The kynurenine pathway and bipolar disorder: intersection of the monoaminergic and glutamatergic systems and immune response

Bashkim Kadriu, Cristan A Farmer, Peixiong Yuan, Lawrence T Park, Zhi-De Deng, Ruin Moaddel, Ioline D Henter, Bridget Shovestul, Elizabeth D Ballard, Cristoph Kraus, Philip W Gold, Rodrigo Machado-Vieira, Carlos A Zarate Jr, Bashkim Kadriu, Cristan A Farmer, Peixiong Yuan, Lawrence T Park, Zhi-De Deng, Ruin Moaddel, Ioline D Henter, Bridget Shovestul, Elizabeth D Ballard, Cristoph Kraus, Philip W Gold, Rodrigo Machado-Vieira, Carlos A Zarate Jr

Abstract

Dysfunction in a wide array of systems-including the immune, monoaminergic, and glutamatergic systems-is implicated in the pathophysiology of depression. One potential intersection point for these three systems is the kynurenine (KYN) pathway. This study explored the impact of the prototypic glutamatergic modulator ketamine on the endogenous KYN pathway in individuals with bipolar depression (BD), as well as the relationship between response to ketamine and depression-related behavioral and peripheral inflammatory markers. Thirty-nine participants with treatment-resistant BD (23 F, ages 18-65) received a single ketamine infusion (0.5 mg/kg) over 40 min. KYN pathway analytes-including plasma concentrations of indoleamine 2,3-dioxygenase (IDO), KYN, kynurenic acid (KynA), and quinolinic acid (QA)-were assessed at baseline (pre-infusion), 230 min, day 1, and day 3 post-ketamine. General linear models with restricted maximum likelihood estimation and robust sandwich variance estimators were implemented. A repeated effect of time was used to model the covariance of the residuals with an unstructured matrix. After controlling for age, sex, and body mass index (BMI), post-ketamine IDO levels were significantly lower than baseline at all three time points. Conversely, ketamine treatment significantly increased KYN and KynA levels at days 1 and 3 versus baseline. No change in QA levels was observed post-ketamine. A lower post-ketamine ratio of QA/KYN was observed at day 1. In addition, baseline levels of proinflammatory cytokines and behavioral measures predicted KYN pathway changes post ketamine. The results suggest that, in addition to having rapid and sustained antidepressant effects in BD participants, ketamine also impacts key components of the KYN pathway.

Trial registration: ClinicalTrials.gov NCT00088699.

Conflict of interest statement

Conflict of Interest

Dr. Zarate is listed as a co-inventor on a patent for the use of ketamine in major depression and suicidal ideation. Drs. Zarate and Moaddel are listed as co-inventors on a patent for the use of (2R,6R)-hydroxynorketamine, (S)-dehydronorketamine, and other stereoisomeric dehydro and hydroxylated metabolites of (R,S)-ketamine metabolites in the treatment of depression and neuropathic pain; and as co-inventors on a patent application for the use of (2R,6R)-hydroxynorketamine and (2S,6S)-hydroxynorketamine in the treatment of depression, anxiety, anhedonia, suicidal ideation, and post-traumatic stress disorders. They have assigned their patent rights to the U.S. government but will share a percentage of any royalties that may be received by the government. All other authors have no conflict of interest to disclose, financial or otherwise.

© 2019. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.

