Elsevier

Journal of Psychiatric Research

Volume 68, September 2015, Pages 316-328
Journal of Psychiatric Research

Review
Kynurenine pathway dysfunction in the pathophysiology and treatment of depression: Evidences from animal and human studies

https://doi.org/10.1016/j.jpsychires.2015.05.007Get rights and content

Highlights

  • Dysregulation in the kynurenine pathway is evident in depression.

  • Kynurenine pathway is associated the severity of depressive symptoms.

  • Kynurenine pathway could represent the link between MDD and medical conditions.

  • Kynurenine modulators could be new therapeutic targets to MDD.

Abstract

Treatment-resistant depression affects up to 20% of individuals suffering from major depressive disorder (MDD). The medications currently available to treat depression, including serotonin re-uptake inhibitors (SSRIs), monoamine oxidase inhibitors (MAOIs) and tricyclic antidepressants (TCAs), fail to produce adequate remission of depressive symptoms for a large number of patients. The monoamine hypothesis upon which these medications are predicated should be expanded and revised as research elucidates alternative mechanisms of depression and effective methods to treat the underlying pathologic consequences. Research into the role of tryptophan degradation and the kynurenine pathway in the setting of inflammation has brought new insight into potential etiologies of MDD. Further investigation into the connection between inflammatory mediators, tryptophan degradation, and MDD can provide many targets for novel antidepressant therapies. Thus, this review will highlight the role of the kynurenine pathway in the pathophysiology of depression, as well as a novel therapeutic target to classic and new modulators to treat depression based on findings from preclinical and clinical studies.

Introduction

Mental illness is a pervasive category of disorders that account for a larger proportion of disability in developed countries than any other illness, including cancer and heart disease (Reeves et al., 2011). Depressive disorders are also a global issue and the leading cause of burden and disability worldwide, according to the 2010 Global Burden of Disease of the World Health Organization (Ferrari et al., 2013). Major Depressive Disorder (MDD) is a significant public health issue within the United States that affects roughly 3% of adults, or approximately 9 million people (Reeves et al., 2011). The economic burden of MDD in the United States was estimated to be $210 billion in 2010, an increase in over 20% since 2005 (Greenberg et al., 2015). The burden this places on the patients, health care system, and economy as a whole indicates the importance of efficacious treatment of mental illnesses and continued research into the biological etiologies of these complex conditions (Table 1, Table 2).

The underlying cause of depression has been difficult to elucidate due to the heterogeneous nature of the disease and is based on cluster of symptoms derived from different etiologies. The monoamine deficiency hypothesis has historically been used to explain how depressive symptoms arise from insufficient levels of monoamine neurotransmitters (Delgado, 2006, Schildkraut, 1965). The serotonergic hypothesis was later developed by Van Praag and Korf (1971), followed by the dopaminergic hypothesis of Willner et al. (1990). However, as evidenced by the latent response to antidepressant medications, specifically reuptake inhibitors, and the prevalence of treatment-resistant depression, it is necessary to continue researching alternative treatment methodologies (Trivedi et al., 2008).

Early generations of antidepressants were the monoamine oxidase inhibitors (MOAIs) and the tricyclic antidepressants (TCAs). The mechanism of action of MOAIs is irreversible inhibition of monoamine oxidase, the enzyme responsible for degradation of serotonin, norepinephrine, and dopamine, thereby increasing synaptic concentrations of these neurotransmitters. TCAs act by decreasing reuptake of serotonin and norepinephrine in the presynaptic neurons, effectively increasing their synaptic concentration (DeBattista, 2012). While MAOIs and TCAs do effectively decrease symptoms of depression in a subset of individuals, they are limited by their considerable side effects and are no longer prescribed as first line treatments for depression (Elhwuegi, 2004).

Current first line antidepressant drugs are selective serotonin reuptake inhibitors (SSRIs) and serotonin and norepinephrine reuptake inhibitors (SNRIs). The selectivity of these drugs has resulted in a treatment option that is better tolerated with fewer side effects, as compared to the MOAIs and TCAs (DeBattista, 2012). However, despite improvements in receptor targeting, SSRIs are no more efficacious than the older TCAs at relieving symptoms of depression (Millan, 2006). Improvements in the side effect profile may increase compliance with treatment programs, but increased efficacy is the ultimate goal for successful pharmacologic management of depression.

