Summary of recent advances
The pathogen recognition receptors (PRR) initiate immediate responses against infection and tissue damage to protect the host from microbial invasion. In response to mucosal damage, intestinal PRR signaling initiates damage repair processes. Recent advances appear to link PRR abnormalities and inflammatory as well as neoplastic intestinal disorders. Emerging evidence suggests a dual role of PRRs in which they may simultaneously induce tumorigenesis and anti-tumor immunity. PRR may induce tumor cell proliferation by activating cell survival signaling mainly via NF-κB, but this signal can activate dendritic cells to promote anti-tumor immunity. TLR signaling within the tumor cells may result in evasion of immune surveillance, propagation of metastatic growth, or rather, induction of tumor cell apoptosis depending on ligands. Epithelial cells induce endogenous PRR ligands when damaged or during neoplastic transformation. Targeted manipulation of PRR signaling may provide emerging opportunities for the development of new therapeutic strategies for many gastrointestinal diseases.
Introduction
Pathogen recognition receptors (PPR)s are a series of innate immune receptors that include membrane bound toll-like receptors (TLR)s, cytoplasmic Nod-like receptors (NLR)s and an RNA helicase family of receptors. PRRs recognize pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs) and initiate immune responses against pathogens or repair responses in damaged tissues. Since the gastrointestinal mucosa is constantly exposed to diverse microorganisms and dietary antigens, the recognition and discrimination of pathogens from normal commensals or nutrients is one of the most important functions of the gastrointestinal tract, and homeostatic maintenance of PRR signaling is largely responsible for this function. Almost all of the TLRs, NLRs, and the RNA helicases (RIG-I and MDA5) are expressed by intestinal epithelial cells (IEC)s as well as in other types of cells in the intestine [1,2]. In the context of PRR-mediated intestinal mucosal homeostasis, TLRs balance inflammatory and anti-inflammatory responses against luminal antigens. Data have suggested that an additional balance of intestinal epithelial proliferation and apoptosis also involves TLR signaling [3,4]. Therefore, loss of these balances may induce dysregulated inflammation or abnormal epithelial regeneration. For instance, genetic studies have identified strong associations of PRR related gene mutations and development of idiopathic inflammatory bowel disease (IBD).
Recently, growing evidence has suggested involvement of PRRs, especially TLRs signaling, in tumor development [5–9]. Based on these reports, it seems that TLR signaling may both promote and prevent tumorigenesis. This paradox may be explained by the existence of different TLRs and different origins of tumor cells. For example, epithelial TLR4 signaling promotes tumorigenesis, but TLR4 signaling in dendritic cells (DCs) may help to promote anti-tumor immunity [5,10]. In fact, some TLR agonists have been tested for cancer immunotherapy and cancer vaccine adjuvant. The themes outlined in this paragraph will be covered in more detail later.
In this review, we discuss how PRR signaling maintains gastrointestinal homeostasis and how involvement of PRR signaling participates in inflammatory and neoplastic conditions of the gastrointestinal tract. We suggest opportunities for targeting of these pathways in inflammatory and neoplastic diseases.
Role of PRRs in the healthy gut
Our gastrointestinal tract is a unique organ that houses ~1014 microorganisms. The microbiota help to maintain our immune system and metabolic homeostasis as well as aid in digestion of nutrients. To maintain this coexistence, the gastrointestinal mucosa has to protect the host from pathogenic invasion while avoiding an excessive immune response against commensal bacteria. In order to avoid an excessive immune response and consequent deregulated inflammation, a variety of mechanisms regulate PRR signaling, especially in membrane bound TLRs.
Almost all TLRs, TLR1 through TLR9, are expressed not only on antigen presenting cells, but also on most cell types in the gastrointestinal mucosa [1,11,12]. IECs are the innermost lining of the mucosa, and TLR signaling is normally down regulated [13,14]. This down-regulation of signaling may be strictly controlled through decreased receptor expression on the epithelial cell surface and increased expression of inhibitors of TLR signaling [13,15,16]. DCs and macrophages in normal gastrointestinal mucosa have also been shown to be hyporesponsive or induce immune tolerance in response to TLR stimulation [17–19]. It has been suggested that the hyporesponsiveness of intestinal antigen presenting cells (APCs) may be due to epithelial or stromal derived factors such as thymic stromal lymphoprotein (TSLP) or TGF-β [19,20]. Therefore, epithelial or other stromal cells crosstalk with APCs to regulate their PRR response to maintain immune tolerance against commensals.
PRRs actively control commensal microbes by inducing antimicrobial peptides and secretory IgA. Defensins are antimicrobial peptides that are divided into two forms, α- and β-defensins. The α-defensins are constitutively expressed by Paneth cells or neutrophils and their expression is associated with NOD2 signaling. In contrast, β-defensins are inducible and mainly expressed by IECs. We have previously demonstrated that TLR4- and TLR2-dependent pathways can stimulate β-defensin-2 expression by human IECs [21]. Mucosal expression of another anti-microbial C-type lectin, Reg IIIγ, is regulated by MyD88-dependent signaling [22]. B cells can secrete non-specific IgA2 to control the load of commensal bacteria in a T cell-independent manner. IECs and DCs have been shown to participate in the IgA2 secretion through expression of the cytokines APRIL (A proliferation-inducing ligand), the BAFF (B cell-activation factor of the tumor necrosis factor family), or TSLP, which is responsible for local IgA class switching [23]. Most TLR ligands have been shown to induce expression of these cytokines, suggesting that PRR signaling culminates in increased IgA production in the intestinal mucosa.
Regulation of epithelial proliferation is an emerging role of PRRs in the gastrointestinal tract. We have shown that healing of injured intestinal epithelium requires the presence of intact TLR signaling [24]. Decreased epithelial proliferation is found in MyD88- and TLR4-deficient mice [24,25]. The defect in epithelial proliferation was attributed to lack of TLR4-mediated regulation of cyclooxygenase-2 (Cox-2) expression and subsequent prostaglandin E2 (PGE2) synthesis [4]. MyD88-dependent signaling is suggested to be involved in redistribution of Cox-2 expressing stromal cells, which can modulate epithelial cell proliferation in response to mucosal injury [3]. In addition, TLR4 signaling activates epidermal growth factor receptor (EGFR) through induction of one of the EGFR ligands, amphiregulin [4]. Therefore, by recognizing commensals, PRRs not only maintain the intestinal epithelial lining in normal mucosa, but also initiate mucosal healing by activating other repair signaling in the case of mucosal damage.
PRRs in gastrointestinal inflammation
During epithelial injury or pathogenic infection, activated PRRs signal to induce local innate immune responses against mucosal microorganisms. The innate immune responses include local pro-inflammatory cytokine and chemokine expression and subsequently an acute inflammatory cell infiltrate to the site of infection or tissue damage. PRR signaling also presents local information to T and B cells to generate antigen specific adaptive immunity and activate regulatory T cells at the same time in the lymphoid tissue. In addition, innate PRR signaling has to initiate a repair process to restore a damaged mucosal barrier so that the host can avoid further exposure to the luminal pathogens or commensals [3]. Therefore, PRR signaling in gastrointestinal mucosa works to clear a dangerous situation in the most efficient way. The initiation of regulatory immune functions is important to diminish unnecessary tissue damage induced by secreted pro-inflammatory substances. Emerging evidence demonstrates the requirement of direct TLR signaling in full activation of regulatory T cells [26,27]. Failure of any of these steps may result in prolonged or uncontrolled gastrointestinal inflammation.
