Abstract
Aromatic amino acid (AAA) metabolites, derived from tryptophan, phenylalanine, and tyrosine through coordinated host and microbial metabolism, have emerged as critical modulators of immune function. We examine the complex journey of AAAs from dietary intake through intestinal absorption and metabolic transformation, highlighting the crucial role of host-microbe metabolic networks in generating diverse immunomodulatory compounds. This review provides a unique integrative perspective by mapping the molecular mechanisms through which these metabolites orchestrate immune responses. Through detailed analysis of metabolite-receptor and metabolite-transporter interactions, we reveal how specific molecular recognition drives cell type-specific immune responses. Our comprehensive examination of signaling networks—from membrane receptor engagement to nuclear receptor activation to post-translational modifications— demonstrates how the same metabolite can elicit distinct functional outcomes in different immune cell populations. The context-dependent nature of these molecular interactions presents both challenges and opportunities for therapeutic development, particularly in inflammatory conditions where metabolite signaling pathways are dysregulated. Understanding the complexity of these regulatory networks and remaining knowledge gaps is fundamental for advancing metabolite-based therapeutic strategies in immune-mediated disorders.
Keywords: Aromatic amino acids, Immunometabolism, Gut microbiome, Immunomodulation, Host-microbe interactions, Molecular mechanisms
INTRODUCTION
The human gut microbiota plays a fundamental role in shaping and maintaining immune homeostasis through complex bidirectional interactions with the host immune system. These microorganisms produce various bioactive metabolites that act as essential mediators in host-microbe communication, influencing both local and systemic immune responses (1,2). Recent advances have revealed that microbiota-derived metabolites significantly influence immune cell function through specific molecular mechanisms. These interactions modulate metabolic programming, gene expression, and cellular function, ultimately affecting immune cell differentiation, activation, and effector functions (3). Dysregulation of these pathways has been implicated in various immune-mediated disorders, including inflammatory bowel disease (IBD), autoimmune conditions, and allergic diseases (4,5), highlighting their clinical significance and potential as therapeutic targets.
Among the diverse array of microbiota-derived metabolites, those derived from aromatic amino acids (AAAs)—tryptophan (Trp), phenylalanine (Phe), and tyrosine (Tyr)— have emerged as critical modulators of immune function. While these compounds have traditionally been recognized for their roles in protein synthesis and as precursors for neurotransmitters (6), recent research has unveiled that AAA catabolism directly influences immune responses through specific molecular mechanisms. These include activation of membrane receptor-mediated signaling cascades, nuclear receptor-mediated signaling pathways, post-translational modification, and reprogramming of cellular metabolism (7,8). Despite their relatively low abundance, these findings have generated renewed interest in AAA-derived metabolites as important immunomodulatory agents essential for maintaining intestinal and systemic immune homeostasis.
Recent research has revealed sophisticated molecular pathways through which AAA metabolites coordinate microbial metabolism and host immune responses (9). However, several fundamental questions remain unresolved, particularly regarding how multiple metabolic pathways interact in different cellular contexts and disease states. This review provides a unique perspective on AAA metabolite-mediated immune regulation by integrating multiple levels of molecular interactions between host and microbes. We examine how dietary AAAs are transformed through coordinated host and microbial metabolic networks to generate diverse immunomodulatory compounds and provide a comprehensive analysis of their mechanisms, focusing on how ligand-specific receptors and transporters contribute to context-appropriate immune responses.
These ligand-specific transporters and receptors enable precise control of both innate and adaptive immunity in a cell type-dependent manner (10). Understanding these molecular mechanisms is particularly crucial for therapeutic development, as altered AAA metabolite profiles correlate with disease severity and show promise in modulating pathogenic immune responses in conditions like IBD (4). However, the same metabolic pathway can have opposing effects in different disease contexts, highlighting the need for careful consideration in therapeutic targeting. This integrated perspective of host-microbe-metabolite interactions is fundamental for developing more effective targeted interventions that can achieve appropriate immune modulation in specific disease contexts.
AAAs AND THEIR METABOLITES: OVERVIEW AND ABSORPTION
AAAs (Trp, Tyr, and Phe) are characterized by their distinctive aromatic ring structures, which enable protein structure stabilization and metabolic functions through their unique chemical properties (11). These essential molecules follow a complex pathway from dietary intake to biological function, involving coordinated processes of digestion, absorption, and metabolic transformation by both host and microbial systems.
Dietary sources and digestion
Trp and Phe are essential amino acids that must be obtained through diet, while Tyr can be synthesized endogenously from Phe. These AAAs are abundant in protein-rich foods and fermented products such as sauerkraut and kimchi (12,13). Fermented foods provide direct sources of AAA metabolites, including phenyllactate, 4-hydroxyphenyllactate and indole-3-lactate, which support immune homeostasis, neurogenesis, and enteric nervous system (ENS) maturation (14,15,16,17,18).
The gastrointestinal (GI) tract processes both dietary and endogenous proteins through a sophisticated digestive system. Initial protein breakdown yields small peptides, followed by further digestion at the brush border of intestinal epithelial cells (IECs) (19). This process achieves highly efficient amino acid absorption, with approximately 90% absorption by the time digestive contents reach the ileum (20).
Absorption
The efficient absorption of AAAs is accomplished through sophisticated transport systems composed of various transporters at IECs that are spatially organized at apical and basolateral membranes (21). In the small intestine, the primary neutral amino acid transporter B0AT1 (Slc6a19) at the apical membrane operates through a Na+-dependent mechanism in complex with angiotensin converting enzyme II to accept all neutral amino acids, branched-chain amino acids, and methionine (22). Additional apical transporters include ASCT2 (Slc1a5), a bidirectional neutral amino acid transporter; b0,+AT (Slc7a9)/rBAT complex, which exchanges cationic with neutral amino acids (KM 100-300 μM); and TAUT (Slc6a6), which facilitates Phe transport (23). The basolateral membrane contains 3 key transporters operating synergistically: 1) LAT2 (Slc7a8), an obligatory amino acid antiporter facilitating the exchange of all neutral amino acids except proline; 2) LAT4 (Slc43a2), a low-affinity facilitative uniporter (KM ≈ 4 mM) specific for Phe, leucine, isoleucine, valine, and methionine; and 3) TAT1 (Slc16a10), a H+-dependent facilitative uniporter selective for aromatic amino acids (KM 2.5-7 mM). Additionally, LAT1 (Slc7a7)/4F2hc complex mediates the exchange of neutral amino acids and Na+ for cationic amino acids (Fig. 1) (24,25).
Figure 1. Microbial and host transport mechanisms for aromatic amino acids in the intestine. Schematic overview of dietary aromatic amino acids (Trp, Phe, Tyr) transport at the intestinal host-microbe interface. Upper panel: Microbial utilization showing specialized transporters and de novo biosynthesis through shikimate pathway. Lower panel: Host absorption mechanisms in the intestinal epithelium, with the small intestine achieving >90% dietary protein uptake through coordinated apical and basal transport systems, while the large intestine processes remaining 2%–7% of unabsorbed proteins and microbial-derived amino acids.
AA0, neutral amino acids; AA+, cationic amino acids; PEP, phosphoenolpyruvate; E4P, erythrose 4-phosphate; PP pathway, pentose phosphate pathway.
In the large intestine, distinct transport systems handle the remaining 2%–7% of dietary proteins and amino acids derived from microbial metabolism and endogenous sources. Key apical transporters include ATB0,+ (Slc6a14), which accepts both neutral and cationic amino acids with high affinity (KM 6-600 μM), in addition to ASCT2 (Slc1a5) and TAUT (Slc6a6). At the basolateral membrane, SNAT2 (Slc38a2) facilitates the uptake of neutral amino acids with Na+ (Fig. 1) (24).
The expression and activity of these transporters are regulated through complex coordinated mechanisms, including spatial gradients of expression along the intestine, temporal control by diurnal rhythm and feeding patterns, and post-translational modifications like phosphorylation. The distinct transport mechanisms work in concert to ensure efficient amino acid absorption and homeostasis in IECs (24).
Microbial utilization of AAAs
The intestinal lumen serves as a shared metabolic environment where host absorption and microbial metabolism of AAAs occur simultaneously. Microbes utilize AAAs through multiple pathways: direct uptake of dietary AAAs, de novo biosynthesis, and metabolism of host-derived substrates. Despite efficient small intestinal absorption, a small fraction of dietary amino acids appears to escape assimilation and becomes available for microbial utilization. Microbes have evolved sophisticated transport systems for AAA uptake. In Escherichia coli, these include the high-affinity Trp permease Mtr, low-affinity Trp permease TnaB, Tyr-specific permease TyrP, and high-affinity Phe permease PheP. Additionally, the general permease AroP and ATP-binding cassette (ABC) transporters facilitate uptake of all three AAAs (26,27,28,29). Other species utilize distinct transporters, such as the Trp permease MhsT identified in Bacillus halodurans (30).
Beyond dietary sources, the host contributes to the amino acid pool through the production of endogenous substrates, particularly glycoproteins such as mucins (31). Some gut bacteria possess the shikimate pathway for de novo AAA biosynthesis, which proceeds through chorismate as a central intermediate. This biosynthetic capability allows certain microbes to thrive independent of dietary AAA availability (32). The bacterial-derived AAAs can be excreted through efflux pumps, like YddG in E. coli, which could affect AAA pools in the gut (33). However, as the extent of activity and the level of bacterial AAA synthesis remain unclear, dietary intake remains the primary source of AAAs for host intake (Fig. 1) (32). The interplay between incomplete absorption, host-derived endogenous substrates, and bacterial de novo AAA synthesis dynamically affects AAA availability in the gut, which further influences microbial community structure and metabolism.
A comparative analysis of blood plasma metabolites between germ-free (GF) and conventional mice has revealed profound differences in AAA metabolism. Trp and Tyr levels are elevated more than 1.4-fold in GF mice, while products of bacterial AAA metabolism, including indoxyl-sulfate, phenyl sulfate, p-Cresol sulfate, and phenylpropionylglycine, are exclusively present in conventional mice (34). These findings highlight the significant impact of gut microbiota on AAA metabolism and availability.
METABOLISM OF AAAs
The metabolic fate of AAAs involves complex networks that generate diverse bioactive compounds. The sophisticated interplay between dietary intake, host metabolism, and microbial processing creates a dynamic system that profoundly influences immune regulation and intestinal homeostasis.
Host metabolic pathways
Following absorption through the small intestinal epithelium, AAAs either enter systemic circulation for distribution to various organs or undergo immediate metabolism in intestinal tissues, establishing distinct yet interconnected metabolic pathways (Fig. 2).
Figure 2. Integrated metabolic network of aromatic amino acid catabolism by host and gut microbiota. Metabolic pathways of aromatic amino acids in the gut microbiota and host cells characterized to date. Blue arrows indicate microbial pathways with representative enzyme names shown in blue, while black arrows and enzymes represent host metabolic processes.
Trp metabolism
Host Trp metabolism proceeds through two principal pathways: kynurenine and serotonin. The kynurenine pathway represents the major route, accounting for approximately 95% of Trp catabolism in the liver (35). This pathway is regulated by three key enzymes that show distinct tissue distribution patterns. Trp 2,3-dioxygenase expression remains specific to the liver, while indoleamine 2,3-dioxygenase (IDO)1 and its paralog IDO2 show broader tissue distribution (36). These enzymes have emerged as critical immunoregulatory molecules (37). IDO1 expression and activity, which are influenced by gut microbiota, serve as a crucial link between immune modulation and microbial regulation of host metabolism. Increased IDO1/2 enzymatic activity leads to a higher kynurenine-to-Trp ratio, which has been reported in various inflammatory diseases and cancers associated with elevated IFN-γ levels (38). The inflammatory response driving IDO1 activation may originate from microbial interactions, highlighting the complex pathways through which microbes influence immune modulation (39).
The serotonin pathway produces crucial signaling molecules including serotonin (5-hydroxytryptamine) that influence multiple physiological systems. This pathway begins with tryptophan hydroxylases (TPH1/2) converting Trp to 5-hydroxytryptophan, followed by decarboxylation via DOPA decarboxylase (DDC) to produce serotonin. The tissue distribution of these enzymes shows clear compartmentalization: TPH1 predominates in peripheral tissues, while TPH2 functions primarily in the central nervous system. Notably, approximately 90%–95% of serotonin production occurs in the intestinal enterochromaffin cells, with additional significant quantities synthesized within the ENS. Since serotonin cannot cross the blood-brain barrier, this gut-derived serotonin serves as a major source for peripheral organs, where it exerts its physiological functions through specific receptor-mediated pathways (40). Beyond its traditional role as a neurotransmitter, serotonin and its metabolites play crucial roles in immune regulation, including T cell activation and dendritic cell (DC) maturation (40). Additionally, serotonin serves as a precursor for melatonin, a hormone essential for circadian rhythm regulation and immune modulation (41).
Phe metabolism
The primary fate of Phe is its conversion to Tyr through phenylalanine hydroxylase (PAH) (42), an enzyme requiring tetrahydrobiopterin (BH4) as a cofactor (43). This conversion occurs primarily in the liver and kidney, providing the majority of the body’s Tyr requirements. The physiological significance of this pathway is evidenced by phenylketonuria, a genetic disease caused by PAH deficiency that leads to elevated Phe concentrations and subsequent severe neurological and developmental complications. Phe can also undergo transamination to phenylpyruvate or decarboxylation to phenethylamine, but these represent minor pathways under normal physiological conditions (44,45). Phe and its derived compounds serve as important regulators of gut barrier function, and Phe itself can directly modulate immune cells function through specific signaling pathways (46).
