Open access peer-reviewed chapter

Pattern Recognition Receptor-Mediated Regulatory T Cell Functions in Diseases

Written By

Ece Oylumlu, Goksu Uzel, Lubeyne Durmus, Meric Tas, Damla Gunes and Ceren Ciraci

Submitted: 19 February 2022 Reviewed: 03 June 2022 Published: 07 July 2022

DOI: 10.5772/intechopen.105693

From the Edited Volume

Regulatory T Cells - New Insights

Edited by Xuehui He

Chapter metrics overview

139 Chapter Downloads

View Full Metrics

Abstract

The advent of new technologies in gene expression, immunology, molecular biology, and computational modeling studies has expedited the discovery process and provided us with a holistic view of host immune responses that are highly regulated. The regulatory mechanisms of the immune system lie not only in weakening the attacker directly but also in fortifying the defender for the development of an efficient adaptive immune response. This chapter reviews a comprehensive set of experimental and bioinformatic studies designed to deepen the current knowledge on the regulatory T cells (Tregs) in the context of Pattern Recognition Receptors (PRRs). Initially, we examined both membrane-bound Toll-like Receptors (TLRs) and C Type Lectin Receptors (CLRs); and cytosolic NOD-like Receptors (NLRs) and RIG-I like Receptors (RLRs) in Tregs. Then, we revisited the disease conditions associated with regulatory T cells by emphasizing the essential roles of PRRs. Expanding our knowledge and strategies on the regulatory mechanisms are likely to provide our best chances for long-term disease control and maintenance of homeostasis.

Keywords

  • pattern recognition receptors
  • regulatory T cells
  • NLRs
  • TLRs
  • CTLs
  • RLRs
  • disease

1. Introduction

Regulatory T cells (Tregs) are a subtype of T cells that are responsible for the maintenance of homeostasis and tolerance to self-molecules. They mediate their action by suppressing the T cell proliferation, and cytokine productions; thereby preventing autoimmunity [1]. In this sense, Tregs can be both helpful to the host by alleviating the immunopathology, and immune system related tissue damages and unfortunately can also be harmful to the host by sabotaging the properly induced immune responses against pathogens [2]. Therefore, harnessing Treg mechanisms could be an efficient therapeutic approach to treat some distinct diseases, including infectious diseases, asthma and allergies, and cancer [3].

It is established that immune cells rely on the germ-line encoded pattern recognition receptors (PRR) that recognize common structural motifs shared by pathogens called pathogen-associated molecular patterns (PAMPs), and also recognize cellular stress and death via molecules known as damage-associated molecular patterns (DAMPs) to initiate inflammation and activate tissue repair mechanisms [4]. As much as the efficacy of PRRs in executing an immune response is critical for the host, it can also be the reason for unintended responses. Fortunately, understanding of how PRRs drive these responses has expanded enormously in the last few decades. In this chapter, we compile the available data using the open-source databases and the current knowledge in an attempt to discern the layers of complex mechanisms from a regulatory T cell standpoint.

Advertisement

2. The expression profiles of Pattern Recognition Receptors (PRRs) in regulatory T cells (Tregs)

2.1 Membrane-bound Toll-Like Receptors (TLRs)

Toll-like receptors (TLRs) have critical roles in the initial defense of innate and adaptive immunity [5]. TLRs which are type I integral membrane receptors have three domains: The N-terminal domain (NTD), which is located either on the outside of the cell membrane or in endosomes, a single helix transmembrane domain that is in the center, and the C-terminal domain (CTD), which is located in the cytoplasm. The N-terminal ectodomains contain a conserved 19–25 tandem leucine-rich repeat (LRR) region leading to the recognition of PAMPs and DAMPs. NTD also contains glycan moieties to bind ligands from different pathogens. On the other hand, the CTD contains the toll-IL-1 receptor (TIR) homologous domain, which enables the interaction with downstream adaptor proteins for signal transduction and thus, activation of the signaling pathway [6, 7, 8]. To date, 13 members of the mammalian TLRs have been identified. 10 members of this receptor family are expressed in humans (TLR1–10), while 12 members are expressed in mice (TLR1–9, TLR11–13). Each TLR can recognize different PAMPs from various pathogens. TLRs divided into two classes according to their localization: cell surface and intracellular [4, 5]. TLR1/TLR2 (triacyl lipopeptides), TLR4 (lipopolysaccharide), TLR5 (flagellin), TLR2/TLR6 (lipoproteins), TLR10 (bacterial 23S rRNA) are expressed in the cell membrane and TLR3 (dsRNA), TLR7/TLR8 (ssRNA), TLR9 (unmethylated CpG DNA), TLR11 (flagellin or profilin-like molecule from T. gondii), TLR12 (profilin from T. gondii), and TLR13 (bacterial 23S rRNA) are expressed in endosomal membranes [4]. TLRs are important for the proper functioning of Tregs because they directly mediate the pathogen sensing. Tregs have higher expression levels of TLR4, TLR5, TLR7, and TLR8 as compared to the effector T cells in humans; however, especially TLR2, TLR4, TLR5, and TLR8 activation has different effects on the differentiation, expansion, and proliferation of Tregs [9]. Although their numbers increased, Tregs lost their suppressive function when treated with PAM3Cys to activate TLR2 signaling, along with T cell receptor (TCR) and interleukin 2 (IL2) stimulation in order for Treg differentiation and function [10, 11]. In this study, the immune response was suppressed neither in vitro nor in vivo in mice who underwent acute infection, as Tregs were not activated by the induction of the TLR2 signaling pathway [9]. However, when the TLR2 ligand was removed, Tregs’ suppressive functions were recovered [11]. MyD88 is an adaptor protein located downstream of TLR2 signaling. Additionally, the effects of TLR2-MyD88 signaling pathway were examined in Tregs isolated from MyD88 deficient mice, and a reduction in suppressive functions of Tregs in the absence of MyD88 was reported [5, 9]. Suppressive functions of Tregs were induced by cell-contact mechanisms and secretion of TGFβ and IL10, which are immunosuppressive cytokines, without an increase in the number of Tregs [11].

Unlike TLR2, LPS indued TLR4 enhanced the suppression effect owing to the increase in FOXP3 expression in both human and murine Tregs. In fact, LPS not only increased the number of Tregs, but also increased the expression of activation markers in cells [12]. As for TLR5, the suppression capacity of CD4 + CD25+ Tregs increased as a result of TLR5 activation by flagellin [13]. TLR8, on the other hand, has been shown to abundantly express in Tregs and upon stimulation with TLR8 ligand, the suppressive function of Tregs was abolished but it had no effect on the Tregs proliferation [14]. Lastly, studies have revealed that TLR9 ligand CpG oligodeoxynucleotide induced proliferation of both effector T cells (Teff) and Tregs and partly inhibits the suppressive activity of regulatory T cells in rats. This combined effect of TLR9 ligand is likely to reinforce the adaptive immunity by not only expanding effector cells but also by mitigating the suppressive activity of regulatory T cells [15]. Taken together, multiple studies suggest that the suppressive properties of regulatory T cells with respect to their proliferation and cytokine production capacities may differ depending on the induction and the differential expression of different TLRs in regulatory T cells (Figure 1).

Figure 1.

TLR expression profile in naive Tregs and memory Tregs: Expression of TLRs in A: naive (CD3+, CD4+, CD25 high, CD45RA+, CD127low); B: memory (CD3+, CD4+, CD25 high, CD45RA-, CD127low) regulatory T cells. C: Differentially expressed genes in between two Treg populations. Expression of genes reported as TPM was normalized to β-actin (TPMgene/TPMβ-actin) for individual representation of naive and memory Tregs. Results are depicted as bar plots in GraphPad Prism.

By examining the open-source databases of immune cell-specific gene expression profiles, we evaluated the results from published literature for TLRs in Tregs and compared their expressions among all available immune cell types. TLRs did not display a Treg specific high expression across the datasets we examined [16, 17, 18]. The DICE database generated by Schmiedel et al contains RNA-seq data of 13 immune cells including naive and memory Tregs (and two ex vivo activated cell types) collected from peripheral blood mononuclear cell fraction of 91 healthy human donors. We used this dataset for a detailed search in Tregs. Expression patterns of TLRs in Tregs are shown as boxplots (Figure 1A and B). Although TLRs had relatively low expression profiles, TLR1, TLR2, TLR5, and TLR6 are the ones with the most prominent representation and differential expression in Tregs (Figure 1C and Table 1). Interestingly, TLR1, TLR2, and TLR5 had higher expression in naive Tregs than memory Tregs, whereas TLR6 expression was higher in memory Tregs than naïve Tregs. Additionally, this dataset presents the sex-biased transcripts for immune cell types. For example, TLR1 in naive Tregs were revealed as one of genes having female bias [16].

BiotypeNaive treg mean expression (TPM)Memory treg mean expression (TPM)Log 2 fold changeAdjusted p-value
TLR1Protein coding7.186.10.160.017
TLR2Protein coding14.712.272.666.90E−233
TLR5Protein coding15.466.591.126.90E−35
TLR6Protein coding1.086.31−2.786.50E−251

Table 1.

Differentially expressed toll-like receptor family members in naïve and memory tregs, p-values (https://dice-database.org/).

2.2 Membrane-bound C-type lectin (CTLs) receptors in tregs

C-type lectin receptors (CLRs) also belong to the family of pattern recognition receptors (PRRs) which recognize PAMPs and induce innate immune responses [19]. CLRs comprise a variety of receptors including selectins, collectins, proteoglycans, and lymphocyte lectins. This receptor family possesses at least one structurally homologous carbohydrate recognition domain (CRD), also known as C-type lectin-like domain (CTLD), that determines the carbohydrate specificity [20]. Based on the protein location site on the cell membrane, CLRs are categorized as transmembrane receptors and secretory receptors [21]. Upon ligand recognition by CLRs, most of them are able to induce intracellular pathways and caspase-recruitment domain-containing domain protein 9 (CARD9) pathway, which are vital, and their dysregulation or malfunctioning may result in critical infections in humans and mice [22, 23].

