51
|
Yu E, Papandreou C, Ruiz-Canela M, Guasch-Ferre M, Clish CB, Dennis C, Liang L, Corella D, Fitó M, Razquin C, Lapetra J, Estruch R, Ros E, Cofán M, Arós F, Toledo E, Serra-Majem L, Sorlí JV, Hu FB, Martinez-Gonzalez MA, Salas-Salvado J. Association of Tryptophan Metabolites with Incident Type 2 Diabetes in the PREDIMED Trial: A Case-Cohort Study. Clin Chem 2018; 64:1211-1220. [PMID: 29884676 PMCID: PMC6218929 DOI: 10.1373/clinchem.2018.288720] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/14/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Metabolites of the tryptophan-kynurenine pathway (i.e., tryptophan, kynurenine, kynurenic acid, quinolinic acid, 3-hydroxyanthranilic) may be associated with diabetes development. Using a case-cohort design nested in the Prevención con Dieta Mediterránea (PREDIMED) study, we studied the associations of baseline and 1-year changes of these metabolites with incident type 2 diabetes (T2D). METHODS Plasma metabolite concentrations were quantified via LC-MS for n = 641 in a randomly selected subcohort and 251 incident cases diagnosed during 3.8 years of median follow-up. Weighted Cox models adjusted for age, sex, body mass index, and other T2D risk factors were used. RESULTS Baseline tryptophan was associated with higher risk of incident T2D (hazard ratio = 1.29; 95% CI, 1.04-1.61 per SD). Positive changes in quinolinic acid from baseline to 1 year were associated with a higher risk of T2D (hazard ratio = 1.39; 95% CI, 1.09-1.77 per SD). Baseline tryptophan and kynurenic acid were directly associated with changes in homeostatic model assessment for insulin resistance (HOMA-IR) from baseline to 1 year. Concurrent changes in kynurenine, quinolinic acid, 3-hydroxyanthranilic acid, and kynurenine/tryptophan ratio were associated with baseline-to-1-year changes in HOMA-IR. CONCLUSIONS Baseline tryptophan and 1-year increases in quinolinic acid were positively associated with incident T2D. Baseline and 1-year changes in tryptophan metabolites predicted changes in HOMA-IR. Tryptophan levels may initially increase and then deplete as diabetes progresses in severity.
Collapse
Affiliation(s)
- Edward Yu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Christopher Papandreou
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institut d'Investigació Sanitària Pere Virgili, Rovira i Virgili University, Reus, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Miguel Ruiz-Canela
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitària de Navarra), Navarra, Spain
| | - Marta Guasch-Ferre
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institut d'Investigació Sanitària Pere Virgili, Rovira i Virgili University, Reus, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Clary B Clish
- Broad Institute of MIT and Harvard University, Cambridge, MA
| | - Courtney Dennis
- Broad Institute of MIT and Harvard University, Cambridge, MA
| | - Liming Liang
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Dolores Corella
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Montserrat Fitó
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Cardiovascular and Nutrition Research Group, Institut de Recerca Hospital del Mar, Barcelona, Spain
| | - Cristina Razquin
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitària de Navarra), Navarra, Spain
| | - José Lapetra
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Family Medicine, Unit Research, Distrito Sanitario Atención Primaria Sevilla, Sevilla, Spain
| | - Ramón Estruch
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Internal Medicine Institut d'Investigacions Biomèdiques August Pi Sunyer (IDI- BAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Emilio Ros
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Lipid Clinic, Department of Endocrinology and Nutrition (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Montserrat Cofán
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Lipid Clinic, Department of Endocrinology and Nutrition (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Fernando Arós
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Cardiology, University Hospital of Álava, Vitoria, Spain
| | - Estefania Toledo
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitària de Navarra), Navarra, Spain
| | - Lluis Serra-Majem
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Research Institute of Biomedical and Health Sciences, University of Las Palmas de Gran Canaria, Las Palmas, Spain
| | - José V Sorlí
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division for Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, MA
| | - Miguel A Martinez-Gonzalez
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitària de Navarra), Navarra, Spain
| | - Jordi Salas-Salvado
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institut d'Investigació Sanitària Pere Virgili, Rovira i Virgili University, Reus, Spain;
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
52
|
Reininghaus B, Riedrich K, Dalkner N, Bengesser SA, Birner A, Platzer M, Hamm C, Gostner JM, Fuchs D, Reininghaus EZ. Changes in the tryptophan-kynurenine axis in association to therapeutic response in clinically depressed patients undergoing psychiatric rehabilitation. Psychoneuroendocrinology 2018; 94:25-30. [PMID: 29753175 DOI: 10.1016/j.psyneuen.2018.04.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 11/30/2022]
Abstract
INTRODUCTION In recent decades a number of studies have shown an association between the Tryptophan (Trp)-Kynurenine (Kyn) axis and neuropsychiatric disorders. However, the role of the Trp-Kyn pathway on the affective status in a general psychiatric cohort requires clarification. This study aimed to measure peripheral changes in Trp, Kyn and the Kyn/Trp-ratio as well as in the inflammatory markers high sensitive C-reactive protein (hsCRP) and interleukine-6 (IL-6) in individuals undergoing a six-week course of intensive treatment program comparing subgroups of treatment responders and non-responders. METHODS In this investigation 87 currently depressed individuals with a life-time history of depressive disorders were divided into treatment responders (n = 48) and non-responders (n = 39). The individuals were selected for an extreme group comparison out of 598 patients undergoing a 6-week psychiatric rehabilitation program in Austria. Responders were defined according to great changes in Becks Depression Inventory (BDI-II) between time of admission and discharge (BDI-II > 29 to BDI-II <14), while non-responders had no or minimal changes (BDI >20, max. 4 points change over time). Differences in the levels of Trp, Kyn, and the Kyn/Trp ratio as well as levels of hsCRP and IL-6, were compared between groups. Differences were analyzed at the time of admission as well as at discharge. RESULTS A significant group x time interaction was found for Kyn [F(1.82) = 5.79; p = 0.018] and the Kyn/Trp ratio [F(1.85) = 4.01, p = 0.048]. Importantly, Kyn increased significantly in the non-responder group, while the Kyn/Trp ratio decreased significantly in the responder group over time. Furthermore, changes in Kyn as well as hsCRP levels correlated significantly with changes in the body mass index over time (Kyn: r=0.24, p = 0.030; hsCRP: r=0.25, p = 0.021). No significant interactions were found for Trp and hsCRP, although they increased significantly over time. DISCUSSION Given the limitations of the study, we could show that the therapeutic response to a multimodal treatment in clinically depressed patients not receiving cytokine treatment is associated with changes in Kyn levels and the Kyn/Trp ratio as well as with hsCRP. However, it is too early to draw any causal conclusion. Future research should clarify relevant clinical and neurobiological parameters associated with changes in Kyn levels and Kyn/Trp ratio, especially in regard to clinical response.
Collapse
Affiliation(s)
- B Reininghaus
- Therapie Zentrum-Justuspark Bad Hall, Austria; Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Auenbruggerplatz 31, 8036, Graz, Austria
| | - K Riedrich
- Therapie Zentrum-Justuspark Bad Hall, Austria; Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Auenbruggerplatz 31, 8036, Graz, Austria
| | - N Dalkner
- Therapie Zentrum-Justuspark Bad Hall, Austria; Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Auenbruggerplatz 31, 8036, Graz, Austria.
| | - S A Bengesser
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Auenbruggerplatz 31, 8036, Graz, Austria
| | - A Birner
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Auenbruggerplatz 31, 8036, Graz, Austria
| | - M Platzer
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Auenbruggerplatz 31, 8036, Graz, Austria
| | - C Hamm
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Auenbruggerplatz 31, 8036, Graz, Austria
| | - J M Gostner
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - D Fuchs
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria; Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - E Z Reininghaus
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Auenbruggerplatz 31, 8036, Graz, Austria
| |
Collapse
|
53
|
Michels N, Clarke G, Olavarria-Ramirez L, Gómez-Martínez S, Díaz LE, Marcos A, Widhalm K, Carvalho LA. Psychosocial stress and inflammation driving tryptophan breakdown in children and adolescents: A cross-sectional analysis of two cohorts. Psychoneuroendocrinology 2018; 94:104-111. [PMID: 29775873 DOI: 10.1016/j.psyneuen.2018.05.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/23/2018] [Accepted: 05/10/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Tryptophan breakdown is an important mechanism in several diseases e.g. inflammation and stress-induced inflammation have been associated with the development of depression via enhanced tryptophan breakdown. Depression is a major public health problem which commonly starts during adolescence, thus identifying underlying mechanisms during early life is crucial in prevention. The aim of this work was to verify whether independent and interacting associations of psychosocial stress and inflammation on tryptophan breakdown already exist in children and adolescents as a vulnerable age group. METHODS Two cross-sectional population-based samples of children/adolescents (8-18 y) were available: 315 from the European HELENA study and 164 from the Belgian ChiBS study. In fasting serum samples, tryptophan, kynurenine, kynurenic acid, C-reactive protein (CRP), interleukin (IL)-6, tumor necrosis factor (TNF)-α, interferon (IFN)-ɣ, soluble vascular adhesion molecule 1 (sVCAM1) and soluble intercellular adhesion molecule 1 (sICAM1) were measured. Psychological stress was measured by stress reports (subjective) and cortisol (objective - awakening salivary cortisol or hair cortisol). Linear regressions with stress or inflammation as predictor were adjusted for age, sex, body mass index, puberty, socio-economic status and country. RESULTS In both cohorts, inflammation as measured by higher levels of CRP, sVCAM1 and sICAM1 was associated with kynurenine/tryptophan ratio and thus enhanced tryptophan breakdown (beta: 0.145-0.429). Psychological stress was only associated with tryptophan breakdown in the presence of higher inflammatory levels (TNF-α in both populations). CONCLUSIONS Inflammatory levels were replicable key in enhancing tryptophan breakdown along the kynurenine pathway, even at young age and in a non-clinical sample. The stress-inflammation interaction indicated that only the stress exposures inducing higher inflammatory levels (or in an already existing inflammatory status) were associated with more tryptophan breakdown. This data further contributes to our understanding of pathways to disease development, and may help identifying those more likely to develop stress or inflammation-related illnesses.
Collapse
Affiliation(s)
- Nathalie Michels
- Department of Public Health, Faculty of Medicine and Health Sciences, Ghent University, Belgium.
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Science and APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Loreto Olavarria-Ramirez
- Department of Psychiatry and Neurobehavioural Science and APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Sonia Gómez-Martínez
- Immunonutrition Research Group, Department of Metabolism & Nutrition, Institute of Food Science, Technology and Nutrition, Madrid, Spain; ImFINE Research Group, Department of Health and Human Performance, Universidad Politécnica de Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Nutrición y la Obesidad (CIBEROBN), Spain
| | - Ligia Esperanza Díaz
- Immunonutrition Research Group, Department of Metabolism & Nutrition, Institute of Food Science, Technology and Nutrition, Madrid, Spain; ImFINE Research Group, Department of Health and Human Performance, Universidad Politécnica de Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Nutrición y la Obesidad (CIBEROBN), Spain
| | - Ascensión Marcos
- Immunonutrition Research Group, Department of Metabolism & Nutrition, Institute of Food Science, Technology and Nutrition, Madrid, Spain; ImFINE Research Group, Department of Health and Human Performance, Universidad Politécnica de Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Nutrición y la Obesidad (CIBEROBN), Spain
| | - Kurt Widhalm
- Department of Pediatric, Division of Clinical Nutrition, Medical University of Vienna, Vienna, Austria
| | - Livia A Carvalho
- Department of Clinical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, Charterhouse Square, EC1 M 6BQ, UK
| |
Collapse
|
54
|
Laurans L, Venteclef N, Haddad Y, Chajadine M, Alzaid F, Metghalchi S, Sovran B, Denis RGP, Dairou J, Cardellini M, Moreno-Navarrete JM, Straub M, Jegou S, McQuitty C, Viel T, Esposito B, Tavitian B, Callebert J, Luquet SH, Federici M, Fernandez-Real JM, Burcelin R, Launay JM, Tedgui A, Mallat Z, Sokol H, Taleb S. Genetic deficiency of indoleamine 2,3-dioxygenase promotes gut microbiota-mediated metabolic health. Nat Med 2018; 24:1113-1120. [PMID: 29942089 DOI: 10.1038/s41591-018-0060-4] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 04/23/2018] [Indexed: 02/07/2023]
Abstract
The association between altered gut microbiota, intestinal permeability, inflammation and cardiometabolic diseases is becoming increasingly clear but remains poorly understood1,2. Indoleamine 2,3-dioxygenase is an enzyme induced in many types of immune cells, including macrophages in response to inflammatory stimuli, and catalyzes the degradation of tryptophan along the kynurenine pathway. Indoleamine 2,3-dioxygenase activity is better known for its suppression of effector T cell immunity and its activation of regulatory T cells3,4. However, high indoleamine 2,3-dioxygenase activity predicts worse cardiovascular outcome5-9 and may promote atherosclerosis and vascular inflammation6, suggesting a more complex role in chronic inflammatory settings. Indoleamine 2,3-dioxygenase activity is also increased in obesity10-13, yet its role in metabolic disease is still unexplored. Here, we show that obesity is associated with an increase of intestinal indoleamine 2,3-dioxygenase activity, which shifts tryptophan metabolism from indole derivative and interleukin-22 production toward kynurenine production. Indoleamine 2,3-dioxygenase deletion or inhibition improves insulin sensitivity, preserves the gut mucosal barrier, decreases endotoxemia and chronic inflammation, and regulates lipid metabolism in liver and adipose tissues. These beneficial effects are due to rewiring of tryptophan metabolism toward a microbiota-dependent production of interleukin-22 and are abrogated after treatment with a neutralizing anti-interleukin-22 antibody. In summary, we identify an unexpected function of indoleamine 2,3-dioxygenase in the fine tuning of intestinal tryptophan metabolism with major consequences on microbiota-dependent control of metabolic disease, which suggests indoleamine 2,3-dioxygenase as a potential therapeutic target.
