1
|
Tanaka M, Szabó Á, Vécsei L. Redefining Roles: A Paradigm Shift in Tryptophan-Kynurenine Metabolism for Innovative Clinical Applications. Int J Mol Sci 2024; 25:12767. [PMID: 39684480 DOI: 10.3390/ijms252312767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/16/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
The tryptophan-kynurenine (KYN) pathway has long been recognized for its essential role in generating metabolites that influence various physiological processes. Traditionally, these metabolites have been categorized into distinct, often opposing groups, such as pro-oxidant versus antioxidant, excitotoxic/neurotoxic versus neuroprotective. This dichotomous framework has shaped much of the research on conditions like neurodegenerative and neuropsychiatric disorders, as well as cancer, where metabolic imbalances are a key feature. The effects are significantly influenced by various factors, including the concentration of metabolites and the particular cellular milieu in which they are generated. A molecule that acts as neuroprotective at low concentrations may exhibit neurotoxic effects at elevated levels. The oxidative equilibrium of the surrounding environment can alter the function of KYN from an antioxidant to a pro-oxidant. This narrative review offers a comprehensive examination and analysis of the contemporary understanding of KYN metabolites, emphasizing their multifaceted biological functions and their relevance in numerous physiological and pathological processes. This underscores the pressing necessity for a paradigm shift in the comprehension of KYN metabolism. Understanding the context-dependent roles of KYN metabolites is vital for novel therapies in conditions like Alzheimer's disease, multiple sclerosis, and cancer. Comprehensive pathway modulation, including balancing inflammatory signals and enzyme regulation, offers promising avenues for targeted, effective treatments.
Collapse
Affiliation(s)
- Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - Ágnes Szabó
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, Korányi fasor 6, H-6720 Szeged, Hungary
| | - László Vécsei
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| |
Collapse
|
2
|
Stone TW, Williams RO. Tryptophan metabolism as a 'reflex' feature of neuroimmune communication: Sensor and effector functions for the indoleamine-2, 3-dioxygenase kynurenine pathway. J Neurochem 2024; 168:3333-3357. [PMID: 38102897 DOI: 10.1111/jnc.16015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/16/2023] [Accepted: 11/08/2023] [Indexed: 12/17/2023]
Abstract
Although the central nervous system (CNS) and immune system were regarded as independent entities, it is now clear that immune system cells can influence the CNS, and neuroglial activity influences the immune system. Despite the many clinical implications for this 'neuroimmune interface', its detailed operation at the molecular level remains unclear. This narrative review focuses on the metabolism of tryptophan along the kynurenine pathway, since its products have critical actions in both the nervous and immune systems, placing it in a unique position to influence neuroimmune communication. In particular, since the kynurenine pathway is activated by pro-inflammatory mediators, it is proposed that physical and psychological stressors are the stimuli of an organismal protective reflex, with kynurenine metabolites as the effector arm co-ordinating protective neural and immune system responses. After a brief review of the neuroimmune interface, the general perception of tryptophan metabolism along the kynurenine pathway is expanded to emphasize this environmentally driven perspective. The initial enzymes in the kynurenine pathway include indoleamine-2,3-dioxygenase (IDO1), which is induced by tissue damage, inflammatory mediators or microbial products, and tryptophan-2,3-dioxygenase (TDO), which is induced by stress-induced glucocorticoids. In the immune system, kynurenic acid modulates leucocyte differentiation, inflammatory balance and immune tolerance by activating aryl hydrocarbon receptors and modulates pain via the GPR35 protein. In the CNS, quinolinic acid activates N-methyl-D-aspartate (NMDA)-sensitive glutamate receptors, whereas kynurenic acid is an antagonist: the balance between glutamate, quinolinic acid and kynurenic acid is a significant regulator of CNS function and plasticity. The concept of kynurenine and its metabolites as mediators of a reflex coordinated protection against stress helps to understand the variety and breadth of their activity. It should also help to understand the pathological origin of some psychiatric and neurodegenerative diseases involving the immune system and CNS, facilitating the development of new pharmacological strategies for treatment.
Collapse
Affiliation(s)
- Trevor W Stone
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Richard O Williams
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Alves LDF, Moore JB, Kell DB. The Biology and Biochemistry of Kynurenic Acid, a Potential Nutraceutical with Multiple Biological Effects. Int J Mol Sci 2024; 25:9082. [PMID: 39201768 PMCID: PMC11354673 DOI: 10.3390/ijms25169082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Kynurenic acid (KYNA) is an antioxidant degradation product of tryptophan that has been shown to have a variety of cytoprotective, neuroprotective and neuronal signalling properties. However, mammalian transporters and receptors display micromolar binding constants; these are consistent with its typically micromolar tissue concentrations but far above its serum/plasma concentration (normally tens of nanomolar), suggesting large gaps in our knowledge of its transport and mechanisms of action, in that the main influx transporters characterized to date are equilibrative, not concentrative. In addition, it is a substrate of a known anion efflux pump (ABCC4), whose in vivo activity is largely unknown. Exogeneous addition of L-tryptophan or L-kynurenine leads to the production of KYNA but also to that of many other co-metabolites (including some such as 3-hydroxy-L-kynurenine and quinolinic acid that may be toxic). With the exception of chestnut honey, KYNA exists at relatively low levels in natural foodstuffs. However, its bioavailability is reasonable, and as the terminal element of an irreversible reaction of most tryptophan degradation pathways, it might be added exogenously without disturbing upstream metabolism significantly. Many examples, which we review, show that it has valuable bioactivity. Given the above, we review its potential utility as a nutraceutical, finding it significantly worthy of further study and development.
Collapse
Affiliation(s)
- Luana de Fátima Alves
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - J. Bernadette Moore
- School of Food Science & Nutrition, University of Leeds, Leeds LS2 9JT, UK;
- Department of Biochemistry, Cell & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
| | - Douglas B. Kell
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
- Department of Biochemistry, Cell & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
| |
Collapse
|
4
|
Stone TW, Darlington LG, Badawy AAB, Williams RO. The Complex World of Kynurenic Acid: Reflections on Biological Issues and Therapeutic Strategy. Int J Mol Sci 2024; 25:9040. [PMID: 39201726 PMCID: PMC11354734 DOI: 10.3390/ijms25169040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
It has been unequivocally established that kynurenic acid has a number of actions in a variety of cells and tissues, raising, in principle, the possibility of targeting its generation, metabolism or sites of action to manipulate those effects to a beneficial therapeutic end. However, many basic aspects of the biology of kynurenic acid remain unclear, potentially leading to some confusion and misinterpretations of data. They include questions of the source, generation, targets, enzyme expression, endogenous concentrations and sites of action. This essay is intended to raise and discuss many of these aspects as a source of reference for more balanced discussion. Those issues are followed by examples of situations in which modulating and correcting kynurenic acid production or activity could bring significant therapeutic benefit, including neurological and psychiatric conditions, inflammatory diseases and cell protection. More information is required to obtain a clear overall view of the pharmacological environment relevant to kynurenic acid, especially with respect to the active concentrations of kynurenine metabolites in vivo and changed levels in disease. The data and ideas presented here should permit a greater confidence in appreciating the sites of action and interaction of kynurenic acid under different local conditions and pathologies, enhancing our understanding of kynurenic acid itself and the many clinical conditions in which manipulating its pharmacology could be of clinical value.
