Fact-checked by Grok 2 weeks ago

Pattern recognition receptor

Pattern recognition receptors (PRRs) are a class of germline-encoded proteins expressed by cells involved in across animals and plants, including immune cells like macrophages, dendritic cells, and neutrophils, as well as non-immune cells, that detect conserved molecular motifs known as pathogen-associated molecular patterns (PAMPs) from microbes and damage-associated molecular patterns (DAMPs) from stressed or damaged host cells. These receptors enable the to rapidly distinguish between self and non-self entities, triggering immediate defensive responses without prior exposure. By recognizing broad structural features rather than specific s, PRRs form the foundational layer of immunity, bridging innate and adaptive responses through production and . The concept of PRRs originated from the work of immunologist Charles A. Janeway Jr., who in 1989 proposed that innate immunity relies on receptors evolved to recognize invariant patterns in pathogens, challenging the prevailing focus on adaptive immunity. This hypothesis gained empirical support in the mid-1990s when identified the receptor in as essential for antifungal defense, revealing a conserved innate sensing mechanism. Shortly thereafter, in 1998, Bruce A. Beutler discovered that the mammalian (TLR4) mediates responses to bacterial (LPS), linking PRRs to and ; these breakthroughs earned Hoffmann, Beutler, and the 2011 in Physiology or Medicine. PRRs are categorized into several families based on their cellular localization, ligand specificity, and signaling domains, including extracellular and endosomal Toll-like receptors (TLRs), cytosolic nucleotide-binding oligomerization domain-like receptors (NLRs), RIG-I-like receptors (RLRs), C-type lectin receptors (CLRs), and absent in melanoma 2-like receptors (ALRs), with additional sensors like cyclic GMP-AMP synthase (cGAS). TLRs, the most studied group, comprise 10 functional members in humans that detect diverse PAMPs such as bacterial lipopeptides, viral double-stranded RNA, and fungal zymosan. Cytosolic PRRs like NLRs and RLRs monitor intracellular threats, forming or activating pathways upon detecting bacterial peptidoglycans or viral RNA, respectively. Upon ligand engagement, PRRs initiate intracellular signaling cascades via adaptor proteins like MyD88, TRIF, or MAVS, culminating in the activation of transcription factors such as and /7 to induce proinflammatory cytokines (e.g., TNF-α, IL-1β), type I interferons, and . These responses not only contain infections but also promote adaptive immunity by enhancing and T-cell priming on dendritic cells. Regulatory mechanisms, including inhibitory receptors and post-translational modifications like ubiquitination, fine-tune PRR activity to prevent excessive . Beyond infection control, PRRs contribute to sterile inflammation in conditions like , neurodegeneration, and cancer, where DAMPs from necrotic cells amplify tissue damage. Dysregulation of PRR pathways underlies autoimmune diseases (e.g., via TLRs) and chronic inflammatory disorders, while their therapeutic modulation—through agonists like TLR4 activators in vaccines or inhibitors targeting —holds promise for treating infections, autoimmunity, and tumors. Recent discoveries since 2021, including cGAS-STING interactions with and epigenetic regulation of "trained immunity," underscore the evolving role of PRRs in systemic and .

Overview

Definition and functions

Pattern recognition receptors (PRRs) are a class of germline-encoded proteins that serve as sentinel sensors in the , enabling host cells to detect and respond to invading pathogens and endogenous danger signals without prior exposure or sensitization. These receptors are constitutively expressed on the surface or within various cell types, including both immune cells like macrophages and dendritic cells, as well as non-immune cells such as epithelial cells, allowing for broad surveillance across tissues. By recognizing highly conserved molecular motifs, PRRs distinguish between self and non-self entities, initiating rapid protective responses that bridge innate and adaptive immunity. The primary ligands for PRRs include pathogen-associated molecular patterns (PAMPs), which are evolutionarily conserved structures unique to microorganisms, such as lipopolysaccharide (LPS) from , flagellin from bacterial flagella, double-stranded RNA (dsRNA) from viruses, from bacterial cell walls, and β-glucans from fungal cell walls. Additionally, PRRs detect damage-associated molecular patterns (DAMPs) released during cellular stress, injury, or necrosis, exemplified by high-mobility group box 1 (HMGB1) protein and extracellular ATP. Upon ligand binding, PRRs trigger multifaceted immune functions, including the production of pro-inflammatory cytokines (e.g., TNF-α, IL-1β, IL-6) to amplify , enhancement of and for pathogen clearance, and activation of antigen-presenting cells to prime adaptive immune responses. These actions collectively promote host defense against infections and sterile tissue damage while maintaining immune homeostasis. PRRs exhibit diverse localization to maximize detection efficiency: membrane-bound forms operate at the extracellular surface or in endosomal compartments to sense external or internalized threats, cytoplasmic variants monitor intracellular invaders, and secreted forms circulate in bodily fluids for systemic surveillance. For instance, Toll-like receptors (TLRs) represent membrane-bound PRRs, while NOD-like receptors (NLRs) function in the . This strategic distribution underscores the evolutionary advantage of PRRs, providing a non-clonal, immediate response that is essential for against diverse microbial challenges and injury, predating adaptive immunity in evolutionary history.

Historical background

In 1989, Charles Janeway Jr. proposed the concept of pattern recognition receptors (PRRs), predicting that germline-encoded receptors in the innate immune system would detect conserved microbial patterns to initiate immune responses, shifting emphasis from the previously dominant adaptive immunity paradigm. This theoretical framework gained experimental validation in 1996 when Bruno Lemaitre and colleagues identified the Toll receptor in Drosophila melanogaster as a key antifungal PRR, demonstrating its role in inducing antimicrobial peptide production against fungal infections. Building on this invertebrate discovery, Ruslan Medzhitov and Charles Janeway Jr. extended the concept to mammals in 1997 by cloning human Toll-like receptor 4 (TLR4) and showing its activation of NF-κB signaling in response to microbial stimuli, with subsequent 1998 studies linking TLR4 specifically to lipopolysaccharide (LPS) recognition from Gram-negative bacteria. Subsequent milestones expanded the PRR repertoire: in 2002, Naohiro Inohara et al. described the (NLR) family as cytosolic sensors of bacterial components, regulating and host defense. In 2004, Mitsutoshi Yoneyama et al. identified RIG-I-like receptors (RLRs) as cytoplasmic detectors of viral double-stranded RNA, triggering type I production. The discovery continued in 2013 with Lijun Sun et al. revealing cyclic GMP-AMP synthase (cGAS) as a cytosolic DNA sensor that generates the second messenger cGAMP to activate pathways. These findings culminated in the 2011 in or awarded to Bruce Beutler and Jules Hoffmann for their contributions to and TLR discoveries, underscoring PRRs' foundational role in innate immunity. The identification of PRRs marked a , redirecting immunological research from adaptive T- and B-cell responses to the innate system's rapid, pattern-based microbial sensing, with NLRs linking to activation and RLRs/cGAS integrating with signaling by the mid-2010s. As of 2025, ongoing research continues to elucidate PRR functions in recognizing damage-associated molecular patterns (DAMPs) in sterile and advances therapeutic targeting of PRRs for immune modulation in diseases like and cancer.

PRR families in animals

Toll-like receptors (TLRs)

Toll-like receptors (TLRs) represent a major family of membrane-bound pattern recognition receptors (PRRs) in animals, pivotal for detecting microbial components and initiating innate immune responses. These type I transmembrane glycoproteins are characterized by a modular : an extracellular ligand-binding domain composed of multiple leucine-rich repeats (LRRs) that form a horseshoe-shaped structure, a single spanning transmembrane helix, and an intracellular Toll/interleukin-1 receptor (TIR) domain responsible for signal propagation. The LRR ectodomain, typically comprising 16–28 repeats, enables specific recognition of diverse pathogen-associated molecular patterns (PAMPs) through concave or convex surfaces that interact with ligands. This allows flexibility in binding various molecular shapes while maintaining specificity. In humans, there are ten functional TLRs (TLR1–10), encoded by genes clustered on chromosomes 4 and X. Their subcellular localization is tightly regulated to match accessibility and prevent aberrant activation by self-molecules. surface TLRs, including TLR1, TLR2, TLR4, TLR5, TLR6, and TLR10, reside on the of various types and primarily sense extracellular and fungal components. In contrast, endosomal TLRs such as TLR3, TLR7, TLR8, and TLR9 are trafficked to intracellular compartments like endosomes and lysosomes, where they detect internalized nucleic acids from viruses or . This compartmentalization is governed by specific trafficking motifs; for instance, endosomal TLRs contain tyrosine-based motifs that mediate via AP-2 adaptors. TLRs exhibit diverse ligand specificities, often requiring accessory proteins for optimal recognition. The following table summarizes key ligands for human TLRs, highlighting their microbial origins:
TLRLocalizationRepresentative LigandsNotes
TLR1Cell surfaceTriacyl lipopeptides (e.g., Pam3CSK4 from bacteria)Forms heterodimers with TLR2
TLR2Cell surfaceDiacyl lipopeptides (e.g., Pam2CSK4 from Gram-positive bacteria), lipoteichoic acid, zymosan (fungi)Heterodimerizes with TLR1 or TLR6; recognizes multiple PAMPs
TLR3EndosomalDouble-stranded RNA (dsRNA, e.g., from viruses like poly(I:C))Homodimerizes
TLR4Cell surfaceLipopolysaccharide (LPS from Gram-negative bacteria), with MD-2 and LBP co-factorsForms homodimers; also senses viral proteins like RSV F protein
TLR5Cell surfaceFlagellin (from bacterial flagella)Recognizes motile bacteria
TLR6Cell surfaceDiacyl lipopeptides (e.g., from mycoplasma), fungal zymosanHeterodimerizes with TLR2
TLR7EndosomalSingle-stranded RNA (ssRNA, e.g., from viruses like imiquimod agonists)Recognizes GU-rich sequences
TLR8EndosomalSingle-stranded RNA (ssRNA, e.g., from viruses like loxoribine)Prefers UG-rich sequences; active in humans and mice
TLR9EndosomalUnmethylated CpG DNA (from bacteria and viruses)Recognizes bacterial DNA motifs
TLR10Cell surfaceMicrobial components (e.g., from Listeria monocytogenes, Streptococcus pneumoniae), HIV gp41, dsRNALeast characterized; acts as anti-inflammatory regulator; interacts with TLR2 ligands
Upon engagement, TLRs typically dimerize—either as homodimers (e.g., TLR4) or heterodimers (e.g., TLR2/TLR1)—inducing conformational changes that bring TIR domains into proximity to recruit signaling molecules. The TIR domain briefly facilitates initiation of intracellular signaling cascades. TLRs are expressed across a wide range of s, including professional immune cells like macrophages, dendritic cells, and B cells, as well as non-immune s such as epithelial cells, endothelial cells, and fibroblasts, enabling surveillance at barrier sites and within tissues. This broad expression pattern supports both detection and . Evolutionarily, TLRs trace back to the fruit fly , where the receptor first demonstrated antimicrobial defense, underscoring their ancient conservation across metazoans. Notably, certain TLRs, such as TLR2, also contribute to symbiotic interactions by recognizing harmless commensal microbes, for instance, in the where they promote tolerance to bacterial lipoproteins from species.