Collapse
Affiliation(s)
- Ludivine Laurans
- Institut National de la Santé et de la Recherche Médicale, U970, Paris Cardiovascular Research Center, and Université Paris-Descartes, Paris, France
| | - Nicolas Venteclef
- Institut National de la Santé et de la Recherche Médicale, UMRS 1138, Sorbonne Universités, UPMC Université Paris 06, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, and Centre de Recherche des Cordeliers, Paris, France
| | - Yacine Haddad
- Institut National de la Santé et de la Recherche Médicale, U970, Paris Cardiovascular Research Center, and Université Paris-Descartes, Paris, France
| | - Mouna Chajadine
- Institut National de la Santé et de la Recherche Médicale, U970, Paris Cardiovascular Research Center, and Université Paris-Descartes, Paris, France
| | - Fawaz Alzaid
- Institut National de la Santé et de la Recherche Médicale, UMRS 1138, Sorbonne Universités, UPMC Université Paris 06, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, and Centre de Recherche des Cordeliers, Paris, France
| | - Sarvenaz Metghalchi
- Institut National de la Santé et de la Recherche Médicale, U970, Paris Cardiovascular Research Center, and Université Paris-Descartes, Paris, France
| | - Bruno Sovran
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Raphael G P Denis
- Unité de Biologie Fonctionnelle et Adaptative, Centre National la Recherche Scientifique, UMR 8251, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Julien Dairou
- UMR 8601 CNRS, Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Marina Cardellini
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Jose-Maria Moreno-Navarrete
- Department of Diabetes and Endocrinology, Hospital de Girona "Dr Josep Trueta", Girona, Spain
- CIBERobn Pathophysiology of Obesity and Nutrition, Instituto de Salud Carlos III, Madrid, Spain
| | - Marjolene Straub
- Sorbonne Université, École normale supérieure, PSL Research University, CNRS, INSERM, AP-HP, Laboratoire de biomolécules, Hôpital Saint-Antoine, Paris, France
| | - Sarah Jegou
- Sorbonne Université, École normale supérieure, PSL Research University, CNRS, INSERM, AP-HP, Laboratoire de biomolécules, Hôpital Saint-Antoine, Paris, France
| | - Claire McQuitty
- Sorbonne Université, École normale supérieure, PSL Research University, CNRS, INSERM, AP-HP, Laboratoire de biomolécules, Hôpital Saint-Antoine, Paris, France
| | - Thomas Viel
- Institut National de la Santé et de la Recherche Médicale, U970, Paris Cardiovascular Research Center, and Université Paris-Descartes, Paris, France
| | - Bruno Esposito
- Institut National de la Santé et de la Recherche Médicale, U970, Paris Cardiovascular Research Center, and Université Paris-Descartes, Paris, France
| | - Bertrand Tavitian
- Institut National de la Santé et de la Recherche Médicale, U970, Paris Cardiovascular Research Center, and Université Paris-Descartes, Paris, France
| | - Jacques Callebert
- Service de Biochimie, Assistance Publique Hôpitaux de Paris, and INSERM UMR942, Hôpital Lariboisière, Paris, France
| | - Serge H Luquet
- Unité de Biologie Fonctionnelle et Adaptative, Centre National la Recherche Scientifique, UMR 8251, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Remy Burcelin
- Institut des maladies métaboliques et cardiovasculaires, INSERM UMR1048, Toulouse, France
| | - Jean-Marie Launay
- Service de Biochimie, Assistance Publique Hôpitaux de Paris, and INSERM UMR942, Hôpital Lariboisière, Paris, France
| | - Alain Tedgui
- Institut National de la Santé et de la Recherche Médicale, U970, Paris Cardiovascular Research Center, and Université Paris-Descartes, Paris, France
| | - Ziad Mallat
- Institut National de la Santé et de la Recherche Médicale, U970, Paris Cardiovascular Research Center, and Université Paris-Descartes, Paris, France
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Harry Sokol
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
- Sorbonne Université, École normale supérieure, PSL Research University, CNRS, INSERM, AP-HP, Laboratoire de biomolécules, Hôpital Saint-Antoine, Paris, France
- Department of Gastroenterology, Saint Antoine Hospital, Assistance Publique - Hopitaux de Paris, Sorbonne Université, Paris, France
| | - Soraya Taleb
- Institut National de la Santé et de la Recherche Médicale, U970, Paris Cardiovascular Research Center, and Université Paris-Descartes, Paris, France.
| |
Collapse
|
55
|
Groer M, Fuchs D, Duffy A, Louis-Jacques A, D’Agata A, Postolache TT. Associations Among Obesity, Inflammation, and Tryptophan Catabolism in Pregnancy. Biol Res Nurs 2018; 20:284-291. [PMID: 29141444 PMCID: PMC6346309 DOI: 10.1177/1099800417738363] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To evaluate relationships among obesity in pregnancy and plasma levels of tryptophan (TRP) and kynurenine (KYN), inflammatory markers, and depressed mood. METHODS Pregnant women ( N = 374) were enrolled, and data were collected at a mean gestation of 20 weeks in this cross-sectional study. Plasma was analyzed for TRP, KYN, neopterin, and nitrite levels. Women completed demographic and mood scales. RESULTS There was a statistically significant inverse correlation between body mass index (BMI) and TRP and positive correlations between BMI and KYN and the kynurenine/tryptophan (KYN/TRP) ratio. Neopterin was correlated with KYN/TRP, suggesting that the indoleamine 2,3-dioxygenase-1 (IDO-1) enzyme was activated. The correlations of neopterin and nitrite with BMI were too small to be clinically meaningful but may provide mechanistic insight. There was a correlation between depressed mood and nitrite levels. Depressed mood was also associated with lower TRP levels. When the sample was divided into pregnant women with or without obesity, TRP was significantly lower and the KYN/TRP ratio was significantly higher in the women with obesity. CONCLUSION The pro-inflammatory state of obesity in pregnancy may drive activation of IDO-1, resulting in diversion of TRP away from serotonin and melatonin production and toward KYN metabolites. This alteration could contribute to depression, impaired sleep, increased production of excitotoxic neurotransmitters, and reinforcement of a pro-inflammatory state in pregnancy.
Collapse
Affiliation(s)
- Maureen Groer
- Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- University of South Florida College of Nursing, Tampa, FL, USA
| | - Dietmar Fuchs
- Division of Biological Chemistry, Innsbruck Medical University, Innsbruck, Austria
| | - Allyson Duffy
- University of South Florida College of Nursing, Tampa, FL, USA
| | - Adetola Louis-Jacques
- Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- University of South Florida College of Nursing, Tampa, FL, USA
| | - Amy D’Agata
- University of South Florida College of Nursing, Tampa, FL, USA
| | | |
Collapse
|
56
|
Cervenka I, Agudelo LZ, Ruas JL. Kynurenines: Tryptophan's metabolites in exercise, inflammation, and mental health. Science 2018; 357:357/6349/eaaf9794. [PMID: 28751584 DOI: 10.1126/science.aaf9794] [Citation(s) in RCA: 771] [Impact Index Per Article: 128.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Kynurenine metabolites are generated by tryptophan catabolism and regulate biological processes that include host-microbiome signaling, immune cell response, and neuronal excitability. Enzymes of the kynurenine pathway are expressed in different tissues and cell types throughout the body and are regulated by cues, including nutritional and inflammatory signals. As a consequence of this systemic metabolic integration, peripheral inflammation can contribute to accumulation of kynurenine in the brain, which has been associated with depression and schizophrenia. Conversely, kynurenine accumulation can be suppressed by activating kynurenine clearance in exercised skeletal muscle. The effect of exercise training on depression through modulation of the kynurenine pathway highlights an important mechanism of interorgan cross-talk mediated by these metabolites. Here, we discuss peripheral mechanisms of tryptophan-kynurenine metabolism and their effects on inflammatory, metabolic, oncologic, and psychiatric disorders.
Collapse
Affiliation(s)
- Igor Cervenka
- Department of Physiology and Pharmacology, Molecular and Cellular Exercise Physiology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Leandro Z Agudelo
- Department of Physiology and Pharmacology, Molecular and Cellular Exercise Physiology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Jorge L Ruas
- Department of Physiology and Pharmacology, Molecular and Cellular Exercise Physiology, Karolinska Institutet, SE-17177 Stockholm, Sweden.
| |
Collapse
|
57
|
Chaves Filho AJM, Lima CNC, Vasconcelos SMM, de Lucena DF, Maes M, Macedo D. IDO chronic immune activation and tryptophan metabolic pathway: A potential pathophysiological link between depression and obesity. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:234-249. [PMID: 28595944 DOI: 10.1016/j.pnpbp.2017.04.035] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 04/03/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022]
Abstract
Obesity and depression are among the most pressing health problems in the contemporary world. Obesity and depression share a bidirectional relationship, whereby each condition increases the risk of the other. By inference, shared pathways may underpin the comorbidity between obesity and depression. Activation of cell-mediated immunity (CMI) is a key factor in the pathophysiology of depression. CMI cytokines, including IFN-γ, TNFα and IL-1β, induce the catabolism of tryptophan (TRY) by stimulating indoleamine 2,3-dioxygenase (IDO) resulting in the synthesis of kynurenine (KYN) and other tryptophan catabolites (TRYCATs). In the CNS, TRYCATs have been related to oxidative damage, inflammation, mitochondrial dysfunction, cytotoxicity, excitotoxicity, neurotoxicity and lowered neuroplasticity. The pathophysiology of obesity is also associated with a state of aberrant inflammation that activates aryl hydrocarbon receptor (AHR), a pathway involved in the detection of intracellular or environmental changes as well as with increases in the production of TRYCATs, being KYN an agonists of AHR. Both AHR and TRYCATS are involved in obesity and related metabolic disorders. These changes in the TRYCAT pathway may contribute to the onset of neuropsychiatric symptoms in obesity. This paper reviews the role of immune activation, IDO stimulation and increased TRYCAT production in the pathophysiology of depression and obesity. Here we suggest that increased synthesis of detrimental TRYCATs is implicated in comorbid obesity and depression and is a new drug target to treat both diseases.
Collapse
Affiliation(s)
- Adriano José Maia Chaves Filho
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Camila Nayane Carvalho Lima
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Silvânia Maria Mendes Vasconcelos
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - David Freitas de Lucena
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Michael Maes
- Impact Strategic Research Center, Deakin University, Geelong, Australia; Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Health Sciences Graduate Program, Health Sciences Center, State University of Londrina, Londrina, Brazil
| | - Danielle Macedo
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
58
|
Polyzos KA, Ketelhuth DFJ. The role of the kynurenine pathway of tryptophan metabolism in cardiovascular disease. Hamostaseologie 2017; 35:128-36. [DOI: 10.5482/hamo-14-10-0052] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 12/18/2014] [Indexed: 12/14/2022] Open
Abstract
SummaryCoronary heart disease and stroke, the deadliest forms of cardiovascular disease (CVD), are mainly caused by atherosclerosis, a chronic inflammatory disease of the artery wall driven by maladaptive immune responses in the vessel wall. Various risk factors for CVD influence this pathogenic process, including diabetes mellitus, hypertension, dyslipidaemia, and obesity. Indoleamine 2,3-dioxygenase (IDO), an enzyme catalyzing the rate-limiting step in the kynurenine pathway of tryptophan degradation, is strongly induced by inflammation in several tissues, including the artery wall. An increasing body of evidence indicates that IDO promotes immune tolerance, decreases inflammation, and functions as a homeostatic mechanism against excessive immune reactions.This review provides an overview of the emerging field of the kynurenine pathway of tryptophan degradation in CVD, emphasizing the role of IDO-mediated tryptophan metabolism and its metabolites in the modulation of ‘classical’ cardiovascular risk factors, such as hypertension, obesity, lipid metabolism, diabetes mellitus, and in the development of atherosclerotic CVD.
