51
|
Polyák H, Cseh EK, Bohár Z, Rajda C, Zádori D, Klivényi P, Toldi J, Vécsei L. Cuprizone markedly decreases kynurenic acid levels in the rodent brain tissue and plasma. Heliyon 2021; 7:e06124. [PMID: 33553777 PMCID: PMC7856478 DOI: 10.1016/j.heliyon.2021.e06124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/13/2020] [Accepted: 01/25/2021] [Indexed: 12/21/2022] Open
Abstract
Background The kynurenine (KYN) pathway (KP) of the tryptophan (TRP) metabolism seems to play a role in the pathomechanism of multiple sclerosis (MS). Cuprizone (CPZ) treated animals develop both demyelination (DEM) and remyelination (REM) in lack of peripheral immune response, such as the lesion pattern type III and IV in MS, representing primary oligodendrogliopathy. Objective To measure the metabolites of the KP in the CPZ treated animals, including TRP, KYN and kynurenic acid (KYNA). We proposed that KYNA levels might be decreased in the CPZ-induced demyelinating phase of the animal model of MS, which model represents the progressive phase of the disease. Methods A total of 64 C57Bl/6J animals were used for the study. Immunohistochemical (IHC) measurements were performed to prove the effect of CPZ, whereas high-performance liquid chromatography (HPLC) was used to quantify the metabolites of the KP (n = 10/4 groups; DEM, CO1, REM, CO2). Results IHC measurements proved the detrimental effects of CPZ. HPLC measurements demonstrated a decrease of KYNA in the hippocampus (p < 0.05), somatosensory cortex (p < 0.01) and in plasma (p < 0.001). Conclusion This is the first evidence of marked reduction in KYNA levels in a non-immune mediated model of MS. Our results suggest an involvement of the KP in the pathomechanism of MS, which needs to be further elucidated.
Collapse
Affiliation(s)
- Helga Polyák
- Department of Neurology, Interdisciplinary Centre of Excellence, Faculty of Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, Szeged, Hungary
| | - Edina Katalin Cseh
- Department of Neurology, Interdisciplinary Centre of Excellence, Faculty of Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, Szeged, Hungary
| | - Zsuzsanna Bohár
- Department of Neurology, Interdisciplinary Centre of Excellence, Faculty of Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, Szeged, Hungary
- MTA-SZTE Neuroscience Research Group, Szeged, Hungary
| | - Cecilia Rajda
- Department of Neurology, Interdisciplinary Centre of Excellence, Faculty of Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, Szeged, Hungary
| | - Dénes Zádori
- Department of Neurology, Interdisciplinary Centre of Excellence, Faculty of Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, Szeged, Hungary
| | - Péter Klivényi
- Department of Neurology, Interdisciplinary Centre of Excellence, Faculty of Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, Szeged, Hungary
| | - József Toldi
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Interdisciplinary Centre of Excellence, Faculty of Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, Szeged, Hungary
- MTA-SZTE Neuroscience Research Group, Szeged, Hungary
- Corresponding author.
| |
Collapse
|
52
|
Lo YC, Lin CL, Fang WY, Lőrinczi B, Szatmári I, Chang WH, Fülöp F, Wu SN. Effective Activation by Kynurenic Acid and Its Aminoalkylated Derivatives on M-Type K + Current. Int J Mol Sci 2021; 22:1300. [PMID: 33525680 PMCID: PMC7865226 DOI: 10.3390/ijms22031300] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 12/24/2022] Open
Abstract
Kynurenic acid (KYNA, 4-oxoquinoline-2-carboxylic acid), an intermediate of the tryptophan metabolism, has been recognized to exert different neuroactive actions; however, the need of how it or its aminoalkylated amide derivative N-(2-(dimethylamino)ethyl)-3-(morpholinomethyl)-4-oxo-1,4-dihydroquinoline-2-carboxamide (KYNA-A4) exerts any effects on ion currents in excitable cells remains largely unmet. In this study, the investigations of how KYNA and other structurally similar KYNA derivatives have any adjustments on different ionic currents in pituitary GH3 cells and hippocampal mHippoE-14 neurons were performed by patch-clamp technique. KYNA or KYNA-A4 increased the amplitude of M-type K+ current (IK(M)) and concomitantly enhanced the activation time course of the current. The EC50 value required for KYNA- or KYNA-A4 -stimulated IK(M) was yielded to be 18.1 or 6.4 μM, respectively. The presence of KYNA or KYNA-A4 shifted the relationship of normalized IK(M)-conductance versus membrane potential to more depolarized potential with no change in the gating charge of the current. The voltage-dependent hysteretic area of IK(M) elicited by long-lasting triangular ramp pulse was observed in GH3 cells and that was increased during exposure to KYNA or KYNA-A4. In cell-attached current recordings, addition of KYNA raised the open probability of M-type K+ channels, along with increased mean open time of the channel. Cell exposure to KYNA or KYNA-A4 mildly inhibited delayed-rectifying K+ current; however, neither erg-mediated K+ current, hyperpolarization-activated cation current, nor voltage-gated Na+ current in GH3 cells was changed by KYNA or KYNA-A4. Under whole-cell, current-clamp recordings, exposure to KYNA or KYNA-A4 diminished the frequency of spontaneous action potentials; moreover, their reduction in firing frequency was attenuated by linopirdine, yet not by iberiotoxin or apamin. In hippocampal mHippoE-14 neurons, the addition of KYNA also increased the IK(M) amplitude effectively. Taken together, the actions presented herein would be one of the noticeable mechanisms through which they modulate functional activities of excitable cells occurring in vivo.
Collapse
Affiliation(s)
- Yi-Ching Lo
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (W.-Y.F.); (W.-H.C.)
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chih-Lung Lin
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan;
- Department of Neurosurgery, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wei-Yu Fang
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (W.-Y.F.); (W.-H.C.)
| | - Bálint Lőrinczi
- Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary; (B.L.); (I.S.); (F.F.)
| | - István Szatmári
- Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary; (B.L.); (I.S.); (F.F.)
| | - Wan-Hsuan Chang
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (W.-Y.F.); (W.-H.C.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ferenc Fülöp
- Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary; (B.L.); (I.S.); (F.F.)
- MTA-SZTE Stereochemistry Research Group, Hungarian Academy of Sciences, Eötvös u. 6, H-6720 Szeged, Hungary
| | - Sheng-Nan Wu
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan City 70101, Taiwan
- Department of Physiology, National Cheng Kung University Medical College, Tainan City 70101, Taiwan
| |
Collapse
|
53
|
Prefrontal α7nAChR Signaling Differentially Modulates Afferent Drive and Trace Fear Conditioning Behavior in Adolescent and Adult Rats. J Neurosci 2021; 41:1908-1916. [PMID: 33478990 DOI: 10.1523/jneurosci.1941-20.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/29/2020] [Accepted: 12/23/2020] [Indexed: 01/22/2023] Open
Abstract
Increased level of kynurenic acid is thought to contribute to the development of cognitive deficits in schizophrenia through an α7nAChR-mediated mechanism in the prefrontal cortex (PFC). However, it remains unclear to what extent disruption of PFC α7nAChR signaling impacts afferent transmission and its modulation of behavior. Using male rats, we found that PFC infusion of methyllycaconitine (MLA; α7nAChR antagonist) shifts ventral hippocampal-induced local field potential (LFP) suppression to LFP facilitation, an effect only observed in adults. Hippocampal stimulation can also elicit a GluN2B-mediated LFP potentiation (when PFC GABAAR is blocked) that is insensitive to MLA. Conversely, PFC infusion of MLA diminished the gain of amygdalar transmission, which is already enabled by postnatal day (P)30. Behaviorally, the impact of prefrontal MLA on trace fear-conditioning and extinction was also age related. While freezing behavior during conditioning was reduced by MLA only in adults, it elicited opposite effects in adolescent and adult rats during extinction as revealed by the level of reduced and increased freezing response, respectively. We next asked whether the late-adolescent onset of α7nAChR modulation of hippocampal inputs contributes to the age-dependent effect of MLA during extinction. Data revealed that the increased freezing behavior elicited by MLA in adult rats could be driven by a dysregulation of the GluN2B transmission in the PFC. Collectively, these results indicate that distinct neural circuits are recruited during the extinction of trace fear memory in adolescents and adults, likely because of the late-adolescent maturation of the ventral hippocampal-PFC functional connectivity and its modulation by α7nAChR signaling.SIGNIFICANCE STATEMENT Abnormal elevation of the astrocyte-derived metabolite kynurenic acid in the prefrontal cortex (PFC) is thought to impair cognitive functions in schizophrenia through an α7nAChR-mediated mechanism. Here, we found that prefrontal α7nAChR signaling is recruited to control the gain of hippocampal and amygdalar afferent transmission in an input-specific, age-related manner during the adolescent transition to adulthood. Behaviorally, prefrontal α7nAChR modulation of trace fear memory was also age-related, likely because of the late-adolescent maturation of the ventral hippocampal pathway and its recruitment of PFC GABAergic transmission enabled by local α7nAChR signaling. Collectively, these results reveal that distinct α7nAChR-sensitive neural circuits contribute to regulate behavior responses in adolescents and adults, particularly those requiring proper integration of hippocampal and amygdalar inputs by the PFC.
Collapse
|
54
|
Morales-Puerto N, Giménez-Gómez P, Pérez-Hernández M, Abuin-Martínez C, Gil de Biedma-Elduayen L, Vidal R, Gutiérrez-López MD, O'Shea E, Colado MI. Addiction and the kynurenine pathway: A new dancing couple? Pharmacol Ther 2021; 223:107807. [PMID: 33476641 DOI: 10.1016/j.pharmthera.2021.107807] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/05/2021] [Indexed: 12/15/2022]
Abstract
Drug use poses a serious threat to health systems throughout the world and the number of consumers rises relentlessly every year. The kynurenine pathway, main pathway of tryptophan degradation, has drawn interest in this field due to its relationship with addictive behaviour. Recently it has been confirmed that modulation of kynurenine metabolism at certain stages of the pathway can reduce, prevent or abolish drug seeking-like behaviours in studies with several different drugs. In this review, we present an up-to-date summary of the evidences of a relationship between drug use and the kynurenine pathway, both the alterations of the pathway due to drug use as well as modulation of the pathway as a potential approach to treat drug addiction. The review discusses ethanol, nicotine, cannabis, amphetamines, cocaine and opioids and new prospects in the drug research field are proposed.
Collapse
Affiliation(s)
- Nuria Morales-Puerto
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Pablo Giménez-Gómez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Mercedes Pérez-Hernández
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Cristina Abuin-Martínez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Leticia Gil de Biedma-Elduayen
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Rebeca Vidal
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - María Dolores Gutiérrez-López
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Esther O'Shea
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain.
| | - María Isabel Colado
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain.
| |
Collapse
|
55
|
Natural Molecules and Neuroprotection: Kynurenic Acid, Pantethine and α-Lipoic Acid. Int J Mol Sci 2021; 22:ijms22010403. [PMID: 33401674 PMCID: PMC7795784 DOI: 10.3390/ijms22010403] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/19/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
The incidence of neurodegenerative diseases has increased greatly worldwide due to the rise in life expectancy. In spite of notable development in the understanding of these disorders, there has been limited success in the development of neuroprotective agents that can slow the progression of the disease and prevent neuronal death. Some natural products and molecules are very promising neuroprotective agents because of their structural diversity and wide variety of biological activities. In addition to their neuroprotective effect, they are known for their antioxidant, anti-inflammatory and antiapoptotic effects and often serve as a starting point for drug discovery. In this review, the following natural molecules are discussed: firstly, kynurenic acid, the main neuroprotective agent formed via the kynurenine pathway of tryptophan metabolism, as it is known mainly for its role in glutamate excitotoxicity, secondly, the dietary supplement pantethine, that is many sided, well tolerated and safe, and the third molecule, α-lipoic acid is a universal antioxidant. As a conclusion, because of their beneficial properties, these molecules are potential candidates for neuroprotective therapies suitable in managing neurodegenerative diseases.
Collapse
|
56
|
Meier TB, España L, Nitta ME, Kent Teague T, Brett BL, Nelson LD, McCrea MA, Savitz J. Positive association between serum quinolinic acid and functional connectivity following concussion. Brain Behav Immun 2021; 91:531-540. [PMID: 33176183 PMCID: PMC7769223 DOI: 10.1016/j.bbi.2020.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/29/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
The molecular mechanisms underlying the diverse psychiatric and neuropathological sequalae documented in subsets of athletes with concussion have not been identified. We have previously reported elevated quinolinic acid (QuinA), a neurotoxic kynurenine pathway metabolite, acutely following concussion in football players with prior concussion. Similarly, work from our group and others has shown that increased functional connectivity strength, assessed using resting state fMRI, occurs following concussion and is associated with worse concussion-related symptoms and outcome. Moreover, other work has shown that repetitive concussion may have cumulative effects on functional connectivity and is a risk factor for adverse outcomes. Understanding the molecular mechanisms underlying these cumulative effects may ultimately be important for therapeutic interventions or the development of prognostic biomarkers. Thus, in this work, we tested the hypothesis that the relationship between QuinA in serum and functional connectivity following concussion would depend on the presence of a prior concussion. Concussed football players with prior concussion (N = 21) and without prior concussion (N = 16) completed a MRI session and provided a blood sample at approximately 1 days, 8 days, 15 days, and 45 days post-injury. Matched, uninjured football players with (N = 18) and without prior concussion (N = 24) completed similar visits. The association between QuinA and global connectivity strength differed based on group (F(3, 127) = 3.46, p = 0.019); post-hoc analyses showed a positive association between QuinA and connectivity strength in concussed athletes with prior concussion (B = 16.05, SE = 5.06, p = 0.002, 95%CI[6.06, 26.03]), but no relationship in concussed athletes without prior concussion or controls. Region-specific analyses showed that this association was strongest in bilateral orbitofrontal cortices, insulae, and basal ganglia. Finally, exploratory analyses found elevated global connectivity strength in concussed athletes with prior concussion who reported depressive symptoms at the 1-day visit compared to those who did not report depressive symptoms (t(15) = 2.37, mean difference = 13.50, SE = 5.69, p = 0.032, 95%CI[1.36, 25.63], Cohen's d = 1.15.). The results highlight a potential role of kynurenine pathway (KP) metabolites in altered functional connectivity following concussion and raise the possibility that repeated concussion has a "priming" effect on KP metabolism.
