Next Article in Journal
The Effect of Impaired Polyamine Transport on Pneumococcal Transcriptome
Previous Article in Journal
Emergence and Characterization of a Novel Reassortant Canine Influenza Virus Isolated from Cats
Previous Article in Special Issue
Host–Pathogen Interactions of Chlamydia trachomatis in Porcine Oviduct Epithelial Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bacteria–Cancer Interface: Awaiting the Perfect Storm

by
Jonathan Pommer Hansen
1,
Waled Mohammed Ali
1,
Rajeeve Sivadasan
2 and
Karthika Rajeeve
1,3,*
1
Department of Biomedicine, The Skou Building, Høegh-Guldbergs Gade 10, 8000 Aarhus, Denmark
2
Neurobiology Laboratory, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695014, India
3
Pathogen Biology Laboratory, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695014, India
*
Author to whom correspondence should be addressed.
Pathogens 2021, 10(10), 1321; https://doi.org/10.3390/pathogens10101321
Submission received: 19 August 2021 / Revised: 11 October 2021 / Accepted: 11 October 2021 / Published: 14 October 2021
(This article belongs to the Special Issue Unraveling Chlamydial Pathogenesis)

Abstract

:
Epidemiological evidence reveal a very close association of malignancies with chronic inflammation as a result of persistent bacterial infection. Recently, more studies have provided experimental evidence for an etiological role of bacterial factors disposing infected tissue towards carcinoma. When healthy cells accumulate genomic insults resulting in DNA damage, they may sustain proliferative signalling, resist apoptotic signals, evade growth suppressors, enable replicative immortality, and induce angiogenesis, thus boosting active invasion and metastasis. Moreover, these cells must be able to deregulate cellular energetics and have the ability to evade immune destruction. How bacterial infection leads to mutations and enriches a tumour-promoting inflammatory response or micro-environment is still not clear. In this review we showcase well-studied bacteria and their virulence factors that are tightly associated with carcinoma and the various mechanisms and pathways that could have carcinogenic properties.

1. Introduction

Genetic alterations that are inherited or acquired during a person’s lifetime can lead to errors in cell division and uncontrolled growth. External factors leading to these genetic mutations largely include exposure to radiation, smoking, and infectious microorganisms such as viruses, bacteria, and parasites. Data from the Global Cancer Observatory (GLOBOCAN) attribute 2.2 million (13%) new cancer cases to 10 carcinogenic pathogens. The International Agency for Research on Cancer (IARC) has classified these carcinogens under six viruses (Epstein—Barr virus, human papillomavirus, hepatitis virus B, hepatitis virus C, human herpesvirus type 8 and human T cell lymphotropic virus type 1), one bacterium (Helicobacter pylori) and three parasites (Opisthorchis viverrine, Clonorchis sinensis, and Schistosoma haematobium). These classifications do not include the pathogens such as the human immunodeficiency virus that results in immunosuppression and enhances the carcinogenic action of viruses and probably bacteria. While only one bacterium has so far been included to the IARC’s list of carcinogenic pathogens, many other bacteria have been discovered to have carcinogenic effect. In this review we will shed light into all the known cancer-associated bacterium (Table 1, Figure 1) and discuss the factors and pathways that the pathogens activate to boost malignancies. The bacterial infection can contribute to carcinoma by favouring inflammatory processes and the release of carcinogenic bacterial effectors.

1.1. Helicobacter pylori

H. pylori is a Gram-negative bacterium, that has established lifelong colonization in the stomach of two-thirds of the world’s population with the highest prevalence in Africa [35]. According to the GLOBOCAN database, H. pylori is responsible for 36.8% of gastric cancers out of the estimated 2.2 million cancers attributed to infections [36]. From the 850,000 non-cardia gastric cancer cases worldwide in 2018, 89% of the total incidence are attributable to H. pylori infections, whereas around 73% of the non-Hodgkin lymphoma of gastric location are attributable to H. pylori infections [36]. Furthermore, H. pylori is the only bacterium classified as a group 1 carcinogen by IARC.
As a result of both intra and intergenomic diversification, H. pylori strains have shown to appear with a broad genetic diversity [37]. Certain strain-specific proteins such as vacuolating cytotoxin A (VacA) and cytotoxin-associated gene A (CagA) are shown to have increased risk towards carcinoma [38,39,40,41]. VacA is a channel forming toxin capable of inducing vacuoles into epithelial cells and has long been associated with the development of gastric inflammation [38]. While the exact oncogenic mechanisms have not yet been established, potential pathways have been suggested: VacA was found to cause apoptosis in gastro-epithelial cells leading to increased cell proliferation or a possible entry point for other carcinogens into the gastric mucosa [38]. VacA can activate the phosphoinositide 3-kinase (PI3K/AKT) pathway, resulting in the inhibition of glycogen synthase kinase 3 beta (GSK3β) via phosphorylation, with the subsequent dissociation of the GSK3β/β-catenin complex [42].
CagA is shown to have a direct oncogenic effect and its expression can lead to atrophic gastritis and gastric cancer [43]. The bacterium uses type IV secretion system (T4SS) to inject CagA into gastric epithelial cells; the protein undergoes tyrosine phosphorylation by the proto-oncogene tyrosine protein kinase (SRC) at the EPIYA phosphorylation motif (Glu-Pro-Ile-Tyr-Ala amino acid sequence). Phosphorylated CagA activates SHP-2 (SRC homology region 2 domain-containing phosphatase-2), a tyrosine phosphatase and known oncoprotein, which potentially activates β-catenin, Janus kinases/signal transducer and activator of transcription proteins (JAK/STAT) [44] and nuclear factor like kappa B (NF-kB) [45] pathways resulting in targeted transcriptional upregulation of genes involved in carcinogenesis [46]. c-Src inactivation induced by CagA leads to tyrosine dephosphorylation of an actin binding protein, cortactin, thus rearranging the actin cytoskeleton [47,48]. Cortactin expression promotes the migration, invasion, and proliferation of cells in vitro and in vivo, conferring malignancy [49,50]. Recently, Helicobacter was reported to upregulate cortactin in a CagA and JNK-dependent pathway which might partially explain the mechanism of Helicobacter driven carcinogenic process [51] (Figure 2).
CagA activates the extracellular signal-regulated kinases/mitogen-activated protein kinase (ERK/MAPK) pathway through SHP2, resulting in morphological changes in the epithelium called hummingbird phenotype in a Ras-independent manner [52]. CagA also interacts with the E-cadherin/β-catenin complex resulting in nuclear accumulation of β-catenin leading to transdifferentiation of gastric epithelial cells [53]. The bacteria can induce the epithelial-to-mesenchymal transition (EMT) mediated by Snail, a transcriptional repressor of E-Cadherin expression, in gastric epithelial cells by GSK3β depletion [54]. The tumour suppressor TP53 undergoes proteasomal degradation leading to DNA damage in the presence of CagA [55]. Most interestingly, the transgenic expression of CagA in mice induces gastrointestinal and hematopoietic neoplasms, revealing the direct carcinogenic potential of the protein [41].
Other proteins that are associated with H. pylori, which increases the risk of gastric cancer, are the outer membrane proteins (OMPs) such as blood-group antigen-binding adhesin (BabA) and outer inflammatory protein A (OipA). BabA binds to human Lewis(b) surface epitopes, which results in increased levels of specific cytokines causing cell proliferation. OipA activates β-catenin through binding of epidermal growth factor receptor and activation of PI3K-AKT signalling [56,57] (Figure 2). The eradication of H. pylori, in both healthy individuals and individuals with gastric neoplasia, reduces the incidence of gastric cancer and hence the mortality from gastric cancer [58].

1.2. Salmonella enterica and Salmonella typhi

Salmonella, a Gram-negative bacterium from the Enterobacteriaceae family of which subspecies Salmonella Enteritidis (S. enteritidis) and Salmonella typhi (S. typhi) have been associated with an increased risk of colon cancer [63] and gallbladder carcinoma, respectively [10,11,64]. Salmonella enterica infection is known to modulate inflammatory response of the host leading to DNA damage, increased cell proliferation, and migration leading to neoplasm and eventually cancer [65,66]. Salmonella effector proteins have been shown to be associated with colon cancer. The first effector protein is typhoid toxin, a cyclomodulin, that favours dysbiosis and leads to increased cell survival, risking the development of inflammatory bowel disease and colon cancer [67,68]. The second effector protein is Avr, secreted by T3SS and detected in the stool of patients with colon cancer [69]. Avr decreases inflammation as it suppresses secretion of cytokines such as IL-12, IL-6, IL-20, TNF-α, and interferon-γ. Avr is shown to directly activate the Wnt/catenin pathway favouring tumour formation in intestinal epithelium [70]. The acetyl transferase activity of Avr targets p53 [71], leading to cell cycle arrest and apoptosis inhibition by depleting the pro-apoptotic protein Bax [72]. Finally, the effector protein AvrA was found to increase the STAT3 signalling pathway in colon cancer [73]. STAT3 is a transcription factor, known to increase cell proliferation and inflammatory regulation and promote tumorigenesis.
Salmonella sp. enters the gall bladder through the bloodstream or the bile [74]. Several studies have shown the presence of Salmonella enterica, S. typhi, S. paratyphi, S. typhimurium in tissue biopsies of gall bladder carcinoma [75]. Salmonella has the ability to form biofilms on gall stones and can persist in the infected tissue [76]. The inflammation caused during infection may cause immune cell recruitment including macrophages that express COX-2, an enzyme that is crucial in the development of gastro-intestinal tumors [77,78]. A study done on murine gallbladder organoids reports that Salmonella enterica was not a single causal factor, but a part of a multistep process in the development of gallbladder carcinoma [10]. Overexpression of the proto-oncogene c-Myc and silencing of tumour suppressor p53, together with the presence of the MAPK and AKT pathways, was necessary to provoke cellular transformation in the organoids [10]. Salmonella enterica was shown to activate MAPK and AKT pathways through effector proteins such as SopB, SopE and SopE2, injected by T3SS into host cells to regulate bacterial uptake and secure intracellular survival through cell transformation [10] (Figure 3). However, more studies are required to study which patients are predisposed to Salmonella-induced carcinoma.