Collapse
|
59
|
Murphy RA, Moore SC, Playdon M, Meirelles O, Newman AB, Milijkovic I, Kritchevsky SB, Schwartz A, Goodpaster BH, Sampson J, Cawthon P, Simonsick EM, Gerszten RE, Clish CB, Harris TB. Metabolites Associated With Lean Mass and Adiposity in Older Black Men. J Gerontol A Biol Sci Med Sci 2017; 72:1352-1359. [PMID: 28052980 DOI: 10.1093/gerona/glw245] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 11/11/2016] [Indexed: 12/12/2022] Open
Abstract
To identify biomarkers of body mass index, body fat, trunk fat, and appendicular lean mass, nontargeted metabolomics was performed in plasma from 319 black men in the Health, Aging and Body Composition study (median age 72 years, median body mass index 26.8 kg/m2). Body mass index was calculated from measured height and weight; percent fat, percent trunk fat, and appendicular lean mass were measured with dual-energy x-ray absorptiometry. Pearson partial correlations between body composition measures and metabolites were adjusted for age, study site, and smoking. Out of 350 metabolites, body mass index, percent fat, percent trunk fat, and appendicular lean mass were significantly correlated with 92, 48, 96, and 43 metabolites at p less than .0014. Metabolites most strongly correlated with body composition included carnitine, a marker of fatty acid oxidation (positively correlated), triacylglycerols (positively correlated), and amino acids including branched-chain amino acids (positively correlated except for acetylglycine and serine). Gaussian Graphical Models of metabolites found that 25 lipid metabolites clustered into a single network. Groups of five amino acids, three plasmalogens, and two carnitines were also observed. Findings confirm prior reports of associations between amino acids, lean mass, and fat mass in addition to associations not previously reported. Future studies should consider whether these metabolites are relevant for metabolic disease processes.
Collapse
Affiliation(s)
- Rachel A Murphy
- Centre of Excellence in Cancer Prevention, School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - Steven C Moore
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Mary Playdon
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Osorio Meirelles
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Bethesda, Maryland
| | - Anne B Newman
- Center for Aging and Population Health, Department of Epidemiology, University of Pittsburgh, Pennsylvania
| | - Iva Milijkovic
- Center for Aging and Population Health, Department of Epidemiology, University of Pittsburgh, Pennsylvania
| | - Stephen B Kritchevsky
- Stitch Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | | | - Joshua Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Peggy Cawthon
- University of California, San Francisco.,California Pacific Medical Center Research Institute, San Francisco
| | - Eleanor M Simonsick
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland
| | | | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Bethesda, Maryland
| | | |
Collapse
|
60
|
Rebnord EW, Strand E, Midttun Ø, Svingen GFT, Christensen MHE, Ueland PM, Mellgren G, Njølstad PR, Tell GS, Nygård OK, Pedersen ER. The kynurenine:tryptophan ratio as a predictor of incident type 2 diabetes mellitus in individuals with coronary artery disease. Diabetologia 2017; 60:1712-1721. [PMID: 28612106 PMCID: PMC5552838 DOI: 10.1007/s00125-017-4329-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 05/15/2017] [Indexed: 12/13/2022]
Abstract
AIMS/HYPOTHESIS The tryptophan metabolite kynurenine has potent immune modulatory and vasoactive properties. Experimental data implicate kynurenine in obesity-related morbidities. Epidemiological studies are, however, sparse. We evaluated associations of the plasma and urine kynurenine:tryptophan ratio (KTR) to incident type 2 diabetes. METHODS We followed 2519 individuals with coronary artery disease (CAD; 73.1% men) without diabetes at baseline for a median of 7.6 years, during which 173 (6.9%) new incidences of type 2 diabetes were identified. Multivariate Cox regression analyses were applied to investigate the prospective relationships of plasma and urine KTR with new onset type 2 diabetes. RESULTS At inclusion, mean (SD) age was 61.3 (10.4) years, BMI was 25.9 (3.71) kg/m2 and median (interquartile range) HbA1c was 5.6% (5.0%-6.0%) (38 [31-42] mmol/mol). Plasma KTR was not significantly related to type 2 diabetes risk. By contrast, urine KTR showed a strong positive association. Comparing quartile 4 with quartile 1, the HRs (95% CIs) were 2.59 (1.56, 4.30) and 2.35 (1.39, 3.96) in the age- and sex-adjusted and multivariate models, respectively. CONCLUSIONS/INTERPRETATION Urine KTR is a strong predictor of incident type 2 diabetes in individuals with CAD. Potential clinical implications and possible pathogenic roles of renal kynurenine excretion in type 2 diabetes development should be further elucidated.
Collapse
Affiliation(s)
- Eirik W Rebnord
- Department of Clinical Science, University of Bergen, Bergen, Norway.
- KG Jebsen Centre for Diabetes Research, University of Bergen, Bergen, Norway.
- Department of Heart Disease, Haukeland University Hospital, Jonas Lies vei 65, 5021, Bergen, Norway.
| | - Elin Strand
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Gard F T Svingen
- Department of Heart Disease, Haukeland University Hospital, Jonas Lies vei 65, 5021, Bergen, Norway
| | - Monika H E Christensen
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Per M Ueland
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Laboratory of Clinical Biochemistry, Haukeland University Hospital, Bergen, Norway
| | - Gunnar Mellgren
- Department of Clinical Science, University of Bergen, Bergen, Norway
- KG Jebsen Centre for Diabetes Research, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Pål R Njølstad
- Department of Clinical Science, University of Bergen, Bergen, Norway
- KG Jebsen Centre for Diabetes Research, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Grethe S Tell
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Ottar K Nygård
- Department of Clinical Science, University of Bergen, Bergen, Norway
- KG Jebsen Centre for Diabetes Research, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Jonas Lies vei 65, 5021, Bergen, Norway
| | - Eva R Pedersen
- KG Jebsen Centre for Diabetes Research, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Jonas Lies vei 65, 5021, Bergen, Norway
| |
Collapse
|
61
|
Oh JS, Seo HS, Kim KH, Pyo H, Chung BC, Lee J. Urinary profiling of tryptophan and its related metabolites in patients with metabolic syndrome by liquid chromatography-electrospray ionization/mass spectrometry. Anal Bioanal Chem 2017; 409:5501-5512. [PMID: 28710517 DOI: 10.1007/s00216-017-0486-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/24/2017] [Accepted: 06/22/2017] [Indexed: 01/09/2023]
Abstract
Tryptophan (Trp) is an essential amino acid that plays an important role in protein synthesis and is a precursor of various substances related to diverse biological functions. An imbalance in Trp metabolites is associated with inflammatory diseases. The accurate and precise measurement of these compounds in biological specimens would provide meaningful information for understanding the biochemical states of various metabolic syndrome-related diseases, such as hyperlipidemia, hypertension, diabetes, and obesity. In this study, we developed a rapid, accurate, and sensitive liquid chromatography-tandem mass spectrometry-based method for the simultaneous targeted analysis of Trp and its related metabolites of the kynurenine (Kyn), serotonin, and tryptamine pathways in urine. The application of the developed method was tested using urine samples after protein precipitation. The detection limits of Trp and its metabolites were in the range of 0.01 to 0.1 μg/mL. The method was successfully validated and applied to urine samples from controls and patients with metabolic syndrome. Our results revealed high concentrations of Kyn, kynurenic acid, xanthurenic acid, and quinolinic acid as well as a high Kyn-to-Trp ratio (KTR) in patients with metabolic syndromes. The levels of urine Kyn and KTR were significantly increased in patients under 60 years old. The profiling of urinary Trp metabolites could be a useful indicator for age-related diseases including metabolic syndrome. ᅟ.
Collapse
Affiliation(s)
- Ji Sun Oh
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea.,Department of Biotechnology, Graduate School, Korea University, Seoul, Republic of Korea
| | - Hong Seong Seo
- Cardiovascular Center, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul, Republic of Korea
| | - Heesoo Pyo
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Bong Chul Chung
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Jeongae Lee
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea.
| |
Collapse
|
62
|
de Vries LV, Minović I, Franssen CFM, van Faassen M, Sanders JSF, Berger SP, Navis G, Kema IP, Bakker SJL. The tryptophan/kynurenine pathway, systemic inflammation, and long-term outcome after kidney transplantation. Am J Physiol Renal Physiol 2017; 313:F475-F486. [PMID: 28490533 DOI: 10.1152/ajprenal.00690.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 04/25/2017] [Accepted: 05/02/2017] [Indexed: 12/14/2022] Open
Abstract
Tryptophan is metabolized along the kynurenine pathway, initially to kynurenine, and subsequently to cytotoxic 3-hydroxykynurenine. There is increasing interest in this pathway because of its proinflammatory nature, and drugs interfering in it have received increasing attention. We aimed to investigate whether serum and urinary parameters of the tryptophan/kynurenine pathway, and particularly cytotoxic 3-hydroxykynurenine, are associated with systemic inflammation and long-term outcome in renal transplant recipients (RTR). Data were collected in outpatient RTR with a functioning graft for >1 yr. Tryptophan, kynurenine, and 3-hydroxykynurenine in serum and urine were measured using LC-MS/MS. A total of 561 RTR (age: 51 ± 12 yr; 56% male) were included at a median of 6.0 (2.6-11.6) yr posttransplantation. Baseline median serum tryptophan was 40.0 (34.5-46.0) µmol/l, serum kynurenine was 1.8 (1.4-2.2) µmol/l, and serum 3-hydroxykynurenine was 42.2 (31.0-61.7) nmol/l. Serum kynurenine and 3-hydroxykynurenine were strongly associated with parameters of systemic inflammation. During follow-up for 7.0 (6.2-7.5) yr, 51 RTR (9%) developed graft failure and 120 RTR (21%) died. Both serum kynurenine and 3-hydroxykynurenine were independently associated with graft failure [HR 1.72 (1.23-2.41), P = 0.002; and HR 2.03 (1.42-2.90), P < 0.001]. Serum 3-hydroxykynurenine was also independently associated with mortality [HR 1.37 (1.08-1.73), P = 0.01], whereas serum kynurenine was not. Urinary tryptophan/kynurenine pathway parameters were not associated with outcome. Of tryptophan metabolites, serum 3-hydroxykynurenine is cross-sectionally most strongly and consistently associated with systemic inflammation and prospectively with adverse long-term outcome after kidney transplantation. Serum 3-hydroxykynurenine may be an interesting biomarker and target for the evaluation of drugs interfering in the tryptophan/kynurenine pathway.
Collapse
Affiliation(s)
- Laura V de Vries
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands;
| | - Isidor Minović
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and.,Top Institute Food and Nutrition, Wageningen, The Netherlands
| | - Casper F M Franssen
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| | - Jan-Stephan F Sanders
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stefan P Berger
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerjan Navis
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Top Institute Food and Nutrition, Wageningen, The Netherlands
| |
Collapse
|
63
|
Oxenkrug G, van der Hart M, Roeser J, Summergrad P. Peripheral kynurenine-3-monooxygenase deficiency as a potential risk factor for metabolic syndrome in schizophrenia patients. ACTA ACUST UNITED AC 2017; 1. [PMID: 28748226 PMCID: PMC5523985 DOI: 10.15761/icm.1000105] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Increased predisposition of schizophrenia patients (SP) to development of obesity and insulin resistance suggested common signaling pathway between metabolic syndrome (MetS) and schizophrenia. Deficiency of kynurenine-3-monooxygenase (KMO), enzyme catalyzing formation of 3-hydroxykynurenine (3-HK) from kynurenine (Kyn), a tryptophan (Trp) metabolite, might contribute to development of MetS as suggested by non-expression of KMO genes in human fat tissue and elevated serum concentrations of Kyn and its metabolites, kynurenic (KYNA) and anthranilic (ANA) acids, in diabetic patients and Zucker fatty rats (ZFR). Markers of KMO deficiency: decreased 3-HK and elevated Kyn, KYNA and ANA, were observed in brains and spinal fluids of SP, and in brains and serum of experimental animals with genetically- or pharmacologically-induced KMO deficiency. However, elevated concentrations of ANA and decreased 3-HK were reported in serum of SP without concurrent increase of Kyn and KYNA. Present study aimed to re-assess serum Kyn metabolites (HPLC-MS) in a sub-group of SP with elevated KYNA. We found increased Kyn concentrations (by 30%) and Kyn:Trp ratio (by 20%) in serum of SP with elevated KYNA concentrations (by 40%). Obtained results and our previous data suggest that peripheral KMO deficiency might be manifested by, at least, two different patterns: elevated ANA with decreased 3-HK; and elevated KYNA and KYN. The latter pattern was previously described in type 2 diabetes patients and might underline increased predisposition of SP to development of MetS. Assessment of peripheral KMO deficiency might identify SP predisposed to MetS. Attenuation of the consequences of peripheral KMO deficiency might be a new target for prevention/treatment of obesity and diabetes in SP.
