Abstract
The ability of the innate and adaptive immune systems to communicate with each other is central to protective immune responses and maintenance of host health. Myeloid cells of the innate immune system are able to sense microbial ligands, perturbations in cellular homeostasis, and virulence factors, thereby allowing them to relay distinct pathogen-specific information to naïve T cells in the form of pathogen-derived peptides and a unique cytokine milieu. Once primed, effector T helper cells produce lineage-defining cytokines to help combat the original pathogen, and a subset of these cells persist as memory or effector-memory populations. These memory T cells then play a dual role in host protection by not only responding rapidly to reinfection, but by also directly instructing myeloid cells to express licensing cytokines. This means there is a bi-directional flow of information first from the innate to the adaptive immune system, and then from the adaptive back to innate immune system. Here, we focus on how signals, first from pathogens and then from primed effector and memory T cells, are integrated by myeloid cells and its consequences for protective immunity or systemic inflammation.
Keywords: dendritic cells, IL-1R, IL-1 family of cytokines, TNF superfamily, auto-immunity, Memory T cells, Th17 cells, Systemic inflammation
The ability of the innate and adaptive immune systems to communicate with each other is central to protective immune responses and maintenance of host health. Myeloid cells of the innate immune system are able to sense microbial ligands, perturbations in cellular homeostasis, and virulence factors, thereby allowing them to relay distinct pathogen-specific information to naïve T cells in the form of pathogen-derived peptides and a unique cytokine milieu. Once primed, effector T helper cells produce lineage-defining cytokines to help combat the original pathogen, and a subset of these cells persist as memory or effector-memory populations. These memory T cells then play a dual role in host protection by not only responding rapidly to reinfection, but by also directly instructing myeloid cells to express licensing cytokines. This means there is a bi-directional flow of information first from the innate to the adaptive immune system, and then from the adaptive back to innate immune system. Here, we focus on how signals, first from pathogens and then from primed effector and memory T cells, are integrated by myeloid cells and its consequences for protective immunity or systemic inflammation.
Introduction
Cells of the immune system constantly interact with and communicate amongst themselves to coordinate responses against invading pathogens. A prime example of this is when myeloid cells, following detection of invading microbes via pattern recognition receptors (PRRs) [1], present pathogen specific information to naïve T cells [2–4]. This process requires myeloid cells, particularly dendritic cells (DCs), to integrate complex pathogen-derived cues [5, 6]. In addition to presentation of pathogen-derived peptides and upregulation of costimulatory molecules, these signals lead to the production of priming cytokines – thus creating a unique cytokine milieu which influences primed T cells to differentiate into the appropriate T helper (Th) cell subset(s) [7, 8]. Pathogen-derived cues can be in the form of microbial structures [5], metabolites [9], and even certain pathogenic behaviors such as invasion, attachment, or production of virulence factors [10–14]. Importantly, the net outcome of the myeloid cell response is dependent on the nature of receptors activated, meaning that unique responses can be induced by different types of pathogens. This innate-to-adaptive relay of information has been broadly termed “innate control of adaptive immunity” and remains a fundamental principle governing our understanding of protective immunity [1, 7, 8].
Following resolution of infection, effector memory T cells (Tem) persist in the host to enable rapid protection in the event of reinfection [15–19]. Reactivation of Tem was initially thought to be independent of original priming signals [20, 21], however recent work has found that Tem continue to rely on APC-derived cues for sustained and optimal effector function [22–24]. In addition to costimulation, IL-1 family cytokines were found to “license” effector cytokine production by Tem following reactivation by cognate antigens [24]. It is critical to note here that both priming and licensing cytokines share a dependency on MyD88 signaling. While myeloid cell intrinsic MyD88, downstream of TLRs, is critical for production of priming cytokines [25], Tem intrinsic MyD88 is integral to their ability to sense IL-1 family of cytokines [26]. This evolutionarily conserved usage of MyD88-dependent signaling in regulating the life of a T cell, spanning from initial priming to memory reactivation, is a remarkable feature of the innate-adaptive cross talk.
The premise behind innate control of adaptive immunity means that, by design, priming cytokines are produced downstream of PRR signaling [1]. While PRR-dependent production of IL-1β aids T cell priming and clonal expansion [27, 28], this cytokine continues to play a critical role in Tem reactivation by licensing the production of their effector cytokines. Interestingly, in the context of Tem reactivation, myeloid production of IL-1β is completely independent of PRR activation, but instead requires TNF receptor super family (TNFRSF) signaling [29]. As Tem were the source for TNFRSF ligands, this means that myeloid cells can sense presence of pathogens (through PRRs) or presence of memory T cells (through TNFRSFs) leading to production of priming or licensing cytokines, respectively. Although beneficial for host protection, licensing cytokines can also have detrimental consequences particularly in the context of T cell mediated autoimmunity [29, 30]. Here, we will focus on how myeloid cells integrate complex cues first from pathogens and then from Tem, and the costs as well as benefits associated with this bidirectional communication network.
Innate recognition of pathogen: signal integration
The concept of innate control of adaptive immunity was proposed in 1989 by Charles Janeway Jr. [1] and has since expanded to encompass sensing of conserved microbial ligands as well as virulence factors [7, 8, 31, 32]. PRR activation relays pathogen-specific information to myeloid cells, including overt pathogenic class, subcellular localization, and life-stage of the microbe [14, 32, 33]. In general, adaptors act downstream of multiple receptors and converge on a defined set of transcription factors [7, 33, 34]. While much is known about signaling pathways downstream of individual PRRs, complex pathogens rarely activate PRRs in isolation. Complexity in pathogen-induced responses therefore arises from the integration of signals downstream of multiple receptors (Figure 1).
