Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
Add more filters

Publication year range
1.
Nature ; 566(7745): 548-552, 2019 02.
Article in English | MEDLINE | ID: mdl-30760924

ABSTRACT

Singlet molecular oxygen (1O2) has well-established roles in photosynthetic plants, bacteria and fungi1-3, but not in mammals. Chemically generated 1O2 oxidizes the amino acid tryptophan to precursors of a key metabolite called N-formylkynurenine4, whereas enzymatic oxidation of tryptophan to N-formylkynurenine is catalysed by a family of dioxygenases, including indoleamine 2,3-dioxygenase 15. Under inflammatory conditions, this haem-containing enzyme is expressed in arterial endothelial cells, where it contributes to the regulation of blood pressure6. However, whether indoleamine 2,3-dioxygenase 1 forms 1O2 and whether this contributes to blood pressure control have remained unknown. Here we show that arterial indoleamine 2,3-dioxygenase 1 regulates blood pressure via formation of 1O2. We observed that in the presence of hydrogen peroxide, the enzyme generates 1O2 and that this is associated with the stereoselective oxidation of L-tryptophan to a tricyclic hydroperoxide via a previously unrecognized oxidative activation of the dioxygenase activity. The tryptophan-derived hydroperoxide acts in vivo as a signalling molecule, inducing arterial relaxation and decreasing blood pressure; this activity is dependent on Cys42 of protein kinase G1α. Our findings demonstrate a pathophysiological role for 1O2 in mammals through formation of an amino acid-derived hydroperoxide that regulates vascular tone and blood pressure under inflammatory conditions.


Subject(s)
Blood Pressure/physiology , Inflammation/blood , Inflammation/physiopathology , Singlet Oxygen/metabolism , Vasodilator Agents/metabolism , Animals , Cell Line , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/chemistry , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Cysteine/metabolism , Enzyme Activation/drug effects , Female , Humans , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation/enzymology , Male , Oxidation-Reduction/drug effects , Rats , Signal Transduction , Singlet Oxygen/chemistry , Tryptophan/chemistry , Tryptophan/metabolism
2.
J Biol Chem ; 299(6): 104753, 2023 06.
Article in English | MEDLINE | ID: mdl-37116709

ABSTRACT

Indoleamine-2, 3-dioxygenase (IDO1) and Tryptophan-2, 3-dioxygenase (TDO) catalyze the conversion of L-tryptophan to N-formyl-kynurenine and thus play primary roles in metabolism, inflammation, and tumor immune surveillance. Because their activities depend on their heme contents, which vary in biological settings and go up or down in a dynamic manner, we studied how their heme levels may be impacted by nitric oxide (NO) in mammalian cells. We utilized cells expressing TDO or IDO1 either naturally or via transfection and determined their activities, heme contents, and expression levels as a function of NO exposure. We found NO has a bimodal effect: a narrow range of low NO exposure promoted cells to allocate heme into the heme-free TDO and IDO1 populations and consequently boosted their heme contents and activities 4- to 6-fold, while beyond this range the NO exposure transitioned to have a negative impact on their heme contents and activities. NO did not alter dioxygenase protein expression levels, and its bimodal impact was observed when NO was released by a chemical donor or was generated naturally by immune-stimulated macrophage cells. NO-driven heme allocations to IDO1 and TDO required participation of a GAPDH-heme complex and for IDO1 required chaperone Hsp90 activity. Thus, cells can up- or downregulate their IDO1 and TDO activities through a bimodal control of heme allocation by NO. This mechanism has important biomedical implications and helps explain why the IDO1 and TDO activities in animals go up and down in response to immune stimulation.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase , Nitric Oxide , Tryptophan Oxygenase , Animals , Heme/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mammals/metabolism , Tryptophan/metabolism , Tryptophan Oxygenase/chemistry , Tryptophan Oxygenase/metabolism
3.
Phys Chem Chem Phys ; 26(23): 16747-16764, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38818624