Tyr metabolism
Tyr serves as a crucial precursor for several physiologically important molecules, primarily in the biosynthesis of dopamine, catecholamines, and melanin. In both intestine and brain, Tyr metabolism begins with Tyr hydroxylase converting Tyr to levodopa (L-3,4-dihydroxyphenylalanine), which subsequently undergoes decarboxylation by DDC to produce dopamine (35). Dysfunctional dopamine signaling in the brain is a hallmark of various neurological disorders, notably schizophrenia and Parkinson’s disease. The gut plays a crucial role in neurotransmitter production, contributing over half of the body’s dopamine production through both host and bacterial pathways (47). Dopamine receptors, extensively expressed throughout the intestinal tract, regulate various functions including intestinal motility, ion transport, mucus secretion, and mucosal blood flow. In specific tissues, particularly the brain, dopamine undergoes further modification by dopamine beta-hydroxylase and phenylethanolamine N-methyltransferase to produce L-noradrenaline and L-adrenaline, respectively. Beyond neurotransmitter synthesis, Tyr also serves as a substrate for melanin biosynthesis through tyrosinase-mediated pathways, contributing to skin, hair, and eye pigmentation, as well as thyroid hormone production (48).
Microbial metabolic pathways
Recent advances in microbiome research have revealed the extensive and specific capacity of gut bacteria to generate unique bioactive compounds from AAAs. These metabolic networks show remarkable complexity and specificity in terms of bacterial species involvement and metabolite production patterns (Fig. 2).
Trp-derived metabolites
The bacterial metabolism of Trp exemplifies the complexity of microbial metabolic capabilities. Multiple bacterial species catalyze the conversion of Trp into various indole derivatives through specific enzymatic cascades. These pathways generate intermediate compounds such as indole, indole-3-acetamide, tryptamine, indole-3-acetaldehyde, and indole-3-pyruvate, which can undergo further transformation to produce final metabolites including indole-3-acetate, indole-3-aldehyde, and indole-3-propionate. Notably, specific bacterial phyla are associated with metabolic pathways; for instance, Proteobacteria are primarily responsible for indole-3-acetamide production, while Firmicutes contribute to the formation of tryptamine and indole-3-pyruvate (49,50,51). Indole, a primary metabolite in this pathway, functions as a well-characterized signaling molecule in microbial communities, affecting bacterial behaviors such as spore formation, plasmid stability, drug resistance, biofilm formation, and virulence (52). Other metabolites, including indole-3-lactate and indole-3-ethanol (tryptophol), have recently been discovered to have anti-fungal and anti-bacterial activity, suggesting their role in microbial community regulation (53,54,55,56).
The bacterial contribution to serotonin metabolism in the gut represents another significant pathway. Several gut bacteria, including Staphylococcus aureus, Clostridium perfringens, Klebsiella grimontii, and Staphylococcus epidermis can produce serotonin, although the exact enzymatic pathways remain unidentified (57). This bacterial production is especially significant in the neonatal gut, where nearly half of bacterial isolates from the small intestine demonstrate serotonin-producing capability (57). Additionally, intestinal bacteria can modulate serotonin availability through β-glucuronidase-mediated deconjugation of host-derived serotonin (58).
Phe-derived metabolites
The microbial metabolism of Phe demonstrates similar complexity and specificity. Different bacterial species orchestrate the conversion of Phe into various metabolites including phenylpyruvate, phenylacetate, phenyllactate, and phenylpropionate. This metabolic network shows clear species-specific patterns: Bacteroides ovatus and Bacteroides fragilis predominantly produce phenylpyruvate, while Clostridium species specialize in phenylacetate formation. Some metabolites, such as phenylacetate, are produced by multiple bacterial species across different phyla, while others like phenyllactate and phenylpropionate are produced by a more limited range of bacteria, with Bifidobacterium species being significant contributor (4). Phenethylamine, produced through bacterial decarboxylation of Phe, has gained particular attention for its effects on mood, energy, and attention regulation, earning its designation as ‘endogenous amphetamine’ (59,60). The physiological roles of many other Phe-derived metabolites remain to be fully elucidated, highlighting an important area for future research.
Tyr-derived metabolites
The bacterial conversion of Tyr yields several bioactive compounds through diverse metabolic pathways (9). Key metabolites include tyramine, 4-hydroxyphenylpyruvate, 4-hydroxyphenylacetate, 4-hydroxyphenyllactate, and p-Cresol. The production of these compounds follows distinct phylogenetic patterns, with Firmicutes specializing in tyramine production and Proteobacteria contributing significantly to 4-hydroxyphenylpyruvate formation. Among these metabolites, p-Cresol has attracted particular attention due to its influence on the immune system, though its potential genotoxicity in the colon necessitates careful investigation of the underlying molecular mechanisms (61).
Host-microbe interactions in AAA metabolism
The interplay between host and microbial metabolism of AAAs involves sophisticated regulatory mechanisms operating at multiple levels, from direct substrate competition to complex metabolic networks. Understanding these interactions is crucial for comprehending the physiological impact of AAA metabolism on immune function and host homeostasis.
Competition for substrates
The intestinal lumen serves as a shared metabolic space where host absorption and microbial metabolism compete for available AAAs (62,63). This competition has significant physiological consequences; intestinal and circulatory AAA levels are notably lower in microbiota-colonized mice compared to GF counterparts. Recent study has identified specific gut microbes and their metabolic genes that efficiently deplete amino acids in the intestine, demonstrating the substantial influence of microbial metabolism on host nutrient homeostasis (64).
Regulation of host metabolic functions
Beyond substrate competition, bacterial activity directly regulates expression or biological function of host enzymes involved in AAA metabolism. For instance, recent studies have demonstrated that gut microbial metabolites, including butyrate, propionate, tryptamine, deoxycholate, and p-aminobenzoate, can upregulate Tph1 expression, thereby influencing systemic serotonin levels (65). Similarly, certain commensal bacteria, including Clostridium, Lachnoclostridium, Ruminoclostridium, and Roseburia species, repress Ido1 expression through butyrate-mediated STAT1 inhibition via its effect on histone deacetylase activity (66). A recent study demonstrated a developmental stage-specific regulation of host serotonin metabolism: neonatal gut bacteria such as Rodentibacter heylii and Enterococcus gallinarum maximize serotonin availability by inducing higher TPH1 expression and maintaining lower monoamine oxidase A levels in the small intestine, effects not observed in adult intestine (57). In addition, the gut microbiota also influences AAA metabolism indirectly through effects on GI motility (67,68). Studies in GF or antibiotic-treated mice have shown slower GI motility and prolonged intestinal transit time, accompanied by the loss of enteric neurons (69,70). These changes in gut physiology can significantly impact the absorption and metabolism of AAAs.
Integration of metabolic networks
The metabolic sharing of AAAs and their derivatives represents a sophisticated form of microbial cooperation that fundamentally shapes gut microbial community structure and function. This metabolic interdependence appears to be an evolutionary adaptation, as many gut bacteria lack complete AAA biosynthetic pathways in their genome, likely due to the high energetic costs and local availability of these compounds from host and other microbial sources (71).
The metabolism of Phe and Tyr exemplifies these complex networks. Certain Firmicutes, particularly Clostridium sporogenes, initiate reductive metabolism of these amino acids through the phenyllactate dehydratase (FldABC) complex, generating phenylpropanoate and 4-hydroxyphenylpropionic acid. These products support auxotrophic bacteria including Clostridioides difficile, Streptococcus pneumoniae, and various Lactobacillus species. The metabolic web extends further through an alternative oxidative pathway, mediated by pyruvate ferredoxin oxidoreductase alpha subunit (PorA), which produces phenylpropanoate and 4-hydroxyphenylacetate. Notably, these pathways respond to glucose availability – when glucose is limited, bacteria primarily use AAAs for energy, and when glucose is abundant, they focus on producing bioactive metabolites that influence community behavior (9).
Trp metabolism provides another compelling example of microbial cooperation. Lactobacillus species initiate the process by converting Trp into indole-3-lactate, which Clostridium species subsequently transform into other bioactive compounds such as indole-3-propionate and indole-3-acetate. This cross-feeding pathway shows enhanced activity under inflammatory conditions, suggesting its potential role in maintaining gut homeostasis during dysbiosis (8). These intricate metabolic relationships ultimately determine the profile of immunomodulatory metabolites available to the host, highlighting how community-level microbial interactions influence host physiology. Understanding these networks is crucial for developing targeted therapeutic approaches that consider the complex web of microbial interactions rather than focusing on individual species or pathways.
MOLECULAR MECHANISMS TARGETED BY AAA METABOLITES FOR IMMUNE REGULATION
AAA metabolites orchestrate immune responses through several distinct but interconnected signaling mechanisms: rapid non-genomic signaling via membrane receptors, sustained genomic regulation through nuclear receptors, unique post-translational modifications, and metabolic signaling pathways (Fig. 3). While our understanding of how these distinct signaling pathways converge in immune cells remains limited, this multi-layered control system enables precise modulation of immune responses across different temporal and spatial scales (4).
Figure 3. Molecular mechanisms of AAA metabolite signaling and receptor distribution across immune cells. Schematic diagrams illustrating signaling pathways and cellular distribution of receptors and transporters. (A) Left panel: Non-genomic signaling mechanisms showing membrane receptor-mediated pathways. AAA metabolites activate various membrane receptors, leading to diverse signaling cascades including cAMP, MAPK, PI3K, and mTOR pathways, ultimately affecting transcription factors (NF-κB, CREB, eIF-4B). Right panel: Genomic regulation mechanisms depicting transporters facilitating cellular uptake of metabolites. Inside the cell, metabolites activate nuclear receptors (AHR, PXR, NRF2), while TGM2 catalyzes histone monoaminylation. Colored boxes indicate predominant metabolite origins: microbe-derived (tan), host-derived (blue), or co-produced (purple). Colored dots denote ligand specificity for different nuclear receptors. (B) Distribution of key receptors and transporters across immune cells and intestinal epithelial cells, highlighting cell type-specific expression patterns.
Ras, rat sarcoma virus; Akt, protein-kinase B; 4E-BP1, eukaryotic initiation factor 4E-binding protein 1; eIF-4B, eukaryotic translation initiation factor 4B.
Non-genomic signaling mechanisms (membrane receptor-mediated pathway)
While nuclear receptor pathways have traditionally dominated our understanding of metabolite sensing, recent evidence highlights the critical importance of rapid non-genomic signaling (72). Through membrane receptors, AAA metabolites can trigger immediate cellular responses within seconds to minutes, far faster than the hours required for genomic regulation. In addition, through second messenger cascades and signal amplification, activation of membrane receptors often allows precise control of immune cell function without requiring new protein synthesis (73,74). The distribution and expression patterns of these receptors across different immune cell populations allow for sophisticated functional regulation. This rapid and sophisticated response system is particularly crucial in dynamic immune environments where quick adaptations can be essential for proper immune function (Fig. 3).
G protein-coupled receptors (GPCRs)
GPCRs constitute the largest family of cell membrane receptors, serving as fundamental mediators of signal transduction. Upon ligand binding, these receptors undergo conformational changes that activate specific G protein subtypes (Gαs, Gαi, Gαq), which in turn trigger distinct intracellular signaling cascades (75). In immune regulation, GPCRs serve as key molecular targets for AAA metabolites, with multiple receptor families orchestrating diverse aspects of immune cell function.
Trace amine-associated receptors (TAARs) represent important targets for bacterial and host-derived AAA metabolites in the immune system. Bacterial metabolites, β-phenethylamine, tyramine, and tryptamine are known ligands for TAARs. These receptors are expressed in various immune cells including B cells, T cells, neutrophils, monocytes, and mononuclear cells, where they mediate distinct immunological responses. Phenethylamine is a potent agonist that activates both TAAR1 and TAAR2, leading to IgE synthesis in B cells and chemosensory migration in polymorphonuclear leukocytes. Tyramine similarly acts through TAAR1, although with lower affinity, and has been shown to induce pro-inflammatory cytokine production in macrophages (76). The clinical significance of these interactions is highlighted by elevated phenethylamine levels serving as potential biomarkers for Crohn’s disease, while tyramine modulates bacterial adhesion to the intestinal epithelium and modulate TNF-α production (76).
Hydroxycarboxylic acid receptor 3 (HCA3) represents another crucial GPCR target in immune regulation. This receptor responds to bacterial phenyllactate produced by Lactobacillus species and host-derived β-hydroxylated fatty acids. Phenyllactate activation of HCA3 promotes monocyte migration through specific signaling cascades (77). Additionally, G-protein coupled receptor 35 (GPR35) responds to Trp metabolites including kynurenic acid and 5-hydroxyindoleacetate, inducing leukocyte adhesion and migration (78,79).
The serotonergic signaling system exemplifies precise immune regulation through cell-type specific receptor expression patterns, with serotonin acting through multiple serotonin receptors (HTRs) subtypes expressed differentially across immune cell populations (10). T cells predominantly express HTR1, HTR2, and HTR7, which coordinate their activation status and cytokine production profiles. In contrast, DCs express a different complement of receptors, including HTR4 and HTR7, enabling distinct responses to serotonin signals (80). Bacterial tryptamine exhibits a unique dual targeting capability, activating both HTR4 and TAAR1. Tryptamine has been shown to induce mucus secretion in colon goblet cells through HTR4 activation (Fig. 3B) (81).
Non-GPCRs
TLR4 expands the repertoire of AAA metabolite signaling through distinct mechanisms. Cinnamic acid, a bacterial Phe derivative, may interact with TLR4 and regulate subsequent NF-κB signaling to regulate inflammatory responses. Cinnamic acid treatment has shown inhibition effect to cytokine production by blocking TLR4 in monocyte (82). Indole-3-acetate, in conjunction with LPS, stimulates the formation of IL-35-producing B cells, which is mediated by NF-κB pathway activation via TLR4 (83). This mechanism highlights the complex interplay between gut microbiota metabolites, bacterial components, and the host immune systems.
Receptor Tyr kinases represent a distinct class of high-affinity cell surface receptors that respond to various polypeptide growth factors, cytokines, and hormones. Within this family, the epidermal growth factor receptor (EGFR) pathway demonstrates particular significance in AAA metabolite signaling. p-Cresol, generated through bacterial Tyr metabolism, interferes with EGFR and TLR4 crosstalk. This blocks synergistic signal transduction and decreases epithelial CCL20 production (84).