Discovery of disease associated gene mutations has significantly improved our understanding of the pathogenesis of IBD. IBD, ulcerative colitis and Crohn’s disease, are idiopathic conditions of sustained and uncontrolled inflammation in the gastrointestinal tract [28]. Ulcerative colitis only affects the large intestine while Crohn’s disease may affect any part of the gastrointestinal tract. Based on findings in the inflamed mucosa, it had been thought that IBD results from aberrant adaptive immunity in the intestine. However, the first gene discovered associated with disease susceptibility in Crohn’s disease encoded NOD2 [29,30]. Further studies have demonstrated several polymorphisms in PRRs as candidate genes in IBD (Table 1). Importantly, most of these PRR-associated polymorphisms in IBD are loss-of-function mutations. Animal studies have provided detailed roles of individual PRRs in intestinal inflammation. Targeted gene knockout mice represent how the loss of function of a particular PRR influences intestinal inflammation. The gastrointestinal phenotype of PRR knockout mice are listed in Table 2. Most of the PRR-knockout mice are susceptible to dextran sodium sulfate (DSS)-induced colitis. [24,25] [25] [31] Subsets of patients with Crohn’s disease express serum antibodies reactive against flagellins derived from commensal bacteria, and truncating mutations in TLR5 prevent the development of these antibodies [32,33]. [34] [35] [36].
Table 1.
Table 2.
Gene Deficiency | Phenotype |
---|---|
TLR2 | Increased susceptibility to DSS colitis [25] |
TLR4 | Increased susceptibility to DSS colitis [24] Bacterial translocation [25] |
TLR5 | 25% of mice develop spontaneous colitis [31] |
TLR9 | Increased susceptibility to DSS colitis [83,84] |
MyD88 | Increased susceptibility to DSS colitis [25] Decreased heat-shock protein (hsp25 and hsp72) in colonic epithelium; Increased susceptibility to radiation injury [25] |
NOD2 | Increased susceptibility to oral infection of Listeria monocytogenes [34] Decreased expression of paneth cell α-defensins (Defcr4 and Defcr-rs10) [34] |
NOD22939ic | Increased susceptibility to DSS colitis [35] |
RIG-I | Spontaneous colitis, susceptible to DSS [36] |
Since PRR signaling may induce both pro- and anti-inflammatory cytokines, the balance of the cytokine induction is important to determine the course of mucosal inflammation. Roles of PRR signaling in cytokine production may differ between epithelial cells and other cell types in the intestinal mucosa. Mucosal DCs from patients with IBD have been shown to express more IL-12 and IL-6 but a similar amount of IL-10 compared to DCs from control subjects [37]. In contrast, IECs have been shown to play a central role for induction of intestinal immune tolerance by producing IL-10 and TGF-β in response to TLR stimulation [38]. TLR2 signaling is known to induce IL-10, and TLR4 can induce IL-10 and TGF-β, cytokines important for induction of regulatory T cells (Tregs). In the clinical setting, patients homozygous for the 3020insC frameshift mutation in the NOD2 gene demonstrate defective release of IL-10 from peripheral blood mononuclear cells after stimulation with TLR2 ligands [39]. Monocytes from patients with the Asp299Gly polymorphism of TLR4 demonstrate defective expression of anti-inflammatory cytokines rather than pro-inflammatory cytokines in response to TLR4 stimulation [40]. Therefore, PRR signaling in intestinal inflammation may act as a double-edged sword: in the early phases of colitis PRR signaling may increase inflammation but it is also responsible for stopping inflammation and repairing the epithelium.
PRRs and gastrointestinal carcinogenesis
Recognition of bacterial and/or viral products by PRRs may induce an injury repair response, which is associated with tumor promotion. Activated PRR signaling may support tumor growth through complex effects on the microenvironment. We have shown using a bone marrow chimera model that induction of chemokines (KC and CCL2) by intestinal epithelial TLR4 signaling is required to create a tumor promoting microenvironment in the setting of chronic colitis [41]. A recent finding demonstrates extracellular matrix proteoglycan produced by primary cancer cells activates TLR2 on macrophages, which facilitates metastatic tumor growth by establishing a microenvironment [42]. Huang et al, have shown that Listeria monocytogenes when present in the microenvironment of tumors accelerate tumor growth via TLR2 signaling in a mouse model of colon cancer xenografts into a BALB/c mouse [43]. In addition, TLR4 expression by colorectal tumor cells may facilitate evasion from immune surveillance perhaps through iNOS-mediated immune modulation [44]. Contrary to the above findings, lack of MyD88 or TLR5 enhanced tumor growth and inhibited tumor necrosis in a mouse model of colon cancer xenografts into a nude mouse [45]. This result was confirmed by tumor regression by stimulating TLR5 with a peritumoral flagellin treatment. An important difference is that the latter experiment is performed in the nude mouse which lacks adaptive immunity. Therefore, different PRRs may work differently in intestinal tumorigenesis.
Emerging evidence has suggested that genetic polymorphisms in PRRs are associated with gastrointestinal cancer. Table 3. These results indicate that abnormal PRR signaling may, at least in part, increase the risk in development of gastrointestinal cancer.
Table 3.
Polymorphisms/Mutations | Cancer |
---|---|
TLR2 (−196 to −174del/del) | Non-cardiac gastric cancer in Japanese population [85] |
TLR4 (Thr135Ala) | Poorly differentiated gastric adenocarcinomas [86] Gastric cancer [87] |
(Asp299Gly) | Gastric cancer [87] |
(Thr399Ile) | Pre-malignant lesions [88] |
NOD2 (3020insC) | Colorectal cancer in older Polish patients [89] |
NOD2 (R702W, G908R, 3020insC) | Colorectal cancer in Greek population [90] Not associated with gastric cancer in Caucasian population [91] Not associated with colorectal cancer in Hungarian subjects [92] |
TLR2 (microsatellite GT) | Colorectal cancer in Croatian patients [93] |
TLR4 (Asp299Gly) |
It has long been thought that cancers represent a “wound that does not heal” and a final stage of an abnormal and uncontrolled tissue repair process [46]. The wound healing process shares some aspects of tumorigenesis. We have found that TLR4 or MyD88-deficient mice have defective intestinal mucosal repair in response to chemically induced mucosal damage due to impaired response of Cox-2 and PGE2 production [4]. This impaired PGE2 production results in reduced transactivation of epidermal growth factor receptor (EGFR) [4]. In IECs, TLR4-deficent mice also have defective expression and secretion of amphiregulin, one of the EGFR ligands [5]. These data indicate that MyD88-dependent TLR4 signaling is largely responsible for intestinal mucosal wound healing. As a result, we have found that TLR4-deficient mice are protected against a murine model of colitis-associated tumor [5]. An animal study has demonstrated involvement of MyD88-dependent signaling in gastrointestinal tumor growth by using the ApcMin/+ mouse model [47]. The ApcMin/+ mice carry a germline mutation in the tumor suppressor Apc gene, which is responsible for familial adenomatous poliposis coli and also frequently mutated in sporadic colorectal cancer. In this study, MyD88-deficiency in ApcMin/+ mice resulted in a significant decrease in visible tumors in the small and large intestine [47]. Importantly, MyD88-deficiency did not influence the overall incidence of tumor development in this model.
