1251
|
AhR sensing of bacterial pigments regulates antibacterial defence. Nature 2014; 512:387-92. [PMID: 25119038 DOI: 10.1038/nature13684] [Citation(s) in RCA: 288] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 07/17/2014] [Indexed: 12/24/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a highly conserved ligand-dependent transcription factor that senses environmental toxins and endogenous ligands, thereby inducing detoxifying enzymes and modulating immune cell differentiation and responses. We hypothesized that AhR evolved to sense not only environmental pollutants but also microbial insults. We characterized bacterial pigmented virulence factors, namely the phenazines from Pseudomonas aeruginosa and the naphthoquinone phthiocol from Mycobacterium tuberculosis, as ligands of AhR. Upon ligand binding, AhR activation leads to virulence factor degradation and regulated cytokine and chemokine production. The relevance of AhR to host defence is underlined by heightened susceptibility of AhR-deficient mice to both P. aeruginosa and M. tuberculosis. Thus, we demonstrate that AhR senses distinct bacterial virulence factors and controls antibacterial responses, supporting a previously unidentified role for AhR as an intracellular pattern recognition receptor, and identify bacterial pigments as a new class of pathogen-associated molecular patterns.
Collapse
|
1252
|
Ott M, Litzenburger UM, Rauschenbach KJ, Bunse L, Ochs K, Sahm F, Pusch S, Opitz CA, Blaes J, von Deimling A, Wick W, Platten M. Suppression of TDO-mediated tryptophan catabolism in glioblastoma cells by a steroid-responsive FKBP52-dependent pathway. Glia 2014; 63:78-90. [PMID: 25132599 DOI: 10.1002/glia.22734] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 07/17/2014] [Indexed: 02/04/2023]
Abstract
Tryptophan catabolism is increasingly recognized as a key and druggable molecular mechanism active in cancer, immune, and glioneural cells and involved in the modulation of antitumor immunity, autoimmunity and glioneural function. In addition to the pivotal rate limiting enzyme indoleamine-2,3-dioxygenase, expression of tryptophan-2,3-dioxygenase (TDO) has recently been described as an alternative pathway responsible for constitutive tryptophan degradation in malignant gliomas and other types of cancer. In addition, TDO has been implicated as a key regulator of neurotoxicity involved in neurodegenerative diseases and ageing. The pathways regulating TDO expression, however, are largely unknown. Here, a siRNA-based transcription factor profiling in human glioblastoma cells revealed that the expression of human TDO is suppressed by endogenous glucocorticoid signaling. Similarly, treatment of glioblastoma cells with the synthetic glucocorticoid dexamethasone led to a reduction of TDO expression and activity in vitro and in vivo. TDO inhibition was dependent on the immunophilin FKBP52, whose FK1 domain physically interacted with the glucocorticoid receptor as demonstrated by bimolecular fluorescence complementation and in situ proximity ligation assays. Accordingly, gene expression profile analyses revealed negative correlation of FKBP52 and TDO in glial and neural tumors and in normal brain. Knockdown of FKBP52 and treatment with the FK-binding immunosuppressant FK506 enhanced TDO expression and activity in glioblastoma cells. In summary, we identify a novel steroid-responsive FKBP52-dependent pathway suppressing the expression and activity of TDO, a central and rate-limiting enzyme in tryptophan metabolism, in human gliomas.
Collapse
Affiliation(s)
- Martina Ott
- German Cancer Consortium (DKTK) Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurooncology, University Hospital Heidelberg and National Center for Tumor Diseases, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1253
|
Genestet C, Le Gouellec A, Chaker H, Polack B, Guery B, Toussaint B, Stasia MJ. Scavenging of reactive oxygen species by tryptophan metabolites helps Pseudomonas aeruginosa escape neutrophil killing. Free Radic Biol Med 2014; 73:400-10. [PMID: 24929180 DOI: 10.1016/j.freeradbiomed.2014.06.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 06/03/2014] [Accepted: 06/04/2014] [Indexed: 10/25/2022]
Abstract
Pseudomonas aeruginosa is responsible for persistent infections in cystic fibrosis patients, suggesting an ability to circumvent innate immune defenses. This bacterium uses the kynurenine pathway to catabolize tryptophan. Interestingly, many host cells also produce kynurenine, which is known to control immune system homeostasis. We showed that most strains of P. aeruginosa isolated from cystic fibrosis patients produce a high level of kynurenine. Moreover, a strong transcriptional activation of kynA (the first gene involved in the kynurenine pathway) was observed upon contact with immune cells and particularly with neutrophils. In addition, using coculture of human neutrophils with various strains of P. aeruginosa producing no (ΔkynA) or a high level of kynurenine (ΔkynU or ΔkynA pkynA), we demonstrated that kynurenine promotes bacterial survival. In addition, increasing the amount kynurenine inhibits reactive oxygen species production by activated neutrophils, as evaluated by chemiluminescence with luminol or isoluminol or SOD-sensitive cytochrome c reduction assay. This inhibition is due neither to a phagocytosis defect nor to direct NADPH oxidase inhibition. Indeed, kynurenine has no effect on oxygen consumption by neutrophils activated by PMA or opsonized zymosan. Using in vitro reactive oxygen species-producing systems, we showed that kynurenine scavenges hydrogen peroxide and, to a lesser extent, superoxide. Kynurenine׳s scavenging effect occurs mainly intracellularly after bacterial stimulation, probably in the phagosome. In conclusion, the kynurenine pathway allows P. aeruginosa to circumvent the innate immune response by scavenging neutrophil reactive oxygen species production.
Collapse
Affiliation(s)
- Charlotte Genestet
- TIMC/Therex Laboratory, UMR 5525 (CNRS-UJF), Faculty of Medicine, University of Grenoble Alpes, Grenoble F-38041, France
| | - Audrey Le Gouellec
- TIMC/Therex Laboratory, UMR 5525 (CNRS-UJF), Faculty of Medicine, University of Grenoble Alpes, Grenoble F-38041, France
| | - Hichem Chaker
- TIMC/Therex Laboratory, UMR 5525 (CNRS-UJF), Faculty of Medicine, University of Grenoble Alpes, Grenoble F-38041, France
| | - Benoit Polack
- TIMC/Therex Laboratory, UMR 5525 (CNRS-UJF), Faculty of Medicine, University of Grenoble Alpes, Grenoble F-38041, France
| | - Benoit Guery
- Recherche translationnelle hôte pathogène, Université Lille 2, Faculté de Médecine, CHRU, Lille, France
| | - Bertrand Toussaint
- TIMC/Therex Laboratory, UMR 5525 (CNRS-UJF), Faculty of Medicine, University of Grenoble Alpes, Grenoble F-38041, France
| | - Marie José Stasia
- TIMC/Therex Laboratory, UMR 5525 (CNRS-UJF), Faculty of Medicine, University of Grenoble Alpes, Grenoble F-38041, France; Chronic Granulomatous Disease Diagnosis and Research Center, Pôle Biologie, CHU de Grenoble, Grenoble F-38043, France.
| |
Collapse
|
1254
|
Wang T, Liu G, Wang R. The Intercellular Metabolic Interplay between Tumor and Immune Cells. Front Immunol 2014; 5:358. [PMID: 25120544 PMCID: PMC4112791 DOI: 10.3389/fimmu.2014.00358] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 07/12/2014] [Indexed: 01/14/2023] Open
Abstract
Functional and effective immune response requires a metabolic rewiring of immune cells to meet their energetic and anabolic demands. Beyond this, the availability of extracellular and intracellular metabolites may serve as metabolic signals interconnecting with cellular signaling events to influence cellular fate and immunological function. As such, tumor microenvironment represents a dramatic example of metabolic derangement, where the highly metabolic demanding tumor cells may compromise the function of some immune cells by competing nutrients (a form of intercellular competition), meanwhile may support the function of other immune cells by forming a metabolic symbiosis (a form of intercellular collaboration). It has been well known that tumor cells harness immune system through information exchanges that are largely attributed to soluble protein factors and intercellular junctions. In this review, we will discuss recent advance on tumor metabolism and immune metabolism, as well as provide examples of metabolic communications between tumor cells and immune system, which may represent a novel mechanism of conveying tumor-immune privilege.
Collapse
Affiliation(s)
- Tingting Wang
- Center for Childhood Cancer and Blood Disease, The Research Institute at Nationwide Children's Hospital , Columbus, OH , USA
| | - Guangwei Liu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Immunology, School of Basic Medical Sciences, Fudan University , Shanghai , China ; Biotherapy Research Center, Fudan University , Shanghai , China
| | - Ruoning Wang
- Center for Childhood Cancer and Blood Disease, The Research Institute at Nationwide Children's Hospital , Columbus, OH , USA ; Hematology/Oncology & BMT, The Research Institute at Nationwide Children's Hospital , Columbus, OH , USA ; Department of Pediatrics, The Ohio State University School of Medicine , Columbus, OH , USA
| |
Collapse
|
1255
|
Chen JY, Li CF, Kuo CC, Tsai KK, Hou MF, Hung WC. Cancer/stroma interplay via cyclooxygenase-2 and indoleamine 2,3-dioxygenase promotes breast cancer progression. Breast Cancer Res 2014; 16:410. [PMID: 25060643 PMCID: PMC4220086 DOI: 10.1186/s13058-014-0410-1] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 07/10/2014] [Indexed: 11/23/2022] Open
Abstract
Introduction Expression of indoleamine 2,3-dioxygenase (IDO) in primary breast cancer increases tumor growth and metastasis. However, the clinical significance of stromal IDO and the regulation of stromal IDO are unclear. Methods Metabolomics and enzyme-linked immunosorbent assay (ELISA) were used to study the effect of cyclooxygenase-2 (COX-2)-overexpressing breast cancer cells on IDO expression in co-cultured human breast fibroblasts. Biochemical inhibitors and short-hairpin RNA (shRNA) were used to clarify how prostaglandin E2 (PGE2) upregulates IDO expression. Associations of stromal IDO with clinicopathologic parameters were tested in tumor specimens. An orthotopic animal model was used to examine the effect of COX-2 and IDO inhibitors on tumor growth. Results Kynurenine, the metabolite generated by IDO, increases in the supernatant of fibroblasts co-cultured with COX-2-overexpressing breast cancer cells. PGE2 released by cancer cells upregulates IDO expression in fibroblasts through an EP4/signal transducer and activator of transcription 3 (STAT3)-dependent pathway. Conversely, fibroblast-secreted kynurenine promotes the formation of the E-cadherin/Aryl hydrocarbon receptor (AhR)/S-phase kinase-associated protein 2 (Skp2) complex, resulting in degradation of E-cadherin to increase breast cancer invasiveness. The enhancement of motility of breast cancer cells induced by co-culture with fibroblasts is suppressed by the IDO inhibitor 1-methyl-tryptophan. Pathological analysis demonstrates that upregulation of stromal IDO is a poor prognosis factor and is associated with of COX-2 overexpression. Co-expression of cancer COX-2 and stromal IDO predicts a worse disease-free and metastasis-free survival. Finally, COX-2 and IDO inhibitors inhibit tumor growth in vivo. Conclusion Integration of metabolomics and molecular and pathological approaches reveals the interplay between cancer and stroma via COX-2, and IDO promotes tumor progression and predicts poor patient survival. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0410-1) contains supplementary material, which is available to authorized users.