Adjuvant therapy with atypical antipsychotics, such as quetiapine, aripiprazole, and olazapine, has been shown to augment the therapeutic effects of SSRIs and has been approved for use in treatment-resistant depression (Bobo and Shelton, 2010, Connolly and Thase, 2011). The detrimental health effects of prolonged atypical antipsychotic use are considered a significant hindrance to the long-term use of this combination, despite the potential benefits. Side effects included significant weight gain, dyslipidemia, and altered glucose metabolism (Davey et al., 2012).

Another important consideration regarding currently available antidepressant treatments is the slow onset of action before clinical improvement is attained. Often, SSRIs and SNRIs require weeks to months of treatment before patients report a diminishment of symptoms (O'Leary et al., 2014). There is also a higher risk of suicide and other deliberate acts of self-harm during the first month of treatment with antidepressants. It has been suggested this is due to an improvement in physical energy that precedes improvements in depressive mood and negative thoughts (Conwell and Heisel, 2012). Identifying faster acting antidepressants is clearly crucial to increasing compliance and decreasing the harmful effects of delayed treatment.

Similarly, related to insufficient therapeutic response of the currently available antidepressants, at least 20% of depressed patients are treatment-resistant—defined as nonresponse to two different pharmacologic classes taken at optimal dose and for sufficient period of time (Berlim and Turecki, 2007). Additionally, approximately 50% of those who are diagnosed with MDD will experience a recurrent or chronic course of the illness (Crown et al., 2002). Major depression is a complex disorder in which gene–environment interactions affects many areas of the body; it stands to reason that manipulating one molecule or neurotransmitter may not effectively cure the disease (Myint, 2012). The insufficient therapeutic effect of the currently available antidepressants has moved the field to focus on alternative mechanisms beyond the monoamine hypothesis. Indeed, over the past two decades there has been a shift from the monoamine hypothesis to pathways involving neuroplasticity impairment. Since patients with autoimmune and inflammatory disorders such as diabetes and fibromyalgia often present with depressive symptoms, it has been proposed that depression may be linked to inflammation (McInnis et al., 2014). Indeed, patients with depression had an increase in serum levels of proinflammatory cytokines, such as interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) (O'Brien et al., 2007). Such pro-inflammatory state has been documented in depressed adolescents as well compared to healthy non-depressed adolescents, suggesting that it may play a role early in the course of illness, suggesting that increased inflammation in MDD is not due to chronicity effects (Gabbay et al., 2010b).

The kynurenine pathway (KP) has been hypothesized to play a key role in processes linking peripheral inflammation and CNS alterations by i) reducing tryptophan availability, and ii) production of oxygen radicals and highly potent neurotoxins (Hochstrasser et al., 2011). Thus, tryptophan degradation and its role in the availability of serotonin have brought attention to the kynurenine pathway as a potential target for future research into alternative treatments for depression.

About 99% of tryptophan (TRP) is metabolized by tryptophan 2,3-dioxygenase (TDO) into kynurenine (KYN) in the liver. However, during active inflammation, indoleamine 2,3 –dioxygenase (IDO) is activated in extrahepatic tissues to convert TRP to KYN (Leklem, 1971). Kynurenine by itself in not neuroactive but instead is compartmentally hydroxylated by kynurenine-3-monooxygenase (KMO) into 3-hydroxykynurenine (OHK). Further cleavage of OHK by kynureninase yields 3-hydroxyanthranilic acid (3-HAA). After production of 3-HAA, there are two possible degradation arms: one proceeds with complete oxidation of 3-HAA to form adenosine triphosphate (ATP) and a small quantity of picolinic acid (PIC), while the other pathway produces quinolinic acid (QUIN) which is eventually degraded into nicotinamide adenine dinucleotide (NAD) (Leklem, 1971). Conversely, an alternative pathway is the conversion of KYN to kynurenic acid (KYNA)—a glutamate and 17-nicotinic acetylcholine receptor antagonist—by kynurenine aminotransferases (KATs) (Lopresti et al., 2014). Fig. 1 portrays the tryptophan degradation pathway.