Collapse
Affiliation(s)
- Timothy B Meier
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States; Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, United States; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States.
| | - Lezlie España
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Morgan E Nitta
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States; Department of Psychology, Marquette University, Milwaukee, WI, United States
| | - T Kent Teague
- Departments of Surgery and Psychiatry, The University of Oklahoma School of Community Medicine, Tulsa, OK, United States; Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, Tulsa, OK, United States
| | - Benjamin L Brett
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States; Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Lindsay D Nelson
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States; Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Michael A McCrea
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States; Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa, OK, United States; Oxley College of Health Sciences, Tulsa, OK, United States
| |
Collapse
|
57
|
Jorratt P, Hoschl C, Ovsepian SV. Endogenous antagonists of N-methyl-d-aspartate receptor in schizophrenia. Alzheimers Dement 2020; 17:888-905. [PMID: 33336545 DOI: 10.1002/alz.12244] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/24/2020] [Indexed: 12/28/2022]
Abstract
Schizophrenia is a chronic neuropsychiatric brain disorder that has devastating personal impact and rising healthcare costs. Dysregulation of glutamatergic neurotransmission has been implicated in the pathobiology of the disease, attributed largely to the hypofunction of the N-methyl-d-aspartate (NMDA) receptor. Currently, there is a major gap in mechanistic analysis as to how endogenous modulators of the NMDA receptors contribute to the onset and progression of the disease. We present a systematic review of the neurobiology and the role of endogenous NMDA receptor antagonists in animal models of schizophrenia, and in patients. We discuss their neurochemical origin, release from neurons and glia with action mechanisms, and functional effects, which might contribute toward the impairment of neuronal processes underlying this complex pathological state. We consider clinical evidence suggesting dysregulations of endogenous NMDA receptor in schizophrenia, and highlight the pressing need in future studies and emerging directions, to restore the NMDA receptor functions for therapeutic benefits.
Collapse
Affiliation(s)
- Pascal Jorratt
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague 10, Czech Republic
| | - Cyril Hoschl
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague 10, Czech Republic
| | - Saak V Ovsepian
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague 10, Czech Republic
| |
Collapse
|
58
|
Seckler JM, Lewis SJ. Advances in D-Amino Acids in Neurological Research. Int J Mol Sci 2020; 21:ijms21197325. [PMID: 33023061 PMCID: PMC7582301 DOI: 10.3390/ijms21197325] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/29/2020] [Accepted: 09/29/2020] [Indexed: 12/16/2022] Open
Abstract
D-amino acids have been known to exist in the human brain for nearly 40 years, and they continue to be a field of active study to today. This review article aims to give a concise overview of the recent advances in D-amino acid research as they relate to the brain and neurological disorders. This work has largely been focused on modulation of the N-methyl-D-aspartate (NMDA) receptor and its relationship to Alzheimer’s disease and Schizophrenia, but there has been a wealth of novel research which has elucidated a novel role for several D-amino acids in altering brain chemistry in a neuroprotective manner. D-amino acids which have no currently known activity in the brain but which have active derivatives will also be reviewed.
Collapse
Affiliation(s)
- James M. Seckler
- Department Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Correspondence:
| | - Stephen J. Lewis
- Department Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA;
| |
Collapse
|
59
|
Ieraci A, Beggiato S, Ferraro L, Barbieri SS, Popoli M. Kynurenine pathway is altered in BDNF Val66Met knock-in mice: Effect of physical exercise. Brain Behav Immun 2020; 89:440-450. [PMID: 32726686 DOI: 10.1016/j.bbi.2020.07.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 07/06/2020] [Accepted: 07/22/2020] [Indexed: 12/18/2022] Open
Abstract
The Brain-Derived Neurotrophic Factor (BDNF) Val66Met polymorphism has been correlated with increased predisposition to develop cognitive and psychiatric disorders, and with a reduced response to some therapeutic treatments. However, the mechanisms underlying these impairments are currently not completely understood. Remarkably, kynurenine pathway alterations have also been implicated in cognitive and psychiatric disorders. Moreover, recent evidence suggests that physical exercise may promote beneficial effects by controlling kynurenine metabolism in the muscle. The aim of the present study was to assess whether the kynurenine pathway was differentially regulated in sedentary and exercising wild-type (BDNFVal/Val) and homozygous knock-in BDNF Val66Met (BDNFMet/Met) mice. We found that plasma and hippocampal levels of kynurenic acid and the hippocampal mRNA levels of IDO1 and KAT2 protein levels were increased in BDNFMet/Met mice and were not modulated by physical exercise. On the contrary, KAT1 protein levels in the gastrocnemius muscle were reduced, whereas MCP1 mRNA in the gastrocnemius muscle and GFAP protein in the hippocampus were increased in BDNFMet/Met mice compared to BDNFVal/Val mice, and reduced by physical exercise. Physical exercise increased plasmatic kynurenine levels only in BDNFMet/Met mice, and protein levels of KAT1 and KAT4 in the gastrocnemius muscle and hippocampus respectively, regardless of the genotype. Finally, we found that physical exercise was able to enhance the hippocampal-dependent memory only in the BDNFVal/Val mice. Overall our results showing an overactivation of the kynurenine pathway in the BDNFMet/Met mice may suggest a possible mechanism underlying the cognitive deficits reported in the BDNF Val66Met carriers.
Collapse
Affiliation(s)
- Alessandro Ieraci
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics - Dipartimento di Scienze Farmaceutiche, Sezione di Fisiologia e Farmacologia, Università di Milano, Milano, Italy.
| | - Sarah Beggiato
- Department of Life Sciences and Biotechnologies, University of Ferrara, Ferrara, Italy; Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Italy
| | - Luca Ferraro
- Department of Life Sciences and Biotechnologies, University of Ferrara, Ferrara, Italy
| | | | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics - Dipartimento di Scienze Farmaceutiche, Sezione di Fisiologia e Farmacologia, Università di Milano, Milano, Italy
| |
Collapse
|
60
|
Mor A, Kalaska B, Pawlak D. Kynurenine Pathway in Chronic Kidney Disease: What’s Old, What’s New, and What’s Next? Int J Tryptophan Res 2020; 13:1178646920954882. [PMID: 35210786 PMCID: PMC8862190 DOI: 10.1177/1178646920954882] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/06/2020] [Indexed: 11/25/2022] Open
Abstract
Impaired kidney function and increased inflammatory process occurring in the course of Chronic Kidney Disease (CKD) contribute to the development of complex amino-acid alterations. The essential amino-acid tryptophan (TRP) undergoes extensive metabolism along several pathways, resulting in the production of many biologically active compounds. The results of many studies have shown that its metabolism via the kynurenine pathway is potently increased in the course of CKD. Metabolites of this pathway exhibit differential, sometimes opposite, roles in several biological processes. Their accumulation in the course of CKD may induce oxidative cell damage which stimulates inflammatory processes. They can also modulate the activity of numerous cellular signaling pathways through activation of the aryl hydrocarbon receptor, leading to the disruption of homeostasis of various organs. As a result, they can contribute to the development of the systemic disorders accompanying the course of chronic renal failure. This review gathers and systematizes reports concerning the knowledge connecting the kynurenine pathway metabolites to systemic disorders accompanying the development of CKD.
Collapse
Affiliation(s)
- Adrian Mor
- Department of Pharmacodynamics, Medical University of Bialystok, Bialystok, Poland
| | - Bartlomiej Kalaska
- Department of Pharmacodynamics, Medical University of Bialystok, Bialystok, Poland
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
61
|
Boros F, Vécsei L. Progress in the development of kynurenine and quinoline-3-carboxamide-derived drugs. Expert Opin Investig Drugs 2020; 29:1223-1247. [DOI: 10.1080/13543784.2020.1813716] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Fanni Boros
- Department of Neurology, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
- MTA-SZTE Neuroscience Research Group of the Hungarian Academy of Sciences and the University of Szeged, Szeged, Hungary
- Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| |
Collapse
|
62
|
Zhuravlev AV, Vetrovoy OV, Ivanova PN, Savvateeva-Popova EV. 3-Hydroxykynurenine in Regulation of Drosophila Behavior: The Novel Mechanisms for Cardinal Phenotype Manifestations. Front Physiol 2020; 11:971. [PMID: 32848886 PMCID: PMC7426499 DOI: 10.3389/fphys.2020.00971] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 07/16/2020] [Indexed: 01/21/2023] Open
Abstract
Dysfunctions of kynurenine pathway of tryptophan metabolism (KPTM) are associated with multiple neuropathologies in vertebrates and invertebrates. Drosophila mutants with altered content of kynurenines are model objects for studying the molecular processes of neurodegeneration and senile dementia. The mutant cardinal (cd1) with accumulation of the redox stress inductor 3-hydroxykynurenine (3-HOK) shows age-dependent impairments of the courtship song and middle-term memory. The molecular mechanisms for 3-HOK accumulation in cd1 are still unknown. Here, we have studied age-dependent differences in spontaneous locomotor activity (SLA) for the wild type strain Canton-S (CS), cd1, and cinnabar (cn1) with an excess of neuroprotective kynurenic acid (KYNA). We have also estimated the level and distribution of protein-bound 3-HOK (PB-3-HOK) in Drosophila brains (Br) and head tissues. The middle-age cd1 show the higher running speed and lower run frequency compared to CS, for cn1 the situation is the opposite. There is a decrease in the index of activity for 40-day-old cd1 that seems to be an effect of the oxidative stress development. Surprisingly, PB-3-HOK level in Drosophila heads, brains, and head capsules (HC) is several times lower for cd1 compared to CS. This complements the traditional hypothesis that cd1 phenotype results from a mutation in phenoxazinone synthase (PHS) gene governing the brown eye pigment xanthommatin synthesis. In addition to 3-HOK dimerization, cd1 mutation affects protein modification by 3-HOK. The accumulation of free 3-HOK in cd1 may result from the impairment of 3-HOK conjugation with some proteins of the brain and head tissues.
Collapse
Affiliation(s)
- Aleksandr V Zhuravlev
- Laboratory of Neurogenetics, Pavlov Institute of Physiology Russian Academy of Sciences, Saint Petersburg, Russia
| | - Oleg V Vetrovoy
- Laboratory of Regulation of Brain Neuron Functions, Pavlov Institute of Physiology Russian Academy of Sciences, Saint Petersburg, Russia.,Department of Biochemistry, Faculty of Biology, Saint Petersburg State University, Saint Petersburg, Russia
| | - Polina N Ivanova
- Laboratory of Neurogenetics, Pavlov Institute of Physiology Russian Academy of Sciences, Saint Petersburg, Russia.,Department of Anatomy and Physiology of Humans and Animals, Faculty of Biology, Herzen State Pedagogical University of Russia, Saint Petersburg, Russia
| | - Elena V Savvateeva-Popova
- Laboratory of Neurogenetics, Pavlov Institute of Physiology Russian Academy of Sciences, Saint Petersburg, Russia
| |
Collapse
|
63
|
Hamed A, Kursa MB. Social deprivation substantially changes multi-structural neurotransmitter signature of social interaction: Glutamate concentration in amygdala and VTA as a key factor in social encounter-induced 50-kHz ultrasonic vocalization. Eur Neuropsychopharmacol 2020; 37:82-99. [PMID: 32651127 DOI: 10.1016/j.euroneuro.2020.06.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 01/03/2023]
Abstract
Ultrasonic vocalizations are important for coordinating social behavior in rats. Examination of the neurochemical mechanisms that govern social behavior and ultrasonic vocalization emission is crucial for understanding the social impairments that occur in many neuropsychiatric disorders. To elucidate neurochemical changes in the brain structures related to social behavior and their mutual relationships, we conducted three-phase experiment. Neurochemicals were measured in the following behavioral situations: without social encounter, with short social encounter, with long social encounter in isolated and non-isolated rats. The aims of this study were to: (1) extract the most important neurotransmitters and their metabolites that are involved in social encounter-induced emission of 50 kHz calls; (2) to elucidate mutual relationships among the neurochemical changes in the selected, six brain structures, and analyze compound relationships by step analysis; (3) create a model of all-to-all neurotransmitter correlations; (4) find the neurochemical basis of 50-kHz USVs emission during social encounter. Our behavioral and neurochemical analysis indicated that social encounter was a triggering factor of the glutamatergic neurotransmission in the ventral tegmental area (VTA), hippocampus, and amygdala; serotonergic neurotransmission in the NAcc, CPu, and amygdala; the dopaminergic neurotransmission in the caudate putamen (CPu) and hippocampus; GABAergic neurotransmission in the hippocampus and VTA. Social encounter-induced 50-kHz USVs were bound up with changes in glutamate in amygdala and VTA, glycine in the amygdala, VTA, hippocampus, nucleus accumbens and CPu, and dopamine metabolites in VTA and CPu.