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO) is a tryptophan (Trp) metabolic enzyme along the kynurenine (NFK) pathway. Under pathological conditions, IDO overexpressed by tumor cells causes depletion of tryptophan and the accumulation of metabolic products, which inhibit the local immune response and form immune escape. Therefore, the suppression of IDO activity is one of the strategies for tumor immunotherapy, and drug design for this target has been the focus of research for more than two decades. Apart from IDO, tryptophan dioxygenase (TDO) of the same family can also catalyze the same biochemical reaction in the human body, but it has different tissue distribution and substrate selectivity from IDO. Based on the principle of drug design with high potency and low cross-reactivity to specific targets, in this subject, the activity and selectivity of IDO and TDO toward small molecular inhibitors were studied from the perspective of thermodynamics and kinetics. The aim was to elucidate the structural requirements for achieving favorable biological activity and selectivity of IDO and TDO inhibitors. Specifically, the interactions of inhibitors from eight families with IDO and TDO were initially investigated through molecular docking and molecular dynamics simulations, and the thermodynamic data for binding of inhibitors were predicted by the molecular mechanics/generalized Born surface area (MM/GBSA) method. Secondly, we explored the free energy landscape of JKloops, the kinetic control element of IDO/TDO, using temperature replica exchange molecular dynamics (T-REMD) simulations and elucidated the connection between the rules of IDO/TDO conformational changes and the inhibitor selectivity mechanism. Furthermore, the binding and dissociation processes of the C1 inhibitor (NLG919) were simulated by the adaptive steering molecular dynamics (ASMD) method, which not only addressed the possible stable, metastable, and transition states for C1 inhibitor-IDO/TDO interactions, but also accurately predicted kinetic data for C1 inhibitor binding and dissociation. In conclusion, we have constructed a complete process from enzyme (IDO/TDO) conformational activation to inhibitor binding/dissociation and used the thermodynamic and kinetic data of each link as clues to verify the control mechanism of IDO/TDO on inhibitor selectivity. This is of great significance for us to understand the design principles of tumor immunotherapy drugs and to avoid drug resistance of immunotherapy drugs.


Subject(s)
Enzyme Inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase , Thermodynamics , Tryptophan Oxygenase , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Tryptophan Oxygenase/metabolism , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan Oxygenase/chemistry , Molecular Dynamics Simulation , Molecular Docking Simulation , Kinetics
4.
Proc Natl Acad Sci U S A ; 117(7): 3848-3857, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32024760

ABSTRACT

l-tryptophan (Trp), an essential amino acid for mammals, is the precursor of a wide array of immunomodulatory metabolites produced by the kynurenine and serotonin pathways. The kynurenine pathway is a paramount source of several immunoregulatory metabolites, including l-kynurenine (Kyn), the main product of indoleamine 2,3-dioxygenase 1 (IDO1) that catalyzes the rate-limiting step of the pathway. In the serotonin pathway, the metabolite N-acetylserotonin (NAS) has been shown to possess antioxidant, antiinflammatory, and neuroprotective properties in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). However, little is known about the exact mode of action of the serotonin metabolite and the possible interplay between the 2 Trp metabolic pathways. Prompted by the discovery that NAS neuroprotective effects in EAE are abrogated in mice lacking IDO1 expression, we investigated the NAS mode of action in neuroinflammation. We found that NAS directly binds IDO1 and acts as a positive allosteric modulator (PAM) of the IDO1 enzyme in vitro and in vivo. As a result, increased Kyn will activate the ligand-activated transcription factor aryl hydrocarbon receptor and, consequently, antiinflammatory and immunoregulatory effects. Because NAS also increased IDO1 activity in peripheral blood mononuclear cells of a significant proportion of MS patients, our data may set the basis for the development of IDO1 PAMs as first-in-class drugs in autoimmune/neuroinflammatory diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Allosteric Regulation , Allosteric Site , Animals , Biocatalysis , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Kynurenine/metabolism , Leukocytes, Mononuclear/metabolism , Male , Mice, Knockout , Multiple Sclerosis/enzymology , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Serotonin/analogs & derivatives , Serotonin/chemistry , Serotonin/metabolism , Tryptophan/metabolism
5.
Pharmazie ; 78(1): 2-5, 2023 04 15.
Article in English | MEDLINE | ID: mdl-37138412

ABSTRACT

To explore potential indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors, we designed a series of compounds incorporating urea and 1,2,3-triazole structures. IDO1 enzymatic activity experiments with the synthesized compounds were used to verify their molecular-level activity; for instance, the half maximal inhibitory concentration value of compound 3c was 0.07 µM. Our research has yielded a series of novel IDO1 inhibitors which may be beneficial in the development of drugs targeting IDO1 for cancer treatment.


