1
|
Lagard C, Vodovar D, Chevillard L, Callebert J, Caillé F, Pottier G, Liang H, Risède P, Tournier N, Mégarbane B. Investigation of the Mechanisms of Tramadol-Induced Seizures in Overdose in the Rat. Pharmaceuticals (Basel) 2022; 15:1254. [PMID: 36297366 PMCID: PMC9607071 DOI: 10.3390/ph15101254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
Tramadol overdose is frequently associated with the onset of seizures, usually considered as serotonin syndrome manifestations. Recently, the serotoninergic mechanism of tramadol-attributed seizures has been questioned. This study’s aim was to identify the mechanisms involved in tramadol-induced seizures in overdose in rats. The investigations included (1) the effects of specific pretreatments on tramadol-induced seizure onset and brain monoamine concentrations, (2) the interaction between tramadol and γ-aminobutyric acid (GABA)A receptors in vivo in the brain using positron emission tomography (PET) imaging and 11C-flumazenil. Diazepam abolished tramadol-induced seizures, in contrast to naloxone, cyproheptadine and fexofenadine pretreatments. Despite seizure abolishment, diazepam significantly enhanced tramadol-induced increase in the brain serotonin (p < 0.01), histamine (p < 0.01), dopamine (p < 0.05) and norepinephrine (p < 0.05). No displacement of 11C-flumazenil brain kinetics was observed following tramadol administration in contrast to diazepam, suggesting that the observed interaction was not related to a competitive mechanism between tramadol and flumazenil at the benzodiazepine-binding site. Our findings do not support the involvement of serotoninergic, histaminergic, dopaminergic, norepinephrine or opioidergic pathways in tramadol-induced seizures in overdose, but they strongly suggest a tramadol-induced allosteric change of the benzodiazepine-binding site of GABAA receptors. Management of tramadol-poisoned patients should take into account that tramadol-induced seizures are mainly related to a GABAergic pathway.
Collapse
Affiliation(s)
- Camille Lagard
- Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, F-75006 Paris, France
| | - Dominique Vodovar
- Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, F-75006 Paris, France
- Department of Medical and Toxicological Critical Care, AP-HP, Lariboisière Hospital, 75010 Paris, France
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Universités Paris-Sud et Paris-Saclay, 91471 Orsay, France
| | - Lucie Chevillard
- Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, F-75006 Paris, France
| | - Jacques Callebert
- Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, F-75006 Paris, France
- Laboratory of Biochemistry and Molecular Biology, AP-HP, Lariboisière Hospital, 75010 Paris, France
| | - Fabien Caillé
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Universités Paris-Sud et Paris-Saclay, 91471 Orsay, France
| | - Géraldine Pottier
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Universités Paris-Sud et Paris-Saclay, 91471 Orsay, France
| | - Hao Liang
- Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, F-75006 Paris, France
| | - Patricia Risède
- Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, F-75006 Paris, France
| | - Nicolas Tournier
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Universités Paris-Sud et Paris-Saclay, 91471 Orsay, France
| | - Bruno Mégarbane
- Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, F-75006 Paris, France
- Department of Medical and Toxicological Critical Care, AP-HP, Lariboisière Hospital, 75010 Paris, France
| |
Collapse
|
2
|
Krämer J, Kang R, Grimm LM, De Cola L, Picchetti P, Biedermann F. Molecular Probes, Chemosensors, and Nanosensors for Optical Detection of Biorelevant Molecules and Ions in Aqueous Media and Biofluids. Chem Rev 2022; 122:3459-3636. [PMID: 34995461 PMCID: PMC8832467 DOI: 10.1021/acs.chemrev.1c00746] [Citation(s) in RCA: 163] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Indexed: 02/08/2023]
Abstract
Synthetic molecular probes, chemosensors, and nanosensors used in combination with innovative assay protocols hold great potential for the development of robust, low-cost, and fast-responding sensors that are applicable in biofluids (urine, blood, and saliva). Particularly, the development of sensors for metabolites, neurotransmitters, drugs, and inorganic ions is highly desirable due to a lack of suitable biosensors. In addition, the monitoring and analysis of metabolic and signaling networks in cells and organisms by optical probes and chemosensors is becoming increasingly important in molecular biology and medicine. Thus, new perspectives for personalized diagnostics, theranostics, and biochemical/medical research will be unlocked when standing limitations of artificial binders and receptors are overcome. In this review, we survey synthetic sensing systems that have promising (future) application potential for the detection of small molecules, cations, and anions in aqueous media and biofluids. Special attention was given to sensing systems that provide a readily measurable optical signal through dynamic covalent chemistry, supramolecular host-guest interactions, or nanoparticles featuring plasmonic effects. This review shall also enable the reader to evaluate the current performance of molecular probes, chemosensors, and nanosensors in terms of sensitivity and selectivity with respect to practical requirement, and thereby inspiring new ideas for the development of further advanced systems.
Collapse
Affiliation(s)
- Joana Krämer
- Institute
of Nanotechnology, Karlsruhe Institute of
Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Rui Kang
- Institute
of Nanotechnology, Karlsruhe Institute of
Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Laura M. Grimm
- Institute
of Nanotechnology, Karlsruhe Institute of
Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Luisa De Cola
- Institute
of Nanotechnology, Karlsruhe Institute of
Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
- Dipartimento
DISFARM, University of Milano, via Camillo Golgi 19, 20133 Milano, Italy
- Department
of Molecular Biochemistry and Pharmacology, Instituto di Ricerche Farmacologiche Mario Negri, IRCCS, 20156 Milano, Italy
| | - Pierre Picchetti
- Institute
of Nanotechnology, Karlsruhe Institute of
Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Frank Biedermann
- Institute
of Nanotechnology, Karlsruhe Institute of
Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
3
|
Hamelin H, Poizat G, Florian C, Kursa MB, Pittaras E, Callebert J, Rampon C, Taouis M, Hamed A, Granon S. Prolonged Consumption of Sweetened Beverages Lastingly Deteriorates Cognitive Functions and Reward Processing in Mice. Cereb Cortex 2021; 32:1365-1378. [PMID: 34491298 DOI: 10.1093/cercor/bhab274] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 12/25/2022] Open
Abstract
We investigated the detrimental effects of chronic consumption of sweet or sweetened beverages in mice. We report that consumption of beverages containing small amounts of sucrose during several weeks impaired reward systems. This is evidenced by robust changes in the activation pattern of prefrontal brain regions associated with abnormal risk-taking and delayed establishment of decision-making strategy. Supporting these findings, we find that chronic consumption of low doses of artificial sweeteners such as saccharin disrupts brain regions' activity engaged in decision-making and reward processes. Consequently, this leads to the rapid development of inflexible decisions, particularly in a subset of vulnerable individuals. Our data also reveal that regular consumption, even at low doses, of sweet or sweeteners dramatically alters brain neurochemistry, i.e., dopamine content and turnover, and high cognitive functions, while sparing metabolic regulations. Our findings suggest that it would be relevant to focus on long-term consequences on the brain of sweet or sweetened beverages in humans, especially as they may go metabolically unnoticed.
Collapse
Affiliation(s)
- Héloïse Hamelin
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91190, Gif-ur-Yvette, France
| | - Ghislaine Poizat
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91190, Gif-ur-Yvette, France
| | - Cédrick Florian
- Research Center on Animal Cognition (CRCA), Center for Integrative Biology, CNRS UMR 5169, Toulouse 31062, France
| | - Miron Bartosz Kursa
- Interdisciplinary Centre for Mathematical and Computational Modelling, University of Warsaw, 02-106 Warsaw, Poland
| | - Elsa Pittaras
- Stanford University, Heller Laboratory, Stanford, CA 94305-5020, USA
| | - Jacques Callebert
- Service of Biochemistry and Molecular Biology, INSERM U942, Hospital Lariboisière, APHP, Paris 75010, France
| | - Claire Rampon
- Research Center on Animal Cognition (CRCA), Center for Integrative Biology, CNRS UMR 5169, Toulouse 31062, France
| | - Mohammed Taouis
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91190, Gif-ur-Yvette, France
| | - Adam Hamed
- Laboratory of Spatial Memory, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Sylvie Granon
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91190, Gif-ur-Yvette, France
| |
Collapse
|
4
|
Bernal-Meléndez E, Callebert J, Bouillaud P, Persuy MA, Olivier B, Badonnel K, Chavatte-Palmer P, Baly C, Schroeder H. Dopaminergic and serotonergic changes in rabbit fetal brain upon repeated gestational exposure to diesel engine exhaust. Arch Toxicol 2021; 95:3085-3099. [PMID: 34189592 DOI: 10.1007/s00204-021-03110-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/17/2021] [Indexed: 11/29/2022]
Abstract
Limited studies in humans and in animal models have investigated the neurotoxic risks related to a gestational exposure to diesel exhaust particles (DEP) on the embryonic brain, especially those regarding monoaminergic systems linked to neurocognitive disorders. We previously showed that exposure to DEP alters monoaminergic neurotransmission in fetal olfactory bulbs and modifies tissue morphology along with behavioral consequences at birth in a rabbit model. Given the anatomical and functional connections between olfactory and central brain structures, we further characterized their impacts in brain regions associated with monoaminergic neurotransmission. At gestational day 28 (GD28), fetal rabbit brains were collected from dams exposed by nose-only to either a clean air or filtered DEP for 2 h/day, 5 days/week, from GD3 to GD27. HPLC dosage and histochemical analyses of the main monoaminergic systems, i.e., dopamine (DA), noradrenaline (NA), and serotonin (5-HT) and their metabolites were conducted in microdissected fetal brain regions. DEP exposure increased the level of DA and decreased the dopaminergic metabolites ratios in the prefrontal cortex (PFC), together with sex-specific alterations in the hippocampus (Hp). In addition, HVA level was increased in the temporal cortex (TCx). Serotonin and 5-HIAA levels were decreased in the fetal Hp. However, DEP exposure did not significantly modify NA levels, tyrosine hydroxylase, tryptophan hydroxylase or AChE enzymatic activity in fetal brain. Exposure to DEP during fetal life results in dopaminergic and serotonergic changes in critical brain regions that might lead to detrimental potential short-term neural disturbances as precursors of long-term neurocognitive consequences.
Collapse
Affiliation(s)
- Estefania Bernal-Meléndez
- NeuroBiologie de l'Olfaction, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France.,CALBINOTOX, EA7488, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Jacques Callebert
- Service de Biochimie et Biologie Moléculaire, Hôpital Lariboisière, Paris, France
| | | | - Marie-Annick Persuy
- NeuroBiologie de l'Olfaction, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France.,Université Paris-Saclay, UVSQ, INRAE, INRAE, BREED UR1198, Bat. 230, Domaine de Vilvert, 78350, Jouy-en-Josas, France
| | - Benoit Olivier
- CALBINOTOX, EA7488, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Karine Badonnel
- NeuroBiologie de l'Olfaction, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France.,Université Paris-Saclay, UVSQ, INRAE, INRAE, BREED UR1198, Bat. 230, Domaine de Vilvert, 78350, Jouy-en-Josas, France
| | - Pascale Chavatte-Palmer
- Université Paris-Saclay, UVSQ, INRAE, INRAE, BREED UR1198, Bat. 230, Domaine de Vilvert, 78350, Jouy-en-Josas, France
| | - Christine Baly
- NeuroBiologie de l'Olfaction, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France. .,Université Paris-Saclay, UVSQ, INRAE, INRAE, BREED UR1198, Bat. 230, Domaine de Vilvert, 78350, Jouy-en-Josas, France.
| | - Henri Schroeder
- CALBINOTOX, EA7488, Université de Lorraine, Vandœuvre-lès-Nancy, France.
| |
Collapse
|
5
|
Piestansky J, Matuskova M, Cizmarova I, Majerova P, Kovac A, Mikus P. Ultrasensitive determination of serotonin in human urine by a two dimensional capillary isotachophoresis-capillary zone electrophoresis hyphenated with tandem mass spectrometry. J Chromatogr A 2021; 1648:462190. [PMID: 33979756 DOI: 10.1016/j.chroma.2021.462190] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022]
Abstract
A two-dimensional capillary isotachophoresis-capillary zone electrophoresis method hyphenated with tandem mass spectrometry was developed and validated for ultrasensitive quantification of serotonin in real human urine samples. Under optimal conditions, the separation was achieved within 12 min (including on-line sample preparation) with the limit of detection of 34 pg mL-1 (due to a large volume sample injection, here 10 µL, and isotachophoretic preconcentration). This concentration limit represents the lowest value for serotonin in comparison to other previously published separation methods employing mass spectrometry detection and applied to urine matrices. Thanks to high orthogonality, on-line concentration and clean-up effects of this approach, other excellent validation parameters such as linearity (coefficient of determination > 0.99), inter-day and intra-day precision (relative standard deviations 3.5-12.2%), accuracy (relative errors within 99-109.4%), and recovery (96-102%) could be easily obtained too. To demonstrate applicability of the method, we monitored serotonin levels in various real samples (from a healthy volunteer and clinical ones). The determined levels, normalized on the creatinine concentrations, were in the range of 6.81-12.86 ng mmol-1 creatinine This advanced method is suggested for an effective, reliable, high sample throughput, and low cost routine clinical screening or targeted metabolomic studies of serotonin in urine samples.
Collapse
Affiliation(s)
- Juraj Piestansky
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovak Republic; Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovak Republic
| | - Michaela Matuskova
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovak Republic
| | - Ivana Cizmarova
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovak Republic
| | - Petra Majerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 84510 Bratislava, Slovak Republic
| | - Andrej Kovac
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 84510 Bratislava, Slovak Republic
| | - Peter Mikus
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovak Republic; Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovak Republic.
| |
Collapse
|
6
|
Decreased phenol sulfotransferase activities associated with hyperserotonemia in autism spectrum disorders. Transl Psychiatry 2021; 11:23. [PMID: 33414449 PMCID: PMC7791095 DOI: 10.1038/s41398-020-01125-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 09/29/2020] [Accepted: 10/27/2020] [Indexed: 11/08/2022] Open
Abstract
Hyperserotonemia is the most replicated biochemical abnormality associated with autism spectrum disorders (ASD). However, previous studies of serotonin synthesis, catabolism, and transport have not elucidated the mechanisms underlying this hyperserotonemia. Here we investigated serotonin sulfation by phenol sulfotransferases (PST) in blood samples from 97 individuals with ASD and their first-degree relatives (138 parents and 56 siblings), compared with 106 controls. We report a deficient activity of both PST isoforms (M and P) in platelets from individuals with ASD (35% and 78% of patients, respectively), confirmed in autoptic tissues (9 pineal gland samples from individuals with ASD-an important source of serotonin). Platelet PST-M deficiency was strongly associated with hyperserotonemia in individuals with ASD. We then explore genetic or pharmacologic modulation of PST activities in mice: variations of PST activities were associated with marked variations of blood serotonin, demonstrating the influence of the sulfation pathway on serotonemia. We also conducted in 1645 individuals an extensive study of SULT1A genes, encoding PST and mapping at highly polymorphic 16p11.2 locus, which did not reveal an association between copy number or single nucleotide variations and PST activity, blood serotonin or the risk of ASD. In contrast, our broader assessment of sulfation metabolism in ASD showed impairments of other sulfation-related markers, including inorganic sulfate, heparan-sulfate, and heparin sulfate-sulfotransferase. Our study proposes for the first time a compelling mechanism for hyperserotonemia, in a context of global impairment of sulfation metabolism in ASD.
Collapse
|
7
|
Pierre C, Pradère N, Froc C, Ornelas-García P, Callebert J, Rétaux S. A mutation in monoamine oxidase (MAO) affects the evolution of stress behavior in the blind cavefish Astyanax mexicanus. J Exp Biol 2020; 223:jeb226092. [PMID: 32737213 DOI: 10.1242/jeb.226092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/24/2020] [Indexed: 08/26/2023]
Abstract
The neurotransmitter serotonin controls a variety of physiological and behavioral processes. In humans, mutations affecting monoamine oxidase (MAO), the serotonin-degrading enzyme, are highly deleterious. Yet, blind cavefish of the species Astyanax mexicanus carry a partial loss-of-function mutation in MAO (P106L) and thrive in their subterranean environment. Here, we established four fish lines, corresponding to the blind cave-dwelling and the sighted river-dwelling morphs of this species, with or without the mutation, in order to decipher the exact contribution of mao P106L in the evolution of cavefish neurobehavioral traits. Unexpectedly, although mao P106L appeared to be an excellent candidate for the genetic determinism of the loss of aggressive and schooling behaviors in cavefish, we demonstrated that it was not the case. Similarly, the anatomical variations in monoaminergic systems observed between cavefish and surface fish brains were independent from mao P106L, and rather due to other, morph-dependent developmental processes. However, we found that mao P106L strongly affected anxiety-like behaviors. Cortisol measurements showed lower basal levels and an increased amplitude of stress response after a change of environment in fish carrying the mutation. Finally, we studied the distribution of the P106L mao allele in wild populations of cave and river A. mexicanus, and discovered that the mutant allele was present - and sometimes fixed - in all populations inhabiting caves of the Sierra de El Abra. The possibility that this partial loss-of-function mao allele evolves under a selective or a neutral regime in the particular cave environment is discussed.
Collapse
Affiliation(s)
- Constance Pierre
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Naomie Pradère
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Cynthia Froc
- Amatrace platform, Institut des Neurosciences Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Patricia Ornelas-García
- Departamento de Zoología, Instituto de Biología, Universidad Autónoma de México, CP 04510, Mexico City, Mexico
| | - Jacques Callebert
- Service Biochimie et Biologie Moléculaire, Hôpital Lariboisière, 75475 Paris, France
| | - Sylvie Rétaux
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91190, Gif-sur-Yvette, France
| |
Collapse
|
8
|
de Hosson LD, Takkenkamp TJ, Kats-Ugurlu G, Bouma G, Bulthuis M, de Vries EGE, van Faassen M, Kema IP, Walenkamp AME. Neuroendocrine tumours and their microenvironment. Cancer Immunol Immunother 2020; 69:1449-1459. [PMID: 32270230 PMCID: PMC7347684 DOI: 10.1007/s00262-020-02556-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 03/26/2020] [Indexed: 02/06/2023]
Abstract
Tumours can escape the immune system by expressing programmed death-ligand-1 (PD-L1), which allows them to bind to PD-1 on T-cells and avoid recognition by the immune system. Regulatory T-cells (Tregs), indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) also play a role in immune suppression. Knowledge about the interaction of neuroendocrine tumours (NETs) with their immune microenvironment and the role of immunotherapy in patients with NET is scarce. Here, we investigated the immune microenvironment of serotonin-producing (SP) and non-serotonin-producing NETs (NSP-NETs). Tumours of 33 patients with SP-NET and 18 patients with NSP-NET were studied. Immunohistochemical analyses were performed for PD-L1, T-cells, IDO, TDO, mismatch repair proteins (MMRp) and activated fibroblasts. PD-L1 expression was seen in < 1% of tumour and T-cells. T-cells were present in 33% of NETs, varying between 1 and 10% T-cells per high power field. IDO was expressed in tumour cells in 55% of SP-NETs and 22% of NSP-NETs (p = 0.039). TDO was expressed in stromal cells in 64% of SP-NETs and 13% of NSP-NETs (p = 0.001). No tumours had loss of MMRp. TDO-expressing stromal cells also strongly expressed α-SMA and were identified as cancer-associated fibroblasts (CAFs). Factors that are associated with a response to checkpoint inhibitor treatment were absent or only present to a limited extent in the tumour microenvironment of NETs. The expression of IDO and TDO in a substantial part of NETs and the presence of CAFs suggest two mechanisms that could be responsible for the cold immune microenvironment, which should be explored to enhance anti-tumour immunity and clinical responses.
Collapse
Affiliation(s)
- Lotte D de Hosson
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, DA11, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Tim J Takkenkamp
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, DA11, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Gursah Kats-Ugurlu
- Department of Pathology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Grietje Bouma
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, DA11, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Marian Bulthuis
- Department of Pathology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, DA11, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Annemiek M E Walenkamp
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, DA11, PO Box 30.001, 9700 RB, Groningen, The Netherlands.
| |
Collapse
|
9
|
Pittaras E, Callebert J, Dorey R, Chennaoui M, Granon S, Rabat A. Mouse Gambling Task reveals differential effects of acute sleep debt on decision-making and associated neurochemical changes. Sleep 2019; 41:5126120. [PMID: 30304537 PMCID: PMC6231521 DOI: 10.1093/sleep/zsy168] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Indexed: 01/19/2023] Open
Abstract
Sleep loss is associated with sleepiness, sustained attention, and memory deficits. However, vulnerability of higher cognitive processes (i.e. decision making) to sleep debt is less understood. Therefore, a major challenge is to understand why and how higher cognitive processes are affected by sleep debt. We had established in mice correlations between individual decision-making strategies, prefrontal activity, and regional monoaminergic levels. Now, we show that acute sleep debt (ASD) disturbs decision-making processes and provokes brain regional modifications of serotonin and dopamine that could explain why ASD promotes inflexible and more risk-prone behaviors. Finally, we highlight, for the first time, that in a large group of healthy inbred mice some of them are more sensitive to ASD by showing inflexible behavior and decision-making deficits. We were also able to predict mice that would be the most vulnerable to ASD depending of their behavior before ASD exposure.
Collapse
Affiliation(s)
- Elsa Pittaras
- Unité Fatigue et Vigilance, Département Environnements Opérationnels, Institut de Recherche Biomédicale des armées, Brétigny-sur-Orge cedex, France.,Equipe 'Neurobiologie de la prise de décision', Neuro-PSI, CNRS UMR 9197, Orsay, France.,Equipe d'accueil VIgilance FAtigue et SOMmeil (VIFASOM) EA 7330 - Université Paris 5 Descartes, Paris, France.,Biology Department, Stanford University, Stanford, CA
| | - Jacques Callebert
- Service de Biochimie et Biologie Moléculaire, Hôpital Lariboisière, Paris, France
| | - Rodolphe Dorey
- Unité Fatigue et Vigilance, Département Environnements Opérationnels, Institut de Recherche Biomédicale des armées, Brétigny-sur-Orge cedex, France.,Equipe d'accueil VIgilance FAtigue et SOMmeil (VIFASOM) EA 7330 - Université Paris 5 Descartes, Paris, France
| | - Mounir Chennaoui
- Unité Fatigue et Vigilance, Département Environnements Opérationnels, Institut de Recherche Biomédicale des armées, Brétigny-sur-Orge cedex, France.,Equipe d'accueil VIgilance FAtigue et SOMmeil (VIFASOM) EA 7330 - Université Paris 5 Descartes, Paris, France
| | - Sylvie Granon
- Equipe 'Neurobiologie de la prise de décision', Neuro-PSI, CNRS UMR 9197, Orsay, France
| | - Arnaud Rabat
- Unité Fatigue et Vigilance, Département Environnements Opérationnels, Institut de Recherche Biomédicale des armées, Brétigny-sur-Orge cedex, France.,Equipe d'accueil VIgilance FAtigue et SOMmeil (VIFASOM) EA 7330 - Université Paris 5 Descartes, Paris, France
| |
Collapse
|
10
|
Bernal-Meléndez E, Lacroix MC, Bouillaud P, Callebert J, Olivier B, Persuy MA, Durieux D, Rousseau-Ralliard D, Aioun J, Cassee F, Couturier-Tarrade A, Valentino S, Chavatte-Palmer P, Schroeder H, Baly C. Repeated gestational exposure to diesel engine exhaust affects the fetal olfactory system and alters olfactory-based behavior in rabbit offspring. Part Fibre Toxicol 2019; 16:5. [PMID: 30654819 PMCID: PMC6335688 DOI: 10.1186/s12989-018-0288-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 12/20/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Airborne pollution, especially from diesel exhaust (DE), is known to have a negative effect on the central nervous system in exposed human populations. However, the consequences of gestational exposure to DE on the fetal brain remain poorly explored, with various effects depending on the conditions of exposure, as well as little information on early developmental stages. We investigated the short-term effects of indirect DE exposure throughout gestation on the developing brain using a rabbit model. We analyzed fetal olfactory tissues at the end of gestation and tested behaviors relevant to pups' survival at birth. Pregnant dams were exposed by nose-only inhalation to either clean air or DE with a content of particles (DEP) adjusted to 1 mg/m3 by diluting engine exhaust, for 2 h/day, 5 days/week, from gestational day 3 (GD3) to day 27 (GD27). At GD28, fetal olfactory mucosa, olfactory bulbs and whole brains were collected for anatomical and neurochemical measurements. At postnatal day 2 (PND2), pups born from another group of exposed or control female were examined for their odor-guided behavior in response to the presentation of the rabbit mammary pheromone 2-methyl-3-butyn-2-ol (2MB2). RESULTS At GD28, nano-sized particles were observed in cilia and cytoplasm of the olfactory sensory neurons in the olfactory mucosa and in the cytoplasm of periglomerular cells in the olfactory bulbs of exposed fetuses. Moreover, cellular and axonal hypertrophies were observed throughout olfactory tissues. Concomitantly, fetal serotoninergic and dopaminergic systems were affected in the olfactory bulbs. Moreover, the neuromodulatory homeostasis was disturbed in a sex-dependent manner in olfactory tissues. At birth, the olfactory sensitivity to 2MB2 was reduced in exposed PND2 pups. CONCLUSION Gestational exposure to DE alters olfactory tissues and affects monoaminergic neurotransmission in fetuses' olfactory bulbs, resulting in an alteration of olfactory-based behaviors at birth. Considering the anatomical and functional continuum between the olfactory system and other brain structures, and due to the importance of monoamine neurotransmission in the plasticity of neural circuits, such alterations could participate to disturbances in higher integrative structures, with possible long-term neurobehavioral consequences.
Collapse
Affiliation(s)
- Estefanía Bernal-Meléndez
- NeuroBiologie de l’Olfaction, INRA, Université Paris-Saclay, 78350 Jouy-en-Josas, France
- CALBINOTOX, Université de Lorraine, EA7488 Vandœuvre-lès-Nancy, France
| | | | | | - Jacques Callebert
- Service de Biochimie et Biologie Moléculaire, Hôpital Lariboisière, Paris, France
| | - Benoit Olivier
- CALBINOTOX, Université de Lorraine, EA7488 Vandœuvre-lès-Nancy, France
| | - Marie-Annick Persuy
- NeuroBiologie de l’Olfaction, INRA, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Didier Durieux
- NeuroBiologie de l’Olfaction, INRA, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | | | - Josiane Aioun
- UMR BDR, INRA, ENVA, Université Paris Saclay, 78350 Jouy-en-Josas, France
| | - Flemming Cassee
- Center for Sustainability, Environment and Health, National Institute for Public Health and the Environment, Bilthoven, Netherlands
- Institute of Risk Assessment Sciences, Utrecht University, Utrecht, Netherlands
| | | | - Sarah Valentino
- UMR BDR, INRA, ENVA, Université Paris Saclay, 78350 Jouy-en-Josas, France
| | | | - Henri Schroeder
- CALBINOTOX, Université de Lorraine, EA7488 Vandœuvre-lès-Nancy, France
| | - Christine Baly
- NeuroBiologie de l’Olfaction, INRA, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| |
Collapse
|
11
|
Riché F, Chousterman BG, Valleur P, Mebazaa A, Launay JM, Gayat E. Protracted immune disorders at one year after ICU discharge in patients with septic shock. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2018; 22:42. [PMID: 29467023 PMCID: PMC5822646 DOI: 10.1186/s13054-017-1934-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 12/27/2017] [Indexed: 01/13/2023]
Abstract
Background Sepsis is a leading cause of mortality and critical illness worldwide and is associated with an increased mortality rate in the months following hospital discharge. The occurrence of persistent or new organ dysfunction(s) after septic shock raises questions about the mechanisms involved in the post-sepsis status. The present study aimed to explore the immune profiles of patients one year after being discharged from the intensive care unit (ICU) following treatment for abdominal septic shock. Methods We conducted a prospective, single-center, observational study in the surgical ICU of a university hospital. Eighty-six consecutive patients admitted for septic shock of abdominal origin were included in this study. Fifteen different plasma biomarkers were measured at ICU admission, at ICU discharge and at one year after ICU discharge. Three different clusters of biomarkers were distinguished according to their functions, namely: (1) inflammatory response, (2) cell damage and apoptosis, (3) immunosuppression and resolution of inflammation. The primary objective was to characterize variations in the immune status of septic shock patients admitted to ICU up to one year after ICU discharge. The secondary objective was to evaluate the relationship between these biomarker variations and patient outcomes. Results At the onset of septic shock, we observed a cohesive pro-inflammatory profile and low levels of inflammation resolution markers. At ICU discharge, the immune status demonstrated decreased but persistent inflammation and increased immunosuppression, with elevated programmed cell death protein-1 (PD-1) levels, and a counterbalanced resolution process, with elevated levels of interleukin-10 (IL-10), resolvin D5 (RvD5), and IL-7. One year after hospital discharge, homeostasis was not completely restored with several markers of inflammation remaining elevated. Remarkably, IL-7 was persistently elevated, with levels comparable to those observed after ICU discharge, and PD-1, while lower, remained in the elevated abnormal range. Conclusions In this study, protracted immune disturbances were observed one year after ICU discharge. The study results suggested the presence of long-lasting immune illness disorders following a long-term septic insult, indicating the need for long-term patient follow up after ICU discharge and questioning the use of immune intervention to restore immune homeostasis after abdominal septic shock. Electronic supplementary material The online version of this article (doi:10.1186/s13054-017-1934-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Florence Riché
- Department of Anesthesiology and Intensive Care Medicine, Saint Louis Lariboisière University Hospital, University Paris Diderot, Assistance Publique - Hôpitaux de Paris, 2 rue Ambroise Paré, 75010, Paris, France.
| | - Benjamin G Chousterman
- Department of Anesthesiology and Intensive Care Medicine, Saint Louis Lariboisière University Hospital, University Paris Diderot, Assistance Publique - Hôpitaux de Paris, 2 rue Ambroise Paré, 75010, Paris, France.,Inserm U1160, Hôpital Saint-Louis, 1 rue Claude Vellefaux, 75010, Paris, France
| | - Patrice Valleur
- Department of Visceral Surgery, Saint Louis Lariboisière University Hospital, University Paris Diderot, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Alexandre Mebazaa
- Department of Anesthesiology and Intensive Care Medicine, Saint Louis Lariboisière University Hospital, University Paris Diderot, Assistance Publique - Hôpitaux de Paris, 2 rue Ambroise Paré, 75010, Paris, France.,Biomarkers in CArdio-Neuro-VAScular diseases (BIOCANVAS), UMR-S 942, Inserm, Paris, France
| | - Jean-Marie Launay
- Biomarkers in CArdio-Neuro-VAScular diseases (BIOCANVAS), UMR-S 942, Inserm, Paris, France.,Department of Biochemistry, Saint Louis Lariboisière University Hospital, University Paris Diderot, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Etienne Gayat
- Department of Anesthesiology and Intensive Care Medicine, Saint Louis Lariboisière University Hospital, University Paris Diderot, Assistance Publique - Hôpitaux de Paris, 2 rue Ambroise Paré, 75010, Paris, France.,Biomarkers in CArdio-Neuro-VAScular diseases (BIOCANVAS), UMR-S 942, Inserm, Paris, France
| |
Collapse
|
12
|
Heritability of the melatonin synthesis variability in autism spectrum disorders. Sci Rep 2017; 7:17746. [PMID: 29255243 PMCID: PMC5735101 DOI: 10.1038/s41598-017-18016-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 12/05/2017] [Indexed: 01/23/2023] Open
Abstract
Autism Spectrum Disorders (ASD) are heterogeneous neurodevelopmental disorders with a complex genetic architecture. They are characterized by impaired social communication, stereotyped behaviors and restricted interests and are frequently associated with comorbidities such as intellectual disability, epilepsy and severe sleep disorders. Hyperserotonemia and low melatonin levels are among the most replicated endophenotypes reported in ASD, but their genetic causes remain largely unknown. Based on the biochemical profile of 717 individuals including 213 children with ASD, 128 unaffected siblings and 376 parents and other relatives, we estimated the heritability of whole-blood serotonin, platelet N-acetylserotonin (NAS) and plasma melatonin levels, as well as the two enzymes arylalkylamine N-acetyltransferase (AANAT) and acetylserotonin O-methyltransferase (ASMT) activities measured in platelets. Overall, heritability was higher for NAS (0.72 ± 0.091) and ASMT (0.59 ± 0.097) compared with serotonin (0.31 ± 0.078), AANAT (0.34 ± 0.077) and melatonin (0.22 ± 0.071). Bivariate analyses showed high phenotypic and genetic correlations between traits of the second step of the metabolic pathway (NAS, ASMT and melatonin) indicating the contribution of shared genetic factors. A better knowledge of the heritability of the melatonin synthesis variability constitutes an important step to identify the factors that perturb this pathway in individuals with ASD.
Collapse
|
13
|
Wei Z, Xu C, Liu S, Song F, Liu Z, Qu X. Metabonomics study of the effects of traditional Chinese medicine formula Ermiaowan on hyperuricemic rats. J Sep Sci 2017; 41:560-570. [DOI: 10.1002/jssc.201700985] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/29/2017] [Accepted: 10/23/2017] [Indexed: 01/04/2023]
Affiliation(s)
- Zhongbao Wei
- Pharmacology of Chinese Traditional Medicine; College of Pharmacy; Changchun University of Chinese Medicine; Changchun P. R.China
| | - Chen Xu
- School of Chemical Engineering; Changchun University of Technology; Changchun P. R. China
| | - Shu Liu
- National Center of Mass Spectrometry in Changchun & Jilin Province Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun P. R. China
| | - Fengrui Song
- National Center of Mass Spectrometry in Changchun & Jilin Province Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun P. R. China
| | - Zhiqiang Liu
- National Center of Mass Spectrometry in Changchun & Jilin Province Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun P. R. China
| | - Xiaobo Qu
- Pharmacology of Chinese Traditional Medicine; College of Pharmacy; Changchun University of Chinese Medicine; Changchun P. R.China
| |
Collapse
|
14
|
Szeitz A, Bandiera SM. Analysis and measurement of serotonin. Biomed Chromatogr 2017; 32. [DOI: 10.1002/bmc.4135] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/27/2017] [Accepted: 11/03/2017] [Indexed: 12/26/2022]
Affiliation(s)
- András Szeitz
- Faculty of Pharmaceutical Sciences; The University of British Columbia; Vancouver British Columbia Canada
| | - Stelvio M. Bandiera
- Faculty of Pharmaceutical Sciences; The University of British Columbia; Vancouver British Columbia Canada
| |
Collapse
|
15
|
Sadok I, Gamian A, Staniszewska MM. Chromatographic analysis of tryptophan metabolites. J Sep Sci 2017; 40:3020-3045. [PMID: 28590049 PMCID: PMC5575536 DOI: 10.1002/jssc.201700184] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/15/2017] [Accepted: 05/24/2017] [Indexed: 12/14/2022]
Abstract
The kynurenine pathway generates multiple tryptophan metabolites called collectively kynurenines and leads to formation of the enzyme cofactor nicotinamide adenine dinucleotide. The first step in this pathway is tryptophan degradation, initiated by the rate-limiting enzymes indoleamine 2,3-dioxygenase, or tryptophan 2,3-dioxygenase, depending on the tissue. The balanced kynurenine metabolism, which has been a subject of multiple studies in last decades, plays an important role in several physiological and pathological conditions such as infections, autoimmunity, neurological disorders, cancer, cataracts, as well as pregnancy. Understanding the regulation of tryptophan depletion provide novel diagnostic and treatment opportunities, however it requires reliable methods for quantification of kynurenines in biological samples with complex composition (body fluids, tissues, or cells). Trace concentrations, interference of sample components, and instability of some tryptophan metabolites need to be addressed using analytical methods. The novel separation approaches and optimized extraction protocols help to overcome difficulties in analyzing kynurenines within the complex tissue material. Recent developments in chromatography coupled with mass spectrometry provide new opportunity for quantification of tryptophan and its degradation products in various biological samples. In this review, we present current accomplishments in the chromatographic methodologies proposed for detection of tryptophan metabolites and provide a guide for choosing the optimal approach.
Collapse
Affiliation(s)
- Ilona Sadok
- Laboratory of Separation and Spectroscopic Method Applications, Centre for Interdisciplinary ResearchThe John Paul II Catholic University of LublinLublinPoland
| | - Andrzej Gamian
- Laboratory of Medical MicrobiologyHirszfeld Institute of Immunology and Experimental TherapyPolish Academy of SciencesWroclawPoland
- Department of Medical BiochemistryWroclaw Medical UniversityWroclawPoland
| | - Magdalena Maria Staniszewska
- Laboratory of Separation and Spectroscopic Method Applications, Centre for Interdisciplinary ResearchThe John Paul II Catholic University of LublinLublinPoland
- Laboratory of Medical MicrobiologyHirszfeld Institute of Immunology and Experimental TherapyPolish Academy of SciencesWroclawPoland
| |
Collapse
|
16
|
Disruption of melatonin synthesis is associated with impaired 14-3-3 and miR-451 levels in patients with autism spectrum disorders. Sci Rep 2017; 7:2096. [PMID: 28522826 PMCID: PMC5437096 DOI: 10.1038/s41598-017-02152-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 04/06/2017] [Indexed: 12/12/2022] Open
Abstract
Autism spectrum disorders (ASD) are characterized by a wide genetic and clinical heterogeneity. However, some biochemical impairments, including decreased melatonin (crucial for circadian regulation) and elevated platelet N-acetylserotonin (the precursor of melatonin) have been reported as very frequent features in individuals with ASD. To address the mechanisms of these dysfunctions, we investigated melatonin synthesis in post-mortem pineal glands - the main source of melatonin (9 patients and 22 controls) - and gut samples - the main source of serotonin (11 patients and 13 controls), and in blood platelets from 239 individuals with ASD, their first-degree relatives and 278 controls. Our results elucidate the enzymatic mechanism for melatonin deficit in ASD, involving a reduction of both enzyme activities contributing to melatonin synthesis (AANAT and ASMT), observed in the pineal gland as well as in gut and platelets of patients. Further investigations suggest new, post-translational (reduced levels of 14-3-3 proteins which regulate AANAT and ASMT activities) and post-transcriptional (increased levels of miR-451, targeting 14-3-3ζ) mechanisms to these impairments. This study thus gives insights into the pathophysiological pathways involved in ASD.
Collapse
|
17
|
Loss-of-function of PTPR γ and ζ, observed in sporadic schizophrenia, causes brain region-specific deregulation of monoamine levels and altered behavior in mice. Psychopharmacology (Berl) 2017; 234:575-587. [PMID: 28025742 DOI: 10.1007/s00213-016-4490-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 11/17/2016] [Indexed: 12/12/2022]
Abstract
RATIONALE The receptor protein tyrosine phosphatase PTPRG has been genetically associated with psychiatric disorders and is a ligand for members of the contactin family, which are themselves linked to autism spectrum disorders. OBJECTIVE Based on our finding of a phosphatase-null de novo mutation in PTPRG associated with a case of sporadic schizophrenia, we used PTPRG knockout (KO) mice to model the effect of a loss-of-function mutation. We compared the results with loss-of-function in its close paralogue PTPRZ, previously associated with schizophrenia. We tested PTPRG -/- , PTPRZ -/- , and wild-type male mice for effects on social behavior, forced swim test, and anxiety, as well as on regional brain neurochemistry. RESULTS The most notable behavioral consequences of PTPRG gene inactivation were reduced immobilization in the forced swim test, suggestive of some negative symptoms of schizophrenia. By contrast, PTPRZ -/- mice demonstrated marked social alteration with increased aggressivity, reminiscent of some positive symptoms of schizophrenia. Both knockouts showed elevated dopamine levels in prefrontal cortex, hippocampus, and most particularly amygdala, but not striatum, accompanied by reduced dopamine beta hydroxylase activity only in amygdala. In addition, PTPRG KO elicited a distinct increase in hippocampal serotonin level not observed in PTPRZ KO. CONCLUSION PTPRG and PTPRZ gene loss therefore induces distinct patterns of behavioral change and region-specific alterations in neurotransmitters, highlighting their usefulness as models for neuropsychiatric disorder mechanisms and making these receptors attractive targets for therapy.
Collapse
|
18
|
Baud A, Mulligan MK, Casale FP, Ingels JF, Bohl CJ, Callebert J, Launay JM, Krohn J, Legarra A, Williams RW, Stegle O. Genetic Variation in the Social Environment Contributes to Health and Disease. PLoS Genet 2017; 13:e1006498. [PMID: 28121987 PMCID: PMC5266220 DOI: 10.1371/journal.pgen.1006498] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 11/21/2016] [Indexed: 11/29/2022] Open
Abstract
Assessing the impact of the social environment on health and disease is challenging. As social effects are in part determined by the genetic makeup of social partners, they can be studied from associations between genotypes of one individual and phenotype of another (social genetic effects, SGE, also called indirect genetic effects). For the first time we quantified the contribution of SGE to more than 100 organismal phenotypes and genome-wide gene expression measured in laboratory mice. We find that genetic variation in cage mates (i.e. SGE) contributes to variation in organismal and molecular measures related to anxiety, wound healing, immune function, and body weight. Social genetic effects explained up to 29% of phenotypic variance, and for several traits their contribution exceeded that of direct genetic effects (effects of an individual's genotypes on its own phenotype). Importantly, we show that ignoring SGE can severely bias estimates of direct genetic effects (heritability). Thus SGE may be an important source of "missing heritability" in studies of complex traits in human populations. In summary, our study uncovers an important contribution of the social environment to phenotypic variation, sets the basis for using SGE to dissect social effects, and identifies an opportunity to improve studies of direct genetic effects.
Collapse
Affiliation(s)
- Amelie Baud
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Megan K. Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Francesco Paolo Casale
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Jesse F. Ingels
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Casey J. Bohl
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Jacques Callebert
- AP-HP, Hôpital Lariboisière, Department of Biochemistry, INSERM U942, Paris, France
| | - Jean-Marie Launay
- AP-HP, Hôpital Lariboisière, Department of Biochemistry, INSERM U942, Paris, France
| | - Jon Krohn
- Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom
| | | | - Robert W. Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Oliver Stegle
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| |
Collapse
|
19
|
Laugeray A, Launay JM, Callebert J, Mutlu O, Guillemin GJ, Belzung C, Barone PR. Chronic Treatment with the IDO1 Inhibitor 1-Methyl-D-Tryptophan Minimizes the Behavioural and Biochemical Abnormalities Induced by Unpredictable Chronic Mild Stress in Mice - Comparison with Fluoxetine. PLoS One 2016; 11:e0164337. [PMID: 27828964 PMCID: PMC5102430 DOI: 10.1371/journal.pone.0164337] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/22/2016] [Indexed: 11/19/2022] Open
Abstract
We demonstrated that confronting mice to the Unpredictable Chronic Mild Stress (UCMS) procedure-a validated model of stress-induced depression-results in behavioural alterations and biochemical changes in the kynurenine pathway (KP), suspected to modify the glutamatergic neurotransmission through the imbalance between downstream metabolites such as 3-hydroxykynurenine, quinolinic and kynurenic acids. We showed that daily treatment with the IDO1 inhibitor 1-methyl-D-tryptophan partially rescues UCMS-induced KP alterations as does the antidepressant fluoxetine. More importantly we demonstrated that 1-methyl-D-tryptophan was able to alleviate most of the behavioural changes resulting from UCMS exposure. We also showed that both fluoxetine and 1-methyl-D-tryptophan robustly reduced peripheral levels of proinflammatory cytokines in UCMS mice suggesting that their therapeutic effects might occur through anti-inflammatory processes. KP inhibition might be involved in the positive effects of fluoxetine on mice behaviour and could be a relevant strategy to counteract depressive-like symptoms.
Collapse
Affiliation(s)
- Anthony Laugeray
- Molecular and Experimental Immunology and Neurogenetics – UMR7355, CNRS - 3b Rue de Férollerie, Orléans La Source, Cedex 2, France
| | - Jean-Marie Launay
- UMRS INSERM U942, Service de Biochimie, Hôpital Lariboisière, Assistance Publique — Hôpitaux de Paris, Paris, France
| | - Jacques Callebert
- UMRS INSERM U942, Service de Biochimie, Hôpital Lariboisière, Assistance Publique — Hôpitaux de Paris, Paris, France
| | - Oguz Mutlu
- UMRS INSERM U930, CNRS ERL 3106, Université François Rabelais, Tours, France
- Department of Pharmacology, Faculty of Medicine, Kocaeli University, 41380 Kocaeli, Turkey
| | - Gilles J. Guillemin
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University, NSW, Sydney, Australia
| | - Catherine Belzung
- UMRS INSERM U930, CNRS ERL 3106, Université François Rabelais, Tours, France
| | - Pascal R. Barone
- UMRS INSERM U930, CNRS ERL 3106, Université François Rabelais, Tours, France
| |
Collapse
|
20
|
Georgin-Lavialle S, Moura DS, Salvador A, Chauvet-Gelinier JC, Launay JM, Damaj G, Côté F, Soucié E, Chandesris MO, Barète S, Grandpeix-Guyodo C, Bachmeyer C, Alyanakian MA, Aouba A, Lortholary O, Dubreuil P, Teyssier JR, Trojak B, Haffen E, Vandel P, Bonin B, Hermine O, Gaillard R. Mast cells' involvement in inflammation pathways linked to depression: evidence in mastocytosis. Mol Psychiatry 2016; 21:1511-1516. [PMID: 26809839 DOI: 10.1038/mp.2015.216] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 10/24/2015] [Accepted: 11/24/2015] [Indexed: 12/17/2022]
Abstract
Converging sources of evidence point to a role for inflammation in the development of depression, fatigue and cognitive dysfunction. More precisely, the tryptophan (TRP) catabolism is thought to play a major role in inflammation-induced depression. Mastocytosis is a rare disease in which chronic symptoms, including depression, are related to mast cell accumulation and activation. Our objectives were to study the correlations between neuropsychiatric features and the TRP catabolism pathway in mastocytosis in order to demonstrate mast cells' potential involvement in inflammation-induced depression. Fifty-four patients with mastocytosis and a mean age of 50.1 years were enrolled in the study and compared healthy age-matched controls. Depression and stress were evaluated with the Beck Depression Inventory revised and the Perceived Stress Scale. All patients had measurements of TRP, serotonin (5-HT), kynurenine (KYN), indoleamine 2,3-dioxygenase 1 (IDO1) activity (ratio KYN/TRP), kynurenic acid (KA) and quinolinic acid (QA). Patients displayed significantly lower levels of TRP and 5-HT without hypoalbuminemia or malabsorption, higher IDO1 activity, and higher levels of KA and QA, with an imbalance towards the latter. High perceived stress and high depression scores were associated with low TRP and high IDO1 activity. In conclusion, TRP metabolism is altered in mastocytosis and correlates with perceived stress and depression, demonstrating mast cells' involvement in inflammation pathways linked to depression.
Collapse
Affiliation(s)
- S Georgin-Lavialle
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders, Hôpital Necker-Enfants malades, Paris, France.,Service de médecine Interne, Hôpital Tenon, Université Pierre et Marie Curie, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - D S Moura
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders, Hôpital Necker-Enfants malades, Paris, France.,Laboratoire de Psychopathologie et Processus de Santé, EA 4057, IUPDP, Institut de Psychologie, Université Paris Descartes, Paris, France
| | - A Salvador
- Laboratoire de "Physiopathologie des maladies Psychiatriques", Centre de Psychiatrie et Neurosciences U894, INSERM, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service de Psychiatrie, Centre Hospitalier Sainte-Anne, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris Descartes, Paris, France
| | - J-C Chauvet-Gelinier
- Service de Psychiatrie, Département de Neurosciences, Dijon, France.,Laboratoire de Psychologie et Psychopathologie Médicale (IFR 100), Université de Bourgogne, Dijon, France
| | - J-M Launay
- Laboratoire de biochimie et biologie moléculaire, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - G Damaj
- Service des Maladies du Sang, Centre Hospitalier Universitaire, Hôpital Sud, Amiens, France
| | - F Côté
- INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders, Hôpital Necker-Enfants malades, Paris, France
| | - E Soucié
- INSERM UMR 891, Centre de Recherche en Cancérologie de Marseille, Laboratoire d'Hématopoïèse Moléculaire et Fonctionnelle, Marseille, France
| | - M-O Chandesris
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France
| | - S Barète
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders, Hôpital Necker-Enfants malades, Paris, France
| | - C Grandpeix-Guyodo
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France
| | - C Bachmeyer
- Service de médecine Interne, Hôpital Tenon, Université Pierre et Marie Curie, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - M-A Alyanakian
- Laboratoire d'immunologie, Hôpital Necker, Paris, France
| | - A Aouba
- Service d'Hématologie Adulte, Université Paris Descartes, Sorbonne, Paris Cité, Assistance Publique-Hôpitaux de Paris, Institut Imagine, Hôpital Necker-Enfants malades, Paris, France
| | - O Lortholary
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,Service des infectieuses et tropicales, Université Paris Descartes, Sorbonne, Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants malades, Paris, France
| | - P Dubreuil
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,INSERM UMR 891, Centre de Recherche en Cancérologie de Marseille, Laboratoire d'Hématopoïèse Moléculaire et Fonctionnelle, Marseille, France
| | - J-R Teyssier
- Laboratoire de génétique, CHU, PTB, 2 rue Angélique Ducoudray, Dijon, France
| | - B Trojak
- Service de Psychiatrie, Centre Hospitalier Sainte-Anne, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris Descartes, Paris, France.,Service de Psychiatrie, Département de Neurosciences, Dijon, France
| | - E Haffen
- Laboratoire de génétique, CHU, PTB, 2 rue Angélique Ducoudray, Dijon, France.,Service de Psychiatrie, CHU de Besançon, Besançon, France.,Laboratoire de Neurosciences EA 481, IFR 133, Université of Franche-Comté, Besançon, France
| | - P Vandel
- Laboratoire de Neurosciences EA 481, IFR 133, Université of Franche-Comté, Besançon, France.,Centre d'Investigation Clinique CIC-IT 808 INSERM, CHU de Besaçon, Besançon, France
| | - B Bonin
- Service de Psychiatrie, Département de Neurosciences, Dijon, France.,Laboratoire de Psychologie et Psychopathologie Médicale (IFR 100), Université de Bourgogne, Dijon, France
| | | | - O Hermine
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders, Hôpital Necker-Enfants malades, Paris, France.,Service d'Hématologie Adulte, Université Paris Descartes, Sorbonne, Paris Cité, Assistance Publique-Hôpitaux de Paris, Institut Imagine, Hôpital Necker-Enfants malades, Paris, France
| | - R Gaillard
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,Laboratoire de "Physiopathologie des maladies Psychiatriques", Centre de Psychiatrie et Neurosciences U894, INSERM, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service de Psychiatrie, Centre Hospitalier Sainte-Anne, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris Descartes, Paris, France.,Human Histopathology and Animal Models, Infection and Epidemiology Department, Institut Pasteur, Paris, France
| |
Collapse
|
21
|
Lagard C, Chevillard L, Malissin I, Risède P, Callebert J, Labat L, Launay JM, Laplanche JL, Mégarbane B. Mechanisms of tramadol-related neurotoxicity in the rat: Does diazepam/tramadol combination play a worsening role in overdose? Toxicol Appl Pharmacol 2016; 310:108-119. [PMID: 27641627 DOI: 10.1016/j.taap.2016.09.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/30/2016] [Accepted: 09/14/2016] [Indexed: 11/29/2022]
Abstract
Poisoning with opioid analgesics including tramadol represents a challenge. Tramadol may induce respiratory depression, seizures and serotonin syndrome, possibly worsened when in combination to benzodiazepines. Our objectives were to investigate tramadol-related neurotoxicity, consequences of diazepam/tramadol combination, and mechanisms of drug-drug interactions in rats. Median lethal-doses were determined using Dixon-Bruce's up-and-down method. Sedation, seizures, electroencephalography and plethysmography parameters were studied. Concentrations of tramadol and its metabolites were measured using liquid-chromatography-high-resolution-mass-spectrometry. Plasma, platelet and brain monoamines were measured using liquid-chromatography coupled to fluorimetry. Median lethal-doses of tramadol and diazepam/tramadol combination did not significantly differ, although time-to-death was longer with combination (P=0.04). Tramadol induced dose-dependent sedation (P<0.05), early-onset seizures (P<0.001) and increase in inspiratory (P<0.01) and expiratory times (P<0.05). The diazepam/tramadol combination abolished seizures but significantly enhanced sedation (P<0.01) and respiratory depression (P<0.05) by reducing tidal volume (P<0.05) in addition to tramadol-related increase in respiratory times, suggesting a pharmacodynamic mechanism of interaction. Plasma M1 and M5 metabolites were mildly increased, contributing additionally to tramadol-related respiratory depression. Tramadol-induced early-onset increase in brain concentrations of serotonin and norepinephrine was not significantly altered by the diazepam/tramadol combination. Interestingly neither pretreatment with cyproheptadine (a serotonin-receptor antagonist) nor a benserazide/5-hydroxytryptophane combination (enhancing brain serotonin) reduced tramadol-induced seizures. Our study shows that diazepam/tramadol combination does not worsen tramadol-induced fatality risk but alters its toxicity pattern with enhanced respiratory depression but abolished seizures. Drug-drug interaction is mainly pharmacodynamic but increased plasma M1 and M5 metabolites may also contribute to enhancing respiratory depression. Tramadol-induced seizures are independent of brain serotonin.
Collapse
Affiliation(s)
- Camille Lagard
- Inserm, U1144, Paris, France; UMR-S 1144, Paris-Descartes University, Paris, France; UMR-S 1144, Paris-Diderot University, Paris, France.
| | - Lucie Chevillard
- Inserm, U1144, Paris, France; UMR-S 1144, Paris-Descartes University, Paris, France; UMR-S 1144, Paris-Diderot University, Paris, France.
| | - Isabelle Malissin
- Assistance Publique - Hôpitaux de Paris, Lariboisière Hospital, Department of Medical and Toxicological Critical Care, Paris, France.
| | - Patricia Risède
- Inserm, U1144, Paris, France; UMR-S 1144, Paris-Descartes University, Paris, France; UMR-S 1144, Paris-Diderot University, Paris, France.
| | - Jacques Callebert
- Inserm, U1144, Paris, France; UMR-S 1144, Paris-Descartes University, Paris, France; UMR-S 1144, Paris-Diderot University, Paris, France; Assistance Publique - Hôpitaux de Paris, Lariboisière Hospital, Laboratory of Biochemistry and Molecular Biology, Paris, France.
| | - Laurence Labat
- Inserm, U1144, Paris, France; UMR-S 1144, Paris-Descartes University, Paris, France; UMR-S 1144, Paris-Diderot University, Paris, France; Assistance Publique - Hôpitaux de Paris, Cochin Hospital, Laboratory of Toxicology, Paris, France.
| | - Jean-Marie Launay
- Assistance Publique - Hôpitaux de Paris, Lariboisière Hospital, Laboratory of Biochemistry and Molecular Biology, Paris, France; Inserm, U942, Paris, France.
| | - Jean-Louis Laplanche
- Inserm, U1144, Paris, France; UMR-S 1144, Paris-Descartes University, Paris, France; UMR-S 1144, Paris-Diderot University, Paris, France; Assistance Publique - Hôpitaux de Paris, Lariboisière Hospital, Laboratory of Biochemistry and Molecular Biology, Paris, France.
| | - Bruno Mégarbane
- Inserm, U1144, Paris, France; UMR-S 1144, Paris-Descartes University, Paris, France; UMR-S 1144, Paris-Diderot University, Paris, France; Assistance Publique - Hôpitaux de Paris, Lariboisière Hospital, Department of Medical and Toxicological Critical Care, Paris, France.
| |
Collapse
|
22
|
Bergwerff CE, Luman M, Blom HJ, Oosterlaan J. No Tryptophan, Tyrosine and Phenylalanine Abnormalities in Children with Attention-Deficit/Hyperactivity Disorder. PLoS One 2016; 11:e0151100. [PMID: 26938936 PMCID: PMC4777504 DOI: 10.1371/journal.pone.0151100] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 02/23/2016] [Indexed: 01/18/2023] Open
Abstract
Background The aim of the current study was to explore the role of aromatic amino acids (AAAs) in blood in relation to attention-deficit/hyperactivity disorder (ADHD). Given their impact on the synthesis of serotonin and dopamine, decreased concentrations of the AAAs tryptophan, tyrosine and phenylalanine in blood may contribute to the expression of ADHD symptoms. Decreased AAA blood concentrations, in turn, may be related to lowered dietary protein intake or to abnormal AAA catabolism, as evidenced by increased urinary AAA concentrations. Methods Eighty-three children with ADHD (75% males) and 72 typically developing (TD) children (51% males), aged 6 to 13 years, participated in the study. AAA concentrations were assessed in blood spots and an 18-hour urinary sample. A nutritional diary was filled out by parents to calculate dietary protein intake. Parent and teacher questionnaires assessed symptoms of ADHD, oppositional defiant disorder, conduct disorder, and autism spectrum disorder. Results Children with ADHD showed normal AAA concentrations in blood spots and urine, as well as normal protein intake compared to controls. No associations between AAA concentrations and symptoms of ADHD or comorbid psychiatric disorders were found. Conclusions This study is the first to explore AAA metabolism in children with ADHD using a well-defined and relatively large sample. We found that AAA deficiencies are not related to ADHD. The results do not support treatment with AAA supplements in children with ADHD. Future studies regarding the cause of serotonin and dopamine alterations in ADHD should focus on other explanations, such as effects of altered transport of AAAs.
Collapse
Affiliation(s)
| | - Marjolein Luman
- Clinical Neuropsychology section, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Henk J. Blom
- Center for Pediatrics and Adolescent Medicine, Medical Center–University of Freiburg, Freiburg, Germany
- Department of Clinical Chemistry, VU University Medical Center Amsterdam, Amsterdam, the Netherlands
| | - Jaap Oosterlaan
- Clinical Neuropsychology section, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
23
|
Georgin-Lavialle S, Launay JM, Côté F, Soucié E, Soria A, Damaj G, Moura DS, Canioni D, Hanssens K, Chandesris MO, Barète S, Dubreuil P, Lortholary O, Hermine O, Sokol H. Decreased tryptophan and increased kynurenine levels in mastocytosis associated with digestive symptoms. Allergy 2016; 71:416-20. [PMID: 26841279 DOI: 10.1111/all.12802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2015] [Indexed: 12/23/2022]
Abstract
The main metabolism pathway of tryptophan is protein formation, but it can also be metabolized into serotonin and kynurenine. Indoleamine 2,3-dioxygenase (IDO) is the enzyme that catalyzes the degradation of tryptophan into kynurenine. Mastocytosis is a heterogeneous disease characterized by mast cell accumulation in various tissues with 57% of patients having gastrointestinal involvement. We studied tryptophan metabolism in mastocytosis patients displaying or not gastrointestinal features and healthy subjects (n = 26 in each group). Mastocytosis patients with digestive symptoms displayed significantly increased kynurenine level and IDO activity as compared to healthy controls and mastocytosis patients without digestive symptoms. This could be linked to mast cell-mediated digestive inflammation among patients with mastocytosis. This work is the first focusing on kynurenine pathway in a mast cell disease and could help to understand the pathogenesis of digestive features in mastocytosis as well as in other mast cell-mediated diseases.
Collapse
Affiliation(s)
- S. Georgin-Lavialle
- Service de Médecine Interne; Hôpital Tenon; Assistance Publique-Hôpitaux de Paris; Université Pierre et Marie Curie-Paris 6; Paris France
- Centre de Référence des Mastocytoses; Sorbonne, Paris Cité; Hôpital Necker Enfants malades; Institut Imagine; Université Paris Descartes; Paris France
| | - J.-M. Launay
- Laboratoire de Biochimie et Biologie Moléculaire; Hôpital Lariboisière; Assistance Publique-Hôpitaux de Paris; INSERM U942; RTRS Santé Mentale; Paris France
| | - F. Côté
- INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders Hôpital Necker-Enfants malades; Institut Imagine; Paris Cedex 15 France
| | - E. Soucié
- INSERM UMR 891; Centre de Recherche en Cancérologie de Marseille; Laboratoire d'Hématopoïèse Moléculaire et Fonctionnelle; Marseille France
| | - A. Soria
- Centre de Référence des Mastocytoses; Sorbonne, Paris Cité; Hôpital Necker Enfants malades; Institut Imagine; Université Paris Descartes; Paris France
| | - G. Damaj
- Centre de Référence des Mastocytoses; Sorbonne, Paris Cité; Hôpital Necker Enfants malades; Institut Imagine; Université Paris Descartes; Paris France
- Hématologie; Faculté de Médecine; Centre Hospitalier Universitaire; Caen France
| | - D. S. Moura
- Centre de Référence des Mastocytoses; Sorbonne, Paris Cité; Hôpital Necker Enfants malades; Institut Imagine; Université Paris Descartes; Paris France
| | - D. Canioni
- Centre de Référence des Mastocytoses; Sorbonne, Paris Cité; Hôpital Necker Enfants malades; Institut Imagine; Université Paris Descartes; Paris France
- Service d'Anatomie-Pathologique; Hôpital Necker-Enfants Malades; Assistance Publique-Hôpitaux de Paris; Université Paris Descartes; Paris France
| | - K. Hanssens
- INSERM UMR 891; Centre de Recherche en Cancérologie de Marseille; Laboratoire d'Hématopoïèse Moléculaire et Fonctionnelle; Marseille France
| | - M.-O. Chandesris
- Centre de Référence des Mastocytoses; Sorbonne, Paris Cité; Hôpital Necker Enfants malades; Institut Imagine; Université Paris Descartes; Paris France
- Service d'hématologie Adulte; Sorbonne, Paris Cité; Assistance Publique-Hôpitaux de Paris; Institut Imagine; Hôpital Necker-Enfants Malades; Université Paris Descartes; Paris France
| | - S. Barète
- Centre de Référence des Mastocytoses; Sorbonne, Paris Cité; Hôpital Necker Enfants malades; Institut Imagine; Université Paris Descartes; Paris France
| | - P. Dubreuil
- Centre de Référence des Mastocytoses; Sorbonne, Paris Cité; Hôpital Necker Enfants malades; Institut Imagine; Université Paris Descartes; Paris France
- INSERM UMR 891; Centre de Recherche en Cancérologie de Marseille; Laboratoire d'Hématopoïèse Moléculaire et Fonctionnelle; Marseille France
| | - O. Lortholary
- Centre de Référence des Mastocytoses; Sorbonne, Paris Cité; Hôpital Necker Enfants malades; Institut Imagine; Université Paris Descartes; Paris France
- Sorbonne, Paris Cité; Service des Maladies Infectieuses et Tropicales; Assistance Publique-Hôpitaux de Paris; Hôpital Necker-Enfants Malades; IHU Imagine; Université Paris Descartes; Paris France
| | - O. Hermine
- Centre de Référence des Mastocytoses; Sorbonne, Paris Cité; Hôpital Necker Enfants malades; Institut Imagine; Université Paris Descartes; Paris France
- Service d'hématologie Adulte; Sorbonne, Paris Cité; Assistance Publique-Hôpitaux de Paris; Institut Imagine; Hôpital Necker-Enfants Malades; Université Paris Descartes; Paris France
| | - H. Sokol
- Service de Gastroentérologie et Nutrition; Hôpital Saint-Antoine; AP-HP; Université Pierre et Marie Curie-Paris 6; Paris France
- Equipe AVENIR; Laboratoire INSERM U1157/UMR CNRS 7203; Université Pierre et Marie Curie 6; Paris France
- Equipe Interactions des Bactéries Commensales et Probiotiques Avec l'hôte; INRA; MICALIS; Jouy en Josas France
| |
Collapse
|
24
|
Hasni Ebou M, Singh-Estivalet A, Launay JM, Callebert J, Tronche F, Ferré P, Gautier JF, Guillemain G, Bréant B, Blondeau B, Riveline JP. Glucocorticoids Inhibit Basal and Hormone-Induced Serotonin Synthesis in Pancreatic Beta Cells. PLoS One 2016; 11:e0149343. [PMID: 26901633 PMCID: PMC4763453 DOI: 10.1371/journal.pone.0149343] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 01/29/2016] [Indexed: 12/31/2022] Open
Abstract
Diabetes is a major complication of chronic Glucocorticoids (GCs) treatment. GCs induce insulin resistance and also inhibit insulin secretion from pancreatic beta cells. Yet, a full understanding of this negative regulation remains to be deciphered. In the present study, we investigated whether GCs could inhibit serotonin synthesis in beta cell since this neurotransmitter has been shown to be involved in the regulation of insulin secretion. To this aim, serotonin synthesis was evaluated in vitro after treatment with GCs of either islets from CD1 mice or MIN6 cells, a beta-cell line. We also explored the effect of GCs on the stimulation of serotonin synthesis by several hormones such as prolactin and GLP 1. We finally studied this regulation in islet in two in vivo models: mice treated with GCs and with liraglutide, a GLP1 analog, and mice deleted for the glucocorticoid receptor in the pancreas. We showed in isolated islets and MIN6 cells that GCs decreased expression and activity of the two key enzymes of serotonin synthesis, Tryptophan Hydroxylase 1 (Tph1) and 2 (Tph2), leading to reduced serotonin contents. GCs also blocked the induction of serotonin synthesis by prolactin or by a previously unknown serotonin activator, the GLP-1 analog exendin-4. In vivo, activation of the Glucagon-like-Peptide-1 receptor with liraglutide during 4 weeks increased islet serotonin contents and GCs treatment prevented this increase. Finally, islets from mice deleted for the GR in the pancreas displayed an increased expression of Tph1 and Tph2 and a strong increased serotonin content per islet. In conclusion, our results demonstrate an original inhibition of serotonin synthesis by GCs, both in basal condition and after stimulation by prolactin or activators of the GLP-1 receptor. This regulation may contribute to the deleterious effects of GCs on beta cells.
Collapse
Affiliation(s)
- Moina Hasni Ebou
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Amrit Singh-Estivalet
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Jean-Marie Launay
- INSERM U942, Assistance Publique–Hôpitaux de Paris (AP-HP), Hôpital Lariboisière, Service de Biochimie, Paris, France
| | - Jacques Callebert
- INSERM U942, Assistance Publique–Hôpitaux de Paris (AP-HP), Hôpital Lariboisière, Service de Biochimie, Paris, France
| | - François Tronche
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- CNRS UMR INSERM 952-CNRS 7224, Paris, France
| | - Pascal Ferré
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Jean-François Gautier
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Department of Diabetes and Endocrinology, Hôpital Lariboisière, AP-HP, Paris, France
- Université Paris Diderot, Paris, France
| | - Ghislaine Guillemain
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Bernadette Bréant
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Bertrand Blondeau
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- * E-mail:
| | - Jean-Pierre Riveline
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Department of Diabetes and Endocrinology, Hôpital Lariboisière, AP-HP, Paris, France
| |
Collapse
|
25
|
Individual behavioral and neurochemical markers of unadapted decision-making processes in healthy inbred mice. Brain Struct Funct 2016; 221:4615-4629. [PMID: 26860089 PMCID: PMC5102946 DOI: 10.1007/s00429-016-1192-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 01/20/2016] [Indexed: 02/03/2023]
Abstract
One of the hallmarks of decision-making processes is the inter-individual variability between healthy subjects. These behavioral patterns could constitute risk factors for the development of psychiatric disorders. Therefore, finding predictive markers of safe or risky decision-making is an important challenge for psychiatry research. We set up a mouse gambling task (MGT)-adapted from the human Iowa gambling task with uncertain contingencies between response and outcome that furthermore enables the emergence of inter-individual differences. Mice (n = 54) were further individually characterized for locomotive, emotional and cognitive behavior. Individual basal rates of monoamines and brain activation after the MGT were assessed in brain regions related to reward, emotion or cognition. In a large healthy mice population, 44 % showed a balanced strategy with limited risk-taking and flexible choices, 29 % showed a safe but rigid strategy, while 27 % adopted risky behavior. Risky mice took also more risks in other apparatus behavioral devices and were less sensitive to reward. No difference existed between groups regarding anxiety, working memory, locomotion and impulsivity. Safe/rigid mice exhibited a hypoactivation of prefrontal subareas, a high level of serotonin in the orbitofrontal cortex combined with a low level of dopamine in the putamen that predicted the emergence of rigid behavior. By contrast, high levels of dopamine, serotonin and noradrenalin in the hippocampus predicted the emergence of more exploratory and risky behaviors. The coping of C57bl/6J mice in MGT enables the determination of extreme patterns of choices either safe/rigid or risky/flexible, related to specific neurochemical and behavioral markers.
Collapse
|
26
|
Bouma G, van Faassen M, Kats-Ugurlu G, de Vries EGE, Kema IP, Walenkamp AME. Niacin (Vitamin B3) Supplementation in Patients with Serotonin-Producing Neuroendocrine Tumor. Neuroendocrinology 2016; 103:489-94. [PMID: 26335390 DOI: 10.1159/000440621] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 08/21/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND/AIMS Tryptophan is the precursor of serotonin and niacin (vitamin B3). The latter is critical for normal cellular metabolism. Tryptophan and niacin can be deficient in patients with serotonin-producing neuroendocrine tumors (NETs). Niacin deficiency may lead to severe symptoms including pellagra. In patients with serotonin-producing NET, data on niacin status are scarce and niacin supplementation hardly receives attention. We aimed to assess the niacin status before and after supplementation in these patients. METHODS We identified serotonin-producing NET patients who had received oral niacin supplementation (mean dose 144 mg daily) for tryptophan deficiency and/or pellagra-associated symptoms. Presupplementation plasma tryptophan levels and niacin status based on the urinary niacin metabolite N1-methylnicotinamide (N1-MN) before (n = 42) and after the start of the supplementation (in 34 paired samples) were assessed. Reference values for urinary N1-MN levels were established in 133 healthy individuals. RESULTS The mean presupplementation plasma tryptophan level was 31.8 ± 9.7 µmol/l (reference value 40.0-70.0). Presupplementation urinary N1-MN levels were lower in patients (median 17.9 µmol/24 h, range 2.6-70.3) compared to healthy controls (median 43.7 µmol/24 h, range 9.5-169.3, p < 0.0001) and below normal in 45% of the patients. Niacin supplementation increased urinary N1-MN levels to high normal levels (median 55.5 µmol/24 h, range 7.4-489.0) in 86% of the niacin-deficient patients. CONCLUSION In serotonin-producing NET patients, niacin deficiency is prevalent. Therefore, urinary N1-MN deserves to be included in their standard biochemical evaluation. Niacin supplementation normalizes the niacin status in most niacin-deficient serotonin-producing NET patients. A prospective study is warranted.
Collapse
|
27
|
Meunier CNJ, Callebert J, Cancela JM, Fossier P. Effect of dopaminergic D1 receptors on plasticity is dependent of serotoninergic 5-HT1A receptors in L5-pyramidal neurons of the prefrontal cortex. PLoS One 2015; 10:e0120286. [PMID: 25775449 PMCID: PMC4361673 DOI: 10.1371/journal.pone.0120286] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 02/02/2015] [Indexed: 01/11/2023] Open
Abstract
Major depression and schizophrenia are associated with dysfunctions of serotoninergic and dopaminergic systems mainly in the prefrontal cortex (PFC). Both serotonin and dopamine are known to modulate synaptic plasticity. 5-HT1A receptors (5-HT1ARs) and dopaminergic type D1 receptors are highly represented on dendritic spines of layer 5 pyramidal neurons (L5PyNs) in PFC. How these receptors interact to tune plasticity is poorly understood. Here we show that D1-like receptors (D1Rs) activation requires functional 5HT1ARs to facilitate LTP induction at the expense of LTD. Using 129/Sv and 5-HT1AR-KO mice, we recorded post-synaptic currents evoked by electrical stimulation in layer 2/3 after activation or inhibition of D1Rs. High frequency stimulation resulted in the induction of LTP, LTD or no plasticity. The D1 agonist markedly enhanced the NMDA current in 129/Sv mice and the percentage of L5PyNs displaying LTP was enhanced whereas LTD was reduced. In 5-HT1AR-KO mice, the D1 agonist failed to increase the NMDA current and orientated the plasticity towards L5PyNs displaying LTD, thus revealing a prominent role of 5-HT1ARs in dopamine-induced modulation of plasticity. Our data suggest that in pathological situation where 5-HT1ARs expression varies, dopaminergic treatment used for its ability to increase LTP could turn to be less and less effective.
Collapse
Affiliation(s)
- Claire Nicole Jeanne Meunier
- Neuroscience Paris-Saclay Institute (NeuroPSI), UMR 8197 CNRS-Université Paris-Sud, Bâtiment 446, Université Paris-Sud, Orsay F-91405, France
| | - Jacques Callebert
- Université Paris Descartes, Laboratoire de Neuropharmacologie des addictions, INSERM U705 CNRS UMR 7157, 4 avenue de l’Observatoire, 75006 Paris, France
| | - José-Manuel Cancela
- Neuroscience Paris-Saclay Institute (NeuroPSI), UMR 8197 CNRS-Université Paris-Sud, Bâtiment 446, Université Paris-Sud, Orsay F-91405, France
| | - Philippe Fossier
- Neuroscience Paris-Saclay Institute (NeuroPSI), UMR 8197 CNRS-Université Paris-Sud, Bâtiment 446, Université Paris-Sud, Orsay F-91405, France
- * E-mail:
| |
Collapse
|
28
|
The serotonin-N-acetylserotonin-melatonin pathway as a biomarker for autism spectrum disorders. Transl Psychiatry 2014; 4:e479. [PMID: 25386956 PMCID: PMC4259991 DOI: 10.1038/tp.2014.120] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/21/2014] [Accepted: 10/05/2014] [Indexed: 12/27/2022] Open
Abstract
Elevated whole-blood serotonin and decreased plasma melatonin (a circadian synchronizer hormone that derives from serotonin) have been reported independently in patients with autism spectrum disorders (ASDs). Here, we explored, in parallel, serotonin, melatonin and the intermediate N-acetylserotonin (NAS) in a large cohort of patients with ASD and their relatives. We then investigated the clinical correlates of these biochemical parameters. Whole-blood serotonin, platelet NAS and plasma melatonin were assessed in 278 patients with ASD, their 506 first-degree relatives (129 unaffected siblings, 199 mothers and 178 fathers) and 416 sex- and age-matched controls. We confirmed the previously reported hyperserotonemia in ASD (40% (35-46%) of patients), as well as the deficit in melatonin (51% (45-57%)), taking as a threshold the 95th or 5th percentile of the control group, respectively. In addition, this study reveals an increase of NAS (47% (41-54%) of patients) in platelets, pointing to a disruption of the serotonin-NAS-melatonin pathway in ASD. Biochemical impairments were also observed in the first-degree relatives of patients. A score combining impairments of serotonin, NAS and melatonin distinguished between patients and controls with a sensitivity of 80% and a specificity of 85%. In patients the melatonin deficit was only significantly associated with insomnia. Impairments of melatonin synthesis in ASD may be linked with decreased 14-3-3 proteins. Although ASDs are highly heterogeneous, disruption of the serotonin-NAS-melatonin pathway is a very frequent trait in patients and may represent a useful biomarker for a large subgroup of individuals with ASD.
Collapse
|
29
|
Simultaneous quantification of 5-hydroxyindoleacetic acid and 5-hydroxytryptamine by capillary electrophoresis with quantum dot and horseradish peroxidase enhanced chemiluminescence detection. J Chromatogr B Analyt Technol Biomed Life Sci 2014; 967:190-4. [DOI: 10.1016/j.jchromb.2014.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 06/09/2014] [Accepted: 07/02/2014] [Indexed: 11/23/2022]
|
30
|
Gauthier C, Hassler C, Mattar L, Launay JM, Callebert J, Steiger H, Melchior JC, Falissard B, Berthoz S, Mourier-Soleillant V, Lang F, Delorme M, Pommereau X, Gerardin P, Bioulac S, Bouvard M, Godart N. Symptoms of depression and anxiety in anorexia nervosa: links with plasma tryptophan and serotonin metabolism. Psychoneuroendocrinology 2014; 39:170-178. [PMID: 24135616 DOI: 10.1016/j.psyneuen.2013.09.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 08/13/2013] [Accepted: 09/08/2013] [Indexed: 11/17/2022]
Abstract
Depressive, anxiety and obsessive symptoms frequently co-occur with anorexia nervosa (AN). The relationship between these clinical manifestations and the biological changes caused by starvation is not well understood. It has been hypothesised that reduced availability of tryptophan (TRP) could reduce serotonin activity and thus trigger these comorbid symptoms. The aim of this study, during re-feeding in individuals with AN, was to analyse covariations across measures of nutritional status, depressive and anxiety symptoms, and peripheral serotonin markers. Depressive and anxiety symptoms, nutritional status and serotonin markers--whole blood serotonin content, plasma TRP and the ratio between TRP and large neutral amino acids--were assessed for 42 AN participants at admission to inpatient treatment and after re-feeding. Biological measures were compared to those obtained in 42 non-eating disordered subjects. For those with AN, psychological, nutritional and biological parameters improved significantly during hospitalisation. Levels of serotonin markers were significantly lower in the AN group compared to the control group, at admission and at discharge. Increase in the TRP/LNAA ratio was correlated with a decrease in depressive symptoms. In addition, there was a positive correlation between serotonin levels and symptoms of both anxiety and depression at discharge. We speculate that enhanced TRP availability during re-feeding, as a result of the increase in the TRP/LNAA ratio, could restore serotonin neurotransmission and lead to a decrease in depressive symptoms. The association between serotonin and anxiety and depressive symptoms would be consistent with numerous observations indicating abnormal functioning of the serotoninergic system in AN.
Collapse
Affiliation(s)
- Claire Gauthier
- Département de Psychiatrie, Institut Mutualiste Montsouris, Paris, France; Faculté de Médecine, Université Paris Descartes, Paris, France; Inserm U669, Paris, Université Paris Descartes, Paris, France.
| | | | - Lama Mattar
- Département de Psychiatrie, Institut Mutualiste Montsouris, Paris, France; Faculté de Médecine, Université Paris Descartes, Paris, France; Inserm U669, Paris, Université Paris Descartes, Paris, France
| | - Jean-Marie Launay
- APHP, Hôpital Lariboisière, Service de Biochimie et de Biologie Moléculaire, Paris, France; INSERM U942, Biomarqueurs et Insuffisance cardiaque, Hôpital Lariboisière, Paris, France
| | - Jacques Callebert
- APHP, Hôpital Lariboisière, Service de Biochimie et de Biologie Moléculaire, Paris, France; INSERM U942, Biomarqueurs et Insuffisance cardiaque, Hôpital Lariboisière, Paris, France
| | - Howard Steiger
- Eating Disorders Program, Douglas University Institute, Verdun, Quebec, Canada; Psychiatry Department, McGill University, Montreal, Quebec, Canada
| | - Jean-Claude Melchior
- Unité de Nutrition clinique-Maladies infectieuses, Département de médecine aiguë spécialisée, Hôpital Raymond Poincarré, Garches, France
| | - Bruno Falissard
- Inserm U669, Paris, Université Paris Descartes, Paris, France
| | - Sylvie Berthoz
- Département de Psychiatrie, Institut Mutualiste Montsouris, Paris, France; Faculté de Médecine, Université Paris Descartes, Paris, France; Inserm U669, Paris, Université Paris Descartes, Paris, France
| | | | - François Lang
- Unité de Psychiatre, CHU Hôpital Nord, Saint-Etienne, France
| | - Marc Delorme
- Unité médico-psychologique de l'adolescent et du jeune adulte, CHU de Bordeaux, Bordeaux, France
| | - Xavier Pommereau
- Unité médico-psychologique de l'adolescent et du jeune adulte, CHU de Bordeaux, Bordeaux, France
| | - Priscille Gerardin
- Département de pédiatrie Médicale - Psychiatrie de l'enfant et de l'adolescent, CHU de Rouen, Rouen, France
| | | | | | - Nathalie Godart
- Département de Psychiatrie, Institut Mutualiste Montsouris, Paris, France; Faculté de Médecine, Université Paris Descartes, Paris, France; Inserm U669, Paris, Université Paris Descartes, Paris, France.
| |
Collapse
|
31
|
Serotonergic modulation of LTP at excitatory and inhibitory synapses in the developing rat visual cortex. Neuroscience 2013; 238:148-58. [DOI: 10.1016/j.neuroscience.2013.02.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 02/06/2013] [Accepted: 02/08/2013] [Indexed: 11/21/2022]
|
32
|
Gedde-Dahl M, Thiis-Evensen E, Tjølsen AM, Mordal KS, Vatn M, Bergestuen DS. Comparison of 24-h and overnight samples of urinary 5-hydroxyindoleacetic acid in patients with intestinal neuroendocrine tumors. Endocr Connect 2013; 2:50-4. [PMID: 23781318 PMCID: PMC3680961 DOI: 10.1530/ec-12-0077] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 12/14/2012] [Indexed: 01/19/2023]
Abstract
Neuroendocrine tumors (NETs) arising in the small intestine are known to produce vasoactive substances, including serotonin, that may result in the carcinoid syndrome (flushing, diarrhea, bronchoconstriction, and carcinoid heart disease). Measurement of the serotonin breakdown product 5-hydroxyindoleacetic acid (5-HIAA) in urine is important in diagnosing and monitoring of patients with intestinal NETs. Our aim was to compare 5-HIAA measurement in 24-h urine sampling with overnight (∼8-h) sampling in patients with known NETs, or at follow-up of patients potentially cured for their NETs. Twenty-four-hour and overnight urine samples were collected from 34 patients and analyzed for urinary 5-HIAA (U5-HIAA) using HPLC. Comparison of the overnight sampling values with the 24-h values showed no difference, P=0.45, and there was a significant direct correlation between the two samples using linear regression (R=0.97, P<0.001). U5-HIAA sample collection during a nightly interval of ∼8 h appears to have the same accuracy as the 24-h collection in this group of patients.
Collapse
|
33
|
Beaune S, Callebert J, Baud FJ, Risède P, Juvin P, Mégarbane B. Mechanisms of high-dose citalopram-induced death in a rat model. Toxicology 2012; 302:248-54. [DOI: 10.1016/j.tox.2012.08.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 08/08/2012] [Accepted: 08/09/2012] [Indexed: 12/25/2022]
|
34
|
Razy-Krajka F, Brown ER, Horie T, Callebert J, Sasakura Y, Joly JS, Kusakabe TG, Vernier P. Monoaminergic modulation of photoreception in ascidian: evidence for a proto-hypothalamo-retinal territory. BMC Biol 2012; 10:45. [PMID: 22642675 PMCID: PMC3414799 DOI: 10.1186/1741-7007-10-45] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 05/29/2012] [Indexed: 12/12/2022] Open
Abstract
Background The retina of craniates/vertebrates has been proposed to derive from a photoreceptor prosencephalic territory in ancestral chordates, but the evolutionary origin of the different cell types making the retina is disputed. Except for photoreceptors, the existence of homologs of retinal cells remains uncertain outside vertebrates. Methods The expression of genes expressed in the sensory vesicle of the ascidian Ciona intestinalis including those encoding components of the monoaminergic neurotransmission systems, was analyzed by in situ hybridization or in vivo transfection of the corresponding regulatory elements driving fluorescent reporters. Modulation of photic responses by monoamines was studied by electrophysiology combined with pharmacological treatments. Results We show that many molecular characteristics of dopamine-synthesizing cells located in the vicinity of photoreceptors in the sensory vesicle of the ascidian Ciona intestinalis are similar to those of amacrine dopamine cells of the vertebrate retina. The ascidian dopamine cells share with vertebrate amacrine cells the expression of the key-transcription factor Ptf1a, as well as that of dopamine-synthesizing enzymes. Surprisingly, the ascidian dopamine cells accumulate serotonin via a functional serotonin transporter, as some amacrine cells also do. Moreover, dopamine cells located in the vicinity of the photoreceptors modulate the light-off induced swimming behavior of ascidian larvae by acting on alpha2-like receptors, instead of dopamine receptors, supporting a role in the modulation of the photic response. These cells are located in a territory of the ascidian sensory vesicle expressing genes found both in the retina and the hypothalamus of vertebrates (six3/6, Rx, meis, pax6, visual cycle proteins). Conclusion We propose that the dopamine cells of the ascidian larva derive from an ancestral multifunctional cell population located in the periventricular, photoreceptive field of the anterior neural tube of chordates, which also gives rise to both anterior hypothalamus and the retina in craniates/vertebrates. It also shows that the existence of multiple cell types associated with photic responses predates the formation of the vertebrate retina.
Collapse
Affiliation(s)
- Florian Razy-Krajka
- Neurobiology and Development, UPR, Institut de Neurobiologie Alfred Fessard, Centre National de la Recherche Scientifique, Gif-sur-Yvette, France
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Zinellu A, Sotgia S, Deiana L, Carru C. Reverse injection capillary electrophoresis UV detection for serotonin quantification in human whole blood. J Chromatogr B Analyt Technol Biomed Life Sci 2012; 895-896:182-5. [DOI: 10.1016/j.jchromb.2012.03.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 03/17/2012] [Accepted: 03/19/2012] [Indexed: 10/28/2022]
|
36
|
Huang GG, Cheng ML, Yang J. Metal Ion-Assisted Infrared Optical Sensor for Selective Determination of Tryptophan in Urine Samples. J CHIN CHEM SOC-TAIP 2011. [DOI: 10.1002/jccs.201190003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
37
|
Zhao J, Chen H, Ni P, Xu B, Luo X, Zhan Y, Gao P, Zhu D. Simultaneous determination of urinary tryptophan, tryptophan-related metabolites and creatinine by high performance liquid chromatography with ultraviolet and fluorimetric detection. J Chromatogr B Analyt Technol Biomed Life Sci 2011; 879:2720-5. [DOI: 10.1016/j.jchromb.2011.07.035] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 07/25/2011] [Accepted: 07/25/2011] [Indexed: 10/17/2022]
|
38
|
Laugeray A, Launay JM, Callebert J, Surget A, Belzung C, Barone PR. Evidence for a key role of the peripheral kynurenine pathway in the modulation of anxiety- and depression-like behaviours in mice: Focus on individual differences. Pharmacol Biochem Behav 2011; 98:161-8. [DOI: 10.1016/j.pbb.2010.12.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 11/26/2010] [Accepted: 12/04/2010] [Indexed: 01/11/2023]
|
39
|
Peripheral and cerebral metabolic abnormalities of the tryptophan-kynurenine pathway in a murine model of major depression. Behav Brain Res 2010; 210:84-91. [PMID: 20153778 DOI: 10.1016/j.bbr.2010.02.014] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 02/02/2010] [Accepted: 02/05/2010] [Indexed: 11/26/2022]
Abstract
Occurring both peripherally and centrally, the kynurenine pathway (KP) - an alternative pathway to 5-HT synthesis from tryptophan (TRP) - could be of particular value to better understand the link between peripheral changes of circulating levels of glucocorticoids (GC)/proinflammatory cytokines and altered neurotransmission observed in depressed patients. Indeed, it is activated by these mediators of stress and can produce several neuroactive compounds like quinolinic acid (QUIN) and kynurenic acid (KYNA) that can respectively increase and decrease glutamate concentration in brain. In order to characterize the role of both the peripheral and cerebral KP in the pathophysiology of depressive disorders, we used the Unpredictable Chronic Mild Stress (UCMS) to induce a depressive-like syndrome and we then measured the level of relevant TRP-KYN pathway metabolites: KYN, 3-hydroxykynurenine (3HK; precursor of QUIN) and KYNA. We also measured TRP-5HT pathway metabolites: TRP, 5-HT, 5-HIAA. We showed that UCMS increased TRP catabolism along the KP in the periphery. 5-HT and KYN were found to be strongly negatively correlated in all brain structures of control mice and of UCMS mice except in the hippocampus. More importantly we found that KYN was preferentially metabolized along the QUIN pathway at the subcortical level (amygdala/striatum) whereas, at the cortical level (cingulate cortex), the QUIN pathway was reduced. Considering the role of these metabolites on the glutamatergic neurotransmission, we propose that such KP alterations could participate to the cortical/subcortical glutamatergic alterations reported in depressed patients.
Collapse
|
40
|
de Jong WHA, Wilkens MHLI, de Vries EGE, Kema IP. Automated mass spectrometric analysis of urinary and plasma serotonin. Anal Bioanal Chem 2010; 396:2609-16. [PMID: 20140664 PMCID: PMC2841759 DOI: 10.1007/s00216-010-3466-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Revised: 11/09/2009] [Accepted: 01/08/2010] [Indexed: 11/25/2022]
Abstract
Serotonin emerges as crucial neurotransmitter and hormone in a growing number of different physiologic processes. Besides extensive serotonin production previously noted in patients with metastatic carcinoid tumors, serotonin now is implicated in liver cell regeneration and bone formation. The aim was to develop a rapid, sensitive, and highly selective automated on-line solid-phase extraction method coupled to high-performance liquid chromatography-tandem mass spectrometry (XLC-MS/MS) to quantify low serotonin concentrations in matrices such as platelet-poor plasma and urine. Fifty microliters plasma or 2.5 microL urine equivalent were pre-purified by automated on-line solid-phase extraction, using weak cation exchange. Chromatography of serotonin and its deuterated internal standard was performed with hydrophilic interaction chromatography. Mass spectrometric detection was operated in multiple reaction monitoring mode using a quadrupole tandem mass spectrometer with positive electrospray ionization. Serotonin concentrations were determined in platelet-poor plasma of metastatic carcinoid patients (n = 23) and healthy controls (n = 22). Urinary reference intervals were set by analyzing 24-h urine collections of 120 healthy subjects. Total run-time was 6 min. Intra- and inter-assay analytical variation were <10%. Linearity in the 0-7300 micromol/L calibration range was excellent (R(2) > 0.99). Quantification limits were 30 and 0.9 nmol/L in urine and plasma, respectively. Platelet-poor serotonin concentrations in metastatic carcinoid patients were significantly higher than in controls. The urinary reference interval was 10-78 micromol/mol creatinine. Serotonin analysis with sensitive and specific XLC-MS/MS overcomes limitations of conventional HPLC. This enables accurate quantification of serotonin for both routine diagnostic procedures and research in serotonin-related disorders.
Collapse
Affiliation(s)
- Wilhelmina H. A. de Jong
- Department of Laboratory Medicine, University Medical Center, Groningen, University of Groningen, P.O. Box 30001, 9700 RB Groningen, The Netherlands
| | - Marianne H. L. I. Wilkens
- Department of Laboratory Medicine, University Medical Center, Groningen, University of Groningen, P.O. Box 30001, 9700 RB Groningen, The Netherlands
| | - Elisabeth G. E. de Vries
- Department of Medical Oncology, University Medical Center, Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Ido P. Kema
- Department of Laboratory Medicine, University Medical Center, Groningen, University of Groningen, P.O. Box 30001, 9700 RB Groningen, The Netherlands
| |
Collapse
|
41
|
Drozdov I, Kidd M, Gustafsson BI, Svejda B, Joseph R, Pfragner R, Modlin IM. Autoregulatory effects of serotonin on proliferation and signaling pathways in lung and small intestine neuroendocrine tumor cell lines. Cancer 2009; 115:4934-45. [PMID: 19634160 DOI: 10.1002/cncr.24533] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND : Survival rates for gastrointestinal (GI) and bronchopulmonary (BP) neuroendocrine tumors (NETs) have not altered significantly (5-year survival rate: GI NETs, 64.1%; BP NETs, 87%-89%) in 30 years (from 1973 to 2004). No effective or specific antineoplastic agents are available to date, although somatostatin analogs inhibit NET 5-hydroxytryptophan (5-HT) secretion. Given the expression of 5-HT receptors on NETs, the authors hypothesized that 5-HT autoregulated NET proliferation. METHODS : Proliferation was evaluated in 3 NET cell lines using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide uptake; in addition, real-time polymerase chain reaction analyses and enzyme-linked immunosorbent assay studies were performed to delineate 5-HT-mediated signaling pathways. To determine the receptor and role of endogenous 5-HT production, the effects of ketanserin (5-HT receptor subtypes 2A and 2C [5-HT(2A/2C)]); ondansetron (5-HT(3)); and the suicide inhibitor of the rate-limiting enzyme for 5-HT synthesis, tryptophan hydroxylase (7-HTP) were investigated. RESULTS : Exogenously added 5-HT stimulated proliferation in atypical BP NET NCI-H720 cells (+50%; half-maximal stimulatory concentration [EC(50)] = 10 nM), in typical BP NET NCI-H727 cells (+40%; EC(50) = 0.01 nM), and in GI NET KRJ-I cells (+60%; EC(50) = 25 nM). In NCI-H720 cells, proliferation was inhibited by ketanserin (half-maximal inhibitory concentration [IC(50)] = 0.06 nM) and ondansetron (IC(50) = 0.4 nM) and also was inhibited by 7-HTP (IC(50) = 0.3 nM). In NCI-H727 cells, ketanserin and 7-HTP inhibited proliferation (IC(50) = 0.3 nM and IC(50) = 2.3 nM, respectively), whereas ondansetron had no effect. In KRJ-I cells, ketanserin (IC(50) = 0.1 nM) and 7-HTP (IC(50) = 0.6 nM), but not ondansetron, inhibited proliferation. In all cell lines, 5-HT activated proliferation through extracellular signal-regulated kinase 1 (ERK1) and ERK2 phosphorylation and c-Jun N-terminal kinase (JNK)-mediated pathways (c-JUN and Ki-67 transcription). An autoregulatory effect was indicated by the 7-HTP-mediated inhibition of extracellular 5-HT and downstream effects on NET proliferation. CONCLUSIONS : Lung and GI NET proliferation was autoregulated by 5-HT through alterations in ERK and JNK signaling. Targeting NET cells with 5-HT(2) receptor antagonists and 7-HTP reversed proliferation. The current results indicated that 5-HT(2) receptor subtype-specific antagonists may represent a viable antiproliferative therapeutic strategy. Cancer 2009. (c) 2009 American Cancer Society.
Collapse
Affiliation(s)
- Ignat Drozdov
- Gastrointestinal Pathobiology Research Group, Yale University School of Medicine, New Haven, Connecticut, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Launay JM, Del Pino M, Chironi G, Callebert J, Peoc'h K, Mégnien JL, Mallet J, Simon A, Rendu F. Smoking induces long-lasting effects through a monoamine-oxidase epigenetic regulation. PLoS One 2009; 4:e7959. [PMID: 19956754 PMCID: PMC2775922 DOI: 10.1371/journal.pone.0007959] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Accepted: 10/17/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Postulating that serotonin (5-HT), released from smoking-activated platelets could be involved in smoking-induced vascular modifications, we studied its catabolism in a series of 115 men distributed as current smokers (S), never smokers (NS) and former smokers (FS) who had stopped smoking for a mean of 13 years. METHODOLOGY/PRINCIPAL FINDINGS 5-HT, monoamine oxidase (MAO-B) activities and amounts were measured in platelets, and 5-hydroxyindolacetic acid (5-HIAA)--the 5-HT/MAO catabolite--in plasma samples. Both platelet 5-HT and plasma 5-HIAA levels were correlated with the 10-year cardiovascular Framingham relative risk (P<0.01), but these correlations became non-significant after adjustment for smoking status, underlining that the determining risk factor among those taken into account in the Framingham risk calculation was smoking. Surprisingly, the platelet 5-HT content was similar in S and NS but lower in FS with a parallel higher plasma level of 5-HIAA in FS. This was unforeseen since MAO-B activity was inhibited during smoking (P<0.00001). It was, however, consistent with a higher enzyme protein concentration found in S and FS than in NS (P<0.001). It thus appears that MAO inhibition during smoking was compensated by a higher synthesis. To investigate the persistent increase in MAO-B protein concentration, a study of the methylation of its gene promoter was undertaken in a small supplementary cohort of similar subjects. We found that the methylation frequency of the MAOB gene promoter was markedly lower (P<0.0001) for S and FS vs. NS due to cigarette smoke-induced increase of nucleic acid demethylase activity. CONCLUSIONS/SIGNIFICANCE This is one of the first reports that smoking induces an epigenetic modification. A better understanding of the epigenome may help to further elucidate the physiopathology and the development of new therapeutic approaches to tobacco addiction. The results could have a larger impact than cardiovascular damage, considering that MAO-dependent 5-HT catabolism is also involved in addiction, predisposition to cancer, behaviour and mental health.
Collapse
Affiliation(s)
- Jean-Marie Launay
- Service de Biochimie et Biologie Moléculaire/Equipe Associée (EA) 3621, Assistance Publique des Hôpitaux de Paris (AP-HP), Hôpital Lariboisière, Faculté de Pharmacie, Université Paris Descartes, Paris, France
| | - Muriel Del Pino
- Signalisation cellulaire, dynamique circulatoire et athérosclérose précoce, Unité Mixte de Recherche (UMR) 7131, Université Pierre et Marie Curie (UPMC) Paris Universitas/Centre Nationale de la Recherche Scientifique (CNRS), Hôpital Broussais, Paris, France
| | - Gilles Chironi
- Signalisation cellulaire, dynamique circulatoire et athérosclérose précoce, Unité Mixte de Recherche (UMR) 7131, Université Pierre et Marie Curie (UPMC) Paris Universitas/Centre Nationale de la Recherche Scientifique (CNRS), Hôpital Broussais, Paris, France
- Centre de médecine préventive cardiovasculaire, AP-HP, Hôpital Européen Georges Pompidou-Broussais, Paris, France
| | - Jacques Callebert
- Service de Biochimie et Biologie Moléculaire/Equipe Associée (EA) 3621, Assistance Publique des Hôpitaux de Paris (AP-HP), Hôpital Lariboisière, Faculté de Pharmacie, Université Paris Descartes, Paris, France
| | - Katell Peoc'h
- Service de Biochimie et Biologie Moléculaire/Equipe Associée (EA) 3621, Assistance Publique des Hôpitaux de Paris (AP-HP), Hôpital Lariboisière, Faculté de Pharmacie, Université Paris Descartes, Paris, France
| | - Jean-Louis Mégnien
- Signalisation cellulaire, dynamique circulatoire et athérosclérose précoce, Unité Mixte de Recherche (UMR) 7131, Université Pierre et Marie Curie (UPMC) Paris Universitas/Centre Nationale de la Recherche Scientifique (CNRS), Hôpital Broussais, Paris, France
- Centre de médecine préventive cardiovasculaire, AP-HP, Hôpital Européen Georges Pompidou-Broussais, Paris, France
| | - Jacques Mallet
- Laboratoire de Génétique Moléculaire de la Neurotransmission et des Processus Neurodégénératifs, UMR 7091, UPMC Paris Universitas/CNRS, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Alain Simon
- Signalisation cellulaire, dynamique circulatoire et athérosclérose précoce, Unité Mixte de Recherche (UMR) 7131, Université Pierre et Marie Curie (UPMC) Paris Universitas/Centre Nationale de la Recherche Scientifique (CNRS), Hôpital Broussais, Paris, France
- Centre de médecine préventive cardiovasculaire, AP-HP, Hôpital Européen Georges Pompidou-Broussais, Paris, France
| | - Francine Rendu
- Signalisation cellulaire, dynamique circulatoire et athérosclérose précoce, Unité Mixte de Recherche (UMR) 7131, Université Pierre et Marie Curie (UPMC) Paris Universitas/Centre Nationale de la Recherche Scientifique (CNRS), Hôpital Broussais, Paris, France
- Faculte de Medecine Pitie-Salpetriere, UMRS 956 Inserm, INSERM, Paris, France
- * E-mail:
| |
Collapse
|
43
|
Trachte GJ, Uncini T, Hinz M. Both stimulatory and inhibitory effects of dietary 5-hydroxytryptophan and tyrosine are found on urinary excretion of serotonin and dopamine in a large human population. Neuropsychiatr Dis Treat 2009; 5:227-35. [PMID: 19557117 PMCID: PMC2695216 DOI: 10.2147/ndt.s5040] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Amino acid precursors of dopamine and serotonin have been administered for decades to treat a variety of clinical conditions including depression, anxiety, insomnia, obesity, and a host of other illnesses. Dietary administration of these amino acids is designed to increase dopamine and serotonin levels within the body, particularly the brain. Convincing evidence exists that these precursors normally elevate dopamine and serotonin levels within critical brain tissues and other organs. However, their effects on urinary excretion of neurotransmitters are described in few studies and the results appear equivocal. The purpose of this study was to define, as precisely as possible, the influence of both 5-hydroxytryptophan (5-HTP) and tyrosine on urinary excretion of serotonin and dopamine in a large human population consuming both 5-HTP and tyrosine. Curiously, only 5-HTP exhibited a marginal stimulatory influence on urinary serotonin excretion when 5-HTP doses were compared to urinary serotonin excretion; however, a robust relationship was observed when alterations in 5-HTP dose were compared to alterations in urinary serotonin excretion in individual patients. The data indicate three statistically discernible components to 5-HTP responses, including inverse, direct, and no relationships between urinary serotonin excretion and 5-HTP doses. The response to tyrosine was more consistent but primarily yielded an unexpected reduction in urinary dopamine excretion. These data indicate that the urinary excretion pattern of neurotransmitters after consumption of their precursors is far more complex than previously appreciated. These data on urinary neurotransmitter excretion might be relevant to understanding the effects of the precursors in other organs.
