Indoleamine 2,3-dioxygenase 1 (IDO1) is a heme-containing enzyme encoded by the IDO1 gene in humans. The term IDO1 Human, Active refers to the recombinant form of this enzyme, produced in Escherichia coli for research and therapeutic applications. It is a non-glycosylated protein fused with an N-terminal His-tag, enabling efficient purification and functional assays . IDO1 catalyzes the first rate-limiting step in tryptophan catabolism, converting L-tryptophan to N-formylkynurenine (NFK) via a kynurenine pathway . Its activity is critical in immune regulation, tumor immune evasion, and inflammation control .
IDO1 Human, Active is produced via bacterial expression and purified using chromatographic techniques:
IDO1 Human, Active performs two key functions:
Tryptophan Catabolism:
Immune Regulation:
IDO1 Human, Active is instrumental in studying:
Enzyme Kinetics: Assessing substrate/inhibitor interactions (e.g., heme competition) .
Therapeutic Target Validation: Testing inhibitors (e.g., β-lapachone, epacadostat) to restore anti-tumor immunity .
Mechanistic Insights: Elucidating heme lability and phosphorylation-dependent regulation .
Parameter | Value |
---|---|
Source | Recombinant E. coli |
Sequence | Human IDO1 (aa 1-403) with N-terminal His-tag |
Activity Assay Protocol | Catalase-dependent NFK production |
Endotoxin | <0.1 EU/μg |
Enzyme | Tissue Distribution | Substrate Specificity | Role in Immunity |
---|---|---|---|
IDO1 | Broad (e.g., placenta, tumors) | L/T-tryptophan, serotonin | Immunosuppression, tumor evasion |
IDO2 | Liver, kidney | L-tryptophan | Modulates IDO1 activity via heme transfer |
TDO | Liver, brain | L-tryptophan | Tryptophan metabolism in non-immune contexts |
Human IDO1 is a monomeric protein consisting of large and small domains connected by a loop (DE-loop; residues 260-268) positioned above the sixth coordination site of the heme cofactor. A critical structural element is the flexible JK-loop (residues 360-382) that controls access to the catalytic site by adopting distinct closed, intermediate, and open conformations . The protein contains a narrow channel formed by α-helices E and F that extends from the solvent-exposed surface to the heme group, facilitating the shuttling of oxygen to the catalytic cleft for oxidative cleavage of the indole ring of L-tryptophan . This structure enables IDO1's versatility in recognizing various substrates beyond L-tryptophan, including serotonin, melatonin, and tryptamine.
IDO1 performs two distinct functions: enzymatic (catalytic) and non-enzymatic (signaling). The enzymatic function occurs in the cytosol and involves the degradation of tryptophan along the kynurenine pathway, producing kynurenine metabolites that have immunomodulatory effects . The non-enzymatic function involves phosphorylation of ITIM (immunoreceptor tyrosine-based inhibitory motif) sequences, which leads to the activation of SHP1/SHP2 phosphatases and subsequent noncanonical NF-κB signaling. This signaling function primarily occurs in early endosomes and reprograms dendritic cells toward a long-term immunoregulatory phenotype . These dual functions allow IDO1 to fulfill distinct environmental needs and explain its complex role in immune regulation.
In normal cells, IDO1 expression is primarily induced by inflammatory signals, with interferon-gamma (IFN-γ) being the most potent inducer. The promoter region of the human IDO1 gene (located on chromosome 8p22) contains IFN-stimulated elements (ISREs) and gamma activation sequences (GAS) that respond to transcription factors such as STAT1 and interferon-regulatory factors (IRFs) .
In contrast, tumor cells often display constitutive IDO1 expression even in the absence of inflammatory signals. This constitutive expression represents a state of intrinsic immune resistance and is driven by oncogenic pathways, particularly mutations in the PI3K and MAPK signaling pathways . Analysis of the Cancer Cell Line Encyclopedia (CCLE) database confirms that IDO1-expressing tumor lines harbor mutations in these pathways more frequently than IDO1-negative lines . This constitutive expression in tumors may contribute to the "cold" or "noninflamed" tumor phenotype that fails to respond to checkpoint inhibitors due to lack of T-cell infiltration.
