Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2021 Jun 30;289(20):6099–6118. doi: 10.1111/febs.16086

Indoleamine 2,3‐dioxygenase 1 (IDO1): an up‐to‐date overview of an eclectic immunoregulatory enzyme

Maria Teresa Pallotta 1, Sofia Rossini 1, Chiara Suvieri 1, Alice Coletti 2, Ciriana Orabona 1, Antonio Macchiarulo 2, Claudia Volpi 1,, Ursula Grohmann 1,
PMCID: PMC9786828  PMID: 34145969

Abstract

Indoleamine 2,3‐dioxygenase 1 (IDO1) catalyzes the initial rate‐limiting step in the degradation of the essential amino acid tryptophan along the kynurenine pathway. When discovered more than 50 years ago, IDO1 was thought to be an effector molecule capable of mediating a survival strategy based on the deprivation of bacteria and tumor cells of the essential amino acid tryptophan. Since 1998, when tryptophan catabolism was discovered to be crucially involved in the maintenance of maternal T‐cell tolerance, IDO1 has become the focus of several laboratories around the world. Indeed, IDO1 is now considered as an authentic immune regulator not only in pregnancy, but also in autoimmune diseases, chronic inflammation, and tumor immunity. However, in the last years, a bulk of new information—including structural, biological, and functional evidence—on IDO1 has come to light. For instance, we now know that IDO1 has a peculiar conformational plasticity and, in addition to a complex and highly regulated catalytic activity, is capable of performing a nonenzymic function that reprograms the expression profile of immune cells toward a highly immunoregulatory phenotype. With this state‐of‐the‐art review, we aimed at gathering the most recent information obtained for this eclectic protein as well as at highlighting the major unresolved questions.

Keywords: dendritic cells; early endosomes; indoleamine 2,3‐dioxygenase 1; moonlighting protein; phosphatidylinositol 3‐kinase; Src kinase; tryptophan metabolism


IDO1 has a complex functional dynamics capable of fulfilling distinct environmental needs. Besides its catalytic activity degrading tryptophan, IDO1 performs a non‐enzymic function that, via activation of SHP1/SHP2 and noncanonical NF‐κB, reprograms the expression profile of dendritic cells (DCs) towards a long‐term immunoregulatory phenotype. The distinct functions occur either in cytosol (enzymic) or in early endosomes (EE; non‐enzymic). In inflammatory conditions, IDO1 is shut down by SOCS3‐mediated proteasomal degradation, and an immunostimulatory program is acquired by the DCs.

graphic file with name FEBS-289-6099-g005.jpg


Abbreviations

IDO1

indoleamine 2,3‐dioxygenase 1

IDO2

indoleamine 2,3‐dioxygenase 2

KP

kynurenine pathway

l‐Kyn

l‐kynurenine

l‐Trp

l‐tryptophan

PI3K

phosphoinositide 3‐kinase

TDO

tryptophan dioxygenase

Introduction

Indoleamine 2,3‐dioxygenase 1 (IDO1) is a cytosolic, monomeric, heme‐containing enzyme that catalyzes the initial rate‐limiting step in the degradation of the essential amino acid l‐tryptophan (l‐Trp) along a pathway known as the kynurenine pathway (KP) (Fig. 1). This pathway is a cascade of enzymatic steps that produces several biologically active molecules, such as l‐kynurenine (l‐Kyn), and can lead to the endogenous production of nicotinamide adenine dinucleotide (NAD+). IDO1 is normally an inducible enzyme and its most important inducer is the cytokine interferon‐γ (IFN‐γ).

Fig. 1.

Fig. 1

The KP in mammals. The first and rate‐limiting step in the KP is catalyzed by IDO1, IDO2, and TDO. N‐formylkynurenine is rapidly metabolized by AFMID into l‐kynurenine, which can be transformed by KATs, KYNU, and KMO into kynurenic acid, anthranilic acid, and 3‐hydroxykynurenine, respectively. 3‐hydroxykynurenine can be then transformed by KATs into xanthurenic acid or KYNU into 3‐hydroxyanthranilic acid, which can be converted by 3‐HAO into 2‐amino‐3‐carboxymuconic‐6‐semialdehyde that spontaneously transforms into quinolinic acid. NAD+, the final product of the pathway, is then obtained by several enzymatic reactions. IDO1, indoleamine 2,3‐dioxygenase 1; TDO, tryptophan dioxygenase; IDO2, indoleamine 2,3‐dioxygenase 2; AFMID, kynurenine formamidase; KATs, kynurenine aminotransferases; KYNU, kynureninase; KMO, kynurenine monooxygenase; 3‐HAO, 3‐hydroxyamino oxidase; NAD+, nicotinamide adenine dinucleotide; QPRT, quinolinate phosphoribosyltransferase.

IDO1 was discovered in the 60' by a Japanese group led by Osamu Hayaishi, along a series of studies on oxygenases in rabbit intestine [1]. However, the year that changed the perception of IDO1 by the scientific community is 1998, when David Munn and Andrew Mellor performed a pioneering experiment demonstrating that IDO1 is crucially involved in the maintenance of maternal T‐cell tolerance [2]. These data raised the interest on this enzyme to such an extent that the number of total publications on IDO1 has increased enormously since the early ‘90s.

Recent evidence indicates that the IDO1 biology is more complex than initially assumed. For instance, in human monocyte‐derived macrophages and tumor cells either as such or stimulated with IFN‐γ, the majority of IDO1 protein molecules do not bind heme, and therefore, a high percentage of IDO1 is in the apo form, whose biology and function are currently unknown [3, 4, 5, 6]. Moreover, IDO1 does not only reside in cytosol but can have a distinct intra‐ and extracellular topology, depending on the cell microenvironment [7]. Perhaps particularly intriguing is the fact that IDO1 is not just an enzyme and, in fact, after phosphorylation of critical tyrosine residues, can act as a mediator of a signaling pathway that profoundly changes the functional phenotype of specific immune cells [8]. Structural studies suggest that the distinct functions of IDO1, that is, catalytic versus signaling, may be associated with distinct protein conformations [9, 10].

Therefore, IDO1 should be considered a moonlighting protein [11, 12], that is, capable of mediating distinct functions in response to distinct cellular needs, and as such should be taken into account as drug target for a more effective immunotherapy.

Insights into IDO1 key structural elements

Several crystallographic structures of human IDO1 are available in different ligand‐bound and unbound forms [13]. Overall, they show a conserved fold of the enzymic primary sequence that is composed of a large domain containing the catalytic cleft and a small domain featuring two functional immunoreceptor tyrosine‐based inhibitory motifs (ITIMs; Y111, Y249) that are associated with the noncatalytic signaling function of the enzyme (Fig. 2) [8, 10, 14]. A further conserved post‐transcriptional regulatory site is located at a YENM motif (residues 145–148), which extends from the small domain toward the large domain. Upon phosphorylation, it triggers the interaction with class IA phosphoinositide 3‐kinases (PI3Ks) that, once activated, promote a localization shift of IDO1 from the cytosol to early endosomes (see below) [7].

Fig. 2.

Fig. 2

Crystal structure of substrate‐bound IDO1 (pdb code: 5WMU). Key structural elements are highlighted and labeled: ITIMs (Tyr111, Tyr249, orange CPK atoms); YENM motif (orange CPK atom sticks); narrow channel (cyan surface). Heme cofactor (green carbon atoms) and l‐Trp (orange carbon atom sticks).

Large and small domains are connected by a loop (DE‐loop; residues 260–268) above the sixth coordination site of the heme cofactor. A large flexible loop (JK‐loop; residues 360–382) controls the access to the catalytic site, adopting distinct closed, intermediate, and open conformations that are involved in mediating substrate recognition [9, 15, 16, 17, 18]. Another structural element of IDO1 is a narrow channel formed by α‐helices E and F that extends from the solvent‐exposed surface of the protein to the heme group [3]. This channel is relevant for the catalytic activity as it is thought to mediate the shuttling of the co‐substrate oxygen to the catalytic cleft for promoting the oxidative cleavage of the indole ring of l‐Trp [3]. At odds with its functionally related enzyme tryptophan dioxygenase (TDO), IDO1 shows a wide recognition of substrates beyond l‐Trp, including many indole‐bearing compounds such as serotonin, melatonin, and tryptamine [19], suggesting that IDO1 activity may translate into additional functions as compared to the l‐Trp‐specific enzyme TDO.

Recent crystallographic studies of human IDO1 in complex with the substrate cast a light on the binding modes of l‐Trp into the catalytic cleft and an accessory site, with the latter being associated with the inhibition by substrate phenomenon [3, 20]. Upon oxygen binding to the ferrous active form of IDO1 [21, 22, 23, 24], l‐Trp binds to the catalytic pocket on the sixth coordination site of the heme cofactor, yielding the catalytically active ternary complex (Fig. 3). According to this binding mode, the indole ring engages S167 with a water‐mediated hydrogen bond and establishes face‐to‐edge π‐stacking and hydrophobic interactions with F163 and Y126. The ammonium group of l‐Trp is hydrogen‐bonded to T379 and the 7‐propionate group of the heme, while the carboxylic moiety makes three hydrogen bonds with R231 and T379. This kind of interactions is frequently observed for primary amine and carboxylic moieties in the crystal structures of ligand‐bound protein complexes [25, 26].

Fig. 3.

Fig. 3

Binding mode of l‐Trp (orange carbon atom sticks) into the catalytic site of IDO1 (pdb code: 5WMU). Hydrogen bond interactions between l‐Trp and binding site residues are shown in black dashed lines. The EF loop is highlighted with a purple cartoon.

High concentration of l‐Trp inhibits the catalytic activity of IDO1, causing the inhibition by substrate phenomenon [19, 27]. This is ascribed to the interaction of l‐Trp to an accessory site that is located below the fifth coordination site of the heme cofactor as observed in the crystal structure of the F270G variant of IDO1 (Fig. 4) [19]. According to this binding mode, l‐Trp engages the side chains of V170, V269, L339, and L342 with extensive hydrophobic contacts, yielding a poor dissociation constant, that is, in the millimolar range of potency (Kd = 26 mm) [21]. Noteworthy, positive allosteric modulators (PAMs) of IDO1 such as indoleamine‐ethanol (IDE) and N‐acetylserotonin (NAS), as well as some uncompetitive inhibitors such as mitomycin C, have been proposed to bind into this accessory site of the enzyme [3, 28, 29].

Fig. 4.

Fig. 4

Binding mode of l‐Trp (orange carbon atom sticks) into the accessory site of the Phe270Gly variant of IDO1 (pdb code: 5WMW). l‐Trp engages a cluster of nonpolar residues (Val170, Val269, Leu342) in hydrophobic contacts, whereas no polar interactions are observed.

Beside the canonical catalytic cleft and the substrate accessory site, other cryptic binding pockets of l‐Trp have been conjectured in the structure of IDO1. However, their functional relevance is still elusive. In this regard, computational and biophysical studies have suggested the presence of a metastable binding pocket that is located on K238 and promotes a first interaction of l‐Trp to the enzyme along the substrate approaching pathway from the solvent to the catalytic cleft [9]. In the same study, another cryptic site was suggested using microscale thermophoresis to investigate binding interactions between l‐Trp and IDO1. Specifically, multiple binding events of the substrate to the enzyme were observed in both the heme‐bound and apo states of IDO1, yielding high and low dissociation constants. Although the high dissociation constants were in agreement with previous studies on the binding interaction of l‐Trp to the catalytic active site [13, 21, 29, 30], the low dissociation constants were clue for the existence of an additional site of l‐Trp in the structure of IDO1, which has still to be characterized.

Overall, these findings suggest a high conformational plasticity of IDO1, with at least two distinct conformations that may be stabilized by the interaction of the substrate and/or ligands to distinct pockets of the enzyme, including the catalytic site. The stabilization of each of these conformations may affect the ability of IDO1 to recruit protein partners, thus driving enzymic or nonenzymic functions.

Cellular mechanisms of enzymic and nonenzymic functions of IDO1

Over the years, the central role of IDO1 in orchestrating immune responses has acquired interesting aspects. l‐Trp starvation and generation of immunoactive kynurenines by IDO1+ dendritic cells (DCs) contribute to create an immunosuppressive microenvironment, characterized by impaired T effector cell functions and enhanced activity of regulatory T (Treg) cells [31, 32]. In particular, the degradation and thus starvation in microenvironments of l‐Trp (i.e., an essential amino acid) activates general control nonderepressible 2 (GCN2) [31], a kinase that shuts down the expression of several genes, including the pro‐inflammatory cytokine interleukin‐6 (IL‐6), via the inactivation of the eukaryotic translation initiation factor 2A (eIF2A). This effect leads to anergy in effector T cells and also blocks the conversion of Treg cells into pro‐inflammatory T helper type 17 (Th17) cells [33, 34]. Moreover, l‐Kyn, the main product of IDO1 catalytic activity, is an endogenous agonist of the aryl hydrocarbon receptor (AhR; Fig. 5) [35], a ligand‐activated transcription factor that, besides induction of xenobiotic metabolism genes in the liver, can promote the differentiation of effector T cells into Treg cells [36] and also upregulation of IDO1 in DCs [37], rendering these potent antigen‐presenting cells immunosuppressive. Very recently, blockade of the AhR was demonstrated to restrict an immunosuppressive axis mediated by Treg cells and tumor‐associated macrophages (TAM) [38].

