Skip to main content
Erschienen in: Infectious Agents and Cancer 1/2008

Open Access 01.12.2008 | Review

The Rho-activating CNF1 toxin from pathogenic E. coli: A risk factor for human cancer development?

verfasst von: Sara Travaglione, Alessia Fabbri, Carla Fiorentini

Erschienen in: Infectious Agents and Cancer | Ausgabe 1/2008

Abstract

Nowadays, there is increasing evidence that some pathogenic bacteria can contribute to specific stages of cancer development. The concept that bacterial infection could be involved in carcinogenesis acquired a widespread interest with the discovery that H. pylori is able to establish chronic infections in the stomach and that this infection is associated with an increased risk of gastric adenocarcinoma and mucosa associated lymphoid tissue lymphoma. Chronic infections triggered by bacteria can facilitate tumor initiation or progression since, during the course of infection, normal cell functions can come under the control of pathogen factors that directly manipulate the host regulatory pathways and the inflammatory reactions.
Renowned publications have recently corroborated the molecular mechanisms that link bacterial infections, inflammation and cancer, indicating certain strains of Escherichia coli as a risk factor for patients with colon cancer. E. coli is a normal inhabitant of the human intestine that becomes highly pathogenic following the acquisition of virulence factors, including a protein toxin named cytotoxic necrotizing factor 1 (CNF1). This toxin permanently activates the small GTP-binding proteins belonging to the Rho family, thus promoting a prominent polymerization of the actin cytoskeleton as well as a number of cellular responses, including changes in protein expression and functional modification of the cell physiology. CNF1 is receiving an increasing attention as a putative factor involved in transformation because of its ability to: (i) induce COX2 expression, an immediate-early gene over-expressed in some type of cancers; (ii) induce a long-lasting activation of the transcription factor NF-kB, a largely accepted marker of tumor cells; (iii) protect epithelial cells from apoptosis; (iv) ensue the release of pro-inflammatory cytokines in epithelial and endothelial cells; and (v) promote cellular motility. As cancer may arise through dysfunction of the same regulatory systems, it seems likely that CNF1-producing E. coli infections can contribute to tumor development.
This review focuses on the aspects of CNF1 activity linked to cell transformation with the aim of contributing to the identification of a possible carcinogenic agent from the microbial world.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1750-9378-3-4) contains supplementary material, which is available to authorized users.

Authors' contributions

ST, AF and CF participated in drafting the manuscript and in its approval.

Introduction

Bacterial infection and cancer

In the last century, cancer research thoroughly established the role of major carcinogenic agents of different nature, including infectious agents. However, although there is a general agreement that some viruses, such as hepatitis B virus (HBV), Epstein-Barr virus (EBV) and human papilloma virus (HPV) can cause cancer, the involvement of bacteria in carcinogenesis remains controversial. The role of viral infections in tumor onset is widely accepted because of the direct action of single viral genes (oncogenes) that result in cell transformation [1]. By contrast, the molecular mechanism(s) by which bacteria might promote tumorigenesis are still poorly characterized. Hence, one of the main challenges, nowadays, is to define the impact of bacterial infections as a cause of cancer and eventually design strategies for their prevention and control.
Bacterial infections are usually believed to cause acute diseases, but it is now becoming clear that some bacteria can contribute to the establishment of chronic diseases, including cancer [2]. The concept that bacterial infection could be involved in carcinogenesis was first proposed in the late nineteenth and early twentieth centuries, based on the discovery of bacteria at the sites of tumors, although there was no proof that the bacteria were in any way causative [3]. Since then, the putative link between chronic infection and cancer acquired a widespread interest with the discovery that Helicobacter pylori is able to establish chronic infections in the stomach and that this infection is associated with an increased risk of gastric adenocarcinoma [4] and mucosa associated lymphoid tissue (MALT) lymphoma [5]. In this context, it is worth noting that H. pylori is classified as a class I carcinogenic factor [6]. Other chronic bacterial infections have been linked to human carcinogenesis although the underlying mechanisms remain to be defined (reviewed in [2]). The strongest epidemiological case is for Salmonella enterica serovar typhi (S. typhi), the agent of typhoid, which can also lead to chronic bacterial carriage in the gallbladder [711]. Surveys of typhoid outbreaks have shown that those who become carriers have an increased risk of developing hepatobiliary carcinoma compared with people who have had acute typhoid and have cleared the infection [9].
A recurring theme in the link between bacterial infection and carcinogenesis is that of chronic inflammation, which is often a common feature of persistent infection [2, 12]. One of the key molecules that link chronic inflammation and cancer is represented by the NF-kB family of transcription factors [12, 13]. In particular, different mouse studies provide strong and direct genetic evidence that the classical, IKK-β dependent NF-kB activation pathway is indeed a crucial mediator of tumor promotion [1416]. This pathway is triggered by bacterial and viral infections, as well as by pro-inflammatory cytokines, such as TNF-α and IL-1, all of which activate the IKK complex [17]. This complex phosphorilates the NF-kB inhibitors IkBs, thereby targeting them for proteosomal degradation and freeing NF-kB to enter the nucleus and mediate transcription of target genes. It is worth noting that many of the genes able to mediate alterations characterizing a tumor cell are under the transcriptional control of NF-kB (reviewed in [18, 19]). For example, the activity and expression of cyclin D1, CDK2 kinase, c-myc, p21, p53 and pRb, which are involved in the control of cell cycle and are altered in several types of cancer, are NF-kB-dependent. The expression of numerous cytokines, that are growth factors for tumor cells (IL-1β, TNF, IL-6, EGF) are also regulated by NF-kB. Tissue invasion and metastasis, two crucial events of tumor progression, are regulated by NF-kB-dependent genes, including metalloproteases (MMPs), urokinase type of plasminogen activator (uPA), IL-8, the adhesion molecules VCAM-1, ICAM-1 and ELAM-1. NF-kB is also involved in the regulation of angiogenesis, the process by which tumor cells promote neo-vascularization. Finally, altered expression of genes involved in suppression of apoptosis (i.e. Bcl-2 family members and IAP proteins), a key feature of cancer cells, is often due to deregulated NF-kB activity.
Concerning this last point, several pathogenic bacteria, particularly those that can establish a persistent intracellular infection, activate NF-kB in the host cell and suppress cell death, thus creating a niche in which the bacterium can survive, in spite of the attempts of the host immune system to destroy the infected cell [20]. As a consequence, the suppression of apoptosis by a pathogen might also allow a partially transformed cell to evade the self-destructive process and to progress to a higher level of transformation.
Another important feature of inflammation-associated cancer is the production of reactive oxygen species (ROS) and nitric oxide (NO) by inflammatory and epithelial cells. This leads to increased mutations and altered functions of important enzymes and proteins in inflamed tissue, thus contributing to the multistage carcinogenetic process [21]. For example, during a chronic H. pylori infection, production of ROS and nitroxides and the associated inflammatory response are assumed to contribute to the induction of a gastric carcinogenic process [22, 23]. These chemical species are mainly produced by inflammatory cells to fight infection and are source of oxidative DNA damage, thus contributing to carcinogenesis [22, 23]. In particular, it has been demonstrated that within 6 hours, H. pylori infection had a mutagenic effect on gastric epithelial cells [24]. The major form of oxidative DNA damage is the formation of 8-oxoG lesions, specifically repaired by the OGG1 DNA glycosylase. The inactivation of this enzyme inhibits the level of inflammatory lesions and abolishes the mutagenic effect induced by the infection at the gastric level, thus strengthening a close relation between chronic inflammation and genotoxicity [24].