Figure 1:

Integration of PRR signaling. Sensing of extracellular or endosomal pathogens by TLRs leads to the recruitment of TLR specific adaptor proteins. Usage of MyD88 by cell-surface TLRs leads to NF-κB activation. Endosomal TLRs that signal through MyD88 predominately activate IRF7, while usage of TRIF by TLR4 and TLR3, in the endosomes, activates IRF3. Intracellular pathogens can be sensed via microbial lipids (NOD), or by nucleic acids exposed during replication (RIG-I, MDA5, cGAS). Adaptors for these receptors are found anchored in the mitochondria (MAVS) or endoplasmic reticulum (ER) (STING). Stimulation through NOD1/2 leads to NF-κB and MAPK activation. STING signaling leads to the transcriptional activation of IRF3, while MAVS can activate both IRF3/7. Pathogen disruption of normal cellular homeostatic processes can also be sensed by host cells. Usage of host transcriptional machinery by pathogens can lead to ER stress, detected by ATF6, PERK, or IRE-1. This leads to the activation of ATF4/6 or XBP1 which results in abrogation of protein synthesis. Perturbations in host ubiquitin homeostasis triggers NOX2-dependent production of reactive oxygen species (ROS). Amino acid scarcity can be sensed through GCN2, leading to phosphorylation of eIF2 and activation of ATF4. Integration of these microbial recognition and homeostatic sensing pathways can occur at either the level of signaling or transcription, leading to responses which are distinct from the sum of their parts.
Signal integration can occur within members of the same or different PRR family [35, 36]. Simultaneous signaling of Toll-like receptor (TLR) 3 or TLR4 with TLR3, TLR7, or TLR9 leads to synergistic induction of TNF, IL-6, and IL-12 [35, 37–39]. This is due to the integration of MyD88 and TRIF signaling pathways, whereby activation of transcription factors NF-κB, c-Jun, and IRF5 are sustained [37, 39, 40]. In vivo, this synergy is beneficial for robust cytokine production and is critical for protection against MCMV, M. tuberculosis, and T. cruzi [41]. Certain viruses have also evolved to exploit this synergy as a method to enhance viral spreading [42].
PRR members from different families can also synergize, as is the case of TLRs and NOD1/2 or NOD-like receptors (NLRs) [43]. Simultaneous stimulation of NOD1/2 and TLR2 or TLR4 leads to the synergistic induction of proinflammatory cytokines and myeloid cell maturation markers [44–46]. Mechanistically, TLR and NOD signaling converges at the level of TAK1 and NEMO, leading to the rapid degradation of IkBα [47]. Further work suggests that NOD/TLR synergy could also be due to the receptors’ differential induction of TNF gene transcription or translation, respectively [48]. NOD/TLR synergy is one mechanism behind systemic inflammation seen during sepsis [49, 50], and antagonists are currently being developed as potential therapeutic options [51]. Signal integration between TLRs and NLRs is fundamentally different from NOD/TLR synergy, as sensing of both extracellular and intracellular virulence factors leads to assembly of a distinct signaling complex called inflammasome [52]. In the case of inflammasome, TLR signals act as a “priming” step allowing for the transcriptional upregulation of NF-κB targets [53, 54], while NLR stimulation triggers “activation” and assembly of the inflammasome with procaspase-1 [52, 55]. Priming-independent inflammasome activation results from simultaneous TLR and NLR stimulation, whereby IRAK1 coordinates rapid secretion of pre-synthesized proIL-18 [56, 57]. This two-step process leading to the production of IL-1β and IL-18 acts as a safeguard against unnecessary production of such tissue-damaging cytokines and ensures that inflammasome is only activated when specific virulent pathogens are detected [58, 59].
Virulence factors perturbing cellular homeostasis can also integrate with microbial cues [31]. Pathogens that rely on host translational machinery, for example, often activate host unfolded protein response (UPR) [60]. UPR directly activates MAPK and NF-κΒ, however simultaneous PRR signaling synergizes with these signals [60, 61]. Multiple pathogens also modulate host ubiquitination machinery, and subsequent ubiquitin homeostasis, to evade or exploit normal immune responses [62–64]. Perturbations in ubiquitin homeostasis were recently found to result in ROS production by macrophages, which potentiated TLR signaling [65]. Accumulation of lipid byproducts [66], or limitations in availability of nutrients [67, 68] and amino acids [69, 70] can also modulate innate immune responses [71]. While signal integration between PRRs and sensors of cellular homeostasis directly influence outcomes of innate immunity, whether they are also important for generating tailored adaptive immune responses against specific pathogens is an area of immense interest.
Dendritic cells as a bridge to adaptive immunity
The innate immune system achieves functional diversity through the division of labor across multiple specialized cells. While macrophages are well suited for rapid pathogen responses [72], type 1 and type 2 conventional DCs (cDC1s, cDC2s) are critical for priming naïve T cells [73–76], and are therefore the predominant node of integration for many pathogen-derived signals. While numerous myeloid subsets, including neutrophils and macrophages, utilize mechanisms of cell death to provide protection against pathogens [77–79], untimely death of cDCs can have negative consequences for the host if adaptive immune responses are not primed [80]. In fact, a virulence strategy utilized by measles and hepatitis C is to trigger premature cDC death [81, 82], effectively blunting priming and Tem development. One method utilized by DCs to avoid premature death is upregulation of c-FLIP during maturation [83], rendering mature DCs resistant to FasL-mediated apoptosis [84–87]. Recent work has also demonstrated that cDCs are particularly resistant to inflammatory death pathways [88–90]. Specifically, cDCs suppress inflammasome activation and pyroptosis, thereby allowing for priming of pathogen-specific T cell responses [89]. Other studies have shown that certain phagocytes can separate inflammasome activation from pyroptosis, allowing for the cell to enter a state of hyperactivation resulting in sustained secretion of IL-1β [91, 92]. While premature cDC death is detrimental to protective immune responses, it is important to note that cDC death is a natural step in limiting the overactivation of lymphocytes [93–95].
The reliance of the immune system on cDCs in eliciting adaptive immune responses is a powerful driving force for their divergence from other myeloid lineages. In fact, the ability of cDCs to suppress inflammasome activation and pyroptosis relies on expression of lineage-defining transcription factors IRF4 and IRF8 [89], suggesting that this feature is intimately intertwined with cDC development itself. Other functional implications of IRF4 and IRF8 expression in cDCs have been reported [96–100]. Although sensing of conserved microbial ligands and virulence factors can lead to qualitatively different responses from macrophages, cDCs’ resistance to specific virulence outcomes preserves their ability to prime T cells against both virulent as well as non-virulent pathogens. This division of labor and cellular segregation of function between macrophages and cDCs is integral for protective immune responses against microbes.
