1
|
Wu C, Li J, Jia H, Zhao J, Qin M, Shi H, Liu C, Lin J, Cai M, Gu Y, Liu B, Gao L. Indoleamine 2,3-dioxygenase 1-mediated iron metabolism in macrophages contributes to lipid deposition in nonalcoholic steatohepatitis. J Gastroenterol 2024; 59:342-356. [PMID: 38402297 DOI: 10.1007/s00535-024-02082-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/17/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) is a rapidly progressing chronic liver disease of global significance. However, the underlying mechanisms responsible for NASH remain unknown. Indoleamine 2,3-dioxygenase 1 (IDO1) has been recognized as essential factor in immune response and metabolic regulation. Here we aimed to investigate the functions and mechanisms of the IDO1 in macrophages on hepatic lipid deposition and iron metabolism in NASH. METHODS The effect of IDO1 in NASH was evaluated by WT and IDO1-/- mice model fed with methionine/choline-deficient (MCD) diet in vivo. Macrophages scavenger clodronate liposomes (CL) and overexpressing of IDO1 in macrophages by virus were employed as well. Lipid deposition was assessed through pathological examination and lipid droplet staining, while iron levels were measured using an iron assay kit and western blotting. Primary hepatocytes and bone marrow-derived macrophages were treated with oleic acid/palmitic acid (OA/PA) to assess IDO1 expression via Oil Red O staining and immunofluorescence staining in vitro. RESULTS Pathological images demonstrated that the increase of IDO1 exacerbated lipid accumulation in the livers of mice with MCD diet, while reduction of iron accumulation was observed in the liver and the serum of MCD-fed mice. Scavenging of macrophages effectively mitigated both lipid and iron accumulation. In addition, the deficiency of IDO1 in macrophages significantly mitigated lipid accumulation and iron overload in hepatic parenchymal cells. Finally, lentivirus-mediated overexpression of IDO1 in liver macrophages exacerbated hepatic steatosis and iron deposition in NASH. CONCLUSIONS Our results demonstrated that effective inhibition of IDO1 expression in macrophages in NASH alleviated hepatic parenchymal cell lipid accumulation and iron deposition, which provided new insights for the future treatment of NASH.
Collapse
Affiliation(s)
- Chaofeng Wu
- Emergency Department, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Junjie Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Hui Jia
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiamin Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Mengchen Qin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Hao Shi
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Chang Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiajie Lin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Min Cai
- Department of Hepatology, Hainan Provincial Hospital of Chinese Medicine, Haikou, China
| | - Yong Gu
- Clinical Research Center, Hainan Provincial Hospital of Chinese Medicine, Haikou, China
| | - Bin Liu
- Emergency Department, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Lei Gao
- Emergency Department, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Matsumoto M, Sawada H, Iwata K, Ibi M, Asaoka N, Katsuyama M, Shintani-Ishida K, Ikegaya H, Takegami S, Umemura A, Yabe-Nishimura C. Bortezomib is an effective enhancer for chemical probe-dependent superoxide detection. Front Med (Lausanne) 2022; 9:941180. [PMID: 36619644 PMCID: PMC9811382 DOI: 10.3389/fmed.2022.941180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Various chemical probes for the detection of reactive oxygen species have been developed to examine oxidative stress associated with different pathologies. L-012, a luminol-based chemiluminescent probe, is widely used to detect extracellular superoxide because of its high sensitivity. We herein demonstrated that the co-application of the peptide boronic acid proteasome inhibitor, bortezomib, with L-012 significantly increased its luminescence without affecting the background. More than a 5-fold increase was detected in the total luminescence of L-012 in both NADPH oxidase-expressing cells and the xanthine oxidase-dependent cell-free superoxide generation system, but not in their background. Therefore, bortezomib increased the signal-to-background ratio and improved the detection of low levels of superoxide. The application of MLN2238, another peptide boronic acid proteasome inhibitor, also enhanced the luminescence of L-012. In contrast, carfilzomib, an epoxyketone proteasome inhibitor, did not increase luminescence, suggesting that the effects of bortezomib depend on the chemical structure of the peptide boronic acid, but not on its pharmacological effects. Bortezomib-induced enhancements appeared to be specific to the detection of superoxide because the detection of H2O2 by Amplex Red/HRP was not affected by the application of bortezomib. In the quantitative detection of the superoxide-specific oxidative product 2-hydroxyethidium (2-OH-E+), the application of bortezomib resulted in a 2-fold increase in the level of 2-OH-E+. Therefore, bortezomib sensitizes the detection of superoxide in both cell-based and cell-free systems, highlighting a novel feature of compounds containing the peptide boronic acid as powerful enhancers for the detection of superoxide.
Collapse
Affiliation(s)
- Misaki Matsumoto
- 1Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, Japan,*Correspondence: Misaki Matsumoto,
| | - Hikari Sawada
- 1Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazumi Iwata
- 1Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masakazu Ibi
- 1Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nozomi Asaoka
- 1Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masato Katsuyama
- 2Radioisotope Center, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kaori Shintani-Ishida
- 3Department of Forensic Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroshi Ikegaya
- 3Department of Forensic Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigehiko Takegami
- 4Department of Analytical Chemistry, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Atsushi Umemura
- 1Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | |
Collapse
|
3
|
Cytochrome B5 type A alleviates HCC metastasis via regulating STOML2 related autophagy and promoting sensitivity to ruxolitinib. Cell Death Dis 2022; 13:623. [PMID: 35851063 PMCID: PMC9293983 DOI: 10.1038/s41419-022-05053-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 06/20/2022] [Accepted: 06/29/2022] [Indexed: 01/21/2023]
Abstract
The incidence of hepatocellular carcinoma (HCC) is increasing in the world. However, its role and underlying molecular mechanism in HCC progression remain unclear. We found that CYB5A plays a key role in HCC metastasis by inhibiting the JAK1/STAT3 pathway through binding to STOML2. CYB5A combined with STOML2 can predict the outcome of patients. To demonstrate the effect of CYB5A on JAK1 inhibitor function, we applied Ruxolitinib in metastatic tumors with high CYB5A expression and found that it slowed disease progression and prolonged survival in mice. To the best of our knowledge, this study is the first to report the Ruxolitinib effect on the metastatic ability of HCC cells in vivo and in vitro.
Collapse
|
4
|
Gáspár R, Halmi D, Demján V, Berkecz R, Pipicz M, Csont T. Kynurenine Pathway Metabolites as Potential Clinical Biomarkers in Coronary Artery Disease. Front Immunol 2022; 12:768560. [PMID: 35211110 PMCID: PMC8861075 DOI: 10.3389/fimmu.2021.768560] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/22/2021] [Indexed: 12/14/2022] Open
Abstract
Coronary artery disease (CAD) is one of the leading cause of mortality worldwide. Several risk factors including unhealthy lifestyle, genetic background, obesity, diabetes, hypercholesterolemia, hypertension, smoking, age, etc. contribute to the development of coronary atherosclerosis and subsequent coronary artery disease. Inflammation plays an important role in coronary artery disease development and progression. Pro-inflammatory signals promote the degradation of tryptophan via the kynurenine pathway resulting in the formation of several immunomodulatory metabolites. An unbalanced kynurenic pathway has been implicated in the pathomechanisms of various diseases including CAD. Significant improvements in detection methods in the last decades may allow simultaneous measurement of multiple metabolites of the kynurenine pathway and such a thorough analysis of the kynurenine pathway may be a valuable tool for risk stratification and determination of CAD prognosis. Nevertheless, imbalance in the activities of different branches of the kynurenine pathway may require careful interpretation. In this review, we aim to summarize clinical evidence supporting a possible use of kynurenine pathway metabolites as clinical biomarkers in various manifestations of CAD.
Collapse
Affiliation(s)
- Renáta Gáspár
- Metabolic Diseases and Cell Signaling Research Group (MEDICS), Department of Biochemistry, University of Szeged Albert Szent-Györgyi Medical School, Szeged, Hungary
- Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| | - Dóra Halmi
- Metabolic Diseases and Cell Signaling Research Group (MEDICS), Department of Biochemistry, University of Szeged Albert Szent-Györgyi Medical School, Szeged, Hungary
- Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| | - Virág Demján
- Metabolic Diseases and Cell Signaling Research Group (MEDICS), Department of Biochemistry, University of Szeged Albert Szent-Györgyi Medical School, Szeged, Hungary
- Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| | - Róbert Berkecz
- Institute of Pharmaceutical Analysis, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - Márton Pipicz
- Metabolic Diseases and Cell Signaling Research Group (MEDICS), Department of Biochemistry, University of Szeged Albert Szent-Györgyi Medical School, Szeged, Hungary
- Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| | - Tamás Csont
- Metabolic Diseases and Cell Signaling Research Group (MEDICS), Department of Biochemistry, University of Szeged Albert Szent-Györgyi Medical School, Szeged, Hungary
- Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| |
Collapse
|
5
|
Zisimopoulos DN, Kalaitzopoulou E, Skipitari M, Papadea P, Panagopoulos NT, Salahas G, Georgiou CD. Detection of superoxide radical in all biological systems by Thin Layer Chromatography. Arch Biochem Biophys 2021; 716:109110. [PMID: 34958749 DOI: 10.1016/j.abb.2021.109110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022]
Abstract
The study presents a new method that detects O2•-, via quantification of 2-hydroxyethidium (2-ΟΗ-Ε+) as low as ∼30 fmoles by High-Performance Thin Layer Chromatography (HPTLC). The method isolates 2-ΟΗ-Ε+ after its extraction by the anionic detergent SDS (at 18-fold higher than its CMC) together with certain organic/inorganic reagents, and its HPTLC-separation from di-ethidium (di-Ε+) and ethidium (Ε+). Quantification of 2-OH-E+ is based on its ex/em maxima at 290/540 nm, and of di-E+ and E+ at 295/545 nm. The major innovations of the present method are the development of protocols for (i) efficient extraction (by SDS) and (ii) sensitive quantification (by HPTLC) for 2-OH-E+ (as well as di-E+ and E+) from most biological systems (animals, plants, cells, subcellular compartments, fluids). The method extracts 2-ΟΗ-Ε+ (by neutralizing the strong binding between its quaternary N+ and negatively charged sites on phospholipids, DNA etc) together with free HE, while protects both from biological oxidases, and also extracts/quantifies total proteins (hydrophilic and hydrophobic) for expressing O2•- levels per protein quantity. The method also uses SDS (at 80-fold lower than its CMC) to extract/remove/wash 2-ΟΗ-Ε+ from cell/organelle exterior membrane sites, for more accurate internal content quantification. The new method is applied on indicative biological systems: (1) artificially stressed (mouse organs and liver mitochondria and nuclei, ±exposed to paraquat, a known O2•- generator), and (2) physiologically stressed (cauliflower plant, exposed to light/dark).