Collapse
Affiliation(s)
- Gregory Oxenkrug
- Department of Psychiatry, Tufts University School of Medicine, USA
| | | | | | - Paul Summergrad
- Department of Psychiatry, Tufts University School of Medicine, USA
| |
Collapse
|
64
|
Abnormal kynurenine pathway of tryptophan catabolism in cardiovascular diseases. Cell Mol Life Sci 2017; 74:2899-2916. [PMID: 28314892 DOI: 10.1007/s00018-017-2504-2] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/26/2017] [Accepted: 03/08/2017] [Indexed: 02/06/2023]
Abstract
Kynurenine pathway (KP) is the primary path of tryptophan (Trp) catabolism in most mammalian cells. The KP generates several bioactive catabolites, such as kynurenine (Kyn), kynurenic acid (KA), 3-hydroxykynurenine (3-HK), xanthurenic acid (XA), and 3-hydroxyanthranilic acid (3-HAA). Increased catabolite concentrations in serum are associated with several cardiovascular diseases (CVD), including heart disease, atherosclerosis, and endothelial dysfunction, as well as their risk factors, including hypertension, diabetes, obesity, and aging. The first catabolic step in KP is primarily controlled by indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO). Following this first step, the KP has two major branches, one branch is mediated by kynurenine 3-monooxygenase (KMO) and kynureninase (KYNU) and is responsible for the formation of 3-HK, 3-HAA, and quinolinic acid (QA); and another branch is controlled by kynurenine amino-transferase (KAT), which generates KA. Uncontrolled Trp catabolism has been demonstrated in distinct CVD, thus, understanding the underlying mechanisms by which regulates KP enzyme expression and activity is paramount. This review highlights the recent advances on the effect of KP enzyme expression and activity in different tissues on the pathological mechanisms of specific CVD, KP is an inflammatory sensor and modulator in the cardiovascular system, and KP catabolites act as the potential biomarkers for CVD initiation and progression. Moreover, the biochemical features of critical KP enzymes and principles of enzyme inhibitor development are briefly summarized, as well as the therapeutic potential of KP enzyme inhibitors against CVD is briefly discussed.
Collapse
|
65
|
Abstract
Untargeted metabolomics using high-resolution liquid chromatography-mass spectrometry (LC-MS) is becoming one of the major areas of high-throughput biology. Functional analysis, that is, analyzing the data based on metabolic pathways or the genome-scale metabolic network, is critical in feature selection and interpretation of metabolomics data. One of the main challenges in the functional analyses is the lack of the feature identity in the LC-MS data itself. By matching mass-to-charge ratio (m/z) values of the features to theoretical values derived from known metabolites, some features can be matched to one or more known metabolites. When multiple matchings occur, in most cases only one of the matchings can be true. At the same time, some known metabolites are missing in the measurements. Current network/pathway analysis methods ignore the uncertainty in metabolite identification and the missing observations, which could lead to errors in the selection of significant subnetworks/pathways. In this paper, we propose a flexible network feature selection framework that combines metabolomics data with the genome-scale metabolic network. The method adopts a sequential feature screening procedure and machine learning-based criteria to select important subnetworks and identify the optimal feature matching simultaneously. Simulation studies show that the proposed method has a much higher sensitivity than the commonly used maximal matching approach. For demonstration, we apply the method on a cohort of healthy subjects to detect subnetworks associated with the body mass index (BMI). The method identifies several subnetworks that are supported by the current literature, as well as detects some subnetworks with plausible new functional implications. The R code is available at http://web1.sph.emory.edu/users/tyu8/MSS.
Collapse
Affiliation(s)
- Qingpo Cai
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia 30322, United States
| | - Jessica A. Alvarez
- Department of Medicine, Emory University, Atlanta, Georgia 30322, United States
| | - Jian Kang
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Tianwei Yu
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
66
|
Bonnefond A, Yengo L, Dechaume A, Canouil M, Castelain M, Roger E, Allegaert F, Caiazzo R, Raverdy V, Pigeyre M, Arredouani A, Borys JM, Lévy-Marchal C, Weill J, Roussel R, Balkau B, Marre M, Pattou F, Brousseau T, Froguel P. Relationship between salivary/pancreatic amylase and body mass index: a systems biology approach. BMC Med 2017; 15:37. [PMID: 28228143 PMCID: PMC5322607 DOI: 10.1186/s12916-017-0784-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/09/2017] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Salivary (AMY1) and pancreatic (AMY2) amylases hydrolyze starch. Copy number of AMY1A (encoding AMY1) was reported to be higher in populations with a high-starch diet and reduced in obese people. These results based on quantitative PCR have been challenged recently. We aimed to re-assess the relationship between amylase and adiposity using a systems biology approach. METHODS We assessed the association between plasma enzymatic activity of AMY1 or AMY2, and several metabolic traits in almost 4000 French individuals from D.E.S.I.R. longitudinal study. The effect of the number of copies of AMY1A (encoding AMY1) or AMY2A (encoding AMY2) measured through droplet digital PCR was then analyzed on the same parameters in the same study. A Mendelian randomization analysis was also performed. We subsequently assessed the association between AMY1A copy number and obesity risk in two case-control studies (5000 samples in total). Finally, we assessed the association between body mass index (BMI)-related plasma metabolites and AMY1 or AMY2 activity. RESULTS We evidenced strong associations between AMY1 or AMY2 activity and lower BMI. However, we found a modest contribution of AMY1A copy number to lower BMI. Mendelian randomization identified a causal negative effect of BMI on AMY1 and AMY2 activities. Yet, we also found a significant negative contribution of AMY1 activity at baseline to the change in BMI during the 9-year follow-up, and a significant contribution of AMY1A copy number to lower obesity risk in children, suggesting a bidirectional relationship between AMY1 activity and adiposity. Metabonomics identified a BMI-independent association between AMY1 activity and lactate, a product of complex carbohydrate fermentation. CONCLUSIONS These findings provide new insights into the involvement of amylase in adiposity and starch metabolism.
Collapse
Affiliation(s)
- Amélie Bonnefond
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8199 - EGID, Lille, 59000, France.
| | - Loïc Yengo
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8199 - EGID, Lille, 59000, France.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4067, Australia
| | - Aurélie Dechaume
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8199 - EGID, Lille, 59000, France
| | - Mickaël Canouil
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8199 - EGID, Lille, 59000, France
| | - Maxime Castelain
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8199 - EGID, Lille, 59000, France
| | - Estelle Roger
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8199 - EGID, Lille, 59000, France
| | - Frédéric Allegaert
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8199 - EGID, Lille, 59000, France
| | - Robert Caiazzo
- University of Lille, Inserm, U1190 - EGID, Lille, 59000, France.,Endocrine Surgery Department, CHU of Lille, Lille, 59000, France
| | - Violeta Raverdy
- University of Lille, Inserm, U1190 - EGID, Lille, 59000, France.,Endocrine Surgery Department, CHU of Lille, Lille, 59000, France
| | - Marie Pigeyre
- University of Lille, Inserm, U1190 - EGID, Lille, 59000, France.,Endocrine Surgery Department, CHU of Lille, Lille, 59000, France
| | | | | | - Claire Lévy-Marchal
- Inserm CIE 05 - Department of Clinical Epidemiology, Robert Debré Hospital, Paris, 75019, France
| | - Jacques Weill
- Pediatric Endocrine Department, CHU of Lille, Lille, 59000, France
| | - Ronan Roussel
- Inserm, U1138, Centre de Recherche des Cordeliers, Paris, 75006, France.,Paris-Diderot University, Sorbonne Paris-Cité, Paris, 75013, France.,Department of Endocrinology-Diabetology and Nutrition, DHU-FIRE, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, 75018, France
| | - Beverley Balkau
- Inserm, U1018, CESP, Team 5 (EpReC, Renal and cardiovascular Epidemiology), UVSQ-UPS, Villejuif, 94807, France
| | - Michel Marre
- Inserm, U1138, Centre de Recherche des Cordeliers, Paris, 75006, France.,Paris-Diderot University, Sorbonne Paris-Cité, Paris, 75013, France.,Department of Endocrinology-Diabetology and Nutrition, DHU-FIRE, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, 75018, France
| | - François Pattou
- University of Lille, Inserm, U1190 - EGID, Lille, 59000, France.,Endocrine Surgery Department, CHU of Lille, Lille, 59000, France
| | - Thierry Brousseau
- UF8832 - Biochimie Automatisée, Pôle de Biologie Pathologie Génétique, CHU of Lille, Lille, 59000, France
| | - Philippe Froguel
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8199 - EGID, Lille, 59000, France. .,Department of Genomics of Common Disease, School of Public Health, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK.
| |
Collapse
|
67
|
Engin AB, Engin A. The Interactions Between Kynurenine, Folate, Methionine and Pteridine Pathways in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:511-527. [PMID: 28585214 DOI: 10.1007/978-3-319-48382-5_22] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Obesity activates both innate and adaptive immune responses in adipose tissue. Elevated levels of eosinophils with depression of monocyte and neutrophil indicate the deficiencies in the immune system of morbidly obese individuals. Actually, adipose tissue macrophages are functional antigen-presenting cells that promote the proliferation of interferon-gamma (IFN-gamma)-producing CD4+ T cells in adipose tissue of obese subjects. Eventually, diet-induced obesity is associated with the loss of tissue homeostasis and development of type 1 inflammatory responses in visceral adipose tissue. Activity of inducible indoleamine 2,3-dioxygenase-1 (IDO-1) plays a major role under pro-inflammatory, IFN-gamma dominated settings. One of the two rate-limiting enzymes which can metabolize tryptophan to kynurenine is IDO-1. Tumor necrosis factor-alpha (TNF-alpha) correlates with IDO-1 in adipose compartments. Actually, IDO-1-mediated tryptophan catabolism due to chronic immune activation is the cause of reduced tryptophan plasma levels and be considered as the driving force for food intake in morbidly obese patients. Thus, decrease in plasma tryptophan levels and subsequent reduction in serotonin (5-HT) production provokes satiety dysregulation that leads to increased caloric uptake and obesity. However, after bariatric surgery, weight reduction does not lead to normalization of IDO-1 activity. Furthermore, there is a connection between arginine and tryptophan metabolic pathways in the generation of reactive nitrogen intermediates. Hence, abdominal obesity is associated with vascular endothelial dysfunction and reduced nitric oxide (NO) availability. IFN-gamma-induced activation of the inducible nitric oxide synthase (iNOS) and dissociation of endothelial adenosine monophosphate activated protein kinase (AMPK)- phosphoinositide 3-kinase (PI3K)-protein kinase B (Akt)- endothelial NO synthase (eNOS) pathway enhances oxidative stress production secondary to high-fat diet. Thus, reduced endothelial NO availability correlates with the increase in plasma non-esterified fatty acids and triglycerides levels. Additionally, in obese patients, folate-deficiency leads to hyperhomocysteinemia. Folic acid confers protection against hyperhomocysteinemia-induced oxidative stress.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- , Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
68
|
Zhao Q, Zhu Y, Best LG, Umans JG, Uppal K, Tran VT, Jones DP, Lee ET, Howard BV, Zhao J. Metabolic Profiles of Obesity in American Indians: The Strong Heart Family Study. PLoS One 2016; 11:e0159548. [PMID: 27434237 PMCID: PMC4951134 DOI: 10.1371/journal.pone.0159548] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 07/04/2016] [Indexed: 01/08/2023] Open
Abstract
Obesity is a typical metabolic disorder resulting from the imbalance between energy intake and expenditure. American Indians suffer disproportionately high rates of obesity and diabetes. The goal of this study is to identify metabolic profiles of obesity in 431 normoglycemic American Indians participating in the Strong Heart Family Study. Using an untargeted liquid chromatography-mass spectrometry, we detected 1,364 distinct m/z features matched to known compounds in the current metabolomics databases. We conducted multivariate analysis to identify metabolic profiles for obesity, adjusting for standard obesity indicators. After adjusting for covariates and multiple testing, five metabolites were associated with body mass index and seven were associated with waist circumference. Of them, three were associated with both. Majority of the obesity-related metabolites belongs to lipids, e.g., fatty amides, sphingolipids, prenol lipids, and steroid derivatives. Other identified metabolites are amino acids or peptides. Of the nine identified metabolites, five metabolites (oleoylethanolamide, mannosyl-diinositol-phosphorylceramide, pristanic acid, glutamate, and kynurenine) have been previously implicated in obesity or its related pathways. Future studies are warranted to replicate these findings in larger populations or other ethnic groups.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States of America
| | - Yun Zhu
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States of America
| | - Lyle G. Best
- Missouri Breaks Industries Research Inc, Timber Lake, SD, United States of America
| | - Jason G. Umans
- MedStar Health Research Institute, Hyattsville, MD, United States of America
| | - Karan Uppal
- Division of Pulmonary Medicine, Emory University School of Medicine, Atlanta, GA, United States of America
| | - ViLinh T. Tran
- Division of Pulmonary Medicine, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Dean P. Jones
- Division of Pulmonary Medicine, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Elisa T. Lee
- Center for American Indian Health Research, University of Oklahoma Health Science Center, Oklahoma City, OK, United States of America
| | - Barbara V. Howard
- Medstar Research Institute and Georgetown and Howard Universities Centers for Translational Sciences, Washington, DC, United States of America
| | - Jinying Zhao
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States of America
| |
Collapse
|
69
|
Post-Bariatric Surgery Changes in Quinolinic and Xanthurenic Acid Concentrations Are Associated with Glucose Homeostasis. PLoS One 2016; 11:e0158051. [PMID: 27327770 PMCID: PMC4915629 DOI: 10.1371/journal.pone.0158051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/09/2016] [Indexed: 12/11/2022] Open
Abstract
Background An increase of plasma kynurenine concentrations, potentially bioactive metabolites of tryptophan, was found in subjects with obesity, resulting from low-grade inflammation of the white adipose tissue. Bariatric surgery decreases low-grade inflammation associated with obesity and improves glucose control. Objective Our goal was to determine the concentrations of all kynurenine metabolites after bariatric surgery and whether they were correlated with glucose control improvement. Design Kynurenine metabolite concentrations, analysed by liquid or gas chromatography coupled with tandem mass spectrometry, circulating inflammatory markers, metabolic traits, and BMI were measured before and one year after bariatric surgery in 44 normoglycemic and 47 diabetic women with obesity. Associations between changes in kynurenine metabolites concentrations and in glucose control and metabolic traits were analysed between baseline and twelve months after surgery. Results Tryptophan and kynurenine metabolite concentrations were significantly decreased one year after bariatric surgery and were correlated with the decrease of the usCRP in both groups. Among all the kynurenine metabolites evaluated, only quinolinic acid and xanthurenic acid were significantly associated with glucose control improvement. The one year delta of quinolinic acid concentrations was negatively associated with the delta of fasting glucose (p = 0.019) and HbA1c (p = 0.014), whereas the delta of xanthurenic acid was positively associated with the delta of insulin sensitivity index (p = 0.0018). Conclusion Bariatric surgery has induced a global down-regulation of kynurenine metabolites, associated with weight loss. Our results suggest that, since kynurenine monoxygenase diverts the kynurenine pathway toward the synthesis of xanthurenic acid, its inhibition may also contribute to glucose homeostasis.