1.3. Escherichia coli

Escherichia coli (E. coli) is a Gram-negative, facultative anaerobe commensal bacteria found in the gut. Studies have frequently found strong associations between mucosa-adherent E. coli strains and colorectal cancer (CRC) [79]. Genotoxins are produced by bacteria as a means of competing with other microbes. The cyclomodulin-positive B2 E. coli strain possesses the polyketide synthase genetic islands (pks +ve), coding for the bacterial genotoxin colibactin. This E. coli strain abnormally colonizes the mucosa of colon cancer patients, promoting low grade inflammation and cell proliferation [80]. Colibactin has shown carcinogenic properties by inducing double-stranded DNA breaks and cell cycle arrest [81]. This activates G2-M DNA damage checkpoint pathway where the mismatch repair proteins MSH2 and MLH1 are effectively depleted in colonic cells through T3SS-induced effector proteins [81]. Colibactin was shown to promote colon tumour growth by induing senescence-associated secretory phenotype through increased p53 SUMOylation [82]. In vitro studies show that the bacteria can alter tumour microenvironment by inducing the three major hallmarks of cancer: EMT, altering the cancer stem cell population, and metabolic reprogramming [83]. Most interestingly the etiological role of colibactin in colorectal carcinoma was evident when the DNA damage signature induced by colibactin corresponded to mutational hotspots in the genome of colorectal cancer patients [5,84]. Nevertheless, whether an already existing colon cancer creates a favourable environment for the B2 E. coli to colonize the colonic epithelial cells [82], or whether E. coli is a causal agent for CRC is still unclear (Figure 4).

1.4. Chlamydia trachomatis and pneumoniae

The Chlamydiaceae family is a group of obligate intracellular bacteria, with the subspecies Chlamydia trachomatis (C. trachomatis) capable of causing sexually transmitted infections and Chlamydia pneumoniae (C. pneumoniae) capable of causing lung infections. Both these bacteria cause asymptomatic and chronic infections. The first associations between C. pneumoniae and lung cancer goes far back, however, newer studies have found varying results when trying to establish a reliable biomarker in measuring chronic C. pneumoniae infection and linking it to lung cancer [85,86]. Most often the measure of titre immunoglobulin A was used, in which some studies found associations, while others found none. Newer studies found associations by measuring Chlamydia heat shock protein-60 antigen, another marker used to indicate chronic Chlamydia infection. The conflicting results could be caused by the modest reliability of micro-immunofluorescence. Despite controversial results, most studies suggest that chronic C. pneumoniae infection did associate with lung cancer [87,88]. Recent studies have indicated that the association may not only be mediated by inflammation, but also epigenetic changes, such as transformation of mesothelial cells and dysregulation in replication, transcription, and DNA repair mechanisms in the host cell [89]. How C. pneumoniae induces these epigenetic changes is unclear and should be looked further into.
While human papillomavirus (HPV) is well known as the most common cause of cervical cancer inflicted by infections, studies indicate that C. trachomatis may not just be a comorbidity to HPV, but instead an independent predictor for cervical cancer. The first evidence of C. trachomatis associated to cancer was based on an epidemiological correlation between C. trachomatis reactive serum antibodies and the presence of cervical cancer [90,91,92]. Studies show the presence of chlamydia HSP60-1 antibodies associated with increased cervical cancer risk [93]. Recently, studies from two independent population showed women seropositive for C. trachomatis had twice the risk of developing ovarian carcinoma [94]. The mechanism by which C. trachomatis may lead to cancer development is described through its ability to cause the host cell DNA damage and an impairment of the host DNA damage response [30]. Increased levels of reactive oxygen species (ROS) [95] lead to oxidative DNA damage, resulting in the production of single-strand breaks [96]. The impairment of the host DNA damage response was caused by histone alterations and activation of other repair inhibitory cell mechanisms. Through inhibitions of base excision repairs, DNA damage was not repaired efficiently, resulting in genomic instability. C. trachomatis is known to strongly activate the PI3/MAPK survival signal and efficiently block cell death [97,98]. It also activates the anti-apoptotic protein cIAP-2. The bacterium is known to possess deubiquitinase Cdu1, which is known to stabilize the anti-apoptotic protein, Mcl-1 [99,100]. Moreover, C. trachomatis also downregulates the tumour suppressor p53 [27,31] via the PI3K and MDM2 pathways. The bacteria induce glutamine addiction in the infected cells by stabilizing the proto-oncogene c-Myc [26]. In addition, the infection remains asymptomatic by paralyzing the neutrophils, the first line of defense [32]. Persistent infection of C. trachomatis-inducing DNA damage, blocking cell death and preventing immune surveillance might lead to tissue damage and malignancies in the infected tissue (Figure 5).

1.5. Fusobacterium nucleatum

F. nucleatum is a Gram-negative obligate anaerobe bacterium commonly found in the oral cavity, known to cause opportunistic infections [101]. This oral commensal is a periodontal pathogen associated with a wide spectrum of human diseases. F. nucleatum has been associated with CRC through detection by quantitative polymerase chain reaction and 16S rRNA sequence [102]. Recently, F. nucleatum has been associated with additional cancer types, including oral, head and neck, oesophageal, cervical, and gastric cancers [103]. However, additional studies should be conducted to further back these associations.
The role of F. nucleatum in CRC is thought to be through multiple different pathways. Fusobacterium adhesin A (FadA) was found to bind and inhibit E-cadherin in xenograft mice. E-cadherin is a tumour suppressor, causing activation of β-catenin signalling, previously mentioned to promote cell proliferation [104]. Additionally, FadA increased expression of nuclear factor-κB (NF-κB) and pro-inflammatory interleukins such as IL-6, IL-8, and IL-18 leading to a proinflammatory microenvironment [104]. Finally, F. nucleatum was shown to induce anticancer immune response evasion through fusobacterial apoptosis protein 2 (Fap2) [105]. Fap2 was found to bind the TIGIT immune receptor, a receptor expressed on all natural killer cells (NK cells) and other immune cells, thus inhibiting NK cell cytotoxicity [105]. Additionally, TIGIT was expressed on tumour-infiltrating lymphocytes and Fap2 was found to inhibit T cell activities, effectively protecting F. nucleatum from immune cell attacks (Figure 6). Interestingly, it was shown that exosomes harbouring miR1246/92b-3p/27a-3p CXCL16 derived from F. nucleatum-infected colorectal cancer cells facilitated tumour metastasis [106]. Moreover, in vitro and in vivo studies show that the bacterial infection leads to upregulation of cytochrome p450 monooxygenase, which increases the invasiveness and migratory ability (EMT) of colorectal cancer cells via the TLR4/Keap1/NRF2 axis [107].

1.6. Streptococcus bovis

S. bovis is a group of Gram-positive, opportunistic bacteria, capable of causing endocarditis and bacteraemia. S. bovis has shown to associate with colorectal cancer (CRC), through a series of retrospective studies [108,109]. Over time it has become clearer, that the association was mostly derived from the subspecies Streptococcus gallolyticus (SG). While it was previously uncertain if the presence of SG in tumour tissue was caused by SG-promoting tumour development, or the result of the tumour enabling SG colonization, recent studies done on mice found that SG may in fact promote colorectal tumour development [110,111].
The study showed a clear indication that SG increased levels of β-catenin, c-Myc, and proliferative cell nuclear antigen (PCNA) resulting in cell proliferation in certain responsive cell lines. Additionally, silencing β-catenin through knockdown cells resulted in a completely abolished effect of SG on the increased cell proliferation, thereby indicating the important role of SG in activating β-catenin and promoting cell proliferation, resulting in potential tumour development. The pathway of how SG upregulated β-catenin is still unclear and should be further investigated [111] (Figure 7).

1.7. Mycoplasma

Mycoplasma is a group of small bacteria with no cell wall, consisting of more than 100 subspecies. Several species of Mycoplasma have been suspected to associate with cancer, with the most prominent being: M. hyorhinis, M. hominis, and M. genitalium, and a few mentions of M. penetrans and M. salivarium [112,113,114].
Few studies have attempted to find these associations, indicating a need for additional large-scale studies. Whether these bacteria can directly lead to malignancies is yet unclear. In relation to cancer, M. hyorhinis is the most widely studied and has shown to be associated with prostate cancer, gastric carcinoma, oesophageal cancer, lung cancer, and breast cancer. In recent studies, several of the bacterial proteins have been found to possibly have carcinogenic mechanisms. One study found a clear indication of M. hyorhinis activating the β-catenin signalling pathway in M. hyorhinis-induced gastric cancer cell motility. Additionally, the activation of β-catenin seemed to be induced by glycogen synthase kinase 3 beta (GSK3β) and the Wnt-receptor lipoprotein-receptor-related protein 6 (LRP6), while also finding interaction between LRP6 and the mycoplasmal membrane protein p37. When GSK3β did not bind and activate LRP6, no increased activation of β-catenin was found [34]. The p37 protein activates carcinogenic effects such as enabling immortality in mammalian cell lines [115] and promoting cell motility, migration, and invasion through the activation of metalloproteinase-2 and epidermal growth factor receptor in vitro [116] (Figure 8).

1.8. Bacteroides fragilis

B. fragilis is an anaerobe bacterium, part of the normal microbiota in the human colon, often categorized into nontoxigenic B. fragilis and enterotoxigenic B. fragilis (ETBF), capable of producing a heat-labile toxin [117]. Recently, gut microbiota have been shown to play a bigger role than previously anticipated in the development of CRC, and ETBF has been found to be prevalent in the colonic mucosa of CRC patients [117,118]. This data was additionally supported by a study on APC+/− mice, which showed the toxin from ETBF had carcinogenic properties through inflammatory cascades. The B. fragilis toxin (BFT) mediated E-cadherin cleavage, through an unidentified receptor, resulting in the activation of the Wnt/β-catenin signalling pathway [119]. Additionally, BFT was able to create a proinflammatory microenvironment through activation of STAT3 and possibly NF-κB. The study additionally found that IL-17 is a key proinflammatory mediator necessary for colon tumour development in the APC+/− mice [119] (Figure 9). The link between IL-17 and tumorigenesis is still unclear.

