801
|
Microbiota Alterations in Alzheimer’s Disease: Involvement of the Kynurenine Pathway and Inflammation. Neurotox Res 2019; 36:424-436. [DOI: 10.1007/s12640-019-00057-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 12/15/2022]
|
802
|
Okabe K, Yaku K, Tobe K, Nakagawa T. Implications of altered NAD metabolism in metabolic disorders. J Biomed Sci 2019; 26:34. [PMID: 31078136 PMCID: PMC6511662 DOI: 10.1186/s12929-019-0527-8] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/25/2019] [Indexed: 12/15/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is an important coenzyme that participates in various energy metabolism pathways, including glycolysis, β-oxidation, and oxidative phosphorylation. Besides, it is a required cofactor for post-translational modifications such as ADP-ribosylation and deacetylation by poly (ADP-ribose) polymerases (PARPs) and sirtuins, respectively. Thus, NAD regulates energy metabolism, DNA damage repair, gene expression, and stress response through these enzymes. Numerous studies have shown that NAD levels decrease with aging and under disturbed nutrient conditions, such as obesity. Additionally, a decline in NAD levels is closely related to the development of various metabolic disorders, including diabetes and fatty liver disease. In addition, many studies have revealed that administration of NAD precursors, such as nicotinamide mononucleotide (NMN) and nicotinamide riboside (NR), efficiently increase NAD levels in various tissues and prevent such metabolic diseases. These NAD precursors are contained in natural foods, such as cow milk, vegetables, and meats. Therefore, altered NAD metabolism can be a practical target for nutritional intervention. Recently, several human clinical trials using NAD precursors have been conducted to investigate the safety, pharmacokinetics, and efficacy against metabolic disorders such as glucose intolerance. In this review, we summarize current knowledge on the implications of NAD metabolism in metabolic diseases and discuss the outcomes of recent human clinical trials.
Collapse
Affiliation(s)
- Keisuke Okabe
- Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194 Japan
- First Department of Internal Medicine, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, 930-0194 Japan
| | - Keisuke Yaku
- Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194 Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, 930-0194 Japan
| | - Takashi Nakagawa
- Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194 Japan
- Institute of Natural Medicine, University of Toyama, Toyama, 930-0194 Japan
| |
Collapse
|
803
|
Herrstedt A, Bay ML, Simonsen C, Sundberg A, Egeland C, Thorsen-Streit S, Djurhuus SS, Magne Ueland P, Midttun Ø, Pedersen BK, Bo Svendsen L, de Heer P, Christensen JF, Hojman P. Exercise-mediated improvement of depression in patients with gastro-esophageal junction cancer is linked to kynurenine metabolism. Acta Oncol 2019; 58:579-587. [PMID: 30696326 DOI: 10.1080/0284186x.2018.1558371] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Exercise may improve depression in cancer patients, yet the molecular mechanism behind this protection is poorly understood. Here, we aimed to explore the link between exercise and regulation of kynurenine (Kyn) metabolism and inflammation in patients with operable gastro-esophageal junction (GEJ) cancer patients, who improved significantly in depression score with exercise training. Material and Methods: Fifty GEJ cancer patients were allocated to 12 weeks of supervised training twice weekly including interval-based aerobic exercise and resistance training, or standard care. Depression score was evaluated by HADS, and blood samples and muscle biopsies were collected for determination of Kyn metabolism and inflammation across the intervention. Results: Depression scores decreased by -1.3 points in the exercise group (p < 0.01), whereas no changes were observed in the control group. Plasma 3-hydroxykynurenine (HK), a Kyn metabolite giving rise to other neurotoxic metabolites, increased by 48% (p <0.001) in the control group, while exercise training attenuated this accumulation. The production of HK is induced by inflammation, and while we observed no differences in systemic pro-inflammatory cytokines, exercise training ameliorated the treatment-induced intramuscular inflammation. Moreover, exercise has been suggested to convert Kyn to the neuroprotective metabolite, kynurenic acid (KA), but despite marked functional and muscular exercise-mediated adaptations, we did not observe any enhancement of KA production and related enzyme expression in the muscles of GEJ cancer patients. Conclusion: Exercise training reduced symptoms of depression in patients with GEJ cancer, and this effect was associated with an exercise-dependent attenuation of the inflammation-induced conversion of Kyn to neurotoxic metabolites.
Collapse
Affiliation(s)
- Anita Herrstedt
- Centre of Inflammation and Metabolism (CIM) and Centre for Physical Activity Research (CFAS), Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Marie L. Bay
- Centre of Inflammation and Metabolism (CIM) and Centre for Physical Activity Research (CFAS), Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Casper Simonsen
- Centre of Inflammation and Metabolism (CIM) and Centre for Physical Activity Research (CFAS), Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Anna Sundberg
- Centre of Inflammation and Metabolism (CIM) and Centre for Physical Activity Research (CFAS), Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte Egeland
- Department of Surgical Gastroenterology C, Rigshospitalet, Copenhagen, Denmark
| | - Sarah Thorsen-Streit
- Centre of Inflammation and Metabolism (CIM) and Centre for Physical Activity Research (CFAS), Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Sissal S. Djurhuus
- Centre of Inflammation and Metabolism (CIM) and Centre for Physical Activity Research (CFAS), Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Per Magne Ueland
- Department of Clinical Science, University of Bergen; Laboratory of Clinical Biochemistry, Haukeland University Hospital, Bergen, Norway
| | | | - Bente K. Pedersen
- Centre of Inflammation and Metabolism (CIM) and Centre for Physical Activity Research (CFAS), Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Lars Bo Svendsen
- Department of Surgical Gastroenterology C, Rigshospitalet, Copenhagen, Denmark
| | - Pieter de Heer
- Department of Surgical Gastroenterology C, Rigshospitalet, Copenhagen, Denmark
| | - Jesper F. Christensen
- Centre of Inflammation and Metabolism (CIM) and Centre for Physical Activity Research (CFAS), Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Pernille Hojman
- Centre of Inflammation and Metabolism (CIM) and Centre for Physical Activity Research (CFAS), Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
804
|
Iwaki T, Bennion BG, Stenson EK, Lynn JC, Otinga C, Djukovic D, Raftery D, Fei L, Wong HR, Liles WC, Standage SW. PPARα contributes to protection against metabolic and inflammatory derangements associated with acute kidney injury in experimental sepsis. Physiol Rep 2019; 7:e14078. [PMID: 31102342 PMCID: PMC6525329 DOI: 10.14814/phy2.14078] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 01/05/2023] Open
Abstract
Sepsis-associated acute kidney injury (AKI) is a significant problem in critically ill children and adults resulting in increased morbidity and mortality. Fundamental mechanisms contributing to sepsis-associated AKI are poorly understood. Previous research has demonstrated that peroxisome proliferator-activated receptor α (PPARα) expression is associated with reduced organ system failure in sepsis. Using an experimental model of polymicrobial sepsis, we demonstrate that mice deficient in PPARα have worse kidney function, which is likely related to reduced fatty acid oxidation and increased inflammation. Ultrastructural evaluation with electron microscopy reveals that the proximal convoluted tubule is specifically injured in septic PPARα deficient mice. In this experimental group, serum metabolomic analysis reveals unanticipated metabolic derangements in tryptophan-kynurenine-NAD+ and pantothenate pathways. We also show that a subgroup of children with sepsis whose genome-wide expression profiles are characterized by repression of the PPARα signaling pathway has increased incidence of severe AKI. These findings point toward interesting associations between sepsis-associated AKI and PPARα-driven fatty acid metabolism that merit further investigation.
Collapse
Affiliation(s)
- Takuma Iwaki
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashington
- Department of PediatricsUniversity HospitalFaculty of MedicineKagawa UniversityKagawaJapan
| | - Brock G. Bennion
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashington
- Department of Pathology and ImmunologyWashington University School of MedicineSt. LouisMissouri
| | - Erin K. Stenson
- Department of PediatricsSection of Critical CareUniversity of Colorado School of MedicineAnschutz Medical CenterChildren's Hospital ColoradoAuroraColorado
- Division of Critical Care MedicineCincinnati Children's Hospital Medical CenterCincinnatiOhio
| | - Jared C. Lynn
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashington
| | - Cynthia Otinga
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashington
| | - Danijel Djukovic
- Department of Chemistry and BiochemistryUniversity of ColoradoBoulderColorado
- Department of Anesthesiology and Pain MedicineUniversity of Washington School of MedicineSeattleWashington
| | - Daniel Raftery
- Department of Anesthesiology and Pain MedicineUniversity of Washington School of MedicineSeattleWashington
| | - Lin Fei
- Division of Biostatistics and EpidemiologyCincinnati Children's Hospital Medical CenterCincinnatiOhio
- Department of PediatricsUniversity of CincinnatiCincinnatiOhio
| | - Hector R. Wong
- Division of Critical Care MedicineCincinnati Children's Hospital Medical CenterCincinnatiOhio
- Department of PediatricsUniversity of CincinnatiCincinnatiOhio
| | - W. Conrad Liles
- Department of MedicineUniversity of Washington School of MedicineSeattleWashington
| | - Stephen W. Standage
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashington
- Division of Critical Care MedicineCincinnati Children's Hospital Medical CenterCincinnatiOhio
- Department of PediatricsUniversity of CincinnatiCincinnatiOhio
| |
Collapse
|
805
|
Sovijit WN, Sovijit WE, Pu S, Usuda K, Inoue R, Watanabe G, Yamaguchi H, Nagaoka K. Ovarian progesterone suppresses depression and anxiety-like behaviors by increasing the Lactobacillus population of gut microbiota in ovariectomized mice. Neurosci Res 2019; 168:76-82. [PMID: 31022413 DOI: 10.1016/j.neures.2019.04.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/15/2019] [Accepted: 04/16/2019] [Indexed: 12/19/2022]
Abstract
Depression and anxiety, which are severe symptoms during menopause, are caused by ceased ovarian activity and declined serum progesterone levels. Studies have demonstrated that gut microbiota can regulate brain function and change the microbiota composition during the perimenopause period. This study investigated whether progesterone affects depressant and anxious behaviors via gut microbiota. In ovariectomized (OVX) mice, treatment with progesterone improved depressive and anxious behaviors, and gut microbiota composition was significantly changed. In particular, increased Lactobacillus spp. were observed in these mice. Reduction of microbiota by antibiotic treatment abolished the effect of progesterone on depression and anxiety. In addition, administration of Lactobacillus (L.) reuteri that was increased by progesterone also reduced the depressant behavior in OVX mice, and BDNF gene expression was elevated by progesterone treatment and L. reuteri administration in the hippocampus. Moreover, we found that progesterone stimulated the growth of L. reuteri in vitro. In summary, our findings indicate that progesterone reduces depression and anxiety through changes in gut microbiota composition, particularly by increasing the Lactobacillus spp. population.
Collapse
Affiliation(s)
- Watcharin N Sovijit
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Japan
| | - Watcharee E Sovijit
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Japan
| | - Shaoxia Pu
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Japan
| | - Kento Usuda
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Agricultural and Life Sciences, Kyoto Prefectural University, Japan
| | - Gen Watanabe
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Japan
| | - Hirohito Yamaguchi
- Cancer research center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar
| | - Kentaro Nagaoka
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Japan.
| |
Collapse
|
806
|
The ‘Yin’ and the ‘Yang’ of the kynurenine pathway: excitotoxicity and neuroprotection imbalance in stress-induced disorders. Behav Pharmacol 2019; 30:163-186. [DOI: 10.1097/fbp.0000000000000477] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
807
|
Clària J, Moreau R, Fenaille F, Amorós A, Junot C, Gronbaek H, Coenraad MJ, Pruvost A, Ghettas A, Chu-Van E, López-Vicario C, Oettl K, Caraceni P, Alessandria C, Trebicka J, Pavesi M, Deulofeu C, Albillos A, Gustot T, Welzel TM, Fernández J, Stauber RE, Saliba F, Butin N, Colsch B, Moreno C, Durand F, Nevens F, Bañares R, Benten D, Ginès P, Gerbes A, Jalan R, Angeli P, Bernardi M, Arroyo V. Orchestration of Tryptophan-Kynurenine Pathway, Acute Decompensation, and Acute-on-Chronic Liver Failure in Cirrhosis. Hepatology 2019; 69:1686-1701. [PMID: 30521097 DOI: 10.1002/hep.30363] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 10/22/2018] [Indexed: 12/12/2022]
Abstract
Systemic inflammation (SI) is involved in the pathogenesis of acute decompensation (AD) and acute-on-chronic liver failure (ACLF) in cirrhosis. In other diseases, SI activates tryptophan (Trp) degradation through the kynurenine pathway (KP), giving rise to metabolites that contribute to multiorgan/system damage and immunosuppression. In the current study, we aimed to characterize the KP in patients with cirrhosis, in whom this pathway is poorly known. The serum levels of Trp, key KP metabolites (kynurenine and kynurenic and quinolinic acids), and cytokines (SI markers) were measured at enrollment in 40 healthy subjects, 39 patients with compensated cirrhosis, 342 with AD (no ACLF) and 180 with ACLF, and repeated in 258 patients during the 28-day follow-up. Urine KP metabolites were measured in 50 patients with ACLF. Serum KP activity was normal in compensated cirrhosis, increased in AD and further increased in ACLF, in parallel with SI; it was remarkably higher in ACLF with kidney failure than in ACLF without kidney failure in the absence of differences in urine KP activity and fractional excretion of KP metabolites. The short-term course of AD and ACLF (worsening, improvement, stable) correlated closely with follow-up changes in serum KP activity. Among patients with AD at enrollment, those with the highest baseline KP activity developed ACLF during follow-up. Among patients who had ACLF at enrollment, those with immune suppression and the highest KP activity, both at baseline, developed nosocomial infections during follow-up. Finally, higher baseline KP activity independently predicted mortality in patients with AD and ACLF. Conclusion: Features of KP activation appear in patients with AD, culminate in patients with ACLF, and may be involved in the pathogenesis of ACLF, clinical course, and mortality.
