Kynurenines in the Pathogenesis of Multiple Sclerosis: Therapeutic Perspectives
Abstract
:1. Introduction
2. The Production and Metabolism of Kynurenines
2.1. Kynurenine Pathway
2.2. Methoxyndole Pathway
3. Neuroactive Metabolites of Tryptophan
3.1. Kynurenic Acid
3.2. N-Acetylserotonin
3.3. Quinolinic Acid
4. The Kynurenine System and Immunoregulation
4.1. Effects on the Immune Cells
4.2. The Effect of Cytokines on Kynurenines
4.3. Gut-Microbiome and Kynurenines
4.4. Disturbances of the Kynurenine System in Neuro-Immunological Conditions
4.5. The Role of the Kynurenine System in Multiple Sclerosis
4.5.1. Kynurenines and Animal Models of MS
4.5.2. Kynurenine Metabolite Changes in MS
4.5.3. The Kynurenine System and Depression in MS
4.5.4. Treatment Effect on the Kynurenine System
4.5.5. Kynurenic Pathway and Redox Disturbances in Neuroinflammation and Multiple Sclerosis
4.5.6. Treating MS in Light of Kynurenines
Laquinimod
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
3-HAO | 3-hydroxyanthranilate oxidase |
3-HK | 3-hydroxykynurenine |
AHR | aryl hydrocarbon receptor |
AMPA | α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid |
ANA | anthralinic acid |
APC | antigen presenting cell |
ATP | adenosine triphosphate |
BBB | blood-brain barrier |
CDxx | cluster of differentiation xx |
CHMP | Committee for Medicinal Products for Human Use |
CNS | central nervous system |
DC | dendritic cell |
EAE | experimental autoimmune encephalitis |
GRP35 | G-protein coupled receptor 35 |
IDO 1,2 | izoenzymes indoleamine 2,3-dioxygenase 1 and 2 |
IFN-β | interferon beta |
IFN-γ | interferon gamma |
IL | interleukin |
KAT 1, 2 | kynurenine aminotransferase 1 and 2 |
KMO | kynure nine 3-monooxygenase |
KP | kynurenine pathway |
KYN | kynurenine |
KYNA | kynurenic acid |
MMP9 | matrix metalloproteinase 9 (MMP9) |
MP | methoxyndole pathway |
MS | multiple sclerosis |
NAD+ | nicotinamide adenine dinucleotide |
NADP+ | nicotinamide adenine dinucleotide phosphate |
NaMN | nicotinic acid mononucleotide |
NAS | N-acetylserotonin |
NK | natural killer cell |
NMAPT | nicotinamide phosphoribosyltransferase |
NMDA | N-methyl-D-aspartate |
NMDA | N-methyl-D-aspartate receptor |
NMN | nicotinamide mononucleotide |
NMNAT | nicotinamide mononucleotide adenyl transferase |
PARP | poly(ADP-ribose) polymerase |
PBMC | peripherial blood monocytes |
PMN | polymorphonuclear cells |
PPMS | primary progressive multiple sclerosis |
QUIN | quinolinic acid |
RRMS | relapsing-remitting multiple sclerosis |
SS | Sjögren’s syndrome |
TDO | tryptophan 2,3-dioxygenase |
TGF-β | transforming growth factor beta |
TNFα | tumor necrosis factor alpha |
Treg | regulatory T cell |
TRP | tryptophan |
VLA-4 | very late antigen-4 |
References
- Schwarcz, R.; Bruno, J.P.; Muchowski, P.J.; Wu, H.Q. Kynurenines in the mammalian brain: When physiology meets pathology. Nat. Revi. Neurosci. 2012, 13, 465–477. [Google Scholar] [CrossRef] [PubMed]
- Leklem, J.E. Quantitative aspects of tryptophan metabolism in humans and other species: A review. Am. J. Clin. Nutr. 1971, 24, 659–672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jones, S.P.; Guillemin, G.J.; Brew, B.J. The kynurenine pathway in stem cell biology. Int. J. Tryptophan Res. 2013, 6, 57–66. [Google Scholar] [CrossRef]
- Moroni, F. Tryptophan metabolism and brain function: Focus on kynurenine and other indole metabolites. Eur. J. Pharmacol. 1999, 375, 87–100. [Google Scholar] [CrossRef]
- Guillemin, G.J. Quinolinic acid, the inescapable neurotoxin. FEBS J. 2012, 279, 1356–1365. [Google Scholar] [CrossRef]
- Guillemin, G.J.; Cullen, K.M.; Lim, C.K.; Smythe, G.A.; Garner, B.; Kapoor, V.; Takikawa, O.; Brew, B.J. Characterization of the kynurenine pathway in human neurons. J. Neurosci. 2007, 27, 12884–12892. [Google Scholar] [CrossRef] [Green Version]
- Hartai, Z.; Klivenyi, P.; Janaky, T.; Penke, B.; Dux, L.; Vecsei, L. Kynurenine metabolism in multiple sclerosis. Acta Neurol. Scand. 2005, 112, 93–96. [Google Scholar] [CrossRef]
- Vecsei, L.; Szalardy, L.; Fulop, F.; Toldi, J. Kynurenines in the CNS: Recent advances and new questions. Nat. Rev. Drug Discov. 2013, 12, 64–82. [Google Scholar] [CrossRef]
- Boros, F.A.; Bohar, Z.; Vecsei, L. Genetic alterations affecting the genes encoding the enzymes of the kynurenine pathway and their association with human diseases. Mutat. Res. 2018, 776, 32–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lovelace, M.D.; Varney, B.; Sundaram, G.; Lennon, M.J.; Lim, C.K.; Jacobs, K.; Guillemin, G.J.; Brew, B.J. Recent evidence for an expanded role of the kynurenine pathway of tryptophan metabolism in neurological diseases. Neuropharmacology 2017, 112, 373–388. [Google Scholar] [CrossRef]
- Braidy, N.; Grant, R.; Adams, S.; Guillemin, G.J. Neuroprotective effects of naturally occurring polyphenols on quinolinic acid-induced excitotoxicity in human neurons. FEBS J. 2010, 277, 368–382. [Google Scholar] [CrossRef] [PubMed]
- Oxenkrug, G.F. Genetic and hormonal regulation of tryptophan kynurenine metabolism: Implications for vascular cognitive impairment, major depressive disorder, and aging. Ann. N. Y. Acad. Sci. 2007, 1122, 35–49. [Google Scholar] [CrossRef]
- Oxenkrug, G.F. Interferon-gamma-inducible kynurenines/pteridines inflammation cascade: Implications for aging and aging-associated psychiatric and medical disorders. J. Neural Transm. 2011, 118, 75–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pardridge, W.M. Blood-brain barrier carrier-mediated transport and brain metabolism of amino acids. Neurochem. Res. 1998, 23, 635–644. [Google Scholar] [CrossRef] [PubMed]
- Mellor, A.L.; Munn, D.H. IDO expression by dendritic cells: Tolerance and tryptophan catabolism. Nat. Rev. Immunol. 2004, 4, 762–774. [Google Scholar] [CrossRef]
- Grohmann, U.; Fallarino, F.; Puccetti, P. Tolerance, DCs and tryptophan: Much ado about IDO. Trends Immunol. 2003, 24, 242–248. [Google Scholar] [CrossRef]
- Platten, M.; Wick, W.; Van den Eynde, B.J. Tryptophan catabolism in cancer: Beyond IDO and tryptophan depletion. Cancer Res. 2012, 72, 5435–5440. [Google Scholar] [CrossRef] [Green Version]
- Van Baren, N.; Van den Eynde, B.J. Tryptophan-degrading enzymes in tumoral immune resistance. Front. Immunol. 2015, 6, 34. [Google Scholar] [CrossRef] [Green Version]
- Katz, J.B.; Muller, A.J.; Prendergast, G.C. Indoleamine 2,3-dioxygenase in T-cell tolerance and tumoral immune escape. Immunol. Rev. 2008, 222, 206–221. [Google Scholar] [CrossRef] [PubMed]
- Mondanelli, G.; Albini, E.; Pallotta, M.T.; Volpi, C.; Chatenoud, L.; Kuhn, C.; Fallarino, F.; Matino, D.; Belladonna, M.L.; Bianchi, R.; et al. The Proteasome Inhibitor Bortezomib Controls Indoleamine 2,3–Dioxygenase 1 Breakdown and Restores Immune Regulation in Autoimmune Diabetes. Front. Immunol. 2017, 8, 428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Llamas-Velasco, M.; Bonay, P.; Jose Concha-Garzon, M.; Corvo-Villen, L.; Vara, A.; Cibrian, D.; Sanguino-Pascual, A.; Sanchez-Madrid, F.; De la Fuente, H.; Dauden, E. Immune cells from patients with psoriasis are defective in inducing indoleamine 2,3-dioxygenase expression in response to inflammatory stimuli. Br. J. Dermatol. 2017, 176, 695–704. [Google Scholar] [CrossRef] [PubMed]
- Cribbs, A.P.; Kennedy, A.; Penn, H.; Read, J.E.; Amjadi, P.; Green, P.; Syed, K.; Manka, S.W.; Brennan, F.M.; Gregory, B.; et al. Treg cell function in rheumatoid arthritis is compromised by ctla-4 promoter methylation resulting in a failure to activate the indoleamine 2,3-dioxygenase pathway. Arthritis Rheumatol. 2014, 66, 2344–2354. [Google Scholar] [CrossRef] [PubMed]
- Negrotto, L.; Correale, J. Amino Acid Catabolism in Multiple Sclerosis Affects Immune Homeostasis. J. Immunol. 2017, 198, 1900–1909. [Google Scholar] [CrossRef] [Green Version]
- Volpi, C.; Mondanelli, G.; Pallotta, M.T.; Vacca, C.; Iacono, A.; Gargaro, M.; Albini, E.; Bianchi, R.; Belladonna, M.L.; Celanire, S.; et al. Allosteric modulation of metabotropic glutamate receptor 4 activates IDO1-dependent, immunoregulatory signaling in dendritic cells. Neuropharmacology 2016, 102, 59–71. [Google Scholar] [CrossRef] [Green Version]
- Miller, C.L.; Llenos, I.C.; Dulay, J.R.; Barillo, M.M.; Yolken, R.H.; Weis, S. Expression of the kynurenine pathway enzyme tryptophan 2,3-dioxygenase is increased in the frontal cortex of individuals with schizophrenia. Neurobiol. Dis. 2004, 15, 618–629. [Google Scholar] [CrossRef]
- Platten, M.; Von Knebel Doeberitz, N.; Oezen, I.; Wick, W.; Ochs, K. Cancer Immunotherapy by Targeting IDO1/TDO and Their Downstream Effectors. Front. Immunol. 2014, 5, 673. [Google Scholar] [CrossRef] [PubMed]
- Grohmann, U.; Bronte, V. Control of immune response by amino acid metabolism. Immunol. Rev. 2010, 236, 243–264. [Google Scholar] [CrossRef] [PubMed]
- Mondanelli, G.; Ugel, S.; Grohmann, U.; Bronte, V. The immune regulation in cancer by the amino acid metabolizing enzymes ARG and IDO. Curr. Opin. Pharmacol. 2017, 35, 30–39. [Google Scholar] [CrossRef] [PubMed]
- Mondanelli, G.; Iacono, A.; Allegrucci, M.; Puccetti, P.; Grohmann, U. Immunoregulatory Interplay between Arginine and Tryptophan Metabolism in Health and Disease. Front. Immunol. 2019, 10, 1565. [Google Scholar] [CrossRef] [PubMed]
- Dang, Y.; Dale, W.E.; Brown, O.R. Comparative effects of oxygen on indoleamine 2,3-dioxygenase and tryptophan 2,3-dioxygenase of the kynurenine pathway. Free Radic. Biol. Med. 2000, 28, 615–624. [Google Scholar] [CrossRef]
- Gal, E.M.; Sherman, A.D. Synthesis and metabolism of L-kynurenine in rat brain. J. Neurochem. 1978, 30, 607–613. [Google Scholar] [CrossRef] [PubMed]
- Speciale, C.; Schwarcz, R. Uptake of kynurenine into rat brain slices. J. Neurochem. 1990, 54, 156–163. [Google Scholar] [CrossRef]