Collapse
Affiliation(s)
- Dimitrios N Zisimopoulos
- Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece.
| | - Electra Kalaitzopoulou
- Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece.
| | - Marianna Skipitari
- Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece.
| | - Polyxeni Papadea
- Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece.
| | | | | | - Christos D Georgiou
- Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece.
| |
Collapse
|
6
|
Mukherjee M, Dey A. Rejigging Electron and Proton Transfer to Transition between Dioxygenase, Monooxygenase, Peroxygenase, and Oxygen Reduction Activity: Insights from Bioinspired Constructs of Heme Enzymes. JACS AU 2021; 1:1296-1311. [PMID: 34604840 PMCID: PMC8479764 DOI: 10.1021/jacsau.1c00100] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Indexed: 05/10/2023]
Abstract
Nature has employed heme proteins to execute a diverse set of vital life processes. Years of research have been devoted to understanding the factors which bias these heme enzymes, with all having a heme cofactor, toward distinct catalytic activity. Among them, axial ligation, distal super structure, and substrate binding pockets are few very vividly recognized ones. Detailed mechanistic investigation of these heme enzymes suggested that several of these enzymes, while functionally divergent, use similar intermediates. Furthermore, the formation and decay of these intermediates depend on proton and electron transfer processes in the enzyme active site. Over the past decade, work in this group, using in situ surface enhanced resonance Raman spectroscopy of synthetic and biosynthetic analogues of heme enzymes, a general idea of how proton and electron transfer rates relate to the lifetime of different O2 derived intermediates has been developed. These findings suggest that the enzymatic activities of all these heme enzymes can be integrated into one general cycle which can be branched out to different catalytic pathways by regulating the lifetime and population of each of these intermediates. This regulation can further be achieved by tuning the electron and proton transfer steps. By strategically populating one of these intermediates during oxygen reduction, one can navigate through different catalytic processes to a desired direction by altering proton and electron transfer steps.
Collapse
Affiliation(s)
- Manjistha Mukherjee
- School of Chemical Science, Indian Association for the Cultivation of Science, 2A Raja SC Mullick Road, Kolkata, WB India, 700032
| | - Abhishek Dey
- School of Chemical Science, Indian Association for the Cultivation of Science, 2A Raja SC Mullick Road, Kolkata, WB India, 700032
| |
Collapse
|
7
|
Yuasa HJ, Stocker R. Methylene blue and ascorbate interfere with the accurate determination of the kinetic properties of IDO2. FEBS J 2021; 288:4892-4904. [PMID: 33686747 DOI: 10.1111/febs.15806] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/16/2021] [Accepted: 03/08/2021] [Indexed: 11/30/2022]
Abstract
Indoleamine 2,3-dioxygenases (IDOs) catalyze the oxidative cleavage of L-tryptophan (Trp) to N-formylkynurenine. Two IDOs, IDO1 and IDO2, are present in vertebrates. IDO1 is a high-affinity Trp-degrading enzyme involved in several physiological processes. By comparison, IDO2 generally has been reported to have low affinity (high Km -value) for Trp, and the enzyme's in vivo function remains unclear. Using IDOs from different species, we show that compared with ferrous-oxy (Fe2+ -O2 ) IDO1, Fe2+ -O2 IDO2 is substantially more stable and engages in multiple turnovers of the reaction in the absence of a reductant. Without reductant, Fe2+ -O2 IDO2 showed Km -values in the range of 80-356 μM, that is, values substantially lower than reported previously and close to the physiological concentrations of Trp. Methylene blue and ascorbate (Asc), used commonly as the reducing system for IDO activity determination, significantly affected the enzymatic activity of IDO2: In combination, the two reductants increased the apparent Km - and kcat -values 8- to 117-fold and 2-fold, respectively. Asc alone both activated and inhibited IDO2 by acting as a source of electrons and as a weak competitive inhibitor, respectively. In addition, ferric (Fe3+ ) IDO1 and IDO2 exhibited weak dioxygenase activity, similar to tryptophan 2,3-dioxygenase. Our results shed new light in the enzymatic activity of IDO2, and they support the view that this isoform of IDO also participates in the metabolism of Trp in vivo.
Collapse
Affiliation(s)
- Hajime J Yuasa
- Laboratory of Biochemistry, Department of Chemistry and Biotechnology, Faculty of Science and Technology, National University Corporation Kochi University, Japan
| | - Roland Stocker
- Arterial Inflammation and Redox Biology Group, Heart Research Institute, Newtown, NSW, Australia
| |
Collapse
|
8
|
Lin W, Shen P, Song Y, Huang Y, Tu S. Reactive Oxygen Species in Autoimmune Cells: Function, Differentiation, and Metabolism. Front Immunol 2021; 12:635021. [PMID: 33717180 PMCID: PMC7946999 DOI: 10.3389/fimmu.2021.635021] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 02/08/2021] [Indexed: 12/19/2022] Open
Abstract
Accumulated reactive oxygen species (ROS) directly contribute to biomacromolecule damage and influence various inflammatory responses. Reactive oxygen species act as mediator between innate and adaptive immune cells, thereby influencing the antigen-presenting process that results in T cell activation. Evidence from patients with chronic granulomatous disease and mouse models support the function of ROS in preventing abnormal autoimmunity; for example, by supporting maintenance of macrophage efferocytosis and T helper 1/T helper 2 and T helper 17/ regulatory T cell balance. The failure of many anti-oxidation treatments indicates that ROS cannot be considered entirely harmful. Indeed, enhancement of ROS may sometimes be required. In a mouse model of rheumatoid arthritis (RA), absence of NOX2-derived ROS led to higher prevalence and more severe symptoms. In patients with RA, naïve CD4+ T cells exhibit inhibited glycolysis and enhanced pentose phosphate pathway (PPP) activity, leading to ROS exhaustion. In this "reductive" state, CD4+ T cell immune homeostasis is disrupted, triggering joint destruction, together with oxidative stress in the synovium.
Collapse
Affiliation(s)
- Weiji Lin
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Shen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaqin Song
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shenghao Tu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Röhrig UF, Reynaud A, Majjigapu SR, Vogel P, Pojer F, Zoete V. Inhibition Mechanisms of Indoleamine 2,3-Dioxygenase 1 (IDO1). J Med Chem 2019; 62:8784-8795. [PMID: 31525930 DOI: 10.1021/acs.jmedchem.9b00942] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) catalyzes the rate-limiting step in the kynurenine pathway of tryptophan metabolism, which is involved in immunity, neuronal function, and aging. Its implication in pathologies such as cancer and neurodegenerative diseases has stimulated the development of IDO1 inhibitors. However, negative phase III clinical trial results of the IDO1 inhibitor epacadostat in cancer immunotherapy call for a better understanding of the role and the mechanisms of IDO1 inhibition. In this work, we investigate the molecular inhibition mechanisms of four known IDO1 inhibitors and of two quinones in detail, using different experimental and computational approaches. We also determine for the first time the X-ray structure of the highly efficient 1,2,3-triazole inhibitor MMG-0358. Based on our results and a comprehensive literature overview, we propose a classification scheme for IDO1 inhibitors according to their inhibition mechanism, which will be useful for further developments in the field.
Collapse
Affiliation(s)
- Ute F Röhrig
- Molecular Modeling Group , SIB Swiss Institute of Bioinformatics , 1015 Lausanne , Switzerland
| | - Aline Reynaud
- Protein Production and Structure Core Facility, School of Life Sciences , École Polytechnique Fédérale de Lausanne (EPFL) , 1015 Lausanne , Switzerland
| | - Somi Reddy Majjigapu
- Molecular Modeling Group , SIB Swiss Institute of Bioinformatics , 1015 Lausanne , Switzerland.,Laboratory of Glycochemistry and Asymmetric Synthesis , École Polytechnique Fédérale de Lausanne (EPFL) , 1015 Lausanne , Switzerland
| | - Pierre Vogel
- Laboratory of Glycochemistry and Asymmetric Synthesis , École Polytechnique Fédérale de Lausanne (EPFL) , 1015 Lausanne , Switzerland
| | - Florence Pojer
- Protein Production and Structure Core Facility, School of Life Sciences , École Polytechnique Fédérale de Lausanne (EPFL) , 1015 Lausanne , Switzerland
| | - Vincent Zoete
- Molecular Modeling Group , SIB Swiss Institute of Bioinformatics , 1015 Lausanne , Switzerland.,Department of Fundamental Oncology , University of Lausanne, Ludwig Lausanne Branch , 1066 Epalinges , Switzerland
| |
Collapse
|
10
|
Nelp MT, Zheng V, Davis KM, Stiefel KJE, Groves JT. Potent Activation of Indoleamine 2,3-Dioxygenase by Polysulfides. J Am Chem Soc 2019; 141:15288-15300. [PMID: 31436417 DOI: 10.1021/jacs.9b07338] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
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.