Fig. 5.

Fig. 5

Intracellular dynamics of IDO1 in DCs. Depending on the microenvironment, IDO1 shapes its conformation and acquires functions suitable for cellular needs. (A) In acute inflammation, the pro‐inflammatory cytokine IFN‐γ induces IDO1 enzymic activity, promoting transformation of l‐Trp into l‐Kyn, an agonist of AhR, a ligand‐activated transcription factor that moves from cytosol to the nucleus and, via a positive feedback loop, upregulates Ido1 gene expression. (B) Pro‐inflammatory contexts driven by IL‐6 are characterized by the upregulation of SOCS3, which associates with phosphorylated ITIM2 via its SH2 domain, recruits the E3 ubiquitin ligase complex, and directs cytosolic IDO1 to proteasomal degradation. (C) In a microenvironment dominated by immunoregulatory TGF‐β, IDO1 is phosphorylated in ITIM (ITIM1 and ITIM2) and YENM domains, which bind and activate SHPs and PI3Ks, respectively. PI3K activation implies IDO1 direct binding to the p85 regulatory subunit that in turn promotes activation of p110, the PI3K catalytic subunit. PI3K binding favors IDO1 anchoring to EE and signaling function. SHP binding promotes IKKα‐dependent activation of the noncanonical pathway of NF‐κB that, via the p52/RelB heterodimer translocating to the nucleus, upregulates the expression of Ido1 and Tgfb1 genes. This mechanism establishes a positive immunoregulatory circuitry that ensures IDO1 long‐term expression in dendritic cells. IDO1, indoleamine 2,3‐dioxygenase 1; IFN‐γ, interferon γ; l‐Trp, l‐tryptophan; l‐Kyn, l‐kynurenine; AhR, aryl hydrocarbon receptor; IL‐6, interleukin‐6; SOCS3, suppressor of cytokine signaling 3; ITIM, immune tyrosine‐based inhibitory motif; TGF‐β, transforming growth factor β; PI3Ks, phosphoinositide 3‐kinases; EE, early endosome; SHPs, Src homology 2 domain phosphatases; IKKα, inhibitory‐κB kinase α.

Approximately 10 years ago, a novel function of IDO1 was discovered [8]. The nonenzymic function of IDO1 depends on the presence of two ITIMs, namely ITIM1 and ITIM2, located in the small, noncatalytic domain of the enzyme. The ITIM sequence, I/V/L/SxYxxL/V/F (where x indicates any amino acids), when tyrosine is phosphorylated, acts as a docking site for diverse molecular partners containing Src homology 2 (SH2) domains [39, 40], thus fulfilling diverse immunological needs. In fact, when DCs face the immunosuppressive cytokine transforming growth factor β (TGF‐β), Src homology 2 domain phosphatase‐1 (SHP1) and SHP2 are upregulated and preferentially associate with ITIM1 rather than ITIM2 (Fig. 5) [8, 14]. This event translates into SHPs’ interaction with the interleukin‐1 receptor‐associated kinase (IRAK), thus favoring the activation of noncanonical, anti‐inflammatory NF‐κB rather than canonical, pro‐inflammatory NF‐κB [8]. In turn, noncanonical NF‐κB, driven by activated inhibitory‐κB kinase α (IKKα) and in the form of p52‐RelB heterodimer, translocates to the nucleus and induces the expression of the Ido1 gene itself [8, 41, 42, 43]. Because the IDO1 induction is accompanied by upregulation of Tgfb1 [7, 8, 44, 45], this mechanism will ultimately establish a positive feedback loop that confers a long‐term immunoregulatory phenotype on both conventional and plasmacytoid mouse DCs (cDCs and pDCs, respectively) [8, 46].

Suppressor of cytokine signaling (SOCS) proteins are critical modulators of immune responses (11) that possess an SH2 domain, binding phosphotyrosine‐containing peptides, and a SOCS box. The latter domain participates in the formation of an E3 ubiquitin ligase complex that targets several signaling proteins for proteasomal degradation [47, 48]. In the presence of a microenvironment dominated by pro‐inflammatory interleukin‐6 (IL‐6), upregulated SOCS3 preferentially associates with phosphorylated IDO1 ITIM2, recruits the E3 ubiquitin ligase complex, and targets IDO1 to proteasomal degradation [14, 49]. Therefore, SOCS3 interaction with phosphorylated ITIM2 will reduce IDO1 half‐life, interrupt tolerogenic mechanisms, and favor a pro‐inflammatory phenotype in the DCs (Fig. 5).

A crucial point in these processes is the tyrosine kinase responsible for the phosphorylation of IDO1 ITIMs. A meta‐analysis of pDC gene expression public data sets indicated that the most widely represented is Fyn, a kinase belonging to the Src family, and that Fyn inhibition decreases IDO1 ITIM phosphorylation by TGF‐β [8]. In cDCs, the most important kinase involved in this mechanism is instead Src [41, 46, 50]. However, the possible existence of a preferential tyrosine phosphorylation of IDO1 ITIM1 versus ITIM2 by these kinases in distinct conditions has not been clarified yet.

Therefore, on the basis of these observations, IDO1 may be considered a moonlighting protein [12], that is, endowed with an additional function besides catalytic activity. Moonlighting proteins are very peculiar in that they will switch between functions by changing their conformational state, as may occur in response to altered environmental conditions [11]. Those include the redox state of the cell, temperature, but also post‐translational modifications (such as phosphorylation), changes in cellular localization, and interactions with other polypeptides. The additional function of IDO1 is compatible with that of a signal transducer leading to genomic effects and reprogramming DC functions. The interesting thing is that phosphorylable ITIMs can either prolong or reduce IDO1 expression, depending on specific cellular microenvironments and distinct partnerships [51], thus underlining the plasticity of IDO1 biology.

Modulation of IDO1 expression

In humans, IDO1 is constitutively expressed in a restricted set of cells, including placental and pulmonary endothelial cells, epithelial cells inside the female genital tract, and antigen‐presenting cells, such as mature DCs in secondary lymphoid organs [52, 53]. IDO1 is also highly expressed in β‐cells of pancreatic islet of healthy individuals but is absent in the residual β‐cells of patients with autoimmune diabetes [54]. Several human tumors constitutively express IDO1. In this regard, the IDO1 protein can be found in neoplastic cells themselves but also in fibroblasts and myeloid and endothelial cells present in the tumor bed [55, 56]. In some tumor cells, prostaglandin E2 (PGE2) and interleukin‐1β (IL‐1β) contribute to the maintenance of IDO1 basal expression [57]. Additionally, constitutive IDO1 expression in human cancer may be sustained by an autocrine signaling loop involving IL‐6 and AhR [58], suggesting that IL‐6 in tumors may behave differently from normal DCs in which the cytokine promotes IDO1 proteasomal degradation [49].

One of the major features of IDO1 is its inducibility [59]. IFN‐γ is considered the main inducer of IDO1 in several types of cells, including but not limited to immunocytes and fibroblasts. Indeed, the promoter region of the IDO1 gene (present in human chromosome 8p22) contains several IFN‐stimulated elements (ISREs) and gamma activation sequences (GAS) [60] that respond to transcription factors such as signal transducer and activator of transcription 1 (STAT1) and interferon‐regulatory factors (IRFs). IDO1 induction by IFN‐γ may represent a counteracting response in inflammatory conditions [61, 62, 63], a mechanism also exploited by tumors to favor their immune escape [63]. Indeed, some neoplastic cells downregulate the expression of Bin1 (encoding a Myc‐interacting protein with features of immunosuppressor), with consequent elevation of STAT1‐dependent expression of IDO1 [64]. Type I IFNs, that is, IFN‐α and IFN‐β, also represent IDO1 inducers, although, at variance with IFN‐γ (type II IFN), activate a pathway that leads to transcription factors that bind ISREs but not GAS and are thus less effective than IFN‐γ [65]. Involvement of type I IFNs is mainly observed in pDCs, which are the cells that produce the highest levels of these cytokines.

Over the years, several other signals have been identified as IDO1 inducers. These include the cytokine TGF‐β that, however, favors the signaling rather than catalytic function of IDO1 [8, 51]. In cDCs, activation of the Src kinase necessary for IDO1 ITIM phosphorylation can be directly promoted by spermidine, a polyamine produced downstream arginase 1 (ARG1; induced by TGF‐β earlier than IDO1) [46], a finding that linked for the first time two potent immunosuppressors such as IDO1 and ARG1 [66, 67] as well as polyamines and kynurenines [68]. The signaling function of IDO1 and consequent immunosuppressive effects can also be triggered by incubation of cDCs with ADX188, a positive allosteric modulator (PAM) of metabotropic glutamate receptor 4 (mGluR4), via potentiation of protein G‐independent mechanisms [41]. Other cytokines, that is, tumor necrosis factor α (TNF‐α), IL‐6, and IL‐10, can synergize with each other to increase IDO1 expression [69].

Pro‐inflammatory signals capable of upregulating IDO1 include pathogen‐associated molecular patterns (PAMPs) [70]. PAMPs are present in diverse organisms but absent in the host and provide exogenous signals that alert the immune system to the presence of pathogens, thereby promoting immunity. They are recognized by and activate Toll‐like receptors (TLRs), normally contributing to inflammatory processes but also immune tolerance [71]. Lipopolysaccharide (LPS) and oligonucleotides containing unmethylated CpG motifs (CpG‐DNA) are PAMPs that can modulate IDO1 activity via activation of TLR4 and TLR9, respectively. TLR4 is an intriguing receptor that, by virtue of its surface membrane or early endosome (EE) localization, can recruit either MyD88 or TRIF signaling adaptors, mediators of inflammatory or tolerogenic effects, respectively [72]. After repeated stimulation, LPS can in fact generate a state of immune suppression, known as endotoxin tolerance. Bessede et al. showed that IDO1 is highly expressed in cDCs repeatedly stimulated with low doses of LPS in vitro and that administration of LPS‐treated DCs or l‐Kyn alone can confer protection in recipient mice from endotoxic shock via activation of AhR and IDO1 upregulation in cDCs [50]. Human DCs conditioned with LPS exhibit upregulation of anti‐inflammatory IL‐10 that in turn activates the noncanonical NF‐κB pathway, with overexpression of RelB in an IDO1‐dependent manner [73]. Activation of TLR9 by CpG‐oligodeoxynucleotides induces the expression of IDO1 in splenic CD19+ DC, which acquire suppressive functions [74]. Moreover, in pDCs, the tolerogenic signaling via TLR9 that leads to IDO1 expression is dependent on the dosage of CpG‐ODN administration (i.e., low dose being immunostimulatory and high dose immunoregulatory) and requires the involvement of the TRIF adaptor [75].

An intriguing issue is the capacity of membrane‐anchored molecules to induce IDO1 in DCs via ‘reverse signaling’. This phenomenon occurs when a molecule normally operating as a receptor and expressed on the surface membrane of another cell activates a signaling pathway via engagement of a membrane molecular counterpart normally acting as a ligand [42]. By binding to CD80 and CD86 on DCs, cytotoxic T lymphocyte antigen 4 (CTLA‐4) engenders an IFN‐γ‐dependent induction of IDO1 [76]. Because Treg cells constitutively express CTLA‐4, this mechanism represents an important functional bridge between Treg cells and regulatory DCs in immune tolerance [77]. In addition to CTLA‐4, Treg cells possess the glucocorticoid‐inducible TNF receptor (GITR) [78]. Engagement of its ligand, GITRL, by a soluble form of GITR activates a reverse signaling in pDCs resulting in the activation of the noncanonical pathway of NF‐κB, increased IDO1 expression, and IDO1‐dependent immunosuppressive effects in vivo [79]. Because GITR and GITRL are under the control of glucocorticoids, these data unveiled an important mechanism of action of such potent compounds.

Therefore, IDO1 expression and functions appear to be under the control of a great variety of stimuli that can act either directly or indirectly and can sometimes behave very differently, depending on the circumstances.