Innate cytokines as drivers of adaptive immunity
Signal integration at the subcellular level and diversity at the cellular level results in the production of a specific cytokine milieu which shapes adaptive immune responses. The identity of priming cytokines leading to differentiation of various Th lineages have been reviewed in depth elsewhere [8, 101, 102]. Importantly, these cues can be derived from myeloid [7, 8], stromal [103, 104], or epithelial origins [105, 106] meaning that T cells can integrate numerous signals necessary for proper effector differentiation. Subsequently, the resulting effector Th cell populations are now poised to neutralize the original invading pathogen. Following clearance of pathogen, a subset of memory T cells persists in secondary lymphoid organs (central memory T cell) or in the periphery (Tem) [107]. Should Tem see cognate peptide again, they are capable of rapidly proliferating and secreting effector cytokines to quickly combat reinfection [17, 18, 108, 109]. While reactivation is largely independent of original priming signals, recent work has demonstrated that Tem reactivation is still enhanced by co-stimulation and innate cytokines provided by APCs [20, 24, 107, 110]. Interestingly, these innate cytokines which we refer to as “licensing” cytokines are distinct from the original priming cytokines and are largely composed of IL-1 family members [24, 111–115].
Despite their different identities, priming and licensing cytokines share conserved dependency on MyD88 signaling. During priming, myeloid-intrinsic MyD88 links TLR ligation to downstream NF-κB activation, resulting in induction of T cell priming cytokines [116]. On the other hand, IL-1 family of cytokines signal through Tem intrinsic MyD88 to license their effector function [25, 117, 118]. The importance of MyD88 signaling downstream of IL-1R has previously been implicated in Tem responses [119, 120], presumably partially due to their lack of response to licensing cytokines. While IL-1β regulates effector cytokine production across all Th lineages, its ability to control murine and human Th17 cytokine production is most striking [24, 121]. We hypothesize that other IL-1 family members, like IL-18 for Th1 and IL-33 for Th2, play similar and more lineage specific roles in amplifying effector function of Tem cells. Whether these cytokines are made by myeloid cells in a MyD88-independent fashion remains an open and exciting area of investigation.
Adaptive instruction of innate immunity
In addition to sensing various pathogenic signals, myeloid cells can also sense cues provided by interacting CD4 T cells (Figure 2). The role of this communication has been suggested to be important during T cell priming where CD40 signaling in myeloid cells enhances their production of priming cytokines, although in these studies it is unclear if CD40L expression by recently activated naïve or newly generated effector T cells engaged CD40 on myeloid cells [122–124]. Upregulation of CD40L by naïve T cells is indeed dependent on their activation [125, 126], meaning that APCs must first be activated (to upregulate pMHC and costimulatory molecules). Therefore, PRR signaling is still a critical first step for APC activation during T cell priming, while TNFRSF signals originating from newly activated T cells may amplify this response [76].
Figure 2:

Crosstalk between innate and adaptive immune cells. During T cell priming, APCs provide three distinct signals to naïve T cells: pathogen-derived peptide on the surface of MHCII (1), costimulatory molecules (2), and priming cytokine production (3). These three signals are PRR-dependent. Newly activated T cells upregulate TNFRSF ligands including CD40L, which signal back to the APC to amplify cytokine production. Following clearance of pathogen, effector memory T cells (Tem) retain expression of TNFRSF ligands. During reactivation, in addition to cognate MHC-TCR engagement (1*), Tem expression of TNFα and FasL (2*) leads to myeloid cell transcription and cleavage of proIL-1β (3*), respectively. This adaptive instruction of innate immunity leads to production of licensing cytokines which are necessary for optimal Tem effector function, but autoreactive T cell engagement of this pathway can also lead to tissue damage and systemic inflammation.
In contrast to naïve T cell priming, Tem reactivation and function are largely independent of innate immune PRR activation [19, 21, 127]. Evolutionarily, this is a sound strategy as Tem have already passed rigorous checkpoints during priming. However, even though PRR activation is not necessary, Tem continue to rely on myeloid-derived licensing cytokines [24]. A mechanism of PRR-independent IL-1β production was recently described, where Tem expression of TNFRSF ligands induces proIL-1β synthesis and cleavage in interacting myeloid cells [29]. This is consistent with reports of rapid upregulation of TNFRSF ligands by Tem following TCR ligation [121, 122, 125, 128]. Together, this suggests that while myeloid cells sense microbial signals through PRRs to prime T cells, they can also sense the presence of pre-existing memory through TNFRSF. Tem essentially appear to replace microbial signals, thereby driving PRR-independent activation of the innate immune system. While PRR-independent production of IL-1β was described to occur for Tem across all Th lineages [29], it is important to note that lineage specific usage of other TNFRSF ligands has been reported [129–133]. It remains an intriguing possibility that lineage specific usage of TNFRSF signaling may provide further specification during adaptive instruction of innate immunity. Invariant NKT cells also utilize FasL expression to induce myeloid production of inflammasome-independent IL-1β, suggesting that TNFRSF-mediated instruction of myeloid cells may play a meaningful role in additional cellular contexts [134].
The physiological significance of Tem instruction of myeloid cells is that protective immunity becomes uncoupled from direct microbial recognition. This is an advantageous strategy, should the pathogen inhibit PRR signaling [135–139]. An inherent cost of this uncoupling, however, is that autoreactive Tem interacting with self-peptide(s) expressed by myeloid cells can trigger widespread production of licensing cytokines, leading to systemic inflammation [29]. In fact, numerous autoimmune diseases including rheumatoid arthritis and psoriasis are linked to pathological IL-1β production, and IL-1β blockade can ameliorate disease symptoms [140]. As there is not a clear link between inflammasome activation and many T cell driven autoimmune diseases [141], we predict that adaptive instruction of innate immune cells via engagement of TNFRSF members is a likely cause of pathology and systemic inflammation during T cell mediated autoimmunity.