Bacterial toxins and cancer

As stated above, there is increasing evidence that some pathogenic bacteria can contribute to specific stages of cancer development. In particular, chronic infections triggered by bacteria can facilitate tumor initiation or progression since, during the course of infection, normal cell functions can undergo the control of factors released by the pathogen [2]. These bacterial factors, namely virulence factors, can directly manipulate the host regulatory pathways and the inflammatory reaction [3].
Bacteria express a wide range of virulence factors, including protein toxins that have evolved to interact with eukaryotic cellular machinery in a precise way. These toxins interfere with key eukaryotic processes, such as cellular signaling components, and some directly attack the genome [2527]. These last can damage DNA via different mechanisms: i) directly by enzymatic attack, ii) indirectly by provoking an inflammatory reaction that produces free radicals, or even iii) by affecting DNA repair mechanisms. Nougayrède and colleagues [28] have recently identified a novel hybrid peptide-polyketide compound from Escherichia coli that leads to DNA damage. This novel compound is produced by pathogenic and, most interestingly, commensal isolates. Although it is not yet clear how the peptide-polyketide compound functions at the molecular level, it is possible that it contributes to bacterial pathogenesis and bacterially-induced carcinogenesis.
Any bacterial product that interferes with signaling, resulting in the disruption of the normal balance of growth, cell division and apoptosis, could facilitate tumor promotion. Similarly, the ability to promote anchorage-independent growth could favor metastatic potential and lead to cancer progression. So far, the best example of potentially carcinogenic toxin is H. pylori CagA that interferes with cellular signaling mechanisms in a way that is characteristic of tumor promoters. Indeed, CagA intracellularly interacts, in a phosphorylation-dependent and independent way, with many host proteins that regulate cell growth, motility and polarity, thus leading to gastric epithelial proliferation, cell-cell dissociation and increased cell scattering and motility (for a review [29]). In particular, Bagnoli and coworkers [30] showed that CagA is sufficient to disrupt the mechanisms that maintain normal epithelial differentiation, including cell adhesion, cell polarity, and the inhibition of migration. Since the cellular behavior induced by CagA is reminiscent of oncogenes that disrupt cytoskeletal signaling, the authors proposed that altered cell-cell and cell matrix interactions may serve as an early event in H. pylori-induced carcinogenesis [30]. Very recently, in experimental studies dealing with H. pylori strains carrying or not CagA, it has been demonstrated that this factor activates host cell survival and anti-apoptotic pathways to overcome self-renewal of the gastric epithelium, thus enhancing bacterial colonization of the stomach and helping sustained H. pylori infection. In this context, it is interesting to note that, patients infected with H. pylori encoding the cag pathogeniticy island (PAI) are associated with an increased risk of gastric cancer [31].
In addition to the link between H. pylori and stomach cancers, very recent studies evidenced that some other toxins may contribute to bowel and urogenital tract cancers [3]. In this context, it has been reported that adherent and invasive strains of Escherichia coli are a risk factor for patients with pre-cancerous and cancerous colon diseases [32, 33]. Interestingly, in one of these studies, 3 out of 8 cancer-associated E. coli were reported to possess the cytotoxic necrotizing factor 1 (cnf1) gene [32], the gene coding for the protein toxin CNF1. The aim of this review is to highlight the main cell responses to CNF1, particularly those related to signaling pathways linked to inflammation and to cell transformation.

CNF1 from E. coli: a protein toxin that activates the Rho GTPases

Although belonging to the normal human intestinal flora, E. coli becomes highly pathogenic following the acquisition of genes coding for virulence factors, one of which being CNF1 [34]. CNF1-producing E. coli strains are occasionally detected in isolates from feces of children with diarrhea [3537], but, more frequently, are responsible of extraintestinal infections, particularly in the urinary tract (UTIs) [3840]. Also, these strains can be detected in cases of bacteraemia [41] and of meningitis in neonates [42]. CNF1, first described in 1983 by Caprioli and coworkers as a toxin capable of causing multinucleation ("cytotoxic") in cultured cells (Fig. 1a, b) and necrosis in rabbit skin ("necrotizing") [43, 44], is a single-chain multidomain protein toxin, containing a binding domain to a cell receptor (N-terminal domain), a translocation domain (middle domain) and an enzymatic one (C-terminal domain) [45, 46], which modifies a specific cellular target in the host cell cytosol. CNF1 binds to the surface of cultured epithelial cells with high affinity [47], and the 67-kDa laminin receptor has been suggested (67LR) as a putative receptor for CNF1 [48]. After binding to its receptor, CNF1 is endocytosed and routed to an endosomal compartment [47], from where the toxin injects its catalytic activity into the cytosol [47], by using two hydrophobic structures present in the middle part of the CNF1 molecule [49].
The cytoplasmic target of CNF1 is represented by the Rho GTPases, important molecular switches belonging to the Ras superfamily, that cycle between an inactive GDP-bound state and an active GTP-bound one, under the strict control of activators (guanine nucleotide exchange factors, GEFs) and inactivators (GTPase-activating proteins, GAPs) [50]. The conformational changes, induced by the binding of GTP or GDP, occur inside two molecular domains of the Rho proteins called switches that are responsible of the coupling of the G-proteins with their downstream effectors (switch 1) and allow the interaction of the activated GTPase with GAPs (switch 2). The enzymatic activity of CNF1 consists in the deamidation of a specific glutamine residue located in the switch 2 domain of the G proteins (glutamine 63 of Rho [51, 52] or glutamine 61 of Rac and Cdc42 [53]). This glutamine residue is essential for the GTPase activity of Rho proteins, either intrinsic or GAP-mediated [54]. By modifying glutamine into glutamic acid, CNF1 impairs the role of Rho GAP, allowing Rho proteins to be permanently locked in their activated GTP-bound state and, thus, enhancing the activity of these proteins on their effectors.
It is worth noting that the modification on Rho proteins induced by CNF1 corresponds to the modification found on Ras proteins in many tumors [55]. In fact, Rho glutamine 63 corresponds to Ras glutamine 61, a well-known residue that, after mutagenization, leads to a permanent activation of the molecule and to tumor onset.

CNF1 and Rho GTPases: actin remodeling and host cell invasion

The Rho GTPases are pivotal in controlling the actin cytoskeleton architecture [50]. In fact, one of the first described cell responses to CNF1 is a remarkable reorganization of the actin cytoskeleton in epithelial cells that consists in the assembly of F-actin in prominent stress fibers, membrane ruffles and filopodia [56] (Fig. 1c, d). The prominent ruffling activity promoted by CNF1 permits epithelial cells to behave as phagocytes, developing a macropinocytotic activity that allows the capture and engulfment of different types of particles, including bacteria [57, 58]. This aspect is of particular relevance for the bacterial pathogenicity since the macropinocytic activity ensued by CNF1 in epithelial cells may possibly represent the route of entry of CNF1-producing E. coli, similarly to what occurs with other intestinal pathogens (Fig. 1e). Furthermore, upon activation by CNF1, the Rho GTPases undergo sensitization to ubiquitylation and subsequent proteosomal degradation [59], a process that would turn off the ruffling process, thus allowing an efficient internalization of bacteria inside the cells (Fig. 1f). It is worth noting that the activity of CNF1, with its ability to switch on the Rho GTPases and then coerce their degradation in the proteasome, is somehow similar to the activity of the intracellular bacterium Salmonella [60], for which a link with cancer has been evidenced [9]. This bacterium first activates the Rho GTPases, by the GEF-like toxin SopE, to promote macropinocytosis that allows its entry into cells and soon after, once inside, deactivates the GTPases via a GAP-mimicking protein (SptP), thus allowing a moderate threshold of Rho protein activation for a high invasion efficiency [61].
By regulating the actin cytoskeleton, the Rho GTPases also play a crucial role in certain aspects of the malignant phenotype, such as tumor cell motility, invasiveness and metastasis [62]. Indeed, RhoC has been implicated as a marker for highly angiogenic and aggressive breast cancer with a high metastatic ability [63]. An additional clue in favor of our hypothesis comes from the observation that CNF1 provokes cell junctions disruption and strongly enhances cellular motility in uroepithelial 804G cells [59]. The augmented motility of epithelial cells highlights another characteristic that somehow links this toxin to cancer.