Intra‐ and extracellular localization of IDO1 and role of PI3Ks

The bulk of literature has repeatedly indicated a cytosolic localization for the IDO1 enzyme in several types of cells [80, 81]. This localization may favor IDO1 intense and transient catalytic activity, possibly because of the easier access to substrate, that is, l‐Trp. However, until recently, no information was available regarding the intracellular topology of IDO1 as a signaling molecule. A critical discovery was that, in addition to ITIMs, IDO1 contains a consensus binding site for the p85 regulatory subunit of PI3K [7]. This motif, namely YENM, is characterized by a YxxM sequence (where ‘x’ indicates any amino acid). Alignment of the amino acid sequences reveals that the YENM sequence of IDO1 in mammals is completely conserved [7] but is not present in mammalian indoleamine 2,3‐dioxygenase 2 (IDO2), the IDO1 paralogue whose gene is considered an ancient version of IDO1 itself (see next paragraph) [82].

When the tyrosine residue within YENM becomes phosphorylated, IDO1 associates with p85 that in turn interacts with the p110 catalytic subunits of PI3Ks [7]. PI3Ks are a family of enzymes subdivided into three classes. Among them, class IA PI3Ks are heterodimeric proteins consisting of a regulatory subunit (p85α, p85β, or p55γ) and a catalytic subunit (p110α, p110β, or p110δ) [83]. Although p110α and p110β subunits are ubiquitously expressed, p110δ expression is generally restricted to cells of the immune system [83] and promotes anti‐inflammatory effects in DCs and macrophages [72]. As a consequence of IDO1 binding to p85, all class IA p110 catalytic subunits can be recruited, although the greater extent is observed for p110δ in cell transfectants expressing mouse IDO1 [7]. Class I PI3Ks possess important roles in several physiologic conditions as well as in disease [84, 85, 86]. By activating multiple signaling pathways and involving a plethora of downstream effectors, they can orchestrate both pro‐inflammatory and anti‐inflammatory mechanisms to maintain effective immunity while protecting host tissues. Interestingly, among their multiple functions, members of the PI3K family can also affect intracellular trafficking of vesicles and proteins [87].

Several activated receptors and signaling proteins accumulate in EE, which therefore can be defined as a platform hosting intracellular signaling [88]. Signals deriving from receptors and other proteins located in EE have different roles, such as control of growth, differentiation, survival, inflammation, and immunity. Efficient TGF‐β signaling—also responsible for IDO1 signaling activation—requires internalization of the TGF‐β receptor in EE [89, 90]. In a very recent study, treatment of pDCs with IFN‐γ or TGF‐β determined a dominant localization of the IDO1 protein in cytosol or EE, respectively [7]. Moreover, IDO1 anchoring to EE was mediated by binding of class I PI3Ks [7], an effect favoring IDO1 signaling rather catalytic activity in pDCs. These observations indicated, for the first time, an IDO1 localization distinct from cytosol and how the intracellular switch can occur. It is interesting to note that, in the absence of any stimulus, in both primary fresh cultures of pDCs and tumor transfectants highly expressing IDO1, IDO1 is mainly present in the cytosol but discrete quantity of the protein can be detected in EE as well, suggesting the a priori existence in IDO1+ cells of a pool of IDO1 proteins with distinct topology and possibly ready for distinct functions. Furthermore, although not demonstrated yet, IDO1 in EE may not contain heme. In fact, in unstimulated human ovarian cancer cells, IDO1 is dynamically bound to its heme cofactor and, perhaps most importantly, at least 85% of IDO1 exists in the apo form, that is, catalytically inactive, that can be nevertheless activated by exogenous heme added in the form of hemin [6]. Likewise, hemin increases IDO1 activity 2‐fold in IFN‐γ‐activated human monocyte‐derived macrophages [4], suggesting that cellular heme is limiting for IDO1 activity and that a significant portion of IDO1 in these cells is present as apoprotein. Along this line of evidence, transfected mouse IDO1 with tyrosine in ITIM1 substituted with glutamate (i.e., mimicking phosphorylated tyrosine) does not show any catalytic activity [14].

IDO1 is not confined into the cells but can be secreted in extracellular vesicles (EVs), including exosomes. In fact, stem cells from amniotic fluid treated with the main IDO1 inducer IFN‐γ produce immunoregulatory EVs containing IDO1 [91]. EVs from human semen express high levels of IDO1 transcripts, suggesting a mechanism whereby these vesicles induce tolerance [92]. Also, malignant glioblastoma cells generate immunosuppressive EVs containing IDO1, suggesting that tumors can exploit both intracellular and extracellular IDO1 as immune escape mechanism [93]. Exosomes derived from IDO1‐overexpressing rat bone marrow mesenchymal stem cells promote immunotolerance of cardiac allografts [94]. Although we do not know how and when IDO1 is transferred outside the cells via vesicles yet, these observations further confirm the complexity and dynamics of IDO1 in physiologic and pathologic conditions.

As a whole, IDO1 expression in cytosol, EE, and EVs—and, perhaps most importantly, the distinct IDO1 functions in cytosol versus EE—further confirm the hypothesis of IDO1 being a moonlighting protein [95]. In fact, the localization of a protein in a new cellular microenvironment could promote unpredicted molecular interactions by many potential new binding partners and contribute to generate new functions, as occurring for IDO1 [7]. Distinct activities mediated by STAT3 and Tgase2 are associated with different localization, including exosomes [96, 97].

In conclusion, the IDO1 catalytic and signaling functions are spatially segregated. Further studies are necessary to define the role of the IDO1 signaling function and its subcellular localization in pathological conditions, such as cancer, in order to optimize the development of new and more effective therapies [98].

IDO1, IDO2, and TDO: similar or distinct enzymes?

In addition to IDO1, there are two other mammalian enzymes capable to convert l‐Trp to l‐Kyn, that is, IDO2 and TDO. Since its discovery in 2007 [82], IDO2 represents the ‘enigmatic version’ of IDO1. In fact, the two IDOs are encoded by genes located in tandem on chromosome 8, in both humans and mice [99], and share a high level of sequence similarity at the amino acid level. Nevertheless, IDO2 catalytic activity is negligible in vitro [82]. Moreover, IDO2 knockout does not contribute to the reduction in l‐Kyn sera levels in vivo as instead shown by IDO1 depletion [100] and the pattern of the expression of the two molecules is consistently different, being IDO2 mainly expressed in the liver under physiologic conditions [101].

Interestingly, lower vertebrates generally possess only IDO2, and many fish species have both IDO1 and IDO2; as a possible explanation, molecular phylogenetic analyses showed that the gene duplication occurred before the divergence of vertebrates, with IDO1 having been lost in a number of lower vertebrate lineages [102]. Sequence analysis of the human IDO2 showed a peculiar and wide distribution of two single nucleotide polymorphisms (SNPs), which are rs10109853, leading to a > 90% reduction in IDO2 catalytic activity (R248W), and rs4503083, generating a premature stop codon (Y359X) [103]. However, the exact functional significance of the two common IDO2 variants is still far from being deciphered, as is the understanding of IDO2 physiological role. In fact, the putative structural analogies between IDO1 and IDO2, suggested by their high amino acid sequence homology, seem to not correspond to similar functions.

In vivo studies demonstrated a contrasting role for IDO2, with experiments in preclinical models of autoimmune arthritis suggesting a pro‐inflammatory role driving the disease [104]. In neoplastic diseases, IDO2 is much less expressed in tumor cells than IDO1 and its function is still matter of debate. In humans, pancreatic ductal adenocarcinoma (PDAC) [105] and non‐small‐cell lung cancer (NSCLC) [106] show particularly high levels of IDO2 expression. Nevertheless, the presence of a genotype compatible with an inactive form of IDO2 significantly associates with an improved disease‐free survival in patients with PDAC [107] but increases the risk of developing NSCLC [108].

Many studies have demonstrated that the knockout of the Ido2 gene can inhibit tumor growth in multiple animal models [109]. However, the use of IDO2 specific inhibitors could be a failure strategy, as it was in case of IDO1 [98]. In fact, the inhibition of IDO1 enzyme activity only may be not sufficient to exploit an effective therapeutic function, considering the complex functional dynamics of IDO1 and its additional signaling function. Regarding IDO2, we might hypothesize that it could possess a completely different function other than the poor enzymatic one, presumably relying on some still unidentified partnership with other proteins and consequent signaling activity. The presence of one ITIM motif (i.e., ITIM2) in both human and mouse IDO2 amino acid sequences pleads for a potential role of this molecule as a signaling molecule, although its function is still unknown.

Unlike IDO2, the physiologic role of TDO is better defined. TDO is mainly located in the liver and uses l‐Trp as substrate, thus showing a higher substrate specificity compared to IDO1 [110]. In the liver, TDO degrades excess dietary l‐Trp in order to maintain systemic stable levels of the amino acid [111]. Interestingly, seric l‐Kyn, the main product of both TDO and IDO1, is increased in TDO knockout mice but is totally absent in mice lacking IDO1, indicating that circulating l‐Kyn is mainly produced by IDO1. This is likely because the l‐Kyn produced by TDO is further degraded in the liver, which expresses all the enzymes of the l‐Kyn pathway, whereas inflammatory and immune cells that express IDO1 in the periphery may not express these enzymes [112].

TDO expression is also detected in the decidualized endometrium, where its function is unknown [113], and in the brain, contributing to the synthesis of neuroactive compounds [114]. TDO is thought to participate in the pathogenesis of several neurodegenerative diseases, such as Alzheimer, Parkinson, and Huntington disease (AD, PD, and HD, respectively), via regulation of proteotoxic events [115]. Significantly high TDO immunoreactivity has been observed in the AD patients' hippocampus [116]. Moreover, in fly models of AD and PD, TDO inhibition improves motor performance and helps survival, and increased kynurenic acid relative to 3‐hydroxykynurenine alleviates HD, indicating that shifting the KP to kynurenic acid rather than 3‐hydroxykynurenine synthesis can reduce neurodegeneration [117].

TDO2 mRNA can be also found in a number of human tumor samples, including hepatocarcinomas, glioblastomas, melanomas, and bladder carcinomas [118] and in many human tumor cell lines, including glioblastoma, colorectal, head and neck, lung, and gall bladder carcinoma cell lines, where its protein expression and activity have been confirmed [119]. Like IDO1, TDO plays an immunoregulatory role in tumors via l‐Kyn activation of AhR [35, 120]. TDO‐transfected mouse mastocytoma P815 cells resist immune rejection by mice immunized against P1A, the dominant antigen expressed in these tumors, and tumor rejection is restored upon treatment of mice with a TDO inhibitor [119], thus pinpointing TDO as a potential target for the development of selective anti‐tumoral drugs.

Physiologic versus pathologic effects of IDO1

Physiologic roles of IDO1

The IDO1 protein is a physiologic checkpoint that guarantees immune homeostasis in our organism by finely modulating a balanced immune response orchestrated by antigen‐presenting cells, such as DCs and macrophages. It plays a key role in supporting immune privilege [121], including the feto‐maternal interface in allogeneic pregnancy to protect the fetus from the maternal immune system [2] and further immune niches as cornea [122] and brain [123]. l‐Trp metabolites generated by IDO1 along the KP serve as signaling molecules for fine‐tuning the host immune response, mainly via AhR engagement of AhR [124, 125, 126]. The large evidence of a nonenzymic function of IDO1 that co‐operates with its catalytic activity renders more intricate the mechanisms of IDO1‐based immune tolerance [8, 51] that, nevertheless, opens the generation of innovative strategies targeting IDO1.

IDO1 catalytic activity also controls the de novo synthesis of NAD+, in addition to the salvage pathway that supplies the cell with the highest levels of NAD+ cofactor. The metabolic relevance of the kynurenine in NAD+ supplementation in both health and disease is not well defined, although the contribution of l‐Trp metabolism to NAD+ formation in the central nervous system (CNS) and in the tumor microenvironment (TME) is more evident than in other tissues [127, 128].

A growing body of evidence has demonstrated that IDO1 becomes a pathogenic factor overarching a wide range of human pathologies, including infections, neoplasia, autoimmunity, and neurodegeneration [55, 129, 130, 131], as outlined below.

IDO1 in infectious diseases

l‐Trp metabolism is an important regulator of immune host‐pathogen interactions. Several PAMPs can induce IDO1 expression by acting as TLR ligands [75, 132].