The KYN pathway also plays a role in the metabolism of glucose. The ATP and HAA formed from this pathway activate glycolysis (Quagliariello et al., 1964), through which glycogen is stored in the cells to be utilized in case of stress or glucose need. QUIN has also been shown to inhibit gluconeogenesis (Lardy, 1971). It appears that under normal physiological conditions in the brain, the KYN pathway serves mainly for glycogen storage and the production of small amounts of NAD required for ATP synthesis in the central nervous system (Myint, 2012).

TRP depletion is therefore the result of enhanced tryptophan catabolism by TDO in the liver (Takikawa, 2005) and IDO in the lungs, placenta, blood, and brain (Heyes et al., 1993, Mellor and Munn, 1999). Furthermore, serotonin is degraded by IDO into formyl-5-hydroxykynuramine, in addition to the degradation by monoamine oxidase (Pertz and Back, 1988). The enhanced degradation of tryptophan towards kynurenine and away from production of serotonin has been termed the “kynurenine shunt” (Lapin and Oxenkrug, 1969, Mangoni, 1974).

The tryptophan degradation pathway and the kynurenine shunt have been connected to a number of psychiatric conditions, suggesting that this biochemical process may have far reaching implications. Patients with BD have shown decreased neuroprotective kynurenine metabolites in their hippocampus and amygdala when compared to control patients (Savitz et al., 2015a). Schizophrenic patients were also identified as having an imbalance between the neuroprotective and neurotoxic metabolites of tryptophan degradation (Kegel et al., 2014). MDD is often associated with a systemic pro-inflammatory state that can be tied to increasing levels of IDO activity in the peripheral tissue and the brain (Heyes et al., 1993). This review will present current research that identifies the kynurenine pathway as a prevalent component of the pathophysiology of depression from studies with animal models and human. In addition, this review will highlight studies that focus on treatment targets associated with the kynurenine pathway and antidepressant responses.

Section snippets

Search strategy

For this narrative review, the PubMed/MEDLINE database was searched with the following Boolean terms: “kynurenine”[Mesh] OR “kynureninase”[Supplementary Concept] OR “kynurenine pathway” OR “indolamine-2,3-dioxygenase” OR “tryptophan 2,3-dioxygenase” OR “kynurenic acid” OR “quinolinic acid” OR “anthranilic acid” OR “3 –Hydroxykynurenine” AND “Depression”[Mesh] OR “Depressive Disorder”[Mesh] OR “Depressive Disorder, Major”[Mesh] OR “Depression”; through March 3rd, 2015.

Observational and

Evidence from human studies: central effects

Recent studies have shown that the kynurenine pathway (KP) plays an important role in depression (Fig. 2) (Myint et al., 2007, Myint et al., 2012). Positive correlations were drawn between KYN, QUIN, KYNA and specific pro-inflammatory immunological variables in the cerebrospinal fluid (CSF), and depressive symptoms in patients with hepatitis on IFN-α treatment (Raison et al., 2009). The pro-inflammatory status of patients with major depression has been associated with increases in

Conclusion

Dysregulation in the kynurenine pathway is evident in depression and has been reported by both animal and human studies. In fact, the kynurenine pathway is associated with not only the diagnosis of MDD but also the severity of depressive symptoms. However, the pathophysiologic mechanisms involved in kynurenine pathway dysfunctions are still not fully understood. Results from human studies are not always reproducible and are sometimes contradictory; the findings are dependent on the stage of

Role of funding source

None.

Contributors

GZR, KJ and SET contributed with writing manuscript. AFC contributes to search strategy. GZR and SET did the figures. GZR and KJ prepared the tables. JQ, AFC, VM and GZR contributed with study design and manuscript revision.

Conflict of interest

None.