Concluding remarks
When studying the interplay between innate and adaptive immune responses, much of the focus has previously been on the relay of information from innate to adaptive immune cells. While we have developed a broad understanding of the inflammatory outcomes of simultaneous engagement of multiple PRRs, the impact of such signal integration on generating tailored adaptive immune responses is not entirely clear. The influence of adaptive immunity on innate immune responses has largely been considered through their production of effector cytokines and chemokines to mobilize innate cells. Here, we have outlined how adaptive instruction of innate immunity, facilitated by cognate interaction between Tem and myeloid cells, makes a critical contribution to this bi-directional communication underlying protective immunity. While PRR signaling is necessary for DC activation during initial T cell priming, here we propose that TNFRSF signaling, initiated by Tem cells, replaces PRR signaling to activate the innate immune system.
Highlights.
Myeloid cells sense and integrate numerous pathogen-derived signals leading to priming of a tailored Th cell response.
Dendritic cells are a critical node for pathogen-derived signal integration, and are resistant to pathogen-induced inflammatory cell death.
Effector CD4 T cells instruct interacting myeloid cells to produce licensing cytokines which support their optimal reactivation.
Footnotes
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 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.
Declaration of Interests
None.
References
References of special interest*
References of outstanding interest**
- [1].Janeway CA Jr., Approaching the asymptote? Evolution and revolution in immunology. Cold Spring Harb Symp Quant Biol, 1989. 54 Pt 1: p. 1–13. [DOI] [PubMed] [Google Scholar]
- [2].Romani N, et al. Presentation of exogenous protein antigens by dendritic cells to T cell clones. Intact protein is presented best by immature, epidermal Langerhans cells. J Exp Med, 1989. 169(3): p. 1169–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Steinman RM and Witmer MD, Lymphoid dendritic cells are potent stimulators of the primary mixed leukocyte reaction in mice. Proc Natl Acad Sci U S A, 1978. 75(10): p. 5132–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Merad M, et al. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol, 2013. 31: p. 563–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Janeway CA Jr. and Medzhitov R, Innate immune recognition. Annu Rev Immunol, 2002. 20: p. 197–216. [DOI] [PubMed] [Google Scholar]
- [6].Gao Y, et al. Transcriptional profiling identifies caspase-1 as a T cell-intrinsic regulator of Th17 differentiation. J Exp Med, 2020. 217(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Iwasaki A and Medzhitov R, Control of adaptive immunity by the innate immune system. Nat Immunol, 2015. 16(4): p. 343–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Jain A and Pasare C, Innate Control of Adaptive Immunity: Beyond the Three-Signal Paradigm. J Immunol, 2017. 198(10): p. 3791–3800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Honda K and Littman DR, The microbiome in infectious disease and inflammation. Annu Rev Immunol, 2012. 30: p. 759–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Atarashi K, et al. Th17 Cell Induction by Adhesion of Microbes to Intestinal Epithelial Cells. Cell, 2015. 163(2): p. 367–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Hajam IA, et al. Bacterial flagellin-a potent immunomodulatory agent. Exp Mol Med, 2017. 49(9): p. e373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Keestra AM, et al. Manipulation of small Rho GTPases is a pathogen-induced process detected by NOD1. Nature, 2013. 496(7444): p. 233–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Sellin ME, et al. Epithelium-intrinsic NAIP/NLRC4 inflammasome drives infected enterocyte expulsion to restrict Salmonella replication in the intestinal mucosa. Cell Host Microbe, 2014. 16(2): p. 237–248. [DOI] [PubMed] [Google Scholar]
- [14].Blander JM and Sander LE, Beyond pattern recognition: five immune checkpoints for scaling the microbial threat. Nat Rev Immunol, 2012. 12(3): p. 215–25. [DOI] [PubMed] [Google Scholar]
- [15].Ahmed R and Gray D, Immunological memory and protective immunity: understanding their relation. Science, 1996. 272(5258): p. 54–60. [DOI] [PubMed] [Google Scholar]
- [16].Cui W and Kaech SM, Generation of effector CD8+ T cells and their conversion to memory T cells. Immunol Rev, 2010. 236: p. 151–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Rogers PR, Dubey C, and Swain SL, Qualitative changes accompany memory T cell generation: faster, more effective responses at lower doses of antigen. J Immunol, 2000. 164(5): p. 2338–46. [DOI] [PubMed] [Google Scholar]
- [18].MacLeod MK, Kappler JW, and Marrack P, Memory CD4 T cells: generation, reactivation and reassignment. Immunology, 2010. 130(1): p. 10–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Chandok MR, et al. A biochemical signature for rapid recall of memory CD4 T cells. J Immunol, 2007. 179(6): p. 3689–98. [DOI] [PubMed] [Google Scholar]
- [20].London CA, Lodge MP, and Abbas AK, Functional responses and costimulator dependence of memory CD4+ T cells. J Immunol, 2000. 164(1): p. 265–72. [DOI] [PubMed] [Google Scholar]
- [21].Pasare C and Medzhitov R, Toll-dependent control mechanisms of CD4 T cell activation. Immunity, 2004. 21(5): p. 733–41. [DOI] [PubMed] [Google Scholar]
- [22].Ndejembi MP, et al. Control of memory CD4 T cell recall by the CD28/B7 costimulatory pathway. J Immunol, 2006. 177(11): p. 7698–706. [DOI] [PubMed] [Google Scholar]
- [23].MacLeod M, et al. CD4 memory T cells survive and proliferate but fail to differentiate in the absence of CD40. J Exp Med, 2006. 203(4): p. 897–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Jain A, et al. T cell-intrinsic IL-1R signaling licenses effector cytokine production by memory CD4 T cells. Nat Commun, 2018. 9(1): p. 3185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Medzhitov R, et al. MyD88 is an adaptor protein in the hToll/IL-1 receptor family signaling pathways. Mol Cell, 1998. 2(2): p. 253–8. [DOI] [PubMed] [Google Scholar]
- [26].Warner N and Nunez G, MyD88: a critical adaptor protein in innate immunity signal transduction. J Immunol, 2013. 190(1): p. 3–4. [DOI] [PubMed] [Google Scholar]
- [27].Ben-Sasson SZ, et al. IL-1 acts directly on CD4 Tcells to enhance their antigen-driven expansion and differentiation. Proc Natl Acad Sci U S A, 2009. 106(17): p. 7119–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Guo L, et al. IL-1 family members and STAT activators induce cytokine production by Th2, Th17, and Th1 cells. Proc Natl Acad Sci U S A, 2009. 106(32): p. 13463–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29]**.Jain A, et al. T cells instruct myeloid cells to produce inflammasome-independent IL-1beta and cause autoimmunity. Nat Immunol, 2020. 21(1): p. 65–74. [DOI] [PMC free article] [PubMed] [Google Scholar]; PRR-independent production of IL-1b by myeloid cells was found to occur during Tem interaction via TNFRSF signaling.