1.9. Neisseria gonorrhoeae

N. gonorrhoeae is an aerobic Gram-negative bacterium, well known for its diplococci shape, being one of the most frequent sexually transmitted diseases, and its capability to cause pelvic inflammatory disease. N. gonorrhoeae has widely been tested for its association with prostate cancer, often leading to controversial results, with large prospective cohort studies failing to find any association [120]. A more recent meta-analysis suggests that despite the varying results, N. gonorrhoea may in fact be associated with prostate cancer [21]. Other studies found associations between N. gonorrhoeae and bladder cancer in men [23]. Due to the vast amount of controversy, large-scale cohort studies should be conducted to support these associations. The mechanism behind the potential carcinogenic effect of N. gonorrhoeae is unclear and not well explored, likely due to its inconsistent associations. However, one study found N. gonorrhoeae caused DNA damage, increased the expression of p21 and p27 and decreased the expression of p53 in non-tumour epithelial cells. This could support the claim that N. gonorrhoeae may be carcinogenic, although this should be further explored [20] (Figure 10).

1.10. Enterococcus faecalis

E. faecalis is a facultative anaerobe Gram-positive bacterium commonly found in the oral cavity and intestines. E. faecalis populations have been found to be significantly increased in faeces of patients with CRC compared to healthy controls; however, the question of whether the role of E. faecalis is protective or carcinogenic remains [121]. One study found E. faecalis to activate Wnt/β-catenin signalling through coculturing with M1 macrophages, thereby inducing cell proliferation in murine primary colon epithelial cells [122]. Another study found the heat-killed E. faecalis EC-12 strain supressed β-catenin signalling, thus having a potential protective role in CRC [123].
A possible explanation could be caused by the isolation of different E. faecalis strains. Different strains, as the result of gene transfers, could lead to the bacterium becoming more or less virulent, resulting in different inflammatory responses [124]. A harmful role of E. faecalis has been suggested to be through its ability to induce intracellular ROS production, causing DNA instability and a lowered DNA repair response. Finally, infection seemed to induce a NF-κB-dominated inflammatory response [125] (Figure 11)

1.11. Clostridium septicum

C. septicum is an anaerobic, Gram-positive, gas-producing bacterium capable of forming spores. Infection with C. septicum is uncommon. Despite the low number of cases, studies have found strong indications of its associated with CRC [126,127]. It is not clear if this association is a coincidence or if any bacterial virulence factors are directly causing malignancy. A possible explanation of this association has been suggested to be that the tumour creates a hypoxic and acidic microenvironment through anaerobic glycolysis, which favours the germination of the C. septicum spores, indicating that the bacterium is not the causal agent of the tumour [128]. A direct involvement of C. septicum in CRC has not yet been found.

1.12. Mycobacterium tuberculosis

Mycobacterium tuberculosis (MBT) is an intracellular pathogenic bacterium, capable of entering the respiratory airways, establishing lifelong infections. MBT has been well documented to be associated with increased risk of lung cancer [129]. A recent study found strong associations with cancer, specifically from the M. tuberculosis L form (MBT-L), a wall defective and pleomorphic form of MBT, which behaves much like carcinogenic viruses. The MBT-L had low activity and pathogenicity, which may often have gone undiagnosed causing possible misclassifications such as chronic lymphadenitis of unknown origin [130]. Recently a study on mice, found MBT inhibited T-cell-mediated cellular immune response by impairing T helper cells in the late stage of infection. MBT is reported to promote tumour metastasis in the lungs through increased programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) pathway activation [11] (Figure 12).

2. Discussion

Although a rather large group of bacteria has been shown to be associated with different cancer types, only H. pylori is currently accepted as a group 1 carcinogen, reflecting how the bacterial cancer field has not received the same amount of attention as the viral field. While some bacteria such as E. coli, S. bovis, and C. trachomatis have long been associated with cancer, studies focused on bacterial factors that directly predispose infected tissue towards carcinoma have only recently been reported. The bacterial effectors that can inflict mutations and result in malignancies in the infected tissue are yet to be discovered.
Several bacteria mentioned are commonly found in the human microbiome, yet they are associated with cancer. If a bacterium is causal for cancer, why is the cancer not seen more frequently? Even though cancer is one of the well-funded and focused research areas, much is still to be discovered. For a cell to turn malignant due to chronic infectious disease, the cell must accumulate the respective mutations in the DNA, or these epigenetic changes might imprint memory to activate indefinite survival signals leading to cell proliferation. Most of the showcased bacteria are capable of inflicting one or more of these hallmarks to an otherwise normal cell, but unless all the hallmarks are acquired, i.e., “the perfect storm has hit”, the cell may not be a fully functional cancer cell. If the bacteria truly are linked to the cause of cancer, they are not likely the only causal factor but instead a contribution in the multistep process required to fulfil the hallmarks of cancer, leaving the rest of the hallmarks to be acquired by other causes, such as radiation, smoking, aging, or inheritance Thus these chronic infections can increase the fitness of cells towards carcinoma.
Since many studies show that bacteria could be located at the site of tumour development, it is also debatable whether the bacteria might colocalize in the transformed tissue due to the excess nutrient availability and increased vascularization. This affinity for tumour tissue is exploited by scientists, by using attenuated strains of bacteria as adjuvants in the treatment of various neoplasms. The bacterial tropism for tumour microenvironment could also activate the innate and adaptive immune response of the host to clear malignant cells.
This review showcases that bacteria likely have a bigger role to play in the category of infectious cancers, and that this category, previously estimated to be 13% of all cancers, is likely underestimated. The bacteria included in this review are the most suggested bacteria to be associated with increased risk of cancer, yet there is still a lack of deeper understanding in how these bacteria may be associated to the cause of cancer. This highlights the need for more research to be done in this field with the likelihood of finding more bacteria, not mentioned in this review, to also have carcinogenic properties.

Author Contributions

This review was conceptualized by K.R., J.P.H., W.M.A., R.S. and K.R. was involved in the original draft preparation, writing, review and editing. K.R. supervised the review and acquired funding. All authors have read and agreed to the published version of the manuscript.