Collapse
Affiliation(s)
- Joan Clària
- European Foundation for the Study of Chronic Liver Failure Consortium and Grifols Chair, Barcelona, Spain.,Hospital Clínic, IDIBAPS and CIBERehd, Barcelona, Spain
| | - Richard Moreau
- European Foundation for the Study of Chronic Liver Failure Consortium and Grifols Chair, Barcelona, Spain.,Inserm, Centre de Recherche sur l'Inflammation, Université Paris Diderot-Paris, Département Hospitalo-Universitaire UNITY; Service d'Hépatologie, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris; Laboratoire d'Excellence Inflamex, ComUE Sorbonne Paris Cité, Paris, France
| | - François Fenaille
- CEA, INRA, Université Paris Saclay, Laboratoire d'Etude du Métabolisme des Médicaments, MetaboHUB-Paris, Gif-Sur-Yvette, France
| | - Alex Amorós
- European Foundation for the Study of Chronic Liver Failure Consortium and Grifols Chair, Barcelona, Spain
| | - Christophe Junot
- CEA, INRA, Université Paris Saclay, Laboratoire d'Etude du Métabolisme des Médicaments, MetaboHUB-Paris, Gif-Sur-Yvette, France
| | - Henning Gronbaek
- Department of Hepatology & Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Minneke J Coenraad
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Alain Pruvost
- CEA, INRA Université Paris Saclay, Service de Pharmacologie et Immunoanalyse, Plateforme SMArt-MS, Gif-sur-Yvette, France
| | - Aurélie Ghettas
- CEA, INRA Université Paris Saclay, Service de Pharmacologie et Immunoanalyse, Plateforme SMArt-MS, Gif-sur-Yvette, France
| | - Emeline Chu-Van
- CEA, INRA, Université Paris Saclay, Laboratoire d'Etude du Métabolisme des Médicaments, MetaboHUB-Paris, Gif-Sur-Yvette, France
| | | | - Karl Oettl
- Medical University of Graz, Graz, Austria
| | - Paolo Caraceni
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Carlo Alessandria
- Division of Gastroenterology and Hepatology, San Giovanni Battista Hospital, Torino, Italy
| | - Jonel Trebicka
- European Foundation for the Study of Chronic Liver Failure Consortium and Grifols Chair, Barcelona, Spain.,Department of Internal Medicine I, University of Bonn, Bonn, Germany.,J.W. Goethe University Hospital, Frankfurt, Germany
| | - Marco Pavesi
- European Foundation for the Study of Chronic Liver Failure Consortium and Grifols Chair, Barcelona, Spain
| | - Carme Deulofeu
- European Foundation for the Study of Chronic Liver Failure Consortium and Grifols Chair, Barcelona, Spain
| | | | - Thierry Gustot
- CUB Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Javier Fernández
- European Foundation for the Study of Chronic Liver Failure Consortium and Grifols Chair, Barcelona, Spain.,Hospital Clínic, IDIBAPS and CIBERehd, Barcelona, Spain
| | | | - Faouzi Saliba
- Hôpital Paul Brousse, Université Paris-Sud, Villejuif, France
| | - Noémie Butin
- CEA, INRA, Université Paris Saclay, Laboratoire d'Etude du Métabolisme des Médicaments, MetaboHUB-Paris, Gif-Sur-Yvette, France
| | - Benoit Colsch
- CEA, INRA, Université Paris Saclay, Laboratoire d'Etude du Métabolisme des Médicaments, MetaboHUB-Paris, Gif-Sur-Yvette, France
| | - Christophe Moreno
- CUB Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - François Durand
- Inserm, Centre de Recherche sur l'Inflammation, Université Paris Diderot-Paris, Département Hospitalo-Universitaire UNITY; Service d'Hépatologie, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris; Laboratoire d'Excellence Inflamex, ComUE Sorbonne Paris Cité, Paris, France
| | | | - Rafael Bañares
- Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | | | - Pere Ginès
- Hospital Clínic, IDIBAPS and CIBERehd, Barcelona, Spain
| | - Alexander Gerbes
- Department of Medicine II, University Hospital LMU Munich, Liver Center Munich, Munich, Germany
| | - Rajiv Jalan
- Liver Failure Group, Institute for Liver Disease Health, University College London, Royal Free Hospital, London, United Kingdom
| | - Paolo Angeli
- European Foundation for the Study of Chronic Liver Failure Consortium and Grifols Chair, Barcelona, Spain.,Unit of Internal Medicine and Hepatology, Department of Medicine, DIMED, University of Padova, Padoa, Italy
| | - Mauro Bernardi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Vicente Arroyo
- European Foundation for the Study of Chronic Liver Failure Consortium and Grifols Chair, Barcelona, Spain
| | | |
Collapse
|
808
|
Whiley L, Nye LC, Grant I, Andreas N, Chappell KE, Sarafian MH, Misra R, Plumb RS, Lewis MR, Nicholson JK, Holmes E, Swann JR, Wilson ID. Ultrahigh-Performance Liquid Chromatography Tandem Mass Spectrometry with Electrospray Ionization Quantification of Tryptophan Metabolites and Markers of Gut Health in Serum and Plasma-Application to Clinical and Epidemiology Cohorts. Anal Chem 2019; 91:5207-5216. [PMID: 30848589 PMCID: PMC6503468 DOI: 10.1021/acs.analchem.8b05884] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
A targeted
ultrahigh-performance liquid chromatography tandem mass
spectrometry with electrospray ionization (UHPLC-ESI-MS/MS) method
has been developed for the quantification of tryptophan and its downstream
metabolites from the kynurenine and serotonin pathways. The assay
coverage also includes markers of gut health and inflammation, including
citrulline and neopterin. The method was designed in 96-well plate
format for application in multiday, multiplate clinical and epidemiology
population studies. A chromatographic cycle time of 7 min enables
the analysis of two 96-well plates in 24 h. To protect chromatographic
column lifespan, samples underwent a two-step extraction, using solvent
protein precipitation followed by delipidation via solid-phase extraction
(SPE). Analytical validation reported accuracy of each analyte <20%
for the lowest limit of quantification and <15% for all other quality
control (QC) levels. The analytical precision for each analyte was
2.1–12.9%. To test the applicability of the method to multiplate
and multiday preparations, a serum pool underwent periodic repeat
analysis during a run consisting of 18 plates. The % CV (coefficient
of variation) values obtained for each analyte were <15%. Additional
biological testing applied the assay to samples collected from healthy
control participants and two groups diagnosed with inflammatory bowel
disease (IBD) (one group treated with the anti-inflammatory 5-aminosalicylic
acid (5-ASA) and one group untreated), with results showing significant
differences in the concentrations of picolinic acid, kynurenine, and
xanthurenic acid. The short analysis time and 96-well plate format
of the assay makes it suitable for high-throughput targeted UHPLC-ESI-MS/MS
metabolomic analysis in large-scale clinical and epidemiological population
studies.
Collapse
Affiliation(s)
- Luke Whiley
- UK Dementia Research Institute, Burlington Danes Building , Imperial College London , Hammersmith Hospital, London W12 0NN , United Kingdom.,MRC-NIHR National Phenome Centre, IRDB Building , Imperial College London , Hammersmith Hospital, London W12 0NN , United Kingdom
| | - Leanne C Nye
- Division of Integrative Systems and Digestive Medicine, Department of Surgery and Cancer , Imperial College London , Sir Alexander Fleming Building, South Kensington Campus , London SW7 2AZ , United Kingdom
| | - Isobelle Grant
- Division of Integrative Systems and Digestive Medicine, Department of Surgery and Cancer , Imperial College London , Sir Alexander Fleming Building, South Kensington Campus , London SW7 2AZ , United Kingdom.,Waters Corporation , Milford , Massachusetts 01757 , United States
| | - Nick Andreas
- Division of Integrative Systems and Digestive Medicine, Department of Surgery and Cancer , Imperial College London , Sir Alexander Fleming Building, South Kensington Campus , London SW7 2AZ , United Kingdom
| | - Katie E Chappell
- MRC-NIHR National Phenome Centre, IRDB Building , Imperial College London , Hammersmith Hospital, London W12 0NN , United Kingdom
| | - Magali H Sarafian
- Division of Integrative Systems and Digestive Medicine, Department of Surgery and Cancer , Imperial College London , Sir Alexander Fleming Building, South Kensington Campus , London SW7 2AZ , United Kingdom
| | - Ravi Misra
- St. Marks Hospital and Academic Institute , Watford Road , Middlesex, London HA1 3UJ , United Kingdom
| | - Robert S Plumb
- Waters Corporation , Milford , Massachusetts 01757 , United States
| | - Matthew R Lewis
- MRC-NIHR National Phenome Centre, IRDB Building , Imperial College London , Hammersmith Hospital, London W12 0NN , United Kingdom
| | - Jeremy K Nicholson
- Australian National Phenome Centre , Murdoch University , Harry Perkins Building , Perth , Western Australia 6150 , Australia
| | - Elaine Holmes
- UK Dementia Research Institute, Burlington Danes Building , Imperial College London , Hammersmith Hospital, London W12 0NN , United Kingdom.,MRC-NIHR National Phenome Centre, IRDB Building , Imperial College London , Hammersmith Hospital, London W12 0NN , United Kingdom.,Division of Integrative Systems and Digestive Medicine, Department of Surgery and Cancer , Imperial College London , Sir Alexander Fleming Building, South Kensington Campus , London SW7 2AZ , United Kingdom.,Australian National Phenome Centre , Murdoch University , Harry Perkins Building , Perth , Western Australia 6150 , Australia
| | - Jonathan R Swann
- Division of Integrative Systems and Digestive Medicine, Department of Surgery and Cancer , Imperial College London , Sir Alexander Fleming Building, South Kensington Campus , London SW7 2AZ , United Kingdom
| | - Ian D Wilson
- Division of Integrative Systems and Digestive Medicine, Department of Surgery and Cancer , Imperial College London , Sir Alexander Fleming Building, South Kensington Campus , London SW7 2AZ , United Kingdom
| |
Collapse
|
809
|
Yuan X, Kang Y, Zhuo C, Huang XF, Song X. The gut microbiota promotes the pathogenesis of schizophrenia via multiple pathways. Biochem Biophys Res Commun 2019; 512:373-380. [PMID: 30898321 DOI: 10.1016/j.bbrc.2019.02.152] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 02/28/2019] [Indexed: 02/07/2023]
Abstract
Schizophrenia is a severe mental disorder with unknown etiology. Many mechanisms, including dysregulation of neurotransmitters, immune disturbance, and abnormal neurodevelopment, are proposed for the pathogenesis of schizophrenia. The significance of communication between intestinal flora and the central nervous system through the gut-brain axis is increasingly being recognized. The intestinal microbiota plays an important role in regulating neurotransmission, immune homeostasis, and brain development. We hypothesize that an imbalance in intestinal flora causes immune activation and dysfunction in the gut-brain axis, contributing to schizophrenia. In this review, we examine recent studies that explore the intestinal flora and immune-mediated neurodevelopment of schizophrenia. We conclude that an imbalance in intestinal flora may reduce protectants and increase neurotoxin and inflammatory mediators, causing neuronal and synaptic damage, which induces schizophrenia.
Collapse
Affiliation(s)
- Xiuxia Yuan
- The First Affiliated Hospital/Zhengzhou University, Zhengzhou, China; Biological Psychiatry International Joint Laboratory of Henan/Zhengzhou University, Zhengzhou, China; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China
| | - Yulin Kang
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Chuanjun Zhuo
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute and School of Medicine, University of Wollongong, NSW, 2522, Australia.
| | - Xueqin Song
- The First Affiliated Hospital/Zhengzhou University, Zhengzhou, China; Biological Psychiatry International Joint Laboratory of Henan/Zhengzhou University, Zhengzhou, China; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
810
|
Joisten N, Rademacher A, Bloch W, Schenk A, Oberste M, Dalgas U, Langdon D, Caminada D, Purde MT, Gonzenbach R, Kool J, Zimmer P, Bansi J. Influence of different rehabilitative aerobic exercise programs on (anti-) inflammatory immune signalling, cognitive and functional capacity in persons with MS - study protocol of a randomized controlled trial. BMC Neurol 2019; 19:37. [PMID: 30849952 PMCID: PMC6407211 DOI: 10.1186/s12883-019-1267-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/04/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Studies have shown positive effects of therapeutic exercise on motor- and cognitive function as well as on psychosocial outcomes in persons with multiple sclerosis (MS). A reduction of inflammatory stress through physical exercise has been suspected as one key mechanism, mediating the positive effects of exercise in the context of MS. The primary objective of this trial is to investigate the acute and chronic effects of different exercise modalities on (anti-)inflammatory immune signalling as well as on cognitive and functional capacity in persons with MS. METHODS A two armed single-blind randomized controlled design will investigate 72 persons with relapsing remitting or secondary progressive MS (EDSS 3.0-6.0), during 3 weeks of inpatient rehabilitation. Participants will be randomized into either a high-intensity interval training (HIIT) or a moderate continuous training group; the latter represents the local standard therapy (ST). Both groups will exercise 3x per week. The HIIT group will perform 5 × 1.5-min high-intensive exercise bouts at 95-100% of their maximum heart rate (HRmax) followed by active breaks of unloaded pedalling (60% HRmax) for 2 min. In contrast, the ST group will exercise for 24 min continuously at 65% of HRmax. The proportion of circulating regulatory T-cells will be measured as primary outcome. Secondary outcomes comprise numbers and proportions of further immune cells including Th17-cells, soluble factors ((anti-) inflammatory cytokines, tryptophan metabolites), endurance capacity, cognitive performance, processing skills for activities of daily living, fatigue, depression and healthcare-related quality of life. Outcomes will be assessed before (T0) and after (T3) the 3-week exercise intervention program. Blood samples of T0 will be taken immediately before the first exercise session. Additionally, blood samples for the soluble factors will be collected immediately after (T1) and three hours (T2) after the first exercise session of each group. DISCUSSION This study will be the first to investigate both acute and chronic effects of aerobic exercise on immune function and disease associated biomarkers in persons with MS. Combining biological analyses with cognitive and functional capacity assessments may contribute to a better understanding of responses to rehabilitative training, needed to improve exercise recommendations for persons with MS. TRIAL REGISTRATION This trial was prospectively registered at ClinicalTrials.gov ( NCT03652519 ; 29 August 2018).
