1
|
Almulla AF, Thipakorn Y, Zhou B, Vojdani A, Paunova R, Maes M. The tryptophan catabolite or kynurenine pathway in long COVID disease: A systematic review and meta-analysis. Neuroscience 2024; 563:268-277. [PMID: 39424264 DOI: 10.1016/j.neuroscience.2024.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/29/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Recent studies confirm the involvement of activated immune-inflammatory responses and increased oxidative and nitrosative stress in Long COVID (LC) disease. However, the influence of these pathways on the metabolism of tryptophan (TRP) through the TRP catabolite (TRYCAT) pathway and their mediating effects on LC pathophysiology, has not been fully explored. OBJECTIVE This meta-analysis investigates peripheral TRP and TRYCAT levels and the TRYCAT pathway in patients with LC disease. METHOD This review utilized systematic searches of PubMed, Google Scholar, SCOPUS and SciFinder, including 14 full-text articles and 1,167 participants, consisting of 480 patients with LC and 687 normal controls. RESULTS The results indicated a significant increase in the kynurenine (KYN)/TRP ratio, with a large effect size (standardized mean difference, SMD = 0.755; confidence intervals, CI: 0.119;1.392), in LC patients compared to normal controls. Additionally, LC patients exhibited a significant decrease in TRP levels (SMD = -0.520, CI: -0.793; -0.246) and an increase in KYN levels after imputing missing studies (SMD = 1.176, CI: 0.474; 1.877), suggesting activation of the indoleamine 2,3-dioxygenase (IDO) enzyme and upregulation of the TRYCAT pathway. No significant elevation in TRYCAT-related neurotoxicity, kynurenic acid (KA)/KYN and 3-hydroxykynurenine (3-HK)/KYN ratios were observed in LC patients compared to normal controls. CONCLUSION The current findings suggest that an activated TRYCAT pathway, characterized by decreased TRP levels and maybe elevated KYN levels, plays a significant role in the pathophysiology of LC.
Collapse
Affiliation(s)
- Abbas F Almulla
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Yanin Thipakorn
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Bo Zhou
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China
| | - Aristo Vojdani
- Immunosciences Lab, Inc., Los Angeles, CA 90035, USA; Cyrex Laboratories, LLC, Phoenix, AZ 85034, USA
| | - Rossitsa Paunova
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria; Research Institute, Medical University of Plovdiv, Plovdiv, Bulgaria; Strategic Research and Innovation Program for the Development of MU - PLOVDIV-(SRIPD-MUP), European Union - NextGenerationEU
| | - Michael Maes
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria; Research Institute, Medical University of Plovdiv, Plovdiv, Bulgaria; Strategic Research and Innovation Program for the Development of MU - PLOVDIV-(SRIPD-MUP), European Union - NextGenerationEU; Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Cognitive Fitness and Biopsychological Technology Research Unit, Faculty of Medicine. Chulalongkorn University, Bangkok, 10330, Thailand, Bangkok, 10330, Thailand; Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, South Korea.
| |
Collapse
|
2
|
Pamart G, Gosset P, Le Rouzic O, Pichavant M, Poulain-Godefroy O. Kynurenine Pathway in Respiratory Diseases. Int J Tryptophan Res 2024; 17:11786469241232871. [PMID: 38495475 PMCID: PMC10943758 DOI: 10.1177/11786469241232871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/28/2024] [Indexed: 03/19/2024] Open
Abstract
The kynurenine pathway is the primary route for tryptophan catabolism and has received increasing attention as its association with inflammation and the immune system has become more apparent. This review provides a broad overview of the kynurenine pathway in respiratory diseases, from the initial observations to the characterization of the different cell types involved in the synthesis of kynurenine metabolites and the underlying immunoregulatory mechanisms. With a focus on respiratory infections, the various attempts to characterize the kynurenine/tryptophan (K/T) ratio as an inflammatory marker are reviewed. Its implication in chronic lung inflammation and its exacerbation by respiratory pathogens is also discussed. The emergence of preclinical interventional studies targeting the kynurenine pathway opens the way for the future development of new therapies.
Collapse
Affiliation(s)
- Guillaume Pamart
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 -CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Philippe Gosset
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 -CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Olivier Le Rouzic
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 -CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Muriel Pichavant
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 -CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Odile Poulain-Godefroy
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 -CIIL - Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
3
|
Gonzalez-Rubio J, Le-Trilling VTK, Baumann L, Cheremkhina M, Kubiza H, Luengen AE, Reuter S, Taube C, Ruetten S, Duarte Campos D, Cornelissen CG, Trilling M, Thiebes AL. SARS-CoV-2 particles promote airway epithelial differentiation and ciliation. Front Bioeng Biotechnol 2023; 11:1268782. [PMID: 38026867 PMCID: PMC10654538 DOI: 10.3389/fbioe.2023.1268782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction: The Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), which caused the coronavirus disease 2019 (COVID-19) pandemic, enters the human body via the epithelial cells of the airway tract. To trap and eject pathogens, the airway epithelium is composed of ciliated and secretory cells that produce mucus which is expelled through a process called mucociliary clearance. Methods: This study examines the early stages of contact between SARS-CoV-2 particles and the respiratory epithelium, utilizing 3D airway tri-culture models exposed to ultraviolet light-irradiated virus particles. These cultures are composed of human endothelial cells and human tracheal mesenchymal cells in a fibrin hydrogel matrix covered by mucociliated human tracheal epithelial cells. Results: We found that SARS-CoV-2 particles trigger a significant increase in ciliation on the epithelial surface instructed through a differentiation of club cells and basal stem cells. The contact with SARS-CoV-2 particles also provoked a loss of cell-cell tight junctions and impaired the barrier integrity. Further immunofluorescence analyses revealed an increase in FOXJ1 expression and PAK1/2 phosphorylation associated with particle-induced ciliation. Discussion: An understanding of epithelial responses to virus particles may be instrumental to prevent or treat respiratory infectious diseases such as COVID-19.
Collapse
Affiliation(s)
- Julian Gonzalez-Rubio
- Department of Biohybrid and Medical Textiles (BioTex), AME—Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | | | - Lea Baumann
- Department of Biohybrid and Medical Textiles (BioTex), AME—Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | - Maria Cheremkhina
- Department of Biohybrid and Medical Textiles (BioTex), AME—Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | - Hannah Kubiza
- Department of Biohybrid and Medical Textiles (BioTex), AME—Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | - Anja E. Luengen
- Department of Biohybrid and Medical Textiles (BioTex), AME—Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Department of Pulmonary Medicine, University Medical Center Essen—Ruhrlandklinik, Essen, Germany
| | - Sebastian Reuter
- Department of Pulmonary Medicine, University Medical Center Essen—Ruhrlandklinik, Essen, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Medical Center Essen—Ruhrlandklinik, Essen, Germany
| | - Stephan Ruetten
- Institute of Pathology, Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany
| | - Daniela Duarte Campos
- Bioprinting and Tissue Engineering Group, Center for Molecular Biology of Heidelberg University (ZMBH), Heidelberg, Germany
| | - Christian G. Cornelissen
- Department of Biohybrid and Medical Textiles (BioTex), AME—Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Clinic for Pneumology and Internal Intensive Care Medicine (Medical Clinic V), RWTH Aachen University Hospital, Aachen, Germany
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Anja Lena Thiebes
- Department of Biohybrid and Medical Textiles (BioTex), AME—Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
4
|
Guo L, Appelman B, Mooij-Kalverda K, Houtkooper RH, van Weeghel M, Vaz FM, Dijkhuis A, Dekker T, Smids BS, Duitman JW, Bugiani M, Brinkman P, Sikkens JJ, Lavell HAA, Wüst RCI, van Vugt M, Lutter R. Prolonged indoleamine 2,3-dioxygenase-2 activity and associated cellular stress in post-acute sequelae of SARS-CoV-2 infection. EBioMedicine 2023; 94:104729. [PMID: 37506544 PMCID: PMC10406961 DOI: 10.1016/j.ebiom.2023.104729] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 06/08/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Post-acute sequela of SARS-CoV-2 infection (PASC) encompass fatigue, post-exertional malaise and cognitive problems. The abundant expression of the tryptophan-catabolizing enzyme indoleamine 2,3-dioxygenase-2 (IDO2) in fatal/severe COVID-19, led us to determine, in an exploratory observational study, whether IDO2 is expressed and active in PASC, and may correlate with pathophysiology. METHODS Plasma or serum, and peripheral blood mononuclear cells (PBMC) were obtained from well-characterized PASC patients and SARS-CoV-2-infected individuals without PASC. We assessed tryptophan and its degradation products by UPLC-MS/MS. IDO2 activity, its potential consequences, and the involvement of the aryl hydrocarbon receptor (AHR) in IDO2 expression were determined in PBMC from another PASC cohort by immunohistochemistry (IHC) for IDO2, IDO1, AHR, kynurenine metabolites, autophagy, and apoptosis. These PBMC were also analyzed by metabolomics and for mitochondrial functioning by respirometry. IHC was also performed on autopsy brain material from two PASC patients. FINDINGS IDO2 is expressed and active in PBMC from PASC patients, as well as in brain tissue, long after SARS-CoV-2 infection. This is paralleled by autophagy, and in blood cells by reduced mitochondrial functioning, reduced intracellular levels of amino acids and Krebs cycle-related compounds. IDO2 expression and activity is triggered by SARS-CoV-2-infection, but the severity of SARS-CoV-2-induced pathology appears related to the generated specific kynurenine metabolites. Ex vivo, IDO2 expression and autophagy can be halted by an AHR antagonist. INTERPRETATION SARS-CoV-2 infection triggers long-lasting IDO2 expression, which can be halted by an AHR antagonist. The specific kynurenine catabolites may relate to SARS-CoV-2-induced symptoms and pathology. FUNDING None.