Collapse
Affiliation(s)
- Adam Hamed
- Laboratory of Spatial Memory, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, Warsaw 02-093, Poland.
| | - Miron Bartosz Kursa
- Interdisciplinary Centre for Mathematical and Computational Modelling, University of Warsaw, Pawinskiego 5A, 02-106 Warsaw, Poland
| |
Collapse
|
64
|
Klausing AD, Fukuwatari T, Bucci DJ, Schwarcz R. Stress-induced impairment in fear discrimination is causally related to increased kynurenic acid formation in the prefrontal cortex. Psychopharmacology (Berl) 2020; 237:1931-1941. [PMID: 32215686 PMCID: PMC7308198 DOI: 10.1007/s00213-020-05507-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/10/2020] [Indexed: 12/11/2022]
Abstract
RATIONALE Stress is related to cognitive impairments which are observed in most major brain diseases. Prior studies showed that the brain concentration of the tryptophan metabolite kynurenic acid (KYNA) is modulated by stress, and that changes in cerebral KYNA levels impact cognition. However, the link between these phenomena has not been tested directly so far. OBJECTIVES To investigate a possible causal relationship between acute stress, KYNA, and fear discrimination. METHODS Adult rats were exposed to one of three acute stressors-predator odor, restraint, or inescapable foot shocks (ISS)-and KYNA in the prefrontal cortex was measured using microdialysis. Corticosterone was analyzed in a subset of rats. Another cohort underwent a fear discrimination procedure immediately after experiencing stress. Different auditory conditioned stimuli (CSs) were either paired with foot shock (CS+) or were non-reinforced (CS-). One week later, fear was assessed by re-exposing rats to each CS. Finally, to test whether stress-induced changes in KYNA causally impacted fear discrimination, a group of rats that received ISS were pre-treated with the selective KYNA synthesis inhibitor PF-04859989. RESULTS ISS caused the greatest increase in circulating corticosterone levels and raised extracellular KYNA levels by ~ 85%. The two other stressors affected KYNA much less (< 25% increase). Moreover, only rats that received ISS were unable to discriminate between CS+ and CS-. PF-04859989 abolished the stress-induced KYNA increase and also prevented the impairment in fear discrimination in animals that experienced ISS. CONCLUSIONS These findings demonstrate a causal connection between stress-induced KYNA increases and cognitive deficits. Pharmacological manipulation of KYNA synthesis therefore offers a novel approach to modulate cognitive processes in stress-related disorders.
Collapse
Affiliation(s)
- Alex D Klausing
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, 55 Wade Avenue, Baltimore, MD, 21228, USA
| | - Tsutomu Fukuwatari
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, 55 Wade Avenue, Baltimore, MD, 21228, USA
- Department of Nutrition, School of Human Cultures, The University of Shiga Prefecture, 2500 Hassaka, Hikone, Shiga, 522-8533, Japan
| | - David J Bucci
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, USA
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, 55 Wade Avenue, Baltimore, MD, 21228, USA.
| |
Collapse
|
65
|
Meier TB, Nitta ME, Teague TK, Nelson LD, McCrea MA, Savitz J. Prospective study of the effects of sport-related concussion on serum kynurenine pathway metabolites. Brain Behav Immun 2020; 87:715-724. [PMID: 32147388 PMCID: PMC7316609 DOI: 10.1016/j.bbi.2020.03.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/18/2020] [Accepted: 03/02/2020] [Indexed: 02/07/2023] Open
Abstract
Reports of neurodegenerative and psychiatric disease in former athletes have increased public concern about the acute and chronic effects of sport-related concussions (SRC). The biological factors underlying individual differences in the psychiatric sequalae of SRC and their role in potential long-term negative outcomes have not been determined. One understudied biological consequence of the known inflammatory response to concussion is the activation of a key immunoregulatory pathway, the kynurenine pathway (KP). Activation of the KP produces several neuroactive metabolites that have been associated with psychiatric and neurodegenerative diseases. We tested the hypothesis that SRC results in an elevation of serum KP metabolites with neurotoxic properties (quinolinic acid [QuinA], 3-hydroxykynurenine [3HK]) together with a reduction in the neuroprotective metabolite kynurenic acid (KynA), and that these metabolites would predict post-concussion psychological symptoms. Additionally, because brain injury is thought to prime the immune system, a secondary goal was to test the hypothesis that athletes with acute SRC and a history of prior SRC would have elevated neurotoxic relative to neuroprotective KP metabolites compared to athletes that were concussed for the first time. High school and collegiate football players (N = 1136) were enrolled at a preseason baseline visit that included clinical testing and blood specimen collection. Athletes that suffered a SRC (N = 59) completed follow-up visits within 6-hours (early-acute), at 24-48 h (late-acute) and at 8, 15, and 45 days post-injury. Uninjured contact sport (CC; N = 54) and non-contact sport athletes completed similar visits and served as controls (NCC; N = 30). SRC athletes had significantly elevated psychological symptoms, assessed using the Brief Symptom Inventory-18 (BSI), acutely following injury relative to both control groups. There was a group-by-visit interaction on the ratio of KynA to 3HK in serum, a neuroprotective index, with elevated KynA/3HK in athletes with SRC at the early-acute visit relative to later visits. Importantly, athletes with greater elevation in this neuroprotective index at the early-acute visit reported fewer depressive symptoms at the late-acute visit. Finally, SRC athletes with prior concussion had significantly lower serum KynA/QuinA at all visits compared to SRC athletes with no prior concussion, an effect driven by elevated QuinA in SRC athletes with prior concussion. These results suggest that early-acute activation of the KynA branch of the KP may protect against the development of depressive symptoms following concussion. Furthermore, they highlight the potential of serum QuinA as a biomarker for repetitive head injury and provide insight into possible mechanisms linking prior concussion with subsequent injury.
Collapse
Affiliation(s)
- Timothy B. Meier
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI,Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI,Corresponding Author: Timothy B. Meier, PhD, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, Phone: 414-955-7310, Fax: 414-955-0115,
| | - Morgan E. Nitta
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI,Department of Psychology, Marquette University, Milwaukee, WI
| | - T. Kent Teague
- Department of Surgery, University of Oklahoma School of Community Medicine, Tulsa, OK,Department of Psychiatry, University of Oklahoma School of Community Medicine, Tulsa, OK,Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, Tulsa, OK.,Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK
| | - Lindsay D. Nelson
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI,Department of Neurology, Medical College of Wisconsin, Milwaukee, WI
| | - Michael A. McCrea
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI,Department of Neurology, Medical College of Wisconsin, Milwaukee, WI
| | - Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa, OK,Oxley College of Health Sciences, The University of Tulsa, Tulsa OK
| |
Collapse
|
66
|
Biernacki T, Sandi D, Bencsik K, Vécsei L. Kynurenines in the Pathogenesis of Multiple Sclerosis: Therapeutic Perspectives. Cells 2020; 9:cells9061564. [PMID: 32604956 PMCID: PMC7349747 DOI: 10.3390/cells9061564] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
Over the past years, an increasing amount of evidence has emerged in support of the kynurenine pathway’s (KP) pivotal role in the pathogenesis of several neurodegenerative, psychiatric, vascular and autoimmune diseases. Different neuroactive metabolites of the KP are known to exert opposite effects on neurons, some being neuroprotective (e.g., picolinic acid, kynurenic acid, and the cofactor nicotinamide adenine dinucleotide), while others are toxic to neurons (e.g., 3-hydroxykynurenine, quinolinic acid). Not only the alterations in the levels of the metabolites but also disturbances in their ratio (quinolinic acid/kynurenic acid) have been reported in several diseases. In addition to the metabolites, the enzymes participating in the KP have been unearthed to be involved in modulation of the immune system, the energetic upkeep of neurons and have been shown to influence redox processes and inflammatory cascades, revealing a sophisticated, intertwined system. This review considers various methods through which enzymes and metabolites of the kynurenine pathway influence the immune system, the roles they play in the pathogenesis of neuroinflammatory diseases based on current evidence with a focus on their involvement in multiple sclerosis, as well as therapeutic approaches.
Collapse
Affiliation(s)
- Tamás Biernacki
- Department of Neurology, Faculty of General Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, H-6725 Szeged, Hungary; (T.B.); (D.S.); (K.B.)
| | - Dániel Sandi
- Department of Neurology, Faculty of General Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, H-6725 Szeged, Hungary; (T.B.); (D.S.); (K.B.)
| | - Krisztina Bencsik
- Department of Neurology, Faculty of General Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, H-6725 Szeged, Hungary; (T.B.); (D.S.); (K.B.)
| | - László Vécsei
- Department of Neurology, Faculty of General Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, H-6725 Szeged, Hungary; (T.B.); (D.S.); (K.B.)
- MTA—SZTE Neuroscience Research Group, H-6725 Szeged, Hungary
- Interdisciplinary Excellence Center, University of Szeged, H-6720 Szeged, Hungary
- Correspondence: ; Tel.: +36-62-545-356; Fax: +36-62-545-597
| |
Collapse
|
67
|
Staats Pires A, Tan VX, Heng B, Guillemin GJ, Latini A. Kynurenine and Tetrahydrobiopterin Pathways Crosstalk in Pain Hypersensitivity. Front Neurosci 2020; 14:620. [PMID: 32694973 PMCID: PMC7338796 DOI: 10.3389/fnins.2020.00620] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 05/19/2020] [Indexed: 12/19/2022] Open
Abstract
Despite the identification of molecular mechanisms associated with pain persistence, no significant therapeutic improvements have been made. Advances in the understanding of the molecular mechanisms that induce pain hypersensitivity will allow the development of novel, effective, and safe therapies for chronic pain. Various pro-inflammatory cytokines are known to be increased during chronic pain, leading to sustained inflammation in the peripheral and central nervous systems. The pro-inflammatory environment activates additional metabolic routes, including the kynurenine (KYN) and tetrahydrobiopterin (BH4) pathways, which generate bioactive soluble metabolites with the potential to modulate neuropathic and inflammatory pain sensitivity. Inflammation-induced upregulation of indoleamine 2,3-dioxygenase 1 (IDO1) and guanosine triphosphate cyclohydrolase I (GTPCH), both rate-limiting enzymes of KYN and BH4 biosynthesis, respectively, have been identified in experimental chronic pain models as well in biological samples from patients affected by chronic pain. Inflammatory inducible KYN and BH4 pathways upregulation is characterized by increase in pronociceptive compounds, such as quinolinic acid (QUIN) and BH4, in addition to inflammatory mediators such as interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α). As expected, the pharmacologic and genetic experimental manipulation of both pathways confers analgesia. Many metabolic intermediates of these two pathways such as BH4, are known to sustain pain, while others, like xanthurenic acid (XA; a KYN pathway metabolite) have been recently shown to be an inhibitor of BH4 synthesis, opening a new avenue to treat chronic pain. This review will focus on the KYN/BH4 crosstalk in chronic pain and the potential modulation of these metabolic pathways that could induce analgesia without dependence or abuse liability.
Collapse
Affiliation(s)
- Ananda Staats Pires
- Neuroinflammation Group, Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- Laboratório de Bioenergética e Estresse Oxidativo, Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Vanessa X. Tan
- Neuroinflammation Group, Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Benjamin Heng
- Neuroinflammation Group, Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Gilles J. Guillemin
- Neuroinflammation Group, Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Alexandra Latini
- Laboratório de Bioenergética e Estresse Oxidativo, Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
68
|
Activation of alpha7 nicotinic and NMDA receptors is necessary for performance in a working memory task. Psychopharmacology (Berl) 2020; 237:1723-1735. [PMID: 32162104 PMCID: PMC7313359 DOI: 10.1007/s00213-020-05495-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/19/2020] [Indexed: 10/24/2022]
Abstract
RATIONALE Working memory deficits are present in schizophrenia (SZ) but remain insufficiently resolved by medications. Similar cognitive dysfunctions can be produced acutely in animals by elevating brain levels of kynurenic acid (KYNA). KYNA's effects may reflect interference with the function of both the α7 nicotinic acetylcholine receptor (α7nAChR) and the glycineB site of the NMDA receptor. OBJECTIVES The aim of the present study was to examine, using pharmacological tools, the respective roles of these two receptor sites on performance in a delayed non-match-to-position working memory (WM) task (DNMTP). METHODS DNMTP consisted of 120 trials/session (5, 10, and 15 s delays). Rats received two doses (25 or 100 mg/kg, i.p.) of L-kynurenine (KYN; bioprecursor of KYNA) or L-4-chlorokynurenine (4-Cl-KYN; bioprecursor of the selective glycineB site antagonist 7-Cl-kynurenic acid). Attenuation of KYN- or 4-Cl-KYN-induced deficits was assessed by co-administration of galantamine (GAL, 3 mg/kg) or PAM-2 (1 mg/kg), two positive modulators of α7nAChR function. Reversal of 4-Cl-KYN-induced deficits was examined using D-cycloserine (DCS; 30 mg/kg), a partial agonist at the glycineB site. RESULTS Both KYN and 4-Cl-KYN administration produced dose-related deficits in DNMTP accuracy that were more severe at the longer delays. In KYN-treated rats, these deficits were reversed to control levels by GAL or PAM-2 but not by DCS. In contrast, DCS eliminated performance deficits in 4-Cl-KYN-treated animals. CONCLUSIONS These experiments reveal that both α7nAChR and NMDAR activity are necessary for normal WM accuracy. They provide substantive new support for the therapeutic potential of positive modulators at these two receptor sites in SZ and other major brain diseases.
Collapse
|
69
|
Possibility of Amino Acid Treatment to Prevent the Psychiatric Disorders via Modulation of the Production of Tryptophan Metabolite Kynurenic Acid. Nutrients 2020; 12:nu12051403. [PMID: 32414200 PMCID: PMC7284450 DOI: 10.3390/nu12051403] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 01/06/2023] Open
Abstract
Kynurenic acid, a metabolite of the kynurenine pathway of tryptophan catabolism, acts as an antagonist for both the α7 nicotinic acetylcholine receptor and glycine coagonist sites of the N-methyl-d-aspartic acid receptor at endogenous brain concentrations. Elevation of brain kynurenic acid levels reduces the release of neurotransmitters such as dopamine and glutamate, and kynurenic acid is considered to be involved in psychiatric disorders such as schizophrenia and depression. Thus, the control of kynurenine pathway, especially kynurenic acid production, in the brain is an important target for the improvement of brain function or the effective treatment of brain disorders. Astrocytes uptake kynurenine, the immediate precursor of kynurenic acid, via large neutral amino acid transporters, and metabolize kynurenine to kynurenic acid by kynurenine aminotransferases. The former transport both branched-chain and aromatic amino acids, and the latter have substrate specificity for amino acids and their metabolites. Recent studies have suggested the possibility that amino acids may suppress kynurenic acid production via the blockade of kynurenine transport or via kynurenic acid synthesis reactions. This approach may be useful in the treatment and prevention of neurological and psychiatric diseases associated with elevated kynurenic acid levels.
Collapse
|
70
|
Cseh EK, Veres G, Körtési T, Polyák H, Nánási N, Tajti J, Párdutz Á, Klivényi P, Vécsei L, Zádori D. Neurotransmitter and tryptophan metabolite concentration changes in the complete Freund's adjuvant model of orofacial pain. J Headache Pain 2020; 21:35. [PMID: 32316909 PMCID: PMC7175490 DOI: 10.1186/s10194-020-01105-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/08/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The neurochemical background of the evolution of headache disorders, still remains partially undiscovered. Accordingly, our aim was to further explore the neurochemical profile of Complete Freund's adjuvant (CFA)-induced orofacial pain, involving finding the shift point regarding small molecule neurotransmitter concentrations changes vs. that of the previously characterized headache-related neuropeptides. The investigated neurotransmitters consisted of glutamate, γ-aminobutyric acid, noradrenalin and serotonin. Furthermore, in light of its influence on glutamatergic neurotransmission, we measured the level of kynurenic acid (KYNA) and its precursors in the kynurenine (KYN) pathway (KP) of tryptophan metabolism. METHODS The effect of CFA was evaluated in male Sprague Dawley rats. Animals were injected with CFA (1 mg/ml, 50 μl/animal) into the right whisker pad. We applied high-performance liquid chromatography to determine the concentrations of the above-mentioned compounds from the trigeminal nucleus caudalis (TNC) and somatosensory cortex (ssCX) of rats. Furthermore, we measured some of these metabolites from the cerebrospinal fluid and plasma as well. Afterwards, we carried out permutation t-tests as post hoc analysis for pairwise comparison. RESULTS Our results demonstrated that 24 h after CFA treatment, the level of glutamate, KYNA and that of its precursor, KYN was still elevated in the TNC, all diminishing by 48 h. In the ssCX, significant concentration increases of KYNA and serotonin were found. CONCLUSION This is the first study assessing neurotransmitter changes in the TNC and ssCX following CFA treatment, confirming the dominant role of glutamate in early pain processing and a compensatory elevation of KYNA with anti-glutamatergic properties. Furthermore, the current findings draw attention to the limited time interval where medications can target the glutamatergic pathways.
Collapse
Affiliation(s)
- Edina K Cseh
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, H-6725, Hungary
| | - Gábor Veres
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, H-6725, Hungary
- MTA-SZTE Neuroscience Research Group, Szeged, Hungary
| | - Tamás Körtési
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, H-6725, Hungary
| | - Helga Polyák
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, H-6725, Hungary
| | - Nikolett Nánási
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, H-6725, Hungary
| | - János Tajti
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, H-6725, Hungary
| | - Árpád Párdutz
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, H-6725, Hungary
| | - Péter Klivényi
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, H-6725, Hungary
| | - László Vécsei
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, H-6725, Hungary
- MTA-SZTE Neuroscience Research Group, Szeged, Hungary
| | - Dénes Zádori
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, H-6725, Hungary.
| |
Collapse
|
71
|
Discovery of sulfonamides and 9-oxo-2,8-diazaspiro[5,5]undecane-2-carboxamides as human kynurenine aminotransferase 2 (KAT2) inhibitors. Bioorg Med Chem Lett 2020; 30:127060. [PMID: 32113843 DOI: 10.1016/j.bmcl.2020.127060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/14/2020] [Accepted: 02/21/2020] [Indexed: 02/02/2023]
Abstract
Human kynurenine aminotransferase 2 (KAT2) inhibitors could be potentially used to treat the cognitive deficits associated with bipolar disease and schizophrenia. Although, there has been active drug research activity by several industrial and academic groups in developing KAT2 inhibitors over the years, no such compound has proceeded to the clinics. Here, we report two different chemical series of reversible KAT2 inhibitors with sub-micromolar activities. The first series was identified by a high-throughput screening of a diverse random library and the second one by structure-based virtual screening. Two novel crystal structures of KAT2 complexed with different reversible inhibitors were also deposited to the Protein databank which could be useful for future drug discovery efforts.
Collapse
|
72
|
Meng Q, Ding Y, Chen L, Li L. The medial agranular cortex mediates attentional enhancement of prepulse inhibition of the startle reflex. Behav Brain Res 2020; 383:112511. [DOI: 10.1016/j.bbr.2020.112511] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 01/15/2020] [Accepted: 01/24/2020] [Indexed: 12/12/2022]
|
73
|
Stone TW. Does kynurenic acid act on nicotinic receptors? An assessment of the evidence. J Neurochem 2020; 152:627-649. [PMID: 31693759 PMCID: PMC7078985 DOI: 10.1111/jnc.14907] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/19/2019] [Accepted: 10/30/2019] [Indexed: 02/06/2023]
Abstract
As a major metabolite of kynurenine in the oxidative metabolism of tryptophan, kynurenic acid is of considerable biological and clinical importance as an endogenous antagonist of glutamate in the central nervous system. It is most active as an antagonist at receptors sensitive to N-methyl-D-aspartate (NMDA) which regulate neuronal excitability and plasticity, brain development and behaviour. It is also thought to play a causative role in hypo-glutamatergic conditions such as schizophrenia, and a protective role in several neurodegenerative disorders, notably Huntington's disease. An additional hypothesis, that kynurenic acid could block nicotinic receptors for acetylcholine in the central nervous system has been proposed as an alternative mechanism of action of kynurenate. However, the evidence for this alternative mechanism is highly controversial, partly because at least eight earlier studies concluded that kynurenic acid blocked NMDA receptors but not nicotinic receptors and five subsequent, independent studies designed to repeat the results have failed to do so. Many studies considered to support the alternative 'nicotinic' hypothesis have been based on the use of analogs of kynurenate such as 7-chloro-kynurenic acid, or putatively nicotinic modulators such as galantamine, but a detailed analysis of the pharmacology of these compounds suggests that the results have often been misinterpreted, especially since the pharmacology of galantamine itself has been disputed. This review examines the evidence in detail, with the conclusion that there is no confirmed, reliable evidence for an antagonist activity of kynurenic acid at nicotinic receptors. Therefore, since there is overwhelming evidence for kynurenate acting at ionotropic glutamate receptors, especially NMDAR glutamate and glycine sites, with some activity at GPR35 sites and Aryl Hydrocarbon Receptors, results with kynurenic acid should be interpreted only in terms of these confirmed sites of action.
Collapse
Affiliation(s)
- Trevor W. Stone
- Institute for Neuroscience and PsychologyUniversity of GlasgowGlasgowG12 8QQUK
- Present address:
Kennedy InstituteNDORMSUniversity of OxfordOxfordOX3 7FYUK
| |
Collapse
|
74
|
Wyckelsma VL, Lindkvist W, Venckunas T, Brazaitis M, Kamandulis S, Pääsuke M, Ereline J, Westerblad H, Andersson DC. Kynurenine aminotransferase isoforms display fiber-type specific expression in young and old human skeletal muscle. Exp Gerontol 2020; 134:110880. [PMID: 32068089 DOI: 10.1016/j.exger.2020.110880] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/12/2020] [Accepted: 02/12/2020] [Indexed: 12/27/2022]
Abstract
Conversion of kynurenine (KYN) to kynurenic acid (KYNA) is the main pathway for free tryptophan degradation in skeletal muscle and has emerged as an important mechanism of how exercise is linked to promotion of mental health. Metabolism of KYN to KYNA mainly depends on the expression of kynurenine aminotransferases (KATs) that is under control of the mitochondria biogenesis regulator PGC-1α. We therefore hypothesized that expression of KATs would vary between muscle fibers that differ in mitochondrial content, i.e. oxidative type I vs more glycolytic type II muscle fibers. Moreover, we tested the hypothesis that KAT expression differs with age. Single muscle fibers were isolated from biopsies taken from the vastus lateralis muscle in young and old healthy subjects. In young and old subjects the abundance of KAT I, KAT III and KAT IV was greater in Type I than Type II fibers without age-dependent difference in the KAT isoform expressions. The link to mitochondrial content was further seen as the expression of KAT IV correlated to mitochondrial cytochrome c oxidase IV (COX IV) abundance in both fiber types. In conclusion, we describe for the first time the expression pattern of KAT isoforms with respect to specific fiber types and age in human skeletal muscle.
Collapse
Affiliation(s)
- V L Wyckelsma
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - W Lindkvist
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - T Venckunas
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - M Brazaitis
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - S Kamandulis
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - M Pääsuke
- Institute of Sport Sciences and Physiotherapy, Faculty of Medicine, University of Tartu, Estonia
| | - J Ereline
- Institute of Sport Sciences and Physiotherapy, Faculty of Medicine, University of Tartu, Estonia
| | - H Westerblad
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - D C Andersson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Heart, Vascular and Neurology Theme, Section for Cardiology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
75
|
Organ-differential responses to ethanol and kynurenic acid, a component of alcoholic beverages in gene transcription. Gene 2020; 737:144434. [PMID: 32018015 DOI: 10.1016/j.gene.2020.144434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/29/2020] [Accepted: 01/31/2020] [Indexed: 11/24/2022]
Abstract
Excessive alcohol (ethanol) use has long been known to affect human health negatively. However, the underlying molecular basis is incompletely understood. Moreover, consumption of alcohol is often mixed with kynurenic acid (KYNA), an abundant tryptophan metabolite produced during fermentation. The combined effect of ethanol and KYNA on host gene expression has not been investigated. The current study used mice as the model to interrogate the impact of ethanol and/or KYNA on global gene transcription. Adult male mice were administered with 2 g/kg ethanol and/or 0.1 mg/kg KYNA by gavage once a day for a week. Three organs: brain, kidney, and liver were collected and their total RNAs extracted for transcriptome sequencing and quantitative real-time PCR. Gene ontology, Kyoto encyclopedia of genes, and genomes pathway analyses revealed that alcohol affects the three organs differentially. Furthermore, the gene expression profile from alcohol and KYNA co-administration was significantly different from that of alcohol or KYNA administration alone. Strikingly, Indolamine 2,3-dioxygenase 1, a rate-limiting enzyme in tryptophan metabolism, was significantly increased in the brain after a combined exposure of alcohol and KYNA, suggesting that Trp metabolism was skewed towards the kynurenine pathway in the brain. Our systemic analysis provides new insights into the mechanism whereby alcohol and KYNA affects organ functions.
Collapse
|
76
|
Tanaka M, Bohár Z, Vécsei L. Are Kynurenines Accomplices or Principal Villains in Dementia? Maintenance of Kynurenine Metabolism. Molecules 2020; 25:molecules25030564. [PMID: 32012948 PMCID: PMC7036975 DOI: 10.3390/molecules25030564] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/24/2020] [Accepted: 01/25/2020] [Indexed: 12/16/2022] Open
Abstract
Worldwide, 50 million people suffer from dementia, a group of symptoms affecting cognitive and social functions, progressing severely enough to interfere with daily life. Alzheimer’s disease (AD) accounts for most of the dementia cases. Pathological and clinical findings have led to proposing several hypotheses of AD pathogenesis, finding a presence of positive feedback loops and additionally observing the disturbance of a branch of tryptophan metabolism, the kynurenine (KYN) pathway. Either causative or resultant of dementia, elevated levels of neurotoxic KYN metabolites are observed, potentially upregulating multiple feedback loops of AD pathogenesis. Memantine is an N-methyl-D-aspartate glutamatergic receptor (NMDAR) antagonist, which belongs to one of only two classes of medications approved for clinical use, but other NMDAR modulators have been explored so far in vain. An endogenous KYN pathway metabolite, kynurenic acid (KYNA), likewise inhibits the excitotoxic NMDAR. Besides its anti-excitotoxicity, KYNA is a multitarget compound that triggers anti-inflammatory and antioxidant activities. Modifying the KYNA level is a potential multitarget strategy to normalize the disturbed KYN pathway and thus to alleviate juxtaposing AD pathogeneses. In this review, the maintenance of KYN metabolism by modifying the level of KYNA is proposed and discussed in search for a novel lead compound against the progression of dementia.
Collapse
Affiliation(s)
- Masaru Tanaka
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary
- Department of Neurology, Interdisciplinary Excellence Centre, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Zsuzsanna Bohár
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary
- Department of Neurology, Interdisciplinary Excellence Centre, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - László Vécsei
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary
- Department of Neurology, Interdisciplinary Excellence Centre, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
- Correspondence: ; Tel.: +36-62-545-351
| |
Collapse
|
77
|
Abstract
The kynurenine pathway (KP) plays a critical role in generating cellular energy in the form of nicotinamide adenine dinucleotide (NAD+). Because energy requirements are substantially increased during an immune response, the KP is a key regulator of the immune system. Perhaps more importantly in the context of psychiatry, many kynurenines are neuroactive, modulating neuroplasticity and/or exerting neurotoxic effects in part through their effects on NMDA receptor signaling and glutamatergic neurotransmission. As such, it is not surprising that the kynurenines have been implicated in psychiatric illness in the context of inflammation. However, because of their neuromodulatory properties, the kynurenines are not just additional members of a list of inflammatory mediators linked with psychiatric illness, but in preclinical studies have been shown to be necessary components of the behavioral analogs of depression and schizophrenia-like cognitive deficits. Further, as the title suggests, the KP is regulated by, and in turn regulates multiple other physiological systems that are commonly disrupted in psychiatric disorders, including endocrine, metabolic, and hormonal systems. This review provides a broad overview of the mechanistic pathways through which the kynurenines interact with these systems, thus impacting emotion, cognition, pain, metabolic function, and aging, and in so doing potentially increasing the risk of developing psychiatric disorders. Novel therapeutic approaches targeting the KP are discussed. Moreover, electroconvulsive therapy, ketamine, physical exercise, and certain non-steroidal anti-inflammatories have been shown to alter kynurenine metabolism, raising the possibility that kynurenine metabolites may have utility as treatment response or therapeutic monitoring biomarkers.
Collapse
Affiliation(s)
- Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa, OK, USA.
- Oxley College of Health Sciences, The University of Tulsa, Tulsa, OK, USA.
| |
Collapse
|
78
|
Huang X, Ding W, Wu F, Zhou S, Deng S, Ning Y. Increased Plasma Kynurenic Acid Levels are Associated with Impaired Attention/Vigilance and Social Cognition in Patients with Schizophrenia. Neuropsychiatr Dis Treat 2020; 16:263-271. [PMID: 32158211 PMCID: PMC6986175 DOI: 10.2147/ndt.s239763] [Citation(s) in RCA: 15] [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: 11/23/2019] [Accepted: 01/13/2020] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE Preclinical studies have reported that abnormal kynurenic acid (KYNA) may play a role in cognitive deficits. Schizophrenia (SCZ) is characterized by a wide range of cognitive deficits that may evolve from abnormal KYNA. This study aimed to explore the relationship between KYNA and cognitive impairment in SCZ, which has not yet been reported. METHODS We recruited 30 SCZ patients and 34 healthy controls, measured clinical symptoms by using the Positive and Negative Syndrome Scale and performed cognitive tests using the MATRICS Consensus Cognitive Battery (MCCB). Plasma levels of tryptophan, kynurenine, and KYNA were determined by high-performance liquid chromatography-tandem mass spectrometry. RESULTS We found that plasma KYNA levels were significantly higher in patients than in healthy controls (p=0.009). The cognitive performance of patients in the total MCCB scores and the scores of all subscales were significantly lower than those in healthy controls (all P < 0.01). Correlation analysis showed that KYNA levels were negatively correlated with attention/vigilance (r=-0.457, p=0.019) and social cognition (r=-0.481, p=0.013) only in SCZ patients. CONCLUSION Our results indicate that elevated plasma KYNA levels may serve as a biomarker of cognitive impairment in SCZ patients.
Collapse
Affiliation(s)
- Xingbing Huang
- First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.,The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, Guangdong, People's Republic of China
| | - Wenhua Ding
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, Guangdong, People's Republic of China
| | - Fengchun Wu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, Guangdong, People's Republic of China
| | - Sumiao Zhou
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, Guangdong, People's Republic of China
| | - Shuhua Deng
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, Guangdong, People's Republic of China
| | - Yuping Ning
- First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.,The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, Guangdong, People's Republic of China.,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
79
|
Wigner P, Saluk-Bijak J, Synowiec E, Miller E, Sliwinski T, Cichon N, Bijak M. Variation of Genes Encoding Tryptophan Catabolites Pathway Enzymes in Stroke. J Clin Med 2019; 8:jcm8122133. [PMID: 31817010 PMCID: PMC6947300 DOI: 10.3390/jcm8122133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 11/16/2022] Open
Abstract
The abnormal activation of the tryptophan catabolites pathway (TRYCATs) is observed in patients suffering from cerebrovascular disease, including stroke. A previous study confirmed that lower bioavailability of tryptophan for serotonin synthesis was characterized in the patients during the acute stroke phase. Interestingly, according to various studies, polymorphisms of the genes involved in the TRYCATs pathway may modulate the risk of stroke occurrence. Therefore, this study aimed to investigate the association between the occurrence of TPH1, TPH2, KAT1, KAT2 and IDO1 polymorphisms and the risk of stroke development.The following 10 polymorphisms of the genes encoding enzymes of the TRYCATs pathway were selected: c.804-7C > A (rs10488682), c.-1668T > A (rs623580), c.803+221C > A (rs1800532), c.-173A > T (rs1799913) - TPH1, c.-1449C > A (rs7963803), and c.-844G > T (rs4570625) - TPH2. c.*456G > A of KAT1 (rs10988134), c.975-7T > C of KAT2 (rs1480544), c.-1849C > A (rs3824259) and c. -1493G > C (rs10089084) of IDO1. The study was carried out on DNA isolated from the peripheral blood taken from 107 patients after a stroke and 107 healthy volunteers. All DNA samples were genotyped using TaqMan probes. The genotypes of eight studied polymorphisms modulated the risk of stroke. No significant difference in genotype and allele frequencies of the c.804-7C > A -TPH1 (rs10488682) and c.*456G > A - KAT1 (rs10988134) polymorphisms were found between patients and controls. Having performed haplotype and gen-gen analyses, it was possible to determine that patients after a stroke and controls differed in terms of the frequency of selected genotypes and haplotypes. Among the studied polymorphisms, eight SNPs were linked with stroke risk modulation. The results obtained confirmed our hypothesis regarding the involvement of the TRYCATs pathway in the pathogenesis of stroke.
Collapse
Affiliation(s)
- Paulina Wigner
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (P.W.)
| | - Joanna Saluk-Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Ewelina Synowiec
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (P.W.)
| | - Elzbieta Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, Poland, Milionowa 14, 93-113 Lodz, Poland;
| | - Tomasz Sliwinski
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (P.W.)
| | - Natalia Cichon
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Michal Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
- Correspondence: ; Tel.: +48-42-635-43-36
| |
Collapse
|
80
|
Westfall S, Pasinetti GM. The Gut Microbiota Links Dietary Polyphenols With Management of Psychiatric Mood Disorders. Front Neurosci 2019; 13:1196. [PMID: 31749681 PMCID: PMC6848798 DOI: 10.3389/fnins.2019.01196] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/22/2019] [Indexed: 12/20/2022] Open
Abstract
The pathophysiology of depression is multifactorial yet generally aggravated by stress and its associated physiological consequences. To effectively treat these diverse risk factors, a broad acting strategy is required and is has been suggested that gut-brain-axis signaling may play a pinnacle role in promoting resilience to several of these stress-induced changes including pathogenic load, inflammation, HPA-axis activation, oxidative stress and neurotransmitter imbalances. The gut microbiota also manages the bioaccessibility of phenolic metabolites from dietary polyphenols whose multiple beneficial properties have known therapeutic efficacy against depression. Although several potential therapeutic mechanisms of dietary polyphenols toward establishing cognitive resilience to neuropsychiatric disorders have been established, only a handful of studies have systematically identified how the interaction of the gut microbiota with dietary polyphenols can synergistically alleviate the biological signatures of depression. The current review investigates several of these potential mechanisms and how synbiotics, that combine probiotics with dietary polyphenols, may provide a novel therapeutic strategy for depression. In particular, synbiotics have the potential to alleviate neuroinflammation by modulating microglial and inflammasome activation, reduce oxidative stress and balance serotonin metabolism therefore simultaneously targeting several of the major pathological risk factors of depression. Overall, synbiotics may act as a novel therapeutic paradigm for neuropsychiatric disorders and further understanding the fundamental mechanisms of gut-brain-axis signaling will allow full utilization of the gut microbiota's as a therapeutic tool.
Collapse
Affiliation(s)
| | - Giulio Maria Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
81
|
Kynurenines and the Endocannabinoid System in Schizophrenia: Common Points and Potential Interactions. Molecules 2019; 24:molecules24203709. [PMID: 31619006 PMCID: PMC6832375 DOI: 10.3390/molecules24203709] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/09/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022] Open
Abstract
Schizophrenia, which affects around 1% of the world’s population, has been described as a complex set of symptoms triggered by multiple factors. However, the exact background mechanisms remain to be explored, whereas therapeutic agents with excellent effectivity and safety profiles have yet to be developed. Kynurenines and the endocannabinoid system (ECS) play significant roles in both the development and manifestation of schizophrenia, which have been extensively studied and reviewed previously. Accordingly, kynurenines and the ECS share multiple features and mechanisms in schizophrenia, which have yet to be reviewed. Thus, the present study focuses on the main common points and potential interactions between kynurenines and the ECS in schizophrenia, which include (i) the regulation of glutamatergic/dopaminergic/γ-aminobutyric acidergic neurotransmission, (ii) their presence in astrocytes, and (iii) their role in inflammatory mechanisms. Additionally, promising pharmaceutical approaches involving the kynurenine pathway and the ECS will be reviewed herein.
Collapse
|
82
|
Bodkin JA, Coleman MJ, Godfrey LJ, Carvalho CM, Morgan CJ, Suckow RF, Anderson T, Ongur D, Kaufman MJ, Lewandowski KE, Siegel AJ, Waldstreicher E, Grochowski CM, Javitt DC, Rujescu D, Hebbring S, Weinshilboum R, Rodriguez SB, Kirchhoff C, Visscher T, Vuckovic A, Fialkowski A, McCarthy S, Malhotra D, Sebat J, Goff DC, Hudson JI, Lupski JR, Coyle JT, Rudolph U, Levy DL. Targeted Treatment of Individuals With Psychosis Carrying a Copy Number Variant Containing a Genomic Triplication of the Glycine Decarboxylase Gene. Biol Psychiatry 2019; 86:523-535. [PMID: 31279534 PMCID: PMC6745274 DOI: 10.1016/j.biopsych.2019.04.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 04/17/2019] [Accepted: 04/17/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND The increased mutational burden for rare structural genomic variants in schizophrenia and other neurodevelopmental disorders has so far not yielded therapies targeting the biological effects of specific mutations. We identified two carriers (mother and son) of a triplication of the gene encoding glycine decarboxylase, GLDC, presumably resulting in reduced availability of the N-methyl-D-aspartate receptor coagonists glycine and D-serine and N-methyl-D-aspartate receptor hypofunction. Both carriers had a diagnosis of a psychotic disorder. METHODS We carried out two double-blind, placebo-controlled clinical trials of N-methyl-D-aspartate receptor augmentation of psychotropic drug treatment in these two individuals. Glycine was used in the first clinical trial, and D-cycloserine was used in the second one. RESULTS Glycine or D-cycloserine augmentation of psychotropic drug treatment each improved psychotic and mood symptoms in placebo-controlled trials. CONCLUSIONS These results provide two independent proof-of-principle demonstrations of symptom relief by targeting a specific genotype and explicitly link an individual mutation to the pathophysiology of psychosis and treatment response.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Dost Ongur
- McLean Hospital, Belmont, MA.,Harvard Medical School, Boston, MA
| | - Marc J. Kaufman
- McLean Hospital, Belmont, MA.,Harvard Medical School, Boston, MA
| | | | - Arthur J. Siegel
- McLean Hospital, Belmont, MA.,Harvard Medical School, Boston, MA
| | | | | | - Daniel C. Javitt
- Columbia University Medical Center, New York, NY.,Nathan Kline Institute, Orangeburg, NY
| | - Dan Rujescu
- Department of Psychiatry, Psychotherapy, and Psychosomatics, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Scott Hebbring
- Center for Human Genetics, Marshfield Clinic Research Institute, Marshfield, WI
| | | | | | | | | | | | | | | | | | | | - Donald C. Goff
- Nathan Kline Institute, Orangeburg, NY.,Department of Psychiatry, New York University Langone Medical Center, New York, NY
| | - James I. Hudson
- McLean Hospital, Belmont, MA.,Harvard Medical School, Boston, MA
| | | | - Joseph T. Coyle
- McLean Hospital, Belmont, MA.,Harvard Medical School, Boston, MA
| | - Uwe Rudolph
- McLean Hospital, Belmont, MA.,Harvard Medical School, Boston, MA
| | - Deborah L. Levy
- McLean Hospital, Belmont, MA.,Harvard Medical School, Boston, MA
| |
Collapse
|
83
|
Fehér E, Szatmári I, Dudás T, Zalatnai A, Farkas T, Lőrinczi B, Fülöp F, Vécsei L, Toldi J. Structural Evaluation and Electrophysiological Effects of Some Kynurenic Acid Analogs. Molecules 2019; 24:molecules24193502. [PMID: 31561643 PMCID: PMC6803921 DOI: 10.3390/molecules24193502] [Citation(s) in RCA: 7] [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: 09/12/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/28/2023] Open
Abstract
Kynurenic acid (KYNA), a metabolite of tryptophan, as an excitatory amino acid receptor antagonist is an effective neuroprotective agent in case of excitotoxicity, which is the hallmark of brain ischemia and several neurodegenerative processes. Therefore, kynurenine pathway, KYNA itself, and its derivatives came into the focus of research. During the past fifteen years, our research group has developed several neuroactive KYNA derivatives, some of which proved to be neuroprotective in preclinical studies. In this study, the synthesis of these KYNA derivatives and their evaluation with divergent molecular characteristics are presented together with their most typical effects on the monosynaptic transmission in CA1 region of the hippocampus of the rat. Their effects on the basic neuronal activity (on the field excitatory postsynaptic potentials: fEPSP) were studied in in vitro hippocampal slices in 1 and 200 μM concentrations. KYNA and its derivative 4 in both 1 and 200 μM concentrations proved to be inhibitory, while derivative 8 only in 200 μM decreased the amplitudes of fEPSPs. Derivative 5 facilitated the fEPSPs in 200 μM concentration. This is the first comparative study which evaluates the structural and functional differences of formerly and newly developed KYNA analogs. Considerations on possible relations between molecular structures and their physiological effects are presented.
Collapse
Affiliation(s)
- Evelin Fehér
- Department of Neurology, Interdisciplinary Excellence Centre, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary.
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary.
| | - István Szatmári
- Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary.
- Stereochemistry Research Group of the Hungarian Academy of Sciences, Eötvös utca 6, H-6720 Szeged, Hungary.
- Institute of Pharmaceutical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary.
| | - Tamás Dudás
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary.
| | - Anna Zalatnai
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary.
| | - Tamás Farkas
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary.
| | - Bálint Lőrinczi
- Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary.
- Stereochemistry Research Group of the Hungarian Academy of Sciences, Eötvös utca 6, H-6720 Szeged, Hungary.
| | - Ferenc Fülöp
- Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary.
- Stereochemistry Research Group of the Hungarian Academy of Sciences, Eötvös utca 6, H-6720 Szeged, Hungary.
- Institute of Pharmaceutical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary.
| | - László Vécsei
- Department of Neurology, Interdisciplinary Excellence Centre, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary.
- MTA-SZTE Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary.
| | - József Toldi
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary.
| |
Collapse
|
84
|
Burgdorf KS, Trabjerg BB, Pedersen MG, Nissen J, Banasik K, Pedersen OB, Sørensen E, Nielsen KR, Larsen MH, Erikstrup C, Bruun-Rasmussen P, Westergaard D, Thørner LW, Hjalgrim H, Paarup HM, Brunak S, Pedersen CB, Torrey EF, Werge T, Mortensen PB, Yolken RH, Ullum H. Large-scale study of Toxoplasma and Cytomegalovirus shows an association between infection and serious psychiatric disorders. Brain Behav Immun 2019; 79:152-158. [PMID: 30685531 DOI: 10.1016/j.bbi.2019.01.026] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/11/2019] [Accepted: 01/23/2019] [Indexed: 10/27/2022] Open
Abstract
BACKGROUND Common infectious pathogens have been associated with psychiatric disorders, self-violence and risk-taking behavior. METHODS This case-control study reviews register data on 81,912 individuals from the Danish Blood Donor Study to identify individuals who have a psychiatric diagnosis (N = 2591), have attempted or committed suicide (N = 655), or have had traffic accidents (N = 2724). For all cases, controls were frequency matched by age and sex, resulting in 11,546 participants. Plasma samples were analyzed for immunoglobulin G (IgG) antibodies against Toxoplasma gondii and cytomegalovirus (CMV). RESULTS T. gondii was detected in 25·9% of the population and was associated with schizophrenia (odds ratio [OR], 1·47; 95% confidence interval [CI], 1·03-2·09). Accounting for temporality, with pathogen exposure preceding outcome, the association was even stronger (IRR, 2·78; 95% CI, 1·27-6·09). A very weak association between traffic accident and toxoplasmosis (OR, 1·11; 95% CI, 1·00-1·23, p = 0.054) was found. CMV was detected in 60·8% of the studied population and was associated with any psychiatric disorder (OR, 1·17; 95% CI, 1·06-1·29), but also with a smaller group of neurotic, stress-related, and somatoform disorders (OR, 1·27; 95% CI, 1·12-1·44), and with attempting or committing suicide (OR, 1·31; 95% CI, 1·10-1·56). Accounting for temporality, any psychiatric disorder (IRR, 1·37; 95% CI, 1·08-1·74) and mood disorders (IRR, 1·43; 95% CI, 1·01-2·04) were associated with exposure to CMV. No association between traffic accident and CMV (OR, 1·06; 95% CI, 0·97-1·17) was found. CONCLUSIONS This large-scale serological study is the first study to examine temporality of pathogen exposure and to provide evidence of a causal relationship between T. gondii and schizophrenia, and between CMV and any psychiatric disorder.
Collapse
Affiliation(s)
| | - Betina B Trabjerg
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Denmark
| | - Marianne Giørtz Pedersen
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Denmark; Centre for Integrated Register-based Research, CIRRAU, Aarhus University, Aarhus, Denmark
| | - Janna Nissen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Kaspar René Nielsen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Margit Hørup Larsen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Peter Bruun-Rasmussen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - David Westergaard
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Lise Wegner Thørner
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Henrik Hjalgrim
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark,; Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Carsten B Pedersen
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Denmark; Centre for Integrated Register-based Research, CIRRAU, Aarhus University, Aarhus, Denmark
| | | | - Thomas Werge
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Denmark; Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Copenhagen University Hospital, Roskilde, Denmark
| | - Preben Bo Mortensen
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Denmark; Centre for Integrated Register-based Research, CIRRAU, Aarhus University, Aarhus, Denmark
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Stanley Neurovirology Laboratory, Johns Hopkins University, USA
| | - Henrik Ullum
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
85
|
Zakhary G, Sherchan P, Li Q, Tang J, Zhang JH. Modification of kynurenine pathway via inhibition of kynurenine hydroxylase attenuates surgical brain injury complications in a male rat model. J Neurosci Res 2019; 98:155-167. [PMID: 31257634 DOI: 10.1002/jnr.24489] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 05/29/2019] [Accepted: 06/14/2019] [Indexed: 01/31/2023]
Abstract
Neurosurgical procedures result in surgically induced brain injury (SBI) that causes postoperative complications including brain edema and neuronal apoptosis in the surrounding brain tissue. SBI leads to the release of cytokines that indirectly cause the stimulation of kynurenine 3-monooxygenase (KMO) and the release of neurotoxic quinolinic acid (QUIN). This study tested a KMO inhibitor, RO 61-8048, to prevent postoperative brain edema and consequent neuronal apoptosis in an in vivo model of SBI. A rodent model of SBI was utilized which involves partial resection of the right frontal lobe. A total of 127 Sprague-Dawley male rats (weight 275-325 g) were randomly divided into the following groups: Sham surgical group, SBI, SBI + DMSO, SBI + RO 61-8048 (10 mg/kg), SBI + RO 61-8048 (40 mg/kg), and SBI + RO 61-8048 (40 mg/kg) + KAT II inhibitor PF-04859989 (5 mg/kg). RO 61-8048 was administered by intraperitoneal injection after SBI. Postoperative assessment at different time points included brain water content (brain edema), neurological scoring, and western blot. SBI increased brain water content (ipsilateral frontal lobe), decreased neurological function, and increased apoptotic markers compared with sham animals. Treatment with RO 61-8048 (40 mg/kg) reduced brain water content and improved long-term neurological function after SBI. RO 61-8048 increased the expression of kynurenic acid while reducing QUIN and apoptotic markers in the surrounding brain tissue after SBI. These neuroprotective effects were reversed by PF-04859989. This study suggests KMO inhibition via RO 61-8048 as a potential postoperative therapy following neurosurgical procedures.
Collapse
Affiliation(s)
- George Zakhary
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California
| | - Qian Li
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California.,Department of Neurosurgery, Loma Linda University, Loma Linda, California.,Department of Anesthesiology, Loma Linda University, Loma Linda, California
| |
Collapse
|
86
|
Duda W, Curzytek K, Kubera M, Connor TJ, Fagan EM, Basta-Kaim A, Trojan E, Papp M, Gruca P, Budziszewska B, Leśkiewicz M, Maes M, Lasoń W. Interaction of the immune-inflammatory and the kynurenine pathways in rats resistant to antidepressant treatment in model of depression. Int Immunopharmacol 2019; 73:527-538. [PMID: 31176083 DOI: 10.1016/j.intimp.2019.05.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 05/10/2019] [Accepted: 05/22/2019] [Indexed: 12/16/2022]
Abstract
The kynurenine pathway (KP), a major route of tryptophan catabolism, may be associated with the pathophysiology of depressive disorders. KP is responsible for ca. 99% of brain tryptophan metabolism via its degradation to kynurenine (KYN) catalyzed by indoleamine 2,3-dioxygenase (IDO). Some cytokines, such as interferon-γ (IFN-γ) and interleukin (IL)-6 are potent inducers of IDO. KYN is further converted by kynurenine aminotransferase (KAT) to the more neuroprotective kynurenic acid or by kynurenine 3-monooxygenase (KMO) to neurotoxic 3-hydroxykynurenine. The aim of the present study was to delineate whether the administration of imipramine (IMI) to rats subjected to chronic mild stress (CMS) may reverse behavioral changes induced by CMS in association with changes in immune-inflammatory markers and KP. We confirmed that the CMS procedure modeled one of the main symptoms of depression, i.e. anhedonia, and administration of IMI for 5 weeks resulted in a significant reduction in anhedonia in a majority of animals (CMS IMI-R animals), whereas 20% of animals did not respond to IMI treatment (CMS IMI-NR animals). We established that CMS procedure increased IFN-γ and IDO mRNA and decreased KAT II mRNA expression in the rat cortex. In the cortex and hippocampus, IMI treatment and non-responsiveness to IMI (in CMS IMI-NR animals) were associated with increased IL-6 mRNA expression. In the spleen, CMS increased production of IFN-γ and IL-6 proteins, while these cytokines were decreased by IMI in CMS IMI-R animals. Chronic IMI administration to CMS rats decreased IDO and KMO mRNA and protein expression and increased KAT II/KMO mRNA and protein ratio in IMI responders (CMS IMI-R) in comparison to CMS rats. In CMS IMI-NR rats, a significant increase in IDO mRNA expression and protein level in comparison with IMI responders was observed. Our findings indicate that resistance to therapeutic action of IMI could be explained by a deficiency of the inhibitory properties of IMI on IDO, KMO and KYN synthesis in the cortex. We conclude that the antidepressant activity of IMI may, at least in part, be explained by modulatory activities on the KAT II/KMO ratio in brain areas.
Collapse
Affiliation(s)
- Weronika Duda
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, PL 31-343 Krakow, Poland
| | - Katarzyna Curzytek
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, PL 31-343 Krakow, Poland
| | - Marta Kubera
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, PL 31-343 Krakow, Poland.
| | - Thomas J Connor
- Neuroimmunology Research Group, Department of Physiology, School of Medicine & Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Eimear M Fagan
- Neuroimmunology Research Group, Department of Physiology, School of Medicine & Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Agnieszka Basta-Kaim
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, PL 31-343 Krakow, Poland
| | - Ewa Trojan
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, PL 31-343 Krakow, Poland
| | - Mariusz Papp
- Behavioural Pharmacology Laboratory, Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, PL 31-343 Krakow, Poland
| | - Piotr Gruca
- Behavioural Pharmacology Laboratory, Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, PL 31-343 Krakow, Poland
| | - Bogusława Budziszewska
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, PL 31-343 Krakow, Poland
| | - Monika Leśkiewicz
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, PL 31-343 Krakow, Poland
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Władysław Lasoń
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, PL 31-343 Krakow, Poland
| |
Collapse
|
87
|
Relationship between toxoplasmosis and obsessive compulsive disorder: A systematic review and meta-analysis. PLoS Negl Trop Dis 2019; 13:e0007306. [PMID: 30969961 PMCID: PMC6457543 DOI: 10.1371/journal.pntd.0007306] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 03/15/2019] [Indexed: 11/19/2022] Open
Abstract
Background A few studies investigated the relationship between toxoplasmosis and mental disorders, such as obsessive compulsive disorder (OCD). However, the specific nature of the association between Toxoplasma gondii (T. gondii) infection and OCD is not yet clear. The aim of this study was to collect information on the relationship between OCD and toxoplasmosis and assess whether patients with toxoplasmosis are prone to OCD. Methods For the purpose of this study, 6 major electronic databases and the Internet search engine Google Scholar were searched for the published articles up to July 30th, 2018 with no restriction of language. The inverse variance method and the random effect model were used to combine the data. The values of odds ratio (OR) were estimated at 95% confidence interval (CI). Results A total of 9 case-control and 3 cross-sectional studies were included in our systematic review. However, 11 of these 12 articles were entered into the meta-analysis containing 9873 participants, out of whom 389 were with OCD (25.96% positive for toxoplasmosis) and 9484 were without OCD (17.12% positive for toxoplasmosis). The estimation of the random effect model indicated a significant common OR of 1.96 [95% CI: 1.32–2.90]. Conclusion This systematic review and meta-analysis revealed that toxoplasmosis could be as an associated factor for OCD (OR = 1.96). However, further prospective investigations are highly recommended to illuminate the underlying pathophysiological mechanisms of T. gondii infection in OCD and to better investigate the relationship between OCD and T. gondii infection. Toxoplasma gondii (T. gondii) is an obligate neurotropic parasite that infected about 25–30% of the total human population in the developed and developing countries. The obsessive compulsive disorder (OCD) is a psychiatric disease that affects the income and quality of life. Some studies confirmed an association between infectious agents as the associated or protective factors specifying the development of psychiatry diseases. Among various pathogens associated with psychological disorders, most of the attention is on T. gondii, which has a life-long asymptomatic latent phase after a short acute stage in healthy individuals. The detrimental effect of T. gondii on immunocompromised people and pregnant women is an important concern for public health. The correlation between toxoplasmosis and OCD is still relatively understudied with a paucity of documented findings. The previous meta-analysis reviewed only two studies and reported a 3.4-fold greater chance of OCD. The results of our study presented stronger evidence of a positive relationship between toxoplasmosis and OCD. Eventually, our research team hopes to present an overview of what is known and encourage more intensive research to determine the real impact of this parasite on the occurrence of OCD that may contribute to the prevention of OCD worldwide.
Collapse
|
88
|
The ‘Yin’ and the ‘Yang’ of the kynurenine pathway: excitotoxicity and neuroprotection imbalance in stress-induced disorders. Behav Pharmacol 2019; 30:163-186. [DOI: 10.1097/fbp.0000000000000477] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
89
|
Zádori D, Veres G, Szalárdy L, Klivényi P, Vécsei L. Alzheimer's Disease: Recent Concepts on the Relation of Mitochondrial Disturbances, Excitotoxicity, Neuroinflammation, and Kynurenines. J Alzheimers Dis 2019; 62:523-547. [PMID: 29480191 DOI: 10.3233/jad-170929] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The pathomechanism of Alzheimer's disease (AD) certainly involves mitochondrial disturbances, glutamate excitotoxicity, and neuroinflammation. The three main aspects of mitochondrial dysfunction in AD, i.e., the defects in dynamics, altered bioenergetics, and the deficient transport, act synergistically. In addition, glutamatergic neurotransmission is affected in several ways. The balance between synaptic and extrasynaptic glutamatergic transmission is shifted toward the extrasynaptic site contributing to glutamate excitotoxicity, a phenomenon augmented by increased glutamate release and decreased glutamate uptake. Neuroinflammation in AD is predominantly linked to central players of the innate immune system, with central nervous system (CNS)-resident microglia, astroglia, and perivascular macrophages having been implicated at the cellular level. Several abnormalities have been described regarding the activation of certain steps of the kynurenine (KYN) pathway of tryptophan metabolism in AD. First of all, the activation of indolamine 2,3-dioxygenase, the first and rate-limiting step of the pathway, is well-demonstrated. 3-Hydroxy-L-KYN and its metabolite, 3-hydroxy-anthranilic acid have pro-oxidant, antioxidant, and potent immunomodulatory features, giving relevance to their alterations in AD. Another metabolite, quinolinic acid, has been demonstrated to be neurotoxic, promoting glutamate excitotoxicity, reactive oxygen species production, lipid peroxidation, and microglial neuroinflammation, and its abundant presence in AD pathologies has been demonstrated. Finally, the neuroprotective metabolite, kynurenic acid, has been associated with antagonistic effects at glutamate receptors, free radical scavenging, and immunomodulation, giving rise to potential therapeutic implications. This review presents the multiple connections of KYN pathway-related alterations to three main domains of AD pathomechanism, such as mitochondrial dysfunction, excitotoxicity, and neuroinflammation, implicating possible therapeutic options.
Collapse
Affiliation(s)
- Dénes Zádori
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Gábor Veres
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Levente Szalárdy
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Péter Klivényi
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary.,MTA-SZTE Neuroscience Research Group, Szeged, Hungary
| |
Collapse
|
90
|
Sekine A, Fukuwatari T. Acute liver failure increases kynurenic acid production in rat brain via changes in tryptophan metabolism in the periphery. Neurosci Lett 2019; 701:14-19. [PMID: 30738081 DOI: 10.1016/j.neulet.2019.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 02/03/2019] [Accepted: 02/04/2019] [Indexed: 12/20/2022]
Abstract
The tryptophan metabolite, kynurenic acid (KYNA), is a preferential antagonist of the α7 nicotinic acetylcholine receptor and N-methyl-d-aspartic acid receptor at endogenous brain concentrations. Recent studies have suggested that increased brain KYNA levels are involved in psychiatric disorders such as schizophrenia and depression. Most of the brain kynurenine (KYN), the KYNA precursor, comes from the periphery, and the liver has a central role in the peripheral tryptophan metabolism. In this study, the effect of acute liver failure (ALF) on brain KYNA production and on the peripheral tryptophan metabolism was investigated in rats. ALF was induced by administration of the hepatotoxin, thioacetamide (TAA). Brain KYNA levels were increased by TAA-induced ALF, and these increases were consistent with KYN levels in the brain, serum and liver. These results suggest that the ALF-induced increase in serum KYN contributes to the increase in brain KYNA via elevated KYN uptake within the brain. This increase in serum KYN level can be caused by the changes in tryptophan-2,3-dioxygenase activity in the liver and the immune-related activation of indoleamine-2,3-dioxygenase in extrahepatic tissues. These findings suggest that hepatic dysfunction may contribute to neurological and psychiatric diseases associated with increased KYNA levels.
Collapse
Affiliation(s)
- Airi Sekine
- Department of Nutrition, School of Human Cultures, The University of Shiga Prefecture, 2500 Hassaka, Hikone, Shiga, 522-8533, Japan
| | - Tsutomu Fukuwatari
- Department of Nutrition, School of Human Cultures, The University of Shiga Prefecture, 2500 Hassaka, Hikone, Shiga, 522-8533, Japan.
| |
Collapse
|
91
|
Linking phencyclidine intoxication to the tryptophan-kynurenine pathway: Therapeutic implications for schizophrenia. Neurochem Int 2019; 125:1-6. [PMID: 30731185 DOI: 10.1016/j.neuint.2019.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/17/2019] [Accepted: 02/04/2019] [Indexed: 12/17/2022]
Abstract
Phencyclidine (PCP) is a dissociative anesthetic that induces psychotic symptoms and neurocognitive deficits in rodents similar to those observed in schizophrenia patients. PCP administration in healthy human subjects induces schizophrenia-like symptoms such as positive and negative symptoms, and a range of cognitive deficits. It has been reported that PCP, ketamine, and related drugs such as N-methyl-D-aspartate-type (NMDA) glutamate receptor antagonists, induce behavioral effects by blocking neurotransmission at NMDA receptors. Further, NMDA receptor antagonists reproduce specific aspects of the symptoms of schizophrenia. Neurochemical models based on the actions of PCP are well established, with increased focus on glutamatergic dysfunction as a basis for both symptoms and cognitive dysfunction in schizophrenia. On the other hand, the endogenous NMDA receptor antagonist, kynurenic acid (KYNA), which is a product of tryptophan-kynurenine pathway (KP) metabolism, is involved in schizophrenia pathogenesis. KYNA concentrations are elevated in the prefrontal cortex and cerebrospinal fluid of patients with schizophrenia. KYNA elevation affects neurotransmitter release in a similar manner to that of psychotomimetic agents such as PCP, underscoring a molecular basis of its involvement in schizophrenia pathophysiology. This review will highlight the relationship between PCP and KP metabolites based on evidence that both exogenous and endogenous NMDA receptor antagonists are involved in the pathogenesis of schizophrenia, and discuss our current understanding of the mechanisms underlying dysfunctional glutamatergic signaling as potential therapeutic targets for schizophrenia.
Collapse
|
92
|
Zhuravlev AV, Vetrovoy OV, Savvateeva-Popova EV. Enzymatic and non-enzymatic pathways of kynurenines' dimerization: the molecular factors for oxidative stress development. PLoS Comput Biol 2018; 14:e1006672. [PMID: 30532237 PMCID: PMC6301705 DOI: 10.1371/journal.pcbi.1006672] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/20/2018] [Accepted: 11/26/2018] [Indexed: 11/19/2022] Open
Abstract
Kynurenines, the products of tryptophan oxidative degradation, are involved in multiple neuropathologies, such as Huntington's chorea, Parkinson's disease, senile dementia, etc. The major cause for hydroxykynurenines's neurotoxicity is the oxidative stress induced by the reactive oxygen species (ROS), the by-products of L-3-hydroxykynurenine (L-3HOK) and 3-hydroxyanthranilic acid (3HAA) oxidative self-dimerization. 2-aminophenol (2AP), a structural precursor of L-3HOK and 3HAA, undergoes the oxidative conjugation to form 2-aminophenoxazinone. There are several modes of 2AP dimerization, including both enzymatic and non-enzymatic stages. In this study, the free energies for 2AP, L-3HOK and 3HAA dimerization stages have been calculated at B3LYP/6-311G(d,p)//6-311+(O)+G(d) level, both in the gas phase and in heptane or water solution. For the intermediates, ionization potentials and electron affinities were calculated, as well as free energy and kinetics of molecular oxygen interaction with several non-enzymatically formed dimers. H-atom donating power of the intermediates increases upon the progress of the oxidation, making possible generation of hydroperoxyl radical or hydrogen peroxide from O2 at the last stages. Among the dimerization intermediates, 2-aminophenoxazinole derivatives have the lowest ionization potential and can reduce O2 to superoxide anion. The rate for O-H homolytic bond dissociation is significantly higher than that for C-H bond in non-enzymatic quinoneimine conjugate. However, the last reaction passes irreversibly, reducing O2 to hydroperoxyl radical. The inorganic ferrous iron and the heme group of Drosophila phenoxazinone synthase significantly reduce the energy cost of 2AP H-atom abstraction by O2. We have also shown experimentally that total antioxidant capacity decreases in Drosophila mutant cardinal with L-3HOK excess relative to the wild type Canton-S, and lipid peroxidation decreases in aged cardinal. Taken together, our data supports the conception of hydroxykynurenines' dual role in neurotoxicity: serving as antioxidants themselves, blocking lipid peroxidation by H-atom donation, they also can easily generate ROS upon dimerization, leading to the oxidative stress development.
Collapse
Affiliation(s)
- Aleksandr V. Zhuravlev
- Laboratory of Neurogenetics, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint-Petersburg, Russia
- * E-mail:
| | - Oleg V. Vetrovoy
- Laboratory of Regulation of the Brain Neuronal Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint-Petersburg, Russia
- Department of Biochemistry, Faculty of Biology, Saint-Petersburg State University, Saint-Petersburg, Russia
| | - Elena V. Savvateeva-Popova
- Laboratory of Neurogenetics, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint-Petersburg, Russia
| |
Collapse
|
93
|
Time and frequency dependent changes in resting state EEG functional connectivity following lipopolysaccharide challenge in rats. PLoS One 2018; 13:e0206985. [PMID: 30418990 PMCID: PMC6231634 DOI: 10.1371/journal.pone.0206985] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022] Open
Abstract
Research has shown that inflammatory processes affect brain function and behavior through several neuroimmune pathways. However, high order brain functions affected by inflammation largely remain to be defined. Resting state functional connectivity of synchronized oscillatory activity is a valid approach to understand network processing and high order brain function under different experimental conditions. In the present study multi-electrode EEG recording in awake, freely moving rats was used to study resting state connectivity after administration of lipopolysaccharides (LPS). Male Wistar rats were implanted with 10 cortical surface electrodes and administered with LPS (2 mg/kg) and monitored for symptoms of sickness at 3, 6 and 24 h. Resting state connectivity and power were computed at baseline, 6 and 24 h. Three prominent connectivity bands were identified using a method resistant to spurious correlation: alpha (5–15 Hz), beta-gamma (20–80 Hz), and high frequency oscillation (150–200 Hz). The most prominent connectivity band, alpha, was strongly reduced 6 h after LPS administration, and returned to baseline at 24 h. Beta-gamma connectivity was also reduced at 6 h and remained reduced at 24 h. Interestingly, high frequency oscillation connectivity remained unchanged at 6 h and was impaired 24 h after LPS challenge. Expected elevations in delta and theta power were observed at 6 h after LPS administration, when behavioral symptoms of sickness were maximal. Notably, gamma and high frequency power were reduced 6 h after LPS and returned to baseline by 24 h, when the effects on connectivity were more evident. Finally, increases in cross-frequency coupling elicited by LPS were detected at 6 h for theta-gamma and at 24 h for theta-high frequency oscillations. These studies show that LPS challenge profoundly affects EEG connectivity across all identified bands in a time-dependent manner indicating that inflammatory processes disrupt both bottom-up and top-down communication across the cortex during the peak and resolution of inflammation.
Collapse
|
94
|
Dadvar S, Ferreira DMS, Cervenka I, Ruas JL. The weight of nutrients: kynurenine metabolites in obesity and exercise. J Intern Med 2018; 284:519-533. [PMID: 30141532 DOI: 10.1111/joim.12830] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Obesity ultimately results from an imbalance between energy intake and expenditure. However, in addition to their bioenergetic value, nutrients and their metabolites can function as important signalling molecules in energy homeostasis. Indeed, macronutrients and their metabolites can be direct regulators of metabolism through their actions on different organs. In turn, target organs can decide to use, store or transform the incoming nutrients depending on their physiological context and in coordination with other cell types. Tryptophan-kynurenine metabolites are an example of a family of compounds that can serve as systemic integrators of energy metabolism by signalling to different cell types. These include adipocytes, immune cells and muscle fibres, in addition to the well-known effects of kynurenine metabolites on the central nervous system. In the context of energy metabolism, several of the effects elicited by kynurenic acid are mediated by the G-protein-coupled receptor, GPR35. As GPR35 is expressed in tissues such as the adipose tissue, immune cells and the gastrointestinal tract, this receptor could be a potential therapeutic target for the treatment of obesity, diabetes and other metabolic diseases. In addition, metabolic disorders often coincide with states of chronic inflammation, which further highlights GPR35 as an integration node in conditions where inflammation skews metabolism. Defining the molecular interplay between different tissues in the regulation of energy homeostasis can help us understand interindividual variability in the response to nutrient intake and develop safe and efficient therapies to fight obesity and metabolic disease.