- Fukui, S.; Schwarcz, R.; Rapoport, S.I.; Takada, Y.; Smith, Q.R. Blood-brain barrier transport of kynurenines: Implications for brain synthesis and metabolism. J. Neurochem. 1991, 56, 2007–2017. [Google Scholar] [CrossRef] [PubMed]
- Han, Q.; Li, J.; Li, J. pH dependence, substrate specificity and inhibition of human kynurenine aminotransferase I. Eur. J. Bochem. 2004, 271, 4804–4814. [Google Scholar] [CrossRef] [PubMed]
- Guillemin, G.J.; Smythe, G.; Takikawa, O.; Brew, B.J. Expression of indoleamine 2,3-dioxygenase and production of quinolinic acid by human microglia, astrocytes, and neurons. Glia 2005, 49, 15–23. [Google Scholar] [CrossRef] [PubMed]
- Parrott, J.M.; O’Connor, J.C. Kynurenine 3-Monooxygenase: An Influential Mediator of Neuropathology. Front. Psychiatry 2015, 6, 116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Y.; Brew, B.J.; Guillemin, G.J. Characterization of the kynurenine pathway in NSC-34 cell line: Implications for amyotrophic lateral sclerosis. J. Neurochem. 2011, 118, 816–825. [Google Scholar] [CrossRef] [PubMed]
- Chiarugi, A.; Calvani, M.; Meli, E.; Traggiai, E.; Moroni, F. Synthesis and release of neurotoxic kynurenine metabolites by human monocyte-derived macrophages. J. Neuroimmunol. 2001, 120, 190–198. [Google Scholar] [CrossRef]
- Heyes, M.P.; Chen, C.Y.; Major, E.O.; Saito, K. Different kynurenine pathway enzymes limit quinolinic acid formation by various human cell types. Biochem. J. 1997, 326, 351–356. [Google Scholar] [CrossRef]
- Kiss, C.; Ceresoli-Borroni, G.; Guidetti, P.; Zielke, C.L.; Zielke, H.R.; Schwarcz, R. Kynurenate production by cultured human astrocytes. J. Neural Transm. 2003, 110, 1–14. [Google Scholar] [CrossRef]
- Kocki, T.; Dolinska, M.; Dybel, A.; Urbanska, E.M.; Turski, W.A.; Albrecht, J. Regulation of kynurenic acid synthesis in C6 glioma cells. J. Neurosci. Res. 2002, 68, 622–626. [Google Scholar] [CrossRef] [PubMed]
- Guidetti, P.; Hoffman, G.E.; Melendez-Ferro, M.; Albuquerque, E.X.; Schwarcz, R. Astrocytic localization of kynurenine aminotransferase II in the rat brain visualized by immunocytochemistry. Glia 2007, 55, 78–92. [Google Scholar] [CrossRef] [PubMed]
- Lehrmann, E.; Molinari, A.; Speciale, C.; Schwarcz, R. Immunohistochemical visualization of newly formed quinolinate in the normal and excitotoxically lesioned rat striatum. Exp. Brain Res. 2001, 141, 389–397. [Google Scholar] [CrossRef]
- Zinger, A.; Barcia, C.; Herrero, M.T.; Guillemin, G.J. The involvement of neuroinflammation and kynurenine pathway in Parkinson’s disease. Parkinson’s Dis. 2011, 2011, 716859. [Google Scholar] [CrossRef] [Green Version]
- Moroni, F.; Russi, P.; Lombardi, G.; Beni, M.; Carla, V. Presence of kynurenic acid in the mammalian brain. J. Neurochem. 1988, 51, 177–180. [Google Scholar] [CrossRef] [PubMed]
- Turski, W.A.; Nakamura, M.; Todd, W.P.; Carpenter, B.K.; Whetsell, W.O., Jr.; Schwarcz, R. Identification and quantification of kynurenic acid in human brain tissue. Brain Res. 1988, 454, 164–169. [Google Scholar] [CrossRef]
- Wu, H.Q.; Guidetti, P.; Goodman, J.H.; Varasi, M.; Ceresoli-Borroni, G.; Speciale, C.; Scharfman, H.E.; Schwarcz, R. Kynurenergic manipulations influence excitatory synaptic function and excitotoxic vulnerability in the rat hippocampus in vivo. Neuroscience 2000, 97, 243–251. [Google Scholar] [CrossRef]
- Wang, J.; Simonavicius, N.; Wu, X.; Swaminath, G.; Reagan, J.; Tian, H.; Ling, L. Kynurenic acid as a ligand for orphan G protein-coupled receptor GPR35. J. Biol. Chem. 2006, 281, 22021–22028. [Google Scholar] [CrossRef] [Green Version]
- Southern, C.; Cook, J.M.; Neetoo-Isseljee, Z.; Taylor, D.L.; Kettleborough, C.A.; Merritt, A.; Bassoni, D.L.; Raab, W.J.; Quinn, E.; Wehrman, T.S.; et al. Screening beta-arrestin recruitment for the identification of natural ligands for orphan G-protein-coupled receptors. J. Biomol. Screen. 2013, 18, 599–609. [Google Scholar] [CrossRef] [Green Version]
- Zhao, P.; Sharir, H.; Kapur, A.; Cowan, A.; Geller, E.B.; Adler, M.W.; Seltzman, H.H.; Reggio, P.H.; Heynen-Genel, S.; Sauer, M.; et al. Targeting of the orphan receptor GPR35 by pamoic acid: A potent activator of extracellular signal-regulated kinase and beta-arrestin2 with antinociceptive activity. Mol. Pharmacol. 2010, 78, 560–568. [Google Scholar] [CrossRef]
- Deng, H.; Hu, H.; Fang, Y. Multiple tyrosine metabolites are GPR35 agonists. Sci. Rep. 2012, 2, 373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, J.; Williams, D.J.; Puhl, H.L., 3rd; Ikeda, S.R. Inhibition of N-type calcium channels by activation of GPR35, an orphan receptor, heterologously expressed in rat sympathetic neurons. J. Pharmacol. Exp. Ther. 2008, 324, 342–351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kimura, E.; Kubo, K.I.; Endo, T.; Nakajima, K.; Kakeyama, M.; Tohyama, C. Excessive activation of AhR signaling disrupts neuronal migration in the hippocampal CA1 region in the developing mouse. J. Toxicol. Sci. 2017, 42, 25–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- DiNatale, B.C.; Murray, I.A.; Schroeder, J.C.; Flaveny, C.A.; Lahoti, T.S.; Laurenzana, E.M.; Omiecinski, C.J.; Perdew, G.H. Kynurenic acid is a potent endogenous aryl hydrocarbon receptor ligand that synergistically induces interleukin-6 in the presence of inflammatory signaling. Toxicol. Sci. 2010, 115, 89–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Opitz, C.A.; Litzenburger, U.M.; Sahm, F.; Ott, M.; Tritschler, I.; Trump, S.; Schumacher, T.; Jestaedt, L.; Schrenk, D.; Weller, M.; et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 2011, 478, 197–203. [Google Scholar] [CrossRef]
- Henderson, G.; Johnson, J.W.; Ascher, P. Competitive antagonists and partial agonists at the glycine modulatory site of the mouse N-methyl-D-aspartate receptor. J. Physiol. 1990, 430, 189–212. [Google Scholar] [CrossRef] [Green Version]
- Kessler, M.; Baudry, M.; Lynch, G. Quinoxaline derivatives are high-affinity antagonists of the NMDA receptor-associated glycine sites. Brain Res. 1989, 489, 377–382. [Google Scholar] [CrossRef]
- Oliver, M.W.; Kessler, M.; Larson, J.; Schottler, F.; Lynch, G. Glycine site associated with the NMDA receptor modulates long-term potentiation. Synapse 1990, 5, 265–270. [Google Scholar] [CrossRef]
- Kloog, Y.; Lamdani-Itkin, H.; Sokolovsky, M. The glycine site of the N-methyl-D-aspartate receptor channel: Differences between the binding of HA-966 and of 7-chlorokynurenic acid. J. Neurochem. 1990, 54, 1576–1583. [Google Scholar] [CrossRef] [PubMed]
- Mayer, M.L.; Westbrook, G.L.; Vyklicky, L., Jr. Sites of antagonist action on N-methyl-D-aspartic acid receptors studied using fluctuation analysis and a rapid perfusion technique. J. Neurophysiol. 1988, 60, 645–663. [Google Scholar] [CrossRef]
- Olverman, H.J.; Jones, A.W.; Mewett, K.N.; Watkins, J.C. Structure/activity relations of N-methyl-D-aspartate receptor ligands as studied by their inhibition of [3H]D-2-amino-5-phosphonopentanoic acid binding in rat brain membranes. Neuroscience 1988, 26, 17–31. [Google Scholar] [CrossRef]
- Moroni, F.; Pellegrini-Giampietro, D.E.; Alesiani, M.; Cherici, G.; Mori, F.; Galli, A. Glycine and kynurenate modulate the glutamate receptors in the myenteric plexus and in cortical membranes. Eur. J. Pharmacol. 1989, 163, 123–126. [Google Scholar] [CrossRef]
- Robinson, M.B.; Schulte, M.K.; Freund, R.K.; Johnson, R.L.; Koerner, J.F. Structure-function relationships for kynurenic acid analogues at excitatory pathways in the rat hippocampal slice. Brain Res. 1985, 361, 19–24. [Google Scholar] [CrossRef]
- Danysz, W.; Fadda, E.; Wroblewski, J.T.; Costa, E. Different modes of action of 3-amino-1-hydroxy-2-pyrrolidone (HA-966) and 7-chlorokynurenic acid in the modulation of N-methyl-D-aspartate-sensitive glutamate receptors. Mol. Pharmacol. 1989, 36, 912–916. [Google Scholar]
- Danysz, W.; Fadda, E.; Wroblewski, J.T.; Costa, E. Kynurenate and 2-amino-5-phosphonovalerate interact with multiple binding sites of the N-methyl-D-aspartate-sensitive glutamate receptor domain. Neurosci. Lett. 1989, 96, 340–344. [Google Scholar] [CrossRef]
- Fisher, J.L.; Mott, D.D. Distinct functional roles of subunits within the heteromeric kainate receptor. J. Neurosci. 2011, 31, 17113–17122. [Google Scholar] [CrossRef]
- Lugo-Huitron, R.; Blanco-Ayala, T.; Ugalde-Muniz, P.; Carrillo-Mora, P.; Pedraza-Chaverri, J.; Silva-Adaya, D.; Maldonado, P.D.; Torres, I.; Pinzon, E.; Ortiz-Islas, E.; et al. On the antioxidant properties of kynurenic acid: Free radical scavenging activity and inhibition of oxidative stress. Neurotoxicol. Teratol. 2011, 33, 538–547. [Google Scholar] [CrossRef]
- Kubicova, L.; Hadacek, F.; Bachmann, G.; Weckwerth, W.; Chobot, V. Coordination Complex Formation and Redox Properties of Kynurenic and Xanthurenic Acid Can Affect Brain Tissue Homeodynamics. Antioxidants 2019, 8, 476. [Google Scholar] [CrossRef] [Green Version]
- Perkins, M.N.; Stone, T.W. An iontophoretic investigation of the actions of convulsant kynurenines and their interaction with the endogenous excitant quinolinic acid. Brain Res. 1982, 247, 184–187. [Google Scholar] [CrossRef]
- Kessler, M.; Terramani, T.; Lynch, G.; Baudry, M. A glycine site associated with N-methyl-D-aspartic acid receptors: Characterization and identification of a new class of antagonists. J. Neurochem. 1989, 52, 1319–1328. [Google Scholar] [CrossRef]
- Parsons, C.G.; Danysz, W.; Quack, G.; Hartmann, S.; Lorenz, B.; Wollenburg, C.; Baran, L.; Przegalinski, E.; Kostowski, W.; Krzascik, P.; et al. Novel systemically active antagonists of the glycine site of the N-methyl-D-aspartate receptor: Electrophysiological, biochemical and behavioral characterization. J. Pharmacol. Exp. Ther. 1997, 283, 1264–1275. [Google Scholar]
