Sunteți pe pagina 1din 14

Cellular Signalling 25 (2013) 403416

Contents lists available at SciVerse ScienceDirect

Cellular Signalling
journal homepage: www.elsevier.com/locate/cellsig

Review

Signaling pathways bridging microbial-triggered inammation and cancer


Maulilio John Kipanyula a, Paul Faustin Seke Etet b, Lorella Vecchio c, Mohammed Farahna b,
Elias Nchiwan Nukenine d, Armel Herv Nwabo Kamdje d, e,
a
Department of Veterinary Anatomy, Sokoine University of Agriculture, P.O. Box 3016, Chuo Kikuu, Morogoro, Tanzania
b
Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, 51452 Al-Qaseem, Saudi Arabia
c
Laboratory of Cytometry, Institute of Molecular Genetics, CNR, University of Pavia, 27100 Pavia, Italy
d
Department of Biomedical Sciences, University of Ngaoundr, P.O. Box 454, Ngaoundr, Cameroon
e
Biomedical Research Center, University of British Columbia, 2222 Health Science Mall, Vancouver BC, Canada, V6T 1Z3

a r t i c l e i n f o a b s t r a c t

Article history: Microbial-triggered inammation protects against pathogens and yet can paradoxically cause considerable sec-
Received 7 October 2012 ondary damage to host tissues that can result in tissue brosis and carcinogenesis, if persistent. In addition to
Accepted 26 October 2012 classical pathogens, gut microbiota bacteria, i.e. a group of mutualistic microorganisms permanently inhabiting
Available online 30 October 2012
the gastrointestinal tract and which plays a key role in digestion, immunity, and cancer prevention, can induce
inammation-associated cancer following the alterations of their microenvironment. Emerging experimental
Keywords:
Signaling pathways
evidence indicates that microbiota members like Escherichia coli and several other genotoxic and mutagenic
Microbe pathogens can cause DNA damage in various cell types. In addition, the inammatory response induced by
Infection chronic infections with pathogens like the microbiota members Helicobacter spp., which have been associated
Inammation with liver, colorectal, cervical cancers and lymphoma, for instance, can also trigger carcinogenic processes. A mi-
Carcinogenesis croenvironment including active immune cells releasing high amounts of inammatory signaling molecules can
Cancer favor the carcinogenic transformation of host cells. Pivotal molecules released during immune response such as
the macrophage migration inhibitory factor (MMIF) and the reactive oxygen and nitrogen species' products
superoxide and peroxynitrite, can further damage DNA and cause the accumulation of oncogenic mutations,
whereas pro-inammatory cytokines, adhesion molecules, and growth factors may create a microenvironment
promoting neoplastic cell survival and proliferation. Recent ndings on the implication of inammatory
signaling pathways in microbial-triggered carcinogenesis as well as the possible role of microbiota modulation
in cancer prevention are herein summarized and discussed.
2012 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 404
2. Pathogens and cancer: protective effects of inammation and mutagenic potential . . . . . . . . . . . . . . . . . . . . . . . . . . . . 404
2.1. Microbiota and susceptibility to infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 404
2.2. Outcome of immune defense against pathogens: mutagenic responses and cancer . . . . . . . . . . . . . . . . . . . . . . . . . 406
3. Pattern-recognition receptors: example of Toll-like receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 406
3.1. TLRs, pathogen-triggered inammation, and cancer: lessons from microbiota . . . . . . . . . . . . . . . . . . . . . . . . . . . . 406
3.2. TLR signaling and inammation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 407
4. NF-B, pathogen-triggered inammation, and cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 408
4.1. NF-B, infection, and cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 408
4.2. NF-B signaling and carcinogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 408
5. Pro-inammatory cytokines and COX-2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 409
5.1. IL-1 and TNF- . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 409
5.2. IL-32 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 410
5.3. IL-8 and IL-29 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 410
5.4. COX-2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 410

Corresponding author at: Department of Biomedical Sciences, University of Ngaoundr, P.O. Box 454, Ngaoundr, Cameroon/Biomedical Research Center, University of British
Columbia, 2222 Health Science Mall, Vancouver BC, Canada, V6T 1Z3. Tel.: +1 604 822 0441(ofce); fax: +1 604 822 7815.
E-mail address: armel.nwabo@gmail.com (A.H. Nwabo Kamdje).

0898-6568/$ see front matter 2012 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.cellsig.2012.10.014
404 M.J. Kipanyula et al. / Cellular Signalling 25 (2013) 403416

6. Oxygen/nitrogen species and microbial-triggered cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 410


6.1. Oxidative and nitrative stress . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 410
6.2. Hypoxia-inducible factor 1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 411
7. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 412
Disclosure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 412
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 412
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 412

1. Introduction In this review, we summarize and discuss the recent ndings on


signaling pathways bridging pathogen-triggered inammation and
In the presence of pathogens or irritants and associated molecules, cancer initiation and progression. The possible role of gut microbiota
the body mounts a strong immune response termed as inamma- modulation for cancer prevention is also considered.
tion aimed at preventing tissue injury and combating infection.
Mononuclear phagocytes residing in tissues are the rst to be activat- 2. Pathogens and cancer: protective effects of inammation and
ed during innate immune response. They recognize pathogens and mutagenic potential
molecules associated with tissue damage through intracellular or
surface-expressed pattern recognition receptors (PRRs) that have in- 2.1. Microbiota and susceptibility to infection
herent capacity to directly or indirectly detect pathogen-associated
molecular patterns (PAMPs) such as microbial nucleic acids, bacterial Although the body is continuously exposed to microorganisms,
cell wall lipopolysaccharide (LPS) and proteoglycans, and fungal cell only a small fraction is pathogenic or can escape/resist immune re-
wall components like -mannan and -glucan; and danger/damage sponse whereas many live in a mutualistic relationship with the
associated molecular patterns (DAMPs), including molecules released body, such as the gut microbiota that is a group of microorganisms
by injured cells such as: nucleic acids, uric acid, ATP, amyloid and permanently inhabiting the gastrointestinal tract [5]. As already men-
cytoplasmic proteins of the S100 family [1,2], for instance. tioned, the commensal microorganisms that inhabit the mammalian
PRR signaling is an important partner of the immune system body surfaces and the gut play a critical role in many key functions
whose pivotal role is to promote the release of factors triggering the of the organism, including immune response, energy metabolism,
activation and recruitment of circulating immune cells like mono- and cancer prevention [15,17]. However, in circumstances that dis-
cytes and neutrophils, and even adaptive immune cells like lympho- turb their microenvironment they can breach the skin barrier or the
cytes, to the infection/damage site in case of persistent challenge intestinal mucosa layer and enter bloodstream, resulting in chronic
[25]: The rst transient changes in vascular wall are caused by se- inammation, and subsequently causing tissue brosis and carcino-
creted factors like prostaglandins, and nitric oxide that act on vascular genesis [24]. Certain commensal bacteria can invade colonic epithelial
smooth muscle to cause vasodilation thus increase blood ow and cells, activating early intracellular signaling systems to trigger host in-
transport more circulating leukocytes. Secondly, vascular permeabili- ammatory reactions. Such bacteria include Fusobacterium varium,
ty is further increased by inammatory mediators such as histamine Bacteroides vulgatus, Escherichia coli and Clostridium clostridioforme,
and leukotrienes that act on endothelial cells to allow plasma proteins for instance, which are commonly isolated from the mucosa of in-
and leukocytes to exit the circulation. Finally pro-inammatory cyto- ammatory bowel disease patients [25]. An in vitro study conducted
kines such as interleukin 1 (IL-1) and tumor necrosis factor alpha by Ohkusa and collaborators [25] have reported that these commen-
(TNF-) promote immune cell inltration to the affected tissue, sal bacteria, but not probiotics, adhere to colonic epithelial cells and
whereas the chemoattractant factors direct them to the inamed invade their cytoplasm, resulting in the expression of mRNAs of
site. The immune cells recruited ght against invading bacteria, virus- IL-8, IL-6, TNF-, and CCL2 (CC motif ligand 2, chemokine), and in
es, or parasites at the infection site, while swelling (i.e. the buildup of the release of signicant amounts of IL-8 and TNF- by invaded
uids in the interstitium) and immune cell phagocytic activity host cells. Furthermore, these authors observed an increased expres-
aimed at eliminating DAMPS protect injured areas from further sion of IL-8, TNF-, and phosphorylated NF-B (nuclear factor B)
harm [6,7]. p65 in inamed epithelia with cryptitis or crypt abscess in ulcerative
Inammatory reactions are ceased following the elimination of colitis patients, in further clinical studies. In addition, H. pylori that
pathogens, but can persist in case of chronic infection or following has been reported protective effects against pathogenic bacteria and
chronic exposure to DAMPs [4,8,9], resulting in tissue brosis and car- esophageal cancer [15], can also play crucial roles in the progression
cinogenesis. The link between inammation and the risk of develop- from inammation to cancer in the stomach [26]. Recent studies in
ing cancer was reported as early as 1863, when Virchow observed a mouse colitis-associated adenocarcinoma model have indicated
that carcinogenesis tends to occur at chronic inammation sites that H. hepaticus promotes tumorigenesis [16]. Similarly, E. coli that
[10]. Subsequently, epidemiologic data have strongly indicated that induces homeostatic mucosal immune responses can also cause
chronic inammation is associated with increased risk of cancer inappropriate immune activation in genetically susceptible hosts,
[11,12]. It is estimated that about two million cancer cases are caused resulting in colitis-associated colorectal cancer [17]. Murine isolate
by infectious agents each year [13]. Agents like human papillomavi- of E. coli chronic intestinal inammation causes functional alterations
ruses, hepatitis B and C viruses, and the Gram-positive bacterium in gene expression in commensal gut bacteria [27]. Further investiga-
Helicobacter pylori, which is a gut microbiota member, are responsible tion of the mechanisms accounting for these effects might identify
for about a third of all cancers attributable to infections [13,14], which therapeutic targets for patients with inammatory bowel diseases
mainly include gastric, liver, colorectal, and cervix uteri cancers and colitis-associated colorectal cancer. Various microbiota bacteria
[15,16]. In addition, both non-mutualistic microorganisms and gut causing inammation-mediated carcinogenesis are presented in Table 1.
microbiota, i.e. mutualistic microorganisms permanently inhabiting Besides, following microenvironment changes, the microbiota mem-
the gastrointestinal tract and which play a key role in digestion, ber E. coli can alter the microbial composition in the intestinal gut in
immunity, and cancer prevention [15,17], may trigger aggravating order to induce the expansion of microorganisms with genotoxic
inammatory responses in various cancer types, including blood capabilities, including itself [13,17,26] (Fig. 1A). This alteration may
cancers such as leukemia [1820] and lymphoma [2123]. also result in an increase in epithelial cell susceptibility of hijacking
M.J. Kipanyula et al. / Cellular Signalling 25 (2013) 403416 405

Table 1 instance. Various non-microbiota pathogens causing inammation-


Various microbiota bacteria causing inammation-associated carcinogenesis. mediated carcinogenesis or aggravating cancer aggressiveness are
Bacteria Types of cancer presented in Table 2.
The question why/how microbiota members can become pathogenic
Bacillus spp. Bladder cancer [66]
Bacteroides spp. Colorectal cancer [74,155,156] and start or favor the establishment of microbial chronic inammatory
Enterobacter spp. Solid [136,157,158] and blood cancers [34,35] diseases eventually resulting in cancer has been puzzling the scientic
Enterococcus spp. Solid cancers [137,138,157] community for decades. It has been hypothesized that the relatively
Escherichia coli Breast [159], bladder [66], and colorectal [17,160] cancers,
low burden of microbes in urban environment may have a profound in-
renal cell carcinoma [33], acute myeloid leukemia [18]
Helicobacter spp. Gastric, liver, cervical [13,26], esophageal [15], uence on the immune function and eventually on inammation-
and colon [16] cancers, lymphoma [21,105] mediated carcinogenesis [37]. A recent systematic review of the litera-
Klebsiella spp. Solid cancers [157,161] ture [37] revealed that up to date, only few studies have addressed the
Pseudomonas spp. Solid [64,157,160,161] and blood cancers [162165] effect of exposure to the environmental microbiota on the risk of cancer
development. Interestingly, from immunological studies it appears that
inammatory cytokines, and more frequently IL-23 released by
by various pathogenic non-microbiota bacteria (e.g. Neisseria spp. T-helper 17 (Th17) cells, are closely linked with tumor-associated in-
[28]), viruses (e.g. Hepatitis C virus [29]), fungi (e.g. Candida albicans ammation; however, the precise mechanisms accounting for the rela-
[30]), and parasites (e.g. Giardia intestinalis [31]), promoting malig- tionship between Th17/IL-23 axis and carcinogenesis are not well
nant transformation of various cell types, including epithelial cells understood. Moreover, a growing body of evidence indicates that Th17
in melanoma [32] and carcinoma [16,33], lymphocytes in several cells are associated with inammation and aberrant angiogenesis in
lymphoma and other blood cancers [22,23,3436] (Fig. 1B,C), for various cancer types, including leukemia [38], for instance. Considering

Fig. 1. Gut microbiota and carcinogenesis. Gut microbiota, a group of microorganisms permanently inhabiting the gastrointestinal tract and which plays a key role in digestion, im-
munity, and cancer prevention [15,17] can promote malignant transformation of various cell types following alterations of their microenvironment. In colorectal carcinoma genesis
(A), epithelial cells invaded by genotoxic bacteria start their transformation following DNA damage and produce inammatory factors favoring neoplastic cell survival. In
lymphomagenesis (B), the transformation of invaded lymphocytes starts following DNA damage induced by genotoxic bacteria that have entered the bloodstream. Affected
immune cells produce a large amount of pro-inammatory factors that favor neoplastic cell survival. Similarly, genotoxic bacteria can infect several other cell types that they
reach transported by the bloodstream, resulting eventually in breast, gastric, cervical, liver, and bladder cancers (C), for instance. MMIF: macrophage migration inhibitory factor.
ROS: reactive oxygen species. TLRs: Toll-like receptors.
406 M.J. Kipanyula et al. / Cellular Signalling 25 (2013) 403416