C-type lectin receptors (CLRs)

receptors (CLRs) are a diverse family of pattern recognition receptors that primarily recognize structures on pathogens and host cells, playing crucial roles in innate immunity through extracellular sensing. These receptors are characterized by the presence of one or more C-type lectin-like domains (CTLDs), which enable calcium-dependent binding to carbohydrates. CLRs can function as membrane-bound or soluble proteins, but this section focuses on their membrane-associated forms specialized for detecting fungal components and self-glycans. The core structural feature of CLRs is the CTLD, a compact globular stabilized by bridges and containing specific motifs for binding, such as EPN for or QPD for recognition in a Ca²⁺-dependent manner. Membrane-bound CLRs are classified into type I and type II transmembrane proteins based on their orientation: type I CLRs, like the (MR), have an extracellular with multiple CTLDs and a single , facilitating endocytic functions; in contrast, type II CLRs, such as dectin-1, feature a short N-terminal cytoplasmic tail and an extracellular C-terminal CTLD, often involved in signaling and . This structural diversity allows CLRs to tether pathogens to immune cells or mediate clearance of glycosylated molecules. CLRs are grouped into families based on phylogeny and function. The mannose receptor family (group I CLRs) includes endocytic receptors like (CD206) and DEC-205 (LY75), which recognize and residues on microbial surfaces, promoting uptake in macrophages and dendritic cells (DCs). The asialoglycoprotein receptor family (group II CLRs) comprises liver-specific receptors such as the (ASGR), which clears desialylated glycoproteins from circulation, maintaining . Additional key subtypes include dectin-1 (CLEC7A) and dectin-2 (CLEC6A), which are type II CLRs specialized for fungal components, with dectin-1 binding β-1,3-glucans and dectin-2 targeting α-mannans. Representative ligands for CLRs include fungal such as β-1,3-glucans bound by dectin-1, mannans recognized by the , and α-mannose structures engaged by DC-SIGN (CD209), a group II CLR on DCs that facilitates viral and bacterial adhesion. CLRs also interact with damage-associated molecular patterns (DAMPs), including and released from damaged cells, linking infection responses to sterile inflammation. These glycan-specific interactions distinguish CLRs from other PRRs by emphasizing motifs over or nucleic acids. CLRs are predominantly expressed on myeloid cells, including macrophages, DCs, and neutrophils, where they drive and of glycan-coated pathogens, enhancing . For instance, the mediates uptake of mycobacterial mannoproteins, while dectin-1 promotes fungal . Beyond immunity, CLRs exhibit dual roles in , such as DC-SIGN inducing T-cell tolerance to self-antigens, thereby preventing . These receptors briefly activate pathways like Syk or CARD9 upon binding to coordinate responses. A unique aspect of the CLR superfamily is its expansion in the , with approximately 100 genes encoding CTLD-containing proteins, reflecting evolutionary adaptation to diverse threats. Additionally, CLR with self-glycans contributes to allergic diseases; for example, dectin-1 and DC-SIGN bind similar motifs on and allergens, promoting Th2-biased responses that exacerbate . This glycan mimicry underscores the receptors' role in balancing protective immunity against pathological reactions.

NOD-like receptors (NLRs)

NOD-like receptors (NLRs) are a family of intracellular receptors that detect microbial motifs and damage-associated molecular patterns in the of host cells, playing a crucial role in innate immunity against bacterial infections. In humans, there are approximately 22-23 NLR proteins, which are primarily expressed in immune cells such as macrophages and epithelial cells, with some members showing tissue-specific distribution like NOD1 in intestinal epithelia. These receptors enable the recognition of intracellular pathogens that evade extracellular sensors, contributing to the maintenance of with the . The canonical structure of NLRs is tripartite, consisting of an N-terminal effector domain for downstream signaling, a central NACHT (domain present in NAIP, CIITA, HET-E, and TP1) oligomerization domain that binds , and a C-terminal (LRR) domain responsible for recognition and autoregulation. The N-terminal effector domains vary across members and include pyrin (PYD), caspase activation and recruitment (), or baculovirus repeat (BIR) domains, which facilitate interactions with adaptor proteins. In the resting state, the LRR domain maintains an autoinhibited conformation; upon binding, it induces a conformational change that promotes NACHT-mediated self-oligomerization, enabling . NLRs are classified into subfamilies based on their N-terminal domains: the NLRC subfamily includes NOD1 and NOD2, which possess CARD domains and sense bacterial peptidoglycans; the NLRP subfamily, with 14 members in humans featuring PYD domains, is involved in inflammasome assembly; and the NAIP subfamily, with one member in humans (NAIP), contains BIR domains and recognizes bacterial proteins such as flagellin. Other subfamilies like NLRB, NLRA, and NLRX exist but are less characterized in humans. Representative ligands include γ-D-glutamyl-meso-diaminopimelic acid (iE-DAP) for NOD1, derived from Gram-negative bacterial peptidoglycan; muramyldipeptide (MDP) for NOD2, a motif common to both Gram-positive and Gram-negative bacteria; and for NLRP3, diverse stimuli such as extracellular ATP, monosodium urate (uric acid) crystals, and the bacterial toxin nigericin, which trigger potassium efflux. NLRs are ubiquitously expressed but enriched in barrier tissues like the and professional such as macrophages, where they monitor cytosolic contents for invasive microbes. Activation leads to oligomerization and recruitment of adaptor molecules, with some NLRPs briefly assembling to activate caspase-1 for inflammatory responses. Notably, NOD1 and play a key role in sensing the by detecting fragments that breach the epithelial barrier, helping to regulate microbial composition and prevent . Mutations in , such as the frameshift 3020insC variant, are strongly associated with increased susceptibility to , impairing bacterial recognition and leading to chronic inflammation.

RIG-I-like receptors (RLRs)

RIG-I-like receptors (RLRs) are a family of cytoplasmic receptors that detect viral in the , playing a crucial role in initiating antiviral innate immune responses. These receptors belong to the DExD/H-box family and are characterized by their ability to recognize specific structural features of non-self , distinguishing them from RNAs. RLRs are essential for establishing an antiviral state in infected cells, primarily through the induction of type I s, though their downstream signaling is mediated via the adaptor protein MAVS to activate interferon regulatory factors. The core structure of RLRs includes a central DExD/H-box domain responsible for ATP-dependent RNA binding and unwinding, flanked by an N-terminal domain containing caspase activation and recruitment () motifs in most members and a C-terminal regulatory domain (CTD) that facilitates autoregulation. The domain consists of two RecA-like subdomains (Hel1 and Hel2) that hydrolyze ATP to translocate along , enabling conformational changes upon ligand . The CTD, often featuring a zinc-binding , serves as a sensor for ligands while maintaining the receptor in an autoinhibited state by masking the domains, preventing spurious activation. The RLR family comprises three main members: retinoic acid-inducible gene I (RIG-I, encoded by DDX58), melanoma differentiation-associated protein 5 (, encoded by IFIH1), and laboratory of genetics and physiology 2 (LGP2, encoded by DHX58). RIG-I preferentially recognizes short double-stranded (dsRNA) molecules bearing a 5'-triphosphate (5' ) end or blunt ends, such as the panhandle structures generated during replication. In contrast, detects longer dsRNA (>1 kb), including replicative intermediates from viruses like encephalomyocarditis virus (EMCV), a . LGP2, lacking domains, acts primarily as a regulatory protein that binds dsRNA termini and modulates the activity of RIG-I and without direct signaling capability. These receptors discriminate viral from host through features like the uncapped 5' on viral genomes—absent in capped or 2'-O-methylated host mRNAs—and RNA length or secondary structure, ensuring specificity. RLRs are broadly expressed at low basal levels in most cell types, including epithelial cells and immune cells like macrophages and dendritic cells, but their expression is rapidly upregulated following viral infection or type I interferon stimulation. Activation requires for RNA unwinding, which induces a conformational shift: in RIG-I, ligand binding to the CTD releases the autoinhibitory , exposing CARDs for oligomerization; forms polar filaments along long ds scaffolds. This auto-regulation via CTD masking of CARDs prevents aberrant activation by self-, highlighting a key unique aspect of RLRs in maintaining while mounting robust antiviral responses. LGP2's regulatory role further fine-tunes this process, either enhancing or dampening RLR signaling depending on the viral context.

AIM2-like receptors (ALRs)

AIM2-like receptors (ALRs) are a family of cytoplasmic pattern recognition receptors characterized by their ability to detect double-stranded DNA (dsDNA) and initiate inflammasome assembly. These receptors typically feature a bipartite domain organization, consisting of an N-terminal pyrin domain (PYD) that facilitates homotypic interactions for inflammasome formation and a C-terminal hematopoietic expression/interferon-inducible nature (HIN) domain responsible for nucleic acid binding. The HIN domain engages dsDNA through electrostatic interactions within positively charged surface grooves, enabling sequence-independent recognition of microbial or self-derived DNA. Prominent members of the ALR family include absent in 2 (AIM2), which senses cytosolic dsDNA from viruses and bacteria, and interferon-inducible protein 16 (IFI16), which also detects dsDNA and can activate pathway in addition to signaling. In humans, the ALR family comprises four members: AIM2, IFI16, myeloid antigen (MNDA), and interferon-inducible protein X (IFIX or PYHIN1). Key ligands for AIM2 include synthetic dsDNA analogs like poly(dA:dT) and viral DNA such as that from herpes simplex virus 1 (HSV-1), with binding affinity being length-dependent—optimal activation occurs with dsDNA longer than 70 base pairs, though shorter fragments (around 24 base pairs) can initiate interactions. ALRs are broadly expressed across immune and non-immune cells, with expression levels upregulated by type I interferons during or . Upon binding, ALRs undergo oligomerization along DNA scaffolds, forming filamentous assemblies that amplify signaling through PYD-mediated of adaptor proteins. This process underpins the role of the AIM2 in inducing as a defense mechanism against intracellular pathogens. Unlike in mice, which possess up to 13 ALRs, the more limited human repertoire highlights species-specific adaptations in DNA sensing.

Cyclic GMP-AMP synthase (cGAS) and STING

Cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) form a key cytosolic pathway for detecting double-stranded DNA (dsDNA), serving as a major surveillance system independent of membrane-bound pattern recognition receptors. This axis enables cells to sense foreign microbial DNA or misplaced self-DNA, initiating innate immune responses through the production of second messengers. Unlike other DNA sensors such as AIM2-like receptors (ALRs), which directly trigger inflammasomes, the cGAS-STING pathway emphasizes the synthesis of cyclic dinucleotides to amplify signaling. cGAS is a soluble characterized by its Mab21 , which facilitates dsDNA binding in a sequence-independent manner. The enables a 2:1 of cGAS molecules to one dsDNA segment, preferentially engaging lengths greater than 40 base pairs to induce conformational changes and enzymatic activation. Upon binding, cGAS exhibits nucleotidyltransferase activity, catalyzing the conversion of ATP and GTP into the second messenger 2'3'-cyclic GMP-AMP (2'3'-cGAMP). This non-canonical cyclic dinucleotide binds with high affinity to downstream effectors, distinguishing it from bacterial cyclic di-GMP or di-AMP. STING is a localized to the (ER), featuring four transmembrane helices and a cytosolic C-terminal tail that includes a TBK1-binding domain. As a cyclic dinucleotide sensor, STING oligomerizes upon engagement, adopting a conformation that exposes interaction sites for signaling partners. The C-terminal domain's structural allows it to detect 2'3'-cGAMP with specificity, triggering conformational rearrangements essential for pathway propagation. Ligands for this pathway include microbial dsDNA from viruses or , as well as endogenous sources such as leaked (mtDNA) during cellular stress or damage. mtDNA, resembling bacterial DNA due to its circular and unmethylated nature, can escape into the and activate cGAS, highlighting the pathway's role in sterile inflammation. 2'3'-cGAMP acts as the primary second messenger, diffusing within cells or even transferring via gap junctions to neighboring cells. Activation begins with dsDNA binding to cGAS, forming a complex that allosterically unlocks its catalytic site to synthesize 2'3'-cGAMP. This second messenger then binds in the , inducing its oligomerization and subsequent trafficking to the Golgi apparatus via the COPII pathway. At the Golgi, STING recruits TBK1 through its C-terminal , positioning the complex for further interactions while undergoing palmitoylation for stability. Human exhibits substantial genetic diversity, with approximately 30% of the population carrying non-reference alleles such as the common (R71H-G230A-R293Q), which can modulate sensitivity and signaling efficiency. Gain-of-function mutations in , such as those causing STING-associated vasculopathy with onset in infancy (SAVI), lead to constitutive pathway activation and autoinflammatory conditions characterized by vasculopathy, , and elevated type I signatures. These variants underscore the pathway's delicate balance in preventing while mounting defenses against pathogens.

