Interactions of Tryptophan and Its Catabolites With Melatonin and the Alpha 7 Nicotinic Receptor in Central Nervous System and Psychiatric Disorders: Role of the Aryl Hydrocarbon Receptor and Direct Mitochondria Regulation

George Anderson, Michael Maes, George Anderson, Michael Maes

Abstract

Recent work indicates an intimate interaction of the tryptophan catabolite (TRYCAT) pathways with the melatonergic pathways, primarily via TRYCAT pathway induction taking tryptophan away from the production of serotonin, which is a necessary precursor for the melatonergic pathways. The alpha 7 nicotinic receptor may be significantly modulated by this interaction, given its inactivation by the TRYCAT, kynurenic acid, and its induction by melatonin. Similarly, the aryl hydrocarbon receptor is activated by both kynurenic acid and kynurenine, leading to CYP1A2 and melatonin metabolism, whereas melatonin may act to inhibit the aryl hydrocarbon receptor. These 2 receptors and pathways may therefore be intimately linked, with relevance to a host of intracellular processes of clinical relevance. In this article, these interactions are reviewed. Interestingly, mitochondria may be a site for direct interactions of these pathways and receptors, suggesting that their differential induction may not only be modulating neuronal, glia, and immune cell processes and activity but also be directly acting to regulate mitochondrial functioning. This is likely to have significant consequences as to how an array of diverse central nervous system and psychiatric conditions are conceptualized and treated.

Keywords: CNS; alpha 7 nicotinic; aryl hydrocarbon; glia; gut-brain axis; melatonin; mitochondria; psychiatry; sirtuins; tryptophan catabolites.

Conflict of interest statement

DECLARATION OF CONFLICTING INTERESTS: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article. Disclosure This manuscript has been read and approved by all authors. This paper is unique and is not under consideration by any other publication and has not been published elsewhere. The authors and peer reviewers of this paper report no conflicts of interest. The authors confirm that they have permission to reproduce any copyrighted material.

Figures

Figure 1.
Figure 1.
The major components of the kynurenine and serotonergic/melatonergic pathways and their interactions. The initial conversion of tryptophan by tryptophan hydroxylase leads to the synthesis of serotonin and melatonin. When stress and pro-inflammatory cytokines are increased, inducing TDO and IDO, respectively, tryptophan is driven down the kynurenine pathway leading to the increased synthesis of tryptophan catabolites. IDO indicates indoleamine 2,3-dioxygenase; TDO, tryptophan 2,3-dioxygenease.
Figure 2.
Figure 2.
The interactions of the AhR and a7nAChR with the TRYCAT and melatonergic pathways. Pro-inflammatory cytokines and chronic stress increase IDO and TDO, respectively, leading to TRYCAT induction, with differential effects on mitochondrial functioning. TRYCAT activation and stress-induced MAO, by decreasing serotonin availability, decrease melatonergic pathway activation. Increased KYNA may also compete with melatonin via the opposing regulation of the a7nAChR. AhR activation, via IDO induction, may positively feedback on TRYCAT synthesis, as well as decrease the availability of melatonin by increasing its metabolism. Some data indicate that the AhR decreases sirtuins, which may also be mediated by decreased melatonin. Both 3-OHK and 3-OH-ANA can modulate mitochondrial functioning. 3-OH-ANA indicates 3-hydroxyanthranillic acid; 3-OHK, 3-hydroxykynurenine; a7nAChR, alpha 7 nicotinic receptor; AhR, aryl hydrocarbon receptor; IDO, indoleamine 2,3-dioxygenase; KYNA, kynurenic acid; MAO, monoamine oxidase; TDO, tryptophan 2,3-dioxygenase; TRYCATs, tryptophan catabolites.

References

    1. Lim CK, Fernández-Gomez FJ, Braidy N, et al. Involvement of the kynurenine pathway in the pathogenesis of Parkinson’s disease [published online ahead of print April 9, 2016]. Prog Neurobiol. doi:10.1016/j.pneurobio.2015.12.009.
    1. Anderson G, Ojala J. Alzheimer’s and seizures: interleukin-18, indoleamine 2,3-dioxygenase and quinolinic acid. Int J Tryptophan Res. 2010;3:169–173.
    1. Anderson G, Maes M. Oxidative/nitrosative stress and immuno-inflammatory pathways in depression: treatment implications. Curr Pharm Des. 2014;20: 3812–3847.
    1. de Bie J, Lim CK, Guillemin GJ. Kynurenines, gender and neuroinflammation; showcase schizophrenia. Neurotox Res. 2016;30:285–294.
    1. Anderson G, Kubera M, Duda W, Lasoń W, Berk M, Maes M. Increased IL-6 trans-signaling in depression: focus on the tryptophan catabolite pathway, melatonin and neuroprogression. Pharmacol Rep. 2013;65:1647–1654.
    1. Anderson G, Maes M. Local melatonin regulates inflammation resolution: a common factor in neurodegenerative, psychiatric and systemic inflammatory disorders. CNS Neurol Disord Drug Targets. 2014;13:817–827.
    1. Tan DX, Manchester LC, Liu X, Rosales-Corral SA, Acuna-Castroviejo D, Reiter RJ. Mitochondria and chloroplasts as the original sites of melatonin synthesis: a hypothesis related to melatonin’s primary function and evolution in eukaryotes. J Pineal Res. 2013;54:127–138.
    1. He C, Wang J, Zhang Z, et al. Mitochondria synthesize melatonin to ameliorate its function and improve mice oocyte’s quality under in vitro conditions. Int J Mol Sci. 2016;17:939.
    1. Markus RP, Silva CL, Franco DG, Barbosa EM, Jr, Ferreira ZS. Is modulation of nicotinic acetylcholine receptors by melatonin relevant for therapy with cholinergic drugs? Pharmacol Ther. 2010;126:251–262.
    1. Sommansson A, Nylander O, Sjöblom M. Melatonin decreases duodenal epithelial paracellular permeability via a nicotinic receptor-dependent pathway in rats in vivo. J Pineal Res. 2013;54:282–291.
    1. Stone TW, Darlington LG. Endogenous kynurenines as targets for drug discovery and development. Nat Rev Drug Discov. 2002;1:609–620.
    1. Ren S, Correia MA. Heme: a regulator of rat hepatic tryptophan 2,3-dioxygenase? Arch Biochem Biophys. 2000;377:195–203.
    1. Foster AC, Collins JF, Schwarcz R. On the excitotoxic properties of quinolinic acid, 2,3-piperidine dicarboxylic acids and structurally related compounds. Neuropharmacology. 1983;22:1331–1342.
    1. Stone TW, Stoy N, Darlington LG. An expanding range of targets for kynurenine metabolites of tryptophan. Trends Pharmacol Sci. 2013;34:136–143.
    1. Schwarz MJ, Guillemin GJ, Teipel SJ, Buerger K, Hampel H. Increased 3-hydroxykynurenine serum concentrations differentiate Alzheimer’s disease patients from controls. Eur Arch Psychiatry Clin Neurosci. 2013;263:345–352.
    1. Young KD, Drevets WC, Dantzer R, Teague TK, Bodurka J, Savitz J. Kynurenine pathway metabolites are associated with hippocampal activity during autobiographical memory recall in patients with depression. Brain Behav Immun. 2016;56:335–342.
    1. Anderson G, Berk M, Maes M. Biological phenotypes underpin the physio-somatic symptoms of somatization, depression, and chronic fatigue syndrome. Acta Psychiatr Scand. 2014;129:83–97.
    1. Anderson G, Jacob A, Bellivier F, Geoffroy PA. Bipolar disorder: the role of the kynurenine and melatonergic pathways. Curr Pharm Des. 2016;22:987–1012.
    1. Maes M, Anderson G. Overlapping the tryptophan catabolite (TRYCAT) and melatoninergic pathways in Alzheimer’s disease. Curr Pharm Des. 2016;22:1074–1085.
    1. Anderson G, Rodriguez M. Multiple sclerosis: the role of melatonin and N-acetylserotonin. Mult Scler Relat Disord. 2015;4:112–123.
    1. 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.
    1. Muxel SM, Pires-Lapa MA, Monteiro AW, et al. NF-κB drives the synthesis of melatonin in RAW 264.7 macrophages by inducing the transcription of the arylalkylamine-N-acetyltransferase (AA-NAT) gene. PLoS ONE. 2012;7:e52010.
    1. Hardeland R, Cardinali DP, Srinivasan V, Spence DW, Brown GM, Pandi-Perumal SR. Melatonin—a pleiotropic, orchestrating regulator molecule. Prog Neurobiol. 2011;93:350–384.
    1. Anderson G, Maes M. The gut-brain axis: the role of melatonin in linking psychiatric, inflammatory and neurodegenerative conditions. Adv Integr Med. 2015;2:31–37.
    1. Jang SW, Liu X, Pradoldej S, et al. N-acetylserotonin activates TrkB receptor in a circadian rhythm. Proc Natl Acad Sci U S A. 2010;107:3876–3881.
    1. Souza-Teodoro LH, Dargenio-Garcia L, Petrilli-Lapa CL, et al. Adenosine triphosphate inhibits melatonin synthesis in the rat pineal gland. J Pineal Res. 2016;60:242–249.
    1. Beischlag TV, Anderson G, Mazzoccoli G. Glioma: tryptophan catabolite and melatoninergic pathways link microRNA, 14-3-3, chromosome 4q35, epigenetic processes and other glioma biochemical changes. Curr Pharm Des. 2016;22:1033–1048.
    1. Dubocovich ML, Delagrange P, Krause DN, Sugden D, Cardinali DP, Olcese J. International Union of Basic and Clinical Pharmacology. LXXV. Nomenclature, classification, and pharmacology of G protein-coupled melatonin receptors. Pharmacol Rev. 2010;62:343–380.
    1. Jockers R, Delagrange P, Dubocovich ML, et al. Update on melatonin receptors: IUPHAR review 20. Br J Pharmacol. 2016;173:2702–2725.
    1. Drolle E, Kučerka N, Hoopes MI, et al. Effect of melatonin and cholesterol on the structure of DOPC and DPPC membranes. Biochim Biophys Acta. 2013;1828:2247–2254.
    1. Obsil T, Ghirlando R, Klein DC, Ganguly S, Dyda F. Crystal structure of the 14-3-3zeta:serotonin N-acetyltransferase complex. A role for scaffolding in enzyme regulation. Cell. 2001;105:257–267.
    1. Choi MR, Hwang S, Park GM, et al. Effect of fluoxetine on the expression of tryptophan hydroxylase and 14-3-3 protein in the dorsal raphe nucleus and hippocampus of rat. J Chem Neuroanat. 2012;43:96–102.
    1. Markus RP, Cecon E, Pires-Lapa MA. Immune-pineal axis: nuclear factor κB (NF-kB) mediates the shift in the melatonin source from pinealocytes to immune competent cells. Int J Mol Sci. 2013;14:10979–10997.
    1. Kalkman HO, Feuerbach D. Modulatory effects of α7 nAChRs on the immune system and its relevance for CNS disorders. Cell Mol Life Sci. 2016;73:2511–2530.
    1. Costantini TW, Krzyzaniak M, Cheadle GA, et al. Targeting α-7 nicotinic acetylcholine receptor in the enteric nervous system: a cholinergic agonist prevents gut barrier failure after severe burn injury. Am J Pathol. 2012;181:478–486.
    1. Yakel JL. Nicotinic ACh receptors in the hippocampal circuit; functional expression and role in synaptic plasticity. J Physiol. 2014;592:4147–4153.
    1. Sharma G, Vijayaraghavan S. Nicotinic cholinergic signaling in hippocampal astrocytes involves calcium-induced calcium release from intracellular stores. Proc Natl Acad Sci U S A. 2001;98:4148–4153.
    1. Noda M, Kobayashi AI. Nicotine inhibits activation of microglial proton currents via interactions with α7 acetylcholine receptors. J Physiol Sci. 2017;67:235–245.
    1. Peña VB, Bonini IC, Antollini SS, Kobayashi T, Barrantes FJ. Alpha 7-type acetylcholine receptor localization and its modulation by nicotine and cholesterol in vascular endothelial cells. J Cell Biochem. 2011;112:3276–3288.
    1. Anderson G, Maes M. Reconceptualizing adult neurogenesis: role for sphingosine-1-phosphate and fibroblast growth factor-1 in co-ordinating astrocyte-neuronal precursor interactions. CNS Neurol Disord Drug Targets. 2014;13:126–136.
    1. Han Z, Shen F, He Y, et al. Activation of α-7 nicotinic acetylcholine receptor reduces ischemic stroke injury through reduction of pro-inflammatory macrophages and oxidative stress. PLoS ONE. 2014;9:e105711.
    1. John D, Shelukhina I, Yanagawa Y, Deuchars J, Henderson Z. Functional alpha7 nicotinic receptors are expressed on immature granule cells of the postnatal dentate gyrus. Brain Res. 2015;1601:15–30.
    1. Anderson G, Seo M, Carvalho A, Berk M, Maes M. Gut permeability and Parkinson’s disease. Curr Pharm Des. 2016;22(40):6142–6151.
    1. Deardorff WJ, Shobassy A, Grossberg GT. Safety and clinical effects of EVP-6124 in subjects with Alzheimer’s disease currently or previously receiving an acetylcholinesterase inhibitor medication. Expert Rev Neurother. 2015;15:7–17.
    1. Marder SR. Alpha-7 nicotinic agonist improves cognition in schizophrenia. Evid Based Ment Health. 2016;19:60.
    1. Damaj MI, Meyer EM, Martin BR. The antinociceptive effects of alpha7 nicotinic agonists in an acute pain model. Neuropharmacology. 2000;39:2785–2791.
    1. Parada E, Buendia I, León R, et al. Neuroprotective effect of melatonin against ischemia is partially mediated by alpha-7 nicotinic receptor modulation and HO-1 overexpression. J Pineal Res. 2014;56:204–212.
    1. Gergalova G, Lykhmus O, Kalashnyk O, et al. Mitochondria express α7 nicotinic acetylcholine receptors to regulate Ca2+ accumulation and cytochrome c release: study on isolated mitochondria. PLoS ONE. 2012;7:e31361.
    1. Pálková L, Vondráček J, Trilecová L, et al. The aryl hydrocarbon receptor-mediated and genotoxic effects of fractionated extract of standard reference diesel exhaust particle material in pulmonary, liver and prostate cells. Toxicol In Vitro. 2015;29:438–448.
    1. Salim SY, Kaplan GG, Madsen KL. Air pollution effects on the gut microbiota: a link between exposure and inflammatory disease. Gut Microbes. 2014;5:215–219.
    1. Sánchez-Martín FJ, Fernández-Salguero PM, Merino JM. Aryl hydrocarbon receptor-dependent induction of apoptosis by 2,3,7,8-tetrachlorodibenzo-p-dioxin in cerebellar granule cells from mouse. J Neurochem. 2011;118:153–162.
    1. Anderson G, Beischlag TV, Vinciguerra M, Mazzoccoli G. The circadian clock circuitry and the AHR signaling pathway in physiology and pathology. Biochem Pharmacol. 2013;85:1405–1416.
    1. Wu WL, Adams CE, Stevens KE, Chow KH, Freedman R, Patterson PH. The interaction between maternal immune activation and alpha 7 nicotinic acetylcholine receptor in regulating behaviors in the offspring. Brain Behav Immun. 2015;46:192–202.
    1. Mohinta S, Kannan AK, Gowda K, Amin SG, Perdew GH, August A. Differential regulation of Th17 and T regulatory cell differentiation by aryl hydrocarbon receptor dependent xenobiotic response element dependent and independent pathways. Toxicol Sci. 2015;145:233–243.
    1. Vogel CF, Chang WL, Kado S, et al. Transgenic overexpression of aryl hydrocarbon receptor repressor (AhRR) and AhR-mediated induction of CYP1A1, cytokines, and acute toxicity. Environ Health Perspect. 2016;124:1071–1083.
    1. Chang TK, Chen J, Yang G, Yeung EY. Inhibition of procarcinogen-bioactivating human CYP1A1, CYP1A2 and CYP1B1 enzymes by melatonin. J Pineal Res. 2010;48:55–64.
    1. Hwang HJ, Dornbos P, Steidemann M, Dunivin TK, Rizzo M, LaPres JJ. Mitochondrial-targeted aryl hydrocarbon receptor and the impact of 2,3,7,8-tetrachlorodibenzo-p-dioxin on cellular respiration and the mitochondrial proteome. Toxicol Appl Pharmacol. 2016;304:121–132.
    1. Baran H, Staniek K, Bertignol-Spörr M, Attam M, Kronsteiner C, Kepplinger B. Effects of various kynurenine metabolites on respiratory parameters of rat brain, liver and heart mitochondria. Int J Tryptophan Res. 2016;9:17–29.
    1. Moyer BJ, Rojas IY, Kerley-Hamilton JS, et al. Inhibition of the aryl hydrocarbon receptor prevents Western diet-induced obesity. Model for AHR activation by kynurenine via oxidized-LDL, TLR2/4, TGFβ and IDO1. Toxicol Appl Pharmacol. 2016;300:13–24.
    1. Chmill S, Kadow S, Winter M, Weighardt H, Esser C. 2,3,7,8-Tetrachlorodibenzo-p-dioxin impairs stable establishment of oral tolerance in mice. Toxicol Sci. 2010;118:98–107.
    1. Roomruangwong C, Kanchanatawan B, Sirivichayakul S, et al. IgA/IgM responses to tryptophan and tryptophan catabolites (TRYCATs) are differently associated with prenatal depression, physio-somatic symptoms at the end of term and premenstrual syndrome [published online ahead of print April 1, 2016]. Mol Neurobiol. doi:10.1007/s12035-016-9877-3.
    1. Ford DE. Zeroing in on depression as a cardiovascular risk factor. Can lifting mood improve outcomes? Postgrad Med. 2003;114:6–13.
    1. Hazar N, Seddigh L, Rampisheh Z, Nojomi M. Population attributable fraction of modifiable risk factors for Alzheimer disease: a systematic review of systematic reviews. Iran J Neurol. 2016;15:164–172.
    1. Carreira VS, Fan Y, Kurita H, et al. Disruption of Ah receptor signaling during mouse development leads to abnormal cardiac structure and function in the adult. PLoS ONE. 2015;10:e0142440.
    1. Campos-Peña V, Toral-Rios D, Becerril-Pérez F, et al. Metabolic syndrome as a risk factor for Alzheimer’s disease: is Aβ a crucial factor in both pathologies? [published online ahead of print August 16, 2016]. Antioxid Redox Signal. doi:10.1089/ars.2016.6768.
    1. Chen YJ, Lin CL, Li CR, et al. Associations among integrated psychoneuroimmunological factors and metabolic syndrome. Psychoneuroendocrinology. 2016;74:342–349.
    1. Schubert KO, Föcking M, Cotter DR. Proteomic pathway analysis of the hippocampus in schizophrenia and bipolar affective disorder implicates 14-3-3 signaling, aryl hydrocarbon receptor signaling, and glucose metabolism: potential roles in GABAergic interneuron pathology. Schizophr Res. 2015;167:64–72.
    1. Foote M, Qiao H, Graham K, Wu Y, Zhou Y. Inhibition of 14-3-3 proteins leads to schizophrenia-related behavioral phenotypes and synaptic defects in mice. Biol Psychiatry. 2015;78:386–395.
    1. Stefanovic B, Spasojevic N, Jovanovic P, Jasnic N, Djordjevic J, Dronjak S. Melatonin mediated antidepressant-like effect in the hippocampus of chronic stress-induced depression rats: regulating vesicular monoamine transporter 2 and monoamine oxidase A levels. Eur Neuropsychopharmacol. 2016;26:1629–1637.
    1. Vikström Bergander L, Cai W, Klocke B, Seifert M, Pongratz I. Tryptamine serves as a proligand of the AhR transcriptional pathway whose activation is dependent of monoamine oxidases. Mol Endocrinol. 2012;26:1542–1551.
    1. He J, Hu B, Shi X, et al. Activation of the aryl hydrocarbon receptor sensitizes mice to nonalcoholic steatohepatitis by deactivating mitochondrial sirtuin deacetylase Sirt3. Mol Cell Biol. 2013;33:2047–2055.
    1. Albani D, Ateri E, Mazzuco S, et al. Modulation of human longevity by SIRT3 single nucleotide polymorphisms in the prospective study “Treviso Longeva (TRELONG).” Age (Dordr). 2014;36:469–478.

Source: PubMed

3
Subscribe