- Szalardy, L.; Zadori, D.; Toldi, J.; Fulop, F.; Klivenyi, P.; Vecsei, L. Manipulating kynurenic acid levels in the brain - on the edge between neuroprotection and cognitive dysfunction. Curr. Top. Med. Chem. 2012, 12, 1797–1806. [Google Scholar] [CrossRef] [PubMed]
- Prescott, C.; Weeks, A.M.; Staley, K.J.; Partin, K.M. Kynurenic acid has a dual action on AMPA receptor responses. Neurosci. Lett. 2006, 402, 108–112. [Google Scholar] [CrossRef] [PubMed]
- Rozsa, E.; Robotka, H.; Vecsei, L.; Toldi, J. The Janus-face kynurenic acid. J. Neural Transm. 2008, 115, 1087–1091. [Google Scholar] [CrossRef]
- Zwilling, D.; Huang, S.Y.; Sathyasaikumar, K.V.; Notarangelo, F.M.; Guidetti, P.; Wu, H.Q.; Lee, J.; Truong, J.; Andrews-Zwilling, Y.; Hsieh, E.W.; et al. Kynurenine 3-monooxygenase inhibition in blood ameliorates neurodegeneration. Cell 2011, 145, 863–874. [Google Scholar] [CrossRef] [Green Version]
- Hilmas, C.; Pereira, E.F.; Alkondon, M.; Rassoulpour, A.; Schwarcz, R.; Albuquerque, E.X. The brain metabolite kynurenic acid inhibits alpha7 nicotinic receptor activity and increases non-alpha7 nicotinic receptor expression: Physiopathological implications. J. Neurosci. 2001, 21, 7463–7473. [Google Scholar] [CrossRef]
- Tanaka, M.; Bohar, Z.; Vecsei, L. Are Kynurenines Accomplices or Principal Villains in Dementia? Maintenance of Kynurenine Metabolism. Molecules 2020, 25, 564. [Google Scholar] [CrossRef] [Green Version]
- Stone, T.W. Kynurenic acid antagonists and kynurenine pathway inhibitors. Expert Opin. Investig. Drugs 2001, 10, 633–645. [Google Scholar] [CrossRef]
- Stone, T.W.; Stoy, N.; Darlington, L.G. An expanding range of targets for kynurenine metabolites of tryptophan. Trends Pharmacol. Sci. 2013, 34, 136–143. [Google Scholar] [CrossRef] [Green Version]
- Mackenzie, A.E.; Lappin, J.E.; Taylor, D.L.; Nicklin, S.A.; Milligan, G. GPR35 as a Novel Therapeutic Target. Front. Endocrinol. 2011, 2, 68. [Google Scholar] [CrossRef] [Green Version]
- Taniguchi, Y.; Tonai-Kachi, H.; Shinjo, K. Zaprinast, a well-known cyclic guanosine monophosphate-specific phosphodiesterase inhibitor, is an agonist for GPR35. FEBS Lett. 2006, 580, 5003–5008. [Google Scholar] [CrossRef] [Green Version]
- Maravillas-Montero, J.L.; Burkhardt, A.M.; Hevezi, P.A.; Carnevale, C.D.; Smit, M.J.; Zlotnik, A. Cutting edge: GPR35/CXCR8 is the receptor of the mucosal chemokine CXCL17. J. Immunol. 2015, 194, 29–33. [Google Scholar] [CrossRef]
- Fallarini, S.; Magliulo, L.; Paoletti, T.; De Lalla, C.; Lombardi, G. Expression of functional GPR35 in human iNKT cells. Biochem. Biophys. Res. Commun. 2010, 398, 420–425. [Google Scholar] [CrossRef]
- Mandi, Y.; Vecsei, L. The kynurenine system and immunoregulation. J. Neural Transm. 2012, 119, 197–209. [Google Scholar] [CrossRef]
- Moroni, F.; Cozzi, A.; Sili, M.; Mannaioni, G. Kynurenic acid: A metabolite with multiple actions and multiple targets in brain and periphery. J. Neural Transm. 2012, 119, 133–139. [Google Scholar] [CrossRef]
- Alkondon, M.; Pereira, E.F.; Todd, S.W.; Randall, W.R.; Lane, M.V.; Albuquerque, E.X. Functional G-protein-coupled receptor 35 is expressed by neurons in the CA1 field of the hippocampus. Biochem. Pharmacol. 2015, 93, 506–518. [Google Scholar] [CrossRef]
- Denison, M.S.; Nagy, S.R. Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Ann. Rev. Pharmacol. Toxicol. 2003, 43, 309–334. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, N.T.; Kimura, A.; Nakahama, T.; Chinen, I.; Masuda, K.; Nohara, K.; Fujii-Kuriyama, Y.; Kishimoto, T. Aryl hydrocarbon receptor negatively regulates dendritic cell immunogenicity via a kynurenine-dependent mechanism. Proc. Natl. Acad. Sci. USA 2010, 107, 19961–19966. [Google Scholar] [CrossRef] [Green Version]
- Stevens, E.A.; Mezrich, J.D.; Bradfield, C.A. The aryl hydrocarbon receptor: A perspective on potential roles in the immune system. Immunology 2009, 127, 299–311. [Google Scholar] [CrossRef]
- Mezrich, J.D.; Fechner, J.H.; Zhang, X.; Johnson, B.P.; Burlingham, W.J.; Bradfield, C.A. An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J. Immunol. 2010, 185, 3190–3198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stephens, G.L.; Wang, Q.; Swerdlow, B.; Bhat, G.; Kolbeck, R.; Fung, M. Kynurenine 3-monooxygenase mediates inhibition of Th17 differentiation via catabolism of endogenous aryl hydrocarbon receptor ligands. Eur. J. Immunol. 2013, 43, 1727–1734. [Google Scholar] [CrossRef]
- Perez-Gonzalez, A.; Alvarez-Idaboy, J.R.; Galano, A. Free-radical scavenging by tryptophan and its metabolites through electron transfer based processes. J. Mol. Model. 2015, 21, 213. [Google Scholar] [CrossRef] [PubMed]
- Goda, K.; Hamane, Y.; Kishimoto, R.; Ogishi, Y. Radical scavenging properties of tryptophan metabolites. Estimation of their radical reactivity. Adv. Exp. Med. Biol. 1999, 467, 397–402. [Google Scholar] [CrossRef]
- Zhou, H.; Wang, J.; Jiang, J.; Stavrovskaya, I.G.; Li, M.; Li, W.; Wu, Q.; Zhang, X.; Luo, C.; Zhou, S.; et al. N-acetyl-serotonin offers neuroprotection through inhibiting mitochondrial death pathways and autophagic activation in experimental models of ischemic injury. J. Neurosci. 2014, 34, 2967–2978. [Google Scholar] [CrossRef]
- Jang, S.W.; Liu, X.; Pradoldej, S.; Tosini, G.; Chang, Q.; Iuvone, P.M.; Ye, K. N-acetylserotonin activates TrkB receptor in a circadian rhythm. Proc. Natl. Acad. Sci. USA 2010, 107, 3876–3881. [Google Scholar] [CrossRef] [Green Version]
- Yoo, J.M.; Lee, B.D.; Sok, D.E.; Ma, J.Y.; Kim, M.R. Neuroprotective action of N-acetyl serotonin in oxidative stress-induced apoptosis through the activation of both TrkB/CREB/BDNF pathway and Akt/Nrf2/Antioxidant enzyme in neuronal cells. Redox Biol. 2017, 11, 592–599. [Google Scholar] [CrossRef] [PubMed]
- Mondanelli, G.; Coletti, A.; Greco, F.A.; Pallotta, M.T.; Orabona, C.; Iacono, A.; Belladonna, M.L.; Albini, E.; Panfili, E.; Fallarino, F.; et al. Positive allosteric modulation of indoleamine 2,3-dioxygenase 1 restrains neuroinflammation. Proc. Natl. Acad. Sci. USA 2020, 117, 3848–3857. [Google Scholar] [CrossRef]
- Gomez-Corvera, A.; Cerrillo, I.; Molinero, P.; Naranjo, M.C.; Lardone, P.J.; Sanchez-Hidalgo, M.; Carrascosa-Salmoral, M.P.; Medrano-Campillo, P.; Guerrero, J.M.; Rubio, A. Evidence of immune system melatonin production by two pineal melatonin deficient mice, C57BL/6 and Swiss strains. J. Pineal Res. 2009, 47, 15–22. [Google Scholar] [CrossRef]
- Jaronen, M.; Quintana, F.J. Immunological Relevance of the Coevolution of IDO1 and AHR. Front. Immunol. 2014, 5, 521. [Google Scholar] [CrossRef] [Green Version]
- Lewis-Ballester, A.; Pham, K.N.; Batabyal, D.; Karkashon, S.; Bonanno, J.B.; Poulos, T.L.; Yeh, S.R. Structural insights into substrate and inhibitor binding sites in human indoleamine 2,3-dioxygenase 1. Nat. Commun. 2017, 8, 1693. [Google Scholar] [CrossRef] [PubMed]
- Mondanelli, G.; Iacono, A.; Carvalho, A.; Orabona, C.; Volpi, C.; Pallotta, M.T.; Matino, D.; Esposito, S.; Grohmann, U. Amino acid metabolism as drug target in autoimmune diseases. Autoimmun. Rev. 2019, 18, 334–348. [Google Scholar] [CrossRef]
- Baumeister, S.H.; Freeman, G.J.; Dranoff, G.; Sharpe, A.H. Coinhibitory Pathways in Immunotherapy for Cancer. Annu. Rev. Immunol. 2016, 34, 539–573. [Google Scholar] [CrossRef]
- Orabona, C.; Mondanelli, G.; Puccetti, P.; Grohmann, U. Immune Checkpoint Molecules, Personalized Immunotherapy, and Autoimmune Diabetes. Trends Mol. Med. 2018, 24, 931–941. [Google Scholar] [CrossRef] [PubMed]
- Brundin, L.; Sellgren, C.M.; Lim, C.K.; Grit, J.; Palsson, E.; Landen, M.; Samuelsson, M.; Lundgren, K.; Brundin, P.; Fuchs, D.; et al. An enzyme in the kynurenine pathway that governs vulnerability to suicidal behavior by regulating excitotoxicity and neuroinflammation. Transl. Psychiatry 2016, 6, e865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stone, T.W.; Perkins, M.N. Quinolinic acid: A potent endogenous excitant at amino acid receptors in CNS. Eur. J. Pharmacol. 1981, 72, 411–412. [Google Scholar] [CrossRef]
- Perkins, M.N.; Stone, T.W. Pharmacology and regional variations of quinolinic acid-evoked excitations in the rat central nervous system. J. Pharmacol. Exp. Ther. 1983, 226, 551–557. [Google Scholar]
- Perkins, M.N.; Stone, T.W. Quinolinic acid: Regional variations in neuronal sensitivity. Brain Res. 1983, 259, 172–176. [Google Scholar] [CrossRef]
- De Carvalho, L.P.; Bochet, P.; Rossier, J. The endogenous agonist quinolinic acid and the non endogenous homoquinolinic acid discriminate between NMDAR2 receptor subunits. Neurochem. Int. 1996, 28, 445–452. [Google Scholar] [CrossRef]
- Monaghan, D.T.; Beaton, J.A. Quinolinate differentiates between forebrain and cerebellar NMDA receptors. Eur. J. Pharmacol. 1991, 194, 123–125. [Google Scholar] [CrossRef]
- Scherzer, C.R.; Landwehrmeyer, G.B.; Kerner, J.A.; Standaert, D.G.; Hollingsworth, Z.R.; Daggett, L.P.; Velicelebi, G.; Penney, J.B., Jr.; Young, A.B. Cellular distribution of NMDA glutamate receptor subunit mRNAs in the human cerebellum. Neurobiol. Dis. 1997, 4, 35–46. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.H.; Bosy, T.Z.; Yasuda, R.P.; Grayson, D.R.; Vicini, S.; Pizzorusso, T.; Wolfe, B.B. Characterization of NMDA receptor subunit-specific antibodies: Distribution of NR2A and NR2B receptor subunits in rat brain and ontogenic profile in the cerebellum. J. Neurochem. 1995, 65, 176–183. [Google Scholar] [CrossRef] [PubMed]
- Santamaria, A.; Rios, C. MK-801, an N-methyl-D-aspartate receptor antagonist, blocks quinolinic acid-induced lipid peroxidation in rat corpus striatum. Neurosci. Lett. 1993, 159, 51–54. [Google Scholar] [CrossRef]