CNF1 hinders apoptosis via the pro-inflammatory Akt/IKK/NF-kB pathway

It is also known that Rho proteins are crucially involved in the development of inflammatory processes and that a key player in the link existing between Rho, chronic inflammation and cancer is the Nuclear Factor-kB (NF-kB) [64]. As mentioned in the Introduction, NF-kB is represented by a group of structurally related and evolutionarily conserved transcription factors involved in regulating the expression of genes that control different aspects of the tumor cell biology, including inflammation, cell growth, and suppression of apoptosis (reviewed in [65]). Tumor cells can show elevated NF-κB-regulated transcription, which can inhibit TNF-α-induced apoptosis through the up-regulation of the anti-apoptotic proteins of the Bcl-2 family.
In this context, we previously reported that CNF1 can activate the nuclear factor-kB (NF-kB) [66] in epithelial cells. Such an activation, is responsible for the ability of the toxin to stimulate the expression of pro-inflammatory factors and to protect host cell from apoptotic stimuli. As concerns the pro-survival activity, we have shown that CNF1 can increase the expression of proteins related to cell adhesion (integrins, Focal Adhesion Kinase, cadherins, catenins), thus improving cell spreading and the ability of cells to adhere to each other and to the extracellular matrix [67]. In fact, prolonged cell survival, together with increased adhesion to matrix components might have significant biological consequences and affect the tumorigenic potential of epithelial cells. Moreover, CNF1 protects epithelial cells against the drop of the mitochondrial membrane potential provoked by UVB radiation and increases the expression of the anti-apoptotic members of the Bcl-2 family, Bcl-2 and Bcl-XL [68]. Although the causal relationship between Bcl-2 and mitochondrial membrane potential has not yet been clarified, we have very recently demonstrated that the up-regulation of the anti-apoptotic protein Bcl-2 somehow controls the mitochondrial morphology, and that this depends on the activation of the pro-inflammatory Rac1/PI3K/Akt/IKK/NF-kB pathway [69]. In fact, besides blocking the activity of the pro-apoptotic members of Bcl-2 family, Bcl-2 is involved in the regulation of mitochondrial morphology, since Bcl-2-over-expressing mitochondria present both increased volume and structural complexity [70]. In living cells, mitochondria continuously divide (fission) and fuse (fusion) with one another [71] and Bcl-2 family members are involved in these processes, the pro-apoptotic members regulating fission whereas the anti-apoptotic regulating fusion of mitochondria [72]. In this context, we very recently demonstrated that CNF1 can induce, in epithelial cells, the formation of a complex network of elongated and interconnected mitochondria with an increased average length [[69]; Fig. 1i, l]. Importantly, Bcl-2 silencing reduces the ability of CNF1 to protect cells against apoptosis and also prevents the CNF1-induced mitochondrial changes. Therefore, since the mitochondrial remodeling is of direct relevance for the role of these organelles in cell physiology and the mitochondrial dysfunction can contribute to a number of human disorders, including cancer [73], the role of CNF1 as a factor favoring transformation can be further supported by this novel finding.
As concerns inflammation, CNF1 ensues the transcription and release of pro-inflammatory cytokines, such as IL-6, IL-8 and TNF-α in uroepithelial [74] and endothelial [75] cells, probably contributing to the establishing of the inflammatory process due to CNF1-producing E. coli. The pro-inflammatory role of CNF1 is also supported by the demonstration that the toxin strongly up-regulate the transcription of cyclooxygenase- 2 (COX-2) [76], an immediate-early gene induced in response to pro-inflammatory cytokines, tumor promoters, and growth factors and over-expressed in cancers of the lung, colon, stomach, and breast [7779].

Conclusion

On the whole, it appears that CNF1 touches some of the signaling pathways that are engaged by carcinogens and tumor promoters, as schematized in Fig. 2. Particularly relevant are the activation of the transcription factor NF-κB, the pro-survival activity, the increased expression of RNA messengers for COX-2 and pro-inflammatory cytokines as well as the augmented cell motility. In addition, CNF1 impairs the cytokinesis, thus leading to multinucleation [44, 56], induces nuclear segmentation, amitotic cell division, multipolar mitosis (Fig. 1g, h) [80], and modulates autophagy [81], cellular phenomena that are frequently observed in different types of cancer cells, and blocks the cell cycle G2/M transition in epithelial cells [82]. The ability of CNF1 to block the cell cycle progression suggests a strategy that permits to contain the host damage, and to induce specific cellular responses rather than rapid cell death. This could in turn facilitate the bacterial invasion of underlying tissues. The activity of CNF1, with its ability to switch on the Rho GTPases and then coerce their degradation in the proteasome, probably renders the CNF1-producing E. coli intracellular parasites, hence permitting their potentially harmful and presumably transforming activity inside the cell, where they can escape the host immune system attack. Finally, it is worth noting that CNF1 activity on cells shares several properties with CagA from the carcinogenic bacterium H. pylori, which can be indicated as the best example of a potentially carcinogenic toxin [29]. In Table 1, the cellular effects of E. coli CNF1 and those of the major virulence factors (VacA and CagA) of H. pylori are compared.
Table 1
Comparison between the cellular effects of E. coli CNF1 and those of the major virulence factors (VacA and CagA) of H. pylori
 
CNF1
CagA
VacA
Effects on cytoskeleton and cell morphology
• Actin polymerization
• Spreading
• Multinucleation
• Actin rearrangement
• "hummingbird" phenotype
• loss of cell polarity
• cell scattering
• Actin disruption
• Vacuolization
Apoptosis regulation
Anti-apoptotic effect
Anti-apoptotic effect
Pro-apoptotic effect
Production of inflammatory mediators
TNF-α
IL-8
TNF-α
 
IL-6
ROS
IL-8
 
IL-8
 
IL-1β
 
ROS
 
IL-6
Activation of transcription factors
NF-kB
NF-kB
NF-kB
  
NFAT
ATF-2
  
SRF
NFAT
  
AP1
 
Effect on cell cycle
Block in G2/M
Inhibition of G1/S progression
Induction of G1/S progression
Effects on mitochondria
Inhibition of UVB-induced mitochondrial membrane depolarization
 
Reduction of mitochondrial membrane potential, cytochrome c release
Hence, taking altogether, we can hypothesize that once secreted by epithelium-associated E. coli, CNF1 switches on the Rho GTPases, initiating a pathway that leads to the actin cytoskeleton reorganization, but also to the activation of NF-κB, which in turn enter the nucleus and causes the transcription of anti apoptotic factors as well as pro-inflammatory cytokines. Production and secretion of pro-inflammatory factors recruits cells of the immune system creating an inflammatory environment. Epithelial cells with the Rho GTPases modified by CNF1 in a way that corresponds to the modification found on Ras proteins in many tumors [55], continue to grow without undergoing apoptosis in an environment rich of inflammatory factors. This can account, finally, for promotion or increase of the neoplastic risk.

Acknowledgements

The authors are grateful to Roberta Terlizzi and Zaira Maroccia for their invaluable technical assistance in preparing the manuscript. The study has been funded by ISS Grants to C.F and A.F.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Authors' contributions

ST, AF and CF participated in drafting the manuscript and in its approval.
Anhänge