CLRs are expressed on antigen presenting cells (APCs), such as dendritic cells (DCs) and macrophages, and play essential roles in antigen uptake and presentation. In this regard, they are divided mainly into two subgroups: type I and type II CLRs. Two subsets of transmembrane CLRs can be classified based on their CRDs; type I and II. Type I CLRs are mannose receptor family (MR) and DEC-205, whereas type II CLRs are sialoglycoprotein receptor family, DC-associated C-type lectin 1 (dectin-1) and macrophage galactose C type lectin (MGL) [24]. MGLs are able to recognize particularly terminal α and β N-acetylglactoseamine (GalNAc or Tn) residues from filovirus, helminths, bacteria, and tumor-associated antigens in humans [25]. Human counterparts of MGL in mice are MGL1 and MGL2, which are expressed on DCs [26] and activated macrophages [27]. The potency of human MGL was shown by Napoletano et al. as an adjuvant for designing novel anticancer vaccines because MGL engagement led to the increased antigen presenting potential in DCs and enhancement in antigen-specific CD8+ cell activation [28]. Dectin-1 is another type II C-type lectin receptor, which is involved in the antifungal immunity by recognizing β-1,3-glucans in the cell wall of several pathogenic fungi. Dectin-1 is able to induce several responses including phagocytosis, through spleen tyrosine kinase (Syk)/CARD9 pathway, which results in the cytokine production [29, 30]. Besides innate immunity, dectin-1 is also capable of triggering adaptive immune responses. For instance, curdlan activated dectin-1 in DCs, skewed the T cell polarization into Th17 and Th1 CD4+ T cell subsets in mice in vitro [31]. Finally, C-type lectin receptor CD69 have been shown to control T cell development and homeostasis in mice along with miR155 as both CD69 and miRNA155 were simultaneously regulated to ensure a balanced immunity [32].

Due to lack of studies focusing on CLRs in Treg populations, we used the DICE database to evaluate the expression profiles of CLRs in naive and memory Tregs collected from PBMC fractions of healthy donors. As described in Figure 2A and B, among all CLRs with low expressions, CLEC4A (DICR) is the one with higher representation in both cell types. Even though CLEC7A (Dectin 1) had low expression in both cell types, it was the only differentially expressed gene (Figure 2C and Table 2). Taken together, CLRs did not have a notable expression profile in Tregs, thus our analysis is in agreement with previously reported data [16, 17, 18].

Figure 2.

C type lectin expression in naive Tregs and memory Tregs: Expression of CLRs in A: naive (CD3+, CD4+, CD25 high, CD45RA+, CD127low); B: memory (CD3+, CD4+, CD25 high, CD45RA-, CD127low) regulatory T cells. C: Expression of genes reported as TPM was normalized to β-actin (TPMgene/TPMβ-actin) for individual representation of naive and memory Tregs. Results are depicted as bar plots in GraphPad Prism.

BiotypeNaive treg mean expression (TPM)Memory treg mean expression (TPM)Log 2 fold changeAdjusted p-value
CLEC7AProtein coding2.052.44−0.420.031

Table 2.

Differentially expressed C type lectin receptor family members in naïve and memory tregs, p-values. (https://dice-database.org/).

2.3 Cytosolic NOD-like receptors (NLRs) in regulatory T cells

Nucleotide binding oligomerization domain (NOD)–like receptors (NLRs) are a family of cytoplasmic PRRs that are known to drive the initial innate immune responses. There are 22 NLR members in human and 34 in mice [33], and they are characterized by a C-terminal domain of leucine rich repeats (LRRs) which senses PAMPs and danger molecules (DAMPs); a central NACHT domain that facilitates NLR oligomerization; and an N-terminal signaling domain [34]. The NLR members have been classified in 5 subfamilies based on their N-terminal domain: i) NLRA (CIITA), which contains acidic transactivation domain; ii) NLRB (NAIP) subfamily having an N-terminal baculovirus inhibition of apoptosis repeat (BIR) domain; iii) NLRC subfamily that contains caspase activation and recruitment domain (CARD) and allows direct interaction of NLR family members; iv) NLRP subfamily that bears a pyrin domain (PYD); and v) NLRX subfamily that has a mitochondria-targeting sequence required for its trafficking [34]. Some of the NLR members including NLRP1, NLRP3, NLRP6, NLRP7, NLRP12, and NLRC4 are reported to assemble large multimeric protein complexes called “Inflammasomes” which regulate the activation of caspases-1 [35, 36]. The signaling pathway where the assembled inflammasome activates pro-caspase-1 into its catalytically active form is generally referred to as the canonical inflammasome whose activation requires two steps: transcription and oligomerization. The first step is regulated by innate immune signaling, primarily by TLR signaling, and/or cytokine receptors such as TNF which leads to the production of biologically inactive pro interleukin-1β (IL1β), IL18, and NLR transcription via nuclear factor-κB (NF-κB) activation. The second step leads to inflammasome oligomerization and eventually caspase-1 activation which, in turn, results in IL1β and IL18 processing and secretion [37]. Biologically active IL1β and IL18 promote inflammatory and antimicrobial responses and activate different helper T cell subsets such as Th1 and Th17 cells [38]. Although NLRs activation leads to numerous signaling cascades which subsequently initiate the appropriate immune responses including the regulation of B and T cell functions [39], studies focusing on NLRs especially in Tregs are limited. Hence, utilizing open-source datasets, we evaluated the expression patterns of NLRs in Treg populations. Firstly, to address whether there is a Treg specific NLR expression, we compared 28 and 29 cell types studied by Ota et al., and Monaco et al., respectively and showed that there is no NLR specifically expressed in Tregs [17, 18]. Based on the current database, NLRC5 and NLRP1 appear to have higher expression levels in memory Tregs among 13 immune cells included by the DICE database. Next, we examined the data obtained from this database for NLR expression by focusing on Tregs separately (Figure 3). As detailed in Figure 3A and B, most NLR family members have low expressions in both I and memory Treg populations. NLRC3, NLRC5, and NLRP1 have higher expression levels than the rest of the NLRs in both cells. Interestingly, Schmiedel et al. listed NLRP2 transcripts as sex-biased (toward females) in both Treg populations [16]. Several NLRs are detected to be differentially expressed naive and memory Tregs are compared with one another (Figure 3C and Table 3). CIITA, NOD2, NLRC5 expressions were significantlyIr in naive Tregs, while NLRP6 has a relatively higher differential expression profile than the remaining NLRs in memory Tregs. Critical roles, if any, of NLRs’ expressional diversity within and in between Treg populations might need further investigation.

Figure 3.

NLR expression profile in naive Tregs and memory Tregs: Expression of NLRs in A: naive (CD3+, CD4+, CD25 high, CD45RA+, CD127low); B: memory (CD3+, CD4+, CD25 high, CD45RA-, CD127low) regulatory T cells. C: Expression of genes reported as TPM was normalized to β-actin (TPMgene/TPMβ-actin) for individual representation of naive and memory Tregs. Results are depicted as bar plots in GraphPad Prism.

BiotypeNaive TREG mean expression (TPM)Memory treg mean expression (TPM)Log 2 fold changeAdjusted p-value
CIITAProtein coding1.616.65−3.898.50E−262
NAIPProtein coding20.7213.320.621.10E−09
NOD1Protein coding16.6126.42−0.792.90E−116
NOD2Protein coding3.111.2−2.15.90E−169
NLRC3Protein coding168237.03−0.617.40E−50
NLRC4Protein coding1.390.960.54.40E−09
NLRC5Protein coding222.01418.41−1.031.50E−81
NLRP1Protein coding272.66376.61−0.576.90E−54
NLRP2Protein coding8.3514.42−0.887.40E−08
NLRP3Protein coding1.331.81−0.597.00E−08
NLRP6Protein coding3.591.331.215.90E−17
NLRX1Protein coding6.789.8−0.652.20E−19

Table 3.

Differentially expressed NOD like receptor family members in naïve and memory tregs, p-values (https://dice-database.org/).

Apart from these, studies concentrating on NLRP3 and NOD2 roles in directing (Treg) differentiation and function demonstrated that NLRP3 negatively regulates Treg differentiation in an inflammasome-independent manner via translocation to the nucleus and subsequently interacting with Kpna2 [40]. Of note, immunoprotective roles were reported for NLRP3 inflammasome in controlling the Th1/Th17 immunity against fungal infection of pulmonary paracoccidioidomycosis by suppressing the expansion and migration of Tregs in mice [41]. In addition to NLRP3, NOD2 has been shown to get activated by muramyl dipeptide (MDP), resulting in NF-κB translocation to nucleus in primary human FOXP3+ T cells thereby protecting from death receptor Fas-mediated apoptosis [42]. Finally, MDP-stimulated migration of Tregs has been shown to suppress the Th17 cells in the lungs of influenza A virus-infected mice [43]. Together, these results suggest that the control of inflammation during fungal and viral infections is mediated by Tregs, with the contribution of NLRs.