Collapse
Affiliation(s)
- George J Trachte
- Department of Physiology and Pharmacology, University of MN Medical School Duluth, Duluth, MN, USA.
| | | | | |
Collapse
|
44
|
Gregersen K, Frøyland L, Berstad A, Araujo P. Direct determination of serotonin in gut lavage fluid by liquid chromatographic ion trap tandem mass spectrometry. Talanta 2008; 75:466-72. [DOI: 10.1016/j.talanta.2007.11.043] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Revised: 11/10/2007] [Accepted: 11/14/2007] [Indexed: 01/20/2023]
|
45
|
Koopmans KP, Neels OC, Kema IP, Elsinga PH, Sluiter WJ, Vanghillewe K, Brouwers AH, Jager PL, de Vries EGE. Improved staging of patients with carcinoid and islet cell tumors with 18F-dihydroxy-phenyl-alanine and 11C-5-hydroxy-tryptophan positron emission tomography. J Clin Oncol 2008; 26:1489-95. [PMID: 18349401 DOI: 10.1200/jco.2007.15.1126] [Citation(s) in RCA: 196] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
PURPOSE To evaluate and compare diagnostic sensitivity of positron emission tomography (PET) scanning in carcinoid and islet cell tumor patients with a serotonin and a catecholamine precursor as tracers. PATIENTS AND METHODS Carcinoid (n = 24) or pancreatic islet cell tumor (n = 23) patients with at least one lesion on conventional imaging including somatostatin receptor scintigraphy (SRS) and computed tomography (CT) scan underwent (11)C-5-hydroxytryptophan ((11)C-5-HTP) PET and 6-[F-18]fluoro-L-dihydroxy-phenylalanine ((18)F-DOPA) PET. PET findings were compared with a composite reference standard derived from all available imaging along with clinical and cytologic/histologic information. RESULTS In carcinoid tumor patients, per-patient analysis showed sensitivities for (11)C-5-HTP PET, (18)F-DOPA PET, SRS, and CT of 100%, 96%, 86%, 96%, respectively, and in islet cell tumors of 100%, 89%, 78%, 87%, respectively. In carcinoid patients, per-lesion analysis revealed sensitivities for (11)C-5-HTP PET, (11)C-5-HTP PET/CT, (18)F-DOPA PET, (18)F-DOPA PET/CT, SRS, SRS/CT, and CT alone of, respectively, 78%, 89%, 87%, 98%, 49%, 73%, and 63% and in islet cell tumors of 67%, 96%, 41%, 80%, 46%, 77%, and 68%, respectively. In all carcinoid patients (18)F-DOPA PET and (11)C-5-HTP PET detected more lesions than SRS (P < .001). (11)C-5-HTP PET was superior to (18)F-DOPA PET in islet cell tumors (P < .0001). In all cases, CT improved the sensitivity of the nuclear scans. CONCLUSION (18)F-DOPA PET/CT is the optimal imaging modality for staging in carcinoid patients and (11)C-5-HTP PET/CT in islet cell tumor patients.
Collapse
Affiliation(s)
- Klaas P Koopmans
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Zuo XL, Li YQ, Yang XZ, Guo M, Guo YT, Lu XF, Li JM, Desmond PV. Plasma and gastric mucosal 5-hydroxytryptamine concentrations following cold water intake in patients with diarrhea-predominant irritable bowel syndrome. J Gastroenterol Hepatol 2007; 22:2330-7. [PMID: 18265445 DOI: 10.1111/j.1440-1746.2006.04772.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND AIM The purpose of the present paper was to investigate the effects of cold water intake on 5-hydroxytryptamine (5-HT) and its metabolite 5-hydroxyindole acetic acid (5-HIAA) in diarrhea-predominant irritable bowel syndrome (d-IBS) patients, and to observe the relationship between 5-HT and symptomatology. METHODS The plasma 5-HT/5-HIAA concentrations at 0, 30 min, 60 min, 90 min, 120 min, 150 min and 180 min following cold or warm water intake were investigated in 32 female subjects with d-IBS and 21 healthy female subjects. Gastric mucosal 5-HT under fasting conditions and following water intake were further investigated in 15 d-IBS patients and nine healthy subjects. Symptomatology was assessed throughout the study. RESULTS The plasma 5-HT concentrations in IBS patients were significantly higher than those of controls at 30 min (P = 0.022), 60 min (P < 0.001), 90 min (P < 0.001), 120 min (P < 0.001) and 150 min (P = 0.001) after cold water intake. The peak plasma 5-HT/5-HIAA and area under the curve for 5-HT/5-HIAA were also higher in d-IBS patients (P < 0.001). Gastric mucosal 5-HT in d-IBS patients and controls did not show any significant differences both under fasting condition (P = 0.596) and after cold water intake (P = 0.426). Last, the d-IBS patients with symptoms had higher 5-HT concentration (P < 0.001) and there was a positive correlation (r = 0.714, P = 0.001)between the symptomatology and plasma 5-HT level. CONCLUSIONS These data suggest that symptomatology following cold water intake may be associated with increased plasma 5-HT concentrations in female subjects with d-IBS.
Collapse
Affiliation(s)
- Xiu Li Zuo
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Real C, Popa D, Seif I, Callebert J, Launay JM, Adrien J, Escourrou P. Sleep apneas are increased in mice lacking monoamine oxidase A. Sleep 2007; 30:1295-302. [PMID: 17969463 PMCID: PMC2266281 DOI: 10.1093/sleep/30.10.1295] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
STUDY OBJECTIVES Alterations in the serotonin (5-HT) system have been suggested as a mechanism of sleep apnea in humans and rodents. The objective is to evaluate the contribution of 5-HT to this disorder. DESIGN We studied sleep and breathing (whole-body plethysmography) in mutant mice that lack monoamine oxidase A (MAOA) and have increased concentrations of monoamines, including 5-HT. MEASUREMENTS AND RESULTS Compared to wild-type mice, the mutants showed similar amounts of slow wave sleep (SWS) and rapid eye movement sleep (REMS), but exhibited a 3-fold increase in SWS and REMS apnea indices. Acute administration of the MAOA inhibitor clorgyline decreased REMS amounts and increased the apnea index in wild-type but not mutant mice. Parachlorophenylalanine, a 5-HT synthesis inhibitor, reduced whole brain concentrations of 5-HT in both strains, and induced a decrease in apnea index in mutant but not wild-type mice. CONCLUSION Our results show that MAOA deficiency is associated with increased sleep apnea in mice and suggest that an acute or chronic excess of 5-HT contributes to this phenotype.
Collapse
Affiliation(s)
- Caroline Real
- Univ Paris-Sud, EA3544, Châtenay-Malabry Cedex, F-92296, Sérotonine et Neuropharmacologie, France.
| | | | | | | | | | | | | |
Collapse
|
48
|
Vitalis T, Cases O, Passemard S, Callebert J, Parnavelas JG. Embryonic depletion of serotonin affects cortical development. Eur J Neurosci 2007; 26:331-44. [PMID: 17650110 DOI: 10.1111/j.1460-9568.2007.05661.x] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Compelling evidence suggests that serotonin (5-HT) is necessary for the refined organization of the cerebral cortex. Here we sought to analyse the short- and long-term consequences of embryonic 5-HT depletion on the development of the cerebral neocortex of the rat. We focused on the migration and differentiation of the pyramidal (projection) and nonpyramidal (interneuron) neuronal populations. Our paradigm used daily injection of DL-P-chlorophenylalanine (PCPA), a reversible inhibitor of 5-HT synthesis, during the E12-17 stage of embryonic development, when major events in corticogenesis take place. We monitored the 5-HT depletion induced by this treatment and showed that it led to subtle alterations in both the pyramidal and nonpyramidal neuronal populations. We found that E12-17 PCPA treatment altered the maturation of pyramidal neurons of layers III and V of the somatosensory cortex, with these cells displaying reduced dendritic arborization and complexity. These long-lasting alterations were not associated with modification of cortical BDNF levels at postnatal stages. We also showed that PCPA treatment transiently altered the incorporation in the cortical plate of interneurons derived from the caudal ganglionic eminence, and persistently affected the differentiation of a subpopulation expressing calretinin and/or cholecystokinin.
Collapse
Affiliation(s)
- Tania Vitalis
- Department of Anatomy and Developmental Biology, University College London, London, UK.
| | | | | | | | | |
Collapse
|
49
|
de Groot JWB, Kema IP, Breukelman H, van der Veer E, Wiggers T, Plukker JTM, Wolffenbuttel BHR, Links TP. Biochemical markers in the follow-up of medullary thyroid cancer. Thyroid 2006; 16:1163-70. [PMID: 17123344 DOI: 10.1089/thy.2006.16.1163] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Medullary thyroid cancer (MTC) shares biochemical features with other neuroendocrine tumors but the particular characteristics are largely unexplored. We investigated the biochemical neuroendocrine profile of MTC and whether specific markers could be useful in follow-up. In addition to the standard tumor marker calcitonin, plasma carcino-embryonic antigen (CEA), plasma catecholamines, (platelet) serotonin, chromogranin A, tryptase, and urinary markers of catecholamine, histamine, and serotonin metabolism were prospectively determined in 46 patients with histologically proven MTC. Patients were divided according to the stage of disease: group 1, no evidence; group 2, stable disease (SD); and group 3, progressive disease (PD). Plasma dopamine was increased in the majority of the patients with SD and PD; however it did not correlate with extent of disease. Elevated plasma platelet levels of serotonin were only present in patients with multiple endocrine neoplasia 2 with SD or PD but did not differ between those groups. Histamine metabolites were elevated in 20% of patients with SD and PD. In addition to plasma calcitonin, only CEA and chromogranin A could differentiate between stable and progressive MTC. MTCs are capable of synthesizing catecholamines, serotonin, and histamine metabolites underscoring that MTCs have metabolic characteristics in common with other neuroendocrine tumors. Thus far, clinical usefulness and relevance seems limited. The most useful markers in the follow-up of MTC are plasma calcitonin and CEA.
Collapse
Affiliation(s)
- Jan Willem B de Groot
- Department of Surgical Oncology, University Medical Center Groningen, University of Groningen, the Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Koopmans KP, de Vries EGE, Kema IP, Elsinga PH, Neels OC, Sluiter WJ, van der Horst-Schrivers ANA, Jager PL. Staging of carcinoid tumours with 18F-DOPA PET: a prospective, diagnostic accuracy study. Lancet Oncol 2006; 7:728-34. [PMID: 16945767 DOI: 10.1016/s1470-2045(06)70801-4] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND To assess individual treatment options for patients with carcinoid tumours, accurate knowledge of tumour localisation is essential. We aimed to test the diagnostic sensitivity of 6-[fluoride-18]fluoro-levodopa ((18)F-DOPA PET), compared with conventional imaging methods, in patients with carcinoid tumours. METHODS In a prospective, single-centre, diagnostic accuracy study, (18)F-DOPA PET with carbidopa pretreatment was compared with somatostatin-receptor scintigraphy (SRS), CT, and combined SRS and CT in 53 patients with a metastatic carcinoid tumour. The performance of all imaging methods was analysed for individual patients, for eight body regions, and for the detection of individual lesions. PET and CT images were fused to improve localisation. To produce a composite reference standard, we used cytological and histological findings; all imaging tests, including secondary assessments for newly found lesions; follow-up; and biochemical data. Sensitivities were calculated and compared. FINDINGS In patient-based analysis, we recorded sensitivities of 100% (95% CI 93-100) for (18)F-DOPA-PET, 92% (82-98) for SRS, 87% (75-95) for CT, and 96% (87-100) for combined SRS and CT (p=0.45 for (18)F-DOPA PET vs combined SRS and CT). However, (18)F-DOPA PET detected more lesions, more positive regions, and more lesions per region than combined SRS and CT. In region-based analysis, sensitivity of (18)F-DOPA PET was 95% (90-98) versus 66% (57-74) for SRS, 57% (48-66) for CT, and 79% (70-86) for combined SRS and CT (p=0.0001, PET vs combined SRS and CT). In individual-lesion analysis, corresponding sensitivities were 96% (95-98), 46% (43-50), 54% (51-58), and 65% (62-69; p<0.0001 for PET vs combined SRS and CT). INTERPRETATION If the improved tumour localisation seen with (18)F-DOPA-PET compared with conventional imaging is confirmed in future studies, this imaging method could replace use of SRS, help improve prediction of prognosis, and be used to assess patients' response to treatment for carcinoid tumours.
Collapse
Affiliation(s)
- Klaas P Koopmans
- Department of Nuclear Medicine and Molecular Imaging, University Medical Centre Groningen, Groningen, Netherlands
| | | | | | | | | | | | | | | |
Collapse
|