Subject(s)
Antineoplastic Agents , Neoplasms , Structure-Activity Relationship , Triazoles/pharmacology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy
6.
Acc Chem Res ; 53(9): 1761-1772, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32819102

ABSTRACT

Cancer immunotherapy, which suppresses tumor relapse and metastasis by boosting host immunity and inducing long-term immune memory effects, is emerging as a vital approach to improve the prognosis of patients. Although remarkable efficacy has been observed in some patients, challenges including low response rate, drug resistance, and immune-related adverse effects still limit the clinical application of cancer immunotherapy in broad types of tumors. Immunotherapeutic agents are used to enhance tumor immunogenicity and reverse the effects of the immunosuppressive tumor microenvironment (ITM), but the benefits of monotherapy are mild and transient due to off-target distribution of drugs. To overcome these issues, smart nanosized drug delivery systems (sNDDS) have been developed to enhance tissue specificity, co-deliver multiple drugs, prime immune cells, and amplify immune responses in tumors. Moreover, accumulating knowledge in cancer biology, immunology, and material science has also greatly promoted the development of sNDDS for enhancing cancer immunotherapy.In this Account, we will discuss the approaches of our group in designing sNDDS to induce immunogenic cell death (ICD) for combination with cancer immunotherapy. We propose a brief overview on the design of nanocarriers, intelligent moieties and immunotherapeutic agents in sNDDS. Then, we discuss the strategies to remodel ITM by leveraging ICD as well as cooperating with programmed cell death protein 1 ligand blockade and indoleamine 2,3-dioxygenase 1 inhibition. We have synthesized a series of stimuli-responsive polymers and prodrugs to fabricate sNDDS and have integrated multiple immunotherapeutic drugs into one platform for combinational immunotherapy. Last, we present an outlook on future design of sNDDS and possible directions for enhancing cancer immunotherapy. Building on the concept of enhancing tumor immunogenicity and reversing ITM, we hope this Account will contribute to the rational design of sNDDS for co-delivery of multiple drugs with amplified immunotherapeutic efficacy.


Subject(s)
Drug Carriers/chemistry , Immunogenic Cell Death , Immunotherapy , Nanostructures/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Immune Checkpoint Inhibitors/chemistry , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunogenic Cell Death/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Ligands , Neoplasms/therapy , Polymers/chemistry , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/therapeutic use , Programmed Cell Death 1 Receptor/chemistry , Programmed Cell Death 1 Receptor/metabolism , Tumor Microenvironment
7.
Bioorg Med Chem Lett ; 40: 127963, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33741464

ABSTRACT

Human indoleamine 2,3-dioxygenase 1 (hIDO1) and tryptophan dioxygenase (hTDO) are rate-limiting enzymes in the kynurenine pathway (KP) of l-tryptophan (l-Trp) metabolism and are becoming key drug targets in the combination therapy of checkpoint inhibitors in immunoncology. To discover a selective and potent IDO1 inhibitor, a structure-activity relationship (SAR) study of N-hydroxybenzofuran-5-carboximidamide as a novel scaffold was investigated in a systematic manner. Among the synthesized compounds, the N-3-bromophenyl derivative 19 showed the most potent inhibition, with an IC50 value of 0.44 µM for the enzyme and 1.1 µM in HeLa cells. The molecular modeling of 19 with the X-ray crystal structure of IDO1 indicated that dipole-ionic interactions with heme iron, halogen bonding with Cys129 and the two hydrophobic interactions were important for the high potency of 19.