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.
Literatur
1.
Zurück zum Zitat Kuper H, Adami H-O, Trichopoulos D: Infections as a major preventable cause of human cancer. J Int Med. 2000, 248: 171-183. 10.1046/j.1365-2796.2000.00742.x.CrossRef Kuper H, Adami H-O, Trichopoulos D: Infections as a major preventable cause of human cancer. J Int Med. 2000, 248: 171-183. 10.1046/j.1365-2796.2000.00742.x.CrossRef
2.
Zurück zum Zitat Lax AJ, Thomas W: How bacteria could cause cancer: one step at a time. Trends Microbiol. 2002, 10: 293-299. 10.1016/S0966-842X(02)02360-0.PubMedCrossRef Lax AJ, Thomas W: How bacteria could cause cancer: one step at a time. Trends Microbiol. 2002, 10: 293-299. 10.1016/S0966-842X(02)02360-0.PubMedCrossRef
3.
Zurück zum Zitat Lax AJ: Opinion: Bacterial toxins and cancer – a case to answer?. Nat Rev Microbiol. 2005, 3: 343-349. 10.1038/nrmicro1130.PubMedCrossRef Lax AJ: Opinion: Bacterial toxins and cancer – a case to answer?. Nat Rev Microbiol. 2005, 3: 343-349. 10.1038/nrmicro1130.PubMedCrossRef
4.
Zurück zum Zitat Parsonnet J, Friedman GD, Vandersteen DP, Chang Y, Vogelman JH, Orentreich N, Sibley RK: Helicobacter pylori infection and the risk of gastric carcinoma. N Eng J Med. 1991, 325 (16): 1127-1131.CrossRef Parsonnet J, Friedman GD, Vandersteen DP, Chang Y, Vogelman JH, Orentreich N, Sibley RK: Helicobacter pylori infection and the risk of gastric carcinoma. N Eng J Med. 1991, 325 (16): 1127-1131.CrossRef
5.
Zurück zum Zitat Wotherspoon AC, Ortiz-Hidalgo C, Falzon MR, Isaacson PG: Helicobacter pylori-associated gastritis and primary B-cell gastric lymphoma. Lancet. 1991, 338: 1175-1176. 10.1016/0140-6736(91)92035-Z.PubMedCrossRef Wotherspoon AC, Ortiz-Hidalgo C, Falzon MR, Isaacson PG: Helicobacter pylori-associated gastritis and primary B-cell gastric lymphoma. Lancet. 1991, 338: 1175-1176. 10.1016/0140-6736(91)92035-Z.PubMedCrossRef
7.
Zurück zum Zitat Welton JC, Marr JS, Friedman SM: Association between hepatobiliary cancer and typhoid carrier state. Lancet. 1979, 1: 791-794. 10.1016/S0140-6736(79)91315-1.PubMedCrossRef Welton JC, Marr JS, Friedman SM: Association between hepatobiliary cancer and typhoid carrier state. Lancet. 1979, 1: 791-794. 10.1016/S0140-6736(79)91315-1.PubMedCrossRef
8.
Zurück zum Zitat Caygill CPJ: Cancer mortality in chronic typhoid and paratyphoid carriers. Lancet. 1994, 343: 83-84. 10.1016/S0140-6736(94)90816-8.PubMedCrossRef Caygill CPJ: Cancer mortality in chronic typhoid and paratyphoid carriers. Lancet. 1994, 343: 83-84. 10.1016/S0140-6736(94)90816-8.PubMedCrossRef
9.
Zurück zum Zitat Caygill CPJ, Braddick M, Hill MJ, Knowles RL, Sharp JCM: The association between typhoid carriage, typhoid infection and subsequent cancer at a number of sites. Eur J Cancer Prev. 1995, 4: 187-193.PubMedCrossRef Caygill CPJ, Braddick M, Hill MJ, Knowles RL, Sharp JCM: The association between typhoid carriage, typhoid infection and subsequent cancer at a number of sites. Eur J Cancer Prev. 1995, 4: 187-193.PubMedCrossRef
10.
Zurück zum Zitat Nath G, Singh H, Shukla VK: Chronic typhoid carraige and carcinoma of the gladdbladder. Eur J Cancer Prev. 1997, 6: 557-559. 10.1097/00008469-199712000-00011.PubMedCrossRef Nath G, Singh H, Shukla VK: Chronic typhoid carraige and carcinoma of the gladdbladder. Eur J Cancer Prev. 1997, 6: 557-559. 10.1097/00008469-199712000-00011.PubMedCrossRef
11.
Zurück zum Zitat Shukla VK, Singh H, Pandey M, Upadhyay SK, Nath G: Carcinoma of the gladdbladder-is a sequel of typhoid?. Dig Dis Sci. 2000, 45: 900-903. 10.1023/A:1005564822630.PubMedCrossRef Shukla VK, Singh H, Pandey M, Upadhyay SK, Nath G: Carcinoma of the gladdbladder-is a sequel of typhoid?. Dig Dis Sci. 2000, 45: 900-903. 10.1023/A:1005564822630.PubMedCrossRef
12.
Zurück zum Zitat Karin M, Greten FR: NF-κB: linking inflammation and immunity to cancer development and progression. Nature Rev. 2005, 5 (10): 749-759. 10.1038/nri1703. Karin M, Greten FR: NF-κB: linking inflammation and immunity to cancer development and progression. Nature Rev. 2005, 5 (10): 749-759. 10.1038/nri1703.
13.
Zurück zum Zitat Karin M, Cao Y, Greten FR, Li ZW: NF-κB in cancer: from innocent bystander to major culprit. Nature Rev Cancer. 2002, 2 (4): 301-310. 10.1038/nrc780.CrossRef Karin M, Cao Y, Greten FR, Li ZW: NF-κB in cancer: from innocent bystander to major culprit. Nature Rev Cancer. 2002, 2 (4): 301-310. 10.1038/nrc780.CrossRef
14.
Zurück zum Zitat Greten FR, Eckmann L, Greten TF, Park JM, Li ZW, Egan LJ, Kagnoff MF, Karin M: IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell. 2004, 118: 285-296. 10.1016/j.cell.2004.07.013.PubMedCrossRef Greten FR, Eckmann L, Greten TF, Park JM, Li ZW, Egan LJ, Kagnoff MF, Karin M: IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell. 2004, 118: 285-296. 10.1016/j.cell.2004.07.013.PubMedCrossRef
15.
Zurück zum Zitat Pikarsky E, Porat RM, Stein I, Abramovitch R, Sharon A, Kasem S, Gutkovich-Pyest E, Uriel-Shoval S, Galun E, Ben-Nerlah Y: NF-κB functions as a tumor promoter in inflammation-associated cancer. Nature. 2004, 43: 461-466. 10.1038/nature02924.CrossRef Pikarsky E, Porat RM, Stein I, Abramovitch R, Sharon A, Kasem S, Gutkovich-Pyest E, Uriel-Shoval S, Galun E, Ben-Nerlah Y: NF-κB functions as a tumor promoter in inflammation-associated cancer. Nature. 2004, 43: 461-466. 10.1038/nature02924.CrossRef
16.
Zurück zum Zitat Luo JL, Maeda S, Hsu LC, Yagita H, Karin M: Inhibition of NF-κB in cancer cells converts inflammation-induced tumor growth mediated by TNF-α to TRAIL-mediated tumor regression. Cancer Cell. 2004, 6: 297-305. 10.1016/j.ccr.2004.08.012.PubMedCrossRef Luo JL, Maeda S, Hsu LC, Yagita H, Karin M: Inhibition of NF-κB in cancer cells converts inflammation-induced tumor growth mediated by TNF-α to TRAIL-mediated tumor regression. Cancer Cell. 2004, 6: 297-305. 10.1016/j.ccr.2004.08.012.PubMedCrossRef
17.
Zurück zum Zitat Häcker H, Karin M: Regulation and function of IKK and IKK-related kinases. Sci STKE. 2006, re13-10.1126/stke.3572006re13. 357 Häcker H, Karin M: Regulation and function of IKK and IKK-related kinases. Sci STKE. 2006, re13-10.1126/stke.3572006re13. 357
18.
Zurück zum Zitat Pacifico F, Leonardi A: NF-κB in solid tumors. Biochem Pharmacol. 2006, 72: 1142-1152. 10.1016/j.bcp.2006.07.032.PubMedCrossRef Pacifico F, Leonardi A: NF-κB in solid tumors. Biochem Pharmacol. 2006, 72: 1142-1152. 10.1016/j.bcp.2006.07.032.PubMedCrossRef
19.
Zurück zum Zitat Karin M: NF-κB and cancer: mechanisms and targets. Mol Carcinogenesis. 2006, 45 (6): 355-361. 10.1002/mc.20217.CrossRef Karin M: NF-κB and cancer: mechanisms and targets. Mol Carcinogenesis. 2006, 45 (6): 355-361. 10.1002/mc.20217.CrossRef
20.
Zurück zum Zitat Van Antwerp DJ, Martin SJ, Kafri T, Green DR, Verma IM: Suppression of TNF-α induced apoptosis by NF-κB. Science. 1996, 274: 787-789. 10.1126/science.274.5288.787.PubMedCrossRef Van Antwerp DJ, Martin SJ, Kafri T, Green DR, Verma IM: Suppression of TNF-α induced apoptosis by NF-κB. Science. 1996, 274: 787-789. 10.1126/science.274.5288.787.PubMedCrossRef
22.