Collapse
|
1256
|
Chen IC, Chiang WF, Chen PF, Chiang HC. STRESS-responsive deacetylase SIRT3 is up-regulated by areca nut extract-induced oxidative stress in human oral keratinocytes. J Cell Biochem 2014; 115:328-39. [PMID: 24339251 DOI: 10.1002/jcb.24667] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 09/06/2013] [Indexed: 12/13/2022]
Abstract
Areca chewing is an important environmental risk factor for development of oral premalignant lesions and cancer. Epidemiological evidence indicates that areca chewing is tightly linked to oral carcinogenesis. However, the pathogenetic impacts of areca nut extract (ANE) on normal human oral keratinocytes (HOKs) are unclear and possibly involve oxidative stress via redox imbalance. Sirtuin 3 (SIRT3) is a member of the sirtuin family of proteins that play an important role in regulating cellular reactive oxygen species (ROS) production. Recent studies have confirmed that ANE and other areca ingredients can induce ROS. In this study, we examined the role of SIRT3 in the regulation of ANE-induced ROS in HOK cells. We examined HOK cell viability following treatment with various ANE concentrations. ANE-induced cytotoxicity increased in a dose-dependent manner and was approximately 48% at a concentration of 50 μg/ml after 24 h. SIRT3 expression and enzyme activity were up-regulated in HOK cells by ANE-induced oxidative stress. Additionally, we identified that SIRT3 controls the enzymatic activity of mitochondrial proteins, such as forkhead box O3a (Foxo3a) transcription factor and antioxidant-encoding gene superoxide dismutase 2 (SOD2), by deacetylation in HOK cells. Moreover, SIRT3-mediated deacetylation and activation of Foxo3a promotes nuclear localization in vivo. These findings suggest that SIRT3 is an endogenous negative regulator in response to ANE-induced oxidative stress and demonstrate an essential role for redox balance in HOK cells.
Collapse
Affiliation(s)
- I-Chieh Chen
- Division of Environmental Health and Occupational Medicine, National Health Research Institutes, Miaoli, Taiwan
| | | | | | | |
Collapse
|
1257
|
Gratz IK, Campbell DJ. Organ-specific and memory treg cells: specificity, development, function, and maintenance. Front Immunol 2014; 5:333. [PMID: 25076948 PMCID: PMC4098124 DOI: 10.3389/fimmu.2014.00333] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 06/30/2014] [Indexed: 12/17/2022] Open
Abstract
Foxp3+ regulatory T cells (Treg cells) are essential for establishing and maintaining self-tolerance, and also inhibit immune responses to innocuous environmental antigens. Imbalances and dysfunction in Treg cells lead to a variety of immune-mediated diseases, as deficits in Treg cell function contribute to the development autoimmune disease and pathological tissue damage, whereas overabundance of Treg cells can promote chronic infection and tumorigenesis. Recent studies have highlighted the fact that Treg cells themselves are a diverse collection of phenotypically and functionally specialized populations, with distinct developmental origins, antigen-specificities, tissue-tropisms, and homeostatic requirements. The signals directing the differentiation of these populations, their specificities and the mechanisms by which they combine to promote organ-specific and systemic tolerance, and how they embody the emerging property of regulatory memory are the focus of this review.
Collapse
Affiliation(s)
- Iris K Gratz
- Department of Molecular Biology, University of Salzburg , Salzburg , Austria ; Department of Dermatology, University of California San Francisco , San Francisco, CA , USA ; Division of Molecular Dermatology and EB House Austria, Department of Dermatology, Paracelsus Medical University , Salzburg , Austria
| | - Daniel J Campbell
- Immunology Program, Benaroya Research Institute , Seattle, WA , USA ; Department of Immunology, University of Washington School of Medicine , Seattle, WA , USA
| |
Collapse
|
1258
|
Röhrig UF, Majjigapu SR, Chambon M, Bron S, Pilotte L, Colau D, Van den Eynde BJ, Turcatti G, Vogel P, Zoete V, Michielin O. Detailed analysis and follow-up studies of a high-throughput screening for indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors. Eur J Med Chem 2014; 84:284-301. [PMID: 25036789 DOI: 10.1016/j.ejmech.2014.06.078] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/26/2014] [Accepted: 06/27/2014] [Indexed: 01/28/2023]
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) is a key regulator of immune responses and therefore an important therapeutic target for the treatment of diseases that involve pathological immune escape, such as cancer. Here, we describe a robust and sensitive high-throughput screen (HTS) for IDO1 inhibitors using the Prestwick Chemical Library of 1200 FDA-approved drugs and the Maybridge HitFinder Collection of 14,000 small molecules. Of the 60 hits selected for follow-up studies, 14 displayed IC50 values below 20 μM under the secondary assay conditions, and 4 showed an activity in cellular tests. In view of the high attrition rate we used both experimental and computational techniques to identify and to characterize compounds inhibiting IDO1 through unspecific inhibition mechanisms such as chemical reactivity, redox cycling, or aggregation. One specific IDO1 inhibitor scaffold, the imidazole antifungal agents, was chosen for rational structure-based lead optimization, which led to more soluble and smaller compounds with micromolar activity.
Collapse
Affiliation(s)
- Ute F Röhrig
- Swiss Institute of Bioinformatics, Molecular Modeling Group, Quartier Sorge - Bâtiment Génopode, CH-1015 Lausanne, Switzerland.
| | - Somi Reddy Majjigapu
- Swiss Institute of Bioinformatics, Molecular Modeling Group, Quartier Sorge - Bâtiment Génopode, CH-1015 Lausanne, Switzerland; Laboratory of Glycochemistry and Asymmetric Synthesis, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| | - Marc Chambon
- Biomolecular Screening Facility, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| | - Sylvian Bron
- Swiss Institute of Bioinformatics, Molecular Modeling Group, Quartier Sorge - Bâtiment Génopode, CH-1015 Lausanne, Switzerland.
| | - Luc Pilotte
- de Duve Institute and the Université catholique de Louvain, B-1200 Brussels, Belgium; Ludwig Institute for Cancer Research, B-1200 Brussels, Belgium.
| | - Didier Colau
- de Duve Institute and the Université catholique de Louvain, B-1200 Brussels, Belgium; Ludwig Institute for Cancer Research, B-1200 Brussels, Belgium.
| | - Benoît J Van den Eynde
- de Duve Institute and the Université catholique de Louvain, B-1200 Brussels, Belgium; Ludwig Institute for Cancer Research, B-1200 Brussels, Belgium.
| | - Gerardo Turcatti
- Biomolecular Screening Facility, 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.
| | - Vincent Zoete
- Swiss Institute of Bioinformatics, Molecular Modeling Group, Quartier Sorge - Bâtiment Génopode, CH-1015 Lausanne, Switzerland.
| | - Olivier Michielin
- Swiss Institute of Bioinformatics, Molecular Modeling Group, Quartier Sorge - Bâtiment Génopode, CH-1015 Lausanne, Switzerland; Department of Oncology, University of Lausanne and Centre Hospitalier Universitaire Vaudois (CHUV), CH-1011 Lausanne, Switzerland; Ludwig Center for Cancer Research of the University of Lausanne, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
1259
|
Hughes T, Briercheck EL, Freud AG, Trotta R, McClory S, Scoville SD, Keller K, Deng Y, Cole J, Harrison N, Mao C, Zhang J, Benson DM, Yu J, Caligiuri MA. The transcription Factor AHR prevents the differentiation of a stage 3 innate lymphoid cell subset to natural killer cells. Cell Rep 2014; 8:150-62. [PMID: 24953655 PMCID: PMC4133146 DOI: 10.1016/j.celrep.2014.05.042] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 04/09/2014] [Accepted: 05/21/2014] [Indexed: 01/16/2023] Open
Abstract
Accumulating evidence indicates that human natural killer (NK) cells develop in secondary lymphoid tissue (SLT) through a so-called "stage 3" developmental intermediate minimally characterized by a CD34(-)CD117(+)CD94(-) immunophenotype that lacks mature NK cell function. This stage 3 population is heterogeneous, potentially composed of functionally distinct innate lymphoid cell (ILC) types that include interleukin-1 receptor (IL-1R1)-positive, IL-22-producing ILC3s. Whether human ILC3s are developmentally related to NK cells is a subject of ongoing investigation. Here, we show that antagonism of the aryl hydrocarbon receptor (AHR) or silencing of AHR gene expression promotes the differentiation of tonsillar IL-22-producing IL-1R1(hi) human ILC3s to CD56(bright)CD94(+) interferon (IFN)-γ-producing cytolytic mature NK cells expressing eomesodermin (EOMES) and T-Box Protein 21 (TBX21 or TBET). Hence, we demonstrate the lineage plasticity of human ILCs by identifying AHR as a transcription factor that prevents IL-1R1(hi) ILC3s from differentiating into NK cells.
Collapse
Affiliation(s)
- Tiffany Hughes
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Edward L Briercheck
- Integrated Biomedical Graduate Program, Medical Scientist Program, The Ohio State University, Columbus, OH 43210, USA
| | - Aharon G Freud
- Department of Pathology, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Rossana Trotta
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Susan McClory
- Integrated Biomedical Graduate Program, Medical Scientist Program, The Ohio State University, Columbus, OH 43210, USA
| | - Steven D Scoville
- Integrated Biomedical Graduate Program, Medical Scientist Program, The Ohio State University, Columbus, OH 43210, USA
| | - Karen Keller
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Youcai Deng
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jordan Cole
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Nicholas Harrison
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Charlene Mao
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jianying Zhang
- Center for Biostatistics, The Ohio State University, Columbus, OH 43210, USA
| | - Don M Benson
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Michael A Caligiuri
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA; Integrated Biomedical Graduate Program, Medical Scientist Program, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
1260
|
Chen J, Shao J, Cai R, Shen Y, Zhang R, Liu L, Qi T, Lu H. Anti-retroviral therapy decreases but does not normalize indoleamine 2,3-dioxygenase activity in HIV-infected patients. PLoS One 2014; 9:e100446. [PMID: 24983463 PMCID: PMC4077698 DOI: 10.1371/journal.pone.0100446] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/22/2014] [Indexed: 01/23/2023] Open
Abstract
Background Indoleamine 2,3-dioxygenase (IDO), which is mainly expressed in activated dendritic cells, catabolizes tryptophan to kynurenine and other downstream catabolites. It is known to be an immune mediator in HIV pathogenesis. The impact of anti-retroviral therapy on its activity has not been well established. Methods We measured systemic IDO activity (the ratio of plasma kynurenine to tryptophan) in HIV-infected patients before and after highly active antiretroviral therapy (HAART) and its association with a microbial translocation marker, soluble CD14 (sCD14). Results Among 76 participants, higher baseline IDO activity was associated with lower CD4+ T cell counts (P<0.05) and higher plasma sCD14 levels (P<0.001). After 1 year of HAART, IDO activity decreased significantly (P<0.01), but was still higher than in healthy controls (P<0.05). The baseline IDO activity did not predict CD4+ T cell recovery after 1 year of therapy. The percentages of myeloid and plasmacytoid dendritic cells were not correlated with IDO activity. Conclusions IDO activity is elevated in HIV-infected patients, which is partially associated with microbial translocation. HAART reduced, but did not normalize the activity of IDO.
Collapse
Affiliation(s)
- Jun Chen
- Department of Infectious Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jiasheng Shao
- Department of Infectious Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Rentian Cai
- Department of Infectious Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yinzhong Shen
- Department of Infectious Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Renfang Zhang
- Department of Infectious Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Li Liu
- Department of Infectious Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Tangkai Qi
- Department of Infectious Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Hongzhou Lu
- Department of Infectious Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Department of Infectious Diseases, HuaShan Hospital, Fudan University, Shanghai, China
- Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, China
- * E-mail:
| |
Collapse
|
1261
|
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]
|
1262
|
Urata Y, Koga K, Hirota Y, Akiyama I, Izumi G, Takamura M, Nagai M, Harada M, Hirata T, Yoshino O, Kawana K, Fujii T, Osuga Y. IL-1β increases expression of tryptophan 2,3-dioxygenase and stimulates tryptophan catabolism in endometrioma stromal cells. Am J Reprod Immunol 2014; 72:496-503. [PMID: 24974860 DOI: 10.1111/aji.12282] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 05/31/2014] [Indexed: 12/31/2022] Open
Abstract
PROBLEM Immune tolerance to endometriotic cells is important to promote endometriosis. Tryptophan 2,3-dioxygenase (TDO) enhances immune tolerance by catabolizing tryptophan to kynurenine. We studied whether interleukin-1β (IL-1β), a typical endometriosis-associated cytokine, affects the expression of TDO and the catabolism of tryptophan in endometrioma stromal cells (ESCs). We also studied whether the expression of TDO is involved in IL-1β-induced secretion of IL-6 and IL-8 in ESCs. METHOD OF STUDY Nineteen endometriotic patients of reproductive age with normal menstrual cycles were recruited. Primary cultures of ESCs were treated with IL-1β and TDO siRNA. TDO mRNA was measured using quantitative PCR. TDO protein was measured using Western blotting. Concentrations of kynurenine in condition media were measured using Ehrlich reagent. Concentrations of tryptophan in conditioned media were measured using tryptophan detection kit. Concentrations of IL-6 and IL-8 in conditioned media were measured using ELISA kits. RESULTS IL-1β (1 ng/mL) increased the expression of TDO mRNA and TDO protein in ESCs. IL-1β-treated ESCs increased the production of kynurenine and the effect was inhibited by TDO siRNA. Treatment with the siRNA also decreased IL-1β-induced secretion of IL-6 and IL-8 from ESCs. CONCLUSION IL-1β is suggested to stimulate tryptophan catabolism and production of IL-6 and IL-8 by increasing TDO expression in endometriosis.