HTR3 represents a unique member of serotonin receptor family, functioning as a ligand-gated ion channel rather than a GPCR. When serotonin binds to the receptor, it triggers a rapid cation influx that leads to membrane depolarization and subsequent cellular activation (85).
Genomic regulation mechanisms (nuclear receptor-mediated and epigenetic regulation)
In contrast to membrane receptor-mediated mechanisms, several AAA metabolites, particularly Trp metabolites, also exert effects through nuclear receptor activation following cellular uptake. While Phe and Tyr metabolites enter cells via specific transporters, their roles in nuclear receptor-mediated pathways have not yet been identified. Additionally, emerging evidence has uncovered that some AAA metabolites can modulate gene expression through histone modifications, broadening their regulatory mechanisms (Fig. 3).
Cellular transport mechanisms
Despite the diverse physiological effects of AAA metabolites, their intracellular transport mechanisms remain incompletely understood. These mechanisms vary depending on the molecular structure of the precursor compounds. For instance, hydrophobic molecules like indole are believed to passively diffuse across cell membranes, whereas others require specific transport proteins for cellular uptake (86). Monoamine transporters (MATs)—including the serotonin transporter (SERT), dopamine transporter (DAT), and norepinephrine transporter (NET)— along with organic cation transporters (OCTs) from the SLC22 family (OCT1, OCT2, OCT3) have been identified as key mediators in the transport of AAA metabolites (87,88). All 3 OCTs (OCT1, OCT2, and OCT3) are predominantly expressed in neuronal cells, whereas OCT1 and OCT3 are also expressed in IECs. Experimental evidence demonstrates that OCT1 facilitates the transport of dopamine, tyramine, phenethylamine, and tryptamine. OCT2 exhibits similar substrate specificity, mediating the transport of dopamine, tyramine, phenethylamine, and serotonin, while OCT3 functions in the transport of dopamine, tyramine, and phenethylamine. SERT, expressed in neurons, intestinal enterocytes, and various immune cells such as monocytes, macrophages, DCs, mast cells, and lymphocytes, is capable of transporting dopamine, tyramine, phenethylamine, serotonin, and tryptamine (40). DAT, predominantly expressed in brain neurons and some immune cells such as monocytes, macrophages, and lymphocytes, shares substantial substrate overlap with NET, a brain-specific transporter (89). Both DAT and NET facilitate the movement of dopamine, tyramine, phenethylamine, serotonin, and tryptamine.
Additionally, L-amino acid transporters, expressed in lymphocytes, macrophages, DCs, and NK cells, mediate transport of phenethylamine and kynurenine (90). The plasma membrane MAT, broadly distributed across various tissues, including intestinal enterocytes, is specifically involved in the transport of tryptamine (91). This diverse array of transport mechanisms highlights the complexity of bacterial metabolite absorption and distribution in mammalian systems (Fig. 3B).
Aryl hydrocarbon receptor (AHR) pathway
Indole metabolites, such as indole-3-acetate, indolelactate, indole-3-propionate, and indolealdehyde, predominantly act through AHR after cellular uptake. As a ligand-dependent transcription factor, AHR responds to both endogenous and exogenous ligands. In its inactive state, AHR exists in the cytoplasm as part of a multi-protein complex, associated with heat shock protein 90, co-chaperone p23, and protein Tyr kinase c-Src (92). Upon ligand binding, AHR undergoes a conformational change that triggers its release from the protein complex and subsequent nuclear translocation and heterodimerization with AHR nuclear translocator (92). This complex recognizes and binds to specific DNA sequences known as xenobiotic response elements (XREs) in target gene promoters, activating transcription of target genes involved in immune regulation, including Il22 and Cyp1a1 (15).
Pregnane X receptor (PXR) pathway
PXR, a member of the nuclear receptor superfamily, provides an additional layer of regulation for certain metabolites, particularly for indole metabolites such as indole, indole-3-acetate, and indole-3-propionate. When ligands bind to PXR, it undergoes a conformational change that promotes coactivator recruitment while displacing corepressors (93,94). The activated PXR functions as a master regulator of xenobiotic metabolism by controlling the transcription of drug-metabolizing enzymes and transport proteins. Additionally, PXR orchestrates diverse immunoregulatory responses in innate and adaptive immune cells—suppressing pro-inflammatory cytokine production in macrophages, modulating T cell differentiation, and regulating B cell responses (95). Notably, many PXR ligands overlap with AHR ligands, suggesting that Trp metabolites coordinate these two pathways to fine-tune immune responses, though the exact mechanisms of their potential crosstalk or cooperation remain to be fully elucidated (8).
Epigenetic modification
A unique aspect of AAA metabolite signaling is exemplified by their involvement in epigenetic modification. Serotonylation, the covalent attachment of serotonin to glutamine residues of target proteins by tissue transglutaminases (TGMs), occurs on histone H3 (H3Q5ser), resulting in long-lasting changes to cellular function and gene expression (96). This modification has emerged as a novel epigenetic regulatory mechanism (97), as demonstrated in neuronal cells where serotonylation of H3Q5 promotes the binding of the transcription factor IID-containing transcription initiation complex to H3K4me3-enriched promoters (98). While histone serotonylation has not been directly demonstrated in immune cells, the presence of serotonin and other monoamines that can serve as TGM2 substrate in immune cells, along with the expression of various serotonin transporters, suggests the potential occurrence of this modification in immune cells (99). The functional significance of protein serotonylation has been demonstrated in CD8+ T cells, where serotonylation of GAPDH at position Q262 promotes its cytoplasmic localization and enhances glycolytic metabolism, supporting T cell activation and antitumor immune responses. The therapeutic potential of this pathway can be enhanced through engineering CAR-T cells to increase serotonin production (100).
Bacterially produced indole-3-propionate provides another example of epigenetic regulation, enhancing H3K27 acetylation at the super-enhancer region of the Tcf7 gene, although the underlying molecular mechanism is not clear. This epigenetic modification increases expression of TCF-1, a master regulator of T cell stemness, promoting the differentiation of progenitor exhausted CD8+ T cells (Tpex) in the tumor microenvironment. The pathway’s essential role was confirmed in Tcf7-knockout mice, where indole-3-propionate failed to enhance immunotherapy efficacy. Therefore, indole-3-propionate represents a molecular connection between microbial metabolites and T cell stemness programming and enhanced antitumor immunity through the maintenance of stem-like CD8+ T cells (101).
Intracellular signaling pathways
cyclic AMP (cAMP)-dependent pathway
The cAMP-dependent signaling serves as a central mechanism through which AAA metabolite modulate immune responses after GPCR activation. The HTR1, HCA3 and GPR35 couple to inhibitory G-proteins (Gαi/o), suppressing adenylyl cyclase activity and reducing intracellular cAMP levels (102,103,104). In contrast, the HTR4, HTR6, and HTR7 couple to the stimulatory G-proteins (Gαs), enhancing adenylyl cyclase activity and elevating cAMP levels. These changes in cAMP levels trigger protein kinase A (PKA) activation, leading to phosphorylation of downstream targets including cAMP response element-binding protein (CREB) and subsequent modulation of gene expression (105). In mature DCs, serotonin-mediated activation of HTR4 and HTR7 modulates cytokine production through cAMP-dependent mechanisms, selectively enhancing IL-1β and IL-8 secretion while suppressing IL-12 and TNF-α production (105).
MAPK signaling pathway
Activation of HTR2 by serotonin promotes cell proliferation through the MAPK signaling pathway (105). Macrophages express HTR2B, which plays a key role in serotonin-mediated regulation of immune function. Activation of HTR2B triggers ERK1/2 phosphorylation via the MAPK signaling pathway, leading to transcriptional regulation of genes such as AP1, c/EBP, and SRF, which drive macrophage activation and polarization. Notably, HTR2B stimulation promotes M2 macrophage polarization while inhibiting M1 polarization and also suppresses the secretion of pro-inflammatory cytokines such as IL-12 in monocyte-derived M2 macrophages through NF-κB signaling upon LPS stimulation (106).
mTOR signaling pathway
The mTOR is a highly conserved serine/threonine protein kinase that functions as a central regulator of cell metabolism, growth, proliferation, and survival in response to nutrients, growth factors, and cellular energy status. Multiple mechanisms have been identified through which AAA metabolites can regulate mTOR activity. Kynurenine, kynurenic acid, and indole-3-lactate have been shown to modulate the mTOR-mediated autophagy pathway, regulating intracellular protein turnover and the intestinal barrier functions (107). IDO, the primary enzyme responsible for host-mediated Trp catabolism, depletes Trp through kynurenine pathway activation and leads to mTOR inhibition, subsequently triggering autophagy (108).
A recent study identified that Trp, Phe, and Try function as direct agonists of mTOR through interaction with Sestrin3, disrupting its association with GATOR2, distinct from the mechanism of previously known mTOR-activating amino acids (glutamine, leucine, and arginine). This activation influences proteasomal localization; under amino acid starvation, mTOR inhibition allows proteasome translocation from nucleus to cytosol for protein breakdown and survival, while AAA-mediated mTOR activation causes nuclear proteasome sequestration, potentially leading to cell death under stress (109). This newly discovered pathway thus emerges as a crucial mechanism for coping with cellular stress, but the ability of other structurally similar metabolites to activate mTOR in the same way remains unexplored.
IMMUNE CELL RESPONSES TO AAA METABOLITES
AAA metabolites exhibit diverse regulatory effects on immune cell populations through multiple signaling pathways, orchestrating both innate and adaptive immune responses. These metabolites fine-tune immune cell function through complex mechanisms that can be broadly categorized by their effects on specific immune cell populations. Table 1 presents a detailed compilation of AAA metabolites and their immunomodulatory effects, highlighting specific target cells, functional changes, and the molecular mechanisms through which they exert their influence (7,40,76,77,78,80,81,82,83,84,100,101,102,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143).
Table 1. Immunomodulatory effects and mechanisms of AAA metabolites.
| Amino acid | Metabolites | Target cells | Function | Mechanism | Reference |
|---|---|---|---|---|---|
| Trp | Indole* | Treg, Th17 | Maintaining balance | AHR signaling pathway | (115) |
| HCT-8 cells (colon epithelial cell model) | NF-κB activation ↓ | TNF-α | (124) | ||
| IL-8 ↓ | - | (124) | |||
| IL-10 ↑ | - | (124) | |||
| Indole-3-pyruvate*,† (IPyA) | Treg1 | Treg1 differentiation ↑ | AHR signaling pathway | (125) | |
| Th17 | (125) | ||||
| DCs | Th1 ↓ | (125) | |||
| NK cells | Formation ↑ | H3K27 acetylation | (101) | ||
| ILCs | (101) | ||||
| Indole-3-acetate* (IAA) | DCs | IL-22 expression and secretion ↑ | AHR signaling pathway | (110) | |
| B cells | IL-35+ B cells ↑ | PXR and TLR4 activation | (83) | ||
| Macrophages | IL-1β ↓, TNF-α and MCP-1 expression and secretion ↓ | AHR signaling pathway | (126) | ||
| Indole-3-lactate* (ILA) | Intraepithelial CD4+ T cells | Reprogram into immunoregulatory T cells | AHR signaling pathway | (127) | |
| Indole-3-propionate* (IPA) | Macrophages | IL-10 ↑, TNF ↓, IL-6 ↓ | PXR activation | (115,128) | |
| CD8+ T cells | Differentiation into Tpex cells | H3K27 acetylation | (101) | ||
| Indole-3-acrylic acid* (IA) | Goblet cells | Muc2 expression ↑ | AHR signaling pathway | (115) | |
| Macrophages | IL-10 ↑, IL-6 ↓ | (115) | |||
| ARE pathway ↑, pro-inflammatory signaling pathway ↓ | NRF2-ARE pathway | (115) | |||
| PBMC | IL-6 ↓, IL-1 β ↓ | NRF2-ARE pathway | (115) | ||
| Tryptamine*,† | Goblet cell | Mucus release ↑ | HTR4 | (81) | |
| Macrophages | IL-1β ↓, TNF-α and MCP-1 secretion ↓ | AHR signaling pathway | (126) | ||
| Treg cells | P4EBP1 expression in Tem cells ↑ | mTOR activation | (120) | ||
| NK cells | Cytotoxicity toward tumor cells ↑ | AHR signaling pathway | (120) | ||
| Indole-3-aldehyde* (IAld) | ILC3 | IL-22 ↑ | AHR signaling pathway | (118,119) | |
| Kynurenine*,† (Kyn) | DCs | IDO1 ↑ | TGF-β signaling pathway | (111,112) | |
| TGF-β ↑ | AHR signaling pathway | (129) | |||
| CD8+ T cells | PD-1 ↑ | (130) | |||
| CD4+ T cells | Foxp3, TGF-β ↑ | (131,132) | |||
| Th17 | Differentiation to IL-10 producing Treg1 | TGF-β signaling pathway | (133) | ||
| AHR signaling pathway | (134,135) | ||||
| Kynurenic acid*,† (KYNA) | Monocytes | TNF, IL-4, IL-23 ↓ | - | (134) | |
| CD4+ T cells | Differentiation to Treg cells ↑ | AHR signaling pathway | (134) | ||
| Differentiation to Th17 cells ↓ | (102) | ||||
| DCs | IL-23 ↓ | GPR35 | (102) | ||
| CD4+ T cells | IL-17 ↓ | (136) | |||
| iNKT | IL-4 ↓ | (137) | |||
| 3-Hydroxyanthranilic acid*,† (3-HAA) | Glial cells, astrocytes | TNF-α ↓, IP-10 ↓ | - | (117) | |
| Macrophages | Immunophenotyping remodeling | CCL2/CCR2 inhibition | (122) | ||
| CD4+ T cells | Apoptosis | NF-kB inhibition | (138) | ||
| DCs | TGF-β ↑ | AHR signaling pathway | (139) | ||
| 3-Hydroxykynurenine*,† (3-HK) | T cells | Suppression | - | (123) | |
| 5-Hydroxytryptophan*,† (5-HTP) | CD8+ T cells | CD8+ T cell exhaustion | AHR signaling pathway | (140) | |
| 5-Hydroxytryptamine*,† (5-HT, Serotonin) | Smooth muscle cells | IL-6 ↑ | HTR2 | (80) | |
| Platelets | T cell activation | IgE receptor | (40) | ||
| Monocytes | TNF-α ↓ | HTR4, HTR7 | (40) | ||
| IL-6, IL-1β ↑ | HTR3, HTR4, HTR7 | (40) | |||
| Macrophages | CCL2 ↑ | HTR2C | (40) | ||
| Phagocytosis | HTR1A, NFkB | (40,113) | |||
| DCs | IL-1β, IL-6, IL-8, IL-10 ↑ | HTR3, HTR4, HTR7 | (114) | ||
| IL-12, TNFα ↓ | HTR7 | (40) | |||
| Eosinophils | Asthmatic response | HTR2 | (40) | ||
| Mast cells | Cell adhesion and migration | HTR1A | (121) | ||
| Lymphocytes | IL-2, IL-16 ↑, T cell proliferation | HTR1A | (100) | ||
| CD8+ T cells | IFN-γ, IL-17 ↓ | Serotonylation | (121) | ||
| CD4+ T cells | IL-10 ↑ | HTR1A, HTR2 | (121) | ||
| Th17 cells | IL-17 ↑ | HTR2A, HTR2B | (141) | ||
| B cells | B-cells proliferation | HTR1A | (78) | ||
| 5-Hydroxyindoleacetate*,† | Neutrophils | Transmigration ↑ | GPR35 | (142) | |
| Phe | Phenylalanine* | Macrophages | Pyroptosis ↑ | NLRP3 inflammasome | (7) |
| M1 macrophage inflammation ↓ | OXPHOS ↑ | (116) | |||
| Phenylpyruvate*,† | Macrophages | IL-1β ↑ | NLRP3 inflammasome | (76) | |
| Phenethylamine*,† | T cells | IL-4 secretion ↑ | TAAR1 signaling | (76) | |
| (PEA) | B cells | IgE expression ↑ | TAAR1 signaling | (143) | |
| Phenylacetate*,† | Neutrophils | Recruitment | - | (82) | |
| Phenylacrylic acid (Cinnamic acid)* | Monocytes | TLR-4 ↑, TLP-2 ↓, HLA-DR ↓, CD80 ↓ | TLR4 blocking | (82) | |
| TNF-α ↓, IL-10 ↓ | (77) | ||||
| Phenyllactate* | Monocytes | Migration ↑ | HCA3 activation | (84) | |
| Tyr | p-Cresol* | Airway epithelial cells | CCL20 production ↓ | EGFR and TLR4 signaling | (76) |
| Tyramine*,† | T cells | IL-4 secretion ↑ | TAAR1 signaling |
*Metabolic compound that can be synthesized by bacteria.