Collapse
Affiliation(s)
- Lihui Guo
- Department Experimental Immunology, Amsterdam Infection and Immunity Center, Amsterdam University Medical Centers (UMC), location Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Brent Appelman
- Center for Experimental and Molecular Medicine, Amsterdam UMC, Amsterdam Institute for Infection and Immunity, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Kirsten Mooij-Kalverda
- Department Pulmonary Medicine, Amsterdam UMC, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology and Metabolism Institute, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences Institute, Amsterdam, the Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology and Metabolism Institute, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences Institute, Amsterdam, the Netherlands; Core Facility Metabolomics, Amsterdam UMC, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Frédéric M Vaz
- Core Facility Metabolomics, Amsterdam UMC, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital, Amsterdam UMC, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Inborn Errors of Metabolism, Amsterdam, the Netherlands
| | - Annemiek Dijkhuis
- Department Experimental Immunology, Amsterdam Infection and Immunity Center, Amsterdam University Medical Centers (UMC), location Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Tamara Dekker
- Department Experimental Immunology, Amsterdam Infection and Immunity Center, Amsterdam University Medical Centers (UMC), location Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Barbara S Smids
- Department Experimental Immunology, Amsterdam Infection and Immunity Center, Amsterdam University Medical Centers (UMC), location Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Jan Willem Duitman
- Department Experimental Immunology, Amsterdam Infection and Immunity Center, Amsterdam University Medical Centers (UMC), location Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Department Pulmonary Medicine, Amsterdam UMC, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Marianna Bugiani
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Paul Brinkman
- Department Pulmonary Medicine, Amsterdam UMC, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Jonne J Sikkens
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - H A Ayesha Lavell
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Rob C I Wüst
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Michèle van Vugt
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - René Lutter
- Department Experimental Immunology, Amsterdam Infection and Immunity Center, Amsterdam University Medical Centers (UMC), location Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Department Pulmonary Medicine, Amsterdam UMC, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| |
Collapse
|
5
|
Gazi MA, Siddique MA, Alam MA, Hossaini F, Hasan MM, Fahim SM, Wahid BZ, Kabir MM, Das S, Mahfuz M, Ahmed T. Plasma Kynurenine to Tryptophan Ratio Is Not Associated with Undernutrition in Adults but Reduced after Nutrition Intervention: Results from a Community-Based Study in Bangladesh. Nutrients 2022; 14:nu14091708. [PMID: 35565678 PMCID: PMC9104876 DOI: 10.3390/nu14091708] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 12/10/2022] Open
Abstract
Infections and persistent immunological activation are linked to increased kynurenine (KYN) and the KYN-to-Tryptophan (TRP) or KT ratio and may be critical factors in undernutrition. We sought to determine the association between the KT ratio and adult malnutrition, as well as investigate if nutritional supplementation had any influence on the decrease of the KT ratio. A total of 525 undernourished adults aged 18–45 years were recruited and provided a nutrition intervention for 60 feeding days. TRP and KYN concentrations were determined from plasma samples using LC-MS/MS. At baseline, the median (interquartile range (IQR)) TRP, KYN and KT ratios were 24.1 (17.6, 34.3) µmol/L, 0.76 (0.53, 1.18) µmol/L and 30.9 (24.5, 41.7), respectively. Following intervention, the median (IQR) KYN and KT ratios were significantly reduced to 0.713 (0.46, 1.12) µmol/L and 27.5 (21.3, 35.8). The KT ratio was found to be inversely linked with adult BMI (coefficient: −0.09; 95% CI: −0.18, 0.004; p-value = 0.06) but not statistically significant. Additionally, Plasma CRP was correlated positively, while LRP1 was inversely correlated with the KT ratio. Our data suggest that in Bangladeshi adults, the KT ratio is not related to the pathophysiology of malnutrition but correlated with inflammatory and anti-inflammatory biomarkers, and the ratio can be reduced by a nutrition intervention.
Collapse
Affiliation(s)
- Md. Amran Gazi
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
| | - Md. Abdullah Siddique
- Emerging Infections and Parasitology Laboratory, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.S.); (M.M.K.)
| | - Md. Ashraful Alam
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
| | - Farzana Hossaini
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
| | - Md. Mehedi Hasan
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
| | - Shah Mohammad Fahim
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
| | - Barbie Zaman Wahid
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
| | - Md. Mamun Kabir
- Emerging Infections and Parasitology Laboratory, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.S.); (M.M.K.)
| | - Subhasish Das
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
- Faculty of Medicine and Life Sciences, University of Tampere, 33100 Tampere, Finland
- Correspondence:
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
- Department of Global Health, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
6
|
Silva ALM, Silva ECO, Botelho RM, Tenorio LPG, Marques ALX, Rodrigues IBAC, Almeida LIM, Sousa AKA, Pires KSN, Tanabe ISB, Allard MJ, Sébire G, Souza ST, Fonseca EJS, Borbely KSC, Borbely AU. Uvaol Prevents Group B Streptococcus-Induced Trophoblast Cells Inflammation and Possible Endothelial Dysfunction. Front Physiol 2021; 12:766382. [PMID: 34925062 PMCID: PMC8678414 DOI: 10.3389/fphys.2021.766382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/02/2021] [Indexed: 12/25/2022] Open
Abstract
Group B Streptococcus (GBS) infection during pregnancy is involved in maternal sepsis, chorioamnionitis, prematurity, fetal infection, neonatal sepsis, and neurodevelopmental alterations. The GBS-induced chorioamnionitis leads to a plethora of immune and trophoblast cells alterations that could influence endothelial cells to respond differently to angiogenic mediators and alter placental vascular structure and function in pregnant women. In this context, preventive measures are needed to reduce such dysfunctions. As such, we evaluated the effects of a non-lethal exposure to inactivated GBS on trophoblast cells and chorionic villi explants, and if the treatment with uvaol would mitigate these effects. The concentration of 106 CFU of GBS was chosen since it was unable to reduce the HTR-8/SVneo cell line nor term chorionic villi explant viability. Raman spectroscopy of trophoblast cells showed significant alterations in their biochemical signature, mostly reverted by uvaol. GBS exposure increased HTR-8/SVneo cells IL-1β and IFN-γ production, phagocytosis, oxidative stress, and decreased trophoblast cell migration. The Ea.hy926 endothelial cell line produced angiopoietin-2, CXCL-8, EGF, FGF-b, IL-6, PlGF, sPECAM-1, and VEGF in culture. When co-cultured in invasion assay with HTR-8/SVneo trophoblast cells, the co-culture had increased production of angiopoietin-2, CXCL-8, FGF-b, and VEGF, while reduced sPECAM-1 and IL-6. GBS exposure led to increased CXCL-8 and IL-6 production, both prevented by uvaol. Chorionic villi explants followed the same patterns of production when exposed to GBS and response to uvaol treatment as well. These findings demonstrate that, even a non-lethal concentration of GBS causes placental inflammation and oxidative stress, reduces trophoblast invasion of endothelial cells, and increases CXCL-8 and IL-6, key factors that participate in vascular dysregulation observed in several diseases. Furthermore, uvaol treatment prevented most of the GBS-provoked changes. Hence, uvaol could prevent the harmful effects of GBS infection for both the mother and the fetus.
Collapse
Affiliation(s)
- Ana Lucia Mendes Silva
- Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, Brazil
| | | | - Rayane Martins Botelho
- Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, Brazil
| | | | - Aldilane Lays Xavier Marques
- Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, Brazil
| | | | | | | | - Keyla Silva Nobre Pires
- Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, Brazil
| | | | | | - Guillaume Sébire
- Department of Pediatrics, McGill University, Montreal, QC, Canada.,Department of Pediatrics, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Samuel Teixeira Souza
- Optics and Nanoscopy Group, Physics Institute, Federal University of Alagoas, Maceio, Brazil
| | | | - Karen Steponavicius Cruz Borbely
- Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, Brazil.,Faculty of Nutrition, Federal University of Alagoas, Maceio, Brazil
| | - Alexandre Urban Borbely
- Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, Brazil
| |
Collapse
|
7
|
Dagenais-Lussier X, Loucif H, Beji C, Telittchenko R, Routy JP, van Grevenynghe J. Latest developments in tryptophan metabolism: Understanding its role in B cell immunity. Cytokine Growth Factor Rev 2021; 59:111-117. [DOI: 10.1016/j.cytogfr.2021.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022]
|
8
|
Pieretti JC, Rubilar O, Weller RB, Tortella GR, Seabra AB. Nitric oxide (NO) and nanoparticles - Potential small tools for the war against COVID-19 and other human coronavirus infections. Virus Res 2021; 291:198202. [PMID: 33086123 PMCID: PMC7568847 DOI: 10.1016/j.virusres.2020.198202] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/11/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022]
Abstract
The endogenous free radical nitric oxide (NO) plays a pivotal role in the immunological system. NO has already been reported as a potential candidate for use in the treatment of human coronavirus infections, including COVID-19. In fact, inhaled NO has been used in clinical settings for its antiviral respiratory action, and in the regulation of blood pressure to avoid clot formation. In this mini-review, we discuss recent progress concerning the antivirus activity of NO in clinical, pre-clinical and research settings, and its beneficial effects in the treatment of clinical complications in patients infected with coronaviruses and other respiratory viral diseases, including COVID-19. We also highlight promising therapeutic effects of NO donors allied to nanomaterials to combat COVID-19 and other human coronavirus infections. Nanomaterials can be designed to deliver sustained, localized NO release directly at the desired application site, enhancing the beneficial effects of NO and minimizing the side effects. Challenges and perspectives are presented to open new fields of research.