Collapse
Affiliation(s)
- Niklas Joisten
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
| | - Annette Rademacher
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
- Deparment of Neurology, Kliniken-Valens, Rehabilitationsklinik-Valens, Taminaplatz 1, 7317 Valens, Switzerland
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
| | - Alexander Schenk
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
| | - Max Oberste
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
| | - Ulrik Dalgas
- Department of Public Health, Section of Sport Science, Århus University, Dalgas Avenue 4, 8000 Århus C, Denmark
| | - Dawn Langdon
- Royal Holloway University of London, Egham, TW20 0EX Surrey UK
| | - Daniel Caminada
- labormedizinisches zentrum Dr Risch, Lagerstrasse 30, 9470 Buchs, Switzerland
| | - Mette-Triin Purde
- labormedizinisches zentrum Dr Risch, Lagerstrasse 30, 9470 Buchs, Switzerland
| | - Roman Gonzenbach
- Deparment of Neurology, Kliniken-Valens, Rehabilitationsklinik-Valens, Taminaplatz 1, 7317 Valens, Switzerland
| | - Jan Kool
- Deparment of Neurology, Kliniken-Valens, Rehabilitationsklinik-Valens, Taminaplatz 1, 7317 Valens, Switzerland
| | - Philipp Zimmer
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
- Division of Physical Activity, Prevention and Cancer, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120 Heidelberg, Germany
| | - Jens Bansi
- Deparment of Neurology, Kliniken-Valens, Rehabilitationsklinik-Valens, Taminaplatz 1, 7317 Valens, Switzerland
| |
Collapse
|
811
|
Byers NM, Fleshman AC, Perera R, Molins CR. Metabolomic Insights into Human Arboviral Infections: Dengue, Chikungunya, and Zika Viruses. Viruses 2019; 11:E225. [PMID: 30845653 PMCID: PMC6466193 DOI: 10.3390/v11030225] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/21/2019] [Accepted: 02/21/2019] [Indexed: 12/14/2022] Open
Abstract
The global burden of arboviral diseases and the limited success in controlling them calls for innovative methods to understand arbovirus infections. Metabolomics has been applied to detect alterations in host physiology during infection. This approach relies on mass spectrometry or nuclear magnetic resonance spectroscopy to evaluate how perturbations in biological systems alter metabolic pathways, allowing for differentiation of closely related conditions. Because viruses heavily depend on host resources and pathways, they present unique challenges for characterizing metabolic changes. Here, we review the literature on metabolomics of arboviruses and focus on the interpretation of identified molecular features. Metabolomics has revealed biomarkers that differentiate disease states and outcomes, and has shown similarities in metabolic alterations caused by different viruses (e.g., lipid metabolism). Researchers investigating such metabolomic alterations aim to better understand host⁻virus dynamics, identify diagnostically useful molecular features, discern perturbed pathways for therapeutics, and guide further biochemical research. This review focuses on lessons derived from metabolomics studies on samples from arbovirus-infected humans.
Collapse
Affiliation(s)
- Nathaniel M Byers
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO 80521, USA.
| | - Amy C Fleshman
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO 80521, USA.
| | - Rushika Perera
- Arthropod-borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology & Pathology, Colorado State University, Fort Collins, CO 80523-1692, USA.
| | - Claudia R Molins
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO 80521, USA.
| |
Collapse
|
812
|
Zhang X, Shao H, Zheng X. Amino acids at the intersection of nutrition and insulin sensitivity. Drug Discov Today 2019; 24:1038-1043. [PMID: 30818029 DOI: 10.1016/j.drudis.2019.02.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/06/2019] [Accepted: 02/19/2019] [Indexed: 01/02/2023]
Abstract
A systems network that is coordinated in the sensing and management of nutrient signals is paramount to energy homeostasis, and its dysfunction induces metabolic stress and insulin resistance. Amino acids have recently emerged as a collection of signaling metabolites that underlie the metabolic impacts of different dietary patterns and life styles. This relationship is beginning to be understood from the close coupling of immune and metabolic systems, and serves to enrich our understanding of metabolic diseases, such as type 2 diabetes mellitus. In this review, we provide an overview of several amino acids or their metabolites that link nutrients with insulin sensitivity and discuss how they integrate into organ crosstalk pathways to influence physiological or pathological metabolic states.
Collapse
Affiliation(s)
- Xueli Zhang
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Hua Shao
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiao Zheng
- School of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
813
|
Bay-Richter C, Buttenschøn HN, Mors O, Eskelund A, Budac D, Kærlev L, Wegener G. Latent toxoplasmosis and psychiatric symptoms - A role of tryptophan metabolism? J Psychiatr Res 2019; 110:45-50. [PMID: 30583085 DOI: 10.1016/j.jpsychires.2018.12.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 02/02/2023]
Abstract
Toxoplasma gondii (TOX) is a common parasite which infects approximately one third of the human population. In recent years, it has been suggested that latent toxoplasmosis may be a risk factor for the development of mental disorders, particularly schizophrenia and anxiety. With regards to depression the results have been varied. The main objective of this study was to examine subpopulations from the Danish PRISME and GENDEP populations for TOX IgG antibodies. These consisted of: a group with symptoms of anxiety, a group suffering from burnout syndrome, as well as two different subpopulations with depression of differing severity. The secondary objective of this study was to examine whether tryptophan metabolism was altered in TOX-positive subjects within each subpopulation. Our results show that the anxiety and burnout populations were more likely to be TOX IgG seropositive. Furthermore, we find that the moderate-severe but not mild-moderate depressive subpopulation were associated with TOX seropositivety, suggesting a possible role of symptom severity. Additionally, we found that TOX positive subjects in the anxiety and burnout subpopulations had altered tryptophan metabolism. This relationship did not exist in the mild-moderate depressive subpopulation. These results suggest that TOX seropositivity may be related to anxiety, burnout and potentially to severity of depression. We furthermore show that the psychiatric symptoms could be associated with an altered tryptophan metabolism.
Collapse
Affiliation(s)
- Cecilie Bay-Richter
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark.
| | | | - Ole Mors
- Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus University, Aarhus, Denmark
| | - Amanda Eskelund
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| | | | - Linda Kærlev
- Research Unit of Clinical Epidemiology, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark; Center for Clinical Epidemiology, Odense University Hospital, Odense, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| |
Collapse
|
814
|
Zheng X, Zhang A, Binnie M, McGuire K, Webster SP, Hughes J, Howie SEM, Mole DJ. Kynurenine 3-monooxygenase is a critical regulator of renal ischemia-reperfusion injury. Exp Mol Med 2019; 51:1-14. [PMID: 30760699 PMCID: PMC6374422 DOI: 10.1038/s12276-019-0210-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 10/18/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023] Open
Abstract
Acute kidney injury (AKI) following ischemia–reperfusion injury (IRI) has a high mortality and lacks specific therapies. Here, we report that mice lacking kynurenine 3-monooxygenase (KMO) activity (Kmonull mice) are protected against AKI after renal IRI. We show that KMO is highly expressed in the kidney and exerts major metabolic control over the biologically active kynurenine metabolites 3-hydroxykynurenine, kynurenic acid, and downstream metabolites. In experimental AKI induced by kidney IRI, Kmonull mice had preserved renal function, reduced renal tubular cell injury, and fewer infiltrating neutrophils compared with wild-type (Kmowt) control mice. Together, these data confirm that flux through KMO contributes to AKI after IRI, and supports the rationale for KMO inhibition as a therapeutic strategy to protect against AKI during critical illness. Inhibition of a metabolic enzyme linked to inflammation could be a novel treatment approach for sudden kidney failure following a “reperfusion” injury caused by blood flow returning to the organ after a period of insufficient blood supply. Damian Mole and colleagues from the University of Edinburgh, UK, temporarily blocked blood vessels leading to the kidneys of mice to induce organ damage. Mice that lacked a working copy of kynurenine 3-monooxygenase (KMO), a gene that encodes an enzyme involved in metabolizing an essential amino acid linked to immune activation, were protected from injury. These KMO-mutant mice experienced less damage to the kidney’s tubular cells and had fewer pro-inflammatory cells than genetically normal animals. The findings support the idea that blocking KMO and its associated metabolic pathway could help mitigate kidney damage following reperfusion injury in humans.
Collapse
Affiliation(s)
- Xiaozhong Zheng
- Centre for Inflammation Research, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Ailiang Zhang
- Centre for Inflammation Research, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Margaret Binnie
- Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Kris McGuire
- Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Scott P Webster
- Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Jeremy Hughes
- Centre for Inflammation Research, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Sarah E M Howie
- Centre for Inflammation Research, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Damian J Mole
- Centre for Inflammation Research, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
815
|
Christensen JJ, Bakke SS, Ulven SM, Retterstøl K, Henriksen T, Bollerslev J, Espevik T, Aukrust P, Halvorsen B, Holven KB. Serum Omega-6 Fatty Acids and Immunology-Related Gene Expression in Peripheral Blood Mononuclear Cells: A Cross-Sectional Analysis in Healthy Children. Mol Nutr Food Res 2019; 63:e1800990. [PMID: 30702198 DOI: 10.1002/mnfr.201800990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/20/2018] [Indexed: 01/07/2023]
Abstract
SCOPE Some studies suggest that a high dietary intake of omega-6 fatty acids is pro-inflammatory. However, whether omega-6 fatty acids actually cause pathogenic inflammation in humans is debated. Therefore, the associations between expression of immunology-related genes in peripheral blood mononuclear cells (PBMCs) and serum total omega-6 PUFA status are investigated. METHODS AND RESULTS Serum fatty acid profile and expression of 460 immunology-related genes in PBMCs from 58 healthy children (6-13 years) is measured, and examined the expression differences between children with high or low total omega-6 PUFA status (upper vs lower tertile). Taken together, both univariate analyses and integrated omics analyses support that while high omega-6 PUFA level associated with higher expressing of genes related to innate immune responses, it also associated with lower expression of several genes related to adaptive immune responses. CONCLUSION Omega-6 PUFA status associated both positively and negatively with expression of specific immunology-related genes in PBMCs in healthy children. The results may suggest a nuanced role for omega-6 fatty acids in the interphase of lipids and inflammation, and warrants further examination in gene-environment studies and randomized controlled trials.
Collapse
Affiliation(s)
- Jacob J Christensen
- Department of Nutrition, University of Oslo, P.O. Box 1046 Blindern, 0317, Oslo, Norway.,The Lipid Clinic, Oslo University Hospital, Rikshospitalet, P.O. Box 4950 Nydalen, 0424, Oslo, Norway
| | - Siril S Bakke
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Stine M Ulven
- Department of Nutrition, University of Oslo, P.O. Box 1046 Blindern, 0317, Oslo, Norway
| | - Kjetil Retterstøl
- Department of Nutrition, University of Oslo, P.O. Box 1046 Blindern, 0317, Oslo, Norway.,The Lipid Clinic, Oslo University Hospital, Rikshospitalet, P.O. Box 4950 Nydalen, 0424, Oslo, Norway
| | - Tore Henriksen
- Faculty of Medicine, University of Oslo, P.O. Box 1046 Blindern, 0317, Oslo, Norway.,Department of Obstetrics, Oslo University Hospital, Rikshospitalet, P.O. Box 4950 Nydalen, 0424, Oslo, Norway
| | - Jens Bollerslev
- Faculty of Medicine, University of Oslo, P.O. Box 1046 Blindern, 0317, Oslo, Norway.,Section of Specialized Endocrinology, Oslo University Hospital, Rikshospitalet, P.O. Box 4950 Nydalen, 0424, Oslo, Norway
| | - Terje Espevik
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, University of Oslo, P.O. Box 1046 Blindern, 0317, Oslo, Norway.,Institute of Clinical Medicine, P.O. Box 1171 Blindern, 0318, Oslo, Norway.,K.G. Jebsen Inflammatory Research Center, P.O. Box 1171 Blindern, 0318, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, P.O. Box 4950 Nydalen, 0424, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, University of Oslo, P.O. Box 1046 Blindern, 0317, Oslo, Norway.,Institute of Clinical Medicine, P.O. Box 1171 Blindern, 0318, Oslo, Norway.,K.G. Jebsen Inflammatory Research Center, P.O. Box 1171 Blindern, 0318, Oslo, Norway
| | - Kirsten B Holven
- Department of Nutrition, University of Oslo, P.O. Box 1046 Blindern, 0317, Oslo, Norway.,Norwegian National Advisory Unit on Familial Hypercholesterolemia, Oslo University Hospital, Rikshospitalet, P.O. Box 4950 Nydalen, 0424, Oslo, Norway
| |
Collapse
|
816
|
Małkiewicz MA, Szarmach A, Sabisz A, Cubała WJ, Szurowska E, Winklewski PJ. Blood-brain barrier permeability and physical exercise. J Neuroinflammation 2019; 16:15. [PMID: 30678702 PMCID: PMC6345022 DOI: 10.1186/s12974-019-1403-x] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 01/09/2019] [Indexed: 12/20/2022] Open
Abstract
In this narrative review, a theoretical framework on the crosstalk between physical exercise and blood-brain barrier (BBB) permeability is presented. We discuss the influence of physical activity on the factors affecting BBB permeability such as systemic inflammation, the brain renin-angiotensin and noradrenergic systems, central autonomic function and the kynurenine pathway. The positive role of exercise in multiple sclerosis and Alzheimer’s disease is described. Finally, the potential role of conditioning as well as the effect of exercise on BBB tight junctions is outlined. There is a body of evidence that regular physical exercise diminishes BBB permeability as it reinforces antioxidative capacity, reduces oxidative stress and has anti-inflammatory effects. It improves endothelial function and might increase the density of brain capillaries. Thus, physical training can be emphasised as a component of prevention programs developed for patients to minimise the risk of the onset of neuroinflammatory diseases as well as an augmentation of existing treatment. Unfortunately, despite a sound theoretical background, it remains unclear as to whether exercise training is effective in modulating BBB permeability in several specific diseases. Further research is needed as the impact of exercise is yet to be fully elucidated.