†Metabolic compound that can be synthesized by host.
Regulation of innate immune cells
DCs integrate multiple AAA metabolite signals through diverse molecular pathways to modulate their function and subsequent T cell responses. Trp metabolites particularly demonstrate potent regulatory effects on DCs through both AHR-dependent and independent mechanisms: Indole-3-acetate enhances IL-22 expression and secretion through AHR signaling pathway activation (110). Kynurenine exhibits dual regulatory mechanisms—inducing IDO1 expression through TGF-β signaling and increasing TGF-β production via AHR signaling pathway, creating a potential feedback loop (111,112). The serotonin signaling system in DCs involves multiple receptor subtypes, including HTR3, HTR4, and HTR7, leading to complex cytokine responses. This signaling network results in increased IL-1β, IL-6, IL-8, and IL-10 production (40,113), while specifically reducing IL-12 and TNF-α through HTR7 activation (114), demonstrating how a single metabolite can orchestrate diverse immune responses through distinct receptor-mediated pathways.
Macrophages respond to AAA metabolites through an intricate network of signaling cascades. Indole-3-propionate and indole-3-acrylic acid reduce pro-inflammatory cytokine production through distinct mechanisms; indole-3-propionate operates through PXR activation to increase IL-10 while decreasing TNF and IL-6, whereas indole-3-acrylic acid employs the NF erythroid 2-related factor 2 (NRF2)-antioxidant response element (ARE) pathway to achieve similar anti-inflammatory effects (115). Tryptamine suppresses inflammatory mediators via AHR signaling, while Phe metabolites like phenethylamine uniquely influences M1 macrophage inflammation through OXPHOS regulation, demonstrating metabolic reprogramming as an additional regulatory mechanism (7). The nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome pathway adds another layer of complexity, mediating phenylpyruvate-induced IL-1β production (116). Furthermore, 3-hydroxyanthranilic acid modulates macrophage function through CCL2/CCR2 inhibition, highlighting the diversity of signaling pathways involved in metabolite-mediated regulation (117).
NK cells and innate lymphoid cells (ILCs) demonstrate specific responses to Trp metabolites through both transcriptional and epigenetic mechanisms. Indole-3-propionate enhances NK cell formation through H3K27 acetylation (101), while indole-3-aldehyde specifically targets ILC3s to promote IL-22 production and intestinal stem cell regeneration through AHR signaling (118,119). Tryptamine increases NK cell cytotoxicity toward tumor cells through AHR pathway activation (120).
Regulation of adaptive immune cells
T cell populations show remarkable sensitivity to AAA metabolites through multiple signaling pathways that influence their differentiation, function, and cytokine production patterns. The regulation of CD4+ T cell differentiation involves complex interplay between different signaling cascades. Multiple Trp metabolites, including indole and indole-3-propionate, regulate the balance between Treg and Th17 cells through both AHR-dependent and independent mechanisms (144). Interestingly, AHR activation has ligand-specific effects for T cell regulation; for example, while 6-formylindolo(3,2-b)carbazole (FICZ) promotes Th17 cell differentiation, 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) preferentially induces Foxp3+ Tregs (145,146). However, the underlying molecular mechanisms of this distinction remain poorly understood.
Serotonin signaling adds another layer of complexity through various receptor subtypes, particularly HTR1A and HTR2, which modulate cytokine production with specific effects on IL-10 and IL-17 production in different T cell subsets (121). Additionally, 3-hydroxyanthranilic acid induces apoptosis through NF-κB inhibition, demonstrating how metabolites can control T cell populations through both functional modification and cell survival pathways (122).
In CD8+ T cells, indole-3-propionate promotes the differentiation of progenitor exhausted CD8+ T cells through epigenetic modification via H3K27 acetylation (101), while 5-hydroxytryptophan influences CD8+ T cell exhaustion through AHR signaling (123). Serotonylation of GAPDH provides an additional regulatory mechanism in CD8+ T cells by enhancing glycolytic metabolism, which in turn promotes T cell proliferation and IFN-γ production for effective antitumor immunity (100).
B cells demonstrate diverse responses to AAA metabolites. Indole-3-acetate coordinates the development of IL-35-producing B cells in the presence of LPS (83), through a coordinated activation of PXR and TLR4 pathways, demonstrating pathway convergence in immune regulation. Phenethylamine increases IgE expression through TAAR1 signaling, while serotonin promotes B cell proliferation via HTR1A activation (76). These diverse effects underscore how different receptor systems can be engaged to modulate various aspects of B cell function.
The complexity of AAA metabolite-mediated immune regulation extends to the tissue microenvironment. For instance, p-Cresol modulates immune responses through EGFR and TLR4 crosstalk in epithelial cells, affecting CCL20 production and subsequent immune cell recruitment (84). This highlights how metabolites can influence immune function through both direct and indirect mechanisms.
Pathway crosstalk for context-dependent regulation
The regulation of immune responses by AAA metabolites involves sophisticated networks of interacting signaling pathways that create context-dependent outcomes. While individual pathways demonstrate distinct regulatory functions, their crosstalk enables more nuanced control of immune responses based on specific tissue environments and physiological conditions.
AHR pathway demonstrates extensive crosstalk with other major signaling networks in response to Trp metabolites. The interaction between AHR and NF-κB pathways represents a crucial regulatory axis, with bidirectional control mechanisms. NF-κB RelA directly controls AHR expression through promoter binding, while AHR activation induces CYP1A1 expression, generating reactive oxygen species that can activate NF-κB signaling (147,148). This reciprocal regulation enables coordinated control of inflammatory responses and oxidative stress. Additionally, indole derivatives like indole-3-acetate inhibit NLRP3 inflammasome, through which suppress inflammation in macrophages and promote tolerogenic phenotypes in DCs (18,51).
AHR signaling also converges with the NRF2 pathway to orchestrate antioxidant responses. NRF2 protein is a transcription factor binding on the ARE. Key target genes such as Nqo1 and Sod1 contain both XRE and ARE sequences in their promoters, enabling integrated regulation by both pathways. Furthermore, AHR activation can directly induce NRF2 expression through XRE elements, establishing a feed-forward loop that amplifies antioxidant responses (149). In addition to this transcriptional crosstalk, indole acrylic acid has been shown to activate both AHR and NRF2-ARE pathways, promoting antioxidant and anti-inflammatory immune responses (115). These mechanisms of pathway convergence demonstrate the multiple levels at which AHR and NRF2 signaling can coordinate cellular responses.
Despite this evidence for pathway crosstalk, our understanding of how these networks integrate signals in different physiological and disease contexts remains limited. Further investigation is needed to fully map these regulatory networks and their context-dependent effects.
DISEASE IMPLICATIONS AND THERAPEUTIC APPLICATIONS
The intricate networks of AAA metabolite signaling pathways play crucial roles in various pathological conditions, offering potential therapeutic opportunities while presenting complex challenges. Understanding these disease-specific mechanisms is essential for developing targeted interventions that can modulate immune responses effectively.
Disease implications
Autoimmune diseases
AHR functions as a central immunological regulator in autoimmune conditions through its modulation of inflammatory and immune pathways. In rheumatoid arthritis (RA), AHR activation exhibits distinct context-dependent effects. AHR activation by TCDD or FICZ promotes disease progression through enhanced osteoclast formation and increased IL-17-expressing cell population within joint tissues. Supporting this pro-inflammatory role, AHR-deficient mice show reduced levels of inflammatory mediators (matrix metalloproteinase 3, IL-1β, and IL-6) and decreased disease severity, primarily through T cell-dependent rather than macrophage-dependent mechanisms (150). Conversely, intestinal AHR activation by oral administration of Norisoboldine promotes Treg differentiation in gut-associated lymphoid tissues, leading to anti-arthritic effects through Th17/Treg balance (151). Mechanistically, TCDD-activated AHR directly bind to the Foxp3 promoter, thereby inducing Foxp3 expression and promoting T cell differentiation (145,146). Similarly, AHR activation by kynurenine also triggers Foxp3 expression in T cells, leading to their differentiation into Foxp3+ Tregs (152). Additionally, microbiota-derived Trp metabolites, particularly indole-3-acrylic acid, indole-3-propionate, and indole-3-acetate, activate AHR signaling in the intestine to regulate Th17/Treg balance and attenuate RA-associated inflammation (Fig. 4A) (153). In psoriasis, AHR’s multifaceted effects manifest through regulation of inflammatory responses, where its antagonism leads to excessive inflammation, while activation by FICZ reduces disease severity through modulation of stromal-immune system interactions. Of clinical relevance, elevated AHR expression in RA patients indicates its role as an environmental response mediator, while in psoriasis, the therapeutic efficacy of the AHR agonist Tapinarof demonstrates its potential as a treatment target (5).
Figure 4. AAA metabolite signaling in inflammatory diseases. (A) In rheumatoid arthritis, dysbiosis leads to disrupted AHR activation, resulting in Th17/Treg imbalance and increased pro-inflammatory cytokine production. (B) During inflammatory bowel disease, deficient AHR activation in the intestinal epithelium causes impaired barrier function through decreased tight junction proteins and mucus production, promoting inflammation. (C) In diabetic foot ulcers, phenylpyruvate triggers M1 macrophage polarization via NLRP3 pathways, leading to increased inflammation and compromised wound healing.
IBD
AHR emerges as a critical mediator in IBD through its pleiotropic effects on intestinal homeostasis, where decreased intestinal AHR expression has been consistently observed in IBD patients, particularly in Crohn’s disease (154). AHR signaling maintains mucosal barrier integrity through multiple mechanisms: regulation of tight junction proteins, promotion of goblet cell differentiation, and enhancement of mucus production, collectively establishing a robust defense against pathogenic invasion (154,155). Within the immune compartment, AHR activation modulates both innate and adaptive immune responses by regulating the differentiation of Th17 cell and Treg, while also inducing the production of anti-inflammatory cytokines such as IL-10 and IL-22 (Fig. 4B) (156).
Notably, AHR plays a crucial role in maintaining vascular homeostasis in the intestine, affecting angiogenesis and vascular repair mechanisms that are often dysregulated in IBD (16). As a sensor for microbiota-derived metabolites, AHR establishes a critical link between microbial metabolism and host immune responses. The integrated effects of AHR signaling on barrier function, immune regulation, and microbial interactions create a complex regulatory network that, when disrupted, contributes to IBD pathogenesis (157).
Diabetic foot ulcer (DFU)
Recent metabolomic profiling of DFUs has revealed a critical pathological feature: the significant accumulation of phenylpyruvate. Initial analyses demonstrated that elevated phenylpyruvate levels positively correlate with NLRP3 inflammasome activation and chronic inflammation, suggesting its potential role in impaired wound healing. This correlation manifests clinically through the persistent activation of proinflammatory M1 macrophages and compromised transition to healing-associated M2 phenotypes, establishing a pathological inflammatory cycle that impedes proper wound resolution (158).
The molecular basis for this pathology has been precisely delineated: phenylpyruvate enters macrophages through the CD36 scavenger receptor and specifically binds to palmitoyl-protein thioesterase 1 (PPT1) protein at residues Lys229 and Gly245. This binding effectively inhibits PPT1’s depalmitoylase activity, resulting in increased palmitoylation of NLRP3 protein, particularly at cysteine residue 6 within its pyrin domain. The enhanced NLRP3 palmitoylation not only augments protein stability and reduces lysosomal degradation but also promotes interaction with the apoptosis-associated speck-like protein containing a caspase recruitment domain adaptor protein, facilitating inflammasome assembly. This stabilized complex trigger sustained release of inflammatory mediators, including IL-1β, IL-18, and TNF-α, perpetuating a self-reinforcing inflammatory state characterized by reduced epidermal thickness, decreased collagen deposition, and compromised angiogenesis (Fig. 4C). Notably, therapeutic intervention through dietary Phe restriction can attenuate phenylpyruvate accumulation and associated inflammation, suggesting a promising treatment strategy for these otherwise refractory wounds (116).
Therapeutic considerations and future directions
The therapeutic application of AAA metabolite pathway modulation represents a complex landscape filled with both promising opportunities and significant challenges. Our growing understanding of these pathways has revealed their potential as therapeutic targets, yet several critical considerations must be carefully weighed as we move forward in this field (159,160).
The context-dependent nature of AAA metabolite signaling presents perhaps the most significant challenge for therapeutic development. The same metabolite or pathway can exhibit opposing effects depending on the disease state or tissue environment. For example, AHR activation in RA can either exacerbate disease progression through enhanced osteoclast differentiation or ameliorate symptoms through suppression of inflammatory responses (161). This dual nature of signaling necessitates more comprehensive research on understanding the underlying molecular mechanisms by which diverse signals are integrated in disease states. Based on that, the development of highly targeted therapeutic approaches that can achieve tissue-specific and context-appropriate modulation.
Furthermore, the redundancy and interconnectedness of metabolic pathways adds another layer of complexity to therapeutic targeting. Multiple pathways often regulate similar immune functions, creating potential compensatory mechanisms that could limit the effectiveness of single-pathway interventions. Our understanding of these compensatory networks remains incomplete, highlighting the need for comprehensive pathway mapping before developing targeted therapies.
Looking ahead, several critical knowledge gaps in host-microbe metabolic interactions must be addressed. Understanding how different bacterial species contribute to the metabolite pool, how dietary factors influence microbial metabolite production, and how host-microbe interactions regulate metabolite availability will be essential. Additionally, investigating the temporal dynamics of microbiota-dependent metabolite production and their influence on immune responses across different developmental stages and disease states will provide crucial insights for therapeutic development. The development of more sophisticated tools for monitoring metabolite concentrations and pathway activity in real-time will be vital for progressing these research directions (160).
The path forward requires a balanced approach that acknowledges both the potential and limitations of targeting AAA metabolite pathways. Success will likely come from therapeutic strategies that can achieve precise, context-specific pathway modulation while maintaining overall metabolic homeostasis. As our understanding of these complex networks continues to evolve, we may uncover new opportunities for therapeutic intervention in inflammatory conditions.
CONCLUDING REMARKS
Recent advances in understanding AAA metabolite-mediated immune regulation have revealed sophisticated molecular mechanisms through which these compounds influence immune cell function and homeostasis. The convergence of multiple signaling pathways—including membrane receptor-mediated rapid responses, nuclear receptor-dependent transcriptional regulation, and post-translational modifications— are considered to enable precise temporal and spatial control of immune responses. However, several critical questions remain unexplored. First, while individual pathways are well-characterized, how multiple signals in response to the same metabolites as well as how signals by multiple distinct metabolites are integrated under physiological conditions remains poorly understood. Second, the tissue microenvironment-specific concentrations and temporal dynamics of many metabolites await precise determination. Third, the mechanisms governing metabolite availability through host-microbe metabolic networks require further investigation, particularly in different disease states. Finally, the therapeutic potential of targeting AAA metabolite pathways, while promising, faces challenges due to their context-dependent effects and pathway redundancy. Future research addressing these knowledge gaps, supported by advanced technologies such as single-cell metabolomics and spatial metabolomics that enable simultaneous analysis of metabolites at the single-cell resolution from the tissue microenvironment (162,163), will be crucial for developing targeted therapeutic strategies that can effectively modulate immune responses while maintaining metabolic homeostasis.
ACKNOWLEDGEMENTS
This study was supported by Creative-Pioneering Researchers Program through Seoul National University, National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (RS-2023-00217157 and RS-2023-00209621).
Abbreviations
- AAA
aromatic amino acid
- ABC
ATP-binding cassette
- AHR
aryl hydrocarbon receptor
- ARE
antioxidant response element
- cAMP
cyclic AMP
- CREB
cAMP response element-binding protein
- DAT
dopamine transporter
- DC
dendritic cell
- DDC
DOPA decarboxylase
- DFU
diabetic foot ulcer
- EGFR
epidermal growth factor receptor
- ENS
enteric nervous system
- FICZ
6-formylindolo(3,2-b)carbazole
- GF
germ-free
- GI
gastrointestinal
- GPCR
G protein-coupled receptor
- GPR35
G-protein coupled receptor 35
- HCA3
hydroxycarboxylic acid receptor 3
- IBD
inflammatory bowel disease
- IDO
indoleamine 2,3-dioxygenase
- IEC
intestinal epithelial cell
- ILC
innate lymphoid cell
- MAT
monoamine transporter
- NET
norepinephrine transporter
- NLRP3
nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3
- Nrf2
NF erythroid 2-related factor 2
- OCT
organic cation transporter
- PAH
phenylalanine hydroxylase
- Phe
phenylalanine
- PKA
protein kinase A
- PPT1
palmitoyl-protein thioesterase 1
- PXR
pregnane X receptor
- RA
rheumatoid arthritis
- SERT
serotonin transporter
- TAAR
trace amine-associated receptor
- TCDD
2,3,7,8-Tetrachlorodibenzo-p-dioxin
- TGM
transglutaminase
- TPH
tryptophan hydroxylase
- Trp
tryptophan
- Tyr
tyrosine
- XRE
xenobiotic response element
Footnotes
Conflict of Interest: The authors declare no potential conflicts of interest.
- Conceptualization: Bang YJ.
- Data curation: Shin HK, Bang YJ.
- Formal analysis: Shin HK, Bang YJ.
- Funding acquisition: Bang YJ.
- Investigation: Shin HK, Bang YJ.
- Supervision: Bang YJ.
- Visualization: Shin HK, Bang YJ.
- Writing - original draft: Shin HK.
- Writing - review & editing: Bang YJ.
References
- 1.Kayama H, Okumura R, Takeda K. Interaction between the microbiota, epithelia, and immune cells in the intestine. Annu Rev Immunol. 2020;38:23–48. doi: 10.1146/annurev-immunol-070119-115104. [DOI] [PubMed] [Google Scholar]
- 2.Lavelle A, Sokol H. Gut microbiota-derived metabolites as key actors in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. 2020;17:223–237. doi: 10.1038/s41575-019-0258-z. [DOI] [PubMed] [Google Scholar]
- 3.O’Neill LAJ, Kishton RJ, Rathmell J. A guide to immunometabolism for immunologists. Nat Rev Immunol. 2016;16:553–565. doi: 10.1038/nri.2016.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Liu Y, Hou Y, Wang G, Zheng X, Hao H. Gut microbial metabolites of aromatic amino acids as signals in host-microbe interplay. Trends Endocrinol Metab. 2020;31:818–834. doi: 10.1016/j.tem.2020.02.012. [DOI] [PubMed] [Google Scholar]
- 5.Neavin DR, Liu D, Ray B, Weinshilboum RM. The role of the aryl hydrocarbon receptor (AHR) in immune and inflammatory diseases. Int J Mol Sci. 2018;19:3851. doi: 10.3390/ijms19123851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Pogson CI, Knowles RG, Salter M. The control of aromatic amino acid catabolism and its relationship to neurotransmitter amine synthesis. Crit Rev Neurobiol. 1989;5:29–64. [PubMed] [Google Scholar]
- 7.Zhang Q, Chen S, Guo Y, He F, Fu J, Ren W. Phenylalanine diminishes M1 macrophage inflammation. Sci China Life Sci. 2023;66:2862–2876. doi: 10.1007/s11427-022-2296-0. [DOI] [PubMed] [Google Scholar]
- 8.Roager HM, Licht TR. Microbial tryptophan catabolites in health and disease. Nat Commun. 2018;9:3294. doi: 10.1038/s41467-018-05470-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Dodd D, Spitzer MH, Van Treuren W, Merrill BD, Hryckowian AJ, Higginbottom SK, Le A, Cowan TM, Nolan GP, Fischbach MA, et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature. 2017;551:648–652. doi: 10.1038/nature24661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zhang Y, Wang Y. The dual roles of serotonin in antitumor immunity. Pharmacol Res. 2024;205:107255. doi: 10.1016/j.phrs.2024.107255. [DOI] [PubMed] [Google Scholar]
- 11.Khemaissa S, Sagan S, Walrant A. Tryptophan, an amino-acid endowed with unique properties and its many roles in membrane proteins. Crystals. 2021;11:1032. [Google Scholar]
- 12.Kumar V, Sharma A, Kaur R, Thukral AK, Bhardwaj R, Ahmad P. Differential distribution of amino acids in plants. Amino Acids. 2017;49:821–869. doi: 10.1007/s00726-017-2401-x. [DOI] [PubMed] [Google Scholar]
- 13.Tessari P, Lante A, Mosca G. Essential amino acids: master regulators of nutrition and environmental footprint? Sci Rep. 2016;6:26074. doi: 10.1038/srep26074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Esser C. Biology and function of the aryl hydrocarbon receptor: report of an international and interdisciplinary conference. Arch Toxicol. 2012;86:1323–1329. doi: 10.1007/s00204-012-0818-2. [DOI] [PubMed] [Google Scholar]
- 15.Rothhammer V, Quintana FJ. The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease. Nat Rev Immunol. 2019;19:184–197. doi: 10.1038/s41577-019-0125-8. [DOI] [PubMed] [Google Scholar]
- 16.Stockinger B, Shah K, Wincent E. AHR in the intestinal microenvironment: safeguarding barrier function. Nat Rev Gastroenterol Hepatol. 2021;18:559–570. doi: 10.1038/s41575-021-00430-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Obata Y, Castaño Á, Boeing S, Bon-Frauches AC, Fung C, Fallesen T, de Agüero MG, Yilmaz B, Lopes R, Huseynova A, et al. Neuronal programming by microbiota regulates intestinal physiology. Nature. 2020;578:284–289. doi: 10.1038/s41586-020-1975-8. [DOI] [PubMed] [Google Scholar]
- 18.Yu K, Li Q, Sun X, Peng X, Tang Q, Chu H, Zhou L, Wang B, Zhou Z, Deng X, et al. Bacterial indole-3-lactic acid affects epithelium-macrophage crosstalk to regulate intestinal homeostasis. Proc Natl Acad Sci U S A. 2023;120:e2309032120. doi: 10.1073/pnas.2309032120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Sterchi EE, Woodley JF. Peptide hydrolases of the human small intestinal mucosa: distribution of activities between brush border membranes and cytosol. Clin Chim Acta. 1980;102:49–56. doi: 10.1016/0009-8981(80)90432-5. [DOI] [PubMed] [Google Scholar]
- 20.Rowan AM, Moughan PJ, Wilson MN, Maher K, Tasman-Jones C. Comparison of the ileal and faecal digestibility of dietary amino acids in adult humans and evaluation of the pig as a model animal for digestion studies in man. Br J Nutr. 1994;71:29–42. doi: 10.1079/bjn19940108. [DOI] [PubMed] [Google Scholar]
- 21.Wang Y, Song W, Wang J, Wang T, Xiong X, Qi Z, Fu W, Yang X, Chen YG. Single-cell transcriptome analysis reveals differential nutrient absorption functions in human intestine. J Exp Med. 2020;217:e20191130. doi: 10.1084/jem.20191130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Jando J, Camargo SMR, Herzog B, Verrey F. Expression and regulation of the neutral amino acid transporter B0AT1 in rat small intestine. PLoS One. 2017;12:e0184845. doi: 10.1371/journal.pone.0184845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Anderson CMH, Howard A, Walters JRF, Ganapathy V, Thwaites DT. Taurine uptake across the human intestinal brush-border membrane is via two transporters: H+-coupled PAT1 (SLC36A1) and Na+- and Cl−-dependent TauT (SLC6A6) J Physiol. 2009;587:731–744. doi: 10.1113/jphysiol.2008.164228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bröer S. Intestinal amino acid transport and metabolic health. Annu Rev Nutr. 2023;43:73–99. doi: 10.1146/annurev-nutr-061121-094344. [DOI] [PubMed] [Google Scholar]