Table 2 factor- [51], prostaglandin E2 [52], and reactive oxygen and nitro-
Various non-microbiota pathogens causing inammation-associated carcinogenesis or gen species [53,54].
aggravating cancer aggressiveness.
Body failure to terminate inammatory response results in its
Types of cancer chronicity, with the establishment of deleterious inammatory cycles
Viruses Cytomegalovirus Glioblastoma [166], prostate cancer [167] [4,8,9]. The hallmarks of chronic inammation are inammatory foci
EBV Immunoblastic lymphoma [36,103], dominated by lymphocytes, plasma cells, and macrophages [55],
Kaposi's sarcoma [104] which produce a high amount of cytokines, chemokines, growth fac-
HBV, HCV Gastric, liver, cervical cancers [13,168]
tors, and reactive oxygen and nitrogen species that may cause contin-
Herpes viruses Kaposi's sarcoma [92,104], melanoma [32]
HIV Plasmablastic lymphoma [22,23,36], uous tissue damage [56]. Interestingly, reactive oxygen and nitrogen
Kaposi's sarcoma [104] species abundantly released in these pathological conditions may
HPV Gastric, liver, cervical [13,113,158], produce mutagenic agents, including peroxynitrite (ONOO ), for in-
prostate [167] cancers stance, which reacts with DNA and causes mutations and damages
Leukemia viruses T-cell leukemia [102], prostate cancer [167]
in proliferating cells [57,58]. The DNA alterations caused may predis-
Bacteria Acinetobacter spp. Solid [6466] and blood cancers [63]
Aeromonas spp. Solid [169] and blood cancers [170,171] pose to neoplasia [59]. In addition, the high concentrations of
Neisseria spp. Prostate [172] and cervical [31] cancers pro-inammatory cytokines and factors like TNF- and macrophage
Propionibacterium spp. Gastric [158], prostate [167,172,173] cancers migration inhibitory factor (MMIF) released by macrophages and
Staphylococcus aureus Solid cancers [64,66,160,161]
T-lymphocytes may exacerbate further DNA damage [60]. MMIF
Parasites Cryptosporidium spp. Colorectal Cancer [174,175]
Plasmodium spp. Burkitt's lymphoma [176,177] contributes to tumorigenesis by interfering with the crucial
Schistosoma spp. Bladder cancer, cholangiocarcinoma [175] cell-cycle signaling pathway cyclin/Rb/E2F [61], and also by impairing
Fungi Candida spp. Leukemia [19,20], other blood cancers [178] p53-dependent protective responses, thus causing the accumulation
Opportunistic yeast Solid and blood cancers [179] of oncogenic mutations [62]. Moreover, the inammatory microenvi-
EBV: Epstein-Barr virus. HBV: hepatitis B virus. HCV: hepatitis C virus. HIV: human im- ronment is favorable to the survival and proliferation of neoplastic
munodeciency virus. HPV: Human papillomavirus. cells [46,47], indicating that the modulation of factors fuelling chronic
inammation may have anticancer effects.
recent reports indicating that regulatory T cells have a counter- Intriguingly, a recent study has revealed that some commonly
regulatory effect on Th17 cells and even inhibiting their functions employed antineoplastic agents causing DNA damage-induced apopto-
[39,40], it can be hypothesized that continuous exposure to environmen- sis in more sensitive cells, such as transforming cells, have antibacterial
tal microbiota strengthens the inammatory cell regulatory network and activities against Acinetobacter spp. [63], non-microbiota pathogens as-
prevents the establishment of chronic inammation. Conversely, limited sociated with various solid [6466] and blood cancers [63]. These
constant exposure to environmental microbiota may favor the establish- agents include the alkaloid vincristine, the alkylating-like agent cisplat-
ment of chronic inammation. Signaling pathways accounting for in, and the small molecule doxorubicin. The current ndings indicate
inammation-mediated carcinogenesis triggered by microbiota bacteria that Acinetobacter spp. become as sensible to DNA damage as
and other pathogens are considered in the following sections. transforming cells during inammation-mediated carcinogenesis, indi-
cating the complexity of the interactions between cancer-related
2.2. Outcome of immune defense against pathogens: mutagenic pathogens and transforming cells. Similarly, eradication of the Gram-
responses and cancer negative bacterium Campylobacter jejuni in immunoproliferative small
bowel disease suppresses the related inammation-mediated lympho-
Many pathogens can cause chronic inammations, including magenesis at its early stage [21]. Future studies addressing the relation-
the protozoan Trypanosoma brucei which causes inammation- ship between bacteria and transforming cells may provide new insights
associated functional alterations [41,42], fungi like Cryptococcus in bacteria-triggered inammation and in the subsequent carcinogenic
neoformans and Aspergillus fumigatus which take advantage of immu- processes.
nocompromised hosts to cause infection [43], and various bacteria
and viruses. As discussed earlier, emerging evidence indicates a 3. Pattern-recognition receptors: example of Toll-like receptors
strong link between cancer and microbiota (Table 1) or other patho-
gens establishing chronic inammation (Table 2). The mechanisms by 3.1. TLRs, pathogen-triggered inammation, and cancer: lessons from
which different microbial pathogens contribute to cancer develop- microbiota
ment are complex. A spate of recent investigations suggests that
these mechanisms involve interplay between chronic inammation, Toll-Like Receptors (TLRs), transmembrane proteins containing
direct microbial effects on host cell physiology and, ultimately, leucine-rich repeats, are involved in sensing endogenous danger sig-
changes in tissue homeostasis. nals and play a critical role in the early innate immune response to in-
The inammatory responses that successfully eliminate PAMPs vading pathogens by sensing PAMPs, predominantly through MyD88
and DAMPs are actively terminated and the healing process starts (myeloid differentiation primary response gene 88) adaptor protein
thereafter. Early studies revealed that the phagocytosis of apoptotic [1,2]. For instance, TLR/MyD88 signaling prevents the dissemination
cells enhances the production of anti-inammatory mediators, of bystander bacteria to deeper tissues during Clostridium difcile in-
promoting anti-inammatory response [44], and conversely, recent fection, by triggering neutrophil and monocyte recruitment to the
ndings indicate that impaired phagocytosis promotes inammation, lamina propria of the large intestine through mechanisms involving
to recruit more immune cells [45], suggesting a crosstalk between chemokine signaling pathways like CCL2/CCR2 (CC receptor type
inamed tissue and immune cells. Such crosstalk would be mediated 2)- and CXCL1 (CXC motif ligand 1)/CXCR2 (CXC motif receptor
by the anti- and pro-inammatory factors abundantly produced 2); mortality is markedly increased in MyD88-decient mice follow-
by both inltrating and resident cells [46,47]. Studies in a rat ing C. difcile infection [2]. Loss of function studies have shown that
carrageenin-sponge implant model have revealed a shift from genetic silencing of PRR signaling protects against chronic inamma-
antibacterial tissue damage to tissue repair involving factors with tory diseases [67], indicating that perturbations of microbiotaPRR
both pro-inammatory and anti-inammatory effects of which the interactions may promote inammation.
effects depend on the microenvironment at the inamed site Translocation of the microbiota from the gut into the systemic
[48,49]. Such factors have been observed in humans as well and in- milieu can cause bacterial sepsis [68,69]. Recent reports suggest that
clude, for instance, metalloproteinases [50], transforming growth small leucine-rich proteoglycans such as decorin and biglycan
M.J. Kipanyula et al. / Cellular Signalling 25 (2013) 403416 407

orchestrate TLR crosstalk during inammation [70,71]. Septic inam- may have an effect on the prevention of colorectal cancer by scaveng-
mation evokes early responses which include activation of the gene ing toxic compounds or preventing their generation in situ [75]. This
encoding decorin; decorin protein level is increased in the plasma of hypothesis should be investigated, considering the importance for co-
septic patients and mouse models, and decorin increases the expres- lorectal cancer prophylaxis.
sion of pro-inammatory molecules in cancer cells [70]. Interestingly, Interestingly, other recent studies have shown that infections with
the ndings from recent studies aiming at unraveling the mecha- H. pylori activate oncogenic signaling pathways like Sonic Hedgehog
nisms used by decorin signaling to control inammation and tumor [26] and Wnt [76,77], which have been reported anti-apoptotic ef-
growth revealed two mechanisms [70]: i) rst, decorin may act as fects and the ability to confer stem-like properties to transforming
an endogenous ligand of TLR2 and TLR4 to stimulate the production cells in various solid and blood cancers (for review see [78,79] and
of pro-inammatory molecules, including programmed cell death [80,81]), respectively. In the APCMin/+ mouse, an animal model of
protein 4 (PDCD4), in macrophages; ii) second, decorin may prevent human colon cancer where a mutation in the murine homolog of
translational repression of PDCD4 by decreasing the activity of the Wnt inhibitor APC gene is induced, a recent report has indicated
transforming growth factor (TGF)-1 and the expression of the key that gut microbiota accelerate tumor growth in comparison with
regulator of oncogenic processes miR-21, which is also a translational wild type mice [82], suggesting a role for the Wnt signaling pathway
inhibitor of PDCD4. In addition, still in these studies, increases in in that context. Rapid tumor growth occurred concomitantly with
PDCD4 levels led to decreases in the expression of the anti- systemic anemia, suggesting a role for intestinal barrier damage and
inammatory cytokine IL-10, thereby making the cytokine prole erythropoiesis-stimulating mitogens. The microbial cell wall compo-
more pro-inammatory. Furthermore, increases in expression of nent LPS accounted for at least part of the cell growth acceleration ob-
pro-inammatory molecules have been associated with tumor served, via c-Jun/JNK signaling pathway. In addition, inltrating
growth promotion and poor patient outcomes in hepatocellular carci- CD11b + myeloid cells released high levels of phosphorylated STAT3
noma [72], for instance. (p-Tyr705) in colonic tumors. These ndings indicate that gut
Experimental evidence suggests that TLRs are key players in microbiota-triggered tumor growth acceleration observed in APC
pathogen-triggered inammation-mediated carcinogenesis. Gut-derived (Min/+) mice is mediated by the c-Jun/JNK and STAT3 signaling
LPS has demonstrated the ability to promote hepatocellular carcinoma pathways in combination with anemia. These ndings point out the
development by activating TLR4 expression on myeloid-derived immune complexity of the mechanisms accounting for the inammatory and
cells in different types of liver injury models, including concanavalin carcinogenic effects of microbiota bacteria.
A-induced hepatitis model, which mimics human viral hepatitis [3], fur-
ther indicating that gut microbiota and TLR4 signaling modulation may 3.2. TLR signaling and inammation
represent potential avenues for therapeutic intervention aimed at treat-
ment of hepatitis virus-induced hepatitis and prevention of hepatocellu- TLRs recognize bacterial, viral, and parasite PAMPs in the extracellu-
lar carcinoma. Similarly, Ochi and collaborators [73] have found that lar environment (TLR1, 2, 46, 11) or in endolysosomes (TLR3, 79,
TLR4 signaling plays a key role in the cellular and molecular mechanisms 10). They can bind several PAMPs/DAMPs and are thought to even
bridging chronic pancreatic broinammatory disease and pancreatic bind common self molecules such as heat shock proteins and brin-
carcinogenesis. These investigators observed that TLR4 signaling ogen, suggesting a link between the stereotypic inammatory response
triggered by LPS exacerbates pancreatic inammation and accelerates triggered by TLRs and autoimmune diseases [83,84]. Stimulation of
tumorigenesis, whereas TLR4 inhibition or blockade of the MyD88- TLRs by the corresponding PAMPs or DAMPs initiates signaling cas-
independent Toll/IL-1 receptor domain-containing adapter-inducing cades leading to the activation of transcription factors, such as AP-1,
interferon- (TRIF) pathway have protective effects against pancreatic NF-B, and interferon regulatory factors (IRFs). TLR signaling is trans-
cancer. Surprisingly, blockade of the MyD88-dependent pathway exacer- duced in the cytoplasm by a Toll/ IL-1 receptor (TIR) domain, which
bated pancreatic inammation and malignant progression, through den- is the docking site of TIR-containing cytoplasmic adaptor proteins
dritic cell (DC)-mediated induction of pancreatic antigen-restricted that are critical in orchestrating the signal transduction pathways
Th2-deviated CD4 (+) T cells. Such pro-tumorigenic and bro- after TLR and IL-1 receptor activation. TIR adaptor molecules mainly in-
inammatory effects of MyD88 inhibition indicate the complexity of clude signaling proteins like MyD88, MyD88 adaptor-like (MAL), TRIF,
TLR signaling in carcinogenesis. TRIF-related adaptor molecule (TRAM), and the negative regulator of
In addition, Uronis and collaborators [74] have demonstrated that TLR pathways SARM (sterile-alpha and Armadillo motif containing
bacterial-induced inammation drives progression from adenoma to protein) [85,86]. Except for TLR3 all TLRs engage the adaptor MyD88,
invasive carcinoma in azoxymethane (AOM oncogene)-exposed however either directly as IL-1R (TLR5, 711, and heterodimeric
IL-10 knockout mice, a model of colitis-associated colorectal cancer. TLR1-TLR2 and TLR2-TLR6) or in association with the adaptor MAL/
These investigators have observed colon tumors in 62% of TIRAP (Toll-IL-1 receptor (TIR) domain containing adaptor protein.
AOM-IL-10(/) mice against only 20% in AOM-wild-type mice; e.g. TLR4, and heterodimeric TLR1-TLR2 and TLR2-TLR6).
and tumor multiplicity directly correlated with the presence of MyD88 signaling is transduced through a death domain (DD)
colitis. Interestingly, IL-10(/) mice mono-associated with the which mediates interactions with the DD of the serine/threonine ki-
mildly colitogenic bacterium B. vulgatus displayed signicantly re- nase IRAK4, the clustering of which would result in its autophospho-
duced colitis and colorectal tumor multiplicity; and germ-free rylation within the receptor complex, allowing a kinase activity-
AOM-treated IL-10(/) mice were devoid of tumors. In addition, dependent binding of IRAK4 DD to the DD of the related kinases
AOM-treated IL-10 (/) or MyD88 (/) mice displayed a re- IRAK1 and 2 [87]. The MyD88IRAK4IRAK1/2 complex nucleate a
duced mRNA expression of TNF- and IL-12p40, a known component larger complex including, E3 ubiquitin ligases like TRAF6 (tumor ne-
of IL-12 and IL-23. These mice showed no signs of tumor develop- crosis factor receptor associated factor 6), cIAP1 (cellular inhibitor
ment, indicating that the TLR/MyD88 pathway is essential for of apoptosis-1), and cIAP2, and the E2 ubiquitin-conjugating enzyme
microbiota-induced development of colitis-associated colorectal can- Ubc13 (ubiquitin-conjugating enzyme E2N), in the case of TLR4
cer. These ndings also indicate that intestinal microbiota modulation [88,89]. TRAF6 catalyzes the formation of polyubiquitin chains on ly-
may decrease cancer risk in inammatory bowel disease. Accordingly, sines within itself and in IL-1 receptor-associated kinase 1 (IRAK1),
it has been hypothesized that probiotics, i.e. live microorganisms con- promoting the K63-linked polyubiquitination of cIAPs and the re-
ferring a health benet on the host when administered in adequate cruitment of adaptor proteins TAB2 and 3, resulting in the activation
amounts, are capable of transient modulation of the microbiota, that of the mitogen-activated protein kinase (MAPK) kinasekinase
have been successfully used to manage inammatory bowel disease (MAPKKK) TAK1 (transforming growth factor--activated kinase 1)
408 M.J. Kipanyula et al. / Cellular Signalling 25 (2013) 403416