Figures

Figure 1.
Figure 1.
The impact of depression on the kynurenine (KYN) pathway in brain and periphery. The figure depicts KYN metabolites and their related effects on neuronal cells (inside boxes) and the enzymes that metabolize them (arrows). The impact of inflammation or stress-related conditions on key rate-limiting enzymes such as indoleamine 2,3-dioxygenase (IDO) shifts KYN metabolism towards microglial byproducts such as 3-hydroxykynurenine (3-HK) and quinolinic acid (QA). This metabolic change is associated with elevated oxidative stress (3-HK and QA) and glutamate excitotoxicity (QA) that could contribute to depressive symptoms (right panel). Conversely, during homeostasis, substantial amounts of KYN are converted to kynurenic acid (KynA), a process mediated by kynurenine aminotransferase II (KAT II) in astrocytes (left panel). At physiologic levels, KynA is an N-methyl-D-aspartate receptor (NMDAR) antagonist and contributes to the clearance of glutamate spillover in the brain. Abbreviations: TNF-α: tumor necrosis factor alpha; IFN-gamma: interferon gamma; IL-6: interleukin-6; α7nAChR: alpha-7-nicotinic acetylcholine receptor.
Figure 2.
Figure 2.
Results of mixed models: post-ketamine change in the kynurenine (KYN) pathway. Least square mean estimated scores (with standard error) are plotted by time point. Significance refers to change from baseline (−60 minutes). IDO: indoleamine 2,3-dioxygenase; KYN: kynurenine; KynA: kynurenic acid; QA: quinolinic acid. Results of the full analysis can be found in Supplementary Table S2.
Figure 3.
Figure 3.
Baseline inflammatory cytokine component scores as moderators of change in the kynurenine (KYN) pathway (Panel A), and baseline KYN levels as moderators of change in depressive symptom ratings (Panel B). Panel A: Results of a mixed model with component scores of pro-inflammatory (top) or anti-inflammatory (bottom) cytokines entered as moderators of change in KYN pathway analytes (Y-axis) at three time points post-ketamine infusion (X-axis). T-values (all df=24) for the simple slope of the moderator for change at each time point are plotted; positive values indicate that higher cytokine component scores were associated with increases in respective KYN pathway analytes, and negative values indicate that higher cytokine component scores were associated with decreases in respective KYN pathway analytes. Panel B: Results of a mixed model with baseline KYN pathway member entered as a moderator of change in depressive symptom ratings (Y-axis) post-ketamine infusion. T-values (all df=33) for the simple slope of the moderator for change at each time point are plotted; positive values indicate that higher baseline KYN pathway concentrations were associated with less improvement in depressive symptoms, and negative values indicate that higher baseline KYN pathway concentrations were associated with more improvement in depressive symptoms. In both models, age, body mass index (BMI), and type of mood stabilizer were included as covariates. Results of the full analysis can be found in Supplementary Table S5 and Supplementary Table S6. IDO: indoleamine 2,3-dioxygenase; KYN: kynurenine; KynA: kynurenic acid; QA: quinolinic acid.

References

    1. Manji HK, Quiroz JA, Payne JL, Singh J, Lopes BP, Viegas JS et al. The underlying neurobiology of bipolar disorder. World Psychiatry 2003; 2: 136–146.
    1. Haroon E, Fleischer CC, Felger JC, Chen X, Woolwine BJ, Patel T et al. Conceptual convergence: increased inflammation is associated with increased basal ganglia glutamate in patients with major depression. Mol Psychiatry 2016; 21: 1351–1357.
    1. Birner A, Platzer M, Bengesser SA, Dalkner N, Fellendorf FT, Queissner R et al. Increased breakdown of kynurenine towards its neurotoxic branch in bipolar disorder. PLoS One 2017; 12: e0172699.
    1. Savitz J, Drevets WC. Bipolar and major depressive disorder: neuroimaging the developmental-degenerative divide. Neurosci Biobehav Rev 2009; 33: 699–771.
    1. Leonard BE. Inflammation and depression: a causal or coincidental link to the pathophysiology? Acta Neuropsychiatr 2017: 1–16.
    1. Strasser B, Becker K, Fuchs D, Gostner JM. Kynurenine pathway metabolism and immune activation: Peripheral measurements in psychiatric and co-morbid conditions. Neuropharmacology 2017; 112: 286–296.
    1. Marazziti D, Baroni S, Picchetti M, Piccinni A, Silvestri S, Dell’Osso L. [New developments on the serotonin hypothesis of depression: shunt of tryptophan]. Riv Psichiatr 2013; 48: 23–34.
    1. Miller AH. Conceptual confluence: the kynurenine pathway as a common target for ketamine and the convergence of the inflammation and glutamate hypotheses of depression. Neuropsychopharmacology 2013; 38: 1607–1608.
    1. Schwarcz R, Stone TW. The kynurenine pathway and the brain: Challenges, controversies and promises. Neuropharmacology 2017; 112: 237–247.
    1. Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW. From inflammation to sickness and depression: when the immune system subjugates the brain. Nat Rev Neurosci 2008; 9: 46–56.
    1. Andre C, O’Connor JC, Kelley KW, Lestage J, Dantzer R, Castanon N. Spatio-temporal differences in the profile of murine brain expression of proinflammatory cytokines and indoleamine 2,3-dioxygenase in response to peripheral lipopolysaccharide administration. J Neuroimmunol 2008; 200: 90–99.
    1. Reus GZ, Jansen K, Titus S, Carvalho AF, Gabbay V, Quevedo J. Kynurenine pathway dysfunction in the pathophysiology and treatment of depression: Evidences from animal and human studies. J Psychiatr Res 2015; 68: 316–328.
    1. Gibney SM, Fagan EM, Waldron AM, O’Byrne J, Connor TJ, Harkin A. Inhibition of stress-induced hepatic tryptophan 2,3-dioxygenase exhibits antidepressant activity in an animal model of depressive behaviour. Int J Neuropsychopharmacol 2014; 17: 917–928.
    1. Heisler JM, O’Connor JC. Indoleamine 2,3-dioxygenase-dependent neurotoxic kynurenine metabolism mediates inflammation-induced deficit in recognition memory. Brain Behav Immun 2015; 50: 115–124.
    1. Guillemin GJ, Kerr SJ, Smythe GA, Smith DG, Kapoor V, Armati PJ et al. Kynurenine pathway metabolism in human astrocytes: a paradox for neuronal protection. J Neurochem 2001; 78: 842–853.
    1. Stone TW. Neuropharmacology of quinolinic and kynurenic acids. Pharmacol Rev 1993; 45: 309–379.
    1. 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.
    1. Potter MC, Elmer GI, Bergeron R, Albuquerque EX, Guidetti P, Wu HQ et al. Reduction of endogenous kynurenic acid formation enhances extracellular glutamate, hippocampal plasticity, and cognitive behavior. Neuropsychopharmacology 2010; 35: 1734–1742.
    1. Ganong AH, Cotman CW. Kynurenic acid and quinolinic acid act at N-methyl-D-aspartate receptors in the rat hippocampus. J Pharmacol Exp Ther 1986; 236: 293–299.