Secreted PRRs

Secreted pattern recognition receptors (PRRs), also known as humoral or soluble PRRs, are soluble proteins that circulate in extracellular fluids such as and mucosal secretions, enabling early detection and neutralization of pathogens before they interact with cellular receptors. These molecules primarily function by recognizing conserved microbial patterns in the , facilitating opsonization for , activation of the via the , and aggregation of pathogens to limit their spread. Unlike membrane-bound PRRs, secreted PRRs operate in the humoral arm of innate immunity, providing a first line of defense in body fluids. Collectins represent a major family of secreted PRRs, characterized by their collagen-like domains and C-type -like carbohydrate recognition domains (CRDs) that enable calcium-dependent binding to sugars. -binding lectin (MBL), a prototypical collectin, is produced in the liver and circulates in as oligomers of three polypeptide chains, each featuring an N-terminal cysteine-rich region, a collagen-like , an α-helical neck, and a C-terminal CRD. MBL binds to microbial carbohydrates such as , , and on , fungi, and parasites, promoting opsonization by enhancing pathogen uptake via and activating complement through association with MBL-associated serine proteases (MASPs). Other collectins include proteins A (SP-A) and D (SP-D), which are synthesized by alveolar type II and cells in the lungs, where they target similar carbohydrate ligands on respiratory pathogens like and house dust mites, aiding aggregation and clearance in mucosal surfaces. MBL deficiency, affecting up to 25% of certain populations with levels below 500 ng/ml, is associated with increased susceptibility to infections, underscoring its role in immune . Ficolins, another class of secreted PRRs, share structural similarities with collectins, featuring collagen-like domains but with fibrinogen-like recognition domains instead of CRDs, allowing binding to acetylated compounds. In humans, three ficolins exist: ficolin-1 (M-ficolin), ficolin-2 (L-ficolin, liver-produced), and ficolin-3 (H-ficolin, expressed in airways and liver). These oligomers recognize ligands such as and lipoteichoic acid on , facilitating opsonization and complement activation via the in a manner analogous to MBL. Ficolins often form complexes with MBL or collectins to amplify immune responses, and deficiencies in ficolin-2 have been linked to heightened risks of allergic and infectious diseases. Pentraxins, the third key family, are multimeric proteins defined by a conserved C-terminal pentraxin domain, existing as short (e.g., , CRP) or long (e.g., PTX3) forms. CRP, an synthesized in the liver, assembles into a pentameric disc structure that binds on bacterial surfaces and damaged cells, promoting opsonization, complement fixation via C1q, and pathogen aggregation during inflammation. PTX3, produced by immune cells like monocytes and endothelial cells rather than the liver, forms octamers with an extended N-terminal domain and recognizes fungal carbohydrates, bacterial lipids, and damage-associated molecular patterns (DAMPs) such as from necrotic cells. Both pentraxins elevate during —CRP serum levels can rise from baseline to over 100 mg/L, while PTX3 increases from ~2 ng/ml to 200-800 ng/ml—and genetic variants in PTX3 are associated with severe infections like . Collectins and ficolins exhibit evolutionary conservation as precursors to receptors (CLRs), sharing domain architectures that bridge soluble and membrane-bound pathogen recognition. These secreted PRRs coordinate briefly with membrane PRRs like CLRs by opsonizing pathogens for enhanced cellular uptake.

PRRs in

Receptor-like kinases (RLKs)

Receptor-like kinases (RLKs) are a major class of plasma membrane-localized pattern recognition receptors (PRRs) in , enabling the direct of microbial patterns and initiation of immune signaling. These transmembrane proteins typically feature an extracellular ligand-binding , a single transmembrane , and an intracellular serine/ that transduces signals upon . The extracellular domains often consist of leucine-rich repeats (LRRs) for recognizing diverse pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), while some RLKs, such as those in the LysM family, bind ligands like . In , the RLK family comprises over 600 members, representing approximately 2.5% of the protein-coding genes, with many implicated in immunity. Prominent examples of immune RLKs include FLAGELLIN SENSING 2 (FLS2), which detects the bacterial flagellin-derived flg22—a conserved 22-amino-acid that elicits pattern-triggered immunity (PTI). Similarly, ELONGATION FACTOR TU RECEPTOR (EFR) recognizes elf18, an 18-amino-acid from the bacterial elongation factor Tu (EF-Tu), triggering responses against bacterial invaders. For fungal pathogens, ELICITOR RECEPTOR KINASE 1 (CERK1), a LysM-RLK, binds chitin oligomers (typically 4-8 units in length) derived from fungal cell walls, essential for chitin-induced immunity in . These RLKs often function through heteromerization with co-receptors, such as BRI1-ASSOCIATED KINASE 1 (BAK1/SERK3), which enhances ligand binding and downstream events.00261-5.pdf) Upon perception, RLKs activate rapid immune responses, including a (ROS) burst via respiratory burst oxidase homologs (RBOHs), (MAPK) cascades that amplify signaling, and stomatal closure to restrict entry. For instance, flg22 binding to FLS2-BAK1 complexes phosphorylates and activates BOTRYTIS-INDUCED 1 (BIK1), leading to RBOHD-mediated ROS and MAPK within minutes. These responses contribute to PTI, restricting microbial proliferation at the cell surface. A distinctive feature of many immune RLKs, such as FLS2 and EFR, is their classification as non--aspartate (non-RD) kinases, where a conserved arginine in the activation loop is replaced, conferring resistance to manipulation by effectors that target RD kinases. This structural adaptation enhances the robustness of plant innate immunity.00067-5)00612-7)00261-5.pdf)

Receptor-like proteins (RLPs)

Receptor-like proteins (RLPs) are a class of pattern recognition receptors (PRRs) in that function as immune sensors at the cell surface, detecting microbial elicitors and initiating defense responses without possessing intrinsic kinase activity. These proteins are single-pass transmembrane receptors characterized by an extracellular for binding, a transmembrane region, and a short intracellular tail lacking a , distinguishing them from receptor-like kinases (RLKs).00111-0) The extracellular domains typically feature leucine-rich repeats (LRRs) or, in some cases, malectin-like domains that facilitate recognition of pathogen-derived molecules. Prominent examples of RLPs include EIX2 from , which binds xylanase elicitors from fungal pathogens to trigger defense signaling; Cf-9, which recognizes the Avr9 effector from the fungus fulvum; and Ve1, which detects the Ave1 effector from the vascular wilt fungus Verticillium dahliae. These RLPs perceive a range of ligands, such as fungal fragments, xylanases, and effectors like Pep1 from Verticillium species, enabling the detection of both conserved pathogen-associated molecular patterns (PAMPs) and specific factors. Upon binding, RLPs initiate pattern-triggered immunity (PTI) by forming heterodimers with co-receptor RLKs, such as SOBIR1, which transduces the signal downstream; this complex often involves additional partners like BAK1 for full activation. In , approximately 57 RLPs have been identified, contributing to both PTI and effector-triggered immunity (ETI), where some RLPs, like those in the Cf family, confer race-specific resistance by directly recognizing effectors. Recent engineering efforts have demonstrated the potential of chimeric RLPs to broaden disease resistance; for instance, modifications to the C-terminal domain of RLP23 enhance PTI responses against bacterial pathogens in without yield penalties, while synthetic immune receptor designs incorporating RLP modules confer durable, spectrum-wide protection against fungal and bacterial threats.

Signaling pathways

TLR signaling

Toll-like receptor (TLR) signaling is mediated by adapter proteins that transduce signals from the cytoplasmic Toll/interleukin-1 receptor (TIR) domains of activated TLRs, culminating in the activation of transcription factors that drive inflammatory and antiviral responses. Upon ligand-induced dimerization, the TIR domains of TLRs recruit specific adapters, primarily myeloid differentiation primary response 88 (MyD88) or TIR-domain-containing adapter-inducing interferon-β (TRIF), to initiate downstream cascades. These pathways are spatially regulated, with plasma membrane signaling favoring proinflammatory outputs and endosomal trafficking enabling interferon production. The MyD88-dependent pathway is utilized by all TLRs except TLR3 and involves TIR-TIR interactions that recruit MyD88, which in turn binds interleukin-1 receptor-associated kinases (IRAKs) via death domains. IRAK4 phosphorylates IRAK1, leading to its ubiquitination and association with TNF receptor-associated factor 6 (TRAF6), a ubiquitin ligase that activates the TAK1 kinase complex. TAK1 then phosphorylates IκB kinase (IKK) and mitogen-activated protein kinase (MAPK) pathways, resulting in nuclear factor kappa B (NF-κB) and activator protein 1 (AP-1) translocation to induce proinflammatory cytokines such as tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6). This pathway is essential for rapid innate immune activation against bacterial and fungal pathogens. In contrast, the TRIF-dependent pathway is engaged directly by TLR3 or, for TLR4, via the bridging adapter TRIF-related adapter molecule (TRAM). TRIF recruits TANK-binding kinase 1 (TBK1) and IκB kinase ε (IKKε), which phosphorylate interferon regulatory factor 3 (IRF3), promoting type I interferon (IFN-α/β) expression for antiviral defense. Concurrently, TRIF interacts with receptor-interacting protein 1 (RIP1) and TRAF6 to activate NF-κB via TAK1, amplifying inflammatory gene expression. Crosstalk between MyD88 and TRIF pathways in TLR4 signaling is spatially controlled, with endosomal localization of TLR4-TRAM-TRIF complexes favoring IRF3-driven IFN responses over plasma membrane MyD88 signaling. Downstream of both pathways, MAPK cascades—including p38, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (ERK)—are activated by TAK1, phosphorylating transcription factors like AP-1 to synergize with and for coordinated gene expression. Recent advances include 2025 structural studies on TIR domains and mechanisms of biased agonism, where ligands selectively favor MyD88 or TRIF pathways, offering potential for targeted . Negative regulation prevents excessive inflammation, with single immunoglobulin IL-1-related receptor (SIGIRR) acting as a decoy by sequestering MyD88 and IRAKs, and suppression of tumorigenicity 2 (ST2) inhibiting TIR interactions in IL-1R/TLR family signaling.