Subject(s)
Amidines/pharmacology , Benzofurans/pharmacology , Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Oximes/pharmacology , Amidines/chemical synthesis , Amidines/metabolism , Benzofurans/chemical synthesis , Benzofurans/metabolism , Catalytic Domain , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Molecular Docking Simulation , Molecular Structure , Oximes/chemical synthesis , Oximes/metabolism , Protein Binding , Static Electricity , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 49: 128314, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34391891

ABSTRACT

A series of IDO1 inhibitors containing a decahydroquinoline, decahydro-1,6-naphthyridine, or octahydro-1H-pyrrolo[3,2-c]pyridine scaffold were identified with good cellular and human whole blood activity against IDO1. These inhibitors contain multiple chiral centers and all diastereomers were separated. The absolute stereochemistry of each isomers were not determined. Compounds 15 and 27 stood out as leads due to their good cellular as well as human whole blood IDO1 inhibition activity, low unbound clearance, and reasonable mean residence time in rat cassette PK studies.


Subject(s)
Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Naphthyridines/pharmacology , Pyrroles/pharmacology , Quinolines/pharmacology , Animals , Catalytic Domain , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , HeLa Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Molecular Docking Simulation , Naphthyridines/chemical synthesis , Naphthyridines/metabolism , Naphthyridines/pharmacokinetics , Pyrroles/chemical synthesis , Pyrroles/metabolism , Pyrroles/pharmacokinetics , Quinolines/chemical synthesis , Quinolines/metabolism , Quinolines/pharmacokinetics , Rats , Stereoisomerism , Structure-Activity Relationship
9.
J Comput Aided Mol Des ; 35(5): 679-694, 2021 05.
Article in English | MEDLINE | ID: mdl-33905074

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO1) is a heme-containing enzyme that catalyzes the first and rate-limiting step in catabolism of tryptophan via the kynurenine pathway, which plays a pivotal role in the proliferation and differentiation of T cells. IDO1 has been proven to be an attractive target for many diseases, such as breast cancer, lung cancer, colon cancer, prostate cancer, etc. In this study, docking-based virtual screening and bioassays were conducted to identify novel inhibitors of IDO1. The cellular assay demonstrated that 24 compounds exhibited potent inhibitory activity against IDO1 at micromolar level, including 8 compounds with IC50 values below 10 µM and the most potent one (compound 1) with IC50 of 1.18 ± 0.04 µM. Further lead optimization based on similarity searching strategy led to the discovery of compound 28 as an excellent inhibitor with IC50 of 0.27 ± 0.02 µM. Then, the structure-activity relationship of compounds 1, 2, 8 and 14 analogues is discussed. The interaction modes of two compounds against IDO1 were further explored through a Python Based Metal Center Parameter Builder (MCPB.py) molecular dynamics simulation, binding free energy calculation and electrostatic potential analysis. The novel IDO1 inhibitors of compound 1 and its analogues could be considered as promising scaffold for further development of IDO1 inhibitors.


Subject(s)
Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Drug Design , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
10.
Proc Natl Acad Sci U S A ; 115(13): 3249-3254, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29531094

ABSTRACT

For cancer cells to survive and proliferate, they must escape normal immune destruction. One mechanism by which this is accomplished is through immune suppression effected by up-regulation of indoleamine 2,3-dioxygenase (IDO1), a heme enzyme that catalyzes the oxidation of tryptophan to N-formylkynurenine. On deformylation, kynurenine and downstream metabolites suppress T cell function. The importance of this immunosuppressive mechanism has spurred intense interest in the development of clinical IDO1 inhibitors. Herein, we describe the mechanism by which a class of compounds effectively and specifically inhibits IDO1 by targeting its apo-form. We show that the in vitro kinetics of inhibition coincide with an unusually high rate of intrinsic enzyme-heme dissociation, especially in the ferric form. X-ray crystal structures of the inhibitor-enzyme complexes show that heme is displaced from the enzyme and blocked from rebinding by these compounds. The results reveal that apo-IDO1 serves as a unique target for inhibition and that heme lability plays an important role in posttranslational regulation.