- Platenik, J.; Stopka, P.; Vejrazka, M.; Stipek, S. Quinolinic acid-iron(ii) complexes: Slow autoxidation, but enhanced hydroxyl radical production in the Fenton reaction. Free Radic. Res. 2001, 34, 445–459. [Google Scholar] [CrossRef]
- Rios, C.; Santamaria, A. Quinolinic acid is a potent lipid peroxidant in rat brain homogenates. Neurochem. Res. 1991, 16, 1139–1143. [Google Scholar] [CrossRef]
- Stipek, S.; Stastny, F.; Platenik, J.; Crkovska, J.; Zima, T. The effect of quinolinate on rat brain lipid peroxidation is dependent on iron. Neurochem. Int. 1997, 30, 233–237. [Google Scholar] [CrossRef]
- St’astny, F.; Hinoi, E.; Ogita, K.; Yoneda, Y. Ferrous iron modulates quinolinate-mediated [3H]MK-801 binding to rat brain synaptic membranes in the presence of glycine and spermidine. Neurosci. Lett. 1999, 262, 105–108. [Google Scholar] [CrossRef]
- Pierozan, P.; Zamoner, A.; Soska, A.K.; Silvestrin, R.B.; Loureiro, S.O.; Heimfarth, L.; Mello e Souza, T.; Wajner, M.; Pessoa-Pureur, R. Acute intrastriatal administration of quinolinic acid provokes hyperphosphorylation of cytoskeletal intermediate filament proteins in astrocytes and neurons of rats. Exp. Neurol. 2010, 224, 188–196. [Google Scholar] [CrossRef]
- Rahman, A.; Ting, K.; Cullen, K.M.; Braidy, N.; Brew, B.J.; Guillemin, G.J. The excitotoxin quinolinic acid induces tau phosphorylation in human neurons. PLoS ONE 2009, 4, e6344. [Google Scholar] [CrossRef]
- Guillemin, G.J.; Kerr, S.J.; Smythe, G.A.; Smith, D.G.; Kapoor, V.; Armati, P.J.; Croitoru, J.; Brew, B.J. Kynurenine pathway metabolism in human astrocytes: A paradox for neuronal protection. J. Neurochem. 2001, 78, 842–853. [Google Scholar] [CrossRef]
- Stone, T.W.; Behan, W.M. Interleukin-1beta but not tumor necrosis factor-alpha potentiates neuronal damage by quinolinic acid: Protection by an adenosine A2A receptor antagonist. J. Neurosci. Res. 2007, 85, 1077–1085. [Google Scholar] [CrossRef]
- Musso, T.; Gusella, G.L.; Brooks, A.; Longo, D.L.; Varesio, L. Interleukin-4 inhibits indoleamine 2,3-dioxygenase expression in human monocytes. Blood 1994, 83, 1408–1411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fallarino, F.; Grohmann, U.; Vacca, C.; Bianchi, R.; Orabona, C.; Spreca, A.; Fioretti, M.C.; Puccetti, P. T cell apoptosis by tryptophan catabolism. Cell Death Differ. 2002, 9, 1069–1077. [Google Scholar] [CrossRef] [PubMed]
- Fallarino, F.; Grohmann, U.; You, S.; McGrath, B.C.; Cavener, D.R.; Vacca, C.; Orabona, C.; Bianchi, R.; Belladonna, M.L.; Volpi, C.; et al. The combined effects of tryptophan starvation and tryptophan catabolites down-regulate T cell receptor zeta-chain and induce a regulatory phenotype in naive T cells. J. Immunol. 2006, 176, 6752–6761. [Google Scholar] [CrossRef] [PubMed]
- Hill, M.; Tanguy-Royer, S.; Royer, P.; Chauveau, C.; Asghar, K.; Tesson, L.; Lavainne, F.; Remy, S.; Brion, R.; Hubert, F.X.; et al. IDO expands human CD4+CD25high regulatory T cells by promoting maturation of LPS-treated dendritic cells. Eur. J. Immunol. 2007, 37, 3054–3062. [Google Scholar] [CrossRef]
- Frumento, G.; Rotondo, R.; Tonetti, M.; Damonte, G.; Benatti, U.; Ferrara, G.B. Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2,3-dioxygenase. J. Exp. Med. 2002, 196, 459–468. [Google Scholar] [CrossRef] [Green Version]
- Song, H.; Park, H.; Kim, Y.S.; Kim, K.D.; Lee, H.K.; Cho, D.H.; Yang, J.W.; Hur, D.Y. L-kynurenine-induced apoptosis in human NK cells is mediated by reactive oxygen species. Int. Immunopharmacol. 2011, 11, 932–938. [Google Scholar] [CrossRef]
- Daubener, W.; MacKenzie, C.R. IFN-gamma activated indoleamine 2,3-dioxygenase activity in human cells is an antiparasitic and an antibacterial effector mechanism. Adv. Exp. Med. Biol. 1999, 467, 517–524. [Google Scholar] [CrossRef]
- Robinson, C.M.; Hale, P.T.; Carlin, J.M. The role of IFN-gamma and TNF-alpha-responsive regulatory elements in the synergistic induction of indoleamine dioxygenase. J. Interferon Cytokine Res. 2005, 25, 20–30. [Google Scholar] [CrossRef]
- O’Connor, J.C.; Andre, C.; Wang, Y.; Lawson, M.A.; Szegedi, S.S.; Lestage, J.; Castanon, N.; Kelley, K.W.; Dantzer, R. Interferon-gamma and tumor necrosis factor-alpha mediate the upregulation of indoleamine 2,3-dioxygenase and the induction of depressive-like behavior in mice in response to bacillus Calmette-Guerin. J. Neurosci. 2009, 29, 4200–4209. [Google Scholar] [CrossRef]
- Belladonna, M.L.; Orabona, C.; Grohmann, U.; Puccetti, P. TGF-beta and kynurenines as the key to infectious tolerance. Trends Mol. Med. 2009, 15, 41–49. [Google Scholar] [CrossRef]
- Forrest, C.M.; Mackay, G.M.; Stoy, N.; Spiden, S.L.; Taylor, R.; Stone, T.W.; Darlington, L.G. Blood levels of kynurenines, interleukin-23 and soluble human leucocyte antigen-G at different stages of Huntington’s disease. J. Neurochem. 2010, 112, 112–122. [Google Scholar] [CrossRef] [PubMed]
- Chaves, A.C.; Ceravolo, I.P.; Gomes, J.A.; Zani, C.L.; Romanha, A.J.; Gazzinelli, R.T. IL-4 and IL-13 regulate the induction of indoleamine 2,3-dioxygenase activity and the control of Toxoplasma gondii replication in human fibroblasts activated with IFN-gamma. Eur. J. Immunol. 2001, 31, 333–344. [Google Scholar] [CrossRef]
- Gomez, A.; Luckey, D.; Taneja, V. The gut microbiome in autoimmunity: Sex matters. Clin. Immunol. 2015, 159, 154–162. [Google Scholar] [CrossRef] [PubMed]
- Brown, C.T.; Davis-Richardson, A.G.; Giongo, A.; Gano, K.A.; Crabb, D.B.; Mukherjee, N.; Casella, G.; Drew, J.C.; Ilonen, J.; Knip, M.; et al. Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes. PLoS ONE 2011, 6, e25792. [Google Scholar] [CrossRef] [Green Version]
- Rosser, E.C.; Mauri, C. A clinical update on the significance of the gut microbiota in systemic autoimmunity. J. Autoimmun. 2016, 74, 85–93. [Google Scholar] [CrossRef]
- Clemente, J.C.; Manasson, J.; Scher, J.U. The role of the gut microbiome in systemic inflammatory disease. BMJ 2018, 360, j5145. [Google Scholar] [CrossRef]
- Ghaisas, S.; Maher, J.; Kanthasamy, A. Gut microbiome in health and disease: Linking the microbiome-gut-brain axis and environmental factors in the pathogenesis of systemic and neurodegenerative diseases. Pharmacol. Ther. 2016, 158, 52–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Westfall, S.; Lomis, N.; Kahouli, I.; Dia, S.Y.; Singh, S.P.; Prakash, S. Microbiome, probiotics and neurodegenerative diseases: Deciphering the gut brain axis. Cell. Mol. Life Sci. 2017, 74, 3769–3787. [Google Scholar] [CrossRef] [PubMed]
- Roy Sarkar, S.; Banerjee, S. Gut microbiota in neurodegenerative disorders. J. Neuroimmunol. 2019, 328, 98–104. [Google Scholar] [CrossRef] [PubMed]
- Bauer, P.V.; Hamr, S.C.; Duca, F.A. Regulation of energy balance by a gut-brain axis and involvement of the gut microbiota. Cell. Mol. Life Sci. 2016, 73, 737–755. [Google Scholar] [CrossRef]
- Ojeda, P.; Bobe, A.; Dolan, K.; Leone, V.; Martinez, K. Nutritional modulation of gut microbiota—The impact on metabolic disease pathophysiology. J. Nutr. Biochem. 2016, 28, 191–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marinoni, I.; Nonnis, S.; Monteferrante, C.; Heathcote, P.; Hartig, E.; Bottger, L.H.; Trautwein, A.X.; Negri, A.; Albertini, A.M.; Tedeschi, G. Characterization of L-aspartate oxidase and quinolinate synthase from Bacillus subtilis. FEBS J. 2008, 275, 5090–5107. [Google Scholar] [CrossRef]
- Barends, T.R.; Dunn, M.F.; Schlichting, I. Tryptophan synthase, an allosteric molecular factory. Curr. Opin. Chem. Biol. 2008, 12, 593–600. [Google Scholar] [CrossRef]
- Reichmann, D.; Coute, Y.; Ollagnier de Choudens, S. Dual activity of quinolinate synthase: Triose phosphate isomerase and dehydration activities play together to form quinolinate. Biochemistry 2015, 54, 6443–6446. [Google Scholar] [CrossRef] [PubMed]
- Sakuraba, H.; Tsuge, H.; Yoneda, K.; Katunuma, N.; Ohshima, T. Crystal structure of the NAD biosynthetic enzyme quinolinate synthase. J. Biol. Chem. 2005, 280, 26645–26648. [Google Scholar] [CrossRef] [Green Version]
- Saunders, A.H.; Griffiths, A.E.; Lee, K.H.; Cicchillo, R.M.; Tu, L.; Stromberg, J.A.; Krebs, C.; Booker, S.J. Characterization of quinolinate synthases from Escherichia coli, Mycobacterium tuberculosis, and Pyrococcus horikoshii indicates that [4Fe-4S] clusters are common cofactors throughout this class of enzymes. Biochemistry 2008, 47, 10999–11012. [Google Scholar] [CrossRef] [Green Version]
- Garavaglia, S.; Perozzi, S.; Galeazzi, L.; Raffaelli, N.; Rizzi, M. The crystal structure of human alpha-amino-beta-carboxymuconate-epsilon-semialdehyde decarboxylase in complex with 1,3-dihydroxyacetonephosphate suggests a regulatory link between NAD synthesis and glycolysis. FEBS J. 2009, 276, 6615–6623. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Dong, Y.; Li, X.; Ren, Y.; Li, Y.; Wang, W.; Wang, L.; Feng, L. Characterization of the anthranilate degradation pathway in Geobacillus thermodenitrificans NG80-2. Microbiology 2010, 156, 589–595. [Google Scholar] [CrossRef] [Green Version]
- Cleaves, H.J.; Miller, S.L. The nicotinamide biosynthetic pathway is a by-product of the RNA world. J. Mol. Evol. 2001, 52, 73–77. [Google Scholar] [CrossRef]
- Shahi, S.K.; Freedman, S.N.; Mangalam, A.K. Gut microbiome in multiple sclerosis: The players involved and the roles they play. Gut Microbes 2017, 8, 607–615. [Google Scholar] [CrossRef] [PubMed]
- Mielcarz, D.W.; Kasper, L.H. The gut microbiome in multiple sclerosis. Curr. Treat. Options Neurol. 2015, 17, 344. [Google Scholar] [CrossRef] [PubMed]
- Jangi, S.; Gandhi, R.; Cox, L.M.; Li, N.; Von Glehn, F.; Yan, R.; Patel, B.; Mazzola, M.A.; Liu, S.; Glanz, B.L.; et al. Alterations of the human gut microbiome in multiple sclerosis. Nat. Commun. 2016, 7, 12015. [Google Scholar] [CrossRef] [PubMed]