Collapse
Affiliation(s)
- Joana C Pieretti
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, SP, Brazil
| | - Olga Rubilar
- Department of Chemical Engineering, Universidad de La Frontera, Temuco, Chile; Centro de Excelencia en Investigación Biotecnologica Aplicada al Medio Ambiente (CIBAMA-BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Richard B Weller
- Centre for Inflammation Research, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Gonzalo R Tortella
- Department of Chemical Engineering, Universidad de La Frontera, Temuco, Chile; Centro de Excelencia en Investigación Biotecnologica Aplicada al Medio Ambiente (CIBAMA-BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Amedea B Seabra
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, SP, Brazil.
| |
Collapse
|
9
|
Keshavarz M, Solaymani-Mohammadi F, Namdari H, Arjeini Y, Mousavi MJ, Rezaei F. Metabolic host response and therapeutic approaches to influenza infection. Cell Mol Biol Lett 2020; 25:15. [PMID: 32161622 PMCID: PMC7059726 DOI: 10.1186/s11658-020-00211-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 02/26/2020] [Indexed: 12/17/2022] Open
Abstract
Based on available metabolomic studies, influenza infection affects a variety of cellular metabolic pathways to ensure an optimal environment for its replication and production of viral particles. Following infection, glucose uptake and aerobic glycolysis increase in infected cells continually, which results in higher glucose consumption. The pentose phosphate shunt, as another glucose-consuming pathway, is enhanced by influenza infection to help produce more nucleotides, especially ATP. Regarding lipid species, following infection, levels of triglycerides, phospholipids, and several lipid derivatives undergo perturbations, some of which are associated with inflammatory responses. Also, mitochondrial fatty acid β-oxidation decreases significantly simultaneously with an increase in biosynthesis of fatty acids and membrane lipids. Moreover, essential amino acids are demonstrated to decline in infected tissues due to the production of large amounts of viral and cellular proteins. Immune responses against influenza infection, on the other hand, could significantly affect metabolic pathways. Mainly, interferon (IFN) production following viral infection affects cell function via alteration in amino acid synthesis, membrane composition, and lipid metabolism. Understanding metabolic alterations required for influenza virus replication has revealed novel therapeutic methods based on targeted inhibition of these cellular metabolic pathways.
Collapse
Affiliation(s)
- Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | | | - Haideh Namdari
- Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaser Arjeini
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Mousavi
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology and Allergy, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Farhad Rezaei
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- National Influenza Center, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Lin YT, Lin CF, Yeh TH. Influenza A virus infection induces indoleamine 2,3-dioxygenase (IDO) expression and modulates subsequent inflammatory mediators in nasal epithelial cells. Acta Otolaryngol 2020; 140:149-156. [PMID: 31852346 DOI: 10.1080/00016489.2019.1700304] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Background: Nasal epithelial cells are the first site of encounter of the influenza virus, and their innate immune response might define subsequent inflammatory direction.Aims/objectives: We used metabolomics analysis to identify metabolic changes and the regulation of inflammatory cytokines in nasal epithelial cells upon influenza virus infection.Material and methods: We cultured nasal epithelial cells using air-liquid interface (ALI) model. Influenza virus (PR8) infection followed by metabolomic analysis was performed. Furthermore, cytokine expression was analyzed by cytokine array and RT-qPCR.Results: Metabolomic analysis revealed depletion of the tryptophan and accumulation of its metabolite, kynurenine, within 48 h. The major enzyme involved in the tryptophan metabolic pathway, indoleamine 2,3-dioxygenase (IDO), was overexpressed after infection. Cytokine expression array after infection showed increased levels of IL-1α, CCL2, IL-6, CXCL10, CCL5, and CXCL11, and after using 1-methyltryptophan (1-MT) as inhibitor, the expression levels of IL-6 and G-CSF were reduced.Conclusions and significance: Viral infection results in depletion of tryptophan and accumulation of kynurenine via increased cellular IDO activity. Inhibition of IDO activity or replenishment of tryptophan by local application may be a good therapeutic strategy for limiting the initial damage caused by influenza virus in nasal epithelial cells.
Collapse
Affiliation(s)
- Yi-Tsen Lin
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
- Graduate School of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chih-Feng Lin
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Te-Huei Yeh
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Otolaryngology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
11
|
Benavente FM, Soto JA, Pizarro-Ortega MS, Bohmwald K, González PA, Bueno SM, Kalergis AM. Contribution of IDO to human respiratory syncytial virus infection. J Leukoc Biol 2019; 106:933-942. [PMID: 31091352 PMCID: PMC7166882 DOI: 10.1002/jlb.4ru0219-051rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/26/2019] [Accepted: 05/05/2019] [Indexed: 12/18/2022] Open
Abstract
IDO is an enzyme that participates in the degradation of tryptophan (Trp), which is an essential amino acid necessary for vital cellular processes. The degradation of Trp and the metabolites generated by the enzymatic activity of IDO can have immunomodulating effects, notably over T cells, which are particularly sensitive to the absence of Trp and leads to the inhibition of T cell activation, cell death, and the suppression of T cell effector functions. Noteworthy, T cells participate in the cellular immune response against the human respiratory syncytial virus (hRSV) and are essential for viral clearance, as well as the total recovery of the host. Furthermore, inadequate or non‐optimal polarization of T cells is often seen during the acute phase of the disease caused by this pathogen. Here, we discuss the capacity of hRSV to exploit the immunosuppressive features of IDO to reduce T cell function, thus acquiring relevant aspects during the biology of the virus. Additionally, we review studies on the influence of IDO over T cell activation and its relationship with hRSV infection.
Collapse
Affiliation(s)
- Felipe M Benavente
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge A Soto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Magdalena S Pizarro-Ortega
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
12
|
Abstract
Our antiviral arsenal to fight influenza viruses is limited and we need novel anti-flu drugs. Recently, cellular drug targets came into focus and omics analysis were instrumental to suggest candidate factors. In this issue of The FEBS Journal, Kainov and colleagues used transcriptome data to investigate virus-induced changes in tryptophan metabolism that may serve as immunomodulatory approach against viruses.
Collapse
Affiliation(s)
- Yvonne Boergeling
- Institute of Virology (IVM), Westfaelische-Wilhelms University, Muenster, Germany
| | - Stephan Ludwig
- Institute of Virology (IVM), Westfaelische-Wilhelms University, Muenster, Germany
| |
Collapse
|
13
|
Xie Z, Zhang M, Xiong W, Wan HY, Zhao XC, Xie T, Lei H, Lin ZC, Luo DS, Liang XL, Chen YH. Immunotolerant indoleamine-2,3-dioxygenase is increased in condyloma acuminata. Br J Dermatol 2017; 177:809-817. [PMID: 28132413 DOI: 10.1111/bjd.15356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND The tryptophan-depleting enzyme indoleamine-2,3-dioxygenase (IDO) is critical for the regulation of immunotolerance and plays an important role in immune-associated skin diseases. OBJECTIVES To analyse the level of IDO in condyloma acuminata (CA) and its role in this condition. METHODS IDO expression was assessed in the skin and peripheral blood of healthy controls and patients with CA. To assess the role of skin IDO in immunity, the ability of isolated epidermal cells to metabolize tryptophan and the influence on polyclonal T-cell mitogen (PHA)-stimulated T-cell proliferation were explored. RESULTS IDO median fluorescence intensities in peripheral blood mononuclear cells from patients with CA were similar to those from healthy controls. Immunohistochemistry showed that IDO+ cells were rare in normal skin and the control skin of patients with CA, but were greatly accumulated in wart tissue. Most fluorescence signals of IDO+ cells did not overlap with those of CD1a+ Langerhans cells. Human papillomavirus (HPV) DNA probe in situ hybridization showed a large number of IDO+ cells in the HPV- site. Keratinocytes in the skin of healthy controls and the circumcised skin of patients with CA could minimally transform tryptophan into kynurenine, but IDO-competent epidermal cells from warts could transform tryptophan. In addition, these IDO-competent epidermal cells could inhibit PHA-stimulated T-cell proliferation. The addition of an IDO inhibitor, 1-methyl-d-tryptophan, restored the inhibited T-cell proliferation. CONCLUSIONS Abnormally localized high IDO expression might be involved in the formation of a local immunotolerant microenvironment.