- [30].Gabay C, Lamacchia C, and Palmer G, IL-1 pathways in inflammation and human diseases. Nat Rev Rheumatol, 2010. 6(4): p. 232–41. [DOI] [PubMed] [Google Scholar]
- [31].Chovatiya R and Medzhitov R, Stress, inflammation, and defense of homeostasis. Mol Cell, 2014. 54(2): p. 281–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Vance RE, Isberg RR, and Portnoy DA, Patterns of pathogenesis: discrimination of pathogenic and nonpathogenic microbes by the innate immune system. Cell Host Microbe, 2009. 6(1): p. 10–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Brubaker SW, et al. Innate immune pattern recognition: a cell biological perspective. Annu Rev Immunol, 2015. 33: p. 257–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34]*.Pope SD and Medzhitov R, Emerging Principles of Gene Expression Programs and Their Regulation. Mol Cell, 2018. 71(3): p. 389–397. [DOI] [PubMed] [Google Scholar]; Defining a framework for understanding complex transcriptional programs.
- [35].Lin B, Dutta B, and Fraser IDC, Systematic Investigation of Multi-TLR Sensing Identifies Regulators of Sustained Gene Activation in Macrophages. Cell Syst, 2017. 5(1): p. 25–37 e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36]**.Pandey S, et al. Pairwise Stimulations of Pathogen-Sensing Pathways Predict Immune Responses to Multi-adjuvant Combinations. Cell Syst, 2020. 11(5): p. 495–508 e10. [DOI] [PMC free article] [PubMed] [Google Scholar]; By performing pairwise in vitro stimulations, authors model and predict outcomes from more complex stimulations.
- [37].Napolitani G, et al. Selected Toll-like receptor agonist combinations synergistically trigger a T helper type 1-polarizing program in dendritic cells. Nat Immunol, 2005. 6(8): p. 769–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Bagchi A, et al. MyD88-dependent and MyD88-independent pathways in synergy, priming, and tolerance between TLR agonists. J Immunol, 2007. 178(2): p. 1164–71. [DOI] [PubMed] [Google Scholar]
- [39].Makela SM, et al. Multiple signaling pathways contribute to synergistic TLR ligand-dependent cytokine gene expression in human monocyte-derived macrophages and dendritic cells. J Leukoc Biol, 2009. 85(4): p. 664–72. [DOI] [PubMed] [Google Scholar]
- [40].Ouyang X, et al. Cooperation between MyD88 and TRIF pathways in TLR synergy via IRF5 activation. Biochem Biophys Res Commun, 2007. 354(4): p. 1045–51. [DOI] [PubMed] [Google Scholar]
- [41].Trinchieri G and Sher A, Cooperation of Toll-like receptor signals in innate immune defence. Nat Rev Immunol, 2007. 7(3): p. 179–90. [DOI] [PubMed] [Google Scholar]
- [42].Wang T, et al. Toll-like receptor 3 mediates West Nile virus entry into the brain causing lethal encephalitis. Nat Med, 2004. 10(12): p. 1366–73. [DOI] [PubMed] [Google Scholar]
- [43].Pashenkov MV, et al. Synergistic interactions between NOD receptors and TLRs: Mechanisms and clinical implications. J Leukoc Biol, 2019. 105(4): p. 669–680. [DOI] [PubMed] [Google Scholar]
- [44].van Heel DA, et al. Synergistic enhancement of Toll-like receptor responses by NOD1 activation. Eur J Immunol, 2005. 35(8): p. 2471–6. [DOI] [PubMed] [Google Scholar]
- [45].Fritz JH, et al. Synergistic stimulation of human monocytes and dendritic cells by Toll-like receptor 4 and NOD1- and NOD2-activating agonists. Eur J Immunol, 2005. 35(8): p. 2459–70. [DOI] [PubMed] [Google Scholar]
- [46].Philpott DJ and Girardin SE, The role of Toll-like receptors and Nod proteins in bacterial infection. Mol Immunol, 2004. 41(11): p. 1099–108. [DOI] [PubMed] [Google Scholar]
- [47].Abbott DW, et al. Coordinated regulation of Toll-like receptor and NOD2 signaling by K63-linked polyubiquitin chains. Mol Cell Biol, 2007. 27(17): p. 6012–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Wolfert MA, et al. The origin of the synergistic effect of muramyl dipeptide with endotoxin and peptidoglycan. J Biol Chem, 2002. 277(42): p. 39179–86. [DOI] [PubMed] [Google Scholar]
- [49].Takada H and Galanos C, Enhancement of endotoxin lethality and generation of anaphylactoid reactions by lipopolysaccharides in muramyl-dipeptide-treated mice. Infect Immun, 1987. 55(2): p. 409–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Murch O, et al. Muramyl dipeptide enhances the response to endotoxin to cause multiple organ injury in the anesthetized rat. Shock, 2008. 29(3): p. 388–94. [DOI] [PubMed] [Google Scholar]
- [51].Opal SM, et al. Effect of eritoran, an antagonist of MD2-TLR4, on mortality in patients with severe sepsis: the ACCESS randomized trial. JAMA, 2013. 309(11): p. 1154–62. [DOI] [PubMed] [Google Scholar]
- [52].Martinon F, Burns K, and Tschopp J, The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell, 2002. 10(2): p. 417–26. [DOI] [PubMed] [Google Scholar]
- [53].Bauernfeind FG, et al. Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol, 2009. 183(2): p. 787–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].He Y, Hara H, and Nunez G, Mechanism and Regulation of NLRP3 Inflammasome Activation. Trends Biochem Sci, 2016. 41(12): p. 1012–1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Martinon F, Mayor A, and Tschopp J, The inflammasomes: guardians of the body. Annu Rev Immunol, 2009. 27: p. 229–65. [DOI] [PubMed] [Google Scholar]
- [56].Lin KM, et al. IRAK-1 bypasses priming and directly links TLRs to rapid NLRP3 inflammasome activation. Proc Natl Acad Sci U S A, 2014. 111(2): p. 775–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Fernandes-Alnemri T, et al. Cutting edge: TLR signaling licenses IRAK1 for rapid activation of the NLRP3 inflammasome. J Immunol, 2013. 191(8): p. 3995–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Lamkanfi M and Dixit VM, Inflammasomes and their roles in health and disease. Annu Rev Cell Dev Biol, 2012. 28: p. 137–61. [DOI] [PubMed] [Google Scholar]
- [59].Strowig T, et al. Inflammasomes in health and disease. Nature, 2012. 481(7381): p. 278–86. [DOI] [PubMed] [Google Scholar]