Collapse
Affiliation(s)
- Micah T Nelp
- Department of Chemistry , Princeton University , Princeton , New Jersey 08544 , United States
| | - Vincent Zheng
- Department of Chemistry , Princeton University , Princeton , New Jersey 08544 , United States
| | - Katherine M Davis
- Department of Chemistry , Princeton University , Princeton , New Jersey 08544 , United States
| | - Katherine J E Stiefel
- Department of Chemistry , Princeton University , Princeton , New Jersey 08544 , United States
| | - John T Groves
- Department of Chemistry , Princeton University , Princeton , New Jersey 08544 , United States
| |
Collapse
|
11
|
Singlet molecular oxygen regulates vascular tone and blood pressure in inflammation. Nature 2019; 566:548-552. [DOI: 10.1038/s41586-019-0947-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 01/10/2019] [Indexed: 11/09/2022]
|
12
|
Lim YJ, Foo TC, Yeung AWS, Tu X, Ma Y, Hawkins CL, Witting PK, Jameson GNL, Terentis AC, Thomas SR. Human Indoleamine 2,3-Dioxygenase 1 Is an Efficient Mammalian Nitrite Reductase. Biochemistry 2019; 58:974-986. [PMID: 30585477 DOI: 10.1021/acs.biochem.8b01231] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
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.
Collapse
Affiliation(s)
| | - Timothy C Foo
- Department of Chemistry and Biochemistry , Florida Atlantic University , Boca Raton , Florida 33431 , United States
| | | | | | | | - Clare L Hawkins
- Department of Biomedical Sciences , University of Copenhagen , Copenhagen N DK-2200 , Denmark
| | - Paul K Witting
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health , University of Sydney , Sydney , NSW 2006 , Australia
| | - Guy N L Jameson
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute , The University of Melbourne , Parkville , VIC 3010 , Australia
| | - Andrew C Terentis
- Department of Chemistry and Biochemistry , Florida Atlantic University , Boca Raton , Florida 33431 , United States
| | | |
Collapse
|
13
|
Bhatt MR, Khatri Y, Rodgers RJ, Martin LL. Role of cytochrome b5 in the modulation of the enzymatic activities of cytochrome P450 17α-hydroxylase/17,20-lyase (P450 17A1). J Steroid Biochem Mol Biol 2017; 170:2-18. [PMID: 26976652 DOI: 10.1016/j.jsbmb.2016.02.033] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/25/2016] [Accepted: 02/27/2016] [Indexed: 12/13/2022]
Abstract
Cytochrome b5 (cyt b5) is a small hemoprotein that plays a significant role in the modulation of activities of an important steroidogenic enzyme, cytochrome P450 17α-hydroxylase/17,20-lyase (P450 17A1, CYP17A1). Located in the zona fasciculata and zona reticularis of the adrenal cortex and in the gonads, P450 17A1 catalyzes two different reactions in the steroidogenic pathway; the 17α-hydroxylation and 17,20-lyase, in the endoplasmic reticulum of these respective tissues. The activities of P450 17A1 are regulated by cyt b5 that enhances the 17,20-lyase reaction by promoting the coupling of P450 17A1 and cytochrome P450 reductase (CPR), allosterically. Cyt b5 can also act as an electron donor to enhance the 16-ene-synthase activity of human P450 17A1. In this review, we discuss the many roles of cyt b5 and focus on the modulation of CYP17A1 activities by cyt b5 and the mechanisms involved.
Collapse
Affiliation(s)
- Megh Raj Bhatt
- Everest Biotech Pvt. Ltd., Khumaltar, Lalitpur, P.O. Box 21608, Kathmandu 44600, Nepal
| | - Yogan Khatri
- Institute of Biochemistry, Saarland University, 66123 Saarbrücken, Germany
| | - Raymond J Rodgers
- School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide SA 5005, Australia
| | - Lisandra L Martin
- School of Chemistry, Monash University, Clayton, 3800, Victoria, Australia.
| |
Collapse
|
14
|
Cantin AM, Hanrahan JW. Thymosin α1: a single drug with multiple targets in cystic fibrosis. Nat Med 2017; 23:536-538. [DOI: 10.1038/nm.4339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
15
|
Watson JD, Prokopec SD, Smith AB, Okey AB, Pohjanvirta R, Boutros PC. 2,3,7,8 Tetrachlorodibenzo-p-dioxin-induced RNA abundance changes identify Ackr3, Col18a1, Cyb5a and Glud1 as candidate mediators of toxicity. Arch Toxicol 2016; 91:325-338. [PMID: 27136898 PMCID: PMC5225275 DOI: 10.1007/s00204-016-1720-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/20/2016] [Indexed: 12/11/2022]
Abstract
2,3,7,8 Tetrachlorodibenzo-p-dioxin (TCDD) is an aromatic, long-lived environmental contaminant. While the pathogenesis of TCDD-induced toxicity is poorly understood, it has been shown that the aryl hydrocarbon receptor (AHR) is required. However, the specific transcriptomic changes that lead to toxic outcomes have not yet been identified. We previously identified a panel of 33 genes that respond to TCDD treatment in two TCDD-sensitive rodent species. To identify genes involved in the onset of hepatic toxicity, we explored 25 of these in-depth using liver from two rat strains: the TCDD-resistant Han/Wistar (H/W) and the TCDD-sensitive Long–Evans (L–E). Time course and dose–response analyses of mRNA abundance following TCDD insult indicate that eight genes are similarly regulated in livers of both strains of rat, suggesting that they are not central to the severe L–E-specific TCDD-induced toxicities. The remaining 17 genes exhibited various divergent mRNA abundances between L–E and H/W strains after TCDD treatment. Several genes displayed a biphasic response where the initial response to TCDD treatment was followed by a secondary response, usually of larger magnitude in L–E liver. This secondary response was most often an exaggeration of the original TCDD-induced response. Only cytochrome b5 type A (microsomal) (Cyb5a) had equivalent TCDD sensitivity to the prototypic AHR-responsive cytochrome P450, family 1, subfamily a, polypeptide 1 (Cyp1a1), while six genes were less sensitive. Four genes showed an early inter-strain difference that was sustained throughout most of the time course (atypical chemokine receptor 3 (Ackr3), collagen, type XVIII, alpha 1 (Col18a1), Cyb5a and glutamate dehydrogenase 1 (Glud1)), and of those genes examined in this study, are most likely to represent genes involved in the pathogenesis of TCDD-induced hepatotoxicity in L–E rats.
Collapse
Affiliation(s)
- John D Watson
- Informatics and Bio-computing Program, MaRS Centre, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, ON, M5G 0A3, Canada
| | - Stephenie D Prokopec
- Informatics and Bio-computing Program, MaRS Centre, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, ON, M5G 0A3, Canada
| | - Ashley B Smith
- Informatics and Bio-computing Program, MaRS Centre, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, ON, M5G 0A3, Canada
| | - Allan B Okey
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Raimo Pohjanvirta
- Laboratory of Toxicology, National Institute for Health and Welfare, Kuopio, Finland.,Department of Food Hygiene and Environmental Health, University of Helsinki, Helsinki, Finland
| | - Paul C Boutros
- Informatics and Bio-computing Program, MaRS Centre, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, ON, M5G 0A3, Canada. .,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, Canada.
| |
Collapse
|
16
|
Reduction potential and heme-pocket polarity in low potential cytochrome b5 of Giardia intestinalis. J Inorg Biochem 2016; 158:110-114. [PMID: 27048807 DOI: 10.1016/j.jinorgbio.2016.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/24/2015] [Accepted: 02/23/2016] [Indexed: 11/20/2022]
Abstract
Although it lacks mitochondria and the ability to synthesize heme, the protozoan parasite Giardia intestinalis encodes several heme proteins. This includes four members of the cytochrome b5 family, three of which are of similar size to mammalian cytochromes b5 but with reduction potentials that are 140 to 180mV lower. While no structures have yet been determined for any of these proteins, homology modeling points to an increase in heme pocket polarity as a reason for their low potentials. To test this we measured the reduction potentials of four mutants of Giardia cytochrome b5 isotype-I (gCYTB5-I) in which polar residues at two candidate positions (C84, Y51) in the heme pocket were changed to nonpolar ones (C84A, C84F; Y51L, Y51F). All mutants were expressed with comparable levels of heme incorporation and had UV-visible spectra consistent with low spin bis-histidyl coordination. These mutations increased the reduction potential by 18 to 57mV and highlight the influence of C84, which is a residue unique to gCYTB5-I and whose mutation to alanine caused the largest increase. The influence of these two residues plus that of Y61 reported previously accounts for much of the reduction potential difference between gCYTB5-I and microsomal cytochrome b5. A complementary triple mutant of the latter with the hydrophilic residues found in gCYTB5-I bound heme less effectively but nonetheless had a reduction potential that was 135mV lower than wild type.
Collapse
|
17
|
Rafferty SP, Dayer G. Heme proteins of Giardia intestinalis. Exp Parasitol 2015; 159:13-23. [PMID: 26297679 DOI: 10.1016/j.exppara.2015.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 08/09/2015] [Accepted: 08/10/2015] [Indexed: 10/23/2022]
Abstract
Among the few organisms that cannot make the iron cofactor heme, some nonetheless possess heme proteins. This includes the protozoan parasite Giardia intestinalis, which encodes five known heme proteins: a flavohemoglobin and four members of the cytochrome b5 family. Giardia flavohemoglobin closely resembles those of the Enterobacteriaceae in structure and function, acting as a nitric oxide dioxygenase that is induced when trophozoites are exposed to reactive nitrogen species. The Giardia cytochromes b5 are soluble proteins having relatively low reduction potentials and lack several features that are expected to promote rapid electron transfer with redox partners. Only one potential electron donor, and no electron acceptors, have yet been identified in the Giardia genome, and the roles of these cytochromes are presently unknown. The answer may lie in the sequences that flank the heme-binding core of these proteins which could serve to localize them within the cell through reversible post-translational modifications and to promote specific protein-protein interactions.