[89,90]. In addition, K63-linked poly-ubiquitin also binds the regula- 4. NF-B, pathogen-triggered inammation, and cancer
tory subunit of the NF-B inhibitor (IB) kinase (IKK) complex
termed as NEMO, allowing its recruitment to the TLR4 signaling 4.1. NF-B, infection, and cancer
complex [91]. TAK1 mediates the activation of the downstream
MAPKs p38 and c-Jun N-terminal kinase (JNK), which are hijacked As TLR pathways, many other inammatory signaling pathways
by activating mutations in pathogen-related cancers like herpes transduce their signal through NF-B activation, including pathways
virus-associated Kaposi's sarcoma [92], and probably participate to mediated by the PRR members NLRs (nucleotide oligomerization
inammation-mediated carcinogenesis. Activated TAK1 complex domain-like receptors) [98] and C-type lectin receptors [100]. NF-B
translocates into the cytosol, where it activates: i) the kinase MKK4 signaling pathway has a critical role in human diseases and carcinogen-
that results in the phosphorylation/activation of JNK [89]; ii) and esis. Not surprisingly, mutations of NF-B signaling molecules have
the related kinases MKK3 and MKK6 for the activation of p38 [93]. been reported in several malignancies. For instance, the somatic
In turn, JNK stimulates the expression of pro-inammatory mediators mutation of TNFAIP3, the gene encoding for the deubiquitinating factor
like TNF- through an AP1 transcription factor-dependent mecha- A20, is frequently reported in human B-cell lymphomas, including
nism [94], whereas p38 activates the cAMP response element- Epstein-Barr virus-associated AIDS-related lymphoma [36] and
binding (CREB), which results in the activation of the CCAAT- Kaposi's sarcoma [101], pointing out A20 as a tumor suppressor in
enhancer-binding proteins (c/EBPs) family of transcription factors, these blood cancers. In addition, in cancer cells collected from adult
and in the subsequent induction of various genes involved in the in- T-cell leukemia patients (a fatal T-cell malignancy associated with
ammatory response, such as: cell adhesion molecules; chemokines human T-cell leukemia virus type I infection), the suppression of
like CXCL-1 and CXCL-2; and cytokines like IL-1, IL-10, IL-12, and NF-B-dependent transcriptional activity by the IB kinase 2 inhibitor
IL-32 [95,96]. IMD-0354 decreased cell survival [102]. Comparably, in EpsteinBarr
On the other hand, IKK induces the activation of the NF-B transcrip- Virus (EBV)-associated immunoblastic lymphoma, which occurs in
tion factor, which results in subsequent transcriptional responses in- immunocompromised patients such as those with AIDS or transplant
volving the MAPKs of the extracellular-signal-regulated kinase (ERK) recipients upon primary EBV infection or following the reactivation
family that, in turn, activate the ATF (activating transcription factors)/ of a pre-existing latent EBV infection, the HIV protease inhibitor
CREB family of transcription factors, resulting in the induction of genes ritonavir inhibits the tumor growth and inltration of EBV-positive
involved in both pro- and anti-inammatory responses such as PTGS2 lymphoblastoid B cells by targeting NF-B signaling [103]. Tang
gene that encodes prostaglandin-endoperoxide synthase 2 or cyclooxy- and collaborators [104] have recently demonstrated that Kaposi's
genase 2 (COX-2), involved in the synthesis of prostaglandins [97,98]. In sarcoma-associated herpesvirus (KSHV) infection is necessary, al-
addition, the canonical IKK/NF-B signaling pathway can also accumu- though, not sufcient for disease development without other cofac-
late in the nucleus and induce the expression of several other genes tors. The authors identied human immunodeciency type 1 (HIV-1)
encoding for inammatory-related molecules, including [98]: PRRs like Tat and herpes simplex virus 1/2 (HSV-1/2) as such cofactors. Interesting-
advanced glycation end product (AGE) receptors, chemokines like ly, the researchers also demonstrated that HSV-2 infection activate NF-B
CCL2 (MCP-1), CCL3 (MIP-1), CCL5 (RANTES) and CXCL1/2; growth signaling pathway, further suggesting a role of HSV-2 in the pathogenesis
factors like GM-CSF (granulocyte macrophage colony-stimulating fac- of Kaposi's sarcoma. Recent studies based on a transgenic mouse model
tor); adhesion molecules like E- and P-selectins , ICAM1 (intercellular have revealed that the prophylactic delivery of IL-15 affords protection
adhesion molecule 1) and VCAM1 (vascular cell adhesion molecule 1); against a lethal HSV-2 challenge and metastasis of B16/F10 melanoma
cytokines like IL-1, IL-12, and TNF-; metalloproteinases like MMP9; cells through NF-B dependent induction of the chemokine CCL5 by my-
the complement factor B; Caspase-11; but also, inhibitors of NF-B sig- eloid immune cells [32]. NF-B may also bridge chronic inammation and
naling like A20 and IB- , the inhibitor of macrophage activation carcinogenesis in bacterial infection-related cancer such as gastric MALT
CD200, the inhibitor of virus proliferation IFN (interferon)-, and the lymphomas associated with H. pylori and Ocular Adnexa MALT lympho-
pleiotropic cytokine IL-6. These observations indicate that NF-B can mas of whose association with Chlamydia psittaci infection has been re-
induce and also regulate the inammatory response. cently demonstrated [105].
Emerging data suggest that TLR signaling role in pathogen-
triggered inammation and carcinogenesis is complex, and therefore,
requires further investigations. For instance, Smolinska and collabo- 4.2. NF-B signaling and carcinogenesis
rators [1] have investigated the role of Src family kinases, i.e. the
largest non-receptor tyrosine kinase family, in TLR signaling, using NF-B may link inammation to cancer through its inherent abil-
primary human macrophages in combination with adenoviral ity to induce the production of adhesion molecules, MMPs, COX-2,
overexpression and small interfering RNA knockdown. The study re- pro-inammatory cytokines, and reactive oxygen and nitrogen spe-
vealed that the Src kinase Hck has a key role in LPS/TLR4-induced cies [98]. In normal inammatory response NF-B-induced adhesion
TNF- and IL-6 production. Furthermore, ndings have suggested molecules, chemokines, and vasomodulatory molecules play a key
that Hck may mediate TLR4-induced transcription of both TNF- role in the processes resulting in immune cell inltration in infected
and IL-6 by a mechanism that does not involve NF-B and MAPK tissue. However, these processes are also crucial in the inammatory
pathways, but rather leads to p38 MAPK-dependent activator protein microenvironment of cancer where adhesion molecules are used
(AP-1) binding with a complex of c-fos and JunD, pointing out the by tumor cells to facilitate migration and positioning during the
possibility of targeting Hck for the alleviation of excessive inam- metastatic process [72]. In addition, MMPs facilitate tumor invasion
mation. More recent studies involving i.p. or s.c. injection of the through their proteolytic activity and consequent ability to modulate
TLR3 interactor polyI:C (Polyinosinicpolycytidylic acid) to Lewis vascular permeability, whereas pro-inammatory and growth factors
lung carcinoma tumor-implant mice have shown that TLR3 signal- create a microenvironment favorable for cancer cell survival and
ing induces tumor regression by converting tumor supporting proliferation, as already mentioned. Interestingly, a feedback loop
macrophages into tumor suppressors [99], anticipating, therefore, between pro-inammatory cytokines, such as TNF-, for instance,
that better characterization and understanding of the signaling and NF-B activation has been suggested; such feedback loop may
pathways mediated by TLR may provide novel pharmacologic tar- be responsible for the constitutive activation of NF-B in inammato-
gets against pathogen-associated cancers. Fig. 2 illustrates how ry diseases, with crucial roles in the link between inammation and
TLR signaling downstream targets integrate to induce inammato- cancers [106,107]. However, the nature of such feedback loop has
ry effects. not yet been characterized.
M.J. Kipanyula et al. / Cellular Signalling 25 (2013) 403416 409

Fig. 2. Toll-like receptor signaling and inammation. The interactions of pattern recognition receptors (PRRs), like toll-like receptors (TLRs) situated on the cell or endolysosome mem-
branes of mononuclear phagocytes, with pathogen-associated or danger associated molecule patterns trigger both the mitogen associated protein kinase (MAPK) kinase-kinase TAK1
(transforming growth factor--activated kinase 1) and the IB (inhibitor of nuclear factor B) kinase IKK signaling pathways. These pathways induce the production of either
pro-inammatory factors, or both pro- and anti-inammatory factors, according to the tissue microenvironment [98], including pro- and anti-inammatory cytokines, chemoattractant
molecules, adhesion molecules, growth factors, other PRR receptors. Activating mutations of the components of these pathways have been reported in various microbe-associated can-
cers (see text). CCL2/3/5: CC motif ligand 2,3 or 5 chemokines. cIAPs: cellular inhibitors of apoptosis. CXCL1/2: CXC motif ligand 1 or 2. ERK: extracellular-signal-regulated kinase. E3:
ubiquitin E3. GM-CSF: granulocyte macrophage colony-stimulating factor. ICAM-1: intercellular adhesion molecule 1. IFN-: interferon beta. IL: interleukin. IRAKs: interleukin-1
receptor-associated kinases. NF-kB: nuclear factor kappa B. TNF-: tumor necrosis factor alpha. TRAF6: tumor necrosis factor receptor associated factor 6. Ubc13: ubiquitin-
conjugating enzyme E2N. VCAM-1: vascular cell adhesion molecule 1.

Interestingly, in squamous epithelium human keratinocyte prolif- transmembrane protein with a cysteine-rich extracellular domains
eration induced by bacterial LPS was found to be dependent on NF-B (CRDs) that binds TNF-. The DD of TNFR1 cytoplasmic tail interacts
activation and subsequent cyclin D1 up-regulation [108]. Thus, with the DD of the adaptor TRADD, which allows the binding of RIP1
NF-B may also contribute to the genomic instability through its to TNFR1 that results in the recruitment of TRAF-2, an adaptor for
antiapoptotic activities that prevent mutated precancerous cells the ubiquitin ligases cIAP1 and cIAP2 [98]. On the other hand, IL-1
from being eliminated [109], and through the promotion of reactive triggers IL-1R/MyD88 signaling that plays key roles in cancer devel-
oxygen species production, which has the potential to cause carcino- opment, as revealed for instance by a recent study investigating ma-
genic mutations [59,109]. In various experimental models where lignant transformation of keratinocytes [112]. More specically,
tumor growth was increased by LPS/TLR4 signaling, the inhibition of genetic and pharmacological approaches showed that the differenti-
NF-B signaling in cancer cells by ancient microbial molecules ation and pro-inammatory effects of oncogenic Ras/MAPK in
resulted in the regression of tumors and metastases [110,111], keratinocytes requires IL-1/IL-1R/MyD88 signaling, which phos-
pointing out NF-B as a new avenue for novel therapies. phorylates the NF-B inhibitor IB, resulting in NF-B activation.
Recent reports have indicated key roles for IL-1R/MyD88 and
5. Pro-inammatory cytokines and COX-2 TNFR1/TRADD signaling pathways in inammation-related carci-
nogenesis. For instance, a population based study performed in
Several pro-inammatory cytokines have been associated with Argentine women has revealed a link between TNF- promoter
inammation-mediated carcinogenesis triggered by pathogenic mi- polymorphisms in human papillomavirus infection and cervical
croorganisms. Numerous data point out IL-1, TNF-, IL-32, IL-23, cancer risk [113]. Another recent population-based study con-
and IL-6 as pivotal. In addition, recent ndings indicate that IL-8 ducted in Brazil has addressed the effect of pro-inammatory
and IL-29 can team up with the pro-inammatory factor COX-2 to cytokine gene polymorphisms on the development of gastric
trigger/sustain infection-related carcinogenesis. cancer in H. pylori-infected patients [114]. Some IL-1 mutations
(IL-1-511 C/C and IL-1-511 C/T alleles) appeared to be
5.1. IL-1 and TNF- associated with chronic gastritis in H. pylori-positive patients,
whereas associations of an IL-1 mutation (IL-1-511 C/C geno-
The pro-inammatory cytokines IL-1 and TNF- released by PRR type) and of a TNF- mutation (TNF--308) with gastric cancer
signaling amplify the inammatory response through signaling were shown. Thus, IL-1 and TNF- gene polymorphisms may be
pathways like NF-B and the oncogenic MAPK. TNF- mediates its associated with chronic gastritis and gastric cancer development
pro-inammatory effects through TNF receptor 1 (TNFR1), a type 1 in H. pylori-infected individuals.
410 M.J. Kipanyula et al. / Cellular Signalling 25 (2013) 403416