Epidemiological studies have suggested that chronic inflammation plays a significant role in the development of gastrointestinal malignancies. A variety of chronic inflammatory conditions, e.g. Barrett’s esophagus, long-standing IBD, and H. pylori -associated chronic gastritis, markedly elevate the risk of developing gastrointestinal malignancies. A great deal of work has been done to identify bacterial factors important in H. pylori pathogenesis. Of these, expression of the CagA protein is thought to confer the greatest oncogenic risk. On the host side, TLR signaling may be quite important in H. pylori -associated gastric cancer. Chronic infection with H. pylori increases TLR4 and MD-2 expression in gastric epithelial cells, and recognition of H. pylori LPS augments NF-κB activation [48,49]. Furthermore, TLR4 is strongly expressed by tumor cells of H. pylori -associated gastric carcinoma [49].
We have reported that tumor cells in human and a mouse model of colitis-associated cancer have increased expression of TLR4 [5]. To assess whether this up-regulation of TLR4 in tumor cells is functionally involved in the pathogenesis of colitis-associated tumor, we challenged TLR4-defficient mice to a mouse model of colitis-associated cancer. We found in this study that TLR4-deficient mice exhibit a significantly decreased incidence and size of colorectal tumors compared to WT mice [5]. More recently, the importance of TLR signaling in colitis-associated tumorigenesis has also been shown in IL-10 knockout mice [50]. In this report, azoxymethane (AOM)-treated IL-10 knockout mice demonstrated development of multiple colorectal tumors including invasive cancer, but tumor development is completely abolished both under germ-free conditions and in the absence of MyD88 (IL-10−/− x MyD88−/− double knockout mice). These results indicate that chronic inflammation up-regulates TLRs, especially TLR4, which signaling through MyD88 promotes tumorigenesis in the gastrointestinal tract.
Several other studies however have demonstrated a protective role of TLR4 signaling against development of neoplastic lesions [8,51]. These conflicting results are observed outside of the gastrointestinal tract, such as lung and skin. It is possible but not yet clear that depending on the organ, TLR signaling plays diverse roles in tumorigenesis. At least the role of TLR signaling in tumor development may differ between the tumor cell and the environment, because the immune environment can either promote or impede tumor growth.
PRRs in anti-tumor immunity
Tumor immunosurveillance, a mechanism to protect hosts against the emergence of a tumor, is established by the interplay between antigen presenting cells (mostly DCs) and natural killer (NK) cells that recognize the transformed cells, followed by the subsequent development of tumor-specific cytotoxic T cell (CTL) responses. In this process, DCs are indispensable in cross-presenting tumor-associated antigens to CTLs, and TLR signaling may contribute to the phagosome maturation that enhances the antigen presentation [52]. However, DCs are not only efficient at generating CTLs, but can also generate tumor-specific regulatory T cells. This generation of tumor-specific Tregs is an obstacle to tumor immunity.
A recent study suggests that TLR4 and MyD88 play an important role in anti-cancer immune responses following chemotherapy and irradiation [53]. TLR4-deficient mice have significantly larger tumors than wild-type mice after doxorubicin and oxalplatin treatment or irradiation, yet displayed similar growth rates of a skin-transplanted colon cancer and thymoma cells in the absence of treatment [53]. Recently, a direct role of PRRs in the induction of anti-tumor immunity has been postulated. PRRs, in particular TLR4, detect endogenous ligands termed DAMPs that are released from injured and inflamed tissues. A wide range of DAMPs including high mobility group B1 (HMGB1), hyaluronan, S100 proteins, heat shock proteins, and fibronectin may activate TLRs to induce danger signaling [54–56]. In the tumor environment, damaged or necrotic tumor cells secrete these DAMPs, which subsequently trigger TLR4 activation in DCs and enhance antigen presentation to promote anti-tumor responses. In fact, TLR4 polymorphisms, manifested by TLR4 loss-of-function allele, lose this antigen cross-presentation process resulting in early tumor relapse and an increase in the frequency of metastasis in patients with breast cancer [53,57]. Therefore, impaired PRR function of TLR4 may attenuate the ability of anti-tumor immunity, thus acting in a tumorigenic manner.
PRRs as new therapeutic targets in gastrointestinal inflammation and cancer
TLR agonists are being developed for the treatment of cancer, allergies and viral infections, and as adjuvants for vaccines to prevent or treat cancer and infectious diseases. Several reports have demonstrated the efficacy of TLR and NOD2 agonists for prevention of murine IBD [58–61]. Notably, most of the inhibitory effects of these agonists are observed when administered prior to induction of intestinal inflammation. A subset of patients with Crohn’s disease showed decreased expression of mucosal TLR3 [62]. Crohn’s disease-associated NOD2 mutations are considered to be loss-of-function mutations [63]. Therefore, stimulation of TLR3 or NOD2 signaling in the mucosa can be a strategy for patients with Crohn’s disease with decreased expression of TLR3 or carrying NOD2 mutations, respectively. On the other hand, blocking TLR signaling is expected to be another strategy for IBD treatment, because TLR2 and TLR4 are up-regulated in IBD [62,64]. TLR4 antagonists are currently being developed [65]. However, when we studied a blocking antibody to TLR4 in acute and chronic murine models of colitis, acute inflammation was dampened and recovery delayed [66]. One aspect of TLR signaling that has not been well appreciated is that they can generate suppressive as well as inflammatory responses in innate immune cells and can promote the induction of regulatory as well as effector T cells. Therefore, to obtain more effective strategies, agonists or antagonists of a targeted function of PRR signaling need to be taken into account for future treatment.
Modulation of TLR signaling as anti-cancer therapy has been more successful than the above-mentioned strategies against inflammatory diseases. Generation of effective immunotherapy or therapeutic anti-cancer vaccines requires breaking the immunological tolerance and generating effector T cell responses specific for the “self” tumor antigens. In addition, we need to overcome the form of immune evasion and subversion by the tumor, which in many ways resembles those evolved by pathogens to circumvent protective immune responses of the host. The potential of TLR ligands to enhance and improve the efficacy of tumor vaccines has been shown. TLR3, TLR4, TLR7 and TLR9 ligands potently activate CD8+ CTLs with increased IFN-γ production and form an immunostimulatory microenvironment [67]. Synthetic ligands for TLR7, TLR4 and TLR9 have been through preclinical evaluation and clinical trials against cancer [68,69]. TLR4 activation by the organic compounds OK-432 or OK-PSA induces strong anti-cancer immunity [10,70]. Studies have shown enhanced chemosensitivity upon TLR stimulation. For example, TLR7 ligation results in sensitization of the chronic lymphocytic leukemia B cells to chemotherapeutic agents (vincristine) in vitro, presumably by enhancing vincristine-induced JNK signaling [71]. Another anti-tumor strategy is induction of apoptosis by TLR signaling. It has been observed that cells stimulated with TLR ligands undergo programmed cell death by Fas-associated death domain protein and subsequent caspase recruitment [72,73]. Studies have shown increased apoptosis of cancer cells by stimulating with the TLR3 agonist, suggesting that the TLR3 agonist may be another option for anti-cancer immunotherapy [74,75]. A recent report has shown a breakthrough in anti-cancer immunotherapy by taking advantage of RIG-I mediated immune response. Poeck et al., designed a small interfering RNA (siRNA), acting as an anti-tumor agent not only to silence the anti-apoptotic Bcl2 but also to activate RIG-I to stimulate anti-tumor immunity [76]. In this study, RIG-I signaling synergized with siRNA-mediated Bcl2 silencing to provoke massive apoptosis of tumor cells in lung metastasis in vivo.