In infectious diseases, l‐Trp depletion caused by IDO1 activity has long been known to induce anti‐microbial actions for auxotroph pathogens [133, 134, 135]. Several in vitro studies reported that measles, influenza, cytomegalovirus, and herpes simplex viral infections are susceptible to l‐Trp levels [132, 135]. However, l‐Trp starvation can reprogram several auxotroph invaders to re‐acquire the capacity to synthesize this essential amino acid, as in the case of Mycobacterium tuberculosis [136]. At the same time, generation of downstream kynurenines by IDO1 acts as a double‐edge sword in infectious diseases, creating a state of immunosuppression that may impair clearance of the microorganism.

Combined antiviral therapy with IDO1 blockade significantly reduces the simian immunodeficiency virus (SIV) load in plasma and lymph nodes of treated rhesus macaques [137], indicating that, in this case, immunoregulatory IDO1 is at work in the disease and its inhibition favors recovery of infected animals. Recently, Gautam et al. [138] demonstrated in macaques affected by active tuberculosis that inhibition of IDO1 activity leads to a reduced M. tuberculosis burden, pathology, and improved animal survival. Currently, it is unclear yet whether elevated IDO1 is causative of the progression to active infection or a compensatory response to the microbe. However, these observations suggest that a potential for using IDO1 inhibitors as an effective anti‐microbial therapy may exist.

In addition to the control of pathogen load, IDO1 activation can restrain pathogenic immune activation that would ultimately worsen the infection. Indeed, in experimental fungal infections, IDO1 blockade greatly exacerbates the disease and the associated inflammatory pathology, as a result of dysregulated innate and adaptive immune responses to the fungi [139]. Interestingly, fungi express IDO1 and produce kynurenines that are involved in fungal but also host fitness [140].

IDO1 in cancer

In tumors, activation of l‐Trp catabolism by any of the l‐Trp‐degrading enzymes leads to a local generation of immunosuppressive kynurenines and l‐Trp depletion that determines a poor immune response. Based on this postulate, a fervid drug discovery activity in the development of anticancer small molecules targeting both IDO1 and TDO enzymes has characterized the last two decades [141, 142, 143]. A variety of catalytic inhibitors of IDO1 have been developed with favorable properties for a drug candidate (i.e., higher selectivity, potency, oral bioavailability, and safe profile) and with different mechanism of inhibition (i.e., competitive, noncompetitive, and heme‐displacing molecules) [69, 144]. An enormous attention has developed around targeting IDO1 in cancer immunotherapy, encouraged by promising results coming from preclinical studies that demonstrated, in a number of animal models, the antitumor effect of IDO1 blockade in monotherapy or in combination with other immune checkpoint inhibitors [145, 146].

Based on the correlation of IDO1 expression with worse prognosis in a wide range of human tumors [147, 148], the most promising compound, that is, epacadostat, reached clinical trial phase 3. Disappointingly, the largest phase 3 clinical trial (ECHO‐301) in advanced melanoma with epacadostat and pembrolizumab (an anti‐PD1 antibody) failed. The trial was suspended because the therapeutic strategy did not show any increased benefit compared with placebo plus pembrolizumab in melanoma patients [149]. To date, none of the IDO1 inhibitors has been approved for anticancer therapy yet.

Despite the failed experience with epacadostat, a strong translational rationale still exists for targeting IDO1 and, more widely, l‐Trp metabolism in cancer immunotherapy. Since it became evident that standalone IDO1 blockade is not effective in tumor disease progression, numerous novel molecules with dual inhibitory activity have been developed. Dual TDO/IDO1 inhibitors have been designed for tumors where IDO1 inhibition can play the by‐pass to l‐Kyn generation by TDO. TDO might provide compensatory mechanisms to l‐Trp depletion and production of l‐Kyn metabolites. Therefore, dual or even pan‐inhibition of l‐Trp‐degrading enzymes (i.e., also targeting IDO2) might be beneficial and complementary for efficacy improvement in immunotherapy [119].

Novel l‐Trp–l‐Kyn–AhR axis inhibitors, as l‐Kyn‐degrading enzymes, direct AhR antagonists, and l‐Trp mimetics are also advancing in preclinical development [150]. Although multitargeting of l‐Trp metabolism in cancer appears a rational strategy, potential side effects should of course be considered. Lastly, the nonenzymatic function of IDO1 that promotes in DCs a long‐term immunosuppressive phenotype [8, 46] could contribute to restrain the antitumor immune response in TME. Conformations that mediate both enzymatic and nonenzymatic (i.e., signaling) activities of IDO1 are in a dynamic balance in the cell [14]. Computational studies performed by Mammoli et al. on the crystal structures of IDO1, with or without bound inhibitors, suggest that diverse inhibitors can induce different conformational changes in the small domain of IDO1 and thus affect not only its catalytic but also the signaling function [10]. In patients with noncurable glioblastoma, the advanced age was associated with increased IDO1 expression, decreased immunotherapeutic efficacy, and was not reversed by IDO1 enzyme inhibition [146], proposing that targeting the signaling activity of IDO1 in combination with its catalytic function may improve IDO1‐targeting immunotherapy. Indeed, the nonenzymatic function could provide either an interpretation of the IDO1 inhibitors' failure in the trials or suggest new strategies for targeting IDO1 more effectively.

IDO1 in autoimmunity

The pleiotropic mechanisms of IDO1 are deeply involved in the pathogenesis of autoimmune conditions featured by an aberrant immune response against self‐antigens. Contrary to what happens in cancer, a poor activity of IDO1 facilitates the over‐activation of immune effectors in autoimmunity [151]. A critical role is played by IDO1+ DCs that, in physiological conditions, may control immune responses by inhibiting effector T cells and promoting Treg lymphocytes in an antigen‐specific manner [42, 61, 62].

In type 1 diabetes (T1D), a defective IDO1 activity has been observed in DCs from nonobese diabetic (NOD) mice [44, 152, 153], in a majority of peripheral blood mononuclear cells (PBMCs) isolated from pediatric diabetic patients [154], and in β‐cells of human pancreas from adult diabetic donors [54]. In both DCs from NOD mice and PBMCs from diabetic children, poor IDO1 activity is caused by a rapid turnover of the protein triggered by an IL‐6‐driven pro‐inflammatory milieu. Indeed, the IDO1 defect can be corrected by blocking the IL‐6 receptor (IL‐6R) by means of tocilizumab (TCZ). The drug inhibits IDO1 degradation and restores normoglycemia in diabetic NOD mice via an IDO1‐dependent mechanism [155]. Similarly, TCZ can restore l‐Trp metabolism in PBMCs from a subgroup of T1D children characterized by a deregulated IL‐6R‐SOCS3 axis and a distinct frequency of a single nucleotide polymorphism (SNP), that is, IDO1 rs7820268. These observations are consistent with the data reported by Antequil et al. that describe a gradual decay of IDO1 in pancreatic β‐cells during T1D pathogenesis, ranging from autoantibody‐positive donors to recent‐onset T1D patients, and a complete absence of IDO1 in insulin‐deficient islets [54].

Recently, Mondanelli et al. [28] proposed an innovative therapeutic indication for NAS, a l‐Trp‐derived metabolite produced along the serotonin pathway, in experimental autoimmune encephalomyelitis (EAE; an animal model of human multiple sclerosis, MS). For the first time, NAS has been described as a PAM of the IDO1 enzyme, capable of implementing the production of l‐Kyn that in turn activates AhR. As a consequence, NAS reinforces the immunoregulatory IDO1‐l‐Kyn‐AhR axis in DCs and restrains the pathogenic neuroinflammation that characterizes MS. NAS protects mice from EAE in an IDO1‐ and AhR‐dependent manner. In PBMCs isolated from patients with relapsing‐remitting MS [28, 156], NAS increases l‐Kyn release in IFN‐γ‐stimulated conditions without modulating IDO1 expression.

The analysis of IDO1 SNPs in both autoimmune diabetes and MS cohorts displayed a different distribution of the rs7820268 SNP among autoimmune patients and healthy subjects. Although several SNPs in IDO1 have been associated with the occurrence of autoimmune/chronic inflammatory diseases, including T1D, Crohn's disease, systemic sclerosis, and MS [28, 151, 157, 158], the majority of them affects intronic regions or the IDO1 promoter and lacks a cause–effect relationship. A bottleneck in developing IDO1 as a predictive biomarker and promising therapeutic target in both T1D and MS is in fact the lack of a genotype‐to‐protein association. To date, a unique rs751360195 single variant in the IDO1 coding sequence has been identified in a 30‐year‐old woman affected by coeliac disorder, selective IgA deficiency, thyroiditis, and thrombocytopenia [159]. Interestingly, the single nucleotide variant (SNV) rs751360195 encodes a missense mutation (K257E) very closed to the ITIM2 motif that shortens IDO1 half‐life in PBMCs. The recent identification of a specific IDO1 genotype encoding for a short‐lived protein IDO1 in a patient affected by a broader dysimmune disorder extends the investigative field of defective IDO1 in further autoimmune conditions and reinforce the perspective to develop IDO1 as a predictive biomarker that will contribute to precision medicine in the autoimmunity field.

IDO1 in neurodegeneration

KP metabolites have been closely linked to the pathogenesis of several neurodegenerative diseases, including AD, PD, and HD [131, 160]. In AD, the amyloid peptide upregulates IDO1 expression and increases the production of quinolinic acid in human macrophages and microglia [161]. Furthermore, human neurons treated with quinolinic acid upregulate genes involved in tau phosphorylation, the mechanism crucially involved in the formation of pathogenetic neurofibrillary tangles [162]. In HD, high levels of l‐Kyn lead to high production of neurotoxic quinolinic acid and very low levels of neuroprotective kynurenic acid [163], thus suggesting the existence of an imbalance between distinct l‐Trp metabolites that may play a causative role in HD pathogenesis. Such imbalance has also been observed in PD patients in dopaminergic neurons and surrounding microglia [164].

As compared to other pathological conditions, targeting IDO1 and other l‐Trp‐metabolizing enzymes in neurodegenerative diseases is still in its infancy. Nevertheless, it appears that efforts in this direction might very likely be worth making.

Concluding remarks

By comparing the effects of carbon monoxide on the conformations of human IDO1 versus human TDO, Syun‐Ru Yeh and colleagues very recently found that IDO1 exhibits remarkable conformational plasticity as compared to the more rigid TDO [165]. The authors hypothesized that this structural fingerprint may be relevant for IDO1 being a multitasking protein under a harsh inflammatory environment [165]. Lacking completely in TDO, the small domain and interconnecting loop may thus represent the main structural parts of IDO1 modulating its conformational as well as functional plasticity. As a matter of fact, all identified motifs allowing IDO1 to establish a partnership with other molecules, that is, SHPs, PI3Ks, and SOCS3, have been localized in its small domain. Therefore, a peculiar overall architecture could imprint IDO1 with the flexible capacity to promote Trp metabolism, its expression, or its proteasomal degradation, meeting specific cellular needs in specific microenvironmental conditions. In this regard, it should be mentioned that several multitasking proteins undergo structural transformation when performing distinct functions [166, 167]. An additional level of complexity derives from the IDO1 ability to catalyze other catalytic reactions in addition to its dioxygenase activity, including peroxygenase [168], peroxidase [169], and nitrite reductase [170] activities as well as the generation of singlet oxygen [171]. Of note, the production of singlet oxygen by IDO1 leads to the formation of a tricyclic hydroperoxide that decreases blood pressure [171]. Therefore, the versatility of IDO1 likely extends to the catalysis of several biologically relevant enzyme reactions.

Although we do not know where binding of SOCS3 and ubiquitination may occur yet (but we may hypothesized the cytosol, given the cytosolic localization of the proteasome complex), IDO1 enzymic and nonenzymic activities take place in the cytosol and EE, respectively, at least in DCs and IDO1‐transfected P815 mouse mastocytoma cells [7]. An important question would be how the apo form of IDO1 influences responsiveness to specific cellular microenvironments (i.e., cytokines, TLR ligands, Trp, and O2 concentration, pH), and thus, the choice of a specific function or fate. Vice versa, specific microenvironments may change the balance between holo‐ and apo‐IDO1. Moreover, which is the intra‐ (and possibly extra‐) cellular topology of apo‐IDO1? Presumably, because tumor transfectants expressing a mouse IDO1 mutant mimicking tyrosine‐phosphorylated ITIM1 perform IDO1 signaling activity but do not bind heme and thus do not produce l‐Kyn [14], the apoprotein may be associated with EE. Therefore, a systematic study of the percentages of apo‐IDO1, dynamics of cellular localization, and performance of a specific function, either in basal conditions or in response to specific stimuli, should be undertaken in both primary cells (including DCs but also macrophages, fibroblasts, and epithelial cells known to express IDO1) and tumors, of mouse and human origin.