NLR and ALR signaling

NLRs and ALRs form multiprotein complexes known as upon recognition of intracellular pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). In the case of , a prominent NLR, ligand-induced oligomerization exposes its pyrin domain (PYD), which recruits the adaptor protein apoptosis-associated speck-like protein containing a (ASC) through PYD-PYD interactions. ASC then oligomerizes into filaments, recruiting pro-caspase-1 via CARD-CARD interactions, leading to proximity-induced autoproteolysis and generation of active caspase-1. Similarly, for ALRs such as AIM2, double-stranded DNA binding triggers AIM2 oligomerization, directly recruiting ASC through PYD-PYD contacts to form the AIM2 , which in turn assembles pro-caspase-1 for activation. Active caspase-1 processes its substrates to drive inflammatory responses. It cleaves the proinflammatory precursors pro-IL-1β and pro-IL-18 at specific aspartate residues, yielding mature IL-1β and IL-18 that are secreted to amplify innate immunity. Additionally, caspase-1 proteolytically activates gasdermin D (GSDMD) by cleaving its autoinhibitory linker, releasing the N-terminal GSDMD fragment that oligomerizes and forms plasma membrane pores, leading to —an inflammatory form of lytic that releases intracellular contents and enhances clearance. is a key outcome of both NLRP3 and AIM2 , distinguishing them from non-lytic immune signaling pathways. NLRP3 activation follows a two-signal model to ensure controlled inflammasome assembly. The priming signal (signal 1), often delivered by (TLR) ligands or cytokines, activates transcription factor, upregulating NLRP3 and pro-IL-1β expression. The activation signal (signal 2) then triggers NLRP3 oligomerization through diverse mechanisms, including (K+) efflux, (ROS) generation, or release of (mtDNA), converging on NEK7 kinase-dependent NLRP3-ASC interactions. This model prevents aberrant activation while responding to a broad array of stimuli, such as crystals, toxins, or metabolic stressors. For ALRs, AIM2 directly integrates with ASC to form a functional , as its HIN domain binds dsDNA while the PYD scaffolds ASC recruitment without requiring additional adaptors. IFI16, another ALR, cooperates with AIM2 in certain contexts, such as during cytosolic DNA sensing in viral infections, where IFI16 enhances AIM2 assembly by facilitating DNA delivery or stabilizing complexes, though it can also exert regulatory effects. This integration allows ALRs to mount robust responses to threats. A non-canonical inflammasome pathway, independent of or AIM2, involves human -4 and -5 (or mouse -11), which directly sense cytosolic (LPS) from via CARD-lipid A interactions. This binding induces caspase oligomerization and autoprocessing, enabling cleavage of GSDMD to form pores and drive , while also indirectly promoting IL-1β release through secondary -1 activation. Unlike canonical pathways, this route can lead to IL-1 without full in some cell types, providing a rapid barrier against intracellular bacterial replication.

RLR and cGAS-STING signaling

Upon activation by viral double-stranded RNA ligands, RIG-I and undergo conformational changes that expose their N-terminal domains, enabling direct interaction with the adaptor protein MAVS on the outer mitochondrial membrane or . MAVS then oligomerizes into prion-like aggregates, recruiting TRAF3 and TRAF6 to form a signaling complex that activates the kinases TBK1 and IKKε, which in turn phosphorylate and dimerize for translocation to the and induction of IFN-β transcription. This pathway specifically detects short 5'-triphosphorylated RNAs for RIG-I and longer double-stranded RNAs for , initiating a robust antiviral response. In parallel, the cGAS-STING pathway responds to cytosolic double-stranded DNA from pathogens or damaged cells, where cGAS synthesizes the second messenger 2'3'-cyclic GMP-AMP (cGAMP). cGAMP binding to STING on the endoplasmic reticulum triggers STING polymerization and translocation to perinuclear sites, where it recruits TBK1 to phosphorylate both STING and IRF3, promoting IRF3 dimerization and type I IFN production, while TRAF6 activation leads to NF-κB-mediated proinflammatory cytokine expression. This mechanism ensures rapid detection of DNA threats, distinct from RNA sensing by RLRs. Both pathways converge on type I IFN signaling through the IFNAR receptor, which activates JAK1 and TYK2 kinases to phosphorylate , forming a complex with IRF9 that drives transcription of interferon-stimulated genes (ISGs) such as MxA and , establishing an antiviral state. is mediated by ISGs like ISG15, which ubiquitinates pathway components to dampen excessive signaling and prevent . Activation of RLR and cGAS-STING pathways induces not only IFN production but also in infected cells via IRF3-mediated upregulation of proapoptotic genes, limiting viral spread. Recent studies highlight metabolic rewiring, including MAVS-dependent upregulation of and shifts to the , supporting energy demands for immune activation. Crosstalk between RLRs and cGAS-STING enhances responses, as RLR activation promotes release into the , priming cGAS for amplified IFN production during infections.

CLR signaling

receptors (CLRs) employ distinct signaling pathways to orchestrate antifungal immunity and , primarily through ITAM-like () and non-ITAM mechanisms. In the ITAM-coupled pathway, exemplified by Dectin-1 and Dectin-2, ligand binding to the receptor's hemITAM motif induces phosphorylation of spleen tyrosine kinase (Syk) by family kinases, initiating downstream signaling. This cascade recruits the adaptor protein CARD9, which complexes with Bcl10 and MALT1 to activate , promoting the production of Th17-polarizing cytokines such as IL-17 and essential for antifungal defense. These responses drive differentiation and recruitment, highlighting the pathway's role in mucosal immunity against fungi like . In contrast, non-ITAM pathways in CLRs like DC-SIGN mediate anti-inflammatory tolerance. Upon ligand engagement, DC-SIGN activates Raf-1 kinase, which acetylates the p65 subunit to upregulate IL-10 production, suppressing excessive inflammation and promoting responses. Similarly, the facilitates endocytosis into lysosomes for degradation without eliciting strong proinflammatory signals, prioritizing clearance over activation in myeloid cells. CLR signaling yields diverse cellular outcomes tailored to pathogen context. ITAM-coupled CLRs such as Dectin-1 trigger reactive oxygen species (ROS) generation and enhance phagocytosis in macrophages and neutrophils, directly combating fungal invasion. Activation of mitogen-activated protein kinase (MAPK) pathways, particularly ERK, further supports dendritic cell maturation and antigen presentation. Group II CLRs, including the asialoglycoprotein receptor (ASGPR), primarily mediate glycoprotein clearance via endocytosis with minimal inflammatory signaling, focusing on homeostasis rather than immunity. Regulation of CLR signaling ensures balanced responses, with unique mechanisms targeting key components. Fungal pathogens deploy phosphatases to inhibit Syk activity, dampening host defenses and facilitating infection persistence. Recent 2025 research reveals epigenetic modifications, such as histone acetylation induced by CLR activation, in the context of trained immunity. CLR pathways also exhibit with Toll-like receptors (TLRs) to amplify adaptive immunity. For instance, Dectin-2 synergizes with TLRs to boost IL-12 and IFN-γ production, enhancing Th1 responses against fungi while integrating and bacterial signals.

Regulation of PRR activity

Positive regulation

Positive regulation of pattern recognition receptors (PRRs) involves multiple mechanisms that enhance their sensitivity, activation, and downstream signaling to amplify innate immune responses. Post-translational modifications play a central role in this process. For instance, K63-linked ubiquitination of TNF receptor-associated factor 6 (TRAF6) occurs through its auto-ubiquitination, which is essential for activating downstream pathways in (TLR) signaling, thereby promoting and MAPK activation. Similarly, tripartite motif-containing protein 25 (TRIM25) mediates K63-linked ubiquitination of the CARD domains of retinoic acid-inducible gene I (RIG-I), facilitating its oligomerization and interaction with (MAVS) to induce type I production. events also contribute, such as the autophosphorylation of interleukin-1 receptor-associated kinase 4 (IRAK4) upon recruitment to the MyD88 complex in TLR signaling, which enables subsequent of IRAK1 and amplification of inflammatory responses. Co-factors further potentiate PRR function by aiding ligand binding and complex assembly. In mammals, lipopolysaccharide-binding protein (LBP) extracts (LPS) from bacterial membranes and transfers it to , which then delivers it to the MD-2/TLR4 complex, enhancing LPS recognition and TLR4 dimerization for robust signaling. In plants, BAK1 (BRASSINOSTEROID INSENSITIVE 1-ASSOCIATED RECEPTOR KINASE 1) acts as a co-receptor for receptor-like kinases (RLKs), while SUPPRESSOR OF BIR1-1 (SOBIR1) stabilizes receptor-like protein (RLP) complexes, both promoting phosphorylation cascades that initiate pattern-triggered immunity (PTI). Priming mechanisms transcriptionally and epigenetically upregulate PRR expression or components to heighten responsiveness. Activation of by TLRs induces transcriptional upregulation of , priming the for subsequent activation by danger signals. Epigenetic modifications, such as histone acetylation at (IFN) promoters following cGAS-STING activation, facilitate opening and enhance type I IFN transcription, amplifying antiviral responses. Feedback loops sustain and intensify PRR signaling. Autocrine type I IFN signaling upregulates expression of RIG-I-like receptors (RLRs) and cGAS through IFN-stimulated response elements, creating a positive amplification circuit for prolonged innate immunity. Recent studies highlight metabolic regulation, where nuclear translocation boosts assembly by inhibiting and enhancing IL-1β processing in macrophages. In plants, hyperactivation of RD-type kinases, such as receptor-like cytoplasmic kinases (RLCKs), during PTI involves rapid bursts that propagate defense signaling, including production and callose deposition.

Negative regulation and tolerance

Negative regulation of receptors (PRRs) is essential to prevent excessive , maintain immune , and avoid damage from overzealous responses to microbial or endogenous ligands. These mechanisms include receptors that sequester ligands or block signaling adaptors, degradative pathways that target PRR components for breakdown, and adaptive processes that dampen repeated stimulations. In both and , such controls ensure balanced immunity, with failures leading to pathological conditions like or autoinflammation. Decoy receptors serve as inhibitory traps for PRR ligands or signaling intermediates. Single immunoglobulin IL-1-related receptor (SIGIRR), also known as TIR8, acts as a by associating with Toll-like receptors (TLRs) and interleukin-1 receptors (IL-1Rs), disrupting their TIR domain interactions and suppressing downstream activation, particularly for TLR4-mediated responses to (LPS). Similarly, IL-1R2 functions as a soluble or membrane-bound that binds IL-1β without , preventing its interaction with signaling-competent IL-1R1. Soluble ectodomains of TLR2 and TLR4, shed from the cell surface, circulate and competitively inhibit to membrane-bound receptors, thereby attenuating TLR signaling in . Degradative mechanisms further limit PRR activity through and . K48-linked ubiquitination targets PRR pathway components for proteasomal degradation; for instance, tripartite motif-containing protein 29 (TRIM29) ubiquitinates (STING) at lysine 370, promoting its degradation and curtailing type I production in the cGAS-STING pathway. Phosphatases also counteract activation by removing phosphate groups; protein tyrosine phosphatase non-receptor type 6 (PTPN6, or SHP-1) dephosphorylates spleen tyrosine kinase (Syk) in C-type lectin receptor (CLR) signaling, thereby inhibiting downstream spleen tyrosine kinase (Syk)-dependent pathways like CARD9-Bcl10-MALT1 activation. Tolerance mechanisms induce hyporesponsiveness to repeated PRR stimulation, preserving during chronic exposure. Endotoxin tolerance, observed after initial LPS challenge, involves interleukin-1 receptor-associated kinase M (IRAK-M) upregulation, which inhibits IRAK1/4 recruitment to MyD88 in TLR4 signaling, reducing and MAPK activation without abolishing detection. Microbiota-induced tolerance employs A20 (TNFAIP3), a deubiquitinase that removes K63-linked from NLR family proteins like , preventing assembly and IL-1β release in the gut to tolerate commensal . In , analogous negative controls modulate PRR responses to prevent growth inhibition from sustained immunity. Receptor-like proteins (RLPs), such as those recognizing fungal , undergo ligand-induced via clathrin-mediated pathways, internalizing and degrading the complexes to terminate signaling and allow receptor . Effector-triggered immunity (ETI), mediated by nucleotide-binding receptors (NLRs), features loops, including by kinases like BIK1 that balance activation and suppression, ensuring rapid but contained hypersensitive responses without systemic overreaction. Dysregulation of these inhibitory processes underlies pathological . Loss of TLR4 negative regulation, such as reduced SIGIRR or IRAK-M expression, exacerbates cytokine storms in by allowing unchecked MyD88-dependent signaling. Gain-of-function in NLRP3 impair its ubiquitination and degradation, leading to spontaneous activation and autoinflammatory syndromes like cryopyrin-associated periodic syndromes (CAPS). Recent studies link metabolic dysregulation to impaired STING inhibition; in , diminished DsbA-L-mediated negative control results in persistent cGAS-STING activation, driving adipose and .

Evolution of PRRs

Origins and conservation

Pattern recognition receptors (PRRs) trace their origins to prokaryotic precursors, where (LRR) domains function in bacterial systems against phages. Bacterial NLR-related proteins, featuring NACHT domains and regions analogous to LRRs, detect phage and abortive to protect host cells. Similarly, Toll/interleukin-1 receptor (TIR) domains in prokaryotic NADase effectors, such as those from species, hydrolyze NAD+ to disrupt host during , highlighting enzymatic roles predating eukaryotic immunity. These domains predate the prokaryote-eukaryote divergence, with LRR and nucleotide-binding site (NBS) motifs evident in ancient bacterial genomes. In early eukaryotes like fungi and protists, PRR-like systems emerge with conserved motifs. The amoebozoan Dictyostelium discoideum employs TirA, a TIR domain-containing protein homologous to Toll-like receptors (TLRs), to recognize bacterial (LPS) and enhance bactericidal activity via MAPK signaling. NACHT domains, central to NLR function, are also conserved in choanoflagellates, the closest unicellular relatives to , where they pair with LRRs in proteins potentially involved in self/non-self , indicating pre-metazoan origins of cytosolic PRR architecture. These findings suggest PRR components evolved in unicellular eukaryotes for microbial sensing before multicellularity. Invertebrate PRRs build on these foundations, with conserved yet simplified repertoires. features a single TLR ortholog, the receptor, which detects fungal and Gram-positive bacterial patterns to activate production. Instead of canonical NLRs, possesses 13 peptidoglycan recognition protein (PGRP) genes encoding LRR-containing sensors that recognize bacterial components and initiate immune signaling. lacks TLRs but expresses a large family of receptors (CLRs), which bind microbial glycans to modulate innate defenses and avoidance. Common downstream pathways include homologs like Relish in , which regulates , and (IFN)-like molecules such as Vago in mosquitoes, a secreted that restricts via JAK-STAT signaling. PRR diversification accelerated around 600 million years ago (Mya), coinciding with the emergence of multicellularity in eukaryotes during the period, enabling coordinated immune responses in complex organisms. Recent phylogenomic analyses, including those from 2025, reveal (HGT) as a key driver in PRR evolution, with microbial-to-plant transfers contributing to the origins of land immune receptors like LRR-RLKs during streptophyte diversification. In vertebrates, PRR families underwent further expansion, adapting to diverse pathogens.

Diversification in metazoans and plants

In metazoans, the diversification of receptors (PRRs) has been marked by extensive events, particularly in vertebrates. Toll-like receptors (TLRs), for instance, expanded from a single ortholog in invertebrates like to 10 functional TLRs in humans, driven by tandem and whole-genome duplications that allowed specialization for diverse microbial ligands. Similarly, NOD-like receptors (NLRs) underwent a dramatic expansion post-chordate divergence, with teleost fish harboring approximately 30–400 NLR genes compared to 22–33 in mammals, reflecting lineage-specific adaptations to aquatic pathogens. AIM2-like receptors (ALRs) are absent in fish and emerged later in mammalian evolution, whereas RIG-I-like receptors (RLRs) originated early in vertebrate evolution and are present across and jawless vertebrates, including non-teleost fish, to enhance intracellular sensing of viral and bacterial nucleic acids. In , PRR repertoires radiated prominently during the transition to land, with receptor-like kinases (RLKs) expanding to over 600 in —contrasting with only a handful in algal ancestors—through serial duplications that enabled of environmental cues. Receptor-like proteins (RLPs), numbering around 50 in , co-evolved with RLKs to form ligand-binding complexes, while LysM-domain RLKs specialized for fungal , recognizing and Nod factors in a manner predating angiosperm diversification. Non-arginine-aspartate (non-RD) kinases, a hallmark of plant immunity, uniquely evolved within RLK signaling to bypass autoinhibition and trigger rapid defense responses, distinguishing them from RD kinases in other contexts. These expansions were propelled by , domain shuffling, and pathogen-driven selection. In , TIR-NB-LRR domains shuffled via unequal recombination to generate resistance (R) genes, enhancing effector amid co-evolutionary arms races. In animals, pathogen pressure, particularly from viruses, accelerated diversification of intracellular sensors like RLRs, with positive selection on RNA-binding domains to counter evolving viral evasion strategies. Comparatively, metazoan PRRs emphasize intracellular surveillance, with NLRs and RLRs detecting endocytosed threats in the , whereas prioritize extracellular RLKs for immediate cell wall contact with microbes, reflecting sessile lifestyles and mobility differences. Recent synthetic biology efforts in 2025 have engineered convergent PRRs across kingdoms, demonstrating how modular designs can recapitulate natural diversification for broad-spectrum immunity.

Clinical and therapeutic implications

Role in infectious diseases

Pattern recognition receptors (PRRs) play a critical role in host defense against infectious diseases by detecting microbial patterns and initiating immune responses. Dysregulation of PRR function, either through genetic deficiencies or pathogen-mediated interference, significantly impacts susceptibility to infections. For instance, polymorphisms in the TLR4 gene, such as Asp299Gly and Thr399Ile, have been associated with hyporesponsiveness to (LPS), leading to increased risk of Gram-negative bacterial . Similarly, variants in the gene, including the Arg587Arg , have been associated with heightened susceptibility to pulmonary in certain populations, such as the Chinese Han, by impairing recognition of muramyl dipeptide from , thereby reducing activation and production. These deficiencies highlight how PRR impairments compromise innate immunity, allowing pathogens to establish persistent infections. Conversely, hyperactivation of PRRs can exacerbate disease severity through excessive . In chronic infection, sustained stimulation of endosomal TLR7 and TLR9 by viral nucleic acids contributes to persistent immune activation, driving dysfunction and ongoing type I production that fuels T-cell exhaustion and disease progression. During severe , overactivation of the by components, such as viroporins and , triggers a characterized by elevated IL-1β and IL-18 levels, leading to acute respiratory distress and multi-organ failure. Pathogens have evolved mechanisms to evade PRR detection, further underscoring their role in infectious outcomes. employs its NS3/4A to cleave (MAVS), thereby inhibiting RIG-I-mediated type I responses and promoting viral persistence in hepatocytes. Likewise, secretes IpaH ubiquitin ligases, such as IpaH9.8, which target components of the NOD1 signaling pathway, including NEMO, to dampen activation and evade peptidoglycan-induced inflammation during epithelial cell invasion. In , analogous PRR dysfunction increases vulnerability to bacterial pathogens. Mutations in the FLS2 receptor-like kinase, which perceives from bacteria like , result in enhanced susceptibility to disease, as seen in and mutants that fail to mount effective bursts and defense gene expression. Recent insights into emerging zoonoses, such as the 2022-2025 outbreaks, reveal the cGAS-STING pathway's pivotal role in sensing monkeypox virus DNA, activating type I interferons to limit in mice and nonhuman primates; disruptions in this pathway correlate with increased pathogenicity, emphasizing PRRs' ongoing relevance in controlling infections.

Involvement in autoimmune and inflammatory disorders

Pattern recognition receptors (PRRs) play a critical role in autoimmune and inflammatory disorders when they are aberrantly activated by endogenous damage-associated molecular patterns (DAMPs), leading to chronic inflammation and loss of self-tolerance. These self-molecules, released during cellular or , mimic pathogen-associated molecular patterns (PAMPs) and trigger PRR signaling pathways, resulting in excessive production of pro-inflammatory cytokines such as type I interferons (IFNs) and interleukin-1β (IL-1β). In autoinflammatory conditions, this activation stems from genetic mutations causing gain-of-function in PRRs or their downstream effectors, while autoimmune diseases often involve environmental or epigenetic factors that prime PRRs for to self-antigens. A prominent mechanism involves mitochondrial DNA (mtDNA) acting as a DAMP to activate the cytosolic PRR cyclic GMP-AMP synthase (cGAS) in systemic lupus erythematosus (SLE). Oxidized mtDNA leaks into the cytosol during mitochondrial dysfunction, binding cGAS to produce cGAMP, which activates the STING pathway and induces type I IFN production, exacerbating autoantibody formation and tissue damage in SLE patients. Similarly, in psoriasis, S100 proteins (e.g., S100A8/A9, also known as calprotectin) serve as DAMPs that bind Toll-like receptor 4 (TLR4) on keratinocytes and immune cells, promoting NF-κB activation and the release of IL-17 and IL-23, which drive epidermal hyperplasia and chronic skin inflammation. Several monogenic autoinflammatory diseases highlight the direct impact of PRR dysregulation. Cryopyrin-associated periodic syndromes (CAPS), a group of cryopyrinopathies, arise from gain-of-function mutations in the NLR family pyrin domain-containing 3 () gene, leading to constitutive activation of the inflammasome, excessive IL-1β secretion, and recurrent fever, urticaria, and joint pain. (FMF) results from mutations in the gene encoding pyrin, an NLR-like protein, which hyperactivates the pyrin in response to effectors or cellular stress, causing episodic and due to unchecked IL-1β release. In Aicardi-Goutières syndrome (AGS), deficiency in the TREX1 leads to accumulation of cytosolic self-DNA, which activates cGAS-STING signaling, mimicking viral and producing high levels of type I IFNs that cause severe and calcifications in infants. Breakdown of immune tolerance often involves defective negative regulation of PRR pathways. In rheumatoid arthritis (RA), hyperactivity of interleukin-1 receptor-associated kinase 1 (IRAK1), a key adaptor in TLR and IL-1R signaling, amplifies and MAPK pathways in synovial fibroblasts and macrophages, sustaining joint inflammation and erosion. Epigenetic modifications contribute to PRR priming in SLE, enhancing TLR7 and TLR9 expression and lowering the threshold for IFN-α production in response to self-nucleic acids. An analogous phenomenon occurs in , where misfiring of (R) genes—encoding NLR proteins similar to animal PRRs—triggers a (HR)-like . Gain-of-function mutations in genes like SNC1 lead to inappropriate recognition of self-components, activating salicylic acid-dependent defenses and localized , mirroring DAMP-driven in metazoans. Recent insights as of 2025 emphasize PRR-metabolic links in (IBD), particularly , where mutations impair bacterial sensing, leading to dysbiosis with reduced Firmicutes and increased Proteobacteria, which perpetuates NOD2-independent PRR activation (e.g., via TLRs) and barrier dysfunction. This dysbiosis amplifies chronic mucosal , highlighting NOD2's role in maintaining microbial to prevent autoinflammatory flares.

Applications in immunotherapy and vaccines

Pattern recognition receptor (PRR) agonists have emerged as potent tools in by mimicking pathogen-associated molecular patterns to stimulate innate immune responses, thereby enhancing adaptive immunity against tumors. For instance, monophosphoryl lipid A (MPL), a detoxified of that acts as a TLR4 , is incorporated into the vaccine for human papillomavirus (HPV), where it promotes robust antibody responses and long-term protection against precursors. Similarly, , a synthetic TLR7/8 , is FDA-approved for topical treatment of and , inducing local and tumor regression through interferon-alpha production and activation of plasmacytoid dendritic cells. In advanced settings, cyclic dinucleotides like ADU-S100 (also known as MIW815), which activate the cGAS-STING pathway, have been evaluated in phase I/II clinical trials for intratumoral injection in solid tumors such as and , demonstrating enhanced T-cell infiltration and partial responses when combined with checkpoint inhibitors. PRR antagonists, conversely, offer therapeutic value by dampening excessive in immunopathologies. Monoclonal antibodies targeting TLR4, such as the investigational agent NI-0101, have shown promise in blocking endotoxin-induced storms in early clinical trials, including phase I studies demonstrating prevention of release in healthy volunteers challenged with LPS. For inflammasome inhibition, MCC950, a selective small-molecule , has demonstrated efficacy in preclinical and early clinical studies for cryopyrin-associated periodic syndromes (CAPS) by preventing IL-1β release and ameliorating autoinflammatory flares, while in models, it suppresses urate crystal-induced without broad . In vaccine development, PRR-targeted s amplify antigen-specific responses, particularly for challenging pathogens. Trehalose-6,6'-dibehenate (TDB), a synthetic of the receptor (CLR) Mincle, is formulated in the CAF01 system for (TB) vaccines, where it induces Th1/Th17 polarization and sustained CD4+ T-cell memory in preclinical models and ongoing phase II trials. Likewise, polyinosinic:polycytidylic acid (poly I:C), a mimic of viral double-stranded that engages RIG-I-like receptors (RLRs) and TLR3, serves as an in vaccines, boosting cross-protective humoral and cellular immunity in human trials by promoting type I secretion and maturation. Beyond human applications, engineered PRRs hold potential in and emerging technologies. In , receptor-like kinases (RLKs), a class of PRRs, have been synthetically modified to broaden ; for example, chimeric LRR-RLK constructs engineered in 2025 studies confer enhanced resistance to fungal and bacterial threats in crops like and by integrating multiple ligand-binding domains for improved specificity and immunity activation. These advancements inspire approaches in design. As of 2025, STING agonists like cyclic dinucleotides continue to advance in phase III trials for , showing improved response rates in combination therapies. Despite these advances, PRR-based therapies face significant challenges, including achieving receptor specificity to prevent off-target activation that could trigger autoimmunity, as nonspecific TLR agonism has been linked to systemic inflammatory adverse events in up to 20% of trial participants. Combination strategies mitigate such risks; for example, pairing TLR9 agonists like CpG oligodeoxynucleotides with PD-1 inhibitors enhances antitumor efficacy in melanoma and non-small cell lung cancer trials by synergistically boosting CD8+ T-cell function while limiting exhaustion, achieving objective response rates of 25-40% in resistant populations. Ongoing research emphasizes dose optimization and delivery systems, such as nanoparticle encapsulation, to balance potency and safety in clinical translation. Pattern recognition receptors (PRRs) play dual roles in cancer, with certain pathways promoting tumorigenesis through chronic inflammation while others exert suppressive effects via immune activation. Toll-like receptors 2 and 4 (TLR2 and TLR4) contribute to tumor progression in colorectal cancer by recognizing damage-associated molecular patterns (DAMPs) released from dying cells, leading to NF-κB activation and the production of pro-inflammatory cytokines that foster a tumor-supportive microenvironment. Similarly, the NLRP3 inflammasome drives inflammation in pancreatic cancer, where its activation by tumor-derived factors enhances IL-1β secretion, promoting tumor cell proliferation and immune evasion. In contrast, the STING pathway suppresses tumor growth by sensing cytosolic DNA and inducing type I interferons (IFNs), which activate antitumor immune responses; this mechanism underpins type I IFN-based therapies in various cancers, including melanoma and lung tumors. In neurodegeneration, PRR dysregulation amplifies and neuronal damage through persistent recognition of misfolded proteins and cellular debris. In , amyloid-β (Aβ) aggregates activate TLR2 and TLR4 on , triggering signaling and IL-1β release, which exacerbates plaque formation and synaptic loss. The is implicated in , where α-synuclein fibrils induce its assembly in , leading to caspase-1 activation, IL-1β maturation, and degeneration; inhibition of reduces α-synuclein aggregates and motor deficits in preclinical models. Chronic cGAS-STING pathway activation contributes to (ALS) by detecting released from damaged motor neurons, resulting in type I IFN-driven and accelerated disease progression. PRRs also link to metabolic diseases by sensing metabolic stressors and microbial signals, perpetuating low-grade inflammation. The NLRP3 inflammasome is central to and (T2D), activated by saturated fatty acids in to produce IL-1β, which impairs insulin signaling and promotes β-cell dysfunction; NLRP3-deficient models resist high-fat diet-induced . TLR4 on adipocytes senses free fatty acids during high-fat feeding, initiating NF-κB-mediated inflammation that drives systemic metabolic dysregulation. In non-alcoholic (NAFLD), PRR activation, including NLRP3 and AIM2 , responds to lipid-derived DAMPs, with epigenetic modifications like miR-204-3p suppression of TLR4 signaling exacerbating hepatic and , as highlighted in recent reviews. Mechanisms underlying these associations involve persistent DAMP signaling and host-microbe interactions. High-mobility group box 1 (), a prototypical DAMP, sustains PRR activation in tumors by TLR4 and , promoting NF-κB-driven and . In , the gut microbiota-PRR axis amplifies , where dysbiosis-derived lipopolysaccharides activate TLR4, contributing to and NAFLD progression via crosstalk. Therapeutically, targeting PRRs holds promise for these conditions. agonists, such as cyclic dinucleotides, enhance antitumor immunity in cancer by boosting type I IFN production and T-cell infiltration, with ongoing clinical trials in solid tumors. inhibitors, including MCC950 and OLT1177, show efficacy in preclinical neurodegeneration models by mitigating IL-1β-driven damage in Parkinson's and , and in metabolic trials for T2D and NAFLD by reducing adipose and improving . As of November 2025, OLT1177 is in phase II trials for , demonstrating reduced without affecting levels.