Funding

This review was funded by the DST SERB POWER grant, India and funding from AUFF Aarhus university Denmark.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tavares, R.; Pathak, S.K. Helicobacter pylori Protein JHP0290 Exhibits Proliferative and Anti-Apoptotic Effects in Gastric Epithelial Cells. PLoS ONE 2015, 10, e0124407. [Google Scholar] [CrossRef]
  2. Wang, H.; Sun, Y.; Liu, S.; Yu, H.; Li, W.; Zeng, J.; Chen, C.; Jia, J. Upregulation of progranulin byHelicobacter pyloriin human gastric epithelial cells via p38MAPK and MEK1/2 signaling pathway: Role in epithelial cell proliferation and migration. FEMS Immunol. Med. Microbiol. 2011, 63, 82–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Chen, Y.-C.; Wang, Y.; Li, J.-Y.; Xu, W.-R.; Zhang, Y.-L. H pyloristimulates proliferation of gastric cancer cells through activating mitogen-activated protein kinase cascade. World J. Gastroenterol. 2006, 12, 5972–5977. [Google Scholar] [CrossRef] [PubMed]
  4. Cuevas-Ramos, G.; Petit, C.R.; Marcq, I.; Boury, M.; Oswald, E.; Nougayrède, J.-P. Escherichia coli induces DNA damage in vivo and triggers genomic instability in mammalian cells. Proc. Natl. Acad. Sci. USA 2010, 107, 11537–11542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Dziubańska-Kusibab, P.J.; Berger, H.; Battistini, F.; Bouwman, B.A.M.; Iftekhar, A.; Katainen, R.; Cajuso, T.; Crosetto, N.; Orozco, M.; Aaltonen, L.A.; et al. Colibactin DNA-damage signature indicates mutational impact in colorectal cancer. Nat. Med. 2020, 26, 1063–1069. [Google Scholar] [CrossRef]
  6. Wilson, M.R.; Jiang, Y.; Villalta, P.W.; Stornetta, A.; Boudreau, P.D.; Carrá, A.; Brennan, C.A.; Chun, E.; Ngo, L.; Samson, L.D.; et al. The human gut bacterial genotoxin colibactin alkylates DNA. Science 2019, 363, eaar7785. [Google Scholar] [CrossRef]
  7. Abdulamir, A.S.; Hafidh, R.R.; Abu Bakar, F. The association of Streptococcus bovis/gallolyticus with colorectal tumors: The nature and the underlying mechanisms of its etiological role. J. Exp. Clin. Cancer Res. 2011, 30, 11. [Google Scholar] [CrossRef] [Green Version]
  8. Ellmerich, S.; Schöller, M.; Duranton, B.; Gossé, F.; Galluser, M.; Klein, J.-P.; Raul, F. Promotion of intestinal carcinogenesis by Streptococcus bovis. Carcinogenesis 2000, 21, 753–756. [Google Scholar] [CrossRef] [Green Version]
  9. Cano, D.A.; Martínez-Moya, M.; Pucciarelli, M.G.; Groisman, E.A.; Casadesús, J.; Portillo, F.G.-D. Salmonella enterica Serovar Typhimurium Response Involved in Attenuation of Pathogen Intracellular Proliferation. Infect. Immun. 2001, 69, 6463–6474. [Google Scholar] [CrossRef] [Green Version]
  10. Scanu, T.; Spaapen, R.; Bakker, J.M.; Pratap, C.B.; Wu, L.-E.; Hofland, I.; Broeks, A.; Shukla, V.; Kumar, M.; Janssen, H.; et al. Salmonella Manipulation of Host Signaling Pathways Provokes Cellular Transformation Associated with Gallbladder Carcinoma. Cell Host Microbe 2015, 17, 763–774. [Google Scholar] [CrossRef] [Green Version]
  11. Mughini-Gras, L.; Schaapveld, M.; Kramers, J.; Mooij, S.; Neefjes-Borst, A.; Van Pelt, W.; Neefjes, J. Increased colon cancer risk after severe Salmonella infection. PLoS ONE 2018, 13, e0189721. [Google Scholar] [CrossRef] [Green Version]
  12. Abed, J.; Maalouf, N.; Manson, A.L.; Earl, A.M.; Parhi, L.; Emgård, J.E.M.; Klutstein, M.; Tayeb, S.; Almogy, G.; Atlan, K.A.; et al. Colon Cancer-Associated Fusobacterium nucleatum May Originate from the Oral Cavity and Reach Colon Tumors via the Circulatory System. Front. Cell. Infect. Microbiol. 2020, 10, 400. [Google Scholar] [CrossRef]
  13. Wu, J.; Li, Q.; Fu, X. Fusobacterium nucleatum Contributes to the Carcinogenesis of Colorectal Cancer by Inducing Inflammation and Suppressing Host Immunity. Transl. Oncol. 2019, 12, 846–851. [Google Scholar] [CrossRef] [PubMed]
  14. Haghi, F.; Goli, E.; Mirzaei, B.; Zeighami, H. The association between fecal enterotoxigenic B. fragilis with colorectal cancer. BMC Cancer 2019, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Cao, Y.; Wang, Z.; Yan, Y.; Ji, L.; He, J.; Xuan, B.; Shen, C.; Ma, Y.; Jiang, S.; Ma, D.; et al. Enterotoxigenic Bacteroides fragilis Promotes Intestinal Inflammation and Malignancy by Inhibiting Exosome-Packaged miR-149-3p. Gastroenterology 2021. [Google Scholar] [CrossRef]
  16. Xie, X.; Jiang, D.; Zhou, X.; Ye, X.; Yang, P.; He, Y. Recombinant Bacteroides fragilis enterotoxin-1 (rBFT-1) promotes proliferation of colorectal cancer via CCL3-related molecular pathways. Open Life Sci. 2021, 16, 408–418. [Google Scholar] [CrossRef] [PubMed]
  17. Khan, Z.; Siddiqui, N.; Saif, M.W. Enterococcus Faecalis Infective Endocarditis and Colorectal Carcinoma: Case of New Association Gaining Ground. Gastroenterol. Res. 2018, 11, 238–240. [Google Scholar] [CrossRef] [Green Version]
  18. Zurmeyer, S.; Fotopoulou, C.; Braicu, E.; Schlichting, U.; Sehouli, J. Clostridium septicum can cause distant myonecrosis in patients with ovarian cancer. Anticancer. Res. 2013, 33, 1585–1589. [Google Scholar]
  19. Sidhu, J.S.; Mandal, A.; Virk, J.; Gayam, V. Early Detection of Colon Cancer Following Incidental Finding of Clostridium septicum Bacteremia. J. Investig. Med. High Impact Case Rep. 2019, 7, 2324709619832050. [Google Scholar] [CrossRef] [Green Version]
  20. Vielfort, K.; Söderholm, N.; Weyler, L.; Vare, D.; Löfmark, S.; Aro, H. Neisseria gonorrhoeae infection causes DNA damage and affects the expression of p21, p27, and p53 in non-tumor epithelial cells. J. Cell Sci. 2013, 126, 339–347. [Google Scholar] [CrossRef] [Green Version]
  21. Lian, W.-Q.; Luo, F.; Song, X.-L.; Lu, Y.-J.; Zhao, S.-C. Gonorrhea and Prostate Cancer Incidence: An Updated Meta-Analysis of 21 Epidemiologic Studies. Med. Sci. Monit. 2015, 21, 1895–1903. [Google Scholar] [CrossRef] [Green Version]
  22. Wang, Y.-C.; Chung, C.-H.; Chen, J.-H.; Chiang, M.-H.; Yin, T.-; Tsao, C.-H.; Lin, F.-H.; Chien, W.-C.; Shang, S.-T.; Chang, F.-Y. Gonorrhea infection increases the risk of prostate cancer in Asian population: A nationwide population-based cohort study. Eur. J. Clin. Microbiol. Infect. Dis. 2016, 36, 813–821. [Google Scholar] [CrossRef]
  23. Michaud, D.S.; Platz, E.A.; Giovannucci, E. Gonorrhoea and male bladder cancer in a prospective study. Br. J. Cancer 2006, 96, 169–171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Cao, S.; Li, J.; Lu, J.; Zhong, R.; Zhong, H. Mycobacterium tuberculosis antigens repress Th1 immune response suppression and promotes lung cancer metastasis through PD-1/PDl-1 signaling pathway. Cell Death Dis. 2019, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Wu, C.-Y.; Hu, H.-Y.; Pu, C.-Y.; Huang, N.; Shen, H.-C.; Li, C.-P.; Chou, Y.-J. Pulmonary tuberculosis increases the risk of lung cancer. Cancer 2010, 117, 618–624. [Google Scholar] [CrossRef]
  26. Rajeeve, K.; Vollmuth, N.; Janaki-Raman, S.; Wulff, T.F.; Baluapuri, A.; Dejure, F.R.; Huber, C.; Fink, J.; Schmalhofer, M.; Schmitz, W.; et al. Author Correction: Reprogramming of host glutamine metabolism during Chlamydia trachomatis infection and its key role in peptidoglycan synthesis. Nat. Microbiol. 2021, 6, 1. [Google Scholar] [CrossRef]
  27. Siegl, C.; Prusty, B.K.; Karunakaran, K.; Wischhusen, J.; Rudel, T. Tumor Suppressor p53 Alters Host Cell Metabolism to Limit Chlamydia trachomatis Infection. Cell Rep. 2014, 9, 918–929. [Google Scholar] [CrossRef] [Green Version]
  28. Sharma, M.; Machuy, N.; Böhme, L.; Karunakaran, K.; Mäurer, A.P.; Meyer, T.F.; Rudel, T. HIF-1α is involved in mediating apoptosis resistance to Chlamydia trachomatis-infected cells. Cell. Microbiol. 2011, 13, 1573–1585. [Google Scholar] [CrossRef] [PubMed]
  29. Kessler, M.; Hoffmann, K.; Fritsche, K.; Brinkmann, V.; Mollenkopf, H.-J.; Thieck, O.; Da Costa, A.R.T.; Braicu, E.I.; Sehouli, J.; Mangler, M.; et al. Chronic Chlamydia infection in human organoids increases stemness and promotes age-dependent CpG methylation. Nat. Commun. 2019, 10, 1194. [Google Scholar] [CrossRef]
  30. Chumduri, C.; Gurumurthy, R.K.; Zadora, P.K.; Mi, Y.; Meyer, T.F. Chlamydia Infection Promotes Host DNA Damage and Proliferation but Impairs the DNA Damage Response. Cell Host Microbe 2013, 13, 746–758. [Google Scholar] [CrossRef] [Green Version]
  31. Zou, Y.; Lei, W.; Su, S.; Bu, J.; Zhu, S.; Huang, Q.; Li, Z. Chlamydia trachomatis plasmid-encoded protein Pgp3 inhibits apoptosis via the PI3K-AKT-mediated MDM2-p53 axis. Mol. Cell. Biochem. 2018, 452, 167–176. [Google Scholar] [CrossRef] [PubMed]
  32. Rajeeve, K.; Das, S.; Prusty, B.K.; Rudel, T. Chlamydia trachomatis paralyses neutrophils to evade the host innate immune response. Nat. Microbiol. 2018, 3, 824–835. [Google Scholar] [CrossRef] [PubMed]