2.4 Cytosolic retinoic acid-inducible Gene (RIG-i) like receptor

Type I interferons are proven to be indispensable during viral infections for their ability to generate an antiviral state. Although they are expressed at low levels, they are induced during the course of an infection which is detected by the presence of the foreign nucleic acids [44]. One example for such sensors is retinoic acid-inducible gene (RIG) I like receptors (RLR) whose activation results in type I interferons [45, 46]. RLRs sense viral RNA in the cytosol. The RLR family comprises three proteins: i) RIG-I; ii) melanoma differentiation-associated antigen 5 (MDA-5); and iii) laboratory of genetics and physiology 2 (LGP2) [47]. All these RLRs share a common structure including a central helicase domain responsible for ATP hydrolysis to unwind RNA and a C-terminal regulatory (CTR) domain adjacent to the helicase core. The CTR domain aids to distinguish the self RNAs from the foreign RNA fragments within the cellular environment. Added to these domains, RIG-I and MDA-5 have N-terminal caspase activation and recruitment domains (CARDs) that are required for downstream signaling through the interaction with CARDs of CARD containing adaptor proteins. Dissimilar to RIG-I and MDA5, LGP2 lacks the CARD domain. Instead, LGP2 is of importance to regulate the RIG-I and MDA5 directed antiviral responses [48, 49, 50].

To investigate the RLR expressions in Treg cells, we used the expression data from the DICE database (Figure 4). Similar to other PRRs, we did not observe a Treg specific expression of RLRs. As depicted in Figure 4A and B, RLRs have similar expression patterns among their members, DDX58 (RIG1) having a relatively higher expression trend as compared to IFIH1 (MDA5) andI8 in both naive and memory Tregs. Next, we listed the differentially expressed genes (Figure 4C and Table 4). Interestingly, Schmiedel et al. identified IFIH1 (MDA5) transcripts to have female-biased sex ratios. Data analysis indicates that all RLRs are expressed in Tregs from healthy donors [16].

Figure 4.

RIG-I expression profile in naive Tregs and memory Tregs: Expression of RLRs in A: naive (CD3+, CD4+, CD25 high, CD45RA+, CD127low); B: memory (CD3+, CD4+, CD25 high, CD45RA-, CD127low) regulatory T cells. C: Expression of genes reported as TPM was normalized to β-actin (TPMgene/TPMβ-actin) for individual representation of naive and memory Tregs. Results are depicted as bar plots in GraphPad Prism.

BiotypeNaive treg mean expression (TPM)Memory treg mean expression (TPM)Log 2 fold changeAdjusted p-value
DDX58Protein coding65.9258.750.090.026
IFIH1Protein coding29.1829.88−0.130.0012

Table 4.

Expression levels of mRNA for each RIG-I like Receptor family member naïve and memory Tregs, p-values (https://dice-database.org/).

RLRs, MDA-5, and RIG-I are ubiquitously expressed in the cytoplasm of immune cells including Tregs. Although exhaustion of Tregs following bacterial ligand treatments has been demonstrated [14, 51, 52], the impact of viral infection on Treg derived suppression remained elusive. A study by Anz et al. suggested a direct suppression mechanism through the activation of RLRs. In this particular study, regulatory and effector T cells from wild-type and MDA-5 deficient mice were cocultured and infected with encephalomyocarditis virus. Results suggested that MDA-5 deficient Tregs lost their ability to suppress the immune responses when compared with wild type counterparts during the viral infection [53].

Because IFN-beta promoter stimulator (IPS-1) is the main adaptor protein of RLR signaling [54], its influence on the RLR signaling during West Nile Virus (WNV) infection were studied with respect to Tregs using IPS-1 deficient mice. Conceivably, uncontrolled inflammatory responses including the more pronounced immune cell responses and failure in virus neutralization were identified with the lack of Tregs expansion which is a characteristic of WNV infection [55]. Moreover, Xu et al. showed the intrinsic suppression ability of RNA stimulated RIG-I in Treg differentiation in a IFN regulatory transcription factor (IRF)-3 dependent manner [56]. Another study investigated the role of RNA-unprimed RIG-I (apo RIG-I) in Treg differentiation. In contrast to the suppression of Treg differentiation by RNA ligand primed RIG-I, this study showed that apo-RIG-I maintains the Treg/Th17 cell balance [57]. Taken together, ever expanding novel findings from large datasets and state-of-the-art approaches may change the way we evaluate PRRs of Tregs in most bewildering ways.

Advertisement

3. Functionality of regulatory T cells in disease in the context of PRRs

3.1 Regulatory T cells in infections

Immune system as a whole represents a quite complex and interacting vast network of cells and biochemical signals circulating in blood and tissues. Therefore, this complexity necessitates a tight regulation. Tregs maintain the homeostasis by suppressing the immune response after the infection is resolved. As discussed earlier, Tregs can be activated by a variety of pathogens and their suppressive functions may differ depending on the pathogen and the progression of the infection. Not only pathogens, but also non-pathogenic environments with endogenous proteins are essential in regulating Treg responses. Heat shock protein gp96 is a chaperone for several TLRs including TLR4 and acts as a ligand as well [58, 59]. Tregs suppress the T cell proliferation and cytokine release to protect the host against excessive immune response. There are few aspects still being investigated, especially which receptors are expressed in Tregs and regulate Tregs during viral, bacterial, and fungal infections [60]. In this part of the chapter, we will continue to discuss the Tregs in terms of infectious and non-infectious disease conditions.

3.1.1 Regulatory T cells in bacterial infections

Helicobacter pylori is a Gram-negative bacterium that infects the stomach and is a highly contagious pathogen [61]. Currently, studies covering PRRs in the context of Tregs are rather sparse when it comes to bacterial infections. However, TLR4 inhibition following H. pylori has been shown to increase the Foxp3 expression and the CD4+CD25+Foxp3+ Treg cell numbers in the gastric mucosa and enhance the bacterial colonization. However, blocking Tregs led to the limited colonization of H. pylori, resulting in the reduced inflammatory responses. This study indicated that the crosstalk between TLR4 signaling pathway and Tregs is crucial for limiting H. pylori colonization and suppressing the inflammation of infected mice [62].

3.1.2 Regulatory T cells in fungus infections

Paracoccidioidomycosis (PCM) is an endemic disease caused by the fungus Paracoccidioides brasiliensis [63]. In PCM disease, regulation of Treg functions is mediated by PRRs such as TLRs, CLRs, and NLRs and downstream proteins like MyD88 [41, 64, 65, 66, 67]. In one study, TLR2-deficient mice had a reduced number of Tregs along with an excessive immune response, suggesting that TLR2 is required for Treg expansion to control the inflammatory response [67]. The study utilized MyD88-deficient mice to further analyze the effect of the downstream effectors of TLR2 signaling pathway in Tregs. The absence of MyD88 resulted in the impaired T cell responses and uncontrolled spread of fungal infection in the murine model of PCM infection [65]. Another study showed that Treg proliferation is decreased in WT mice compared to TLR4 deficient mice, therefore, the level of infiltration of activated T cells and macrophages into the lung increased, resulting in severe infection [64]. Dectin-1 is a CTL receptor involved in the antifungal immune response [68]. In Dectin-1-deficient mice with P. brasiliensis, low levels of activated effector/memory CD4+ and CD8+ T cells along with increased CD4 + CD25 + Foxp3+ Treg levels let the infection spread to tissues, eventually controlling the severity of the disease and causing the increased mortality in mice [66]. Besides the membrane bound Dectin-1 receptor, NLRP3 inflammasome complex mediated the activation and secretion of IL1β and IL18 have been reported in P. brasiliensis infected mice. NLRP3 deficient mice infected with P. brasiliensis had an increased Th1/Th17 immune response and reduced Treg response [41].

During candidiasis, a fungal infection mediated by Candida albicans, TLR2 is of great importance to control Treg survival. In this study, Netea et al. reported that TLR2-deficient mice, but not TLR4, had increased Th1 immune response and a reduced IL10 producing CD4 + CD25+ Treg population as compared to the WT mice infected with C. albicans. Tregs were further stimulated with a TLR2 ligand, peptidoglycan, to assess the significance of TLR2 signaling. Findings from this study highlights the importance of TLR2 signaling in the maintenance of survival, expansion and suppression capacity of Treg and IL10 production in candidiasis [69].

3.1.3 Regulatory T cells in viral infections

One of the immune system mechanisms that are used to protect the host from viral infections is the recognition of viral nucleic acids by PRRs such as TLRs and RLRs [70]. The human genome encodes 10 different TLRs, four of which are responsible for the recognition of viral genome, and these are TLR3, TLR7, TLR8, and TLR9. Interestingly, unlike other TLRs located on the outer cell membrane, they are located in the endosomal membranes and induce downstream molecules through adaptor proteins [4]. RLRs sense viral RNAs in the cytosol. Among RLRs, RIG-I, and MDA5 have RNA helicase activity which give them the ability to bind viral RNA and induce immune response [46]. It is well established in the literature that innate immune cells are activated through these PRRs as the first line of host defense against viral attacks. Activated innate immune cells then phagocytose and process the virus to present it to naive T cells in the draining lymph nodes. Primed T cells eventually differentiate into different types of helper T cells including Tregs [60].

TLR2 and TLR4 have been the focus of numerous studies which emphasized the effects on regulatory T cells in the course of viral infections. During hepatitis C virus infection, Tregs were suggested to suppress the HCV-specific antiviral responses resulting in viral persistence [71, 72]. In a different study by Zhai et al., the core protein of hepatitis C virus (HCVc) in the blood of HCV-infected patients has been shown to induce the proliferation of Tregs which subsequently hampered the CD4+ T cell proliferation and IFN-γ production. Since HCVc binds TLR2 on Kupffer cells and dendritic cells, just like the TLR2 agonist lipoteichoic acid, which leading to the similar processes, they proposed that HCVc mediated Treg expansion was TLR2 dependent [73]. In a separate study, the number of Tregs, TLR2 and TLR4 expression levels in the peripheral blood monocytes of chronic hepatitis C patients have been shown to elevate in parallel to viral load [74]. To this end, literature on TLRs seem to report consistent results with respect to Treg regulation.