The delineation of the biologic and functional profile of IDO1 in tumor contexts may lead to the generation of novel drugs targeting this important protein in neoplastic patients and, perhaps, most importantly, could be more effective than the currently available catalytic inhibitors. In this regard, it would be very interesting to know whether the catalytic inhibitors targeting apo‐IDO1 [6]—and preventing heme binding—do also influence the IDO1 signaling activity, which may be more important in a chronic disease like neoplasia. In chronic inflammation and autoimmunity, drug targeting of IDO1 (and enzymes in general) appears to be a more difficult task. In fact, in these conditions, IDO1 function/s should be potentiated rather than inhibited. However, the very recent discovery of VIS351 [172], an inducer of IDO1 signaling, and NAS [28], an enhancer of IDO1 catalytic activity protecting from neuroinflammation, may bring some fresh air in IDO1‐based immunotherapies.

Therefore, the great amount of work that has been performed on IDO1 in the most recent years raises new biologic questions but also opens new exciting perspectives for therapeutic advances in several disease areas.

Conflict of interest

The authors declare no conflict of interest.

Author contributions

All authors contributed to the article and approved the submitted version.

Acknowledgements

This work was supported by Associazione Italiana per la Ricerca sul Cancro (AIRC 2019‐23084 to U.G.), the Italian Ministry of Education, University, and Research (PRIN 20173EAZ2Z to C.V.; PRIN 2017WJZ9W9 to M.T.P.; and PRIN 2017BA9LM5 to C.O.), and the European Research Council (899838‐ENHANCIDO to U.G. and A.M.). Open Access Funding provided by Universita degli Studi di Perugia within the CRUI‐CARE Agreement. [Correction added on 20 May 2022, after first online publication: CRUI funding statement has been added.]

Claudia Volpi and Ursula Grohmann share senior authorship

Contributor Information

Claudia Volpi, Email: claudia.volpi@unipg.it.

Ursula Grohmann, Email: ursula.grohmann@unipg.it.