Collapse
Affiliation(s)
- Marta A Małkiewicz
- Department of Human Physiology, Faculty of Health Sciences, Medical University of Gdansk, Tuwima Str. 15, 80-210, Gdansk, Poland.,Department of Psychiatry, Faculty of Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Arkadiusz Szarmach
- 2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| | - Agnieszka Sabisz
- 2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| | - Wiesław J Cubała
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Edyta Szurowska
- 2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| | - Paweł J Winklewski
- Department of Human Physiology, Faculty of Health Sciences, Medical University of Gdansk, Tuwima Str. 15, 80-210, Gdansk, Poland. .,2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland. .,Department of Clinical Anatomy and Physiology, Faculty of Health Sciences, Pomeranian University of Slupsk, Slupsk, Poland.
| |
Collapse
|
817
|
Biochemical Differences in Cerebrospinal Fluid between Secondary Progressive and Relapsing⁻Remitting Multiple Sclerosis. Cells 2019; 8:cells8020084. [PMID: 30678351 PMCID: PMC6406712 DOI: 10.3390/cells8020084] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/16/2019] [Accepted: 01/22/2019] [Indexed: 11/29/2022] Open
Abstract
To better understand the pathophysiological differences between secondary progressive multiple sclerosis (SPMS) and relapsing-remitting multiple sclerosis (RRMS), and to identify potential biomarkers of disease progression, we applied high-resolution mass spectrometry (HRMS) to investigate the metabolome of cerebrospinal fluid (CSF). The biochemical differences were determined using partial least squares discriminant analysis (PLS-DA) and connected to biochemical pathways as well as associated to clinical and radiological measures. Tryptophan metabolism was significantly altered, with perturbed levels of kynurenate, 5-hydroxytryptophan, 5-hydroxyindoleacetate, and N-acetylserotonin in SPMS patients compared with RRMS and controls. SPMS patients had altered kynurenine compared with RRMS patients, and altered indole-3-acetate compared with controls. Regarding the pyrimidine metabolism, SPMS patients had altered levels of uridine and deoxyuridine compared with RRMS and controls, and altered thymine and glutamine compared with RRMS patients. Metabolites from the pyrimidine metabolism were significantly associated with disability, disease activity and brain atrophy, making them of particular interest for understanding the disease mechanisms and as markers of disease progression. Overall, these findings are of importance for the characterization of the molecular pathogenesis of SPMS and support the hypothesis that the CSF metabolome may be used to explore changes that occur in the transition between the RRMS and SPMS pathologies.
Collapse
|
818
|
Xuan W, Pow R, Watfa N, Zheng Q, Surman AJ, Long DL, Cronin L. Stereoselective Assembly of Gigantic Chiral Molybdenum Blue Wheels Using Lanthanide Ions and Amino Acids. J Am Chem Soc 2019; 141:1242-1250. [PMID: 30495944 PMCID: PMC6351008 DOI: 10.1021/jacs.8b09750] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Indexed: 01/05/2023]
Abstract
The synthesis of chiral polyoxometalates (POMs) is a challenge because of the difficulty to induce the formation of intrinsically chiral metal-oxo frameworks. Herein we report the stereoselective synthesis of a series of gigantic chiral Mo Blue (MB) POM clusters 1-5 that are formed by exploiting the synergy between coordinating lanthanides ions as symmetry breakers to produce MBs with chiral frameworks decorated with amino acids ligands; these promote the selective formation of enantiopure MBs. All the compounds share the same framework archetype, based on {Mo124Ce4}, which forms an intrinsically chiral Δ or Λ configurations, controlled by the configurations of functionalized chiral amino acids. The chirality and stability of 1-5 in solution are confirmed by circular dichroism, 1H NMR, and electrospray ion mobility-mass spectrometry studies. In addition, the framework of the {Mo124Ce4} MB not only behaves as a host able to trap a chiral {Mo8} cluster that is not accessible by traditional synthesis but also promotes the transformation of tryptophan to kynurenine in situ. This work demonstrates the potential and applicability of our synthetic strategy to produce gigantic chiral POM clusters capable of host-guest chemistry and selective synthetic transformations.
Collapse
Affiliation(s)
- Weimin Xuan
- WestCHEM, School of Chemistry, The University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Robert Pow
- WestCHEM, School of Chemistry, The University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Nancy Watfa
- WestCHEM, School of Chemistry, The University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Qi Zheng
- WestCHEM, School of Chemistry, The University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Andrew J. Surman
- WestCHEM, School of Chemistry, The University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - De-Liang Long
- WestCHEM, School of Chemistry, The University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Leroy Cronin
- WestCHEM, School of Chemistry, The University of Glasgow, Glasgow G12 8QQ, United Kingdom
| |
Collapse
|
819
|
Chen Z, Leinisch F, Greco I, Zhang W, Shu N, Chuang CY, Lund MN, Davies MJ. Characterisation and quantification of protein oxidative modifications and amino acid racemisation in powdered infant milk formula. Free Radic Res 2019; 53:68-81. [DOI: 10.1080/10715762.2018.1554250] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Zhifei Chen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Fabian Leinisch
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ines Greco
- Department of Food Science, Faculty of Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Wei Zhang
- Department of Food Science, Faculty of Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nan Shu
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christine Y. Chuang
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marianne N. Lund
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Food Science, Faculty of Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael J. Davies
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
820
|
Abstract
The gut microbiota is a crucial actor in human physiology. Many of these effects are mediated by metabolites that are either produced by the microbes or derived from the transformation of environmental or host molecules. Among the array of metabolites at the interface between these microorganisms and the host is the essential aromatic amino acid tryptophan (Trp). In the gut, the three major Trp metabolism pathways leading to serotonin (5-hydroxytryptamine), kynurenine (Kyn), and indole derivatives are under the direct or indirect control of the microbiota. In this review, we gather the most recent advances concerning the central role of Trp metabolism in microbiota-host crosstalk in health and disease. Deciphering the complex equilibrium between these pathways will facilitate a better understanding of the pathogenesis of human diseases and open therapeutic opportunities.
Collapse
|
821
|
Lent-Schochet D, McLaughlin M, Ramakrishnan N, Jialal I. Exploratory metabolomics of metabolic syndrome: A status report. World J Diabetes 2019; 10:23-36. [PMID: 30697368 PMCID: PMC6347655 DOI: 10.4239/wjd.v10.i1.23] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 02/05/2023] Open
Abstract
Metabolic syndrome (MetS) is as a cluster of cardio-metabolic factors that greatly increase the risk of chronic diseases such as type II diabetes mellitus and atherosclerotic cardiovascular disease. In the United States, obesity, physical inactivity, aging, and genetics (to a minor extent) have arisen as risk factors for developing MetS. Although 35% of American adults suffer from MetS, its pathogenesis largely remains unknown. Worse, there is a lack of screening and optimum therapy for this disease. Researchers have consequently turned towards metabolomics to identify biomarkers to better understand MetS. The purpose of this review is to characterize various metabolites and their potential connections to MetS. Numerous studies have also characterized MetS as a disease of increased inflammation, and therefore this review also explores how metabolites play a role in various inflammatory pathways. Our review explores a broad range of metabolites including biogenic amines, branched chain amino acids, aromatic amines, phosphatidylcholines, as well as a variety of other molecules. We will explore their biochemical pathways and their potential role in serving as biomarkers.
Collapse
Affiliation(s)
- Daniella Lent-Schochet
- Metabolism and Clinical Pathology, College of Medicine, California Northstate University, Elk Grove, CA 95757, United States
| | - Matthew McLaughlin
- Metabolism and Clinical Pathology, College of Medicine, California Northstate University, Elk Grove, CA 95757, United States
| | - Neeraj Ramakrishnan
- Metabolism and Clinical Pathology, College of Medicine, California Northstate University, Elk Grove, CA 95757, United States
| | - Ishwarlal Jialal
- Metabolism and Clinical Pathology, College of Medicine, California Northstate University, Elk Grove, CA 95757, United States
- VA Medical Center, Mather CA 95655, United States
| |
Collapse
|
822
|
Solvang SEH, Nordrehaug JE, Tell GS, Nygård O, McCann A, Ueland PM, Midttun Ø, Meyer K, Vedeler CA, Aarsland D, Refsum H, Smith AD, Giil LM. The kynurenine pathway and cognitive performance in community-dwelling older adults. The Hordaland Health Study. Brain Behav Immun 2019; 75:155-162. [PMID: 30675874 DOI: 10.1016/j.bbi.2018.10.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/17/2018] [Accepted: 10/23/2018] [Indexed: 01/16/2023] Open
Abstract
INTRODUCTION Tryptophan, its downstream metabolites in the kynurenine pathway and neopterin have been associated with inflammation and dementia. We aimed to study the associations between plasma levels of these metabolites and cognitive function in community-dwelling, older adults. METHODS This cross-sectional study included 2174 participants aged 70-72 years of the community-based Hordaland Health Study. Tryptophan, kynurenine, neopterin and eight downstream kynurenines were measured in plasma. Kendrick Object Learning Test (KOLT), Digit Symbol Test (DST) and the Controlled Oral Word Association Test (COWAT) were all outcomes in standardized Zellner's regression. The Wald test of a composite linear hypothesis of an association with each metabolite was adjusted by the Bonferroni method. Age, body mass index, C-reactive protein, depressive symptoms, diabetes, education, glomerular filtration rate, hypertension, previous myocardial infarction, prior stroke, pyridoxal 5'phosphate, sex and smoking were considered as potential confounders. RESULTS Higher levels of the kynurenine-to-tryptophan ratio (KTR) and neopterin were significantly associated with poorer, overall cognitive performance (p < 0.002). Specifically, KTR was negatively associated with KOLT (β -0.08, p = 0.001) and COWAT (β -0.08, p = 0.001), but not with DST (β -0.03, p = 0.160). This pattern was also seen for neopterin (KOLT: β -0.07; p = 0.001; COWAT: β -0.06, p = 0.010; DST: β -0.01, p = 0.800). The associations were not confounded by the examined variables. No significant associations were found between the eight downstream kynurenines and cognition. CONCLUSION Higher KTR and neopterin levels, biomarkers of cellular immune activation, were associated with reduced cognitive performance, implying an association between the innate immune system, memory, and language.
Collapse
Affiliation(s)
- Stein-Erik Hafstad Solvang
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway; Institute of Clinical Science, University of Bergen, Norway.
| | - Jan Erik Nordrehaug
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway; Institute of Clinical Science, University of Bergen, Norway
| | - Grethe S Tell
- Department of Global Public Health and Primary Care, University of Bergen, Norway; Division of Mental and Physical Health, Norwegian Institute of Public Health, Bergen, Norway
| | - Ottar Nygård
- Institute of Clinical Science, University of Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | | | | | | | | | | | - Dag Aarsland
- Department of Old Age Psychiatry, King's College University, London, UK
| | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway; Department of Pharmacology, University of Oxford, UK
| | - A David Smith
- Department of Pharmacology, University of Oxford, UK
| | - Lasse Melvaer Giil
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway; Institute of Clinical Science, University of Bergen, Norway
| |
Collapse
|
823
|
Wang M, Xie Z, Li L, Chen Y, Li Y, Wang Y, Lu B, Zhang S, Ma F, Ma C, Lin L, Liao Q. Supplementation with compound polysaccharides contributes to the development and metabolic activity of young rat intestinal microbiota. Food Funct 2019; 10:2658-2675. [DOI: 10.1039/c8fo02565g] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Compound polysaccharides may be used as a functional food to modulate the composition and metabolism of gut microbiota, and to help maintain the health of the intestinal microecosystem.