Collapse
Affiliation(s)
- Z Xie
- Department of Dermatology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, China
| | - M Zhang
- Department of Gastroenterology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - W Xiong
- Division of Urology and Transplantation, Department of Surgery, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, China
| | - H Y Wan
- Department of Dermatology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, China
| | - X C Zhao
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Nanfang Medical University, Guangzhou, China
| | - T Xie
- Department of Dermatology, Guangdong Provincial Hospital of Chinese Hospital, Guangzhou, China
| | - H Lei
- Department of Dermatology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, China
| | - Z C Lin
- Department of Dermatology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, China
| | - D S Luo
- Department of Dermatology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, China
| | - X L Liang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Y H Chen
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
14
|
Kim SB, Choi JY, Uyangaa E, Patil AM, Hossain FMA, Hur J, Park SY, Lee JH, Kim K, Eo SK. Blockage of indoleamine 2,3-dioxygenase regulates Japanese encephalitis via enhancement of type I/II IFN innate and adaptive T-cell responses. J Neuroinflammation 2016; 13:79. [PMID: 27090635 PMCID: PMC4835894 DOI: 10.1186/s12974-016-0551-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/12/2016] [Indexed: 11/24/2022] Open
Abstract
Background Japanese encephalitis (JE), a leading cause of viral encephalitis, is characterized by extensive neuroinflammation following infection with neurotropic JE virus (JEV). Indoleamine 2,3-dioxygenase (IDO) has been identified as an enzyme associated with immunoregulatory function. Although the regulatory role of IDO in viral replication has been postulated, the in vivo role of IDO activity has not been fully addressed in neurotropic virus-caused encephalitis. Methods Mice in which IDO activity was inhibited by genetic ablation or using a specific inhibitor were examined for mortality and clinical signs after infection. Neuroinflammation was evaluated by central nervous system (CNS) infiltration of leukocytes and cytokine expression. IDO expression, viral burden, JEV-specific T-cell, and type I/II interferon (IFN-I/II) innate responses were also analyzed. Results Elevated expression of IDO activity in myeloid and neuron cells of the lymphoid and CNS tissues was closely associated with clinical signs of JE. Furthermore, inhibition of IDO activity enhanced resistance to JE, reduced the viral burden in lymphoid and CNS tissues, and resulted in early and increased CNS infiltration by Ly-6Chi monocytes, NK, CD4+, and CD8+ T-cells. JE amelioration in IDO-ablated mice was also associated with enhanced NK and JEV-specific T-cell responses. More interestingly, IDO ablation induced rapid enhancement of type I IFN (IFN-I) innate responses in CD11c+ dendritic cells (DCs), including conventional and plasmacytoid DCs, following JEV infection. This enhanced IFN-I innate response in IDO-ablated CD11c+ DCs was coupled with strong induction of PRRs (RIG-I, MDA5), transcription factors (IRF7, STAT1), and antiviral ISG genes (Mx1, Mx2, ISG49, ISG54, ISG56). IDO ablation also enhanced the IFN-I innate response in neuron cells, which may delay the spread of virus in the CNS. Finally, we identified that IDO ablation in myeloid cells derived from hematopoietic stem cells (HSCs) dominantly contributed to JE amelioration and that HSC-derived leukocytes played a key role in the enhanced IFN-I innate responses in the IDO-ablated environment. Conclusions Inhibition of IDO activity ameliorated JE via enhancement of antiviral IFN-I/II innate and adaptive T-cell responses and increased CNS infiltration of peripheral leukocytes. Therefore, our data provide valuable insight into the use of IDO inhibition by specific inhibitors as a promising tool for therapeutic and prophylactic strategies against viral encephalitis caused by neurotropic viruses.
Collapse
Affiliation(s)
- Seong Bum Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Jin Young Choi
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Erdenebileg Uyangaa
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Ajit Mahadev Patil
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Ferdaus Mohd Altaf Hossain
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Jin Hur
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Sang-Youel Park
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea.,Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, 54896, Republic of Korea
| | - John-Hwa Lee
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea.,Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, 54896, Republic of Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Seong Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea. .,Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, 54896, Republic of Korea.
| |
Collapse
|
15
|
Abstract
IDO1 (indoleamine 2,3-dioxygenase 1) is a member of a unique class of mammalian haem dioxygenases that catalyse the oxidative catabolism of the least-abundant essential amino acid, L-Trp (L-tryptophan), along the kynurenine pathway. Significant increases in knowledge have been recently gained with respect to understanding the fundamental biochemistry of IDO1 including its catalytic reaction mechanism, the scope of enzyme reactions it catalyses, the biochemical mechanisms controlling IDO1 expression and enzyme activity, and the discovery of enzyme inhibitors. Major advances in understanding the roles of IDO1 in physiology and disease have also been realised. IDO1 is recognised as a prominent immune regulatory enzyme capable of modulating immune cell activation status and phenotype via several molecular mechanisms including enzyme-dependent deprivation of L-Trp and its conversion into the aryl hydrocarbon receptor ligand kynurenine and other bioactive kynurenine pathway metabolites, or non-enzymatic cell signalling actions involving tyrosine phosphorylation of IDO1. Through these different modes of biochemical signalling, IDO1 regulates certain physiological functions (e.g. pregnancy) and modulates the pathogenesis and severity of diverse conditions including chronic inflammation, infectious disease, allergic and autoimmune disorders, transplantation, neuropathology and cancer. In the present review, we detail the current understanding of IDO1’s catalytic actions and the biochemical mechanisms regulating IDO1 expression and activity. We also discuss the biological functions of IDO1 with a focus on the enzyme's immune-modulatory function, its medical implications in diverse pathological settings and its utility as a therapeutic target.
Collapse
|
16
|
Fox JM, Crabtree JM, Sage LK, Tompkins SM, Tripp RA. Interferon Lambda Upregulates IDO1 Expression in Respiratory Epithelial Cells After Influenza Virus Infection. J Interferon Cytokine Res 2015; 35:554-62. [PMID: 25756191 PMCID: PMC4507134 DOI: 10.1089/jir.2014.0052] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 01/19/2015] [Indexed: 01/10/2023] Open
Abstract
Influenza infection causes an increase in indoleamine 2, 3-dioxygenase (IDO) activity in the lung parenchyma. IDO catabolizes tryptophan into kynurenine, leading to immune dampening. Multiple cell types express IDO, and while IFN-γ upregulates IDO in dendritic cells and macrophages, it is unclear how IDO is affected in respiratory epithelial cells during influenza infection. In this study, the role of IFN-λ in IDO regulation was investigated after influenza infection of respiratory epithelial cells. IDO1 expression increased concurrently with IFN-λ expression. In differentiated NHBE cells, the IDO metabolite was released basolaterally. Recombinant IFN-λ upregulated IDO1 activity, and silencing of IFN-λ decreased IDO1 expression during influenza infection. During IFN-λ stimulation, most differentiated cell types are able to express IDO but during influenza infection, IDO is primarily expressed in uninfected cells. These studies show a role for IDO in the host response to influenza infection, and they provide insights into novel approaches for enhancing vaccine responses and therapeutic approaches.
Collapse
Affiliation(s)
- Julie M Fox
- Department of Infectious Diseases, University of Georgia , Athens, Georgia
| | | | - Leo K Sage
- Department of Infectious Diseases, University of Georgia , Athens, Georgia
| | - S Mark Tompkins
- Department of Infectious Diseases, University of Georgia , Athens, Georgia
| | - Ralph A Tripp
- Department of Infectious Diseases, University of Georgia , Athens, Georgia
| |
Collapse
|
17
|
Takegawa D, Nakanishi T, Hirao K, Yumoto H, Takahashi K, Matsuo T. Modulatory roles of interferon-γ through indoleamine 2, 3-dioxygenase induction in innate immune response of dental pulp cells. J Endod 2014; 40:1382-7. [PMID: 25146019 DOI: 10.1016/j.joen.2014.03.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 01/31/2014] [Accepted: 03/21/2014] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Marked infiltration of inflammatory cells such as activated T cells producing interferon-γ (IFN-γ) is observed in severe pulpitis. However, the roles of IFN-γ in the innate immune response of dental pulp have not been reported. Indoleamine 2, 3-dioxygenase (IDO) is a regulator of immune responses, and the IDO expression is induced by IFN-γ in many cells whose expression in dental pulp is unknown. The purpose of this study was to determine the role of IFN-γ in the immune response through microbial pattern recognition receptors (PRRs) such as Toll-like receptors or nucleotide-binding oligomerization domain-like receptors on the production of proinflammatory cytokines such as CXCL10 and interleukin (IL)-6 and the expression of IDO in cultured human dental pulp cells (HDPCs). METHODS HDPCs were established from explant cultures of healthy pulp tissues. CXCL10 and IL-6 production was determined using enzyme-linked immunosorbent assay. Confirmation of IDO localization in dental pulp tissues was examined using immunohistochemistry. IDO expression in HDPCs was analyzed by immunoblot. RESULTS IFN-γ significantly up-regulated CXCL10 and IL-6 production in the HDPCs stimulated with ligands for PRRs in a concentration-dependent manner. The expression of IDO was detected in inflamed pulp tissue. In addition, IFN-γ in combination with the PRR ligands enhanced IDO expression in HDPCs compared with IFN-γ alone. Moreover, CXCL10 production in IFN-γ-stimulated HDPCs was inhibited by an IDO inhibitor. CONCLUSIONS This study showed the synergistic effects by IFN-γ on cytokine production and IDO expression in HDPCs, suggesting that IFN-γ may modulate the innate immune response of dental pulp.