Collapse
Affiliation(s)
- Trevor W. Stone
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK;
| | - L. Gail Darlington
- Worthing Hospital, University Hospitals Sussex NHS Foundation Trust, Worthing BN11 2DH, UK
| | - Abdulla A.-B. Badawy
- Formerly School of Health Sciences, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| | - Richard O. Williams
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK;
| |
Collapse
|
5
|
Alarcan H, Bruno C, Emond P, Raoul C, Vourc'h P, Corcia P, Camu W, Veyrune JL, Garlanda C, Locati M, Juntas-Morales R, Saker S, Suehs C, Masseguin C, Kirby J, Shaw P, Malaspina A, De Vos J, Al-Chalabi A, Leigh PN, Tree T, Bensimon G, Blasco H. Pharmacometabolomics applied to low-dose interleukin-2 treatment in amyotrophic lateral sclerosis. Ann N Y Acad Sci 2024; 1536:82-91. [PMID: 38771698 DOI: 10.1111/nyas.15147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating motor neuron disease. The immunosuppressive functions of regulatory T lymphocytes (Tregs) are impaired in ALS, and correlate to disease progression. The phase 2a IMODALS trial reported an increase in Treg number in ALS patients following the administration of low-dose (ld) interleukin-2 (IL-2). We propose a pharmacometabolomics approach to decipher metabolic modifications occurring in patients treated with ld-IL-2 and its relationship with Treg response. Blood metabolomic profiles were determined on days D1, D64, and D85 from patients receiving 2 MIU of IL-2 (n = 12) and patients receiving a placebo (n = 12). We discriminated the three time points for the treatment group (average error rate of 42%). Among the important metabolites, kynurenine increased between D1 and D64, followed by a reduction at D85. The percentage increase of Treg number from D1 to D64, as predicted by the metabolome at D1, was highly correlated with the observed value. This study provided a proof of concept for metabolic characterization of the effect of ld-IL-2 in ALS. These data could present advances toward a personalized medicine approach and present pharmacometabolomics as a key tool to complement genomic and transcriptional data for drug characterization, leading to systems pharmacology.
Collapse
Affiliation(s)
- Hugo Alarcan
- Service de Biochimie et Biologie Moléculaire, CHRU Bretonneau, Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, Tours, France
| | - Clément Bruno
- Service de Pharmacologie Médicale, CHRU Bretonneau, Tours, France
| | - Patrick Emond
- UMR 1253 iBrain, Université de Tours, Inserm, Tours, France
- Laboratoire de Médecine nucléaire in vitro, CHRU Bretonneau, Tours, France
| | - Cédric Raoul
- INM, University of Montpellier, INSERM, Montpellier, France
- ALS Reference Center, University of Montpellier, CHU Montpellier, Montpellier, France
| | - Patrick Vourc'h
- Service de Biochimie et Biologie Moléculaire, CHRU Bretonneau, Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, Tours, France
| | - Philippe Corcia
- UMR 1253 iBrain, Université de Tours, Inserm, Tours, France
- Service de Neurologie, CHRU Bretonneau, Tours, France
| | - William Camu
- INM, University of Montpellier, INSERM, Montpellier, France
- ALS Reference Center, University of Montpellier, CHU Montpellier, Montpellier, France
| | - Jean-Luc Veyrune
- Institute of Human Genetics, University of Montepllier, Montpellier, France
| | - Cecilia Garlanda
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Italy
| | | | - Raúl Juntas-Morales
- Neuromuscular Diseases Unit, European Reference Network on Rare Neuromuscular Diseases (ERN EURO-NMD), Department of Neurology, Vall d'Hebron University Hospital, Barcelona, Spain
| | | | - Carey Suehs
- Laboratoire de Biostatistique, Epidémiologie clinique, Santé Publique, Innovation et Méthodologie (BESPIM), Université de Nîmes, Nîmes, France
| | - Christophe Masseguin
- Delegation for Clinical Research and Innovation, Nîmes University Hospital, Nîmes, France
| | - Janine Kirby
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Pamela Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Andrea Malaspina
- UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, University College London, Queen Square, London, UK
| | - John De Vos
- Department of Cell and Tissue Engineering, University Montpellier, CHU Montpellier, Montpellier, France
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | | | - Timothy Tree
- Department of Computer Science, University of Sheffield, Sheffield, UK
| | - Gilbert Bensimon
- Laboratoire de Biostatistique, Epidémiologie clinique, Santé Publique, Innovation et Méthodologie (BESPIM), Université de Nîmes, Nîmes, France
| | - Hélène Blasco
- Service de Biochimie et Biologie Moléculaire, CHRU Bretonneau, Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, Tours, France
| |
Collapse
|
6
|
Pathak S, Nadar R, Kim S, Liu K, Govindarajulu M, Cook P, Watts Alexander CS, Dhanasekaran M, Moore T. The Influence of Kynurenine Metabolites on Neurodegenerative Pathologies. Int J Mol Sci 2024; 25:853. [PMID: 38255925 PMCID: PMC10815839 DOI: 10.3390/ijms25020853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
As the kynurenine pathway's links to inflammation, the immune system, and neurological disorders became more apparent, it attracted more and more attention. It is the main pathway through which the liver breaks down Tryptophan and the initial step in the creation of nicotinamide adenine dinucleotide (NAD+) in mammals. Immune system activation and the buildup of potentially neurotoxic substances can result from the dysregulation or overactivation of this pathway. Therefore, it is not shocking that kynurenines have been linked to neurological conditions (Depression, Parkinson's, Alzheimer's, Huntington's Disease, Schizophrenia, and cognitive deficits) in relation to inflammation. Nevertheless, preclinical research has demonstrated that kynurenines are essential components of the behavioral analogs of depression and schizophrenia-like cognitive deficits in addition to mediators associated with neurological pathologies due to their neuromodulatory qualities. Neurodegenerative diseases have been extensively associated with neuroactive metabolites of the kynurenine pathway (KP) of tryptophan breakdown. In addition to being a necessary amino acid for protein synthesis, Tryptophan is also transformed into the important neurotransmitters tryptamine and serotonin in higher eukaryotes. In this article, a summary of the KP, its function in neurodegeneration, and the approaches being used currently to target the route therapeutically are discussed.
Collapse
Affiliation(s)
- Suhrud Pathak
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Rishi Nadar
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Shannon Kim
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Keyi Liu
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Manoj Govindarajulu
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Preston Cook
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | | | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Timothy Moore
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
7
|
Bian X, Wang Q, Wang Y, Lou S. The function of previously unappreciated exerkines secreted by muscle in regulation of neurodegenerative diseases. Front Mol Neurosci 2024; 16:1305208. [PMID: 38249295 PMCID: PMC10796786 DOI: 10.3389/fnmol.2023.1305208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/05/2023] [Indexed: 01/23/2024] Open
Abstract
The initiation and progression of neurodegenerative diseases (NDs), distinguished by compromised nervous system integrity, profoundly disrupt the quality of life of patients, concurrently exerting a considerable strain on both the economy and the social healthcare infrastructure. Exercise has demonstrated its potential as both an effective preventive intervention and a rehabilitation approach among the emerging therapeutics targeting NDs. As the largest secretory organ, skeletal muscle possesses the capacity to secrete myokines, and these myokines can partially improve the prognosis of NDs by mediating the muscle-brain axis. Besides the well-studied exerkines, which are secreted by skeletal muscle during exercise that pivotally exert their beneficial function, the physiological function of novel exerkines, e.g., apelin, kynurenic acid (KYNA), and lactate have been underappreciated previously. Herein, this review discusses the roles of these novel exerkines and their mechanisms in regulating the progression and improvement of NDs, especially the significance of their functions in improving NDs' prognoses through exercise. Furthermore, several myokines with potential implications in ameliorating ND progression are proposed as the future direction for investigation. Elucidation of the function of exerkines secreted by skeletal muscle in the regulation of NDs advances the understanding of its pathogenesis and facilitates the development of therapeutics that intervene in these processes to cure NDs.