Subject(s)
Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Apoproteins/chemistry , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , HeLa Cells , Heme/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inhibitory Concentration 50 , Myoglobin/chemistry
11.
Chembiochem ; 21(11): 1593-1596, 2020 06 02.
Article in English | MEDLINE | ID: mdl-31944493

ABSTRACT

Human indoleamine 2,3-dioxygenase 1 (IDO1) has become an increasingly valuable target for cancer immunotherapy because it promotes immune escape by tumor cells. To date, the function of post-translational modifications (PTMs) on IDO1 has not been fully elucidated. Among the many forms of PTMs, it has been identified that three tyrosine sites (Y15, Y345, and Y353) on IDO1 are nitrated and play important roles in catalytic function. Herein, by genetically encoding 3-nitro-l-tyrosine into the tyrosine nitration sites of IDO1, the homogeneous and native nitrated IDO1 have been obtained. It is found that the nitration of different tyrosine sites has different effects on the IDO1 structure and enzyme activity. Nitration at position Y15 has a negligible effect, but nitration at Y345 or Y353 decreases the enzyme activity, especially Y353. Furthermore, these results demonstrate that the regulation of the catalytic function caused by tyrosine nitration is related to perturbation of the protein structure and heme-binding disruption.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Nitrates/chemistry , Protein Processing, Post-Translational , Tryptophan/chemistry , Tyrosine/analogs & derivatives , Amino Acid Sequence , Binding Sites , Biocatalysis , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kinetics , Models, Molecular , Nitrates/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Substrate Specificity , Tryptophan/metabolism , Tyrosine/chemistry , Tyrosine/metabolism
12.
J Enzyme Inhib Med Chem ; 35(1): 1240-1257, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32466694

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO1) as a key rate-limiting enzyme in the kynurenine pathway of tryptophan metabolism plays an important role in tumour immune escape. Herein, a variety of secondary sulphonamides were synthesised and evaluated in the HeLa cell-based IDO1/kynurenine assay, leading to the identification of new IDO1 inhibitors. Among them, compounds 5d, 5l and 8g exhibited the strongest inhibitory effect with significantly improved activity over the hit compound BS-1. The in vitro results showed that these compounds could restore the T cell proliferation and inhibit the differentiation of naïve CD4+ T cell into highly immunosuppressive FoxP3+ regulatory T (Treg) cell without affecting the viability of HeLa cells and the expression of IDO1 protein. Importantly, the pharmacodynamic assay showed that compound 5d possessed potent antitumour effect in both CT26 and B16F1 tumours bearing immunocompetent mice but not in immunodeficient mice. Functionally, subsequent experiments demonstrated that compound 5d could effectively inhibit tumour cell proliferation, induce apoptosis, up-regulate the expression of IFN-γ and granzyme B, and suppress FoxP3+ Treg cell differentiation, thereby activate the immune system. Thus, compound 5d could be a potential and efficacious agent for further evaluation.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Discovery , Enzyme Inhibitors/chemistry , HeLa Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Male , Mice , Mice, Inbred BALB C , Protein Conformation , Proton Magnetic Resonance Spectroscopy , Spectrometry, Mass, Electrospray Ionization , Sulfonamides/chemistry , T-Lymphocytes/drug effects
13.
Molecules ; 25(6)2020 Mar 23.
Article in English | MEDLINE | ID: mdl-32210078

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO1) is a heme-containing intracellular enzyme that catalyzes the first and rate-determining step of tryptophan metabolism and is an important immunotherapeutic target for the treatment of cancer. In this study, we designed and synthesized a new series of compounds as potential IDO1 inhibitors. These compounds were then evaluated for inhibitory activity against IDO1 and tryptophan 2,3-dioxygenase (TDO). Among them, the three phenyl urea derivatives i12, i23, i24 as showed potent IDO1 inhibition, with IC50 values of 0.1-0.6 µM and no compound exhibited TDO inhibitory activity. Using molecular docking, we predicted the binding mode of compound i12 within IDO1. Compound i12 was further investigated by determining its in vivo pharmacokinetic profile and anti-tumor efficacy. The pharmacokinetic study revealed that compound i12 had satisfactory properties in mice, with moderate plasma clearance (22.45 mL/min/kg), acceptable half-life (11.2 h) and high oral bioavailability (87.4%). Compound i12 orally administered at 15 mg/kg daily showed tumor growth inhibition (TGI) of 40.5% in a B16F10 subcutaneous xenograft model and 30 mg/kg daily showed TGI of 34.3% in a PAN02 subcutaneous xenograft model. In addition, the body weight of i12-treated mice showed no obvious reduction compared with the control group. Overall, compound i12 is a potent lead compound for developing IDO1 inhibitors and anti-tumor agents.