    1. Heyes MP, Saito K, Crowley JS, Davis LE, Demitrack MA, Der M et al. Quinolinic acid and kynurenine pathway metabolism in inflammatory and non-inflammatory neurological disease. Brain 1992; 115: 1249–1273.
    1. Dantzer R, O’Connor JC, Lawson MA, Kelley KW. Inflammation-associated depression: from serotonin to kynurenine. Psychoneuroendocrinology 2011; 36: 426–436.
    1. Myint AM, Kim YK, Verkerk R, Scharpe S, Steinbusch H, Leonard B. Kynurenine pathway in major depression: evidence of impaired neuroprotection. J Affect Disord 2007; 98: 143–151.
    1. Anderson G, Maes M. Bipolar disorder: role of immune-inflammatory cytokines, oxidative and nitrosative stress and tryptophan catabolites. Curr Psychiatry Rep 2015; 17: 8.
    1. Parrott JM, O’Connor JC. Kynurenine 3-monooxygenase: an influential mediator of neuropathology. Front Psychiatry 2015; 6: 116.
    1. Harrison NA, Brydon L, Walker C, Gray MA, Steptoe A, Critchley HD. Inflammation causes mood changes through alterations in subgenual cingulate activity and mesolimbic connectivity. Biol Psychiatry 2009; 66: 407–414.
    1. Savitz J The kynurenine pathway: a finger in every pie. Mol Psychiatry 2019; April 12 [epub ahead of print].
    1. Prinz M, Priller J. The role of peripheral immune cells in the CNS in steady state and disease. Nat Neurosci 2017; 20: 136–144.
    1. Rao JS, Harry GJ, Rapoport SI, Kim HW. Increased excitotoxicity and neuroinflammatory markers in postmortem frontal cortex from bipolar disorder patients. Mol Psychiatry 2010; 15: 384–392.
    1. Bay-Richter C, Linderholm KR, Lim CK, Samuelsson M, Traskman-Bendz L, Guillemin GJ et al. A role for inflammatory metabolites as modulators of the glutamate N-methyl-D-aspartate receptor in depression and suicidality. Brain Behav Immun 2015; 43: 110–117.
    1. Bryleva EY, Brundin L. Kynurenine pathway metabolites and suicidality. Neuropharmacology 2017; 112: 324–330.
    1. Erhardt S, Lim CK, Linderholm KR, Janelidze S, Lindqvist D, Samuelsson M et al. Connecting inflammation with glutamate agonism in suicidality. Neuropsychopharmacology 2013; 38: 743–752.
    1. Busse M, Busse S, Myint AM, Gos T, Dobrowolny H, Muller UJ et al. Decreased quinolinic acid in the hippocampus of depressive patients: evidence for local anti-inflammatory and neuroprotective responses? Eur Arch Psychiatry Clin Neurosci 2015; 265: 321–329.
    1. Steiner J, Walter M, Gos T, Guillemin GJ, Bernstein HG, Sarnyai Z et al. Severe depression is associated with increased microglial quinolinic acid in subregions of the anterior cingulate gyrus: evidence for an immune-modulated glutamatergic neurotransmission? J Neuroinflammation 2011; 8: 94.
    1. Zarate CA Jr., Brutsche NE, Ibrahim L, Franco-Chaves J, Diazgranados N, Cravchik A et al. Replication of ketamine’s antidepressant efficacy in bipolar depression: a randomized controlled add-on trial. Biol Psychiatry 2012; 71: 939–946.
    1. Diazgranados N, Ibrahim L, Brutsche NE, Newberg A, Kronstein P, Khalife S et al. A randomized add-on trial of an N-methyl-D-aspartate antagonist in treatment-resistant bipolar depression. Arch Gen Psychiatry 2010; 67: 793–802.
    1. Autry AE, Adachi M, Nosyreva E, Na ES, Los MF, Cheng PF et al. NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses. Nature 2011; 475: 91–95.
    1. Duman RS, Aghajanian GK, Sanacora G, Krystal JH. Synaptic plasticity and depression: new insights from stress and rapid-acting antidepressants. Nat Med 2016; 22: 238–249.
    1. Kiraly DD, Horn SR, Van Dam NT, Costi S, Schwartz J, Kim-Schulze S et al. Altered peripheral immune profiles in treatment-resistant depression: response to ketamine and prediction of treatment outcome. Transl Psychiatry 2017; 7: e1065.
    1. Kadriu B, Gold PW, Luckenbaugh DA, Lener MS, Ballard ED, Niciu MJ et al. Acute ketamine administration corrects abnormal inflammatory bone markers in major depressive disorder. Mol Psychiatry 2017.
    1. Machado-Vieira R, Gold PW, Luckenbaugh DA, Ballard ED, Richards EM, Henter ID et al. The role of adipokines in the rapid antidepressant effects of ketamine. Mol Psychiatry 2016.
    1. Walker AK, Budac DP, Bisulco S, Lee AW, Smith RA, Beenders B et al. NMDA receptor blockade by ketamine abrogates lipopolysaccharide-induced depressive-like behavior in C57BL/6J mice. Neuropsychopharmacology 2013; 38: 1609–1616.
    1. Moaddel R, Shardell M, Khadeer M, Lovett J, Kadriu B, Ravichandran S et al. Plasma metabolomic profiling of a ketamine and placebo crossover trial of major depressive disorder and healthy control subjects. Psychopharmacology (Berl) 2018; Aug 16 [epub ahead of print].
    1. Beck AT, Ward CH, Mendelson M, Mock J, Erbaugh J. An inventory for measuring depression. Arch Gen Psychiatry 1961; 4: 561–571.
    1. Hamilton M A rating scale for depression. J Neurol Neurosurg Psychiatry 1960; 23: 56–62.
    1. Montgomery SA, Asberg M. A new depression scale designed to be sensitive to change. Br J Psychiatry 1979; 134: 382–389.
    1. Snaith RP, Hamilton M, Morley S, Humayan A, Hargreaves D, Trigwell P. A scale for the assessment of hedonic tone the Snaith-Hamilton Pleasure Scale. Br J Psychiatry 1995; 167: 99–103.
    1. Ballard ED, Yarrington JS, Farmer CA, Lener MS, Kadriu B, Lally N et al. Parsing the heterogeneity of depression: an exploratory factor analysis across commonly used depression rating scales. J Affect Disord 2018; 231: 51–57.
    1. Dobos N, de Vries EF, Kema IP, Patas K, Prins M, Nijholt IM et al. The role of indoleamine 2,3-dioxygenase in a mouse model of neuroinflammation-induced depression. J Alzheimers Dis 2012; 28: 905–915.
    1. O’Connor JC, Lawson MA, Andre C, Briley EM, Szegedi SS, Lestage J et al. Induction of IDO by bacille Calmette-Guerin is responsible for development of murine depressive-like behavior. J Immunol 2009; 182: 3202–3212.
    1. Fatokun AA, Hunt NH, Ball HJ. Indoleamine 2,3-dioxygenase 2 (IDO2) and the kynurenine pathway: characteristics and potential roles in health and disease. Amino Acids 2013; 45: 1319–1329.
    1. Hughes MM, Connor TJ, Harkin A. Stress-related immune markers in depression: implications for treatment. Int J Neuropsychopharmacol 2016; 19: pii: pyw001.
    1. Wigner P, Czarny P, Galecki P, Su KP, Sliwinski T. The molecular aspects of oxidative & nitrosative stress and the tryptophan catabolites pathway (TRYCATs) as potential causes of depression. Psychiatry Res 2018; 262: 566–574.
    1. Moffett JR, Blinder KL, Venkateshan CN, Namboodiri MA. Differential effects of kynurenine and tryptophan treatment on quinolinate immunoreactivity in rat lymphoid and non-lymphoid organs. Cell Tissue Res 1998; 293: 525–534.
    1. Kita T, Morrison PF, Heyes MP, Markey SP. Effects of systemic and central nervous system localized inflammation on the contributions of metabolic precursors to the L-kynurenine and quinolinic acid pools in brain. J Neurochem 2002; 82: 258–268.
    1. Smith AK, Simon JS, Gustafson EL, Noviello S, Cubells JF, Epstein MP 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.
    1. Lawson MA, Parrott JM, McCusker RH, Dantzer R, Kelley KW, O’Connor JC. Intracerebroventricular administration of lipopolysaccharide induces indoleamine-2,3-dioxygenase-dependent depression-like behaviors. J Neuroinflammation 2013; 10: 87.
    1. Quak J, Doornbos B, Roest AM, Duivis HE, Vogelzangs N, Nolen WA et al. Does tryptophan degradation along the kynurenine pathway mediate the association between pro-inflammatory immune activity and depressive symptoms?. Psychoneuroendocrinology 2014; 45: 202–210.