- O’Mahony, S.M.; Clarke, G.; Borre, Y.E.; Dinan, T.G.; Cryan, J.F. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav. Brain Res. 2015, 277, 32–48. [Google Scholar] [CrossRef] [PubMed]
- Kennedy, P.J.; Cryan, J.F.; Dinan, T.G.; Clarke, G. Kynurenine pathway metabolism and the microbiota-gut-brain axis. Neuropharmacology 2017, 112, 399–412. [Google Scholar] [CrossRef]
- Suhs, K.W.; Novoselova, N.; Kuhn, M.; Seegers, L.; Kaever, V.; Muller-Vahl, K.; Trebst, C.; Skripuletz, T.; Stangel, M.; Pessler, F. Kynurenine Is a Cerebrospinal Fluid Biomarker for Bacterial and Viral Central Nervous System Infections. J. Infect. Dis. 2019, 220, 127–138. [Google Scholar] [CrossRef]
- Jeltsch-David, H.; Muller, S. Neuropsychiatric systemic lupus erythematosus: Pathogenesis and biomarkers. Nat. Rev. Neurol. 2014, 10, 579–596. [Google Scholar] [CrossRef]
- Akesson, K.; Pettersson, S.; Stahl, S.; Surowiec, I.; Hedenstrom, M.; Eketjall, S.; Trygg, J.; Jakobsson, P.J.; Gunnarsson, I.; Svenungsson, E.; et al. Kynurenine pathway is altered in patients with SLE and associated with severe fatigue. Lupus Sci. Med. 2018, 5, e000254. [Google Scholar] [CrossRef] [Green Version]
- Vitali, C.; Bombardieri, S.; Jonsson, R.; Moutsopoulos, H.M.; Alexander, E.L.; Carsons, S.E.; Daniels, T.E.; Fox, P.C.; Fox, R.I.; Kassan, S.S.; et al. Classification criteria for Sjogren’s syndrome: A revised version of the European criteria proposed by the American-European Consensus Group. Ann. Rheum. Dis. 2002, 61, 554–558. [Google Scholar] [CrossRef] [Green Version]
- Brito-Zeron, P.; Theander, E.; Baldini, C.; Seror, R.; Retamozo, S.; Quartuccio, L.; Bootsma, H.; Bowman, S.J.; Dorner, T.; Gottenberg, J.E.; et al. Early diagnosis of primary Sjogren’s syndrome: EULAR-SS task force clinical recommendations. Exp. Rev. Clin. Immunol. 2016, 12, 137–156. [Google Scholar] [CrossRef]
- De Oliveira, F.R.; Fantucci, M.Z.; Adriano, L.; Valim, V.; Cunha, T.M.; Louzada-Junior, P.; Rocha, E.M. Neurological and Inflammatory Manifestations in Sjogren’s Syndrome: The Role of the Kynurenine Metabolic Pathway. Int. J. Mol. Sci. 2018, 19, 3953. [Google Scholar] [CrossRef] [Green Version]
- Furuzawa-Carballeda, J.; Hernandez-Molina, G.; Lima, G.; Rivera-Vicencio, Y.; Ferez-Blando, K.; Llorente, L. Peripheral regulatory cells immunophenotyping in primary Sjogren’s syndrome: A cross-sectional study. Arthritis Res. Ther. 2013, 15, R68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Legany, N.; Berta, L.; Kovacs, L.; Balog, A.; Toldi, G. The role of B7 family costimulatory molecules and indoleamine 2,3-dioxygenase in primary Sjogren’s syndrome and systemic sclerosis. Immunol. Res. 2017, 65, 622–629. [Google Scholar] [CrossRef] [PubMed]
- Raine, C.S.; Scheinberg, L.C. On the immunopathology of plaque development and repair in multiple sclerosis. J. Neuroimmunol. 1988, 20, 189–201. [Google Scholar] [CrossRef]
- O’Connor, R.A.; Prendergast, C.T.; Sabatos, C.A.; Lau, C.W.; Leech, M.D.; Wraith, D.C.; Anderton, S.M. Cutting edge: Th1 cells facilitate the entry of Th17 cells to the central nervous system during experimental autoimmune encephalomyelitis. J. Immunol. 2008, 181, 3750–3754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miller, S.D.; Karpus, W.J. Experimental autoimmune encephalomyelitis in the mouse. Curr. Protoc. Immunol. 2007, 15, 15.1. [Google Scholar] [CrossRef] [Green Version]
- Gold, R.; Linington, C.; Lassmann, H. Understanding pathogenesis and therapy of multiple sclerosis via animal models: 70 years of merits and culprits in experimental autoimmune encephalomyelitis research. Brain 2006, 129, 1953–1971. [Google Scholar] [CrossRef]
- Fuvesi, J.; Rajda, C.; Bencsik, K.; Toldi, J.; Vecsei, L. The role of kynurenines in the pathomechanism of amyotrophic lateral sclerosis and multiple sclerosis: Therapeutic implications. J. Neural Transm. 2012, 119, 225–234. [Google Scholar] [CrossRef] [PubMed]
- Lassmann, H.; Ransohoff, R.M. The CD4-Th1 model for multiple sclerosis: A critical [correction of crucial re-appraisal. Trends Immunol. 2004, 25, 132–137. [Google Scholar] [CrossRef]
- Barnett, M.H.; Prineas, J.W. Relapsing and remitting multiple sclerosis: Pathology of the newly forming lesion. Ann. Neurol. 2004, 55, 458–468. [Google Scholar] [CrossRef]
- Lodygin, D.; Hermann, M.; Schweingruber, N.; Flugel-Koch, C.; Watanabe, T.; Schlosser, C.; Merlini, A.; Korner, H.; Chang, H.F.; Fischer, H.J.; et al. beta-Synuclein-reactive T cells induce autoimmune CNS grey matter degeneration. Nature 2019, 566, 503–508. [Google Scholar] [CrossRef]
- Dutta, R.; McDonough, J.; Yin, X.; Peterson, J.; Chang, A.; Torres, T.; Gudz, T.; Macklin, W.B.; Lewis, D.A.; Fox, R.J.; et al. Mitochondrial dysfunction as a cause of axonal degeneration in multiple sclerosis patients. Ann. Neurol. 2006, 59, 478–489. [Google Scholar] [CrossRef] [PubMed]
- Lu, F.; Selak, M.; O’Connor, J.; Croul, S.; Lorenzana, C.; Butunoi, C.; Kalman, B. Oxidative damage to mitochondrial DNA and activity of mitochondrial enzymes in chronic active lesions of multiple sclerosis. J. Neurol. Sci. 2000, 177, 95–103. [Google Scholar] [CrossRef]
- Mahad, D.; Ziabreva, I.; Lassmann, H.; Turnbull, D. Mitochondrial defects in acute multiple sclerosis lesions. Brain 2008, 131, 1722–1735. [Google Scholar] [CrossRef]
- Trapp, B.D.; Stys, P.K. Virtual hypoxia and chronic necrosis of demyelinated axons in multiple sclerosis. Lancet Neurol. 2009, 8, 280–291. [Google Scholar] [CrossRef]
- Witte, M.E.; Bo, L.; Rodenburg, R.J.; Belien, J.A.; Musters, R.; Hazes, T.; Wintjes, L.T.; Smeitink, J.A.; Geurts, J.J.; De Vries, H.E.; et al. Enhanced number and activity of mitochondria in multiple sclerosis lesions. J. Pathol. 2009, 219, 193–204. [Google Scholar] [CrossRef]
- Veto, S.; Acs, P.; Bauer, J.; Lassmann, H.; Berente, Z.; Setalo, G., Jr.; Borgulya, G.; Sumegi, B.; Komoly, S.; Gallyas, F., Jr.; et al. Inhibiting poly(ADP-ribose) polymerase: A potential therapy against oligodendrocyte death. Brain 2010, 133, 822–834. [Google Scholar] [CrossRef] [Green Version]
- Ziabreva, I.; Campbell, G.; Rist, J.; Zambonin, J.; Rorbach, J.; Wydro, M.M.; Lassmann, H.; Franklin, R.J.; Mahad, D. Injury and differentiation following inhibition of mitochondrial respiratory chain complex IV in rat oligodendrocytes. Glia 2010, 58, 1827–1837. [Google Scholar] [CrossRef] [Green Version]
- Mahad, D.; Lassmann, H.; Turnbull, D. Review: Mitochondria and disease progression in multiple sclerosis. Neuropathol. Appl. Neurobiol. 2008, 34, 577–589. [Google Scholar] [CrossRef] [PubMed]
- Mahad, D.J.; Ziabreva, I.; Campbell, G.; Lax, N.; White, K.; Hanson, P.S.; Lassmann, H.; Turnbull, D.M. Mitochondrial changes within axons in multiple sclerosis. Brain 2009, 132, 1161–1174. [Google Scholar] [CrossRef] [Green Version]
- Sharma, R.; Fischer, M.T.; Bauer, J.; Felts, P.A.; Smith, K.J.; Misu, T.; Fujihara, K.; Bradl, M.; Lassmann, H. Inflammation induced by innate immunity in the central nervous system leads to primary astrocyte dysfunction followed by demyelination. Acta Neuropathol. 2010, 120, 223–236. [Google Scholar] [CrossRef] [Green Version]
- Kwidzinski, E.; Bunse, J.; Aktas, O.; Richter, D.; Mutlu, L.; Zipp, F.; Nitsch, R.; Bechmann, I. Indolamine 2,3-dioxygenase is expressed in the CNS and down-regulates autoimmune inflammation. FASEB J. 2005, 19, 1347–1349. [Google Scholar] [CrossRef] [PubMed]
- Sakurai, K.; Zou, J.P.; Tschetter, J.R.; Ward, J.M.; Shearer, G.M. Effect of indoleamine 2,3-dioxygenase on induction of experimental autoimmune encephalomyelitis. J. Neuroimmunol. 2002, 129, 186–196. [Google Scholar] [CrossRef]
- Yan, Y.; Zhang, G.X.; Gran, B.; Fallarino, F.; Yu, S.; Li, H.; Cullimore, M.L.; Rostami, A.; Xu, H. IDO upregulates regulatory T cells via tryptophan catabolite and suppresses encephalitogenic T cell responses in experimental autoimmune encephalomyelitis. J. Immunol. 2010, 185, 5953–5961. [Google Scholar] [CrossRef] [PubMed]
- Gonsette, R.E. Self-tolerance in multiple sclerosis. Acta Neurol. Belg. 2012, 112, 133–140. [Google Scholar] [CrossRef] [Green Version]
- Fazio, F.; Zappulla, C.; Notartomaso, S.; Busceti, C.; Bessede, A.; Scarselli, P.; Vacca, C.; Gargaro, M.; Volpi, C.; Allegrucci, M.; et al. Cinnabarinic acid, an endogenous agonist of type-4 metabotropic glutamate receptor, suppresses experimental autoimmune encephalomyelitis in mice. Neuropharmacology 2014, 81, 237–243. [Google Scholar] [CrossRef]
- Xiao, B.G.; Liu, X.; Link, H. Antigen-specific T cell functions are suppressed over the estrogen-dendritic cell-indoleamine 2,3-dioxygenase axis. Steroids 2004, 69, 653–659. [Google Scholar] [CrossRef] [PubMed]
- Platten, M.; Ho, P.P.; Youssef, S.; Fontoura, P.; Garren, H.; Hur, E.M.; Gupta, R.; Lee, L.Y.; Kidd, B.A.; Robinson, W.H.; et al. Treatment of autoimmune neuroinflammation with a synthetic tryptophan metabolite. Science 2005, 310, 850–855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chiarugi, A.; Cozzi, A.; Ballerini, C.; Massacesi, L.; Moroni, F. Kynurenine 3-mono-oxygenase activity and neurotoxic kynurenine metabolites increase in the spinal cord of rats with experimental allergic encephalomyelitis. Neuroscience 2001, 102, 687–695. [Google Scholar] [CrossRef]
- Flanagan, E.M.; Erickson, J.B.; Viveros, O.H.; Chang, S.Y.; Reinhard, J.F., Jr. Neurotoxin quinolinic acid is selectively elevated in spinal cords of rats with experimental allergic encephalomyelitis. J. Neurochem. 1995, 64, 1192–1196. [Google Scholar] [CrossRef] [PubMed]
- Cammer, W. Oligodendrocyte killing by quinolinic acid in vitro. Brain Res. 2001, 896, 157–160. [Google Scholar] [CrossRef]
- Guillemin, G.J.; Wang, L.; Brew, B.J. Quinolinic acid selectively induces apoptosis of human astrocytes: Potential role in AIDS dementia complex. J. Neuroinflamm. 2005, 2, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kerr, S.J.; Armati, P.J.; Guillemin, G.J.; Brew, B.J. Chronic exposure of human neurons to quinolinic acid results in neuronal changes consistent with AIDS dementia complex. Aids 1998, 12, 355–363. [Google Scholar] [CrossRef] [PubMed]
- Rajda, C.; Majlath, Z.; Pukoli, D.; Vecsei, L. Kynurenines and Multiple Sclerosis: The Dialogue between the Immune System and the Central Nervous System. Int. J. Mol. Sci. 2015, 16, 18270–18282. [Google Scholar] [CrossRef]
- Behan, W.M.; McDonald, M.; Darlington, L.G.; Stone, T.W. Oxidative stress as a mechanism for quinolinic acid-induced hippocampal damage: Protection by melatonin and deprenyl. Br. J. Pharmacol. 1999, 128, 1754–1760. [Google Scholar] [CrossRef] [PubMed]
- Santamaria, A.; Jimenez-Capdeville, M.E.; Camacho, A.; Rodriguez-Martinez, E.; Flores, A.; Galvan-Arzate, S. In vivo hydroxyl radical formation after quinolinic acid infusion into rat corpus striatum. Neuroreport 2001, 12, 2693–2696. [Google Scholar] [CrossRef] [PubMed]
- Leipnitz, G.; Schumacher, C.; Scussiato, K.; Dalcin, K.B.; Wannmacher, C.M.; Wyse, A.T.; Dutra-Filho, C.S.; Wajner, M.; Latini, A. Quinolinic acid reduces the antioxidant defenses in cerebral cortex of young rats. Int. J. Dev. Neurosci. 2005, 23, 695–701. [Google Scholar] [CrossRef]
- Rodriguez-Martinez, E.; Camacho, A.; Maldonado, P.D.; Pedraza-Chaverri, J.; Santamaria, D.; Galvan-Arzate, S.; Santamaria, A. Effect of quinolinic acid on endogenous antioxidants in rat corpus striatum. Brain Res. 2000, 858, 436–439. [Google Scholar] [CrossRef]
- Rodriguez, E.; Mendez-Armenta, M.; Villeda-Hernandez, J.; Galvan-Arzate, S.; Barroso-Moguel, R.; Rodriguez, F.; Rios, C.; Santamaria, A. Dapsone prevents morphological lesions and lipid peroxidation induced by quinolinic acid in rat corpus striatum. Toxicology 1999, 139, 111–118. [Google Scholar] [CrossRef]
- Santamaria, A.; Galvan-Arzate, S.; Lisy, V.; Ali, S.F.; Duhart, H.M.; Osorio-Rico, L.; Rios, C.; St’astny, F. Quinolinic acid induces oxidative stress in rat brain synaptosomes. Neuroreport 2001, 12, 871–874. [Google Scholar] [CrossRef]
- Baran, H.; Staniek, K.; Kepplinger, B.; Gille, L.; Stolze, K.; Nohl, H. Kynurenic acid influences the respiratory parameters of rat heart mitochondria. Pharmacology 2001, 62, 119–123. [Google Scholar] [CrossRef]
- Bordelon, Y.M.; Chesselet, M.F.; Nelson, D.; Welsh, F.; Erecinska, M. Energetic dysfunction in quinolinic acid-lesioned rat striatum. J. Neurochem. 1997, 69, 1629–1639. [Google Scholar] [CrossRef] [PubMed]
- Blumenthal, A.; Nagalingam, G.; Huch, J.H.; Walker, L.; Guillemin, G.J.; Smythe, G.A.; Ehrt, S.; Britton, W.J.; Saunders, B.M.M. tuberculosis induces potent activation of IDO-1, but this is not essential for the immunological control of infection. PLoS ONE 2012, 7, e37314. [Google Scholar] [CrossRef] [Green Version]
- Suzuki, Y.; Suda, T.; Asada, K.; Miwa, S.; Suzuki, M.; Fujie, M.; Furuhashi, K.; Nakamura, Y.; Inui, N.; Shirai, T.; et al. Serum indoleamine 2,3-dioxygenase activity predicts prognosis of pulmonary tuberculosis. Clin. Vaccine Immunol. 2012, 19, 436–442. [Google Scholar] [CrossRef] [PubMed]
- Watzlawik, J.O.; Wootla, B.; Rodriguez, M. Tryptophan Catabolites and Their Impact on Multiple Sclerosis Progression. Curr. Pharma. Des. 2016, 22, 1049–1059. [Google Scholar] [CrossRef]
- Pemberton, L.A.; Kerr, S.J.; Smythe, G.; Brew, B.J. Quinolinic acid production by macrophages stimulated with IFN-gamma, TNF-alpha, and IFN-alpha. J. Interferon Cytokine Res. 1997, 17, 589–595. [Google Scholar] [CrossRef]
- Beck, J.; Rondot, P.; Catinot, L.; Falcoff, E.; Kirchner, H.; Wietzerbin, J. Increased production of interferon gamma and tumor necrosis factor precedes clinical manifestation in multiple sclerosis: Do cytokines trigger off exacerbations? Acta Neurol. Scand. 1988, 78, 318–323. [Google Scholar] [CrossRef]
- Rajda, C.; Galla, Z.; Polyak, H.; Maroti, Z.; Babarczy, K.; Pukoli, D.; Vecsei, L. Cerebrospinal Fluid Neurofilament Light Chain Is Associated with Kynurenine Pathway Metabolite Changes in Multiple Sclerosis. Int. J. Mol. Sci. 2020, 21, 2665. [Google Scholar] [CrossRef] [Green Version]
- Monaco, F.; Fumero, S.; Mondino, A.; Mutani, R. Plasma and cerebrospinal fluid tryptophan in multiple sclerosis and degenerative diseases. J. Neurol. Neurosurg. Psychiatry 1979, 42, 640–641. [Google Scholar] [CrossRef]
- Ott, M.; Demisch, L.; Engelhardt, W.; Fischer, P.A. Interleukin-2, soluble interleukin-2-receptor, neopterin, L-tryptophan and beta 2-microglobulin levels in CSF and serum of patients with relapsing-remitting or chronic-progressive multiple sclerosis. J. Neurol. 1993, 241, 108–114. [Google Scholar] [CrossRef]
- Rudzite, V.; Berzinsh, J.; Grivane, I.; Fuchs, D.; Baier-Bitterlich, G.; Wachter, H. Serum tryptophan, kynurenine, and neopterin in patients with Guillain-Barre-syndrome (GBS) and multiple sclerosis (MS). Adv. Exp. Med. Biol. 1996, 398, 183–187. [Google Scholar] [CrossRef]
- Amirkhani, A.; Rajda, C.; Arvidsson, B.; Bencsik, K.; Boda, K.; Seres, E.; Markides, K.E.; Vecsei, L.; Bergquist, J. Interferon-beta affects the tryptophan metabolism in multiple sclerosis patients. Eur. J. Neurol. 2005, 12, 625–631. [Google Scholar] [CrossRef]
- Rothhammer, V.; Borucki, D.M.; Garcia Sanchez, M.I.; Mazzola, M.A.; Hemond, C.C.; Regev, K.; Paul, A.; Kivisakk, P.; Bakshi, R.; Izquierdo, G.; et al. Dynamic regulation of serum aryl hydrocarbon receptor agonists in MS. Neurol. Neuroimmunol. Neuroinflamm. 2017, 4, e359. [Google Scholar] [CrossRef] [PubMed]
- Rejdak, K.; Petzold, A.; Kocki, T.; Kurzepa, J.; Grieb, P.; Turski, W.A.; Stelmasiak, Z. Astrocytic activation in relation to inflammatory markers during clinical exacerbation of relapsing-remitting multiple sclerosis. J. Neural Transm. 2007, 114, 1011–1015. [Google Scholar] [CrossRef] [PubMed]
- Rejdak, K.; Bartosik-Psujek, H.; Dobosz, B.; Kocki, T.; Grieb, P.; Giovannoni, G.; Turski, W.A.; Stelmasiak, Z. Decreased level of kynurenic acid in cerebrospinal fluid of relapsing-onset multiple sclerosis patients. Neurosci. Lett. 2002, 331, 63–65. [Google Scholar] [CrossRef]
- Anderson, J.M.; Hampton, D.W.; Patani, R.; Pryce, G.; Crowther, R.A.; Reynolds, R.; Franklin, R.J.; Giovannoni, G.; Compston, D.A.; Baker, D.; et al. Abnormally phosphorylated tau is associated with neuronal and axonal loss in experimental autoimmune encephalomyelitis and multiple sclerosis. Brain 2008, 131, 1736–1748. [Google Scholar] [CrossRef]
- Lim, C.K.; Brew, B.J.; Sundaram, G.; Guillemin, G.J. Understanding the roles of the kynurenine pathway in multiple sclerosis progression. Int. J. Tryptophan Res. 2010, 3, 157–167. [Google Scholar] [CrossRef] [PubMed]
- Lim, C.K.; Bilgin, A.; Lovejoy, D.B.; Tan, V.; Bustamante, S.; Taylor, B.V.; Bessede, A.; Brew, B.J.; Guillemin, G.J. Kynurenine pathway metabolomics predicts and provides mechanistic insight into multiple sclerosis progression. Sci. Rep. 2017, 7, 41473. [Google Scholar] [CrossRef]
- Mancuso, R.; Hernis, A.; Agostini, S.; Rovaris, M.; Caputo, D.; Fuchs, D.; Clerici, M. Indoleamine 2,3 Dioxygenase (IDO) Expression and Activity in Relapsing-Remitting Multiple Sclerosis. PLoS ONE 2015, 10, e0130715. [Google Scholar] [CrossRef] [Green Version]
- Aeinehband, S.; Brenner, P.; Stahl, S.; Bhat, M.; Fidock, M.D.; Khademi, M.; Olsson, T.; Engberg, G.; Jokinen, J.; Erhardt, S.; et al. Cerebrospinal fluid kynurenines in multiple sclerosis; relation to disease course and neurocognitive symptoms. Brain Behav. Immun. 2016, 51, 47–55. [Google Scholar] [CrossRef] [Green Version]
- Boeschoten, R.E.; Braamse, A.M.J.; Beekman, A.T.F.; Cuijpers, P.; Van Oppen, P.; Dekker, J.; Uitdehaag, B.M.J. Prevalence of depression and anxiety in Multiple Sclerosis: A systematic review and meta-analysis. J. Neurol. Sci. 2017, 372, 331–341. [Google Scholar] [CrossRef]
- Anderson, G.; Maes, M. Oxidative/nitrosative stress and immuno-inflammatory pathways in depression: Treatment implications. Curr. Pharm. Des. 2014, 20, 3812–3847. [Google Scholar] [CrossRef]
- Anderson, G.; Maes, M. TRYCAT pathways link peripheral inflammation, nicotine, somatization and depression in the etiology and course of Parkinson’s disease. CNS Neurol. Disord. Drug Targets 2014, 13, 137–149. [Google Scholar] [CrossRef] [PubMed]
- Maes, M.; Kubera, M.; Obuchowiczwa, E.; Goehler, L.; Brzeszcz, J. Depression’s multiple comorbidities explained by (neuro)inflammatory and oxidative & nitrosative stress pathways. Neuro Endocrinol. Lett. 2011, 32, 7–24. [Google Scholar] [PubMed]
- Campbell, B.M.; Charych, E.; Lee, A.W.; Moller, T. Kynurenines in CNS disease: Regulation by inflammatory cytokines. Front. Neurosci. 2014, 8, 12. [Google Scholar] [CrossRef] [Green Version]
- Anderson, G.; Maes, M.; Berk, M. Inflammation-related disorders in the tryptophan catabolite pathway in depression and somatization. Adv. Protein Chem. Struct. Biol. 2012, 88, 27–48. [Google Scholar] [CrossRef] [PubMed]
- Durastanti, V.; Lugaresi, A.; Bramanti, P.; Amato, M.; Bellantonio, P.; De Luca, G.; Picconi, O.; Fantozzi, R.; Locatelli, L.; Solda, A.; et al. Neopterin production and tryptophan degradation during 24-months therapy with interferon beta-1a in multiple sclerosis patients. J. Transl. Med. 2011, 9, 42. [Google Scholar] [CrossRef] [Green Version]
- Sadowska-Bartosz, I.; Adamczyk-Sowa, M.; Gajewska, A.; Bartosz, G. Oxidative modification of blood serum proteins in multiple sclerosis after interferon or mitoxantrone treatment. J. Neuroimmunol. 2014, 266, 67–74. [Google Scholar] [CrossRef]
- Kappos, L.; Polman, C.H.; Freedman, M.S.; Edan, G.; Hartung, H.P.; Miller, D.H.; Montalban, X.; Barkhof, F.; Bauer, L.; Jakobs, P.; et al. Treatment with interferon beta-1b delays conversion to clinically definite and McDonald MS in patients with clinically isolated syndromes. Neurology 2006, 67, 1242–1249. [Google Scholar] [CrossRef]
- O’Connor, P.; Filippi, M.; Arnason, B.; Comi, G.; Cook, S.; Goodin, D.; Hartung, H.P.; Jeffery, D.; Kappos, L.; Boateng, F.; et al. 250 microg or 500 microg interferon beta-1b versus 20 mg glatiramer acetate in relapsing-remitting multiple sclerosis: A prospective, randomised, multicentre study. Lancet Neurol. 2009, 8, 889–897. [Google Scholar] [CrossRef] [Green Version]
- Reder, A.T.; Ebers, G.C.; Traboulsee, A.; Li, D.; Langdon, D.; Goodin, D.S.; Bogumil, T.; Beckmann, K.; Konieczny, A. Investigators of the 16-Year Long-Term Follow-Up, S. Cross-sectional study assessing long-term safety of interferon-beta-1b for relapsing-remitting MS. Neurology 2010, 74, 1877–1885. [Google Scholar] [CrossRef]
- Smith, A.K.; Simon, J.S.; Gustafson, E.L.; Noviello, S.; Cubells, J.F.; Epstein, M.P.; Devlin, D.J.; Qiu, P.; Albrecht, J.K.; Brass, C.A.; et al. Association of a polymorphism in the indoleamine- 2,3-dioxygenase gene and interferon-alpha-induced depression in patients with chronic hepatitis C. Mol. Psychiatry 2012, 17, 781–789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Capuron, L.; Neurauter, G.; Musselman, D.L.; Lawson, D.H.; Nemeroff, C.B.; Fuchs, D.; Miller, A.H. Interferon-alpha-induced changes in tryptophan metabolism. relationship to depression and paroxetine treatment. Biol. Psychiatry 2003, 54, 906–914. [Google Scholar] [CrossRef]
- Wichers, M.C.; Koek, G.H.; Robaeys, G.; Verkerk, R.; Scharpe, S.; Maes, M. IDO and interferon-alpha-induced depressive symptoms: A shift in hypothesis from tryptophan depletion to neurotoxicity. Mol. Psychiatry 2005, 10, 538–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galvao-de Almeida, A.; Quarantini, L.C.; Sampaio, A.S.; Lyra, A.C.; Parise, C.L.; Parana, R.; De Oliveira, I.R.; Koenen, K.C.; Miranda-Scippa, A.; Guindalini, C. Lack of association of indoleamine 2,3-dioxygenase polymorphisms with interferon-alpha-related depression in hepatitis C. Brain Behav. Immun. 2011, 25, 1491–1497. [Google Scholar] [CrossRef] [Green Version]
- Guillemin, G.J.; Kerr, S.J.; Pemberton, L.A.; Smith, D.G.; Smythe, G.A.; Armati, P.J.; Brew, B.J. IFN-beta1b induces kynurenine pathway metabolism in human macrophages: Potential implications for multiple sclerosis treatment. J. Interferon Cytokine Res. 2001, 21, 1097–1101. [Google Scholar] [CrossRef]
- Ratajczak, J.; Joffraud, M.; Trammell, S.A.; Ras, R.; Canela, N.; Boutant, M.; Kulkarni, S.S.; Rodrigues, M.; Redpath, P.; Migaud, M.E.; et al. NRK1 controls nicotinamide mononucleotide and nicotinamide riboside metabolism in mammalian cells. Nat. Commun. 2016, 7, 13103. [Google Scholar] [CrossRef]
- Revollo, J.R.; Grimm, A.A.; Imai, S. The regulation of nicotinamide adenine dinucleotide biosynthesis by Nampt/PBEF/visfatin in mammals. Curr. Opin. Gastroenterol. 2007, 23, 164–170. [Google Scholar] [CrossRef]
- Penberthy, W.T.; Tsunoda, I. The importance of NAD in multiple sclerosis. Curr. Pharm. Des. 2009, 15, 64–99. [Google Scholar] [CrossRef] [Green Version]
- Braidy, N.; Grant, R. Kynurenine pathway metabolism and neuroinflammatory disease. Neural Regen. Res. 2017, 12, 39–42. [Google Scholar] [CrossRef]
- Massudi, H.; Grant, R.; Guillemin, G.J.; Braidy, N. NAD+ metabolism and oxidative stress: The golden nucleotide on a crown of thorns. Redox Rep. Commun. Free Radic. Res. 2012, 17, 28–46. [Google Scholar] [CrossRef]
- Abeti, R.; Duchen, M.R. Activation of PARP by oxidative stress induced by beta-amyloid: Implications for Alzheimer’s disease. Neurochem. Res. 2012, 37, 2589–2596. [Google Scholar] [CrossRef] [PubMed]
- Braidy, N.; Poljak, A.; Grant, R.; Jayasena, T.; Mansour, H.; Chan-Ling, T.; Guillemin, G.J.; Smythe, G.; Sachdev, P. Mapping NAD(+) metabolism in the brain of ageing Wistar rats: Potential targets for influencing brain senescence. Biogerontology 2014, 15, 177–198. [Google Scholar] [CrossRef] [PubMed]
- Verdin, E. NAD(+) in aging, metabolism, and neurodegeneration. Science 2015, 350, 1208–1213. [Google Scholar] [CrossRef]
- Chiarugi, A. Intrinsic mechanisms of poly(ADP-ribose) neurotoxicity: Three hypotheses. Neurotoxicology 2005, 26, 847–855. [Google Scholar] [CrossRef]
- Braidy, N.; Rossez, H.; Lim, C.K.; Jugder, B.E.; Brew, B.J.; Guillemin, G.J. Characterization of the Kynurenine Pathway in CD8(+) Human Primary Monocyte-Derived Dendritic Cells. Neurotox. Res. 2016, 30, 620–632. [Google Scholar] [CrossRef]
- Schwarcz, R.; Stone, T.W. The kynurenine pathway and the brain: Challenges, controversies and promises. Neuropharmacology 2017, 112, 237–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grant, R.S.; Passey, R.; Matanovic, G.; Smythe, G.; Kapoor, V. Evidence for increased de novo synthesis of NAD in immune-activated RAW264.7 macrophages: A self-protective mechanism? Arch. Biochem. Biophys. 1999, 372, 1–7. [Google Scholar] [CrossRef]
- Oh, G.S.; Pae, H.O.; Choi, B.M.; Chae, S.C.; Lee, H.S.; Ryu, D.G.; Chung, H.T. 3-Hydroxyanthranilic acid, one of metabolites of tryptophan via indoleamine 2,3-dioxygenase pathway, suppresses inducible nitric oxide synthase expression by enhancing heme oxygenase-1 expression. Biochem. Biophys. Res. Commun. 2004, 320, 1156–1162. [Google Scholar] [CrossRef] [PubMed]
- Sekkai, D.; Guittet, O.; Lemaire, G.; Tenu, J.P.; Lepoivre, M. Inhibition of nitric oxide synthase expression and activity in macrophages by 3-hydroxyanthranilic acid, a tryptophan metabolite. Arch. Biochem. Biophys. 1997, 340, 117–123. [Google Scholar] [CrossRef]
- Krause, D.; Suh, H.S.; Tarassishin, L.; Cui, Q.L.; Durafourt, B.A.; Choi, N.; Bauman, A.; Cosenza-Nashat, M.; Antel, J.P.; Zhao, M.L.; et al. The tryptophan metabolite 3-hydroxyanthranilic acid plays anti-inflammatory and neuroprotective roles during inflammation: Role of hemeoxygenase-1. Am. J. Pathol. 2011, 179, 1360–1372. [Google Scholar] [CrossRef]
- Lowe, M.M.; Mold, J.E.; Kanwar, B.; Huang, Y.; Louie, A.; Pollastri, M.P.; Wang, C.; Patel, G.; Franks, D.G.; Schlezinger, J.; et al. Identification of cinnabarinic acid as a novel endogenous aryl hydrocarbon receptor ligand that drives IL-22 production. PLoS ONE 2014, 9, e87877. [Google Scholar] [CrossRef]
- Suh, S.W.; Hamby, A.M.; Swanson, R.A. Hypoglycemia, brain energetics, and hypoglycemic neuronal death. Glia 2007, 55, 1280–1286. [Google Scholar] [CrossRef] [PubMed]
- Schrocksnadel, K.; Wirleitner, B.; Winkler, C.; Fuchs, D. Monitoring tryptophan metabolism in chronic immune activation. Clin. Chim. Acta 2006, 364, 82–90. [Google Scholar] [CrossRef]
- Penberthy, W.T. Pharmacological targeting of IDO-mediated tolerance for treating autoimmune disease. Curr. Drug Metabol. 2007, 8, 245–266. [Google Scholar] [CrossRef] [PubMed]
- Munn, D.H.; Shafizadeh, E.; Attwood, J.T.; Bondarev, I.; Pashine, A.; Mellor, A.L. Inhibition of T cell proliferation by macrophage tryptophan catabolism. J. Exp. Med. 1999, 189, 1363–1372. [Google Scholar] [CrossRef] [PubMed]
- Munn, D.H.; Sharma, M.D.; Mellor, A.L. Ligation of B7-1/B7-2 by human CD4+ T cells triggers indoleamine 2,3-dioxygenase activity in dendritic cells. J. Immunol. 2004, 172, 4100–4110. [Google Scholar] [CrossRef] [Green Version]
- Iribarren, P.; Cui, Y.H.; Le, Y.; Wang, J.M. The role of dendritic cells in neurodegenerative diseases. Archiv. Immunol. Ther. Exp. 2002, 50, 187–196. [Google Scholar]
- Pashenkov, M.; Teleshova, N.; Link, H. Inflammation in the central nervous system: The role for dendritic cells. Brain Pathol. 2003, 13, 23–33. [Google Scholar] [CrossRef]
- Matysiak, M.; Stasiolek, M.; Orlowski, W.; Jurewicz, A.; Janczar, S.; Raine, C.S.; Selmaj, K. Stem cells ameliorate EAE via an indoleamine 2,3-dioxygenase (IDO) mechanism. J. Neuroimmunol. 2008, 193, 12–23. [Google Scholar] [CrossRef] [Green Version]
- Belladonna, M.L.; Grohmann, U.; Guidetti, P.; Volpi, C.; Bianchi, R.; Fioretti, M.C.; Schwarcz, R.; Fallarino, F.; Puccetti, P. Kynurenine pathway enzymes in dendritic cells initiate tolerogenesis in the absence of functional IDO. J. Immunol. 2006, 177, 130–137. [Google Scholar] [CrossRef] [Green Version]
- Sharma, M.D.; Baban, B.; Chandler, P.; Hou, D.Y.; Singh, N.; Yagita, H.; Azuma, M.; Blazar, B.R.; Mellor, A.L.; Munn, D.H. Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2,3-dioxygenase. J. Clin. Investig. 2007, 117, 2570–2582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kauppinen, T.M.; Suh, S.W.; Genain, C.P.; Swanson, R.A. Poly(ADP-ribose) polymerase-1 activation in a primate model of multiple sclerosis. J. Neurosci. Res. 2005, 81, 190–198. [Google Scholar] [CrossRef] [PubMed]
- Opitz, C.A.; Wick, W.; Steinman, L.; Platten, M. Tryptophan degradation in autoimmune diseases. Cell. Mol. Life Sci. 2007, 64, 2542–2563. [Google Scholar] [CrossRef]
- Braidy, N.; Lim, C.K.; Grant, R.; Brew, B.J.; Guillemin, G.J. Serum nicotinamide adenine dinucleotide levels through disease course in multiple sclerosis. Brain Res. 2013, 1537, 267–272. [Google Scholar] [CrossRef]
- Kaneko, S.; Wang, J.; Kaneko, M.; Yiu, G.; Hurrell, J.M.; Chitnis, T.; Khoury, S.J.; He, Z. Protecting axonal degeneration by increasing nicotinamide adenine dinucleotide levels in experimental autoimmune encephalomyelitis models. J. Neurosci. 2006, 26, 9794–9804. [Google Scholar] [CrossRef] [PubMed]
- Esquifino, A.I.; Cano, P.; Jimenez, V.; Cutrera, R.A.; Cardinali, D.P. Experimental allergic encephalomyelitis in male Lewis rats subjected to calorie restriction. J. Physiol. Biochem. 2004, 60, 245–252. [Google Scholar] [CrossRef] [PubMed]
- Piccio, L.; Stark, J.L.; Cross, A.H. Chronic calorie restriction attenuates experimental autoimmune encephalomyelitis. J. Leukocyte Biol. 2008, 84, 940–948. [Google Scholar] [CrossRef] [Green Version]
- Esquifino, A.I.; Cano, P.; Jimenez-Ortega, V.; Fernandez-Mateos, M.P.; Cardinali, D.P. Immune response after experimental allergic encephalomyelitis in rats subjected to calorie restriction. J. Neuroinflamm. 2007, 4, 6. [Google Scholar] [CrossRef] [Green Version]
- Grant, R.S.; Naif, H.; Espinosa, M.; Kapoor, V. IDO induction in IFN-gamma activated astroglia: A role in improving cell viability during oxidative stress. Redox Rep. Communic. Free Radic. Res. 2000, 5, 101–104. [Google Scholar] [CrossRef]
- Grant, R.; Kapoor, V. Inhibition of indoleamine 2,3-dioxygenase activity in IFN-gamma stimulated astroglioma cells decreases intracellular NAD levels. Biochem. Pharmacol. 2003, 66, 1033–1036. [Google Scholar] [CrossRef]
- Kujundzic, R.N.; Lowenthal, J.W. The role of tryptophan metabolism in iNOS transcription and nitric oxide production by chicken macrophage cells upon treatment with interferon gamma. Immunol. Lett. 2008, 115, 153–159. [Google Scholar] [CrossRef]
- Adams, O.; Besken, K.; Oberdorfer, C.; MacKenzie, C.R.; Russing, D.; Daubener, W. Inhibition of human herpes simplex virus type 2 by interferon gamma and tumor necrosis factor alpha is mediated by indoleamine 2,3-dioxygenase. Microbes Infect. 2004, 6, 806–812. [Google Scholar] [CrossRef] [PubMed]
- Adams, O.; Besken, K.; Oberdorfer, C.; MacKenzie, C.R.; Takikawa, O.; Daubener, W. Role of indoleamine-2,3-dioxygenase in alpha/beta and gamma interferon-mediated antiviral effects against herpes simplex virus infections. J. Virol. 2004, 78, 2632–2636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bodaghi, B.; Goureau, O.; Zipeto, D.; Laurent, L.; Virelizier, J.L.; Michelson, S. Role of IFN-gamma-induced indoleamine 2,3 dioxygenase and inducible nitric oxide synthase in the replication of human cytomegalovirus in retinal pigment epithelial cells. J. Immunol. 1999, 162, 957–964. [Google Scholar]
- Sanni, L.A.; Thomas, S.R.; Tattam, B.N.; Moore, D.E.; Chaudhri, G.; Stocker, R.; Hunt, N.H. Dramatic changes in oxidative tryptophan metabolism along the kynurenine pathway in experimental cerebral and noncerebral malaria. Am. Pathol. 1998, 152, 611–619. [Google Scholar]
- Pfefferkorn, E.R. Interferon gamma blocks the growth of Toxoplasma gondii in human fibroblasts by inducing the host cells to degrade tryptophan. Proc. Natl. Acad. Sci. USA 1984, 81, 908–912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Byrne, G.I.; Lehmann, L.K.; Landry, G.J. Induction of tryptophan catabolism is the mechanism for gamma-interferon-mediated inhibition of intracellular Chlamydia psittaci replication in T24 cells. Infect. Immun. 1986, 53, 347–351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jonsson, S.; Andersson, G.; Fex, T.; Fristedt, T.; Hedlund, G.; Jansson, K.; Abramo, L.; Fritzson, I.; Pekarski, O.; Runstrom, A.; et al. Synthesis and biological evaluation of new 1,2-dihydro-4-hydroxy-2-oxo-3-quinolinecarboxamides for treatment of autoimmune disorders: Structure-activity relationship. J. Med. Chem. 2004, 47, 2075–2088. [Google Scholar] [CrossRef]
- Majlath, Z.; Annus, A.; Vecsei, L. Kynurenine System and Multiple Sclerosis, Pathomechanism and Drug Targets with an Emphasis on Laquinimod. Curr. Drug Targ. 2018, 19, 805–814. [Google Scholar] [CrossRef]
- Varrin-Doyer, M.; Zamvil, S.S.; Schulze-Topphoff, U. Laquinimod, an up-and-coming immunomodulatory agent for treatment of multiple sclerosis. Exp. Neurol. 2014, 262, 66–71. [Google Scholar] [CrossRef] [Green Version]
- Bruck, W.; Wegner, C. Insight into the mechanism of laquinimod action. J. Neurol. Sci. 2011, 306, 173–179. [Google Scholar] [CrossRef] [PubMed]
- Ali, R.; Nicholas, R.S.; Muraro, P.A. Drugs in development for relapsing multiple sclerosis. Drugs 2013, 73, 625–650. [Google Scholar] [CrossRef]
- Gurevich, M.; Gritzman, T.; Orbach, R.; Tuller, T.; Feldman, A.; Achiron, A. Laquinimod suppress antigen presentation in relapsing-remitting multiple sclerosis: In-vitro high-throughput gene expression study. J. Neuroimmunol. 2010, 221, 87–94. [Google Scholar] [CrossRef] [PubMed]
- Toubi, E.; Nussbaum, S.; Staun-Ram, E.; Snir, A.; Melamed, D.; Hayardeny, L.; Miller, A. Laquinimod modulates B cells and their regulatory effects on T cells in multiple sclerosis. J. Neuroimmunol. 2012, 251, 45–54. [Google Scholar] [CrossRef] [PubMed]
- Bjork, P.; Bjork, A.; Vogl, T.; Stenstrom, M.; Liberg, D.; Olsson, A.; Roth, J.; Ivars, F.; Leanderson, T. Identification of human S100A9 as a novel target for treatment of autoimmune disease via binding to quinoline-3-carboxamides. PLoS Biol. 2009, 7, e97. [Google Scholar] [CrossRef] [Green Version]
- Mishra, M.K.; Wang, J.; Silva, C.; Mack, M.; Yong, V.W. Kinetics of proinflammatory monocytes in a model of multiple sclerosis and its perturbation by laquinimod. Am. J. Pathol. 2012, 181, 642–651. [Google Scholar] [CrossRef]
- Wegner, C.; Stadelmann, C.; Pfortner, R.; Raymond, E.; Feigelson, S.; Alon, R.; Timan, B.; Hayardeny, L.; Bruck, W. Laquinimod interferes with migratory capacity of T cells and reduces IL-17 levels, inflammatory demyelination and acute axonal damage in mice with experimental autoimmune encephalomyelitis. J. Neuroimmunol. 2010, 227, 133–143. [Google Scholar] [CrossRef]
- Schulze-Topphoff, U.; Shetty, A.; Varrin-Doyer, M.; Molnarfi, N.; Sagan, S.A.; Sobel, R.A.; Nelson, P.A.; Zamvil, S.S. Laquinimod, a quinoline-3-carboxamide, induces type II myeloid cells that modulate central nervous system autoimmunity. PLoS ONE 2012, 7, e33797. [Google Scholar] [CrossRef] [Green Version]
- Nyamoya, S.; Steinle, J.; Chrzanowski, U.; Kaye, J.; Schmitz, C.; Beyer, C.; Kipp, M. Laquinimod Supports Remyelination in Non-Supportive Environments. Cells 2019, 8, 1363. [Google Scholar] [CrossRef] [Green Version]
- Jolivel, V.; Luessi, F.; Masri, J.; Kraus, S.H.; Hubo, M.; Poisa-Beiro, L.; Klebow, S.; Paterka, M.; Yogev, N.; Tumani, H.; et al. Modulation of dendritic cell properties by laquinimod as a mechanism for modulating multiple sclerosis. Brain 2013, 136, 1048–1066. [Google Scholar] [CrossRef] [Green Version]
- Kalb, R. The protean actions of neurotrophins and their receptors on the life and death of neurons. Trends Neurosci. 2005, 28, 5–11. [Google Scholar] [CrossRef] [PubMed]
- Aharoni, R.; Saada, R.; Eilam, R.; Hayardeny, L.; Sela, M.; Arnon, R. Oral treatment with laquinimod augments regulatory T-cells and brain-derived neurotrophic factor expression and reduces injury in the CNS of mice with experimental autoimmune encephalomyelitis. J. Neuroimmunol. 2012, 251, 14–24. [Google Scholar] [CrossRef] [PubMed]
- Thone, J.; Ellrichmann, G.; Seubert, S.; Peruga, I.; Lee, D.H.; Conrad, R.; Hayardeny, L.; Comi, G.; Wiese, S.; Linker, R.A.; et al. Modulation of autoimmune demyelination by laquinimod via induction of brain-derived neurotrophic factor. Am. J. Pathol. 2012, 180, 267–274. [Google Scholar] [CrossRef] [PubMed]
- Comi, G.; Jeffery, D.; Kappos, L.; Montalban, X.; Boyko, A.; Rocca, M.A.; Filippi, M.; Group, A.S. Placebo-controlled trial of oral laquinimod for multiple sclerosis. N. Engl. J. Med. 2012, 366, 1000–1009. [Google Scholar] [CrossRef] [Green Version]
- Vollmer, T.L.; Sorensen, P.S.; Selmaj, K.; Zipp, F.; Havrdova, E.; Cohen, J.A.; Sasson, N.; Gilgun-Sherki, Y.; Arnold, D.L.; Group, B.S. A randomized placebo-controlled phase III trial of oral laquinimod for multiple sclerosis. J. Neurol. 2014, 261, 773–783. [Google Scholar] [CrossRef] [PubMed]
- Matsumoto, K.; Kinoshita, K.; Yoshimizu, A.; Kurauchi, Y.; Hisatsune, A.; Seki, T.; Katsuki, H. Laquinimod and 3,3’-diindolylemethane alleviate neuropathological events and neurological deficits in a mouse model of intracerebral hemorrhage. J. Neuroimmunol. 2020, 342, 577195. [Google Scholar] [CrossRef]
Receptor | Ligand | Action | IC/EC50 | Effect |
---|---|---|---|---|
GPR35 | cGMP, LPA, T3, rT3, DHICA | Agonist | 1–10 µM [48,49,50,51,52] | hyperpolarisation, adenylate cyclase inhibition |
AHR | Xenobiotic chemicals | Agonist | 10-100 µM [53,54,55] | migration, proliferation, immunmodulation |
NMDAR | Glycine, D-serine (glycine-2 co agonist NR1 site) | Antagonist | ~8–10 µM [56,57,58,59,60] | excitation, plasticity, neurodegeneration, depolarization, Ca2+ influx |
NMDAR | Glutamate, NMDA (glutamate/NMDA NR2 site) | Antagonist | ~200–500 µM [57,58,60,61,62,63,64,65] | excitation, neurodegeneration, depolarization, Ca2+ influx |
AMPA/ Kainate | Glutamate | Antagonist | ~250 µM [57,58,64,65,66] | excitation, depolarization |
Free radicals | n/a | n/a | >200 µM [67,68] | hydroxyl, superoxide radical complexation |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Biernacki, T.; Sandi, D.; Bencsik, K.; Vécsei, L. Kynurenines in the Pathogenesis of Multiple Sclerosis: Therapeutic Perspectives. Cells 2020, 9, 1564. https://doi.org/10.3390/cells9061564
Biernacki T, Sandi D, Bencsik K, Vécsei L. Kynurenines in the Pathogenesis of Multiple Sclerosis: Therapeutic Perspectives. Cells. 2020; 9(6):1564. https://doi.org/10.3390/cells9061564
Chicago/Turabian StyleBiernacki, Tamás, Dániel Sandi, Krisztina Bencsik, and László Vécsei. 2020. "Kynurenines in the Pathogenesis of Multiple Sclerosis: Therapeutic Perspectives" Cells 9, no. 6: 1564. https://doi.org/10.3390/cells9061564
APA StyleBiernacki, T., Sandi, D., Bencsik, K., & Vécsei, L. (2020). Kynurenines in the Pathogenesis of Multiple Sclerosis: Therapeutic Perspectives. Cells, 9(6), 1564. https://doi.org/10.3390/cells9061564