Subject(s)
Drug Design , Enzyme Inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase , Melanoma, Experimental , Neoplasm Proteins , Phenylurea Compounds , Animals , Cell Line, Tumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Melanoma, Experimental/drug therapy , Melanoma, Experimental/enzymology , Melanoma, Experimental/pathology , Mice , Molecular Docking Simulation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/pharmacology , Structure-Activity Relationship
14.
Biophys J ; 117(4): 706-716, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31405517

ABSTRACT

Human indoleamine 2,3-dioxygenase 1 (IDO) is a heme enzyme that catalyzes the first reaction of the main metabolic pathway of L-tryptophan (Trp) to produce N-formylkynurenin. The reaction involves cleavage of the C2=C3 bond in the Trp indole ring and insertion of two atomic oxygens from the iron-bound O2 into the indole 2 and 3 position. For establishment of the chemical mechanism of this unique enzymatic reaction, it is necessary to determine the conformation and electronic state of the substrate Trp bound to IDO. In this study, we measured the ultraviolet resonance Raman spectra of IDO in the presence of Trp to detect the vibrational modes of the substrate Trp. We compared the ultraviolet resonace Raman spectra of Trp in a ternary complex (Trp-bound cyanide enzyme) and a binary complex (Trp-bound reduced enzyme) of IDO with that of free Trp in solution and found that binding to IDO influences the conformation of Trp, resulting in similar changes in the two complexes, especially around the C3-Cß bond. However, the presence of the diatomic ligand at the heme sixth coordination site in the ternary complex significantly alters the mobility and electronic structure of Trp, most likely resulting in the C2=C3 bond cleavage in the enzymatic reaction.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Tryptophan/chemistry , Heme/chemistry , Heme/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Oxygen/chemistry , Oxygen/metabolism , Protein Binding , Protein Conformation , Spectrum Analysis, Raman , Tryptophan/metabolism
15.
Biochemistry ; 58(7): 974-986, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30585477

ABSTRACT

The heme enzyme indoleamine 2,3-dioxygenase-1 (IDO1) catalyzes the first reaction of l-tryptophan oxidation along the kynurenine pathway. IDO1 is a central immunoregulatory enzyme with important implications for inflammation, infectious disease, autoimmune disorders, and cancer. Here we demonstrate that IDO1 is a mammalian nitrite reductase capable of chemically reducing nitrite to nitric oxide (NO) under hypoxia. Ultraviolet-visible absorption and resonance Raman spectroscopy showed that incubation of dithionite-reduced, ferrous-IDO1 protein (FeII-IDO1) with nitrite under anaerobic conditions resulted in the time-dependent formation of an FeII-nitrosyl IDO1 species, which was inhibited by substrate l-tryptophan, dependent on the concentration of nitrite or IDO1, and independent of the concentration of the reductant, dithionite. The bimolecular rate constant for IDO1 nitrite reductase activity was determined as 5.4 M-1 s-1 (pH 7.4, 23 °C), which was comparable to that measured for myoglobin (3.6 M-1 s-1; pH 7.4, 23 °C), an efficient and biologically important mammalian heme-based nitrite reductase. IDO1 nitrite reductase activity was pH-dependent but differed with myoglobin in that it showed a reduced proton dependency at pH >7. Electron paramagnetic resonance studies measuring NO production showed that the conventional IDO1 dioxygenase reducing cofactors, ascorbate and methylene blue, enhanced IDO1's nitrite reductase activity and the time- and IDO1 concentration-dependent release of NO in a manner inhibited by l-tryptophan or the IDO inhibitor 1-methyl-l-tryptophan. These data identify IDO1 as an efficient mammalian nitrite reductase that is capable of generating NO under anaerobic conditions. IDO1's nitrite reductase activity may have important implications for the enzyme's biological actions when expressed within hypoxic tissues.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Nitrite Reductases/metabolism , Anaerobiosis , Electron Spin Resonance Spectroscopy , Heme/chemistry , Heme/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Nitrite Reductases/chemistry , Nitrites/chemistry , Nitrites/metabolism , Protons , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrophotometry, Ultraviolet , Spectrum Analysis, Raman
16.
J Am Chem Soc ; 141(38): 15288-15300, 2019 09 25.
Article in English | MEDLINE | ID: mdl-31436417