Conclusions
The roles of PRRs in gastrointestinal inflammation and tumorigenesis are currently being elucidated. Extensive studies into the role of PRR signaling in gastrointestinal homeostasis have increased our understanding of the complicated and delicate balance of the PRR functions in the gastrointestinal mucosa. Very interestingly, defects in any part of PRR signaling in the gastrointestinal mucosa results in either inflammation or tumor development. In particular, epithelial TLR4 signaling via MyD88 may play a pivotal role in the development of cancer in the gastrointestinal tract. A better understanding of this complicated receptor family will provide clues for a future generation of effective strategies to treat many gastrointestinal diseases.
Acknowledgments
This work was supported by National Institutes of Health grants AI052266 and DK078682 for Maria T. Abreu, and a Career Development Award from the Crohn’s Colitis Foundation of America (CCFA) for Masayuki Fukata.
Abbreviations
- PAMP
pathogen-associated molecular pattern
- DAMP
danger-associated molecular pattern
- RIG-I
retinoic acid inducible gene I
- MDA5
melanoma differentiation-associated gene 5
Footnotes
There are no conflicts of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Abreu MT, Fukata M, Arditi M. TLR signaling in the gut in health and disease. J Immunol. 2005;174:4453–4460. doi: 10.4049/jimmunol.174.8.4453. [DOI] [PubMed] [Google Scholar]
- 2.Kufer TA, Banks DJ, Philpott DJ. Innate immune sensing of microbes by Nod proteins. Ann N Y Acad Sci. 2006;1072:19–27. doi: 10.1196/annals.1326.020. [DOI] [PubMed] [Google Scholar]
- 3•.Brown SL, Riehl TE, Walker MR, Geske MJ, Doherty JM, Stenson WF, Stappenbeck TS. Myd88-dependent positioning of Ptgs2-expressing stromal cells maintains colonic epithelial proliferation during injury. J Clin Invest. 2007;117:258–269. doi: 10.1172/JCI29159. Using Myd88- and prostaglandin-endoperoxide synthase 2 (Ptgs2)-deficient mice, the authors demonstrate an important role of MyD88- dependent signaling in relocation of a Ptgs2-expressing lamina propria stromal cells in response to mucosal injury, which is involved in the regulation of epithelial proliferation and cellular organization within the crypts. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Fukata M, Chen A, Klepper A, Krishnareddy S, Vamadevan AS, Thomas LS, Xu R, Inoue H, Arditi M, Dannenberg AJ, et al. Cox-2 is regulated by Toll-like receptor-4 (TLR4) signaling: Role in proliferation and apoptosis in the intestine. Gastroenterology. 2006;131:862–877. doi: 10.1053/j.gastro.2006.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5•.Fukata M, Chen A, Vamadevan AS, Cohen J, Breglio K, Krishnareddy S, Hsu D, Xu R, Harpaz N, Dannenberg AJ, et al. Toll-like receptor-4 promotes the development of colitis-associated colorectal tumors. Gastroenterology. 2007;133:1869–1881. doi: 10.1053/j.gastro.2007.09.008. The authors demonstrate an increased expression of TLR4 in human and murine model of colitis-associated dysplasia. TLR4-deficient mice show significantly lower incidence and size of colorectal tumors in the colitis-associated cancer model. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Chen R, Alvero AB, Silasi DA, Mor G. Inflammation, cancer and chemoresistance: taking advantage of the toll-like receptor signaling pathway. Am J Reprod Immunol. 2007;57:93–107. doi: 10.1111/j.1600-0897.2006.00441.x. [DOI] [PubMed] [Google Scholar]
- 7.Szczepanski MJ, Czystowska M, Szajnik M, Harasymczuk M, Boyiadzis M, Kruk-Zagajewska A, Szyfter W, Zeromski J, Whiteside TL. Triggering of Toll-like receptor 4 expressed on human head and neck squamous cell carcinoma promotes tumor development and protects the tumor from immune attack. Cancer Res. 2009;69:3105–3113. doi: 10.1158/0008-5472.CAN-08-3838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Yusuf N, Nasti TH, Long JA, Naseemuddin M, Lucas AP, Xu H, Elmets CA. Protective role of Toll-like receptor 4 during the initiation stage of cutaneous chemical carcinogenesis. Cancer Res. 2008;68:615–622. doi: 10.1158/0008-5472.CAN-07-5219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.El Andaloussi A, Sonabend AM, Han Y, Lesniak MS. Stimulation of TLR9 with CpG ODN enhances apoptosis of glioma and prolongs the survival of mice with experimental brain tumors. Glia. 2006;54:526–535. doi: 10.1002/glia.20401. [DOI] [PubMed] [Google Scholar]
- 10.Okamoto M, Oshikawa T, Tano T, Ahmed SU, Kan S, Sasai A, Akashi S, Miyake K, Moriya Y, Ryoma Y, et al. Mechanism of anticancer host response induced by OK-432, a streptococcal preparation, mediated by phagocytosis and Toll-like receptor 4 signaling. J Immunother. 2006;29:78–86. doi: 10.1097/01.cji.0000192106.32206.30. [DOI] [PubMed] [Google Scholar]
- 11.Bogunovic M, Dave SH, Tilstra JS, Chang DT, Harpaz N, Xiong H, Mayer LF, Plevy SE. Enteroendocrine cells express functional Toll-like receptors. Am J Physiol Gastrointest Liver Physiol. 2007;292:G1770–1783. doi: 10.1152/ajpgi.00249.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Rock FL, Hardiman G, Timans JC, Kastelein RA, Bazan JF. A family of human receptors structurally related to Drosophila Toll. Proc Natl Acad Sci U S A. 1998;95:588–593. doi: 10.1073/pnas.95.2.588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Melmed G, Thomas LS, Lee N, Tesfay SY, Lukasek K, Michelsen KS, Zhou Y, Hu B, Arditi M, Abreu MT. Human intestinal epithelial cells are broadly unresponsive to Toll-like receptor 2-dependent bacterial ligands: implications for host-microbial interactions in the gut. J Immunol. 2003;170:1406–1415. doi: 10.4049/jimmunol.170.3.1406. [DOI] [PubMed] [Google Scholar]
- 14.Otte JM, Cario E, Podolsky DK. Mechanisms of cross hyporesponsiveness to Toll-like receptor bacterial ligands in intestinal epithelial cells. Gastroenterology. 2004;126:1054–1070. doi: 10.1053/j.gastro.2004.01.007. [DOI] [PubMed] [Google Scholar]