Collapse
Affiliation(s)
- Yoko Urata
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1263
|
MPINet: metabolite pathway identification via coupling of global metabolite network structure and metabolomic profile. BIOMED RESEARCH INTERNATIONAL 2014; 2014:325697. [PMID: 25057481 PMCID: PMC4095715 DOI: 10.1155/2014/325697] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/18/2014] [Indexed: 01/11/2023]
Abstract
High-throughput metabolomics technology, such as gas chromatography mass spectrometry, allows the analysis of hundreds of metabolites. Understanding that these metabolites dominate the study condition from biological pathway perspective is still a significant challenge. Pathway identification is an invaluable aid to address this issue and, thus, is urgently needed. In this study, we developed a network-based metabolite pathway identification method, MPINet, which considers the global importance of metabolites and the unique character of metabolomic profile. Through integrating the global metabolite functional network structure and the character of metabolomic profile, MPINet provides a more accurate metabolomic pathway analysis. This integrative strategy simultaneously captures the global nonequivalence of metabolites in a pathway and the bias from metabolomic experimental technology. We then applied MPINet to four different types of metabolite datasets. In the analysis of metastatic prostate cancer dataset, we demonstrated the effectiveness of MPINet. With the analysis of the two type 2 diabetes datasets, we show that MPINet has the potentiality for identifying novel pathways related with disease and is reliable for analyzing metabolomic data. Finally, we extensively applied MPINet to identify drug sensitivity related pathways. These results suggest MPINet's effectiveness and reliability for analyzing metabolomic data across multiple different application fields.
Collapse
|
1264
|
Salisbury TB, Tomblin JK, Primerano DA, Boskovic G, Fan J, Mehmi I, Fletcher J, Santanam N, Hurn E, Morris GZ, Denvir J. Endogenous aryl hydrocarbon receptor promotes basal and inducible expression of tumor necrosis factor target genes in MCF-7 cancer cells. Biochem Pharmacol 2014; 91:390-9. [PMID: 24971714 DOI: 10.1016/j.bcp.2014.06.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 06/13/2014] [Accepted: 06/16/2014] [Indexed: 01/11/2023]
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that upon activation by the toxicant 2,3,7,8 tetrachlorodibenzo-p-dioxin (TCDD) stimulates gene expression and toxicity. AHR is also important for normal mouse physiology and may play a role in cancer progression in the absence of environmental toxicants. The objective of this report was to identify AHR-dependent genes (ADGs) whose expression is regulated by AHR in the absence of toxicants. RNA-Seq analysis revealed that AHR regulated the expression of over 600 genes at an FDR<10% in MCF-7 breast cancer cells upon knockdown with short interfering RNA. Pathway analysis revealed that a significant number of ADGs were components of TCDD and tumor necrosis factor (TNF) pathways. We also demonstrated that siRNA knockdown of AHR modulated TNF induction of MNSOD and cytotoxicity in MCF-7 cells. Collectively, the major new findings of this report are: (1) endogenous AHR promotes the expression of xenobiotic metabolizing enzymes even in the absence of toxicants and drugs, (2) AHR by modulating the basal expression of a large fraction of TNF target genes may prime them for TNF stimulation and (3) AHR is required for TNF induction of MNSOD and the cellular response to cytotoxicity in MCF-7 cells. This latter result provides a potentially new role for AHR in MCF-7 cancer progression as a mediator of TNF and antioxidant responses.
Collapse
Affiliation(s)
- Travis B Salisbury
- Departments of Pharmacology, Physiology and Toxicology Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA.
| | - Justin K Tomblin
- Departments of Pharmacology, Physiology and Toxicology Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA.
| | - Donald A Primerano
- Biochemistry and Microbiology and Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA.
| | - Goran Boskovic
- Biochemistry and Microbiology and Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA
| | - Jun Fan
- Biochemistry and Microbiology and Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA.
| | - Inderjit Mehmi
- Medical Oncology and Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA
| | - Jackie Fletcher
- Department of Biology, West Virginia State University, Institute, WV 25112, USA.
| | - Nalini Santanam
- Departments of Pharmacology, Physiology and Toxicology Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA.
| | - Estil Hurn
- Departments of Pharmacology, Physiology and Toxicology Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA.
| | - Gary Z Morris
- Department of Science and Mathematics, Glenville State College, Glenville, WV 26351, USA.
| | - James Denvir
- Biochemistry and Microbiology and Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA.
| |
Collapse
|
1265
|
Endler A, Chen L, Shibasaki F. Coactivator recruitment of AhR/ARNT1. Int J Mol Sci 2014; 15:11100-10. [PMID: 24950180 PMCID: PMC4100201 DOI: 10.3390/ijms150611100] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 05/27/2014] [Accepted: 06/07/2014] [Indexed: 01/03/2023] Open
Abstract
A common feature of nuclear receptors (NRs) is the transformation of external cell signals into specific transcriptions of the signal molecule. Signal molecules function as ligands for NRs and, after their uptake, activated NRs form homo- or heterodimers at promoter recognition sequences of the specific genes in the nucleus. Another common feature of NRs is their dependence on coactivators, which bridge the basic transcriptional machinery and other cofactors to the target genes, in order to initiate transcription and to unwind histone-bound DNA for exposing additional promoter recognition sites via their histone acetyltransferase (HAT) function. In this review, we focus on our recent findings related to the recruitment of steroid receptor coactivator 1 (SRC1/NCoA1) by the estrogen receptor-α (ERα) and by the arylhydrocarbon receptor/arylhydrocarbon receptor nuclear translocator 1 (AhR/ARNT1) complex. We also describe the extension of our previously published findings regarding the binding between ARNT1.1 exon16 and SRC1e exon 21, via in silico analyses of androgen receptor (AR) NH2-carboxyl-terminal interactions, the results of which were verified by in vitro experiments. Based on these data, we suggest a newly derived tentative binding site of nuclear coactivator 2/glucocorticoid receptor interacting protein-1/transcriptional intermediary factor 2 (NCOA-2/ GRIP-1/TIF-2) for ARNT1.1 exon 16. Furthermore, results obtained by immunoprecipitation have revealed a second leucine-rich binding site for hARNT1.1 exon 16 in SRC1e exon 21 (LSSTDLL). Finally, we discuss the role of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) as an endocrine disruptor for estrogen related transcription.
Collapse
Affiliation(s)
- Alexander Endler
- Department of Molecular Medical Research, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| | - Li Chen
- Department of Molecular Medical Research, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| | - Futoshi Shibasaki
- Department of Molecular Medical Research, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| |
Collapse
|
1266
|
Zuo H, Tell GS, Vollset SE, Ueland PM, Nygård O, Midttun Ø, Meyer K, Ulvik A, Eussen SJPM. Interferon-γ-induced inflammatory markers and the risk of cancer: the Hordaland Health Study. Cancer 2014; 120:3370-7. [PMID: 24948355 PMCID: PMC4283722 DOI: 10.1002/cncr.28869] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 04/30/2014] [Accepted: 05/21/2014] [Indexed: 12/17/2022]
Abstract
BACKGROUND It has been reported that interferon-γ (IFN-γ)-induced inflammatory markers, such as circulating neopterin and kynurenine-to-tryptophan ratio (KTR), are increased in patients with cancer and are also a predictor of poor prognosis. However, whether baseline levels of these makers are associated with subsequent cancer risk in the general population remains unknown. METHODS We conducted a prospective analysis of the Hordaland Health Study in 6594 adults without known cancer at baseline who were enrolled between April 1998 and June 1999. Hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated using multivariate Cox proportional hazards regression models adjusted for sex, age, body mass index, smoking status, and renal function. RESULTS A total of 971 incident cancer cases (507 men and 464 women) were identified over a median follow-up time of 12 years. Baseline plasma neopterin, KTR and C-reactive protein (CRP) were significantly associated with an increased risk of overall cancer in models adjusted for covariates (P for trend across quartiles = .006 for neopterin, .022 for KTR, and .005 for CRP). The multivariate-adjusted HR (95% CI) per SD increment in similar models were 1.09 (1.03-1.16) for neopterin, 1.07 (1.01-1.14) for KTR, and 1.04 (0.98-1.10) for CRP. The associations between the inflammatory markers and risk of major specific cancer types were also provided. CONCLUSIONS Our findings indicate that plasma neopterin, KTR, and CRP are associated with a significantly increased risk of overall cancer. Our study revealed novel evidence regarding the role of IFN-γ-induced inflammation in human carcinogenesis.
Collapse
Affiliation(s)
- Hui Zuo
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway; Section for Pharmacology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
1267
|
Cella M, Miller H, Song C. Beyond NK cells: the expanding universe of innate lymphoid cells. Front Immunol 2014; 5:282. [PMID: 24982658 PMCID: PMC4058828 DOI: 10.3389/fimmu.2014.00282] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 05/30/2014] [Indexed: 12/20/2022] Open
Abstract
For a long time, natural killer (NK) cells were thought to be the only innate immune lymphoid population capable of responding to invading pathogens under the influence of changing environmental cues. In the last few years, an increasing amount of evidence has shown that a number of different innate lymphoid cell (ILC) populations found at mucosal sites rapidly respond to locally produced cytokines in order to establish or maintain homeostasis. These ILC populations closely mirror the phenotype of adaptive T helper subsets in their repertoire of secreted soluble factors. Early in the immune response, ILCs are responsible for setting the stage to mount an adaptive T cell response that is appropriate for the incoming insult. Here, we review the diversity of ILC subsets and discuss similarities and differences between ILCs and NK cells in function and key transcriptional factors required for their development.
Collapse
Affiliation(s)
- Marina Cella
- Department of Pathology and Immunology, Washington University School of Medicine , St. Louis, MO , USA
| | - Hannah Miller
- Department of Pathology and Immunology, Washington University School of Medicine , St. Louis, MO , USA
| | - Christina Song
- Department of Pathology and Immunology, Washington University School of Medicine , St. Louis, MO , USA
| |
Collapse
|
1268
|
Depletion of the aryl hydrocarbon receptor in MDA-MB-231 human breast cancer cells altered the expression of genes in key regulatory pathways of cancer. PLoS One 2014; 9:e100103. [PMID: 24932473 PMCID: PMC4059751 DOI: 10.1371/journal.pone.0100103] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 05/22/2014] [Indexed: 11/19/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR), a transcription factor that is best known for its role in mediating the toxic responses elicited by poly aromatic hydrocarbons as well as many other environmental factors; is also involved in breast cancer progression. We previously reported that stable knockdown of AhR decreased the tumorigenic properties of the highly metastatic MDA-MB-231 breast cancer cell line; whereas ectopic overexpression of AhR was sufficient to transform immortalized human mammary epithelial cells to exhibit malignant phenotypes. In the present study we investigated the genes that are differentially regulated by AhR and are controlling cellular processes linked to breast cancer. We used Affymetrix Human GeneChip 1.0-ST whole transcriptome arrays to analyze alterations of gene expression resulting from stable AhR knockdown in the MDA-MB-231 breast cancer cell line. The expression of 144 genes was significantly altered with a ≥2.0-fold change and a multiple test corrected p-value ≤0.05, as a result of AhR knockdown. We demonstrate that AhR knockdown alters the expression of several genes known to be linked to cancer. These genes include those involved in tryptophan metabolism (KYNU), cell growth (MUC1 and IL8), cell survival (BIRC3 and BCL3), cell migration and invasion (S100A4 and ABI3), multi-drug resistance (ABCC3) and angiogenesis (VEGFA and CCL2). The identification of the genes and pathways affected by AhR depletion provides new insight into possible molecular events that could explain the reported phenotypic changes. In conclusion AhR knockdown alters the expression of genes known to enhance or inhibit cancer progression; tipping the balance towards a state that counteracts tumor progression.