Collapse
Affiliation(s)
- Daisuke Takegawa
- Department of Conservative Dentistry, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Tadashi Nakanishi
- Department of Conservative Dentistry, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan.
| | - Kouji Hirao
- Department of Conservative Dentistry, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Hiromichi Yumoto
- Department of Conservative Dentistry, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Kanako Takahashi
- Department of Conservative Dentistry, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Takashi Matsuo
- Department of Conservative Dentistry, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| |
Collapse
|
18
|
Fox JM, Sage LK, Poore S, Johnson S, Tompkins SM, Tripp RA. Drug analog inhibition of indoleamine 2,3-dioxygenase (IDO) activity modifies pattern recognition receptor expression and proinflammatory cytokine responses early during influenza virus infection. J Leukoc Biol 2014; 96:447-52. [PMID: 24799604 DOI: 10.1189/jlb.3ab0114-046rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Influenza virus is recognized by PRRs, which are critical in the early response to virus infection and induction of proinflammatory cytokines. IDO is increased in the lung of mice immediately following influenza infection, and the presence of IDO has been shown to mediate immune suppression through depletion of trp and reduction in IL-6 production. To determine the role of IDO activity in the early immune response to influenza infection, IDO activity was inhibited using the synthetic analog, 1MT. The results show that IDO inhibition enhanced proinflammatory cytokine gene and protein expression at 24 and 48 h postinfection, respectively, compared with control-treated mice and affected PRR expression. The enhanced proinflammatory response in the presence of 1MT was attributed to macrophages in the airways, as Raw264.7 and primary AMs showed enhanced production of IFN-β, IL-1β, IL-6, and TNF-α in the presence of 1MT. These findings provide important knowledge for the role of IDO during initial host response to influenza infection.
Collapse
Affiliation(s)
- Julie M Fox
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Leo K Sage
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Spencer Poore
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Scott Johnson
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - S Mark Tompkins
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Ralph A Tripp
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
19
|
The expression of the eotaxins IL-6 and CXCL8 in human epithelial cells from various levels of the respiratory tract. Cell Mol Biol Lett 2013; 18:612-30. [PMID: 24297684 PMCID: PMC6275597 DOI: 10.2478/s11658-013-0107-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 11/27/2013] [Indexed: 11/23/2022] Open
Abstract
Airway epithelium acts as multifunctional site of response in the respiratory tract. Epithelial activity plays an important part in the pathophysiology of obstructive lung disease. In this study, we compare normal human epithelial cells from various levels of the respiratory tract in terms of their reactivity to pro-allergic and pro-inflammatory stimulation. Normal human nasal, bronchial and small airway epithelial cells were stimulated with IL-4 and IL-13. The expressions of the eotaxins IL-6 and CXCL8 were evaluated at the mRNA and protein levels. The effects of pre-treatment with IFN-γ on the cell reactivity were measured, and the responses to TNF-α, LPS and IFN-γ were evaluated. All of the studied primary cells expressed CCL26, IL-6 and IL-8 after IL-4 or IL-13 stimulation. IFN-γ pre-treatment resulted in decreased CCL26 and increased IL-6 expression in the nasal and small airway cells, but this effect was not observed in the bronchial cells. IL-6 and CXCL8 were produced in varying degrees by all of the epithelial primary cells in cultures stimulated with TNF-α, LPS or IFN-γ. We showed that epithelial cells from the various levels of the respiratory tract act in a united way, responding in a similar manner to stimulation with IL-4 and IL-13, showing similar reactivity to TNF-α and LPS, and giving an almost unified response to IFN-γ pre-stimulation.
Collapse
|
20
|
Chowdhury S, Dijkhuis A, Steiert S, Lutter R. IL-17 attenuates degradation of ARE-mRNAs by changing the cooperation between AU-binding proteins and microRNA16. PLoS Genet 2013; 9:e1003747. [PMID: 24086143 PMCID: PMC3784493 DOI: 10.1371/journal.pgen.1003747] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 07/10/2013] [Indexed: 01/25/2023] Open
Abstract
Interleukin 17A (IL-17), a mediator implicated in chronic and severe inflammatory diseases, enhances the production of pro-inflammatory mediators by attenuating decay of the encoding mRNAs. The decay of many of these mRNAs depends on proteins (AUBps) that target AU-rich elements in the 3′-untranslated region of mRNAs and facilitate either mRNA decay or stabilization. Here we show that AUBps and the target mRNA assemble in a novel ribonucleoprotein complex in the presence of microRNA16 (miR16), which leads to the degradation of the target mRNA. Notably, IL-17 attenuates miR16 expression and promotes the binding of stabilizing AUBps over that of destabilizing AUBps, reducing mRNA decay. These findings indicate that miR16 independently of a seed sequence, directs the competition between degrading and stabilizing AUBps for target mRNAs. Since AUBps affect expression of about 8% of the human transcriptome and miR16 is ubiquitously expressed, IL-17 may in addition to inflammation affect many other cellular processes. Inflammation is driven by inflammatory mediators. Interleukin 17A (IL-17) is implicated in chronic and severe inflammation and exaggerates production of inflammatory mediators. This is due, at least in part, to the IL-17-attenuated degradation of mRNAs encoding these inflammatory mediators, but the underlying mechanism has remained elusive. Most of these mRNAs contain AU-rich elements in their 3′-untranslated region and are targeted by AU-binding proteins (AUBps) that promote either mRNA degradation or stabilization. Here we show that IL-17 directs the AU-mediated mRNA degradation (AMD pathway) by modulating the interaction of degrading and stabilizing AUBps via microRNA16 (miR16). Whereas microRNAs target mRNAs to the RISC pathway for degradation by binding to a seed sequence, miR16 drives degradation by the AMD pathway without an apparent seed sequence. Transcriptome analyses have revealed that the expression of 8% of all eukaryotic transcripts is dependent on the AMD pathway. Therefore, the impact of IL-17 on inflammatory diseases may extend beyond the production of inflammatory mediators to processes like tissue repair, cell cycle, etc. In addition, targeting AUBp and/or miR16 may provide a novel therapeutic option to combat the IL-17 axis of inflammation.
Collapse
Affiliation(s)
- Saheli Chowdhury
- Department of Respiratory Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- * E-mail:
| | - Annemiek Dijkhuis
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sabrina Steiert
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - René Lutter
- Department of Respiratory Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Inflammation and repeated infections in CGD: two sides of a coin. Cell Mol Life Sci 2011; 69:7-15. [PMID: 22083605 PMCID: PMC3249194 DOI: 10.1007/s00018-011-0834-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 09/13/2011] [Accepted: 09/13/2011] [Indexed: 01/13/2023]
Abstract
Chronic granulomatous disease (CGD) is an uncommon congenital immunodeficiency seen approximately in 1 of 250,000 individuals. It is caused by a profound defect in a burst of oxygen consumption that normally accompanies phagocytosis in all myeloid cells (neutrophils, eosinophils, monocytes, and macrophages). This “respiratory burst” involves the catalytic conversion of molecular oxygen to the oxygen free-radical superoxide, which in turn gives rise to hydrogen peroxide, hypochlorous acid, and hydroxyl radicals. These oxygen derivatives play a critical role in the killing of pathogenic bacteria and fungi. As a result of the failure to activate the respiratory burst in their phagocytes, the majority of CGD patients suffer from severe recurrent infections and rather unexplained prolonged inflammatory reactions that may result in granulomatous lesions. Both may cause severe organ dysfunction depending on the tissues involved. Preventive measures as well as rapid (invasive) diagnostic procedures are required to successfully treat CGD. Hematopoietic stem cell transplantation may be a serious option in some of the patients.
Collapse
|
22
|
Darcy CJ, Davis JS, Woodberry T, McNeil YR, Stephens DP, Yeo TW, Anstey NM. An observational cohort study of the kynurenine to tryptophan ratio in sepsis: association with impaired immune and microvascular function. PLoS One 2011; 6:e21185. [PMID: 21731667 PMCID: PMC3120841 DOI: 10.1371/journal.pone.0021185] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Accepted: 05/23/2011] [Indexed: 12/16/2022] Open
Abstract
Both endothelial and immune dysfunction contribute to the high mortality rate in human sepsis, but the underlying mechanisms are unclear. In response to infection, interferon-γ activates indoleamine 2,3-dioxygenase (IDO) which metabolizes the essential amino acid tryptophan to the toxic metabolite kynurenine. IDO can be expressed in endothelial cells, hepatocytes and mononuclear leukocytes, all of which contribute to sepsis pathophysiology. Increased IDO activity (measured by the kynurenine to tryptophan [KT] ratio in plasma) causes T-cell apoptosis, vasodilation and nitric oxide synthase inhibition. We hypothesized that IDO activity in sepsis would be related to plasma interferon-γ, interleukin-10, T cell lymphopenia and impairment of microvascular reactivity, a measure of endothelial nitric oxide bioavailability. In an observational cohort study of 80 sepsis patients (50 severe and 30 non-severe) and 40 hospital controls, we determined the relationship between IDO activity (plasma KT ratio) and selected plasma cytokines, sepsis severity, nitric oxide-dependent microvascular reactivity and lymphocyte subsets in sepsis. Plasma amino acids were measured by high performance liquid chromatography and microvascular reactivity by peripheral arterial tonometry. The plasma KT ratio was increased in sepsis (median 141 [IQR 64–235]) compared to controls (36 [28–52]); p<0.0001), and correlated with plasma interferon-γ and interleukin-10, and inversely with total lymphocyte count, CD8+ and CD4+ T-lymphocytes, systolic blood pressure and microvascular reactivity. In response to treatment of severe sepsis, the median KT ratio decreased from 162 [IQR 100–286] on day 0 to 89 [65–139] by day 7; p = 0.0006) and this decrease in KT ratio correlated with a decrease in the Sequential Organ Failure Assessment score (p<0.0001). IDO-mediated tryptophan catabolism is associated with dysregulated immune responses and impaired microvascular reactivity in sepsis and may link these two fundamental processes in sepsis pathophysiology.
Collapse
Affiliation(s)
- Christabelle J Darcy
- Global Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Northern Territory, Australia
| | | | | | | | | | | | | |
Collapse
|
23
|
Fan XY, van den Berg A, Snoek M, van der Flier LG, Smids B, Jansen HM, Liu RY, Lutter R. Arginine deficiency augments inflammatory mediator production by airway epithelial cells in vitro. Respir Res 2009; 10:62. [PMID: 19575800 PMCID: PMC2714041 DOI: 10.1186/1465-9921-10-62] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Accepted: 07/03/2009] [Indexed: 11/25/2022] Open
Abstract
Background Previously we showed that reduced availability of the essential amino acid tryptophan per se attenuates post-transcriptional control of interleukin (IL)-6 and IL-8 leading to hyperresponsive production of these inflammatory mediators by airway epithelial cells. Availability of the non-essential amino acid arginine in the inflamed airway mucosa of patients with asthma is reduced markedly, but it is not known whether this can also lead to an exaggerated production of IL-6 and IL-8. Methods IL-6 and IL-8 were determined by ELISA in culture supernatants of NCI-H292 airway epithelial-like cells and normal bronchial epithelial (NHBE) cells that were exposed to TNF-α, LPS or no stimulus, in medium with or without arginine. Arginine deficiency may also result from exposure to poly-L-arginine or major basic protein (MBP), which can block arginine uptake. Epithelial cells were exposed to these polycationic proteins and L-14C-arginine uptake was assessed as well as IL-6 and IL-8 production. To determine the mode of action, IL-6 and IL-8 mRNA profiles over time were assessed as were gene transcription and post-transcriptional mRNA degradation. Results For both NCI-H292 and NHBE cells, low arginine concentrations enhanced basal epithelial IL-6 and IL-8 production and synergized with TNF-α-induced IL-6 and IL-8 production. Poly-L-arginine enhanced the stimulus-induced IL-6 and IL-8 production, however, blocking arginine uptake and the enhanced IL-6 and IL-8 production appeared unrelated. The exaggerated IL-6 and IL-8 production due to arginine deficiency and to poly-L-arginine depend on a post-transcriptional and a transcriptional process, respectively. Conclusion We conclude that both reduced arginine availability per se and the presence of polycationic proteins may promote airway inflammation by enhanced pro-inflammatory mediator production in airway epithelial cells, but due to distinct mechanisms.
Collapse
Affiliation(s)
- Xiao-Yun Fan
- Department of Pulmonology, The Geriatric Institute of Anhui, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Nisapakultorn K, Makrudthong J, Sa-Ard-Iam N, Rerkyen P, Mahanonda R, Takikawa O. Indoleamine 2,3-dioxygenase expression and regulation in chronic periodontitis. J Periodontol 2009; 80:114-21. [PMID: 19228097 DOI: 10.1902/jop.2009.080315] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Indoleamine 2,3-dioxygenase (IDO) is an intracellular tryptophan-oxidizing enzyme with immunosuppressive characteristics. Its expression and regulation in periodontal tissues are unknown. The aim of this study was to determine IDO expression in healthy gingiva and chronic periodontitis lesions. In addition, the effect of inflammatory cytokines and bacterial products on the expression and activity of DOI in human gingival fibroblasts (HGFs) was assessed. METHODS Human gingival tissue samples were obtained from patients who underwent periodontal surgery. IDO expression in healthy gingiva and periodontitis lesions was determined by immunohistochemistry. HGF cells were treated with interferon-gamma (IFN-gamma), interleukin (IL)-1beta, tumor necrosis factor-alpha (TNF-alpha), and lipopolysaccharides from Porphyromonas gingivalis (PgLPS). IDO mRNA expression was determined by reverse transcription-polymerase chain reaction. The IDO enzymatic activity was determined by measuring the kynurenine level using a colorimetric method. RESULTS In gingival tissues, IDO expression was detected in epithelial cells, fibroblasts, endothelial cells, and inflammatory mononuclear cells. IDO expression was higher in periodontitis lesions than in healthy gingiva. HGFs did not constitutively express IDO. IFN-gamma strongly induced IDO expression and activity in HGFs, in a dose-dependent manner. IL-1beta, TNF-alpha, and PgLPS were also able to induce IDO expression in HGF cells. IFN-gamma in combination with IL-1beta, TNF-alpha, or PgLPS showed enhanced IDO expression. CONCLUSIONS IDO was expressed in human gingiva, and the expression was upregulated in chronic periodontitis. The increased IDO expression in periodontitis lesions may be due, in part, to the activation of HGFs by inflammatory cytokines and bacterial products.
Collapse
|
25
|
Immunosuppression routed via the kynurenine pathway: a biochemical and pathophysiologic approach. Adv Clin Chem 2008; 45:155-97. [PMID: 18429497 DOI: 10.1016/s0065-2423(07)00007-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the past years, it has been shown that kynurenines pathway is a regulator of both the innate and the adaptive immune responses. Particularly, the initial enzyme of this pathway, indoleamine 2,3-dioxygenase (IDO), is implicated in maintaining tolerance during pregnancy, and also can be expressed in tumors to avoid the immune attack. In this chapter, we will describe how the kynurenine pathway affects the immune system with important implications both in physiology and in pathology. The incorrect activation or blockade suppressive properties of the kynurenine pathway are also implicated in a number of other diseases such as AIDS or autoimmune diseases.
Collapse
|
26
|
Keslacy S, Tliba O, Baidouri H, Amrani Y. Inhibition of tumor necrosis factor-alpha-inducible inflammatory genes by interferon-gamma is associated with altered nuclear factor-kappaB transactivation and enhanced histone deacetylase activity. Mol Pharmacol 2007; 71:609-18. [PMID: 17108260 DOI: 10.1124/mol.106.030171] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Airway smooth muscle (ASM) cells can act as effector cells in the initiation and/or perpetuation of airway inflammation in asthma by producing various inflammatory chemokines or cytokines. Previous studies from our laboratory and others showed that the combination of tumor necrosis factor-alpha (TNFalpha) and interferon-gamma (IFNgamma) or endogenous IFNbeta results in a synergistic induction of various pro-inflammatory genes, including CD38 and regulated upon activation normal T-cell expressed and secreted (RANTES), in ASM cells. In contrast to these studies, we found that IFNgamma (1000 U/ml) markedly inhibited TNFalpha-induced expression of interleukin (IL)-6, IL-8, and eotaxin by 66.29+/-3.33, 43.86+/-7.11, and 63.25+/-6.46%, respectively. These genes were also found to be NF-kappaB-dependent in that TNFalpha-induced expression of IL-6, IL-8, and eotaxin was dose-dependently inhibited by the selective IKKbeta inhibitor 4-(2'-aminoethyl)amino-1,8-dimethylimidazo[1,2-a]quinoxaline (BMS-345541) (1-30 microM). Using a luciferase reporter construct containing kappaB sites, we found that IFNgamma (10-1000 U/ml) inhibits NF-kappaB-dependent gene transcription in a dose-dependent manner. Moreover, IFNgamma failed to affect TNFalpha-induced IkappaKbeta phosphorylation or IkappaB degradation as well as nuclear NF-kappaB/DNA interaction. It is noteworthy that IFNgamma decreases TNFalpha-induced histone acetyl transferase (HAT) and increases histone deacetylase (HDAC) activities. Finally, trichostatin A, an HDAC inhibitor, prevents IFNgamma inhibitory action on TNFalpha-induced gene expression. Together, our data indicate that IFNgamma is a potent inhibitor of specific TNFalpha-inducible inflammatory genes by acting on NF-kappaB transactivation via the modulation of HDAC function.
Collapse
Affiliation(s)
- Stefan Keslacy
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, University of Pennsylvania Medical Center, University of Pennsylvania School of Medicine, Translational Research Laboratories Building, Philadelphia PA 19104-3403, USA
| | | | | | | |
Collapse
|
27
|
van den Berg A, Freitas J, Keles F, Snoek M, van Marle J, Jansen HM, Lutter R. Cytoskeletal architecture differentially controls post-transcriptional processing of IL-6 and IL-8 mRNA in airway epithelial-like cells. Exp Cell Res 2006; 312:1496-506. [PMID: 16499908 DOI: 10.1016/j.yexcr.2006.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2005] [Revised: 01/12/2006] [Accepted: 01/12/2006] [Indexed: 11/18/2022]
Abstract
Airway epithelial cells are critically dependent on an intact cytoskeleton for innate defense functions. There are various pathophysiological conditions that affect the cytoskeletal architecture. We studied the effect of cytoskeletal distortion in polarized airway epithelial-like NCI-H292 cells on inflammatory gene expression, exemplified by interleukin(IL)-6 and IL-8. Disruption of microtubule structure with vinblastin and of actin with cytochalasin D did not affect TNF-alpha-induced IL-6 and IL-8 gene transcription but stabilized IL-8 and IL-6 mRNA. In line with previous studies, IL-8 mRNA stabilization was paralleled by hyperresponsive IL-8 production, but surprisingly, IL-6 production was reduced despite IL-6 mRNA stabilization. Polysome profiling revealed that, in cells with a disrupted cytoskeleton, translational efficiency of IL-6 mRNA was reduced, whereas that of IL-8 mRNA remained unaffected. Our findings indicate that distortion of the cytoskeleton in airway epithelial cells differentially affects both degradation and translation of IL-6 and IL-8 mRNA, modifying inflammatory gene expression and thus their innate defense function.