- 15.Gewirtz AT, Navas TA, Lyons S, Godowski PJ, Madara JL. Cutting edge: bacterial flagellin activates basolaterally expressed TLR5 to induce epithelial proinflammatory gene expression. J Immunol. 2001;167:1882–1885. doi: 10.4049/jimmunol.167.4.1882. [DOI] [PubMed] [Google Scholar]
- 16.Naik S, Kelly EJ, Meijer L, Pettersson S, Sanderson IR. Absence of Toll-like receptor 4 explains endotoxin hyporesponsiveness in human intestinal epithelium. J Pediatr Gastroenterol Nutr. 2001;32:449–453. doi: 10.1097/00005176-200104000-00011. [DOI] [PubMed] [Google Scholar]
- 17.Rescigno M, Matteoli G. Lamina propria dendritic cells: for whom the bell TOLLs? Eur J Immunol. 2008;38:1483–1486. doi: 10.1002/eji.200838435. [DOI] [PubMed] [Google Scholar]
- 18.Sun CM, Hall JA, Blank RB, Bouladoux N, Oukka M, Mora JR, Belkaid Y. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med. 2007;204:1775–1785. doi: 10.1084/jem.20070602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Smythies LE, Sellers M, Clements RH, Mosteller-Barnum M, Meng G, Benjamin WH, Orenstein JM, Smith PD. Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity. J Clin Invest. 2005;115:66–75. doi: 10.1172/JCI19229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Iliev ID, Matteoli G, Rescigno M. The yin and yang of intestinal epithelial cells in controlling dendritic cell function. J Exp Med. 2007;204:2253–2257. doi: 10.1084/jem.20062535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Vora P, Youdim A, Thomas LS, Fukata M, Tesfay SY, Lukasek K, Michelsen KS, Wada A, Hirayama T, Arditi M, et al. Beta-defensin-2 expression is regulated by TLR signaling in intestinal epithelial cells. J Immunol. 2004;173:5398–5405. doi: 10.4049/jimmunol.173.9.5398. [DOI] [PubMed] [Google Scholar]
- 22.Brandl K, Plitas G, Schnabl B, DeMatteo RP, Pamer EG. MyD88-mediated signals induce the bactericidal lectin RegIII gamma and protect mice against intestinal Listeria monocytogenes infection. J Exp Med. 2007;204:1891–1900. doi: 10.1084/jem.20070563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23•.He B, Xu W, Santini PA, Polydorides AD, Chiu A, Estrella J, Shan M, Chadburn A, Villanacci V, Plebani A, et al. Intestinal bacteria trigger T cell-independent immunoglobulin A(2) class switching by inducing epithelial-cell secretion of the cytokine APRIL. Immunity. 2007;26:812–826. doi: 10.1016/j.immuni.2007.04.014. This paper demonstrates that TLR signaling in human IEC induces IgA class switching in lamina propria B cells by inducing APRIL and TSLP. [DOI] [PubMed] [Google Scholar]
- 24.Fukata M, Michelsen KS, Eri R, Thomas LS, Hu B, Lukasek K, Nast CC, Lechago J, Xu R, Naiki Y, et al. Toll-like receptor-4 is required for intestinal response to epithelial injury and limiting bacterial translocation in a murine model of acute colitis. Am J Physiol Gastrointest Liver Physiol. 2005;288:G1055–1065. doi: 10.1152/ajpgi.00328.2004. [DOI] [PubMed] [Google Scholar]
- 25.Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004;118:229–241. doi: 10.1016/j.cell.2004.07.002. [DOI] [PubMed] [Google Scholar]
- 26.Sutmuller RP, den Brok MH, Kramer M, Bennink EJ, Toonen LW, Kullberg BJ, Joosten LA, Akira S, Netea MG, Adema GJ. Toll-like receptor 2 controls expansion and function of regulatory T cells. J Clin Invest. 2006;116:485–494. doi: 10.1172/JCI25439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Fukata M, Breglio K, Chen A, Vamadevan AS, Goo T, Hsu D, Conduah D, Xu R, Abreu MT. The myeloid differentiation factor 88 (MyD88) is required for CD4+ T cell effector function in a murine model of inflammatory bowel disease. J Immunol. 2008;180:1886–1894. doi: 10.4049/jimmunol.180.3.1886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Young Y, Abreu MT. Advances in the pathogenesis of inflammatory bowel disease. Curr Gastroenterol Rep. 2006;8:470–477. doi: 10.1007/s11894-006-0037-1. [DOI] [PubMed] [Google Scholar]
- 29.Ogura Y, Bonen DK, Inohara N, Nicolae DL, Chen FF, Ramos R, Britton H, Moran T, Karaliuskas R, Duerr RH, et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease. Nature. 2001;411:603–606. doi: 10.1038/35079114. [DOI] [PubMed] [Google Scholar]
- 30.Hugot JP, Chamaillard M, Zouali H, Lesage S, Cezard JP, Belaiche J, Almer S, Tysk C, O’Morain CA, Gassull M, et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature. 2001;411:599–603. doi: 10.1038/35079107. [DOI] [PubMed] [Google Scholar]
- 31.Vijay-Kumar M, Sanders CJ, Taylor RT, Kumar A, Aitken JD, Sitaraman SV, Neish AS, Uematsu S, Akira S, Williams IR, et al. Deletion of TLR5 results in spontaneous colitis in mice. J Clin Invest. 2007;117:3909–3921. doi: 10.1172/JCI33084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Lodes MJ, Cong Y, Elson CO, Mohamath R, Landers CJ, Targan SR, Fort M, Hershberg RM. Bacterial flagellin is a dominant antigen in Crohn disease. J Clin Invest. 2004;113:1296–1306. doi: 10.1172/JCI20295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Sitaraman SV, Klapproth JM, Moore DA, 3rd, Landers C, Targan S, Williams IR, Gewirtz AT. Elevated flagellin-specific immunoglobulins in Crohn’s disease. Am J Physiol Gastrointest Liver Physiol. 2005;288:G403–406. doi: 10.1152/ajpgi.00357.2004. [DOI] [PubMed] [Google Scholar]
- 34.Kobayashi KS, Chamaillard M, Ogura Y, Henegariu O, Inohara N, Nunez G, Flavell RA. Nod2-dependent regulation of innate and adaptive immunity in the intestinal tract. Science. 2005;307:731–734. doi: 10.1126/science.1104911. [DOI] [PubMed] [Google Scholar]
- 35.Maeda S, Hsu LC, Liu H, Bankston LA, Iimura M, Kagnoff MF, Eckmann L, Karin M. Nod2 mutation in Crohn’s disease potentiates NF-kappaB activity and IL-1beta processing. Science. 2005;307:734–738. doi: 10.1126/science.1103685. [DOI] [PubMed] [Google Scholar]