Collapse
|
70
|
Inhibition of the aryl hydrocarbon receptor prevents Western diet-induced obesity. Model for AHR activation by kynurenine via oxidized-LDL, TLR2/4, TGFβ, and IDO1. Toxicol Appl Pharmacol 2016; 300:13-24. [PMID: 27020609 DOI: 10.1016/j.taap.2016.03.011] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/11/2016] [Accepted: 03/21/2016] [Indexed: 12/25/2022]
Abstract
Obesity is an increasingly urgent global problem, yet, little is known about its causes and less is known how obesity can be effectively treated. We showed previously that the aryl hydrocarbon receptor (AHR) plays a role in the regulation of body mass in mice fed Western diet. The AHR is a ligand-activated nuclear receptor that regulates genes involved in a number of biological pathways, including xenobiotic metabolism and T cell polarization. This study was an investigation into whether inhibition of the AHR prevents Western diet-based obesity. Male C57Bl/6J mice were fed control and Western diets with and without the AHR antagonist α-naphthoflavone or CH-223191, and a mouse hepatocyte cell line was used to delineate relevant cellular pathways. Studies are presented showing that the AHR antagonists α-naphthoflavone and CH-223191 significantly reduce obesity and adiposity and ameliorates liver steatosis in male C57Bl/6J mice fed a Western diet. Mice deficient in the tryptophan metabolizing enzyme indoleamine 2,3-dioxygenase 1 (IDO1) were also resistant to obesity. Using an AHR-directed, luciferase-expressing mouse hepatocyte cell line, we show that the transforming growth factor β1 (TGFβ1) signaling pathway via PI3K and NF-κB and the toll-like receptor 2/4 (TLR2/4) signaling pathway stimulated by oxidized low-density lipoproteins via NF-κB, each induce luciferase expression; however, TLR2/4 signaling was significantly reduced by inhibition of IDO1. At physiological levels, kynurenine but not kynurenic acid (both tryptophan metabolites and known AHR agonists) activated AHR-directed luciferase expression. We propose a hepatocyte-based model, in which kynurenine production is increased by enhanced IDO1 activity stimulated by TGFβ1 and TLR2/4 signaling, via PI3K and NF-κB, to perpetuate a cycle of AHR activation to cause obesity; and inhibition of the AHR, in turn, blocks the cycle's output to prevent obesity. The AHR with its broad ligand binding specificity is a promising candidate for a potentially simple therapeutic approach for the prevention and treatment of obesity and associated complications.
Collapse
|
71
|
Stepien M, Duarte-Salles T, Fedirko V, Floegel A, Barupal DK, Rinaldi S, Achaintre D, Assi N, Tjønneland A, Overvad K, Bastide N, Boutron-Ruault MC, Severi G, Kühn T, Kaaks R, Aleksandrova K, Boeing H, Trichopoulou A, Bamia C, Lagiou P, Saieva C, Agnoli C, Panico S, Tumino R, Naccarati A, Bueno-de-Mesquita HBA, Peeters PH, Weiderpass E, Quirós JR, Agudo A, Sánchez MJ, Dorronsoro M, Gavrila D, Barricarte A, Ohlsson B, Sjöberg K, Werner M, Sund M, Wareham N, Khaw KT, Travis RC, Schmidt JA, Gunter M, Cross A, Vineis P, Romieu I, Scalbert A, Jenab M. Alteration of amino acid and biogenic amine metabolism in hepatobiliary cancers: Findings from a prospective cohort study. Int J Cancer 2016; 138:348-60. [PMID: 26238458 DOI: 10.1002/ijc.29718] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 07/08/2015] [Indexed: 12/25/2022]
Abstract
Perturbations in levels of amino acids (AA) and their derivatives are observed in hepatocellular carcinoma (HCC). Yet, it is unclear whether these alterations precede or are a consequence of the disease, nor whether they pertain to anatomically related cancers of the intrahepatic bile duct (IHBC), and gallbladder and extrahepatic biliary tract (GBTC). Circulating standard AA, biogenic amines and hexoses were measured (Biocrates AbsoluteIDQ-p180Kit) in a case-control study nested within a large prospective cohort (147 HCC, 43 IHBC and 134 GBTC cases). Liver function and hepatitis status biomarkers were determined separately. Multivariable conditional logistic regression was used to calculate odds ratios and 95% confidence intervals (OR; 95%CI) for log-transformed standardised (mean = 0, SD = 1) serum metabolite levels and relevant ratios in relation to HCC, IHBC or GBTC risk. Fourteen metabolites were significantly associated with HCC risk, of which seven metabolites and four ratios were the strongest predictors in continuous models. Leucine, lysine, glutamine and the ratio of branched chain to aromatic AA (Fischer's ratio) were inversely, while phenylalanine, tyrosine and their ratio, glutamate, glutamate/glutamine ratio, kynurenine and its ratio to tryptophan were positively associated with HCC risk. Confounding by hepatitis status and liver enzyme levels was observed. For the other cancers no significant associations were observed. In conclusion, imbalances of specific AA and biogenic amines may be involved in HCC development.
Collapse
Affiliation(s)
- Magdalena Stepien
- Nutrition and Metabolism Section, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Talita Duarte-Salles
- Nutrition and Metabolism Section, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Veronika Fedirko
- Department of Epidemiology, Rollins School of Public Health, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Anne Floegel
- Department of Epidemiology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany
| | - Dinesh Kumar Barupal
- Nutrition and Metabolism Section, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Sabina Rinaldi
- Nutrition and Metabolism Section, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - David Achaintre
- Nutrition and Metabolism Section, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Nada Assi
- Nutrition and Metabolism Section, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Anne Tjønneland
- Diet, Genes and Enviroment Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Kim Overvad
- Department of Public Health, Section for Epidemiology, Aarhus University, Aarhus, Denmark
| | - Nadia Bastide
- Inserm, Centre for Research in Epidemiology and Population Health (CESP), U1018, Nutrition, Hormones and Women's Health Team, Villejuif, F-94805, France
- Université Paris Sud, UMRS 1018, Villejuif, F-94805, France
- Institut Gustave Roussy, Villejuif, F-94805, France
| | - Marie-Christine Boutron-Ruault
- Inserm, Centre for Research in Epidemiology and Population Health (CESP), U1018, Nutrition, Hormones and Women's Health Team, Villejuif, F-94805, France
- Université Paris Sud, UMRS 1018, Villejuif, F-94805, France
- Institut Gustave Roussy, Villejuif, F-94805, France
| | | | - Tilman Kühn
- Department of Cancer Epidemiology, German Cancer Research Centre, Heidelberg, Germany
| | - Rudolf Kaaks
- Department of Cancer Epidemiology, German Cancer Research Centre, Heidelberg, Germany
| | - Krasimira Aleksandrova
- Department of Epidemiology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany
| | - Antonia Trichopoulou
- Hellenic Health Foundation, Athens, Greece
- Bureau of Epidemiologic Research, Academy of Athens, Athens, Greece
| | - Christina Bamia
- Hellenic Health Foundation, Athens, Greece
- Department of Hygiene, Epidemiology and Medical Statistics, University of Athens Medical School, Athens, Greece
| | - Pagona Lagiou
- Department of Hygiene, Epidemiology and Medical Statistics, University of Athens Medical School, Athens, Greece
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
| | - Calogero Saieva
- Molecular and Nutritional Epidemiology Unit, Cancer Research and Prevention Institute-ISPO, Florence, Italy
| | - Claudia Agnoli
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Salvatore Panico
- Dipartamento Di Medicina Clinicae Chirurgias, Federico II University, Naples, Italy
| | - Rosario Tumino
- Cancer Registry and Histopathology Unit, "Civic-M.P. Arezzo" Hospital, ASP Ragusa, Italy
| | - Alessio Naccarati
- Molecular and Genetic Epidemiology Unit, HuGeF, Human Genetics Foundation, Torino, Italy
| | - H B As Bueno-de-Mesquita
- Department for Determinants of Chronic Diseases (DCD), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Centre, Utrecht, The Netherlands
- Department of Epidemiology and Biostatistics, the School of Public Health, Imperial College London, London, United Kingdom
- Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Petra H Peeters
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center, Utrecht, The Netherlands
| | - Elisabete Weiderpass
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, the Arctic University of Norway, Tromsø, Norway
- Department of Research, Cancer Registry of Norway, Oslo, Norway
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Samfundet Folkhälsan, Helsinki, Finland
| | | | - Antonio Agudo
- Unit of Nutrition, Environment and Cancer, IDIBELL, Catalan Institute of Oncology, Barcelona, Spain
| | - María-José Sánchez
- Escuela Andaluza De Salud Pública, Instituto De Investigación Biosanitaria Ibs, GRANADA, Hospitales Universitarios De Granada/Universidad De Granada, Granada, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER- CIBERESP), Spain
| | - Miren Dorronsoro
- Public Health Direction and Biodonostia-Ciberesp, Basque Regional Health Department, San Sebastian, Spain
| | - Diana Gavrila
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER- CIBERESP), Spain
- Department of Epidemiology, Regional Health Council, Murcia, Spain
- IMIB-Arrixaca, Murcia, Spain
| | - Aurelio Barricarte
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER- CIBERESP), Spain
- Navarre Public Health Institute, Pamplona, Spain
| | - Bodil Ohlsson
- Department of Clinical Sciences, Division of Internal Medicine, Skåne University Hospital Malmö, Sweden
| | - Klas Sjöberg
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Gastroenterology and Nutrition, Skåne University Hospital, Malmö, Sweden
| | - Mårten Werner
- Department of Public Health and Clinical Medicine, Umeå University, Umea, Sweden
| | - Malin Sund
- Department of Surgical and Perioperative Sciences, Umeå University, Umea, Sweden
| | - Nick Wareham
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Kay-Tee Khaw
- Clinical Gerontology, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Ruth C Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Julie A Schmidt
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Marc Gunter
- Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Amanda Cross
- Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Paolo Vineis
- Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Isabelle Romieu
- Nutrition and Metabolism Section, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Augustin Scalbert
- Nutrition and Metabolism Section, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Mazda Jenab
- Nutrition and Metabolism Section, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| |
Collapse
|
72
|
Boulet MM, Chevrier G, Grenier-Larouche T, Pelletier M, Nadeau M, Scarpa J, Prehn C, Marette A, Adamski J, Tchernof A. Alterations of plasma metabolite profiles related to adipose tissue distribution and cardiometabolic risk. Am J Physiol Endocrinol Metab 2015; 309:E736-46. [PMID: 26306599 DOI: 10.1152/ajpendo.00231.2015] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 08/11/2015] [Indexed: 12/17/2022]
Abstract
Metabolomic profiling of obese individuals revealed altered concentrations of many metabolites, especially branched-chain amino acids (BCAA), possibly linked to altered adipose tissue BCAA catabolism. We tested the hypothesis that some features of this metabolite signature relate closely to visceral obesity and concomitant alterations in cardiometabolic risk factors. We also postulated that alterations in BCAA-catabolizing enzymes are predominant in visceral adipose tissue. Fifty-nine women (BMI 20-41 kg/m(2)) undergoing gynecologic surgery were recruited and characterized for overall and regional adiposity, blood metabolite levels using targeted metabolomics, and cardiometabolic risk factors. Adipose samples (visceral and subcutaneous) were obtained and used for gene expression and Western blot analyses. Obese women had significantly higher circulating BCAA and kynurenine/tryptophan (Kyn/Trp) ratio than lean or overweight women (P < 0.01). Principal component analysis confirmed that factors related to AA and the Kyn/Trp ratio were positively associated with BMI, fat mass, visceral or subcutaneous adipose tissue area, and subcutaneous adipocyte size (P ≤ 0.05). AA-related factor was positively associated with HOMA-IR (P ≤ 0.01). Factors reflecting glycerophospholipids and sphingolipids levels were mostly associated with altered blood lipid concentrations (P ≤ 0.05). Glutamate level was the strongest independent predictor of visceral adipose tissue area (r = 0.46, P < 0.001). Obese women had lower expression and protein levels of BCAA-catabolizing enzymes in visceral adipose tissue than overweight or lean women (P ≤ 0.05). We conclude that among metabolites altered in obesity plasma concentrations of BCAA and the Kyn/Trp ratio are closely related to increased adiposity. Alterations in expression and protein levels of BCAA-catabolizing enzymes are predominant in visceral adipose tissue.