Measuring IDO1 enzymatic activity typically involves quantifying either tryptophan consumption or kynurenine production. For reliable measurements, a combination of approaches is recommended:
HPLC-based assays: High-performance liquid chromatography offers precise quantification of both tryptophan and kynurenine. The assay typically includes:
Sample preparation with protein extraction and potential deproteinization
Chromatographic separation of tryptophan and kynurenine
Detection using fluorescence for tryptophan and UV absorbance for kynurenine
Calculation of the kynurenine/tryptophan ratio as an indicator of IDO1 activity
Colorimetric assays: These measure kynurenine levels after reaction with p-dimethylaminobenzaldehyde (Ehrlich's reagent), which produces a yellow color measurable at 490 nm. While less sensitive than HPLC, this method is suitable for high-throughput screening.
Isotope labeling: Using isotope-labeled tryptophan (e.g., 13C-tryptophan) followed by mass spectrometry provides highly accurate measurements and can distinguish between different metabolic pathways.
When conducting these assays, researchers should include appropriate controls to account for potential interfering factors, such as the presence of tryptophan dioxygenase (TDO) or indoleamine 2,3-dioxygenase 2 (IDO2) , which can also catalyze tryptophan degradation.
Differentiating between IDO1's enzymatic and signaling functions requires specific experimental approaches:
Producing active human IDO1 protein presents several challenges due to its heme-binding properties and complex folding requirements. Optimal expression systems include:
Mammalian expression systems:
HEK293 cells generally yield properly folded human IDO1 with appropriate post-translational modifications
CHO cells can be used for larger-scale production with good activity retention
These systems typically require the addition of hemin to the culture medium to ensure proper heme incorporation
Insect cell/baculovirus systems:
Sf9 or High Five insect cells provide high yields of active protein
This system balances higher expression levels than mammalian cells with better folding than bacterial systems
Addition of hemin is essential for obtaining enzymatically active protein
E. coli systems with optimization:
Although challenging, bacterial expression can be optimized by:
Using specialized strains that enhance disulfide bond formation (e.g., Origami)
Co-expressing molecular chaperones
Inclusion of hemin in the growth medium
Controlled induction at lower temperatures (16-20°C)
Careful refolding from inclusion bodies when necessary
For crystallography studies, as shown in multiple structural investigations, specific constructs with optimized solubility and stability may be necessary . When evaluating protein activity, researchers should assess both heme incorporation (via UV-visible spectroscopy) and enzymatic function, as a significant fraction of expressed IDO1 may be in the apo form lacking heme .
Distinguishing between constitutive and IFN-γ-induced IDO1 expression in tumor samples requires a multi-parameter approach:
Immunohistochemical analysis combining IDO1 staining with:
T-cell infiltration markers (CD3, CD8)
IFN-γ signaling markers (phospho-STAT1, IRF1)
Assessment of spatial relationships between IDO1+ cells and T cells
Gene expression profiling:
Quantify IDO1 mRNA alongside IFN-γ-responsive genes
Analyze expression of genes associated with constitutive IDO1 expression (PI3K/MAPK pathway components)
Calculate gene signature scores for inflammatory versus non-inflammatory tumors
Molecular pathway analysis:
Ex vivo culture systems:
Tumors with constitutive IDO1 expression typically show IDO1 positivity in the absence of T-cell infiltration and maintain expression when IFN-γ signaling is blocked. These tumors often harbor oncogenic mutations in PI3K or MAPK pathways, similar to patterns observed with constitutive PD-L1 expression in certain cancers .
IDO1 protein exhibits significant structural plasticity that affects inhibitor binding. Key factors include:
JK-loop conformation (residues 360-382):
This flexible loop controls access to the catalytic site and can adopt closed, intermediate, or open conformations
In substrate-free structures, the JK-loop is often unresolved, suggesting high flexibility
Substrate binding can stabilize closed loop conformations when the ligand makes specific interactions with loop residues
Smaller ligands often cannot stabilize closed-loop conformations, while larger ligands may occupy pocket B and impede loop closure
Heme redox state:
The ferric (Fe3+) state is the resting state
The ferrous (Fe2+) state binds O2 and is catalytically active
Different inhibitors may preferentially bind to specific redox states
The redox state affects the conformation of surrounding residues and access to binding pockets
Binding pocket architecture:
Pocket A accommodates the indole ring of tryptophan and is the primary binding site
Pocket B is adjacent to pocket A and accommodates the amino acid portion of tryptophan
Pocket C extends beyond pocket A and is utilized by some inhibitors
Pocket D is a secondary binding site opened by conformational changes in Phe270
Allosteric effects:
These factors are critical considerations when designing IDO1 inhibitors, as they determine which conformational states and binding pockets are accessible to different chemical scaffolds.