- 36••.Wang Y, Zhang HX, Sun YP, Liu ZX, Liu XS, Wang L, Lu SY, Kong H, Liu QL, Li XH, et al. Rig-I−/− mice develop colitis associated with downregulation of G alpha i2. Cell Res. 2007;17:858–868. doi: 10.1038/cr.2007.81. Compared to TLRs, little is known about the role of RNA helicases in gastrointestinal inflammation and cancer. This paper demonstrates spontaneous development of colitis in RIG-I-deficient mice. RIG-I regulates T cell expression of Gαi2, which is important to maintain T cell activation. The authors show increased effector T cells in the RIG-I-deficient mice as a mechanism by which scolitis is induced. [DOI] [PubMed] [Google Scholar]
- 37.Hart AL, Al-Hassi HO, Rigby RJ, Bell SJ, Emmanuel AV, Knight SC, Kamm MA, Stagg AJ. Characteristics of intestinal dendritic cells in inflammatory bowel diseases. Gastroenterology. 2005;129:50–65. doi: 10.1053/j.gastro.2005.05.013. [DOI] [PubMed] [Google Scholar]
- 38•.Jarry A, Bossard C, Bou-Hanna C, Masson D, Espaze E, Denis MG, Laboisse CL. Mucosal IL-10 and TGF-beta play crucial roles in preventing LPS-driven, IFN-gamma-mediated epithelial damage in human colon explants. J Clin Invest. 2008;118:1132–1142. doi: 10.1172/JCI32140. Using human intestinal mucosal explants, the authors demonstrate how the mucosal immune balance is regulated. Blocking IL-10 or TGF-β by a neutralizing antibody results in epithelial disruption due to increased production of IFN-γ and TNF-α. Further blocking of TLR4 signaling prevents epithelial damage by suppressing the production of IFN-γ and TNF-α. Importantly, they show that the main source of mucosal IL-10 production is IECs. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Pauleau AL, Murray PJ. Role of nod2 in the response of macrophages to toll-like receptor agonists. Mol Cell Biol. 2003;23:7531–7539. doi: 10.1128/MCB.23.21.7531-7539.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Dehus O, Bunk S, von Aulock S, Hermann C. IL-10 release requires stronger toll-like receptor 4-triggering than TNF: a possible explanation for the selective effects of heterozygous TLR4 polymorphism Asp(299)Gly on IL-10 release. Immunobiology. 2008;213:621–627. doi: 10.1016/j.imbio.2008.03.001. [DOI] [PubMed] [Google Scholar]
- 41.Fukata M, Hernandez Y, Conduah D, Cohen J, Chen A, Breglio K, Goo T, Hsu D, Xu R, Abreu MT. Innate immune signaling by Toll-like receptor-4 (TLR4) shapes the inflammatory microenvironment in colitis-associated tumors. Inflamm Bowel Dis. 2009;15:997–1006. doi: 10.1002/ibd.20880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42••.Kim S, Takahashi H, Lin WW, Descargues P, Grivennikov S, Kim Y, Luo JL, Karin M. Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature. 2009;457:102–106. doi: 10.1038/nature07623. The authors clearly show importance of TLR2 signaling in macrophages for establishment of a tumor microenvironment. Human Lewis lung carcinoma (LLC) secrets the extracellular matrix proteoglycan, versican, which activates macrophages via TLR2 and TLR6, resulting in metastatic tumor growth in a murine exograft tumor model. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Huang B, Zhao J, Shen S, Li H, He KL, Shen GX, Mayer L, Unkeless J, Li D, Yuan Y, et al. Listeria monocytogenes promotes tumor growth via tumor cell toll-like receptor 2 signaling. Cancer Res. 2007;67:4346–4352. doi: 10.1158/0008-5472.CAN-06-4067. [DOI] [PubMed] [Google Scholar]
- 44.Huang B, Zhao J, Li H, He KL, Chen Y, Chen SH, Mayer L, Unkeless JC, Xiong H. Toll-like receptors on tumor cells facilitate evasion of immune surveillance. Cancer Res. 2005;65:5009–5014. doi: 10.1158/0008-5472.CAN-05-0784. [DOI] [PubMed] [Google Scholar]
- 45.Rhee SH, Im E, Pothoulakis C. Toll-like receptor 5 engagement modulates tumor development and growth in a mouse xenograft model of human colon cancer. Gastroenterology. 2008;135:518–528. doi: 10.1053/j.gastro.2008.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315:1650–1659. doi: 10.1056/NEJM198612253152606. [DOI] [PubMed] [Google Scholar]
- 47.Rakoff-Nahoum S, Medzhitov R. Regulation of spontaneous intestinal tumorigenesis through the adaptor protein MyD88. Science. 2007;317:124–127. doi: 10.1126/science.1140488. [DOI] [PubMed] [Google Scholar]
- 48.Ishihara S, Rumi MA, Kadowaki Y, Ortega-Cava CF, Yuki T, Yoshino N, Miyaoka Y, Kazumori H, Ishimura N, Amano Y, et al. Essential role of MD-2 in TLR4-dependent signaling during Helicobacter pylori-associated gastritis. J Immunol. 2004;173:1406–1416. doi: 10.4049/jimmunol.173.2.1406. [DOI] [PubMed] [Google Scholar]
- 49.Schmausser B, Andrulis M, Endrich S, Muller-Hermelink HK, Eck M. Toll-like receptors TLR4, TLR5 and TLR9 on gastric carcinoma cells: an implication for interaction with Helicobacter pylori. Int J Med Microbiol. 2005;295:179–185. doi: 10.1016/j.ijmm.2005.02.009. [DOI] [PubMed] [Google Scholar]
- 50.Uronis JM, Muhlbauer M, Herfarth HH, Rubinas TC, Jones GS, Jobin C. Modulation of the intestinal microbiota alters colitis-associated colorectal cancer susceptibility. PLoS One. 2009;4:e6026. doi: 10.1371/journal.pone.0006026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Bauer AK, Dixon D, DeGraff LM, Cho HY, Walker CR, Malkinson AM, Kleeberger SR. Toll-like receptor 4 in butylated hydroxytoluene-induced mouse pulmonary inflammation and tumorigenesis. J Natl Cancer Inst. 2005;97:1778–1781. doi: 10.1093/jnci/dji403. [DOI] [PubMed] [Google Scholar]
- 52.Blander JM, Medzhitov R. Regulation of phagosome maturation by signals from toll-like receptors. Science. 2004;304:1014–1018. doi: 10.1126/science.1096158. [DOI] [PubMed] [Google Scholar]
- 53.Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, Mignot G, Maiuri MC, Ullrich E, Saulnier P, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13:1050–1059. doi: 10.1038/nm1622. [DOI] [PubMed] [Google Scholar]
- 54.Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418:191–195. doi: 10.1038/nature00858. [DOI] [PubMed] [Google Scholar]
- 55.Somersan S, Larsson M, Fonteneau JF, Basu S, Srivastava P, Bhardwaj N. Primary tumor tissue lysates are enriched in heat shock proteins and induce the maturation of human dendritic cells. J Immunol. 2001;167:4844–4852. doi: 10.4049/jimmunol.167.9.4844. [DOI] [PubMed] [Google Scholar]