Additionally, cytosolic RLRs have roles during viral infections. Amphiregulin, known as EGFR ligand, is produced mainly by Tregs in lungs during influenza A virus infection and it is important for tissue protection [75, 76, 77]. Interestingly, EGFR signaling has been suggested to suppress RIG-I signaling during viral infections [78, 79]. Thus, amphiregulin produced by regulatory T cells may reduce RIG-I signaling to increase survival during viral infections.

3.2 Regulatory T cells in autoimmune diseases

Autoimmunity can be defined as immunologic aberrations which exclusively exhibit abnormal self-antigen tolerance. PRRs can govern autoimmunity by playing pivotal roles in distinct immunological mechanisms [80, 81]. Autoimmune diseases have been associated with viral, bacterial and, more recently, fungal infections after detection by PRRs because of the reduced number of Treg cells and increased proinflammatory cytokines, such as IL17, IL22 and IL23, which drive the differentiation into CD4 Th17 T cells [82].

As we discussed previously in this chapter (Figure 1), TLRs are expressed and have functions in adaptive immune cells such as TCR alpha beta cells, TCR gamma beta T cells and regulatory T cells [83]. LPS induced TLR4 in CD4+CD25+ T cells have been shown to lead to activation and proliferation of Treg cells [12]. Although controversial, other TLRs including TLR5, TLR7, and TLR8 have been shown to express in human and murine CD4+CD45+ Tregs [11]. With regards to autoimmunity, using a cohort of MS patients who were helminth-infected or non-infected, Correale and Farez investigated the roles of retinoic acid (RA) and TLR2 in parasite mediated protection in MS patients. Helminth-activated DCs not only inhibited IL-17 and IFN-γ production via autoreactive T cells but also led to the immunoprotection which was attributed to the involvement of TLR2 and RA and the augmentation of CD4+CD25+FOXP3+ Treg cells [84].

Multiple sclerosis (MS) is a central nervous system autoimmune disease [85] which is characterized by demyelination [86]. When healthy individuals were compared to MS patients, it was found that Tregs of the healthy group displayed higher TLR2 expression. Furthermore, the PBMC samples from these two separate groups were stimulated with an agonist of TLR1/2, Pam3Cys, which lowered Treg functions and induced Th17 in MS groups samples [87]. Another example of autoimmune disease is type 1 diabetes mellitus (T1DM) that is associated with pancreatic β cell deficiency which results in abnormal sugar level [88]. High mobility-group box (HMGB) proteins have a role to induce the innate immunity by interacting with nucleic acids and recruiting them to PRRs and they engage receptors for advanced glycation end products (RAGE) [89]. Wild et al. showed that HMGB1 enhanced IL-10 levels and prolonged survival of Treg cells [90]. Furthermore, the inhibition of HMGB1 during beta cell mass turnover at an early stage in NOD mice is followed by reduced incidence of diabetes. Additionally, TLR4 and RAGE were shown to be predominant HMGB1 receptors in Treg cells and blockade of either one diminished Treg instability whilst stimulation with recombinant HMGB1 remarkably increased the amount of phosphorylated downstream targets including PI3K, Akt and mTOR in Tregs [91]. Overall, PRRs and Tregs axis in certain immune conditions has been pending to be investigated more elaborately.

3.3 Regulatory T cells in asthma and allergy

Persistent inflammation with the excessive production of cytokines by the immune cells can be harmful which is associated with numerous diseases including asthma and allergy [92]. Asthma is an inflammatory disease of airways which is linked to excessive T helper cell type-2 (Th2) immunity. Both allergic and non-allergic stimuli including house dust mites (HDM), pollens, viral infections and tobacco smoke trigger a cascade of events resulting in chronic airway inflammation which then leads to the airway hyperresponsiveness (AHR) [93]. Th2 cells in the airway release specific cytokines including IL4, IL5, IL9, and IL13; thereby promoting eosinophilic inflammation and immunoglobulin E (IgE) production which in turn, triggers the release of other inflammatory mediators, such as leukotrienes and histamines [94]. One of the hallmarks of asthma pathogenesis is the enhanced Th2 response and the inadequate differentiation and functional defects of Tregs. Baatjes et al. has reported that CD4+CD25highFoxp3+ Tregs were lower in the peripheral blood of the asthma patients than non-asthmatic individuals [95]. In vivo animal model studies have shown that IL10 and TGFβ secreted by Treg remarkably suppressed the airway inflammation and AHR [96] while blocking IL10 and TGFβ worsened the airway inflammation and AHR [97].

Several studies also revealed the involvement of PRRs, especially TLRs and NLRs in asthma susceptibility. Simpson et al. firstly discovered the upregulation of TLR2, TLR4, and pro-inflammatory cytokines, IL1β and IL18, in neutrophilic asthma [98]. Another study reported that TLR2 activation induces Treg and long-term suppression of asthma symptoms in OVA-sensitized mice [99]. Moreover, the important roles of TLR7 in alleviation of airway inflammation, promoting Th1 immune responses and reversing AHR have been shown [100]. Meng et al. reported that TLR7 stimulation suppressed eosinophilic inflammation by reducing Th2 cytokines IL4 and IL5, eotaxin and IgE in numerous animal models of asthma [101]. Yet another study added that treatment with TLR7 agonist R848 induced Treg cell-mediated suppression of asthma symptoms in OVA-sensitized and challenged mice [102].

Although limited, the involvement of inflammasome activation in asthmatic airway inflammations has been studied as well. The prolonged administration of IL1β, an inflammasome dependent cytokine, has been shown to induce AHR [103]. Also, increased levels of IL1β in the serum and BALF of asthmatic patients were decreased after glucocorticoids inhalation [104]. Significantly higher inflammasome depent IL18 levels in the serum of asthma patients were detected [105]. Moreover, Simpson et al. reported the elevated expression of the NLRP3 inflammasome in patients with neutrophilic asthma [106]. Another recent study using HDM-induced mouse models of allergic airway inflammation reported elevated expression of NLRP3, NLRC4, NLRC5, and caspase-1 genes as well as pro- IL1β levels in the lungs, while mature IL1β was not observed which suggested that inflammasome components are upregulated even if they do not form functional inflammasome complexes [107]. Unfortunately, studies as to the effects of inflammasomes on Treg functions in allergy and asthma conditions are limited. One study demonstrated that intranasal stimulation with NOD2 ligand disrupted the generation of Tregs and subsequently induced the development of eosinophil-associated airway inflammation [108]. Altogether, targeting NLRs and inflammasome components can be another potential therapeutic approach for the control of airway inflammation.

3.4 Regulatory T cells in cancer

Countless pathological conditions involve infections and tissue damage leading to chronic inflammation after the activation of PRRs. Innate immunity and PRRs in cancer initiation and progression are extensively studied because PRRs are expressed in different tumor tissues, such as lung, breast, colon, gastric cancer, and melanoma [21, 109]. The PRR activation in cancer cells can stimulate the production of many cytokines, chemokines, hormones, and vascular-promoting factors to induce the formation of an inflammatory tumor microenvironment that promotes the tumor progression [110]. The activation of PRRs on antigen presenting immune cells can induce dendritic cells, tumor-associated macrophages, and B cells for the generation of tumor-specific T cell responses. Tregs are found in tumor microenvironment and are able to suppress anti-tumor immune responses which is required for escaping immune system thereby cancer progression.

Signaling through PRRs results in robust pro-inflammatory responses by promoting antigen presenting cells and orchestrating adaptive immunity against tumor associated antigens [110]. Indeed, PRR ligands can both stimulate tumors and tumor-infiltrating immune cells to secrete cytokines and chemokines which modulate immune cell polarization and reprogramming the tumor microenvironment to reinforce innate and adaptive anti-tumor immunity [111]. Even though the roles of NLRs and RLRs in tumor immunity still largely unknown, TLRs have significant roles in stimulating DC maturation, antigen uptake and presentation, and the differentiation of CD4+ T cells. Additionally, Nyirenda et al. have reported the reversion of immunosuppressive skills of Tregs upon TLR stimulation and this is especially interesting for cancer research due to their activity in tumor microenvironment [112]. Given what we know about TLRs and their ligands currently, different TLR agonists have been used in anticancer therapies. Studies on TLR8 have demonstrated that adoptive transfer of TLR8 ligand-stimulated Treg cells reduced the tumor growth in mice [14] by reprogramming Treg glucose metabolism [113]. On the contrary, peritumoral administration of TLR5 ligand flagellin did not affect the growth of murine breast carcinoma D2F2 [114].

Several inflammasome forming NLRs including NLRP1, NLRP3, NLRP6, and NLRC4 may both have protective and detrimental roles in tumor development by their modulation of innate and adaptive immunity, apoptosis and differentiation [115]. On one hand, Janowski et al. has reported the protective role of NLRC4 in melanoma progression independent of the inflammasome components ASC and caspase-1 [116]. On the other hand, a recent study has shown that in metastatic melanoma and sarcoma models, NLRP3 inflammasome activation increased Treg population while inhibiting both NK and T-cell mediated anti-tumor immunity [117]. Additionally, recent studies demonstrated that NLRP3 inflammasome inhibitor (MCC950) reduced IL1β production and Tregs in head and neck squamous cell carcinoma mouse model [118]; and NLRP3 inhibitor (OLT1177) reduced melanoma growth and Foxp3+ cells in tumor microenvironment, and when given in combination with anti-PD-1 therapy, its efficacy increased as compared to monotherapy [119].