  33. Xu, Y.; Li, H.; Chen, W.; Yao, X.; Xing, Y.; Wang, X.; Zhong, J.; Meng, G. Mycoplasma hyorhinis Activates the NLRP3 Inflammasome and Promotes Migration and Invasion of Gastric Cancer Cells. PLoS ONE 2013, 8, e77955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Liu, X.; Rong, Z.; Shou, C. Mycoplasma hyorhinis infection promotes gastric cancer cell motility via β-catenin signaling. Cancer Med. 2019, 8, 5301–5312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Hooi, J.K.Y.; Lai, W.Y.; Ng, W.K.; Suen, M.M.; Underwood, F.E.; Tanyingoh, D.; Malfertheiner, P.; Graham, D.Y.; Wong, V.W.; Wu, J.C.; et al. Global Prevalence of Helicobacter pylori Infection: Systematic Review and Meta-Analysis. Gastroenterology 2017, 153, 420–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. de Martel, C.; Georges, D.; Bray, F.; Ferlay, J.; Clifford, G.M. Global burden of cancer attributable to infections in 2018: A worldwide incidence analysis. Lancet Glob. Health 2020, 8, e180–e190. [Google Scholar] [CrossRef] [Green Version]
  37. Dorer, M.S.; Talarico, S.; Salama, N.R. Helicobacter pylori’s Unconventional Role in Health and Disease. PLoS Pathog. 2009, 5, e1000544. [Google Scholar] [CrossRef]
  38. McClain, M.S.; Beckett, A.C.; Cover, T.L. Helicobacter pylori Vacuolating Toxin and Gastric Cancer. Toxins 2017, 9, 316. [Google Scholar] [CrossRef] [Green Version]
  39. Polk, D.B.; Peek, R.M., Jr. Helicobacter pylori: Gastric cancer and beyond. Nat. Rev. Cancer 2010, 10, 403–414. [Google Scholar] [CrossRef] [Green Version]
  40. Schulz, C.; Schütte, K.; Mayerle, J.; Malfertheiner, P. The role of the gastric bacterial microbiome in gastric cancer: Helicobacter pylori and beyond. Ther. Adv. Gastroenterol. 2019, 12, 1756284819894062. [Google Scholar] [CrossRef] [Green Version]
  41. Ohnishi, N.; Yuasa, H.; Tanaka, S.; Sawa, H.; Miura, M.; Matsui, A.; Higashi, H.; Musashi, M.; Iwabuchi, K.; Suzuki, M.; et al. Transgenic expression of Helicobacter pylori CagA induces gastrointestinal and hematopoietic neoplasms in mouse. Proc. Natl. Acad. Sci. USA 2008, 105, 1003–1008. [Google Scholar] [CrossRef] [Green Version]
  42. Nakayama, M.; Hisatsune, J.; Yamasaki, E.; Isomoto, H.; Kurazono, H.; Hatakeyama, M.; Azuma, T.; Yamaoka, Y.; Yahiro, K.; Moss, J.; et al. Helicobacter pylori VacA-induced Inhibition of GSK3 through the PI3K/Akt Signaling Pathway. J. Biol. Chem. 2009, 284, 1612–1619. [Google Scholar] [CrossRef] [Green Version]
  43. Hatakeyama, M. Helicobacter pylori CagA and Gastric Cancer: A Paradigm for Hit-and-Run Carcinogenesis. Cell Host Microbe 2014, 15, 306–316. [Google Scholar] [CrossRef] [Green Version]
  44. Umehara, S.; Higashi, H.; Ohnishi, N.; Asaka, M.; Hatakeyama, M. Effects of Helicobacter pylori CagA protein on the growth and survival of B lymphocytes, the origin of MALT lymphoma. Oncogene 2003, 22, 8337–8342. [Google Scholar] [CrossRef] [Green Version]
  45. Lamb, A.; Yang, X.-D.; Tsang, Y.N.; Li, J.-D.; Higashi, H.; Hatakeyama, M.; Peek, R.M.; Blanke, S.R.; Chen, L. Helicobacter pylori CagA activates NF-κB by targeting TAK1 for TRAF6-mediated Lys 63 ubiquitination. EMBO Rep. 2009, 10, 1242–1249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Higashi, H.; Tsutsumi, R.; Muto, S.; Sugiyama, T.; Azuma, T.; Asaka, M.; Hatakeyama, M. SHP-2 Tyrosine Phosphatase as an Intracellular Target of Helicobacter pylori CagA Protein. Science 2002, 295, 683–686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Sharafutdinov, I.; Backert, S.; Tegtmeyer, N. Cortactin: A Major Cellular Target of the Gastric Carcinogen Helicobacter pylori. Cancers 2020, 12, 159. [Google Scholar] [CrossRef] [Green Version]
  48. Selbach, M.; Moese, S.; Hurwitz, R.; Hauck, C.R.; Meyer, T.F.; Backert, S. The Helicobacter pylori CagA protein induces cortactin dephosphorylation and actin rearrangement by c-Src inactivation. EMBO J. 2003, 22, 515–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Wei, J.; Zhao, Z.-X.; Li, Y.; Zhou, Z.-Q.; You, T.-G. Cortactin expression confers a more malignant phenotype to gastric cancer SGC-7901 cells. World J. Gastroenterol. 2014, 20, 3287–3300. [Google Scholar] [CrossRef] [PubMed]
  50. Wei, J.; Wang, Y.; Xie, B.; Ma, J.; Wang, Y. Cortactin and HER2 as potential markers for dural-targeted therapy in advanced gastric cancer. Clin. Exp. Med. 2021. [Google Scholar] [CrossRef]
  51. Sharafutdinov, I.; Backert, S.; Tegtmeyer, N. The Helicobacter pylori type IV secretion system upregulates epithelial cortactin expression by a CagA - and JNK -dependent pathway. Cell. Microbiol. 2021, 23, e13376. [Google Scholar] [CrossRef] [PubMed]
  52. Higashi, H.; Nakaya, A.; Tsutsumi, R.; Yokoyama, K.; Fujii, Y.; Ishikawa, S.; Higuchi, M.; Takahashi, A.; Kurashima, Y.; Teishikata, Y.; et al. Helicobacter pylori CagA Induces Ras-independent Morphogenetic Response through SHP-2 Recruitment and Activation. J. Biol. Chem. 2004, 279, 17205–17216. [Google Scholar] [CrossRef] [Green Version]
  53. Muratakamiya, N.; Kurashima, Y.; Teishikata, Y.; Yamahashi, Y.; Saito, Y.; Higashi, H.; Aburatani, H.; Akiyama, T.; Peek, R.M.; Azuma, T.; et al. Helicobacter pylori CagA interacts with E-cadherin and deregulates the β-catenin signal that promotes intestinal transdifferentiation in gastric epithelial cells. Oncogene 2007, 26, 4617–4626. [Google Scholar] [CrossRef] [Green Version]
  54. Lee, D.-G.; Kim, H.S.; Lee, Y.S.; Kim, S.; Cha, S.; Ota, I.; Kim, N.H.; Cha, Y.H.; Yang, D.H.; Lee, Y.; et al. Helicobacter pylori CagA promotes Snail-mediated epithelial–mesenchymal transition by reducing GSK-3 activity. Nat. Commun. 2014, 5, 4423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Buti, L.; Spooner, E.; Van der Veen, A.G.; Rappuoli, R.; Covacci, A.; Ploegh, H.L. Helicobacter pylori cytotoxin-associated gene A (CagA) subverts the apoptosis-stimulating protein of p53 (ASPP2) tumor suppressor pathway of the host. Proc. Natl. Acad. Sci. USA 2011, 108, 9238–9243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Tabassam, F.H.; Graham, D.Y.; Yamaoka, Y. Helicobacter pyloriactivate epidermal growth factor receptor- and phosphatidylinositol 3-OH kinase-dependent Akt and glycogen synthase kinase 3β phosphorylation. Cell. Microbiol. 2009, 11, 70–82. [Google Scholar] [CrossRef] [Green Version]
  57. Franco, A.; Johnston, E.; Krishna, U.; Yamaoka, Y.; Israel, D.A.; Nagy, T.A.; Wroblewski, L.E.; Piazuelo, M.B.; Correa, P.; Peek, R.M. Regulation of Gastric Carcinogenesis by Helicobacter pylori Virulence Factors. Cancer Res. 2008, 68, 379–387. [Google Scholar] [CrossRef] [Green Version]
  58. Ford, A.C.; Yuan, Y.; Moayyedi, P. Helicobacter pylori eradication therapy to prevent gastric cancer: Systematic review and meta-analysis. Gut 2020, 69, 2113–2121. [Google Scholar] [CrossRef]
  59. Boren, T.; Falk, P.; Roth, K.; Larson, G.; Normark, S. Attachment of Helicobacter pylori to human gastric epithelium mediated by blood group antigens. Science 1993, 262, 1892–1895. [Google Scholar] [CrossRef]
  60. Ilver, D.; Arnqvist, A.; Ögren, J.; Frick, I.-M.; Kersulyte, D.; Incecik, E.T.; Berg, D.E.; Covacci, A.; Engstrand, L.; Borén, T. Helicobacter pylori Adhesin Binding Fucosylated Histo-Blood Group Antigens Revealed by Retagging. Science 1998, 279, 373–377. [Google Scholar] [CrossRef]
  61. Ishijima, N.; Suzuki, M.; Ashida, H.; Ichikawa, Y.; Kanegae, Y.; Saito, I.; Borén, T.; Haas, R.; Sasakawa, C.; Mimuro, H. BabA-mediated Adherence Is a Potentiator of the Helicobacter pylori Type IV Secretion System Activity. J. Biol. Chem. 2011, 286, 25256–25264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Bolitho, C.; Hahn, M.A.; Baxter, R.; Marsh, D.J. The chemokine CXCL1 induces proliferation in epithelial ovarian cancer cells by transactivation of the epidermal growth factor receptor. Endocr.-Relat. Cancer 2010, 17, 929–940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Lu, R.; Wu, S.; Zhang, Y.-G.; Xia, Y.; Liu, X.; Zheng, Y.; Chen, H.; Schaefer, K.L.; Zhou, Z.; Bissonnette, M.; et al. Enteric bacterial protein AvrA promotes colonic tumorigenesis and activates colonic beta-catenin signaling pathway. Oncogenesis 2014, 3, e105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Shukla, V.; Singh, H.; Pandey, M.; Upadhyay, S.; Nath, G. Carcinoma of the Gallbladder—Is It a Sequel of Typhoid? Dig. Dis. Sci. 2000, 45, 900–903. [Google Scholar] [CrossRef]
  65. LaRock, D.; Chaudhary, A.; Miller, S.I. Salmonellae interactions with host processes. Nat. Rev. Genet. 2015, 13, 191–205. [Google Scholar] [CrossRef]
  66. Kuper, H.; Adami, H.-O.; Trichopoulos, D. Infections as a major preventable cause of human cancer. J. Intern. Med. 2000, 248, 171–183. [Google Scholar] [CrossRef]
  67. Belluz, L.D.B.; Guidi, R.; Pateras, I.S.; Levi, L.; Mihaljevic, B.; Rouf, S.F.; Wrande, M.; Candela, M.; Turroni, S.; Nastasi, C.; et al. The Typhoid Toxin Promotes Host Survival and the Establishment of a Persistent Asymptomatic Infection. PLoS Pathog. 2016, 12, e1005528. [Google Scholar] [CrossRef]
  68. Kang, M.; Martin, A. Microbiome and colorectal cancer: Unraveling host-microbiota interactions in colitis-associated colorectal cancer development. Semin. Immunol. 2017, 32, 3–13. [Google Scholar] [CrossRef]
  69. Lu, R.; Bosland, M.; Xia, Y.; Zhang, Y.-G.; Kato, I.; Sun, J. Presence of Salmonella AvrA in colorectal tumor and its precursor lesions in mouse intestine and human specimens. Oncotarget 2017, 8, 55104–55115. [Google Scholar] [CrossRef] [Green Version]
  70. Wang, Z.; Vogelstein, B.; Kinzler, K.W. Phosphorylation of beta-catenin at S33, S37, or T41 can occur in the absence of phosphorylation at T45 in colon cancer cells. Cancer Res. 2003, 63, 5234–5235. [Google Scholar]