Collapse
Affiliation(s)
- Steven Patrick Rafferty
- Department of Chemistry, Trent University, 1600 West Bank Drive, Peterborough, Ontario, Canada, K9J 7B8; Environmental and Life Sciences Graduate Program, Trent University, 1600 West Bank Drive, Peterborough, Ontario, Canada, K9J 7B8.
| | - Guillem Dayer
- Environmental and Life Sciences Graduate Program, Trent University, 1600 West Bank Drive, Peterborough, Ontario, Canada, K9J 7B8
| |
Collapse
|
18
|
Abstract
IDO1 (indoleamine 2,3-dioxygenase 1) is a member of a unique class of mammalian haem dioxygenases that catalyse the oxidative catabolism of the least-abundant essential amino acid, L-Trp (L-tryptophan), along the kynurenine pathway. Significant increases in knowledge have been recently gained with respect to understanding the fundamental biochemistry of IDO1 including its catalytic reaction mechanism, the scope of enzyme reactions it catalyses, the biochemical mechanisms controlling IDO1 expression and enzyme activity, and the discovery of enzyme inhibitors. Major advances in understanding the roles of IDO1 in physiology and disease have also been realised. IDO1 is recognised as a prominent immune regulatory enzyme capable of modulating immune cell activation status and phenotype via several molecular mechanisms including enzyme-dependent deprivation of L-Trp and its conversion into the aryl hydrocarbon receptor ligand kynurenine and other bioactive kynurenine pathway metabolites, or non-enzymatic cell signalling actions involving tyrosine phosphorylation of IDO1. Through these different modes of biochemical signalling, IDO1 regulates certain physiological functions (e.g. pregnancy) and modulates the pathogenesis and severity of diverse conditions including chronic inflammation, infectious disease, allergic and autoimmune disorders, transplantation, neuropathology and cancer. In the present review, we detail the current understanding of IDO1’s catalytic actions and the biochemical mechanisms regulating IDO1 expression and activity. We also discuss the biological functions of IDO1 with a focus on the enzyme's immune-modulatory function, its medical implications in diverse pathological settings and its utility as a therapeutic target.
Collapse
|
19
|
Röhrig UF, Majjigapu SR, Vogel P, Zoete V, Michielin O. Challenges in the Discovery of Indoleamine 2,3-Dioxygenase 1 (IDO1) Inhibitors. J Med Chem 2015; 58:9421-37. [DOI: 10.1021/acs.jmedchem.5b00326] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Ute F. Röhrig
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Somi Reddy Majjigapu
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
- Laboratory
of Glycochemistry and Asymmetric Synthesis, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Pierre Vogel
- Laboratory
of Glycochemistry and Asymmetric Synthesis, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
- Ludwig Center for Cancer Research of the University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Vincent Zoete
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Olivier Michielin
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
- Ludwig Center for Cancer Research of the University of Lausanne, CH-1015 Lausanne, Switzerland
- Department of Oncology, University of Lausanne and Centre Hospitalier Universitaire Vaudois (CHUV), CH-1011 Lausanne, Switzerland
| |
Collapse
|
20
|
Kolawole AO, Hixon BP, Dameron LS, Chrisman IM, Smirnov VV. Catalytic activity of human indoleamine 2,3-dioxygenase (hIDO1) at low oxygen. Arch Biochem Biophys 2015; 570:47-57. [PMID: 25712221 PMCID: PMC4412315 DOI: 10.1016/j.abb.2015.02.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 02/12/2015] [Accepted: 02/15/2015] [Indexed: 11/30/2022]
Abstract
A cytokine-inducible extrahepatic human indoleamine 2,3-dioxygenase (hIDO1) catalyzes the first step of the kynurenine pathway. Immunosuppressive activity of hIDO1 in tumor cells weakens host T-cell immunity, contributing to the progression of cancer. Here we report on enzyme kinetics and catalytic mechanism of hIDO1, studied at varied levels of dioxygen (O2) and L-tryptophan (L-Trp). Using a cytochrome b5-based activating system, we measured the initial rates of O2 decay with a Clark-type oxygen electrode at physiologically-relevant levels of both substrates. Kinetics was also studied in the presence of two substrate analogs: 1-methyl-L-tryptophan and norharmane. Quantitative analysis supports a steady-state rather than a rapid equilibrium kinetic mechanism, where the rates of individual pathways, leading to a ternary complex, are significantly different, and the overall rate of catalysis depends on contributions of both routes. One path, where O2 binds to ferrous hIDO1 first, is faster than the second route, which starts with the binding of L-Trp. However, L-Trp complexation with free ferrous hIDO1 is more rapid than that of O2. As the level of L-Trp increases, the slower route becomes a significant contributor to the overall rate, resulting in observed substrate inhibition.
Collapse
Affiliation(s)
- Ayodele O Kolawole
- Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, United States
| | - Brian P Hixon
- Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, United States
| | - Laura S Dameron
- Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, United States
| | - Ian M Chrisman
- Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, United States
| | - Valeriy V Smirnov
- Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, United States.
| |
Collapse
|
21
|
Lysophosphatidylcholine exacerbates Leishmania major-dendritic cell infection through interleukin-10 and a burst in arginase1 and indoleamine 2,3-dioxygenase activities. Int Immunopharmacol 2015; 25:1-9. [DOI: 10.1016/j.intimp.2015.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 01/01/2015] [Accepted: 01/06/2015] [Indexed: 02/08/2023]
|
22
|
Tryptophan catabolism is unaffected in chronic granulomatous disease. Nature 2014; 514:E16-7. [DOI: 10.1038/nature13844] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 08/04/2014] [Indexed: 12/18/2022]
|
23
|
Ball HJ, Jusof FF, Bakmiwewa SM, Hunt NH, Yuasa HJ. Tryptophan-catabolizing enzymes - party of three. Front Immunol 2014; 5:485. [PMID: 25346733 PMCID: PMC4191572 DOI: 10.3389/fimmu.2014.00485] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 09/22/2014] [Indexed: 11/13/2022] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) are tryptophan-degrading enzymes that have independently evolved to catalyze the first step in tryptophan catabolism via the kynurenine pathway (KP). The depletion of tryptophan and formation of KP metabolites modulates the activity of the mammalian immune, reproductive, and central nervous systems. IDO and TDO enzymes can have overlapping or distinct functions depending on their expression patterns. The expression of TDO and IDO enzymes in mammals differs not only by tissue/cellular localization but also by their induction by distinct stimuli. To add to the complexity, these genes also have undergone duplications in some organisms leading to multiple isoforms of IDO or TDO. For example, many vertebrates, including all mammals, have acquired two IDO genes via gene duplication, although the IDO1-like gene has been lost in some lower vertebrate lineages. Gene duplications can allow the homologs to diverge and acquire different properties to the original gene. There is evidence for IDO enzymes having differing enzymatic characteristics, signaling properties, and biological functions. This review analyzes the evolutionary convergence of IDO and TDO enzymes as tryptophan-catabolizing enzymes and the divergent evolution of IDO homologs to generate an enzyme family with diverse characteristics not possessed by TDO enzymes, with an emphasis on the immune system.
Collapse
Affiliation(s)
- Helen J Ball
- Molecular Immunopathology Unit, School of Medical Sciences and Bosch Institute, University of Sydney , Sydney, NSW , Australia
| | - Felicita F Jusof
- Molecular Immunopathology Unit, School of Medical Sciences and Bosch Institute, University of Sydney , Sydney, NSW , Australia ; Department of Physiology, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| | - Supun M Bakmiwewa
- Molecular Immunopathology Unit, School of Medical Sciences and Bosch Institute, University of Sydney , Sydney, NSW , Australia
| | - Nicholas H Hunt
- Molecular Immunopathology Unit, School of Medical Sciences and Bosch Institute, University of Sydney , Sydney, NSW , Australia
| | - Hajime J Yuasa
- Laboratory of Biochemistry, Faculty of Science, Department of Applied Science, National University Corporation Kochi University , Kochi , Japan
| |
Collapse
|
24
|
The Fe-heme structure of met-indoleamine 2,3-dioxygenase-2 determined by X-ray absorption fine structure. Biochem Biophys Res Commun 2014; 450:25-9. [DOI: 10.1016/j.bbrc.2014.05.054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 05/14/2014] [Indexed: 01/20/2023]
|
25
|
Sedlmayr P, Blaschitz A, Stocker R. The role of placental tryptophan catabolism. Front Immunol 2014; 5:230. [PMID: 24904580 PMCID: PMC4032907 DOI: 10.3389/fimmu.2014.00230] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 05/05/2014] [Indexed: 01/22/2023] Open
Abstract
This review discusses the mechanisms and consequences of degradation of tryptophan (Trp) in the placenta, focusing mainly on the role of indoleamine 2,3-dioxygenase-1 (IDO1), one of three enzymes catalyzing the first step of the kynurenine pathway of Trp degradation. IDO1 has been implicated in regulation of feto-maternal tolerance in the mouse. Local depletion of Trp and/or the presence of metabolites of the kynurenine pathway mediate immunoregulation and exert antimicrobial functions. In addition to the decidual glandular epithelium, IDO1 is localized in the vascular endothelium of the villous chorion and also in the endothelium of spiral arteries of the decidua. Possible consequences of IDO1-mediated catabolism of Trp in the endothelium encompass antimicrobial activity and immunosuppression, as well as relaxation of the placental vasotonus, thereby contributing to placental perfusion and growth of both placenta and fetus. It remains to be evaluated whether other enzymes mediating Trp oxidation, such as indoleamine 2,3-dioxygenase-2, Trp 2,3-dioxygenase, and Trp hydroxylase-1 are of relevance to the biology of the placenta.