5.2. IL-32 where pathogenic roles of mutations affecting the IL-8/CXCR2/P53


signaling pathway have been shown [121]; ovary cancer, where IL-8
The role of the new pro-inammatory cytokine IL-32 over- overexpression is associated with increased tumor growth and
expressed in inammatory diseases and gastric cancer has been in- metastases [122]; and breast cancer, where the production of IL-8 ac-
vestigated in H. pylori infection [115]. In this study, IL-32 expression tivates matrix metalloproteinases and promotes invasion of breast
paralleled human gastric tissue pathology, with higher expressions epithelial and cancer cells [123].
in H. pylori-associated gastritis and gastric cancer tissue. Interestingly,
H. pylori-induced expression of IL-32 was NF-B-dependent, and as- 5.4. COX-2
sociated with the expression of pro-inammatory molecules, includ-
ing CXCL1/2, and IL-8, suggesting a role for IL-32 in the regulation COX-2 expression can also be induced by a wide range of stimuli,
of cytokine expression in H. pylori-infected gastric mucosa. Similarly, including LPS, pro-inammatory cytokines, such as IL-1 and TNF-,
in studies assessing IL-32 role in hepatitis C virus-related liver inam- and a growth factor like EGF (epidermal growth factor). The products
mation, IFN- exerted a signicant additive effect on TNF--induced of COX-2 enzyme are prostaglandins, which are key mediators of in-
but not IL-1-induced expression of IL-32 in CD14 + monocytes, ammation. Various non-steroidal anti-inammatory drugs (NSAIDs)
through a mechanism dependent on both NF-B and Janus kinase/ affect COX-2 activity by covalent modication or competition for a
signal transducers and activators of transcription (JAK/STAT) signal- substrate binding site. Indeed the long-term use of NSAIDs was
ing [116]. IL-1 and TNF- signicantly induced IL-32 expression in shown by population-based studies to reduce the risk of several can-
Huh-7.5 liver cells. Viral infection of Huh-7.5 cells resulted in an cers [124]. COX-2 is also overexpressed in various types of cancer and
11-fold increase of IL-32 mRNA expression. In addition, IL-32 involved in cellular proliferation, antiapoptotic activity, angiogenesis,
overexpression has been also observed in human hepatoma cells, and an increase of metastasis. A recent report demonstrates that
and its expression was increased by pro-inammatory stimuli. COX-2 pathway can also be induced by cigarette smoke in human
These observations point out IL-32 as a novel pro-inammatory cyto- lung broblasts, suggesting a possible mechanism for the cigarette
kine involved in HCV-associated liver inammation and cancer. Fur- smoking-triggered cancer-prone inammatory lung diseases [125].
ther investigation on the mechanism controlling the interaction of The functions of COX-2 in linking inammation to cancer are now be-
this cytokine with viral infection may provide better understanding coming a subject of intense investigation. However, there are contro-
of the link between infection-triggered inammation and carcinogen- versies regarding the exact role of COX-2 in the transformation from
esis in gastric and hepatic tissue. Interestingly, IL-32 overexpression inammation to cancer. A recent study using an esophageal rat
studies in surgically resected or biopsied specimens from patients model indirectly supports a notion that COX-2 induction might con-
with localized clear cell renal cell carcinoma (CCRCC) have suggested tribute to the progression of cancers from inammation [126].
that IL-32 is also associated with high recurrence rates of renal cell COX-2 inhibitor, celecoxib inhibits the COX-2 pathway and delays
carcinoma (RCC) further suggesting that it may be a novel prognostic the development and progression from esophageal inammation,
factor that may be useful for predicting the outcomes of the disease in metaplasia, to adenocarcinoma. In addition, another recent study
such patients [117]. demonstrated that COX-2 may be sufcient to cause inammation
and the subsequent cancerous aberrations. Within this study, it was
5.3. IL-8 and IL-29 found that the forced expression of COX-2 transgene under the con-
trol of a keratin-5 promoter causes spontaneous inammation-
Data from recent reports have demonstrated that hepatitis B virus associated transitional cell hyperplasia and transitional cell carcino-
(HBV), a major causative factor associated with the development of ma in urinary bladders of transgenic mice [127]. Nevertheless,
cirrhosis and hepatocellular carcinoma, induces a novel inammation COX-2 expression would be independent of the degree of inamma-
network involving at least three inammatory factors, i.e. IL-29 (or tion, but related to the premalignant cells that already existed in the
IFN-1), IL-8, and COX-2. Yu and collaborators [118] reported that tissue, in the context of Barrett's esophageal epithelium [128], there-
IL-29 inhibits HBV replication and IL-8 attenuates the expression of fore suggesting that COX-2 expression plays an important role in en-
IL-10R2 and the anti-HBV activity of IL-29, favoring the establishment hancing cancer development but is not an indispensable factor for the
of persistent viral infection. It increased expressions of IL-29, IL-8, and transformation of epithelial cells in the scenario of chronic inamma-
COX-2 genes in HBV-infected patients and in HBV-expressing cells tion. This notion is also supported by a recent immunohistochemical
was also observed. Furthermore, in HBV-transfected human lympho- study of human prostate cancers in which the local chronic inamma-
cytes and hepatocytes, the expression of COX-2 was enhanced by IL-8 tion has been found to be able to up-regulate COX-2 expression in ad-
production, which in turn appeared to be IL-29-dependent. Interest- jacent tumor cells and induced angiogenesis [127]. Further research
ingly, COX-2 decreased the production of IL-8, and the latter in this area is needed to understand the specic role of COX-2 in
decreased the expression of IL-29. From these ndings they have hy- linking microbial-triggered inammation and cancer. Interestingly,
pothesized that HBV infection induces a novel inammation cytokine arachidonic acids, substrates for COX-2 to produce prostaglandins,
network involving at least the three inammatory factors IL-29, IL-8, can be converted by another enzyme lipoxygenases to leukotrienes,
and COX-2, which regulate each other through positive regulation which were suggested to be another missing link between inamma-
and negative feedback. Still in these studies, the enhancement of tion and cancer [129].
COX-2 expression by IL-8 proved to be mediated through the tran-
scription factors CREB and c/EBP, and in addition MAPK signaling 6. Oxygen/nitrogen species and microbial-triggered cancer
pathways such as ERK and JNK were also involved in the regulation
of COX-2. These ndings provide new insights for the understanding 6.1. Oxidative and nitrative stress
of the mechanism by which inammatory factors regulate each other
in response to HBV infection and probably in the infection-related Recent studies have strongly indicated that oxidative and nitrative
carcinogenesis. Overexpression of IL-8 has been reported in several stress is a major player in inammation-associated carcinogenesis.
human cancers, including for instance, colon cancer, where serum Various oxygen and nitrogen species and products have been associ-
IL-8 levels is decreased in patients following anticancer treatment ated with microbial triggered inammation-associated cancer. For in-
[119]; melanoma, where the suppression of cancer cell invasion prop- stance, genital infection by high risk HPV, the main etiologic factor of
erties correlates with a downregulation of IL-8 and MAPK phosphor- cervical cancer, is not per se sufcient to induce tumor development
ylation, including p38, JNK, and ERK-1/2 [120]; prostate cancer, and oxidative stress was recently pointed out as a key pathogenic
M.J. Kipanyula et al. / Cellular Signalling 25 (2013) 403416 411

factor cooperating with this pathogenic agent to induce carcinogene- the unique cells making use of glycolysis mediated by HIF-1 for ATP
sis. A study involving tissues collected from patients revealed that in generation constitutively even during normoxia, whereas other cell
dysplastic tissues the oxidative modication of DNA and proteins types use this metabolic system only during hypoxia [141]. Recent
involved in cell morphogenesis and terminal differentiation may pro- studies from Staples et al. [142] have demonstrated that severe hyp-
vide the conditions for the neoplastic progression; in addition, cancer oxia during monocyte to macrophage differentiation as observed in
tissues seem to attain an improved control on oxidative damage and tumors, wounds, arthritic joints, and other sites of inammation, re-
selectively reduced carbonyl adducts on key detoxifying/pro-survival sults in macrophages with a pattern of gene expression distinct
proteins [130]. Inducible nitric oxide synthase (iNOS) is an enzyme- from those maturing in normoxia, characterized by a transcriptionally
catalyzing nitric oxide (NO) production induced by TNF-, IL-1, up-regulated HIF-1 mRNA, pointing out HIF-1 as an important play-
and NF-B, among other inammatory factors [131], which was er in chronic inammation. Hara and colleagues [141] reported that
found to be overexpressed in chronic inammatory diseases and var- the deletion of the Mint3/Apba3 gene in mice abrogates macrophage
ious types of cancer [132]. Similarly, in the chronic inammation in- functions and increases resistance to LPS-induced septic shock, indi-
duced by liver uke (Opisthorchis viverrini) infection, the major risk cating a malfunction of TLR4 response. Interestingly, the ATP level
factor for cholangiocarcinoma in Northeastern Thailand, increases in macrophages of mutant mice lacking Apba3 gene was reduced to
levels of pro-inammatory cytokines, NF-B, COX-2, iNOS activity, 60% of the level observed in wild type cells: Reductions of
and disturb the homeostasis of oxidants/anti-oxidants and DNA re- ATP-dependent activities such as glycolysis, cytokine production,
pair enzymes [133]. Thus, oxidative and nitrative stress-related DNA and motility were observed as well, indicating that unraveling the
damage may occur as the consequence of the overproduction of reac- specic function of APBA3 in macrophages might allow the develop-
tive oxygen and nitrogen species in inamed target cells. Similarly, ment of therapeutics regulating aberrant macrophage function during
nitrative DNA damage via NF-B activation leading to tumor develop- infection and the subsequent inammatory response, and will allow
ment have been reported in urinary bladder cancer associated with further understanding of mechanisms governing septic shock associ-
infections with the parasite Schistosoma haematobium [134]. ated TLR4 inammatory response.
Another recent study [135] has revealed that NO derived iNOS can HIF-1 is expressed in acute and chronically inamed site [143].
induce apoptosis in H. pylori through a pERK pc-Fos/c-Jun HIF-1 regulates the transcription of several genes in response to hyp-
c-Myc ornithine decarboxylase (ODC) spermine oxidase (SMO) oxic stress and changes in growth factors, and plays an important role
pathway. Indeed alterations in the availability and metabolism into in tumor angiogenesis and growth. Crohn disease-associated E. coli
polyamines of L-arginine (L-Arg), controlled by NOS and arginase promote gastrointestinal inammatory disorders by activation of
enzymes, through mechanisms like competition for L-Arg substrate HIF-dependent responses. Mimouna et al. [143] have reported that
by H. pylori arginase and induction of host macrophage arginase II HIF-1 is maximally expressed in inamed ileal epithelium of
(Arg2) may explain failure of this apoptotic process, and contribute Crohn disease patients, in concomitance with the expression of
therefore to the deregulation of the host immune response to the in- CEACAM6 (carcinoembryonic antigen-related cell adhesion molecule
fections. Targeting such signaling pathways may serve as potential 6 or CD66c), a protein that acts as a receptor of adherent-invasive
therapeutic targets in H. pylori-associated carcinogenesis. E. coli. The expression of CEACAM6 in transgenic mice expressing
The intestinal commensal Gram-negative bacteria of the Entero- this receptor induced the production of HIF-1 and the activation of
bacter spp. have been signicantly associated with solid cancer VEGF receptor signaling, and downstream analyses on human intesti-
development [136] and more severe disease in cancer patients nal epithelial cells silenced for hif-1, highlighted the crucial role of
[137]. Interestingly, E. faecalis produces extracellular superoxide and CEACAM6 in the production of the latter pro-angiogenic factors,
promotes chromosome instability via infected macrophage-induced highlighting the crucial role of adherent-invasive E. coli as promoter
bystander effects mediated by 4-hydroxy-2-nonenal (4-HNE), a dif- of inammatory disorders and neoplastic cell favoring environment
fusible breakdown product of -6 polyunsaturated fatty acids; and in the gastrointestinal tract. HIF-1 protein plays a major role in
IL-10 knockout mice colonized with superoxide-producing E faecalis mediating this effect, and therefore may represent a good target for
developed inammation and colorectal cancer, whereas colonization colorectal cancer treatment.
with a superoxide-decient strain resulted in inammation but not A phase II study trial in patients diagnosed with HPV revealed the
cancer [138]. The potential of antioxidants as anti-tumorigenic agents presence of non-HIF-1 ligand-induced VEGF, which was associated
of has been revealed by a transgenic Rac1 model of Kaposi's sarcoma with a high risk head and neck squamous cell carcinomas' [144], indi-
[139], the major HIV-associated malignancy, whose hallmark is the cating the presence of activating mutations hijacking the HIF-1 recep-
proliferation of spindle cells, inammatory inltrate, and aberrant an- tor signaling pathway. Comparably, angiogenesis promotion through
giogenesis caused by Kaposi's sarcoma herpesvirus (KSHV) infection the enhancement of HIF-1 and VEGF expressions induced by the
[104]. The expression of Rac1, a small GTPase triggering reactive oxy- overexpression of HPV type 16 oncoproteins has been reported in
gen species (ROS) production by NADPH-oxidases, is sufcient to non-small cell lung cancer cells [145]. HPV E7 oncoprotein enhances
cause Kaposi's sarcoma-like tumors in transgenic mice through HIF-1-mediated transcription by inhibiting binding of histone
mechanisms involving ROS-driven proliferation mechanisms, AKT deacetylases HDAC1, HDAC4, and HDAC7 to increase the expression
signaling, and hypoxia-inducible factor 1 (HIF-1)-related angio- of pro-angiogenic factors in cervical cancer cells [146], indicating
genesis, pointing out host and viral genes triggering Rac1 or ROS pro- that HPV oncoproteins may induce a microenvironment favorable to
duction as key determinants of Kaposi's sarcoma onset and potential their replication through epigenetic control of various genes, includ-
chemopreventive or therapeutic targets [139]. A growing body of ing genes encoding for HIF-1 and the genes it regulates.
evidence has pointed out the ROS product HIF-1 as a major player Hepatitis C virus (HCV) infection is a major risk factor for the de-
in microbial-triggered carcinogenesis. velopment of hepatocellular carcinoma [13]. HCV gene expression in-
duces oxidative stress, which in turn induces the stabilization of
6.2. Hypoxia-inducible factor 1 HIF-1, followed by VEGF and angiogenic cytokine release, leading
to tumorigenic neovascularization in vivo [147]. Such HIF-1 stabili-
HIF-1 is a heterodimeric transcription factor and key regulator zation requests the activation of NF-B, STAT-3, PI3K/Akt, and
responsible for the induction of genes that facilitate adaptation and p42/44 mitogen-activated protein kinase. A study aimed at clarifying
survival of cells during hypoxia. HIF-1 consists of a constitutively whether human papillomavirus (HPV) directly affects the oncogene-
expressed subunit HIF-1 and an oxygen sensitive subunit HIF-1 sis and biologic behavior of tonsillar cancer revealed that compared
(or its paralogs HIF-2 and HIF-3) [140]. Macrophages would be with HPV-negative patients, HPV-positive tonsillar cancer patients
412 M.J. Kipanyula et al. / Cellular Signalling 25 (2013) 403416

have a HIF-1- overexpression [148]. Furthermore, recent studies Disclosure


have suggested that HCV glycoproteins would perturb tight and
adherens junction protein expression, and increase hepatoma migra- All authors have participated to the bibliographic research, to
tion and expression of epithelial to mesenchymal transition markers manuscript preparation, and have approved the nal version of the
Snail and Twist via stabilizing HIF-1 and genes it regulates. As men- manuscript.
tioned earlier, HIF- regulates various genes involved in tumor Authors declare no competing nancial interest.
growth and metastasis, including VEGF and transforming growth
factor-beta (TGF-), whose neutralization showed different roles for
Acknowledgments
VEGF and TGF- in regulating hepatoma polarity and migration, re-
spectively, indicating a dual role for HIF-1 in the cellular processes
Authors are grateful to their respective institutions for constant
that are deregulated in many human cancers and in the viral life
support and particularly to Dr. Mazzini for his insightful observations.
cycle [149]. Similarly, hypoxia and HIF-1 up-regulate both latent
and lytic Kaposi's sarcoma-associated herpesvirus replication and
may play a central role in the life cycle of this virus [150]. From References
these observations, it appears that HIF-1 expressed during carcino-
genesis is aimed at oncogenic virus maintenance, and carcinogenic [1] M.J. Smolinska, T.H. Page, A.M. Urbaniak, B.E. Mutch, N.J. Horwood, Hck tyrosine
kinase regulates TLR4-induced TNF and IL-6 production via AP-1, Journal of
effects would just be bystander effects. Therefore, targeting HIF-1 Immunology 187 (2011) 60436051.
may alter oncogenic virus maintenance and suppress carcinogenesis. [2] I. Jarchum, M. Liu, C. Shi, M. Equinda, E.G. Pamer, Critical role for
HIF-1alpha is overexpressed in a majority of human cancers [151], MyD88-mediated neutrophil recruitment during Clostridium difcile colitis,
Infection and Immunity 80 (2012) 29892996.
and accordingly, may represent a potent therapeutic agent for cancer. [3] Y. Lin, L.X. Yu, H.X. Yan, W. Yang, L. Tang, H.L. Zhang, Q. Liu, S.S. Zou, Y.Q. He, C.
The natural compound deguelin, which inhibits tumor growth and Wang, M.C. Wu, H.Y. Wang, Gut-derived lipopolysaccharide promotes
blocks tumor angiogenesis in mice, decreases the expression of T-cell-mediated hepatitis in mice through Toll-like receptor 4, Cancer Preven-
tion Research (Philadelphia, Pa.) 5 (2012) 10901102.
HIF-1 and of its target genes, including VEGF, in H1299 non-small [4] C.A. Duckworth, D. Clyde, D.M. Pritchard, CD24 is expressed in gastric parietal
cell lung cancer cells in normoxic and hypoxic conditions [151]. In cells and regulates apoptosis and the response to Helicobacter felis infection in
study addressing the possibility to use the oncolytic reovirus as a the murine stomach, American Journal of Physiology. Gastrointestinal and
Liver Physiology (2012), http://dx.doi.org/10.1152/ajpgi.00068.
cancer therapeutic agent, reovirus infection suppressed HIF-1 levels
[5] O. Rossi, P. van Baarlen, J.M. Wells, Host-recognition of pathogens and commensals
in renal carcinoma and colon cancer cells through a proteosomal in the mammalian intestine, Current Topics in Microbiology and Immunology
activity-dependent mechanism, resulting in cancer cell apoptosis (2011), http://dx.doi.org/10.1007/82_2011_191.
[6] C.J. Karvellas, F. Pink, M. McPhail, M. Austin, G. Auzinger, W. Bernal, E. Sizer, D.J.
[152]. Knockdown of HIF-1 expression constructed by adenovirus-
Kutsogiannis, I. Eltringham, J.A. Wendon, Bacteremia, acute physiology and
mediated RNA interference tools inhibits the invasion and growth of chronic health evaluation II and modied end stage liver disease are indepen-
hepatocellular carcinoma cell lines cultured under hypoxia condi- dent predictors of mortality in critically ill nontransplanted patients with
tions; angiogenesis as well as mobility of vascular cells to tumor acute on chronic liver failure, Critical Care Medicine 38 (2010) 121126.
[7] S.M. Farooq, A.W. Stadnyk, Neutrophil inltration of the colon is independent of
were suppressed as well [153]. In addition, a more recent study the FPR1 yet FPR1 decient mice show differential susceptibilities to acute
revealed that HIF-1 and NF-B targeting induces an overexpression versus chronic induced colitis, Digestive Diseases and Sciences 57 (2012)
of the von Hippel-Lindau protein, which in turn enhances the efcacy 18021812.
[8] C. Narayan, A. Kumar, Constitutive over expression of IL-1beta, IL-6, NF-kappaB,
of the anticancer drug doxorubicin in hepatocellular carcinoma cell and Stat3 is a potential cause of lung tumorgenesis in urethane (ethyl
lines [154]. carbamate) induced Balb/c mice, Journal of Carcinogenesis 11 (2012) 9.
[9] K. Saito, K. Kihara, Role of C-reactive protein in urological cancers: a useful
biomarker for predicting outcomes, International Journal of Urology (2012),
http://dx.doi.org/10.1111/j.1442-2042.2012.03121.x.
7. Concluding remarks [10] F. Balkwill, A. Mantovani, Inammation and cancer: back to Virchow? Lancet
357 (2001) 539545.
[11] V.A. Grote, R. Kaaks, A. Nieters, A. Tjonneland, J. Halkjaer, K. Overvad, M.R.
Pathogen entry in the body classically triggers inammatory re-
Skjelbo Nielsen, M.C. Boutron-Ruault, F. Clavel-Chapelon, A. Racine, B. Teucher,
sponse, which is a defensive mechanism aimed at destroying the S. Becker, T. Pischon, H. Boeing, A. Trichopoulou, C. Cassapa, V. Stratigakou, D.
pathogen itself and eliminating damage/danger-associated mole- Palli, V. Krogh, R. Tumino, P. Vineis, S. Panico, L. Rodriguez, E.J. Duell, M.J.
Sanchez, M. Dorronsoro, C. Navarro, A.B. Gurrea, P.D. Siersema, P.H. Peeters,
cules, to prevent further tissue injury. However, experimental and
W. Ye, M. Sund, B. Lindkvist, D. Johansen, K.T. Khaw, N. Wareham, N.E. Allen,
clinical data indicate that a signicant fraction of cancer cases are R.C. Travis, V. Fedirko, M. Jenab, D.S. Michaud, S.C. Chuang, D. Romaguera, H.B.
caused by infectious agents mediating chronic inammation. Bueno-de-Mesquita, S. Rohrmann, Inammation marker and risk of pancreatic
During chronic inammation immune cells release agents, such as cancer: a nested casecontrol study within the EPIC cohort, British Journal of
Cancer 106 (2012) 18661874.
peroxynitrite, superoxide, and MMIF, which are able to trigger or sus- [12] L. Wang, Y. Jiang, Y. Zhang, Y. Wang, S. Huang, Z. Wang, B. Tian, Y. Yang, W. Jiang,
tain carcinogenic DNA damage induced in host cells by genotoxic D. Pang, Association analysis of IL-17A and IL-17F polymorphisms in Chinese
pathogens. In addition, the high amounts of pro-inammatory fac- Han women with breast cancer, PloS one 7 (2012) e34400.
[13] C. de Martel, J. Ferlay, S. Franceschi, J. Vignat, F. Bray, D. Forman, M. Plummer,
tors, adhesion molecules, and growth factors produced may favor Global burden of cancers attributable to infections in 2008: a review and
neoplastic cell survival. A growing body of evidence has strongly synthetic analysis, The Lancet Oncology 13 (2012) 607615.
linked a spate of inammatory signaling pathways as bridging factors [14] L.V. Lopes, A.V. Conceicao, J.B. Oliveira, A. Tavares, C. Domingos, L.L. Santos,
Cancer in Angola, resources and strategy for its control, The Pan African Medical
between pathogen-triggered inammation and cancer. Inammatory Journal 12 (2012) 13.
signaling pathways such as TLR/MyD88, pro-inammatory cytokine, [15] F. Islami, F. Kamangar, Helicobacter pylori and esophageal cancer risk: a
NF-B, and COX-2 pathways have been implicated in forging the meta-analysis, Cancer Prevention Research (Philadelphia, Pa.) 1 (2008)
329338.
link between inammation and carcinogenesis. Nonetheless, it is
[16] C.M. Nagamine, A.B. Rogers, J.G. Fox, D.B. Schauer, Helicobacter hepaticus pro-
conceivable that these signaling pathways could become likely thera- motes azoxymethane-initiated colon tumorigenesis in BALB/c-IL10-decient
peutic targets to break the growing link between inammation and mice, International Journal of Cancer 122 (2008) 832838.
[17] J.C. Arthur, E. Perez-Chanona, M. Muhlbauer, S. Tomkovich, J.M. Uronis, T.J. Fan, B.J.
different types of cancer. It is thus anticipated that vigorous future
Campbell, T. Abujamel, B. Dogan, A.B. Rogers, J.M. Rhodes, A. Stintzi, K.W. Simpson,
investigations of these pathways and their interactions may provide J.J. Hansen, T.O. Keku, A.A. Fodor, C. Jobin, Intestinal inammation targets cancer-
better understanding of the association between chronic inammation inducing activity of the microbiota, Science 338 (6103) (2012) 120123.
and cancer, particularly in microbiota associated cancers, consider- [18] H. Ozdemir, E. Ciftci, A. Karbuz, G. Oktay, D. Aysev, G. Yavuz, E. Ince, Successful
treatment of multidrug-resistant Escherichia coli bacteremia with tigecycline in
ing that these microorganisms inhabit the body all over the an acute myeloid leukemia child, The Turkish Journal of Pediatrics 54 (2012)
lifetime. 5960.
M.J. Kipanyula et al. / Cellular Signalling 25 (2013) 403416 413

[19] A.M. de Carvalho Parahym, C.M. da Silva, M.P. Leao, M.C. Macario, G.A. Filho, N.T. granulomatosis with polyangiitis, associates with calreticulin on apoptotic
de Oliveira, R.P. Neves, Invasive infection in an acute myeloblastic leukemia neutrophils, impairs macrophage phagocytosis, and promotes inammation,
patient due to triazole-resistant Candida tropicalis, Diagnostic Microbiology Journal of Immunology 189 (2012) 25742583.
and Infectious Disease 71 (2011) 291293. [46] M.P. Protti, M.L. De, Cross-talk within the tumor microenvironment mediates
[20] N. Kurucu, S. Kul, I. Tosun, E. Erduran, I. Koksal, Fungemia and renal fungus ball Th2-type inammation in pancreatic cancer, Oncoimmunology 1 (2012) 8991.
formation with Candida norvegensis in a child with acute lymphoblastic leuke- [47] S. Li, N. Wang, P. Brodt, Metastatic cells can escape the proapoptotic effects of
mia, The Turkish Journal of Pediatrics 53 (2011) 448451. TNF-alpha through increased autocrine IL-6/STAT3 signaling, Cancer Research
[21] P. Marteau, U. Chaput, Bacteria as trigger for chronic gastrointestinal disorders, 72 (2012) 865875.
Digestive Diseases 29 (2011) 166171. [48] L.T. Sorensen, Wound healing and infection in surgery: the pathophysiological
[22] A. Gogia, S. Bakhshi, Plasmablastic lymphoma of oral cavity in a HIV-negative impact of smoking, smoking cessation, and nicotine replacement therapy: a
child, Pediatric Blood & Cancer 55 (2010) 390391. systematic review, Annals of Surgery 255 (2012) 10691079.
[23] M.A. Khan, S. Jakate, S. Komanduri, Rare AIDS-associated plasmablastic lympho- [49] M.C. Maiuri, G. Tajana, T. Iuvone, S.D. De, G. Mele, M.T. Ribecco, M.P. Cinelli, M.F.
ma as the initial presentation of AIDS, Clinical Advances in Hematology and Romano, M.C. Turco, R. Carnuccio, Nuclear factor-kappaB regulates inammato-
Oncology 8 (2010) 5557. ry cell apoptosis and phagocytosis in rat carrageenin-sponge implant model,
[24] D. Compare, G. Nardone, Contribution of gut microbiota to colonic and American Journal of Pathology 165 (2004) 115126.
extracolonic cancer development, Digestive Diseases 29 (2011) 554561. [50] C.K. Joo, Y. Seomun, Matrix metalloproteinase (MMP) and TGF beta 1-stimulated
[25] T. Ohkusa, T. Yoshida, N. Sato, S. Watanabe, H. Tajiri, I. Okayasu, Commensal cell migration in skin and cornea wound healing, Cell Adhesion & Migration 2
bacteria can enter colonic epithelial cells and induce proinammatory cytokine (2008) 252253.
secretion: a possible pathogenic mechanism of ulcerative colitis, Journal of [51] S.V. Ward, G. Cadby, J.S. Heyworth, M.W. Fear, H.J. Wallace, J.M. Cole, F.M. Wood,
Medical Microbiology 58 (2009) 535545. L.J. Palmer, Association of TGFbeta1 and clinical factors with scar outcome fol-
[26] A.E. Sherman, Y. Zavros, Role of sonic hedgehog signaling during progression lowing melanoma excision, Archives for Dermatological Research 304 (2012)
from inammation to cancer in the stomach, World Journal of Gastrointestinal 343351.
Pathophysiology 2 (2011) 103108. [52] K. Ganesh, A. Das, R. Dickerson, S. Khanna, N.L. Parinandi, G.M. Gordillo, C.K. Sen,
[27] L.G. Patwa, T.J. Fan, S. Tchaptchet, Y. Liu, Y.A. Lussier, R.B. Sartor, J.J. Hansen, S. Roy, Prostaglandin E2 induces oncostatin M expression in human chronic
Chronic intestinal inammation induces stress-response genes in commensal wound macrophages through Axl receptor tyrosine kinase pathway, Journal of
Escherichia coli, Gastroenterology 141 (2011) 18421851. Immunology 189 (2012) 25632573.
[28] S. Lofmark, N. de Klerk, H. Aro, Neisseria gonorrhoeae infection induces altered [53] M.S. Bitar, F. Al-Mulla, ROS constitute a convergence nexus in the development
amphiregulin processing and release, PloS one 6 (2011) e16369. of IGF1 resistance and impaired wound healing in a rat model of type 2 diabetes,
[29] Z.H. Li, Q.B. Tang, J. Wang, L. Zhou, W.L. Huang, R.Y. Liu, R.F. Chen, Hepatitis C Disease Models & Mechanisms 5 (2012) 375388.
virus core protein induces malignant transformation of biliary epithelial cells [54] R.T. Lakshmi, T. Priyanka, J. Meenakshi, K.R. Mathangi, V. Jeyaraman, M. Babu,
by activating nuclear factor-kappaB pathway, Journal of Gastroenterology and Low molecular weight heparin mediated regulation of nitric oxide synthase dur-
Hepatology 25 (2010) 13151320. ing burn wound healing, Annals of Burns and Fire Disasters 24 (2011) 2429.
[30] B. Wachtler, F. Citiulo, N. Jablonowski, S. Forster, F. Dalle, M. Schaller, D. Wilson, [55] M. Philip, D.A. Rowley, H. Schreiber, Inammation as a tumor promoter in can-
B. Hube, Candida albicansepithelial interactions: dissecting the roles of active cer induction, Seminars in Cancer Biology 14 (2004) 433439.
penetration, induced endocytosis and host factors on the infection process, [56] M. Macarthur, G.L. Hold, E.M. El-Omar, Inammation and cancer II. Role of
PloS one 7 (2012) e36952. chronic inammation and cytokine gene polymorphisms in the pathogenesis
[31] B. Stadelmann, M.C. Merino, L. Persson, S.G. Svard, Arginine consumption by the of gastrointestinal malignancy, American Journal of Physiology. Gastrointestinal
intestinal parasite Giardia intestinalis reduces proliferation of intestinal epitheli- and Liver Physiology 286 (2004) G515G520.
al cells, PloS one 7 (2012) e45325. [57] N.Y. Tretyakova, S. Burney, B. Pamir, J.S. Wishnok, P.C. Dedon, G.N. Wogan, S.R.
[32] M.J. Chenoweth, M.F. Mian, N.G. Barra, T. Alain, N. Sonenberg, J. Bramson, B.D. Tannenbaum, Peroxynitrite-induced DNA damage in the supF gene: correlation
Lichty, C.D. Richards, A. Ma, A.A. Ashkar, IL-15 can signal via IL-15Ralpha, JNK, with the mutational spectrum, Mutation Research 447 (2000) 287303.
and NF-kappaB to drive RANTES production by myeloid cells, Journal of Immu- [58] J.T. Mutamba, D. Svilar, S. Prasongtanakij, X.H. Wang, Y.C. Lin, P.C. Dedon, R.W.
nology 188 (2012) 41494157. Sobol, B.P. Engelward, XRCC1 and base excision repair balance in response to
[33] Z. Custovic, S. Sosa, Focal bacterial nephritis masquerading as renal cell carcinoma: nitric oxide, DNA Repair 10 (2011) 12821293.
case report, Acta Clinica Croatica 50 (2011) 113114. [59] T. Shimizu, H. Marusawa, Y. Endo, T. Chiba, Inammation-mediated genomic in-
[34] B. Odeh, D. Bareford, Colonic carcinoma presenting as repeated episodes of stability: roles of activation-induced cytidine deaminase in carcinogenesis,
enterobacter septicaemia during induction of remission in acute myeloblastic Cancer Science (2012), http://dx.doi.org/10.1111/j.1349-7006.2012.02293.x.
leukaemia, BMJ 339 (2009) b4027. [60] J.W. Pollard, Tumour-educated macrophages promote tumour progression and
[35] P. Koulocheris, M.C. Metzger, M.R. Kesting, B. Hohlweg-Majert, Life-threatening metastasis, Nature Reviews. Cancer 4 (2004) 7178.
complications associated with acute monocytic leukaemia after dental treat- [61] O. Petrenko, U.M. Moll, Macrophage migration inhibitory factor MIF interferes
ment, Australian Dental Journal 54 (2009) 4548. with the Rb-E2F pathway, Molecular Cell 17 (2005) 225236.
[36] L. Giulino, S. Mathew, G. Ballon, A. Chadburn, S. Barouk, G. Antonicelli, L. [62] B. Gesser, M.K. Rasmussen, L. Raaby, C. Rosada, C. Johansen, R.B. Kjellerup, K.
Leoncini, Y.F. Liu, S. Gogineni, W. Tam, E. Cesarman, A20 (TNFAIP3) genetic Kragballe, L. Iversen, Dimethylfumarate inhibits MIF-induced proliferation of
alterations in EBV-associated AIDS-related lymphoma, Blood 117 (2011) keratinocytes by inhibiting MSK1 and RSK1 activation and by inducing nuclear
48524854. p-c-Jun (S63) and p-p53 (S15) expression, Inammation Research 60 (2011)
[37] L.C. von Hertzen, H. Joensuu, T. Haahtela, Microbial deprivation, inammation 643653.
and cancer, Cancer and Metastasis Reviews 30 (2011) 211223. [63] A.J. McCarron, C. Armstrong, G. Glynn, B.C. Millar, P.J. Rooney, C.E. Goldsmith, J.
[38] E. Ersvaer, G.K. Melve, O. Bruserud, Future perspectives: should Th17 cells be Xu, J.E. Moore, Antibacterial effects on acinetobacter species of commonly
considered as a possible therapeutic target in acute myeloid leukemia patients employed antineoplastic agents used in the treatment of haematological malig-
receiving allogeneic stem cell transplantation? Cancer Immunology, Immuno- nancies: an in vitro laboratory evaluation, British Journal of Biomedical Science
therapy 60 (2011) 16691681. 69 (2012) 1417.
[39] T. Maruyama, K. Kono, Y. Mizukami, Y. Kawaguchi, K. Mimura, M. Watanabe, S. [64] K. Prabhash, A. Medhekar, N. Ghadyalpatil, V. Noronha, S. Biswas, P. Kurkure, R.
Izawa, H. Fujii, Distribution of Th17 cells and FoxP3(+) regulatory T cells in Nair, R. Kelkar, Blood stream infections in cancer patients: a single center expe-
tumor-inltrating lymphocytes, tumor-draining lymph nodes and peripheral rience of isolates and sensitivity pattern, Indian Journal of Cancer 47 (2010)
blood lymphocytes in patients with gastric cancer, Cancer Science 101 (2010) 184188.
19471954. [65] M.C. Chiang, S.C. Kuo, S.J. Chen, S.P. Yang, Y.T. Lee, T.L. Chen, C.P. Fung, Clinical
[40] A. Fialova, S. Partlova, L. Sojka, H. Hromadkova, T. Brtnicky, J. Fucikova, P. Kocian, characteristics and outcomes of bacteremia due to different genomic species
L. Rob, J. Bartunkova, R. Spisek, Dynamics of T-cell inltration during the course of Acinetobacter baumannii complex in patients with solid tumors, Infection 40
of ovarian cancer: the gradual shift from a Th17 effector cell response to a (2012) 1926.
predominant inltration by regulatory T-cells, International Journal of Cancer [66] S. Roperto, G.G. Di, L. Leonardi, U. Pagnini, E. Manco, O. Paciello, I. Esposito, G.
(2012), http://dx.doi.org/10.1002/ijc.27759. Borzacchiello, V. Russo, P. Maiolino, F. Roperto, Bacterial isolates from the
[41] P.F. Seke Etet, M. Palomba, V. Colavito, G. Grassi-Zucconi, M. Bentivoglio, G. Bertini, urine of cattle affected by urothelial tumors of the urinary bladder, Research
Sleep and rhythm changes at the time of Trypanosoma brucei invasion of the brain in Veterinary Science 93 (3) (2012) 13611366.
parenchyma in the rat, Chronobiology International 29 (2012) 469481. [67] F.A. Carvalho, J.D. Aitken, M. Vijay-Kumar, A.T. Gewirtz, Toll-like receptorgut
[42] D.N. Amin, S.K. Vodnala, W. Masocha, B. Sun, K. Kristensson, M.E. Rottenberg, microbiota interactions: perturb at your own risk! Annual Review of Physiology
Distinct Toll-like receptor signals regulate cerebral parasite load and interferon 74 (2012) 177198.
alpha/beta and tumor necrosis factor alpha-dependent T-cell inltration in the [68] J. Kinross, A.C. von Roon, N. Penney, E. Holmes, D. Silk, J.K. Nicholson, A. Darzi,
brains of Trypanosoma brucei-infected mice, Journal of Infectious Diseases 205 The gut microbiota as a target for improved surgical outcome and improved
(2012) 320332. patient care, Current Pharmaceutical Design 15 (2009) 15371545.
[43] L. Romani, P. Puccetti, Controlling pathogenic inammation to fungi, Expert [69] C. Ubeda, Y. Taur, R.R. Jenq, M.J. Equinda, T. Son, M. Samstein, A. Viale, N.D. Socci,
Review of Anti-Infective Therapy 5 (2007) 10071017. M.R. van den Brink, M. Kamboj, E.G. Pamer, Vancomycin-resistant Enterococcus
[44] M.L. Huynh, V.A. Fadok, P.M. Henson, Phosphatidylserine-dependent ingestion domination of intestinal microbiota is enabled by antibiotic treatment in mice
of apoptotic cells promotes TGF-beta1 secretion and the resolution of inamma- and precedes bloodstream invasion in humans, The Journal of Clinical Investiga-
tion, The Journal of Clinical Investigation 109 (2002) 4150. tion 120 (2010) 43324341.
[45] J. Gabillet, A. Millet, M. Pederzoli-Ribeil, P. Tacnet-Delorme, L. Guillevin, L. [70] R. Merline, K. Moreth, J. Beckmann, M.V. Nastase, J. Zeng-Brouwers, J.G. Tralhao,
Mouthon, P. Frachet, V. Witko-Sarsat, Proteinase 3, the autoantigen in P. Lemarchand, J. Pfeilschifter, R.M. Schaefer, R.V. Iozzo, L. Schaefer, Signaling by
414 M.J. Kipanyula et al. / Cellular Signalling 25 (2013) 403416