Even though CLRs are expressed by dendritic cells, they trigger distinct signaling pathways which induce the expression of cytokines and ultimately determine the T cell differentiation. There are several CLR agonists or antagonists that can be used as anti-cancer drugs, such as β-glucan as dectin-1 agonists [120]. With this, Osorio et al. have shown that a bacterial β-glucan, curdlan, skewed Tregs toward Th17 cells both in vitro and in vivo using 4 T1 mouse mammary tumor models [121]. Also, another study has demonstrated that LSECtin, a type II transmembrane protein, which belongs to the C-type lectin receptor superfamily inhibited the proliferation of tumor-specific effector T cells and induce more IL10 production from Treg cells [122].

In addition to these, cancer cells may mimic viral infections to activate interferon response pathway, and activation of RLR signaling in cancer cells may trigger cell death, activation of innate immune cells in tumor microenvironment or increased recruitment of adaptive immune cells into poorly immunogenic tumors [123]. RLRs could inhibit growth or induce apoptosis of different types of cancer cells upon recognition of RNA ligands. Jiang et al. has noted that intratumoral delivery of SLR14, RIG-I agonist induced strong anti-tumor immune responses through the reduction of CD4 + FoxP3+ Treg cells and induction of CD8+ T lymphocytes and NK cells [124]. In another study, high RIG-I expression in ovarian cancer was associated with increased FoxP3 expression and enriched PD-L1 and PD-1 mRNA expression [125]. Taken together, it is noteworthy that even though cancer disease and tumor microenvironment are highly heterogeneous, findings from completed and ongoing research raise the possibility of new targets for the treatment of cancer.

Advertisement

4. Conclusions

To study infectious and immune system related diseases is specifically difficult due to the genetic diversity of hosts and pathogens, the ever-changing nature of infection as it progresses, and the secession of host responses during the course of infection and the disease progression. Despite these challenges, utilization of more sophisticated, contemporary immunogenetic methods and tools such as single cell sequencing, high throughput screenings, computational modeling along with the availability of novel in vivo disease models, 3D organoid cultures perhaps lead to exciting outcomes for the long-term control of both infectious diseases and non-infectious immune system related diseases. These approaches will eventually lay the foundation of a framework to understand the interactive relationship between PRRs and regulatory T cells.