Collapse
|
1269
|
Bjerregaard-Andersen K, Sommer T, Jensen JK, Jochimsen B, Etzerodt M, Morth JP. A proton wire and water channel revealed in the crystal structure of isatin hydrolase. J Biol Chem 2014; 289:21351-9. [PMID: 24917679 DOI: 10.1074/jbc.m114.568824] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The high resolution crystal structures of isatin hydrolase from Labrenzia aggregata in the apo and the product state are described. These are the first structures of a functionally characterized metal-dependent hydrolase of this fold. Isatin hydrolase converts isatin to isatinate and belongs to a novel family of metalloenzymes that include the bacterial kynurenine formamidase. The product state, mimicked by bound thioisatinate, reveals a water molecule that bridges the thioisatinate to a proton wire in an adjacent water channel and thus allows the proton released by the reaction to escape only when the product is formed. The functional proton wire present in isatin hydrolase isoform b represents a unique catalytic feature common to all hydrolases is here trapped and visualized for the first time. The local molecular environment required to coordinate thioisatinate allows stronger and more confident identification of orthologous genes encoding isatin hydrolases within the prokaryotic kingdom. The isatin hydrolase orthologues found in human gut bacteria raise the question as to whether the indole-3-acetic acid degradation pathway is present in human gut flora.
Collapse
Affiliation(s)
- Kaare Bjerregaard-Andersen
- From the Norwegian Center of Molecular Medicine, Nordic EMBL Partnership University of Oslo, Gaustadalléen 21, 0349 Oslo, Norway, the Department for Molecular Biology and Genetics, Aarhus University, Gustav Wieds vej 10C, DK-8000 Aarhus, Denmark
| | - Theis Sommer
- From the Norwegian Center of Molecular Medicine, Nordic EMBL Partnership University of Oslo, Gaustadalléen 21, 0349 Oslo, Norway
| | - Jan K Jensen
- the Department for Molecular Biology and Genetics, Aarhus University, Gustav Wieds vej 10C, DK-8000 Aarhus, Denmark
| | - Bjarne Jochimsen
- the Department for Molecular Biology and Genetics, Aarhus University, Gustav Wieds vej 10C, DK-8000 Aarhus, Denmark
| | - Michael Etzerodt
- the Department for Molecular Biology and Genetics, Aarhus University, Gustav Wieds vej 10C, DK-8000 Aarhus, Denmark
| | - J Preben Morth
- From the Norwegian Center of Molecular Medicine, Nordic EMBL Partnership University of Oslo, Gaustadalléen 21, 0349 Oslo, Norway, the Institute for Experimental Medical Research, Oslo University Hospital, N-0424 Oslo, Norway, and
| |
Collapse
|
1270
|
Zeng J, Yin P, Tan Y, Dong L, Hu C, Huang Q, Lu X, Wang H, Xu G. Metabolomics study of hepatocellular carcinoma: discovery and validation of serum potential biomarkers by using capillary electrophoresis-mass spectrometry. J Proteome Res 2014; 13:3420-31. [PMID: 24853826 DOI: 10.1021/pr500390y] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal malignancies. The lack of effective screening methods for early diagnosis has been a longstanding bottleneck to improve the survival rate. In the present study, a capillary electrophoresis-time-of-flight mass spectrometry (CE-TOF/MS)-based metabolomics method was employed to discover novel biomarkers for HCC. A total of 183 human serum specimens (77 sera in discovery set and 106 sera in external validation set) were enrolled in this study, and a "serum biomarker model" including tryptophan, glutamine, and 2-hydroxybutyric acid was finally established based on the comprehensive screening and validation workflow. This model was evaluated as an effective tool in that area under the receiver operating characteristic curve reached 0.969 in the discovery set and 0.99 in the validation set for diagnosing HCC from non-HCC (health and cirrhosis). Furthermore, this model enabled the discrimination of small HCC from precancer cirrhosis with an AUC of 0.976, highlighting the potential of early diagnosis. The biomarker model is effective for those a-fetoprotein (AFP) false-negative and false-postive subjects, indicating the complementary function to conventional tumor marker AFP. This study demonstrates the promising potential of CE-MS-based metabolomics approach in finding biomarkers for disease diagnosis and providing special insights into tumor metabolism.
Collapse
Affiliation(s)
- Jun Zeng
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences , 457 Zhongshan Road, Dalian 116023, China
| | | | | | | | | | | | | | | | | |
Collapse
|
1271
|
Yu TY, Kondo T, Matsumoto T, Fujii-Kuriyama Y, Imai Y. Aryl hydrocarbon receptor catabolic activity in bone metabolism is osteoclast dependent in vivo. Biochem Biophys Res Commun 2014; 450:416-22. [PMID: 24938130 DOI: 10.1016/j.bbrc.2014.05.114] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 05/27/2014] [Indexed: 10/25/2022]
Abstract
Bone mass is regulated by various molecules including endogenous factors as well as exogenous factors, such as nutrients and pollutants. Aryl hydrocarbon receptor (AhR) is known as a dioxin receptor and is responsible for various pathological and physiological processes. However, the role of AhR in bone homeostasis remains elusive because the cell type specific direct function of AhR has never been explored in vivo. Here, we show the cell type specific function of AhR in vivo in bone homeostasis. Systemic AhR knockout (AhRKO) mice exhibit increased bone mass with decreased resorption and decreased formation. Meanwhile, osteoclast specific AhRKO (AhR(ΔOc/ΔOc)) mice have increased bone mass with reduced bone resorption, although the mice lacking AhR in osteoblasts have a normal bone phenotype. Even under pathological conditions, AhR(ΔOc/ΔOc) mice are resistant to sex hormone deficiency-induced bone loss resulting from increased bone resorption. Furthermore, 3-methylcholanthrene, an AhR agonist, induces low bone mass with increased bone resorption in control mice, but not in AhR(ΔOc/ΔOc) mice. Taken together, cell type specific in vivo evidence for AhR functions indicates that osteoclastic AhR plays a significant role in maintenance of bone homeostasis, suggesting that inhibition of AhR in osteoclasts can be beneficial in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Tai-yong Yu
- Division of Integrative Pathophysiology, Proteo-Science Center, Graduate School of Medicine, Ehime University, Ehime 791-0295, Japan; Laboratory of Epigenetic Skeletal Diseases, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-0032, Japan
| | - Takeshi Kondo
- Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8503, Japan
| | - Takahiro Matsumoto
- Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8503, Japan
| | | | - Yuuki Imai
- Division of Integrative Pathophysiology, Proteo-Science Center, Graduate School of Medicine, Ehime University, Ehime 791-0295, Japan; Laboratory of Epigenetic Skeletal Diseases, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-0032, Japan.
| |
Collapse
|
1272
|
Heiland I. Was hat Tryptophan mit Krebs und Parkinson zu tun? CHEM UNSERER ZEIT 2014. [DOI: 10.1002/ciuz.201480013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
1273
|
Kochhar A, Kopelovich L, Sue E, Guttenplan JB, Herbert BS, Dannenberg AJ, Subbaramaiah K. p53 modulates Hsp90 ATPase activity and regulates aryl hydrocarbon receptor signaling. Cancer Prev Res (Phila) 2014; 7:596-606. [PMID: 24736433 PMCID: PMC4074578 DOI: 10.1158/1940-6207.capr-14-0051] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The aryl hydrocarbon receptor (AhR), a client protein of heat shock protein 90 (Hsp90), is a ligand-activated transcription factor that plays a role in polycyclic aromatic hydrocarbon (PAH)-induced carcinogenesis. Tobacco smoke activates AhR signaling leading to increased transcription of CYP1A1 and CYP1B1, which encode proteins that convert PAHs to mutagens. Recently, p53 was found to regulate Hsp90 ATPase activity via effects on activator of Hsp90 ATPase (Aha1). It is possible, therefore, that AhR-dependent expression of CYP1A1 and CYP1B1 might be affected by p53 status. The main objective of this study was to determine whether p53 modulated AhR-dependent gene expression and PAH metabolism. Here, we show that silencing p53 led to elevated Aha1 levels, increased Hsp90 ATPase activity, and enhanced CYP1A1 and CYP1B1 expression. Overexpression of wild-type p53 suppressed levels of CYP1A1 and CYP1B1. The significance of Aha1 in mediating these p53-dependent effects was determined. Silencing of Aha1 led to reduced Hsp90 ATPase activity and downregulation of CYP1A1 and CYP1B1. In contrast, overexpressing Aha1 was associated with increased Hsp90 ATPase activity and elevated levels of CYP1A1 and CYP1B1. Using p53 heterozygous mutant epithelial cells from patients with Li-Fraumeni syndrome, we show that monoallelic mutation of p53 was associated with elevated levels of CYP1A1 and CYP1B1 under both basal conditions and following treatment with benzo[a]pyrene. Treatment with CP-31398, a p53 rescue compound, suppressed benzo[a]pyrene-mediated induction of CYP1A1 and CYP1B1 and the formation of DNA adducts. Collectively, our results suggest that p53 affects AhR-dependent gene expression, PAH metabolism, and possibly carcinogenesis.
Collapse
Affiliation(s)
- Amit Kochhar
- Authors' Affiliations: Department of Medicine, Weill Cornell Medical College; Department of Basic Sciences, College of Dentistry; and Department of Environmental Medicine, School of Medicine, New York University, New York; Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins Medical Institutions, Baltimore, Maryland; and Department of Medical and Molecular Genetics, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IndianaAuthors' Affiliations: Department of Medicine, Weill Cornell Medical College; Department of Basic Sciences, College of Dentistry; and Department of Environmental Medicine, School of Medicine, New York University, New York; Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins Medical Institutions, Baltimore, Maryland; and Department of Medical and Molecular Genetics, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Levy Kopelovich
- Authors' Affiliations: Department of Medicine, Weill Cornell Medical College; Department of Basic Sciences, College of Dentistry; and Department of Environmental Medicine, School of Medicine, New York University, New York; Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins Medical Institutions, Baltimore, Maryland; and Department of Medical and Molecular Genetics, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Erika Sue
- Authors' Affiliations: Department of Medicine, Weill Cornell Medical College; Department of Basic Sciences, College of Dentistry; and Department of Environmental Medicine, School of Medicine, New York University, New York; Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins Medical Institutions, Baltimore, Maryland; and Department of Medical and Molecular Genetics, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Joseph B Guttenplan
- Authors' Affiliations: Department of Medicine, Weill Cornell Medical College; Department of Basic Sciences, College of Dentistry; and Department of Environmental Medicine, School of Medicine, New York University, New York; Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins Medical Institutions, Baltimore, Maryland; and Department of Medical and Molecular Genetics, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IndianaAuthors' Affiliations: Department of Medicine, Weill Cornell Medical College; Department of Basic Sciences, College of Dentistry; and Department of Environmental Medicine, School of Medicine, New York University, New York; Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins Medical Institutions, Baltimore, Maryland; and Department of Medical and Molecular Genetics, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Brittney-Shea Herbert
- Authors' Affiliations: Department of Medicine, Weill Cornell Medical College; Department of Basic Sciences, College of Dentistry; and Department of Environmental Medicine, School of Medicine, New York University, New York; Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins Medical Institutions, Baltimore, Maryland; and Department of Medical and Molecular Genetics, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Andrew J Dannenberg
- Authors' Affiliations: Department of Medicine, Weill Cornell Medical College; Department of Basic Sciences, College of Dentistry; and Department of Environmental Medicine, School of Medicine, New York University, New York; Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins Medical Institutions, Baltimore, Maryland; and Department of Medical and Molecular Genetics, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kotha Subbaramaiah
- Authors' Affiliations: Department of Medicine, Weill Cornell Medical College; Department of Basic Sciences, College of Dentistry; and Department of Environmental Medicine, School of Medicine, New York University, New York; Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins Medical Institutions, Baltimore, Maryland; and Department of Medical and Molecular Genetics, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
1274
|
Seegers N, van Doornmalen AM, Uitdehaag JCM, de Man J, Buijsman RC, Zaman GJR. High-Throughput Fluorescence-Based Screening Assays for Tryptophan-Catabolizing Enzymes. ACTA ACUST UNITED AC 2014; 19:1266-74. [DOI: 10.1177/1087057114536616] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Indoleamine 2,3-dioxygenase (IDO1) and tryptophan 2,3-dioxygenase (TDO) are two structurally different enzymes that have a different tissue distribution and physiological roles, but both catalyze the conversion of tryptophan to N-formylkynurenine (NFK). IDO1 has been clinically validated as a small-molecule drug target for cancer, while preclinical studies indicate that TDO may be a target for cancer immunotherapy and neurodegenerative disease. We have developed a high-throughput screening assay for IDO1 and TDO based on a novel chemical probe, NFK Green, that reacts specifically with NFK to form a green fluorescent molecule with an excitation wavelength of 400 nm and an emission wavelength of 510 nm. We provide the first side-by-side comparison of a number of published inhibitors of IDO1 and TDO and reveal that the preclinical IDO1 inhibitor Compound 5l shows significant cross-reactivity with TDO, while the relative selectivity of other published inhibitors was confirmed. The suitability for high-throughput screening of the assays was demonstrated by screening a library of 87,000 chemical substances in 384- or 1536-well format. Finally, we demonstrate that the assay can also be used to measure the capacity of cells to metabolize tryptophan and to measure the cellular potency of IDO1 and TDO inhibitors.