- 56.Scheibner KA, Lutz MA, Boodoo S, Fenton MJ, Powell JD, Horton MR. Hyaluronan fragments act as an endogenous danger signal by engaging TLR2. J Immunol. 2006;177:1272–1281. doi: 10.4049/jimmunol.177.2.1272. [DOI] [PubMed] [Google Scholar]
- 57.Apetoh L, Ghiringhelli F, Tesniere A, Criollo A, Ortiz C, Lidereau R, Mariette C, Chaput N, Mira JP, Delaloge S, et al. The interaction between HMGB1 and TLR4 dictates the outcome of anticancer chemotherapy and radiotherapy. Immunol Rev. 2007;220:47–59. doi: 10.1111/j.1600-065X.2007.00573.x. [DOI] [PubMed] [Google Scholar]
- 58.Obermeier F, Dunger N, Strauch UG, Grunwald N, Herfarth H, Scholmerich J, Falk W. Contrasting activity of cytosin-guanosin dinucleotide oligonucleotides in mice with experimental colitis. Clin Exp Immunol. 2003;134:217–224. doi: 10.1046/j.1365-2249.2003.02288.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Vijay-Kumar M, Aitken JD, Sanders CJ, Frias A, Sloane VM, Xu J, Neish AS, Rojas M, Gewirtz AT. Flagellin treatment protects against chemicals, bacteria, viruses, and radiation. J Immunol. 2008;180:8280–8285. [Google Scholar]
- 60.Vijay-Kumar M, Wu H, Aitken J, Kolachala VL, Neish AS, Sitaraman SV, Gewirtz AT. Activation of toll-like receptor 3 protects against DSS-induced acute colitis. Inflamm Bowel Dis. 2007;13:856–864. doi: 10.1002/ibd.20142. [DOI] [PubMed] [Google Scholar]
- 61.Watanabe T, Asano N, Murray PJ, Ozato K, Tailor P, Fuss IJ, Kitani A, Strober W. Muramyl dipeptide activation of nucleotide-binding oligomerization domain 2 protects mice from experimental colitis. J Clin Invest. 2008;118:545–559. doi: 10.1172/JCI33145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Cario E, Podolsky DK. Differential alteration in intestinal epithelial cell expression of toll-like receptor 3 (TLR3) and TLR4 in inflammatory bowel disease. Infect Immun. 2000;68:7010–7017. doi: 10.1128/iai.68.12.7010-7017.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Netea MG, Ferwerda G, de Jong DJ, Werts C, Boneca IG, Jehanno M, Van Der Meer JW, Mengin-Lecreulx D, Sansonetti PJ, Philpott DJ, et al. The frameshift mutation in Nod2 results in unresponsiveness not only to Nod2- but also Nod1-activating peptidoglycan agonists. J Biol Chem. 2005;280:35859–35867. doi: 10.1074/jbc.M504924200. [DOI] [PubMed] [Google Scholar]
- 64.Hausmann M, Kiessling S, Mestermann S, Webb G, Spottl T, Andus T, Scholmerich J, Herfarth H, Ray K, Falk W, et al. Toll-like receptors 2 and 4 are up-regulated during intestinal inflammation. Gastroenterology. 2002;122:1987–2000. doi: 10.1053/gast.2002.33662. [DOI] [PubMed] [Google Scholar]
- 65.Rossignol DP, Wasan KM, Choo E, Yau E, Wong N, Rose J, Moran J, Lynn M. Safety, pharmacokinetics, pharmacodynamics, and plasma lipoprotein distribution of eritoran (E5564) during continuous intravenous infusion into healthy volunteers. Antimicrob Agents Chemother. 2004;48:3233–3240. doi: 10.1128/AAC.48.9.3233-3240.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ungaro R, Fukata M, Hsu D, Hernandez Y, Breglio K, Chen A, Xu R, Sotolongo J, Espana C, Zaias J, et al. A novel Toll-like receptor 4 antagonist antibody ameliorates inflammation but impairs mucosal healing in murine colitis. Am J Physiol Gastrointest Liver Physiol. 2009;296:G1167–1179. doi: 10.1152/ajpgi.90496.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Hamdy S, Molavi O, Ma Z, Haddadi A, Alshamsan A, Gobti Z, Elhasi S, Samuel J, Lavasanifar A. Co-delivery of cancer-associated antigen and Toll-like receptor 4 ligand in PLGA nanoparticles induces potent CD8+ T cell-mediated anti-tumor immunity. Vaccine. 2008;26:5046–5057. doi: 10.1016/j.vaccine.2008.07.035. [DOI] [PubMed] [Google Scholar]
- 68.Krieg AM. Therapeutic potential of Toll-like receptor 9 activation. Nat Rev Drug Discov. 2006;5:471–484. doi: 10.1038/nrd2059. [DOI] [PubMed] [Google Scholar]
- 69.Kanzler H, Barrat FJ, Hessel EM, Coffman RL. Therapeutic targeting of innate immunity with Toll-like receptor agonists and antagonists. Nat Med. 2007;13:552–559. doi: 10.1038/nm1589. [DOI] [PubMed] [Google Scholar]
- 70.Homma S, Sagawa Y, Komita H, Koido S, Nagasaki E, Ryoma Y, Okamoto M. Mechanism of antitumor effect on mouse hepatocellular carcinoma by intratumoral injection of OK-432, a streptococcal preparation. Cancer Immunol Immunother. 2007;56:1265–1274. doi: 10.1007/s00262-006-0277-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Shi Y, White D, He L, Miller RL, Spaner DE. Toll-like receptor-7 tolerizes malignant B cells and enhances killing by cytotoxic agents. Cancer Res. 2007;67:1823–1831. doi: 10.1158/0008-5472.CAN-06-2381. [DOI] [PubMed] [Google Scholar]
- 72.Into T, Kiura K, Yasuda M, Kataoka H, Inoue N, Hasebe A, Takeda K, Akira S, Shibata K. Stimulation of human Toll-like receptor (TLR) 2 and TLR6 with membrane lipoproteins of Mycoplasma fermentans induces apoptotic cell death after NF-kappa B activation. Cell Microbiol. 2004;6:187–199. doi: 10.1046/j.1462-5822.2003.00356.x. [DOI] [PubMed] [Google Scholar]
- 73.Jung DY, Lee H, Jung BY, Ock J, Lee MS, Lee WH, Suk K. TLR4, but not TLR2, signals autoregulatory apoptosis of cultured microglia: a critical role of IFN-beta as a decision maker. J Immunol. 2005;174:6467–6476. doi: 10.4049/jimmunol.174.10.6467. [DOI] [PubMed] [Google Scholar]
- 74.Paone A, Starace D, Galli R, Padula F, De Cesaris P, Filippini A, Ziparo E, Riccioli A. Toll-like receptor 3 triggers apoptosis of human prostate cancer cells through a PKC-alpha-dependent mechanism. Carcinogenesis. 2008;29:1334–1342. doi: 10.1093/carcin/bgn149. [DOI] [PubMed] [Google Scholar]
- 75.Salaun B, Lebecque S, Matikainen S, Rimoldi D, Romero P. Toll-like receptor 3 expressed by melanoma cells as a target for therapy? Clin Cancer Res. 2007;13:4565–4574. doi: 10.1158/1078-0432.CCR-07-0274. [DOI] [PubMed] [Google Scholar]
- 76••.Poeck H, Besch R, Maihoefer C, Renn M, Tormo D, Morskaya SS, Kirschnek S, Gaffal E, Landsberg J, Hellmuth J, et al. 5′-Triphosphate-siRNA: turning gene silencing and Rig-I activation against melanoma. Nat Med. 2008;14:1256–1263. doi: 10.1038/nm.1887. To achieve effective anti-cancer immunotherapy, efficient induction of anti-cancer immunity is required to overcome immune evasion and subversion by the tumor. The authors generate siRNA against Bcl2 to induce tumor apoptosis. To simultaneously stimulate anti-cancer immunity, the authors modified the siRNA with 5′-triphosphate ends (3p-siRNA). 5′-triphosphate successfully activates RIG-I, induces expression of IFN and assists tumor apoptosis in vivo. [DOI] [PubMed] [Google Scholar]
- 77.McGovern DP, Hysi P, Ahmad T, van Heel DA, Moffatt MF, Carey A, Cookson WO, Jewell DP. Association between a complex insertion/deletion polymorphism in NOD1 (CARD4) and susceptibility to inflammatory bowel disease. Hum Mol Genet. 2005;14:1245–1250. doi: 10.1093/hmg/ddi135. [DOI] [PubMed] [Google Scholar]
- 78.Pierik M, Joossens S, Van Steen K, Van Schuerbeek N, Vlietinck R, Rutgeerts P, Vermeire S. Toll-like receptor-1, -2, and -6 polymorphisms influence disease extension in inflammatory bowel diseases. Inflamm Bowel Dis. 2006;12:1–8. doi: 10.1097/01.mib.0000195389.11645.ab. [DOI] [PubMed] [Google Scholar]
- 79.Franchimont D, Vermeire S, El Housni H, Pierik M, Van Steen K, Gustot T, Quertinmont E, Abramowicz M, Van Gossum A, Deviere J, et al. Deficient host-bacteria interactions in inflammatory bowel disease? The toll-like receptor (TLR)-4 Asp299gly polymorphism is associated with Crohn’s disease and ulcerative colitis. Gut. 2004;53:987–992. doi: 10.1136/gut.2003.030205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Torok HP, Glas J, Tonenchi L, Mussack T, Folwaczny C. Polymorphisms of the lipopolysaccharide-signaling complex in inflammatory bowel disease: association of a mutation in the Toll-like receptor 4 gene with ulcerative colitis. Clin Immunol. 2004;112:85–91. doi: 10.1016/j.clim.2004.03.002. [DOI] [PubMed] [Google Scholar]
- 81.Brand S, Staudinger T, Schnitzler F, Pfennig S, Hofbauer K, Dambacher J, Seiderer J, Tillack C, Konrad A, Crispin A, et al. The role of Toll-like receptor 4 Asp299Gly and Thr399Ile polymorphisms and CARD15/NOD2 mutations in the susceptibility and phenotype of Crohn’s disease. Inflamm Bowel Dis. 2005;11:645–652. doi: 10.1097/01.mib.0000168372.94907.d2. [DOI] [PubMed] [Google Scholar]
- 82.Hong J, Leung E, Fraser AG, Merriman TR, Vishnu P, Krissansen GW. TLR2, TLR4 and TLR9 polymorphisms and Crohn’s disease in a New Zealand Caucasian cohort. J Gastroenterol Hepatol. 2007;22:1760–1766. doi: 10.1111/j.1440-1746.2006.04727.x. [DOI] [PubMed] [Google Scholar]
- 83.Obermeier F, Dunger N, Strauch UG, Hofmann C, Bleich A, Grunwald N, Hedrich HJ, Aschenbrenner E, Schlegelberger B, Rogler G, et al. CpG motifs of bacterial DNA essentially contribute to the perpetuation of chronic intestinal inflammation. Gastroenterology. 2005;129:913–927. doi: 10.1053/j.gastro.2005.06.061. [DOI] [PubMed] [Google Scholar]
- 84.Lee J, Mo JH, Katakura K, Alkalay I, Rucker AN, Liu YT, Lee HK, Shen C, Cojocaru G, Shenouda S, et al. Maintenance of colonic homeostasis by distinctive apical TLR9 signalling in intestinal epithelial cells. Nat Cell Biol. 2006;8:1327–1336. doi: 10.1038/ncb1500. [DOI] [PubMed] [Google Scholar]
- 85.Tahara T, Arisawa T, Wang F, Shibata T, Nakamura M, Sakata M, Hirata I, Nakano H. Toll-like receptor 2 −196 to 174del polymorphism influences the susceptibility of Japanese people to gastric cancer. Cancer Sci. 2007;98:1790–1794. doi: 10.1111/j.1349-7006.2007.00590.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Ohara T, Morishita T, Suzuki H, Hibi T. Heterozygous Thr 135 Ala polymorphism at leucine-rich repeat (LRR) in genomic DNA of toll-like receptor 4 in patients with poorly-differentiated gastric adenocarcinomas. Int J Mol Med. 2006;18:59–63. [PubMed] [Google Scholar]
- 87.Hold GL, Rabkin CS, Chow WH, Smith MG, Gammon MD, Risch HA, Vaughan TL, McColl KE, Lissowska J, Zatonski W, et al. A functional polymorphism of toll-like receptor 4 gene increases risk of gastric carcinoma and its precursors. Gastroenterology. 2007;132:905–912. doi: 10.1053/j.gastro.2006.12.026. [DOI] [PubMed] [Google Scholar]
- 88.Achyut BR, Ghoshal UC, Moorchung N, Mittal B. Association of Toll-like receptor-4 (Asp299Gly and Thr399Ileu) gene polymorphisms with gastritis and precancerous lesions. Hum Immunol. 2007;68:901–907. doi: 10.1016/j.humimm.2007.10.006. [DOI] [PubMed] [Google Scholar]
- 89.Kurzawski G, Suchy J, Kladny J, Grabowska E, Mierzejewski M, Jakubowska A, Debniak T, Cybulski C, Kowalska E, Szych Z, et al. The NOD2 3020insC mutation and the risk of colorectal cancer. Cancer Res. 2004;64:1604–1606. doi: 10.1158/0008-5472.can-03-3791. [DOI] [PubMed] [Google Scholar]
- 90.Papaconstantinou I, Theodoropoulos G, Gazouli M, Panoussopoulos D, Mantzaris GJ, Felekouras E, Bramis J. Association between mutations in the CARD15/NOD2 gene and colorectal cancer in a Greek population. Int J Cancer. 2005;114:433–435. doi: 10.1002/ijc.20747. [DOI] [PubMed] [Google Scholar]
- 91.Wex T, Ebert MP, Kropf S, Dierkes J, Schuttler K, Rocken C, Hocker M, Malfertheiner P. Gene polymorphisms of the NOD-2/CARD-15 gene and the risk of gastric cancer in Germany. Anticancer Res. 2008;28:757–762. [PubMed] [Google Scholar]
- 92.Lakatos PL, Hitre E, Szalay F, Zinober K, Fuszek P, Lakatos L, Fischer S, Osztovits J, Gemela O, Veres G, et al. Common NOD2/CARD15 variants are not associated with susceptibility or the clinicopathologic characteristics of sporadic colorectal cancer in Hungarian patients. BMC Cancer. 2007;7:54. doi: 10.1186/1471-2407-7-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Boraska Jelavic T, Barisic M, Drmic Hofman I, Boraska V, Vrdoljak E, Peruzovic M, Hozo I, Puljiz Z, Terzic J. Microsatelite GT polymorphism in the toll-like receptor 2 is associated with colorectal cancer. Clin Genet. 2006;70:156–160. doi: 10.1111/j.1399-0004.2006.00651.x. [DOI] [PubMed] [Google Scholar]