Collapse
Affiliation(s)
- Marie Michèle Boulet
- Endocrinology and Nephrology, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada; School of Nutrition, Laval University, Quebec City, Canada; Quebec Heart and Lung Institute, Quebec City, Canada
| | | | | | - Mélissa Pelletier
- Endocrinology and Nephrology, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada
| | | | - Julia Scarpa
- Helmholtz Zentrum München, Institute of Experimental Genetics, Genome Analysis Center, Neuherberg, Germany
| | - Cornelia Prehn
- Helmholtz Zentrum München, Institute of Experimental Genetics, Genome Analysis Center, Neuherberg, Germany
| | - André Marette
- Quebec Heart and Lung Institute, Quebec City, Canada
| | - Jerzy Adamski
- Helmholtz Zentrum München, Institute of Experimental Genetics, Genome Analysis Center, Neuherberg, Germany; Lehrstuhl für Experimentelle Genetik, Technische Universität München, Freising-Weihenstephan, Germany; and German Center for Diabetes Research, Neuherberg, Germany
| | - André Tchernof
- Endocrinology and Nephrology, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada; School of Nutrition, Laval University, Quebec City, Canada; Quebec Heart and Lung Institute, Quebec City, Canada;
| |
Collapse
|
73
|
Favennec M, Hennart B, Caiazzo R, Leloire A, Yengo L, Verbanck M, Arredouani A, Marre M, Pigeyre M, Bessede A, Guillemin GJ, Chinetti G, Staels B, Pattou F, Balkau B, Allorge D, Froguel P, Poulain-Godefroy O. The kynurenine pathway is activated in human obesity and shifted toward kynurenine monooxygenase activation. Obesity (Silver Spring) 2015; 23:2066-74. [PMID: 26347385 DOI: 10.1002/oby.21199] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/20/2015] [Accepted: 06/01/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study characterized the kynurenine pathway (KP) in human obesity by evaluating circulating levels of kynurenines and the expression of KP enzymes in adipose tissue. METHODS Tryptophan and KP metabolite levels were measured in serum of individuals from the D.E.S.I.R. cohort (case-cohort study: 212 diabetic, 836 randomly sampled) and in women with obesity, diabetic or normoglycemic, from the ABOS cohort (n = 100). KP enzyme gene expressions were analyzed in omental and subcutaneous adipose tissue of women from the ABOS cohort, in human primary adipocytes and in monocyte-derived macrophages. RESULTS In the D.E.S.I.R. cohort, kynurenine levels were positively associated with body mass index (BMI) (P = 4.68 × 10(-19) ) and with a higher HOMA2-IR insulin resistance index (P = 6.23 × 10(-4) ). The levels of kynurenine, kynurenic acid, and quinolinic acid were associated with higher BMI (P < 0.05). The expression of several KP enzyme genes (indoleamine 2,3-dioxygenase 1 [IDO1], kynureninase [KYNU], kynurenine 3-monooxygenase [KMO], and kynurenine aminotransferase III [CCBL2]) was increased in the omental adipose tissue of women with obesity compared to lean (P < 0.05), and their expression was induced by proinflammatory cytokines in human primary adipocytes (P < 0.05), except for KMO that is not expressed in these cells. The expressions of IDO1, KYNU, KMO, and CCBL2 were higher in proinflammatory than in anti-inflammatory macrophages (P < 0.05). CONCLUSIONS In the context of obesity, the presence of macrophages in adipose tissue may contribute to diverting KP toward KMO activation.
Collapse
Affiliation(s)
- Marie Favennec
- CNRS UMR 8199, Lille, France
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- CHRU De Lille, Lille, France
| | - Benjamin Hennart
- University of Lille, Lille, France
- CHRU De Lille, Lille, France
- EA4483, Lille, France
| | - Robert Caiazzo
- University of Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- CHRU De Lille, Lille, France
- INSERM UMR 1190, Lille, France
| | - Audrey Leloire
- CNRS UMR 8199, Lille, France
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | - Loïc Yengo
- CNRS UMR 8199, Lille, France
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | - Marie Verbanck
- CNRS UMR 8199, Lille, France
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | | | | | - Marie Pigeyre
- University of Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- CHRU De Lille, Lille, France
- INSERM UMR 1190, Lille, France
| | | | - Gilles J Guillemin
- Neuroinflammation Group, Macquarie University, Sydney, New South Wales, Australia
| | - Giulia Chinetti
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- INSERM UMR 1011, Lille, France
| | - Bart Staels
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- INSERM UMR 1011, Lille, France
| | - François Pattou
- University of Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- CHRU De Lille, Lille, France
- INSERM UMR 1190, Lille, France
| | | | - Delphine Allorge
- University of Lille, Lille, France
- CHRU De Lille, Lille, France
- EA4483, Lille, France
| | - Philippe Froguel
- CNRS UMR 8199, Lille, France
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- CHRU De Lille, Lille, France
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London, UK
| | - Odile Poulain-Godefroy
- CNRS UMR 8199, Lille, France
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| |
Collapse
|
74
|
Oxenkrug G, van der Hart M, Summergrad P. Elevated anthranilic acid plasma concentrations in type 1 but not type 2 diabetes mellitus. ACTA ACUST UNITED AC 2015; 2:365-368. [PMID: 26523229 DOI: 10.15761/imm.1000169] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Experimental data suggested involvement of tryptophan (Trp) - kynurenine (Kyn) pathway (TKP) in mechanisms of autoimmune, type 1 (T1D), and metabolic, type 2 (T2D), diabetes. However, clinical evaluations of TKP metabolites were limited to T2D. We assessed Trp, Kyn and TKP metabolites: anthranilic (AA), kynurenic (KYNA) and xanthurenic (XA) acids, in plasma samples of fifteen T1D, thirty T2D patients and twenty eight non-diabetic subjects by HPLC-mass spectrometry. Trp concentrations were higher in T1D than in T2D and controls while Kyn concentrations were not changed suggesting down-regulation of indoleamine-2,3-dioxygenase (IDO), a rate-limiting enzyme of TKP, in T1D. AA concentrations were 2.3-fold higher in T1D than in T2D and in controls. KYNA and XA concentrations were higher in T1D than in controls, and in previously reported T2D. AA elevation might be a specific feature of T1D. TKP shift towards AA formation in T1D may result from riboflavin deficiency, that increases AA in rats and baboons, and is highly associated with T1D but not T2D. AA augments autoimmune-induced apoptosis of pancreatic cells (PC) by increasing formation of antibodies to PC auto-antigen. Marked increase of AA was reported in rheumatoid arthritis, another autoimmune disorder. Trp, an essential amino acid for humans, is synthesized from AA by diabetogenic intestinal microbiome. AA down-regulates IDO by inhibition of Trp entry into cells. Resulting elevation of Trp attenuates Trp depletion-induced protection of PC against autoimmunity. Further studies of TKP might offer new tools for prevention and treatment of T1D and other autoimmune disorders.
Collapse
Affiliation(s)
- Gregory Oxenkrug
- Psychiatry and Inflammation Program, Department of Psychiatry, Tufts University School of Medicine/Tufts Medical Center, Boston, MA, 02111, USA
| | | | - Paul Summergrad
- Psychiatry and Inflammation Program, Department of Psychiatry, Tufts University School of Medicine/Tufts Medical Center, Boston, MA, 02111, USA
| |
Collapse
|
75
|
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
|
76
|
CD4+ROR γ t++ and Tregs in a Mouse Model of Diet-Induced Nonalcoholic Steatohepatitis. Mediators Inflamm 2015; 2015:239623. [PMID: 26229237 PMCID: PMC4503578 DOI: 10.1155/2015/239623] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 06/07/2015] [Accepted: 06/14/2015] [Indexed: 01/30/2023] Open
Abstract
Background and Aims. Inflammatory mediators that cross-talk in different metabolically active organs are thought to play a crucial role in the pathogenesis of Nonalcoholic Steatohepatitis (NASH). This study was aimed at investigating the CD4+RORγt+ T-helper cells and their counterpart, the CD4+CD25+FOXP3+ regulatory T cells in the liver, subcutaneous adipose tissue (SAT), and abdominal adipose tissue (AAT) in a high fat diet (HFD) mouse model. Methods. C57BL6 mice were fed a HFD or a normal diet (ND). Liver enzymes, metabolic parameters, and liver histology were assessed. The expression of CD4+RORγt+ cells and regulatory T cells in different organs (blood, liver, AAT, and SAT) were analyzed by flow cytometry. Cytokine and adipokine tissue expression were studied by RT-PCR. Results. Mice fed a HFD developed NASH and metabolic alterations compared to normal diet. CD4+RORγt++ cells were significantly increased in the liver and the AAT while an increase of regulatory T cells was observed in the SAT of mice fed HFD compared to ND. Inflammatory cytokines were also upregulated. Conclusions. CD4+RORγt++ cells and regulatory T cells are altered in NASH with a site-specific pattern and correlate with the severity of the disease. These site-specific differences are associated with increased cytokine expression.
Collapse
|
77
|
Kynurenines as predictors of acute coronary events in the Hordaland Health Study. Int J Cardiol 2015; 189:18-24. [DOI: 10.1016/j.ijcard.2015.03.413] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/27/2015] [Accepted: 03/28/2015] [Indexed: 11/21/2022]
|
78
|
Liu JJ, Raynal S, Bailbé D, Gausseres B, Carbonne C, Autier V, Movassat J, Kergoat M, Portha B. Expression of the kynurenine pathway enzymes in the pancreatic islet cells. Activation by cytokines and glucolipotoxicity. Biochim Biophys Acta Mol Basis Dis 2015; 1852:980-91. [PMID: 25675848 DOI: 10.1016/j.bbadis.2015.02.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 01/29/2015] [Accepted: 02/03/2015] [Indexed: 12/19/2022]
Abstract
The tryptophan/kynurenine pathway (TKP) is the main route of tryptophan degradation and generates several neuroactive and immunomodulatory metabolites. Experimental and clinical data have clearly established that besides fat, muscle and liver, pancreatic islet tissue itself is a site of inflammation during obesity and type 2 diabetes. Therefore it is conceivable that pancreatic islet exposure to increased levels of cytokines may induce upregulation of islet kynurenine metabolism in a way resembling that seen in the brain in many neurodegenerative disorders. Using normal rat islets and the INS-1 β-cell line, we have demonstrated for the first time that: 1/only some TKP genes are constitutively expressed, both in β-cells as well as non β-cells; 2/ the regulatory enzyme indoleamine 2,3-dioxygenase (IDO1) is not constitutively expressed; 3/ IDO1 and kynurenine 3-monoxygenase (KMO) expression are potently activated by proinflammatory cytokines (IFN-γ, IL-1β) and glucolipotoxicity respectively, rather in β-cells than in non β-cells; 4/ Islet kynurenine/kynurenic acid production ratio is enhanced following IFN-γ and glucolipotoxicity; 5/ acute exposure to KYN potentiates glucose-induced insulin secretion by normal islets; and 6/ oxidative stress or glucocorticoid modulates TKP genes only marginally. Pancreatic islets may represent a new target tissue for inflammation and glucolipotoxicity to activate the TKP. Since inflammation is now recognized as a crucial mechanism in the development of the metabolic syndrome and more specifically at the islet level, it is needed to evaluate the potential induction of the TKP in the endocrine pancreas during obesity and/or diabetes and its relationship to the islet cell functional alterations.