Collapse
Affiliation(s)
- Nicole Seegers
- Netherlands Translational Research Center B.V., Oss, The Netherlands
| | | | | | - Jos de Man
- Netherlands Translational Research Center B.V., Oss, The Netherlands
| | | | - Guido J. R. Zaman
- Netherlands Translational Research Center B.V., Oss, The Netherlands
| |
Collapse
|
1275
|
Moylan S, Berk M, Dean OM, Samuni Y, Williams LJ, O'Neil A, Hayley AC, Pasco JA, Anderson G, Jacka FN, Maes M. Oxidative & nitrosative stress in depression: why so much stress? Neurosci Biobehav Rev 2014; 45:46-62. [PMID: 24858007 DOI: 10.1016/j.neubiorev.2014.05.007] [Citation(s) in RCA: 293] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 04/17/2014] [Accepted: 05/13/2014] [Indexed: 12/29/2022]
Abstract
Many studies support a crucial role for oxidative & nitrosative stress (O&NS) in the pathophysiology of unipolar and bipolar depression. These disorders are characterized inter alia by lowered antioxidant defenses, including: lower levels of zinc, coenzyme Q10, vitamin E and glutathione; increased lipid peroxidation; damage to proteins, DNA and mitochondria; secondary autoimmune responses directed against redox modified nitrosylated proteins and oxidative specific epitopes. This review examines and details a model through which a complex series of environmental factors and biological pathways contribute to increased redox signaling and consequently increased O&NS in mood disorders. This multi-step process highlights the potential for future interventions that encompass a diverse range of environmental and molecular targets in the treatment of depression.
Collapse
Affiliation(s)
- Steven Moylan
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia; Barwon Health, Geelong, Victoria, Australia.
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia; Florey Institute for Neuroscience and Mental Health University of Melbourne, Parkville, Victoria, Australia; School of Public Health and Preventive Medicine, Monash University, Prahran, Victoria, Australia; University of Melbourne, Department of Psychiatry, Level 1 North, Main Block, Royal Melbourne Hospital, Parkville 3052, Australia; Barwon Health, Geelong, Victoria, Australia; Orygen Youth Health Research Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Olivia M Dean
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia; Florey Institute for Neuroscience and Mental Health University of Melbourne, Parkville, Victoria, Australia; University of Melbourne, Department of Psychiatry, Level 1 North, Main Block, Royal Melbourne Hospital, Parkville 3052, Australia
| | - Yuval Samuni
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Lana J Williams
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia; University of Melbourne, Department of Psychiatry, Level 1 North, Main Block, Royal Melbourne Hospital, Parkville 3052, Australia
| | - Adrienne O'Neil
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia; School of Public Health and Preventive Medicine, Monash University, Prahran, Victoria, Australia
| | - Amie C Hayley
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Julie A Pasco
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia; Northwest Academic Centre, University of Melbourne, St. Albans, Victoria, Australia
| | | | - Felice N Jacka
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia; Department of Psychiatry, Chulalongkorn University, Faculty of Medicine, Bangkok, Thailand; Department of Psychiatry, State University of Londrina, Londrina, Brazil
| |
Collapse
|
1276
|
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: 75] [Impact Index Per Article: 6.8] [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
|
1277
|
Piro RM, Wiesberg S, Schramm G, Rebel N, Oswald M, Eils R, Reinelt G, König R. Network topology-based detection of differential gene regulation and regulatory switches in cell metabolism and signaling. BMC SYSTEMS BIOLOGY 2014; 8:56. [PMID: 24886210 PMCID: PMC4031158 DOI: 10.1186/1752-0509-8-56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 04/29/2014] [Indexed: 11/10/2022]
Abstract
Background Common approaches to pathway analysis treat pathways merely as lists of genes disregarding their topological structures, that is, ignoring the genes' interactions on which a pathway's cellular function depends. In contrast, PathWave has been developed for the analysis of high-throughput gene expression data that explicitly takes the topology of networks into account to identify both global dysregulation of and localized (switch-like) regulatory shifts within metabolic and signaling pathways. For this purpose, it applies adjusted wavelet transforms on optimized 2D grid representations of curated pathway maps. Results Here, we present the new version of PathWave with several substantial improvements including a new method for optimally mapping pathway networks unto compact 2D lattice grids, a more flexible and user-friendly interface, and pre-arranged 2D grid representations. These pathway representations are assembled for several species now comprising H. sapiens, M. musculus, D. melanogaster, D. rerio, C. elegans, and E. coli. We show that PathWave is more sensitive than common approaches and apply it to RNA-seq expression data, identifying crucial metabolic pathways in lung adenocarcinoma, as well as microarray expression data, identifying pathways involved in longevity of Drosophila. Conclusions PathWave is a generic method for pathway analysis complementing established tools like GSEA, and the update comprises efficient new features. In contrast to the tested commonly applied approaches which do not take network topology into account, PathWave enables identifying pathways that are either known be involved in or very likely associated with such diverse conditions as human lung cancer or aging of D. melanogaster. The PathWave R package is freely available at http://www.ichip.de/software/pathwave.html.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rainer König
- Division of Theoretical Bioinformatics, German Cancer Research Center (Deutsches Krebsforschungszentrum, DKFZ), Heidelberg, Germany.
| |
Collapse
|
1278
|
Shin D, Kim IS, Lee JM, Shin SY, Lee JH, Baek SH, Cho KH. The hidden switches underlying RORα-mediated circuits that critically regulate uncontrolled cell proliferation. J Mol Cell Biol 2014; 6:338-48. [PMID: 24831657 DOI: 10.1093/jmcb/mju023] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Prostaglandin E2 (PGE2) is known to have a key role in the development of colorectal cancer, but previous experiments showed its contrasting (i.e. tumor-promoting or tumor-suppressive) roles depending on experimental conditions. To elucidate the mechanisms underlying such contrasting roles of PGE2 in tumorigenesis, we investigated all the previous experiments and found a new signal transduction pathway mediated by retinoic acid receptor-related orphan receptor (ROR)α, in which PGE2/PKCα-dependent phosphorylation of RORα attenuates Wnt target gene expression in colon cancer cells. From mathematical simulations combined with biochemical experimentation, we revealed that RORα induces a biphasic response of Wnt target genes to PGE2 stimulation through a regulatory switch formed by an incoherent feedforward loop, which provides a mechanistic explanation on the contrasting roles of PGE2 observed in previous experiments. More interestingly, we found that RORα constitutes another regulatory switch formed by coupled positive and negative feedback loops, which regulates the hysteretic response of Wnt signaling and eventually converts a proliferative cellular state into an anti-proliferative state in a very delicate way. Our results indicate that RORα is the key regulator at the center of these hidden switches that critically regulate cancer cell proliferation and thereby being a promising anti-cancer therapeutic target.
Collapse
Affiliation(s)
- Dongkwan Shin
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Ik Soo Kim
- Department of Biological Sciences, Creative Research Initiative Center for Chromatin Dynamics, Seoul National University, Seoul 151-742, Republic of Korea
| | - Ji Min Lee
- Department of Biological Sciences, Creative Research Initiative Center for Chromatin Dynamics, Seoul National University, Seoul 151-742, Republic of Korea
| | - Sung-Young Shin
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Jong-Hoon Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Sung Hee Baek
- Department of Biological Sciences, Creative Research Initiative Center for Chromatin Dynamics, Seoul National University, Seoul 151-742, Republic of Korea
| | - Kwang-Hyun Cho
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| |
Collapse
|
1279
|
Pinne M, Raucy JL. Advantages of cell-based high-volume screening assays to assess nuclear receptor activation during drug discovery. Expert Opin Drug Discov 2014; 9:669-86. [DOI: 10.1517/17460441.2014.913019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
1280
|
Moretti S, Menicali E, Voce P, Morelli S, Cantarelli S, Sponziello M, Colella R, Fallarino F, Orabona C, Alunno A, de Biase D, Bini V, Mameli MG, Filetti S, Gerli R, Macchiarulo A, Melillo RM, Tallini G, Santoro M, Puccetti P, Avenia N, Puxeddu E. Indoleamine 2,3-dioxygenase 1 (IDO1) is up-regulated in thyroid carcinoma and drives the development of an immunosuppressant tumor microenvironment. J Clin Endocrinol Metab 2014; 99:E832-40. [PMID: 24517146 DOI: 10.1210/jc.2013-3351] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CONTEXT Indoleamine 2,3-dioxygenase 1 (IDO1) is a single chain oxidoreductase that catalyzes tryptophan degradation to kynurenine. In cancer, it appears to exert an immunosuppressive function as part of an acquired mechanism of immune escape mediated by the inhibition of lymphocyte proliferation and survival and by the induction of FoxP3+ T regulatory cells. OBJECTIVE The objective of the study was to evaluate IDO1 expression in thyroid carcinoma and demonstrate its immunosuppressive function in the context of thyroid tumors. SETTING IDO1 expression was evaluated by quantitative PCR in 105 papillary thyroid carcinomas (PTCs), 11 medullary thyroid carcinomas, six anaplastic thyroid carcinomas, and five thyroid carcinoma cell lines (TCCLs), by immunohistochemistry in 55 PTCs and by Western blotting in five TCCLs. FoxP3+ Treg lymphocyte density was evaluated by immunohistochemistry in 29 PTCs. IDO1 inhibitory effect on lymphocyte proliferation was tested in coculture experiments of TCCLs and activated lymphocytes. RESULTS IDO1 mRNA expression resulted significantly higher in all the analyzed thyroid carcinoma histotypes compared with normal thyroid. Interestingly, an increase of IDO1 mRNA expression magnitude could be observed with gain of aggressiveness (PTCs and medullary thyroid carcinomas ≪ anaplastic thyroid carcinomas). In PTCs, IDO1 mRNA expression magnitude correlated with IDO1 immunostaining intensity in cancer cells and with FoxP3+ Treg lymphocyte density in the tumor microenvironment. IDO1 was expressed in human thyroid cancer cell lines in vitro, and FTC-133 cells showed high kynurenine concentration in the conditioned medium and a strong suppressive action on the proliferation of activated lymphocytes in coculture experiments. CONCLUSIONS For the first time, this study demonstrates a pivotal role of IDO1 in the suppression of lymphocyte function in thyroid carcinoma microenvironment.