- 25.Fotiadis D, Kanai Y, Palacín M. The SLC3 and SLC7 families of amino acid transporters. Mol Aspects Med. 2013;34:139–158. doi: 10.1016/j.mam.2012.10.007. [DOI] [PubMed] [Google Scholar]
- 26.Saier MH., Jr Families of transmembrane transporters selective for amino acids and their derivatives. Microbiology (Reading) 2000;146:1775–1795. doi: 10.1099/00221287-146-8-1775. [DOI] [PubMed] [Google Scholar]
- 27.Burkovski A, Krämer R. Bacterial amino acid transport proteins: occurrence, functions, and significance for biotechnological applications. Appl Microbiol Biotechnol. 2002;58:265–274. doi: 10.1007/s00253-001-0869-4. [DOI] [PubMed] [Google Scholar]
- 28.Steglich M, Hofmann JD, Helmecke J, Sikorski J, Spröer C, Riedel T, Bunk B, Overmann J, Neumann-Schaal M, Nübel U. Convergent loss of ABC transporter genes from Clostridioides difficile genomes is associated with impaired tyrosine uptake and p-Cresol production. Front Microbiol. 2018;9:901. doi: 10.3389/fmicb.2018.00901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Wang L, Chen J, Zhang J, Xu F, Luo X, Pang H, Zhang M, Duan Y, Cai Y, Wu G, et al. Changes in intestinal microbiota, immunity and metabolism caused by mixed Lactiplantibacillus plantarum and Bacillus subtilis-fermented feed in Bamei pigs. Chem Biol Technol Agric. 2024;11:76. [Google Scholar]
- 30.Malinauskaite L, Quick M, Reinhard L, Lyons JA, Yano H, Javitch JA, Nissen P. A mechanism for intracellular release of Na+ by neurotransmitter/sodium symporters. Nat Struct Mol Biol. 2014;21:1006–1012. doi: 10.1038/nsmb.2894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Neis EPJG, Dejong CHC, Rensen SS. The role of microbial amino acid metabolism in host metabolism. Nutrients. 2015;7:2930–2946. doi: 10.3390/nu7042930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mesnage R, Antoniou MN. Computational modelling provides insight into the effects of glyphosate on the shikimate pathway in the human gut microbiome. Curr Res Toxicol. 2020;1:25–33. doi: 10.1016/j.crtox.2020.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Doroshenko V, Airich L, Vitushkina M, Kolokolova A, Livshits V, Mashko S. YddG from Escherichia coli promotes export of aromatic amino acids. FEMS Microbiol Lett. 2007;275:312–318. doi: 10.1111/j.1574-6968.2007.00894.x. [DOI] [PubMed] [Google Scholar]
- 34.Wikoff WR, Anfora AT, Liu J, Schultz PG, Lesley SA, Peters EC, Siuzdak G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci U S A. 2009;106:3698–3703. doi: 10.1073/pnas.0812874106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bender DA. Biochemistry of tryptophan in health and disease. Mol Aspects Med. 1983;6:101–197. doi: 10.1016/0098-2997(83)90005-5. [DOI] [PubMed] [Google Scholar]
- 36.Hayaishi O. Properties and function of indoleamine 2,3-dioxygenase. J Biochem. 1976;79:13P–21P. doi: 10.1093/oxfordjournals.jbchem.a131115. [DOI] [PubMed] [Google Scholar]
- 37.Pallotta MT, Rossini S, Suvieri C, Coletti A, Orabona C, Macchiarulo A, Volpi C, Grohmann U. Indoleamine 2,3-dioxygenase 1 (IDO1): an up-to-date overview of an eclectic immunoregulatory enzyme. FEBS J. 2022;289:6099–6118. doi: 10.1111/febs.16086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Valladares R, Bojilova L, Potts AH, Cameron E, Gardner C, Lorca G, Gonzalez CF. Lactobacillus johnsonii inhibits indoleamine 2,3-dioxygenase and alters tryptophan metabolite levels in BioBreeding rats. FASEB J. 2013;27:1711–1720. doi: 10.1096/fj.12-223339. [DOI] [PubMed] [Google Scholar]
- 39.Dehhaghi M, Kazemi Shariat Panahi H, Guillemin GJ. Microorganisms, tryptophan metabolism, and kynurenine pathway: a complex interconnected loop influencing human health status. Int J Tryptophan Res. 2019;12:1178646919852996. doi: 10.1177/1178646919852996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Herr N, Bode C, Duerschmied D. The effects of serotonin in immune cells. Front Cardiovasc Med. 2017;4:48. doi: 10.3389/fcvm.2017.00048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Bondy SC, Campbell A. Melatonin and regulation of immune function: impact on numerous diseases. Curr Aging Sci. 2020;13:92–101. doi: 10.2174/1874609813666200711153223. [DOI] [PubMed] [Google Scholar]
- 42.Møller N, Meek S, Bigelow M, Andrews J, Nair KS. The kidney is an important site for in vivo phenylalanine-to-tyrosine conversion in adult humans: a metabolic role of the kidney. Proc Natl Acad Sci U S A. 2000;97:1242–1246. doi: 10.1073/pnas.97.3.1242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Hufton SE, Jennings IG, Cotton RG. Structure and function of the aromatic amino acid hydroxylases. Biochem J. 1995;311:353–366. doi: 10.1042/bj3110353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Clarke JTR, Bier DM. The conversion of phenylalanine to tyrosine in man. Direct measurement by continuous intravenous tracer infusions of L-[ring-2H5]phenylalanine and L-[1-13C] tyrosine in the postabsorptive state. Metabolism. 1982;31:999–1005. doi: 10.1016/0026-0495(82)90142-1. [DOI] [PubMed] [Google Scholar]
- 45.Flydal MI, Martinez A. Phenylalanine hydroxylase: function, structure, and regulation. IUBMB Life. 2013;65:341–349. doi: 10.1002/iub.1150. [DOI] [PubMed] [Google Scholar]
- 46.Chen L, Yang P, Hu L, Yang L, Chu H, Hou X. Modulating phenylalanine metabolism by L. acidophilus alleviates alcohol-related liver disease through enhancing intestinal barrier function. Cell Biosci. 2023;13:24. doi: 10.1186/s13578-023-00974-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Eisenhofer G, Aneman A, Friberg P, Hooper D, Fåndriks L, Lonroth H, Hunyady B, Mezey E. Substantial production of dopamine in the human gastrointestinal tract. J Clin Endocrinol Metab. 1997;82:3864–3871. doi: 10.1210/jcem.82.11.4339. [DOI] [PubMed] [Google Scholar]
- 48.Rohrbach JM, Süsskind D, Grüb M. The melanocyte and the eye: a review with special emphasis on the cornea. Klin Monatsbl Augenheilkd. 2012;229:42–47. doi: 10.1055/s-0031-1273400. [DOI] [PubMed] [Google Scholar]
- 49.Russell WR, Duncan SH, Scobbie L, Duncan G, Cantlay L, Calder AG, Anderson SE, Flint HJ. Major phenylpropanoid-derived metabolites in the human gut can arise from microbial fermentation of protein. Mol Nutr Food Res. 2013;57:523–535. doi: 10.1002/mnfr.201200594. [DOI] [PubMed] [Google Scholar]
- 50.Hubbard TD, Murray IA, Perdew GH. Indole and tryptophan metabolism: endogenous and dietary routes to ah receptor activation. Drug Metab Dispos. 2015;43:1522–1535. doi: 10.1124/dmd.115.064246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Alexeev EE, Lanis JM, Kao DJ, Campbell EL, Kelly CJ, Battista KD, Gerich ME, Jenkins BR, Walk ST, Kominsky DJ, et al. Microbiota-derived indole metabolites promote human and murine intestinal homeostasis through regulation of interleukin-10 receptor. Am J Pathol. 2018;188:1183–1194. doi: 10.1016/j.ajpath.2018.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Lee JH, Lee J. Indole as an intercellular signal in microbial communities. FEMS Microbiol Rev. 2010;34:426–444. doi: 10.1111/j.1574-6976.2009.00204.x. [DOI] [PubMed] [Google Scholar]
- 53.Honoré AH, Aunsbjerg SD, Ebrahimi P, Thorsen M, Benfeldt C, Knøchel S, Skov T. Metabolic footprinting for investigation of antifungal properties of Lactobacillus paracasei . Anal Bioanal Chem. 2016;408:83–96. doi: 10.1007/s00216-015-9103-6. [DOI] [PubMed] [Google Scholar]
- 54.Lee JH, Kim YG, Kim M, Kim E, Choi H, Kim Y, Lee J. Indole-associated predator-prey interactions between the nematode Caenorhabditis elegans and bacteria. Environ Microbiol. 2017;19:1776–1790. doi: 10.1111/1462-2920.13649. [DOI] [PubMed] [Google Scholar]
- 55.Narayanan TK, Rao GR. Beta-indoleethanol and beta-indolelactic acid production by Candida species: their antibacterial and autoantibiotic action. Antimicrob Agents Chemother. 1976;9:375–380. doi: 10.1128/aac.9.3.375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Bommarius B, Anyanful A, Izrayelit Y, Bhatt S, Cartwright E, Wang W, Swimm AI, Benian GM, Schroeder FC, Kalman D. A family of indoles regulate virulence and Shiga toxin production in pathogenic E. coli . PLoS One. 2013;8:e54456. doi: 10.1371/journal.pone.0054456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Sanidad KZ, Rager SL, Carrow HC, Ananthanarayanan A, Callaghan R, Hart LR, Li T, Ravisankar P, Brown JA, Amir M, et al. Gut bacteria-derived serotonin promotes immune tolerance in early life. Sci Immunol. 2024;9:eadj4775. doi: 10.1126/sciimmunol.adj4775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Walsh J, Olavarria-Ramirez L, Lach G, Boehme M, Dinan TG, Cryan JF, Griffin BT, Hyland NP, Clarke G. Impact of host and environmental factors on β-glucuronidase enzymatic activity: implications for gastrointestinal serotonin. Am J Physiol Gastrointest Liver Physiol. 2020;318:G816–G826. doi: 10.1152/ajpgi.00026.2020. [DOI] [PubMed] [Google Scholar]
- 59.Marcobal A, De las Rivas B, Landete JM, Tabera L, Muñoz R. Tyramine and phenylethylamine biosynthesis by food bacteria. Crit Rev Food Sci Nutr. 2012;52:448–467. doi: 10.1080/10408398.2010.500545. [DOI] [PubMed] [Google Scholar]
- 60.Shimazu S, Miklya I. Pharmacological studies with endogenous enhancer substances: beta-phenylethylamine, tryptamine, and their synthetic derivatives. Prog Neuropsychopharmacol Biol Psychiatry. 2004;28:421–427. doi: 10.1016/j.pnpbp.2003.11.016. [DOI] [PubMed] [Google Scholar]
- 61.Al Hinai EA, Kullamethee P, Rowland IR, Swann J, Walton GE, Commane DM. Modelling the role of microbial p-Cresol in colorectal genotoxicity. Gut Microbes. 2019;10:398–411. doi: 10.1080/19490976.2018.1534514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Zeng X, Xing X, Gupta M, Keber FC, Lopez JG, Lee YJ, Roichman A, Wang L, Neinast MD, Donia MS, et al. Gut bacterial nutrient preferences quantified in vivo . Cell. 2022;185:3441–3456.e19. doi: 10.1016/j.cell.2022.07.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Whitt DD, Demoss RD. Effect of microflora on the free amino acid distribution in various regions of the mouse gastrointestinal tract. Appl Microbiol. 1975;30:609–615. doi: 10.1128/am.30.4.609-615.1975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Li TT, Chen X, Huo D, Arifuzzaman M, Qiao S, Jin WB, Shi H, Li XV, Iliev ID, et al. JRI Live Cell Bank Consortium. Microbiota metabolism of intestinal amino acids impacts host nutrient homeostasis and physiology. Cell Host Microbe. 2024;32:661–675.e10. doi: 10.1016/j.chom.2024.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, Nagler CR, Ismagilov RF, Mazmanian SK, Hsiao EY. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell. 2015;161:264–276. doi: 10.1016/j.cell.2015.02.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Martin-Gallausiaux C, Larraufie P, Jarry A, Béguet-Crespel F, Marinelli L, Ledue F, Reimann F, Blottière HM, Lapaque N. Butyrate produced by commensal bacteria down-regulates indolamine 2,3-dioxygenase 1 (IDO-1) expression via a dual mechanism in human intestinal epithelial cells. Front Immunol. 2018;9:2838. doi: 10.3389/fimmu.2018.02838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Vicentini FA, Keenan CM, Wallace LE, Woods C, Cavin JB, Flockton AR, Macklin WB, Belkind-Gerson J, Hirota SA, Sharkey KA. Intestinal microbiota shapes gut physiology and regulates enteric neurons and glia. Microbiome. 2021;9:210. doi: 10.1186/s40168-021-01165-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Anitha M, Vijay-Kumar M, Sitaraman SV, Gewirtz AT, Srinivasan S. Gut microbial products regulate murine gastrointestinal motility via Toll-like receptor 4 signaling. Gastroenterology. 2012;143:1006–16.e4. doi: 10.1053/j.gastro.2012.06.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Husebye E, Hellström PM, Sundler F, Chen J, Midtvedt T. Influence of microbial species on small intestinal myoelectric activity and transit in germ-free rats. Am J Physiol Gastrointest Liver Physiol. 2001;280:G368–G380. doi: 10.1152/ajpgi.2001.280.3.G368. [DOI] [PubMed] [Google Scholar]
- 70.Zheng Z, Tang J, Hu Y, Zhang W. Role of gut microbiota-derived signals in the regulation of gastrointestinal motility. Front Med (Lausanne) 2022;9:961703. doi: 10.3389/fmed.2022.961703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Starke S, Harris DMM, Zimmermann J, Schuchardt S, Oumari M, Frank D, Bang C, Rosenstiel P, Schreiber S, Frey N, et al. Amino acid auxotrophies in human gut bacteria are linked to higher microbiome diversity and long-term stability. ISME J. 2023;17:2370–2380. doi: 10.1038/s41396-023-01537-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ordóñez-Morán P, Muñoz A. Nuclear receptors: genomic and non-genomic effects converge. Cell Cycle. 2009;8:1675–1680. doi: 10.4161/cc.8.11.8579. [DOI] [PubMed] [Google Scholar]