Collapse
Affiliation(s)
- J J Liu
- UnivParisDiderot, Sorbonne-Paris-Cité, Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptive), CNRS UMR 8251 CNRS, Paris, France; MetaBrain Research, Chilly-Mazarin, France
| | - S Raynal
- MetaBrain Research, Chilly-Mazarin, France
| | - D Bailbé
- UnivParisDiderot, Sorbonne-Paris-Cité, Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptive), CNRS UMR 8251 CNRS, Paris, France
| | - B Gausseres
- UnivParisDiderot, Sorbonne-Paris-Cité, Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptive), CNRS UMR 8251 CNRS, Paris, France
| | - C Carbonne
- MetaBrain Research, Chilly-Mazarin, France
| | - V Autier
- MetaBrain Research, Chilly-Mazarin, France
| | - J Movassat
- UnivParisDiderot, Sorbonne-Paris-Cité, Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptive), CNRS UMR 8251 CNRS, Paris, France
| | - M Kergoat
- MetaBrain Research, Chilly-Mazarin, France
| | - B Portha
- UnivParisDiderot, Sorbonne-Paris-Cité, Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptive), CNRS UMR 8251 CNRS, Paris, France.
| |
Collapse
|
79
|
Pedersen ER, Tuseth N, Eussen SJ, Ueland PM, Strand E, Svingen GFT, Midttun Ø, Meyer K, Mellgren G, Ulvik A, Nordrehaug JE, Nilsen DW, Nygård O. Associations of Plasma Kynurenines With Risk of Acute Myocardial Infarction in Patients With Stable Angina Pectoris. Arterioscler Thromb Vasc Biol 2015; 35:455-62. [DOI: 10.1161/atvbaha.114.304674] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Objective—
Enhanced tryptophan degradation, induced by the proinflammatory cytokine interferon-γ, has been related to cardiovascular disease progression and insulin resistance. We assessed downstream tryptophan metabolites of the kynurenine pathway as predictors of acute myocardial infarction in patients with suspected stable angina pectoris. Furthermore, we evaluated potential effect modifications according to diagnoses of pre-diabetes mellitus or diabetes mellitus.
Approach and Results—
Blood samples were obtained from 4122 patients (median age, 62 years; 72% men) who underwent elective coronary angiography. During median follow-up of 56 months, 8.3% had acute myocardial infarction. Comparing the highest quartile to the lowest, for the total cohort, multivariable adjusted hazard ratios (95% confidence intervals) were 1.68 (1.21–2.34), 1.81 (1.33–2.48), 1.68 (1.21–2.32), and 1.48 (1.10–1.99) for kynurenic acid, hydroxykynurenine, anthranilic acid, and hydroxyanthranilic acid, respectively. The kynurenines correlated with phenotypes of the metabolic syndrome, and risk associations were generally stronger in subgroups classified with pre-diabetes mellitus or diabetes mellitus at inclusion (
P
int
≤0.05). Evaluated in the total population, hydroxykynurenine and anthranilic acid provided statistically significant net reclassification improvements (0.21 [0.08–0.35] and 0.21 [0.07–0.35], respectively).
Conclusions—
In patients with suspected stable angina pectoris, elevated levels of plasma kynurenines predicted increased risk of acute myocardial infarction, and risk estimates were generally stronger in subgroups with evidence of impaired glucose homeostasis. Future studies should aim to clarify roles of the kynurenine pathway in atherosclerosis and glucose metabolism.
Collapse
Affiliation(s)
- Eva Ringdal Pedersen
- From the Department of Clinical Science (E.R.P., N.T., P.M.U., E.S., G.F.T.S., G.M., J.E.N., D.W.N., O.N.) and Department of Global Public Health and Primary Health Care (S.J.P.M.E.), University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway (N.T., O.N.); Department of Epidemiology, School for Public Health and Primary Care-CAPHRI, Maastricht University, Maastricht, The Netherlands (S.J.P.M.E.); Laboratory of Clinical Biochemistry (P.M.U.) and
| | - Nora Tuseth
- From the Department of Clinical Science (E.R.P., N.T., P.M.U., E.S., G.F.T.S., G.M., J.E.N., D.W.N., O.N.) and Department of Global Public Health and Primary Health Care (S.J.P.M.E.), University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway (N.T., O.N.); Department of Epidemiology, School for Public Health and Primary Care-CAPHRI, Maastricht University, Maastricht, The Netherlands (S.J.P.M.E.); Laboratory of Clinical Biochemistry (P.M.U.) and
| | - Simone J.P.M. Eussen
- From the Department of Clinical Science (E.R.P., N.T., P.M.U., E.S., G.F.T.S., G.M., J.E.N., D.W.N., O.N.) and Department of Global Public Health and Primary Health Care (S.J.P.M.E.), University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway (N.T., O.N.); Department of Epidemiology, School for Public Health and Primary Care-CAPHRI, Maastricht University, Maastricht, The Netherlands (S.J.P.M.E.); Laboratory of Clinical Biochemistry (P.M.U.) and
| | - Per Magne Ueland
- From the Department of Clinical Science (E.R.P., N.T., P.M.U., E.S., G.F.T.S., G.M., J.E.N., D.W.N., O.N.) and Department of Global Public Health and Primary Health Care (S.J.P.M.E.), University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway (N.T., O.N.); Department of Epidemiology, School for Public Health and Primary Care-CAPHRI, Maastricht University, Maastricht, The Netherlands (S.J.P.M.E.); Laboratory of Clinical Biochemistry (P.M.U.) and
| | - Elin Strand
- From the Department of Clinical Science (E.R.P., N.T., P.M.U., E.S., G.F.T.S., G.M., J.E.N., D.W.N., O.N.) and Department of Global Public Health and Primary Health Care (S.J.P.M.E.), University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway (N.T., O.N.); Department of Epidemiology, School for Public Health and Primary Care-CAPHRI, Maastricht University, Maastricht, The Netherlands (S.J.P.M.E.); Laboratory of Clinical Biochemistry (P.M.U.) and
| | - Gard Frodahl Tveitevåg Svingen
- From the Department of Clinical Science (E.R.P., N.T., P.M.U., E.S., G.F.T.S., G.M., J.E.N., D.W.N., O.N.) and Department of Global Public Health and Primary Health Care (S.J.P.M.E.), University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway (N.T., O.N.); Department of Epidemiology, School for Public Health and Primary Care-CAPHRI, Maastricht University, Maastricht, The Netherlands (S.J.P.M.E.); Laboratory of Clinical Biochemistry (P.M.U.) and
| | - Øivind Midttun
- From the Department of Clinical Science (E.R.P., N.T., P.M.U., E.S., G.F.T.S., G.M., J.E.N., D.W.N., O.N.) and Department of Global Public Health and Primary Health Care (S.J.P.M.E.), University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway (N.T., O.N.); Department of Epidemiology, School for Public Health and Primary Care-CAPHRI, Maastricht University, Maastricht, The Netherlands (S.J.P.M.E.); Laboratory of Clinical Biochemistry (P.M.U.) and
| | - Klaus Meyer
- From the Department of Clinical Science (E.R.P., N.T., P.M.U., E.S., G.F.T.S., G.M., J.E.N., D.W.N., O.N.) and Department of Global Public Health and Primary Health Care (S.J.P.M.E.), University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway (N.T., O.N.); Department of Epidemiology, School for Public Health and Primary Care-CAPHRI, Maastricht University, Maastricht, The Netherlands (S.J.P.M.E.); Laboratory of Clinical Biochemistry (P.M.U.) and
| | - Gunnar Mellgren
- From the Department of Clinical Science (E.R.P., N.T., P.M.U., E.S., G.F.T.S., G.M., J.E.N., D.W.N., O.N.) and Department of Global Public Health and Primary Health Care (S.J.P.M.E.), University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway (N.T., O.N.); Department of Epidemiology, School for Public Health and Primary Care-CAPHRI, Maastricht University, Maastricht, The Netherlands (S.J.P.M.E.); Laboratory of Clinical Biochemistry (P.M.U.) and
| | - Arve Ulvik
- From the Department of Clinical Science (E.R.P., N.T., P.M.U., E.S., G.F.T.S., G.M., J.E.N., D.W.N., O.N.) and Department of Global Public Health and Primary Health Care (S.J.P.M.E.), University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway (N.T., O.N.); Department of Epidemiology, School for Public Health and Primary Care-CAPHRI, Maastricht University, Maastricht, The Netherlands (S.J.P.M.E.); Laboratory of Clinical Biochemistry (P.M.U.) and
| | - Jan Erik Nordrehaug
- From the Department of Clinical Science (E.R.P., N.T., P.M.U., E.S., G.F.T.S., G.M., J.E.N., D.W.N., O.N.) and Department of Global Public Health and Primary Health Care (S.J.P.M.E.), University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway (N.T., O.N.); Department of Epidemiology, School for Public Health and Primary Care-CAPHRI, Maastricht University, Maastricht, The Netherlands (S.J.P.M.E.); Laboratory of Clinical Biochemistry (P.M.U.) and
| | - Dennis W. Nilsen
- From the Department of Clinical Science (E.R.P., N.T., P.M.U., E.S., G.F.T.S., G.M., J.E.N., D.W.N., O.N.) and Department of Global Public Health and Primary Health Care (S.J.P.M.E.), University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway (N.T., O.N.); Department of Epidemiology, School for Public Health and Primary Care-CAPHRI, Maastricht University, Maastricht, The Netherlands (S.J.P.M.E.); Laboratory of Clinical Biochemistry (P.M.U.) and
| | - Ottar Nygård
- From the Department of Clinical Science (E.R.P., N.T., P.M.U., E.S., G.F.T.S., G.M., J.E.N., D.W.N., O.N.) and Department of Global Public Health and Primary Health Care (S.J.P.M.E.), University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway (N.T., O.N.); Department of Epidemiology, School for Public Health and Primary Care-CAPHRI, Maastricht University, Maastricht, The Netherlands (S.J.P.M.E.); Laboratory of Clinical Biochemistry (P.M.U.) and
| |
Collapse
|
80
|
Navrotskaya V, Oxenkrug G, Vorobyova L, Summergrad P. Attenuation of high sucrose diet-induced insulin resistance in tryptophan 2,3-dioxygenase deficient Drosophila melanogaster vermilion mutants. ACTA ACUST UNITED AC 2015; 1:93-95. [PMID: 26191458 DOI: 10.15761/iod.1000120] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Exposure to high sugar diet (HSD) serves as an experimental model of insulin resistance (IR) and type 2 diabetes (T2D) in mammals and insects. Peripheral IR induced by HSD delays emergence of pupae from larvae and decreases body weight of Drosophila imago. Understanding of mechanisms of IR/T2D is essential for refining T2D prevention and treatment strategies. Dysregulation of tryptophan (TRP) - kynurenine (KYN) pathway was suggested as one of the mechanisms of IR development. Rate-limiting enzyme of TRP - KYN pathway in Drosophila is TRP 2,3-dioxygenase (TDO), an evolutionary conserved ortholog of human TDO. In insects TDO is encoded by vermilion gene. TDO is not active in vermilion mutants. In order to evaluate the possible impact of deficient formation of KYN from TRP on the inducement of IR by HSD, we compared the effect of HSD in wild type (Oregon) and vermilion mutants of Drosophila melanogaster by assessing the time of white pupae emergence from larva and body weight of imago. Delay of emergence of pupae from larvae induced by high sucrose diet was less pronounced in vermilion (1.4 days) than in Oregon flies (3.3 days) in comparison with flies maintained on standard diet. Exposure to high sucrose diet decreased body weight of Oregon (but not vermilion) imago. Attenuation of high sucrose diet-induced IR/T2D in vermilion flies might depend on deficiency of TRP - KYN pathway. Besides IR/T2D, HSD induces obesity in Drosophila. Future studies of HSD-induced obesity and IR/T2D in TDO deficient vermilion mutants of Drosophila might help to understand the mechanisms of high association between IR/T2D and obesity. Modulation of TRP - KYN metabolism might be utilized for prevention and treatment of IR/T2D.