Acknowledgments

Laboratory of Neurosciences (Brazil) is a center within the National Institute for Molecular Medicine (INCT-MM) and a member of the Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC). This research was supported by grants from CNPq (JQ, AFC, KJ, and GZR), FAPESC (JQ), Instituto Cérebro e Mente, UNESC (JQ), and LÓréal/UNESCO/ABC Brazil Fellowship for Women in Science 2011 (GZR). JQ, KJ, and AFC are CNPq Research Fellows. Center for Translational Psychiatry (USA) is funded

References (133)

  • A. Dairam et al.

    Non-steroidal anti-inflammatory agents, tolmetin and sulindac, inhibit liver tryptophan 2,3-dioxygenase activityand alter brain neurotransmitter levels

    Life Sci

    (2006)
  • A.L. Dinel et al.

    Lipopolysaccharide-induced brain activation of the indoleamine 2,3-dioxygenase and depressive-like behaviorare impaired in a mouse model of metabolic syndrome

    Psychoneuroendocrinology

    (2014)
  • A.S. Elgarf et al.

    Lipopolysaccharide repeated challenge followed by chronic mild stress protocol introduces a combined model ofdepression in rats: reversibility by imipramine and pentoxifylline

    Pharmacol Biochem Behav

    (2014)
  • A.S. Elhwuegi

    Central monoamines and their role in major depression

    Prog Neuropsychopharmacol Biol Psychiatry

    (2004)
  • M. Elovainio et al.

    Moderating effect of indoleamine 2,3-dioxygenase (IDO) activation in the association between depressivesymptoms and carotid atherosclerosis: evidence from the Young Finns study

    J Affect Disord

    (2011)
  • C.M. Forrest et al.

    Prenatal inhibition of the tryptophan-kynurenine pathway alters synaptic plasticity and protein expression in the rat hippocampus

    Brain Res

    (2013)
  • C.M. Forrest et al.

    Changes in synaptic transmission and protein expression in the brains of adult offspring after prenatal inhibition of the kynurenine pathway

    Neuroscience

    (2013)
  • M. Franklin et al.

    Sub-chronic dietary tryptophan depletion an animal model of depression with improved face and good constructvalidity

    J Psychiatr Res

    (2012)
  • V. Gabbay et al.

    The kynurenine pathway in adolescent depression: preliminary findings from a proton MR spectroscopy study

    Prog Neuropsychopharmacol Biol Psychiatry

    (2010)
  • A. Galvão-de Almeida et al.

    Lack of association of indoleamine 2,3-dioxygenase polymorphisms with interferon-alpha-related depression inhepatitis C

    Brain Behav Immun

    (2011)
  • S.M. Gibney et al.

    Poly I: C-induced activation of the immune response is accompanied by depression and anxiety-like behaviours,kynurenine pathway activation and reduced BDNF expression

    Brain Behav Immun

    (2013)
  • C.J. Henry et al.

    Peripheral lipopolysaccharide (lps) challenge promotes microglial hyperactivity in aged mice that is associated with exaggerated induction of both pro-inflammatory IL-1beta and anti-inflammatory IL-10 cytokines

    Brain Behav Immun

    (2009)
  • T. Hochstrasser et al.

    Inflammatory stimuli reduce survival of serotonergic neurons and induce neuronal expression of indoleamine 2,3-dioxygenase in rat dorsal raphe nucleus organotypic brain slices

    Neuroscience

    (2011)
  • M.M. Hughes et al.

    Tryptophan depletion in depressed patients occurs independent of kynurenine pathway activation

    Brain Behav Immun

    (2012)
  • Z.M. Ignácio et al.

    Epigenetic and epistatic interactions between serotonin transporter and brain-derived neurotrophic factor genetic polymorphism: insights in depression

    Neuroscience

    (2014)
  • K.W. Kelley et al.

    Aging leads to prolonged duration of inflammation-induced depression-like behavior caused by BacillusCalmette-Guérin

    Brain Behav Immun

    (2013)
  • J.P. Kim et al.

    Quinolinate neurotoxicity in cortical cell culture

    Neuroscience

    (1987)
  • I.P. Lapin et al.