Zurück zum Zitat Farinati F, Cardin R, Degan P, Rugge M, Di Mario F, Bonvicini P, Naccarato R: Oxidative DNA damage accumulation in gastric carcinogenesis. Gut. 1998, 42: 351-356.PubMedPubMedCentralCrossRef Farinati F, Cardin R, Degan P, Rugge M, Di Mario F, Bonvicini P, Naccarato R: Oxidative DNA damage accumulation in gastric carcinogenesis. Gut. 1998, 42: 351-356.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Obst B, Wagner S, Sewing K, Beil W: Helicobacter pylori causes DNA damage in gastric epithelial cells. Carcinogenesis. 2000, 21: 1111-1115. 10.1093/carcin/21.6.1111.PubMedCrossRef Obst B, Wagner S, Sewing K, Beil W: Helicobacter pylori causes DNA damage in gastric epithelial cells. Carcinogenesis. 2000, 21: 1111-1115. 10.1093/carcin/21.6.1111.PubMedCrossRef
24.
Zurück zum Zitat Touati E, Michel V, Thiberge J-M, Avé P, Huerre M, Bourgade F, Klungland A, Labigne A: Deficiency in OGG1 protects against inflammation and mutagenic effects associated with H. pylori infection in mouse. Helicobacter. 2006, 11: 494-505. 10.1111/j.1523-5378.2006.00442.x.PubMedCrossRef Touati E, Michel V, Thiberge J-M, Avé P, Huerre M, Bourgade F, Klungland A, Labigne A: Deficiency in OGG1 protects against inflammation and mutagenic effects associated with H. pylori infection in mouse. Helicobacter. 2006, 11: 494-505. 10.1111/j.1523-5378.2006.00442.x.PubMedCrossRef
25.
Zurück zum Zitat Schiavo G, van der Goot FG: The bacterial toxin toolkit. Nat Rev Mol Cell Biol. 2001, 2: 530-537. 10.1038/35080089.PubMedCrossRef Schiavo G, van der Goot FG: The bacterial toxin toolkit. Nat Rev Mol Cell Biol. 2001, 2: 530-537. 10.1038/35080089.PubMedCrossRef
26.
Zurück zum Zitat Fiorentini C, Falzano L, Travaglione S, Fabbri A: Hijacking Rho GTPases by protein toxins and apoptosis: molecular strategies of pathogenic bacteria. Cell Death Differ. 2003, 10: 147-152. 10.1038/sj.cdd.4401151.PubMedCrossRef Fiorentini C, Falzano L, Travaglione S, Fabbri A: Hijacking Rho GTPases by protein toxins and apoptosis: molecular strategies of pathogenic bacteria. Cell Death Differ. 2003, 10: 147-152. 10.1038/sj.cdd.4401151.PubMedCrossRef
27.
Zurück zum Zitat Lax AJ: New genotoxin shows diversity of bacterial attack mechanisms. Trends Mol Med. 2007, 13: 91-93. 10.1016/j.molmed.2007.01.001.PubMedCrossRef Lax AJ: New genotoxin shows diversity of bacterial attack mechanisms. Trends Mol Med. 2007, 13: 91-93. 10.1016/j.molmed.2007.01.001.PubMedCrossRef
28.
Zurück zum Zitat Nougayrède J-P, Homburg S, Taieb F, Boury M, Brzuszkiewicz E, Gottschalk G, Buchrieser C, Hacker J, Dobrindt U, Oswald E: Escherichia coli induces DNA double-strand breaks in eukaryotic cells. Science. 313: 848-851. 10.1126/science.1127059. Nougayrède J-P, Homburg S, Taieb F, Boury M, Brzuszkiewicz E, Gottschalk G, Buchrieser C, Hacker J, Dobrindt U, Oswald E: Escherichia coli induces DNA double-strand breaks in eukaryotic cells. Science. 313: 848-851. 10.1126/science.1127059.
29.
Zurück zum Zitat Hatakeyama M, Higashi H: Helicobacter pylori CagA: a new paradigm for bacterial carcinogenesis. Cancer Sci. 2005, 96: 835-43. 10.1111/j.1349-7006.2005.00130.x.PubMedCrossRef Hatakeyama M, Higashi H: Helicobacter pylori CagA: a new paradigm for bacterial carcinogenesis. Cancer Sci. 2005, 96: 835-43. 10.1111/j.1349-7006.2005.00130.x.PubMedCrossRef
30.
Zurück zum Zitat Bagnoli F, Buti L, Tompkins L, Covacci A, Amieva MR: Helicobacter pylori CagA induces a transition from polarized to invasive phenotypes in MDCK cells. Proc Natl Acad Sci USA. 2005, 102: 16339-16344. 10.1073/pnas.0502598102.PubMedPubMedCentralCrossRef Bagnoli F, Buti L, Tompkins L, Covacci A, Amieva MR: Helicobacter pylori CagA induces a transition from polarized to invasive phenotypes in MDCK cells. Proc Natl Acad Sci USA. 2005, 102: 16339-16344. 10.1073/pnas.0502598102.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Blaser MJ, Perez-Perez GI, Kleanthous H, Cover TL, Peek RM, Chyou PH, Stemmermann GN, Nomura A: Infection with Helicobacter pylori strains possessing cagA is associated with an increased risk of developing adenocarcinoma of the stomach. Cancer Res. 1995, 55: 2111-2115.PubMed Blaser MJ, Perez-Perez GI, Kleanthous H, Cover TL, Peek RM, Chyou PH, Stemmermann GN, Nomura A: Infection with Helicobacter pylori strains possessing cagA is associated with an increased risk of developing adenocarcinoma of the stomach. Cancer Res. 1995, 55: 2111-2115.PubMed
32.
Zurück zum Zitat Martin HM, Campbell BJ, Hart CA, Mpofu C, Nayar M, Singh R, Englyst H, Williams HF, Rhodes JM: Enhanced Escherichia coli adherence and invasion in Crohn's disease and colon cancer. Gastroenterology. 2004, 127: 80-93. 10.1053/j.gastro.2004.03.054.PubMedCrossRef Martin HM, Campbell BJ, Hart CA, Mpofu C, Nayar M, Singh R, Englyst H, Williams HF, Rhodes JM: Enhanced Escherichia coli adherence and invasion in Crohn's disease and colon cancer. Gastroenterology. 2004, 127: 80-93. 10.1053/j.gastro.2004.03.054.PubMedCrossRef
33.
Zurück zum Zitat Darfeuille-Michaud A, Boudeau J, Bulois P, Neut C, Glasser AL, Barnich N, Bringer MA, Swidsinski A, Beaugerie L, Colombel JF: High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn's disease. Gastroenterology. 2004, 127: 412-421. 10.1053/j.gastro.2004.04.061.PubMedCrossRef Darfeuille-Michaud A, Boudeau J, Bulois P, Neut C, Glasser AL, Barnich N, Bringer MA, Swidsinski A, Beaugerie L, Colombel JF: High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn's disease. Gastroenterology. 2004, 127: 412-421. 10.1053/j.gastro.2004.04.061.PubMedCrossRef
34.
Zurück zum Zitat Boquet P, Fiorentini C: The cytotoxic necrotizing factor 1 from Escherichia coli. Bacterial Protein toxins. Handbook of Experimental Pharmacology. Edited by: Aktories K, Just I. 2000, Berlin: Springer, 145: 361-379.CrossRef Boquet P, Fiorentini C: The cytotoxic necrotizing factor 1 from Escherichia coli. Bacterial Protein toxins. Handbook of Experimental Pharmacology. Edited by: Aktories K, Just I. 2000, Berlin: Springer, 145: 361-379.CrossRef
35.
Zurück zum Zitat Elliott SJ, Srinivas S, Albert MJ, Alam K, Robins-Browne RM, Gunzburg ST, Mee BJ, Chang BJ: Characterization of the roles of hemolysin and other toxins in enteropathy caused by alpha-hemolytic Escherichia coli linked to human diarrhea. Infect Immun. 1998, 66: 2040-2051.PubMedPubMedCentral Elliott SJ, Srinivas S, Albert MJ, Alam K, Robins-Browne RM, Gunzburg ST, Mee BJ, Chang BJ: Characterization of the roles of hemolysin and other toxins in enteropathy caused by alpha-hemolytic Escherichia coli linked to human diarrhea. Infect Immun. 1998, 66: 2040-2051.PubMedPubMedCentral
36.
Zurück zum Zitat Okeke IN, Lamikanra A, Steinruck H, Kaper JB: Characterization of Escherichia coli strains from cases of childhood diarrhea in provincial southwestern Nigeria. J Clin Microbiol. 2000, 38: 7-12.PubMedPubMedCentral Okeke IN, Lamikanra A, Steinruck H, Kaper JB: Characterization of Escherichia coli strains from cases of childhood diarrhea in provincial southwestern Nigeria. J Clin Microbiol. 2000, 38: 7-12.PubMedPubMedCentral
37.
Zurück zum Zitat Paciorek J: Virulence properties of Escherichia coli faecal strains isolated in Poland from healthy children and strains belonging to serogroups O18, O26, O44, O86, O126 and O127 isolated from children with diarrhoea. J Med Microbiol. 2002, 51: 548-556.PubMedCrossRef Paciorek J: Virulence properties of Escherichia coli faecal strains isolated in Poland from healthy children and strains belonging to serogroups O18, O26, O44, O86, O126 and O127 isolated from children with diarrhoea. J Med Microbiol. 2002, 51: 548-556.PubMedCrossRef
38.
Zurück zum Zitat De Rycke J, Milon A, Oswald E: Necrotoxic Escherichia coli (NTEC): two emerging categories of human and animal pathogens. Vet Res. 1999, 30: 221-233.PubMed De Rycke J, Milon A, Oswald E: Necrotoxic Escherichia coli (NTEC): two emerging categories of human and animal pathogens. Vet Res. 1999, 30: 221-233.PubMed
39.
Zurück zum Zitat Landraud L, Gauthier M, Fosse T, Boquet P: Frequency of Escherichia coli strains producing the cytotoxic necrotizing factor (CNF1) in nosocomial urinary tract infections. Lett Appl Microbiol. 2000, 30: 213-216. 10.1046/j.1472-765x.2000.00698.x.PubMedCrossRef Landraud L, Gauthier M, Fosse T, Boquet P: Frequency of Escherichia coli strains producing the cytotoxic necrotizing factor (CNF1) in nosocomial urinary tract infections. Lett Appl Microbiol. 2000, 30: 213-216. 10.1046/j.1472-765x.2000.00698.x.PubMedCrossRef
40.
Zurück zum Zitat Boquet P: The cytotoxic necrotizing factor 1 (CNF1) from Escherichia coli. Toxicon. 2001, 39: 1673-1680. 10.1016/S0041-0101(01)00154-4.PubMedCrossRef Boquet P: The cytotoxic necrotizing factor 1 (CNF1) from Escherichia coli. Toxicon. 2001, 39: 1673-1680. 10.1016/S0041-0101(01)00154-4.PubMedCrossRef
41.
Zurück zum Zitat Blanco J, Blanco M, Gonzalez EA, Alonso MP, Garabal JI: Comparative evaluation of three tests for the detection of Escherichia coli cytotoxic necrotizing factors (CNF1 and CNF2) using filtrates of cultures treated with mitomycin C. FEMS Microbiol Lett. 1990, 57: 311-316. 10.1111/j.1574-6968.1990.tb04250.x.PubMedCrossRef Blanco J, Blanco M, Gonzalez EA, Alonso MP, Garabal JI: Comparative evaluation of three tests for the detection of Escherichia coli cytotoxic necrotizing factors (CNF1 and CNF2) using filtrates of cultures treated with mitomycin C. FEMS Microbiol Lett. 1990, 57: 311-316. 10.1111/j.1574-6968.1990.tb04250.x.PubMedCrossRef
42.
Zurück zum Zitat Houdouin V, Bonacorsi S, Brahimi N, Clermont O, Nassif X, Bingen E: A uropathogenicity island contributes to the pathogenicity of Escherichia coli strains that cause neonatal meningitis. Infect Immun. 2002, 70: 5865-5869. 10.1128/IAI.70.10.5865-5869.2002.PubMedPubMedCentralCrossRef Houdouin V, Bonacorsi S, Brahimi N, Clermont O, Nassif X, Bingen E: A uropathogenicity island contributes to the pathogenicity of Escherichia coli strains that cause neonatal meningitis. Infect Immun. 2002, 70: 5865-5869. 10.1128/IAI.70.10.5865-5869.2002.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Caprioli A, Falbo V, Roda LG, Ruggeri FM, Zona C: Partial purification and characterization of an Escherichia coli toxic factor that induces morphological cell alterations. Infect Immun. 1983, 39: 1300-1306.PubMedPubMedCentral Caprioli A, Falbo V, Roda LG, Ruggeri FM, Zona C: Partial purification and characterization of an Escherichia coli toxic factor that induces morphological cell alterations. Infect Immun. 1983, 39: 1300-1306.PubMedPubMedCentral
44.
Zurück zum Zitat Caprioli A, Donelli G, Falbo V, Possenti R, Roda LG, Roscetti G, Ruggeri FM: A cell division-active protein from E. coli. Biochem Biophys Res Commun. 1984, 118: 587-593. 10.1016/0006-291X(84)91343-3.PubMedCrossRef Caprioli A, Donelli G, Falbo V, Possenti R, Roda LG, Roscetti G, Ruggeri FM: A cell division-active protein from E. coli. Biochem Biophys Res Commun. 1984, 118: 587-593. 10.1016/0006-291X(84)91343-3.PubMedCrossRef
45.
Zurück zum Zitat Lemichez E, Flatau G, Bruzzone M, Boquet P, Gauthier M: Molecular localization of the Escherichia coli cytotoxic necrotizing factor CNF1 cell-binding and catalytic domains. Mol Microbiol. 1997, 24: 1061-1070. 10.1046/j.1365-2958.1997.4151781.x.PubMedCrossRef Lemichez E, Flatau G, Bruzzone M, Boquet P, Gauthier M: Molecular localization of the Escherichia coli cytotoxic necrotizing factor CNF1 cell-binding and catalytic domains. Mol Microbiol. 1997, 24: 1061-1070. 10.1046/j.1365-2958.1997.4151781.x.PubMedCrossRef
46.
Zurück zum Zitat Fabbri A, Gauthier M, Boquet P: The 5' region of cnf1 harbours a translational regulatory mechanism for CNF1 synthesis and encodes the cell-binding domain of the toxin. Mol Microbiol. 1999, 33: 108-118. 10.1046/j.1365-2958.1999.01453.x.PubMedCrossRef Fabbri A, Gauthier M, Boquet P: The 5' region of cnf1 harbours a translational regulatory mechanism for CNF1 synthesis and encodes the cell-binding domain of the toxin. Mol Microbiol. 1999, 33: 108-118. 10.1046/j.1365-2958.1999.01453.x.PubMedCrossRef
47.
Zurück zum Zitat Contamin S, Galmiche A, Doye A, Flatau G, Benmerah A, Boquet P: The p21 Rho-activating toxin cytotoxic necrotizing factor 1 is endocytosed by a clathrin-independent mechanism and enters the cytosol by an acidic-dependent membrane translocation step. Mol Biol Cell. 2000, 11: 1775-1787.PubMedPubMedCentralCrossRef Contamin S, Galmiche A, Doye A, Flatau G, Benmerah A, Boquet P: The p21 Rho-activating toxin cytotoxic necrotizing factor 1 is endocytosed by a clathrin-independent mechanism and enters the cytosol by an acidic-dependent membrane translocation step. Mol Biol Cell. 2000, 11: 1775-1787.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Kim KJ, Chung JW, Kim KS: 67-kDa laminin receptor promotes internalization of cytotoxic necrotizing factor 1-expressing Escherichia coli K1 into human brain microvascular endothelial cells. J Biol Chem. 2005, 280: 1360-1368. 10.1074/jbc.M410176200.PubMedCrossRef Kim KJ, Chung JW, Kim KS: 67-kDa laminin receptor promotes internalization of cytotoxic necrotizing factor 1-expressing Escherichia coli K1 into human brain microvascular endothelial cells. J Biol Chem. 2005, 280: 1360-1368. 10.1074/jbc.M410176200.PubMedCrossRef
49.
Zurück zum Zitat Pei S, Doye A, Boquet P: Mutation of specific acidic residues of the CNF1 T domain into lysine alters cell membrane translocation of the toxin. Mol Microbiol. 2001, 41: 1237-1247. 10.1046/j.1365-2958.2001.02596.x.PubMedCrossRef Pei S, Doye A, Boquet P: Mutation of specific acidic residues of the CNF1 T domain into lysine alters cell membrane translocation of the toxin. Mol Microbiol. 2001, 41: 1237-1247. 10.1046/j.1365-2958.2001.02596.x.PubMedCrossRef
50.
Zurück zum Zitat Etienne-Manneville S, Hall A: Rho GTPases in cell biology. Nature. 2002, 420: 629-635. 10.1038/nature01148.PubMedCrossRef Etienne-Manneville S, Hall A: Rho GTPases in cell biology. Nature. 2002, 420: 629-635. 10.1038/nature01148.PubMedCrossRef
51.
Zurück zum Zitat Flatau G, Lemichez E, Gauthier M, Chardin P, Paris S, Fiorentini C, Boquet P: Toxin-induced activation of the G protein p21 Rho by deamidation of glutamine. J Biol Chem. 1997, 272: 19532-19537. 10.1074/jbc.272.31.19532.PubMedCrossRef Flatau G, Lemichez E, Gauthier M, Chardin P, Paris S, Fiorentini C, Boquet P: Toxin-induced activation of the G protein p21 Rho by deamidation of glutamine. J Biol Chem. 1997, 272: 19532-19537. 10.1074/jbc.272.31.19532.PubMedCrossRef
52.