Collapse
Affiliation(s)
- S Dadvar
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - D M S Ferreira
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - I Cervenka
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - J L Ruas
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| |
Collapse
|
95
|
Lodge D, Watkins JC, Bortolotto ZA, Jane DE, Volianskis A. The 1980s: D-AP5, LTP and a Decade of NMDA Receptor Discoveries. Neurochem Res 2018; 44:516-530. [PMID: 30284673 PMCID: PMC6420420 DOI: 10.1007/s11064-018-2640-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/12/2018] [Accepted: 09/17/2018] [Indexed: 01/02/2023]
Abstract
In the 1960s and 70s, biochemical and pharmacological evidence was pointing toward glutamate as a synaptic transmitter at a number of distinct receptor classes, known as NMDA and non-NMDA receptors. The field, however, lacked a potent and highly selective antagonist to block these putative postsynaptic receptors. So, the discoveries in the early 1980s of d-AP5 as a selective NMDA receptor antagonist and of its ability to block synaptic events and plasticity were a major breakthrough leading to an explosion of knowledge about this receptor subtype. During the next 10 years, the role of NMDA receptors was established in synaptic transmission, long-term potentiation, learning and memory, epilepsy, pain, among others. Hints at pharmacological heterogeneity among NMDA receptors were followed by the cloning of separate subunits. The purpose of this review is to recognize the important contributions made in the 1980s by Graham L. Collingridge and other key scientists to the advances in our understanding of the functions of NMDA receptors throughout the central nervous system.
Collapse
Affiliation(s)
- D Lodge
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - J C Watkins
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Z A Bortolotto
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - D E Jane
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - A Volianskis
- School of Clinical Sciences, University of Bristol, Bristol, UK.
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
96
|
Sharma R, Razdan K, Bansal Y, Kuhad A. Rollercoaster ride of kynurenines: steering the wheel towards neuroprotection in Alzheimer's disease. Expert Opin Ther Targets 2018; 22:849-867. [PMID: 30223691 DOI: 10.1080/14728222.2018.1524877] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is associated with cerebral cognitive deficits exhibiting two cardinal hallmarks: accruement of extracellular amyloid plaques and intracellular neurofibrillary tangles composed of hyperphosphorylated tau protein. The currently accessible therapeutic armamentarium merely provides symptomatic relief. Therefore, the cry for prospective neuroprotective strategies seems to be the need of the hour. Areas covered: This review comprehensively establishes correlation between kynurenine pathway (KP) metabolites and AD with major emphasis on its two functionally contrasting neuroactive metabolites i.e. kynurenic acid (KYNA) and quinolinic acid (QUIN) and enlists various clinical studies which hold a potential for future therapeutics in AD. Also, major hypotheses of AD and mechanisms underlying them have been scrutinized with the aim to brush up the readers with basic pathology of AD. Expert opinion: KP is unique in itself as it holds two completely different domains i.e. neurotoxic QUIN and neuroprotective KYNA and disrupted equilibrium between the two has a hand in neurodegeneration. KYNA has long been demonstrated to be neuroprotective but lately being disparaged for cognitive side effects. But we blaze a trail by amalgamating the pharmacological mechanistic studies of KYNA in kinship with α7nAChRs, NMDARs and GABA which lends aid in favour of KA.
Collapse
Affiliation(s)
- Radhika Sharma
- a Pharmacology Research Laboratory , University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University , Chandigarh , India
| | - Karan Razdan
- b Pharmaceutics division , University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University , Chandigarh , India
| | - Yashika Bansal
- a Pharmacology Research Laboratory , University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University , Chandigarh , India
| | - Anurag Kuhad
- a Pharmacology Research Laboratory , University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University , Chandigarh , India
| |
Collapse
|
97
|
Secci ME, Mascia P, Sagheddu C, Beggiato S, Melis M, Borelli AC, Tomasini MC, Panlilio LV, Schindler CW, Tanda G, Ferré S, Bradberry CW, Ferraro L, Pistis M, Goldberg SR, Schwarcz R, Justinova Z. Astrocytic Mechanisms Involving Kynurenic Acid Control Δ 9-Tetrahydrocannabinol-Induced Increases in Glutamate Release in Brain Reward-Processing Areas. Mol Neurobiol 2018; 56:3563-3575. [PMID: 30151725 DOI: 10.1007/s12035-018-1319-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/14/2018] [Indexed: 12/27/2022]
Abstract
The reinforcing effects of Δ9-tetrahydrocannabinol (THC) in rats and monkeys, and the reinforcement-related dopamine-releasing effects of THC in rats, can be attenuated by increasing endogenous levels of kynurenic acid (KYNA) through systemic administration of the kynurenine 3-monooxygenase inhibitor, Ro 61-8048. KYNA is a negative allosteric modulator of α7 nicotinic acetylcholine receptors (α7nAChRs) and is synthesized and released by astroglia, which express functional α7nAChRs and cannabinoid CB1 receptors (CB1Rs). Here, we tested whether these presumed KYNA autoreceptors (α7nAChRs) and CB1Rs regulate glutamate release. We used in vivo microdialysis and electrophysiology in rats, RNAscope in situ hybridization in brain slices, and primary culture of rat cortical astrocytes. Acute systemic administration of THC increased extracellular levels of glutamate in the nucleus accumbens shell (NAcS), ventral tegmental area (VTA), and medial prefrontal cortex (mPFC). THC also reduced extracellular levels of KYNA in the NAcS. These THC effects were prevented by administration of Ro 61-8048 or the CB1R antagonist, rimonabant. THC increased the firing activity of glutamatergic pyramidal neurons projecting from the mPFC to the NAcS or to the VTA in vivo. These effects were averted by pretreatment with Ro 61-8048. In vitro, THC elicited glutamate release from cortical astrocytes (on which we demonstrated co-localization of the CB1Rs and α7nAChR mRNAs), and this effect was prevented by KYNA and rimonabant. These results suggest a key role of astrocytes in interactions between the endocannabinoid system, kynurenine pathway, and glutamatergic neurotransmission, with ramifications for the pathophysiology and treatment of psychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- Maria E Secci
- Behavioral Neuroscience Research Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Paola Mascia
- Behavioral Neuroscience Research Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Claudia Sagheddu
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Sarah Beggiato
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Andrea C Borelli
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Maria C Tomasini
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Leigh V Panlilio
- Behavioral Neuroscience Research Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Charles W Schindler
- Behavioral Neuroscience Research Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Gianluigi Tanda
- Molecular Targets and Medications Discovery Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Sergi Ferré
- Molecular Targets and Medications Discovery Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Charles W Bradberry
- Behavioral Neuroscience Research Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Luca Ferraro
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Marco Pistis
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
- National Research Council of Italy (CNR), Section of Cagliari, Neuroscience Institute, Monserrato, Italy
| | - Steven R Goldberg
- Behavioral Neuroscience Research Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zuzana Justinova
- Behavioral Neuroscience Research Branch, Intramural Research Program, Department of Health and Human Services, National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA.
| |
Collapse
|
98
|
Chang C, Fonseca KR, Li C, Horner W, Zawadzke LE, Salafia MA, Welch KA, Strick CA, Campbell BM, Gernhardt SS, Rong H, Sawant-Basak A, Liras J, Dounay A, Tuttle JB, Verhoest P, Maurer TS. Quantitative Translational Analysis of Brain Kynurenic Acid Modulation via Irreversible Kynurenine Aminotransferase II Inhibition. Mol Pharmacol 2018; 94:823-833. [PMID: 29853495 DOI: 10.1124/mol.118.111625] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/24/2018] [Indexed: 11/22/2022] Open
Abstract
Kynurenic acid (KYNA) plays a significant role in maintaining normal brain function, and abnormalities in KYNA levels have been associated with various central nervous system disorders. Confirmation of its causality in human diseases requires safe and effective modulation of central KYNA levels in the clinic. The kynurenine aminotransferases (KAT) II enzyme represents an attractive target for pharmacologic modulation of central KYNA levels; however, KAT II and KYNA turnover kinetics, which could contribute to the duration of pharmacologic effect, have not been reported. In this study, the kinetics of central KYNA-lowering effect in rats and nonhuman primates (NHPs, Cynomolgus macaques) was investigated using multiple KAT II irreversible inhibitors as pharmacologic probes. Mechanistic pharmacokinetic-pharmacodynamic analysis of in vivo responses to irreversible inhibition quantitatively revealed that 1) KAT II turnover is relatively slow [16-76 hours' half-life (t1/2)], whereas KYNA is cleared more rapidly from the brain (<1 hour t1/2) in both rats and NHPs, 2) KAT II turnover is slower in NHPs than in rats (76 hours vs. 16 hours t1/2, respectively), and 3) the percent contribution of KAT II to KYNA formation is constant (∼80%) across rats and NHPs. Additionally, modeling results enabled establishment of in vitro-in vivo correlation for both enzyme turnover rates and drug potencies. In summary, quantitative translational analysis confirmed the feasibility of central KYNA modulation in humans. Model-based analysis, where system-specific properties and drug-specific properties are mechanistically separated from in vivo responses, enabled quantitative understanding of the KAT II-KYNA pathway, as well as assisted development of promising candidates to test KYNA hypothesis in humans.
Collapse
Affiliation(s)
- Cheng Chang
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Kari R Fonseca
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Cheryl Li
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Weldon Horner
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Laura E Zawadzke
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Michelle A Salafia
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Kathryn A Welch
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Christine A Strick
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Brian M Campbell
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Steve S Gernhardt
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Haojing Rong
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Aarti Sawant-Basak
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Jennifer Liras
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Amy Dounay
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Jamison B Tuttle
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Patrick Verhoest
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| | - Tristan S Maurer
- Systems Modeling and Simulation Group, Pharmacokinetics, Dynamics and Metabolism, Medicine Design (C.C., C.L., T.S.M.), Neuroscience and Pain Research Unit (W.H., L.E.Z., M.A.S., K.A.W., C.A.S., B.M.C., A.D., J.B.T., P.V.), and Pharmacokinetics, Dynamics and Metabolism, Medicine Design (K.R.F., S.S.G., H.R., A.S.-B., J.L.), Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts
| |
Collapse
|
99
|
Relevance of Alternative Routes of Kynurenic Acid Production in the Brain. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:5272741. [PMID: 29977455 PMCID: PMC5994304 DOI: 10.1155/2018/5272741] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/24/2018] [Indexed: 01/24/2023]
Abstract
The catabolism of tryptophan has gained great importance in recent years due to the fact that the metabolites produced during this process, with neuroactive and redox properties, are involved in physiological and pathological events. One of these metabolites is kynurenic acid (KYNA), which is considered as a neuromodulator since it can interact with NMDA, nicotinic, and GPR35 receptors among others, modulating the release of neurotransmitters as glutamate, dopamine, and acetylcholine. Kynureninate production is attributed to kynurenine aminotransferases. However, in some physiological and pathological conditions, its high production cannot be explained just with kynurenine aminotransferases. This review focuses on the alternative mechanism whereby KYNA can be produced, either from D-amino acids or by means of other enzymes as D-amino acid oxidase or by the participation of free radicals. It is important to mention that an increase in KYNA levels in processes as brain development, aging, neurodegenerative diseases, and psychiatric disorders, which share common factors as oxidative stress, inflammation, immune response activation, and participation of gut microbiota that can also be related with the alternative routes of KYNA production, has been observed.
Collapse
|
100
|
Parthasarathy A, Cross PJ, Dobson RCJ, Adams LE, Savka MA, Hudson AO. A Three-Ring Circus: Metabolism of the Three Proteogenic Aromatic Amino Acids and Their Role in the Health of Plants and Animals. Front Mol Biosci 2018; 5:29. [PMID: 29682508 PMCID: PMC5897657 DOI: 10.3389/fmolb.2018.00029] [Citation(s) in RCA: 193] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 03/21/2018] [Indexed: 12/19/2022] Open
Abstract
Tyrosine, phenylalanine and tryptophan are the three aromatic amino acids (AAA) involved in protein synthesis. These amino acids and their metabolism are linked to the synthesis of a variety of secondary metabolites, a subset of which are involved in numerous anabolic pathways responsible for the synthesis of pigment compounds, plant hormones and biological polymers, to name a few. In addition, these metabolites derived from the AAA pathways mediate the transmission of nervous signals, quench reactive oxygen species in the brain, and are involved in the vast palette of animal coloration among others pathways. The AAA and metabolites derived from them also have integral roles in the health of both plants and animals. This review delineates the de novo biosynthesis of the AAA by microbes and plants, and the branching out of AAA metabolism into major secondary metabolic pathways in plants such as the phenylpropanoid pathway. Organisms that do not possess the enzymatic machinery for the de novo synthesis of AAA must obtain these primary metabolites from their diet. Therefore, the metabolism of AAA by the host animal and the resident microflora are important for the health of all animals. In addition, the AAA metabolite-mediated host-pathogen interactions in general, as well as potential beneficial and harmful AAA-derived compounds produced by gut bacteria are discussed. Apart from the AAA biosynthetic pathways in plants and microbes such as the shikimate pathway and the tryptophan pathway, this review also deals with AAA catabolism in plants, AAA degradation via the monoamine and kynurenine pathways in animals, and AAA catabolism via the 3-aryllactate and kynurenine pathways in animal-associated microbes. Emphasis will be placed on structural and functional aspects of several key AAA-related enzymes, such as shikimate synthase, chorismate mutase, anthranilate synthase, tryptophan synthase, tyrosine aminotransferase, dopachrome tautomerase, radical dehydratase, and type III CoA-transferase. The past development and current potential for interventions including the development of herbicides and antibiotics that target key enzymes in AAA-related pathways, as well as AAA-linked secondary metabolism leading to antimicrobials are also discussed.
Collapse
Affiliation(s)
- Anutthaman Parthasarathy
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY, United States
| | - Penelope J. Cross
- Biomolecular Interaction Centre and School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| | - Renwick C. J. Dobson
- Biomolecular Interaction Centre and School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| | - Lily E. Adams
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY, United States
| | - Michael A. Savka
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY, United States
| | - André O. Hudson
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY, United States
| |
Collapse
|