References

  1. [1]
    Pattern recognition receptors in health and diseases - Nature
    Aug 4, 2021 · Pattern recognition receptors (PRRs) are a class of receptors that can directly recognize the specific molecular structures on the surface of pathogens.
  2. [2]
    Pattern recognition receptors: function, regulation and therapeutic ...
    Jul 11, 2025 · Pattern recognition receptors (PRRs) are sensors in the immune system, detecting pathogen-associated molecular patterns (PAMPs) and ...
  3. [3]
    The Nobel Prize in Physiology or Medicine 2011 - Advanced ...
    Janeway to postulate the existence of a recognition system for microbial components (“pattern recognition receptors”) (12), different from the antigen ...
  4. [4]
    An Overview of Pathogen Recognition Receptors for Innate ... - NIH
    Pathogen recognition receptors (PRRs) are a class of germ line-encoded receptors that recognize pathogen-associated molecular patterns (PAMPs).
  5. [5]
    Pattern recognition receptors: function, regulation and therapeutic ...
    Jul 11, 2025 · Building on this, Janeway and Medzhitov identified a human counterpart to Toll, initially termed hToll and later known as toll-like receptor 4 ( ...Missing: history | Show results with:history
  6. [6]
    Approaching the asymptote? Evolution and revolution in immunology
    Approaching the asymptote? Evolution and revolution in immunology ... Cold Spring Harb Symp Quant Biol. 1989:54 Pt 1:1-13. doi: 10.1101/sqb.1989.054.01.003.
  7. [7]
    The dorsoventral regulatory gene cassette spätzle/Toll ... - PubMed
    The dorsoventral regulatory gene cassette spätzle/Toll/cactus controls the potent antifungal response in Drosophila adults ... Cell. 1996 Sep 20;86(6):973-83. doi ...
  8. [8]
    A human homologue of the Drosophila Toll protein signals activation ...
    We report here the cloning and characterization of a human homologue of the Drosophila toll protein (Toll) which has been shown to induce the innate immune ...
  9. [9]
    Cyclic GMP-AMP synthase is a cytosolic DNA sensor that ... - PubMed
    Feb 15, 2013 · These results indicate that cGAS is a cytosolic DNA sensor that induces interferons by producing the second messenger cGAMP.
  10. [10]
    Innate immune recognition - PubMed
    Innate immune recognition relies on a limited number of germline-encoded receptors. These receptors evolved to recognize conserved products of microbial ...
  11. [11]
  12. [12]
    The Structural Biology of Toll-Like Receptors - PMC - PubMed Central
    The receptors share a common structural framework in their extracellular, ligand-binding domains. These domains all adopt horseshoe-shaped structures built ...
  13. [13]
  14. [14]
    Toll-Like Receptors and Relevant Emerging Therapeutics with ...
    Expression, localization, and ligands of Toll-like receptors (TLRs). Receptor, Microbial Ligand(s), Microbial Ligand Source, Endogenous Ligand(s), Adaptor, TLR ...
  15. [15]
    Signalling through C-type lectin receptors: shaping immune responses
    In this Review we discuss the transmembrane CLRs that function as PRRs. These CLRs can be divided into two groups: group I CLRs belong to the mannose receptor ...
  16. [16]
  17. [17]
    C-Type Lectin Receptor Mediated Modulation of T2 Immune ...
    Feb 1, 2023 · In this review, we highlight the capacity of CLRs to recognize carbohydrates in common allergens triggering different signaling pathways ...
  18. [18]
    Various Tastes of Sugar: The Potential of Glycosylation in Targeting ...
    C-type lectin receptors (CLRs) are important in several immune regulatory processes. These receptors recognize glycans expressed by host cells or by pathogens.
  19. [19]
    The NLRP3 Inflammasome: An Overview of Mechanisms of ... - MDPI
    The NLRP3 inflammasome is a critical component of the innate immune system that mediates caspase-1 activation and the secretion of proinflammatory cytokines.
  20. [20]
    A frameshift mutation in NOD2 associated with susceptibility ... - Nature
    A frameshift mutation caused by a cytosine insertion, 3020insC, which is expected to encode a truncated NOD2 protein, is associated with Crohn's disease.
  21. [21]
    RIG-I-like receptors: their regulation and roles in RNA sensing - Nature
    Mar 13, 2020 · RIG-I-like receptors (RLRs) are key sensors of virus infection, mediating the transcriptional induction of type I interferons and other genes that collectively ...
  22. [22]
    Immune signaling by RIG-I-like receptors - PMC - PubMed Central
    The RIG-I-like receptors (RLRs) RIG-I, MDA5, and LGP2 play a major role in pathogen sensing of RNA virus infection to initiate and modulate antiviral immunity.Missing: seminal papers
  23. [23]
    Comparative Structure and Function Analysis of the RIG-I-Like ...
    This review article discusses the similarities and differences between RIG-I and MDA5 from multiple perspectives, including their structures, evolution and ...Missing: seminal papers
  24. [24]
    Length-dependent recognition of double-stranded ribonucleic acids ...
    Jun 30, 2008 · We found that the MDA5 ligand, poly I:C, was converted to a RIG-I ligand after shortening of the dsRNA length. RIG-I and MDA5 preferentially ...
  25. [25]
    AIM2 inflammasome activation and regulation: A structural perspective
    ALRs are usually bipartite proteins: an N-terminal PYD domain followed by one or two HIN domains with the exception of p202, which lacks the PYD domain (Fig. 1A) ...Missing: seminal papers
  26. [26]
    Filament assembly underpins the double-stranded DNA specificity of ...
    Mar 2, 2023 · Absent-in-melanoma-2 (AIM2)-like receptors (ALRs) are a family of innate immune sensors that play vital yet versatile roles in defense ...Missing: seminal | Show results with:seminal
  27. [27]
    Immunobiology and Structural Biology of AIM2 Inflammasome - PMC
    In this article, we briefly review the identification of AIM2 as a cytoplasmic DNA sensor, summarize the importance of AIM2 inflammasome in infections and ...Missing: seminal | Show results with:seminal
  28. [28]
    AIM2-Like Receptors Positively and Negatively Regulate the ...
    Jul 5, 2017 · Structures of the HIN domain:DNA complexes reveal ligand binding and activation mechanisms of the AIM2 inflammasome and IFI16 receptor.
  29. [29]
    Nuclear AIM2‐Like Receptors Drive Genotoxic Tissue Injury by ...
    Oct 18, 2021 · The ALR family constitutes four members in humans (AIM2, IFI16, IFIX (PYHIN1), and MNDA) and thirteen in mice (including Aim2, p202, p203, p204 ...Introduction · Results · Conclusion and Discussion · Experimental Section
  30. [30]
    The AIM2 inflammasome is essential for host defense ... - Nature
    Mar 28, 2010 · These receptors recognize microbial products from bacteria, viruses, fungi and parasites and in most cases trigger signaling pathways that ...<|control11|><|separator|>
  31. [31]
    Assembly mechanism of the inflammasome sensor AIM2 revealed ...
    Dec 2, 2023 · An increase in assembly rates close to 700-fold has been observed when extending dsDNA from 24 to 600 bp. However, we lack information on ...
  32. [32]
    AIM2 in health and disease: inflammasome and beyond - PMC
    The AIM2 inflammasome is responsible for sensing DNA in the cytosol and is a critical host defense mechanism against bacterial and viral infections.(18) AIM2 ...Aim2 In Health And Disease... · 3. Aim2 Sensing Of Microbial... · 4. Aim2 Sensing Of Self-Dna...Missing: seminal | Show results with:seminal
  33. [33]
    AIM2 Inflammasome's First Decade of Discovery: Focus on Oral ...
    Aug 12, 2020 · AIM2 consists of one N-terminal PYD domain and one C-terminal HIN domain (Figure 1B), which provides a structural basis for its function as a ...
  34. [34]
  35. [35]
    Soluble pattern recognition molecules: Guardians and regulators of ...
    Pentraxins are in addition complement activating proteins that lead to increased C3b levels and inflammatory signalling via C3a. The short pentraxins CRP and ...
  36. [36]
    Receptor-like kinases from Arabidopsis form a monophyletic gene ...
    Plant RLKs can be distinguished from animal RTKs by the finding that all RLKs examined to date show serine/threonine kinase specificity, whereas animal receptor ...
  37. [37]
    The receptor-like kinase SERK3/BAK1 is a central regulator ... - PNAS
    Although roles for most RLKs are unknown, many are implicated in plant growth and development, whereas others function in plant defense as so-called pattern- ...
  38. [38]
    CERK1, a LysM receptor kinase, is essential for chitin elicitor ... - PNAS
    Here, we identified a receptor-like kinase, designated CERK1, which is essential for chitin elicitor signaling in Arabidopsis.
  39. [39]
    Regulation of reactive oxygen species during plant immunity ...
    Apr 15, 2020 · ROS production upon pathogen challenge regulates multiple processes, including callose deposition, systemic signaling, and stomatal closure.
  40. [40]
    Receptor-like cytoplasmic kinases of different subfamilies ... - Nature
    May 21, 2024 · Both RLKs and RLPs detect extracellular immunogenic patterns (ExIPs), leading to extracellularly-triggered immunity (ExTI). ExTI is associated ...
  41. [41]
    Plant receptor-like protein activation by a microbial glycoside ...
    Sep 21, 2022 · Receptor-like proteins (RLPs) with a leucine-rich repeat (LRR) ectodomain constitute a subgroup of pattern recognition receptors and play a ...Missing: RLPs structure immunity
  42. [42]
    The Receptor-Like Protein ReMAX of Arabidopsis Detects the ...
    In turn, RLPs like Cf1 to Cf9, Ve1, and Eix2 found in Solanales have no functional counterpart in Arabidopsis (Fradin et al., 2011).
  43. [43]
    A receptor‐like protein from Nicotiana benthamiana mediates ...
    Oct 9, 2020 · By contrast, various RLPs function as PRRs to mediate plant innate immunity as well. For example, the typical RLPs Eix2, Ve1 and Cfs from ...
  44. [44]
    Receptor-like kinase SOBIR1/EVR interacts with receptor ... - PNAS
    Cell surface receptors are represented by receptor-like kinases (RLKs) that frequently contain extracellular leucine-rich repeats and an intracellular kinase ...
  45. [45]
    Multi-omics approach highlights differences between RLP classes in ...
    Jul 20, 2021 · Of the 57 annotated RLPs in Arabidopsis thaliana, 2 RLPs are experimentally validated to be associated with developmental functions (RLP10/CLV2 ...
  46. [46]
    Engineered pattern recognition receptors enhance broad-spectrum ...
    Oct 13, 2025 · Leucine-rich repeat receptor-like proteins (LRR-RLPs) also show potential for improving disease resistance. For example, transferring the ...
  47. [47]
    Systematic discovery and engineering of synthetic immune ...
    Sep 4, 2025 · In plants, PRRs are classified as either receptor-like kinases (RLKs) or receptor-like proteins (RLPs), which lack kinase domains (3). RLKs and ...
  48. [48]
    Toll-Like Receptor Signaling Pathways - Frontiers
    Intracellular localization of TLRs is thought to be critical for ligand recognition as well as for preventing TLRs from coming into contact with self-nucleic ...
  49. [49]
    TLR signaling | Cell Death & Differentiation - Nature
    Jan 20, 2006 · This review outlines the recent advances in our understanding of TLR-signaling pathways and their roles in immune responses.
  50. [50]
    MyD88: a central player in innate immune signaling - PMC
    Nov 4, 2014 · MyD88 is the canonical adaptor for inflammatory signaling pathways downstream of members of the Toll-like receptor (TLR) and interleukin-1 (IL-1) receptor ...Missing: seminal | Show results with:seminal
  51. [51]
    Toll-like receptor–mediated NF-κB activation - PubMed Central - NIH
    In addition to NF-κB activation, TLRs can also initiate mitogen-activated protein kinase (MAPK) signaling cascades and thus activate multiple transcription ...
  52. [52]
    TRIF‐dependent TLR signaling, its functions in host defense and ...
    May 9, 2016 · The MyD88-dependent signaling pathways are responsible for the production of inflammatory cytokines. The activated TLR4 recruits MyD88 through ...
  53. [53]
    Toll-Like Receptor Signaling Pathways - PMC - NIH
    TLR4 activates both the MyD88-dependent and TRIF-dependent pathways. Activation of these pathways is controlled by several molecules to induce appropriate ...Missing: seminal | Show results with:seminal
  54. [54]
    Structural basis for TIR domain–mediated innate immune signaling ...
    Jan 9, 2025 · This study elucidates the structural basis of Toll-like receptor (TLR) signaling through the TIR domain–containing adaptor proteins TRIF and TRAM.
  55. [55]
    Label-free biosensor assay decodes the dynamics of Toll-like ...
    Nov 12, 2024 · We present a label-free optical biosensor-based assay as a method for detecting TLR activation in a native and label-free environment and defining the dynamics ...
  56. [56]
    SIGIRR Inhibits Interleukin-1 Receptor- and Toll-like Receptor 4 ...
    T1/ST2 (3) and SIGIRR (4), also known as TIR8 (5), have been shown to function as negative regulators for Toll-IL-1R-mediated signaling. The leucine-rich repeat ...
  57. [57]
    Role of TLRs as signaling cascades to combat infectious diseases
    Mar 19, 2025 · MyD88 is crucial in most TLR-mediated signaling pathways. Initially, MyD88 was identified as a gene that is quickly activated when M1 ...Missing: seminal | Show results with:seminal
  58. [58]
    Unified Polymerization Mechanism for the Assembly of ASC ...
    AIM2 and NLRP3 are representative sensor proteins in two major families of inflammasomes. The adaptor protein ASC bridges the sensor proteins and caspase-1 to ...
  59. [59]
    Mechanisms governing inflammasome activation, assembly ... - NIH
    This inflammasome assembly consists of NLR or ALR and a bipartite protein ... AIM2 acts as a scaffold to assemble the AIM2 inflammasome (108). A recent ...
  60. [60]
    Gasdermin D is an executor of pyroptosis and required for ... - Nature
    Nov 27, 2015 · Caspase-11 can trigger pyroptosis but its effect on IL-1β/18 maturation is indirect because this process requires NLRP3-dependent activation of ...
  61. [61]
    Molecular mechanisms regulating NLRP3 inflammasome activation
    Nov 9, 2015 · Activation of the NLRP3 inflammasome requires at least two signals: signal 1, also known as the priming signal, is mediated by microbial ...
  62. [62]
    Human caspase-4 and caspase-5 regulate the one-step non ...
