in F6Publishing: 2989]  [Article Influence: 130.0]  [Reference Citation Analysis (0)]
98.  Fox JG, Ge Z, Whary MT, Erdman SE, Horwitz BH. Helicobacter hepaticus infection in mice: models for understanding lower bowel inflammation and cancer. Mucosal Immunol. 2011;4:22-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 139]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
99.  Zhou D, Zhang Y, Gong W, Mohamed SO, Ogbomo H, Wang X, Liu Y, Quan Z. Are Helicobacter pylori and other Helicobacter species infection associated with human biliary lithiasis? A meta-analysis. PLoS One. 2011;6:e27390.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Randi G, Franceschi S, La Vecchia C. Gallbladder cancer worldwide: geographical distribution and risk factors. Int J Cancer. 2006;118:1591-1602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 523]  [Cited by in F6Publishing: 525]  [Article Influence: 29.2]  [Reference Citation Analysis (0)]
101.  Pandey M. Helicobacter species are associated with possible increase in risk of biliary lithiasis and benign biliary diseases. World J Surg Oncol. 2007;5:94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 11]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
102.  Rossi M, Bolz C, Revez J, Javed S, El-Najjar N, Anderl F, Hyytiäinen H, Vuorela P, Gerhard M, Hänninen ML. Evidence for conserved function of γ-glutamyltranspeptidase in Helicobacter genus. PLoS One. 2012;7:e30543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 449]  [Reference Citation Analysis (0)]
103.  Javed S, Mejías-Luque R, Kalali B, Bolz C, Gerhard M. Helicobacter bilis gamma-glutamyltranspeptidase enhances inflammatory stress response via oxidative stress in colon epithelial cells. PLoS One. 2013;8:e73160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
104.  Peek RM, Crabtree JE. Helicobacter infection and gastric neoplasia. J Pathol. 2006;208:233-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 368]  [Cited by in F6Publishing: 417]  [Article Influence: 21.9]  [Reference Citation Analysis (0)]
105.  Myung SJ, Kim MH, Shim KN, Kim YS, Kim EO, Kim HJ, Park ET, Yoo KS, Lim BC, Seo DW. Detection of Helicobacter pylori DNA in human biliary tree and its association with hepatolithiasis. Dig Dis Sci. 2000;45:1405-1412.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 48]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
106.  Ananieva O, Nilsson I, Vorobjova T, Uibo R, Wadström T. Immune responses to bile-tolerant helicobacter species in patients with chronic liver diseases, a randomized population group, and healthy blood donors. Clin Diagn Lab Immunol. 2002;9:1160-1164.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Xuan SY, Li N, Qiang X, Zhou RR, Shi YX, Jiang WJ. Helicobacter infection in hepatocellular carcinoma tissue. World J Gastroenterol. 2006;12:2335-2340.  [PubMed]  [DOI]  [Cited in This Article: ]