Collapse
MESH Headings
- Adenocarcinoma, Follicular/genetics
- Adenocarcinoma, Follicular/immunology
- Adenocarcinoma, Follicular/metabolism
- Adenocarcinoma, Follicular/pathology
- Carcinoma, Medullary/genetics
- Carcinoma, Medullary/immunology
- Carcinoma, Medullary/metabolism
- Carcinoma, Medullary/pathology
- Carcinoma, Papillary/genetics
- Carcinoma, Papillary/immunology
- Carcinoma, Papillary/metabolism
- Carcinoma, Papillary/pathology
- Cell Line, Tumor
- Forkhead Transcription Factors/metabolism
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/immunology
- Thyroid Neoplasms/metabolism
- Thyroid Neoplasms/pathology
- Tumor Microenvironment/immunology
- Up-Regulation/physiology
Collapse
Affiliation(s)
- Sonia Moretti
- Departments of Internal Medicine (S.Moret., E.M., P.V., S.Morel., S.C., V.B., E.P.), Experimental Medicine and Biochemical Sciences (R.C., F.F., C.O., M.G.M., P.P.), Clinical and Experimental Medicine (A.A., R.G.), Chemistry and Drug Technology (A.M.), and Surgery (N.A.), University of Perugia, 06132 Perugia, Italy; Research Centre of Thyroid Proteomics and Genomics (S.Moret., E.M., P.V., S.Morel., N.A., E.P.), University of Perugia, 05100 Terni, Italy; Department of Clinical Sciences (M.S., S.F.), University of Rome "Sapienza," 00161 Rome, Italy; Department of Medicine (D.d.B., G.T.), Anatomic Pathology Unit, Bellaria Hospital, University of Bologna, 40139 Bologna, Italy; and Department of Molecular Medicine and Biotechnological Sciences (R.M.M., M.S.), University of Naples "Federico II," 80138 Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1281
|
Nguyen-Khac F, Della Valle V, Lopez RG, Ghysdael J, Bernard OA. Deregulation of AhR function by the human acute leukemia TEL-ARNT fusion protein. Am J Hematol 2014; 89:566-7. [PMID: 24469860 DOI: 10.1002/ajh.23680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 01/22/2014] [Accepted: 01/23/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Florence Nguyen-Khac
- Service d'Hématologie Biologique, Hôpital Pitié-Salpêtrière, Paris, France; INSERM U872, Centre de Recherche des Cordeliers, Paris, France; UPMC, Paris, France
| | | | | | | | | |
Collapse
|
1282
|
Jin UH, Lee SO, Sridharan G, Lee K, Davidson LA, Jayaraman A, Chapkin RS, Alaniz R, Safe S. Microbiome-derived tryptophan metabolites and their aryl hydrocarbon receptor-dependent agonist and antagonist activities. Mol Pharmacol 2014; 85:777-88. [PMID: 24563545 PMCID: PMC3990014 DOI: 10.1124/mol.113.091165] [Citation(s) in RCA: 229] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 02/21/2014] [Indexed: 12/17/2022] Open
Abstract
The tryptophan metabolites indole, indole-3-acetate, and tryptamine were identified in mouse cecal extracts and fecal pellets by mass spectrometry. The aryl hydrocarbon receptor (AHR) agonist and antagonist activities of these microbiota-derived compounds were investigated in CaCo-2 intestinal cells as a model for understanding their interactions with colonic tissue, which is highly aryl hydrocarbon (Ah)-responsive. Activation of Ah-responsive genes demonstrated that tryptamine and indole 3-acetate were AHR agonists, whereas indole was an AHR antagonist that inhibited TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin)-induced CYP1A1 expression. In contrast, the tryptophan metabolites exhibited minimal anti-inflammatory activities, whereas TCDD decreased phorbol ester-induced CXCR4 [chemokine (C-X-C motif) receptor 4] gene expression, and this response was AHR dependent. These results demonstrate that the tryptophan metabolites indole, tryptamine, and indole-3-acetate modulate AHR-mediated responses in CaCo-2 cells, and concentrations of indole that exhibit AHR antagonist activity (100-250 μM) are detected in the intestinal microbiome.
Collapse
Affiliation(s)
- Un-Ho Jin
- Institute of Biosciences and Technology, Texas A&M Health Sciences Center, Houston, Texas (U.-H.J., S.-O.L., S.S.); Department of Microbial and Molecular Pathogenesis, Texas A&M University Health Sciences Center (A.J., R.A.), Department of Veterinary Physiology and Pharmacology (S.S.), Department of Chemical Engineering (A.J.), and Department of Nutrition and Food Science (L.A.D., R.S.C.), Texas A&M University, College Station, Texas; Department of Food Science and Technology, Keimyung University, Daegu, Republic of Korea (S.-O.L.); and Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts (G.S., K.L.)
| | | | | | | | | | | | | | | | | |
Collapse
|
1283
|
Prendergast GC, Smith C, Thomas S, Mandik-Nayak L, Laury-Kleintop L, Metz R, Muller AJ. Indoleamine 2,3-dioxygenase pathways of pathogenic inflammation and immune escape in cancer. Cancer Immunol Immunother 2014; 63:721-35. [PMID: 24711084 DOI: 10.1007/s00262-014-1549-4] [Citation(s) in RCA: 390] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 03/26/2014] [Indexed: 12/15/2022]
Abstract
Genetic and pharmacological studies of indoleamine 2,3-dioxygenase (IDO) have established this tryptophan catabolic enzyme as a central driver of malignant development and progression. IDO acts in tumor, stromal and immune cells to support pathogenic inflammatory processes that engender immune tolerance to tumor antigens. The multifaceted effects of IDO activation in cancer include the suppression of T and NK cells, the generation and activation of T regulatory cells and myeloid-derived suppressor cells, and the promotion of tumor angiogenesis. Mechanistic investigations have defined the aryl hydrocarbon receptor, the master metabolic regulator mTORC1 and the stress kinase Gcn2 as key effector signaling elements for IDO, which also exerts a non-catalytic role in TGF-β signaling. Small-molecule inhibitors of IDO exhibit anticancer activity and cooperate with immunotherapy, radiotherapy or chemotherapy to trigger rapid regression of aggressive tumors otherwise resistant to treatment. Notably, the dramatic antitumor activity of certain targeted therapeutics such as imatinib (Gleevec) in gastrointestinal stromal tumors has been traced in part to IDO downregulation. Further, antitumor responses to immune checkpoint inhibitors can be heightened safely by a clinical lead inhibitor of the IDO pathway that relieves IDO-mediated suppression of mTORC1 in T cells. In this personal perspective on IDO as a nodal mediator of pathogenic inflammation and immune escape in cancer, we provide a conceptual foundation for the clinical development of IDO inhibitors as a novel class of immunomodulators with broad application in the treatment of advanced human cancer.
Collapse
Affiliation(s)
- George C Prendergast
- Lankenau Institute for Medical Research (LIMR), 100 Lancaster Avenue, Wynnewood, PA, 19096, USA,
| | | | | | | | | | | | | |
Collapse
|
1284
|
Kawasaki H, Chang HW, Tseng HC, Hsu SC, Yang SJ, Hung CH, Zhou Y, Huang SK. A tryptophan metabolite, kynurenine, promotes mast cell activation through aryl hydrocarbon receptor. Allergy 2014; 69:445-52. [PMID: 24397465 DOI: 10.1111/all.12346] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2013] [Indexed: 02/06/2023]
Abstract
BACKGROUND Tryptophan metabolites have been suggested to play a role in immune modulation, wherein those have recently been shown to be endogenous ligands of aryl hydrocarbon receptor (AhR; a unique cellular chemical sensor). However, the involvement of tryptophan metabolites and AhR in modulating mast cell function remains to be fully defined. We therefore investigated that the functional impacts of tryptophan metabolites on human and mouse mast cell responses in vitro and their functional importance in vivo. METHODS Three tryptophan metabolites, kynurenine (KYN), kynurenic acid (KA) and quinolinic acid (QA), were examined in terms of their effect on IgE-mediated responses in mouse bone marrow-derived mast cells (BMMCs) and in human peripheral blood-derived cultured mast cells (HCMCs) and on in vivo anaphylactic responses. For evaluation of AhR involvement, we examined the responses of mast cells from AhR-null or AhR-wild-type mice with the use of a known AhR antagonist, CH223191. RESULTS Kynurenine, but not KA and QA, enhanced IgE-mediated responses, including degranulation, LTC4 release, and IL-13 production in BMMCs through the activation of PLCγ1, Akt, MAPK p38, and the increase of intracellular calcium. KYN also enhanced cutaneous anaphylaxis in vivo. These enhancing effects of KYN were not observed in AhR-deficient BMMCs and could be inhibited by CH223191 in BMMCs. Further, KYN had similar enhancing effects on HCMCs, which were inhibited by CH223191. CONCLUSION The AhR-KYN axis is potentially important in modulating mast cell responses and represents an example of AhR's critical involvement in the regulation of allergic responses.
Collapse
Affiliation(s)
- H. Kawasaki
- National Health Research Institutes; Zhu-Nan Taiwan
| | - H.-W. Chang
- National Health Research Institutes; Zhu-Nan Taiwan
| | - H.-C. Tseng
- National Health Research Institutes; Zhu-Nan Taiwan
| | - S.-C. Hsu
- National Health Research Institutes; Zhu-Nan Taiwan
| | - S.-J. Yang
- National Health Research Institutes; Zhu-Nan Taiwan
| | - C.-H. Hung
- Department of Pediatrics; Kaohsiung Medical University Hospital; Kaohsiung Taiwan
| | - Y. Zhou
- Johns Hopkins Asthma and Allergy Center; Johns Hopkins University School of Medicine; Baltimore MD USA
| | - S.-K. Huang
- National Health Research Institutes; Zhu-Nan Taiwan
- Johns Hopkins Asthma and Allergy Center; Johns Hopkins University School of Medicine; Baltimore MD USA
| |
Collapse
|
1285
|
Yong-Gang X, Ming-Zhe W, Jin-Yan Z, Zhi-Hai P, Jun-Ming X. Combination of N-(3׳,4׳-dimethoxycinnamoyl) anthranilic acid with cyclosporin A treatment preserves immunosuppressive effect and reduces the side effect of cyclosporin A in rat. Eur J Pharmacol 2014; 728:16-23. [DOI: 10.1016/j.ejphar.2014.01.055] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 01/22/2014] [Accepted: 01/22/2014] [Indexed: 01/31/2023]
|
1286
|
Becker JC, thor Straten P, Andersen MH. Self-reactive T cells: suppressing the suppressors. Cancer Immunol Immunother 2014; 63:313-9. [PMID: 24368340 PMCID: PMC11029163 DOI: 10.1007/s00262-013-1512-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 12/11/2013] [Indexed: 01/22/2023]
Abstract
The immune system is a tightly regulated and complex system. An important part of this immune regulation is the assurance of tolerance toward self-antigens to maintain immune homeostasis. However, in recent years, antigen-specific cellular immune responses toward several normal self-proteins expressed in regulatory immune cells have been reported, especially in patients with cancer. The seemingly lack of tolerance toward such proteins is interesting, as it suggests a regulatory function of self-reactive T (srT) cells, which may be important for the fine tuning of the immune system. In particular, surprising has been the description of cytotoxic srT cells that are able to eliminate normal regulatory immune cells. Such srT cells may be important as effector cells that suppress regulatory suppressor cells. The current knowledge of the nature and function of srT cells is still limited. Still, the therapeutic targeting of srT cells offers a novel approach to harness immune-regulatory networks in cancer.