- [60].Celli J and Tsolis RM, Bacteria, the endoplasmic reticulum and the unfolded protein response: friends or foes? Nat Rev Microbiol, 2015. 13(2): p. 71–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Janssens S, Pulendran B, and Lambrecht BN, Emerging functions of the unfolded protein response in immunity. Nat Immunol, 2014. 15(10): p. 910–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Alomairi J, et al. Alterations of host cell ubiquitination machinery by pathogenic bacteria. Front Cell Infect Microbiol, 2015. 5: p. 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Ashida H and Sasakawa C, Bacterial E3 ligase effectors exploit host ubiquitin systems. Curr Opin Microbiol, 2017. 35: p. 16–22. [DOI] [PubMed] [Google Scholar]
- [64].Lin YH and Machner MP, Exploitation of the host cell ubiquitin machinery by microbial effect or proteins. J Cell Sci, 2017. 130(12): p. 1985–1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65]**.Charbonneau ME, et al. Perturbation of ubiquitin homeostasis promotes macrophage oxidative defenses. Sci Rep, 2019. 9(1): p. 10245. [DOI] [PMC free article] [PubMed] [Google Scholar]; Manipulation of host ubiquitin homeostasis leads to ROS production and potentiates IL-6 secretion downstream of TLR4.
- [66]*.Cubillos-Ruiz JR, et al. ER Stress Sensor XBP1 Controls Anti-tumor Immunity by Disrupting Dendritic Cell Homeostasis. Cell, 2015. 161(7): p. 1527–38. [DOI] [PMC free article] [PubMed] [Google Scholar]; Lipid peroxidation byproducts drive XBP1 activation and result in immunosuppressive DC phenotype.
- [67].Weichhart T, Hengstschlager M, and Linke M, Regulation of innate immune cell function by mTOR. Nat Rev Immunol, 2015. 15(10): p. 599–614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].York AG, et al. Limiting Cholesterol Biosynthetic Flux Spontaneously Engages Type I IFN Signaling. Cell, 2015. 163(7): p. 1716–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Tattoli I, et al. Amino acid starvation induced by invasive bacterial pathogens triggers an innate host defense program. Cell Host Microbe, 2012. 11(6): p. 563–75. [DOI] [PubMed] [Google Scholar]
- [70].Ravindran R, et al. Vaccine activation of the nutrient sensor GCN2 in dendritic cells enhances antigen presentation. Science, 2014. 343(6168): p. 313–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Pulendran B, The varieties of immunological experience: of pathogens, stress, and dendritic cells. Annu Rev Immunol, 2015. 33: p. 563–606. [DOI] [PubMed] [Google Scholar]
- [72].Fairn GD and Grinstein S, How nascent phagosomes mature to become phagolysosomes. Trends Immunol, 2012. 33(8): p. 397–405. [DOI] [PubMed] [Google Scholar]
- [73].Thery C and Amigorena S, The cell biology of antigen presentation in dendritic cells. Curr Opin Immunol, 2001. 13(1): p. 45–51. [DOI] [PubMed] [Google Scholar]
- [74].Trombetta ES and Mellman I, Cell biology of antigen processing in vitro and in vivo. Annu Rev Immunol, 2005. 23: p. 975–1028. [DOI] [PubMed] [Google Scholar]
- [75].Guilliams M, et al. Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny. Nat Rev Immunol, 2014. 14(8): p. 571–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76]*.Ferris ST, et al. cDC1 prime and are licensed by CD4(+) T cells to induce anti-tumour immunity. Nature, 2020. 584(7822): p. 624–629. [DOI] [PMC free article] [PubMed] [Google Scholar]; Authors find that cDC1s play critical role in both CD4 and CD8 T cell priming, which hinges on cDC1 expression of CD40.
- [77].Jorgensen I, Rayamajhi M, and Miao EA, Programmed cell death as a defence against infection. Nat Rev Immunol, 2017. 17(3): p. 151–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Jorgensen I, et al. Pyroptosis triggers pore-induced intracellular traps (PITs) that capture bacteria and lead to their clearance by efferocytosis. J Exp Med, 2016. 213(10): p. 2113–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Lawrence SM, Corriden R, and Nizet V, How Neutrophils Meet Their End. Trends Immunol, 2020. 41(6): p. 531–544. [DOI] [PubMed] [Google Scholar]
- [80].Ashida H, et al. Cell death and infection: a double-edged sword for host and pathogen survival. J Cell Biol, 2011. 195(6): p. 931–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Servet-Delprat C, et al. Consequences of Fas-mediated human dendritic cell apoptosis induced by measles virus. J Virol, 2000. 74(9): p. 4387–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Siavoshian S, et al. Hepatitis C virus core, NS3, NS5A, NS5B proteins induce apoptosis in mature dendritic cells. J Med Virol, 2005. 75(3): p. 402–11. [DOI] [PubMed] [Google Scholar]
- [83].Leverkus M, et al. Maturation of dendritic cells leads to up-regulation of cellular FLICE-inhibitory protein and concomitant down-regulation of death ligand-mediated apoptosis. Blood, 2000. 96(7): p. 2628–31. [PubMed] [Google Scholar]
- [84].Ashany D, et al. Dendritic cells are resistant to apoptosis through the Fas (CD95/APO-1) pathway. J Immunol, 1999. 163(10): p. 5303–11. [PubMed] [Google Scholar]
- [85].Willems F, et al. Expression of c-FLIP(L) and resistance to CD95-mediated apoptosis of monocyte-derived dendritic cells: inhibition by bisindolylmaleimide. Blood, 2000. 95(11): p. 3478–82. [PubMed] [Google Scholar]
- [86].Lundqvist A, et al. Mature dendritic cells are protected from Fas/CD95-mediated apoptosis by upregulation of Bcl-X(L). Cancer Immunol Immunother, 2002. 51(3): p. 139–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Riol-Blanco L, et al. Immunological synapse formation inhibits, via NF-kappaB and FOXO1, the apoptosis of dendritic cells. Nat Immunol, 2009. 10(7): p. 753–60. [DOI] [PubMed] [Google Scholar]
- [88]**.Erlich Z, et al. Macrophages, rather than DCs, are responsible for inflammasome activity in the GM-CSF BMDC model. Nat Immunol, 2019. 2019. [DOI] [PubMed] [Google Scholar]; Demonstrates that cDCs are resistant to inflammasome activation.