  71. Wu, S.; Ye, Z.; Liu, X.; Zhao, Y.; Xia, Y.; Steiner, A.; Petrof, E.O.; Claud, E.C.; Sun, J. Salmonella typhimurium infection increases p53 acetylation in intestinal epithelial cells. Am. J. Physiol. Liver Physiol. 2010, 298, G784–G794. [Google Scholar] [CrossRef] [Green Version]
  72. Yamaguchi, H.; Woods, N.T.; Piluso, L.G.; Lee, H.-H.; Chen, J.; Bhalla, K.N.; Monteiro, A.; Liu, X.; Hung, M.-C.; Wang, H.-G. p53 Acetylation Is Crucial for Its Transcription-independent Proapoptotic Functions. J. Biol. Chem. 2009, 284, 11171–11183. [Google Scholar] [CrossRef] [Green Version]
  73. Lu, R.; Wu, S.; Zhang, Y.-G.; Xia, Y.; Zhou, Z.; Kato, I.; Dong, H.; Bissonnette, M.; Sun, J. Salmonella Protein AvrA Activates the STAT3 Signaling Pathway in Colon Cancer. Neoplasia 2016, 18, 307–316. [Google Scholar] [CrossRef] [Green Version]
  74. Parry, C.M.; Hien, T.T.; Dougan, G.; White, N.J.; Farrar, J.J. Typhoid Fever. N. Engl. J. Med. 2002, 347, 1770–1782. [Google Scholar] [CrossRef] [Green Version]
  75. Iyer, P.; Barreto, S.G.; Sahoo, B.; Chandrani, P.; Ramadwar, M.R.; Shrikhande, S.V.; Dutt, A. Non-typhoidal Salmonella DNA traces in gallbladder cancer. Infect. Agents Cancer 2016, 11, 12. [Google Scholar] [CrossRef] [Green Version]
  76. Dongol, S.; Thompson, C.N.; Clare, S.; Nga, T.V.T.; Duy, P.T.; Karkey, A.; Arjyal, A.; Koirala, S.; Khatri, N.S.; Maskey, P.; et al. The Microbiological and Clinical Characteristics of Invasive Salmonella in Gallbladders from Cholecystectomy Patients in Kathmandu, Nepal. PLoS ONE 2012, 7, e47342. [Google Scholar] [CrossRef] [Green Version]
  77. Ye, Y.; Ning, S.; Guo, Q.; Liu, M.; Yuan, H.; Wang, Y.; Chen, Z.; Ji, R.; Li, Q.; Zhou, Y. COX-2 regulates Snail expression in gastric cancer via the Notch1 signaling pathway. Int. J. Mol. Med. 2017, 40, 512–522. [Google Scholar] [CrossRef] [Green Version]
  78. Sorski, L.; Melamed, R.; Matzner, P.; Lavon, H.; Shaashua, L.; Rosenne, E.; Ben-Eliyahu, S. Reducing liver metastases of colon cancer in the context of extensive and minor surgeries through β-adrenoceptors blockade and COX2 inhibition. Brain Behav. Immun. 2016, 58, 91–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Swidsinski, A.; Khilkin, M.; Kerjaschki‡, D.; Schreiber, S.; Ortner, M.; Weber, J.; Lochs, H. Association between intraepithelial Escherichia coli and colorectal cancer. Gastroenterology 1998, 115, 281–286. [Google Scholar] [CrossRef]
  80. Raisch, J. Colon cancer-associated B2Escherichia colicolonize gut mucosa and promote cell proliferation. World J. Gastroenterol. 2014, 20, 6560–6572. [Google Scholar] [CrossRef] [PubMed]
  81. Maddocks, O.D.K.; Scanlon, K.M.; Donnenberg, M.S. An Escherichia coli Effector Protein Promotes Host Mutation via Depletion of DNA Mismatch Repair Proteins. mBio 2013, 4, e00152-13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Cougnoux, A.; Dalmasso, G.; Martinez, R.; Buc, E.; Delmas, J.; Gibold, L.; Sauvanet, P.; Darcha, C.; Déchelotte, P.; Bonnet, M.; et al. Bacterial genotoxin colibactin promotes colon tumour growth by inducing a senescence-associated secretory phenotype. Gut 2014, 63, 1932–1942. [Google Scholar] [CrossRef]
  83. Abd-El-Raouf, R.; Ouf, S.A.; Gabr, M.M.; Zakaria, M.M.; El-Yasergy, K.F.; Ali-El-Dein, B. Escherichia coli foster bladder cancer cell line progression via epithelial mesenchymal transition, stemness and metabolic reprogramming. Sci. Rep. 2020, 10, 18024. [Google Scholar] [CrossRef]
  84. Pleguezuelos-Manzano, C.; Puschhof, J.; Huber, A.R.; Van Hoeck, A.; Wood, H.M.; Nomburg, J.; Gurjao, C.; Manders, F.; Dalmasso, G.; Stege, P.B.; et al. Mutational signature in colorectal cancer caused by genotoxic pks+ E. coli. Nature 2020, 580, 269–273. [Google Scholar] [CrossRef]
  85. Laurila, A.L.; Anttila, T.; Läärä, E.; Bloigu, A.; Virtamo, J.; Albanes, D.; Leinonen, M.; Saikku, P. Serological evidence of an association betweenChlamydia pneumoniae infection and lung cancer. Int. J. Cancer 1997, 74, 31–34. [Google Scholar] [CrossRef]
  86. Littman, A.J.; Jackson, L.A.; Vaughan, T.L. Chlamydia pneumoniae and Lung Cancer: Epidemiologic Evidence. Cancer Epidemiol. Biomark. Prev. 2005, 14, 773–778. [Google Scholar] [CrossRef] [Green Version]
  87. Chaturvedi, A.K.; Gaydos, C.A.; Agreda, P.; Holden, J.P.; Chatterjee, N.; Goedert, J.J.; Caporaso, N.E.; Engels, E.A. Chlamydia pneumoniae Infection and Risk for Lung Cancer. Cancer Epidemiol. Biomark. Prev. 2010, 19, 1498–1505. [Google Scholar] [CrossRef] [Green Version]
  88. Alshamsan, A.; Khan, S.; Imran, A.; Aljuffali, I.A.; Alsaleh, K. Prediction of Chlamydia pneumoniae protein localization in host mitochondria and cytoplasm and possible involvements in lung cancer etiology: A computational approach. Saudi Pharm. J. 2017, 25, 1151–1157. [Google Scholar] [CrossRef]
  89. Rizzo, A.; Carratelli, C.R. Transforming activities of Chlamydia pneumoniae in human mesothelial cells. Int. Microbiol. 2014, 17, 185–193. [Google Scholar] [CrossRef] [PubMed]
  90. Zhu, H.; Shen, Z.; Luo, H.; Zhang, W.; Zhu, X. Chlamydia Trachomatis Infection-Associated Risk of Cervical Cancer. Medicine 2016, 95, e3077. [Google Scholar] [CrossRef] [PubMed]
  91. Koskela, P.; Anttila, T.; Bjørge, T.; Brunsvig, A.; Dillner, J.; Hakama, M.; Hakulinen, T.; Jellum, E.; Lehtinen, M.; Lenner, P.; et al. Chlamydia trachomatis infection as a risk factor for invasive cervical cancer. Int. J. Cancer 2000, 85, 35–39. [Google Scholar] [CrossRef]
  92. Smith, J.S.; Bosetti, C.; Muñoz, N.; Herrero, R.; Bosch, F.X.; Eluf-Neto, J.; Meijer, C.J.; Brule, A.J.V.D.; Franceschi, S.; Peeling, R.W. Chlamydia trachomatisand invasive cervical cancer: A pooled analysis of the IARC multicentric case-control study. Int. J. Cancer 2004, 111, 431–439. [Google Scholar] [CrossRef] [PubMed]
  93. Paavonen, J.; Karunakaran, K.P.; Noguchi, Y.; Anttila, T.; Bloigu, A.; Dillner, J.; Hallmans, G.; Hakulinen, T.; Jellum, E.; Koskela, P.; et al. Serum antibody response to the heat shock protein 60 of Chlamydia trachomatis in women with developing cervical cancer. Am. J. Obstet. Gynecol. 2003, 189, 1287–1292. [Google Scholar] [CrossRef]
  94. Trabert, B.; Waterboer, T.; Idahl, A.; Brenner, N.; Brinton, L.A.; Butt, J.; Coburn, S.B.; Hartge, P.; Hufnagel, K.; Inturrisi, F.; et al. Antibodies Against Chlamydia trachomatis and Ovarian Cancer Risk in Two Independent Populations. J. Natl. Cancer Inst. 2018, 111, 129–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Abdul-Sater, A.A.; Saïd-Sadier, N.; Lam, V.M.; Singh, B.; Pettengill, M.A.; Soares, F.; Tattoli, I.; Lipinski, S.; Girardin, S.E.; Rosenstiel, P.; et al. Enhancement of Reactive Oxygen Species Production and Chlamydial Infection by the Mitochondrial Nod-like Family Member NLRX. J. Biol. Chem. 2010, 285, 41637–41645. [Google Scholar] [CrossRef] [Green Version]
  96. Mi, Y.; Gurumurthy, R.K.; Zadora, P.K.; Meyer, T.F.; Chumduri, C. Chlamydia trachomatis Inhibits Homologous Recombination Repair of DNA Breaks by Interfering with PP2A Signaling. mBio 2018, 9, e01465-18. [Google Scholar] [CrossRef] [Green Version]
  97. Weber, M.; Lam, J.L.; Dooley, C.A.; Noriea, N.F.; Hansen, B.T.; Hoyt, F.H.; Carmody, A.B.; Sturdevant, G.L.; Hackstadt, T. Absence of Specific Chlamydia trachomatis Inclusion Membrane Proteins Triggers Premature Inclusion Membrane Lysis and Host Cell Death. Cell Rep. 2017, 19, 1406–1417. [Google Scholar] [CrossRef] [Green Version]
  98. Ying, S.; Fischer, S.F.; Pettengill, M.; Conte, D.; Paschen, S.A.; Ojcius, D.; Häcker, G. Characterization of Host Cell Death Induced by Chlamydia trachomatis. Infect. Immun. 2006, 74, 6057–6066. [Google Scholar] [CrossRef] [Green Version]
  99. Rajalingam, K.; Sharma, M.; Lohmann, C.; Oswald, M.; Thieck, O.; Froelich, C.J.; Rudel, T. Mcl-1 Is a Key Regulator of Apoptosis Resistance in Chlamydia trachomatis-Infected Cells. PLoS ONE 2008, 3, e3102. [Google Scholar] [CrossRef]
  100. Fischer, A.; Harrison, K.S.; Ramirez, Y.; Auer, D.; Chowdhury, S.R.; Prusty, B.K.; Sauer, F.; Dimond, Z.; Kisker, C.; Hefty, P.S.; et al. Chlamydia trachomatis-containing vacuole serves as deubiquitination platform to stabilize Mcl-1 and to interfere with host defense. eLife 2017, 6, 6192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Brennan, C.A.; Garrett, W.S. Fusobacterium nucleatum—Symbiont, opportunist and oncobacterium. Nat. Rev. Microbiol. 2019, 17, 156–166. [Google Scholar] [CrossRef]
  102. Kostic, A.D.; Gevers, D.; Pedamallu, C.S.; Michaud, M.; Duke, F.; Earl, A.M.; Ojesina, A.I.; Jung, J.; Bass, A.J.; Tabernero, J.; et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012, 22, 292–298. [Google Scholar] [CrossRef] [Green Version]
  103. Shang, F.-M.; Liu, H.-L. Fusobacterium nucleatum and colorectal cancer: A review. World J. Gastrointest. Oncol. 2018, 10, 71–81. [Google Scholar] [CrossRef]
  104. Rubinstein, M.R.; Wang, X.; Liu, W.; Hao, Y.; Cai, G.; Han, Y.W. Fusobacterium nucleatum Promotes Colorectal Carcinogenesis by Modulating E-Cadherin/β-Catenin Signaling via its FadA Adhesin. Cell Host Microbe 2013, 14, 195–206. [Google Scholar] [CrossRef] [Green Version]