- 73.Newton AC, Bootman MD, Scott JD. Second messengers. Cold Spring Harb Perspect Biol. 2016;8:a005926. doi: 10.1101/cshperspect.a005926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Zhong XP, Shin J, Gorentla BK, O’Brien T, Srivatsan S, Xu L, Chen Y, Xie D, Pan H. Receptor signaling in immune cell development and function. Immunol Res. 2011;49:109–123. doi: 10.1007/s12026-010-8175-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Laeremans T, Sands ZA, Claes P, De Blieck A, De Cesco S, Triest S, Busch A, Felix D, Kumar A, Jaakola VP, et al. Accelerating GPCR drug discovery with conformation-stabilizing VHHs. Front Mol Biosci. 2022;9:863099. doi: 10.3389/fmolb.2022.863099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Moiseenko VI, Apryatina VA, Gainetdinov RR, Apryatin SA. Trace amine-associated receptors’ role in immune system functions. Biomedicines. 2024;12:893. doi: 10.3390/biomedicines12040893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Peters A, Krumbholz P, Jäger E, Heintz-Buschart A, Çakir MV, Rothemund S, Gaudl A, Ceglarek U, Schöneberg T, Stäubert C. Metabolites of lactic acid bacteria present in fermented foods are highly potent agonists of human hydroxycarboxylic acid receptor 3. PLoS Genet. 2019;15:e1008145. doi: 10.1371/journal.pgen.1008145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.De Giovanni M, Tam H, Valet C, Xu Y, Looney MR, Cyster JG. GPR35 promotes neutrophil recruitment in response to serotonin metabolite 5-HIAA. Cell. 2022;185:815–830.e19. doi: 10.1016/j.cell.2022.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Barth MC, Ahluwalia N, Anderson TJT, Hardy GJ, Sinha S, Alvarez-Cardona JA, Pruitt IE, Rhee EP, Colvin RA, Gerszten RE. Kynurenic acid triggers firm arrest of leukocytes to vascular endothelium under flow conditions. J Biol Chem. 2009;284:19189–19195. doi: 10.1074/jbc.M109.024042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Arreola R, Becerril-Villanueva E, Cruz-Fuentes C, Velasco-Velázquez MA, Garcés-Alvarez ME, Hurtado-Alvarado G, Quintero-Fabian S, Pavón L. Immunomodulatory effects mediated by serotonin. J Immunol Res. 2015;2015:354957. doi: 10.1155/2015/354957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Bhattarai Y, Jie S, Linden DR, Ghatak S, Mars RAT, Williams BB, Pu M, Sonnenburg JL, Fischbach MA, Farrugia G, et al. Bacterially derived tryptamine increases mucus release by activating a host receptor in a mouse model of inflammatory bowel disease. iScience. 2020;23:101798. doi: 10.1016/j.isci.2020.101798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Conti BJ, Búfalo MC, Golim MA, Bankova V, Sforcin JM. Cinnamic acid is partially involved in propolis immunomodulatory action on human monocytes. Evid Based Complement Alternat Med. 2013;2013:109864. doi: 10.1155/2013/109864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Su X, Zhang M, Qi H, Gao Y, Yang Y, Yun H, Zhang Q, Yang X, Zhang Y, He J, et al. Gut microbiota-derived metabolite 3-idoleacetic acid together with LPS induces IL-35+ B cell generation. Microbiome. 2022;10:13. doi: 10.1186/s40168-021-01205-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Wypych TP, Pattaroni C, Perdijk O, Yap C, Trompette A, Anderson D, Creek DJ, Harris NL, Marsland BJ. Microbial metabolism of L-tyrosine protects against allergic airway inflammation. Nat Immunol. 2021;22:279–286. doi: 10.1038/s41590-020-00856-3. [DOI] [PubMed] [Google Scholar]
- 85.Niesler B, Kapeller J, Hammer C, Rappold G. Serotonin type 3 receptor genes: HTR3A, B, C, D, E. Pharmacogenomics. 2008;9:501–504. doi: 10.2217/14622416.9.5.501. [DOI] [PubMed] [Google Scholar]
- 86.Gaede HC, Yau WM, Gawrisch K. Electrostatic contributions to indole-lipid interactions. J Phys Chem B. 2005;109:13014–13023. doi: 10.1021/jp0511000. [DOI] [PubMed] [Google Scholar]
- 87.Gozal EA, O’Neill BE, Sawchuk MA, Zhu H, Halder M, Chou CC, Hochman S. Anatomical and functional evidence for trace amines as unique modulators of locomotor function in the mammalian spinal cord. Front Neural Circuits. 2014;8:134. doi: 10.3389/fncir.2014.00134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Gebauer L, Jensen O, Neif M, Brockmöller J, Dücker C. Overlap and specificity in the substrate spectra of human monoamine transporters and organic cation transporters 1, 2, and 3. Int J Mol Sci. 2021;22:12816. doi: 10.3390/ijms222312816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Gopinath A, Mackie PM, Phan LT, Mirabel R, Smith AR, Miller E, Franks S, Syed O, Riaz T, Law BK, et al. Who knew? Dopamine transporter activity is critical in innate and adaptive immune responses. Cells. 2023;12:269. doi: 10.3390/cells12020269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Yang L, Chu Z, Liu M, Zou Q, Li J, Liu Q, Wang Y, Wang T, Xiang J, Wang B. Amino acid metabolism in immune cells: essential regulators of the effector functions, and promising opportunities to enhance cancer immunotherapy. J Hematol Oncol. 2023;16:59. doi: 10.1186/s13045-023-01453-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Estudante M, Morais JG, Soveral G, Benet LZ. Intestinal drug transporters: an overview. Adv Drug Deliv Rev. 2013;65:1340–1356. doi: 10.1016/j.addr.2012.09.042. [DOI] [PubMed] [Google Scholar]
- 92.Cox MB, Miller CA., 3rd Cooperation of heat shock protein 90 and p23 in aryl hydrocarbon receptor signaling. Cell Stress Chaperones. 2004;9:4–20. doi: 10.1379/460.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Giguère V. Orphan nuclear receptors: from gene to function. Endocr Rev. 1999;20:689–725. doi: 10.1210/edrv.20.5.0378. [DOI] [PubMed] [Google Scholar]
- 94.Vyhlídalová B, Bartoňková I, Jiskrová E, Li H, Mani S, Dvořák Z. Differential activation of human pregnane X receptor PXR by isomeric mono-methylated indoles in intestinal and hepatic in vitro models. Toxicol Lett. 2020;324:104–110. doi: 10.1016/j.toxlet.2020.02.010. [DOI] [PubMed] [Google Scholar]
- 95.Dubrac S, Elentner A, Ebner S, Horejs-Hoeck J, Schmuth M. Modulation of T lymphocyte function by the pregnane X receptor. J Immunol. 2010;184:2949–2957. doi: 10.4049/jimmunol.0902151. [DOI] [PubMed] [Google Scholar]
- 96.Bader M. Serotonylation: serotonin signaling and epigenetics. Front Mol Neurosci. 2019;12:288. doi: 10.3389/fnmol.2019.00288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Jiang SH, Wang YH, Hu LP, Wang X, Li J, Zhang XL, Zhang ZG. The physiology, pathology and potential therapeutic application of serotonylation. J Cell Sci. 2021;134:jcs257337. doi: 10.1242/jcs.257337. [DOI] [PubMed] [Google Scholar]
- 98.Farrelly LA, Thompson RE, Zhao S, Lepack AE, Lyu Y, Bhanu NV, Zhang B, Loh YE, Ramakrishnan A, Vadodaria KC, et al. Histone serotonylation is a permissive modification that enhances TFIID binding to H3K4me3. Nature. 2019;567:535–539. doi: 10.1038/s41586-019-1024-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ivashkin E, Melnikova V, Kurtova A, Brun NR, Obukhova A, Khabarova MY, Yakusheff A, Adameyko I, Gribble KE, Voronezhskaya EE. Transglutaminase activity determines nuclear localization of serotonin immunoreactivity in the early embryos of invertebrates and vertebrates. ACS Chem Neurosci. 2019;10:3888–3899. doi: 10.1021/acschemneuro.9b00346. [DOI] [PubMed] [Google Scholar]
- 100.Wang X, Fu SQ, Yuan X, Yu F, Ji Q, Tang HW, Li RK, Huang S, Huang PQ, Qin WT, et al. A GAPDH serotonylation system couples CD8+ T cell glycolytic metabolism to antitumor immunity. Mol Cell. 2024;84:760–775.e7. doi: 10.1016/j.molcel.2023.12.015. [DOI] [PubMed] [Google Scholar]
- 101.Jia D, Wang Q, Qi Y, Jiang Y, He J, Lin Y, Sun Y, Xu J, Chen W, Fan L, et al. Microbial metabolite enhances immunotherapy efficacy by modulating T cell stemness in pan-cancer. Cell. 2024;187:1651–1665.e21. doi: 10.1016/j.cell.2024.02.022. [DOI] [PubMed] [Google Scholar]
- 102.Salimi Elizei S, Poormasjedi-Meibod MS, Wang X, Kheirandish M, Ghahary A. Kynurenic acid downregulates IL-17/1L-23 axis in vitro . Mol Cell Biochem. 2017;431:55–65. doi: 10.1007/s11010-017-2975-3. [DOI] [PubMed] [Google Scholar]
- 103.Suzuki S, Tanaka K, Nishikawa K, Suzuki H, Oshima A, Fujiyoshi Y. Structural basis of hydroxycarboxylic acid receptor signaling mechanisms through ligand binding. Nat Commun. 2023;14:5899. doi: 10.1038/s41467-023-41650-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Wu Y, Zhang P, Fan H, Zhang C, Yu P, Liang X, Chen Y. GPR35 acts a dual role and therapeutic target in inflammation. Front Immunol. 2023;14:1254446. doi: 10.3389/fimmu.2023.1254446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Karmakar S, Lal G. Role of serotonin receptor signaling in cancer cells and anti-tumor immunity. Theranostics. 2021;11:5296–5312. doi: 10.7150/thno.55986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.de las Casas-Engel M, Domínguez-Soto A, Sierra-Filardi E, Bragado R, Nieto C, Puig-Kroger A, Samaniego R, Loza M, Corcuera MT, Gómez-Aguado F, et al. Serotonin skews human macrophage polarization through HTR2B and HTR7. J Immunol. 2013;190:2301–2310. doi: 10.4049/jimmunol.1201133. [DOI] [PubMed] [Google Scholar]
- 107.Yue Y, Ke Y, Zheng J, Wang Z, Liu H, Liu S. Microbiota-derived tryptophan metabolism and AMPK/mTOR pathway mediate antidepressant-like effect of Shugan Hewei Decoction. Front Pharmacol. 2024;15:1466336. doi: 10.3389/fphar.2024.1466336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Metz R, Rust S, Duhadaway JB, Mautino MR, Munn DH, Vahanian NN, Link CJ, Prendergast GC. IDO inhibits a tryptophan sufficiency signal that stimulates mTOR: a novel IDO effector pathway targeted by D-1-methyl-tryptophan. OncoImmunology. 2012;1:1460–1468. doi: 10.4161/onci.21716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Livneh I, Cohen-Kaplan V, Fabre B, Abramovitch I, Lulu C, Nataraj NB, Lazar I, Ziv T, Yarden Y, Zohar Y, et al. Regulation of nucleo-cytosolic 26S proteasome translocation by aromatic amino acids via mTOR is essential for cell survival under stress. Mol Cell. 2023;83:3333–3346.e5. doi: 10.1016/j.molcel.2023.08.016. [DOI] [PubMed] [Google Scholar]
- 110.Ihekweazu FD, Engevik MA, Ruan W, Shi Z, Fultz R, Engevik KA, Chang-Graham AL, Freeborn J, Park ES, Venable S, et al. Bacteroides ovatus promotes IL-22 production and reduces trinitrobenzene sulfonic acid-driven colonic inflammation. Am J Pathol. 2021;191:704–719. doi: 10.1016/j.ajpath.2021.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Gargaro M, Scalisi G, Manni G, Briseño CG, Bagadia P, Durai V, Theisen DJ, Kim S, Castelli M, Xu CA, et al. Indoleamine 2,3-dioxygenase 1 activation in mature cDC1 promotes tolerogenic education of inflammatory cDC2 via metabolic communication. Immunity. 2022;55:1032–1050.e14. doi: 10.1016/j.immuni.2022.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Pallotta MT, Orabona C, Volpi C, Vacca C, Belladonna ML, Bianchi R, Servillo G, Brunacci C, Calvitti M, Bicciato S, et al. Indoleamine 2,3-dioxygenase is a signaling protein in long-term tolerance by dendritic cells. Nat Immunol. 2011;12:870–878. doi: 10.1038/ni.2077. [DOI] [PubMed] [Google Scholar]
- 113.Müller T, Dürk T, Blumenthal B, Grimm M, Cicko S, Panther E, Sorichter S, Herouy Y, Di Virgilio F, Ferrari D, et al. 5-Hydroxytryptamine modulates migration, cytokine and chemokine release and T-cell priming capacity of dendritic cells in vitro and in vivo . PLoS One. 2009;4:e6453. doi: 10.1371/journal.pone.0006453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Idzko M, Panther E, Stratz C, Müller T, Bayer H, Zissel G, Dürk T, Sorichter S, Di Virgilio F, Geissler M, et al. The serotoninergic receptors of human dendritic cells: identification and coupling to cytokine release. J Immunol. 2004;172:6011–6019. doi: 10.4049/jimmunol.172.10.6011. [DOI] [PubMed] [Google Scholar]
- 115.Wlodarska M, Luo C, Kolde R, d’Hennezel E, Annand JW, Heim CE, Krastel P, Schmitt EK, Omar AS, Creasey EA, et al. Indoleacrylic acid produced by commensal peptostreptococcus species suppresses inflammation. Cell Host Microbe. 2017;22:25–37.e6. doi: 10.1016/j.chom.2017.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Lv D, Cao X, Zhong L, Dong Y, Xu Z, Rong Y, Xu H, Wang Z, Yang H, Yin R, et al. Targeting phenylpyruvate restrains excessive NLRP3 inflammasome activation and pathological inflammation in diabetic wound healing. Cell Rep Med. 2023;4:101129. doi: 10.1016/j.xcrm.2023.101129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Xue C, Gu X, Zheng Q, Shi Q, Yuan X, Chu Q, Jia J, Su Y, Bao Z, Lu J, et al. Effects of 3-HAA on HCC by regulating the heterogeneous macrophages-a scRNA-seq analysis. Adv Sci (Weinh) 2023;10:e2207074. doi: 10.1002/advs.202207074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Hou Q, Ye L, Liu H, Huang L, Yang Q, Turner JR, Yu Q. Lactobacillus accelerates ISCs regeneration to protect the integrity of intestinal mucosa through activation of STAT3 signaling pathway induced by LPLs secretion of IL-22. Cell Death Differ. 2018;25:1657–1670. doi: 10.1038/s41418-018-0070-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Zelante T, Iannitti RG, Cunha C, De Luca A, Giovannini G, Pieraccini G, Zecchi R, D’Angelo C, Massi-Benedetti C, Fallarino F, et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity. 2013;39:372–385. doi: 10.1016/j.immuni.2013.08.003. [DOI] [PubMed] [Google Scholar]