Collapse
Affiliation(s)
- Jürgen C. Becker
- Department of General Dermatology, Medical University of Graz, Graz, Austria
| | - Per thor Straten
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Mads Hald Andersen
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
| |
Collapse
|
1287
|
Wang W, Li M, Wei Z, Wang Z, Bu X, Lai W, Yang S, Gong H, Zheng H, Wang Y, Liu Y, Li Q, Fang Q, Hu Z. Bimodal Imprint Chips for Peptide Screening: Integration of High-Throughput Sequencing by MS and Affinity Analyses by Surface Plasmon Resonance Imaging. Anal Chem 2014; 86:3703-7. [DOI: 10.1021/ac500465e] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Weizhi Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Menglin Li
- Department
of Biomedical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Zewen Wei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Zihua Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Xiangli Bu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Wenjia Lai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Shu Yang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - He Gong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Hui Zheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Yuqiao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Ying Liu
- Beijing
Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, China
| | - Qin Li
- Department
of Biomedical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Qiaojun Fang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Zhiyuan Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
- Beijing
Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, China
- Institute for Systems Biology, 401 Terry Avenue N, Seattle, Washington 98109, United States
| |
Collapse
|
1288
|
Poormasjedi-Meibod MS, Hartwell R, Taghi Kilani R, Ghahary A. Anti-scarring properties of different tryptophan derivatives. PLoS One 2014; 9:e91955. [PMID: 24637853 PMCID: PMC3956813 DOI: 10.1371/journal.pone.0091955] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 02/18/2014] [Indexed: 12/03/2022] Open
Abstract
Hypertrophic scars are associated with prolonged extracellular matrix (ECM) production, aberrant ECM degradation and high tissue cellularity. Routinely used antifibrotic strategies aim to reduce ECM deposition and enhance matrix remodeling. Our previous study investigating the antifibrotic effects of indoleamine2, 3 dioxygenase (IDO) led to the identification of kynurenine (Kyn) as an antiscarring agent. A topical antifibrogenic therapy using Kyn is very attractive; however, it is well established that Kyn passes the blood brain barrier (BBB) which causes complications including excitatory neuronal death. Here we investigated the antiscarring properties of kynurenic acid (KynA), a downstream end product of Kyn that is unlikely to pass the BBB, as an effective and safe replacement for Kyn. Our results indicated that while not having any adverse effect on dermal cell viability, KynA significantly increases the expression of matrix metalloproteinases (MMP1 and MMP3) and suppresses the production of type-I collagen and fibronectin by fibroblasts. Topical application of cream containing KynA in fibrotic rabbit ear significantly decreased scar elevation index (1.13±0.13 vs. 1.61±0.12) and tissue cellularity (221.38±21.7 vs. 314.56±8.66 cells/hpf) in KynA treated wounds compared to controls. KynA treated wounds exhibited lower levels of collagen deposition which is accompanied with a significant decrease in type-I collagen and fibronectin expression, as well as an increase in MMP1 expression compared to untreated wounds or wounds treated with cream only. The results of this study provided evidence for the first time that KynA is promising candidate antifibrogenic agent to improve healing outcome in patients at risk of hypertrophic scarring.
Collapse
Affiliation(s)
| | - Ryan Hartwell
- Division of plastic surgery, Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ruhangiz Taghi Kilani
- Division of plastic surgery, Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Aziz Ghahary
- Division of plastic surgery, Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
1289
|
Sallée M, Dou L, Cerini C, Poitevin S, Brunet P, Burtey S. The aryl hydrocarbon receptor-activating effect of uremic toxins from tryptophan metabolism: a new concept to understand cardiovascular complications of chronic kidney disease. Toxins (Basel) 2014; 6:934-49. [PMID: 24599232 PMCID: PMC3968369 DOI: 10.3390/toxins6030934] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 02/19/2014] [Accepted: 02/24/2014] [Indexed: 12/13/2022] Open
Abstract
Patients with chronic kidney disease (CKD) have a higher risk of cardiovascular diseases and suffer from accelerated atherosclerosis. CKD patients are permanently exposed to uremic toxins, making them good candidates as pathogenic agents. We focus here on uremic toxins from tryptophan metabolism because of their potential involvement in cardiovascular toxicity: indolic uremic toxins (indoxyl sulfate, indole-3 acetic acid, and indoxyl-β-d-glucuronide) and uremic toxins from the kynurenine pathway (kynurenine, kynurenic acid, anthranilic acid, 3-hydroxykynurenine, 3-hydroxyanthranilic acid, and quinolinic acid). Uremic toxins derived from tryptophan are endogenous ligands of the transcription factor aryl hydrocarbon receptor (AhR). AhR, also known as the dioxin receptor, interacts with various regulatory and signaling proteins, including protein kinases and phosphatases, and Nuclear Factor-Kappa-B. AhR activation by 2,3,7,8-tetrachlorodibenzo-p-dioxin and some polychlorinated biphenyls is associated with an increase in cardiovascular disease in humans and in mice. In addition, this AhR activation mediates cardiotoxicity, vascular inflammation, and a procoagulant and prooxidant phenotype of vascular cells. Uremic toxins derived from tryptophan have prooxidant, proinflammatory, procoagulant, and pro-apoptotic effects on cells involved in the cardiovascular system, and some of them are related with cardiovascular complications in CKD. We discuss here how the cardiovascular effects of these uremic toxins could be mediated by AhR activation, in a “dioxin-like” effect.
Collapse
Affiliation(s)
- Marion Sallée
- Aix Marseille Université, Inserm, VRCM, UMR_S 1076, Marseille13005, France.
| | - Laetitia Dou
- Aix Marseille Université, Inserm, VRCM, UMR_S 1076, Marseille13005, France.
| | - Claire Cerini
- Aix Marseille Université, Inserm, VRCM, UMR_S 1076, Marseille13005, France.
| | - Stéphane Poitevin
- Aix Marseille Université, Inserm, VRCM, UMR_S 1076, Marseille13005, France.
| | - Philippe Brunet
- Aix Marseille Université, Inserm, VRCM, UMR_S 1076, Marseille13005, France.
| | - Stéphane Burtey
- Aix Marseille Université, Inserm, VRCM, UMR_S 1076, Marseille13005, France.
| |
Collapse
|
1290
|
Ligand promiscuity of aryl hydrocarbon receptor agonists and antagonists revealed by site-directed mutagenesis. Mol Cell Biol 2014; 34:1707-19. [PMID: 24591650 DOI: 10.1128/mcb.01183-13] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-dependent transcription factor that can be activated by structurally diverse chemicals. To examine the mechanisms responsible for the promiscuity in AhR ligand binding, we determined the effects of mutations within the AhR ligand-binding domain (LBD) on the activity of diverse AhR ligands. Site-directed mutagenesis identified Ile319 of the mouse AhR and, to a lesser extent, Phe318 as residues involved in ligand-selective modulation of AhR transformation using a panel of 12 AhR ligands. These ligands could be categorized into four distinct structurally related groups based on their ability to activate AhR mutants at position 319 in vitro. The mutation I319K was selectively activated by FICZ and not by other examined ligands in vitro and in cell culture. F318L and F318A mutations resulted in the conversion of AhR agonists β-naphthoflavone and 3-methylcholanthrene, respectively, into partial agonists/antagonists. Hsp90 binding to the AhR was decreased with several mutations and was inversely correlated with AhR ligand-binding promiscuity. Together, these data define overlapping amino acid residues within the AhR LBD involved in the selectivity of ligand binding, the agonist or antagonist mode of ligand binding, and hsp90 binding and provide insights into the ligand diversity of AhR activators.
Collapse
|
1291
|
|
1292
|
Chuang SC, Fanidi A, Ueland PM, Relton C, Midttun O, Vollset SE, Gunter MJ, Seckl MJ, Travis RC, Wareham N, Trichopoulou A, Lagiou P, Trichopoulos D, Peeters PHM, Bueno-de-Mesquita HB, Boeing H, Wientzek A, Kuehn T, Kaaks R, Tumino R, Agnoli C, Palli D, Naccarati A, Aicua EA, Sánchez MJ, Quirós JR, Chirlaque MD, Agudo A, Johansson M, Grankvist K, Boutron-Ruault MC, Clavel-Chapelon F, Fagherazzi G, Weiderpass E, Riboli E, Brennan PJ, Vineis P, Johansson M. Circulating biomarkers of tryptophan and the kynurenine pathway and lung cancer risk. Cancer Epidemiol Biomarkers Prev 2014; 23:461-8. [PMID: 24357106 DOI: 10.1158/1055-9965.epi-13-0770] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Imbalances in tryptophan metabolism have been linked to cancer-related immune escape and implicated in several cancers, including lung cancer. METHODS We conducted a nested case-control study within the European Prospective Investigation into Cancer and Nutrition (EPIC) that included 893 incident lung cancer cases and 1,748 matched controls. Circulating levels of tryptophan and six of its metabolites were measured and evaluated in relation to lung cancer risk. RESULTS Tryptophan (Ptrend = 2 × 10(-5)) and the kynurenine/tryptophan ratio (KTR; Ptrend = 4 × 10(-5)) were associated with lung cancer risk overall after adjusting for established risk factors. The ORs comparing the fifth and first quintiles (OR5th vs. 1st) were 0.52 [95% confidence interval (CI), 0.37-0.74] for tryptophan and 1.74 (95% CI, 1.24-2.45) for KTR. After adjusting for plasma methionine (available from previous work, which was strongly correlated with tryptophan), the associations of tryptophan (adjusted Ptrend = 0.13) and KTR (Ptrend = 0.009) were substantially attenuated. KTR was positively associated with squamous cell carcinoma, the OR5th vs. 1st being 2.83 (95% CI, 1.62-4.94, Ptrend = 3 × 10(-5)) that was only marginally affected by adjusting for methionine. CONCLUSIONS This study indicates that biomarkers of tryptophan metabolism are associated with subsequent lung cancer risk. Although this result would seem consistent with the immune system having a role in lung cancer development, the overall associations were dependent on methionine, and further studies are warranted to further elucidate the importance of these metabolites in lung cancer etiology. IMPACT This is the first prospective study investigating the tryptophan pathway in relation to lung cancer risk.
Collapse
Affiliation(s)
- Shu-Chun Chuang
- Authors' Affiliations: School of Public Health; Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London; Institute for Ageing and Health, Newcastle University, Newcastle; Cancer Epidemiology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom; International Agency for Research on Cancer, Lyon; Inserm U1018, Centre for Research in Epidemiology and Population Health (CESP); University Paris Sud, UMRS 1018; IGR, Villejuif, France; Section of Pharmacology, Institute of Medicine; Department of Public Health and Primary Health Care, University of Bergen; Laboratory of Clinical Biochemistry, Haukeland University Hospital; Bevital AS; Division of Epidemiology, Norwegian Institute of Public Health, Bergen; Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway; WHO Collaborating Center for Food and Nutrition Policies, Department of Hygiene, Epidemiology and Medical Statistics, University of Athens Medical School; Bureau of Epidemiologic Research, Academy of Athens; Hellenic Health Foundation, Athens, Greece; Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts; Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center; Department of Gastroenterology and Hepatology, University Medical Centre, Utrecht; National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal; Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Cancer Registry and Histopathology Unit, "Civile M.P.Arezzo" Hospital, ASP Ragusa; Nutritional Epidemiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan; Molecular and Nutritional Epidemiology Unit, Cancer Research an
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1293
|
Duan Z, Duan Y, Lei H, Hu N, Shi J, Shen D, Wang X, Hu Y. Attenuation of antigenic immunogenicity by kynurenine, a novel suppressive adjuvant. Hum Vaccin Immunother 2014; 10:1295-305. [PMID: 24583631 DOI: 10.4161/hv.28099] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A novel therapeutic strategy is required for autoimmune diseases characterized by the production of autoantibody, because current clinical strategies have limitations. Vaccination against autoimmune diseases is a feasible strategy because vaccines induce immune response memory and the antigen specificity. However, no suitable adjuvant is available to direct the immune response toward tolerance or suppression. In the current study, we evaluated whether kynurenine (Kyn) could serve as a novel suppressive adjuvant to decrease the humoral immune responses against hepatitis A virus (HAV) in the ICR mouse model in vivo and lipopolysaccharide (LPS) in B cells in vitro. The underlying mechanisms of Kyn-mediated suppression of LPS-induced IgM responses were explored. The results showed that Kyn significantly decreased HAV immunogenicity when co-administered with HAV, and that Kyn (100 μM/1000 μM) impaired IgM generation compared with that induced by LPS alone. We also demonstrated that microRNA30b (miR30b) played a critical role in the process of Kyn-mediated suppression of IgM responses induced by LPS, and that Bach2, a transcriptional repressor of B cell terminal differentiation, was a novel target of miR30b. These findings suggest that Kyn can serve as a novel and effective suppressive adjuvant for vaccines.