  105. Gur, C.; Ibrahim, Y.; Isaacson, B.; Yamin, R.; Abed, J.; Gamliel, M.; Enk, J.; Bar-On, Y.; Stanietsky-Kaynan, N.; Coppenhagen-Glazer, S.; et al. Binding of the Fap2 Protein of Fusobacterium nucleatum to Human Inhibitory Receptor TIGIT Protects Tumors from Immune Cell Attack. Immunity 2015, 42, 344–355. [Google Scholar] [CrossRef] [Green Version]
  106. Guo, S.; Chen, J.; Chen, F.; Zeng, Q.; Liu, W.-L.; Zhang, G. Exosomes derived from Fusobacterium nucleatum-infected colorectal cancer cells facilitate tumour metastasis by selectively carrying miR-1246/92b-3p/27a-3p and CXCL. Gut 2021, 70, 1507–1519. [Google Scholar] [CrossRef]
  107. Kong, C.; Yan, X.; Zhu, Y.; Zhu, H.; Luo, Y.; Liu, P.; Ferrandon, S.; Kalady, M.F.; Gao, R.; He, J.; et al. Fusobacterium Nucleatum Promotes the Development of Colorectal Cancer by Activating a Cytochrome P450/Epoxyoctadecenoic Acid Axis via TLR4/Keap1/NRF2 Signaling. Cancer Res. 2021, 81, 4485–4498. [Google Scholar] [CrossRef] [PubMed]
  108. Klein, R.S.; Recco, R.A.; Catalano, M.T.; Edberg, S.C.; Casey, J.I.; Steigbigel, N. Association ofStreptococcus boviswith Carcinoma of the Colon. N. Engl. J. Med. 1977, 297, 800–802. [Google Scholar] [CrossRef] [PubMed]
  109. Gupta, A.; Madani, R.; Mukhtar, H. Streptococcus bovisendocarditis, a silent sign for colonic tumour. Color. Dis. 2010, 12, 164–171. [Google Scholar] [CrossRef] [PubMed]
  110. Aymeric, L.; Donnadieu, F.; Mulet, C.; du Merle, L.; Nigro, G.; Saffarian, A.; Bérard, M.; Poyart, C.; Robine, S.; Regnault, B.; et al. Colorectal cancer specific conditions promote Streptococcus gallolyticus gut colonization. Proc. Natl. Acad. Sci. USA 2018, 115, E283–E291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Kumar, R.; Herold, J.L.; Schady, D.; Davis, J.; Kopetz, S.; Martinez-Moczygemba, M.; Murray, B.E.; Han, F.; Li, Y.; Callaway, E.; et al. Streptococcus gallolyticus subsp. gallolyticus promotes colorectal tumor development. PLoS Pathog. 2017, 13, e1006440. [Google Scholar] [CrossRef]
  112. Huang, S.; Li, J.-Y.; Wu, J.; Meng, L.; Shou, C.-C. Mycoplasma infections and different human carcinomas. World J. Gastroenterol. 2001, 7, 266–269. [Google Scholar] [CrossRef]
  113. Barykova, Y.A.; Logunov, D.Y.; Shmarov, M.; Vinarov, A.; Fiev, D.N.; Vinarova, N.A.; Rakovskaya, I.V.; Baker, P.S.; Shyshynova, I.; Stephenson, A.J.; et al. Association of Mycoplasma hominis infection with prostate cancer. Oncotarget 2011, 2, 289–297. [Google Scholar] [CrossRef] [Green Version]
  114. Miyake, M.; Ohnishi, K.; Hori, S.; Nakano, A.; Nakano, R.; Yano, H.; Ohnishi, S.; Owari, T.; Morizawa, Y.; Itami, Y.; et al. Mycoplasma genitalium Infection and Chronic Inflammation in Human Prostate Cancer: Detection Using Prostatectomy and Needle Biopsy Specimens. Cells 2019, 8, 212. [Google Scholar] [CrossRef] [Green Version]
  115. Liu, W.; Ren, T.; Jiang, B.; Gong, M.; Shou, C. Mycoplasmal membrane protein p37 promotes malignant changes in mammalian cells. Can. J. Microbiol. 2007, 53, 270–276. [Google Scholar] [CrossRef]
  116. Gong, M.; Meng, L.; Jiang, B.; Zhang, J.; Yang, H.; Wu, J.; Shou, C. p37 fromMycoplasma hyorhinispromotes cancer cell invasiveness and metastasis through activation of MMP-2 and followed by phosphorylation of EGFR. Mol. Cancer Ther. 2008, 7, 530–537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Gagnière, J.; Raisch, J.; Veziant, J.; Barnich, N.; Bonnet, R.; Buc, E.; Bringer, M.-A.; Pezet, D.; Bonnet, M. Gut microbiota imbalance and colorectal cancer. World J. Gastroenterol. 2016, 22, 501–518. [Google Scholar] [CrossRef] [PubMed]
  118. Boleij, A.; Hechenbleikner, E.M.; Goodwin, A.C.; Badani, R.; Stein, E.M.; Lazarev, M.G.; Ellis, B.; Carroll, K.C.; Albesiano, E.; Wick, E.C.; et al. The Bacteroides fragilis Toxin Gene Is Prevalent in the Colon Mucosa of Colorectal Cancer Patients. Clin. Infect. Dis. 2015, 60, 208–215. [Google Scholar] [CrossRef]
  119. Chung, L.; Orberg, E.T.; Geis, A.L.; Chan, J.L.; Fu, K.; Shields, C.E.D.; Dejea, C.M.; Fathi, P.; Chen, J.; Finard, B.B.; et al. Bacteroides fragilis Toxin Coordinates a Pro-carcinogenic Inflammatory Cascade via Targeting of Colonic Epithelial Cells. Cell Host Microbe 2018, 23, 421. [Google Scholar] [CrossRef] [PubMed]
  120. Sutcliffe, S.; Giovannucci, E.; De Marzo, A.M.; Leitzmann, M.F.; Willett, W.C.; Platz, E.A. Gonorrhea, Syphilis, Clinical Prostatitis, and the Risk of Prostate Cancer. Cancer Epidemiol. Biomark. Prev. 2006, 15, 2160–2166. [Google Scholar] [CrossRef] [Green Version]
  121. Balamurugan, R.; Rajendiran, E.; George, S.; Samuel, G.V.; Ramakrishna, B.S. Real-time polymerase chain reaction quantification of specific butyrate-producing bacteria, DesulfovibrioandEnterococcus faecalisin the feces of patients with colorectal cancer. J. Gastroenterol. Hepatol. 2008, 23, 1298–1303. [Google Scholar] [CrossRef]
  122. Wang, X.; Yang, Y.; Huycke, M.M. Commensal-infected macrophages induce dedifferentiation and reprogramming of epithelial cells during colorectal carcinogenesis. Oncotarget 2017, 8, 102176–102190. [Google Scholar] [CrossRef] [PubMed]
  123. Miyamoto, S.; Komiya, M.; Fujii, G.; Hamoya, T.; Nakanishi, R.; Fujimoto, K.; Tamura, S.; Kurokawa, Y.; Takahashi, M.; Ijichi, T.; et al. Preventive Effects of Heat-Killed Enterococcus faecalis Strain EC-12 on Mouse Intestinal Tumor Development. Int. J. Mol. Sci. 2017, 18, 826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. de Almeida, C.V.; Taddei, A.; Amedei, A. The controversial role of Enterococcus faecalis in colorectal cancer. Ther. Adv. Gastroenterol. 2018, 11, 1756284818783606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Strickertsson, J.A.B.; Desler, C.; Martin-Bertelsen, T.; Machado, A.M.D.; Wadstrøm, T.; Winther, O.; Rasmussen, L.J.; Friis-Hansen, L. Enterococcus faecalis Infection Causes Inflammation, Intracellular Oxphos-Independent ROS Production, and DNA Damage in Human Gastric Cancer Cells. PLoS ONE 2013, 8, e63147. [Google Scholar] [CrossRef] [Green Version]
  126. Hermsen, J.L.; Schurr, M.J.; Kudsk, K.A.; Faucher, L.D. Phenotyping Clostridium septicum Infection: A Surgeon’s Infectious Disease. J. Surg. Res. 2008, 148, 67–76. [Google Scholar] [CrossRef]
  127. Chew, S.S.B.; Lubowski, D.Z. Clostridium septicumand malignancy. ANZ J. Surg. 2001, 71, 647–649. [Google Scholar] [CrossRef]
  128. Dylewski, J.; Luterman, L. Septic arthritis and Clostridium septicum: A clue to colon cancer. Can. Med. Assoc. J. 2010, 182, 1446–1447. [Google Scholar] [CrossRef]
  129. Engels, E.A.; Shen, M.; Chapman, R.S.; Pfeiffer, R.M.; Yu, Y.-Y.; He, X.; Lan, Q. Tuberculosis and subsequent risk of lung cancer in Xuanwei, China. Int. J. Cancer 2009, 124, 1183–1187. [Google Scholar] [CrossRef]
  130. Tian, Y.; Hao, T.; Cao, B.; Zhang, W.; Ma, Y.; Lin, Q.; Li, X. Clinical End-Points Associated withMycobacterium tuberculosisand Lung Cancer: Implications into Host-Pathogen Interaction and Coevolution. BioMed Res. Int. 2015, 2015, 827829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Graphical overview of bacteria associated with carcinoma.
Figure 1. Graphical overview of bacteria associated with carcinoma.
Pathogens 10 01321 g001
Figure 2. Oncogenic pathways activated by H. pylori: VacA can cause apoptosis, resulting in increased cell proliferation or serve as a possible entry point for other possible carcinogens. CagA enters the cell through T4SS and is phosphorylated by Src, causing it to activate SHP-2, resulting in cellular morphological changes. Additionally, CagA can stabilize β-catenin, resulting in increased cell proliferation. Outer membrane protein BabA binds to human Lewis (b) surface epitopes [59,60], thus increasing specific proinflammatory cytokines like CXCL1 [61], that might boost cell proliferation [62]. Outer membrane protein OipA binds to EGFR, leading to activated PI3K-AKT signalling and increased β-catenin.
Figure 2. Oncogenic pathways activated by H. pylori: VacA can cause apoptosis, resulting in increased cell proliferation or serve as a possible entry point for other possible carcinogens. CagA enters the cell through T4SS and is phosphorylated by Src, causing it to activate SHP-2, resulting in cellular morphological changes. Additionally, CagA can stabilize β-catenin, resulting in increased cell proliferation. Outer membrane protein BabA binds to human Lewis (b) surface epitopes [59,60], thus increasing specific proinflammatory cytokines like CXCL1 [61], that might boost cell proliferation [62]. Outer membrane protein OipA binds to EGFR, leading to activated PI3K-AKT signalling and increased β-catenin.
Pathogens 10 01321 g002
Figure 3. Oncogenic pathways activated by Salmonella enterica: Salmonella protein AvrA activates Wnt-signalling, stabilizing β-catenin, leading to an increased expression of c-Myc and decreased expression of p53 resulting in increased cell proliferation. AvrA also activates STAT3-signaling, further increasing cell proliferation and inflammatory regulation. Salmonella proteins SopB, SopE and SopE2, injected through type III secretion system activate MAPK/AKT pathway provoking cell transformation and carcinoma.