Collapse
Affiliation(s)
- Mengxia Wang
- School of Pharmaceutical Sciences
- Guangzhou University of Chinese Medicine
- Guangzhou
- P. R. China
| | - Zhiyong Xie
- School of Pharmaceutical Sciences (Shenzhen)
- Sun Yat-sen University
- Guangzhou
- P. R. China
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation
| | - Lin Li
- School of Pharmaceutical Sciences
- Guangzhou University of Chinese Medicine
- Guangzhou
- P. R. China
| | - Yongxiong Chen
- School of Pharmaceutical Sciences
- Guangzhou University of Chinese Medicine
- Guangzhou
- P. R. China
| | - Yuan Li
- School of Pharmaceutical Sciences
- Guangzhou University of Chinese Medicine
- Guangzhou
- P. R. China
| | | | - Biyu Lu
- School of Pharmaceutical Sciences
- Guangzhou University of Chinese Medicine
- Guangzhou
- P. R. China
| | - Shaobao Zhang
- School of Pharmaceutical Sciences (Shenzhen)
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Fangli Ma
- Infinitus (China) Company Ltd
- Guangzhou
- China
| | | | - Lei Lin
- School of Pharmaceutical Sciences (Shenzhen)
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Qiongfeng Liao
- School of Pharmaceutical Sciences
- Guangzhou University of Chinese Medicine
- Guangzhou
- P. R. China
| |
Collapse
|
824
|
Zhang HL, Zhang AH, Miao JH, Sun H, Yan GL, Wu FF, Wang XJ. Targeting regulation of tryptophan metabolism for colorectal cancer therapy: a systematic review. RSC Adv 2019; 9:3072-3080. [PMID: 35518968 PMCID: PMC9060217 DOI: 10.1039/c8ra08520j] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/23/2018] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most malignant cancers resulting from abnormal metabolism alterations. As one of the essential amino acids, tryptophan has a variety of physiological functions, closely related to regulation of immune system, central nervous system, gastrointestinal nervous system and intestinal microflora. Colorectal cancer, a type of high-grade malignancy disease, stems from a variety of factors and often accompanies inflammatory reactions, dysbacteriosis, and metabolic disorders. Colorectal cancer accompanies inflammation and imbalance of intestinal microbiota and affects tryptophan metabolism. It is known that metabolites, rate-limiting enzymes, and ARH in tryptophan metabolism are associated with the development of CRC. Specifically, IDO1 may be a potential therapeutic target in colorectal cancer treatment. Furthermore, the reduction of tryptophan amount is proportional to the poor quality of life for colorectal cancer patients. This paper aims to discuss the role of tryptophan metabolism in a normal organism and investigate the relationship between this amino acid and colorectal cancer. This study is expected to provide theoretical support for research related to targeted therapy for colorectal cancer. Furthermore, strategies that modify tryptophan metabolism, effectively inhibiting tumor progression, may be more effective for CRC treatment. Colorectal cancer (CRC) is one of the most malignant cancers resulting from abnormal metabolism alterations.![]()
Collapse
Affiliation(s)
- Hong-lian Zhang
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials
- Guangxi Botanical Garden of Medicinal Plant
- Nanning
- China
- Sino-America Chinmedomics Technology Collaboration Center
| | - Ai-hua Zhang
- Sino-America Chinmedomics Technology Collaboration Center
- National TCM Key Laboratory of Serum Pharmacochemistry
- Chinmedomics Research Center of State Administration of TCM
- Laboratory of Metabolomics
- Department of Pharmaceutical Analysis
| | - Jian-hua Miao
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials
- Guangxi Botanical Garden of Medicinal Plant
- Nanning
- China
| | - Hui Sun
- Sino-America Chinmedomics Technology Collaboration Center
- National TCM Key Laboratory of Serum Pharmacochemistry
- Chinmedomics Research Center of State Administration of TCM
- Laboratory of Metabolomics
- Department of Pharmaceutical Analysis
| | - Guang-li Yan
- Sino-America Chinmedomics Technology Collaboration Center
- National TCM Key Laboratory of Serum Pharmacochemistry
- Chinmedomics Research Center of State Administration of TCM
- Laboratory of Metabolomics
- Department of Pharmaceutical Analysis
| | - Fang-fang Wu
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials
- Guangxi Botanical Garden of Medicinal Plant
- Nanning
- China
- Sino-America Chinmedomics Technology Collaboration Center
| | - Xi-jun Wang
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials
- Guangxi Botanical Garden of Medicinal Plant
- Nanning
- China
- Sino-America Chinmedomics Technology Collaboration Center
| |
Collapse
|
825
|
Hahn D, Stokes CS, Kaiser R, Meyer MR, Lammert F, Gruenhage F. Antidepressant effects of direct-acting antivirals against hepatitis C virus-Results from a pilot study. Eur J Clin Invest 2018; 48:e13024. [PMID: 30175442 DOI: 10.1111/eci.13024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 07/30/2018] [Accepted: 08/29/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIMS The new direct-acting antiviral agents (DAA) have revolutionized the treatment of patients with chronic hepatitis C virus (HCV) infection. This study investigates to which extent DAA affect fatigue and mood and, if so, whether this results from changes to tryptophan (TRP) metabolism, as reflected by two critical biosynthetic pathways, serotonin (SRT) generation from TRP and TRP degradation through kynurenines (KYN) via indoleamine 2,3-dioxygenase (IDO). METHODS This study assessed 24 patients with chronic HCV infection, before (T1), during (T2: at 4 weeks) and 12 weeks post-treatment with DAA (T3) with respect to viral load, fatigue and depressive symptoms (BDI-II questionnaire), physical activity (actigraph) and plasma serotonin-tryptophan metabolites (LC/MS). The KYN:TRP ratio reflected IDO activity. RESULTS All participants achieved sustained virological response (SVR12) with DAA treatment (79% sofosbuvir-based). Fatigue (scores at T1:0.83 ± 0.70, T2:0.48 ± 0.70, T3:0.30 ± 0.50; P = 0.023) and depressive symptoms (scores at T1:9.8 ± 10.2, T2:6.0 ± 7.3, T3:5.0 ± 7.6; P = 0.005) improved significantly on therapy, whereas no changes were noted in five untreated controls. TRP plasma concentrations markedly decreased (T1:306 ± 179 mg/L, T2:283 ± 84 mg/L), whereas 5-HTP levels increased (T1:0.08 ± 0.01 mg/L, T2:0.10 ± 0.06 mg/L). KYN concentrations (T1:2.4 ± 2.0 mg/L, T2:3.7 ± 1.4 mg/L, P = 0.003) increased significantly during treatment, as did IDO activity (T1:0.008 ± 0.006 mg/L, T2:0.014 ± 0.004 mg/L; P < 0.001). CONCLUSIONS In this study, DAA exert positive and persistent effects on both fatigue and mood in patients with chronic HCV infection. These extrahepatic benefits are, at least in part, related to the modulation of TRP metabolism. The robust elevation of KYN concentrations challenges the current paradigm of low KYN levels as prerequisite for mental health.
Collapse
Affiliation(s)
- Daphne Hahn
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Caroline S Stokes
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Ralf Kaiser
- Department of Medicine V, Saarland University Medical Center, Saarland University Homburg, Homburg, Germany
| | - Markus R Meyer
- Department of Experimental and Clinical Toxicology, Saarland University, Homburg, Germany
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Frank Gruenhage
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany.,Department of Internal Medicine, RKN-Clinics, St. Elisabeth Hospital, Grevenbroich, Germany
| |
Collapse
|
826
|
Metabolite and lipoprotein responses and prediction of weight gain during breast cancer treatment. Br J Cancer 2018; 119:1144-1154. [PMID: 30401977 PMCID: PMC6220113 DOI: 10.1038/s41416-018-0211-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 07/06/2018] [Accepted: 07/10/2018] [Indexed: 12/18/2022] Open
Abstract
Background Breast cancer treatment has metabolic side effects, potentially affecting risk of cardiovascular disease (CVD) and recurrence. We aimed to compare alterations in serum metabolites and lipoproteins during treatment between recipients and non-recipients of chemotherapy, and describe metabolite profiles associated with treatment-related weight gain. Methods This pilot study includes 60 stage I/II breast cancer patients who underwent surgery and were treated according to national guidelines. Serum sampled pre-surgery and after 6 and 12 months was analysed by MR spectroscopy and mass spectrometry. In all, 170 metabolites and 105 lipoprotein subfractions were quantified. Results The metabolite and lipoprotein profiles of chemotherapy recipients and non-recipients changed significantly 6 months after surgery (p < 0.001). Kynurenine, the lipid signal at 1.55–1.60 ppm, ADMA, 2 phosphatidylcholines (PC aa C38:3, PC ae C42:1), alpha-aminoadipic acid, hexoses and sphingolipids were increased in chemotherapy recipients after 6 months. VLDL and small dense LDL increased after 6 months, while HDL decreased, with triglyceride enrichment in HDL and LDL. At baseline, weight gainers had less acylcarnitines, phosphatidylcholines, lyso-phosphatidylcholines and sphingolipids, and showed an inflammatory lipid profile. Conclusion Chemotherapy recipients exhibit metabolic changes associated with inflammation, altered immune response and increased risk of CVD. Altered lipid metabolism may predispose for treatment-related weight gain.
Collapse
|
827
|
Dadvar S, Ferreira DMS, Cervenka I, Ruas JL. The weight of nutrients: kynurenine metabolites in obesity and exercise. J Intern Med 2018; 284:519-533. [PMID: 30141532 DOI: 10.1111/joim.12830] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Obesity ultimately results from an imbalance between energy intake and expenditure. However, in addition to their bioenergetic value, nutrients and their metabolites can function as important signalling molecules in energy homeostasis. Indeed, macronutrients and their metabolites can be direct regulators of metabolism through their actions on different organs. In turn, target organs can decide to use, store or transform the incoming nutrients depending on their physiological context and in coordination with other cell types. Tryptophan-kynurenine metabolites are an example of a family of compounds that can serve as systemic integrators of energy metabolism by signalling to different cell types. These include adipocytes, immune cells and muscle fibres, in addition to the well-known effects of kynurenine metabolites on the central nervous system. In the context of energy metabolism, several of the effects elicited by kynurenic acid are mediated by the G-protein-coupled receptor, GPR35. As GPR35 is expressed in tissues such as the adipose tissue, immune cells and the gastrointestinal tract, this receptor could be a potential therapeutic target for the treatment of obesity, diabetes and other metabolic diseases. In addition, metabolic disorders often coincide with states of chronic inflammation, which further highlights GPR35 as an integration node in conditions where inflammation skews metabolism. Defining the molecular interplay between different tissues in the regulation of energy homeostasis can help us understand interindividual variability in the response to nutrient intake and develop safe and efficient therapies to fight obesity and metabolic disease.
Collapse
Affiliation(s)
- S Dadvar
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - D M S Ferreira
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - I Cervenka
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - J L Ruas
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| |
Collapse
|
828
|
Moaddel R, Shardell M, Khadeer M, Lovett J, Kadriu B, Ravichandran S, Morris PJ, Yuan P, Thomas CJ, Gould TD, Ferrucci L, Zarate CA. Plasma metabolomic profiling of a ketamine and placebo crossover trial of major depressive disorder and healthy control subjects. Psychopharmacology (Berl) 2018; 235:3017-3030. [PMID: 30116859 PMCID: PMC6193489 DOI: 10.1007/s00213-018-4992-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 07/30/2018] [Indexed: 12/28/2022]
Abstract
(R,S)-Ketamine produces rapid, robust, and sustained antidepressant effects in major depressive disorder. Specifically, its pharmacological efficacy in treatment refractory depression is considered a major breakthrough in the field. However, the mechanism of action of ketamine's rapid effect remains to be determined. In order to identify pathways that are responsible for ketamine's effect, a targeted metabolomic approach was carried out using a double-blind, placebo-controlled crossover design, with infusion order randomized with medication-free patients with treatment-resistant major depressive disorder (29 subjects) and healthy controls (25 subjects). The metabolomic profile of these subjects was characterized at multiple time points, and a comprehensive analysis was investigated between the following: MDD and healthy controls, treatment and placebo in both groups and the corresponding response to ketamine treatment. Ketamine treatment resulted in a general increase in circulating sphingomyelins, levels which were not correlated with response. Ketamine response resulted in more pronounced effects in the kynurenine pathway and the arginine pathway at 4 h post-infusion, where a larger decrease in circulating kynurenine levels and a larger increase in the bioavailability of arginine were observed in responders to ketamine treatment, suggesting possible mechanisms for response to ketamine treatment.
Collapse
Affiliation(s)
- Ruin Moaddel
- Biomedical Research Center, Intramural Research Program, National Institute on Aging, National Institutes, Bethesda, MD, USA.
| | - Michelle Shardell
- Biomedical Research Center, Intramural Research Program, National Institute on Aging, National Institutes, Bethesda, MD, USA
| | - Mohammed Khadeer
- Biomedical Research Center, Intramural Research Program, National Institute on Aging, National Institutes, Bethesda, MD, USA
| | - Jacqueline Lovett
- Biomedical Research Center, Intramural Research Program, National Institute on Aging, National Institutes, Bethesda, MD, USA
| | - Bashkim Kadriu
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Sarangan Ravichandran
- Advanced Biomedical and Computational Sciences, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Leidos Biomedical Research Inc, Fredrick, MD 21702, USA
| | - Patrick J. Morris
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Intramural Research Program, National Institutes of Health, Rockville, MD, USA
| | - Peixiong Yuan
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Craig J. Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Intramural Research Program, National Institutes of Health, Rockville, MD, USA
| | - Todd D. Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Luigi Ferrucci
- Biomedical Research Center, Intramural Research Program, National Institute on Aging, National Institutes, Bethesda, MD, USA
| | - Carlos A. Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
829
|
Metcalfe AJ, Koliamitra C, Javelle F, Bloch W, Zimmer P. Acute and chronic effects of exercise on the kynurenine pathway in humans – A brief review and future perspectives. Physiol Behav 2018; 194:583-587. [DOI: 10.1016/j.physbeh.2018.07.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/12/2018] [Accepted: 07/18/2018] [Indexed: 01/17/2023]
|
830
|
Duval N, Vacano GN, Patterson D. Rapamycin Treatment Ameliorates Age-Related Accumulation of Toxic Metabolic Intermediates in Brains of the Ts65Dn Mouse Model of Down Syndrome and Aging. Front Aging Neurosci 2018; 10:263. [PMID: 30237765 PMCID: PMC6135881 DOI: 10.3389/fnagi.2018.00263] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/14/2018] [Indexed: 01/22/2023] Open
Abstract
Down syndrome (DS), caused by trisomy of chromosome 21, is the most common genetic cause of intellectual disability. Individuals with DS exhibit changes in neurochemistry and neuroanatomy that worsen with age, neurological delay in learning and memory, and predisposition to Alzheimer's disease. The Ts65Dn mouse is the best characterized model of DS and has many features reminiscent of DS, including developmental anomalies and age-related neurodegeneration. The mouse carries a partial triplication of mouse chromosome 16 containing roughly 100 genes syntenic to human chromosome 21 genes. We hypothesized that there would be differences in brain metabolites with trisomy and age, and that long-term treatment with rapamycin, mechanistic target of rapamycin (mTOR) inhibitor and immunosuppressant, would correct these differences. Using HPLC coupled with electrochemical detection, we identified differences in levels of metabolites involved in dopaminergic, serotonergic, and kynurenine pathways in trisomic mice that are exacerbated with age. These include homovanillic acid, norepinephrine, and kynurenine. In addition, we demonstrate that prolonged treatment with rapamycin reduces accumulation of toxic metabolites (such as 6-hydroxymelatonin and 3-hydroxykynurenine) in aged mice.
Collapse
Affiliation(s)
- Nathan Duval
- Department of Biological Sciences, Knoebel Institute for Healthy Aging, and Eleanor Roosevelt Institute, University of Denver, Denver, CO, United States
| | | | | |
Collapse
|
831
|
4-Chloro-l-kynurenine as fluorescent amino acid in natural peptides. Amino Acids 2018; 50:1697-1705. [DOI: 10.1007/s00726-018-2642-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/27/2018] [Indexed: 11/26/2022]
|
832
|
Abstract
Typical and atypical antipsychotics are the first-line treatments for schizophrenia, but these classes of drugs are not universally effective, and they can have serious side effects that impact compliance. Antipsychotic drugs generally target the dopamine pathways with some variation. As research of schizophrenia pathophysiology has shifted away from a strictly dopamine-centric focus, the development of new pharmacotherapies has waned. A field of inquiry with centuries-old roots is gaining traction in psychiatric research circles and may represent a new frontier for drug discovery in schizophrenia. At the forefront of this investigative effort is the immune system and its many components, pathways and phenotypes, which are now known to actively engage the brain. Studies in schizophrenia reveal an intricate association of environmentally-driven immune activation in concert with a disrupted genetic template. A consistent conduit through this gene-environmental milieu is the gut-brain axis, which when dysregulated can generate pathological autoimmunity. In this review, we present epidemiological and biochemical evidence in support of an autoimmune component in schizophrenia and depict gut processes and a dysbiotic microbiome as a source and perpetuator of autoimmune dysfunction in the brain. Within this framework, we review the role of infectious agents, inflammation, gut dysbioses and autoantibody propagation on CNS pathologies such as neurotransmitter receptor hypofunction and complement pathway-mediated synaptic pruning. We then review the new pharmacotherapeutic horizon and novel agents directed to impact these pathological conditions. At the core of this discourse is the understanding that schizophrenia is etiologically and pathophysiologically heterogeneous and thus its treatment requires individualized attention with disease state variants diagnosed with objective biomarkers.
Collapse
Affiliation(s)
| | | | - Robert H Yolken
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
833
|
Arnone D, Saraykar S, Salem H, Teixeira AL, Dantzer R, Selvaraj S. Role of Kynurenine pathway and its metabolites in mood disorders: A systematic review and meta-analysis of clinical studies. Neurosci Biobehav Rev 2018; 92:477-485. [PMID: 29940237 PMCID: PMC6686193 DOI: 10.1016/j.neubiorev.2018.05.031] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 12/16/2022]
Abstract
Activation of the kynurenine pathway is one of the described mechanisms by which inflammation can induce depression. It involves multiple pathways including interference with the bioavailability of tryptophan central to the synthesis of the neurotransmitter serotonin. In this systematic review, we examine the relationship between kynurenine metabolites (kynurenine, kynurenic acid, tryptophan, quinolinic acid, the ratio of kynurenine and tryptophan) and mood disorders by conducting a meta-analysis. Fifty-six studies were identified, 21 met inclusion criteria and 14 were deemed suitable (9 investigating unipolar depression and 5 bipolar disorder). We found decreased levels of kynurenine in unipolar major depression vs. healthy controls but studies were significantly heterogeneous in nature. No significant differences were found in tryptophan levels or kynurenine/tryptophan ratios. Kynurenine metabolites are likely to play a role in major depression but an exact etiological role in mood disorder seem complex and requires further research.
Collapse
Affiliation(s)
- Danilo Arnone
- Department of Psychological Medicine, Centre for Affective Disorders, Institute of Psychiatry, Psychology & Neuroscience, King's College London, and South London and Maudsley NHS Foundation Trust, London, UK
| | - Smita Saraykar
- The University of Texas Health Science Center at Houston, Department of Psychiatry and Behavioral Sciences, 1941 East Rd., Houston, TX 77054, United States
| | - Haitham Salem
- The University of Texas Health Science Center at Houston, Department of Psychiatry and Behavioral Sciences, 1941 East Rd., Houston, TX 77054, United States
| | - Antonio L Teixeira
- The University of Texas Health Science Center at Houston, Department of Psychiatry and Behavioral Sciences, 1941 East Rd., Houston, TX 77054, United States
| | - Robert Dantzer
- The University of Texas MD Anderson Cancer Center, Department of Symptom Research, 1515 Holcombe Blvd, Unit # 1450, Houston, TX 77030, United States
| | - Sudhakar Selvaraj
- The University of Texas Health Science Center at Houston, Department of Psychiatry and Behavioral Sciences, 1941 East Rd., Houston, TX 77054, United States; The University of Texas MD Anderson Cancer Center, Department of Symptom Research, 1515 Holcombe Blvd, Unit # 1450, Houston, TX 77030, United States.
| |
Collapse
|
834
|
Sorgdrager FJH, Werumeus Buning J, Bos EH, Van Beek AP, Kema IP. Hydrocortisone Affects Fatigue and Physical Functioning Through Metabolism of Tryptophan: A Randomized Controlled Trial. J Clin Endocrinol Metab 2018; 103:3411-3419. [PMID: 29982583 DOI: 10.1210/jc.2018-00582] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/27/2018] [Indexed: 12/19/2022]
Abstract
CONTEXT Hydrocortisone (HC) treatment influences health-related quality of life (HRQOL) in secondary adrenal insufficiency (AI). Glucocorticoids regulate tryptophan metabolism through the kynurenine pathway, which modulates mood and energy homeostasis. OBJECTIVE This study investigated whether tryptophan metabolism mediated the effect of HC dose on HRQOL in patients with secondary AI. DESIGN, SETTING, AND PATIENTS Forty-seven patients with secondary AI participated in this double-blind randomized controlled cross-over trial in the University Medical Center Groningen. INTERVENTION Patients were treated for two 10-week periods with a daily HC dose of 0.2 to 0.3 mg/kg and 0.4 to 0.6 mg/kg body weight, respectively. MAIN OUTCOME MEASURES Diary data and questionnaires were used to assess HRQOL. Tryptophan, kynurenine and 3-hydroxykynurenine were measured in serum and dialyzed plasma and the kynurenine-to-tryptophan ratio (Kyn/Trp ratio) ratio was calculated. RESULTS A higher dose HC was associated with increased levels of tryptophan (95% CI for mean difference 0.37 to 12.5, P = 0.038), reduced levels of kynurenine (95% CI, -0.49 to -0.10, P = 0.004) and 3-hydroxykynurenine (95% CI, -10.6 to -2.35, P = 0.003), and a reduced Kyn/Trp ratio (95% CI, -0.84 to -0.50, P < 0.001). The Kyn/Trp ratio mediated the effect of a higher dose HC on fatigue (P = 0.041) and physical functioning (P = 0.005). CONCLUSION Metabolism of tryptophan through the kynurenine pathway is reduced after a 10-week treatment with a higher dose HC and plays a role in the effect of HC on fatigue and physical functioning in patients with secondary AI.
Collapse
Affiliation(s)
- Freek J H Sorgdrager
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, RB Groningen, Netherlands
| | - Jorien Werumeus Buning
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, RB Groningen, Netherlands
| | - Elske H Bos
- Interdisciplinary Center Psychopathology and Emotion Regulation, University of Groningen, University Medical Center Groningen, RB Groningen, Netherlands
- Department of Developmental Psychology, University of Groningen, TS Groningen, Netherlands
| | - André P Van Beek
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, RB Groningen, Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, RB Groningen, Netherlands
| |
Collapse
|
835
|
Gutierrez DB, Gant-Branum RL, Romer CE, Farrow MA, Allen JL, Dahal N, Nei YW, Codreanu SG, Jordan AT, Palmer LD, Sherrod SD, McLean JA, Skaar EP, Norris JL, Caprioli RM. An Integrated, High-Throughput Strategy for Multiomic Systems Level Analysis. J Proteome Res 2018; 17:3396-3408. [PMID: 30114907 DOI: 10.1021/acs.jproteome.8b00302] [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] [Indexed: 12/16/2022]
Abstract
Proteomics, metabolomics, and transcriptomics generate comprehensive data sets, and current biocomputational capabilities allow their efficient integration for systems biology analysis. Published multiomics studies cover methodological advances as well as applications to biological questions. However, few studies have focused on the development of a high-throughput, unified sample preparation approach to complement high-throughput omic analytics. This report details the automation, benchmarking, and application of a strategy for transcriptomic, proteomic, and metabolomic analyses from a common sample. The approach, sample preparation for multi-omics technologies (SPOT), provides equivalent performance to typical individual omic preparation methods but greatly enhances throughput and minimizes the resources required for multiomic experiments. SPOT was applied to a multiomics time course experiment for zinc-treated HL-60 cells. The data reveal Zn effects on NRF2 antioxidant and NFkappaB signaling. High-throughput approaches such as these are critical for the acquisition of temporally resolved, multicondition, large multiomic data sets such as those necessary to assess complex clinical and biological concerns. Ultimately, this type of approach will provide an expanded understanding of challenging scientific questions across many fields.
Collapse
|
836
|
De novo NAD + biosynthetic impairment in acute kidney injury in humans. Nat Med 2018; 24:1351-1359. [PMID: 30127395 PMCID: PMC6129212 DOI: 10.1038/s41591-018-0138-z] [Citation(s) in RCA: 264] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/03/2018] [Indexed: 12/21/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) extends longevity in experimental organisms, raising interest in its impact on human health. De novo NAD+ biosynthesis from tryptophan is evolutionarily conserved yet considered supplanted among higher species by biosynthesis from nicotinamide (Nam). Here we show that a bottleneck enzyme in de novo biosynthesis, quinolinate phosphoribosyltransferase (QPRT), defends renal NAD+ and mediates resistance to acute kidney injury (AKI). Following murine AKI, renal NAD+ fell, quinolinate rose, and QPRT declined. QPRT+/− mice exhibited higher quinolinate, lower NAD+, and higher AKI susceptibility. Metabolomics proposed elevated urinary quinolinate/tryptophan (uQ:T) as an indicator of reduced QPRT. Elevated uQ:T predicted AKI and other adverse outcomes in critically ill patients. A Phase 1 placebo-controlled study of oral Nam demonstrated dose-related increase in circulating NAD+ metabolites. Nam was well-tolerated and was associated with less AKI. Impaired NAD+ biosynthesis may therefore be a feature of high-risk hospitalizations for which NAD+ augmentation could be beneficial.
Collapse
|
837
|
Liang H, Dai Z, Liu N, Ji Y, Chen J, Zhang Y, Yang Y, Li J, Wu Z, Wu G. Dietary L-Tryptophan Modulates the Structural and Functional Composition of the Intestinal Microbiome in Weaned Piglets. Front Microbiol 2018; 9:1736. [PMID: 30131777 PMCID: PMC6090026 DOI: 10.3389/fmicb.2018.01736] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 07/11/2018] [Indexed: 12/29/2022] Open
Abstract
Background: Intestinal microbiota plays an important role in regulating metabolism, physiology, and immune response of the host. L-Tryptophan (Trp) are metabolized by several genera of bacteria. It remains largely unknown whether Trp can regulate the composition and diversity of the intestinal microbiota and contribute to intestinal homeostasis. Methods: A total of 126 weaning piglets were fed a corn- and soybean meal-based diet supplemented with 0, 0.2, or 0.4% Trp for 4 weeks. The intestinal microbiota was measured by using bacterial 16S rRNA gene-based high-throughput sequencing methods. Metabolites of Trp and short-chain fatty acids (SCFAs) in the hindgut were determined by high-performance liquid chromatography and gas chromatography, respectively. The mRNA levels for aromatic hydrocarbon receptor (AhR), tumor necrotic factor-α (TNF-α), interleukin-8 (IL-8), and protein abundances of tight junction proteins were determined. Results: Compared with the control group, Trp supplementation enhanced piglet growth performance and markedly altered the intestinal microbial composition as evidenced by enhanced alpha and beta diversity in the microbiome (P < 0.05). The abundances of Prevotella, Roseburia, and Succinivibrio genera were enriched, but those of Clostridium sensu stricto and Clostridium XI, opportunistic pathogens, were decreased with dietary Trp supplementation. Analysis of metabolic pathways indicated enhanced indole alkaloid biosynthesis and Trp metabolism, which was validated by elevated concentrations of 3-indoleacetic acid and indole in the intestinal contents of Trp-supplemented piglets (P < 0.05). These changes in Trp metabolites were correlated with activation of AhR and cytochrome p4501 A1 (CYP1A1) in cecum and colonic tissues, and with a decrease in the intestinal mucosal IL-8 mRNA level. Moreover, the protein abundances for zonula occluden (ZO)-1 and occludin were upregulated by Trp supplementation in colonic tissues. Conclusion: Dietary Trp supplementation altered intestinal microbial composition and diversity, improved intestinal mucosal barrier function, activated AhR signaling, and downregulated expression of inflammatory cytokines in the large intestine of weaned piglets. These results indicate a crosstalk between dietary Trp and intestine in nutrition, microbial metabolism, and mucosal immunity.
Collapse
Affiliation(s)
- Haiwei Liang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Ning Liu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Yun Ji
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Jingqing Chen
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Yunchang Zhang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Ju Li
- Henan Yinfa Animal Husbandry Co., Xinzheng, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Guoyao Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China.,Department of Animal Science, Texas A&M University, College Station, TX, United States
| |
Collapse
|
838
|
Reversal of indoleamine 2,3-dioxygenase-mediated cancer immune suppression by systemic kynurenine depletion with a therapeutic enzyme. Nat Biotechnol 2018; 36:758-764. [PMID: 30010674 PMCID: PMC6078800 DOI: 10.1038/nbt.4180] [Citation(s) in RCA: 230] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 06/01/2018] [Indexed: 12/25/2022]
Abstract
Increased tryptophan (Trp) catabolism in the tumor microenvironment (TME) can mediate immune suppression by upregulation of interferon (IFN)-γ-inducible indoleamine 2,3-dioxygenase (IDO1) and/or ectopic expression of the predominantly liver-restricted enzyme tryptophan 2,3-dioxygenase (TDO). Whether these effects are due to Trp depletion in the TME or mediated by the accumulation of the IDO1 and/or TDO (hereafter referred to as IDO1/TDO) product kynurenine (Kyn) remains controversial. Here we show that administration of a pharmacologically optimized enzyme (PEGylated kynureninase; hereafter referred to as PEG-KYNase) that degrades Kyn into immunologically inert, nontoxic and readily cleared metabolites inhibits tumor growth. Enzyme treatment was associated with a marked increase in the tumor infiltration and proliferation of polyfunctional CD8+ lymphocytes. We show that PEG-KYNase administration had substantial therapeutic effects when combined with approved checkpoint inhibitors or with a cancer vaccine for the treatment of large B16-F10 melanoma, 4T1 breast carcinoma or CT26 colon carcinoma tumors. PEG-KYNase mediated prolonged depletion of Kyn in the TME and reversed the modulatory effects of IDO1/TDO upregulation in the TME.
Collapse
|
839
|
Popov DV. Adaptation of Skeletal Muscles to Contractile Activity of Varying Duration and Intensity: The Role of PGC-1α. BIOCHEMISTRY (MOSCOW) 2018; 83:613-628. [DOI: 10.1134/s0006297918060019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
840
|
Early Maternal Deprivation Induces Microglial Activation, Alters Glial Fibrillary Acidic Protein Immunoreactivity and Indoleamine 2,3-Dioxygenase during the Development of Offspring Rats. Mol Neurobiol 2018; 56:1096-1108. [PMID: 29873040 DOI: 10.1007/s12035-018-1161-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 05/29/2018] [Indexed: 02/07/2023]
Abstract
Maternal deprivation (MD) induces behavioral changes and impacts brain circuits that could be associated with the pathophysiology of depression. This study investigated the markers of microglia and astrocyte activation as well as indoleamine 2,3-dioxygenase (IDO) expression in developmental programming after early life MD (on postnatal days (PNDs) 20, 30, 40, and 60). On PND 60, the rats that were subjected to MD displayed depressive-like behavior. On PND 10, it was found that there was a decrease in the level of glial fibrillary acidic protein (GFAP) immunopositive cells, a decrease in the level of IDO expression, and an increase in the level of Iba-1 (microglial marker) in the hippocampus of rats that were subjected to MD. On PND 20, levels of GFAP were also found to have decreased in the hippocampus, and there was an increase in the level of Iba-1 in the hippocampus. AIF-1 (microglial marker) expression was observed in the PFC following MD. On PND 30, the levels of Iba-1 remained elevated. On PND 40, the levels of GFAP were found to have increased in the hippocampus of rats that were subjected to MD. On PND 60, the levels of GFAP and AIF-1 remained elevated following MD. These results suggest that early life stress induces negative developmental programming in rats, as demonstrated by depressive-like behavior in adult life. Moreover, MD increases microglial activation in both early and late developmental phases. The levels of GFAP and IDO decreased in the early stages but were found to be higher in later developmental periods. These findings suggest that MD could differentially affect the expression of the IDO enzyme, astrocytes, and microglial activation depending on the neurodevelopmental period. The onset of an inflammatory state from resident brain cells could be associated with the activation of the kynurenine pathway and the development of depressive behavior in adulthood.
Collapse
|
841
|
Cao B, Wang D, Brietzke E, McIntyre RS, Pan Z, Cha D, Rosenblat JD, Zuckerman H, Liu Y, Xie Q, Wang J. Characterizing amino-acid biosignatures amongst individuals with schizophrenia: a case-control study. Amino Acids 2018; 50:1013-1023. [PMID: 29796929 DOI: 10.1007/s00726-018-2579-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 04/27/2018] [Indexed: 01/25/2023]
Abstract
Amino acids and derivatives participate in the biosynthesis and downstream effects of numerous neurotransmitters. Variations in specific amino acids have been implicated in the pathophysiology of schizophrenia. Herein, we sought to compare levels of amino acids and derivatives between subjects with schizophrenia and healthy controls (HC). Two hundred and eight subjects with Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition criteria (DSM-IV)-defined schizophrenia and 175 age- and sex-matched HC were enrolled. The levels of twenty-five amino acids and seven related derivatives were measured in plasma samples using hydrophilic interaction liquid chromatography (HILIC) liquid chromatography-tandem mass spectrometry (LC-MS). After controlling for age, sex and body mass index (BMI), four amino acids and derivatives (i.e., cysteine, GABA, glutamine and sarcosine) were observed to be higher in the schizophrenia group when compared with HC; seven amino acids and derivatives were lower in the schizophrenia group (i.e., arginine, L-ornithine, threonine, taurine, tryptophan, methylcysteine, and kynurenine). Statistically significant differences in plasma amino-acid profiles between subjects with first-episode vs. recurrent schizophrenia for aspartate and glutamine were also demonstrated using generalized linear models controlling for age, sex, and BMI. The differences in amino acids and derivatives among individuals with schizophrenia when compared to HC may represent underlying pathophysiology, including but not limited to dysfunctional proteinogenic processes, alterations in excitatory and inhibitory neurotransmission, changes in ammonia metabolism and the urea cycle. Taken together, amino-acid profiling may provide a novel stratification approach among individuals with schizophrenia.
Collapse
Affiliation(s)
- Bing Cao
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, 38 Xue-Yuan Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Dongfang Wang
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, 38 Xue-Yuan Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Elisa Brietzke
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, University Health Network, Toronto, ON, Canada.,Department of Psychiatry, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, University Health Network, Toronto, ON, Canada.,Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Zihang Pan
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Danielle Cha
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, University Health Network, Toronto, ON, Canada.,Faculty of Medicine, School of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Joshua D Rosenblat
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Hannah Zuckerman
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Yaqiong Liu
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, 38 Xue-Yuan Road, Haidian District, Beijing, 100191, People's Republic of China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China.,Peking University Medical and Health Analysis Center, Peking University, Beijing, 100191, People's Republic of China
| | - Qing Xie
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, 38 Xue-Yuan Road, Haidian District, Beijing, 100191, People's Republic of China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China.,Peking University Medical and Health Analysis Center, Peking University, Beijing, 100191, People's Republic of China
| | - Jingyu Wang
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, 38 Xue-Yuan Road, Haidian District, Beijing, 100191, People's Republic of China. .,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China. .,Peking University Medical and Health Analysis Center, Peking University, Beijing, 100191, People's Republic of China.
| |
Collapse
|
842
|
Allen AP, Naughton M, Dowling J, Walsh A, O'Shea R, Shorten G, Scott L, McLoughlin DM, Cryan JF, Clarke G, Dinan TG. Kynurenine pathway metabolism and the neurobiology of treatment-resistant depression: Comparison of multiple ketamine infusions and electroconvulsive therapy. J Psychiatr Res 2018; 100:24-32. [PMID: 29475018 DOI: 10.1016/j.jpsychires.2018.02.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 11/19/2022]
Abstract
Current first-line antidepressants can take weeks or months to decrease depressive symptoms. Low dose ketamine, an N-methyl-D-aspartate (NMDA) receptor antagonist, shows potential for a more rapid antidepressant effect, with efficacy also evident in previously treatment-resistant populations. However, a greater understanding of the physiological mechanisms underlying such effects is required. We assessed the potential impact of ketamine infusion on neurobiological drivers of kynurenine pathway metabolism in major depression (HPA axis hyperactivity, inflammation) in patients with treatment-resistant depression compared to gender-matched healthy controls. Furthermore, we assessed these biomarkers before and after electroconvulsive therapy (ECT), which is currently the gold standard for management of treatment-resistant depression. As previously demonstrated, treatment with ketamine and ECT was associated with improved depressive symptoms in patients. At baseline, waking cortisol output was greater in the ECT cohort, kynurenine was greater in the ketamine cohort, and kynurenic acid was lower in patients compared to healthy controls, although inflammatory markers (IL-6, IL-8, IL-10 or IFN-γ) were similar in patients and controls. Furthermore, in patients who responded to ECT, the cortisol awakening response was decreased following treatment. Despite a trend towards reduced kynurenine concentrations in those who responded to ketamine, ketamine was not associated with significant alterations in any of the biomarkers assessed.
Collapse
Affiliation(s)
- A P Allen
- Department of Psychiatry & Neurobehavioural Science, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - M Naughton
- Department of Psychiatry & Neurobehavioural Science, University College Cork, Cork, Ireland
| | - J Dowling
- Department of Anaesthesia and Intensive Care Medicine, University College Cork, Cork, Ireland
| | - A Walsh
- Department of Anaesthesia and Intensive Care Medicine, University College Cork, Cork, Ireland
| | - R O'Shea
- School of Medicine, University College Cork, Cork, Ireland
| | - G Shorten
- Department of Anaesthesia and Intensive Care Medicine, University College Cork, Cork, Ireland
| | - L Scott
- Department of Psychiatry & Neurobehavioural Science, University College Cork, Cork, Ireland
| | - D M McLoughlin
- St. Patrick's University Hospital, Dublin 8, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - J F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
| | - G Clarke
- Department of Psychiatry & Neurobehavioural Science, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - T G Dinan
- Department of Psychiatry & Neurobehavioural Science, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
843
|
Ball HC, levari-Shariati S, Cooper LN, Aliani M. Comparative metabolomics of aging in a long-lived bat: Insights into the physiology of extreme longevity. PLoS One 2018; 13:e0196154. [PMID: 29715267 PMCID: PMC5929510 DOI: 10.1371/journal.pone.0196154] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 04/06/2018] [Indexed: 12/21/2022] Open
Abstract
Vespertilionid bats (Mammalia: Order Chiroptera) live 3–10 times longer than other mammals of an equivalent body size. At present, nothing is known of how bat fecal metabolic profiles shift with age in any taxa. This study established the feasibility of using a non-invasive, fecal metabolomics approach to examine age-related differences in the fecal metabolome of young and elderly adult big brown bats (Eptesicus fuscus) as an initial investigation into using metabolomics for age determination. Samples were collected from captive, known-aged big brown bats (Eptesicus fuscus) from 1 to over 14 years of age: these two ages represent age groups separated by approximately 75% of the known natural lifespan of this taxon. Results showed 41 metabolites differentiated young (n = 22) and elderly (n = 6) Eptesicus. Significant differences in metabolites between young and elderly bats were associated with tryptophan metabolism and incomplete protein digestion. Results support further exploration of the physiological mechanisms bats employ to achieve exceptional longevity.
Collapse
Affiliation(s)
- Hope C. Ball
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, Ohio, The United States of America
- Musculoskeletal Biology Group, Northeast Ohio Medical University, Rootstown, Ohio, The United States of America
| | - Shiva levari-Shariati
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - Lisa Noelle Cooper
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, Ohio, The United States of America
- Musculoskeletal Biology Group, Northeast Ohio Medical University, Rootstown, Ohio, The United States of America
- * E-mail: (LNC); (MA)
| | - Michel Aliani
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
- Department of Foods and Human Nutritional Sciences, University of Manitoba, Duff Roblin Building, Winnipeg, Canada
- * E-mail: (LNC); (MA)
| |
Collapse
|
844
|
van Baar ACG, Prodan A, Wahlgren CD, Poulsen SS, Knop FK, Groen AK, Bergman JJ, Nieuwdorp M, Levin E. Duodenal L cell density correlates with features of metabolic syndrome and plasma metabolites. Endocr Connect 2018; 7:673-680. [PMID: 29669802 PMCID: PMC5952241 DOI: 10.1530/ec-18-0094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 04/18/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Enteroendocrine cells are essential for the regulation of glucose metabolism, but it is unknown whether they are associated with clinical features of metabolic syndrome (MetS) and fasting plasma metabolites. OBJECTIVE We aimed to identify fasting plasma metabolites that associate with duodenal L cell, K cell and delta cell densities in subjects with MetS with ranging levels of insulin resistance. RESEARCH DESIGN AND METHODS In this cross-sectional study, we evaluated L, K and delta cell density in duodenal biopsies from treatment-naïve males with MetS using machine-learning methodology. RESULTS We identified specific clinical biomarkers and plasma metabolites associated with L cell and delta cell density. L cell density was associated with increased plasma metabolite levels including symmetrical dimethylarginine, 3-aminoisobutyric acid, kynurenine and glycine. In turn, these L cell-linked fasting plasma metabolites correlated with clinical features of MetS. CONCLUSIONS Our results indicate a link between duodenal L cells, plasma metabolites and clinical characteristics of MetS. We conclude that duodenal L cells associate with plasma metabolites that have been implicated in human glucose metabolism homeostasis. Disentangling the causal relation between L cells and these metabolites might help to improve the (small intestinal-driven) pathophysiology behind insulin resistance in human obesity.