Zurück zum Zitat Schmidt G, Sher P, Wilm M, Selzer J, Mann M, Aktories K: Gln 63 of Rho is deamidated by Escherichia coli cytotoxic necrotizing factor-1. Nature. 1997, 387: 725-729. 10.1038/42735.PubMedCrossRef Schmidt G, Sher P, Wilm M, Selzer J, Mann M, Aktories K: Gln 63 of Rho is deamidated by Escherichia coli cytotoxic necrotizing factor-1. Nature. 1997, 387: 725-729. 10.1038/42735.PubMedCrossRef
53.
Zurück zum Zitat Lerm M, Selzer J, Hoffmeyer A, Rapp UR, Aktories K, Schmidt G: Deamidation of Cdc42 and Rac by Escherichia coli cytotoxic necrotizing factor 1: activation of the C-Jun N-terminal kinase in HeLa cells. Infect Immun. 1999, 67: 496-503.PubMedPubMedCentral Lerm M, Selzer J, Hoffmeyer A, Rapp UR, Aktories K, Schmidt G: Deamidation of Cdc42 and Rac by Escherichia coli cytotoxic necrotizing factor 1: activation of the C-Jun N-terminal kinase in HeLa cells. Infect Immun. 1999, 67: 496-503.PubMedPubMedCentral
54.
Zurück zum Zitat Rittinger K, Walker PA, Eccleston JF, Nurmahomed K, Owen D, Laue E, Gamblin SJ, Smerdon SJ: Crystal structure of a small G protein in complex with the GTPase-activating protein rhoGAP. Nature. 1997, 388: 693-697. 10.1038/41805.PubMedCrossRef Rittinger K, Walker PA, Eccleston JF, Nurmahomed K, Owen D, Laue E, Gamblin SJ, Smerdon SJ: Crystal structure of a small G protein in complex with the GTPase-activating protein rhoGAP. Nature. 1997, 388: 693-697. 10.1038/41805.PubMedCrossRef
55.
Zurück zum Zitat Bos JL: Ras oncogenes in human cancer: a review. Cancer Res. 1989, 49: 4682-4689.PubMed Bos JL: Ras oncogenes in human cancer: a review. Cancer Res. 1989, 49: 4682-4689.PubMed
56.
Zurück zum Zitat Fiorentini C, Arancia G, Caprioli A, Falbo V, Ruggeri FM, Donelli G: Cytoskeletal changes induced in HEp-2 cells by the cytotoxic necrotizing factor of Escherichia coli. Toxicon. 1988, 26: 1047-1056. 10.1016/0041-0101(88)90203-6.PubMedCrossRef Fiorentini C, Arancia G, Caprioli A, Falbo V, Ruggeri FM, Donelli G: Cytoskeletal changes induced in HEp-2 cells by the cytotoxic necrotizing factor of Escherichia coli. Toxicon. 1988, 26: 1047-1056. 10.1016/0041-0101(88)90203-6.PubMedCrossRef
57.
Zurück zum Zitat Falzano L, Fiorentini C, Donelli G, Michel E, Kocks C, Cossart P, Cabanié L, Oswald E, Boquet P: Induction of phagocytic behaviour in human epithelial cells by Escherichia coli cytotoxic necrotizing factor type 1. Mol Microbiol. 1993, 9: 1247-1254. 10.1111/j.1365-2958.1993.tb01254.x.PubMedCrossRef Falzano L, Fiorentini C, Donelli G, Michel E, Kocks C, Cossart P, Cabanié L, Oswald E, Boquet P: Induction of phagocytic behaviour in human epithelial cells by Escherichia coli cytotoxic necrotizing factor type 1. Mol Microbiol. 1993, 9: 1247-1254. 10.1111/j.1365-2958.1993.tb01254.x.PubMedCrossRef
58.
Zurück zum Zitat Fiorentini C, Falzano L, Fabbri A, Stringaro A, Logozzi M, Travaglione S, Contamin S, Arancia G, Malorni W, Fais S: Activation of rho GTPases by cytotoxic necrotizing factor 1 induces macropinocytosis and scavenging activity in epithelial cells. Mol Biol Cell. 2001, 12: 2061-2073.PubMedPubMedCentralCrossRef Fiorentini C, Falzano L, Fabbri A, Stringaro A, Logozzi M, Travaglione S, Contamin S, Arancia G, Malorni W, Fais S: Activation of rho GTPases by cytotoxic necrotizing factor 1 induces macropinocytosis and scavenging activity in epithelial cells. Mol Biol Cell. 2001, 12: 2061-2073.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Doye A, Mettouchi A, Bossis G, Clement R, Buisson-Touati C, Flatau G, Gagnoux L, Piechaczyk M, Boquet P, Lemichez E: CNF1 exploits the ubiquitin-proteasome machinery to restrict Rho GTPase activation for bacterial host cell invasion. Cell. 2002, 111: 553-564. 10.1016/S0092-8674(02)01132-7.PubMedCrossRef Doye A, Mettouchi A, Bossis G, Clement R, Buisson-Touati C, Flatau G, Gagnoux L, Piechaczyk M, Boquet P, Lemichez E: CNF1 exploits the ubiquitin-proteasome machinery to restrict Rho GTPase activation for bacterial host cell invasion. Cell. 2002, 111: 553-564. 10.1016/S0092-8674(02)01132-7.PubMedCrossRef
60.
Zurück zum Zitat Fu Y, Galàn JE: A Salmonella protein antagonizes Rac1 and Cdc42 to mediate host-cell recovery after bacterial invasion. Nature. 1999, 401: 293-297. 10.1038/45829.PubMedCrossRef Fu Y, Galàn JE: A Salmonella protein antagonizes Rac1 and Cdc42 to mediate host-cell recovery after bacterial invasion. Nature. 1999, 401: 293-297. 10.1038/45829.PubMedCrossRef
61.
Zurück zum Zitat Schlumberger MC, Hardt WD: Triggered phagocytosis by Salmonella : bacterial molecular mimicry of RhoGTPase activation/deactivation. Curr Top Microbiol Immunol. 2005, 291: 29-42.PubMed Schlumberger MC, Hardt WD: Triggered phagocytosis by Salmonella : bacterial molecular mimicry of RhoGTPase activation/deactivation. Curr Top Microbiol Immunol. 2005, 291: 29-42.PubMed
62.
Zurück zum Zitat Ridley AJ: Rho proteins and cancer. Breast Cancer res Treat. 2004, 84: 13-19. 10.1023/B:BREA.0000018423.47497.c6.PubMedCrossRef Ridley AJ: Rho proteins and cancer. Breast Cancer res Treat. 2004, 84: 13-19. 10.1023/B:BREA.0000018423.47497.c6.PubMedCrossRef
63.
Zurück zum Zitat Kleer CG, van Golen KL, Zhang Y, Wu ZF, Rubin MA, Merajver SD: Characterization of RhoC expression in benign and malignant breast disease. Am J Pathol. 2002, 160: 579-584.PubMedPubMedCentralCrossRef Kleer CG, van Golen KL, Zhang Y, Wu ZF, Rubin MA, Merajver SD: Characterization of RhoC expression in benign and malignant breast disease. Am J Pathol. 2002, 160: 579-584.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Marx J: Inflammation and cancer: the link grows stronger. Cancer Res. 2004, 306 (5698): 966-968. Marx J: Inflammation and cancer: the link grows stronger. Cancer Res. 2004, 306 (5698): 966-968.
65.
Zurück zum Zitat Ghosh S, May MJ, Kopp EB: NF-kappa B and Rel proteins: evolutionarily conserved mediators of immune responses. Annu Rev Immunol. 1998, 16: 225-260. 10.1146/annurev.immunol.16.1.225.PubMedCrossRef Ghosh S, May MJ, Kopp EB: NF-kappa B and Rel proteins: evolutionarily conserved mediators of immune responses. Annu Rev Immunol. 1998, 16: 225-260. 10.1146/annurev.immunol.16.1.225.PubMedCrossRef
66.
Zurück zum Zitat Boyer L, Travaglione S, Falzano L, Gauthier NC, Popoff MR, Lemichez E, Fiorentini C, Fabbri A: Rac GTPase instructs Nuclear Factor-kB activating by conveying the SCF complex and IkB-alpha to the ruffling membranes. Mol Biol Cell. 2004, 15: 1124-1133. 10.1091/mbc.E03-05-0301.PubMedPubMedCentralCrossRef Boyer L, Travaglione S, Falzano L, Gauthier NC, Popoff MR, Lemichez E, Fiorentini C, Fabbri A: Rac GTPase instructs Nuclear Factor-kB activating by conveying the SCF complex and IkB-alpha to the ruffling membranes. Mol Biol Cell. 2004, 15: 1124-1133. 10.1091/mbc.E03-05-0301.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Fiorentini C, Matarrese P, Straface E, Falzano L, Donelli G, Boquet P, Malorni W: Rho-dependent cell spreading activated by E. coli cytotoxic necrotizing factor 1 hinders apoptosis in epithelial cells. Cell Death Differ. 1998, 5: 720-728. 10.1038/sj.cdd.4400412.PubMedCrossRef Fiorentini C, Matarrese P, Straface E, Falzano L, Donelli G, Boquet P, Malorni W: Rho-dependent cell spreading activated by E. coli cytotoxic necrotizing factor 1 hinders apoptosis in epithelial cells. Cell Death Differ. 1998, 5: 720-728. 10.1038/sj.cdd.4400412.PubMedCrossRef
68.