the matrix proteoglycan decorin controls inammation and cancer through [96] S.K. Polumuri, G.G. Jayakar, K.A. Shirey, Z.J. Roberts, D.J. Perkins, P.M. Pitha, S.N.
PDCD4 and MicroRNA-21, Science Signaling 4 (2011) ra75. Vogel, Transcriptional regulation of murine IL-33 by TLR and non-TLR agonists,
[71] K. Moreth, R.V. Iozzo, L. Schaefer, Small leucine-rich proteoglycans orchestrate Journal of Immunology 189 (2012) 5060.
receptor crosstalk during inammation, Cell Cycle 11 (2012) 20842091. [97] A. Banerjee, S. Gerondakis, Coordinating TLR-activated signaling pathways in
[72] Q. Gao, Y.J. Zhao, X.Y. Wang, S.J. Qiu, Y.H. Shi, J. Sun, Y. Yi, J.Y. Shi, G.M. Shi, Z.B. cells of the immune system, Immunology and Cell Biology 85 (2007) 420424.
Ding, Y.S. Xiao, Z.H. Zhao, J. Zhou, X.H. He, J. Fan, CXCR6 upregulation contributes [98] K. Newton, V.M. Dixit, Signaling in innate immunity and inammation, Cold
to a proinammatory tumor microenvironment that drives metastasis and poor Spring Harbor Perspectives in Biology 4 (2012).
patient outcomes in hepatocellular carcinoma, Cancer Research 72 (2012) [99] H. Shime, M. Matsumoto, H. Oshiumi, S. Tanaka, A. Nakane, Y. Iwakura, H.
35463556. Tahara, N. Inoue, T. Seya, Toll-like receptor 3 signaling converts tumor-
[73] A. Ochi, A.H. Nguyen, A.S. Bedrosian, H.M. Mushlin, S. Zarbakhsh, R. Barilla, C.P. supporting myeloid cells to tumoricidal effectors, Proceedings of the National
Zambirinis, N.C. Fallon, A. Rehman, Y. Pylayeva-Gupta, S. Badar, C.H. Hajdu, Academy of Sciences of the United States of America 109 (2012) 20662071.
A.B. Frey, D. Bar-Sagi, G. Miller, MyD88 inhibition amplies dendritic cell capac- [100] L.M. Kingeter, X. Lin, C-type lectin receptor-induced NF-kappaB activation in in-
ity to promote pancreatic carcinogenesis via Th2 cells, The Journal of Experi- nate immune and inammatory responses, Cellular & Molecular Immunology 9
mental Medicine (2012) 16711687. (2012) 105112.
[74] J.M. Uronis, M. Muhlbauer, H.H. Herfarth, T.C. Rubinas, G.S. Jones, C. Jobin, Mod- [101] H. Matta, R. Gopalakrishnan, V. Punj, H. Yi, Y. Suo, P.M. Chaudhary, A20 is in-
ulation of the intestinal microbiota alters colitis-associated colorectal cancer duced by Kaposi sarcoma-associated herpesvirus-encoded viral FLICE inhibitory
susceptibility, PloS one 4 (2009) e6026. protein (vFLIP) K13 and blocks K13-induced nuclear factor-kappaB in a negative
[75] M.A. Azcarate-Peril, M. Sikes, J.M. Bruno-Barcena, The intestinal microbiota, feedback manner, Journal of Biological Chemistry 286 (2011) 2155521564.
gastrointestinal environment and colorectal cancer: a putative role for [102] S. Uota, D.M. Zahidunnabi, Y. Saitoh, S. Muto, A. Itai, A. Utsunomiya, T. Watanabe,
probiotics in prevention of colorectal cancer? American Journal of Physiology. N. Yamamoto, S. Yamaoka, An IkappaB kinase 2 inhibitor IMD-0354 suppresses
Gastrointestinal and Liver Physiology 301 (2011) G401G424. the survival of adult T-cell leukemia cells, Cancer Science 103 (2012) 100106.
[76] A.B. Rogers, Distance burning: how gut microbes promote extraintestinal [103] M.Z. Dewan, M. Tomita, H. Katano, N. Yamamoto, S. Ahmed, M. Yamamoto, T.
cancers, Gut Microbes 2 (2011) 5257. Sata, N. Mori, N. Yamamoto, An HIV protease inhibitor, ritonavir targets the
[77] A.K. Shenoy, R.C. Fisher, E.A. Butterworth, L. Pi, L.J. Chang, H.D. Appelman, M. nuclear factor-kappaB and inhibits the tumor growth and inltration of
Chang, E.W. Scott, E.H. Huang, Transition from colitis to cancer: high Wnt activity EBV-positive lymphoblastoid B cells, International Journal of Cancer 124
sustains the tumor-initiating potential of colon cancer stem cell precursors, Cancer (2009) 622629.
Research 72 (19) (2012) 50915100. [104] Q. Tang, D. Qin, Z. Lv, X. Zhu, X. Ma, Q. Yan, Y. Zeng, Y. Guo, N. Feng, C. Lu, Herpes
[78] D. Subramaniam, S. Ramalingam, C.W. Houchen, S. Anant, Cancer stem cells: a simplex virus type 2 triggers reactivation of Kaposi's sarcoma-associated her-
novel paradigm for cancer prevention and treatment, Mini Reviews in Medicinal pesvirus from latency and collaborates with HIV-1 Tat, PloS one 7 (2012)
Chemistry 10 (2010) 359371. e31652.
[79] Z.F. Zimmerman, R.T. Moon, A.J. Chien, Targeting Wnt pathways in disease. [105] F. Collina, C.A. De, R.A. De, R.G. De, G. Botti, R. Franco, Chlamydia psittaci in ocu-
Cold Spring Harb, Perspectives in Biology (2012), http://dx.doi.org/10.1101/ lar adnexa MALT lymphoma: a possible role in lymphomagenesis and a different
cshperspect.a008086. geographical distribution, Infectious Agents and Cancer 7 (2012) 8.
[80] P.F. Seke Etet, L. Vecchio, A.H. Nwabo Kamdje, Interactions between bone mar- [106] E. Pikarsky, R.M. Porat, I. Stein, R. Abramovitch, S. Amit, S. Kasem, E.
row stromal microenvironment and B-chronic lymphocytic leukemia cells: any Gutkovich-Pyest, S. Urieli-Shoval, E. Galun, Y. Ben-Neriah, NF-kappaB functions
role for Notch, Wnt and Hh signaling pathways? Cellular Signalling 24 (2012) as a tumour promoter in inammation-associated cancer, Nature 431 (2004)
14331443. 461466.
[81] P.F. Seke Etet, L. Vecchio, K.P. Bogne, N.E. Nchiwan, M. Krampera, A.H. Nwabo [107] F.R. Greten, L. Eckmann, T.F. Greten, J.M. Park, Z.W. Li, L.J. Egan, M.F. Kagnoff, M.
Kamdje, Normal hematopoiesis and hematologic malignancies: role of canonical Karin, IKKbeta links inammation and tumorigenesis in a mouse model of
Wnt signaling pathway and stromal microenvironment, Biochimica et colitis-associated cancer, Cell 118 (2004) 285296.
Biophysica Acta (2012), http://dx.doi.org/10.1016/j.bbcan.2012.08.002. [108] D. Preciado, E. Caicedo, R. Jhanjee, R. Silver, G. Harris, S.K. Juhn, D.I. Choo, F.
[82] Y. Li, P. Kundu, S.W. Seow, C.T. de Matos, L. Aronsson, K.C. Chin, K. Karre, S. Ondrey, Pseudomonas aeruginosa lipopolysaccharide induction of keratinocyte
Pettersson, G. Greicius, Gut microbiota accelerate tumor growth via c-jun proliferation, NF-kappa B, and cyclin D1 is inhibited by indomethacin, Journal
and STAT3 phosphorylation in APCMin/+ mice, Carcinogenesis 33 (2012) of Immunology 174 (2005) 29642973.
12311238. [109] M. Karin, Y. Cao, F.R. Greten, Z.W. Li, NF-kappaB in cancer: from innocent by-
[83] H. Naglova, M. Bucova, HMGB1 and its physiological and pathological roles, stander to major culprit, Nature Reviews. Cancer 2 (2002) 301310.
Bratislavsk Lekrske Listy 113 (2012) 163171. [110] J.L. Luo, S. Maeda, L.C. Hsu, H. Yagita, M. Karin, Inhibition of NF-kappaB in cancer
[84] Y.Z. Wang, M. Yan, F.F. Tian, J.M. Zhang, Q. Liu, H. Yang, W.B. Zhou, J. Li, Possible cells converts inammation- induced tumor growth mediated by TNFalpha to
involvement of Toll-like receptors in the pathogenesis of Myasthenia gravis, In- TRAIL-mediated tumor regression, Cancer Cell 6 (2004) 297305.
ammation (2012), http://dx.doi.org/10.1007/s10753-012-9526-6. [111] F. Yan, D.B. Polk, Disruption of NF-kappaB signalling by ancient microbial molecules:
[85] L.W. Belinda, W.X. Wei, B.T. Hanh, L.X. Lei, H. Bow, D.J. Ling, SARM: a novel novel therapies of the future? Gut 59 (2010) 421426.
Toll-like receptor adaptor, is functionally conserved from arthropod to human, [112] C. Cataisson, R. Salcedo, S. Hakim, B.A. Moftt, L. Wright, M. Yi, R. Stephens, R.M.
Molecular Immunology 45 (2008) 17321742. Dai, L. Lyakh, D. Schenten, H.S. Yuspa, G. Trinchieri, IL-1R-MyD88 signaling in
[86] E. Valkov, A. Stamp, F. Dimaio, D. Baker, B. Verstak, P. Roversi, S. Kellie, M.J. keratinocyte transformation and carcinogenesis, The Journal of Experimental
Sweet, A. Mansell, N.J. Gay, J.L. Martin, B. Kobe, Crystal structure of Toll-like re- Medicine 209 (9) (2012) 16891702.
ceptor adaptor MAL/TIRAP reveals the molecular basis for signal transduction [113] G. Barbisan, L.O. Perez, A. Contreras, C.D. Golijow, TNF-alpha and IL-10 promoter
and disease protection, Proceedings of the National Academy of Sciences of polymorphisms, HPV infection, and cervical cancer risk, Tumour Biology 33 (5)
the United States of America 108 (2011) 1487914884. (2012) 15491556.
[87] S.C. Lin, Y.C. Lo, H. Wu, Helical assembly in the MyD88-IRAK4-IRAK2 complex in [114] J.C. Santos, M.S. Ladeira, J. Pedrazzoli Jr., M.L. Ribeiro, Relationship of IL-1 and
TLR/IL-1R signalling, Nature 465 (2010) 885890. TNF-alpha polymorphisms with Helicobacter pylori in gastric diseases in a Brazilian
[88] W. Piao, C. Song, H. Chen, M.A. Diaz, L.M. Wahl, K.A. Fitzgerald, L. Li, A.E. population, Brazilian Journal of Medical and Biological Research 45 (2012) 811817.
Medvedev, Endotoxin tolerance dysregulates MyD88- and Toll/IL-1R [115] K. Sakitani, Y. Hirata, Y. Hayakawa, T. Serizawa, W. Nakata, R. Takahashi, H.
domain-containing adapter inducing IFN-beta-dependent pathways and in- Kinoshita, K. Sakamoto, H. Nakagawa, M. Akanuma, H. Yoshida, S. Maeda, K.
creases expression of negative regulators of TLR signaling, Journal of Leukocyte Koike, The role of Interleukin-32 in Helicobacter pylori-induced gastric
Biology 86 (2009) 863875. inammation, Infection and Immunity 80 (11) (2012) 37953803.
[89] P.H. Tseng, A. Matsuzawa, W. Zhang, T. Mino, D.A. Vignali, M. Karin, Different modes [116] A.R. Moschen, T. Fritz, A.D. Clouston, I. Rebhan, O. Bauhofer, H.D. Barrie, E.E.
of ubiquitination of the adaptor TRAF3 selectively activate the expression of type I Powell, S.H. Kim, C.A. Dinarello, R. Bartenschlager, J.R. Jonsson, H. Tilg, Interleu-
interferons and proinammatory cytokines, Nature Immunology 11 (2010) 7075. kin-32: a new proinammatory cytokine involved in hepatitis C virus-related
[90] E.W. Harhaj, V.M. Dixit, Regulation of NF-kappaB by deubiquitinases, Immuno- liver inammation and brosis, Hepatology 53 (2011) 18191829.
logical Reviews 246 (2012) 107124. [117] H.J. Lee, Z.L. Liang, S.M. Huang, J.S. Lim, D.Y. Yoon, H.J. Lee, J.M. Kim,
[91] K. Clark, M. Peggie, L. Plater, R.J. Sorcek, E.R. Young, J.B. Madwed, J. Hough, E.G. Overexpression of IL-32 is a novel prognostic factor in patients with localized
McIver, P. Cohen, Novel cross-talk within the IKK family controls innate immu- clear cell renal cell carcinoma, Oncology Letters 3 (2012) 490496.
nity, Biochemical Journal 434 (2011) 93104. [118] Y. Yu, R. Gong, Y. Mu, Y. Chen, C. Zhu, Z. Sun, M. Chen, Y. Liu, Y. Zhu, J. Wu,
[92] H. Matta, R. Gopalakrishnan, C. Graham, B. Tolani, A. Khanna, H. Yi, Y. Suo, P.M. Hepatitis B virus induces a novel inammation network involving three inam-
Chaudhary, Kaposi's sarcoma associated herpesvirus encoded viral FLICE inhib- matory factors, IL-29, IL-8, and cyclooxygenase-2, Journal of Immunology 187
itory protein K13 activates NF-kappaB pathway independent of TRAF6, TAK1 (2011) 48444860.
and LUBAC, PloS one 7 (2012) e36601. [119] A. Kargi, A.D. Yalcin, N. Erin, B. Savas, H.H. Polat, R.M. Gorczynski, IL8 and
[93] M. Guma, D. Hammaker, K. Topolewski, M. Corr, D.L. Boyle, M. Karin, G.S. serum soluble TRAIL levels following anti-VEGF monoclonal antibody treatment
Firestein, Pro- and anti-inammatory functions of the p38 pathway in rheuma- in patients with metastatic colon cancer, Clinical Laboratory 58 (2012)
toid arthritis: advantages of targeting upstream kinases MKK3 or MKK6, 501505.
Arthritis and Rheumatism (2012), http://dx.doi.org/10.1002/art.34489. [120] X. Liu, H. Fang, H. Chen, X. Jiang, D. Fang, Y. Wang, D. Zhu, An articial miRNA
[94] M. Das, G. Sabio, F. Jiang, M. Rincon, R.A. Flavell, R.J. Davis, Induction of hepatitis against HPSE suppresses melanoma invasion properties, correlating with a
by JNK-mediated expression of TNF-alpha, Cell 136 (2009) 249260. down-regulation of chemokines and MAPK phosphorylation, PloS one 7
[95] M. Mellett, P. Atzei, R. Jackson, L.A. O'Neill, P.N. Moynagh, Mal mediates (2012) e38659.
TLR-induced activation of CREB and expression of IL-10, Journal of Immunology [121] H. Chen, Y. Sun, C. Wu, C.E. Magyar, X. Li, L. Cheng, J.L. Yao, S. Shen, A.O.
186 (2011) 49254935. Osunkoya, C. Liang, J. Huang, Pathogenesis of prostatic small cell carcinoma
M.J. Kipanyula et al. / Cellular Signalling 25 (2013) 403416 415