    Oct 28, 2015 · In mice, a non-canonical inflammasome pathway is regulated by caspase-11 activated by forced introduction of LPS into the cytoplasm via non ...
  63. [63]
    Structures and Mechanisms in the cGAS-STING Innate Immunity ...
    Jul 14, 2020 · This review focuses on the structural and mechanistic insights into the roles of cGAS and STING in immunity and diseases revealed by these recent studies.Missing: seminal | Show results with:seminal<|separator|>
  64. [64]
    Regulation of RIG-I-like receptor-mediated signaling - Nature
    Jan 18, 2021 · In this review, we focus on RLRs that detect cytoplasmic viral RNAs and the molecular mechanisms underlying RNA-mediated activation of RLRs and ...<|control11|><|separator|>
  65. [65]
    MAVS integrates glucose metabolism and RIG-I-like receptor signaling
    Sep 2, 2023 · This study reports that RLR action drives a switch from glycolysis to the pentose phosphate pathway (PPP) and the hexosamine biosynthesis ...
  66. [66]
    Interplay between RNA viruses and cGAS/STING axis in innate ...
    This review summarizes the characterized cGAS/STING escape strategies to date, together with the proposed mechanisms of STING signalling activation perpetrated ...
  67. [67]
    Regulation of C-Type Lectin Receptor-Mediated Antifungal Immunity
    Jan 31, 2018 · Signaling from Dectin-2, Dectin-3, and Mincle is mediated via the Syk/PKCδ-dependent CARD9/Bcl-10/MALT1 pathway, resulting in the activation of ...
  68. [68]
    Myeloid C-type Lectin Receptors in Pathogen Recognition and Host ...
    This review focuses on CLRs that signal in myeloid cells to initiate or tune immunity to pathogens. This subset of C-type lectins cuts across existing ...
  69. [69]
    Regulation of C-Type Lectin Receptor-Mediated Antifungal Immunity
    Feb 1, 2018 · In this review, we will focus on the roles and mechanisms of membrane-bound CLR-mediated-signaling pathways in host defense against fungal infections.
  70. [70]
    The Role of Pattern Recognition Receptors in Epigenetic and ...
    Jun 13, 2025 · C-Type Lectin Receptors (CLRs). CLRs, as specialized PRRs, utilize conserved carbohydrate-recognition domains (CRDs) to detect pathogenic ...
  71. [71]
    SIGIRR, a negative regulator of Toll-like receptor-interleukin 1 ...
    SIGIRR, a negative regulator of Toll-like receptor ... Our data show that SIGIRR functions as a biologically important modulator of TLR-IL-1R signaling.Missing: primary paper
  72. [72]
    TRIM29 negatively controls antiviral immune response through ...
    Mar 20, 2018 · TRIM29 negatively controls antiviral immune response through targeting STING for degradation. Qijie Li
  73. [73]
    Physical and functional interaction between A20 and ATG16L1 ...
    Apr 23, 2019 · Our data show that A20 and Atg16l1 cooperatively control intestinal homeostasis by acting at the intersection of inflammatory, autophagy and cell death ...
  74. [74]
    NLRP3 inflammasome and NLRP3-related autoinflammatory diseases
    This review provides a broad overview of NLRP3 inflammasome biology with particular emphasis on CAPS, whose clinical, genetic, and therapeutic aspects will be ...
  75. [75]
    The role of the cGAS-STING pathway in metabolic diseases
    Jun 30, 2024 · Activation of the cGAS-STING pathway has been found to be a major factor in metabolic disorders, including obesity, insulin resistance, and ...
  76. [76]
    Bacterial NLR-related proteins protect against phage - PMC
    There are two types of antigen receptor-type domains: FGS (Formylglycine-generating enzyme sulfatase) domains and supersecondary structure-forming tandem ...
  77. [77]
    The role of TIR domain-containing proteins in bacterial defense ...
    Aug 27, 2024 · The filament structure coordinates the activation of the NADase activity of the TIR domains, resulting in NAD+ depletion and abortive infection ...
  78. [78]
    Plant and prokaryotic TIR domains generate distinct cyclic ADPR ...
    Mar 17, 2023 · This study demonstrates that the TIR signaling requirements for plant and prokaryotic immune systems are distinct and that TIRs across kingdoms generate a ...
  79. [79]
    Lipopolysaccharide enhances bactericidal activity in Dictyostelium ...
    Receptors homologous to the mammalian TLRs and a family of cytosolic PRRs, the nucleotide binding and oligomerization domain-like receptors (Nod-like receptors ...
  80. [80]
    Gene family innovation, conservation and loss on the animal stem ...
    May 31, 2018 · ... domain (NACHT) and multiple LRRs. Although we detected proteins encoding both NACHT and LRR domains in multiple choanoflagellates, neither ...
  81. [81]
    Toll-Like Receptors, Associated Biological Roles, and Signaling ...
    Toll-like receptors (TLRs) are a class of pattern recognition receptors (PRRs) that initiate the innate immune response by sensing conserved molecular patterns ...
  82. [82]
    Peptidoglycan recognition proteins in Drosophila immunity - PMC
    For example, Drosophila has 13 PGRP genes encoding 19 proteins, mosquito (Anopheles gambiae) has 7 PGRP genes encoding 9 proteins, and humans and mouse have 4 ...
  83. [83]
    An organismal understanding of C. elegans innate immune ...
    These molecules are detected by hosts using various pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs), C-type lectin receptors (CLRs), ...
  84. [84]
    Origin and evolution of the plant immune system - Han - 2019
    Dec 21, 2018 · One uses cell surface pattern-recognition receptors (PRRs) to recognize microbe-associated molecular patterns (MAMPs) present in a large group ...
  85. [85]
    Major episodes of horizontal gene transfer drove the evolution of ...
    May 2, 2022 · Two major episodes of HGT events occurred in land plant evolution, corresponding to the early evolution of streptophytes and the origin of land plants, ...
  86. [86]
    Mechanisms, detection, and impact of horizontal gene transfer in ...
    Plants not only acquire genes from other plant species but also engage in HGT with a diverse set of organisms across both prokaryotic and eukaryotic domains, ...
  87. [87]
    The evolution of vertebrate Toll-like receptors - PNAS
    Jul 5, 2005 · Most vertebrates have exactly one gene ortholog for each TLR family. The family including TLR1 has more species-specific adaptations than other ...
  88. [88]
    [PDF] The genome of the early diverged amphioxus, Asymmetron ... - bioRxiv
    Jul 22, 2025 · Our phylogenetic analysis of NLRs from diverse metazoan lineages suggests a strong pattern of lineage-specific expansion, with. NLRs from ...
  89. [89]
    Progresses on three pattern recognition receptor families (TLRs ...
    Meanwhile, new ligands as well as signaling pathways do occur during evolution of teleost. This review summarizes progresses on the TLRs/RLRs/NLRs in teleost.Missing: ALRs | Show results with:ALRs
  90. [90]
    Evolutionary trajectory of pattern recognition receptors in plants - PMC
    Feb 1, 2024 · These receptors can be categorised as either receptor-like proteins (RLPs), which possess a transmembrane domain (TM), or receptor-like kinases ...
  91. [91]
    A plant Lysin Motif Receptor-Like Kinase plays an ancestral ... - PNAS
    We conclude that LysM-RLK-mediated perception of arbuscular mycorrhiza (AM) fungi is an ancestral requirement for AM symbiosis.
  92. [92]
    Activation loop phosphorylation of a non-RD receptor kinase ...
    Sep 16, 2021 · We propose that leucine-rich repeat-receptor kinase complexes containing a non-RD kinase are activated through phosphorylation-dependent conformational changes.
  93. [93]
    Sequence Exchange between Homologous NB-LRR Genes ...
    Domain swapping between the NB-LRR immune receptors Rx1 and Gpa2 converts extreme resistance to potato virus X in the shoots into potato cyst nematode ...
  94. [94]
    Recent insights into the evolution of innate viral sensing in animals
    Jul 18, 2014 · Viral sensors evolve under contrasting selective pressures: the conserved nature of viral PAMPs may tend to constrain evolution, whereas ...
  95. [95]
    An Overview of PRR- and NLR-Mediated Immunities - PubMed Central
    Of note, RLKs form a monophyletic gene family and their origin predates the divergence of plants and animals [13,14], whereas RLPs are less conserved [15,16].Missing: RLR | Show results with:RLR
  96. [96]
    Toll-Like Receptor 4 Polymorphisms and Risk of Gram-Negative ...
    These polymorphic forms of TLR4 have been associated with an increased incidence of Gram-negative infections and sepsis, and more recently with an increased ...
  97. [97]
    NOD2 Polymorphisms and Pulmonary Tuberculosis Susceptibility
    The association between NOD2 and tuberculosis (TB) risk has been reported widely, but the results of previous studies remained controversial and ambiguous.
  98. [98]
    Immunopathogenesis of Asymptomatic Chronic HIV Infection
    SIV leads to pDC activation via TLR7 and TLR9 stimulation with subsequent IFNα production in human and RM PBMC that is markedly attenuated in SMs. This ...<|separator|>
  99. [99]
    SARS-CoV-2 infection and overactivation of Nlrp3 inflammasome as ...
    A cytokine “storm,” which in many cases ends in severe acute respiratory syndrome, fulminant myocarditis and kidney injury.
  100. [100]
    Hepatitis C Virus Evasion from RIG-I-Dependent Hepatic Innate ...
    Our current understanding of RIG-I regulation by HCV is that the viral NS3/4A protease is the major inhibitor of RIG-I signaling through its ability to target ...
  101. [101]
  102. [102]
    Heterologous expression of Arabidopsis FLS2 in Nicotiana ...
    GmFLS2 contributes quantitatively to soybean resistance to Ralstonia, since soybean Gmfls2 loss‐of‐function mutants exhibit enhanced bacterial wilt ...
  103. [103]
    MPXV infection activates cGAS-STING signaling and IFN-I treatment ...
    May 20, 2025 · MPXV infection activates cGAS-STING signaling and IFN-I treatment reduces pathogenicity of mpox in CAST/EiJ mice and rhesus macaques.
  104. [104]
    mtDNA activation of the cGAS-STING pathway - PubMed
    May 30, 2024 · This article aims to explore the role of the mtDNA-triggered cGAS-STING pathway and its mechanisms in SLE, with the goal of providing novel ...
  105. [105]
    Pathogenic role of S100 proteins in psoriasis - PMC - NIH
    Jun 6, 2023 · S100A8 interacts with TLR4-MD2 complex, enhancing inflammatory response in target cells (122) ( Figure 3 ). Activation of TLR4 signaling pathway ...
  106. [106]
    CAPS and NLRP3 - PMC - PubMed Central - NIH
    Gain-of-function mutations in NLRP3 in CAPS patients lead to activation of the cryopyrin inflammasome resulting in the inappropriate release of inflammatory ...
  107. [107]
    cGAS Is Required for Lethal Autoimmune Disease in the Trex1 ...
    Sep 1, 2015 · In this study, we show that Trex1 (-/-) mice lacking cGAS are completely protected from lethality, exhibit dramatically reduced tissue inflammation, and fail ...
  108. [108]
    Identification of IRAK1 as a risk gene with critical role in the ... - PNAS
    Apr 14, 2009 · The likely role of IRAK1 in driving the second checkpoint, myeloid cell hyperactivity, is quite apparent given the central role of IRAK1 in ...
  109. [109]
    Plant autoimmunity—fresh insights into an old phenomenon - PMC
    Increasing evidence indicates that plant autoimmunity is in many cases the consequence of the inappropriate activation of cytoplasmic immune sensors.Missing: PRR misfiring
  110. [110]
    The Effect of NOD2 on the Microbiota in Crohn's Disease - PMC
    Mutations to NOD2 promote low diversity and dysbiosis in the microbiome, leading to impaired mucosal barrier function.
  111. [111]
    Gut Microbiome Dysbiosis and Inflammatory Bowel Disease ...
    Feb 25, 2025 · Mutations in the NOD2 gene increase the risk of developing CD. A lack of functional NOD2 leads to an increased abundance of the Bacteroidetes ...
  112. [112]
    Vaccine adjuvants: mechanisms and platforms - Nature
    Jul 19, 2023 · Agonists of TLRs as adjuvants have shown promising results by activating TLRs that lead to enhanced antigen presentation, upregulation of co- ...Missing: skin | Show results with:skin
  113. [113]
    Pattern recognition receptors: immune targets to enhance cancer ...
    Ongoing Clinical Trials using PRR agonists as cancer vaccine adjuvants or for intratumoral delivery. PRR targeted, PRR agonist, Tumors, Phase, Title, Clinical ...
  114. [114]
    MCC950 in the treatment of NLRP3-mediated inflammatory diseases
    Feb 3, 2022 · Evidence suggests that various innate immune system components are involved in pathological inflammatory conditions.
  115. [115]
    The NLRP3 inhibitor MCC950 inhibits... - Wellcome Open Research
    Oct 21, 2020 · The NLRP3 inflammasome inhibitor MCC950 blocked constitutive activation of NLRP3 observed in the PBMCs of CAPS patients.
  116. [116]
    A novel NLRP3 inhibitor as a therapeutic agent against ... - Frontiers
    SLC3037, a NLRP3 inhibitor blocking NEK7 binding to NLRP3, could be a novel agent against diseases associated with NLRP3 inflammasome activation such as gout.
  117. [117]
    Targeting the Mincle and TLR3 receptor using the dual agonist ...
    Jul 31, 2018 · Cationic Adjuvant Formulation 01 (CAF01)—an adjuvant based on trehalose dibehenate (TDB) and targeting the Mincle receptor—has entered human ...
  118. [118]
    Selection of adjuvants for vaccines targeting specific pathogens - PMC
    Liposomes are used in vaccine formulations against influenza, chlamydia, malaria, and tuberculosis (TB). ... Vaccine adjuvant uses of poly-IC and derivatives.
  119. [119]
  120. [120]
    Systematic Discovery and Design of Synthetic Immune Receptors in ...
    Mar 17, 2025 · We describe a strategy to map the recognition landscape of LRR-RLK-XIIs in plants, paving the way for the discovery of novel PRRs that can be synthetically ...Missing: vaccines | Show results with:vaccines
  121. [121]
    Pattern recognition receptors: immune targets to enhance cancer ...
    Aug 13, 2019 · The aim of this review is to gather the scientific rationale for the use of pattern recognition receptor (PRR) agonists in cancer therapy, and ...
  122. [122]
    Toll-like receptor 9 agonists and combination therapies - Nature
    Jul 28, 2022 · It has also been shown that inhaled TLR9 agonist administration combined with PD-1 antibody induces CD8+ T cells to become highly functional ...