Figure 3. Oncogenic pathways activated by Salmonella enterica: Salmonella protein AvrA activates Wnt-signalling, stabilizing β-catenin, leading to an increased expression of c-Myc and decreased expression of p53 resulting in increased cell proliferation. AvrA also activates STAT3-signaling, further increasing cell proliferation and inflammatory regulation. Salmonella proteins SopB, SopE and SopE2, injected through type III secretion system activate MAPK/AKT pathway provoking cell transformation and carcinoma.
Pathogens 10 01321 g003
Figure 4. E. coli, through colibactin, caused cell cycle arrest and double-stranded DNA breaks resulting in genomic instability. Additionally, colibactin was found to increase cell proliferation and induce tumour-promoting inflammation. Colibactin also resulted in increased p53 SUMOylation resulting in emergence of senescent cells. Finally, E. coli downregulated mismatch repair proteins MSH2 and MLH1 leading to further genomic instability.
Figure 4. E. coli, through colibactin, caused cell cycle arrest and double-stranded DNA breaks resulting in genomic instability. Additionally, colibactin was found to increase cell proliferation and induce tumour-promoting inflammation. Colibactin also resulted in increased p53 SUMOylation resulting in emergence of senescent cells. Finally, E. coli downregulated mismatch repair proteins MSH2 and MLH1 leading to further genomic instability.
Pathogens 10 01321 g004
Figure 5. C. trachomatis downregulates the tumour suppressor P53 leading to increased cell proliferation, while also increasing levels of reactive oxygen species, causing oxidative DNA damage, which together with inhibition of cell DNA repair mechanisms causes genomic instability. C. pneumoniae, through unknown mechanisms, leads to inflammation and dysregulation of replication, transcription, and DNA repair mechanisms.
Figure 5. C. trachomatis downregulates the tumour suppressor P53 leading to increased cell proliferation, while also increasing levels of reactive oxygen species, causing oxidative DNA damage, which together with inhibition of cell DNA repair mechanisms causes genomic instability. C. pneumoniae, through unknown mechanisms, leads to inflammation and dysregulation of replication, transcription, and DNA repair mechanisms.
Pathogens 10 01321 g005
Figure 6. Pathways activated by F. nucleatum. FadA was found to inhibit E-cadherin, activating β-catenin and increasing cell proliferation. Additionally, increased levels of NF-κB and proinflammatory interleukins IL-6, IL-8, and IL-18 resulted in a possible tumour promoting inflammation. Fap2 showed interactions with the TIGIT immune receptor, causing inhibition of immune cells, and thereby creating an immunosuppressive microenvironment.
Figure 6. Pathways activated by F. nucleatum. FadA was found to inhibit E-cadherin, activating β-catenin and increasing cell proliferation. Additionally, increased levels of NF-κB and proinflammatory interleukins IL-6, IL-8, and IL-18 resulted in a possible tumour promoting inflammation. Fap2 showed interactions with the TIGIT immune receptor, causing inhibition of immune cells, and thereby creating an immunosuppressive microenvironment.
Pathogens 10 01321 g006
Figure 7. S. bovis, through an unknown pathway, increased levels of β-catenin and c-Myc, resulting in increased cell proliferation.
Figure 7. S. bovis, through an unknown pathway, increased levels of β-catenin and c-Myc, resulting in increased cell proliferation.
Pathogens 10 01321 g007
Figure 8. M. hyorhinis increases β-catenin through GSK3β, Wnt-signalling, and activation of LRP6. This resulted in increased cell proliferation. Without LRP6 activation, GSK3β had an inhibitory effect on β-catenin. LRP6 was additionally able to interact with p37, leading to increased levels of cell immortality, cell motility, migration, and invasion.
Figure 8. M. hyorhinis increases β-catenin through GSK3β, Wnt-signalling, and activation of LRP6. This resulted in increased cell proliferation. Without LRP6 activation, GSK3β had an inhibitory effect on β-catenin. LRP6 was additionally able to interact with p37, leading to increased levels of cell immortality, cell motility, migration, and invasion.
Pathogens 10 01321 g008
Figure 9. B. fragilis (ETBF) cleaves E-cadherin, leading to activated β-catenin levels, resulting in increased cell proliferation. ETBF also creates tumour-promoting inflammation through upregulation of the STAT3 pathway, IL-17, and NF-κB.
Figure 9. B. fragilis (ETBF) cleaves E-cadherin, leading to activated β-catenin levels, resulting in increased cell proliferation. ETBF also creates tumour-promoting inflammation through upregulation of the STAT3 pathway, IL-17, and NF-κB.
Pathogens 10 01321 g009
Figure 10. N. gonorrhoeae, through unclear mechanisms, caused DNA damage and increased the expression of p21 and p27 while decreasing the levels of p53 in non-tumour epithelial cells, resulting in genomic instability and cell cycle dysregulation.
Figure 10. N. gonorrhoeae, through unclear mechanisms, caused DNA damage and increased the expression of p21 and p27 while decreasing the levels of p53 in non-tumour epithelial cells, resulting in genomic instability and cell cycle dysregulation.
Pathogens 10 01321 g010
Figure 11. E. faecalis increased intracellular levels of ROS, leading to DNA instability and reduced DNA repair response resulting in genomic instability. E. Faecalis increased NF-κB levels, thereby causing a tumour promoting inflammation. When cocultured with M1 macrophages, β-catenin levels increased, leading to increased cell proliferation. Coculturing with heat-killed E. faecalis EC-12 strain, β-catenin was reduced, showing a possible protective role in CRC.
Figure 11. E. faecalis increased intracellular levels of ROS, leading to DNA instability and reduced DNA repair response resulting in genomic instability. E. Faecalis increased NF-κB levels, thereby causing a tumour promoting inflammation. When cocultured with M1 macrophages, β-catenin levels increased, leading to increased cell proliferation. Coculturing with heat-killed E. faecalis EC-12 strain, β-catenin was reduced, showing a possible protective role in CRC.
Pathogens 10 01321 g011
Figure 12. Mycobacterium tuberculosis (MBT) increased levels of PD-1/PD-L1 resulting in increased tumour metastasis while also inhibiting T cell immune response causing an immunosuppressive microenvironment favourable for potential tumour progression.
Figure 12. Mycobacterium tuberculosis (MBT) increased levels of PD-1/PD-L1 resulting in increased tumour metastasis while also inhibiting T cell immune response causing an immunosuppressive microenvironment favourable for potential tumour progression.
Pathogens 10 01321 g012
Table 1. Overview of bacteria associated with carcinoma.
Table 1. Overview of bacteria associated with carcinoma.
BacteriaPotential Hallmarks of Cancer and Other Associated PathologiesType of Carcinoma Associated
Helicobacter pyloriIncrease proliferation signalling [1,2,3]Gastric cancer [1,2,3]
Escherichia coli(pks+ve strain)Genomic instability (DNA damage), tumour-promoting inflammation, and increased cell proliferation [4,5,6]Colorectal cancer [4,5,6]
Streptococcus bovisIncrease proliferation signallingColorectal cancer [7,8]
Salmonella enterica (serovar Enteritidis and Typhi) Increased cell proliferation, transformation, and tumour-promoting inflammation [9,10,11]Colorectal cancer and gallbladder carcinoma [9,10,11]
Fusobacterium nucleatumIncreased cell proliferation, avoid immune destruction, and tumour-promoting inflammation [12,13]Colorectal cancer (potentially oral, head, neck, oesophageal, cervical, and gastric cancer tissues) [12,13]
Enterotoxigenic Bacteroides fragilis (ETBF)Increased cell proliferation and tumour-promoting inflammation [14,15,16]Colorectal cancer [14,15,16]
Enterococcus faecalisGenomic instability, tumour-promoting inflammation, and increased cell proliferation [17]Colorectal cancer [17]
Clostridium septicumMyonecrosis [18,19]Potential involvement in colorectal cancer [19]
Neisseria gonorrhoeaeDNA damage [20], Genomic instabilityProstate cancer [21,22] and bladder cancer [23]
Mycobacterium tuberculosisTumour metastasis and avoiding immune destruction [24,25]Lung cancer [24,25]
Chlamydia trachomatis and Chlamydia pneumoniaeIncreased cell proliferation, tumour-promoting [26,27,28] inflammation [29], genomic instability [30], and evasion of apoptosis [31], evade immune system [32]Cervical cancer [26,27,28] and lung cancer
Mycoplasma hyorhinisCell immortality, invasion, and increased cell proliferation [33,34]Prostate cancer, gastric carcinoma, oesophageal cancer, lung cancer, and breast cancer [33,34]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hansen, J.P.; Ali, W.M.; Sivadasan, R.; Rajeeve, K. Bacteria–Cancer Interface: Awaiting the Perfect Storm. Pathogens 2021, 10, 1321. https://doi.org/10.3390/pathogens10101321

AMA Style

Hansen JP, Ali WM, Sivadasan R, Rajeeve K. Bacteria–Cancer Interface: Awaiting the Perfect Storm. Pathogens. 2021; 10(10):1321. https://doi.org/10.3390/pathogens10101321

Chicago/Turabian Style

Hansen, Jonathan Pommer, Waled Mohammed Ali, Rajeeve Sivadasan, and Karthika Rajeeve. 2021. "Bacteria–Cancer Interface: Awaiting the Perfect Storm" Pathogens 10, no. 10: 1321. https://doi.org/10.3390/pathogens10101321

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop