TDO2 Human

Tryptophan 2,3-Dioxygenase Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure

TDO2 exists as a homotetramer (190 kDa) composed of four identical 48 kDa monomers . Each monomer contains a heme group, essential for catalytic activity, and adopts an all-α-helical structure . Crystallographic studies of bacterial homologs (e.g., Xanthomonas campestris) reveal a substrate-binding loop that facilitates L-tryptophan interaction, while structures of human TDO2 (hTDO) complexed with substrates and inhibitors have elucidated oxygen binding and catalytic mechanisms .

Catalytic Mechanism

TDO2 oxidizes L-tryptophan via a dioxygenation reaction:

L-tryptophan+O2N-formyl-L-kynurenine\text{L-tryptophan} + \text{O}_2 \rightarrow \text{N-formyl-L-kynurenine}

Tissue Distribution

TDO2 is predominantly expressed in the liver and brain, with lower levels in peripheral tissues . Its expression in the liver regulates systemic tryptophan levels, while brain-specific activity influences neurotransmitter metabolism (e.g., serotonin) .

TissueExpression LevelKey Functions
LiverHighTryptophan homeostasis, kynurenine production, detoxification
BrainModerateNeurotransmitter regulation (serotonin), neuroinflammation modulation
Immune CellsLowMinimal role under basal conditions; upregulated in pathological states

Kynurenine Pathway Significance

TDO2-mediated tryptophan catabolism generates metabolites with diverse roles:

  • Neuroactive Compounds: Kynurenine, quinolinic acid, and 3-hydroxykynurenine modulate neurotransmission and neuroinflammation .

  • Immune Regulation: Downstream metabolites (e.g., kynurenine) suppress T-cell proliferation and promote immune tolerance .

Cancer Prognosis

TDO2 overexpression correlates with poor outcomes in malignant tumors, including hepatocellular carcinoma (HCC), colorectal cancer, and melanoma. Meta-analyses reveal:

Mechanisms in Hepatocellular Carcinoma

TDO2 promotes epithelial-to-mesenchymal transition (EMT) by activating the kynurenine-aryl hydrocarbon receptor (AhR) pathway, enhancing metastasis and invasion . Preclinical studies demonstrate that TDO2 inhibition (e.g., 680C91) reduces migration and invasion in HCC cell lines (P < 0.01) .

Neuropsychiatric Disorders

Genetic variants in TDO2 are linked to schizophrenia, depression, and autism:

DisorderGenetic AssociationFunctional Impact
SchizophreniaSNPs in intron 6 disrupting YY-1 binding sites Altered tryptophan metabolism, dopamine signaling
AutismAggregation in AGRE cohort (Nabi et al., 2004) Impaired synaptic plasticity, social behavior
DepressionPolymorphisms associated with reduced enzymatic activitySerotonin depletion, neuroinflammation

Small-Molecule Inhibitors

TDO2 is a priority target in oncology and neurology. Notable inhibitors include:

InhibitorTargetIC50 (TDO2)Applications
680C91TDO2~10 μMHCC metastasis suppression
EpacadostatIDO1/TDO2Dual inhibitionCancer immunotherapy (clinical trials)
PF-06840018TDO2<100 nMPreclinical glioma models

Animal Models

  • TDO2 Knockout Mice: Reduced anxiety-like behavior, enhanced cognitive performance, and elevated brain serotonin levels .

  • HCC Xenografts: TDO2 inhibition with 680C91 reduces tumor growth and metastasis (P < 0.01) .

Biomarker Potential

TDO2 expression correlates with:

  • Cancer Staging: Higher levels in advanced TNM stages (HR = 0.65, P = 0.002) .

  • Immune Evasion: Upregulation in tumors with high PD-L1 expression, suggesting combinatorial immunotherapy potential .

Future Directions

  1. Precision Oncology: Combining TDO2 inhibitors with checkpoint inhibitors to enhance anti-tumor immunity.

  2. Neurodegenerative Diseases: Exploring TDO2 modulation to balance kynurenine pathway metabolites in Alzheimer’s and Parkinson’s.