Advertisement

Acknowledgments

Authors thank ITU Scientific Research Projects Department Project No 43336 and 40713.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Kondělková K, Vokurková D, Krejsek J, Borská L, Fiala Z, Ctirad A. Regulatory T Cells (TREG) and their Roles in Immune System with Respect to Immunopathological Disorders. Facultas Medica Hradec Králové: Acta medica (Hradec Králové)/Universitas Carolina; 2010. DOI: 10.14712/18059694.2016.63
  2. 2. Mills KHG. Regulatory T cells: Friend or foe in immunity to infection? Nature Reviews Immunology. 2004;4:841-855. DOI: 10.1038/nri1485
  3. 3. Belkaid Y, Rouse BT. Natural regulatory T cells in infectious disease. Nature Immunology. 2005;6:353-360. DOI: 10.1038/ni1181
  4. 4. Kawasaki T, Kawai T. Toll-like receptor signaling pathways. Frontiers in Immunology. 2014;5:1-8. DOI: 10.3389/fimmu.2014.00461
  5. 5. Dai J, Liu B, Li Z. Regulatory T cells and Toll-like receptors: What is the missing link? International Immunopharmacology. 2009;9:528-533. DOI: 10.1016/j.intimp.2009.01.027
  6. 6. Bell JK, Mullen GED, Leifer CA, Mazzoni A, Davies DR, Segal DM. Leucine-rich repeats and pathogen recognition in Toll-like receptors. Trends in Immunology. 2003;24:528-533. DOI: 10.1016/S1471-4906(03)00242-4
  7. 7. Botos I, Segal DM, Davies DR. The structural biology of Toll-like receptors. Structure. 2011;19:447-459. DOI: 10.1016/j.str.2011.02.004
  8. 8. Sameer AS, Nissar S. Toll-Like Receptors (TLRs): Structure, functions, signaling, and role of their polymorphisms in colorectal cancer susceptibility. BioMed Research International. 2021;2021:1-14. DOI: 10.1155/2021/1157023
  9. 9. Sutmuller RPM, Garritsen A, Adema GJ. Regulatory T cells and toll-like receptors: Regulating the regulators. Annals of the Rheumatic Diseases. 2007;66(Suppl 3):iii91-iii95. DOI: 10.1136/ard.2007.078535
  10. 10. Li MO, Rudensky AY. T cell receptor signalling in the control of regulatory T cell differentiation and function. Nature Reviews Immunology. 2016;16:220-233. DOI: 10.1038/nri.2016.26
  11. 11. Van Maren WWC, Jacobs JFM, De Vries IJM, Nierkens S, Adema GJ. Toll-like receptor signalling on tregs: To suppress or not to suppress? Immunology. 2008;124:445-452. DOI: 10.1111/j.1365-2567.2008.02871.x
  12. 12. Caramalho I, Lopes-Carvalho T, Ostler D, Zelenay S, Haury M, Demengeot J. Regulatory T cells selectively express toll-like receptors and are activated by lipopolysaccharide. The Journal of Experimental Medicine. 2003;197:403-411. DOI: 10.1084/jem.20021633
  13. 13. Crellin NK, Garcia RV, Hadisfar O, Allan SE, Steiner TS, Levings MK. Human CD4 + T cells express TLR5 and its ligand flagellin enhances the suppressive capacity and expression of FOXP3 in CD4 + CD25 + T regulatory cells. Journal of Immunology. 2005:175. DOI: 10.4049/jimmunol.175.12.8051
  14. 14. Peng G, Guo Z, Kiniwa Y, Voo KS, Peng W, Fu T, et al. Immunology: Toll-like receptor 8-mediated reversal of CD4+ regulatory T cell function. Science (80-). 2005:309. DOI: 10.1126/science.1113401
  15. 15. Chiffoleau E, Heslan JM, Heslan M, Louvet C, Condamine T, Cuturi MC. TLR9 ligand enhances proliferation of rat CD4+ T cell and modulates suppressive activity mediated by CD4+ CD25+ T cell. International Immunology. 2007;19:193-201. DOI: 10.1093/intimm/dxl136
  16. 16. Schmiedel BJ, Singh D, Madrigal A, Valdovino-Gonzalez AG, White BM, Zapardiel-Gonzalo J, et al. Impact of genetic polymorphisms on human immune cell gene expression. Cell. 2018;175:1701-1715. DOI: 10.1016/j.cell.2018.10.022
  17. 17. Monaco G, Lee B, Xu W, Mustafah S, Hwang YY, Carré C, et al. RNA-Seq signatures normalized by mRNA abundance allow absolute deconvolution of human immune cell types. Cell Reports. 2019;26:1627-1640. DOI: 10.1016/j.celrep.2019.01.041
  18. 18. Ota M, Nagafuchi Y, Hatano H, Ishigaki K, Terao C, Takeshima Y, et al. Dynamic landscape of immune cell-specific gene regulation in immune-mediated diseases. Cell. 2021;184:3006-3021. DOI: 10.1016/j.cell.2021.03.056
  19. 19. Geijtenbeek TBH, Gringhuis SI. Signalling through C-type lectin receptors: Shaping immune responses. Nature Reviews Immunology. 2009;9:465-479. DOI: 10.1038/nri2569
  20. 20. Lepenies B, Lee J, Sonkaria S. Targeting C-type lectin receptors with multivalent carbohydrate ligands. Advanced Drug Delivery Reviews. 2013;65:1271-1281. DOI: 10.1016/j.addr.2013.05.007
  21. 21. Li D, Wu M. Pattern recognition receptors in health and diseases. Signal Transduction and Targeted Therapy. 2021;6:1-24. DOI: 10.1038/s41392-021-00687-0
  22. 22. Rieber N, Gazendam RP, Freeman AF, Hsu AP, Collar AL, Sugui JA, et al. Extrapulmonary Aspergillus infection in patients with CARD9 deficiency. JCI Insight. 2021;1:1-13. DOI: 10.1172/jci.insight.89890
  23. 23. Speakman EA, Dambuza IM, Salazar F, Brown GD. T cell antifungal immunity and the role of C-type lectin receptors. Trends in Immunology. 2020;41:61-76. DOI: 10.1016/j.it.2019.11.007
  24. 24. Singh SK, Streng-Ouwehand I, Litjens M, Weelij DR, García-Vallejo JJ, van Vliet SJ, et al. Characterization of murine MGL1 and MGL2 C-type lectins: Distinct glycan specificities and tumor binding properties. Molecular Immunology. 2009;46:1240-1249. DOI: 10.1016/j.molimm.2008.11.021
  25. 25. Zizzari IG, Napoletano C, Battisti F, Rahimi H, Caponnetto S, Pierelli L, et al. MGL receptor and immunity: When the ligand can make the difference. Journal of Immunology Research. 2015;2015:1-8. DOI: 10.1155/2015/450695
  26. 26. van Vliet SJ, van Liempt E, Saeland E, Aarnoudse CA, Appelmelk B, Irimura T, et al. Carbohydrate profiling reveals a distinctive role for the C-type lectin MGL in the recognition of helminth parasites and tumor antigens by dendritic cells. International Immunology. 2005;17:261-669. DOI: 10.1093/intimm/dxh246
  27. 27. Raes G, Brys L, Dahal BK, Brandt J, Grooten J, Brombacher F, et al. Macrophage galactose-type C-type lectins as novel markers for alternatively activated macrophages elicited by parasitic infections and allergic airway inflammation. Journal of Leukocyte Biology. 2005;77:321-327. DOI: 10.1189/jlb.0304212
  28. 28. Napoletano C, Zizzari IG, Rughetti A, Rahimi H, Irimura T, Clausen H, et al. Targeting of macrophage galactose-type C-type lectin (MGL) induces DC signaling Regulatory T Cells and activation. European Journal of Immunology. 2012;42:936-945. DOI: 10.1002/eji.201142086
  29. 29. Saijo S, Iwakura Y. Dectin-1 and Dectin-2 in innate immunity against fungi. International Immunology. 2011;23:467-472. DOI: 10.1093/intimm/dxr046
  30. 30. Wevers BA, Kaptein TM, Zijlstra-Willems EM, Theelen B, Boekhout T, Geijtenbeek TBH, et al. Fungal engagement of the C-type lectin mincle suppresses dectin-1-induced antifungal immunity. Cell Host & Microbe. 2014;15:494-505. DOI: 10.1016/j.chom.2014.03.008
  31. 31. LeibundGut-Landmann S, Groß O, Robinson MJ, Osorio F, Slack EC, Tsoni SV, et al. Syk- and CARD9-dependent coupling of innate immunity to the induction of T helper cells that produce interleukin 17. Nature Immunology. 2007;8:630-638. DOI: 10.1038/ni1460
  32. 32. Sánchez-Díaz R, Blanco-Dominguez R, Lasarte S, Tsilingiri K, Martín-Gayo E, Linillos-Pradillo B, et al. Thymus-derived regulatory T cell development is regulated by C-type lectin-mediated BIC/MicroRNA 155 expression. Molecular and Cellular Biology. 2017;37:1-18. DOI: 10.1128/mcb.00341-16
  33. 33. Motta V, Soares F, Sun T, Philpott DJ. Nod-like receptors: Versatile cytosolic sentinels. Physiological Reviews. 2015;95:149-178. DOI: 10.1152/physrev.00009.2014
  34. 34. Werts C, Rubino S, Ling A, Girardin SE, Philpott DJ. Nod-like receptors in intestinal homeostasis, inflammation, and cancer. Journal of Leukocyte Biology. 2011;90:9594-9627. DOI: 10.1189/jlb.0411183
  35. 35. Lamkanfi M, Dixit VM. Mechanisms and functions of inflammasomes. Cell. 2014;157:1013-1022. DOI: 10.1016/j.cell.2014.04.007
  36. 36. Davis BK, Wen H, Ting JP-Y. The inflammasome NLRs in immunity, inflammation, and associated diseases. Annual Review of Immunology. 2011;29:707-735. DOI: 10.1146/annurev-immunol-031210-101405
  37. 37. Pellegrini C, Antonioli L, Lopez-Castejon G, Blandizzi C, Fornai M. Canonical and non-canonical activation of NLRP3 inflammasome at the crossroad between immune tolerance and intestinal inflammation. Frontiers in Immunology. 2017;8:1-12. DOI: 10.3389/fimmu.2017.00036
  38. 38. Ciraci C, Janczy JR, Jain N, Haasken S, Silva CP, Benjamim CF, et al. Immune complexes indirectly suppress the generation of Th17 responses In Vivo. PLoS One. 2016;11:1-15. DOI: 10.1371/journal.pone.0151252
  39. 39. Liu D, Rhebergen AM, Eisenbarth SC. Licensing adaptive immunity by NOD-like receptors. Frontiers in Immunology. 2013;4:1-17. DOI: 10.3389/fimmu.2013.00486
  40. 40. Park SH, Ham S, Lee A, Möller A, Kim TS. NLRP3 negatively regulates Treg differentiation through Kpna2-mediated nuclear translocation. The Journal of Biological Chemistry. 2019;294:17951-17961. DOI: 10.1074/jbc.RA119.010545
  41. 41. Feriotti C, de Araújo EF, Loures FV, Costa TA da, Galdino NA de L, Zamboni DS, et al. NOD-like receptor P3 inflammasome controls protective Th1/Th17 immunity against pulmonary paracoccidioidomycosis. Frontiers in Immunology 2017;8:1-15. DOI:10.3389/fimmu.2017.00786
  42. 42. Rahman MK, Midtling EH, Svingen PA, Xiong Y, Bell MP, Tung J, et al. The pathogen recognition receptor NOD2 regulates human FOXP3 + T cell survival. Journal of Immunology. 2010;184:7247-7256. DOI: 10.4049/jimmunol.0901479
  43. 43. Egarnes B, Gosselin J. Contribution of regulatory T cells in nucleotide-binding oligomerization domain 2 response to influenza virus infection. Frontiers in Immunology. 2018;9:1-12. DOI: 10.3389/fimmu.2018.00132
  44. 44. Loo YM, Gale M. Immune signaling by RIG-I-like receptors. Immunity. 2011;34:680-692. DOI: 10.1016/j.immuni.2011.05.003
  45. 45. McNab F, Mayer-Barber K, Sher A, Wack A, O’Garra A. Type I interferons in infectious disease. Nature Reviews Immunology. 2015;15:87-103. DOI: 10.1038/nri3787
  46. 46. Rehwinkel J, Gack MU. RIG-I-like receptors: Their regulation and roles in RNA sensing. Nature Reviews Immunology. 2020;20:537-551. DOI: 10.1038/s41577-020-0288-3
  47. 47. Zou J, Chang M, Nie P, Secombes CJ. Origin and evolution of the RIG-I like RNA helicase gene family. BMC Evolutionary Biology. 2009;9:1-14. DOI: 10.1186/1471-2148-9-85
  48. 48. Childs KS, Randall RE, Goodbourn S. LGP2 plays a critical role in sensitizing mda-5 to activation by double-stranded RNA. PLoS One. 2013;8:1-8. DOI: 10.1371/journal.pone.0064202
  49. 49. Rawling DC, Pyle AM. Parts, assembly and operation of the RIG-I family of motors. Current Opinion in Structural Biology. 2014;25:25-33. DOI: 10.1016/j.sbi.2013.11.011
  50. 50. Sellati TJ, Sahay B. Cells of innate immunity: Mechanisms of activation. Pathobiology of Human Disease: A Dynamic Encyclopedia of Disease Mechanisms. 2014:258-274. DOI: 10.1016/B978-0-12-386456-7.01804-9
  51. 51. Kubo T, Hatton RD, Oliver J, Liu X, Elson CO, Weaver CT. Regulatory T cell suppression and anergy are differentially regulated by proinflammatory cytokines produced by TLR-activated dendritic cells. Journal of Immunology. 2004;173:7249-7258. DOI: 10.4049/jimmunol.173.12.7249
  52. 52. Sutmuller RPM, Den Brok MHMGM, Kramer M, Bennink EJ, Toonen LWJ, Kullberg BJ, et al. Toll-like receptor 2 controls expansion and function of regulatory T cells. The Journal of Clinical Investigation. 2006;116:485-494. DOI: 10.1172/JCI25439
  53. 53. Anz D, Koelzer VH, Moder S, Thaler R, Schwerd T, Lahl K, et al. Immunostimulatory RNA Blocks Suppression by regulatory T Cells. Journal of Immunology. 2010;184:939-946. DOI: 10.4049/jimmunol.0901245
  54. 54. Kawai T, Takahashi K, Sato S, Coban C, Kumar H, Kato H, et al. IPS-1, an adaptor triggering RIG-I- and Mda5-mediated type I interferon induction. Nature Immunology. 2005;6:981-988. DOI: 10.1038/ni1243
  55. 55. Suthar MS, Ma DY, Thomas S, Lund JM, Zhang N, Daffis S, et al. IPS-1 is essential for the control of west nile virus infection and immunity. PLoS Pathogens. 2010;6:1-15. DOI: 10.1371/journal.ppat.1000757
  56. 56. Xu P, Bailey-Bucktrout S, Xi Y, Xu D, Du D, Zhang Q , et al. Innate antiviral host defense attenuates TGF-β function through IRF3-mediated suppression of smad signaling. Molecular Cell. 2014;56:723-737. DOI: 10.1016/j.molcel.2014.11.027
  57. 57. Yang H, Guo H-Z, Li X-Y, Lin J, Zhang W, Zhao J-M, et al. Viral RNA–unprimed Rig-I restrains Stat3 activation in the modulation of regulatory T Cell/Th17 cell balance. Journal of Immunology. 2017;199:119-128. DOI: 10.4049/jimmunol.1700366
  58. 58. Vabulas RM, Wagner H, Schild H. Heat shock proteins as ligands of Toll-like receptors. Current Topics in Microbiology and Immunology. 2002;270:169-184. DOI: 10.1007/978-3-642-59430-4_11
  59. 59. Dai J, Liu B, Ngoi SM, Sun S, Vella AT, Li Z. TLR4 hyperresponsiveness via cell surface expression of heat shock protein gp96 potentiates suppressive function of regulatory T cells. Journal of Immunology. 2007;178:3219-3225. DOI: 10.4049/jimmunol.178.5.3219
  60. 60. Boer MC, Joosten SA, Ottenhoff THM. Regulatory T-cells at the interface between human host and pathogens in infectious diseases and vaccination. Frontiers in Immunology. 2015;6:1-15. DOI: 10.3389/fimmu.2015.00217
  61. 61. Hooi JKY, Lai WY, Ng WK, Suen MMY, Underwood FE, Tanyingoh D, et al. Global prevalence of helicobacter pylori infection: Systematic review and meta-analysis. Gastroenterology. 2017;153:420-429. DOI: 10.1053/j.gastro.2017.04.022
  62. 62. Gong Y, Tao L, Jing L, Liu D, Hu S, Liu W, et al. Association of TLR4 and treg in Helicobacter pylori colonization and inflammation in mice. PLoS One. 2016;11:1-16. DOI: 10.1371/journal.pone.0149629
  63. 63. Marques-da-Silva SH, Rodrigues AM, De Hoog GS, Silveira-Gomes F, De Camargo ZP. Case report: Occurrence of paracoccidioides lutzii in the amazon region: Description of two cases. The American Journal of Tropical Medicine and Hygiene. 2012;87:710-714. DOI: 10.4269/ajtmh.2012.12-0340
  64. 64. Loures FV, Pina A, Felonato M, Araújo EF, Leite KRM, Calich VLG. Toll-like receptor 4 signaling leads to severe fungal infection associated with enhanced proinflammatory immunity and impaired expansion of regulatory T cells. Infection and Immunity. 2010:78. DOI: 10.1128/IAI.01198-09
  65. 65. Loures FV, Pina A, Felonato M, Feriotti C, de Araújo EF, Calich VLG. MyD88 signaling is required for efficient innate and adaptive immune responses to Paracoccidioides brasiliensis infection. Infection and Immunity. 2011:79. DOI: 10.1128/IAI.00375-10
  66. 66. Loures FV, Araújo EF, Feriotti C, Bazan SB, Costa TA, Brown GD, et al. Dectin-1 induces M1 macrophages and prominent expansion of CD8 +IL-17+ cells in pulmonary paracoccidioidomycosis. The Journal of Infectious Diseases. 2014:210. DOI: 10.1093/infdis/jiu136
  67. 67. Loures FV, Pina A, Felonato M, Calich VLG. TLR2 is a negative regulator of Th17 cells and tissue pathology in a pulmonary model of fungal infection. Journal of Immunology. 2009:183. DOI: 10.4049/jimmunol.0801599
  68. 68. Tang J, Lin G, Langdon WY, Tao L, Zhang J. Regulation of C-type lectin receptor-mediated antifungal immunity. Frontiers in Immunology. 2018;9:1-12. DOI: 10.3389/fimmu.2018.0012
  69. 69. Netea MG, Sutmuller R, Hermann C, Van der Graaf CAA, Van der Meer JWM, van Krieken JH, et al. Toll-like receptor 2 suppresses immunity against candida albicans through induction of IL-10 and regulatory T cells. Journal of Immunology. 2004;172:3712-3718. DOI: 10.4049/jimmunol.172.6.3712
  70. 70. Hotz C, Roetzer LC, Huber T, Sailer A, Oberson A, Treinies M, et al. TLR and RLR signaling are reprogrammed in opposite directions after detection of viral infection. Journal of Immunology. 2015;195:4387-4395. DOI: 10.4049/jimmunol.1500079
  71. 71. Burton JR, Klarquist J, Im KA, Smyk-Pearson S, Golden-Mason L, Castelblanco N, et al. Prospective analysis of effector and regulatory CD4+ T cells in chronic HCV patients undergoing combination antiviral therapy. Journal of Hepatology. 2008;49:329-338. DOI: 10.1016/j.jhep.2008.05.020
  72. 72. Cabrera R, Tu Z, Xu Y, Firpi RJ, Rosen HR, Liu C, et al. An immunomodulatory role for CD4+CD25+ regulatory T lymphocytes in hepatitis C virus infection. Hepatology. 2004;40:1062-1071. DOI: 10.1002/hep.20454
  73. 73. Zhai N, Chi X, Li T, Song H, Li H, Jin X, et al. Hepatitis C virus core protein triggers expansion and activation of CD4+CD25+regulatory T cells in chronic hepatitis C patients. Cellular & Molecular Immunology. 2015;12:743-749. DOI: 10.1038/cmi.2014.119
  74. 74. Wang JP, Zhang Y, Wei X, Li J, Nan XP, Yu HT, et al. Circulating toll-like receptor (TLR) 2, TLR4, and regulatory T cells in patients with chronic hepatitis C. APMIS. 2010;118:261-270. DOI: 10.1111/j.1600-0463.2010.02586.x
  75. 75. Arpaia N, Green JA, Moltedo B, Arvey A, Hemmers S, Yuan S, et al. A distinct function of regulatory T cells in tissue protection. Cell. 2015;162:1078-1089. DOI: 10.1016/j.cell.2015.08.021
  76. 76. Burzyn D, Kuswanto W, Kolodin D, Shadrach JL, Cerletti M, Jang Y, et al. A special population of regulatory T cells potentiates muscle repair. Cell. 2013;155:1282-1295. DOI: 10.1016/j.cell.2013.10.054
  77. 77. Harb H, Benamar M, Lai PS, Contini P, Griffith JW, Crestani E, et al. Notch4 signaling limits regulatory T-cell-mediated tissue repair and promotes severe lung inflammation in viral infections. Immunity. 2021;54:1186-1199. DOI: 10.1016/j.immuni.2021.04.002
  78. 78. Oshiumi H, Miyashita M, Okamoto M, Morioka Y, Okabe M, Matsumoto M, et al. DDX60 is involved in RIG-I-dependent and independent antiviral responses, and its function is attenuated by virus-induced EGFR activation. Cell Reports. 2015;11:1193-1207. DOI: 10.1016/j.celrep.2015.04.047
  79. 79. Ueki IF, Min-Oo G, Kalinowski A, Ballon-Landa E, Lanier LL, Nadel JA, et al. Respiratory virus-induced EGFR activation suppresses IRF1-dependent interferon λ and antiviral defense in airway epithelium. The Journal of Experimental Medicine. 2013;210:1929-1936. DOI: 10.1084/jem.20121401
  80. 80. Chen L, Qi CS, Min LZ, Jun HS, Ping ZJ. NOD-like receptors in autoimmune diseases. Acta Pharmacologica Sinica. 2021. DOI: 10.1038/s41401-020-00603-2
  81. 81. Ciraci C. Physiology and pathology of autoinflammation: NOD like receptors in autoinflammation and autoimmunity. Physiology and Pathology of Immunology. 2017. DOI: 10.5772/intechopen.70484
  82. 82. Roe K. How major fungal infections can initiate severe autoimmune diseases. Microbial Pathogenesis. 2021;161:1-5. DOI: 10.1016/j.micpath.2021.105200
  83. 83. Gibson J, Gow N, Wong SYC. Expression and functions of innate pattern recognition receptors in T and B cells. Immunology, Endocrine & Metabolic Agents in Medicinal Chemistry. 2012;10:11-20. DOI: 10.2174/187152210791171304
  84. 84. Correale J, Farez MF. Parasite infections in multiple sclerosis modulate immune responses through a retinoic acid–dependent pathway. Journal of Immunology. 2013;191:3827-3837. DOI: 10.4049/jimmunol.1301110
  85. 85. Castrop F, Haslinger B, Hemmer B, Buck D. Review of the pharmacoeconomics of early treatment of multiple sclerosis using interferon beta. Neuropsychiatric Disease and Treatment. 2013;9:1339-1349. DOI: 10.2147/NDT.S33949
  86. 86. Podda G, Nyirenda M, Crooks J, Gran B. Innate immune responses in the CNS: Role of toll-like receptors, mechanisms, and therapeutic opportunities in multiple sclerosis. Journal of Neuroimmune Pharmacology. 2013. DOI: 10.1007/s11481-013-9483-3