Collapse
Affiliation(s)
- Peter Sedlmayr
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz , Graz , Austria
| | - Astrid Blaschitz
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz , Graz , Austria
| | - Roland Stocker
- Victor Chang Cardiac Research Institute , Darlinghurst, NSW , Australia
| |
Collapse
|
26
|
Geng J, Liu A. Heme-dependent dioxygenases in tryptophan oxidation. Arch Biochem Biophys 2013; 544:18-26. [PMID: 24295960 DOI: 10.1016/j.abb.2013.11.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 11/19/2013] [Accepted: 11/20/2013] [Indexed: 12/29/2022]
Abstract
L-Tryptophan is an essential amino acid for mammals. It is utilized not only for protein synthesis but also for the biosynthesis of serotonin and melatonin by the serotonin pathway as well as nicotinamide adenine dinucleotide by the kynurenine pathway. Although the kynurenine pathway is responsible for the catabolism of over 90% of l-tryptophan in the mammalian intracellular and extracellular pools, the scientific field was dominated in the last century by studies of the serotonin pathway, due to the physiological significance of the latter's catabolic intermediates and products. However, in the past decade, the focus gradually reversed as the link between the kynurenine pathway and various neurodegenerative disorders and immune diseases is increasingly highlighted. Notably, the first step of this pathway, which is catalyzed by heme-dependent dioxygenases, has been proven to be a potential target for immune regulation and cancer treatment. A thorough understanding of the intriguing chemistry of the heme-dependent dioxygenases may yield insight for the drug discovery of these prevalent illnesses. In this review, we survey enzymatic and mechanistic studies, initially started by Kotake and Masayama over 70 years ago, at the molecular level on the heme-dependent tryptophan dioxygenation reactions.
Collapse
Affiliation(s)
- Jiafeng Geng
- Department of Chemistry, Georgia State University, 50 Decatur Street SE, Atlanta, GA 30303, United States
| | - Aimin Liu
- Department of Chemistry, Georgia State University, 50 Decatur Street SE, Atlanta, GA 30303, United States.
| |
Collapse
|
27
|
Fatokun AA, Hunt NH, Ball HJ. Indoleamine 2,3-dioxygenase 2 (IDO2) and the kynurenine pathway: characteristics and potential roles in health and disease. Amino Acids 2013; 45:1319-29. [PMID: 24105077 DOI: 10.1007/s00726-013-1602-1] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 09/25/2013] [Indexed: 12/21/2022]
Abstract
The kynurenine pathway is the major route for the oxidative degradation of the amino acid tryptophan. Activity of the pathway is involved in several disease conditions, both in the periphery and the central nervous system, including cancer, inflammatory disorders, neurological conditions, psychiatric disorders and neurodegenerative diseases. Three enzymes are now known to catalyze the first and rate-limiting step in the catabolism of tryptophan along this pathway: tryptophan 2,3-dioxygenase (TDO) and indoleamine 2,3-dioxygenase (IDO, subsequently named IDO1), both of which have been extensively studied, and a third enzyme, indoleamine 2,3-dioxygenase 2 (IDO2), a relative newcomer to the kynurenine pathway field. The adjuvant chemotherapeutic agent, 1-methyl-D-tryptophan, was intially suggested to target IDO2, implying involvement of IDO2 in tumorigenesis. Subsequently this compound has been suggested to have alternative actions and the physiological and pathophysiological roles of IDO2 are unclear. Targeted genetic interventions and selective inhibitors provide approaches for investigating the biology of IDO2. This review focuses on the current knowledge of IDO2 biology and discusses tools that will assist in further characterizing the enzymes of the kynurenine pathway.
Collapse
Affiliation(s)
- Amos A Fatokun
- School of Biomedical Sciences, Institute of Cell Signalling, The University of Nottingham, Nottingham, NG7 2UH, UK
| | | | | |
Collapse
|
28
|
Alam S, Yee J, Couture M, Takayama SIJ, Tseng WH, Mauk AG, Rafferty S. Cytochrome b5 from Giardia lamblia. Metallomics 2013; 4:1255-61. [PMID: 23151674 DOI: 10.1039/c2mt20152f] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The protozoan intestinal parasite Giardia lamblia lacks mitochondria and the ability to make haem yet encodes several putative haem-binding proteins, including three of the cytochrome b(5) family. We cloned one of these (gCYTb5-I) and expressed it within Escherichia coli as a soluble holoprotein. UV-visible and resonance Raman spectra of gCYTb5-I resemble those of microsomal cytochrome b(5), and homology modelling supports a structure in which a pair of invariant histidine residues act as axial ligands to the haem iron. The reduction potential of gCYTb5-I is -165 mV vs. SHE and is relatively low compared to most values (-110 to +80 mV) for this class of protein. The amino- and carboxy-terminal sequences that flank the central haem-binding core of the Giardia cytochromes are highly charged and differ from those of other family members. A core gCYTb5-I variant lacking these flanking sequences was also able to bind haem. The presence of one actual and two probable functional cytochromes b(5) in Giardia is evidence of uncharacterized cytochrome-mediated metabolic processes within this medically important protist.
Collapse
Affiliation(s)
- Samiah Alam
- Environmental and Life Sciences Graduate Program, Trent University, Peterborough, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
29
|
McGaha TL, Huang L, Lemos H, Metz R, Mautino M, Prendergast GC, Mellor AL. Amino acid catabolism: a pivotal regulator of innate and adaptive immunity. Immunol Rev 2013; 249:135-57. [PMID: 22889220 DOI: 10.1111/j.1600-065x.2012.01149.x] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Enhanced amino acid catabolism is a common response to inflammation, but the immunologic significance of altered amino acid consumption remains unclear. The finding that tryptophan catabolism helped maintain fetal tolerance during pregnancy provided novel insights into the significance of amino acid metabolism in controlling immunity. Recent advances in identifying molecular pathways that enhance amino acid catabolism and downstream mechanisms that affect immune cells in response to inflammatory cues support the notion that amino acid catabolism regulates innate and adaptive immune cells in pathologic settings. Cells expressing enzymes that degrade amino acids modulate antigen-presenting cell and lymphocyte functions and reveal critical roles for amino acid- and catabolite-sensing pathways in controlling gene expression, functions, and survival of immune cells. Basal amino acid catabolism may contribute to immune homeostasis that prevents autoimmunity, whereas elevated amino acid catalytic activity may reinforce immune suppression to promote tumorigenesis and persistence of some pathogens that cause chronic infections. For these reasons, there is considerable interest in generating novel drugs that inhibit or induce amino acid consumption and target downstream molecular pathways that control immunity. In this review, we summarize recent developments and highlight novel concepts and key outstanding questions in this active research field.
Collapse
Affiliation(s)
- Tracy L McGaha
- Immunotherapy Center, Georgia Health Sciences University, Augusta, GA 30912, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Segal BH, Grimm MJ, Khan ANH, Han W, Blackwell TS. Regulation of innate immunity by NADPH oxidase. Free Radic Biol Med 2012; 53:72-80. [PMID: 22583699 PMCID: PMC3377837 DOI: 10.1016/j.freeradbiomed.2012.04.022] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 03/26/2012] [Accepted: 04/06/2012] [Indexed: 11/29/2022]
Abstract
NADPH oxidase is a critical regulator of both antimicrobial host defense and inflammation. Activated in nature by microbes and microbial-derived products, the phagocyte NADPH oxidase is rapidly assembled, and generates reactive oxidant intermediates (ROIs) in response to infectious threat. Chronic granulomatous disease (CGD) is an inherited disorder of the NADPH oxidase characterized by recurrent and severe bacterial and fungal infections, and pathology related to excessive inflammation. Studies in CGD patients and CGD mouse models indicate that NADPH oxidase plays a key role in modulating inflammation and injury that is distinct from its antimicrobial function. The mechanisms by which NADPH oxidase mediates killing of pathogens and regulation of inflammation have broad relevance to our understanding of normal physiological immune responses and pathological states, such as acute lung injury and bacterial or fungal infections.