References

  • 1. Yamamoto S & Hayaishi O (1967) Tryptophan pyrrolase of rabbit intestine. D‐ and L‐tryptophan‐cleaving enzyme or enzymes. J Biol Chem 242, 5260–5266. [PubMed] [Google Scholar]
  • 2. Munn DH, Zhou M, Attwood JT, Bondarev I, Conway SJ, Marshall B, Brown C & Mellor AL (1998) Prevention of allogeneic fetal rejection by tryptophan catabolism. Science 281, 1191–1193. [DOI] [PubMed] [Google Scholar]
  • 3. Lewis‐Ballester A, Pham KN, Batabyal D, Karkashon S, Bonanno JB, Poulos TL & Yeh SR (2017) Structural insights into substrate and inhibitor binding sites in human indoleamine 2,3‐dioxygenase 1. Nat Commun 8, 1693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Thomas SR, Salahifar H, Mashima R, Hunt NH, Richardson DR & Stocker R (2001) Antioxidants inhibit indoleamine 2,3‐dioxygenase in IFN‐gamma‐activated human macrophages: posttranslational regulation by pyrrolidine dithiocarbamate. J Immunol 166, 6332–6340. [DOI] [PubMed] [Google Scholar]
  • 5. Behmoaras J (2020) The versatile biochemistry of iron in macrophage effector functions. FEBS J. 10.1111/febs.15682 [DOI] [PubMed] [Google Scholar]
  • 6. Nelp MT, Kates PA, Hunt JT, Newitt JA, Balog A, Maley D, Zhu X, Abell L, Allentoff A, Borzilleri R et al. (2018) Immune‐modulating enzyme indoleamine 2,3‐dioxygenase is effectively inhibited by targeting its apo‐form. Proc Natl Acad Sci USA 115, 3249–3254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Iacono A, Pompa A, De Marchis F, Panfili E, Greco FA, Coletti A, Orabona C, Volpi C, Belladonna ML, Mondanelli G et al. (2020) Class IA PI3Ks regulate subcellular and functional dynamics of IDO1. EMBO Rep 21, e49756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Pallotta MT, Orabona C, Volpi C, Vacca C, Belladonna ML, Bianchi R, Servillo G, Brunacci C, Calvitti M, Bicciato S et al. (2011) Indoleamine 2,3‐dioxygenase is a signaling protein in long‐term tolerance by dendritic cells. Nat Immunol 12, 870–878. [DOI] [PubMed] [Google Scholar]
  • 9. Greco FA, Albini E, Coletti A, Dolciami D, Carotti A, Orabona C, Grohmann U & Macchiarulo A (2019) Tracking hidden binding pockets along the molecular recognition path of l‐Trp to Indoleamine 2,3‐Dioxygenase 1. ChemMedChem 14, 2084–2092. [DOI] [PubMed] [Google Scholar]
  • 10. Mammoli A, Coletti A, Ballarotto M, Riccio A, Carotti A, Grohmann U, Camaioni E & Macchiarulo A (2020) New insights from crystallographic data: diversity of structural motifs and molecular recognition properties between groups of IDO1 structures. ChemMedChem 15, 891–899. [DOI] [PubMed] [Google Scholar]
  • 11. Jeffery CJ (2003) Moonlighting proteins: old proteins learning new tricks. Trends Genet 19, 415–417. [DOI] [PubMed] [Google Scholar]
  • 12. Jeffery CJ (2020) Enzymes, pseudoenzymes, and moonlighting proteins: diversity of function in protein superfamilies. FEBS J 287, 4141–4149. [DOI] [PubMed] [Google Scholar]
  • 13. Sugimoto H, Oda S, Otsuki T, Hino T, Yoshida T & Shiro Y (2006) Crystal structure of human indoleamine 2,3‐dioxygenase: catalytic mechanism of O2 incorporation by a heme‐containing dioxygenase. Proc Natl Acad Sci USA 103, 2611–2616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Albini E, Rosini V, Gargaro M, Mondanelli G, Belladonna ML, Pallotta MT, Volpi C, Fallarino F, Macchiarulo A, Antognelli C et al. (2017) Distinct roles of immunoreceptor tyrosine‐based motifs in immunosuppressive indoleamine 2,3‐dioxygenase 1. J Cell Mol Med 21, 165–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Macchiarulo A, Nuti R, Bellocchi D, Camaioni E & Pellicciari R (2007) Molecular docking and spatial coarse graining simulations as tools to investigate substrate recognition, enhancer binding and conformational transitions in indoleamine‐2,3‐dioxygenase (IDO). Biochem Biophys Acta 1774, 1058–1068. [DOI] [PubMed] [Google Scholar]
  • 16. Greco FA, Bournique A, Coletti A, Custodi C, Dolciami D, Carotti A & Macchiarulo A (2016) Docking studies and molecular dynamic simulations reveal different features of IDO1 structure. Mol Informat 35, 449–459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Alvarez L, Lewis‐Ballester A, Roitberg A, Estrin DA, Yeh SR, Marti MA & Capece L (2016) Structural study of a flexible active site loop in human indoleamine 2,3‐dioxygenase and its functional implications. Biochemistry 55, 2785–2793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Mirgaux M, Leherte L & Wouters J (2020) Influence of the presence of the heme cofactor on the JK‐loop structure in indoleamine 2,3‐dioxygenase 1. Acta Crystallogr D 76, 1211–1221. [DOI] [PubMed] [Google Scholar]
  • 19. Sono M, Roach MP, Coulter ED & Dawson JH (1996) Heme‐Containing Oxygenases. Chem Rev 96, 2841–2888. [DOI] [PubMed] [Google Scholar]
  • 20. Luo S, Xu K, Xiang S, Chen J, Chen C, Guo C, Tong Y & Tong L (2018) High‐resolution structures of inhibitor complexes of human indoleamine 2,3‐dioxygenase 1 in a new crystal form. Acta Crystallogr F 74, 717–724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Lu C, Lin Y & Yeh SR (2010) Spectroscopic studies of ligand and substrate binding to human indoleamine 2,3‐dioxygenase. Biochemistry 49, 5028–5034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Efimov I, Basran J, Sun X, Chauhan N, Chapman SK, Mowat CG & Raven EL (2012) The mechanism of substrate inhibition in human indoleamine 2,3‐dioxygenase. J Am Chem Soc 134, 3034–3041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Kolawole AO, Hixon BP, Dameron LS, Chrisman IM & Smirnov VV (2015) Catalytic activity of human indoleamine 2,3‐dioxygenase (hIDO1) at low oxygen. Arch Biochem Biophys 570, 47–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Lewis‐Ballester A, Karkashon S, Batabyal D, Poulos TL & Yeh SR (2018) Inhibition Mechanisms of Human Indoleamine 2,3 Dioxygenase 1. J Am Chem Soc 140, 8518–8525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Macchiarulo A, Nuti R, Eren G & Pellicciari R (2009) Charting the chemical space of target sites: insights into the binding modes of amine and amidine groups. J Chem Inf Model 49, 900–912. [DOI] [PubMed] [Google Scholar]
  • 26. Macchiarulo A & Pellicciari R (2007) Exploring the other side of biologically relevant chemical space: insights into carboxylic, sulfonic and phosphonic acid bioisosteric relationships. J Mol Graph Model 26, 728–739. [DOI] [PubMed] [Google Scholar]
  • 27. Sono M, Taniguchi T, Watanabe Y & Hayaishi O (1980) Indoleamine 2,3‐dioxygenase. Equilibrium studies of the tryptophan binding to the ferric, ferrous, and CO‐bound enzymes. J Biol Chem 255, 1339–1345. [PubMed] [Google Scholar]
  • 28. Mondanelli G, Coletti A, Greco FA, Pallotta MT, Orabona C, Iacono A, Belladonna ML, Albini E, Panfili E, Fallarino F et al. (2020) Positive allosteric modulation of indoleamine 2,3‐dioxygenase 1 restrains neuroinflammation. Proc Natl Acad Sci USA 117, 3848–3857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Lu C, Lin Y & Yeh SR (2009) Inhibitory substrate binding site of human indoleamine 2,3‐dioxygenase. J Am Chem Soc 131, 12866–12867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Papadopoulou ND, Mewies M, McLean KJ, Seward HE, Svistunenko DA, Munro AW & Raven EL (2005) Redox and spectroscopic properties of human indoleamine 2,3‐dioxygenase and a His303Ala variant: implications for catalysis. Biochemistry 44, 14318–14328. [DOI] [PubMed] [Google Scholar]
  • 31. Munn DH, Sharma MD, Baban B, Harding HP, Zhang Y, Ron D & Mellor AL (2005) GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3‐dioxygenase. Immunity 22, 633–642. [DOI] [PubMed] [Google Scholar]
  • 32. Fallarino F, Grohmann U, You S, McGrath BC, Cavener DR, Vacca C, Orabona C, Bianchi R, Belladonna ML, Volpi C et al. (2006) The combined effects of tryptophan starvation and tryptophan catabolites down‐regulate T cell receptor zeta‐chain and induce a regulatory phenotype in naive T cells. J Immunol 176, 6752–6761. [DOI] [PubMed] [Google Scholar]
  • 33. Baban B, Chandler PR, Sharma MD, Pihkala J, Koni PA, Munn DH & Mellor AL (2009) IDO activates regulatory T cells and blocks their conversion into Th17‐like T cells. J Immunol 183, 2475–2483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Sharma MD, Hou DY, Liu Y, Koni PA, Metz R, Chandler P, Mellor AL, He Y & Munn DH (2009) Indoleamine 2,3‐dioxygenase controls conversion of Foxp3+ Tregs to TH17‐like cells in tumor‐draining lymph nodes. Blood 113, 6102–6111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Opitz CA, Litzenburger UM, Sahm F, Ott M, Tritschler I, Trump S, Schumacher T, Jestaedt L, Schrenk D, Weller M et al. (2011) An endogenous tumour‐promoting ligand of the human aryl hydrocarbon receptor. Nature 478, 197–203. [DOI] [PubMed] [Google Scholar]
  • 36. Mezrich JD, Fechner JH, Zhang X, Johnson BP, Burlingham WJ & Bradfield CA (2010) An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J Immunol 185, 3190–3198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Nguyen NT, Kimura A, Nakahama T, Chinen I, Masuda K, Nohara K, Fujii‐Kuriyama Y & Kishimoto T (2010) Aryl hydrocarbon receptor negatively regulates dendritic cell immunogenicity via a kynurenine‐dependent mechanism. Proc Natl Acad Sci USA 107, 19961–19966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Campesato LF, Budhu S, Tchaicha J, Weng CH, Gigoux M, Cohen IJ, Redmond D, Mangarin L, Pourpe S, Liu C et al. (2020) Blockade of the AHR restricts a Treg‐macrophage suppressive axis induced by L‐Kynurenine. Nat Commun 11, 4011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Paul SP, Taylor LS, Stansbury EK & McVicar DW (2000) Myeloid specific human CD33 is an inhibitory receptor with differential ITIM function in recruiting the phosphatases SHP‐1 and SHP‐2. Blood 96, 483–490. [PubMed] [Google Scholar]
  • 40. Unkeless JC & Jin J (1997) Inhibitory receptors, ITIM sequences and phosphatases. Curr Opin Immunol 9, 338–343. [DOI] [PubMed] [Google Scholar]
  • 41. Volpi C, Mondanelli G, Pallotta MT, Vacca C, Iacono A, Gargaro M, Albini E, Bianchi R, Belladonna ML, Celanire S et al. (2016) Allosteric modulation of metabotropic glutamate receptor 4 activates IDO1‐dependent, immunoregulatory signaling in dendritic cells. Neuropharmacology 102, 59–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Puccetti P & Grohmann U (2007) IDO and regulatory T cells: a role for reverse signalling and non‐canonical NF‐kappaB activation. Nat Rev Immunol 7, 817–823. [DOI] [PubMed] [Google Scholar]
  • 43. Tas SW, Vervoordeldonk MJ, Hajji N, Schuitemaker JH, van der Sluijs KF , May MJ, Ghosh S, Kapsenberg ML, Tak PP & de Jong EC (2007) Noncanonical NF‐kappaB signaling in dendritic cells is required for indoleamine 2,3‐dioxygenase (IDO) induction and immune regulation. Blood 110, 1540–1549. [DOI] [PubMed] [Google Scholar]
  • 44. Pallotta MT, Orabona C, Bianchi R, Vacca C, Fallarino F, Belladonna ML, Volpi C, Mondanelli G, Gargaro M, Allegrucci M et al. (2014) Forced IDO1 expression in dendritic cells restores immunoregulatory signalling in autoimmune diabetes. J Cell Mol Med 18, 2082–2091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Chen W (2011) IDO: more than an enzyme. Nat Immunol 12, 809–811. [DOI] [PubMed] [Google Scholar]
  • 46. Mondanelli G, Bianchi R, Pallotta MT, Orabona C, Albini E, Iacono A, Belladonna ML, Vacca C, Fallarino F, Macchiarulo A et al. (2017) A relay pathway between arginine and tryptophan metabolism confers immunosuppressive properties on dendritic cells. Immunity 46, 233–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Wong PK, Egan PJ, Croker BA, O'Donnell K, Sims NA, Drake S, Kiu H, McManus EJ, Alexander WS, Roberts AW et al. (2006) SOCS‐3 negatively regulates innate and adaptive immune mechanisms in acute IL‐1‐dependent inflammatory arthritis. J Clin Invest 116, 1571–1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Orr SJ, Morgan NM, Elliott J, Burrows JF, Scott CJ, McVicar DW & Johnston JA (2007) CD33 responses are blocked by SOCS3 through accelerated proteasomal‐mediated turnover. Blood 109, 1061–1068. [DOI] [PubMed] [Google Scholar]
  • 49. Orabona C, Pallotta MT, Volpi C, Fallarino F, Vacca C, Bianchi R, Belladonna ML, Fioretti MC, Grohmann U & Puccetti P (2008) SOCS3 drives proteasomal degradation of indoleamine 2,3‐dioxygenase (IDO) and antagonizes IDO‐dependent tolerogenesis. Proc Natl Acad Sci USA 105, 20828–20833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Bessede A, Gargaro M, Pallotta MT, Matino D, Servillo G, Brunacci C, Bicciato S, Mazza EM, Macchiarulo A, Vacca C et al. (2014) Aryl hydrocarbon receptor control of a disease tolerance defence pathway. Nature 511, 184–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Orabona C, Pallotta MT & Grohmann U (2012) Different partners, opposite outcomes: a new perspective of the immunobiology of indoleamine 2,3‐dioxygenase. Mol Med 18, 834–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Theate I, van Baren N , Pilotte L, Moulin P, Larrieu P, Renauld JC, Herve C, Gutierrez‐Roelens I, Marbaix E, Sempoux C et al. (2015) Extensive profiling of the expression of the indoleamine 2,3‐dioxygenase 1 protein in normal and tumoral human tissues. Cancer Immunol Res 3, 161–172. [DOI] [PubMed] [Google Scholar]
  • 53. Zhai L, Bell A, Ladomersky E, Lauing KL, Bollu L, Sosman JA, Zhang B, Wu JD, Miller SD, Meeks JJ et al. (2020) Immunosuppressive IDO in cancer: mechanisms of action, animal models, and targeting strategies. Front Immunol 11, 1185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Anquetil F, Mondanelli G, Gonzalez N, Rodriguez Calvo T, Zapardiel Gonzalo J, Krogvold L, Dahl‐Jorgensen K, Van den Eynde B, Orabona C, Grohmann U et al. (2018) Loss of IDO1 expression from human pancreatic beta‐cells precedes their destruction during the development of type 1 diabetes. Diabetes 67, 1858–1866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. van Baren N & Van den Eynde BJ (2015) Tryptophan‐degrading enzymes in tumoral immune resistance. Front Immunol 6, 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Meireson A, Devos M & Brochez L (2020) IDO expression in cancer: different compartment, different functionality? Front Immunol 11, 531491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Hennequart M, Pilotte L, Cane S, Hoffmann D, Stroobant V, Plaen E & Van den Eynde BJ (2017) Constitutive IDO1 expression in human tumors is driven by cyclooxygenase‐2 and mediates intrinsic immune resistance. Cancer Immunol Res 5, 695–709. [DOI] [PubMed] [Google Scholar]