Collapse
Affiliation(s)
- Arjen van den Berg
- Department of Pulmonology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
28
|
van den Berg A, Snoek M, Jansen HM, Lutter R. E1A expression dysregulates IL-8 production and suppresses IL-6 production by lung epithelial cells. Respir Res 2005; 6:111. [PMID: 16185356 PMCID: PMC1261537 DOI: 10.1186/1465-9921-6-111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2005] [Accepted: 09/26/2005] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND The adenoviral protein E1A has been proposed to play a role in the pathophysiology of COPD, in particular by increasing IL-8 gene transcription of lung epithelial cells in response to cigarette smoke-constituents such as LPS. As IL-8 production is also under tight post-transcriptional control, we planned to study whether E1A affected IL-8 production post-transcriptionally. The production of IL-6 by E1A-positive cells had not been addressed and was studied in parallel. Based on our previous work into the regulation of IL-8 and IL-6 production in airway epithelial cells, we used the lung epithelial-like cell line NCI-H292 to generate stable transfectants expressing either E1A and/or E1B, which is known to frequently co-integrate with E1A. We analyzed IL-8 and IL-6 production and the underlying regulatory processes in response to LPS and TNF-alpha. METHODS Stable transfectants were generated and characterized with immunohistochemistry, western blot and flow cytometry. IL-8 and IL-6 protein production was measured by ELISA. Levels of IL-8 and IL-6 mRNA were measured using specific radiolabeled probes. EMSA was used to assess transcriptional activation of relevant transcription factors. Post-transcriptional regulation of mRNA half-life was measured by Actinomycin D chase experiments. RESULTS Most of the sixteen E1A-expressing transfectants showed suppression of IL-6 production, indicative of biologically active E1A. Significant but no uniform effects on IL-8 production, nor on transcriptional and post-transcriptional regulation of IL-8 production, were observed in the panel of E1A-expressing transfectants. E1B expression exerted similar effects as E1A on IL-8 production. CONCLUSION Our results indicate that integration of adenoviral DNA and expression of E1A and E1B can either increase or decrease IL-8 production. Furthermore, we conclude that expression of E1A suppresses IL-6 production. These findings question the unique role of E1A protein in the pathophysiology of COPD, but do not exclude a role for adenoviral E1A/E1B DNA in modulating inflammatory responses nor in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Arjen van den Berg
- Department of Pulmonology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Mieke Snoek
- Department of Pulmonology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Henk M Jansen
- Department of Pulmonology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - René Lutter
- Department of Pulmonology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
29
|
Djouad F, Bony C, Häupl T, Uzé G, Lahlou N, Louis-Plence P, Apparailly F, Canovas F, Rème T, Sany J, Jorgensen C, Noël D. Transcriptional profiles discriminate bone marrow-derived and synovium-derived mesenchymal stem cells. Arthritis Res Ther 2005; 7:R1304-15. [PMID: 16277684 PMCID: PMC1297577 DOI: 10.1186/ar1827] [Citation(s) in RCA: 155] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2005] [Revised: 07/26/2005] [Accepted: 08/24/2005] [Indexed: 01/01/2023] Open
Abstract
Previous studies have reported that mesenchymal stem cells (MSC) may be isolated from the synovial membrane by the same protocol as that used for synovial fibroblast cultivation, suggesting that MSC correspond to a subset of the adherent cell population, as MSC from the stromal compartment of the bone marrow (BM). The aims of the present study were, first, to better characterize the MSC derived from the synovial membrane and, second, to compare systematically, in parallel, the MSC-containing cell populations isolated from BM and those derived from the synovium, using quantitative assays. Fluorescent-activated cell sorting analysis revealed that both populations were negative for CD14, CD34 and CD45 expression and that both displayed equal levels of CD44, CD73, CD90 and CD105, a phenotype currently known to be characteristic of BM-MSC. Comparable with BM-MSC, such MSC-like cells isolated from the synovial membrane were shown for the first time to suppress the T-cell response in a mixed lymphocyte reaction, and to express the enzyme indoleamine 2,3-dioxygenase activity to the same extent as BM-MSC, which is a possible mediator of this suppressive activity. Using quantitative RT-PCR these data show that MSC-like cells from the synovium and BM may be induced to chondrogenic differentiation and, to a lesser extent, to osteogenic differentiation, but the osteogenic capacities of the synovium-derived MSC were significantly reduced based on the expression of the markers tested (collagen type II and aggrecan or alkaline phosphatase and osteocalcin, respectively). Transcription profiles, determined with the Atlas Human Cytokine/Receptor Array, revealed discrimination between the MSC-like cells from the synovial membrane and the BM-MSC by 46 of 268 genes. In particular, activin A was shown to be one major upregulated factor, highly secreted by BM-MSC. Whether this reflects a different cellular phenotype, a different amount of MSC in the synovium-derived population compared with BM-MSC adherent cell populations or the impact of a different microenvironment remains to be determined. In conclusion, although the BM-derived and synovium-derived MSC shared similar phenotypic and functional properties, both their differentiation capacities and transcriptional profiles permit one to discriminate the cell populations according to their tissue origin.
Collapse
Affiliation(s)
| | | | | | | | - Najiba Lahlou
- Hormonal Biology Laboratory, St Vincent de Paul Hospital, Paris, France
| | | | | | | | | | - Jacques Sany
- Immuno-Rhumatologie, Lapeyronie Hospital, Montpellier, France
| | | | | |
Collapse
|
30
|
Pawliczak R, Logun C, Madara P, Barb J, Suffredini AF, Munson PJ, Danner RL, Shelhamer JH. Influence of IFN-gamma on gene expression in normal human bronchial epithelial cells: modulation of IFN-gamma effects by dexamethasone. Physiol Genomics 2005; 23:28-45. [PMID: 15985639 DOI: 10.1152/physiolgenomics.00011.2005] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Interferon gamma (IFN-gamma) plays a role in a variety of lung inflammatory responses, and corticosteroids are frequently employed as a treatment in these conditions. Therefore, the effect of IFN-gamma, of the corticosteroid dexamethasone (Dex), or of both on gene expression was studied in normal human bronchial epithelial (NHBE) cells. NHBE cells were exposed to medium alone, IFN-gamma (300 U/ml), Dex (10(-7) M), or both IFN-gamma and Dex for 8 or 24 h. Gene expression was examined using oligonucleotide microarrays. A principal components analysis demonstrated that the IFN-gamma treatment effect was the primary source of differences in the data. With a 5% false discovery rate, of the 66 genes upregulated by IFN-gamma by twofold or greater at 8 h and 287 genes upregulated at 24 h, coincubation with Dex inhibited the expression of 2 genes at 8 h and 45 genes at 24 h. Prominent among these were cytokines and secreted proteins. Dex cotreatment increased expression of 65 of the 376 genes that were inhibited by IFN-gamma by 50% at 24 h. The majority of these genes encode cell cycle or nuclear proteins. Dex alone increased the expression of only 22 genes and inhibited the expression of 7 genes compared with controls at 24 h. The effect of Dex on IFN-gamma-induced changes suggests a specific, targeted effect on IFN-gamma responses that is substantially greater than the effect of Dex alone. Dex had little effect on the immediate early response to IFN-gamma but a significant effect on the late responses.
Collapse
Affiliation(s)
- Rafal Pawliczak
- Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Munn DH, Sharma MD, Baban B, Harding HP, Zhang Y, Ron D, Mellor AL. GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. Immunity 2005; 22:633-42. [PMID: 15894280 DOI: 10.1016/j.immuni.2005.03.013] [Citation(s) in RCA: 952] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2004] [Revised: 03/09/2005] [Accepted: 03/23/2005] [Indexed: 01/27/2023]
Abstract
Indoleamine 2,3 dioxygenase (IDO) catabolizes the amino acid tryptophan. IDO-expressing immunoregulatory dendritic cells (DCs) have been implicated in settings including tumors, autoimmunity, and transplant tolerance. However, the downstream molecular mechanisms by which IDO functions to regulate T cell responses remain unknown. We now show that IDO-expressing plasmacytoid DCs activate the GCN2 kinase pathway in responding T cells. GCN2 is a stress-response kinase that is activated by elevations in uncharged tRNA. T cells with a targeted disruption of GCN2 were not susceptible to IDO-mediated suppression of proliferation in vitro. In vivo, proliferation of GCN2-knockout T cells was not inhibited by IDO-expressing DCs from tumor-draining lymph nodes. IDO induced profound anergy in responding wild-type T cells, but GCN2-knockout cells were refractory to IDO-induced anergy. We hypothesize that GCN2 acts as a molecular sensor in T cells, allowing them to detect and respond to conditions created by IDO.
Collapse
Affiliation(s)
- David H Munn
- Department of Pediatrics, Medical College of Georgia, Augusta, GA 30912, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
van den Berg A, Kuiper M, Snoek M, Timens W, Postma DS, Jansen HM, Lutter R. Interleukin-17 induces hyperresponsive interleukin-8 and interleukin-6 production to tumor necrosis factor-alpha in structural lung cells. Am J Respir Cell Mol Biol 2005; 33:97-104. [PMID: 15845864 DOI: 10.1165/rcmb.2005-0022oc] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Lung epithelial cells contribute to local inflammation by the production of pro-inflammatory mediators like interleukin (IL)-8 and IL-6. Although their production depends on gene transcription, previous studies showed that post-transcriptional mechanisms modulate IL-8 and IL-6 production. Human lung epithelial cells turn from normoresponsive into hyperresponsive IL-8- and IL-6-producing cells when their IL-8 and IL-6 mRNA degradation is reduced. We hypothesized that IL-17, a mediator predominantly released by memory T cells and present in airways of individuals with asthma, would modulate rather than induce IL-8 and IL-6 production by both human lung epithelial cells and fibroblasts. We show here for both cell types that IL-17 was a weak stimulus of IL-8 and IL-6 production, but markedly enhanced IL-8 and IL-6 responses to another stimulus, such as tumor necrosis factor-alpha. This modulatory effect of IL-17 was paralleled by a reduced IL-8 and IL-6 mRNA degradation, with no effect on IL-8 and IL-6 gene transcription. In conclusion, IL-17 particularly affects post-transcriptional regulation of IL-8 and IL-6 expression leading to enhanced IL-8 and IL-6 responses to secondary stimuli, and is only a weak proinflammatory stimulus by itself. This poses the interesting concept that by releasing IL-17 from memory T cells, the adaptive immune system instructs lung structural cells as part of the innate immune system to respond more vigorously.