Collapse
Affiliation(s)
- Brahm H Segal
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 37232-2650, USA
| | | | | | | | | |
Collapse
|
31
|
Röhrig UF, Majjigapu SR, Grosdidier A, Bron S, Stroobant V, Pilotte L, Colau D, Vogel P, Van den Eynde BJ, Zoete V, Michielin O. Rational Design of 4-Aryl-1,2,3-Triazoles for Indoleamine 2,3-Dioxygenase 1 Inhibition. J Med Chem 2012; 55:5270-90. [DOI: 10.1021/jm300260v] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ute F. Röhrig
- Ludwig Center
for Cancer Research
of the University of Lausanne, CH-1015 Lausanne, Switzerland
- Swiss Institute of Bioinformatics,
Molecular Modeling Group, CH-1015 Lausanne, Switzerland
| | - Somi Reddy Majjigapu
- Ludwig Center
for Cancer Research
of the University of Lausanne, CH-1015 Lausanne, Switzerland
- Laboratory of Glycochemistry and
Asymmetric Synthesis, Ecole Polytechnique Fédérale de
Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Aurélien Grosdidier
- Swiss Institute of Bioinformatics,
Molecular Modeling Group, CH-1015 Lausanne, Switzerland
| | - Sylvian Bron
- Swiss Institute of Bioinformatics,
Molecular Modeling Group, CH-1015 Lausanne, Switzerland
- Pluridisciplinary Centre for
Clinical Oncology (CePO), Centre Hospitalier Universitaire Vaudois
(CHUV), Lausanne, Switzerland
| | - Vincent Stroobant
- Ludwig Institute for Cancer
Research, Brussels Branch, and de Duve Institute, Université
Catholique de Louvain, B-1200 Brussels, Belgium
| | - Luc Pilotte
- Ludwig Institute for Cancer
Research, Brussels Branch, and de Duve Institute, Université
Catholique de Louvain, B-1200 Brussels, Belgium
| | - Didier Colau
- Ludwig Institute for Cancer
Research, Brussels Branch, and de Duve Institute, Université
Catholique de Louvain, B-1200 Brussels, Belgium
| | - Pierre Vogel
- Ludwig Center
for Cancer Research
of the University of Lausanne, CH-1015 Lausanne, Switzerland
- Laboratory of Glycochemistry and
Asymmetric Synthesis, Ecole Polytechnique Fédérale de
Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Benoît J. Van den Eynde
- Ludwig Institute for Cancer
Research, Brussels Branch, and de Duve Institute, Université
Catholique de Louvain, B-1200 Brussels, Belgium
| | - Vincent Zoete
- Swiss Institute of Bioinformatics,
Molecular Modeling Group, CH-1015 Lausanne, Switzerland
| | - Olivier Michielin
- Ludwig Center
for Cancer Research
of the University of Lausanne, CH-1015 Lausanne, Switzerland
- Swiss Institute of Bioinformatics,
Molecular Modeling Group, CH-1015 Lausanne, Switzerland
- Pluridisciplinary Centre for
Clinical Oncology (CePO), Centre Hospitalier Universitaire Vaudois
(CHUV), Lausanne, Switzerland
| |
Collapse
|
32
|
Millett ES, Efimov I, Basran J, Handa S, Mowat CG, Raven EL. Heme-containing dioxygenases involved in tryptophan oxidation. Curr Opin Chem Biol 2012; 16:60-6. [DOI: 10.1016/j.cbpa.2012.01.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Revised: 01/18/2012] [Accepted: 01/25/2012] [Indexed: 10/28/2022]
|
33
|
Idkowiak J, Randell T, Dhir V, Patel P, Shackleton CHL, Taylor NF, Krone N, Arlt W. A missense mutation in the human cytochrome b5 gene causes 46,XY disorder of sex development due to true isolated 17,20 lyase deficiency. J Clin Endocrinol Metab 2012; 97:E465-75. [PMID: 22170710 PMCID: PMC3388247 DOI: 10.1210/jc.2011-2413] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 11/11/2011] [Indexed: 11/29/2022]
Abstract
CONTEXT Isolated 17,20 lyase deficiency is commonly defined by apparently normal 17α-hydroxylase activity but severely reduced 17,20 lyase activity of the bifunctional enzyme cytochrome P450 (CYP) enzyme 17A1 (CYP17A1), resulting in sex steroid deficiency but normal glucocorticoid and mineralocorticoid reserve. Cytochrome b5 (CYB5A) is thought to selectively enhance 17,20 lyase activity by facilitating the allosteric interaction of CYP17A1 with its electron donor P450 oxidoreductase (POR). OBJECTIVE We investigated a large consanguineous family including three siblings with 46,XY disorder of sex development (DSD) presenting with isolated 17,20 lyase deficiency. DESIGN We investigated the clinical and biochemical phenotype, conducted genetic analyses, and functionally characterized the identified CYB5A mutation in cell-based CYP17A1 coexpression assays. RESULTS All three siblings presented with 46,XY DSD, sex steroid deficiency, normal mineralocorticoids and glucocorticoids, and a urine steroid metabolome suggestive of isolated 17,20 lyase deficiency. CYP17A1 and POR sequences were normal, but we detected a homozygous CYB5A missense mutation (g.28,400A→T; p.H44L). Functional in vitro analysis revealed normal CYP17A1 17α-hydroxylase activity but severely impaired 17,20 lyase activity. In silico analysis suggested the disruption of CYB5A heme binding by p.H44L. CONCLUSION We have identified the first human CYB5A missense mutation as the cause of isolated 17,20 lyase deficiency in three individuals with 46,XY DSD. Detailed review of previously reported cases with apparently isolated 17,20 lyase deficiency due to mutant CYP17A1 and POR reveals impaired 17α-hydroxylase activity as assessed by steroid metabolome analysis and short cosyntropin testing. This suggests that truly isolated 17,20 lyase deficiency is observed only in individuals with inactivating CYB5A mutations.
Collapse
Affiliation(s)
- Jan Idkowiak
- Centre for Endocrinology, Diabetes, and Metabolism, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Hunt NH. Redox pioneer: professor Roland Stocker. Antioxid Redox Signal 2011; 15:3101-5. [PMID: 21609251 DOI: 10.1089/ars.2010.3842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Dr. Roland Stocker (Ph.D. 1985) is recognized here as a Redox Pioneer, because he has published one article on antioxidant/redox biology as first author that has been cited over 1000 times and has published another 32 articles, each cited over 100 times. Dr. Stocker received his undergraduate education at the Federal Institute of Technology Zürich, Switzerland (1975-1981), followed by postgraduate training at the Australian National University Canberra, Australia (1982-1985) and postdoctoral training at the University of California, Berkeley (1986-1987), and the University of Berne, Switzerland (1987-1988). Dr. Stocker's top scientific contributions are in the following areas: (i) molecular action and interaction of nonproteinaceous antioxidants, particularly bilirubin, α-tocopherol, and ubiquinol-10; (ii) lipoprotein lipid oxidation and its inhibition, with a particular focus on how α-tocopherol affects these processes; (iii) the role of arterial lipoprotein lipid oxidation in atherosclerosis and related diseases; (iv) modes of antiatherosclerotic action of probucol and the involvement of heme oxygenase-1 in vascular protection; and (v) the regulation of indoleamine 2,3-dioxygenase and its contribution to vascular tone and blood pressure in inflammatory diseases.
Collapse
Affiliation(s)
- Nicholas H Hunt
- Molecular Immunopathology Unit, Bosch Institute and School of Medical Sciences, University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
35
|
Tryptophan metabolism to kynurenine is a potential novel contributor to hypotension in human sepsis*. Crit Care Med 2011; 39:2678-83. [DOI: 10.1097/ccm.0b013e31822827f2] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
36
|
Soleymaninejadian E, Pramanik K, Samadian E. Immunomodulatory Properties of Mesenchymal Stem Cells: Cytokines and Factors. Am J Reprod Immunol 2011; 67:1-8. [DOI: 10.1111/j.1600-0897.2011.01069.x] [Citation(s) in RCA: 175] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
37
|
Rosell FI, Kuo HH, Mauk AG. NADH oxidase activity of indoleamine 2,3-dioxygenase. J Biol Chem 2011; 286:29273-29283. [PMID: 21690092 PMCID: PMC3190733 DOI: 10.1074/jbc.m111.262139] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 06/10/2011] [Indexed: 12/12/2022] Open
Abstract
The heme enzyme indoleamine 2,3-dioxygenase (IDO) was found to oxidize NADH under aerobic conditions in the absence of other enzymes or reactants. This reaction led to the formation of the dioxygen adduct of IDO and supported the oxidation of Trp to N-formylkynurenine. Formation of the dioxygen adduct and oxidation of Trp were accelerated by the addition of small amounts of hydrogen peroxide, and both processes were inhibited in the presence of either superoxide dismutase or catalase. Anaerobic reaction of IDO with NADH proceeded only in the presence of a mediator (e.g. methylene blue) and resulted in formation of the ferrous form of the enzyme. We propose that trace amounts of peroxide previously proposed to occur in NADH solutions as well as solid NADH activate IDO and lead to aerobic formation of superoxide and the reactive dioxygen adduct of the enzyme.
Collapse
Affiliation(s)
- Federico I Rosell
- Department of Biochemistry and Molecular Biology and Centre for Blood Research, Life Sciences Centre, 2350 Health Sciences Mall, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Hsin H Kuo
- Department of Biochemistry and Molecular Biology and Centre for Blood Research, Life Sciences Centre, 2350 Health Sciences Mall, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - A Grant Mauk
- Department of Biochemistry and Molecular Biology and Centre for Blood Research, Life Sciences Centre, 2350 Health Sciences Mall, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.
| |
Collapse
|
38
|
Watts SW, Shaw S, Burnett R, Dorrance AM. Indoleamine 2,3-diooxygenase in periaortic fat: mechanisms of inhibition of contraction. Am J Physiol Heart Circ Physiol 2011; 301:H1236-47. [PMID: 21841011 DOI: 10.1152/ajpheart.00384.2011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Indoleamine 2,3-dioxygenase (IDO) metabolizes L-tryptophan to L-kynurenine, promotes immunosuppression, and has been described as a consumer of superoxide. We discovered IDO expression in periaortic fat and tested the hypothesis that periarterial IDO functionally reduces agonist-induced contraction. Our model was the thoracic aorta, abdominal aorta, and superior mesenteric artery of the male Sprague-Dawley rat. Periaortic fat from the thoracic aorta stained intensely for IDO, the brown fat marker uncoupling protein-1, and oil red O as a general lipid marker. White fat around the mesenteric artery and abdominal aorta stained less for IDO; brown fat was less abundant. IDO activity (kynurenine-to-tryptophan ratio via HPLC) was detected in visceral and mesenteric artery fat (ratio: ∼4) but was highest in perithoracic aortic fat (ratio: 10 ± 1.1). In isometric contractile experiments, periadventitial fat reduced ANG II-induced thoracic aortic (with fat: 34% of without fat) and mesenteric artery (with fat: 63% of without fat) maximal contraction. In contrast, periadventitial fat did not reduce agonist-induced contraction in the abdominal aorta. The IDO inhibitor 1-L-methyltryptophan (1-MT) reversed the fat-induced reduction of ANG II-induced contraction in the thoracic aorta but not in the mesenteric artery. The IDO metabolite kynurenine relaxed the thoracic aorta only at high (9 mM) concentrations, whereas the downstream metabolite quinolinic acid (1 mM) relaxed the contracted thoracic aorta (∼80%). 1-MT did not correct the reduction in basal superoxide levels observed in the presence of perithoracic aortic fat. We conclude that IDO is an enzyme active primarily in brown fat surrounding the thoracic aorta and depresses aortic contractility.