Zurück zum Zitat Fiorentini C, Matarrese P, Straface E, Falzano L, Fabbri A, Donelli G, Cossarizza A, Boquet P, Malorni W: Toxin-induced activation of Rho GTP-binding protein increases Bcl-2 expression and influences mitochondrial homeostasis. Exp Cell Res. 1998, 242: 341-350. 10.1006/excr.1998.4057.PubMedCrossRef Fiorentini C, Matarrese P, Straface E, Falzano L, Fabbri A, Donelli G, Cossarizza A, Boquet P, Malorni W: Toxin-induced activation of Rho GTP-binding protein increases Bcl-2 expression and influences mitochondrial homeostasis. Exp Cell Res. 1998, 242: 341-350. 10.1006/excr.1998.4057.PubMedCrossRef
69.
Zurück zum Zitat Giamboi Miraglia A, Travaglione S, Meschini S, Falzano L, Matarrese P, Quaranta MG, Viora M, Fiorentini C, Fabbri A: Cytotoxic necrotizing factor 1 prevents apoptosis via the Akt/IkappaB kinase pathway: role of nuclear factor-kappaB and Bcl-2. Mol Biol Cell. 2007, 18: 2735-2744. 10.1091/mbc.E06-10-0910.CrossRef Giamboi Miraglia A, Travaglione S, Meschini S, Falzano L, Matarrese P, Quaranta MG, Viora M, Fiorentini C, Fabbri A: Cytotoxic necrotizing factor 1 prevents apoptosis via the Akt/IkappaB kinase pathway: role of nuclear factor-kappaB and Bcl-2. Mol Biol Cell. 2007, 18: 2735-2744. 10.1091/mbc.E06-10-0910.CrossRef
70.
Zurück zum Zitat Kowaltowski AJ, Cosso RG, Campos CB, Fiskum G: Effect of bcl-2 overexpression on mitochondrial structure and function. J Biol Chem. 2002, 277: 42802-42807. 10.1074/jbc.M207765200.PubMedCrossRef Kowaltowski AJ, Cosso RG, Campos CB, Fiskum G: Effect of bcl-2 overexpression on mitochondrial structure and function. J Biol Chem. 2002, 277: 42802-42807. 10.1074/jbc.M207765200.PubMedCrossRef
71.
Zurück zum Zitat Cereghetti GM, Scorrano L: The many shape of mitochondrial death. Oncogene. 2006, 25: 4717-4724. 10.1038/sj.onc.1209605.PubMedCrossRef Cereghetti GM, Scorrano L: The many shape of mitochondrial death. Oncogene. 2006, 25: 4717-4724. 10.1038/sj.onc.1209605.PubMedCrossRef
72.
Zurück zum Zitat Delivani P, Adrain C, Taylor RC, Duriez PJ, Martin SJ: Role for CED-9 and Egl-1 as regulators of mitochondrial fission and fusion dynamics. Mol Cell. 2006, 21: 761-773. 10.1016/j.molcel.2006.01.034.PubMedCrossRef Delivani P, Adrain C, Taylor RC, Duriez PJ, Martin SJ: Role for CED-9 and Egl-1 as regulators of mitochondrial fission and fusion dynamics. Mol Cell. 2006, 21: 761-773. 10.1016/j.molcel.2006.01.034.PubMedCrossRef
73.
Zurück zum Zitat Alirol E, Martinou JC: Mitochondria and cancer: is there a morphological connection?. Oncogene. 2006, 25: 4706-4716. 10.1038/sj.onc.1209600.PubMedCrossRef Alirol E, Martinou JC: Mitochondria and cancer: is there a morphological connection?. Oncogene. 2006, 25: 4706-4716. 10.1038/sj.onc.1209600.PubMedCrossRef
74.
Zurück zum Zitat Falzano L, Quaranta MG, Travaglione S, Filippini P, Fabbri A, Viora M, Donelli G, Fiorentini C: Cytotoxic necrotizing factor 1 enhances reactive oxygen species-dependent transcription and secretion of proinflammatory cytokines in human uroepithelial cells. Infect Immun. 2003, 71: 4178-4181. 10.1128/IAI.71.7.4178-4181.2003.PubMedPubMedCentralCrossRef Falzano L, Quaranta MG, Travaglione S, Filippini P, Fabbri A, Viora M, Donelli G, Fiorentini C: Cytotoxic necrotizing factor 1 enhances reactive oxygen species-dependent transcription and secretion of proinflammatory cytokines in human uroepithelial cells. Infect Immun. 2003, 71: 4178-4181. 10.1128/IAI.71.7.4178-4181.2003.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Munro P, Flatau G, Doye A, Boyer L, Oregioni O, Mege JL, Landraud L, Lemichez E: Activation and proteasomal degradation of Rho GTPases by cytotoxic necrotizing factor-1 elicit a controlled inflammatory response. J Biol Chem. 2004, 279: 35849-35857. 10.1074/jbc.M401580200.PubMedCrossRef Munro P, Flatau G, Doye A, Boyer L, Oregioni O, Mege JL, Landraud L, Lemichez E: Activation and proteasomal degradation of Rho GTPases by cytotoxic necrotizing factor-1 elicit a controlled inflammatory response. J Biol Chem. 2004, 279: 35849-35857. 10.1074/jbc.M401580200.PubMedCrossRef
76.
Zurück zum Zitat Thomas W, Ascott ZK, Harmey D, Slice LW, Rozengurt E, Lax AJ: Cytotoxic necrotizing factor from Escherichia coli induces RhoA-dependent expression of the cyclooxygenase-2 gene. Infect Immun. 2001, 69: 6839-6845. 10.1128/IAI.69.11.6839-6845.2001.PubMedPubMedCentralCrossRef Thomas W, Ascott ZK, Harmey D, Slice LW, Rozengurt E, Lax AJ: Cytotoxic necrotizing factor from Escherichia coli induces RhoA-dependent expression of the cyclooxygenase-2 gene. Infect Immun. 2001, 69: 6839-6845. 10.1128/IAI.69.11.6839-6845.2001.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Mann JR, Dubois RN: Cyclooxygenase-2 and gastrointestinal cancer. Cancer J. 2004, 10: 145-152.PubMedCrossRef Mann JR, Dubois RN: Cyclooxygenase-2 and gastrointestinal cancer. Cancer J. 2004, 10: 145-152.PubMedCrossRef
78.
Zurück zum Zitat Wang D, Dubois RN: Cyclooxygenase-2: a potential target in breast cancer. Semin Oncol. 2004, 31: 64-73. 10.1053/j.seminoncol.2004.01.008.PubMedCrossRef Wang D, Dubois RN: Cyclooxygenase-2: a potential target in breast cancer. Semin Oncol. 2004, 31: 64-73. 10.1053/j.seminoncol.2004.01.008.PubMedCrossRef
79.
Zurück zum Zitat Ristimaki A: Cyclooxygenase 2: from inflammation to carcinogenesis. Novartis Found Symp. 2004, 256: 215-221.PubMedCrossRef Ristimaki A: Cyclooxygenase 2: from inflammation to carcinogenesis. Novartis Found Symp. 2004, 256: 215-221.PubMedCrossRef
80.
Zurück zum Zitat Malorni W, Fiorentini C: Is the Rac GTPase-activating toxin CNF1 a smart hijacker of host cell fate?. FASEB J. 2006, 20: 606-609. 10.1096/fj.05-4706hyp.PubMedCrossRef Malorni W, Fiorentini C: Is the Rac GTPase-activating toxin CNF1 a smart hijacker of host cell fate?. FASEB J. 2006, 20: 606-609. 10.1096/fj.05-4706hyp.PubMedCrossRef
81.
Zurück zum Zitat Fiorentini C, Malorni W: Exploiting cell death pathways by an E. coli cytotoxin: autophagy as a double-edged sword for the host. Autophagy. 2006, 2: 310-311.PubMedCrossRef Fiorentini C, Malorni W: Exploiting cell death pathways by an E. coli cytotoxin: autophagy as a double-edged sword for the host. Autophagy. 2006, 2: 310-311.PubMedCrossRef
82.
Zurück zum Zitat Falzano L, Filippini P, Travaglione S, Miraglia AG, Fabbri A, Fiorentini C: Escherichia coli cytotoxic necrotizing factor 1 blocks cell cycle G2/M transition in uroepithelial cells. Infect Immun. 2006, 74: 3765-3772. 10.1128/IAI.01413-05.PubMedPubMedCentralCrossRef Falzano L, Filippini P, Travaglione S, Miraglia AG, Fabbri A, Fiorentini C: Escherichia coli cytotoxic necrotizing factor 1 blocks cell cycle G2/M transition in uroepithelial cells. Infect Immun. 2006, 74: 3765-3772. 10.1128/IAI.01413-05.PubMedPubMedCentralCrossRef
Metadaten
Titel
The Rho-activating CNF1 toxin from pathogenic E. coli: A risk factor for human cancer development?
verfasst von
Sara Travaglione
Alessia Fabbri
Carla Fiorentini
Publikationsdatum
01.12.2008
Verlag
BioMed Central
Erschienen in
Infectious Agents and Cancer / Ausgabe 1/2008
Elektronische ISSN: 1750-9378
DOI
https://doi.org/10.1186/1750-9378-3-4

Weitere Artikel der Ausgabe 1/2008

Infectious Agents and Cancer 1/2008 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.