Collapse
Affiliation(s)
- Xuepeng Bian
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Qian Wang
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Yibing Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Shujie Lou
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
8
|
Uceda S, Echeverry-Alzate V, Reiriz-Rojas M, Martínez-Miguel E, Pérez-Curiel A, Gómez-Senent S, Beltrán-Velasco AI. Gut Microbial Metabolome and Dysbiosis in Neurodegenerative Diseases: Psychobiotics and Fecal Microbiota Transplantation as a Therapeutic Approach-A Comprehensive Narrative Review. Int J Mol Sci 2023; 24:13294. [PMID: 37686104 PMCID: PMC10487945 DOI: 10.3390/ijms241713294] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
The comprehensive narrative review conducted in this study delves into the mechanisms of communication and action at the molecular level in the human organism. The review addresses the complex mechanism involved in the microbiota-gut-brain axis as well as the implications of alterations in the microbial composition of patients with neurodegenerative diseases. The pathophysiology of neurodegenerative diseases with neuronal loss or death is analyzed, as well as the mechanisms of action of the main metabolites involved in the bidirectional communication through the microbiota-gut-brain axis. In addition, interventions targeting gut microbiota restructuring through fecal microbiota transplantation and the use of psychobiotics-pre- and pro-biotics-are evaluated as an opportunity to reduce the symptomatology associated with neurodegeneration in these pathologies. This review provides valuable information and facilitates a better understanding of the neurobiological mechanisms to be addressed in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sara Uceda
- BRABE Group, Psychology Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | - Víctor Echeverry-Alzate
- BRABE Group, Psychology Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | - Manuel Reiriz-Rojas
- BRABE Group, Psychology Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | - Esther Martínez-Miguel
- Health Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | - Ana Pérez-Curiel
- Health Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | - Silvia Gómez-Senent
- Health Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | | |
Collapse
|
9
|
Zhao L, Zhang F, Wang K, Zhang X, Hu G, Chen E, Qiu J, Yuan C, He J. Quinolinic acid catabolism is initiated by a novel four-component hydroxylase QuiA in Alcaligenes faecalis JQ191. ENVIRONMENTAL RESEARCH 2023; 216:114421. [PMID: 36162464 DOI: 10.1016/j.envres.2022.114421] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 06/16/2023]
Abstract
Quinolinic acid (QA) is an essential nitrogen-containing aromatic heterocyclic compounds in organisms and it also acts as an important intermediate in chemical industry, which has strong neurotoxicity and cytotoxicity. The wide range of sources and applications caused the release and accumulation of QA in the environment which might poses a hazard to ecosystems and human health. However, few research on the degradation of QA by microorganisms and toxicity of QA and its metabolites were reported. Alcaligenes faecalis JQ191 could degrade QA but the genetic foundation of QA degradation has not been studied. In this study, the gene cluster quiA1A2A3A4 was identified from A. faecalis JQ191, which was responsible for the initial catabolism step of QA. The quiA1A2A3A4 gene cluster encodes a novel cytoplasmic four-component hydroxylase QuiA. The 1H nuclear magnetic resonance indicated that QuiA catalyzed QA to 6-hydroxyquinolinic acid (6HQA) and the H218O-labeling analysis confirmed that the hydroxyl group incorporating into 6HQA was derived from water. Toxicity tests showed that the QA could approximately inhibit 20%-80% growth of Chlorella ellipsoidea, and 6HQA could relieve at least 50% QA growth inhibition of Chlorella ellipsoidea, indicating that the 6-hydroxylation of QA by QuiA is a detoxification process. This research provides new insights into the metabolism of QA by microorganism and potential application in the bioremediation of toxic pyridine derivatives-contaminated environments.
Collapse
Affiliation(s)
- Lingling Zhao
- Department of Microbiology, College of Life Sciences, Nanjing Agricultural University, Key Laboratory of Agricultural and Environmental Microbiology, Ministry of Agriculture and Rural Affairs, Nanjing, 210095, China
| | - Fuyin Zhang
- Department of Microbiology, College of Life Sciences, Nanjing Agricultural University, Key Laboratory of Agricultural and Environmental Microbiology, Ministry of Agriculture and Rural Affairs, Nanjing, 210095, China
| | - Kexin Wang
- Department of Microbiology, College of Life Sciences, Nanjing Agricultural University, Key Laboratory of Agricultural and Environmental Microbiology, Ministry of Agriculture and Rural Affairs, Nanjing, 210095, China
| | - Xuan Zhang
- Department of Microbiology, College of Life Sciences, Nanjing Agricultural University, Key Laboratory of Agricultural and Environmental Microbiology, Ministry of Agriculture and Rural Affairs, Nanjing, 210095, China
| | - Gang Hu
- Laboratory Centre of Life Science, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - E Chen
- The Environmental Monitoring Center of Gansu Province, Lanzhou, China
| | - Jiguo Qiu
- Department of Microbiology, College of Life Sciences, Nanjing Agricultural University, Key Laboratory of Agricultural and Environmental Microbiology, Ministry of Agriculture and Rural Affairs, Nanjing, 210095, China
| | - Cansheng Yuan
- College of Rural Revitalization, Jiangsu Open University, Nanjing, Jiangsu, 210036, China
| | - Jian He
- Department of Microbiology, College of Life Sciences, Nanjing Agricultural University, Key Laboratory of Agricultural and Environmental Microbiology, Ministry of Agriculture and Rural Affairs, Nanjing, 210095, China; College of Rural Revitalization, Jiangsu Open University, Nanjing, Jiangsu, 210036, China.
| |
Collapse
|
10
|
Stone TW, Clanchy FIL, Huang YS, Chiang NY, Darlington LG, Williams RO. An integrated cytokine and kynurenine network as the basis of neuroimmune communication. Front Neurosci 2022; 16:1002004. [PMID: 36507331 PMCID: PMC9729788 DOI: 10.3389/fnins.2022.1002004] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022] Open
Abstract
Two of the molecular families closely associated with mediating communication between the brain and immune system are cytokines and the kynurenine metabolites of tryptophan. Both groups regulate neuron and glial activity in the central nervous system (CNS) and leukocyte function in the immune system, although neither group alone completely explains neuroimmune function, disease occurrence or severity. This essay suggests that the two families perform complementary functions generating an integrated network. The kynurenine pathway determines overall neuronal excitability and plasticity by modulating glutamate receptors and GPR35 activity across the CNS, and regulates general features of immune cell status, surveillance and tolerance which often involves the Aryl Hydrocarbon Receptor (AHR). Equally, cytokines and chemokines define and regulate specific populations of neurons, glia or immune system leukocytes, generating more specific responses within restricted CNS regions or leukocyte populations. In addition, as there is a much larger variety of these compounds, their homing properties enable the superimposition of dynamic variations of cell activity upon local, spatially limited, cell populations. This would in principle allow the targeting of potential treatments to restricted regions of the CNS. The proposed synergistic interface of 'tonic' kynurenine pathway affecting baseline activity and the superimposed 'phasic' cytokine system would constitute an integrated network explaining some features of neuroimmune communication. The concept would broaden the scope for the development of new treatments for disorders involving both the CNS and immune systems, with safer and more effective agents targeted to specific CNS regions.
Collapse
Affiliation(s)
- Trevor W. Stone
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom,*Correspondence: Trevor W. Stone,
| | - Felix I. L. Clanchy
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Yi-Shu Huang
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Nien-Yi Chiang
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - L. Gail Darlington
- Department of Internal Medicine, Ashtead Hospital, Ashtead, United Kingdom
| | - Richard O. Williams
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
11
|
Zheng H, Teague TK, Yeh FC, Burrows K, Figueroa-Hall LK, Aupperle RL, Khalsa SS, Paulus MP, Savitz J. C-Reactive protein and the kynurenic acid to quinolinic acid ratio are independently associated with white matter integrity in major depressive disorder. Brain Behav Immun 2022; 105:180-189. [PMID: 35853557 PMCID: PMC9983279 DOI: 10.1016/j.bbi.2022.07.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/01/2022] [Accepted: 07/14/2022] [Indexed: 02/09/2023] Open
Abstract
Kynurenic acid (KynA) and quinolinic acid (QA) are neuroactive kynurenine pathway (KP) metabolites that have neuroprotective and neurotoxic properties, respectively. At least partly as a result of immune activation, the ratio of KynA to QA in the blood is reduced in major depressive disorder (MDD) and has been reported to be positively correlated with gray matter volume in depression. This study examined whether the inflammatory mediator, C-reactive protein (CRP) and the putative neuroprotective index, KynA/QA, were associated with white matter integrity in MDD, and secondly, whether any such associations were independent of each other or whether the effect of CRP was mediated by KynA/QA. One hundred and sixty-six participants in the Tulsa 1000 study with a DSM-V diagnosis of MDD completed diffusion tensor imaging and provided a serum sample for the quantification of CRP, KynA, and QA. Correlational tractography was performed using DSI Studio to map the specific white matter pathways that correlated with CRP and KynA/QA. CRP was negatively related to KynA/QA (standardized beta coefficient, SBC = -0.35 with standard error, Std.E = 0.13, p < 0.01) after controlling for nine possible confounders, i.e., age, sex, body mass index (BMI), medication status, lifetime alcohol use, severity of depression, severity of anxiety, length of illness, and smoking status. Higher concentrations of CRP were associated with decreased white matter integrity (fractional anisotropy, FA) of the bilateral cingulum and fornix after controlling for the nine potential confounders (SBC = -0.43, Std.E = 0.13, p = 0.002). Greater serum KynA/QA was associated with increased white matter integrity of the bilateral fornix, bilateral superior thalamic radiations, corpus callosum, and bilateral cingulum bundles after controlling for the same possible confounders (SBC = 0.26, Std.E = 0.09, p = 0.005). The relationship between CRP and FA was not mediated by KynA/QA. Exploratory analyses also showed that KynA/QA but not CRP was associated with self-reported positive affect, attentiveness, and fatigue measured with the PANASX (SBCs = 0.17-0.23). Taken together, these results are consistent with the hypothesis that within a subgroup of MDD patients, a higher level of systemic inflammation alters the balance of KP metabolism but also raise the possibility that CRP and neuroactive KP metabolites represent independent molecular mechanisms underlying white matter alterations in MDD.
Collapse
Affiliation(s)
- Haixia Zheng
- Laureate Institute for Brain Research, Tulsa, OK 74136, USA
| | - T Kent Teague
- Department of Surgery, University of Oklahoma School of Community Medicine, Tulsa, OK 74135, USA; Department of Psychiatry, University of Oklahoma School of Community Medicine, Tulsa, OK 74135, USA; Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK 74107, USA
| | - Fang-Cheng Yeh
- Department of Neurological Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | | | - Robin L Aupperle
- Laureate Institute for Brain Research, Tulsa, OK 74136, USA; Oxley College of Health Sciences, The University of Tulsa, Tulsa, OK 74119, USA
| | - Sahib S Khalsa
- Laureate Institute for Brain Research, Tulsa, OK 74136, USA; Oxley College of Health Sciences, The University of Tulsa, Tulsa, OK 74119, USA
| | - Martin P Paulus
- Laureate Institute for Brain Research, Tulsa, OK 74136, USA; Oxley College of Health Sciences, The University of Tulsa, Tulsa, OK 74119, USA
| | - Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa, OK 74136, USA; Oxley College of Health Sciences, The University of Tulsa, Tulsa, OK 74119, USA.
| |
Collapse
|
12
|
The Role of Tryptophan Dysmetabolism and Quinolinic Acid in Depressive and Neurodegenerative Diseases. Biomolecules 2022; 12:biom12070998. [PMID: 35883554 PMCID: PMC9313172 DOI: 10.3390/biom12070998] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/04/2022] [Accepted: 07/14/2022] [Indexed: 02/04/2023] Open
Abstract
Emerging evidence suggests that neuroinflammation is involved in both depression and neurodegenerative diseases. The kynurenine pathway, generating metabolites which may play a role in pathogenesis, is one of several competing pathways of tryptophan metabolism. The present article is a narrative review of tryptophan metabolism, neuroinflammation, depression, and neurodegeneration. A disturbed tryptophan metabolism with increased activity of the kynurenine pathway and production of quinolinic acid may result in deficiencies in tryptophan and derived neurotransmitters. Quinolinic acid is an N-methyl-D-aspartate receptor agonist, and raised levels in CSF, together with increased levels of inflammatory cytokines, have been reported in mood disorders. Increased quinolinic acid has also been observed in neurodegenerative diseases, including Parkinson’s disease, Alzheimer’s disease, amyotrophic lateral sclerosis, and HIV-related cognitive decline. Oxidative stress in connection with increased indole-dioxygenase (IDO) activity and kynurenine formation may contribute to inflammatory responses and the production of cytokines. Increased formation of quinolinic acid may occur at the expense of kynurenic acid and neuroprotective picolinic acid. While awaiting ongoing research on potential pharmacological interventions on tryptophan metabolism, adequate protein intake with appropriate amounts of tryptophan and antioxidants may offer protection against oxidative stress and provide a balanced set of physiological receptor ligands.
Collapse
|
13
|
Sanz I, Altomare A, Mondanelli G, Protti M, Valsecchi V, Mercolini L, Volpi C, Regazzoni L. Chromatographic measurement of 3-hydroxyanthranilate 3,4-dioxygenase activity reveals that edaravone can mitigate the formation of quinolinic acid through a direct enzyme inhibition. J Pharm Biomed Anal 2022; 219:114948. [PMID: 35907317 DOI: 10.1016/j.jpba.2022.114948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 10/17/2022]
Abstract
Herein it is reported the development and application of two chromatographic assays for the measurement of the activity of 3-Hydroxyanthranilate-3,4-dioxygenase (3HAO). Such an enzyme converts 3-Hydroxyanthranilic acid (3HAA) to 2-amino-3-carboxymuconic semialdehyde (ACMS), which undergo a spontaneous, non-enzymatic cyclization to produce quinolinic acid (QUIN). The enzyme activity was measured by quantitation of the substrate consumption over time either with spectrophotometric (UV) or mass spectrometric (MS) detection upon reversed-phase chromatographic separation. MS detection resulted more selective and sensitive, but less accurate and precise. However, both methods have sufficient sensitivity to allow the measurement of enzyme activity with consistent results compared to literature data. Since MS detection allowed less sample consumption it was used to calculate the kinetics parameters (i.e., Vmax and Kd) of recombinant 3HAO. Another MS-based method was then developed to measure the amount of QUIN produced, revealing an incomplete conversion of 3HAA to QUIN. As suggested by previous studies, the enzyme activity was apparently sensitive to the redox state of the enzyme thiols. In fact, thiol reducing agents such as dithiothreitol (DTT) and glutathione (GSH), can alter the enzyme activity although the investigation on the exact mechanism involved in such effect was beyond the scope of the research. Interestingly, edaravone (EDA) induced an in vitro suppression of QUIN production through direct, competitive 3HAO inhibition. EDA is a molecule approved for the treatment of amyotrophic lateral sclerosis (ALS), a neurodegenerative disease associated with an increase of QUIN concentrations in both serum and cerebrospinal fluid. Although EDA was reported to mitigate ALS progression its mode of action is still largely unknown. Some studies reported antioxidant and radical scavenger properties of EDA, but none confirm a direct activity as 3HAO enzyme inhibitor. Since QUIN is reported to be a neurotoxic metabolite, 3HAO inhibition can contribute to the beneficial effect of EDA in ALS, although such a mechanism must be then confirmed in vivo. However, EDA might be a convenient scaffold for the design of selective 3HAO inhibitors with potential applications in ALS treatment.
Collapse
Affiliation(s)
- Ines Sanz
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133, Milan, Italy
| | - Alessandra Altomare
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133, Milan, Italy
| | - Giada Mondanelli
- Department of Medicine and Surgery, University of Perugia, Via Gambuli 1, 06132, Perugia, Italy
| | - Michele Protti
- Research group of Pharmaco-Toxicological Analysis (PTA Lab), Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Valeria Valsecchi
- Department of Neuroscience and Reproductive and Odontostomatological Sciences, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Laura Mercolini
- Research group of Pharmaco-Toxicological Analysis (PTA Lab), Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Claudia Volpi
- Department of Medicine and Surgery, University of Perugia, Via Gambuli 1, 06132, Perugia, Italy
| | - Luca Regazzoni
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133, Milan, Italy.
| |
Collapse
|
14
|
Huang Y, Zhao M, Chen X, Zhang R, Le A, Hong M, Zhang Y, Jia L, Zang W, Jiang C, Wang J, Fan X, Wang J. Tryptophan Metabolism in Central Nervous System Diseases: Pathophysiology and Potential Therapeutic Strategies. Aging Dis 2022; 14:858-878. [PMID: 37191427 DOI: 10.14336/ad.2022.0916] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 09/16/2022] [Indexed: 11/19/2022] Open
Abstract
The metabolism of L-tryptophan (TRP) regulates homeostasis, immunity, and neuronal function. Altered TRP metabolism has been implicated in the pathophysiology of various diseases of the central nervous system. TRP is metabolized through two main pathways, the kynurenine pathway and the methoxyindole pathway. First, TRP is metabolized to kynurenine, then kynurenic acid, quinolinic acid, anthranilic acid, 3-hydroxykynurenine, and finally 3-hydroxyanthranilic acid along the kynurenine pathway. Second, TRP is metabolized to serotonin and melatonin along the methoxyindole pathway. In this review, we summarize the biological properties of key metabolites and their pathogenic functions in 12 disorders of the central nervous system: schizophrenia, bipolar disorder, major depressive disorder, spinal cord injury, traumatic brain injury, ischemic stroke, intracerebral hemorrhage, multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease. Furthermore, we summarize preclinical and clinical studies, mainly since 2015, that investigated the metabolic pathway of TRP, focusing on changes in biomarkers of these neurologic disorders, their pathogenic implications, and potential therapeutic strategies targeting this metabolic pathway. This critical, comprehensive, and up-to-date review helps identify promising directions for future preclinical, clinical, and translational research on neuropsychiatric disorders.
Collapse
|
15
|
Fifita JA, Chan Moi Fat S, McCann EP, Williams KL, Twine NA, Bauer DC, Rowe DB, Pamphlett R, Kiernan MC, Tan VX, Blair IP, Guillemin GJ. Genetic Analysis of Tryptophan Metabolism Genes in Sporadic Amyotrophic Lateral Sclerosis. Front Immunol 2021; 12:701550. [PMID: 34194442 PMCID: PMC8236844 DOI: 10.3389/fimmu.2021.701550] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 05/31/2021] [Indexed: 01/17/2023] Open
Abstract
The essential amino acid tryptophan (TRP) is the initiating metabolite of the kynurenine pathway (KP), which can be upregulated by inflammatory conditions in cells. Neuroinflammation-triggered activation of the KP and excessive production of the KP metabolite quinolinic acid are common features of multiple neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). In addition to its role in the KP, genes involved in TRP metabolism, including its incorporation into proteins, and synthesis of the neurotransmitter serotonin, have also been genetically and functionally linked to these diseases. ALS is a late onset neurodegenerative disease that is classified as familial or sporadic, depending on the presence or absence of a family history of the disease. Heritability estimates support a genetic basis for all ALS, including the sporadic form of the disease. However, the genetic basis of sporadic ALS (SALS) is complex, with the presence of multiple gene variants acting to increase disease susceptibility and is further complicated by interaction with potential environmental factors. We aimed to determine the genetic contribution of 18 genes involved in TRP metabolism, including protein synthesis, serotonin synthesis and the KP, by interrogating whole-genome sequencing data from 614 Australian sporadic ALS cases. Five genes in the KP (AFMID, CCBL1, GOT2, KYNU, HAAO) were found to have either novel protein-altering variants, and/or a burden of rare protein-altering variants in SALS cases compared to controls. Four genes involved in TRP metabolism for protein synthesis (WARS) and serotonin synthesis (TPH1, TPH2, MAOA) were also found to carry novel variants and/or gene burden. These variants may represent ALS risk factors that act to alter the KP and lead to neuroinflammation. These findings provide further evidence for the role of TRP metabolism, the KP and neuroinflammation in ALS disease pathobiology.
Collapse
Affiliation(s)
- Jennifer A. Fifita
- Macquarie University Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Sandrine Chan Moi Fat
- Macquarie University Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Emily P. McCann
- Macquarie University Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Kelly L. Williams
- Macquarie University Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Natalie A. Twine
- Macquarie University Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- Australian e-Health Research Centre, Commonwealth Scientific and Industrial Research Organization, Health & Biosecurity Flagship, Sydney, NSW, Australia
| | - Denis C. Bauer
- Australian e-Health Research Centre, Commonwealth Scientific and Industrial Research Organization, Health & Biosecurity Flagship, Sydney, NSW, Australia
- Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- Applied BioSciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Dominic B. Rowe
- Macquarie University Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Roger Pamphlett
- Discipline of Pathology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- Department of Neuropathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - Matthew C. Kiernan
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Vanessa X. Tan
- Macquarie University Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Ian P. Blair
- Macquarie University Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Gilles J. Guillemin
- Macquarie University Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
16
|
Grifka-Walk HM, Jenkins BR, Kominsky DJ. Amino Acid Trp: The Far Out Impacts of Host and Commensal Tryptophan Metabolism. Front Immunol 2021; 12:653208. [PMID: 34149693 PMCID: PMC8213022 DOI: 10.3389/fimmu.2021.653208] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
Tryptophan (Trp) is an essential amino acid primarily derived from the diet for use by the host for protein synthesis. The intestinal tract is lined with cells, both host and microbial, that uptake and metabolize Trp to also generate important signaling molecules. Serotonin (5-HT), kynurenine and its downstream metabolites, and to a lesser extent other neurotransmitters are generated by the host to signal onto host receptors and elicit physiological effects. 5-HT production by neurons in the CNS regulates sleep, mood, and appetite; 5-HT production in the intestinal tract by enterochromaffin cells regulates gastric motility and inflammation in the periphery. Kynurenine can signal onto the aryl hydrocarbon receptor (AHR) to elicit pleiotropic responses from several cell types including epithelial and immune cells, or can be further metabolized into bioactive molecules to influence neurodegenerative disease. There is a remarkable amount of cross-talk with the microbiome with regard to tryptophan metabolites as well. The gut microbiome can regulate the production of host tryptophan metabolites and can use dietary or recycled trp to generate bioactive metabolites themselves. Trp derivatives like indole are able to signal onto xenobiotic receptors, including AHR, to elicit tolerogenic effects. Here, we review studies that demonstrate that tryptophan represents a key intra-kingdom signaling molecule.
Collapse
Affiliation(s)
| | | | - Douglas J. Kominsky
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| |
Collapse
|
17
|
Some CSF Kynurenine Pathway Intermediates Associated with Disease Evolution in Amyotrophic Lateral Sclerosis. Biomolecules 2021; 11:biom11050691. [PMID: 34063031 PMCID: PMC8147980 DOI: 10.3390/biom11050691] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 12/15/2022] Open
Abstract
The aim of this study was to evaluate the kynurenine pathway (KP) and amino acids profile, using mass spectrometry, in the cerebrospinal fluid (CSF) of 42 amyotrophic lateral sclerosis (ALS) patients at the diagnosis and 40 controls to detect early disorders of these pathways. Diagnostic and predictive ability (based on weight loss, forced vital capacity, ALS Functional Rating Scale-Revised evolution over 12 months, and survival time) of these metabolites were evaluated using univariate followed by supervised multivariate analysis. The multivariate model between ALS and controls was not significant but highlighted some KP metabolites (kynurenine (KYN), kynurenic acid (KYNA), 3-Hydroxynurenine (3-HK)/KYNA ratio), and amino acids (Lysine, asparagine) as involved in the discrimination between groups (accuracy 62%). It revealed a probable KP impairment toward neurotoxicity in ALS patients and in bulbar forms. Regarding the prognostic effect of metabolites, 12 were commonly discriminant for at least 3 of 4 disease evolution criteria. This investigation was crucial as it did not show significant changes in CSF concentrations of amino acids and KP intermediates in early ALS evolution. However, trends of KP modifications suggest further exploration. The unclear kinetics of neuroinflammation linked to KP support the interest in exploring these pathways during disease evolution through a longitudinal strategy.
Collapse
|
18
|
Garcez ML, Tan VX, Heng B, Guillemin GJ. Sodium Butyrate and Indole-3-propionic Acid Prevent the Increase of Cytokines and Kynurenine Levels in LPS-induced Human Primary Astrocytes. Int J Tryptophan Res 2021; 13:1178646920978404. [PMID: 33447046 PMCID: PMC7780186 DOI: 10.1177/1178646920978404] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 11/10/2020] [Indexed: 12/14/2022] Open
Abstract
The crosstalk between central nervous system (CNS) and gut microbiota plays key roles in neuroinflammation and chronic immune activation that are common features of all neurodegenerative diseases. Imbalance in the microbiota can lead to an increase in the intestinal permeability allowing toxins to diffuse and reach the CNS, as well as impairing the production of neuroprotective metabolites such as sodium butyrate (SB) and indole-3-propionic acid (IPA). The aim of the present study was to evaluate the effect of SB and IPA on LPS-induced production of cytokines and tryptophan metabolites in human astrocytes. Primary cultures of human astrocytes were pre-incubated with SB or IPA for 1 hour before treatment with LPS. Cell viability was not affected at 24, 48 or 72 hours after pre-treatment with SB, IPA or LPS treatment. SB was able to significantly prevent the increase of GM-CSF, MCP-1, IL-6 IL-12, and IL-13 triggered by LPS. SB and IPA also prevented inflammation indicated by the increase in kynurenine and kynurenine/tryptophan ratio induced by LPS treatment. IPA pre-treatment prevented the LPS-induced increase in MCP-1, IL-12, IL-13, and TNF-α levels 24 hours after pre-treatment, but had no effect on tryptophan metabolites. The present study showed for the first time that bacterial metabolites SB and IPA have potential anti-inflammatory effect on primary human astrocytes with potential therapeutic benefit in neurodegenerative disease characterized by the presence of chronic low-grade inflammation.
Collapse
Affiliation(s)
- Michelle L Garcez
- Neurochemistry Laboratory, Department of Biochemistry, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil.,Neurodegenerative diseases Research Group, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Vanessa X Tan
- Neurodegenerative diseases Research Group, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Benjamin Heng
- Neurodegenerative diseases Research Group, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Gilles J Guillemin
- Neurodegenerative diseases Research Group, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia.,PANDIS.org, Little Collins St, Melbourne VIC, Australia
| |
Collapse
|
19
|
Zhang S, Collier MEW, Heyes DJ, Giorgini F, Scrutton NS. Advantages of brain penetrating inhibitors of kynurenine-3-monooxygenase for treatment of neurodegenerative diseases. Arch Biochem Biophys 2020; 697:108702. [PMID: 33275878 DOI: 10.1016/j.abb.2020.108702] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/18/2020] [Accepted: 11/24/2020] [Indexed: 01/16/2023]
Abstract
Kynurenine-3-monooxygenase (KMO) is an important therapeutic target for several brain disorders that has been extensively studied in recent years. Potent inhibitors towards KMO have been developed and tested within different disease models, showing great therapeutic potential, especially in models of neurodegenerative disease. The inhibition of KMO reduces the production of downstream toxic kynurenine pathway metabolites and shifts the flux to the formation of the neuroprotectant kynurenic acid. However, the efficacy of KMO inhibitors in neurodegenerative disease has been limited by their poor brain permeability. Combined with virtual screening and prodrug strategies, a novel brain penetrating KMO inhibitor has been developed which dramatically decreases neurotoxic metabolites. This review highlights the importance of KMO as a drug target in neurological disease and the benefits of brain permeable inhibitors in modulating kynurenine pathway metabolites in the central nervous system.
Collapse
Affiliation(s)
- Shaowei Zhang
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - Mary E W Collier
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, UK
| | - Derren J Heyes
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, UK
| | - Nigel S Scrutton
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK.
| |
Collapse
|
20
|
Bhat A, Ray B, Mahalakshmi AM, Tuladhar S, Nandakumar DN, Srinivasan M, Essa MM, Chidambaram SB, Guillemin GJ, Sakharkar MK. Phosphodiesterase-4 enzyme as a therapeutic target in neurological disorders. Pharmacol Res 2020; 160:105078. [PMID: 32673703 DOI: 10.1016/j.phrs.2020.105078] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 02/08/2023]
Abstract
Phosphodiesterases (PDE) are a diverse family of enzymes (11 isoforms so far identified) responsible for the degradation of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) which are involved in several cellular and biochemical functions. Phosphodiesterase 4 (PDE4) is the major isoform within this group and is highly expressed in the mammalian brain. An inverse association between PDE4 and cAMP levels is the key mechanism in various pathophysiological conditions like airway inflammatory diseases-chronic obstruction pulmonary disease (COPD), asthma, psoriasis, rheumatoid arthritis, and neurological disorders etc. In 2011, roflumilast, a PDE4 inhibitor (PDE4I) was approved for the treatment of COPD. Subsequently, other PDE4 inhibitors (PDE4Is) like apremilast and crisaborole were approved by the Food and Drug Administration (FDA) for psoriasis, atopic dermatitis etc. Due to the adverse effects like unbearable nausea and vomiting, dose intolerance and diarrhoea, PDE4 inhibitors have very less clinical compliance. Efforts are being made to develop allosteric modulation with high specificity to PDE4 isoforms having better efficacy and lesser adverse effects. Interestingly, repositioning PDE4Is towards neurological disorders including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS) and sleep disorders, is gaining attention. This review is an attempt to summarize the data on the effects of PDE4 overexpression in neurological disorders and the use of PDE4Is and newer allosteric modulators as therapeutic options. We have also compiled a list of on-going clinical trials on PDE4 inhibitors in neurological disorders.
Collapse
Affiliation(s)
- Abid Bhat
- Dept. of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Bipul Ray
- Dept. of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | | | - Sunanda Tuladhar
- Dept. of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - D N Nandakumar
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Malathi Srinivasan
- Department of Lipid Science, CSIR - Central Food Technological Research Institute (CFTRI), CFTRI Campus, Mysuru, 570020, India
| | - Musthafa Mohamed Essa
- Ageing and Dementia Research Group, Sultan Qaboos University, Muscat, Oman; Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman.
| | - Saravana Babu Chidambaram
- Dept. of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India; Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India.
| | - Gilles J Guillemin
- Neuroinflammation group, Faculty of Medicine and Health Sciences, Macquarie University, NSW, 2109, Australia.
| | - Meena Kishore Sakharkar
- College of Pharmacy and Nutrition, University of Saskatchewan, 107, Wiggins Road, Saskatoon, SK, S7N 5C9, Canada
| |
Collapse
|
21
|
Lu Y, Shao M, Wu T. Kynurenine-3-monooxygenase: A new direction for the treatment in different diseases. Food Sci Nutr 2020; 8:711-719. [PMID: 32148781 PMCID: PMC7020307 DOI: 10.1002/fsn3.1418] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 11/24/2019] [Accepted: 12/21/2019] [Indexed: 12/14/2022] Open
Abstract
Kynurenine-3-monooxygenase (KMO) is an enzyme that relies on nicotinamide adenine dinucleotide phosphate (NADP), a key site in the kynurenine pathway (KP), which has great effects on neurological diseases, cancer, and peripheral inflammation. This review mainly pay attention to the research of KMO mechanism for the treatment of different diseases, and hopes to provide assistance for clinical and drug use. KMO controlling the chief division of the KP, which directly controls downstream product quinolinic acid (QUIN) and indirectly controls kynurenic acid (KYNA), plays an important role in many diseases, especially neurological diseases.
Collapse
Affiliation(s)
- Yifei Lu
- Institute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Mingmei Shao
- Institute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
22
|
Inoue H, Matsushige T, Ichiyama T, Okuno A, Takikawa O, Tomonaga S, Anlar B, Yüksel D, Otsuka Y, Kohno F, Hoshide M, Ohga S, Hasegawa S. Elevated quinolinic acid levels in cerebrospinal fluid in subacute sclerosing panencephalitis. J Neuroimmunol 2019; 339:577088. [PMID: 31733567 DOI: 10.1016/j.jneuroim.2019.577088] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 10/04/2019] [Accepted: 10/17/2019] [Indexed: 12/13/2022]
Abstract
Subacute sclerosing panencephalitis (SSPE) is a rare neurodegenerative disorder caused by a persistent infection with aberrant measles virus. Indoleamine-2, 3-dioxygenase (IDO) initiates the increased production of kynurenine pathway (KP) metabolites quinolinic acid (QUIN), which has an excitotoxic effect for neurons. We measured serum IDO activity and cerebrospinal fluid (CSF) levels of QUIN. The CSF QUIN levels were significantly higher in SSPE patients than in controls, and increased according as neurological disability in a patient studied. Elevation of CSF QUIN and progression of SSPE indicate a pathological role of KP metabolism in the inflammatory neurodestruction.
Collapse
Affiliation(s)
- Hirofumi Inoue
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan
| | - Takeshi Matsushige
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan.
| | - Takashi Ichiyama
- Division of Pediatrics, Tsudumigaura medical center for children with disabilities, Yamaguchi 745-0801, Japan
| | - Alato Okuno
- Faculty of Medical and Health Sciences, Tsukuba International University, Ibaraki 300-0051, Japan; Division of Medical Informatics and Bioinformatics, Kobe University Hospital, Hyogo 650-0017, Japan
| | - Osamu Takikawa
- Department of Research Promotion, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| | - Shozo Tomonaga
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8501, Japan
| | - Banu Anlar
- Department of Pediatric Neurology, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Deniz Yüksel
- Department of Pediatric Neurology, Dr. Sami Ulus Children's Hospital, Ankara, Turkey
| | - Yasushi Otsuka
- Department of Neurology, Toki General Hospital, Gifu 509-5193, Japan
| | - Fumitaka Kohno
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan; Division of Pediatrics, Tsudumigaura medical center for children with disabilities, Yamaguchi 745-0801, Japan
| | - Madoka Hoshide
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Shunji Hasegawa
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan
| |
Collapse
|
23
|
Tan VX, Guillemin GJ. Kynurenine Pathway Metabolites as Biomarkers for Amyotrophic Lateral Sclerosis. Front Neurosci 2019; 13:1013. [PMID: 31616242 PMCID: PMC6764462 DOI: 10.3389/fnins.2019.01013] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/06/2019] [Indexed: 12/19/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) currently lacks a robust and well-defined biomarker that can 1) assess the progression of the disease, 2) predict and/or delineate the various clinical subtypes, and 3) evaluate or predict a patient's response to treatments. The kynurenine Pathway (KP) of tryptophan degradation represent a promising candidate as it is involved with several neuropathological features present in ALS including neuroinflammation, excitotoxicity, oxidative stress, immune system activation and dysregulation of energy metabolism. Some of the KP metabolites (KPMs) can cross the blood brain barrier, and many studies have shown their levels are dysregulated in major neurodegenerative diseases including ALS. The KPMs can be easily analyzed in body fluids and tissue and as they are small molecules, and are stable. KPMs have a Janus face action, they can be either or both neurotoxic and/or neuroprotective depending of their levels. This mini review examines and presents evidence supporting the use of KPMs as a relevant set of biomarkers for ALS, and highlights the criteria required to achieve a valid biomarker set for ALS.
Collapse
Affiliation(s)
| | - Gilles J. Guillemin
- Macquarie University Centre for MND Research, Department of Biological Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
24
|
Zhang S, Sakuma M, Deora GS, Levy CW, Klausing A, Breda C, Read KD, Edlin CD, Ross BP, Wright Muelas M, Day PJ, O’Hagan S, Kell DB, Schwarcz R, Leys D, Heyes DJ, Giorgini F, Scrutton NS. A brain-permeable inhibitor of the neurodegenerative disease target kynurenine 3-monooxygenase prevents accumulation of neurotoxic metabolites. Commun Biol 2019; 2:271. [PMID: 31372510 PMCID: PMC6656724 DOI: 10.1038/s42003-019-0520-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 06/28/2019] [Indexed: 12/16/2022] Open
Abstract
Dysregulation of the kynurenine pathway (KP) leads to imbalances in neuroactive metabolites associated with the pathogenesis of several neurodegenerative disorders, including Huntington's disease (HD). Inhibition of the enzyme kynurenine 3-monooxygenase (KMO) in the KP normalises these metabolic imbalances and ameliorates neurodegeneration and related phenotypes in several neurodegenerative disease models. KMO is thus a promising candidate drug target for these disorders, but known inhibitors are not brain permeable. Here, 19 new KMO inhibitors have been identified. One of these (1) is neuroprotective in a Drosophila HD model but is minimally brain penetrant in mice. The prodrug variant (1b) crosses the blood-brain barrier, releases 1 in the brain, thereby lowering levels of 3-hydroxykynurenine, a toxic KP metabolite linked to neurodegeneration. Prodrug 1b will advance development of targeted therapies against multiple neurodegenerative and neuroinflammatory diseases in which KP likely plays a role, including HD, Alzheimer's disease, and Parkinson's disease.
Collapse
Affiliation(s)
- Shaowei Zhang
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Michiyo Sakuma
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Girdhar S. Deora
- School of Pharmacy, The University of Queensland, Brisbane, Queensland 4072 Australia
| | - Colin W. Levy
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Alex Klausing
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228 USA
| | - Carlo Breda
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH UK
| | - Kevin D. Read
- Drug Discovery Unit, School of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH UK
| | | | - Benjamin P. Ross
- School of Pharmacy, The University of Queensland, Brisbane, Queensland 4072 Australia
| | - Marina Wright Muelas
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Philip J. Day
- Manchester Institute of Biotechnology and Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL UK
| | - Stephen O’Hagan
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Douglas B. Kell
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228 USA
| | - David Leys
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Derren J. Heyes
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH UK
| | - Nigel S. Scrutton
- Manchester Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN UK
| |
Collapse
|
25
|
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: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
26
|
Abstract
OBJECTIVE The objective of this paper was to link the phytochemical and metabolic research treating quinolinic acid induced oxidative stress in neurodegenerative disorders. METHODS Quinolinic acid, a metabolite of the kynurenine pathway of tryptophan catabolism, plays a role in the oxidative stress associated with many neurological disorders and is used to simulate disorders such as Parkinson's disease. RESULTS In these models, phytochemicals have been shown to reduce striatal lesion size, reduce inflammation and prevent lipid peroxidation caused by quinolinic acid. CONCLUSION These results suggest that phenolic compounds, a class of phytochemicals, including flavonoids and diarylheptanoids, should be further studied to develop new treatments for oxidative stress related neurological disorders.
Collapse
Affiliation(s)
- K. Parasram
- Department of Biology, University of Windsor, Windsor, Canada
| |
Collapse
|
27
|
Quinolinic Acid Amyloid-like Fibrillar Assemblies Seed α-Synuclein Aggregation. J Mol Biol 2018; 430:3847-3862. [PMID: 30098337 DOI: 10.1016/j.jmb.2018.08.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 07/17/2018] [Accepted: 08/01/2018] [Indexed: 01/08/2023]
Abstract
Quinolinic acid (QA), a downstream neurometabolite in the kynurenine pathway, the biosynthetic pathway of tryptophan, is associated with neurodegenerative diseases pathology. Mutations in genes encoding kynurenine pathway enzymes, which control the level of QA production, are linked with elevated risk of developing Parkinson's disease. Recent findings have revealed the accumulation and deposition of QA in post-mortem samples, as well as in cellular models of Alzheimer's disease and related disorders. Furthermore, intrastriatal inoculation of mice with QA results in increased levels of phosphorylated α-synuclein and neurodegenerative pathological and behavioral characteristics. However, the cellular and molecular mechanisms underlying the involvement of QA accumulation in protein aggregation and neurodegeneration remain elusive. We recently established that self-assembled ordered structures are formed by various metabolites and hypothesized that these "metabolite amyloids" may seed amyloidogenic proteins. Here we demonstrate the formation of QA amyloid-like fibrillar assemblies and seeding of α-synuclein aggregation by these nanostructures both in vitro and in cell culture. Notably, α-synuclein aggregation kinetics was accelerated by an order of magnitude. Additional amyloid-like properties of QA assemblies were demonstrated using thioflavin T assay, powder X-ray diffraction and cell apoptosis analysis. Moreover, fluorescently labeled QA assemblies were internalized by neuronal cells and co-localized with α-synuclein aggregates. In addition, we observed cell-to-cell propagation of fluorescently labeled QA assemblies in a co-culture of treated and untreated cells. Our findings suggest that excess QA levels, due to mutations in the kynurenine pathway, for example, may lead to the formation of metabolite assemblies that seed α-synuclein aggregation, resulting in neuronal toxicity and induction of Parkinson's disease.
Collapse
|
28
|
Bo L, Guojun T, Li G. An Expanded Neuroimmunomodulation Axis: sCD83-Indoleamine 2,3-Dioxygenase-Kynurenine Pathway and Updates of Kynurenine Pathway in Neurologic Diseases. Front Immunol 2018; 9:1363. [PMID: 29963055 PMCID: PMC6013554 DOI: 10.3389/fimmu.2018.01363] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/01/2018] [Indexed: 12/30/2022] Open
Abstract
Many neurologic diseases are related to autoimmune dysfunction and a variety of molecules or reaction pathways are involved in the regulation of immune function of the nervous system. Soluble CD83 (sCD83) is the soluble form of CD83, a specific marker of mature dendritic cell, which has recently been shown to have an immunomodulatory effect. Indoleamine 2,3-dioxygenase (IDO; corresponding enzyme intrahepatic, tryptophan 2,3-dioxygenase, TDO), a rate-limiting enzyme of extrahepatic tryptophan kynurenine pathway (KP) participates in the immunoregulation through a variety of mechanisms solely or with the synergy of sCD83, and the imbalances of metabolites of KP were associated with immune dysfunction. With the complement of sCD83 to IDO-KP, a previously known immunomodulatory axis, this review focused on an expanded neuroimmunomodulation axis: sCD83-IDO-KP and its involvement in nervous system diseases.
Collapse
Affiliation(s)
- Li Bo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tan Guojun
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guo Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
29
|
Liang H, Wu C, Deng Y, Zhu L, Zhang J, Gan W, Tang C, Xu R. Aldehyde Dehydrogenases 1A2 Expression and Distribution are Potentially Associated with Neuron Death in Spinal Cord of Tg(SOD1*G93A)1Gur Mice. Int J Biol Sci 2017; 13:574-587. [PMID: 28539831 PMCID: PMC5441175 DOI: 10.7150/ijbs.19150] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 02/20/2017] [Indexed: 12/11/2022] Open
Abstract
The pathogenesis of amyotrophic lateral sclerosis (ALS) has not been unclear yet, it might be associated with the abnormal expression and distribution of certain proteins. Aldehyde dehydrogenases 1A2 (ALDH1A2) was thought to be one of potential candidates. Therefore, in this study we observed and analyzed the alteration of the expression and distribution of ALDH1A2 in the spinal cord of wild-type (WT) and Tg(SOD1*G93A)1Gur mice. We compared the expression and distribution of ALDH1A2 in the different segments, anatomic regions and neural cells of spinal cord at the different stages of WT and Tg(SOD1*G93A)1Gur mice applied the methods of fluorescent immunohistochemistry and western blot. Results revealed that ALDH1A2 extensively expressed and distributed in the spinal cord of adult WT and Tg(SOD1*G93A)1Gur mice. The expression and distribution of ALDH1A2 in the white matter including the anterior, posterior and lateral funiculus were more than that in the gray matter including the central canal, the anterior and dorsal horn. ALDH1A2 majorly expressed and distributed in the astrocyte, microglial, oligodendrocyte and neuron cells. The ALDH1A2 expression significantly decreased and redistributed in some anatomic regions of spinal cord at the onset and progression stages of Tg(SOD1*G93A)1Gur mice. The expression decrease of ALDH1A2 followed with the increase of neuron cells death. This study suggested that the alteration of expression and distribution of ALDH1A2 was potentially associated with the pathogenesis of ALS.
Collapse
Affiliation(s)
- Huiting Liang
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Chengsi Wu
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Youqing Deng
- Department of Neurology, Third Affiliated Hospital of Nanchang University, Nanchang 330008, Jiangxi, China
| | - Lei Zhu
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Jie Zhang
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Weiming Gan
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Chunyan Tang
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Renshi Xu
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| |
Collapse
|
30
|
Onay A, Onay M, Abul O. Classification of nervous system withdrawn and approved drugs with ToxPrint features via machine learning strategies. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2017; 142:9-19. [PMID: 28325450 DOI: 10.1016/j.cmpb.2017.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 01/20/2017] [Accepted: 02/08/2017] [Indexed: 06/06/2023]
Abstract
BACKGROUND AND OBJECTIVES Early-phase virtual screening of candidate drug molecules plays a key role in pharmaceutical industry from data mining and machine learning to prevent adverse effects of the drugs. Computational classification methods can distinguish approved drugs from withdrawn ones. We focused on 6 data sets including maximum 110 approved and 110 withdrawn drugs for all and nervous system diseases to distinguish approved drugs from withdrawn ones. METHODS In this study, we used support vector machines (SVMs) and ensemble methods (EMs) such as boosted and bagged trees to classify drugs into approved and withdrawn categories. Also, we used CORINA Symphony program to identify Toxprint chemotypes including over 700 predefined chemotypes for determination of risk and safety assesment of candidate drug molecules. In addition, we studied nervous system withdrawn drugs to determine the key fragments with The ParMol package including gSpan algorithm. RESULTS According to our results, the descriptors named as the number of total chemotypes and bond CN_amine_aliphatic_generic were more significant descriptors. The developed Medium Gaussian SVM model reached 78% prediction accuracy on test set for drug data set including all disease. Here, bagged tree and linear SVM models showed 89% of accuracies for phycholeptics and psychoanaleptics drugs. A set of discriminative fragments in nervous system withdrawn drug (NSWD) data sets was obtained. These fragments responsible for the drugs removed from market were benzene, toluene, N,N-dimethylethylamine, crotylamine, 5-methyl-2,4-heptadiene, octatriene and carbonyl group. CONCLUSION This paper covers the development of computational classification methods to distinguish approved drugs from withdrawn ones. In addition, the results of this study indicated the identification of discriminative fragments is of significance to design a new nervous system approved drugs with interpretation of the structures of the NSWDs.
Collapse
Affiliation(s)
- Aytun Onay
- Department of Computer Engineering, TOBB University of Economics & Technology, 06560, Ankara, Turkey
| | - Melih Onay
- Department of Environmental Engineering, Computational & Experimental Biochemistry Lab, Yuzuncu Yil University, 65080, Van, Turkey.
| | - Osman Abul
- Department of Computer Engineering, TOBB University of Economics & Technology, 06560, Ankara, Turkey
| |
Collapse
|
31
|
Schwarcz R, Stone TW. The kynurenine pathway and the brain: Challenges, controversies and promises. Neuropharmacology 2017; 112:237-247. [PMID: 27511838 PMCID: PMC5803785 DOI: 10.1016/j.neuropharm.2016.08.003] [Citation(s) in RCA: 270] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/29/2016] [Accepted: 08/05/2016] [Indexed: 12/29/2022]
Abstract
Research on the neurobiology of the kynurenine pathway has suffered years of relative obscurity because tryptophan degradation, and its involvement in both physiology and major brain diseases, was viewed almost exclusively through the lens of the well-established metabolite serotonin. With increasing recognition that kynurenine and its metabolites can affect and even control a variety of classic neurotransmitter systems directly and indirectly, interest is expanding rapidly. Moreover, kynurenine pathway metabolism itself is modulated in conditions such as infection and stress, which are known to induce major changes in well-being and behaviour, so that kynurenines may be instrumental in the etiology of psychiatric and neurological disorders. It is therefore likely that the near future will not only witness the discovery of additional physiological and pathological roles for brain kynurenines, but also ever-increasing interest in drug development based on these roles. In particular, targeting the kynurenine pathway with new specific agents may make it possible to prevent disease by appropriate pharmacological or genetic manipulations. The following overview focuses on areas of kynurenine research which are either controversial, of major potential therapeutic interest, or just beginning to receive the degree of attention which will clarify their relevance to neurobiology and medicine. It also highlights technical issues so that investigators entering the field, and new research initiatives, are not misdirected by inappropriate experimental approaches or incorrect interpretations at this time of skyrocketing interest in the subject matter. This article is part of the Special Issue entitled 'The Kynurenine Pathway in Health and Disease'.
Collapse
Affiliation(s)
- Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Trevor W Stone
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
32
|
The kynurenine pathway: Towards metabolic equilibrium. Neuropharmacology 2017; 112:235-236. [DOI: 10.1016/j.neuropharm.2016.08.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 08/19/2016] [Indexed: 12/15/2022]
|