involves the inactivation of the P53 pathway, Endocrine-Related Cancer 19 inammatory disorders by activation of HIF-dependent responses, Gut Microbes 2
(2012) 321331. (2011) 335346.
[122] M.M. Shahzad, J.M. Arevalo, G.N. Armaiz-Pena, C. Lu, R.L. Stone, M. [144] S. Jo, A. Juhasz, K. Zhang, C. Ruel, S. Loera, S.P. Wilczynski, Y. Yen, X. Liu, J.
Moreno-Smith, M. Nishimura, J.W. Lee, N.B. Jennings, J. Bottsford-Miller, P. Ellenhorn, D. Lim, B. Paz, G. Somlo, N. Vora, S. Shibata, Human papillomavirus in-
Vivas-Mejia, S.K. Lutgendorf, G. Lopez-Berestein, M. Bar-Eli, S.W. Cole, A.K. fection as a prognostic factor in oropharyngeal squamous cell carcinomas treat-
Sood, Stress effects on FosB- and interleukin-8 (IL8)-driven ovarian cancer ed in a prospective phase II clinical trial, Anticancer Research 29 (2009)
growth and metastasis, Journal of Biological Chemistry 285 (2010) 14671474.
3546235470. [145] G. Li, L. He, E. Zhang, J. Shi, Q. Zhang, A.D. Le, K. Zhou, X. Tang, Overexpression of
[123] S. Li, S.E. Kendall, R. Raices, J.B. Finlay, M. Covarrubias, Z. Liu, G. Lowe, Y.H. Lin, human papillomavirus (HPV) type 16 oncoproteins promotes angiogenesis via
Y.H. Teh, V. Leigh, S. Dhillon, S. Flanagan, K.S. Aboody, C.A. Glackin, TWIST1 enhancing HIF-1alpha and VEGF expression in non-small cell lung cancer cells,
associates with NF-kappaB subunit RELA via carboxyl-terminal WR domain to Cancer Letters 311 (2011) 160170.
promote cell autonomous invasion through IL8 production, BMC Biology 10 [146] J.M. Bodily, K.P. Mehta, L.A. Laimins, Human papillomavirus E7 enhances
(2012) 73. hypoxia-inducible factor 1-mediated transcription by inhibiting binding of his-
[124] C.J. Buskens, B.P. Van Rees, A. Sivula, J.B. Reitsma, C. Haglund, P.J. Bosma, G.J. tone deacetylases, Cancer Research 71 (2011) 11871195.
Offerhaus, J.J. Van Lanschot, A. Ristimaki, Prognostic signicance of elevated cy- [147] M. Nasimuzzaman, G. Waris, D. Mikolon, D.G. Stupack, A. Siddiqui, Hepatitis C
clooxygenase 2 expression in patients with adenocarcinoma of the esophagus, virus stabilizes hypoxia-inducible factor 1alpha and stimulates the synthesis of
Gastroenterology 122 (2002) 18001807. vascular endothelial growth factor, Journal of Virology 81 (2007) 1024910257.
[125] C.A. Martey, S.J. Pollock, C.K. Turner, K.M. O'Reilly, C.J. Baglole, R.P. Phipps, P.J. [148] S.H. Kim, B.S. Koo, S. Kang, K. Park, H. Kim, K.R. Lee, M.J. Lee, J.M. Kim, E.C. Choi,
Sime, Cigarette smoke induces cyclooxygenase-2 and microsomal prostaglandin N.H. Cho, HPV integration begins in the tonsillar crypt and leads to the alteration
E2 synthase in human lung broblasts: implications for lung inammation and of p16, EGFR and c-myc during tumor formation, International Journal of Cancer
cancer, American Journal of Physiology. Lung Cellular and Molecular Physiology 120 (2007) 14181425.
287 (2004) L981L991. [149] G.K. Wilson, C.L. Brimacombe, I.A. Rowe, G.M. Reynolds, N.F. Fletcher, Z.
[126] K. Oyama, T. Fujimura, I. Ninomiya, T. Miyashita, S. Kinami, S. Fushida, T. Ohta, M. Stamataki, R.H. Bhogal, M.L. Simoes, M. Ashcroft, S.C. Afford, R.R. Mitry, A.
Koichi, A COX-2 inhibitor prevents the esophageal inammation-metaplasia- Dhawan, C.J. Mee, S.G. Hubscher, P. Balfe, J.A. McKeating, A dual role for hypoxia
adenocarcinoma sequence in rats, Carcinogenesis 26 (2005) 565570. inducible factor-1alpha in the hepatitis C virus lifecycle and hepatoma migra-
[127] R.D. Klein, C.S. Van Pelt, A.L. Sabichi, C.J. Dela, S.M. Fischer, G. Furstenberger, K. tion, Journal of Hepatology 56 (2012) 803809.
Muller-Decker, Transitional cell hyperplasia and carcinomas in urinary bladders [150] R.P. Veeranna, M. Haque, D.A. Davis, M. Yang, R. Yarchoan, Kaposi's
of transgenic mice with keratin 5 promoter-driven cyclooxygenase-2 sarcoma-associated herpesvirus latency-associated nuclear antigen induction
overexpression, Cancer Research 65 (2005) 18081813. by hypoxia and hypoxia-inducible factors, Journal of Virology 86 (2012)
[128] S.I. Abdalla, I.R. Sanderson, R.C. Fitzgerald, Effect of inammation on cyclooxy- 10971108.
genase (COX)-2 expression in benign and malignant oesophageal cells, Carcino- [151] S.H. Oh, J.K. Woo, Q. Jin, H.J. Kang, J.W. Jeong, K.W. Kim, W.K. Hong, H.Y. Lee,
genesis 26 (2005) 16271633. Identication of novel antiangiogenic anticancer activities of deguelin targeting
[129] R.N. DuBois, Leukotriene A4 signaling, inammation, and cancer, Journal of the hypoxia-inducible factor-1 alpha, International Journal of Cancer 122 (2008)
National Cancer Institute 95 (2003) 10281029. 514.
[130] F. De Marco, E. Bucaj, C. Foppoli, A. Fiorini, C. Blarzino, K. Filipi, A. Giorgi, M.E. [152] I.R. Cho, S.S. Koh, H.J. Min, E.H. Park, S. Ratakorn, B.H. Jhun, S.H. Jeong, Y.H. Yoo,
Schinina, D.F. Di, R. Coccia, D.A. Buttereld, M. Perluigi, Oxidative stress in H.D. Youn, R.N. Johnston, Y.H. Chung, Down-regulation of HIF-1alpha by
HPV-driven viral carcinogenesis: redox proteomics analysis of HPV-16 dysplas- oncolytic reovirus infection independently of VHL and p53, Cancer Gene
tic and neoplastic tissues, PloS one 7 (2012) e34366. Therapy 17 (2010) 365372.
[131] S.P. Hussain, G.E. Trivers, L.J. Hofseth, P. He, I. Shaikh, L.E. Mechanic, S. Doja, W. [153] S.H. Choi, H.W. Shin, J.Y. Park, J.Y. Yoo, Y. Kim do, W.S. Ro, C.O. Yun, K.H. Han, Effects
Jiang, J. Subleski, L. Shorts, D. Haines, V.E. Laubach, R.H. Wiltrout, D. Djurickovic, of the knockdown of hypoxia inducible factor-1alpha expression by adenovirus-
C.C. Harris, Nitric oxide, a mediator of inammation, suppresses tumorigenesis, mediated shRNA on angiogenesis and tumor growth in hepatocellular carcinoma
Cancer Research 64 (2004) 68496853. cell lines, The Korean Journal of Hepatology 16 (2010) 280287.
[132] Y.H. Kim, K.J. Woo, J.H. Lim, S. Kim, T.J. Lee, E.M. Jung, J.M. Lee, J.W. Park, T.K. [154] J. Wang, Y. Ma, H. Jiang, H. Zhu, L. Liu, B. Sun, S. Pan, G.W. Krissansen, X. Sun,
Kwon, 8-Hydroxyquinoline inhibits iNOS expression and nitric oxide production Overexpression of von HippelLindau protein synergizes with doxorubicin to
by down-regulating LPS-induced activity of NF-kappaB and C/EBPbeta in Raw suppress hepatocellular carcinoma in mice, Journal of Hepatology 55 (2011)
264.7 cells, Biochemical and Biophysical Research Communications 329 (2005) 359368.
591597. [155] T. Wang, G. Cai, Y. Qiu, N. Fei, M. Zhang, X. Pang, W. Jia, S. Cai, L. Zhao, Structural
[133] P. Yongvanit, S. Pinlaor, H. Bartsch, Oxidative and nitrative DNA damage: key segregation of gut microbiota between colorectal cancer patients and healthy
events in opisthorchiasis-induced carcinogenesis, Parasitology International 61 volunteers, ISME Journal 6 (2012) 320329.
(2012) 130135. [156] Y. Yoshino, T. Kitazawa, M. Ikeda, K. Tatsuno, S. Yanagimoto, S. Okugawa, Y. Ota,
[134] N. Ma, R. Thanan, H. Kobayashi, O. Hammam, M. Wishahi, T. El Leithy, Y. Hiraku, H. Yotsuyanagi, Clinical features of Bacteroides bacteremia and their association
E. Amro, S. Oikawa, S. Ohnishi, M. Murata, S. Kawanishi, Nitrative DNA damage with colorectal carcinoma, Infection 40 (2012) 6367.
and Oct3/4 expression in urinary bladder cancer with Schistosoma haematobium [157] M. Ortega, F. Marco, A. Soriano, M. Almela, J.A. Martinez, J. Lopez, C. Pitart, J.
infection, Biochemical and Biophysical Research Communications 414 (2011) Mensa, Epidemiology and prognostic determinants of bacteraemic biliary tract
344349. infection, Journal of Antimicrobial Chemotherapy 67 (2012) 15081513.
[135] R. Chaturvedi, S.T. de, L.A. Coburn, A.P. Gobert, K.T. Wilson, Arginine and poly- [158] M. Kato, M. Kaise, T. Obata, J. Yonezawa, H. Toyoizumi, N. Yoshimura, Y. Yoshida,
amines in Helicobacter pylori-induced immune dysregulation and gastric carci- M. Kawamura, H. Tajiri, Bacteremia and endotoxemia after endoscopic submu-
nogenesis, Amino Acids 42 (2012) 627640. cosal dissection for gastric neoplasia: pilot study, Gastric Cancer 15 (2012)
[136] C.I. Kang, D.R. Chung, K.S. Ko, K.R. Peck, J.H. Song, Clinical predictors of 1520.
Enterobacter bacteremia among patients admitted to the ED, The American Jour- [159] S. Gehmert, S. Gehmert, X. Bai, S. Klein, O. Ortmann, L. Prantl, Limitation of in
nal of Emergency Medicine 30 (2012) 165169. vivo models investigating angiogenesis in breast cancer, Clinical Hemorheology
[137] D. Conde-Estevez, S. Grau, J. Albanell, R. Terradas, M. Salvado, H. Knobel, Clinical and Microcirculation 49 (2011) 519526.
characteristics and outcomes of patients with vancomycin-susceptible [160] M.E. Santolaya, M.J. Farfan, L.M. De, V.M. Cocina, F. Santelices, A.M. Alvarez, C.L.
Enterococcus faecalis and Enterococcus faecium bacteraemia in cancer patients, Aviles, A. Becker, M. O'Ryan, P. Roman, C. Salgado, P. Silva, S. Topelberg, J.
European Journal of Clinical Microbiology and Infectious Diseases 30 (2011) Tordecilla, M. Varas, M. Villarroel, T. Viviani, M. Zubieta, J.P. Torres, Diagnosis of
103108. bacteremia in febrile neutropenic episodes in children with cancer: microbiologic
[138] X. Wang, Y. Yang, D.R. Moore, S.L. Nimmo, S.A. Lightfoot, M.M. Huycke, and molecular approach, Pediatric Infectious Disease Journal 30 (2011) 957961.
4-hydroxy-2-nonenal mediates genotoxicity and bystander effects caused by [161] C.C. Huang, C.J. Wu, L.R. Wang, H.C. Lee, C.M. Chang, N.Y. Lee, T.Y. Chen, W.C. Ko,
Enterococcus faecalis-infected macrophages, Gastroenterology 142 (2012) Antimicrobial susceptibility of bacteremic isolates from cancer patients with or
543551. without neutropenia at a medical center in southern Taiwan, Journal of Microbi-
[139] Q. Ma, L.E. Cavallin, B. Yan, S. Zhu, E.M. Duran, H. Wang, L.P. Hale, C. Dong, E. ology, Immunology and Infection 44 (2011) 376381.
Cesarman, E.A. Mesri, P.J. Goldschmidt-Clermont, Antitumorigenesis of antioxi- [162] E.M. Trecarichi, M. Tumbarello, M. Caira, A. Candoni, C. Cattaneo, D. Pastore, R.
dants in a transgenic Rac1 model of Kaposi's sarcoma, Proceedings of the National Fanci, A. Nosari, N. Vianelli, A. Busca, A. Spadea, L. Pagano, Multidrug resistant
Academy of Sciences of the United States of America 106 (2009) 86838688. Pseudomonas aeruginosa bloodstream infection in adult patients with hemato-
[140] Q. Ke, M. Costa, Hypoxia-inducible factor-1 (HIF-1), Molecular Pharmacology 70 logic malignancies, Haematologica 96 (2011) e1e3.
(2006) 14691480. [163] K. Katsibardi, V. Papadakis, A. Charisiadou, A. Pangalis, S. Polychronopoulou,
[141] T. Hara, K. Mimura, T. Abe, G. Shioi, M. Seiki, T. Sakamoto, Deletion of the Blood stream infections throught the entire course of acute lymphoblastic
Mint3/Apba3 gene in mice abrogates macrophage functions and increases leukemia treatment, Neoplasma 58 (2011) 326330.
resistance to lipopolysaccharide-induced septic shock, Journal of Biological [164] R. Jeddi, H. Ghedira, A.R. Ben, A. Turki, K. Kacem, A.Y. Ben, L.R. Ben, L. Aissaoui,
Chemistry 286 (2011) 3254232551. A.H. Ben, H.Z. Bel, B. Meddeb, Risk factors of septic shock in patients with hema-
[142] K.J. Staples, F. Sotoodehnejadnematalahi, H. Pearson, M. Frankenberger, L. tologic malignancies and Pseudomonas infections, Hematology 16 (2011)
Francescut, L. Ziegler-Heitbrock, B. Burke, Monocyte-derived macrophages ma- 160165.
tured under prolonged hypoxia transcriptionally up-regulate HIF-1alpha [165] T. Horino, A. Chiba, S. Kawano, T. Kato, F. Sato, Y. Maruyama, Y. Nakazawa, K.
mRNA, Immunobiology 216 (2011) 832839. Yoshikawa, M. Yoshida, S. Hori, Clinical characteristics and risk factors for mor-
[143] S. Mimouna, D. Goncalves, N. Barnich, A. Darfeuille-Michaud, P. Hofman, V. tality in patients with bacteremia caused by Pseudomonas aeruginosa, Internal
Vouret-Craviari, Crohn disease-associated Escherichia coli promote gastrointestinal Medicine 51 (2012) 5964.
416 M.J. Kipanyula et al. / Cellular Signalling 25 (2013) 403416

[166] L. Soroceanu, L. Matlaf, V. Bezrookove, L. Harkins, R. Martinez, M. Greene, P. in diseased prostates and its inammatory and transforming activity on prostate
Soteropoulos, C.S. Cobbs, Human cytomegalovirus US28 found in glioblastoma epithelial cells, International Journal of Medical Microbiology 301 (2011) 6978.
promotes an invasive and angiogenic phenotype, Cancer Research 71 (2011) [174] V. Sulzyc-Bielicka, L. Kolodziejczyk, S. Jaczewska, D. Bielicki, J. Kladny, K.
66436653. Safranow, Prevalence of Cryptosporidium sp. in patients with colorectal cancer,
[167] K.S. Sfanos, J. Sauvageot, H.L. Fedor, J.D. Dick, A.M. De Marzo, W.B. Isaacs, A Polski Przeglad Chirurgiczny 84 (2012) 348351.
molecular analysis of prokaryotic and viral DNA sequences in prostate tissue [175] S. Benamrouz, V. Conseil, C. Creusy, E. Calderon, E. Dei-Cas, G. Certad, Parasites
from patients with prostate cancer indicates the presence of multiple and and malignancies, a review, with emphasis on digestive cancer induced
diverse microorganisms, Prostate 68 (2008) 306320. by Cryptosporidium parvum (Alveolata: Apicomplexa), Parasite 19 (2012)
[168] N. Nishida, A. Goel, Genetic and epigenetic signatures in human hepatocellular 101115.
carcinoma: a systematic review, Current Genomics 12 (2011) 130137. [176] A. Chene, D. Donati, A.O. Guerreiro-Cacais, V. Levitsky, Q. Chen, K.I. Falk, J. Orem,
[169] C.J. Lay, H.J. Zhuang, Y.H. Ho, Y.S. Tsai, L.S. Wang, C.C. Tsai, Different clinical F. Kironde, M. Wahlgren, M.T. Bejarano, A molecular link between malaria and
characteristics between polymicrobial and monomicrobial Aeromonas bacter- EpsteinBarr virus reactivation, PLoS Pathogens 3 (2007) e80.
emiaa study of 216 cases, Internal Medicine 49 (2010) 24152421. [177] A. Chene, D. Donati, J. Orem, E.R. Mbidde, F. Kironde, M. Wahlgren, M.T.
[170] M.S. Tsai, C.Y. Kuo, M.C. Wang, H.C. Wu, C.C. Chien, J.W. Liu, Clinical features and Bejarano, Endemic Burkitt's lymphoma as a polymicrobial disease: new insights
risk factors for mortality in Aeromonas bacteremic adults with hematologic on the interaction between Plasmodium falciparum and EpsteinBarr virus, Sem-
malignancies, Journal of Microbiology, Immunology and Infection 39 (2006) inars in Cancer Biology 19 (2009) 411420.
150154. [178] A. Tragiannidis, W. Fegeler, G. Rellensmann, V. Debus, V. Muller, I. Hoernig-Franz,
[171] V. Papadakis, N. Poniros, K. Katsibardi, A.E. Charissiadou, J. Anastasopoulos, S. K. Siam, Z.D. Pana, H. Jurgens, A.H. Groll, Candidaemia in a European Paediatric
Polychronopoulou, Fulminant Aeromonas hydrophila infection during acute University Hospital: a 10-year observational study, Clinical Microbiology and
lymphoblastic leukemia treatment, Journal of Microbiology, Immunology and Infection 18 (2012) E27E30.
Infection 45 (2012) 154157. [179] M.N. Chitasombat, D.P. Kofteridis, Y. Jiang, J. Tarrand, R.E. Lewis, D.P.
[172] S. Sutcliffe, E.A. Platz, Inammation and prostate cancer: a focus on infections, Kontoyiannis, Rare opportunistic (non-Candida, non-Cryptococcus) yeast
Current Urology Reports 9 (2008) 243249. bloodstream infections in patients with cancer, Journal of Infection 64 (2012)
[173] F.L. Fassi, T.N. Mak, B. Laube, V. Brinkmann, L.A. Ogilvie, H. Mollenkopf, M. Lein, 6875.
T. Schmidt, T.F. Meyer, H. Bruggemann, Prevalence of Propionibacterium acnes

S-ar putea să vă placă și