IDO1 integrates with multiple immunoregulatory pathways in the tumor microenvironment, creating complex networks that support immune evasion:
IDO1-AhR-PD-1 axis:
Kynurenine produced by IDO1 acts as a ligand for the aryl hydrocarbon receptor (AhR)
AhR activation upregulates PD-1 expression on T cells
This creates a feed-forward loop where IDO1 activity enhances PD-1-mediated T cell exhaustion
This interaction provides rationale for combination therapies targeting both IDO1 and PD-1/PD-L1
IDO1-COX-2-PGE2 autocrine signaling:
In many tumors, IDO1 expression is maintained through an autocrine loop involving cyclooxygenase-2 (COX-2) and prostaglandin E2 (PGE2)
PGE2 activates prostaglandin E receptors, triggering PI3K activation
PI3K activation leads to IDO1 expression through various downstream effectors
COX-2 inhibitors can disrupt this loop and prevent constitutive IDO1 expression
IDO1-TGF-β regulatory circuit:
IDO1 signaling induces TGF-β expression
TGF-β promotes the signaling function of IDO1 rather than its catalytic activity
This creates a positive feedback loop that establishes long-term immunoregulatory phenotypes in dendritic cells
TGF-β also induces arginase 1 (ARG1), another potent immunosuppressor that acts synergistically with IDO1
IDO1 and myeloid-derived suppressor cells (MDSCs):
IDO1 activity promotes MDSC accumulation and activation in the tumor microenvironment
MDSCs express additional immunosuppressive factors that complement IDO1 function
This interaction contributes to layered immunosuppression mechanisms in tumors
Understanding these interactions is essential for designing effective combination immunotherapy strategies that can overcome the complex immunosuppressive networks in tumors.
Designing robust screening assays for IDO1 inhibitors requires careful consideration of several factors:
Selection of appropriate enzyme source:
Recombinant human IDO1 with confirmed heme incorporation and enzymatic activity
Evaluation of specific activity and stability under assay conditions
Consideration of full-length versus truncated constructs depending on assay goals
Assay format considerations:
Biochemical assays measuring direct enzymatic activity:
Colorimetric kynurenine detection (high throughput but lower sensitivity)
HPLC-based methods (higher sensitivity but lower throughput)
Oxygen consumption measurements (real-time kinetics)
Cell-based assays:
IDO1-expressing tumor cell lines (especially those with constitutive expression)
Induced IDO1 expression systems (e.g., IFN-γ-treated cells)
Reporter systems linking IDO1 activity to fluorescent or luminescent readouts
Redox considerations:
Maintenance of proper redox environment for IDO1 activity
Inclusion of reducing agents (e.g., methylene blue, ascorbic acid)
Counter-screening against compounds with redox-cycling properties that may give false positives
Control compounds:
Inclusion of established IDO1 inhibitors as positive controls (e.g., 1-methyl-tryptophan)
Testing against related enzymes (TDO, IDO2) to assess selectivity
Evaluation of compound interference with detection methods
Structure-informed screening:
These methodological considerations help ensure that screening campaigns identify genuine IDO1 inhibitors rather than artifacts, and that the inhibitors discovered have properties suitable for further development.
Evaluating IDO1 inhibitors in humanized mouse models presents unique challenges but offers valuable insights into human-specific responses. Effective evaluation requires:
Selection of appropriate humanized model:
CD34+ hematopoietic stem cell-engrafted NSG mice (provide human immune cell development)
PBMC-engrafted NSG mice (faster reconstitution but limited lifespan)
BLT (bone marrow, liver, thymus) mice (more complete immune reconstitution including T-cell education)
Tumor model considerations:
Human tumor cell lines with defined IDO1 expression patterns (constitutive vs. inducible)
Patient-derived xenografts that maintain original tumor microenvironment features
Confirmation of IDO1 expression in established tumors before treatment initiation
Treatment regimen design:
Comprehensive endpoint analyses:
Tumor growth and survival measurements
Immunophenotyping of tumor-infiltrating lymphocytes
Assessment of tryptophan and kynurenine levels in tumor and plasma
Measurement of inflammatory cytokines and immune activation markers
Spatial analysis of immune cell distribution within tumors
Validation in multiple models:
Testing in both "hot" (T-cell inflamed) and "cold" (non-inflamed) tumor models
Evaluation across tumors with different mechanisms of IDO1 expression
Cross-validation with complementary in vitro human immune cell assays
The SKOV3 tumor model in NSG mice reconstituted with human allogeneic lymphocytes has been validated for testing IDO1 inhibitors. In this model, tumors were not rejected unless mice were treated with either a COX-2 inhibitor or an IDO1 inhibitor, demonstrating the relevance of the IDO1 pathway in preventing immune-mediated tumor rejection .
Research on IDO1 structure has produced some apparently contradictory data regarding conformational states. Resolving these contradictions requires sophisticated analytical approaches:
Critical evaluation of structural data quality:
Assessment of resolution, R-free values, and diffraction-component precision index (DPI)
Evaluation of electron density quality for ligands using metrics like EDIAm
Consideration of crystal packing effects on protein conformation
Identification of structures with high coordinate uncertainty (DPI > 0.8 Å)
Integrative structural biology approaches:
Combining X-ray crystallography with solution-based techniques:
Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to assess conformational dynamics
Small-angle X-ray scattering (SAXS) for solution-state conformations
Nuclear magnetic resonance (NMR) for dynamic regions like the JK-loop
Molecular dynamics simulations:
Simulation of IDO1 conformational changes in different environments
Assessment of energy landscapes for different conformational states
Calculation of transition pathways between observed crystallographic conformations
Evaluation of ligand effects on protein dynamics
Functional correlation studies:
Mutagenesis of residues involved in conformational changes
Correlation of structural states with enzymatic or signaling activities
Analysis of structure-activity relationships across multiple inhibitor series
Development of conformation-specific antibodies or nanobodies
Standardized experimental conditions:
Performing structural and functional studies under identical conditions
Controlling redox state, pH, and buffer composition
Using consistent protein constructs across different studies
Implementing rigorous quality control for protein samples
These approaches can help reconcile apparently contradictory data by identifying conditions that favor specific conformational states and understanding the dynamic nature of IDO1 structure in relation to its dual enzymatic and signaling functions.
Recent evidence indicates that a significant proportion of IDO1 protein exists in the apo form (without bound heme) in human monocyte-derived macrophages and tumor cells, even after IFN-γ stimulation . The biology and function of this apo form remain largely unknown, raising important research questions:
Understanding the role of apo-IDO1 could provide insights into the complex biology of this enzyme beyond its canonical tryptophan-catabolizing function and might explain some of the context-dependent effects observed in different disease models.
Differentiating between the three enzymes that catalyze the first step of tryptophan degradation (IDO1, IDO2, and TDO) is crucial for understanding their specific contributions in complex biological samples. Recommended approaches include:
Gene expression analysis with high specificity:
Quantitative PCR with carefully validated primer sets specific for each enzyme
RNA-seq analysis with bioinformatic pipelines that can distinguish between highly similar transcripts
In situ hybridization with gene-specific probes for spatial localization
Protein-level differentiation:
Western blotting with validated antibodies that do not cross-react
Immunoprecipitation followed by mass spectrometry for definitive identification
Targeted proteomics approaches using enzyme-specific peptides
Enzyme-specific activity assays based on differential inhibitor sensitivity
Genetic manipulation strategies:
CRISPR/Cas9-mediated knockout of individual enzymes
siRNA knockdown with rescue experiments using inhibitor-resistant mutants
Cell line models with controlled expression of each enzyme individually
Pharmacological approaches:
Use of selective inhibitors:
1-methyl-L-tryptophan (1-MT): more selective for IDO1 than IDO2
TDO-specific inhibitors (e.g., 680C91, LM10)
Emerging IDO2-selective compounds
Dose-response curves with multiple inhibitors to establish contribution profiles
Metabolic tracing:
Isotopically labeled tryptophan combined with mass spectrometry
Analysis of enzyme-specific metabolic products or intermediates
Compartment-specific metabolic analysis to distinguish spatially separated activities
These approaches, used in combination, can provide a comprehensive understanding of the relative contributions of IDO1, IDO2, and TDO to tryptophan catabolism in complex biological samples, avoiding misattribution of effects in experimental and clinical studies.
Addressing both the enzymatic and signaling functions of IDO1 requires sophisticated targeting strategies:
Dual-function inhibitor development:
Structure-based design of compounds that:
Block the catalytic site to inhibit enzymatic function
Interfere with ITIM phosphorylation or SHP1/SHP2 recruitment to inhibit signaling
Screening pipelines with assays for both functions to identify dual-action compounds
Combination approaches:
Pairing enzymatic inhibitors with signaling pathway inhibitors:
IDO1 catalytic inhibitors + SHP1/SHP2 inhibitors
IDO1 catalytic inhibitors + noncanonical NF-κB pathway inhibitors
Evaluation of synergistic effects in both in vitro and in vivo models
Alternative targeting strategies:
Degrader technologies (PROTACs) to eliminate the IDO1 protein entirely
Antisense oligonucleotides or siRNA approaches to prevent IDO1 expression
Targeting upstream regulators common to both functions
Development of conformation-specific inhibitors that lock IDO1 in inactive states
Context-specific targeting:
Identifying cellular contexts where one function predominates
Developing tissue-targeted delivery systems for inhibitors
Exploiting differences in subcellular localization of enzymatic versus signaling functions
Biomarker development:
Establishing assays to determine predominant IDO1 function in patient samples
Identifying patient subgroups likely to benefit from different targeting approaches
Monitoring both tryptophan/kynurenine levels and signaling pathway activation
Effective targeting of both IDO1 functions could potentially overcome resistance mechanisms observed with first-generation IDO1 inhibitors and provide more comprehensive modulation of IDO1-mediated immunosuppression in cancer and other diseases.
IDO1 exists as a monomer and is composed of a large catalytic domain, which contains the heme group, and a small non-catalytic domain with two immunoreceptor tyrosine-based inhibition motifs (ITIMs) that regulate intracellular signaling . The enzyme catalyzes the oxidation of L-tryptophan to N-formylkynurenine, the first and rate-limiting step of the kynurenine pathway . This pathway leads to the production of several metabolites, including 3-hydroxy anthranilic acid, which have immunoprotective and immunosuppressive effects .
IDO1 is widely expressed in several lymphoid and non-lymphoid organs, including lymph nodes, spleen, tonsils, lungs, and intestine, as well as immune cells such as macrophages and dendritic cells (DCs), and endothelial cells . The activity of IDO1 is regulated at the transcriptional level, where its expression is increased by transcription factors such as NF-κB, aryl hydrocarbon receptor (AhR), and CCTF. These factors are induced by stimulation with toll-like receptor (TLR) ligands or cytokines and inhibited by hypoxia . Additionally, IDO1 activity can be inhibited at the protein level by peroxynitrite, leading to IDO1 inactivation, or by suppressor of cytokine signaling 3 (SOCS3), which binds to and targets IDO1 for proteasomal degradation .
IDO1 plays a significant role in immune regulation by modulating T-cell behavior through the catabolism of tryptophan. The enzyme’s activity leads to a local reduction in tryptophan availability and an increase in bioactive metabolites such as kynurenine, which mediate immune regulation and immune tolerance . This mechanism is involved in various pathophysiological processes, including antimicrobial and antitumor defense, neuropathology, immunoregulation, and antioxidant activity .
IDO1 has been identified as a promising therapeutic target, particularly in the context of cancer immunotherapy. The enzyme’s ability to create an immunosuppressive microenvironment by depleting tryptophan and producing immunosuppressive metabolites makes it an attractive target for therapeutic intervention . Inhibitors targeting IDO1 are being explored as potential treatments to enhance anti-tumor immunity and improve the efficacy of existing cancer therapies .