Collapse
Affiliation(s)
- Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824-1317, USA.
| | | | | | | |
Collapse
|
39
|
Maes M, Leonard BE, Myint AM, Kubera M, Verkerk R. The new '5-HT' hypothesis of depression: cell-mediated immune activation induces indoleamine 2,3-dioxygenase, which leads to lower plasma tryptophan and an increased synthesis of detrimental tryptophan catabolites (TRYCATs), both of which contribute to the onset of depression. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:702-21. [PMID: 21185346 DOI: 10.1016/j.pnpbp.2010.12.017] [Citation(s) in RCA: 475] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 12/07/2010] [Accepted: 12/16/2010] [Indexed: 02/07/2023]
Abstract
This paper reviews the body of evidence that not only tryptophan and consequent 5-HT depletion, but also induction of indoleamine 2,3-dioxygenase (IDO) and the detrimental effects of tryptophan catabolites (TRYCATs) play a role in the pathophysiology of depression. IDO is induced by interferon (IFN)γ, interleukin-6 and tumor necrosis factor-α, lipopolysaccharides and oxidative stress, factors that play a role in the pathophysiology of depression. TRYCATs, like kynurenine and quinolinic acid, are depressogenic and anxiogenic; activate oxidative pathways; cause mitochondrial dysfunctions; and have neuroexcitatory and neurotoxic effects that may lead to neurodegeneration. The TRYCAT pathway is also activated following induction of tryptophan 2,3-dioxygenase (TDO) by glucocorticoids, which are elevated in depression. There is evidence that activation of IDO reduces plasma tryptophan and increases TRYCAT synthesis in depressive states and that TDO activation may play a role as well. The development of depressive symptoms during IFNα-based immunotherapy is strongly associated with IDO activation, increased production of detrimental TRYCATs and lowered levels of tryptophan. Women show greater IDO activation and TRYCAT production following immune challenge than men. In the early puerperium, IDO activation and TRYCAT production are associated with the development of affective symptoms. Clinical depression is accompanied by lowered levels of neuroprotective TRYCATs or increased levels or neurotoxic TRYCATs, and lowered plasma tryptophan, which is associated with indices of immune activation and glucocorticoid hypersecretion. Lowered tryptophan and increased TRYCATs induce T cell unresponsiveness and therefore may exert a negative feedback on the primary inflammatory response in depression. It is concluded that activation of the TRYCAT pathway by IDO and TDO may be associated with the development of depressive symptoms through tryptophan depletion and the detrimental effects of TRYCATs. Therefore, the TRYCAT pathway should be a new drug target in depression. Direct inhibitors of IDO are less likely to be useful drugs than agents, such as kynurenine hydroxylase inhibitors; drugs which block the primary immune response; compounds that increase the protective effects of kynurenic acid; and specific antioxidants that target IDO activation, the immune and oxidative pathways, and 5-HT as well.
Collapse
Affiliation(s)
- M Maes
- Maes Clinics @ TRIA, Piyavate Hospital, 998 Rimklongsamsen Road, Bangkok 10310, Thailand.
| | | | | | | | | |
Collapse
|
40
|
Efimov I, Basran J, Thackray SJ, Handa S, Mowat CG, Raven EL. Structure and reaction mechanism in the heme dioxygenases. Biochemistry 2011; 50:2717-24. [PMID: 21361337 PMCID: PMC3092302 DOI: 10.1021/bi101732n] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
![]()
As members of the family of heme-dependent enzymes, the heme dioxygenases are differentiated by virtue of their ability to catalyze the oxidation of l-tryptophan to N-formylkynurenine, the first and rate-limiting step in tryptophan catabolism. In the past several years, there have been a number of important developments that have meant that established proposals for the reaction mechanism in the heme dioxygenases have required reassessment. This focused review presents a summary of these recent advances, written from a structural and mechanistic perspective. It attempts to present answers to some of the long-standing questions, to highlight as yet unresolved issues, and to explore the similarities and differences of other well-known catalytic heme enzymes such as the cytochromes P450, NO synthase, and peroxidases.
Collapse
Affiliation(s)
- Igor Efimov
- Department of Chemistry, George Porter Building, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
| | | | | | | | | | | |
Collapse
|
41
|
Yanagisawa S, Horitani M, Sugimoto H, Shiro Y, Okada N, Ogura T. Resonance Raman study on the oxygenated and the ferryl-oxo species of indoleamine 2,3-dioxygenase during catalytic turnover. Faraday Discuss 2011; 148:239-47; discussion 299-314. [PMID: 21322487 DOI: 10.1039/c004552g] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Resonance Raman (RR) spectra of the oxygenated and Fe(IV)=O reaction intermediates of indoleamine 2,3-dioxygenase (IDO) are reported. Absorption and RR spectra reveal that the electronic and geometric structures of the two respective species at pH 6.5 and pH 8.0 are the same, although the enzymatic activity at pH 6.5 is 6 times higher than at pH 8.0. The results thus further support our current understanding that the Fe(IV)=O heme species is the active species in the IDO reaction cycle, although its presence was unexpected. The Fe-O2 and the O-O stretching frequencies of the IDO-Trp-O2 ternary complex at Trp concentrations of 50 microM and 8 mM are essentially identical. These results suggest that "substrate inhibition" of enzymatic activity occurs by binding of a second substrate molecule to an unknown binding site and not to the heme pocket.
Collapse
Affiliation(s)
- Sachiko Yanagisawa
- Picobiology Institute, Graduate School of Life Science, University of Hyogo, Koto 3-2-1, Kamigori, Ako, Hyogo 678-1297, Japan
| | | | | | | | | | | |
Collapse
|
42
|
Vascular expression, activity and function of indoleamine 2,3-dioxygenase-1 following cerebral ischaemia-reperfusion in mice. Naunyn Schmiedebergs Arch Pharmacol 2011; 383:471-81. [PMID: 21359968 DOI: 10.1007/s00210-011-0611-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Accepted: 02/02/2011] [Indexed: 01/24/2023]
Abstract
Indoleamine 2,3-dioxygenases-1 (Ido1) and -2 initiate the kynurenine pathway of tryptophan metabolism. In addition to the established immune regulatory effects of Ido1 and the ability of nitric oxide to regulate Ido1 activity, it is now also known that Ido1-mediated metabolism of tryptophan to kynurenine can modulate vascular tone. Ido activity is reportedly elevated in stroke patients and correlates with increased risk of death. Thus, the present goals were to test whether, following cerebral ischaemia, Ido activity and cerebrovascular Ido1 expression are altered and whether expression of Ido1 contributes to stroke outcome. Transient cerebral ischaemia was induced in wild-type and Ido1 gene-deficient (Ido1 (-/-)) mice. Mice were pre-treated with vehicle, the Ido1 inhibitor, 1-methyl-D-tryptophan (1-MT; 50 mg/kg i.p.) or the inducible nitric oxide synthase (Nos2) inhibitor, aminoguanidine (AG, 100 mg/kg i.p.). At 24 h, neurological function, brain infarct size and swelling were assessed. In addition, Ido activity was estimated by plasma kynurenine and tryptophan, and Ido1 expression was examined in cerebral arterioles. Cerebral ischaemia-reperfusion in wild-type mice increased Ido activity and its expression in cerebral arterioles. Ido1 (-/-) and 1-MT-treated wild-type mice had lower Ido activity but similar post-stroke neurological function and similar total brain infarct volume and swelling, relative to control mice. Inhibition of Nos2 with AG also did not affect Ido activity or outcome following stroke. This study provides molecular and pharmacological evidence that the expression and the activity of Ido1 increase following stroke. However, such Ido1 expression does not appear to affect overall outcome following acute ischaemic stroke, and furthermore, a regulatory role of Nos2-derived nitric oxide on Ido activity following cerebral ischaemia-reperfusion appears unlikely.
Collapse
|
43
|
Wink DA, Hines HB, Cheng RYS, Switzer CH, Flores-Santana W, Vitek MP, Ridnour LA, Colton CA. Nitric oxide and redox mechanisms in the immune response. J Leukoc Biol 2011; 89:873-91. [PMID: 21233414 DOI: 10.1189/jlb.1010550] [Citation(s) in RCA: 499] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The role of redox molecules, such as NO and ROS, as key mediators of immunity has recently garnered renewed interest and appreciation. To regulate immune responses, these species trigger the eradication of pathogens on the one hand and modulate immunosuppression during tissue-restoration and wound-healing processes on the other. In the acidic environment of the phagosome, a variety of RNS and ROS is produced, thereby providing a cauldron of redox chemistry, which is the first line in fighting infection. Interestingly, fluctuations in the levels of these same reactive intermediates orchestrate other phases of the immune response. NO activates specific signal transduction pathways in tumor cells, endothelial cells, and monocytes in a concentration-dependent manner. As ROS can react directly with NO-forming RNS, NO bioavailability and therefore, NO response(s) are changed. The NO/ROS balance is also important during Th1 to Th2 transition. In this review, we discuss the chemistry of NO and ROS in the context of antipathogen activity and immune regulation and also discuss similarities and differences between murine and human production of these intermediates.