  • 58. Litzenburger UM, Opitz CA, Sahm F, Rauschenbach KJ, Trump S, Winter M, Ott M, Ochs K, Lutz C, Liu X et al. (2014) Constitutive IDO expression in human cancer is sustained by an autocrine signaling loop involving IL‐6, STAT3 and the AHR. Oncotarget 5, 1038–1051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Taylor MW & Feng GS (1991) Relationship between interferon‐gamma, indoleamine 2,3‐dioxygenase, and tryptophan catabolism. FASEB J 5, 2516–2522. [PubMed] [Google Scholar]
  • 60. Konan KV & Taylor MW (1996) Importance of the two interferon‐stimulated response element (ISRE) sequences in the regulation of the human indoleamine 2,3‐dioxygenase gene. J Biol Chem 271, 19140–19145. [DOI] [PubMed] [Google Scholar]
  • 61. Grohmann U, Fallarino F & Puccetti P (2003) Tolerance, DCs and tryptophan: much ado about IDO. Trends Immunol 24, 242–248. [DOI] [PubMed] [Google Scholar]
  • 62. Mellor AL & Munn DH (2004) IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nat Rev Immunol 4, 762–774. [DOI] [PubMed] [Google Scholar]
  • 63. Munn DH & Mellor AL (2016) IDO in the tumor microenvironment: inflammation, counter‐regulation, and tolerance. Trends Immunol 37, 193–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Muller AJ, DuHadaway JB, Donover PS, Sutanto‐Ward E & Prendergast GC (2005) Inhibition of indoleamine 2,3‐dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy. Nat Med 11, 312–319. [DOI] [PubMed] [Google Scholar]
  • 65. Puccetti P (2007) On watching the watchers: IDO and type I/II IFN. Eur J Immunol 37, 876–879. [DOI] [PubMed] [Google Scholar]
  • 66. Mondanelli G, Iacono A, Allegrucci M, Puccetti P & Grohmann U (2019) Immunoregulatory interplay between arginine and tryptophan metabolism in health and disease. Front Immunol 10, 1565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Grohmann U & Bronte V (2010) Control of immune response by amino acid metabolism. Immunol Rev 236, 243–264. [DOI] [PubMed] [Google Scholar]
  • 68. Proietti E, Rossini S, Grohmann U & Mondanelli G (2020) Polyamines and kynurenines at the intersection of immune modulation. Trends Immunol 41, 1037–1050. [DOI] [PubMed] [Google Scholar]
  • 69. Zhai L, Spranger S, Binder DC, Gritsina G, Lauing KL, Giles FJ & Wainwright DA (2015) Molecular pathways: targeting IDO1 and other tryptophan dioxygenases for cancer immunotherapy. Clin Cancer Res 21, 5427–5433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Tang D, Kang R, Coyne CB, Zeh HJ & Lotze MT (2012) PAMPs and DAMPs: signal 0s that spur autophagy and immunity. Immunol Rev 249, 158–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Kumar H, Kawai T & Akira S (2011) Pathogen recognition by the innate immune system. Int Rev Immunol 30, 16–34. [DOI] [PubMed] [Google Scholar]
  • 72. Aksoy E, Taboubi S, Torres D, Delbauve S, Hachani A, Whitehead MA, Pearce WP, Berenjeno IM, Nock G, Filloux A et al. (2012) The p110delta isoform of the kinase PI(3)K controls the subcellular compartmentalization of TLR4 signaling and protects from endotoxic shock. Nat Immunol 13, 1045–1054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Salazar F, Awuah D, Negm OH, Shakib F & Ghaemmaghami AM (2017) The role of indoleamine 2,3‐dioxygenase‐aryl hydrocarbon receptor pathway in the TLR4‐induced tolerogenic phenotype in human DCs. Sci Rep 7, 43337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Mellor AL, Baban B, Chandler PR, Manlapat A, Kahler DJ & Munn DH (2005) Cutting edge: CpG oligonucleotides induce splenic CD19+ dendritic cells to acquire potent indoleamine 2,3‐dioxygenase‐dependent T cell regulatory functions via IFN Type 1 signaling. J Immunol 175, 5601–5605. [DOI] [PubMed] [Google Scholar]
  • 75. Volpi C, Fallarino F, Pallotta MT, Bianchi R, Vacca C, Belladonna ML, Orabona C, De Luca A, Boon L, Romani L et al. (2013) High doses of CpG oligodeoxynucleotides stimulate a tolerogenic TLR9‐TRIF pathway. Nat Commun 4, 1852. [DOI] [PubMed] [Google Scholar]
  • 76. Grohmann U, Orabona C, Fallarino F, Vacca C, Calcinaro F, Falorni A, Candeloro P, Belladonna ML, Bianchi R, Fioretti MC et al. (2002) CTLA‐4‐Ig regulates tryptophan catabolism in vivo. Nat Immunol 3, 1097–1101. [DOI] [PubMed] [Google Scholar]
  • 77. Fallarino F, Grohmann U, Hwang KW, Orabona C, Vacca C, Bianchi R, Belladonna ML, Fioretti MC, Alegre ML & Puccetti P (2003) Modulation of tryptophan catabolism by regulatory T cells. Nat Immunol 4, 1206–1212. [DOI] [PubMed] [Google Scholar]
  • 78. Ayroldi E & Grohmann U (2020) Exemplifying complexity of immune suppression by a "canonical" speech: A glimpse into TNFRSF‐activated signaling pathways in Treg cells. Eur J Immunol 50, 944–948. [DOI] [PubMed] [Google Scholar]
  • 79. Grohmann U, Volpi C, Fallarino F, Bozza S, Bianchi R, Vacca C, Orabona C, Belladonna ML, Ayroldi E, Nocentini G et al. (2007) Reverse signaling through GITR ligand enables dexamethasone to activate IDO in allergy. Nat Med 13, 579–586. [DOI] [PubMed] [Google Scholar]
  • 80. Honig A, Rieger L, Kapp M, Sutterlin M, Dietl J & Kammerer U (2004) Indoleamine 2,3‐dioxygenase (IDO) expression in invasive extravillous trophoblast supports role of the enzyme for materno‐fetal tolerance. J Reprod Immunol 61, 79–86. [DOI] [PubMed] [Google Scholar]
  • 81. Littlejohn TK, Takikawa O, Truscott RJ & Walker MJ (2003) Asp274 and his346 are essential for heme binding and catalytic function of human indoleamine 2,3‐dioxygenase. J Biol Chem 278, 29525–29531. [DOI] [PubMed] [Google Scholar]
  • 82. Ball HJ, Sanchez‐Perez A, Weiser S, Austin CJ, Astelbauer F, Miu J, McQuillan JA, Stocker R, Jermiin LS & Hunt NH (2007) Characterization of an indoleamine 2,3‐dioxygenase‐like protein found in humans and mice. Gene 396, 203–213. [DOI] [PubMed] [Google Scholar]
  • 83. Okkenhaug K & Vanhaesebroeck B (2003) PI3K in lymphocyte development, differentiation and activation. Nat Rev Immunol 3, 317–330. [DOI] [PubMed] [Google Scholar]
  • 84. Burke JE (2018) Structural basis for regulation of phosphoinositide kinases and their involvement in human disease. Mol Cell 71, 653–673. [DOI] [PubMed] [Google Scholar]
  • 85. Dornan GL & Burke JE (2018) Molecular mechanisms of human disease mediated by oncogenic and primary immunodeficiency mutations in class IA phosphoinositide 3‐kinases. Front Immunol 9, 575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Fruman DA, Chiu H, Hopkins BD, Bagrodia S, Cantley LC & Abraham RT (2017) The PI3K Pathway in Human Disease. Cell 170, 605–635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Vanhaesebroeck B, Guillermet‐Guibert J, Graupera M & Bilanges B (2010) The emerging mechanisms of isoform‐specific PI3K signalling. Nat Rev Mol Cell Biol 11, 329–341. [DOI] [PubMed] [Google Scholar]
  • 88. Murphy JE, Padilla BE, Hasdemir B, Cottrell GS & Bunnett NW (2009) Endosomes: a legitimate platform for the signaling train. Proc Natl Acad Sci USA 106, 17615–17622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Hayes S, Chawla A & Corvera S (2002) TGF beta receptor internalization into EEA1‐enriched early endosomes: role in signaling to Smad2. J Cell Biol 158, 1239–1249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Penheiter SG, Mitchell H, Garamszegi N, Edens M, Dore JJ Jr & Leof EB (2002) Internalization‐dependent and ‐independent requirements for transforming growth factor beta receptor signaling via the Smad pathway. Mol Cell Biol 22, 4750–4759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Romani R, Pirisinu I, Calvitti M, Pallotta MT, Gargaro M, Bistoni G, Vacca C, Di Michele A, Orabona C, Rosati J et al. (2015) Stem cells from human amniotic fluid exert immunoregulatory function via secreted indoleamine 2,3‐dioxygenase1. J Cell Mol Med 19, 1593–1605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Vojtech L, Zhang M, Dave V, Levy C, Hughes SM, Wang R, Calienes F, Prlic M, Nance E & Hladik F (2019) Extracellular vesicles in human semen modulate antigen‐presenting cell function and decrease downstream antiviral T cell responses. PLoS ONE 14, e0223901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Ricklefs FL, Alayo Q, Krenzlin H, Mahmoud AB, Speranza MC, Nakashima H, Hayes JL, Lee K, Balaj L, Passaro C et al. (2018) Immune evasion mediated by PD‐L1 on glioblastoma‐derived extracellular vesicles. Sci Adv 4, eaar2766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. He JG, Xie QL, Li BB, Zhou L & Yan D (2018) Exosomes derived from IDO1‐overexpressing rat bone marrow mesenchymal stem cells promote immunotolerance of cardiac allografts. Cell Transplant 27, 1657–1683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Adamo A, Frusteri C, Pallotta MT, Pirali T, Sartoris S & Ugel S (2020) Moonlighting proteins are important players in cancer immunology. Front Immunol 11, 613069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Huberts DH & van der Klei IJ (2010) Moonlighting proteins: an intriguing mode of multitasking. Biochem Biophys Acta 1803, 520–525. [DOI] [PubMed] [Google Scholar]
  • 97. Diaz‐Hidalgo L, Altuntas S, Rossin F, D'Eletto M, Marsella C, Farrace MG, Falasca L, Antonioli M, Fimia GM & Piacentini M (2016) Transglutaminase type 2‐dependent selective recruitment of proteins into exosomes under stressful cellular conditions. Biochem Biophys Acta 1863, 2084–2092. [DOI] [PubMed] [Google Scholar]
  • 98. Eynde BJV, Baren NV & Baurain JF (2020) Is there a clinical future for IDO1 inhibitors after the failure of epacadostat in melanoma? Ann Rev Cancer Biol 4, 241–256. [Google Scholar]
  • 99. Yuasa HJ, Takubo M, Takahashi A, Hasegawa T, Noma H & Suzuki T (2007) Evolution of vertebrate indoleamine 2,3‐dioxygenases. J Mol Evol 65, 705–714. [DOI] [PubMed] [Google Scholar]
  • 100. Metz R, Smith C, DuHadaway JB, Chandler P, Baban B, Merlo LM, Pigott E, Keough MP, Rust S, Mellor AL et al. (2014) IDO2 is critical for IDO1‐mediated T‐cell regulation and exerts a non‐redundant function in inflammation. Int Immunol 26, 357–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Jusof FF, Bakmiwewa SM, Weiser S, Too LK, Metz R, Prendergast GC, Fraser ST, Hunt NH & Ball HJ (2017) Investigation of the tissue distribution and physiological roles of indoleamine 2,3‐dioxygenase‐2. Int J Tryptophan Res. 10, 1178646917735098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Yuasa HJ, Mizuno K & Ball HJ (2015) Low efficiency IDO2 enzymes are conserved in lower vertebrates, whereas higher efficiency IDO1 enzymes are dispensable. FEBS J 282, 2735–2745. [DOI] [PubMed] [Google Scholar]
  • 103. Metz R, Duhadaway JB, Kamasani U, Laury‐Kleintop L, Muller AJ & Prendergast GC (2007) Novel tryptophan catabolic enzyme IDO2 is the preferred biochemical target of the antitumor indoleamine 2,3‐dioxygenase inhibitory compound D‐1‐methyl‐tryptophan. Cancer Res 67, 7082–7087. [DOI] [PubMed] [Google Scholar]
  • 104. Merlo LMF, DuHadaway JB, Montgomery JD, Peng WD, Murray PJ, Prendergast GC, Caton AJ, Muller AJ & Mandik‐Nayak L (2020) Differential roles of IDO1 and IDO2 in T and B cell inflammatory immune responses. Front Immunol 11, 1861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Witkiewicz AK, Costantino CL, Metz R, Muller AJ, Prendergast GC, Yeo CJ & Brody JR (2009) Genotyping and expression analysis of IDO2 in human pancreatic cancer: a novel, active target. J Am Coll Surg 208, 781–787. discussion 787–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Mandarano M, Bellezza G, Belladonna ML, Vannucci J, Gili A, Ferri I, Lupi C, Ludovini V, Falabella G, Metro G et al. (2020) Indoleamine 2,3‐dioxygenase 2 immunohistochemical expression in resected human non‐small cell lung cancer: a potential new prognostic tool. Front Immunol 11, 839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Nevler A, Muller AJ, Sutanto‐Ward E, DuHadaway JB, Nagatomo K, Londin E, O'Hayer K, Cozzitorto JA, Lavu H, Yeo TP et al. (2019) Host IDO2 gene status influences tumor progression and radiotherapy response in KRAS‐driven sporadic pancreatic cancers. Clin Cancer Res 25, 724–734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Mondanelli G, Mandarano M, Belladonna ML, Suvieri C, Pelliccia C, Bellezza G, Sidoni A, Carvalho A, Grohmann U & Volpi C (2021) Current challenges for IDO2 as target in cancer immunotherapy. Front Immunol 12, 679953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Li P, Xu W, Liu F, Zhu H, Zhang L, Ding Z, Liang H & Song J (2021) The emerging roles of IDO2 in cancer and its potential as a therapeutic target. Biomed Pharmacother 137, 111295. [DOI] [PubMed] [Google Scholar]
  • 110. Capece L, Arrar M, Roitberg AE, Yeh SR, Marti MA & Estrin DA (2010) Substrate stereo‐specificity in tryptophan dioxygenase and indoleamine 2,3‐dioxygenase. Proteins 78, 2961–2972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Kanai M, Funakoshi H, Takahashi H, Hayakawa T, Mizuno S, Matsumoto K & Nakamura T (2009) Tryptophan 2,3‐dioxygenase is a key modulator of physiological neurogenesis and anxiety‐related behavior in mice. Mol Brain 2, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Schramme F, Crosignani S, Frederix K, Hoffmann D, Pilotte L, Stroobant V, Preillon J, Driessens G & Van den Eynde BJ (2020) Inhibition of tryptophan‐dioxygenase activity increases the antitumor efficacy of immune checkpoint inhibitors. Cancer Immunol Res 8, 32–45. [DOI] [PubMed] [Google Scholar]
  • 113. Li DD, Gao YJ, Tian XC, Yang ZQ, Cao H, Zhang QL, Guo B & Yue ZP (2014) Differential expression and regulation of Tdo2 during mouse decidualization. J Endocrinol 220, 73–83. [DOI] [PubMed] [Google Scholar]
  • 114. Yu CP, Pan ZZ & Luo DY (2016) TDO as a therapeutic target in brain diseases. Metab Brain Dis 31, 737–747. [DOI] [PubMed] [Google Scholar]