- 120.Li S. Modulation of immunity by tryptophan microbial metabolites. Front Nutr. 2023;10:1209613. doi: 10.3389/fnut.2023.1209613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Wan M, Ding L, Wang D, Han J, Gao P. Serotonin: a potent immune cell modulator in autoimmune diseases. Front Immunol. 2020;11:186. doi: 10.3389/fimmu.2020.00186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Hayashi T, Mo JH, Gong X, Rossetto C, Jang A, Beck L, Elliott GI, Kufareva I, Abagyan R, Broide DH, et al. 3-Hydroxyanthranilic acid inhibits PDK1 activation and suppresses experimental asthma by inducing T cell apoptosis. Proc Natl Acad Sci U S A. 2007;104:18619–18624. doi: 10.1073/pnas.0709261104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Liu Y, Zhou N, Zhou L, Wang J, Zhou Y, Zhang T, Fang Y, Deng J, Gao Y, Liang X, et al. IL-2 regulates tumor-reactive CD8+ T cell exhaustion by activating the aryl hydrocarbon receptor. Nat Immunol. 2021;22:358–369. doi: 10.1038/s41590-020-00850-9. [DOI] [PubMed] [Google Scholar]
- 124.Bansal T, Alaniz RC, Wood TK, Jayaraman A. The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation. Proc Natl Acad Sci U S A. 2010;107:228–233. doi: 10.1073/pnas.0906112107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Aoki R, Aoki-Yoshida A, Suzuki C, Takayama Y. Indole-3-pyruvic acid, an aryl hydrocarbon receptor activator, suppresses experimental colitis in mice. J Immunol. 2018;201:3683–3693. doi: 10.4049/jimmunol.1701734. [DOI] [PubMed] [Google Scholar]
- 126.Krishnan S, Ding Y, Saedi N, Choi M, Sridharan GV, Sherr DH, Yarmush ML, Alaniz RC, Jayaraman A, Lee K. Gut microbiota-derived tryptophan metabolites modulate inflammatory response in hepatocytes and macrophages. Cell Reports. 2018;23:1099–1111. doi: 10.1016/j.celrep.2018.03.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Cervantes-Barragan L, Chai JN, Tianero MD, Di Luccia B, Ahern PP, Merriman J, Cortez VS, Caparon MG, Donia MS, Gilfillan S, et al. Lactobacillus reuteri induces gut intraepithelial CD4+CD8αα+ T cells. Science. 2017;357:806–810. doi: 10.1126/science.aah5825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Venkatesh M, Mukherjee S, Wang H, Li H, Sun K, Benechet AP, Qiu Z, Maher L, Redinbo MR, Phillips RS, et al. Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4. Immunity. 2014;41:296–310. doi: 10.1016/j.immuni.2014.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Chalise JP, Pallotta MT, Narendra SC, Carlsson B, Iacono A, Namale J, Boon L, Grohmann U, Magnusson M. IDO1 and TGF-β mediate protective effects of IFN-α in antigen-induced arthritis. J Immunol. 2016;197:3142–3151. doi: 10.4049/jimmunol.1502125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Liu Y, Liang X, Dong W, Fang Y, Lv J, Zhang T, Fiskesund R, Xie J, Liu J, Yin X, et al. Tumor-repopulating cells induce PD-1 expression in CD8+ T cells by transferring kynurenine and AhR activation. Cancer Cell. 2018;33:480–494.e7. doi: 10.1016/j.ccell.2018.02.005. [DOI] [PubMed] [Google Scholar]
- 131.Nguyen NT, Kimura A, Nakahama T, Chinen I, Masuda K, Nohara K, Fujii-Kuriyama Y, Kishimoto T. Aryl hydrocarbon receptor negatively regulates dendritic cell immunogenicity via a kynurenine-dependent mechanism. Proc Natl Acad Sci U S A. 2010;107:19961–19966. doi: 10.1073/pnas.1014465107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Campesato LF, Budhu S, Tchaicha J, Weng CH, Gigoux M, Cohen IJ, Redmond D, Mangarin L, Pourpe S, Liu C, et al. Blockade of the AHR restricts a Treg-macrophage suppressive axis induced by L-Kynurenine. Nat Commun. 2020;11:4011. doi: 10.1038/s41467-020-17750-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Gagliani N, Amezcua Vesely MC, Iseppon A, Brockmann L, Xu H, Palm NW, de Zoete MR, Licona-Limón P, Paiva RS, Ching T, et al. Th17 cells transdifferentiate into regulatory T cells during resolution of inflammation. Nature. 2015;523:221–225. doi: 10.1038/nature14452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Stone TW, Williams RO. Modulation of T cells by tryptophan metabolites in the kynurenine pathway. Trends Pharmacol Sci. 2023;44:442–456. doi: 10.1016/j.tips.2023.04.006. [DOI] [PubMed] [Google Scholar]
- 135.Kiank C, Zeden JP, Drude S, Domanska G, Fusch G, Otten W, Schuett C. Psychological stress-induced, IDO1-dependent tryptophan catabolism: implications on immunosuppression in mice and humans. PLoS One. 2010;5:e11825. doi: 10.1371/journal.pone.0011825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Fallarini S, Magliulo L, Paoletti T, de Lalla C, Lombardi G. Expression of functional GPR35 in human iNKT cells. Biochem Biophys Res Commun. 2010;398:420–425. doi: 10.1016/j.bbrc.2010.06.091. [DOI] [PubMed] [Google Scholar]
- 137.Krause D, Suh HS, Tarassishin L, Cui QL, Durafourt BA, Choi N, Bauman A, Cosenza-Nashat M, Antel JP, Zhao ML, et al. The tryptophan metabolite 3-hydroxyanthranilic acid plays anti-inflammatory and neuroprotective roles during inflammation: role of hemeoxygenase-1. Am J Pathol. 2011;179:1360–1372. doi: 10.1016/j.ajpath.2011.05.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Gargaro M, Vacca C, Massari S, Scalisi G, Manni G, Mondanelli G, Mazza EMC, Bicciato S, Pallotta MT, Orabona C, et al. Engagement of nuclear coactivator 7 by 3-hydroxyanthranilic acid enhances activation of aryl hydrocarbon receptor in immunoregulatory dendritic cells. Front Immunol. 2019;10:1973. doi: 10.3389/fimmu.2019.01973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Terness P, Bauer TM, Röse L, Dufter C, Watzlik A, Simon H, Opelz G. Inhibition of allogeneic T cell proliferation by indoleamine 2,3-dioxygenase-expressing dendritic cells: mediation of suppression by tryptophan metabolites. J Exp Med. 2002;196:447–457. doi: 10.1084/jem.20020052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Ito T, Ikeda U, Shimpo M, Yamamoto K, Shimada K. Serotonin increases interleukin-6 synthesis in human vascular smooth muscle cells. Circulation. 2000;102:2522–2527. doi: 10.1161/01.cir.102.20.2522. [DOI] [PubMed] [Google Scholar]
- 141.Iken K, Chheng S, Fargin A, Goulet AC, Kouassi E. Serotonin upregulates mitogen-stimulated B lymphocyte proliferation through 5-HT1A receptors. Cell Immunol. 1995;163:1–9. doi: 10.1006/cimm.1995.1092. [DOI] [PubMed] [Google Scholar]
- 142.Tang Y, Yu Y, Li R, Tao Z, Zhang L, Wang X, Qi X, Li Y, Meng T, Qu H, et al. Phenylalanine promotes alveolar macrophage pyroptosis via the activation of CaSR in ARDS. Front Immunol. 2023;14:1114129. doi: 10.3389/fimmu.2023.1114129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Bhuiyan MS, Ellett F, Murray GL, Kostoulias X, Cerqueira GM, Schulze KE, Mahamad Maifiah MH, Li J, Creek DJ, Lieschke GJ, et al. Acinetobacter baumannii phenylacetic acid metabolism influences infection outcome through a direct effect on neutrophil chemotaxis. Proc Natl Acad Sci U S A. 2016;113:9599–9604. doi: 10.1073/pnas.1523116113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Kim WH, Lillehoj HS, Min W. Indole treatment alleviates intestinal tissue damage induced by chicken coccidiosis through activation of the aryl hydrocarbon receptor. Front Immunol. 2019;10:560. doi: 10.3389/fimmu.2019.00560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Quintana FJ, Basso AS, Iglesias AH, Korn T, Farez MF, Bettelli E, Caccamo M, Oukka M, Weiner HL. Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor. Nature. 2008;453:65–71. doi: 10.1038/nature06880. [DOI] [PubMed] [Google Scholar]
- 146.Lee GR. Molecular mechanisms of T helper cell differentiation and functional specialization. Immune Netw. 2023;23:e4. doi: 10.4110/in.2023.23.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Vogel CFA, Khan EM, Leung PSC, Gershwin ME, Chang WLW, Wu D, Haarmann-Stemmann T, Hoffmann A, Denison MS. Cross-talk between aryl hydrocarbon receptor and the inflammatory response: a role for nuclear factor-κB. J Biol Chem. 2014;289:1866–1875. doi: 10.1074/jbc.M113.505578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Dong H, Hao L, Zhang W, Zhong W, Guo W, Yue R, Sun X, Zhou Z. Activation of AhR-NQO1 signaling pathway protects against alcohol-induced liver injury by improving redox balance. Cell Mol Gastroenterol Hepatol. 2021;12:793–811. doi: 10.1016/j.jcmgh.2021.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Vogel CFA, Van Winkle LS, Esser C, Haarmann-Stemmann T. The aryl hydrocarbon receptor as a target of environmental stressors - implications for pollution mediated stress and inflammatory responses. Redox Biol. 2020;34:101530. doi: 10.1016/j.redox.2020.101530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Nakahama T, Kimura A, Nguyen NT, Chinen I, Hanieh H, Nohara K, Fujii-Kuriyama Y, Kishimoto T. Aryl hydrocarbon receptor deficiency in T cells suppresses the development of collagen-induced arthritis. Proc Natl Acad Sci U S A. 2011;108:14222–14227. doi: 10.1073/pnas.1111786108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Tong B, Yuan X, Dou Y, Wu X, Chou G, Wang Z, Xia Y, Dai Y. Norisoboldine, an isoquinoline alkaloid, acts as an aryl hydrocarbon receptor ligand to induce intestinal Treg cells and thereby attenuate arthritis. Int J Biochem Cell Biol. 2016;75:63–73. doi: 10.1016/j.biocel.2016.03.014. [DOI] [PubMed] [Google Scholar]
- 152.Mezrich JD, Fechner JH, Zhang X, Johnson BP, Burlingham WJ, Bradfield CA. An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J Immunol. 2010;185:3190–3198. doi: 10.4049/jimmunol.0903670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Jiang ZM, Zeng SL, Huang TQ, Lin Y, Wang FF, Gao XJ, Li J, Li P, Liu EH. Sinomenine ameliorates rheumatoid arthritis by modulating tryptophan metabolism and activating aryl hydrocarbon receptor via gut microbiota regulation. Sci Bull (Beijing) 2023;68:1540–1555. doi: 10.1016/j.scib.2023.06.027. [DOI] [PubMed] [Google Scholar]
- 154.Marafini I, Monteleone I, Laudisi F, Monteleone G. Aryl hydrocarbon receptor signalling in the control of gut inflammation. Int J Mol Sci. 2024;25:4527. doi: 10.3390/ijms25084527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Zhou X, Chakraborty D, Murray IA, Coslo D, Kehs Z, Vijay A, Ton C, Desai D, Amin SG, Patterson AD, et al. Aryl hydrocarbon receptor activation coordinates mouse small intestinal epithelial cell programming. Lab Invest. 2023;103:100012. doi: 10.1016/j.labinv.2022.100012. [DOI] [PubMed] [Google Scholar]
- 156.Huang W, Rui K, Wang X, Peng N, Zhou W, Shi X, Lu L, Hu D, Tian J. The aryl hydrocarbon receptor in immune regulation and autoimmune pathogenesis. J Autoimmun. 2023;138:103049. doi: 10.1016/j.jaut.2023.103049. [DOI] [PubMed] [Google Scholar]
- 157.Hou JJ, Ma AH, Qin YH. Activation of the aryl hydrocarbon receptor in inflammatory bowel disease: insights from gut microbiota. Front Cell Infect Microbiol. 2023;13:1279172. doi: 10.3389/fcimb.2023.1279172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Dawson RE, Jenkins BJ. The role of inflammasome-associated innate immune receptors in cancer. Immune Netw. 2024;24:e38. doi: 10.4110/in.2024.24.e38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Loh JS, Mak WQ, Tan LKS, Ng CX, Chan HH, Yeow SH, Foo JB, Ong YS, How CW, Khaw KY. Microbiota-gut-brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct Target Ther. 2024;9:37. doi: 10.1038/s41392-024-01743-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Zhang Y, Chen R, Zhang D, Qi S, Liu Y. Metabolite interactions between host and microbiota during health and disease: which feeds the other? Biomed Pharmacother. 2023;160:114295. doi: 10.1016/j.biopha.2023.114295. [DOI] [PubMed] [Google Scholar]
- 161.Salminen A. Aryl hydrocarbon receptor (AhR) reveals evidence of antagonistic pleiotropy in the regulation of the aging process. Cell Mol Life Sci. 2022;79:489. doi: 10.1007/s00018-022-04520-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Hu T, Allam M, Cai S, Henderson W, Yueh B, Garipcan A, Ievlev AV, Afkarian M, Beyaz S, Coskun AF. Single-cell spatial metabolomics with cell-type specific protein profiling for tissue systems biology. Nat Commun. 2023;14:8260. doi: 10.1038/s41467-023-43917-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Ali A, Davidson S, Fraenkel E, Gilmore I, Hankemeier T, Kirwan JA, Lane AN, Lanekoff I, Larion M, McCall LI, et al. Single cell metabolism: current and future trends. Metabolomics. 2022;18:77. doi: 10.1007/s11306-022-01934-3. [DOI] [PMC free article] [PubMed] [Google Scholar]