    Intensification of the central serotoninergic processes as a possible determinant of the thymoleptic effect

    Lancet

    (1969)
  • H.A. Lardy

    The role of tryptophan metabolites in regulating gluconeogenesis

    Am J Clin Nutr

    (1971)
  • A. Laugeray et al.

    Peripheral and cerebral metabolic abnormalities of the tryptophan-kynurenine pathway in a murine model of major depression

    Behav Brain Res

    (2010)
  • A. Laugeray et al.

    Evidence for a key role of the peripheral kynurenine pathway in the modulation of anxiety- and depression-likebehaviours in mice: focus on individual differences

    Pharmacol Biochem Behav

    (2011)
  • M.A. Lawson et al.

    Intracerebroventricular administration of HIV-1 Tat induces brain cytokine and indoleamine 2,3-dioxygenaseexpression: a possible mechanism for AIDS comorbid depression

    Brain Behav Immun

    (2011)
  • J.E. Leklem

    Quantitative aspects of tryptophan metabolism in humans and other species: a review

    Am J Clin Nutr

    (1971)
  • A.L. Lopresti et al.

    A review of peripheral biomarkers in major depression: the potential of inflammatory and oxidative stress biomarkers

    Prog Neuropsychopharmacol Biol Psychiatry

    (2014)
  • A.L. Mellor et al.

    Tryptophan catabolism and t-cell tolerance: immunosuppression by starvation?

    Immunol Today

    (1999)
  • M.J. Millan

    Multi-target strategies for the improved treatment of depressive states: conceptual foundations and neuronal substrates, drug discovery and therapeutic application

    Pharmacol Ther

    (2006)
  • C.L. Miller et al.

    Upregulation of the initiating step of the kynurenine pathway in postmortem anterior cingulated cortex from individuals with schizophrenia and bipolar disorder

    Brain Res

    (2006)
  • R. Molteni et al.

    Modulation of the inflammatory response in rats chronically treated with the antidepressant agomelatine

    Eur Neuropsychopharmacol

    (2013)
  • M. Moreau et al.

    Inoculation of Bacillus Calmette-Guerin to mice induces an acute episode of sickness behavior followed bychronic depressive-like behavior

    Brain Behav Immun

    (2008)
  • A.M. Myint et al.

    The role of the kynurenine metabolism in major depression: evidence of impaired neuroprotection

    J Affect Disord

    (2007)
  • A.M. Myint et al.

    Tryptophan metabolism and immunogenetics in major depression: a role for interferon-γ gene

    Brain Behav Immun

    (2013)
  • S.M. O'Brien et al.

    Plasma cytokine profiles in depressed patients who fail to respond to selective serotonin reuptake inhibitor therapy

    J Psychiatr Res

    (2007)
  • M.N. Perkins et al.

    An iontophoretic investigation of the actions of convulsant kynurenines and their interaction with the endogenous excitant quinolinic acid

    Brain Res

    (1982)
  • M.M. Pertl et al.

    C-reactive protein predicts fatigue independently of depression in breast cancer patients prior to chemotherapy

    Brain Behav Immun

    (2013)
  • C.G. Abdallah et al.

    Ketamine as a promising prototype for a new generation of rapid-acting antidepressants

    Ann N Y Acad Sci

    (2015)
  • A. Baranyi et al.

    A biopsychosocial model of interferon-alpha-induced depression in patients with chronic hepatitis C infection

    Psychother Psychosom

    (2013)
  • M.T. Berlim et al.

    Definition, assessment, and staging of treatment-resistant refractory major depression: a review of current concepts and methods

    Can J Psychiatry

    (2007)
  • O. Berton et al.

    New approaches to antidepressant drug discovery: beyond monoamines

    Nat Rev Neurosci

    (2006)
  • W.V. Bobo et al.

    Efficacy, safety and tolerability of Symbyax for acute-phase management of treatment-resistant depression

    Expert Rev Neurother

    (2010)
  • M. Busse 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)
  • Cited by (0)

    View full text