Collapse
Affiliation(s)
- Annieke C G van Baar
- Department of Gastroenterology and HepatologyAcademic Medical Center, Amsterdam, the Netherlands
| | - Andrei Prodan
- Department of Vascular MedicineAcademic Medical Center, Amsterdam, the Netherlands
| | - Camilla D Wahlgren
- Center for Diabetes ResearchGentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Steen S Poulsen
- Department of Biomedical SciencesFaculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic ResearchFaculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Filip K Knop
- Center for Diabetes ResearchGentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic ResearchFaculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical MedicineFaculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Albert K Groen
- Department of Vascular MedicineAcademic Medical Center, Amsterdam, the Netherlands
- Department of Laboratory MedicineUniversity of Groningen, University Medical Center, Groningen, the Netherlands
| | - Jacques J Bergman
- Department of Gastroenterology and HepatologyAcademic Medical Center, Amsterdam, the Netherlands
| | - Max Nieuwdorp
- Department of Vascular MedicineAcademic Medical Center, Amsterdam, the Netherlands
- Department of Internal MedicineVUMC Free University, Amsterdam, the Netherlands
- Wallenberg LaboratorySahlgrenska Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Evgeni Levin
- Department of Vascular MedicineAcademic Medical Center, Amsterdam, the Netherlands
- Horaizon BVDelft, the Netherlands
| |
Collapse
|
845
|
Davis I, Yang Y, Wherritt D, Liu A. Reassignment of the human aldehyde dehydrogenase ALDH8A1 (ALDH12) to the kynurenine pathway in tryptophan catabolism. J Biol Chem 2018; 293:9594-9603. [PMID: 29703752 DOI: 10.1074/jbc.ra118.003320] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 04/25/2018] [Indexed: 12/17/2022] Open
Abstract
The kynurenine pathway is the primary route for l-tryptophan degradation in mammals. Intermediates and side products of this pathway are involved in immune response and neurodegenerative diseases. This makes the study of enzymes, especially those from mammalian sources, of the kynurenine pathway worthwhile. Recent studies on a bacterial version of an enzyme of this pathway, 2-aminomuconate semialdehyde (2-AMS) dehydrogenase (AMSDH), have provided a detailed understanding of the catalytic mechanism and identified residues conserved for muconate semialdehyde recognition and activation. Findings from the bacterial enzyme have prompted the reconsideration of the function of a previously identified human aldehyde dehydrogenase, ALDH8A1 (or ALDH12), which was annotated as a retinal dehydrogenase based on its ability to preferentially oxidize 9-cis-retinal over trans-retinal. Here, we provide compelling bioinformatics and experimental evidence that human ALDH8A1 should be reassigned to the missing 2-AMS dehydrogenase of the kynurenine metabolic pathway. For the first time, the product of the semialdehyde oxidation by AMSDH is also revealed by NMR and high-resolution MS. We found that ALDH8A1 catalyzes the NAD+-dependent oxidation of 2-AMS with a catalytic efficiency equivalent to that of AMSDH from the bacterium Pseudomonas fluorescens Substitution of active-site residues required for substrate recognition, binding, and isomerization in the bacterial enzyme resulted in human ALDH8A1 variants with 160-fold increased Km or no detectable activity. In conclusion, this molecular study establishes an additional enzymatic step in an important human pathway for tryptophan catabolism.
Collapse
Affiliation(s)
- Ian Davis
- From the Department of Chemistry, University of Texas, San Antonio, Texas 78249
| | - Yu Yang
- From the Department of Chemistry, University of Texas, San Antonio, Texas 78249
| | - Daniel Wherritt
- From the Department of Chemistry, University of Texas, San Antonio, Texas 78249
| | - Aimin Liu
- From the Department of Chemistry, University of Texas, San Antonio, Texas 78249
| |
Collapse
|
846
|
Leppik L, Kriisa K, Koido K, Koch K, Kajalaid K, Haring L, Vasar E, Zilmer M. Profiling of Amino Acids and Their Derivatives Biogenic Amines Before and After Antipsychotic Treatment in First-Episode Psychosis. Front Psychiatry 2018; 9:155. [PMID: 29740359 PMCID: PMC5928450 DOI: 10.3389/fpsyt.2018.00155] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 04/06/2018] [Indexed: 12/13/2022] Open
Abstract
Schizophrenia (SCH) is a heterogeneous disorder, deriving from a potential multitude of etiopathogenetic factors. During the past few years there has been an increasing interest in the role of circulating amino acids (AAs) and biogenic amines (BAs) in the pathophysiology of SCH. In the present study, we aimed to provide an insight into the potential role of alterations in levels of AAs and BAs as well as examine their more specific metabolic shifts in relation to early stage of SCH. We measured 21 AAs and 17 BAs in serum samples of patients with first-episode psychosis (FEP) before and after 7-month antipsychotic treatment in comparison to control subjects (CSs). According to multivariate analysis, antipsychotic-naïve FEP patients had significantly higher levels of taurine and spermine, whereas values of proline (Pro), alpha-aminoadipic acid (alpha-AAA), kynurenine (Kyn), valine (Val), tyrosine (Tyr), citrulline (Citr), tryptophan (Trp), and histidine (His) were diminished compared to CSs. Increased levels of taurine and spermine, as well as reduced levels of alpha-AAA and Kyn probably reflect the compromised function of N-methyl-D-aspartate (NMDA) receptors in patients. The decreased levels of Pro (AA modulating the function of glutamate decarboxylase) likely reflect the imbalanced function of gamma-aminobutyric acid (GABA) system in the brain of FEP patients. The alterations in ratio between Tyr and phenylalanine (Phe) can be taken as a sign of compromised function of dopaminergic system. These metabolic shifts were reinstated by 7-month antipsychotic treatment. Serum metabolic profiles can be regarded as important indicators to investigate clinical course of SCH and treatment response.
Collapse
Affiliation(s)
- Liisa Leppik
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
- Psychiatry Clinic of Tartu University Hospital, Tartu, Estonia
| | - Kärt Kriisa
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Kati Koido
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Kadri Koch
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
- Psychiatry Clinic of Tartu University Hospital, Tartu, Estonia
| | - Kärolin Kajalaid
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
- Psychiatry Clinic of Tartu University Hospital, Tartu, Estonia
| | - Liina Haring
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
- Psychiatry Clinic of Tartu University Hospital, Tartu, Estonia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Mihkel Zilmer
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
847
|
Schobersberger W, Blank C, Hanser F, Griesmacher A, Canazei M, Leichtfried V. Impact of a single, short morning bright light exposure on tryptophan pathways and visuo- and sensorimotor performance: a crossover study. J Physiol Anthropol 2018; 37:12. [PMID: 29685169 PMCID: PMC5913807 DOI: 10.1186/s40101-018-0173-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 04/16/2018] [Indexed: 11/29/2022] Open
Abstract
Background Bright light (BL) has been shown to be effective in enhancing both cognitive and physical performances. Alterations in nighttime melatonin levels have also been observed. However, evaluations of light-induced changes in the preceding biochemical processes are absent. Therefore, the impact of a single morning BL exposure on sensorimotor and visuomotor performance, as well as tryptophan (trp) and trp metabolites, was evaluated in this study. Methods In a crossover design, 33 healthy volunteers were randomly exposed to 30 min of < 150 lx at eye level (office light, OL) and 5000 lx at eye level (bright light, BL) of 6500 K in the morning hours. Trp, sulfatoxymelatonin (aMT6s), and kynurenine (kyn) courses over the morning hours were analyzed, and changes in sensori- and visuomotor measures were examined. Results Motoric performance increased in both setups, independent of light intensity. aMT6s and kyn decreased equally under both lighting conditions. Trp levels decreased from a mean (95% confidence interval) of 82.0 (77.2–86.9) to 66.5 (62.5–70.1) in the OL setup only. Conclusion These data suggest that BL in the morning hours has a limited effect on visuo- and sensorimotor performance. Nevertheless, trp degradation pathways in the morning show diverse courses after OL and BL exposure. This suggests that trp courses can potentially be altered by BL exposure.
Collapse
Affiliation(s)
- Wolfgang Schobersberger
- Institute for Sports Medicine, Alpine Medicine and Health Tourism, UMIT - University for Health Sciences, Medical Informatics and Technology, Eduard Wallnöfer Zentrum 1, 6060, Hall in Tyrol, Austria. .,Tirol Kliniken GmbH, Anichstraße 35, 6020, Innsbruck, Austria.
| | - Cornelia Blank
- Institute for Sports Medicine, Alpine Medicine and Health Tourism, UMIT - University for Health Sciences, Medical Informatics and Technology, Eduard Wallnöfer Zentrum 1, 6060, Hall in Tyrol, Austria
| | - Friedrich Hanser
- Department of Biomedical Computer Science and Mechatronics, Institute of Electrical and Biomedical Engineering, UMIT - University for Health Sciences, Medical Informatics and Technology, Eduard Wallnöfer Zentrum 1, 6060, Hall in Tyrol, Austria
| | - Andrea Griesmacher
- Central Institute of Medical and Chemical Diagnostics, LKH - University Hospital of Innsbruck, Anichstraße 35, 6020, Innsbruck, Tyrol, Austria
| | - Markus Canazei
- Department of Visual Perception, Bartenbach GmbH, Rinner Strasse 14, 6071, Aldrans, Tyrol, Austria
| | - Veronika Leichtfried
- Institute for Sports Medicine, Alpine Medicine and Health Tourism, UMIT - University for Health Sciences, Medical Informatics and Technology, Eduard Wallnöfer Zentrum 1, 6060, Hall in Tyrol, Austria
| |
Collapse
|
848
|
Groen RN, de Clercq NC, Nieuwdorp M, Hoenders HJR, Groen AK. Gut microbiota, metabolism and psychopathology: A critical review and novel perspectives. Crit Rev Clin Lab Sci 2018; 55:283-293. [PMID: 29673295 DOI: 10.1080/10408363.2018.1463507] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Psychiatric disorders are often associated with metabolic comorbidities. However, the mechanisms through which metabolic and psychiatric disorders are connected remain unclear. Pre-clinical studies in rodents indicate that the bidirectional signaling between the intestine and the brain, the so-called microbiome-gut-brain axis, plays an important role in the regulation of both metabolism and behavior. The gut microbiome produces a vast number of metabolites that may be transported into the host and play a part in homeostatic control of metabolism as well as brain function. In addition to short chain fatty acids, many of these metabolites have been identified in recent years. To what extent both microbiota and their products control human metabolism and behavior is a subject of intense investigation. In this review, we will discuss the most recent findings concerning alterations in the gut microbiota as a possible pathophysiological factor for the co-occurrence of metabolic comorbidities in psychiatric disorders.
Collapse
Affiliation(s)
- Robin N Groen
- a Department of Psychiatry, Interdisciplinary Center Psychopathology and Emotion Regulation , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Nicolien C de Clercq
- b Department of Internal and Vascular Medicine , Academic Medical Center , Amsterdam , The Netherlands
| | - Max Nieuwdorp
- b Department of Internal and Vascular Medicine , Academic Medical Center , Amsterdam , The Netherlands.,c Department of Internal Medicine, VUmc Diabetes Center , Free University Medical Center , Amsterdam , The Netherlands.,d Wallenberg Laboratory , University of Gothenburg , Gothenburg , Sweden
| | | | - Albert K Groen
- b Department of Internal and Vascular Medicine , Academic Medical Center , Amsterdam , The Netherlands.,c Department of Internal Medicine, VUmc Diabetes Center , Free University Medical Center , Amsterdam , The Netherlands.,f Department of Pediatrics , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| |
Collapse
|
849
|
Shin I, Ambler BR, Wherritt D, Griffith WP, Maldonado AC, Altman RA, Liu A. Stepwise O-Atom Transfer in Heme-Based Tryptophan Dioxygenase: Role of Substrate Ammonium in Epoxide Ring Opening. J Am Chem Soc 2018; 140:4372-4379. [PMID: 29506384 PMCID: PMC5874177 DOI: 10.1021/jacs.8b00262] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Heme-based tryptophan dioxygenases are established immunosuppressive metalloproteins with significant biomedical interest. Here, we synthesized two mechanistic probes to specifically test if the α-amino group of the substrate directly participates in a critical step of the O atom transfer during catalysis in human tryptophan 2,3-dioxygenase (TDO). Substitution of the nitrogen atom of the substrate to a carbon (probe 1) or oxygen (probe 2) slowed the catalytic step following the first O atom transfer such that transferring the second O atom becomes less likely to occur, although the dioxygenated products were observed with both probes. A monooxygenated product was also produced from probe 2 in a significant quantity. Analysis of this new product by HPLC coupled UV-vis spectroscopy, high-resolution mass spectrometry, 1H NMR, 13C NMR, HSQC, HMBC, and infrared (IR) spectroscopies concluded that this monooxygenated product is a furoindoline compound derived from an unstable epoxyindole intermediate. These results prove that small molecules can manipulate the stepwise O atom transfer reaction of TDO and provide a showcase for a tunable mechanism by synthetic compounds. The product analysis results corroborate the presence of a substrate-based epoxyindole intermediate during catalysis and provide the first substantial experimental evidence for the involvement of the substrate α-amino group in the epoxide ring-opening step during catalysis. This combined synthetic, biochemical, and biophysical study establishes the catalytic role of the α-amino group of the substrate during the O atom transfer reactions and thus represents a substantial advance to the mechanistic comprehension of the heme-based tryptophan dioxygenases.
Collapse
Affiliation(s)
- Inchul Shin
- Department of Chemistry, University of Texas at San Antonio, San Antonio, Texas 78249, United States
| | - Brett R. Ambler
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Daniel Wherritt
- Department of Chemistry, University of Texas at San Antonio, San Antonio, Texas 78249, United States
| | - Wendell P. Griffith
- Department of Chemistry, University of Texas at San Antonio, San Antonio, Texas 78249, United States
| | - Amanda C. Maldonado
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Ryan A. Altman
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Aimin Liu
- Department of Chemistry, University of Texas at San Antonio, San Antonio, Texas 78249, United States
| |
Collapse
|
850
|
Cheong JE, Sun L. Targeting the IDO1/TDO2–KYN–AhR Pathway for Cancer Immunotherapy – Challenges and Opportunities. Trends Pharmacol Sci 2018; 39:307-325. [PMID: 29254698 DOI: 10.1016/j.tips.2017.11.007] [Citation(s) in RCA: 312] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 11/15/2017] [Accepted: 11/21/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Jae Eun Cheong
- Center for Drug Discovery and Translational Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Lijun Sun
- Center for Drug Discovery and Translational Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|