Collapse
Affiliation(s)
- Zhiqing Duan
- Institute of Medical Biology; Chinese Academy of Medical Sciences and Peking Union Medical College; Kunming, PR China
| | - Yunqing Duan
- Shanxi Agricultural University; Taigu, Shanxi, PR China
| | - Huangui Lei
- Shanxi Agricultural University; Taigu, Shanxi, PR China
| | - Ningzhu Hu
- Institute of Medical Biology; Chinese Academy of Medical Sciences and Peking Union Medical College; Kunming, PR China
| | - Jiandong Shi
- Institute of Medical Biology; Chinese Academy of Medical Sciences and Peking Union Medical College; Kunming, PR China
| | - Dong Shen
- Institute of Medical Biology; Chinese Academy of Medical Sciences and Peking Union Medical College; Kunming, PR China
| | - Xi Wang
- Institute of Medical Biology; Chinese Academy of Medical Sciences and Peking Union Medical College; Kunming, PR China
| | - Yunzhang Hu
- Institute of Medical Biology; Chinese Academy of Medical Sciences and Peking Union Medical College; Kunming, PR China
| |
Collapse
|
1294
|
Pabst C, Krosl J, Fares I, Boucher G, Ruel R, Marinier A, Lemieux S, Hébert J, Sauvageau G. Identification of small molecules that support human leukemia stem cell activity ex vivo. Nat Methods 2014; 11:436-42. [PMID: 24562423 DOI: 10.1038/nmeth.2847] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 01/12/2014] [Indexed: 11/09/2022]
Abstract
Leukemic stem cells (LSCs) are considered a major cause of relapse in acute myeloid leukemia (AML). Defining pathways that control LSC self-renewal is crucial for a better understanding of underlying mechanisms and for the development of targeted therapies. However, currently available culture conditions do not prevent spontaneous differentiation of LSCs, which greatly limits the feasibility of cell-based assays. To overcome these constraints we conducted a high-throughput chemical screen and identified small molecules that inhibit differentiation and support LSC activity in vitro. Similar to reports with cord blood stem cells, several of these compounds suppressed the aryl-hydrocarbon receptor (AhR) pathway, which we show to be inactive in vivo and rapidly activated ex vivo in AML cells. We also identified a compound, UM729, that collaborates with AhR suppressors in preventing AML cell differentiation. Together, these findings provide newly defined culture conditions for improved ex vivo culture of primary human AML cells.
Collapse
Affiliation(s)
- Caroline Pabst
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, Canada
| | - Jana Krosl
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, Canada
| | - Iman Fares
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, Canada
| | - Geneviève Boucher
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, Canada
| | - Réjean Ruel
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, Canada
| | - Anne Marinier
- 1] Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, Canada. [2] Chemistry Department, University of Montreal, Montreal, Quebec, Canada
| | - Sébastien Lemieux
- 1] Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, Canada. [2] Department of Computer Science and Operations Research, University of Montreal, Montreal, Quebec, Canada
| | - Josée Hébert
- 1] Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, Canada. [2] Leukemia Cell Bank of Quebec, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada. [3] Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada. [4] Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Guy Sauvageau
- 1] Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, Canada. [2] Leukemia Cell Bank of Quebec, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada. [3] Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada. [4] Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
1295
|
Campbell BM, Charych E, Lee AW, Möller T. Kynurenines in CNS disease: regulation by inflammatory cytokines. Front Neurosci 2014; 8:12. [PMID: 24567701 PMCID: PMC3915289 DOI: 10.3389/fnins.2014.00012] [Citation(s) in RCA: 274] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 01/20/2014] [Indexed: 12/27/2022] Open
Abstract
The kynurenine pathway (KP) metabolizes the essential amino acid tryptophan and generates a number of neuroactive metabolites collectively called the kynurenines. Segregated into at least two distinct branches, often termed the “neurotoxic” and “neuroprotective” arms of the KP, they are regulated by the two enzymes kynurenine 3-monooxygenase and kynurenine aminotransferase, respectively. Interestingly, several enzymes in the pathway are under tight control of inflammatory mediators. Recent years have seen a tremendous increase in our understanding of neuroinflammation in CNS disease. This review will focus on the regulation of the KP by inflammatory mediators as it pertains to neurodegenerative and psychiatric disorders.
Collapse
Affiliation(s)
- Brian M Campbell
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA Paramus, NJ, USA
| | - Erik Charych
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA Paramus, NJ, USA
| | - Anna W Lee
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA Paramus, NJ, USA
| | - Thomas Möller
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA Paramus, NJ, USA
| |
Collapse
|
1296
|
Lowe MM, Mold JE, Kanwar B, Huang Y, Louie A, Pollastri MP, Wang C, Patel G, Franks DG, Schlezinger J, Sherr DH, Silverstone AE, Hahn ME, McCune JM. Identification of cinnabarinic acid as a novel endogenous aryl hydrocarbon receptor ligand that drives IL-22 production. PLoS One 2014; 9:e87877. [PMID: 24498387 PMCID: PMC3912126 DOI: 10.1371/journal.pone.0087877] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 12/30/2013] [Indexed: 01/17/2023] Open
Abstract
The aryl hydrocarbon receptor (AHR) binds to environmental toxicants including synthetic halogenated aromatic hydrocarbons and is involved in a diverse array of biological processes. Recently, the AHR was shown to control host immunity by affecting the balance between inflammatory T cells that produce IL-17 (Th17) and IL-22 versus regulatory T cells (Treg) involved in tolerance. While environmental AHR ligands can mediate this effect, endogenous ligands are likely to be more relevant in host immune responses. We investigated downstream metabolites of tryptophan as potential AHR ligands because (1) tryptophan metabolites have been implicated in regulating the balance between Th17 and Treg cells and (2) many of the AHR ligands identified thus far are derivatives of tryptophan. We characterized the ability of tryptophan metabolites to bind and activate the AHR and to increase IL-22 production in human T cells. We report that the tryptophan metabolite, cinnabarinic acid (CA), is an AHR ligand that stimulates the differentiation of human and mouse T cells producing IL-22. We compare the IL-22-stimulating activity of CA to that of other tryptophan metabolites and define stimulation conditions that lead to CA production from immune cells. Our findings link tryptophan metabolism to AHR activation and define a novel endogenous AHR agonist with potentially broad biological functions.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cells, Cultured
- Chromatography, Liquid
- Cytochrome P-450 CYP1A1/metabolism
- Humans
- Interleukins/metabolism
- Ligands
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Oxazines/metabolism
- Receptors, Aryl Hydrocarbon/physiology
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th17 Cells
- Tryptophan/metabolism
- Interleukin-22
Collapse
Affiliation(s)
- Margaret M. Lowe
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Jeff E. Mold
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Bittoo Kanwar
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- Division of Gastroenterology, Department of Pediatrics, University of California San Francisco, San Francisco, California, United States of America
| | - Yong Huang
- Drug Studies Unit, Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Alexander Louie
- Drug Studies Unit, Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Michael P. Pollastri
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, United States of America
| | - Cuihua Wang
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, United States of America
| | - Gautam Patel
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, United States of America
| | - Diana G. Franks
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts, United States of America
| | - Jennifer Schlezinger
- Department of Environmental Health, School of Public Health, Boston University, Boston, Massachusetts, United States of America
| | - David H. Sherr
- Department of Environmental Health, School of Public Health, Boston University, Boston, Massachusetts, United States of America
| | - Allen E. Silverstone
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| | - Mark E. Hahn
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts, United States of America
| | - Joseph M. McCune
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
1297
|
Affiliation(s)
- Charity Duran
- From the Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA
| | | |
Collapse
|
1298
|
Hukkanen J. Induction of cytochrome P450 enzymes: a view on humanin vivofindings. Expert Rev Clin Pharmacol 2014; 5:569-85. [PMID: 23121279 DOI: 10.1586/ecp.12.39] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Janne Hukkanen
- Department of Internal Medicine, Institute of Clinical Medicine, and Biocenter Oulu, University of Oulu, Oulu, Finland.
| |
Collapse
|
1299
|
Metz R, Smith C, DuHadaway JB, Chandler P, Baban B, Merlo LMF, Pigott E, Keough MP, Rust S, Mellor AL, Mandik-Nayak L, Muller AJ, Prendergast GC. IDO2 is critical for IDO1-mediated T-cell regulation and exerts a non-redundant function in inflammation. Int Immunol 2014; 26:357-67. [PMID: 24402311 DOI: 10.1093/intimm/dxt073] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IDO2 is implicated in tryptophan catabolism and immunity but its physiological functions are not well established. Here we report the characterization of mice genetically deficient in IDO2, which develop normally but exhibit defects in IDO-mediated T-cell regulation and inflammatory responses. Construction of this strain was prompted in part by our discovery that IDO2 function is attenuated in macrophages from Ido1 (-/-) mice due to altered message splicing, generating a functional mosaic with implications for interpreting findings in Ido1 (-/-) mice. No apparent defects were observed in Ido2 (-/-) mice in embryonic development or hematopoietic differentiation, with wild-type profiles documented for kynurenine in blood serum and for immune cells in spleen, lymph nodes, peritoneum, thymus and bone marrow of naive mice. In contrast, upon immune stimulation we determined that IDO1-dependent T regulatory cell generation was defective in Ido2 (-/-) mice, supporting Ido1-Ido2 genetic interaction and establishing a functional role for Ido2 in immune modulation. Pathophysiologically, both Ido1 (-/-) and Ido2 (-/-) mice displayed reduced skin contact hypersensitivity responses, but mechanistic distinctions were apparent, with only Ido2 deficiency associated with a suppression of immune regulatory cytokines that included GM-CSF, G-CSF, IFN-γ, TNF-α, IL-6 and MCP-1/CCL2. Different contributions to inflammation were likewise indicated by the finding that Ido2 (-/-) mice did not phenocopy Ido1 (-/-) mice in the reduced susceptibility of the latter to inflammatory skin cancer. Taken together, our results offer an initial glimpse into immune modulation by IDO2, revealing its genetic interaction with IDO1 and distinguishing its non-redundant contributions to inflammation.
Collapse
Affiliation(s)
- Richard Metz
- New Link Genetics Corporation, Ames, IA 50010, USA
| | - Courtney Smith
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | | | - Phillip Chandler
- Immunotherapy Center, Georgia Regents University, Augusta, GA 30912, USA
| | - Babak Baban
- Immunotherapy Center, Georgia Regents University, Augusta, GA 30912, USA
| | - Lauren M F Merlo
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | - Elizabeth Pigott
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | - Martin P Keough
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | - Sonja Rust
- New Link Genetics Corporation, Ames, IA 50010, USA
| | - Andrew L Mellor
- Immunotherapy Center, Georgia Regents University, Augusta, GA 30912, USA
| | - Laura Mandik-Nayak
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Alexander J Muller
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - George C Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA Department of Pathology, Anatomy and Cell Biology, Jefferson Medical School, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
1300
|
Li DD, Gao YJ, Tian XC, Yang ZQ, Cao H, Zhang QL, Guo B, Yue ZP. Differential expression and regulation of Tdo2 during mouse decidualization. J Endocrinol 2014; 220:73-83. [PMID: 24190896 DOI: 10.1530/joe-13-0429] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tryptophan 2,3-dioxygenase (Tdo2) is a rate-limiting enzyme which directs the conversion of tryptophan to kynurenine. The aim of this study was to examine the expression and regulation of Tdo2 in mouse uterus during decidualization. Tdo2 mRNA was mainly expressed in the decidua on days 6-8 of pregnancy. By real-time PCR, a high level of Tdo2 expression was observed in the uteri from days 6 to 8 of pregnancy, although Tdo2 expression was observed on days 1-8. Simultaneously, Tdo2 mRNA was also detected under in vivo and in vitro artificial decidualization. Estrogen, progesterone, and 8-bromoadenosine-cAMP could induce the expression of Tdo2 in the ovariectomized mouse uterus and uterine stromal cells. Tdo2 could regulate cell proliferation and stimulate the expression of decidual marker Dtprp in the uterine stromal cells and decidual cells. Overexpression of Tdo2 could upregulate the expression of Ahr, Cox2, and Vegf genes in uterine stromal cells, while Tdo2 inhibitor 680C91 could downregulate the expression of Cox2 and Vegf genes in uterine decidual cells. These data indicate that Tdo2 may play an important role during mouse decidualization and be regulated by estrogen, progesterone, and cAMP.
Collapse
Affiliation(s)
- Dang-Dang Li
- College of Veterinary Medicine, Jilin University, Changchun 130062, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|