  87. 87. Nyirenda MH, Morandi E, Vinkemeier U, Constantin-Teodosiu D, Drinkwater S, Mee M, et al. TLR2 stimulation regulates the balance between regulatory T cell and Th17 function: A novel mechanism of reduced regulatory T cell function in multiple sclerosis. Journal of Immunology. 2015;194:5761-5774. DOI: 10.4049/jimmunol.1400472
  88. 88. Katsarou A, Gudbjörnsdottir S, Rawshani A, Dabelea D, Bonifacio E, Anderson BJ, et al. Type 1 diabetes mellitus. Nature Reviews Disease Primers. 2017;3:1-17. DOI: 10.1038/nrdp.2017.16
  89. 89. Sims GP, Rowe DC, Rietdijk ST, Herbst R, Coyle AJ. HMGB1 and RAGE in inflammation and cancer. Annual Review of Immunology. 2010;28:367-388. DOI: 10.1146/annurev.immunol.021908.132603
  90. 90. Wild CA, Bergmann C, Fritz G, Schuler P, Hoffmann TK, Lotfi R, et al. HMGB1 conveys immunosuppressive characteristics on regulatory and conventional T cells. International Immunology. 2012;24:485-494. DOI: 10.1093/intimm/dxs051
  91. 91. Zhang J, Chen L, Wang F, Zou Y, Li J, Luo J, et al. Extracellular HMGB1 exacerbates autoimmune progression and recurrence of type 1 diabetes by impairing regulatory T cell stability. Diabetologia. 2020;63:987-1001. DOI: 10.1007/s00125-020-05105-8
  92. 92. Dong Z, Xiong L, Zhang W, Gibson PG, Wang T, Lu Y, et al. Holding the inflammatory system in check: TLRs and their targeted therapy in asthma. Mediators of Inflammation. 2016;2. DOI: 10.1155/2016/2180417
  93. 93. Quirt J, Hildebrand KJ, Mazza J, Noya F, Kim H. Asthma. Allergy, Asthma and Clinical Immunology. 2018. DOI: 10.1186/s13223-018-0279-0
  94. 94. Lemanske RF, Busse WW. Asthma: Clinical expression and molecular mechanisms. The Journal of Allergy and Clinical Immunology. 2010;125:S95-S102. DOI: 10.1016/j.jaci.2009.10.047
  95. 95. Baatjes AJ, Smith SG, Watson R, Howie K, Murphy D, Larché M, et al. T regulatory cell phenotypes in peripheral blood and bronchoalveolar lavage from non-asthmatic and asthmatic subjects. Clinical and Experimental Allergy. 2015;45:1654-1662. DOI: 10.1111/cea.12594
  96. 96. Fu CL, Chuang YH, Chau LY, Chiang BL. Effects of adenovirus-expressing IL-10 in alleviating airway inflammation in asthma. The Journal of Gene Medicine. 2006;8:1393-1399. DOI: 10.1002/jgm.974
  97. 97. Oh JW, Seroogy CM, Meyer EH, Akbari O, Berry G, Fathman CG, et al. CD4 T-helper cells engineered to produce IL-10 prevent allergen-induced airway hyperreactivity and inflammation. The Journal of Allergy and Clinical Immunology. 2002;110:460-468. DOI: 10.1067/mai.2002.127512
  98. 98. Simpson JL, Grissell TV, Douwes J, Scott RJ, Boyle MJ, Gibson PG. Innate immune activation in neutrophilic asthma and bronchiectasis. Thorax. 2007:62. DOI: 10.1136/thx.2006.061358
  99. 99. Nawijn MC, Motta AC, Gras R, Shirinbak S, Maazi H, van Oosterhout AJM. TLR-2 activation induces regulatory T cells and long-term suppression of asthma manifestations in mice. PLoS One. 2013;8:1-9. DOI: 10.1371/journal.pone.0055307
  100. 100. Duechs MJ, Hahn C, Benediktus E, Werner-Klein M, Braun A, Hoymann HG, et al. TLR agonist mediated suppression of allergic responses is associated with increased innate inflammation in the airways. Pulmonary Pharmacology & Therapeutics. 2011;24:203-214. DOI: 10.1016/j.pupt.2010.12.009
  101. 101. Meng L, He X, Zhu W, Yang X, Jiang C, Sun Q , et al. TLR3 and TLR7 modulate IgE production in antigen induced pulmonary inflammation via influencing IL-4 expression in immune organs. PLoS One. 2011;6:1-11. DOI: 10.1371/journal.pone.0017252
  102. 102. Pham Van L, Bardel E, Gregoire S, Vanoirbeek J, Schneider E, Dy M, et al. Treatment with the TLR7 agonist R848 induces regulatory T-cell-mediated suppression of established asthma symptoms. European Journal of Immunology. 2011;41:1992-1999. DOI: 10.1002/eji.201040914
  103. 103. Zhang Y, Xu CB, Cardell LO. Long-term exposure to IL-1β enhances Toll-IL-1 receptor-mediated inflammatory signaling in murine airway hyperresponsiveness. European Cytokine Network. 2009;20:148-156. DOI: 10.1684/ecn.2009.0156
  104. 104. Sousa AR, Trigg CJ, Lane SJ, Hawksworth R, Nakhosteen JA, Poston RN, et al. Effect of inhaled glucocorticoids on IL-1β and IL-1 receptor antagonist (IL- 1ra) expression in asthmatic bronchial epithelium. Thorax. 1997;52:407-410. DOI: 10.1136/thx.52.5.407
  105. 105. Tanaka H, Miyazaki N, Oashi K, Teramoto S, Shiratori M, Hashimoto M, et al. IL-18 might reflect disease activity in mild and moderate asthma exacerbation. The Journal of Allergy and Clinical Immunology. 2001;107:331-336. DOI: 10.1067/mai.2001.112275
  106. 106. Simpson JL, Phipps S, Baines KJ, Oreo KM, Gunawardhana L, Gibson PG. Elevated expression of the NLRP3 inflammasome in neutrophilic asthma. The European Respiratory Journal. 2014;43:1067-1076. DOI: 10.1183/09031936.00105013
  107. 107. Tan HTT, Hagner S, Ruchti F, Radzikowska U, Tan G, Altunbulakli C, et al. Tight junction, mucin, and inflammasome-related molecules are differentially expressed in eosinophilic, mixed, and neutrophilic experimental asthma in mice. Allergy Eur J Allergy Clin Immunol. 2019;74:294-307. DOI: 10.1111/all.13619
  108. 108. Duan W, Mehta AK, Magalhaes JG, Ziegler SF, Dong C, Philpott DJ, et al. Innate signals from Nod2 block respiratory tolerance and program T H2-driven allergic inflammation. The Journal of Allergy and Clinical Immunology. 2010;126:1284-1293. DOI: 10.1016/j.jaci.2010.09.021
  109. 109. Żeromski J, Kaczmarek M, Boruczkowski M, Kierepa A, Kowala-Piaskowska A, Mozer-Lisewska I. Significance and role of pattern recognition receptors in malignancy. Archivum Immunologiae et Therapiae Experimentalis. 2019;67:133-141. DOI: 10.1007/s00005-019-00540-x
  110. 110. Burn OK, Prasit KK, Hermans IF. Modulating the tumour microenvironment by intratumoural injection of pattern recognition receptor agonists. Cancers. 2020;12:1-22. DOI: 10.3390/cancers12123824
  111. 111. Bai L, Li W, Zheng W, Xu D, Chen N, Cui J. Promising targets based on pattern recognition receptors for cancer immunotherapy. Pharmacological Research. 2020;159:1-13. DOI: 10.1016/j.phrs.2020.105017
  112. 112. Nyirenda MH, Sanvito L, Darlington PJ, O’Brien K, Zhang G-X, Constantinescu CS, et al. TLR2 stimulation drives human naive and effector regulatory T cells into a Th17-like phenotype with reduced suppressive function. Journal of Immunology. 2011;187:2278-2290. DOI: 10.4049/jimmunol.1003715
  113. 113. Li L, Liu X, Sanders KL, Edwards JL, Ye J, Si F, et al. TLR8-mediated metabolic control of human treg function: A mechanistic target for cancer immunotherapy. Cell Metabolism. 2019;29:103-123. DOI: 10.1016/j.cmet.2018.09.020
  114. 114. Sfondrini L, Rossini A, Besusso D, Merlo A, Tagliabue E, Mènard S, et al. Antitumor activity of the TLR-5 ligand flagellin in mouse models of cancer. Journal of Immunology. 2006;176:6624-6630. DOI: 10.4049/jimmunol.176.11.6624
  115. 115. Di Virgilio F. The therapeutic potential of modifying inflammasomes and NOD-like receptors. Pharmacological Reviews. 2013;65:872-905. DOI: 10.1124/pr.112.006171
  116. 116. Janowski AM, Colegio OR, Hornick EE, McNiff JM, Martin MD, Badovinac VP, et al. NLRC4 suppresses melanoma tumor progression independently of inflammasome activation. The Journal of Clinical Investigation. 2016;126:3917-3928. DOI: 10.1172/JCI86953
  117. 117. Lin TY, Tsai MC, Tu W, Yeh HC, Wang SC, Huang SP, et al. Role of the NLRP3 inflammasome: Insights into cancer hallmarks. Frontiers in Immunology. 2021;11:1-15. DOI: 10.3389/fimmu.2020.610492
  118. 118. Chen L, Huang CF, Li YC, Deng WW, Mao L, Wu L, et al. Blockage of the NLRP3 inflammasome by MCC950 improves anti-tumor immune responses in head and neck squamous cell carcinoma. Cellular and Molecular Life Sciences. 2018;75:2045-2058. DOI: 10.1007/s00018-017-2720-9
  119. 119. Tengesdal IW, Menon DR, Osborne DG, Neff CP, Powers NE, Gamboni F, et al. Targeting tumor-derived NLRP3 reduces melanoma progression by limiting MDSCs expansion. Proceedings of the National Academy of Sciences of the United States of America. 2021;118:1-11. DOI: 10.1073/pnas.2000915118
  120. 120. Yan H, Kamiya T, Suabjakyong P, Tsuji NM. Targeting C-type lectin receptors for cancer immunity. Frontiers in Immunology. 2015;6:1-9. DOI: 10.3389/fimmu.2015.00408
  121. 121. Osorio F, LeibundGut-Landmann S, Lochner M, Lahl K, Sparwaser T, Eberl G, et al. DC activated via dectin-1 convert Treg into IL-17 producers. European Journal of Immunology. 2008;38:3274-3281. DOI: 10.1002/eji.200838950
  122. 122. Xu F, Liu J, Liu D, Liu B, Wang M, Hu Z, et al. LSECtin expressed on melanoma cells promotes tumor progression by inhibiting antitumor T-cell responses. Cancer Research. 2014;74:3418-3428. DOI: 10.1158/0008-5472.CAN-13-2690
  123. 123. Bufalieri F, Basili I, Di Marcotullio L, Infante P. Harnessing the activation of RIG-I like receptors to inhibit glioblastoma tumorigenesis. Frontiers in Molecular Neuroscience. 2021;14:1-9. DOI: 10.3389/fnmol.2021.710171
  124. 124. Jiang X, Muthusamy V, Fedorova O, Kong Y, Kim DJ, Bosenberg M, et al. Intratumoral delivery of RIG-I agonist SLR14 induces robust antitumor responses. The Journal of Experimental Medicine. 2019;216:2854-2868. DOI: 10.1084/jem.20190801
  125. 125. Wolf D, Fiegl H, Zeimet AG, Wieser V, Marth C, Sprung S, et al. High RIG-I expression in ovarian cancer associates with an immune-escape signature and poor clinical outcome. International Journal of Cancer. 2020;146. DOI: 10.1002/ijc.32818

Written By

Ece Oylumlu, Goksu Uzel, Lubeyne Durmus, Meric Tas, Damla Gunes and Ceren Ciraci

Submitted: 19 February 2022 Reviewed: 03 June 2022 Published: 07 July 2022