Collapse
Affiliation(s)
- Arjen van den Berg
- Department of Pulmonology and Laboratory of Experimental Immunology, Academic Medical Center, University of Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
33
|
Lavigne MC, Thakker P, Gunn J, Wong A, Miyashiro JS, Wasserman AM, Wei SQ, Pelker JW, Kobayashi M, Eppihimer MJ. Human bronchial epithelial cells express and secrete MMP-12. Biochem Biophys Res Commun 2004; 324:534-46. [PMID: 15474460 DOI: 10.1016/j.bbrc.2004.09.080] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2004] [Indexed: 11/18/2022]
Abstract
Matrix metalloproteinases (MMPs) degrade extracellular matrix proteins, which may be responsible for enlargement of alveoli in chronic obstructive pulmonary disease (COPD) and remodeling of pulmonary tissue associated with chronic asthma. Here, we provide novel evidence that MMP-12 is expressed and secreted by normal human bronchial epithelial cell cultures (NHBECs) and reveal the regulation of MMP-12 gene expression by tumor necrosis factor-alpha (TNF-alpha), epidermal growth factor (EGF), and interferon gamma (IFN-gamma). Reverse transcription-polymerase chain reaction analyses demonstrated MMP-12 mRNA presence in unstimulated differentiated NHBEC cultures. Cultures stimulated independently with EGF or IFN-gamma failed to alter MMP-12 mRNA abundance, while TNF-alpha, TNF-alpha+EGF, or TNF-alpha+IFN-gamma elicited relatively early (6 h) peak increases in MMP-12 mRNA levels. Western blot analyses specifically indicated the presence of MMP-12 in differentiated NHBEC-conditioned media. These findings indicate that the bronchial epithelium may be an important source of elastolytic activity in COPD and tissue remodeling in chronic asthma.
Collapse
Affiliation(s)
- Mark C Lavigne
- Wyeth Research, Cardiovascular and Metabolic Diseases, Cambridge, MA 02140, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Mellor AL, Munn DH. IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nat Rev Immunol 2004; 4:762-74. [PMID: 15459668 DOI: 10.1038/nri1457] [Citation(s) in RCA: 1759] [Impact Index Per Article: 83.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Indoleamine 2,3-dioxygenase (IDO) is an enzyme that degrades the essential amino acid tryptophan. The concept that cells expressing IDO can suppress T-cell responses and promote tolerance is a relatively new paradigm in immunology. Considerable evidence now supports this hypothesis, including studies of mammalian pregnancy, tumour resistance, chronic infections and autoimmune diseases. In this review, we summarize key recent developments and propose a unifying model for the role of IDO in tolerance induction.
Collapse
Affiliation(s)
- Andrew L Mellor
- Program in Molecular Immunology, Institute of Molecular Medicine and Genetics, Department of Medicine, Medical College of Georgia, Augusta, Georgia 30912, USA.
| | | |
Collapse
|
35
|
Roger T, Bresser P, Snoek M, van der Sluijs K, van den Berg A, Nijhuis M, Jansen HM, Lutter R. Exaggerated IL-8 and IL-6 responses to TNF-alpha by parainfluenza virus type 4-infected NCI-H292 cells. Am J Physiol Lung Cell Mol Physiol 2004; 287:L1048-55. [PMID: 15273081 DOI: 10.1152/ajplung.00396.2003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Respiratory viruses induce and potentiate airway inflammation, which is related to the induction of proinflammatory mediators such as interleukin (IL)-8 and IL-6. Here we report on mechanisms implicated in IL-8 and IL-6 production by airway epithelium-like NCI-H292 cells exposed to parainfluenza virus type 4a (PIV-4). PIV-4 readily infected NCI-H292 cells as reflected by intracellular PIV-4 antigen expression. PIV-4 infection triggered a biphasic IL-8 and IL-6 mRNA response. Transient transfection with truncated and mutated promoter constructs identified NF-kappaB and activator protein (AP)-1, and CCAAT-enhancer binding protein (C/EBP) as the relevant transcription factors for PIV-4-induced IL-8 and IL-6 gene transcription, respectively. An increase of DNA-binding activities for NF-kappaB and C/EBP paralleled the induction of the first and second IL-8 and IL-6 mRNA peaks, whereas the onset of AP-1 paralleled the first IL-8 mRNA peak only. The second mRNA peak, apparently dependent on viral replication, coincided also with a marked reduction of IL-8 and IL-6 mRNA degradation. Importantly, cells at the time of the reduced mRNA degradation displayed an exaggerated IL-8 and IL-6 protein production to a secondary stimulus, as exemplified by steeper dose-response curves to TNF-alpha. Thus PIV-4 infection enhances epithelial IL-8 and IL-6 production by transcriptional and posttranscriptional mechanisms. The previously unrecognized phase of reduced IL-8 and IL-6 mRNA degradation and the concurrent amplified epithelial IL-8 and IL-6 responses may play an important role in virus-induced potentiation of airway inflammation.
Collapse
Affiliation(s)
- Thierry Roger
- Department of Pulmonology, Academic Medical Center, University of Amsterdam, 1100 DE Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Zegarra-Moran O, Folli C, Manzari B, Ravazzolo R, Varesio L, Galietta LJV. Double Mechanism for Apical Tryptophan Depletion in Polarized Human Bronchial Epithelium. THE JOURNAL OF IMMUNOLOGY 2004; 173:542-9. [PMID: 15210815 DOI: 10.4049/jimmunol.173.1.542] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Indoleamine 2,3-dioxygenase is an enzyme that catabolizes tryptophan to kynurenine. We investigated the consequences of IDO induction by IFN-gamma in polarized human bronchial epithelium. IDO mRNA expression was undetectable in resting conditions, but strongly induced by IFN-gamma. We determined the concentration of tryptophan and kynurenine in the extracellular medium, and we found that apical tryptophan concentration was lower than the basolateral in resting cells. IFN-gamma caused a decrease in tryptophan concentration on both sides of the epithelium. Kynurenine was absent in control conditions, but increased in the basolateral medium after IFN-gamma treatment. The asymmetric distribution of tryptophan and kynurenine suggested the presence of a transepithelial amino acid transport. Uptake experiments with radiolabeled amino acids demonstrated the presence of a Na(+)-dependent amino acid transporter with broad specificity that was responsible for the tryptophan/kynurenine transport. We confirmed these data by measuring the short-circuit currents elicited by direct application of tryptophan or kynurenine to the apical surface. The rate of amino acid transport was dependent on the transepithelial potential, and we established that in cystic fibrosis epithelia, in which the transepithelial potential is significantly more negative than in noncystic fibrosis epithelia, amino acid uptake was reduced. This work suggests that human airway epithelial cells maintain low apical tryptophan concentrations by two mechanisms, a removal through a Na(+)-dependent amino acid transporter and an IFN-gamma-inducible degradation by IDO.
Collapse
Affiliation(s)
- Olga Zegarra-Moran
- Laboratorio di Genetica Molecolare, Istituto Giannina Gaslini, Largo G. Gaslini 5, Genoa 16148, Italy.
| | | | | | | | | | | |
Collapse
|
37
|
Adams O, Besken K, Oberdörfer C, MacKenzie CR, Takikawa O, Däubener W. Role of indoleamine-2,3-dioxygenase in alpha/beta and gamma interferon-mediated antiviral effects against herpes simplex virus infections. J Virol 2004; 78:2632-6. [PMID: 14963171 PMCID: PMC369218 DOI: 10.1128/jvi.78.5.2632-2636.2004] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Gamma interferon (IFN-gamma)-mediated indoleamine-2,3-dioxygenase (IDO) activity in human astrocytoma cells and in native astrocytes was found to be responsible for the inhibition of herpes simplex virus replication. The effect is abolished in the presence of excess amounts of L-tryptophan. Both IFN-alpha and IFN-beta restricted herpes simplex virus replication in both cell types, but (in contrast to the results seen with IFN-gamma) the addition of an excess amount of L-tryptophan did not inhibit the induced antiviral effect.
Collapse
Affiliation(s)
- O Adams
- Institut für Virologie. Institut für Medizinische Mikrobiologie, Heinrich-Heine-Universität, D-40225 Dusseldorf, Germany. Department of Cell Pharmacology, Hokkaido University, Sapporo 060-8638, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Lutter R, van Wissen M, Roger T, Bresser P, van der Sluijs K, Nijhuis M, Jansen HM. Mechanisms that potentially underlie virus-induced exaggerated inflammatory responses by airway epithelial cells. Chest 2003; 123:391S-2S. [PMID: 12628999 DOI: 10.1378/chest.123.3_suppl.391s] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Affiliation(s)
- René Lutter
- Departments of Pulmonology, Academic Medical Center, University of Amsterdam, the Netherlands.
| | | | | | | | | | | | | |
Collapse
|