Collapse
Affiliation(s)
- Valeriya Navrotskaya
- Department of Genetics and Cytology, V.N. Karazin Kharkiv National University, Kharkiv, Ukraine
| | - Gregory Oxenkrug
- Psychiatry and Inflammation Program, Department of Psychiatry, Tufts University/Tufts Medical Center, Boston, MA, USA
| | - Lyudmila Vorobyova
- Department of Genetics and Cytology, V.N. Karazin Kharkiv National University, Kharkiv, Ukraine
| | - Paul Summergrad
- Psychiatry and Inflammation Program, Department of Psychiatry, Tufts University/Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
81
|
Diabetes and Tryptophan Metabolism. TRYPTOPHAN METABOLISM: IMPLICATIONS FOR BIOLOGICAL PROCESSES, HEALTH AND DISEASE 2015. [DOI: 10.1007/978-3-319-15630-9_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
82
|
3-Hydroxykynurenic Acid and Type 2 Diabetes: Implications for Aging, Obesity, Depression, Parkinson’s Disease, and Schizophrenia. TRYPTOPHAN METABOLISM: IMPLICATIONS FOR BIOLOGICAL PROCESSES, HEALTH AND DISEASE 2015. [DOI: 10.1007/978-3-319-15630-9_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
83
|
Anti-sRAGE autoimmunity in obesity: Downturn after bariatric surgery is independent of previous diabetic status. DIABETES & METABOLISM 2014; 40:356-62. [DOI: 10.1016/j.diabet.2014.04.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/23/2014] [Accepted: 04/25/2014] [Indexed: 12/15/2022]
|
84
|
Reininghaus EZ, McIntyre RS, Reininghaus B, Geisler S, Bengesser SA, Lackner N, Hecht K, Birner A, Kattnig F, Unterweger R, Kapfhammer HP, Zelzer S, Fuchs D, Mangge H. Tryptophan breakdown is increased in euthymic overweight individuals with bipolar disorder: a preliminary report. Bipolar Disord 2014; 16:432-40. [PMID: 24330408 DOI: 10.1111/bdi.12166] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 08/27/2013] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Individuals with bipolar disorder (BD) are disproportionately affected by symptoms of being overweight and metabolic syndrome when compared to the general population. The pertinence of this observation is underscored by observations that excess weight is associated with a more complex illness presentation, course, and outcome in BD. We present the first preliminary report of our BIPFAT study, which explored shared hypothesized pathophysiological pathways between being overweight and having BD. METHODS We investigated the tryptophan-kynurenine metabolism pathway as a proxy of dysregulated inflammatory homeostasis in euthymic, overweight individuals with BD (n = 78) compared to healthy controls (n = 156). RESULTS Both blood kynurenine concentrations and the kynurenine to tryptophan ratio [(Kyn:Trp); an estimate of tryptophan breakdown] were significantly higher in the total sample of euthymic patients with BD, with greater increases noted in both parameters in the subsample of overweight patients with BD. When compared to controls, peripheral neopterin concentrations were significantly lower. Within the BD group, there were also significant between-group differences in neopterin concentrations, with higher levels in those who were overweight and in subjects with BD in the later stages of illness compared to earlier stages. CONCLUSIONS Increased tryptophan breakdown, as well as neopterin levels in BD, may be an indirect mediator of immune-mediated inflammation. In BD, this may account for the high prevalence of medical comorbidities and increased mortality. The observation of increased kynurenine levels and Kyn:Trp, and altered circulating neopterin levels provides indirect evidence of increased activity of tryptophan-degrading indoleamine 2,3-dioxygenase in euthymic individuals with BD, underscoring the role of inflammatory mediators as a causative and/or consequent factor. More robust abnormalities in the overweight subsample underscore the additional inflammatory burden of medical comorbidity and suggest a shared pathophysiology as well as a mechanism mediating BD and cardiovascular disease.
Collapse
|
85
|
Mangge H, Summers KL, Meinitzer A, Zelzer S, Almer G, Prassl R, Schnedl WJ, Reininghaus E, Paulmichl K, Weghuber D, Fuchs D. Obesity-related dysregulation of the tryptophan-kynurenine metabolism: role of age and parameters of the metabolic syndrome. Obesity (Silver Spring) 2014; 22:195-201. [PMID: 23625535 DOI: 10.1002/oby.20491] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 04/01/2013] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Obesity-related immune mediated systemic inflammation was associated with the development of the metabolic syndrome by induction of the tryptophan (TRP)-kynurenine (KYN) pathway. The study aimed to assess whether this holds true across the lifespan from juvenility to adulthood. DESIGN AND METHODS Five hundred twenty-seven participants aged between 10 and 65 years were analyzed. Standard anthropometric measures, carotid ultrasound, and laboratory analysis including interleukin-6, ultra-sensitive C-reactive protein, lipids, glucose metabolism, neopterin, TRP, KYN levels, and the KYN/TRP ratio were performed. RESULTS Overweight/obese (ow/ob) adults had significantly increased KYN serum levels and a significantly increased KYN/TRP ratio. In sharp contrast, ow/ob juvenile males aged ≤18 years showed decreased, females similar KYN and KYN/TRP ratio in comparison to their control counterparts. Also, adult ow/ob subjects with metabolic syndrome showed markedly increased KYN/TRP ratios contrary to decreased KYN/TRP ratios in ow/ob juveniles. Abdominal fat content, characterized by age normalized waist circumference, and not body mass index, had the strongest effect for an increase of the KYN/TRP ratio in adults. CONCLUSIONS TRP metabolism and obesity-related immune mediated inflammation differs markedly between juveniles and adults. While childhood obesity seems to be dominated by a Th2-driven activation, an accelerated production of Th1-type cytokines may pave the way for later atherosclerotic endpoints.
Collapse
Affiliation(s)
- Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Research Unit on Lifestyle and Inflammation-associated Risk Biomarkers, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Oxenkrug G. Insulin resistance and dysregulation of tryptophan-kynurenine and kynurenine-nicotinamide adenine dinucleotide metabolic pathways. Mol Neurobiol 2013; 48:294-301. [PMID: 23813101 PMCID: PMC3779535 DOI: 10.1007/s12035-013-8497-4] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 06/16/2013] [Indexed: 11/25/2022]
Abstract
Insulin resistance (IR) underlines aging and aging-associated medical (diabetes, obesity, dyslipidemia, hypertension) and psychiatric (depression, cognitive decline) disorders. Molecular mechanisms of IR in genetically or metabolically predisposed individuals remain uncertain. Current review of the literature and our data presents the evidences that dysregulation of tryptophan (TRP)-kynurenine (KYN) and KYN-nicotinamide adenine dinucleotide (NAD) metabolic pathways is one of the mechanisms of IR. The first and rate-limiting step of TRP-KYN pathway is regulated by enzymes inducible by pro-inflammatory factors and/or stress hormones. The key enzymes of KYN-NAD pathway require pyridoxal-5-phosphate (P5P), an active form of vitamin B6, as a cofactor. Deficiency of P5P diverts KYN-NAD metabolism from production of NAD to the excessive formation of xanthurenic acid (XA). Human and experimental studies suggested that XA and some other KYN metabolites might impair production, release, and biological activity of insulin. We propose that one of the mechanisms of IR is inflammation- and/or stress-induced upregulation of TRP-KYN metabolism in combination with P5P deficiency-induced diversion of KYN-NAD metabolism towards formation of XA and other KYN derivatives affecting insulin activity. Monitoring of KYN/P5P status and formation of XA might help to identify subjects at risk for IR. Pharmacological regulation of the TRP-KYN and KYN-NAD pathways and maintaining of adequate vitamin B6 status might contribute to prevention and treatment of IR in conditions associated with inflammation/stress-induced excessive production of KYN and deficiency of vitamin B6, e.g., type 2 diabetes, obesity, cardiovascular diseases, aging, menopause, pregnancy, and hepatitis C virus infection.
Collapse
Affiliation(s)
- Gregory Oxenkrug
- Psychiatry and Inflammation Program, Department of Psychiatry, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA,
| |
Collapse
|
87
|
Nagano J, Shimizu M, Hara T, Shirakami Y, Kochi T, Nakamura N, Ohtaki H, Ito H, Tanaka T, Tsurumi H, Saito K, Seishima M, Moriwaki H. Effects of indoleamine 2,3-dioxygenase deficiency on high-fat diet-induced hepatic inflammation. PLoS One 2013; 8:e73404. [PMID: 24039933 PMCID: PMC3767792 DOI: 10.1371/journal.pone.0073404] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 07/23/2013] [Indexed: 12/18/2022] Open
Abstract
Hepatic immune regulation is associated with the progression from simple steatosis to non-alcoholic steatohepatitis, a severe condition of inflamed fatty liver. Indoleamine 2,3-dioxygenase (IDO), an intracellular enzyme that mediates the catabolism of L-tryptophan to L-kynurenine, plays an important role in hepatic immune regulation. In the present study, we examined the effects of IDO gene silencing on high-fat diet (HFD)-induced liver inflammation and fibrosis in mice. After being fed a HFD for 26 weeks, the IDO-knockout (KO) mice showed a marked infiltration of inflammatory cells, especially macrophages and T lymphocytes, in the liver. The expression levels of F4/80, IFNγ, IL-1β, and IL-6 mRNA in the liver and the expression levels of F4/80 and TNF-α mRNA in the white adipose tissue were significantly increased in IDO-KO mice, although hepatic steatosis, the accumulation of intrahepatic triglycerides, and the amount of oxidative stress were lower than those in IDO-wild-type mice. IDO-KO mice also developed marked pericellular fibrosis in the liver, accumulated hepatic hydroxyproline, and exhibited increased expression levels of hepatic TGF-β1 mRNA. These findings suggest that IDO-KO renders the mice more susceptible to HFD-induced hepatic inflammation and fibrosis. Therefore, IDO may have a protective effect against hepatic fibrosis, at least in this HFD-induced liver injury model.
Collapse
Affiliation(s)
- Junji Nagano
- Department of Internal Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masahito Shimizu
- Department of Internal Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
- * E-mail:
| | - Takeshi Hara
- Department of Internal Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yohei Shirakami
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Takahiro Kochi
- Department of Internal Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Nobuhiko Nakamura
- Department of Internal Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hirofumi Ohtaki
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hiroyasu Ito
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Takuji Tanaka
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hisashi Tsurumi
- Department of Internal Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Kuniaki Saito
- Human Health Sciences, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Mitsuru Seishima
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hisataka Moriwaki
- Department of Internal Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
88
|
Poulain-Godefroy O, Eury E, Leloire A, Hennart B, Guillemin GJ, Allorge D, Froguel P. Induction of TDO2 and IDO2 in Liver by High-Fat Feeding in Mice: Discrepancies with Human Obesity. Int J Tryptophan Res 2013; 6:29-37. [PMID: 26882470 PMCID: PMC3729279 DOI: 10.4137/ijtr.s11717] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Low-grade and chronic inflammation is elicited in white adipose tissue in human obesity. The presence of inflammatory molecules leads to an increased tryptophan catabolism through the induction of indoleamine-2,3-dioxygenase-1 (IDO1). In order to characterize the mechanisms underlying this dysregulation, we have studied 2 mouse models of obesity. Unexpectedly, we did not detect any IDO1 expression in obese or lean mice adipose tissue. In a previous study, we did not find any significant difference in the liver for IDO2 and tryptophan-2,3-dioxygenase (TDO2) gene expression between normal weight and obese patients. IDO2 and TDO2 expression was increased in the liver of high-fat fed mice, but not in ob/ob mice, and was strongly correlated with hydroxysteroid-(11-beta) dehydrogenase-1 (HSD11B1) expression, an enzyme that generates active cortisol within tissues. In conclusion, despite a dysregulation of tryptophan metabolism, obese mice display discrepancies with human obesity metabolism, rendering them inappropriate for further investigations in this animal model.
Collapse
Affiliation(s)
- Odile Poulain-Godefroy
- European Genomic Institute for Diabetes (EGID), Lille, France.; University of Lille, Lille, France.; CNRS UMR 8199, Lille, France.; CHRU Lille, Lille, France.; IPL, Lille, France
| | - Elodie Eury
- European Genomic Institute for Diabetes (EGID), Lille, France.; University of Lille, Lille, France.; CNRS UMR 8199, Lille, France.; CHRU Lille, Lille, France.; IPL, Lille, France
| | - Audrey Leloire
- European Genomic Institute for Diabetes (EGID), Lille, France.; University of Lille, Lille, France.; CNRS UMR 8199, Lille, France.; CHRU Lille, Lille, France.; IPL, Lille, France
| | - Benjamin Hennart
- University of Lille, Lille, France.; CHRU Lille, Lille, France.; EA4483, Faculty of Medicine, Lille, France
| | - Gilles J Guillemin
- MND and Neurodegenerative Diseases Research Group, Australian School of Advanced Medicine, Macquarie University, Australia
| | - Delphine Allorge
- University of Lille, Lille, France.; CHRU Lille, Lille, France.; EA4483, Faculty of Medicine, Lille, France
| | - Philippe Froguel
- European Genomic Institute for Diabetes (EGID), Lille, France.; University of Lille, Lille, France.; CNRS UMR 8199, Lille, France.; CHRU Lille, Lille, France.; IPL, Lille, France.; Department of Genomics of Common Disease, School of Public Health, Imperial College London, United Kingdom
| |
Collapse
|