- [89].McDaniel MM, et al. Suppression of Inflammasome Activation by IRF8 and IRF4 in cDCs Is Critical for T Cell Priming. Cell Rep, 2020. 31(5): p. 107604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Cella M, et al. Maturation, activation, and protection of dendritic cells induced by double-stranded RNA. J Exp Med, 1999. 189(5): p. 821–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Zanoni I, et al. By Capturing Inflammatory Lipids Released from Dying Cells, the Receptor CD14 Induces Inflammasome-Dependent Phagocyte Hyperactivation. Immunity, 2017. 47(4): p. 697–709 e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Evavold CL, et al. The Pore-Forming Protein Gasdermin D Regulates Interleukin-1 Secretion from Living Macrophages. Immunity, 2018. 48(1): p. 35–44 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Chen M and Wang J, Programmed cell death of dendritic cells in immune regulation. Immunol Rev, 2010. 236: p. 11–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Stranges PB, et al. Elimination of antigen-presenting cells and autoreactive T cells by Fas contributes to prevention of autoimmunity. Immunity, 2007. 26(5): p. 629–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Patel R, et al. Culling of APCs by inflammatory cell death pathways restricts TIM3 and PD-1 expression and promotes the survival of primed CD8 T cells. Cell Death Differ, 2017. 24(11): p. 1900–1911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Bajana S, et al. IRF4 and IRF8 Act in CD11c+ Cells To Regulate Terminal Differentiation of Lung Tissue Dendritic Cells. J Immunol, 2016. 196(4): p. 1666–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Ainsua-Enrich E, et al. IRF4-dependent dendritic cells regulate CD8(+) T-cell differentiation and memory responses in influenza infection. Mucosal Immunol, 2019. 12(4): p. 1025–1037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Sichien D, et al. IRF8 Transcription Factor Controls Survival and Function of Terminally Differentiated Conventional and Plasmacytoid Dendritic Cells, Respectively. Immunity, 2016. 45(3): p. 626–640. [DOI] [PubMed] [Google Scholar]
- [99]**.Vander Lugt B, et al. Transcriptional determinants of tolerogenic and immunogenic states during dendritic cell maturation. J Cell Biol, 2017. 216(3): p. 779–792. [DOI] [PMC free article] [PubMed] [Google Scholar]; IRF4-deficient cDCs are more immunogenic and inflammatory, resulting in loss of tolerogenic T cell priming.
- [100].Vander Lugt B, et al. Transcriptional programming of dendritic cells for enhanced MHC class II antigen presentation. Nat Immunol, 2014. 15(2): p. 161–7. [DOI] [PubMed] [Google Scholar]
- [101].Zhu J, Yamane H, and Paul WE, Differentiation of effector CD4 T cell populations (*). Annu Rev Immunol, 2010. 28: p. 445–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Kaiko GE, et al. Immunological decision-making: how does the immune system decide to mount a helper T-cell response? Immunology, 2008. 123(3): p. 326–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Nowarski R, Jackson R, and Flavell RA, The Stromal Intervention: Regulation of Immunity and Inflammation at the Epithelial-Mesenchymal Barrier. Cell, 2017. 168(3): p. 362–375. [DOI] [PubMed] [Google Scholar]
- [104].Tan AM, et al. TLR4 signaling in stromal cells is critical for the initiation of allergic Th2 responses to inhaled antigen. J Immunol, 2010. 184(7): p. 3535–44. [DOI] [PubMed] [Google Scholar]
- [105].Rochman Y, et al. TSLP signaling in CD4(+) T cells programs a pathogenic T helper 2 cell state. Sci Signal, 2018. 11(521). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Van Dyken SJ, et al. A tissue checkpoint regulates type 2 immunity. Nat Immunol, 2016. 17(12): p. 1381–1387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Sallusto F, Geginat J, and Lanzavecchia A, Central memory and effector memory T cell subsets:function, generation, and maintenance. Annu Rev Immunol, 2004. 22: p. 745–63. [DOI] [PubMed] [Google Scholar]
- [108]**.Amezcua Vesely MC, et al. Effector TH17 Cells Give Rise to Long-Lived TRM Cells that Are Essential for an Immediate Response against Bacterial Infection. Cell, 2019. 178(5): p. 1176–1188 e15. [DOI] [PMC free article] [PubMed] [Google Scholar]; Defines the orgin of resident memory CD4 T cells and explores their function in protective immunity against microbial rechallenge.
- [109].Yu SF, et al. Human memory, but not naive, CD4+ T cells expressing transcription factor T-bet might drive rapid cytokine production. J Biol Chem, 2014. 289(51): p. 35561–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Boesteanu AC and Katsikis PD, Memory T cells need CD28 costimulation to remember. Semin Immunol, 2009. 21(2): p. 69–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Berg RE, et al. Memory CD8+ T cells provide innate immune protection against Listeria monocytogenes in the absence of cognate antigen. J Exp Med, 2003. 198(10): p. 1583–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Pham OH, et al. T cell expression of IL-18R and DR3 is essential for non-cognate stimulation of Th1 cells and optimal clearance of intracellular bacteria. PLoS Pathog, 2017. 13(8): p. e1006566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Lee HG, et al. Pathogenic function of bystander-activated memory-like CD4(+) T cells in autoimmune encephalomyelitis. Nat Commun, 2019. 10(1): p. 709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114]*.Lee PH, et al. Host conditioning with IL-1beta improves the antitumor function of adoptively transferred T cells. J Exp Med, 2019. 216(11): p. 2619–2634. [DOI] [PMC free article] [PubMed] [Google Scholar]; IL1b signaling in CD8 T cells can also enhance proliferation and cytotoxicity.