Collapse
Affiliation(s)
- David A Wink
- Radiation Biology Branch, National Cancer Institute/National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Dolušić E, Larrieu P, Blanc S, Sapunaric F, Norberg B, Moineaux L, Colette D, Stroobant V, Pilotte L, Colau D, Ferain T, Fraser G, Galleni M, Galeni M, Frère JM, Masereel B, Van den Eynde B, Wouters J, Frédérick R. Indol-2-yl ethanones as novel indoleamine 2,3-dioxygenase (IDO) inhibitors. Bioorg Med Chem 2010; 19:1550-61. [PMID: 21269836 DOI: 10.1016/j.bmc.2010.12.032] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 12/07/2010] [Accepted: 12/13/2010] [Indexed: 01/24/2023]
Abstract
Indoleamine 2,3-dioxygenase (IDO) is a heme dioxygenase which has been shown to be involved in the pathological immune escape of diseases such as cancer. The synthesis and structure-activity relationships (SAR) of a novel series of IDO inhibitors based on the indol-2-yl ethanone scaffold is described. In vitro and in vivo biological activities have been evaluated, leading to compounds with IC(50) values in the micromolar range in both tests. Introduction of small substituents in the 5- and 6-positions of the indole ring, indole N-methylation and variations of the aromatic side chain are all well tolerated. An iron coordinating group on the linker is a prerequisite for biological activity, thus corroborating the virtual screening results.
Collapse
Affiliation(s)
- Eduard Dolušić
- Drug design and Discovery Center, University of Namur, 61 Rue de Bruxelles, 5000 Namur, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Induction of regulatory T cells by macrophages is dependent on production of reactive oxygen species. Proc Natl Acad Sci U S A 2010; 107:17686-91. [PMID: 20861446 DOI: 10.1073/pnas.1012016107] [Citation(s) in RCA: 205] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The phagocyte NAPDH-oxidase complex consists of several phagocyte oxidase (phox) proteins, generating reactive oxygen species (ROS) upon activation. ROS are involved in the defense against microorganisms and also in immune regulation. Defective ROS formation leads to chronic granulomatous disease (CGD) with increased incidence of autoimmunity and disturbed resolution of inflammation. Because regulatory T cells (Tregs) suppress autoimmune T-cell responses and are crucial in down-regulating immune responses, we hypothesized that ROS deficiency may lead to decreased Treg induction. Previously, we showed that in p47(phox)-mutated mice, reconstitution of macrophages (Mph) with ROS-producing capacity was sufficient to protect the mice from arthritis. Now, we present evidence that Mph-derived ROS induce Tregs. In vitro, we showed that Mph ROS-dependently induce Treg, using an NADPH-oxidase inhibitor. This finding was confirmed genetically: rat or human CGD Mph with mutated p47(phox) or gp91(phox) displayed hampered Treg induction and T-cell suppression. However, basal Treg numbers in these subjects were comparable to those in controls, indicating a role for ROS in induction of peripheral Tregs. Induction of allogeneic delayed-type hypersensitivity with p47(phox)-mutated Mph confirmed the importance of Mph-derived ROS in Treg induction in vivo. We conclude that NAPDH oxidase activity in Mph is important for the induction of Tregs to regulate T cell-mediated inflammation.
Collapse
|
46
|
De Ravin SS, Zarember KA, Long-Priel D, Chan KC, Fox SD, Gallin JI, Kuhns DB, Malech HL. Tryptophan/kynurenine metabolism in human leukocytes is independent of superoxide and is fully maintained in chronic granulomatous disease. Blood 2010; 116:1755-60. [PMID: 20511543 PMCID: PMC2947395 DOI: 10.1182/blood-2009-07-233734] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 05/03/2010] [Indexed: 11/20/2022] Open
Abstract
In chronic granulomatous disease (CGD), defective phagocytic nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity causes reduced superoxide anion (O(2)(·)) radical production leading to frequent infections as well as granulomas and impaired wound healing indicative of excessive inflammation. Based on recent mouse studies, the lack of O(2)(·)-dependent interferon γ (IFNγ)-induced synthesis of kynurenine (kyn), an anti-inflammatory tryptophan metabolite produced by indolamine 2,3 deoxygenase (IDO), was proposed as a cause of hyperinflammation in CGD and this pathway has been considered for clinical intervention. Here, we show that IFNγ induces normal levels of kynurenine in cultures of O(2)(·)-deficient monocytes, dendritic cells, and polymorphonuclear leukocytes from gp91(PHOX)- or p47(PHOX)-deficient human CGD donors. Kynurenine accumulation was dose- and time-dependent as was that of a downstream metabolite, anthranilic acid. Furthermore, urinary and serum levels of kynurenine and a variety of other tryptophan metabolites were elevated rather than suppressed in CGD donors. Although we did not specifically evaluate kyn metabolism in local tissue or inflamed sites in humans, our data demonstrates that O(2)(·) anion is dispensable for the rate-limiting step in tryptophan degradation, and CGD patients do not appear to have either hematopoietic cell or systemic deficits in the production of the anti-inflammatory kynurenine molecule.
Collapse
MESH Headings
- Cells, Cultured
- Chromatography, Liquid
- Dose-Response Relationship, Drug
- Granulomatous Disease, Chronic/blood
- Granulomatous Disease, Chronic/genetics
- Granulomatous Disease, Chronic/urine
- Humans
- Immunoblotting
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Interferon-gamma/pharmacology
- Kinetics
- Kynurenine/metabolism
- Leukocytes/cytology
- Leukocytes/drug effects
- Leukocytes/metabolism
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Monocytes/cytology
- Monocytes/drug effects
- Monocytes/metabolism
- NADPH Oxidase 2
- NADPH Oxidases/deficiency
- NADPH Oxidases/genetics
- Neutrophils/cytology
- Neutrophils/drug effects
- Neutrophils/metabolism
- Spectrometry, Mass, Electrospray Ionization
- Superoxides/metabolism
- Time Factors
- Tryptophan/metabolism
- Tryptophan/urine
Collapse
Affiliation(s)
- Suk See De Ravin
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Affiliation(s)
- Laura Crisa
- Department of Medicine, Diabetes and Obesity Center of Excellence, and the Institute of Stem Cells and Regenerative Medicine, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
48
|
Huang L, Baban B, Johnson BA, Mellor AL. Dendritic cells, indoleamine 2,3 dioxygenase and acquired immune privilege. Int Rev Immunol 2010; 29:133-55. [PMID: 20367139 DOI: 10.3109/08830180903349669] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Dendritic cells (DCs) are specialized to stimulate T cell immunity. Paradoxically, some DCs suppress T cell responses and activate regulatory T cells. In this review, we focus on a potent counter-regulatory pathway mediated by plasmacytoid DCs (pDCs) expressing the immunosuppressive enzyme indoleamine 2,3 dioxygenase (IDO). IDO-expressing pDCs inhibit effector T cell responses, activate regulatory T cells, and attenuate pro-inflammatory responses in settings of chronic inflammation that manifest in clinical syndromes, such as infectious, allergic, and autoimmune diseases; cancer; and transplantation. Thus, IDO-expressing pDCs create immune privilege and provide novel opportunities to improve immunotherapy in multiple disease syndromes.
Collapse
Affiliation(s)
- Lei Huang
- Immunotherapy Center and Department of Radiology, Medical College of Georgia, Augusta, GA, USA
| | | | | | | |
Collapse
|
49
|
Grimm MJ, Vethanayagam RR, Almyroudis NG, Lewandowski D, Rall N, Blackwell TS, Segal BH. Role of NADPH oxidase in host defense against aspergillosis. Med Mycol 2010; 49 Suppl 1:S144-9. [PMID: 20560866 DOI: 10.3109/13693786.2010.487077] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
NADPH oxidase plays a critical role in antimicrobial host defense, as evident in chronic granulomatous disease (CGD), an inherited disorder of the NADPH oxidase characterized by severe bacterial and fungal diseases. Invasive aspergillosis and other moulds are the major cause of mortality in CGD. We also learn from CGD patients that NADPH oxidase plays an important role in regulating inflammation; CGD patients are prone to developing inflammatory diseases such as inflammatory bowel disease, obstructive granulomata of the genitourinary tract, and hypersensitivity pneumonitis. Indeed, the NADPH oxidase plays an essential role in calibrating innate and T-cell responses to control the growth of inhaled fungi while protecting against excessive and injurious inflammation. Knowledge gained on the mechanisms by which NADPH oxidase kills fungi and regulates inflammation may lead to new therapeutics for CGD and will have broad relevance to understanding host-pathogen interactions between mammals and ubiquitous moulds to which we are continually exposed.
Collapse
Affiliation(s)
- Melissa J Grimm
- Department of Medicine, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Intact indoleamine 2,3-dioxygenase activity in human chronic granulomatous disease. Clin Immunol 2010; 137:1-4. [PMID: 20570568 PMCID: PMC2946558 DOI: 10.1016/j.clim.2010.05.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 05/12/2010] [Accepted: 05/12/2010] [Indexed: 12/26/2022]
Abstract
Chronic granulomatous disease (CGD) is characterized by a disability to produce reactive oxygen intermediates (ROI) caused by a defect of phagocyte nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. A hyperinflammatory response to immune activation has been reported to contribute to the pathology of CGD. The tryptophan catabolizing enzyme indoleamine 2,3-dioxygenase (IDO) is considered critical for regulating immune responses and suppression of inflammation. IDO is generally believed to require ROI for enzymatic activity and was found to be inactive in murine CGD. Here, we report that, strikingly, in human CGD IDO metabolic activity is intact. Monocyte-derived dendritic cells generated from CGD patients, harbouring X-linked and autosomal recessive forms of CGD, and from healthy controls produced similar amounts of the tryptophan metabolite kynurenine upon activation with lipopolysaccharide and interferon-γ. Thus, in humans, ROI apparently are dispensable for IDO activity. Hyperinflammation in human CGD cannot be attributed to disabled IDO activation.
Collapse
|