    1. Ogyu K, Kubo K, Noda Y, Iwata Y, Tsugawa S, Omura Y et al. Kynurenine pathway in depression: A systematic review and meta-analysis. Neurosci Biobehav Rev 2018; 90: 16–25.
    1. Gabbay V, Klein RG, Katz Y, Mendoza S, Guttman LE, Alonso CM et al. The possible role of the kynurenine pathway in adolescent depression with melancholic features. J Child Psychol Psychiatry 2010; 51: 935–943.
    1. Krause D, Myint AM, Schuett C, Musil R, Dehning S, Cerovecki A et al. High kynurenine (a tryptophan metabolite) predicts remission in patients with major depression to add-on treatment with celecoxib. Front Psychiatry 2017; 8: 16.
    1. Eskelund A, Li Y, Budac DP, Muller HK, Gulinello M, Sanchez C et al. Drugs with antidepressant properties affect tryptophan metabolites differently in rodent models with depression-like behavior. J Neurochem 2017; 142: 118–131.
    1. Muller N, Myint AM, Schwarz MJ. The impact of neuroimmune dysregulation on neuroprotection and neurotoxicity in psychiatric disorders--relation to drug treatment. Dialogues Clin Neurosci 2009; 11: 319–332.
    1. Ongur D, Drevets WC, Price JL. Glial reduction in the subgenual prefrontal cortex in mood disorders. Proc Natl Acad Sci U S A 1998; 95: 13290–13295.
    1. Rajkowska G, Stockmeier CA. Astrocyte pathology in major depressive disorder: insights from human postmortem brain tissue. Curr Drug Targets 2013; 14: 1225–1236.
    1. de Carvalho LP, 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.
    1. Savitz J, Drevets WC, Smith CM, Victor TA, Wurfel BE, Bellgowan PS et al. Putative neuroprotective and neurotoxic kynurenine pathway metabolites are associated with hippocampal and amygdalar volumes in subjects with major depressive disorder. Neuropsychopharmacology 2015; 40: 463–471.
    1. Maes M, Leonard BE, Myint AM, Kubera M, Verkerk R. The new ‘5-HT’ hypothesis of depression: cell-mediated immune activation induces indoleamine 2,3-dioxygenase, which leads to lower plasma tryptophan and an increased synthesis of detrimental tryptophan catabolites (TRYCATs), both of which contribute to the onset of depression. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35: 702–721.
    1. Della Chiesa M, Carlomagno S, Frumento G, Balsamo M, Cantoni C, Conte R et al. The tryptophan catabolite L-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK-cell function. Blood 2006; 108: 4118–4125.
    1. Krishnan V, Nestler EJ. The molecular neurobiology of depression. Nature 2008; 455: 894–902.
    1. Veldic M, Millischer V, Port JD, Ho AM, Jia YF, Geske JR et al. Genetic variant in SLC1A2 is associated with elevated anterior cingulate cortex glutamate and lifetime history of rapid cycling. Transl Psychiatry 2019; 23: 149.
    1. Giorgini F, Huang SY, Sathyasaikumar KV, Notarangelo FM, Thomas MA, Tararina M et al. Targeted deletion of kynurenine 3-monooxygenase in mice: a new tool for studying kynurenine pathway metabolism in periphery and brain. J Biol Chem 2013; 288: 36554–36566.
    1. Amaral M, Levy C, Heyes DJ, Lafite P, Outeiro TF, Giorgini F et al. Structural basis of kynurenine 3-monooxygenase inhibition. Nature 2013; 496: 382–385.
    1. Smith JR, Jamie JF, Guillemin GJ. Kynurenine-3-monooxygenase: a review of structure, mechanism, and inhibitors. Drug Discov Today 2016; 21: 315–324.
    1. Choi M, Lee SH, Wang SE, Ko SY, Song M, Choi JS et al. Ketamine produces antidepressant-like effects through phosphorylation-dependent nuclear export of histone deacetylase 5 (HDAC5) in rats. Proc Natl Acad Sci U S A 2015; 112: 15755–15760.
    1. Duman RS. Pathophysiology of depression: the concept of synaptic plasticity. Eur Psychiatry 2002; 17 (Suppl 3): 306–310.

Source: PubMed

3
Předplatit