- [115]*.Tuncel J, Benoist C, and Mathis D, T cell anergy in perinatal mice is promoted by T reg cells and prevented by IL-33. J Exp Med, 2019. 216(6): p. 1328–1344. [DOI] [PMC free article] [PubMed] [Google Scholar]; IL-33 signaling in CD4+ T cells extended their activation and prevented anergy.
- [116].Kawai T and Akira S, Signaling to NF-kappaB by Toll-like receptors. Trends Mol Med, 2007. 13(11): p. 460–9. [DOI] [PubMed] [Google Scholar]
- [117].Wesche H, et al. MyD88: an adapter that recruits IRAK to the IL-1 receptor complex. Immunity, 1997. 7(6): p. 837–47. [DOI] [PubMed] [Google Scholar]
- [118]*.Griesenauer B, et al. ST2/MyD88 Deficiency Protects Mice against Acute Graft-versus-Host Disease and Spares Regulatory T Cells. J Immunol, 2019. 202(10): p. 3053–3064. [DOI] [PMC free article] [PubMed] [Google Scholar]; ST2/MyD88-deficiency in T cells reduces GVHD severity due to abrogation of ST2, not TLR, signaling.
- [119].Schenten D, et al. Signaling through the adaptor molecule MyD88 in CD4+ T cells is required to overcome suppression by regulatory T cells. Immunity, 2014. 40(1): p. 78–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Hu W, et al. Priming microenvironments dictate cytokine requirements for T helper 17 cell lineage commitment. Immunity, 2011. 35(6): p. 1010–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Liu H and Rohowsky-Kochan C, Regulation of IL-17 in human CCR6+ effector memory T cells. J Immunol, 2008. 180(12): p. 7948–57. [DOI] [PubMed] [Google Scholar]
- [122]**.Muller J, Baeyens A, and Dustin ML, Tumor Necrosis Factor Receptor Superfamily in T Cell Priming and Effector Function. Adv Immunol, 2018. 140: p. 21–57. [DOI] [PubMed] [Google Scholar]; Excellent review on TNFRSF members in T cell priming/effector function.
- [123].Grewal IS and Flavell RA, The role of CD40 ligand in costimulation and T-cell activation. Immunol Rev, 1996. 153: p. 85–106. [DOI] [PubMed] [Google Scholar]
- [124].Cella M, et al. Ligation of CD40 on dendritic cells triggers production of high levels of interleukin-12 and enhances T cell stimulatory capacity: T-Thelp via APC activation. J Exp Med, 1996. 184(2): p. 747–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [125].Casamayor-Palleja M, Khan M, and MacLennan IC, A subset of CD4+ memory T cells contains preformed CD40 ligand that is rapidly but transiently expressed on their surface after activation through the T cell receptor complex. J Exp Med, 1995. 181(4): p. 1293–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Lee BO, et al. The biological outcome of CD40 signaling is dependent on the duration of CD40 ligand expression: reciprocal regulation by interleukin (IL)-4 and IL-12. J Exp Med, 2002. 196(5): p. 693–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Lai W, et al. Transcriptional control of rapid recall by memory CD4 T cells. J Immunol, 2011. 187(1): p. 133–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Koguchi Y, et al. Preformed CD40 ligand exists in secretory lysosomes in effector and memory CD4+ T cells and is quickly expressed on the cell surface in an antigen-specific manner. Blood, 2007. 110(7): p. 2520–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Han BK, Olsen NJ, and Bottaro A, The CD27-CD70 pathway and pathogenesis of autoimmune disease. Semin Arthritis Rheum, 2016. 45(4): p. 496–501. [DOI] [PubMed] [Google Scholar]
- [130].van Oosterwijk MF, et al. CD27-CD70 interactions sensitise naive CD4+ T cells for IL-12-induced Th1 cell development. Int Immunol, 2007. 19(6): p. 713–8. [DOI] [PubMed] [Google Scholar]
- [131].Salek-Ardakani S, et al. OX40 (CD134) controls memory T helper 2 cells that drive lung inflammation. J Exp Med, 2003. 198(2): p. 315–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Jember AG, et al. Development of allergic inflammation in a murine model of asthma is dependent on the costimulatory receptor OX40. J Exp Med, 2001. 193(3): p. 387–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Iezzi G, et al. CD40-CD40L cross-talk integrates strong antigenic signals and microbial stimuli to induce development of IL-17-producing CD4+ T cells. Proc Natl Acad Sci U S A, 2009. 106(3): p. 876–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [134].Donado CA, et al. A Two-Cell Model for IL-1beta Release Mediated by Death-Receptor Signaling.Cell Rep, 2020. 31(1): p. 107466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Silke J and Hartland EL, Masters, marionettes and modulators: intersection of pathogen virulence factors and mammalian death receptor signaling. Curr Opin Immunol, 2013. 25(4): p. 436–40. [DOI] [PubMed] [Google Scholar]
- [136].Hodgson A and Wan F, Interference with nuclear factor kappaB signaling pathway by pathogen-encoded proteases: global and selective inhibition. Mol Microbiol, 2016. 99(3): p. 439–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Peterson LW, et al. RIPK1-dependent apoptosis bypasses pathogen blockade of innate signaling to promote immune defense. J Exp Med, 2017. 214(11): p. 3171–3182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Demarco B, et al. Caspase-8-dependent gasdermin D cleavage promotes antimicrobial defense but confers susceptibility to TNF-induced lethality. Sci Adv, 2020. 6(47). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Orning P, et al. Pathogen blockade of TAK1 triggers caspase-8-dependent cleavage of gasdermin D and cell death. Science, 2018. 362(6418): p. 1064–1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Dinarello CA, Simon A, and van der Meer JW, Treating inflammation by blocking interleukin-1 in a broad spectrum of diseases. Nat Rev Drug Discov, 2012. 11(8): p. 633–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Shaw PJ, McDermott MF, and Kanneganti TD, Inflammasomes and autoimmunity. Trends Mol Med, 2011. 17(2): p. 57–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