  3. Diagnostic Biomarkers: Validating TDO2 as a prognostic marker in clinical trials.

Product Specs

Introduction
TDO2, a ferrous heme enzyme, catalyzes the initial and rate-limiting step in the kynurenine pathway, the primary metabolic route for tryptophan. This enzyme incorporates oxygen into the indole ring of tryptophan. TDO2 exhibits broad substrate specificity, acting on tryptamine and its derivatives, including D- and L-tryptophan, 5-hydroxytryptophan, and serotonin.
Description
Recombinant human TDO2, expressed in E. coli, is a monomeric, non-glycosylated polypeptide chain. This protein, with a molecular weight of 50 kDa, consists of 426 amino acids (residues 1-406) and includes an N-terminal 20 amino acid His-tag. Purification of TDO2 is achieved using proprietary chromatographic methods.
Physical Appearance
A sterile, colorless solution, free of particulates.
Formulation
The TDO2 solution is supplied at a concentration of 0.25 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 0.2 M NaCl, 5 mM DTT, 1 mM EDTA, and 30% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freezing and thawing should be avoided.
Purity
Purity is determined to be greater than 90.0% using SDS-PAGE analysis.
Synonyms
Tryptophan 2,3-dioxygenase, TDO, Tryptamin 2,3-dioxygenase, Tryptophan oxygenase, TO, TRPO, Tryptophan pyrrolase, Tryptophanase, TDO2, TPH2.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSGCPFLGNN FGYTFKKLPV EGSEEDKSQT GVNRASKGGL IYGNYLHLEK VLNAQELQSE TKGNKIHDEH LFIITHQAYE LWFKQILWEL DSVREIFQNG HVRDERNMLK VVSRMHRVSV ILKLLVQQFS ILETMTALDF NDFREYLSPA SGFQSLQFRL LENKIGVLQN MRVPYNRRHY RDNFKGEENE LLLKSEQEKT LLELVEAWLE RTPGLEPHGF NFWGKLEKNI TRGLEEEFIR IQAKEESEEK EEQVAEFQKQ KEVLLSLFDE KRHEHLLSKG ERRLSYRALQ GALMIYFYRE EPRFQVPFQL LTSLMDIDSL MTKWRYNHVC MVHRMLGSKA GTGGSSGYHY LRSTVSDRYK VFVDLFNLST YLIPRHWIPK MNPTIHKFLY TAEYCDSSYF SSDESD.

Q&A

What is TDO2 and what is its primary function in human biology?

TDO2 (Tryptophan 2,3-dioxygenase) is a rate-limiting enzyme in the kynurenine pathway that catalyzes the conversion of L-tryptophan to N-formyl-kynurenine. Unlike its counterpart IDO1 (indoleamine-2,3-dioxygenase), TDO2 exists as a tetrameric structure with two independent TDO2 molecules contributing to each of its four active sites, while IDO1 is monomeric . TDO2 plays essential roles in tryptophan metabolism, inflammation regulation, and tumor immune surveillance. The enzyme requires heme for substrate binding and catalysis, though it can exist in both heme-containing (holo) and heme-free (apo) states, with the balance between these forms varying according to biological conditions . TDO2 expression is predominantly high in the liver, which serves as the major metabolic location of tryptophan, but is also found in various other tissues including the brain .

How does TDO2 expression vary across normal human tissues?

TDO2 expression demonstrates distinctive patterns across human tissues. Analysis of tissue databases reveals that TDO2 expression is generally deficient across most normal tissues, with notable exceptions being the liver and pituitary, where expression is markedly elevated . This tissue-specific expression pattern correlates with TDO2's function in tryptophan metabolism, as the liver serves as the primary site for tryptophan degradation. The differential expression across tissues suggests specialized roles in different organ systems, potentially related to local regulation of tryptophan levels and downstream kynurenine pathway metabolites. When examining expression patterns, researchers should consider using combined data from multiple databases (such as TCGA and GTEx) to achieve more comprehensive tissue coverage for accurate assessment of normal TDO2 expression profiles .

What experimental models are available for studying TDO2 function?

Multiple experimental models have been developed to study TDO2 function, with TDO2 knockout (KO) mice being among the most extensively characterized. These knockout models have been generated using gene-targeting techniques, such as those employing the R1 ES cell line, with subsequent backcrossing to control genetic background (e.g., C57BL/6Cr mice for at least 8 generations) . Cell line models expressing various levels of TDO2 are also available, as TDO2 expression has been documented across numerous cancer cell lines with varying expression levels . For structural and biochemical studies, recombinant TDO2 protein has been used to determine enzyme kinetics and for crystallography studies, particularly in developing and testing TDO2 inhibitors . Researchers should carefully select models based on their specific research questions, considering factors such as species differences, genetic background effects, and whether cellular or organismal contexts are most appropriate for their investigation.

How do apo-TDO2 and holo-TDO2 forms differ in their regulatory mechanisms?

The distinction between apo-TDO2 (heme-free) and holo-TDO2 (heme-containing) forms represents a complex regulatory mechanism controlling enzyme activity. These forms exist in a dynamic equilibrium that responds to various biological signals, with nitric oxide (NO) levels being particularly significant in this regulation . Historical studies from approximately fifty years ago demonstrated that healthy rat liver TDO2 typically shows low (25-50%) heme saturation, which can rapidly increase to 80-100% saturation within 2 hours under certain physiological conditions . The apo form presents distinct advantages as a drug target compared to the holo form, including potentially different binding pockets and regulatory mechanisms. Recent crystallographic analysis of inhibitor binding to apo-TDO2 reveals that small molecules can interact with the large, hydrophobic heme binding pocket within the active site . Researchers investigating TDO2 regulation should account for the dynamic interconversion between these forms and consider how experimental conditions might alter this balance, potentially affecting experimental outcomes and interpretations.

What is the relationship between TDO2 and immune checkpoint genes in cancer?

TDO2 demonstrates significant associations with various immune checkpoint genes across multiple cancer types. Correlation analyses from pancancer studies have identified particularly strong relationships between TDO2 expression and immune checkpoint-related gene markers including LAIR1, CD276, NRP1, CD80, and CD86 . These correlations suggest that TDO2 may participate in tumor immune evasion mechanisms that involve checkpoint regulation. Mechanistically, overexpression of TDO2 can activate the aryl hydrocarbon receptor (AhR) pathway in immune cells, contributing to immune escape . This relationship has significant therapeutic implications, as treatment with TDO2 inhibitors has been shown to enhance dendritic cell function and improve T-cell-mediated immune responses, potentially reducing tumor metastasis in experimental models . Researchers investigating cancer immunotherapy approaches should consider the interconnected nature of TDO2 and immune checkpoint pathways when designing intervention strategies or interpreting resistance mechanisms to existing immunotherapies.

How does TDO2 interact with DNA repair mechanisms and genomic stability?

TDO2 expression demonstrates significant correlations with DNA mismatch repair (MMR) gene mutation rates across multiple cancer types. Pancancer analysis has revealed that TDO2 expression is closely associated with mutation rates in five critical MMR genes . Since MMR deficiency can lead to genomic errors and microsatellite instability (MSI), this relationship suggests TDO2 may influence tumorigenesis through pathways affecting genomic stability. Additionally, TDO2 expression correlates with tumor mutational burden (TMB) and DNA methyltransferase (DNMT) activity in various cancers . The correlation with DNMTs is particularly noteworthy, as it indicates DNA methylation may play a role in regulating TDO2 expression or that TDO2-related metabolic changes might influence epigenetic programming. These findings suggest TDO2 operates within a broader network of genome maintenance mechanisms, potentially contributing to tumor development through effects on mutational processes or epigenetic regulation. When investigating TDO2 in cancer contexts, researchers should consider assessing MMR status, TMB, and DNA methylation patterns to fully characterize potential mechanisms.

What mechanisms explain TDO2's contribution to tumor immune evasion?

TDO2 contributes to tumor immune evasion through several interconnected mechanisms centered on tryptophan metabolism and kynurenine pathway activation. By catalyzing tryptophan degradation, TDO2 creates a tryptophan-depleted microenvironment that can suppress T-cell proliferation and induce T-cell apoptosis, thereby altering immune responses . Simultaneously, the production of kynurenine activates the aryl hydrocarbon receptor (AhR) in immune cells, which has been confirmed to mediate tumoral immune resistance . This activation alters immune cell function and contributes to immunosuppression. Pancancer analyses have demonstrated that TDO2 expression levels are significantly associated with immune cell infiltration patterns across various tumors, particularly with dendritic cell infiltration . Treatment with TDO2 inhibitors has been shown to enhance dendritic cell function and improve T-cell-mediated immune responses, potentially reducing tumor metastasis in experimental models . When investigating TDO2-related immune evasion, researchers should assess multiple aspects of the tumor immune microenvironment, including tryptophan and kynurenine levels, immune cell infiltration patterns, and AhR pathway activation to comprehensively understand the mechanisms involved.

How can TDO2 inhibition strategies be optimized for cancer therapy?

Optimizing TDO2 inhibition requires consideration of the enzyme's structural states and inhibitor binding mechanisms. Recent advances have identified the first apo-TDO2 binding inhibitors that demonstrate inhibition of cellular TDO2 activity at low nanomolar concentrations . The crystal structure of a potent small molecule inhibitor bound to apo-TDO2 has revealed detailed binding interactions within the large, hydrophobic heme binding pocket of the active site . This structural information provides a foundation for structure-guided drug design. When developing inhibition strategies, researchers should consider targeting both apo and holo forms, as the enzyme exists in a dynamic equilibrium between these states in cellular environments. Combination approaches targeting TDO2 alongside immune checkpoint inhibitors may offer synergistic benefits, given the established correlation between TDO2 and immune checkpoint genes like LAIR1, CD276, NRP1, CD80, and CD86 . Experimental studies have already demonstrated that TDO2 inhibitors can enhance dendritic cell function and improve T-cell-mediated immune responses . Future optimization should explore tissue-specific delivery methods, biomarkers for patient selection (potentially including MMR status, TMB, or DNMT expression), and combination strategies that address potential resistance mechanisms.

How is TDO2 implicated in psychiatric disorders based on genetic and molecular evidence?

TDO2 has been implicated in various psychiatric disorders through both genetic association studies and molecular pathway analyses. Single nucleotide polymorphisms (SNPs) in the TDO2 gene have been associated with psychiatric conditions including schizophrenia, depression, and attention deficit hyperactivity disorder . Mechanistically, TDO2's role in tryptophan metabolism positions it at a critical junction for serotonin availability, as it diverts tryptophan away from serotonin synthesis and toward the kynurenine pathway. TDO2 knockout mice show increased levels of tryptophan and serotonin in the hippocampus and midbrain, supporting this mechanistic link . Furthermore, TDO2 expression is regulated by stress hormones and immune stimulation, with corticosteroids known to enhance TDO2 gene expression, and immune system activation by lipopolysaccharide (LPS) or polyinosinic-polycytidylic acid (pI:C) increasing TDO2 mRNA expression . Intriguingly, the TDO2 gene has highly selective expression in the dentate gyrus of the hippocampus, and its expression is dramatically reduced in the "immature dentate gyrus" (iDG) phenotype, which has been observed postmortem in brains of patients with schizophrenia and bipolar disorder . When investigating TDO2's role in psychiatric disorders, researchers should consider integrating genetic, neurochemical, and behavioral approaches, and examine interactions with environmental stressors and immune activation.

What neurochemical changes result from TDO2 modulation in brain tissue?

TDO2 modulation produces significant neurochemical changes in brain tissue, primarily affecting tryptophan and serotonin metabolism. In TDO2 knockout mice, concentrations of tryptophan and serotonin (5-HT) in the hippocampus and midbrain are significantly elevated compared to wild-type mice . This neurochemical alteration results from reduced tryptophan catabolism through the kynurenine pathway, allowing more tryptophan to be available for serotonin synthesis. Beyond direct effects on serotonin, TDO2 modulation likely influences levels of kynurenine pathway metabolites, including kynurenic acid (a glutamate receptor antagonist) and quinolinic acid (an NMDA receptor agonist), which have neuroactive properties that can affect glutamatergic neurotransmission . These neurochemical changes may underlie the behavioral phenotypes observed in TDO2 knockout models, including reduced anxiety-like behavior and enhanced exploratory activity. When studying neurochemical effects of TDO2 modulation, researchers should employ techniques that can measure multiple neurotransmitters and metabolites simultaneously (such as high-performance liquid chromatography or mass spectrometry) and examine region-specific changes, particularly in areas with high TDO2 expression like the dentate gyrus of the hippocampus or regions involved in emotional and cognitive processing.

What are the most effective approaches for measuring TDO2 activity in biological samples?

Measuring TDO2 activity in biological samples requires consideration of multiple technical approaches, each with specific advantages. For enzyme kinetic studies, spectrophotometric assays that monitor the formation of N-formylkynurenine (NFK) at 321 nm provide direct measurement of TDO2 catalytic activity. When working with complex biological samples, high-performance liquid chromatography (HPLC) or liquid chromatography-mass spectrometry (LC-MS) methods offer higher sensitivity and specificity by quantifying tryptophan depletion and kynurenine formation. For cellular models, researchers can implement dual approaches measuring both tryptophan consumption and kynurenine production in culture media. Additionally, isotope-labeled tryptophan can be used to track metabolic flux through the kynurenine pathway with high precision. When assessing the ratio of apo to holo forms, researchers should consider spectroscopic techniques that can distinguish heme-bound from heme-free protein. It's essential to control for assay conditions like pH, temperature, and cofactor availability (particularly heme concentration), as these significantly affect enzyme activity. Researchers should also be aware that nitric oxide levels can influence the apo/holo equilibrium, potentially affecting activity measurements in biological systems .

How can researchers effectively evaluate the role of TDO2 in tumor immune microenvironment?

Evaluating TDO2's role in the tumor immune microenvironment requires integrated approaches spanning multiple analytical platforms. Flow cytometry and single-cell RNA sequencing enable characterization of immune cell populations and their functional states in relation to TDO2 expression. Multiplex immunohistochemistry or immunofluorescence can reveal spatial relationships between TDO2-expressing cells and immune infiltrates within the tumor microstructure. Metabolomic analysis of tryptophan and kynurenine levels in tumor tissue and surrounding microenvironment provides direct evidence of TDO2 activity impact. For mechanistic studies, co-culture systems combining TDO2-expressing tumor cells with various immune cell populations can reveal direct effects on immune cell function. The implementation of TDO2 inhibitors in these systems serves as valuable functional validation. Researchers should also assess correlations with immune checkpoint molecules, particularly LAIR1, CD276, NRP1, CD80, and CD86, which show significant associations with TDO2 expression . When conducting in vivo studies, immune-competent mouse models are essential, as they preserve the complex interactions between tumor cells, stromal components, and immune elements that may be influenced by TDO2 activity.

What are the most promising strategies for developing next-generation TDO2 inhibitors?

Development of next-generation TDO2 inhibitors should capitalize on recent structural insights and target state-specific inhibition mechanisms. The recent discovery of apo-TDO2 binding inhibitors with nanomolar potency represents a significant advance, as these compounds target the enzyme's heme-free state . Future inhibitor development should leverage the crystal structure data showing detailed binding interactions within the large, hydrophobic heme binding pocket of the active site . Dual-state inhibitors targeting both apo and holo forms could potentially offer more comprehensive enzyme inhibition across varying physiological conditions, considering the dynamic equilibrium between these states in cellular environments. Structure-based drug design approaches should focus on optimizing binding interactions while addressing pharmacokinetic properties to ensure adequate tissue distribution, particularly for targeting central nervous system applications. Novel delivery approaches, including antibody-drug conjugates or nanoparticle formulations, could enable more selective targeting of TDO2 in specific tissue contexts. Additionally, researchers should explore the development of bifunctional molecules that simultaneously inhibit TDO2 and modulate complementary immune checkpoints, given the established correlations between TDO2 and immune checkpoint genes . Comprehensive preclinical evaluation should include assessment of effects on immune cell function, particularly dendritic cells and T cells, which have shown sensitivity to TDO2 inhibition .

How might TDO2 function as a biomarker in personalized cancer treatment approaches?

TDO2 shows considerable promise as a biomarker for personalized cancer treatment, particularly in guiding immunotherapy decisions. Pancancer analysis has established significant correlations between TDO2 expression and prognosis in multiple cancer types, with high expression associated with poor outcomes in kidney renal papillary cell carcinoma (KIRP), brain lower grade glioma (LGG), testicular germ cell tumors (TGCT), and uveal melanoma (UVM) . For clinical implementation, immunohistochemical assessment of TDO2 protein expression in tumor biopsies could provide a straightforward approach for patient stratification. Gene expression profiling of TDO2 alongside immune checkpoint molecules (particularly LAIR1, CD276, NRP1, CD80, and CD86) might identify patients likely to benefit from combined TDO2 and immune checkpoint inhibition . Additionally, liquid biopsy approaches measuring circulating tumor DNA methylation patterns affecting TDO2 could offer less invasive monitoring capabilities. Metabolomic profiling of kynurenine pathway metabolites in patient serum might serve as functional biomarkers of TDO2 activity. Future clinical trials involving TDO2-targeting agents should incorporate biomarker analyses correlating TDO2 expression, mismatch repair status, tumor mutational burden, and microsatellite instability with treatment response . This would enable identification of patient subgroups most likely to benefit from TDO2-targeted interventions and guide combination therapy strategies.

Product Science Overview

Introduction

Tryptophan 2,3-dioxygenase (TDO2) is a heme-containing enzyme that plays a crucial role in the metabolism of the essential amino acid tryptophan. This enzyme catalyzes the first and rate-limiting step in the kynurenine pathway, which is the oxidative cleavage of the indole ring of tryptophan to form N-formylkynurenine .

Gene and Protein Structure

The TDO2 gene is located on chromosome 4q32.1 in humans . The gene encodes a protein that forms a homo-tetrameric structure, with each monomer having a molecular weight of approximately 48 kDa . The active enzyme complex is around 190 kDa in size . The enzyme contains a heme prosthetic group, which is essential for its catalytic activity .

Function and Mechanism

TDO2 is primarily expressed in the liver, but it is also found in other tissues such as the brain and placenta . The enzyme’s primary function is to regulate systemic tryptophan levels by catalyzing its degradation along the kynurenine pathway . This pathway is significant for the production of several bioactive metabolites, including kynurenine, which can be further metabolized into nicotinamide adenine dinucleotide (NAD+), an essential coenzyme in cellular metabolism .

The reaction catalyzed by TDO2 involves the incorporation of molecular oxygen into tryptophan, resulting in the formation of N-formylkynurenine. This reaction is the first and rate-limiting step in the kynurenine pathway .

Clinical Significance

TDO2 has been implicated in various physiological and pathological processes. Increased activity of TDO2 and subsequent production of kynurenine have been associated with immune suppression in cancer . This is because kynurenine and its metabolites can modulate immune responses, potentially aiding tumor cells in evading immune surveillance .

Additionally, single nucleotide polymorphisms (SNPs) in the TDO2 gene have been linked to disorders such as autism and hypertryptophanemia . The enzyme’s role in tryptophan metabolism also makes it a potential target for therapeutic interventions in diseases where tryptophan catabolism is dysregulated .

Recombinant TDO2

Recombinant human TDO2 is produced using genetic engineering techniques, where the TDO2 gene is cloned and expressed in suitable host cells. This allows for the production of large quantities of the enzyme for research and therapeutic purposes . Recombinant TDO2 retains the same structural and functional properties as the native enzyme, making it a valuable tool for studying tryptophan metabolism and developing potential therapeutic agents .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.