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO1) is a heme enzyme that catalyzes the oxygenation of the indole ring of tryptophan to afford N-formylkynurenine. This activity significantly suppresses the immune response, mediating inflammation and autoimmune reactions. These consequential effects are regulated through redox changes in the heme cofactor of IDO1, which autoxidizes to the inactive ferric state during turnover. This change in redox status increases the lability of the heme cofactor leading to further suppression of activity. The cell can thus regulate IDO1 activity through the supply of heme and reducing agents. We show here that polysulfides bind to inactive ferric IDO1 and reduce it to the oxygen-binding ferrous state, thus activating IDO1 to maximal turnover even at low, physiologically significant concentrations. The on-rate for hydrogen disulfide binding to ferric IDO1 was found to be >106 M-1 s-1 at pH 7 using stopped-flow spectrometry. Fe K-edge XANES and EPR spectroscopy indicated initial formation of a low-spin ferric sulfur-bound species followed by reduction to the ferrous state. The µM affinity of polysulfides for IDO1 implicates these polysulfides as important signaling factors in immune regulation through the kynurenine pathway. Tryptophan significantly enhanced the relatively lower-affinity binding of hydrogen sulfide to IDO1, inspiring the use of the small molecule 3-mercaptoindole (3MI), which selectively binds to and activates ferric IDO1. 3MI sustains turnover by catalytically transferring reducing equivalents from glutathione to IDO1, representing a novel strategy of upregulating innate immunosuppression for treatment of autoimmune disorders. Reactive sulfur species are thus likely unrecognized immune-mediators with potential as therapeutic agents through these interactions with IDO1.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Sulfides/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Molecular Structure , Sulfides/chemistry
17.
J Am Chem Soc ; 141(47): 18771-18779, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31682426

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (hIDO1) and tryptophan dioxygenase (hTDO) are two of the only three heme-based dioxygenases in humans. They have recently been identified as key cancer immunotherapeutic drug targets. While structures of hIDO1 in complex with inhibitors have been documented, so far there are no structures of hTDO-inhibitor complexes available. Here we use PF-06840003 (IPD), a hIDO1-selective inhibitor in clinical trials, as a structural probe to elucidate inhibitor-selectivity in hIDO1 versus hTDO. Spectroscopic studies show that IPD exhibits 400-fold higher inhibition activity toward hIDO1 with respect to hTDO. Crystallographic structures reveal that the binding pocket of IPD in the active site in hIDO1 is much more flexible as compared to that in hTDO, which offers a molecular explanation for the superior inhibition activity of IPD in hIDO1 with respect to hTDO. In addition to the IPD bound in the active site, a second IPD molecule was identified in an inhibitory site on the proximal side of the heme in hIDO1 and in an exosite that is ∼40 Šaway from the active site in hTDO. Taken together the data provide new insights into structure-based design of mono and dual inhibitors targeting hIDO1 and/or hTDO.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Tryptophan Oxygenase/antagonists & inhibitors , Crystallography, X-Ray , Enzyme Inhibitors/metabolism , Heme/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Models, Molecular , Protein Domains , Substrate Specificity , Tryptophan Oxygenase/chemistry , Tryptophan Oxygenase/metabolism
18.
Nucleic Acids Res ; 45(W1): W253-W259, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28498966