  • 115. Michels H, Seinstra RI, Uitdehaag JC, Koopman M, van Faassen M , Martineau CN, Kema IP, Buijsman R & Nollen EA (2016) Identification of an evolutionary conserved structural loop that is required for the enzymatic and biological function of tryptophan 2,3‐dioxygenase. Sci Rep 6, 39199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Wu W, Nicolazzo JA, Wen L, Chung R, Stankovic R, Bao SS, Lim CK, Brew BJ, Cullen KM & Guillemin GJ (2013) Expression of tryptophan 2,3‐dioxygenase and production of kynurenine pathway metabolites in triple transgenic mice and human Alzheimer's disease brain. PLoS ONE 8, e59749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Breda C, Sathyasaikumar KV, Sograte Idrissi S, Notarangelo FM, Estranero JG, Moore GG, Green EW, Kyriacou CP, Schwarcz R & Giorgini F (2016) Tryptophan‐2,3‐dioxygenase (TDO) inhibition ameliorates neurodegeneration by modulation of kynurenine pathway metabolites. Proc Natl Acad Sci USA 113, 5435–5440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Hoffmann D, Dvorakova T, Stroobant V, Bouzin C, Daumerie A, Solvay M, Klaessens S, Letellier MC, Renauld JC, van Baren N et al. (2020) Tryptophan 2,3‐dioxygenase expression identified in human hepatocellular carcinoma cells and in intratumoral pericytes of most cancers. Cancer Immunol Res 8, 19–31. [DOI] [PubMed] [Google Scholar]
  • 119. Pilotte L, Larrieu P, Stroobant V, Colau D, Dolusic E, Frederick R, De Plaen E, Uyttenhove C, Wouters J, Masereel B et al. (2012) Reversal of tumoral immune resistance by inhibition of tryptophan 2,3‐dioxygenase. Proc Natl Acad Sci USA 109, 2497–2502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Dolusic E, Larrieu P, Moineaux L, Stroobant V, Pilotte L, Colau D, Pochet L, Van den Eynde B, Masereel B, Wouters J et al. (2011) Tryptophan 2,3‐dioxygenase (TDO) inhibitors. 3‐(2‐(pyridyl)ethenyl)indoles as potential anticancer immunomodulators. J Med Chem 54, 5320–5334. [DOI] [PubMed] [Google Scholar]
  • 121. Routy JP, Routy B, Graziani GM & Mehraj V (2016) The kynurenine pathway is a double‐edged sword in immune‐privileged sites and in cancer: implications for immunotherapy. Int J Tryptophan Res 9, 67–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Beutelspacher SC, Pillai R, Watson MP, Tan PH, Tsang J, McClure MO, George AJ & Larkin DF (2006) Function of indoleamine 2,3‐dioxygenase in corneal allograft rejection and prolongation of allograft survival by over‐expression. Eur J Immunol 36, 690–700. [DOI] [PubMed] [Google Scholar]
  • 123. Guillemin GJ, Kerr SJ, Smythe GA, Smith DG, Kapoor V, Armati PJ, Croitoru J & Brew BJ (2001) Kynurenine pathway metabolism in human astrocytes: a paradox for neuronal protection. J Neurochem 78, 842–853. [DOI] [PubMed] [Google Scholar]
  • 124. Li Q, Harden JL, Anderson CD & Egilmez NK (2016) Tolerogenic phenotype of IFN‐gamma‐induced IDO+ dendritic cells is maintained via an autocrine IDO‐kynurenine/AhR‐IDO loop. J Immunol 197, 962–970. [DOI] [PubMed] [Google Scholar]
  • 125. Gargaro M, Vacca C, Massari S, Scalisi G, Manni G, Mondanelli G, Mazza EMC, Bicciato S, Pallotta MT, Orabona C et al. (2019) Engagement of nuclear coactivator 7 by 3‐hydroxyanthranilic acid enhances activation of Aryl hydrocarbon receptor in immunoregulatory dendritic cells. Front Immunol 10, 1973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Manni G, Mondanelli G, Scalisi G, Pallotta MT, Nardi D, Padiglioni E, Romani R, Talesa VN, Puccetti P, Fallarino F et al. (2020) Pharmacologic induction of endotoxin tolerance in dendritic cells by L‐kynurenine. Front Immunol 11, 292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Braidy N, Guillemin GJ & Grant R (2011) Effects of kynurenine pathway inhibition on NAD metabolism and cell viability in human primary astrocytes and neurons. Int J Tryptophan Res 4, 29–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Grant R, Nguyen S & Guillemin G (2010) Kynurenine pathway metabolism is involved in the maintenance of the intracellular NAD concentration in human primary astrocytes. Int J Tryptophan Res 3, 151–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Orabona C, Mondanelli G, Puccetti P & Grohmann U (2018) Immune checkpoint molecules, personalized immunotherapy, and autoimmune diabetes. Trends Mol Med 24, 931–941. [DOI] [PubMed] [Google Scholar]
  • 130. Zelante T, Iannitti RG, Fallarino F, Gargaro M, De Luca A, Moretti S, Bartoli A & Romani L (2014) Tryptophan feeding of the IDO1‐AhR axis in host‐microbial symbiosis. Front Immunol 5, 640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Platten M, Nollen EAA, Rohrig UF, Fallarino F & Opitz CA (2019) Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat Rev Drug Discov 18, 379–401. [DOI] [PubMed] [Google Scholar]
  • 132. Schmidt SV & Schultze JL (2014) New Insights into IDO Biology in Bacterial and Viral Infections. Front Immunol 5, 384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Beatty WL, Belanger TA, Desai AA, Morrison RP & Byrne GI (1994) Tryptophan depletion as a mechanism of gamma interferon‐mediated chlamydial persistence. Infect Immun 62, 3705–3711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Russell DG (2013) Trp'ing tuberculosis. Cell 155, 1209–1210. [DOI] [PubMed] [Google Scholar]
  • 135. Zhang YJ & Rubin EJ (2013) Feast or famine: the host‐pathogen battle over amino acids. Cell Microbiol 15, 1079–1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Zhang YJ, Reddy MC, Ioerger TR, Rothchild AC, Dartois V, Schuster BM, Trauner A, Wallis D, Galaviz S, Huttenhower C et al. (2013) Tryptophan biosynthesis protects mycobacteria from CD4 T‐cell‐mediated killing. Cell 155, 1296–1308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Boasso A, Vaccari M, Fuchs D, Hardy AW, Tsai WP, Tryniszewska E, Shearer GM & Franchini G (2009) Combined effect of antiretroviral therapy and blockade of IDO in SIV‐infected rhesus macaques. J Immunol 182, 4313–4320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Gautam US, Foreman TW, Bucsan AN, Veatch AV, Alvarez X, Adekambi T, Golden NA, Gentry KM, Doyle‐Meyers LA, Russell‐Lodrigue KE et al. (2018) In vivo inhibition of tryptophan catabolism reorganizes the tuberculoma and augments immune‐mediated control of Mycobacterium tuberculosis. Proc Natl Acad Sci USA 115, E62–E71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Romani L & Puccetti P (2006) Protective tolerance to fungi: the role of IL‐10 and tryptophan catabolism. Trends Microbiol 14, 183–189. [DOI] [PubMed] [Google Scholar]
  • 140. Zelante T, Choera T, Beauvais A, Fallarino F, Paolicelli G, Pieraccini G, Pieroni M, Galosi C, Beato C, De Luca A et al. (2021) Aspergillus fumigatus tryptophan metabolic route differently affects host immunity. Cell Rep 34, 108673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Platten M, von Knebel Doeberitz N , Oezen I, Wick W & Ochs K (2014) Cancer immunotherapy by targeting IDO1/TDO and their downstream effectors. Front Immunol 5, 673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Prendergast GC, Malachowski WP, DuHadaway JB & Muller AJ (2017) Discovery of IDO1 inhibitors: from bench to bedside. Cancer Res 77, 6795–6811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Vacchelli E, Aranda F, Eggermont A, Sautes‐Fridman C, Tartour E, Kennedy EP, Platten M, Zitvogel L, Kroemer G & Galluzzi L (2014) Trial watch: IDO inhibitors in cancer therapy. Oncoimmunology. 3, e957994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Sun L (2020) Advances in the discovery and development of selective heme‐displacing IDO1 inhibitors. Expert Opin Drug Discov 15, 1223–1232. [DOI] [PubMed] [Google Scholar]
  • 145. Khair DO, Bax HJ, Mele S, Crescioli S, Pellizzari G, Khiabany A, Nakamura M, Harris RJ, French E, Hoffmann RM et al. (2019) Combining immune checkpoint inhibitors: established and emerging targets and strategies to improve outcomes in melanoma. Front Immunol 10, 453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Ladomersky E, Zhai L, Lenzen A, Lauing KL, Qian J, Scholtens DM, Gritsina G, Sun X, Liu Y, Yu F et al. (2018) IDO1 inhibition synergizes with radiation and PD‐1 blockade to durably increase survival against advanced glioblastoma. Clin Cancer Res 24, 2559–2573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Brandacher G, Perathoner A, Ladurner R, Schneeberger S, Obrist P, Winkler C, Werner ER, Werner‐Felmayer G, Weiss HG, Gobel G et al. (2006) Prognostic value of indoleamine 2,3‐dioxygenase expression in colorectal cancer: effect on tumor‐infiltrating T cells. Clin Cancer Res 12, 1144–1151. [DOI] [PubMed] [Google Scholar]
  • 148. Inaba T, Ino K, Kajiyama H, Yamamoto E, Shibata K, Nawa A, Nagasaka T, Akimoto H, Takikawa O & Kikkawa F (2009) Role of the immunosuppressive enzyme indoleamine 2,3‐dioxygenase in the progression of ovarian carcinoma. Gynecol Oncol 115, 185–192. [DOI] [PubMed] [Google Scholar]
  • 149. Long GV, Dummer R, Hamid O, Gajewski TF, Caglevic C, Dalle S, Arance A, Carlino MS, Grob JJ, Kim TM et al. (2019) Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO‐301/KEYNOTE‐252): a phase 3, randomised, double‐blind study. Lancet Oncol 20, 1083–1097. [DOI] [PubMed] [Google Scholar]
  • 150. Labadie BW, Bao R & Luke JJ (2019) Reimagining IDO pathway inhibition in cancer immunotherapy via downstream focus on the Tryptophan‐Kynurenine‐Aryl hydrocarbon axis. Clin Cancer Res 25, 1462–1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Mondanelli G, Iacono A, Carvalho A, Orabona C, Volpi C, Pallotta MT, Matino D, Esposito S & Grohmann U (2019) Amino acid metabolism as drug target in autoimmune diseases. Autoimmun Rev 18, 334–348. [DOI] [PubMed] [Google Scholar]
  • 152. Grohmann U, Fallarino F, Bianchi R, Orabona C, Vacca C, Fioretti MC & Puccetti P (2003) A defect in tryptophan catabolism impairs tolerance in nonobese diabetic mice. J Exp Med 198, 153–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Mondanelli G, Albini E, Pallotta MT, Volpi C, Chatenoud L, Kuhn C, Fallarino F, Matino D, Belladonna ML, Bianchi R et al. (2017) The proteasome inhibitor bortezomib controls indoleamine 2,3‐dioxygenase 1 breakdown and restores immune regulation in autoimmune diabetes. Front Immunol 8, 428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Orabona C, Mondanelli G, Pallotta MT, Carvalho A, Albini E, Fallarino F, Vacca C, Volpi C, Belladonna ML, Berioli MG et al. (2018) Deficiency of immunoregulatory indoleamine 2,3‐dioxygenase 1in juvenile diabetes. JCI insight. 3 e96244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Arif S, Leete P, Nguyen V, Marks K, Nor NM, Estorninho M, Kronenberg‐Versteeg D, Bingley PJ, Todd JA, Guy C et al. (2014) Blood and islet phenotypes indicate immunological heterogeneity in type 1 diabetes. Diabetes 63, 3835–3845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Negrotto L & Correale J (2017) Amino acid catabolism in multiple sclerosis affects immune homeostasis. J Immunol 198, 1900–1909. [DOI] [PubMed] [Google Scholar]
  • 157. Boros FA, Bohar Z & Vecsei L (2018) Genetic alterations affecting the genes encoding the enzymes of the kynurenine pathway and their association with human diseases. Mutat Res 776, 32–45. [DOI] [PubMed] [Google Scholar]
  • 158. Tardito S, Negrini S, Conteduca G, Ferrera F, Parodi A, Battaglia F, Kalli F, Fenoglio D, Cutolo M & Filaci G (2013) Indoleamine 2,3 dioxygenase gene polymorphisms correlate with CD8+ Treg impairment in systemic sclerosis. Hum Immunol 74, 166–169. [DOI] [PubMed] [Google Scholar]
  • 159. Mondanelli G, Di Battista V, Pellanera F, Mammoli A, Macchiarulo A, Gargaro M, Mavridou E, Matteucci C, Ruggeri L, Orabona C et al. (2020) A novel mutation of indoleamine 2,3‐dioxygenase 1 causes a rapid proteasomal degradation and compromises protein function. J Autoimmun 115, 102509. [DOI] [PubMed] [Google Scholar]
  • 160. Maddison DC & Giorgini F (2015) The kynurenine pathway and neurodegenerative disease. Semin Cell Dev Biol 40, 134–141. [DOI] [PubMed] [Google Scholar]
  • 161. Guillemin GJ, Smythe GA, Veas LA, Takikawa O & Brew BJ (2003) A beta 1–42 induces production of quinolinic acid by human macrophages and microglia. NeuroReport 14, 2311–2315. [DOI] [PubMed] [Google Scholar]
  • 162. Rahman A, Ting K, Cullen KM, Braidy N, Brew BJ & Guillemin GJ (2009) The excitotoxin quinolinic acid induces tau phosphorylation in human neurons. PLoS ONE 4, e6344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Jauch D, Urbanska EM, Guidetti P, Bird ED, Vonsattel JP, Whetsell WO Jr & Schwarcz R (1995) Dysfunction of brain kynurenic acid metabolism in Huntington's disease: focus on kynurenine aminotransferases. J Neurol Sci 130, 39–47. [DOI] [PubMed] [Google Scholar]
  • 164. Zinger A, Barcia C, Herrero MT & Guillemin GJ (2011) The involvement of neuroinflammation and kynurenine pathway in Parkinson's disease. Parkinsons Dis 2011, 716859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Pham KN, Lewis‐Ballester A & Yeh SR (2021) Conformational plasticity in human heme‐based dioxygenases. J Am Chem Soc 143, 1836–1845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Lun CM, Bishop BM & Smith LC (2017) Multitasking immune Sp185/333 protein, Rsptransformer‐E1, and Its recombinant fragments undergo secondary structural transformation upon binding targets. J Immunol 198, 2957–2966. [DOI] [PubMed] [Google Scholar]
  • 167. Kursula P (2014) The many structural faces of calmodulin: a multitasking molecular jackknife. Amino Acids 46, 2295–2304. [DOI] [PubMed] [Google Scholar]
  • 168. Kuo HH & Mauk AG (2012) Indole peroxygenase activity of indoleamine 2,3‐dioxygenase. Proc Natl Acad Sci USA 109, 13966–13971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Freewan M, Rees MD, Plaza TS, Glaros E, Lim YJ, Wang XS, Yeung AW, Witting PK, Terentis AC & Thomas SR (2013) Human indoleamine 2,3‐dioxygenase is a catalyst of physiological heme peroxidase reactions: implications for the inhibition of dioxygenase activity by hydrogen peroxide. J Biol Chem 288, 1548–1567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Lim YJ, Foo TC, Yeung AWS, Tu X, Ma Y, Hawkins CL, Witting PK, Jameson GNL, Terentis AC & Thomas SR (2019) Human indoleamine 2,3‐dioxygenase 1 is an efficient mammalian nitrite reductase. Biochemistry 58, 974–986. [DOI] [PubMed] [Google Scholar]
  • 171. Stanley CP, Maghzal GJ, Ayer A, Talib J, Giltrap AM, Shengule S, Wolhuter K, Wang Y, Chadha P, Suarna C et al. (2019) Singlet molecular oxygen regulates vascular tone and blood pressure in inflammation. Nature 566, 548–552. [DOI] [PubMed] [Google Scholar]
  • 172. Albini E, Coletti A, Greco F, Pallotta MT, Mondanelli G, Gargaro M, Belladonna ML, Volpi C, Bianchi R, Grohmann U et al. (2018) Identification of a 2‐propanol analogue modulating the non‐enzymatic function of indoleamine 2,3‐dioxygenase 1. Biochem Pharmacol 158, 286–297. [DOI] [PubMed] [Google Scholar]

Articles from The Febs Journal are provided here courtesy of Wiley

RESOURCES