ABSTRACT

Discovery of potentially deleterious sequence variants is important and has wide implications for research and generation of new hypotheses in human and veterinary medicine, and drug discovery. The GenProBiS web server maps sequence variants to protein structures from the Protein Data Bank (PDB), and further to protein-protein, protein-nucleic acid, protein-compound, and protein-metal ion binding sites. The concept of a protein-compound binding site is understood in the broadest sense, which includes glycosylation and other post-translational modification sites. Binding sites were defined by local structural comparisons of whole protein structures using the Protein Binding Sites (ProBiS) algorithm and transposition of ligands from the similar binding sites found to the query protein using the ProBiS-ligands approach with new improvements introduced in GenProBiS. Binding site surfaces were generated as three-dimensional grids encompassing the space occupied by predicted ligands. The server allows intuitive visual exploration of comprehensively mapped variants, such as human somatic mis-sense mutations related to cancer and non-synonymous single nucleotide polymorphisms from 21 species, within the predicted binding sites regions for about 80 000 PDB protein structures using fast WebGL graphics. The GenProBiS web server is open and free to all users at http://genprobis.insilab.org.


Subject(s)
Genetic Variation , Proteins/chemistry , Proteins/genetics , Software , Binding Sites , Brain Neoplasms/genetics , Databases, Protein , Enzyme Inhibitors/chemistry , Genes, p53 , Genome-Wide Association Study , Glioblastoma/genetics , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Internet , Ligands , Mutation, Missense , Polymorphism, Single Nucleotide , Proteins/metabolism
19.
Int J Mol Sci ; 20(13)2019 Jun 28.
Article in English | MEDLINE | ID: mdl-31261636

ABSTRACT

Protein-ligand docking is a widely used method to generate solutions for the binding of a small molecule with its target in a short amount of time. However, these methods provide identification of physically sound protein-ligand complexes without a complete view of the binding process dynamics, which has been recognized to be a major discriminant in binding affinity and ligand selectivity. In this paper, a novel piece of open-source software to approach this problem is presented, called GPathFinder. It is built as an extension of the modular GaudiMM platform and is able to simulate ligand diffusion pathways at atomistic level. The method has been benchmarked on a set of 20 systems whose ligand-binding routes were studied by other computational tools or suggested from experimental "snapshots". In all of this set, GPathFinder identifies those channels that were already reported in the literature. Interestingly, the low-energy pathways in some cases indicate novel possible binding routes. To show the usefulness of GPathFinder, the analysis of three case systems is reported. We believe that GPathFinder is a software solution with a good balance between accuracy and computational cost, and represents a step forward in extending protein-ligand docking capacities, with implications in several fields such as drug or enzyme design.


Subject(s)
Molecular Docking Simulation/methods , Software , Algorithms , Aquaporins/chemistry , Aquaporins/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Binding Sites , Cytochrome P-450 CYP2C19/chemistry , Cytochrome P-450 CYP2C19/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Ligands , Protein Binding
20.
Molecules ; 24(10)2019 May 15.
Article in English | MEDLINE | ID: mdl-31096672

ABSTRACT

IDO1, a key dioxygenase in tryptophan-kynurenine metabolism, appeared in the last 10 years at the vanguard of druggable targets in cancer therapy due to its well-established role both in immune escape and inflammatory neovascularization. Among the pool of IDO1 inhibitors that have entered clinical trials, none have reached approval. The identification of novel inhibitors endowed with better clinical profile, together with the further comprehension of the interactions with residues in IDO1 active site, are still a need. In this context, we have synthesized a novel class of imidazothiazole derivatives as IDO1 inhibitors and identified three compounds with inhibitory potency in the low micromolar range. This report strengthens the role played by pocket C in the active site of IDO1, providing novel directions in the design of IDO1 inhibitors.


Subject(s)
Imidazoles/chemical synthesis , Imidazoles/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Molecular Docking Simulation , Thiazoles/chemical synthesis , Thiazoles/pharmacology , Binding Sites , Catalytic Domain , Click Chemistry , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/chemistry , Molecular Conformation , Molecular Dynamics Simulation , Molecular Structure , Protein Binding , Structure-Activity Relationship , Thiazoles/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL