Next Article in Journal
Gonadotropin Cell Transduction Mechanisms
Next Article in Special Issue
Melatonin and the Brain–Heart Crosstalk in Neurocritically Ill Patients—From Molecular Action to Clinical Practice
Previous Article in Journal
Molecular Dynamics Simulation of Association Processes in Aqueous Solutions of Maleate Salts of Drug-like Compounds: The Role of Counterion
Previous Article in Special Issue
Biological Effects of Indole-3-Propionic Acid, a Gut Microbiota-Derived Metabolite, and Its Precursor Tryptophan in Mammals’ Health and Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Emerging Cross-Species Marker for Organismal Health: Tryptophan-Kynurenine Pathway

1
Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4K1, Canada
2
Department of Chemistry, Centre for Oil and Gas Research and Development (COGRAD), University of Manitoba, 586 Parker Building, 144 Dysart Rd., Winnipeg, MB R3T 2N2, Canada
3
Quesnel River Research Centre, University of Northern British Columbia, Prince George, BC V2N 4Z9, Canada
4
Environment and Climate Change Canada, National Wildlife Research Centre, Ottawa, ON K1A 0H3, Canada
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(11), 6300; https://doi.org/10.3390/ijms23116300
Submission received: 28 February 2022 / Revised: 26 May 2022 / Accepted: 30 May 2022 / Published: 4 June 2022
(This article belongs to the Special Issue Tryptophan in Nutrition and Health 2.0)

Abstract

:
Tryptophan (TRP) is an essential dietary amino acid that, unless otherwise committed to protein synthesis, undergoes metabolism via the Tryptophan-Kynurenine (TRP-KYN) pathway in vertebrate organisms. TRP and its metabolites have key roles in diverse physiological processes including cell growth and maintenance, immunity, disease states and the coordination of adaptive responses to environmental and dietary cues. Changes in TRP metabolism can alter the availability of TRP for protein and serotonin biosynthesis as well as alter levels of the immune-active KYN pathway metabolites. There is now considerable evidence which has shown that the TRP-KYN pathway can be influenced by various stressors including glucocorticoids (marker of chronic stress), infection, inflammation and oxidative stress, and environmental toxicants. While there is little known regarding the role of TRP metabolism following exposure to environmental contaminants, there is evidence of linkages between chemically induced metabolic perturbations and altered TRP enzymes and KYN metabolites. Moreover, the TRP-KYN pathway is conserved across vertebrate species and can be influenced by exposure to xenobiotics, therefore, understanding how this pathway is regulated may have broader implications for environmental and wildlife toxicology. The goal of this narrative review is to (1) identify key pathways affecting Trp-Kyn metabolism in vertebrates and (2) highlight consequences of altered tryptophan metabolism in mammals, birds, amphibians, and fish. We discuss current literature available across species, highlight gaps in the current state of knowledge, and further postulate that the kynurenine to tryptophan ratio can be used as a novel biomarker for assessing organismal and, more broadly, ecosystem health.

Graphical Abstract

1. Introduction

The aromatic amino acid L-tryptophan (TRP) is one of nine essential amino acids required by all living organisms for protein synthesis. Although bacteria, fungi and plants can synthesize TRP from phosphoenolpyruvic acid via the shikimate pathway [1], vertebrates must obtain TRP from dietary sources [2,3], such as plants and plant products (i.e., fruit, nuts, oats, chocolate) and other animals and animal products (i.e., fish, turkey, dairy). In addition to its role in protein synthesis, TRP is the precursor of the monoaminergic neurotransmitter, serotonin (5-hydroxytryptamine, 5-HT) [2,3,4,5]. 5-HT is known to regulate behaviour and adaptive responses towards environmental stress. These responses include mood (anxiety) [6,7,8], cognition [9,10], nociception, aggressiveness, appetite [11,12], and temperature homeostasis [13]. Furthermore, there is evidence that 5-HT and its derivative melatonin regulate reproductive processes in vertebrates through the hypothalamo-hypophyseal system and by direct effects on reproductive organs [14,15,16]. Although the role of TRP and its metabolites may differ among mammals, birds, and fish, all vertebrates produce 5-HT in the central nervous system [4,5,17,18,19]. More recently, peripheral 5-HT synthesis and signaling has been identified in mammals [20,21] and fish [15,22]. Peripheral 5-HT has been reported to modulate gonadal hormone secretion, immune and inflammatory responses, and metabolic homeostasis in the gut [4,23]. Although TRP is a critical precursor for serotonin synthesis, only 1–2% of dietary TRP is converted to 5-HT; approximately 95% of dietary TRP is metabolized through the tryptophan-kynurenine (TRP-KYN) pathway [3].

1.1. Tryptophan Metabolites

The kynurenine (KYN) pathway of TRP metabolism produces biologically active metabolites involved in inflammation, immune responses, and excitatory neurotransmission [3,24,25]. The first step involves the oxidation of the indole ring and is catalyzed by two enzymes that are differentially distributed: tryptophan 2,3-dioxygenase (TDO) (predominantly expressed in the liver and brain) and indoleamine 2,3-dioxygenease (IDO) (expressed in most peripheral tissues) [3,26,27] (Figure 1). These heme-containing enzymes are structurally distinct proteins that have evolved to catalyze the conversion of TRP to N-formylkynurenine. N-formylkynurenine is rapidly converted to KYN by N-formylkynurenine formamidase. At this point, KYN metabolism can follow one of two branches: (1) it can be metabolized to kynurenic acid (KYNA) and anthranilic acid (AA) via kynurenine aminotransferase (KAT) and kynureninase (KYNU), respectively, or (2) it can be converted to the neurotoxic and free-radical generator 3-hydroxykynurenine (3-HK), which is further transaminated to xanthurenic acid (XA). KYNU catalyzes the hydrolysis of 3-HK to form the immunomodulatory and free-radical generator 3-hydroxyanthranilic acid (3-HAA), which can spontaneously transform to nicotinic acid (NIC) via 3-hydroxyanthranilate 3,4-dioxygenase (3-HAAO) or be completely oxidized to carbon dioxide (CO2). Initially, 3-HAAO converts 3-HAA to the unstable intermediate 2-amino-3-carboxymuconic semialdehyde (ACMS), which spontaneously rearranges to the excitotoxin and N-methyl-D-aspartate (NMDA) receptor agonist quinolinic acid (QUIN) or is further converted to the neuroprotective picolinic acid (PIC) after enzymatic decarboxylation via 2-amino-3-carboxymuconate semialdehyde decarboxylase (ACMSD). QUIN is decarboxylated to form NIC, the NAD+ coenzyme precursor. QUIN is also the substrate for quinolinate phosphoribosyltransferase (QPRT), which initiates several metabolic steps to ultimately produce the essential cofactor, NAD+ [3,4,25].

1.2. Kynurenine Metabolites as Biomarkers of Human Disease

TRP and its metabolites have key roles in diverse physiological processes such as cell growth and maintenance (where TRP serves as a building block for proteins) and the coordination of adaptive responses to environmental and dietary cues (where TRP metabolites serve as neurotransmitters and signaling molecules). For example, many KYN metabolites are neuroactive and are considered cytoprotective (such as KYNA) or cytotoxic (such as 3-HK, 3-HAA, QUIN, and NIC) [3,28]. These effects are highly dependent on the accumulation of metabolites within specific tissues [Reviewed In: [29]]. KYNA acts as an antagonist to α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), NMDA and kainate glutamate receptors, while QUIN acts as an NMDA receptor agonist [30,31]. NAD+ is an important coenzyme in many energy metabolism pathways including glycolysis, β-oxidation, and oxidative phosphorylation [32], and picolinic acid modulates immune function and antimicrobial activity [33].
Over the past 15 years, there has been increasing interest in the role of KYN metabolites in human disease models (Table S1). Indeed, altered levels of TRP-KYN pathway metabolites have been reported in aging and sleep disorders [34,35], metabolic syndrome [23,24,36], cardiovascular disease [37,38], cancer [39], autoimmune disease [40,41,42], anxiety and depression [6,7], neurodegenerative diseases [30], as well as obesity, anorexia and bulimia nervosa, and other diseases presenting peripheral symptoms [19,42]. Changes in TRP metabolism can alter the availability of TRP for protein and 5-HT biosynthesis and dysregulate the levels of immuno- and neuro-active KYN metabolites. Recently, research has begun to assess KYN metabolites as biomarkers to many neuro- and immune-associated illnesses, including Alzheimer’s Disease [43,44,45], ALS [46], and Major Depressive Disorder [47].

1.3. Objective of Study

However, to assess KYN enzymes and metabolites as markers of altered homeostasis, it is critical to understand and appreciate the integration of TRP catabolism. KYN pathway activation, enzymatic activity, and metabolite formation and function is dependent on exogenous stimuli and endogenous ligand binding. There is now considerable evidence showing that the KYN pathway of TRP metabolism can be influenced by various stressors including glucocorticoids (GCs) [24,47,48,49,50,51], infection [52,53,54,55,56], inflammation and oxidative stress [57,58,59,60,61,62,63], and environmental contaminants [3,64,65,66].
Importantly, the TRP-KYN pathway is conserved across vertebrate species, understanding how this pathway is regulated may have broader implications for environmental and wildlife toxicology. In mammalian species and birds with monogastric systems including duck (Anatidae) and chicken (Gallus gallus), the KYN pathway is the central catabolic route for TRP’s indole ring [2,3]. As reviewed by Ball and colleagues [67], the genes of the major TRP catabolizing enzymes IDO and TDO have undergone gene duplications leading to multiple isoforms of IDO. For example, all mammals have two IDO genes (IDO1 and IDO2) via gene duplication, whereas fish and amphibians have IDO2-like genes. Interestingly, while IDO and TDO enzymes show functional convergence, TDO has a higher catalytic efficiency for TRP catabolism, and as such, its conservation across metazoans and vertebrates is clear [27]. More importantly, the expression of these enzymes differ across species by tissue/organ/cellular localization, by enzymatic characteristics, signaling properties, and by biological function following induction by distinct stimuli [27,67].
The goal of this narrative review is to identify (1) key pathways affecting TRP-KYN metabolism in vertebrates and (2) consequences of altered TRP metabolism in mammals, birds, amphibians, and fish. We provide evidence of studies linking exposure to environmental contaminants and altered TRP-KYN metabolites, which contribute to a range of adverse health outcomes. Furthermore, we postulate that alterations in TRP metabolism, KYN metabolites, and the TRP:KYN ratio may be an indication of exposure to environmental contaminants. Thus, we suggest that levels of KYN metabolites and the TRP:KYN ratio can be used as a novel integrative biomarker for assessing organismal exposure to environmental contaminants and, more broadly, ecosystem health.

2. Methods

A literature search was conducted in NCBI using MeSH on “Tryptophan” OR “Kynurenine” AND the terms: “Glucocorticoids”, “Stress, Physiological”, “Infections”, “Inflammation”, “Oxidative Stress”, “Environmental Pollution/adverse effects”, “Environmental Pollution/genetics”, “Environmental Pollution/immunology”, “Environmental Pollution/metabolism”, “Environmental Pollution/pathogenicity”, “Environmental Pollution/pathology”, “Environmental Pollution/physiology”, “Environmental Pollution/physiopathology”, and “Environmental Pollution/toxicity”. Collectively, between the years 1970–2021, a total of 1959 publications were identified. From these, we selected publications that specifically identified changes in key cellular/physiological pathways that led to alterations in TRP-KYN metabolites in vertebrates (i.e., mammals, birds, fish, amphibians), and publications that identified altered tryptophan metabolites and their physiological consequences. A total of 265 papers were selected, and of those 185 were used in this review to focus on (1) glucocorticoids and stress, (2) infection, inflammation, and oxidative stress and (3) environmental contaminants. The break down by species and by physiological processes/pathways is depicted in Figure 2.
An additional screening was completed to identify enzyme and metabolite presence across vertebrate animal kingdoms (Figure 3). Both gene detection (i.e., PCR of mRNA, RNA, and DNA) and protein levels (i.e., HPLC, LC, MS, Western Blots) were methodologically screened using literature searches and databases. Gene data was collected through PubMed and NCBI searches specified for “gene name” (i.e., IDO, TDO, TPH, KYN, KYNA etc.) AND “homo sapiens” OR for mammals [“mammal”, “rat”, “mouse”, “rabbit”] OR for fish [“fish”, “medaka”, “salmon”, “trout”, “carp”] OR for birds [“bird”, “pigeon”, “chicken”], OR for amphibians [“amphibian”, “frog”]. It is important to note that when screening for enzyme and gene names, all isoforms of IDO and KAT were identified and used as “gene name”. If genomic information was present, it was considered “detected”. For protein levels, the UNIPROT database was screened with “kingdom” (i.e., mammal, bird, fish, frog, amphibian) and “Enzyme Name”. Entries were then confirmed by identifying protein name, organism, and whether or not it was reviewed (Swiss-Prot) or unreviewed (TrEMBL, computational curation). If it was reviewed, it was considered “detected”. If it was unreviewed, a PubMed and NCBI check was conducted, using MeSH terms for “Enzyme Name”, selecting “Other Animals” and a species name if needed (e.g., Gallus gallus). If an enzyme was unreviewed, and PubMed and NCBI identified primary articles that showed protein levels of the enzyme, it was considered “detected”. If an enzyme was unreviewed and no additional information could be found, it was considered “orthologs may be present but have not been confirmed”. If no information was found, it was considered “unknown”. Metabolites could not be screened using UNIPROT, so literature data was used primarily to identify the presence across animal kingdoms.

3. Discussion

In the following sections, we discuss critical stress-inducing pathways that may play a role in driving TRP metabolism down the KYN pathway. As the TRP-KYN pathway is altered in response to glucocorticoids and chronic stress, infection, inflammation, and oxidative stress, and environmental toxicants, and these stressors are ubiquitous phenomenon across animal kingdoms, we explore the current literature available with regard to mammals, birds, fish, and amphibians.

3.1. Glucocorticoids and Chronic Stress

Glucocorticoids (GCs) are hormones of the endocrine system responsible for regulating stress responses. GCs are modulated by the hypothalamic-pituitary-adrenal (HPA) axis through the release of tropic hormones and negative feedback regulation. Although GCs exhibit circadian variation, they can be induced in response to physical, chemical, and psychological stressors. The molecular mechanism begins with the binding of GCs to the nuclear glucocorticoid receptor (GR), which functions as a transcription factor and modulates downstream gene expression. In general, increased GC signaling has anti-inflammatory and immunosuppressive activity. When responding to stress, the body mobilizes energy through sympathetic activation, which governs the “fight-or-flight” response. For this reason, the stress response sits at the interface of neuroendocrine, immune, and behavioural systems.
The degree of the stress response often depends on the duration and intensity of the stressor. Acute, immediate stress responses tend to be protective and allow an organism to adapt to a changing environment [63,68]. Conversely, chronic, prolonged stress activation can lead to the accumulation of wear-and-tear on physiological systems and result in impaired health outcomes. For this reason, it is important to manage the balance between protective and damaging stress responses. In environmental toxicology, biomarkers such as metabolic cytochrome P450 (CYP) enzymes and heat shock proteins are often used as indicators for exposure to acute stressors and environmental insults [69]. Not only are these markers sensitive to contaminants and inexpensive for use in field monitoring, they are reliable in a wide variety of organisms. While acute stress-induced GC increase in plasma is associated with increased 5-HT production through TRP hydroxylase activity, chronic stress reduces 5-HT turnover and release and favours the KYN pathway [7]. Given the role of the TRP-KYN pathway in stress responses, its metabolites and enzymes may be useful biomarkers for stress across species.
It is generally accepted that GCs increase the expression and activity of hepatic TDO in rats and mice [29,70,71,72,73]. GC signaling involves de novo synthesis of TDO at the transcriptional level, which is mediated by GC-responsive elements on the promoter region of TDO [72,73]. TDO activity can be increased through substrate activation by TRP and cofactor activation by heme, neither of which involve de novo synthesis. Chronic stress responses, which involve both HPA signaling and sympathetic activation, increase the metabolism of TRP to KYN by TDO, which reduces the availability and transport of TRP to the brain [3] for 5-HT production. While IDO expression is not directly induced by GCs, HPA tropic hormones and/or stress can increase the expression and activity of IDO. This has been shown in the hippocampus of mice treated with adrenocorticotropic hormone [74], and in the serum and frontal cortex of rats exposed to chronic mild stress [75,76]. Some studies have suggested that TRP uptake into the brain is increased with acute stress and HPA activation. For example, in rats, foot shock stress increased levels of TRP, KYN, KYNA, and 3-HK in the brain. Similarly, acute stress induced by physical restraint in mice increased TRP metabolism to KYN in the brain and plasma, along with an increase in IDO1, IDO2, and TDO2 [77]. Moreover, following exposure to a novel stressor, brain TRP and KYN levels were raised in mice with no change in the TRP:KYN ratio and a reduction in the 5-HT:TRP ratio [78]. This suggests that while brain TRP levels increase with stress, brain IDO activity may also be induced, which increases KYN levels and shifts metabolism away from the 5-HT pathway [5].
Stress-induced alterations in TRP metabolism in the brain and periphery in mammals can also be due to an interaction between the immune system and the HPA axis [78]. Exposure to stressors induces the release of proinflammatory cytokines, which further activate the HPA axis. For example, proinflammatory interleukin-1 (IL-1) is a potent stimulant for corticotropin releasing hormone (CRH) synthesis. By increasing CRH levels, GC production can be increased. Additionally, the proinflammatory cytokine interferon-gamma (IFN-γ) induces IDO expression in hypothalamic and pituitary neurons, which produce 3-HK and QUIN [78]. Induction of IDO by IFN-γ can be potentiated by dexamethasone, which has no effect when administered alone in human monocytes [79]. In mice, stimulation of the HPA axis through immune activation by IL-1 and lipopolysaccharide has been shown to increase brain TRP levels. However, since stress-induced changes in brain TRP levels occur in adrenalectomized rats and mice, this is not necessarily dependent on adrenocortical activation [78]. Similarly, Rose et al. (2020) reported increased KYN and decreased TRP levels in plasma, and decreased expression of monoamine oxidase A (MAOA) following exposure to ozone; the addition of metyrapone, a GC inhibitor, was able to reverse the effects of ozone on MAOA and kynurenine monooxygenase (KMO), but did not change the effects on TRP and KYN levels [80]. Interestingly, it has been shown that GC release is inhibited with increased KYNA levels in some areas of the central nervous system [81]. Furthermore, the enol tautomer of indole-3-pyruvic acid (IPA), another metabolite in the TRP-KYN pathway, has been reported to reduce plasma GC levels and GR activity in the hippocampus following repeated stress [81]. Taken together, these data suggest that stress plays a key role in mammalian TRP-KYN metabolism through both GC-dependent and -independent mechanisms.
There is little known regarding the molecular effects of stress on the regulation of the TRP-KYN pathway in birds. However, in avian species, the TRP-KYN metabolic pathway is associated with undesirable behavioural patterns. For example, when chickens were under socially disruptive stress due to housing changes and manual restraint, it was found that increased destructive feather-pecking behaviour was associated with decreased plasma TRP:KYN ratios. In the same study, the socially disrupted group had gained more weight. Since plasma metabolite levels can vary with food intake, and stress can either increase or decrease feeding behaviours, it is unclear whether the observed results were due to a stress response or increased food intake [82]. Similar to rats, it was found that chickens with a TRP-supplemented diet showed reduced aggressive behaviour such as feather pecking; however, it is not clear if this is due to alterations in TRP-KYN pathway metabolites or increased 5-HT production [82].
In fish, stress responses involve the release of catecholamines from chromaffin tissue in the head, kidney and adrenergic nerves, as well as cortisol from the interrenal tissue into the circulation. Chronic, prolonged stress responses can lead to metabolic dysfunction and immunosuppression, which are characteristic of HPA hyperactivity [83]. The majority of work in fish has focused on the impact of TRP-supplemented diets on stress response reduction [84,85,86,87,88,89]. For example, studies in meagre (Argyosomus regius), rainbow trout (Oncorhynchus mykiss), Sengalese soles (Solea sengalensis kaup), Atlantic salmon (Salmo salar) and Atlantic cod (Gadus morhua) species have investigated the effects of a TRP-supplemented diet and chronic stress on TRP-KYN metabolism. Chronic stress in fish fed a control diet did not affect brain levels of 5-HT or TRP concentrations, but liver TRP, KYN, and QUIN were increased; in fish given a TRP-supplemented diet, brain 5-HT and liver TRP were reduced, and hepatic KYN and QUIN were increased [84]. Similarly, Wish et al. (2022) have found increased KYN levels in the brain and liver of rainbow trout following exposure to an acute stressor [90]. Taken together, this suggests that the KYN pathway is enhanced in stressed fish, regardless of diet.
Across species, it appears that mammals, birds, and fish have similar responses to stress with respect to changes in TRP-KYN metabolism (Figure 4). Increased GC and catecholamine signaling, which are characteristic of stress responses, elevate TRP metabolism through the KYN pathway, as indicated by the levels of downstream metabolites. While it is generally accepted that this occurs through hepatic TDO induction in mammals, the molecular mechanism is not yet clear in other vertebrate species. In bird and fish species, TRP-enhanced diets have been shown to reduce stress-induced plasma GC levels and aggressive behaviours.

3.2. Infection, Inflammation and Oxidative Stress

In all species, there is an interaction between the neuroendocrine and immune systems. Both immune and endocrine cells share common receptors, and different hormones and cytokines are involved in many of the same physiological processes. TRP functions similarly in all higher vertebrates to regulate (1) the activation, proliferation, and migration of immune-surveillant cells (i.e., T- and B-lymphocytes, macrophages, and natural killer cells) and (2) the production of inflammatory signaling molecules, cytokines, nitric oxides and superoxides (Figure 5). In mammals, KYN metabolites are reported to be involved in inflammation, immune response, and excitatory neurotransmission [24]. In recent years, metabolites including KYN, KYNA, and QUIN are emerging as key targets in diseases such as diabetes, HIV [91], atherosclerosis [92], neurodegenerative diseases including schizophrenia, Alzheimer’s and Huntington’s [93,94,95], and cancer [24,96]. Primarily, these pathologies converge on inflammatory events which include target organ infiltration of circulating immune cells, activation of pro-inflammatory signaling pathways, expression of cytokines [24]/chemokines [97], and the production of reactive oxygen species (ROS) [92]. Recent studies have reported that TRP deficiencies result in immunosuppression and a significant increase in the susceptibility of humans [60], pigs, and teleost fish [98] to disease, infection, morbidity, and mortality. As such, the TRP-KYN pathway is recognized as an important player in inflammation and immune response [94,99].
Of the rate-limiting enzymes in the TRP-KYN pathway, the IDO enzymes are known to play a role in modulating infection and autoimmunity [100]. In fact, the biological role of IDO on the infiltration of circulating immune cells has been coined the TRP-depletion hypothesis; IDO suppresses microbial infection by reducing TRP availability in infected tissues [101,102]. Furthermore, IDO is highly regulated by superoxides and inflammatory mediators, where its expression is increased by IFN-γ[102,103,104,105], tumor necrosis factor-alpha (TNFα) [105,106], tumour growth factor-beta (TGF-β) [96], interleukins 1-beta, 2, 12, 18 (IL1β, IL2, IL12, and IL18) [96], and prostaglandins (PGE2) [105,107,108], as well as pathogenic infections including parasites, viruses, and bacteria. Downstream of IDO activation, the metabolite KYNA promotes monocyte extravasation and IL6 production and controls cytokine release [24,109], while the metabolite 3-HAA can induce apoptosis in T-cells through glutathione depletion [110]. In line with this, high KYN levels can increase the proliferation and migratory capacity of cancer cells and aid tumours in evading immune surveillance. In addition, KYN metabolites can then act via the aryl hydrocarbon receptor (AhR) to mediate T-cell anergy and apoptosis, proliferation of T-regulatory and T-helper(Th)17 cells, and the Th1/Th2 response [100]. Similarly, KYN metabolite KYNA has also been found to activate human, mouse, and rat GPR35—a G protein-couple receptor with orthologs found in a variety of mammals and one amphibian species (Xenopus tropicalis). While KYNA has been found to be an endogenous activator of GPR35 in humans and rats altering immune responses in inflammation, pain, cancer, cardiovascular disease and energy homeostasis [Reviewed In: [111]], its role as an activator of GPR35 in other species remains unknown.
There is increasing evidence that IDO’s role in immune function may begin in gestation. Histochemical studies of the human decidua have reported alterations in the expression of IDO and KMO throughout pregnancy. In the first trimester, IDO and KMO expression is present in stromal and glandular epithelial cells of the decidua. In both the first and second trimester placenta, they become localized to the syncytiotrophoblast, stroma, and macrophages before shifting to fetal endothelial cells and macrophages in terminal villi of term placenta [112,113,114,115]. This shift in expression suggests that the function of these enzymes may change from a role of immunosuppression at the maternal-fetal interface in early pregnancy to one associated with the regulation of fetoplacental blood flow or placental metabolism in late gestation [112]. Work done by Williams and colleagues found that 24 h following inflammation induced by intrauterine endotoxin administration, there was a significant upregulation of IDO in the placenta and fetal brain, which was associated with increased IFN-γ expression and increased in KYN, KYNA and QUIN levels. These increases occurred in parallel with decreased levels of 5-hydroxyindole acetic acid, a precursor for 5-HT [105]. Taken together, these results indicate that maternal inflammation can shunt TRP metabolism away from 5-HT and down the KYN pathway.
IFN-γ also upregulates other enzymes in the KYN pathway, including KMO, KYNU, and 3-HAAO activities [116]. For example, in adipose tissues, which have resident macrophages with high levels of KMO, the catabolism of TRP via KMO induction following immune system activation leads to the increased production of QUIN rather than KYNA, which is reported to have anti-inflammatory properties [117]. Interestingly, plasma neopterin levels, an indicator of IFN-γ activity, have been paralleled by increased levels of the TRP:KYN ratio, which is reflective of IDO activity in patients with metabolic disorders such as type 2 diabetes [118] and neurodegenerative disorders such as Huntington’s Disease [95]. In fact, in Huntington’s patients, the TRP:KYN ratio was higher and associated with elevated levels of C-reactive protein, neopterin and lipid peroxidation products. Increased plasma KYN levels and TRP:KYN ratios have been found in patients with systemic inflammatory response syndrome, sepsis and septic shock, but the biological significance and prognostic value of these findings have remained uncertain [119]. Together, TRP:KYN and plasma neopterin levels are considered systemic markers of inflammation and oxidative stress [92,95].
Similar to mammalian IFN-γ, chicken IFN-γ is a known regulator of the immune response through the regulation of proinflammatory cytokines that correlate to immune activation and antiviral status. In fact, studies have previously shown that immunocompromised chickens have downregulated IFN-γ [120], which resulted in higher mortality following viral infection than normal chickens. Yuk and colleagues (2016) showed that siRNA suppression of IFN-γ in chicken embryo fibroblasts results in increased replication of viral genes during infection [121]. Taken together, these studies suggest that the immune state of chickens and other avian species are mediated by interferons [122,123]. Interestingly, Emadi and colleagues (2010) have shown that supplementing broiler chicken feed with double the National Research Council’s recommended levels of TRP enhanced the IFN-α, IFN-γ, and immunoglobulin G response to infection [124]. Taken together, these studies show that TRP may also be playing a vital role in the immunity of avian species.
In fish, TRP plays a critical role in macrophage and lymphocyte function. When faced with infection, TRP and IDO levels are related to the induction of anti-inflammatory signaling molecules, the main effectors being T-cells (Reviewed in: [125,126]). Similar to the other vertebrates discussed, interferons that regulate IDO play analogous antiviral roles in teleost fish (Reviewed in: [127,128]). Notably, fish interferons possess the same exon/intron structure as mammalian IFN-γ [127], also playing a similar critical role in adaptive cell-mediated immune responses produced by Th1 and cytotoxic T-lymphocytes [127,129]. Interestingly, TRP supplementation has not shown improvement in immune status in European seabass (Dicentrarchus labrax) [130] or Persian sturgeon (Acipenser persicus) [131]. Furthermore, Machado et al. (2015) have shown that following TRP supplementation and subsequent increases in cortisol, European seabass had decreased monocyte/macrophage activation in response to infection. The decreased levels of lymphocytes and monocytes/macrophages were accompanied with decreased mRNA expression of proinflammatory cytokines (IL1β, IL8 and TGFβ), diminished IFN-γ, and lowered levels of superoxide dismutase (SOD), an enzyme involved in antioxidant defence [130]. Conversely, in mice [132] and broiler chickens [124], dietary TRP supplementation was able to alleviate inflammatory responses by attenuating the migration of inflammatory cells. While an interesting difference amongst vertebrates, it has been suggested that these differences may be a result of teleost fish requiring an optimal amount of dietary TRP for growth—in fact, alterations to dietary TRP has resulted in osmotic-based acute stress [98,133,134], where TRP supplementation in non-stressed fish has resulted in increased plasma cortisol levels, while the opposite effect has been observed in stressed groups [13,85]. Moreover, exogenous TRP supplementation varies depending on fish species and size, as well as exposure to stressful environments (i.e., crowding, high stocking density, pollutants, etc.) [98,134,135,136].
Oxidative stress (OS) is another mechanism by which cell and tissue damage can occur. OS is an increase between the production and accumulation of ROS, which are generated as by-products of oxygen metabolism in processes including immune activation, apoptosis, cell differentiation, and protein phosphorylation, as well as through exposure to environmental stressors and xenobiotics [137]. The consequences of increased cell exposure to ROS include reduced levels of adenosine triphosphate (ATP), lipid peroxidation, cell membrane depolarization, morphological changes in cell surfaces, and DNA damage [138]. Cellular damage marked by OS includes lipid peroxidation, increased levels of protein carbonyl, and decreased levels of antioxidant enzymes (SOD, catalase [CAT] and glutathione peroxidase [GPx]) [137]. The TRP-KYN pathway plays a role in the onset of OS (Reviewed in: [139]) and it has been proposed that TRP exhibits antioxidant activity as it reacts with free radicals and modulates antioxidant enzyme activities. In fact, there is accumulating evidence to support the use of TRP and its metabolites as antioxidants [140]. In rabbits, dietary TRP supplementation was effective in protecting against free radical generation and lipid oxidative damage produced by hypoxic myocardial injury [141]. Similarly, in weaned piglets, increasing dietary TRP levels resulted in enhanced antioxidant capacity (SOD and GPx), which attenuated the OS response induced by diquat injection [142]. In line with the work done in mammals, dietary TRP supplementation in ducks alleviated stress and improved growth performance and antioxidant activity (GPx and CAT). Similarly, (Reviewed In: [143,144]) it has been shown in fish that while increased supplementation of dietary TRP increases total SOD, CAT, glutathione activity, and total antioxidant capacity, an optimal range of TRP supplementation, is dependent on species [145].
Some TRP metabolites including 5-hydroxytryptophan (5-HTP), indole-3-acetic acid (IAA), 3-HAA, 3-HK and XA can act as ROS scavengers and modulate antioxidant enzymes. Interestingly, other KYN metabolites including QUIN, 3-HK and 3-HAA can generate oxygen radicals and ROS, thereby modulating OS [146,147,148]. Accordingly, the pharmacological inhibition of the KYN pathway can decrease levels of OS. For example, in a rat model of schizophrenia, inhibitors of TDO, IDO and KMO were able to prevent lipid peroxidation, decrease protein carbonyl levels, and increase SOD levels and catalase activity [149]. There is also considerable evidence linking aging with OS; in many aging-associated disorders, there are consistent reports of upregulated IDO (Reviewed in: [150,151]). Currently, reliable markers of OS include carbonylated proteins, malondialdehyde, 4-hydroxy-2-nonenal, and F2-isoprostanes analyzed by LC-MS/MS [152]. However, given that TRP and KYN metabolite levels have been repeatedly shown to be sensitive and specific to antioxidant capacity and associated gene changes, TRP-KYN levels, or varying ratios of the metabolites (i.e., 3-HAA/AA), may be surrogate markers for the organism’s response to insults causing increased ROS.

3.3. Environmental Contaminants

Environmental stressors are factors that constrain the productivity, survival, and reproductive success of organisms. These stressors can range from biological or social stress (i.e., predation, competition, and disease); physical disturbances to the landscape (i.e., weather events like tsunamis, hurricanes, volcanic eruptions, and wildfires, or anthropogenic deforestation, machinery trampling, and hikers); and chemical pollution (i.e., pesticides, flame retardants, personal care products, waste chemicals from industrial development, and air pollution). Increasingly, stressors due to anthropogenic activities have become the most critical influence on species and ecosystems [153]. Rapid industrialization and urbanization can dramatically change the composition and diversity of biotic communities. Alongside urbanization comes the increased distribution of environmental toxins through human activity. Some of these contaminants include heavy metals; polycyclic aromatic compounds (PACs) which include benzo[a]pyrene (BaP), polycyclic aromatic hydrocarbons (PAHs) and their heterocyclic, alkylated, halogenated, oxygenated, sulphated and nitrated analogs (Reviewed In: [154]); dioxins, including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), polychlorinated dibenzofurans (PCDFs), polychlorinated dibenzo-p-dioxins (PCDDs); organochlorine compounds including polychlorinated biphenyls (PCBs), hexachlorocyclohexane isomers, dichlorodiphenyltrichloroethane (DDT) compounds, and hexachlorobenzenes (Reviewed In: [155]). Many of these contaminants can be released into the environment from natural and anthropogenic sources and have become ubiquitous in recent years. For example, PACs can be found in sea ice in the Arctic, industrialized harbors, the oil sands region in Canada, and major oil spills (e.g., the Exxon Valdez oil spill, Alaska; Prestige oil spill, Spain; Deepwater Horizon oil spill, Gulf of Mexico) [154,156]. Similarly, exposure to dioxins and furans can occur naturally through wildfires and through industrial processes such as waste incineration, the burning of fuels (i.e., wood, coal, and oil), and accidental/residential/structural fires. Other contaminants such as pesticides have an even greater public health concern; while they are designed to kill specific plants and insects, they often have harmful effects on non-target species. In mammals, exposure to pesticides has been linked to cancer, neurotoxicity and immunotoxicity [157]; endocrine disruption [158], reproductive effects, and birth and developmental defects (Reviewed In: [159]), and changes in energy homeostatic organs such as the liver and adipose tissue [160]. Similarly, in birds, organophosphates, neonicotinoids [161], carbamates and second-generation insecticides can result in decreased food consumption, weight loss, delayed migration, and decreases in the production, fertility, and hatchability of eggs (Reviewed In: [157,162]). Fish and other aquatic organisms are the most exposed in their environments due to the runoff of pesticides [163,164,165], oil spills [166,167,168,169], wastewaters from sewage containing personal care products [170,171,172,173], and chemical plant disposals; and have been extensively reviewed. Animal and human exposure to these chemicals can occur directly through pollution, or indirectly through food chain effects [174,175,176].
Many physiological homeostatic responses to environmental contaminants can be attributed to perturbations in pathways associated with GC signaling, inflammation, OS and, ultimately, alterations in TRP metabolism (Figure 6). For example, rats exposed to pyrethroid pesticides (deltamethrin and fenpropathrin) had significantly lowered KYNA production in cortical brain slices [177]. In chicken embryos, exposure to organophosphate and methylcarbamate insecticides was associated with lowered embryo NAD+ levels [178]; this effect was attributed to the inhibition of KYN formamidase, which impairs conversion of TRP to essential pyridine nucleotide cofactors. The majority of research in this area has focused on the link between environmental contaminants that act as AhR ligands and perturbations in the TRP-KYN pathway. Many environmental contaminants, particularly PACs, are known to act as AhR ligands; these compounds are also known to regulate IDO and TDO expression, thereby affecting the production of immunomodulatory TRP metabolites. For example, in mammals, it is well established that IDO expression and activity can be induced by AhR ligands including TCDD, BaP, and several PAHs [179,180]. Interestingly, KYN metabolites, including L-KYN, KYNA, XA, cinnabarinic acid, indigo, indirubin, and ultraviolet (UV) photoproducts of TRP such as 6-formylindolo[3,2-b]carbazole (FICZ) can act as AhR ligands and induce AhR target genes (CYP1A1, CYP1A2, and CYP1B1) (Reviewed In: [181]). Novikov et al. (2016) investigated the role of TRP-derived metabolites within malignant and non-malignant breast cancer cell lines and showed that (1) cell lines that expressed TDO produced sufficient intracellular KYN and XA concentrations to activate the AhR; (2) TDO overexpression led to excess KYN and XA, which accelerated the migration of tumour cells in an AhR-dependent manner; and (3) the environmental ligands TCDD and BaP, as well as the endogenous TRP-derivative FICZ mimic this effect [182]. Furthermore, AhR knockdown or inhibition significantly reduced TDO2 expression. KYN has also been shown to activate AhR in an autocrine/paracrine fashion, resulting in the suppression of antitumour immune responses and the promotion of tumour cell survival and motility [179]. Furthermore, FICZ—the UV photoproduct of TRP—binds to AhR with the highest affinity known to date of endogenous ligands; at high concentrations, FICZ behaves similarly to TCDD, exhibiting toxicity in fish and bird embryos, and playing a role in immunosuppression [183]. In birds, including the chicken (Gallus gallus domesticus), ring-necked pheasant (Phasianus colchicus), Japanese quail, and common tern (Sterna hirundo), FICZ has been identified as an avian AhR ligand [184], However, with the exception of changes in the avian CYP1A gene, other pathways mediated by AhR remain to be studied. 2-(1′H-indole-3′-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE)—a dietary TRP derivative—was discovered in porcine lung tissues and has been shown to be a high-affinity AhR ligand in humans, mice, and fish [185]. Taken together, these data suggest that across vertebrates, environmental exposure to certain contaminants can result in altered TRP-KYN metabolites that can contribute to a range of adverse health outcomes.

4. Conclusions

Many physiological responses to environmental contaminants can be attributed to perturbations in pathways associated with GC signaling, inflammation, oxidative stress, and, ultimately, alterations in TRP metabolism. Notably, environmental contaminants and their responses in vertebrate species are strikingly similar. Given that AhR and TRP metabolism are evolutionarily conserved across vertebrates (i.e., mammals, birds, fish), the crosstalk between xenobiotic receptors such as AhR, IDO/TDO immunoregulatory pathways, and altered stress indicators (i.e., antioxidant levels, ROS levels, and cortisol) suggest that alterations in TRP-KYN metabolism, metabolite levels, and ratio can be a cross-species marker of environmental exposure to chemical contaminants. Moreover, as ecotoxicological assessments are slowly moving away from evaluating the effects of a single compound to complex environmental mixtures, the TRP-KYN pathway provides a promising avenue to model the impacts of exposure to complex mixtures. Given its role in other biological processes, the TRP-KYN pathway provides many integrative biomarkers; both enzymatic ratios and metabolite levels can link environmental exposures to animal health, and broadly, ecosystem health. These integrative markers can be used as part of environmental effects monitoring (EEM) programs, where a few biomarkers could be monitored over a larger spatial area, across multiple species, trigger “investigation of cause” (IOC) studies or lead to focused studies to aid in the identification of mixtures and environmental factors that cause sublethal effects over acute and chronic time points.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms23116300/s1. References [186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226,227,228,229,230,231,232,233,234,235,236,237,238,239,240,241,242,243,244,245,246,247,248] are cited in the supplementary materials.

Author Contributions

Conceptualization, L.J., J.V.W., J.C.R., G.T.T., P.J.T. and A.C.H.; methodology and writing—original draft preparation, L.J., A.D., S.J. and J.V.W.; writing—review and editing, J.C.R., G.T.T., P.J.T. and A.C.H.; visualization, L.J. and S.J.; supervision, G.T.T. and A.C.H.; funding acquisition, L.J. and A.C.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Natural Science and Engineering Research Council (NSERC) of Canada a Discovery Grant to A.C.H., grant number RGPIN-2020-06358. Additional funding was provided by NSERC Vanier Canada Doctoral Scholarship to L.J.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare that they have no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

Abbreviations

3-HAA3-hydroxyanthranilic acid
3-HAAO3-hydroxyanthranilate 3,4-dioxygenase
3-HK3-hydroxykynurenine
5-HT5-hydroxytryptamine
5-HTP5-hydroxytryptophan
AAAnthranilic acid
ACMS2-amino-3-carboxymuconic semialdehyde
ACMSD2-amino-3-carboxymuconate semialdehyde decarboxylase
AhRAryl hydrocarbon receptor
AMPAα-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
ATPAdenosine triphosphate
BaPBenzo[a]pyrene
CATCatalase
CO2Carbon dioxide
CRHCorticotropin releasing hormone
CYPCytochrome P450
DDTDichlorodiphenyltrichloroethane
FICZ6-formylindolo[3,2-b]carbazole
GCGlucocorticoid
GPxGlutathione peroxide
GRGlucocorticoid receptor
HPAHypothalamic-pituitary-adrenocortical
IAAIndole-3-acetic Acid
IDOIndoleamine 2,3-dioxygenase
IFN-γProinflammatory interferon-gamma
ILInterleukin
IPAIndole-3-pyruvic acid
ITE2-(1′H-indole-3′-carbonyl)-thiazole-4-carboxylic acid methyl ester
KATKynurenine aminotransferase
KMOKynurenine monooxygenase
KYNKynurenine
KYNAKynurenic acid
KYNUKynureninase
MAOAMonoamine oxidase A
NAD+Nicotinamide adenine dinucleotide
NICNicotinic acid
NMDAN-methyl-D-aspartate
OSOxidative stress
PACPolycyclic aromatic compound
PAHPolycyclic aromatic hydrocarbon
PICPicolinic acid
PCBPolychlorinated biphenyl
PCDDPolychlorinated dibenzo-p-dioxin
PCDFPolychlorinated dibenzofuran
PGE3Prostaglandin
QPRTQuinolinate phosphoribosyltransferase
QUINQuinolinic acid
ROSReactive oxygen species
SODSuperoxide dismutase
TCDD2,3,7,8-tetrachlorodibenzo-p-dioxin
TDOTryptophan 2,3-dioxygenase
TGF-bTumor growth factor-beta
ThT-helper
TNFaTumor necrosis factor-alpha
TRPTryptophan
TRP-KYNTryptophan-kynurenine
UVUltraviolet
XAXanthurenic acid

References

  1. Francenia Santos-Sánchez, N.; Salas-Coronado, R.; Hernández-Carlos, B.; Villanueva-Cañongo, C. Shikimic Acid Pathway in Biosynthesis of Phenolic Compounds. In Plant Physiological Aspects of Phenolic Compounds; Soto-Hernández, M., García-Mateos, R., Palma-Tenango, M., Eds.; IntechOpen: London, UK, 2019; ISBN 978-1-78984-033-9. [Google Scholar]
  2. Yin, Y. Tryptophan Metabolism in Animals Important Roles in Nutrition and Health. Front. Biosci. 2011, S3, 286–297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Badawy, A.A.-B. Kynurenine Pathway of Tryptophan Metabolism: Regulatory and Functional Aspects. Int. J. Tryptophan Res. 2017, 10, 117864691769193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Palego, L.; Betti, L.; Rossi, A.; Giannaccini, G. Tryptophan Biochemistry: Structural, Nutritional, Metabolic, and Medical Aspects in Humans. J. Amino Acids 2016, 2016, 952520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Höglund, E.; Øverli, Ø.; Winberg, S. Tryptophan Metabolic Pathways and Brain Serotonergic Activity: A Comparative Review. Front. Endocrinol. 2019, 10, 158. [Google Scholar] [CrossRef]
  6. Albert, P.R.; Benkelfat, C.; Descarries, L. The Neurobiology of Depression—Revisiting the Serotonin Hypothesis. I. Cellular and Molecular Mechanisms. Phil. Trans. R. Soc. B 2012, 367, 2378–2381. [Google Scholar] [CrossRef] [Green Version]
  7. Oxenkrug, G.F. Tryptophan Kynurenine Metabolism as a Common Mediator of Genetic and Environmental Impacts in Major Depressive Disorder: The Serotonin Hypothesis Revisited 40 Years Later. Isr. J. Psychiatry Relat. Sci. 2010, 47, 56–63. [Google Scholar]
  8. Birkl, P.; Chow, J.; Forsythe, P.; Gostner, J.M.; Kjaer, J.B.; Kunze, W.A.; McBride, P.; Fuchs, D.; Harlander-Matauschek, A. The Role of Tryptophan-Kynurenine in Feather Pecking in Domestic Chicken Lines. Front. Vet. Sci. 2019, 6, 209. [Google Scholar] [CrossRef] [Green Version]
  9. Muller, C.L.; Anacker, A.M.J.; Veenstra-VanderWeele, J. The Serotonin System in Autism Spectrum Disorder: From Biomarker to Animal Models. Neuroscience 2016, 321, 24–41. [Google Scholar] [CrossRef] [Green Version]
  10. Walsh, J.J.; Christoffel, D.J.; Heifets, B.D.; Ben-Dor, G.A.; Selimbeyoglu, A.; Hung, L.W.; Deisseroth, K.; Malenka, R.C. 5-HT Release in Nucleus Accumbens Rescues Social Deficits in Mouse Autism Model. Nature 2018, 560, 589–594. [Google Scholar] [CrossRef]
  11. Bordukalo-Niksic, T.; Cicin-Sain, L.; Jernej, B. Expression of Brain and Platelet Serotonin Transporters in Sublines of Rats with Constitutionally Altered Serotonin Homeostasis. Neurosci. Lett. 2004, 369, 44–49. [Google Scholar] [CrossRef]
  12. Kesić, M.; Baković, P.; Horvatiček, M.; Proust, B.L.J.; Štefulj, J.; Čičin-Šain, L. Constitutionally High Serotonin Tone Favors Obesity: Study on Rat Sublines With Altered Serotonin Homeostasis. Front. Neurosci. 2020, 14, 219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Hoseini, S.M.; Hosseini, S.A. Effect of Dietary L-Tryptophan on Osmotic Stress Tolerance in Common Carp, Cyprinus Carpio, Juveniles. Fish Physiol. Biochem. 2010, 36, 1061–1067. [Google Scholar] [CrossRef] [PubMed]
  14. Sirotkin, A.V.; Schaeffer, H.-J. Direct Regulation of Mammalian Reproductive Organs by Serotonin and Melatonin. J. Endocrinol. 1997, 154, 1–5. [Google Scholar] [CrossRef] [PubMed]
  15. Prasad, P.; Ogawa, S.; Parhar, I.S. Role of Serotonin in Fish Reproduction. Front. Neurosci. 2015, 9, 195. [Google Scholar] [CrossRef]
  16. Sower, S.A.; Freamat, M.; Kavanaugh, S.I. The Origins of the Vertebrate Hypothalamic–Pituitary–Gonadal (HPG) and Hypothalamic–Pituitary–Thyroid (HPT) Endocrine Systems: New Insights from Lampreys. Gen. Comp. Endocrinol. 2009, 161, 20–29. [Google Scholar] [CrossRef]
  17. Holland, L.Z.; Albalat, R.; Azumi, K.; Benito-Gutierrez, E.; Blow, M.J.; Bronner-Fraser, M.; Brunet, F.; Butts, T.; Candiani, S.; Dishaw, L.J.; et al. The Amphioxus Genome Illuminates Vertebrate Origins and Cephalochordate Biology. Genome Res. 2008, 18, 1100–1111. [Google Scholar] [CrossRef] [Green Version]
  18. Kanda, S. Evolution of the Regulatory Mechanisms for the Hypothalamic-Pituitary-Gonadal Axis in Vertebrates–Hypothesis from a Comparative View. Gen. Comp. Endocrinol. 2019, 284, 113075. [Google Scholar] [CrossRef]
  19. Martin, A.M.; Young, R.L.; Leong, L.; Rogers, G.B.; Spencer, N.J.; Jessup, C.F.; Keating, D.J. The Diverse Metabolic Roles of Peripheral Serotonin. Endocrinology 2017, 158, 1049–1063. [Google Scholar] [CrossRef]
  20. Namkung, Jun; Kim, Hail; Park, Sangkyu Peripheral Serotonin: A New Player in Systemic Energy Homeostasis. Mol. Cells 2015, 38, 1023–1028. [CrossRef]
  21. Watanabe, H.; Rose, M.; Kanayama, Y.; Shirakawa, H.; Aso, H. Energy Homeostasis by the Peripheral Serotonergic System. In Serotonin—A Chemical Messenger Between All Types of Living Cells; Shad, K.F., Ed.; IntechOpen: London, UK, 2017; ISBN 978-953-51-3361-2. [Google Scholar]
  22. Khan, N.; Deschaux, P. Role of Serotonin in Fish Immunomodulation. J. Exp. Biol. 1997, 200, 1833–1838. [Google Scholar] [CrossRef]
  23. Yabut, J.M.; Crane, J.D.; Green, A.E.; Keating, D.J.; Khan, W.I.; Steinberg, G.R. Emerging Roles for Serotonin in Regulating Metabolism: New Implications for an Ancient Molecule. Endocr. Rev. 2019, 40, 1092–1107. [Google Scholar] [CrossRef] [PubMed]
  24. Cervenka, I.; Agudelo, L.Z.; Ruas, J.L. Kynurenines: Tryptophan’s Metabolites in Exercise, Inflammation, and Mental Health. Science 2017, 357, eaaf9794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Badawy, A.A.-B. Tryptophan Metabolism: A Versatile Area Providing Multiple Targets for Pharmacological Intervention. Egypt. J. Basic Clin. Pharmacol. 2019, 9. [Google Scholar] [CrossRef] [PubMed]
  26. Percudani, R.; Peracchi, A. A Genomic Overview of Pyridoxal-phosphate-dependent Enzymes. EMBO Rep. 2003, 4, 850–854. [Google Scholar] [CrossRef] [PubMed]
  27. Yuasa, H.J.; Ball, H.J. Efficient Tryptophan-Catabolizing Activity Is Consistently Conserved through Evolution of TDO Enzymes, but Not IDO Enzymes. J. Exp. Zool. B. Mol. Dev. Evol. 2015, 324, 128–140. [Google Scholar] [CrossRef]
  28. Chen, Y.; Guillemin, G.J. Kynurenine Pathway Metabolites in Humans: Disease and Healthy States. Int. J. Tryptophan Res. 2009, 2, IJTR.S2097. [Google Scholar] [CrossRef] [Green Version]
  29. Wirthgen, E.; Hoeflich, A.; Rebl, A.; Günther, J. Kynurenic Acid: The Janus-Faced Role of an Immunomodulatory Tryptophan Metabolite and Its Link to Pathological Conditions. Front. Immunol. 2018, 8, 1957. [Google Scholar] [CrossRef] [Green Version]
  30. 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]
  31. 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]
  32. Okabe, K.; Yaku, K.; Tobe, K.; Nakagawa, T. Implications of Altered NAD Metabolism in Metabolic Disorders. J. Biomed. Sci. 2019, 26, 34. [Google Scholar] [CrossRef] [Green Version]
  33. Grant, R.S.; Coggan, S.E.; Smythe, G.A. The Physiological Action of Picolinic Acid in the Human Brain. Int. J. Tryptophan Res. 2009, 2, IJTR.S2469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Paredes, S.D.; Barriga, C.; Reiter, R.J.; Rodríguez, A.B. Assessment of the Potential Role of Tryptophan as the Precursor of Serotonin and Melatonin for the Aged Sleep-Wake Cycle and Immune Function: Streptopelia Risoria as a Model. Int. J. Tryptophan Res. 2009, 2, IJTR.S1129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Bravo, R.; Matito, S.; Cubero, J.; Paredes, S.D.; Franco, L.; Rivero, M.; Rodríguez, A.B.; Barriga, C. Tryptophan-Enriched Cereal Intake Improves Nocturnal Sleep, Melatonin, Serotonin, and Total Antioxidant Capacity Levels and Mood in Elderly Humans. Geroscience 2013, 35, 1277–1285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Mangge, H.; Summers, K.L.; Meinitzer, A.; Zelzer, S.; Almer, G.; Prassl, R.; Schnedl, W.J.; Reininghaus, E.; Paulmichl, K.; Weghuber, D.; et al. Obesity-Related Dysregulation of the Tryptophan-Kynurenine Metabolism: Role of Age and Parameters of the Metabolic Syndrome: Tryptophan Metabolism and Inflammation in Obesity. Obesity 2014, 22, 195–201. [Google Scholar] [CrossRef] [PubMed]
  37. Liu, G.; Chen, S.; Zhong, J.; Teng, K.; Yin, Y. Crosstalk between Tryptophan Metabolism and Cardiovascular Disease, Mechanisms, and Therapeutic Implications. Oxid. Med. Cell. Longev. 2017, 2017, 1602074. [Google Scholar] [CrossRef] [PubMed]
  38. Mangge, H.; Stelzer, I.; Reininghaus, E.Z.; Weghuber, D.; Postolache, T.T.; Fuchs, D. Disturbed Tryptophan Metabolism in Cardiovascular Disease. Curr. Med. Chem. 2014, 21, 1931–1937. [Google Scholar] [CrossRef] [PubMed]
  39. 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] [PubMed] [Green Version]
  40. Choi, S.-C.; Brown, J.; Gong, M.; Ge, Y.; Zadeh, M.; Li, W.; Croker, B.P.; Michailidis, G.; Garrett, T.J.; Mohamadzadeh, M.; et al. Gut Microbiota Dysbiosis and Altered Tryptophan Catabolism Contribute to Autoimmunity in Lupus-Susceptible Mice. Sci. Transl. Med. 2020, 12, eaax2220. [Google Scholar] [CrossRef]
  41. Brown, R.R.; Ozaki, Y.; Datta, S.P.; Borden, E.C.; Sondel, P.M.; Malone, D.G. Implications of Interferon-Induced Tryptophan Catabolism in Cancer, Autoimmune Diseases and Aids. In Kynurenine and Serotonin Pathways; Schwarcz, R., Young, S.N., Brown, R.R., Eds.; Advances in Experimental Medicine and Biology; Springer New York: Boston, MA, USA, 1991; Volume 294, pp. 425–435. ISBN 978-1-4684-5954-8. [Google Scholar]
  42. 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]
  43. Kałużna-Czaplińska, J.; Gątarek, P.; Chirumbolo, S.; Chartrand, M.S.; Bjørklund, G. How Important Is Tryptophan in Human Health? Crit. Rev. Food Sci. Nutr. 2019, 59, 72–88. [Google Scholar] [CrossRef]
  44. Huang, Y.-S.; Ogbechi, J.; Clanchy, F.I.; Williams, R.O.; Stone, T.W. IDO and Kynurenine Metabolites in Peripheral and CNS Disorders. Front. Immunol. 2020, 11, 388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Liang, Y.; Xie, S.; He, Y.; Xu, M.; Qiao, X.; Zhu, Y.; Wu, W. Kynurenine Pathway Metabolites as Biomarkers in Alzheimer’s Disease. Dis. Markers 2022, 2022, e9484217. [Google Scholar] [CrossRef] [PubMed]
  46. Tan, V.X.; Guillemin, G.J. Kynurenine Pathway Metabolites as Biomarkers for Amyotrophic Lateral Sclerosis. Front. Neurosci. 2019, 13, 1013. [Google Scholar] [CrossRef] [PubMed]
  47. Erabi, H.; Okada, G.; Shibasaki, C.; Setoyama, D.; Kang, D.; Takamura, M.; Yoshino, A.; Fuchikami, M.; Kurata, A.; Kato, T.A.; et al. Kynurenic Acid Is a Potential Overlapped Biomarker between Diagnosis and Treatment Response for Depression from Metabolome Analysis. Sci. Rep. 2020, 10, 16822. [Google Scholar] [CrossRef]
  48. Badawy, A.A.-B. Modulation of Tryptophan and Serotonin Metabolism as a Biochemical Basis of the Behavioral Effects of Use and Withdrawal of Androgenic-Anabolic Steroids and Other Image- and Performance-Enhancing Agents. Int. J. Tryptophan Res. 2018, 11, 117864691775342. [Google Scholar] [CrossRef] [Green Version]
  49. Brooks, A.K.; Lawson, M.A.; Smith, R.A.; Janda, T.M.; Kelley, K.W.; McCusker, R.H. Interactions between Inflammatory Mediators and Corticosteroids Regulate Transcription of Genes within the Kynurenine Pathway in the Mouse Hippocampus. J. Neuroinflammation 2016, 13, 98. [Google Scholar] [CrossRef] [Green Version]
  50. Sorgdrager, F.J.H.; Werumeus Buning, J.; Bos, E.H.; Van Beek, A.P.; Kema, I.P. Hydrocortisone Affects Fatigue and Physical Functioning Through Metabolism of Tryptophan: A Randomized Controlled Trial. J. Clin. Endocrinol. Metab. 2018, 103, 3411–3419. [Google Scholar] [CrossRef]
  51. Lestage, J.; Verrier, D.; Palin, K.; Dantzer, R. The Enzyme Indoleamine 2,3-Dioxygenase Is Induced in the Mouse Brain in Response to Peripheral Administration of Lipopolysaccharide and Superantigen. Brain Behav. Immun. 2002, 16, 596–601. [Google Scholar] [CrossRef]
  52. Miura, H.; Ando, Y.; Noda, Y.; Isobe, K.; Ozaki, N. Long-Lasting Effects of Inescapable-Predator Stress on Brain Tryptophan Metabolism and the Behavior of Juvenile Mice. Stress 2011, 14, 262–272. [Google Scholar] [CrossRef]
  53. Mehraj, V.; Routy, J.-P. Tryptophan Catabolism in Chronic Viral Infections: Handling Uninvited Guests. Int. J. Tryptophan Res. 2015, 8, IJTR.S26862. [Google Scholar] [CrossRef]
  54. Dehhaghi, M.; Kazemi Shariat Panahi, H.; Guillemin, G.J. Microorganisms, Tryptophan Metabolism, and Kynurenine Pathway: A Complex Interconnected Loop Influencing Human Health Status. Int. J. Tryptophan Res. 2019, 12, 117864691985299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Majumdar, T.; Sharma, S.; Kumar, M.; Hussain, M.A.; Chauhan, N.; Kalia, I.; Sahu, A.K.; Rana, V.S.; Bharti, R.; Haldar, A.K.; et al. Tryptophan-Kynurenine Pathway Attenuates β-Catenin-Dependent pro-Parasitic Role of STING-TICAM2-IRF3-IDO1 Signalosome in Toxoplasma Gondii Infection. Cell Death Dis. 2019, 10, 161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Qi, Q.; Hua, S.; Clish, C.B.; Scott, J.M.; Hanna, D.B.; Wang, T.; Haberlen, S.A.; Shah, S.J.; Glesby, M.J.; Lazar, J.M.; et al. Plasma Tryptophan-Kynurenine Metabolites Are Altered in Human Immunodeficiency Virus Infection and Associated With Progression of Carotid Artery Atherosclerosis. Clin. Infect. Dis. 2018, 67, 235–242. [Google Scholar] [CrossRef] [PubMed]
  57. Mackay, G.M.; Forrest, C.M.; Stoy, N.; Christofides, J.; Egerton, M.; Stone, T.W.; Darlington, L.G. Tryptophan Metabolism and Oxidative Stress in Patients with Chronic Brain Injury. Eur. J. Neurol. 2006, 13, 30–42. [Google Scholar] [CrossRef] [PubMed]
  58. Pedraz-Petrozzi, B.; Elyamany, O.; Rummel, C.; Mulert, C. Effects of Inflammation on the Kynurenine Pathway in Schizophrenia—A Systematic Review. J. Neuroinflamm. 2020, 17, 56. [Google Scholar] [CrossRef] [PubMed]
  59. Sorgdrager, F.J.H.; Naudé, P.J.W.; Kema, I.P.; Nollen, E.A.; Deyn, P.P.D. Tryptophan Metabolism in Inflammaging: From Biomarker to Therapeutic Target. Front. Immunol. 2019, 10, 2565. [Google Scholar] [CrossRef]
  60. Agudelo, L.Z.; Ferreira, D.M.S.; Cervenka, I.; Bryzgalova, G.; Dadvar, S.; Jannig, P.R.; Pettersson-Klein, A.T.; Lakshmikanth, T.; Sustarsic, E.G.; Porsmyr-Palmertz, M.; et al. Kynurenic Acid and Gpr35 Regulate Adipose Tissue Energy Homeostasis and Inflammation. Cell Metab. 2018, 27, 378–392.e5. [Google Scholar] [CrossRef] [Green Version]
  61. Coccaro, E.F.; Lee, R.; Fanning, J.R.; Fuchs, D.; Goiny, M.; Erhardt, S.; Christensen, K.; Brundin, L.; Coussons-Read, M. Tryptophan, Kynurenine, and Kynurenine Metabolites: Relationship to Lifetime Aggression and Inflammatory Markers in Human Subjects. Psychoneuroendocrinology 2016, 71, 189–196. [Google Scholar] [CrossRef] [Green Version]
  62. Jaronen, M.; Quintana, F.J. Immunological Relevance of the Coevolution of IDO1 and AHR. Front. Immunol. 2014, 5, 521. [Google Scholar] [CrossRef] [Green Version]
  63. Yao, J.; Wang, J.; Wu, L.; Lu, H.; Wang, Z.; Yu, P.; Xiao, H.; Gao, R.; Yu, J. Perinatal Exposure to Bisphenol A Causes a Disturbance of Neurotransmitter Metabolic Pathways in Female Mouse Offspring: A Focus on the Tryptophan and Dopamine Pathways. Chemosphere 2020, 254, 126715. [Google Scholar] [CrossRef]
  64. Lin, Z.; Roede, J.R.; He, C.; Jones, D.P.; Filipov, N.M. Short-Term Oral Atrazine Exposure Alters the Plasma Metabolome of Male C57BL/6 Mice and Disrupts α-Linolenate, Tryptophan, Tyrosine and Other Major Metabolic Pathways. Toxicology 2014, 326, 130–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Laffon, B.; Aguilera, F.; Ríos-Vázquez, J.; García-Lestón, J.; Fuchs, D.; Valdiglesias, V.; Pásaro, E. Endocrine and Immunological Parameters in Individuals Involved in Prestige Spill Cleanup Tasks Seven Years after the Exposure. Environ. Int. 2013, 59, 103–111. [Google Scholar] [CrossRef] [PubMed]
  66. Ball, H.J.; Jusof, F.F.; Bakmiwewa, S.M.; Hunt, N.H.; Yuasa, H.J. Tryptophan-Catabolizing Enzymes—Party of Three. Front. Immunol. 2014, 5, 485. [Google Scholar] [CrossRef] [Green Version]
  67. Nold, V.; Sweatman, C.; Karabatsiakis, A.; Böck, C.; Bretschneider, T.; Lawless, N.; Fundel-Clemens, K.; Kolassa, I.-T.; Allers, K.A. Activation of the Kynurenine Pathway and Mitochondrial Respiration to Face Allostatic Load in a Double-Hit Model of Stress. Psychoneuroendocrinology 2019, 107, 148–159. [Google Scholar] [CrossRef] [PubMed]
  68. McEwen, B.S. Neurobiological and Systemic Effects of Chronic Stress. Chronic Stress 2017, 1, 2470547017692328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Ryan, J.A.; Hightower, L.E. Stress Proteins as Molecular Biomarkers for Environmental Toxicology. In Stress-Inducible Cellular Responses; Feige, U., Yahara, I., Morimoto, R.I., Polla, B.S., Eds.; EXS; Birkhäuser: Basel, Switzerland, 1996; pp. 411–424. ISBN 978-3-0348-9088-5. [Google Scholar]
  70. Leonard, B.E. HPA and Immune Axes in Stress: Involvement of the Serotonergic System. Neuroimmunomodulation 2006, 13, 268–276. [Google Scholar] [CrossRef] [PubMed]
  71. Isenović, E.R.; Zakula, Z.; Koricanac, G.; Ribarac-Stepić, N. Comparative Analysis of Tryptophan Oxygenase Activity and Glucocorticoid Receptor under the Influence of Insulin. Physiol. Res. 2008, 57, 101–107. [Google Scholar] [CrossRef] [PubMed]
  72. NIIMI, S.; NAKAMURA, T.; NAWA, K.; ICHIHARA, A. Hormonal Regulation of Translatable MRNA of Tryptophan 2,3-Dioxygenase in Primary Cultures of Adult Rat Hepatocytes1. J. Biochem. 1983, 94, 1697–1706. [Google Scholar] [CrossRef]
  73. Schubart, U.K. Regulation of Gene Expression in Rat Hepatocytes and Hepatoma Cells by Insulin: Quantitation of Messenger Ribonucleic Acid’s Coding for Tyrosine Aminotransferase, Tryptophan Oxygenase, and Phosphoenolpyruvate Carboxykinase. Endocrinology 1986, 119, 1741–1749. [Google Scholar] [CrossRef]
  74. Antunes, M.S.; Ruff, J.R.; de Oliveira Espinosa, D.; Piegas, M.B.; de Brito, M.L.O.; Rocha, K.A.; de Gomes, M.G.; Goes, A.T.R.; Souza, L.C.; Donato, F.; et al. Neuropeptide Y Administration Reverses Tricyclic Antidepressant Treatment-Resistant Depression Induced by ACTH in Mice. Horm. Behav. 2015, 73, 56–63. [Google Scholar] [CrossRef]
  75. Martín-Hernández, D.; Tendilla-Beltrán, H.; Madrigal, J.L.M.; García-Bueno, B.; Leza, J.C.; Caso, J.R. Chronic Mild Stress Alters Kynurenine Pathways Changing the Glutamate Neurotransmission in Frontal Cortex of Rats. Mol. Neurobiol. 2019, 56, 490–501. [Google Scholar] [CrossRef] [PubMed]
  76. Liu, W.; Sheng, H.; Xu, Y.; Liu, Y.; Lu, J.; Ni, X. Swimming Exercise Ameliorates Depression-like Behavior in Chronically Stressed Rats: Relevant to Proinflammatory Cytokines and IDO Activation. Behav. Brain Res. 2013, 242, 110–116. [Google Scholar] [CrossRef] [PubMed]
  77. Dostal, C.R.; Carson Sulzer, M.; Kelley, K.W.; Freund, G.G.; MCusker, R.H. Glial and Tissue-Specific Regulation of Kynurenine Pathway Dioxygenases by Acute Stress of Mice. Neurobiol. Stress 2017, 7, 1–15. [Google Scholar] [CrossRef] [PubMed]
  78. Miura, H.; Ozaki, N.; Sawada, M.; Isobe, K.; Ohta, T.; Nagatsu, T. A Link between Stress and Depression: Shifts in the Balance between the Kynurenine and Serotonin Pathways of Tryptophan Metabolism and the Etiology and Pathophysiology of Depression. Stress 2008, 11, 198–209. [Google Scholar] [CrossRef] [PubMed]
  79. Ozaki, Y.; Edelstein, M.P.; Duch, D.S. The Actions of Interferon and Antiinflammatory Agents on Induction of Indoleamine 2,3-Dioxygenase in Human Peripheral Blood Monocytes. Biochem. Biophys. Res. Commun. 1987, 144, 1147–1153. [Google Scholar] [CrossRef]
  80. Rose, M.; Filiatreault, A.; Guénette, J.; Williams, A.; Thomson, E.M. Ozone Increases Plasma Kynurenine-Tryptophan Ratio and Impacts Hippocampal Serotonin Receptor and Neurotrophic Factor Expression: Role of Stress Hormones. Environ. Res. 2020, 185, 109483. [Google Scholar] [CrossRef]
  81. Bartolini, B.; Corniello, C.; Sella, A.; Somma, F.; Politi, V. The Enol Tautomer of Indole-3-Pyruvic Acid as A Biological Switch in Stress Responses. In Developments in Tryptophan and Serotonin Metabolism; Allegri, G., Costa, C.V.L., Ragazzi, E., Steinhart, H., Varesio, L., Eds.; Advances in Experimental Medicine and Biology; Springer USA: Boston, MA, USA, 2003; pp. 601–608. ISBN 978-1-4615-0135-0. [Google Scholar]
  82. van Hierden, Y.M.; Koolhaas, J.M.; Korte, S.M. Chronic Increase of Dietary L-Tryptophan Decreases Gentle Feather Pecking Behaviour. Appl. Anim. Behav. Sci. 2004, 89, 71–84. [Google Scholar] [CrossRef] [Green Version]
  83. Iwama, G.K. Stress in Fish. Ann. N. Y. Acad. Sci. 1998, 851, 304–310. [Google Scholar] [CrossRef]
  84. Herrera, M.; Fernández-Alacid, L.; Sanahuja, I.; Ibarz, A.; Salamanca, N.; Morales, E.; Giráldez, I. Physiological and Metabolic Effects of a Tryptophan-Enriched Diet to Face up Chronic Stress in Meagre (Argyrosomus regius). Aquaculture 2020, 522, 735102. [Google Scholar] [CrossRef]
  85. Lepage, O.; Tottmar, O.; Winberg, S. Elevated Dietary Intake of L-Tryptophan Counteracts the Stress-Induced Elevation of Plasma Cortisol in Rainbow Trout (Oncorhynchus mykiss). J. Exp. Biol. 2002, 205, 3679–3687. [Google Scholar] [CrossRef]
  86. Winberg, S.; Lepage, O. Elevation of Brain 5-HT Activity, POMC Expression, and Plasma Cortisol in Socially Subordinate Rainbow Trout. Am. J. Physiol. Regul. Integr. Comp. Physiol. 1998, 274, R645–R654. [Google Scholar] [CrossRef] [PubMed]
  87. Herrera, M.; Miró, J.M.; Giráldez, I.; Salamanca, N.; Martos-Sitcha, J.A.; Mancera, J.M.; López, J.R. Metabolic and Stress Responses in Senegalese Soles (Solea Senegalensis Kaup) Fed Tryptophan Supplements: Effects of Concentration and Feeding Period. Animals 2019, 9, 320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Basic, D.; Krogdahl, Å.; Schjolden, J.; Winberg, S.; Vindas, M.A.; Hillestad, M.; Mayer, I.; Skjerve, E.; Höglund, E. Short- and Long-Term Effects of Dietary l-Tryptophan Supplementation on the Neuroendocrine Stress Response in Seawater-Reared Atlantic Salmon (Salmo salar). Aquaculture 2013, 388–391, 8–13. [Google Scholar] [CrossRef]
  89. Basic, D.; Schjolden, J.; Krogdahl, Å.; von Krogh, K.; Hillestad, M.; Winberg, S.; Mayer, I.; Skjerve, E.; Höglund, E. Changes in Regional Brain Monoaminergic Activity and Temporary Down-Regulation in Stress Response from Dietary Supplementation with l-Tryptophan in Atlantic Cod (Gadus Morhua). Br. J. Nutr. 2013, 109, 2166–2174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Wish, J.; Bulloch, P.; Oswald, L.; Halldorson, T.; Raine, J.C.; Jamshed, L.; Marvin, C.; Thomas, P.J.; Holloway, A.C.; Tomy, G.T. Kynurenine to Tryptophan Ratio as a Biomarker of Acute Stress in Fish. Chemosphere 2022, 288, 132522. [Google Scholar] [CrossRef] [PubMed]
  91. Gelpi, M.; Ueland, P.M.; Trøseid, M.; Mocroft, A.; Lebech, A.-M.; Ullum, H.; Midttun, Ø.; Lundgren, J.; Nielsen, S.D.; for the Copenhagen Comorbidity in HIV Infection (COCOMO) Study. Abdominal Adipose Tissue Is Associated With Alterations in Tryptophan-Kynurenine Metabolism and Markers of Systemic Inflammation in People With Human Immunodeficiency Virus. J. Infect. Dis. 2020, 221, 419–427. [Google Scholar] [CrossRef] [PubMed]
  92. Hajsl, M.; Hlavackova, A.; Broulikova, K.; Sramek, M.; Maly, M.; Dyr, J.E.; Suttnar, J. Tryptophan Metabolism, Inflammation, and Oxidative Stress in Patients with Neurovascular Disease. Metabolites 2020, 10, 208. [Google Scholar] [CrossRef]
  93. Beal, M.F.; Kowall, N.W.; Ellison, D.W.; Mazurek, M.F.; Swartz, K.J.; Martin, J.B. Replication of the Neurochemical Characteristics of Huntington’s Disease by Quinolinic Acid. Nature 1986, 321, 168–171. [Google Scholar] [CrossRef]
  94. Wu, W.; Nicolazzo, J.A.; Wen, L.; Chung, R.; Stankovic, R.; Bao, S.S.; Lim, C.K.; Brew, B.J.; Cullen, K.M.; Guillemin, G.J. Expression of Tryptophan 2,3-Dioxygenase and Production of Kynurenine Pathway Metabolites in Triple Transgenic Mice and Human Alzheimer’s Disease Brain. PLoS ONE 2013, 8, e59749. [Google Scholar] [CrossRef] [Green Version]
  95. Stoy, N.; Mackay, G.M.; Forrest, C.M.; Christofides, J.; Egerton, M.; Stone, T.W.; Darlington, L.G. Tryptophan Metabolism and Oxidative Stress in Patients with Huntington’s Disease. J. Neurochem. 2005, 93, 611–623. [Google Scholar] [CrossRef]
  96. Lanser, L.; Kink, P.; Egger, E.M.; Willenbacher, W.; Fuchs, D.; Weiss, G.; Kurz, K. Inflammation-Induced Tryptophan Breakdown Is Related With Anemia, Fatigue, and Depression in Cancer. Front. Immunol. 2020, 11, 249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Dantzer, R.; O’Connor, J.C.; Lawson, M.A.; Kelley, K.W. Inflammation-Associated Depression: From Serotonin to Kynurenine. Psychoneuroendocrinology 2011, 36, 426–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Machado, M.; Azeredo, R.; Domingues, A.; Fernandez-Boo, S.; Dias, J.; Conceição, L.E.C.; Costas, B. Dietary Tryptophan Deficiency and Its Supplementation Compromises Inflammatory Mechanisms and Disease Resistance in a Teleost Fish. Sci. Rep. 2019, 9, 7689. [Google Scholar] [CrossRef] [PubMed]
  99. Bai, M.; Liu, H.; Xu, K.; Oso, A.O.; Wu, X.; Liu, G.; Tossou, M.C.B.; Al-Dhabi, N.A.; Duraipandiyan, V.; Xi, Q.; et al. A Review of the Immunomodulatory Role of Dietary Tryptophan in Livestock and Poultry. Amino Acids 2017, 49, 67–74. [Google Scholar] [CrossRef]
  100. Van der Leek, A.P.; Yanishevsky, Y.; Kozyrskyj, A.L. The Kynurenine Pathway As a Novel Link between Allergy and the Gut Microbiome. Front. Immunol. 2017, 8, 1374. [Google Scholar] [CrossRef] [Green Version]
  101. Mellor, A.L.; Munn, D.H. Tryptophan Catabolism and Regulation of Adaptive Immunity. J. Immunol. 2003, 170, 5809–5813. [Google Scholar] [CrossRef] [Green Version]
  102. Taylor, M.W.; Feng, G.S. Relationship between Interferon-Gamma, Indoleamine 2,3-Dioxygenase, and Tryptophan Catabolism. FASEB J. 1991, 5, 2516–2522. [Google Scholar] [CrossRef]
  103. Widner, B.; Ledochowski, M.; Fuchs, D. Interferon-Gamma-Induced Tryptophan Degradation: Neuropsychiatric and Immunological Consequences. Curr. Drug Metab. 2000, 1, 193–204. [Google Scholar] [CrossRef]
  104. Varma, T.K.; Lin, C.Y.; Toliver-Kinsky, T.E.; Sherwood, E.R. Endotoxin-Induced Gamma Interferon Production: Contributing Cell Types and Key Regulatory Factors. Clin. Diagn. Lab Immunol. 2002, 9, 530–543. [Google Scholar] [CrossRef] [Green Version]
  105. Williams, M.; Zhang, Z.; Nance, E.; Drewes, J.L.; Lesniak, W.G.; Singh, S.; Chugani, D.C.; Rangaramanujam, K.; Graham, D.R.; Kannan, S. Maternal Inflammation Results in Altered Tryptophan Metabolism in Rabbit Placenta and Fetal Brain. Dev. Neurosci. 2017, 39, 399–412. [Google Scholar] [CrossRef]
  106. 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]
  107. 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]
  108. Liebau, C.; Merk, H.; Schmidt, S.; Roesel, C.; Karreman, C.; Prisack, J.B.; Bojar, H.; Baltzer, A.W. Interleukin-12 and Interleukin-18 Change ICAM-I Expression, and Enhance Natural Killer Cell Mediated Cytolysis of Human Osteosarcoma Cells. Cytokines Cell. Mol. Ther. 2002, 7, 135–142. [Google Scholar] [CrossRef]
  109. Wu, H.; Gong, J.; Liu, Y. Indoleamine 2, 3-Dioxygenase Regulation of Immune Response (Review). Mol. Med. Rep. 2018, 17, 4867–4873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. 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]
  111. Quon, T.; Lin, L.-C.; Ganguly, A.; Tobin, A.B.; Milligan, G. Therapeutic Opportunities and Challenges in Targeting the Orphan G Protein-Coupled Receptor GPR35. ACS Pharmacol. Transl. Sci. 2020, 3, 801–812. [Google Scholar] [CrossRef] [PubMed]
  112. Ligam, P.; Manuelpillai, U.; Wallace, E.M.; Walker, D. Localisation of Indoleamine 2,3-Dioxygenase and Kynurenine Hydroxylase in the Human Placenta and Decidua: Implications for Role of the Kynurenine Pathway in Pregnancy. Placenta 2005, 26, 498–504. [Google Scholar] [CrossRef]
  113. Sedlmayr, P.; Blaschitz, A.; Wintersteiger, R.; Semlitsch, M.; Hammer, A.; MacKenzie, C.R.; Walcher, W.; Reich, O.; Takikawa, O.; Dohr, G. Localization of Indoleamine 2,3-Dioxygenase in Human Female Reproductive Organs and the Placenta. Mol. Hum. Reprod. 2002, 8, 385–391. [Google Scholar] [CrossRef] [Green Version]
  114. von Rango, U.; Krusche, C.A.; Beier, H.M.; Classen-Linke, I. Indoleamine-Dioxygenase Is Expressed in Human Decidua at the Time Maternal Tolerance Is Established. J. Reprod. Immunol. 2007, 74, 34–45. [Google Scholar] [CrossRef]
  115. McGaha, T.L.; Huang, L.; Lemos, H.; Metz, R.; Mautino, M.; Prendergast, G.C.; Mellor, A.L. Amino Acid Catabolism: A Pivotal Regulator of Innate and Adaptive Immunity. Immunol. Rev. 2012, 249, 135–157. [Google Scholar] [CrossRef] [Green Version]
  116. Baumgartner, R.; Forteza, M.J.; Ketelhuth, D.F.J. The Interplay between Cytokines and the Kynurenine Pathway in Inflammation and Atherosclerosis. Cytokine 2019, 122, 154148. [Google Scholar] [CrossRef] [PubMed]
  117. Moffett, J.R.; Arun, P.; Puthillathu, N.; Vengilote, R.; Ives, J.A.; Badawy, A.A.-B.; Namboodiri, A.M. Quinolinate as a Marker for Kynurenine Metabolite Formation and the Unresolved Question of NAD+ Synthesis During Inflammation and Infection. Front. Immunol. 2020, 11, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Gürcü, S.; Girgin, G.; Yorulmaz, G.; Kılıçarslan, B.; Efe, B.; Baydar, T. Neopterin and Biopterin Levels and Tryptophan Degradation in Patients with Diabetes. Sci. Rep. 2020, 10, 17025. [Google Scholar] [CrossRef]
  119. Changsirivathanathamrong, D.; Wang, Y.; Rajbhandari, D.; Maghzal, G.J.; Mak, W.M.; Woolfe, C.; Duflou, J.; Gebski, V.; dos Remedios, C.G.; Celermajer, D.S.; et al. Tryptophan Metabolism to Kynurenine Is a Potential Novel Contributor to Hypotension in Human Sepsis. Crit. Care Med. 2011, 39, 2678–2683. [Google Scholar] [CrossRef] [PubMed]
  120. Kwon, J.-S.; Lee, H.-J.; Lee, D.-H.; Lee, Y.-J.; Mo, I.-P.; Nahm, S.-S.; Kim, M.-J.; Lee, J.-B.; Park, S.-Y.; Choi, I.-S.; et al. Immune Responses and Pathogenesis in Immunocompromised Chickens in Response to Infection with the H9N2 Low Pathogenic Avian Influenza Virus. Virus Res. 2008, 133, 187–194. [Google Scholar] [CrossRef]
  121. Yuk, S.-S.; Lee, D.-H.; Park, J.-K.; Tseren-Ochir, E.-O.; Kwon, J.-H.; Noh, J.-Y.; Lee, J.-B.; Park, S.-Y.; Choi, I.-S.; Song, C.-S. Pre-Immune State Induced by Chicken Interferon Gamma Inhibits the Replication of H1N1 Human and H9N2 Avian Influenza Viruses in Chicken Embryo Fibroblasts. Virol. J. 2016, 13, 71. [Google Scholar] [CrossRef] [Green Version]
  122. Santhakumar, D.; Rubbenstroth, D.; Martinez-Sobrido, L.; Munir, M. Avian Interferons and Their Antiviral Effectors. Front. Immunol. 2017, 8, 49. [Google Scholar] [CrossRef] [Green Version]
  123. Jiang, H.; Yang, H.; Kapczynski, D.R. Chicken Interferon Alpha Pretreatment Reduces Virus Replication of Pandemic H1N1 and H5N9 Avian Influenza Viruses in Lung Cell Cultures from Different Avian Species. Virol. J. 2011, 8, 447. [Google Scholar] [CrossRef] [Green Version]
  124. Emadi, M.; Jahanshiri, F.; Kaveh, K.; Hair-Bejo, M.; Ideris, A.; Alimon, R. Tryptophan Stimulates Immune Response in Broiler Chickens Challenged with Infectious Bursal Disease Vaccine. J. Anim. Vet. Adv. 2010, 9, 610–616. [Google Scholar] [CrossRef] [Green Version]
  125. Grohmann, U.; Fallarino, F.; Puccetti, P. Tolerance, DCs and Tryptophan: Much Ado about IDO. Trends Immunol. 2003, 24, 242–248. [Google Scholar] [CrossRef]
  126. Le Floc’h, N.; Otten, W.; Merlot, E. Tryptophan Metabolism, from Nutrition to Potential Therapeutic Applications. Amino Acids 2011, 41, 1195–1205. [Google Scholar] [CrossRef] [PubMed]
  127. Robertsen, B. The Interferon System of Teleost Fish. Fish Shellfish Immunol. 2006, 20, 172–191. [Google Scholar] [CrossRef] [PubMed]
  128. Zou, J.; Secombes, C.J. Teleost Fish Interferons and Their Role in Immunity. Dev. Comp. Immunol. 2011, 35, 1376–1387. [Google Scholar] [CrossRef] [PubMed]
  129. Biron, C.A.; Sen, G.C. Interferons and Other Cytokines. In Fields’ Virology; Lippincott Williams and Wilkins: Philadelphia, PA, USA, 2001; pp. 321–351. [Google Scholar]
  130. Machado, M.; Azeredo, R.; Díaz-Rosales, P.; Afonso, A.; Peres, H.; Oliva-Teles, A.; Costas, B. Dietary Tryptophan and Methionine as Modulators of European Seabass (Dicentrarchus Labrax) Immune Status and Inflammatory Response. Fish Shellfish Immunol. 2015, 42, 353–362. [Google Scholar] [CrossRef] [Green Version]
  131. Hoseini, S.M.; Mirghaed, A.T.; Mazandarani, M.; Zoheiri, F. Serum Cortisol, Glucose, Thyroid Hormones’ and Non-Specific Immune Responses of Persian Sturgeon, Acipenser Persicus to Exogenous Tryptophan and Acute Stress. Aquaculture 2016, 462, 17–23. [Google Scholar] [CrossRef]
  132. Qiu, S.; Fang, Z.; Wu, D.; Lin, Y.; Che, L. Tryptophan Supplements Promote Pregnancy Success in Mice Challenged with Pseudorabies Virus (PRV) by Regulating the Expression of Systemic Cytokines, Immunoglobulins, PRV-Specific Protein Profiles, and Toll-Like Receptors. J. Med. Food 2011, 14, 857–865. [Google Scholar] [CrossRef]
  133. Engelsma, M.Y.; Hougee, S.; Nap, D.; Hofenk, M.; Rombout, J.H.W.M.; van Muiswinkel, W.B.; Lidy Verburg-van Kemenade, B.M. Multiple Acute Temperature Stress Affects Leucocyte Populations and Antibody Responses in Common carp, Cyprinus carpio L. Fish Shellfish Immunol. 2003, 15, 397–410. [Google Scholar] [CrossRef]
  134. Barton, B.A. Stress in Fishes: A Diversity of Responses with Particular Reference to Changes in Circulating Corticosteroids. Integr. Comp. Biol. 2002, 42, 517–525. [Google Scholar] [CrossRef]
  135. Cerqueira, M.; Schrama, D.; Silva, T.S.; Colen, R.; Engrola, S.A.D.; Conceição, L.E.C.; Rodrigues, P.M.L.; Farinha, A.P. How Tryptophan Levels in Plant-Based Aquafeeds Affect Fish Physiology, Metabolism and Proteome. J. Proteomics 2020, 221, 03782. [Google Scholar] [CrossRef]
  136. Kaczorek, E.; Szarek, J.; Mikiewicz, M.; Terech-Majewska, E.; Schulz, P.; Małaczewska, J.; Wójcik, R.; Siwicki, A.K. Effect of Feed Supplementation with Kynurenic Acid on the Morphology of the Liver, Kidney and Gills in Rainbow Trout (Oncorhynchus Mykiss Walbaum, 1792), Healthy and Experimentally Infected with Yersinia Ruckeri. J. Fish Dis. 2017, 40, 873–884. [Google Scholar] [CrossRef]
  137. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative Stress: Harms and Benefits for Human Health. Oxid. Med. Cell. Longev. 2017, 2017, e8416763. [Google Scholar] [CrossRef] [PubMed]
  138. Dandekar, A.; Mendez, R.; Zhang, K. Cross Talk between ER Stress, Oxidative Stress, and Inflammation in Health and Disease. Methods Mol. Biol. 2015, 1292, 205–214. [Google Scholar] [CrossRef] [PubMed]
  139. González Esquivel, D.; Ramírez-Ortega, D.; Pineda, B.; Castro, N.; Ríos, C.; Pérez de la Cruz, V. Kynurenine Pathway Metabolites and Enzymes Involved in Redox Reactions. Neuropharmacology 2017, 112, 331–345. [Google Scholar] [CrossRef]
  140. Xu, K.; Liu, H.; Bai, M.; Gao, J.; Wu, X.; Yin, Y. Redox Properties of Tryptophan Metabolism and the Concept of Tryptophan Use in Pregnancy. Int. J. Mol. Sci. 2017, 18, 1595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Narïn, F.; Narin, N.; Başarslan, F.; Baykan, A.; Sezer, S.; Akgün, H.; Akin, A.; Akçakuş, M.; Ceyran, H. The Effect of L-Tryptophan on the Heart in Rabbits via Chronic Hypoxia. Turk. J. Med. Sci. 2010, 40, 257–263. [Google Scholar] [CrossRef]
  142. Mao, X.; Lv, M.; Yu, B.; He, J.; Zheng, P.; Yu, J.; Wang, Q.; Chen, D. The Effect of Dietary Tryptophan Levels on Oxidative Stress of Liver Induced by Diquat in Weaned Piglets. J. Anim. Sci. Biotechnol. 2014, 5, 49. [Google Scholar] [CrossRef] [Green Version]
  143. Hoseini, S.M.; Pérez-Jiménez, A.; Costas, B.; Azeredo, R.; Gesto, M. Physiological Roles of Tryptophan in Teleosts: Current Knowledge and Perspectives for Future Studies. Rev. Aquac. 2019, 11, 3–24. [Google Scholar] [CrossRef] [Green Version]
  144. Tripathy, A. Oxidative Stress, Reactive Oxygen Species (ROS) and Antioxidative Defense System, with Special Reference to Fish. Int. J. Curr. Res. Biosci. Plant Biol. 2016, 3, 79–89. [Google Scholar] [CrossRef] [Green Version]
  145. Ji, K.; Liang, H.; Ren, M.; Ge, X.; Liu, B.; Xi, B.; Pan, L.; Yu, H. Effects of Dietary Tryptophan Levels on Antioxidant Status and Immunity for Juvenile Blunt Snout Bream (Megalobrama Amblycephala) Involved in Nrf2 and TOR Signaling Pathway. Fish Shellfish Immunol. 2019, 93, 474–483. [Google Scholar] [CrossRef]
  146. Forrest, C.M.; Mackay, G.M.; Stoy, N.; Egerton, M.; Christofides, J.; Stone, T.W.; Darlington, L.G. Tryptophan Loading Induces Oxidative Stress. Free Radic. Res. 2004, 38, 1167–1171. [Google Scholar] [CrossRef]
  147. Lugo-Huitrón, R.; Blanco-Ayala, T.; Ugalde-Muñiz, P.; Carrillo-Mora, P.; Pedraza-Chaverrí, J.; Silva-Adaya, D.; Maldonado, P.D.; Torres, I.; Pinzón, 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] [PubMed]
  148. Wang, Q.; Liu, D.; Song, P.; Zou, M.-H. Tryptophan-Kynurenine Pathway Is Dysregulated in Inflammation, and Immune Activation. Front. Biosci. (Landmark Ed) 2015, 20, 1116–1143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Réus, G.Z.; Becker, I.R.T.; Scaini, G.; Petronilho, F.; Oses, J.P.; Kaddurah-Daouk, R.; Ceretta, L.B.; Zugno, A.I.; Dal-Pizzol, F.; Quevedo, J.; et al. The Inhibition of the Kynurenine Pathway Prevents Behavioral Disturbances and Oxidative Stress in the Brain of Adult Rats Subjected to an Animal Model of Schizophrenia. Prog. Neuropsychopharmacol. Biol. Psychiatry 2018, 81, 55–63. [Google Scholar] [CrossRef] [PubMed]
  150. Sas, K.; Szabó, E.; Vécsei, L. Mitochondria, Oxidative Stress and the Kynurenine System, with a Focus on Ageing and Neuroprotection. Molecules 2018, 23, 191. [Google Scholar] [CrossRef] [Green Version]
  151. Reyes Ocampo, J.; Lugo Huitrón, R.; González-Esquivel, D.; Ugalde-Muñiz, P.; Jiménez-Anguiano, A.; Pineda, B.; Pedraza-Chaverri, J.; Ríos, C.; Pérez de la Cruz, V. Kynurenines with Neuroactive and Redox Properties: Relevance to Aging and Brain Diseases. Oxid. Med. Cell. Longev. 2014, 2014, e646909. [Google Scholar] [CrossRef]
  152. Gianazza, E.; Brioschi, M.; Fernandez, A.M.; Banfi, C. Lipoxidation in Cardiovascular Diseases. Redox Biol. 2019, 23, 101119. [Google Scholar] [CrossRef]
  153. Monosson, E. Chemical Mixtures: Considering the Evolution of Toxicology and Chemical Assessment. Environ. Health Perspect. 2005, 113, 383–390. [Google Scholar] [CrossRef] [Green Version]
  154. Wallace, S.J.; de Solla, S.R.; Head, J.A.; Hodson, P.V.; Parrott, J.L.; Thomas, P.J.; Berthiaume, A.; Langlois, V.S. Polycyclic Aromatic Compounds (PACs) in the Canadian Environment: Exposure and Effects on Wildlife. Environ. Pollut. 2020, 265, 114863. [Google Scholar] [CrossRef]
  155. Hoshi, H.; Minamoto, N.; Iwata, H.; Shiraki, K.; Tatsukawa, R.; Tanabe, S.; Fujita, S.; Hirai, K.; Kinjo, T. Organochlorine Pesticides and Polychlorinated Biphenyl Congeners in Wild Terrestrial Mammals and Birds from Chubu Region, Japan: Interspecies Comparison of the Residue Levels and Compositions. Chemosphere 1998, 36, 3211–3221. [Google Scholar] [CrossRef]
  156. Lee, K.; Boufadel, M.; Chen, B.; Foght, J.; Hodson, P.; Swanson, S.; Venosa, A. Expert Panel Report on the Behaviour and Environmental Impacts of Crude Oil Released into Aqueous Environments; Royal Society of Canada: Ottawa, ON, Canada, 2015; ISBN 978-1-928140-02-3. [Google Scholar]
  157. Mitra, A.; Sarkar, M.; Chatterjee, C. Modulation of Immune Response by Organophosphate Pesticides: Mammals as Potential Model. Proc. Zool. Soc. 2019, 72, 13–24. [Google Scholar] [CrossRef]
  158. Santi, D.; Marca, A.L.; Michelangeli, M.; Casonati, A.; Grassi, R.; Baraldi, E.; Simoni, M. Ovarian Reserve and Exposure to Environmental Pollutants (ORExPo Study). In Proceedings of the Endocrine Abstracts, Bioscientifica, Bristol, UK, 1 May 2019; Volume 63. [Google Scholar]
  159. Tiemann, U. In Vivo and in Vitro Effects of the Organochlorine Pesticides DDT, TCPM, Methoxychlor, and Lindane on the Female Reproductive Tract of Mammals: A Review. Reprod. Toxicol. 2008, 25, 316–326. [Google Scholar] [CrossRef] [PubMed]
  160. Gutgesell, R.M.; Tsakiridis, E.E.; Jamshed, S.; Steinberg, G.R.; Holloway, A.C. Impact of Pesticide Exposure on Adipose Tissue Development and Function. Biochem J. 2020, 477, 2639–2653. [Google Scholar] [CrossRef] [PubMed]
  161. Eng, M.L.; Stutchbury, B.J.M.; Morrissey, C.A. A Neonicotinoid Insecticide Reduces Fueling and Delays Migration in Songbirds. Science 2019, 365, 1177–1180. [Google Scholar] [CrossRef] [PubMed]
  162. Tassin de Montaigu, C.; Goulson, D. Identifying Agricultural Pesticides That May Pose a Risk for Birds. PeerJ 2020, 8, e9526. [Google Scholar] [CrossRef]
  163. Murthy, K.S.; Kiran, B.; Venkateshwarlu, M. A Review on Toxicity of Pesticides in Fish. Int. J. Open Scient. Res. 2013, 1, 15–36. [Google Scholar]
  164. Slaninova, A.; Smutna, M.; Modra, H.; Svobodova, Z. A Review: Oxidative Stress in Fish Induced by Pesticides. Neuro. Endocrinol. Lett. 2009, 30 (Suppl. 1), 2–12. [Google Scholar] [PubMed]
  165. Ullah, S.; Zorriehzahra, J. Ecotoxicology: A Review of Pesticides Induced Toxicity in Fish. Adv. Anim. Vet. Sci. 2015, 3, 40–57. [Google Scholar] [CrossRef]
  166. Grosell, M.; Pasparakis, C. Physiological Responses of Fish to Oil Spills. Ann. Rev. Mar. Sci. 2021, 13, 137–160. [Google Scholar] [CrossRef]
  167. Gong, Y.; Zhao, X.; Cai, Z.; O’Reilly, S.E.; Hao, X.; Zhao, D. A Review of Oil, Dispersed Oil and Sediment Interactions in the Aquatic Environment: Influence on the Fate, Transport and Remediation of Oil Spills. Mar. Pollut. Bull. 2014, 79, 16–33. [Google Scholar] [CrossRef]
  168. Langangen, Ø.; Olsen, E.; Stige, L.C.; Ohlberger, J.; Yaragina, N.A.; Vikebø, F.B.; Bogstad, B.; Stenseth, N.C.; Hjermann, D.Ø. The Effects of Oil Spills on Marine Fish: Implications of Spatial Variation in Natural Mortality. Mar. Pollut. Bull. 2017, 119, 102–109. [Google Scholar] [CrossRef]
  169. Gracia, A.; Murawski, S.A.; Vázquez-Bader, A.R. Impacts of Deep Oil Spills on Fish and Fisheries. In Deep Oil Spills: Facts, Fate, and Effects; Murawski, S.A., Ainsworth, C.H., Gilbert, S., Hollander, D.J., Paris, C.B., Schlüter, M., Wetzel, D.L., Eds.; Springer International Publishing: Cham, Switzerland, 2020; pp. 414–430. ISBN 978-3-030-11605-7. [Google Scholar]
  170. Overturf, M.D.; Anderson, J.C.; Pandelides, Z.; Beyger, L.; Holdway, D.A. Pharmaceuticals and Personal Care Products: A Critical Review of the Impacts on Fish Reproduction. Crit. Rev. Toxicol. 2015, 45, 469–491. [Google Scholar] [CrossRef] [PubMed]
  171. Xie, H.; Hao, H.; Xu, N.; Liang, X.; Gao, D.; Xu, Y.; Gao, Y.; Tao, H.; Wong, M. Pharmaceuticals and Personal Care Products in Water, Sediments, Aquatic Organisms, and Fish Feeds in the Pearl River Delta: Occurrence, Distribution, Potential Sources, and Health Risk Assessment. Sci. Total Environ. 2019, 659, 230–239. [Google Scholar] [CrossRef] [PubMed]
  172. Wardrop, P.; Shimeta, J.; Nugegoda, D.; Morrison, P.D.; Miranda, A.; Tang, M.; Clarke, B.O. Chemical Pollutants Sorbed to Ingested Microbeads from Personal Care Products Accumulate in Fish. Environ. Sci. Technol. 2016, 50, 4037–4044. [Google Scholar] [CrossRef] [PubMed]
  173. Kim, J.-W.; Ishibashi, H.; Yamauchi, R.; Ichikawa, N.; Takao, Y.; Hirano, M.; Koga, M.; Arizono, K. Acute Toxicity of Pharmaceutical and Personal Care Products on Freshwater Crustacean (Thamnocephalus Platyurus) and Fish (Oryzias Latipes). J. Toxicol. Sci. 2009, 34, 227–232. [Google Scholar] [CrossRef] [Green Version]
  174. Thompson, L.A.; Darwish, W.S. Environmental Chemical Contaminants in Food: Review of a Global Problem. J. Toxicol. 2019, 2019, e2345283. [Google Scholar] [CrossRef] [Green Version]
  175. Rather, I.A.; Koh, W.Y.; Paek, W.K.; Lim, J. The Sources of Chemical Contaminants in Food and Their Health Implications. Front. Pharmacol. 2017, 8, 830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Loonen, H.; van de Guchte, C.; Parsons, J.R.; de Voogt, P.; Govers, H.A.J. Ecological Hazard Assessment of Dioxins: Hazards to Organisms at Different Levels of Aquatic Food Webs (Fish-Eating Birds and Mammals, Fish and Invertebrates). Sci. Total Environ. 1996, 182, 93–103. [Google Scholar] [CrossRef]
  177. Zielińska, E.; Kocki, T.; Saran, T.; Borbely, S.; Kuc, D.; Vilagi, I.; Urbańska, E.M.; Turski, W.A. Effect of Pesticides on Kynurenic Acid Production in Rat Brain Slices. Ann. Agric. Environ. Med. 2005, 12, 177–179. [Google Scholar]
  178. Seifert, J.; Casida, J.E. Relation of Yolk Sac Membrane Kynurenine Formamidase Inhibition to Certain Teratogenic Effects of Organophosphorus Insecticides and of Carbaryl and Eserine in Chicken Embryos. Biochem. Pharmacol. 1978, 27, 2611–2615. [Google Scholar] [CrossRef]
  179. 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]
  180. Dharwadkar, S. Implications of Biotransformation of Benzo (a) Pyrene: A Review. Bionano Frontier 2011, 63–65. [Google Scholar]
  181. Hubbard, T.D.; Murray, I.A.; Perdew, G.H. Indole and Tryptophan Metabolism: Endogenous and Dietary Routes to Ah Receptor Activation. Drug Metab. Dispos. 2015, 43, 1522–1535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Novikov, O.; Wang, Z.; Stanford, E.A.; Parks, A.J.; Ramirez-Cardenas, A.; Landesman, E.; Laklouk, I.; Sarita-Reyes, C.; Gusenleitner, D.; Li, A.; et al. An Aryl Hydrocarbon Receptor-Mediated Amplification Loop That Enforces Cell Migration in ER-/PR-/Her2- Human Breast Cancer Cells. Mol. Pharmacol. 2016, 90, 674–688. [Google Scholar] [CrossRef] [Green Version]
  183. Rannug, A.; Rannug, U. The Tryptophan Derivative 6-Formylindolo [3,2-b]Carbazole, FICZ, a Dynamic Mediator of Endogenous Aryl Hydrocarbon Receptor Signaling, Balances Cell Growth and Differentiation. Crit. Rev. Toxicol. 2018, 48, 555–574. [Google Scholar] [CrossRef] [Green Version]
  184. Farmahin, R.; Crump, D.; Kennedy, S.W. Sensitivity of Avian Species to the Aryl Hydrocarbon Receptor Ligand 6-Formylindolo [3,2-b] Carbazole (FICZ). Chem. Biol. Interact. 2014, 221, 61–69. [Google Scholar] [CrossRef]
  185. Song, J.; Clagett-Dame, M.; Peterson, R.E.; Hahn, M.E.; Westler, W.M.; Sicinski, R.R.; DeLuca, H.F. A Ligand for the Aryl Hydrocarbon Receptor Isolated from Lung. Proc. Natl. Acad. Sci. USA 2002, 99, 14694–14699. [Google Scholar] [CrossRef] [Green Version]
  186. Sordillo, P.P.; Sordillo, L.A.; Helson, L. The Kynurenine Pathway: A Primary Resistance Mechanism in Patients with Glioblastoma. Anticancer Res. 2017, 37, 2159–2171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Morales-Puerto, N.; Giménez-Gómez, P.; Pérez-Hernández, M.; Abuin-Martínez, C.; Gil de Biedma-Elduayen, L.; Vidal, R.; Gutiérrez-López, M.D.; O’Shea, E.; Colado, M.I. Addiction and the Kynurenine Pathway: A New Dancing Couple? Pharmacol. Ther. 2021, 223, 107807. [Google Scholar] [CrossRef]
  188. Zhang, P.; Huang, H.; Gao, X.; Jiang, J.; Xi, C.; Wu, L.; Fu, Y.; Lai, J.; Hu, S. Involvement of Kynurenine Metabolism in Bipolar Disorder: An Updated Review. Front. Psychiatry 2021, 12, 677039. [Google Scholar] [CrossRef] [PubMed]
  189. Bartoli, F.; Misiak, B.; Callovini, T.; Cavaleri, D.; Cioni, R.M.; Crocamo, C.; Savitz, J.B.; Carrà, G. The Kynurenine Pathway in Bipolar Disorder: A Meta-Analysis on the Peripheral Blood Levels of Tryptophan and Related Metabolites. Mol. Psychiatry 2021, 26, 3419–3429. [Google Scholar] [CrossRef]
  190. Marx, W.; McGuinness, A.J.; Rocks, T.; Ruusunen, A.; Cleminson, J.; Walker, A.J.; Gomes-da-Costa, S.; Lane, M.; Sanches, M.; Diaz, A.P.; et al. The Kynurenine Pathway in Major Depressive Disorder, Bipolar Disorder, and Schizophrenia: A Meta-Analysis of 101 Studies. Mol. Psychiatry 2021, 26, 4158–4178. [Google Scholar] [CrossRef] [PubMed]
  191. Erhardt, S.; Schwieler, L.; Imbeault, S.; Engberg, G. The Kynurenine Pathway in Schizophrenia and Bipolar Disorder. Neuropharmacology 2017, 112, 297–306. [Google Scholar] [CrossRef]
  192. Hunt, C.; Macedo, E.; Cordeiro, T.; Suchting, R.; de Dios, C.; Cuellar Leal, V.A.; Soares, J.C.; Dantzer, R.; Teixeira, A.L.; Selvaraj, S. Effect of Immune Activation on the Kynurenine Pathway and Depression Symptoms—A Systematic Review and Meta-Analysis. Neurosci. Biobehav. Rev. 2020, 118, 514–523. [Google Scholar] [CrossRef]
  193. Brown, S.J.; Huang, X.-F.; Newell, K.A. The Kynurenine Pathway in Major Depression: What We Know and Where to Next. Neurosci. Biobehav. Rev. 2021, 127, 917–927. [Google Scholar] [CrossRef]
  194. Serafini, G.; Adavastro, G.; Canepa, G.; Capobianco, L.; Conigliaro, C.; Pittaluga, F.; Murri, M.B.; Valchera, A.; De Berardis, D.; Pompili, M.; et al. Abnormalities in Kynurenine Pathway Metabolism in Treatment-Resistant Depression and Suicidality: A Systematic Review. CNS Neurol. Disord. Drug Targets 2017, 16, 440–453. [Google Scholar] [CrossRef]
  195. Cao, B.; Chen, Y.; Ren, Z.; Pan, Z.; McIntyre, R.S.; Wang, D. Dysregulation of Kynurenine Pathway and Potential Dynamic Changes of Kynurenine in Schizophrenia: A Systematic Review and Meta-Analysis. Neurosci. Biobehav. Rev. 2021, 123, 203–214. [Google Scholar] [CrossRef]
  196. Pukoli, D.; Polyák, H.; Rajda, C.; Vécsei, L. Kynurenines and Neurofilament Light Chain in Multiple Sclerosis. Front. Neurosci. 2021, 15, 658202. [Google Scholar] [CrossRef]
  197. Sharma, V.K.; Singh, T.G.; Prabhakar, N.K.; Mannan, A. Kynurenine Metabolism and Alzheimer’s Disease: The Potential Targets and Approaches. Neurochem. Res. 2022, 47, 1459–1476. [Google Scholar] [CrossRef] [PubMed]
  198. Tanaka, M.; Bohár, Z.; Vécsei, L. Are Kynurenines Accomplices or Principal Villains in Dementia? Maintenance of Kynurenine Metabolism. Molecules 2020, 25, 564. [Google Scholar] [CrossRef] [Green Version]
  199. Venkatesan, D.; Iyer, M.; Narayanasamy, A.; Siva, K.; Vellingiri, B. Kynurenine Pathway in Parkinson’s Disease-an Update. Eneurologicalsci 2020, 21, 100270. [Google Scholar] [CrossRef] [PubMed]
  200. Behl, T.; Kaur, I.; Sehgal, A.; Singh, S.; Bhatia, S.; Al-Harrasi, A.; Zengin, G.; Bumbu, A.G.; Andronie-Cioara, F.L.; Nechifor, A.C.; et al. The Footprint of Kynurenine Pathway in Neurodegeneration: Janus-Faced Role in Parkinson’s Disorder and Therapeutic Implications. Int. J. Mol. Sci. 2021, 22, 6737. [Google Scholar] [CrossRef]
  201. Mor, A.; Tankiewicz-Kwedlo, A.; Krupa, A.; Pawlak, D. Role of Kynurenine Pathway in Oxidative Stress during Neurodegenerative Disorders. Cells 2021, 10, 1603. [Google Scholar] [CrossRef] [PubMed]
  202. Török, N.; Tanaka, M.; Vécsei, L. Searching for Peripheral Biomarkers in Neurodegenerative Diseases: The Tryptophan-Kynurenine Metabolic Pathway. Int. J. Mol. Sci. 2020, 21, 9338. [Google Scholar] [CrossRef]
  203. Curto, M.; Lionetto, L.; Fazio, F.; Mitsikostas, D.-D.; Martelletti, P. Fathoming the Kynurenine Pathway in Migraine: Why Understanding the Enzymatic Cascades Is Still Critically Important. Intern. Emerg. Med. 2015, 10, 413–421. [Google Scholar] [CrossRef] [PubMed]
  204. Fila, M.; Chojnacki, J.; Pawlowska, E.; Szczepanska, J.; Chojnacki, C.; Blasiak, J. Kynurenine Pathway of Tryptophan Metabolism in Migraine and Functional Gastrointestinal Disorders. Int. J. Mol. Sci. 2021, 22, 10134. [Google Scholar] [CrossRef]
  205. Martos, D.; Tuka, B.; Tanaka, M.; Vécsei, L.; Telegdy, G. Memory Enhancement with Kynurenic Acid and Its Mechanisms in Neurotransmission. Biomedicines 2022, 10, 849. [Google Scholar] [CrossRef]
  206. Tanaka, M.; Vécsei, L. Monitoring the Kynurenine System: Concentrations, Ratios or What Else? Adv. Clin. Exp. Med. 2021, 30, 775–778. [Google Scholar] [CrossRef] [PubMed]
  207. Dogrul, B.N. Indolamine 2,3-Dioxygenase (IDO) Inhibitors as a Potential Treatment for Somatic Symptoms. Med. Hypotheses 2022, 160, 110777. [Google Scholar] [CrossRef]
  208. Tanaka, M.; Török, N.; Tóth, F.; Szabó, Á.; Vécsei, L. Co-Players in Chronic Pain: Neuroinflammation and the Tryptophan-Kynurenine Metabolic Pathway. Biomedicines 2021, 9, 897. [Google Scholar] [CrossRef]
  209. Ciapała, K.; Mika, J.; Rojewska, E. The Kynurenine Pathway as a Potential Target for Neuropathic Pain Therapy Design: From Basic Research to Clinical Perspectives. Int. J. Mol. Sci. 2021, 22, 11055. [Google Scholar] [CrossRef]
  210. Jovanovic, F.; Candido, K.D.; Knezevic, N.N. The Role of the Kynurenine Signaling Pathway in Different Chronic Pain Conditions and Potential Use of Therapeutic Agents. Int. J. Mol. Sci. 2020, 21, 6045. [Google Scholar] [CrossRef] [PubMed]
  211. Heng, B.; Lim, C.K.; Lovejoy, D.B.; Bessede, A.; Gluch, L.; Guillemin, G.J. Understanding the Role of the Kynurenine Pathway in Human Breast Cancer Immunobiology. Oncotarget 2016, 7, 6506–6520. [Google Scholar] [CrossRef] [Green Version]
  212. Kiluk, M.; Lewkowicz, J.; Pawlak, D.; Tankiewicz-Kwedlo, A. Crosstalk between Tryptophan Metabolism via Kynurenine Pathway and Carbohydrate Metabolism in the Context of Cardio-Metabolic Risk-Review. J. Clin. Med. 2021, 10, 2484. [Google Scholar] [CrossRef]
  213. Song, P.; Ramprasath, T.; Wang, H.; Zou, M.-H. Abnormal Kynurenine Pathway of Tryptophan Catabolism in Cardiovascular Diseases. Cell. Mol. Life Sci. 2017, 74, 2899–2916. [Google Scholar] [CrossRef]
  214. Gáspár, R.; Halmi, D.; Demján, V.; Berkecz, R.; Pipicz, M.; Csont, T. Kynurenine Pathway Metabolites as Potential Clinical Biomarkers in Coronary Artery Disease. Front. Immunol. 2021, 12, 768560. [Google Scholar] [CrossRef]
  215. Razquin, C.; Ruiz-Canela, M.; Toledo, E.; Hernández-Alonso, P.; Clish, C.B.; Guasch-Ferré, M.; Li, J.; Wittenbecher, C.; Dennis, C.; Alonso-Gómez, A.; et al. Metabolomics of the Tryptophan-Kynurenine Degradation Pathway and Risk of Atrial Fibrillation and Heart Failure: Potential Modification Effect of Mediterranean Diet. Am. J. Clin. Nutr. 2021, 114, 1646–1654. [Google Scholar] [CrossRef] [PubMed]
  216. Melhem, N.J.; Taleb, S. Tryptophan: From Diet to Cardiovascular Diseases. Int. J. Mol. Sci. 2021, 22, 9904. [Google Scholar] [CrossRef] [PubMed]
  217. Engin, A.B.; Engin, A. The Interactions Between Kynurenine, Folate, Methionine and Pteridine Pathways in Obesity. In Obesity and Lipotoxicity; Engin, A.B., Engin, A., Eds.; Advances in Experimental Medicine and Biology; Springer International Publishing: Cham, Switzerland, 2017; pp. 511–527. ISBN 978-3-319-48382-5. [Google Scholar]
  218. Dadvar, S.; Ferreira, D.M.S.; Cervenka, I.; Ruas, J.L. The Weight of Nutrients: Kynurenine Metabolites in Obesity and Exercise. J. Intern. Med. 2018, 284, 519–533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Agus, A.; Planchais, J.; Sokol, H. Gut Microbiota Regulation of Tryptophan Metabolism in Health and Disease. Cell Host Microbe 2018, 23, 716–724. [Google Scholar] [CrossRef] [Green Version]
  220. 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]
  221. Sun, M.; Ma, N.; He, T.; Johnston, L.J.; Ma, X. Tryptophan (Trp) Modulates Gut Homeostasis via Aryl Hydrocarbon Receptor (AhR). Crit. Rev. Food Sci. Nutr. 2020, 60, 1760–1768. [Google Scholar] [CrossRef] [PubMed]
  222. Chen, L.-M.; Bao, C.-H.; Wu, Y.; Liang, S.-H.; Wang, D.; Wu, L.-Y.; Huang, Y.; Liu, H.-R.; Wu, H.-G. Tryptophan-Kynurenine Metabolism: A Link between the Gut and Brain for Depression in Inflammatory Bowel Disease. J. Neuroinflamm. 2021, 18, 135. [Google Scholar] [CrossRef]
  223. Krupa, A.; Kowalska, I. The Kynurenine Pathway-New Linkage between Innate and Adaptive Immunity in Autoimmune Endocrinopathies. Int. J. Mol. Sci. 2021, 22, 9879. [Google Scholar] [CrossRef]
  224. Haq, S.; Grondin, J.A.; Khan, W.I. Tryptophan-Derived Serotonin-Kynurenine Balance in Immune Activation and Intestinal Inflammation. FASEB J. 2021, 35, e21888. [Google Scholar] [CrossRef]
  225. Collier, M.E.; Zhang, S.; Scrutton, N.S.; Giorgini, F. Inflammation Control and Improvement of Cognitive Function in COVID-19 Infections: Is There a Role for Kynurenine 3-Monooxygenase Inhibition? Drug Discov. Today 2021, 26, 1473–1481. [Google Scholar] [CrossRef]
  226. Lawler, N.G.; Gray, N.; Kimhofer, T.; Boughton, B.; Gay, M.; Yang, R.; Morillon, A.-C.; Chin, S.-T.; Ryan, M.; Begum, S.; et al. Systemic Perturbations in Amine and Kynurenine Metabolism Associated with Acute SARS-CoV-2 Infection and Inflammatory Cytokine Responses. J. Proteome Res. 2021, 20, 2796–2811. [Google Scholar] [CrossRef] [PubMed]
  227. Wee, H.N.; Liu, J.-J.; Ching, J.; Kovalik, J.-P.; Lim, S.C. The Kynurenine Pathway in Acute Kidney Injury and Chronic Kidney Disease. Am. J. Nephrol. 2021, 52, 771–787. [Google Scholar] [CrossRef]
  228. Mor, A.; Kalaska, B.; Pawlak, D. Kynurenine Pathway in Chronic Kidney Disease: What’s Old, What’s New, and What’s Next? Int. J. Tryptophan Res. IJTR 2020, 13, 1178646920954882. [Google Scholar] [CrossRef] [PubMed]
  229. Krishnamurthy, S.; Gilot, D.; Ahn, S.B.; Lam, V.; Shin, J.-S.; Guillemin, G.J.; Heng, B. Involvement of Kynurenine Pathway in Hepatocellular Carcinoma. Cancers 2021, 13, 5180. [Google Scholar] [CrossRef]
  230. Miyazaki, T.; Chung, S.; Sakai, H.; Ohata, H.; Obata, Y.; Shiokawa, D.; Mizoguchi, Y.; Kubo, T.; Ichikawa, H.; Taniguchi, H.; et al. Stemness and Immune Evasion Conferred by the TDO2-AHR Pathway Are Associated with Liver Metastasis of Colon Cancer. Cancer Sci. 2022, 113, 170–181. [Google Scholar] [CrossRef]
  231. Martin, K.S.; Azzolini, M.; Lira Ruas, J. The Kynurenine Connection: How Exercise Shifts Muscle Tryptophan Metabolism and Affects Energy Homeostasis, the Immune System, and the Brain. Am. J. Physiol. Cell Physiol. 2020, 318, C818–C830. [Google Scholar] [CrossRef] [PubMed]
  232. Sui, G.; Jia, L.; Quan, D.; Zhao, N.; Yang, G. Activation of the Gut Microbiota-Kynurenine-Liver Axis Contributes to the Development of Nonalcoholic Hepatic Steatosis in Nondiabetic Adults. Aging 2021, 13, 21309–21324. [Google Scholar] [CrossRef] [PubMed]
  233. Dhillon, A.K.; Rupp, C.; Bergquist, A.; Voitl, R.; Folseraas, T.; Trøseid, M.; Midttun, Ø.; Ueland, P.M.; Karlsen, T.H.; Vesterhus, M.; et al. Associations of Neopterin and Kynurenine–Tryptophan Ratio with Survival in Primary Sclerosing Cholangitis. Scand. J. Gastroenterol. 2021, 56, 443–452. [Google Scholar] [CrossRef] [PubMed]
  234. Clària, J.; Moreau, R.; Fenaille, F.; Amorós, A.; Junot, C.; Gronbaek, H.; Coenraad, M.J.; Pruvost, A.; Ghettas, A.; Chu-Van, E.; et al. Orchestration of Tryptophan-Kynurenine Pathway, Acute Decompensation, and Acute-on-Chronic Liver Failure in Cirrhosis. Hepatology 2019, 69, 1686–1701. [Google Scholar] [CrossRef]
  235. Kardashian, A.; Ma, Y.; Yin, M.T.; Scherzer, R.; Nolan, O.; Aweeka, F.; Tien, P.C.; Price, J.C. High Kynurenine:Tryptophan Ratio Is Associated With Liver Fibrosis in HIV-Monoinfected and HIV/Hepatitis C Virus–Coinfected Women. Open Forum Infect. Dis. 2019, 6, ofz281. [Google Scholar] [CrossRef]
  236. Ogbechi, J.; Clanchy, F.I.; Huang, Y.-S.; Topping, L.M.; Stone, T.W.; Williams, R.O. IDO Activation, Inflammation and Musculoskeletal Disease. Exp. Gerontol. 2020, 131, 110820. [Google Scholar] [CrossRef]
  237. Vyavahare, S.; Kumar, S.; Cantu, N.; Kolhe, R.; Bollag, W.B.; McGee-Lawrence, M.E.; Hill, W.D.; Hamrick, M.W.; Isales, C.M.; Fulzele, S. Tryptophan-Kynurenine Pathway in COVID-19-Dependent Musculoskeletal Pathology: A Minireview. Mediators Inflamm. 2021, 2021, 2911578. [Google Scholar] [CrossRef]
  238. Broekhuizen, M.; Danser, A.H.J.; Reiss, I.K.M.; Merkus, D. The Function of the Kynurenine Pathway in the Placenta: A Novel Pharmacotherapeutic Target? Int. J. Environ. Res. Public. Health 2021, 18, 11545. [Google Scholar] [CrossRef]
  239. Silvano, A.; Seravalli, V.; Strambi, N.; Cecchi, M.; Tartarotti, E.; Parenti, A.; Di Tommaso, M. Tryptophan Metabolism and Immune Regulation in the Human Placenta. J. Reprod. Immunol. 2021, 147, 103361. [Google Scholar] [CrossRef]
  240. Badawy, A.A.-B. Tryptophan Metabolism, Disposition and Utilization in Pregnancy. Biosci. Rep. 2015, 35, e00261. [Google Scholar] [CrossRef]
  241. Xu, K.; Liu, G.; Fu, C. The Tryptophan Pathway Targeting Antioxidant Capacity in the Placenta. Oxid. Med. Cell. Longev. 2018, 2018, 1054797. [Google Scholar] [CrossRef] [PubMed]
  242. Worton, S.A.; Greenwood, S.L.; Wareing, M.; Heazell, A.E.; Myers, J. The Kynurenine Pathway; A New Target for Treating Maternal Features of Preeclampsia? Placenta 2019, 84, 44–49. [Google Scholar] [CrossRef]
  243. Gumusoglu, S.; Scroggins, S.; Vignato, J.; Santillan, D.; Santillan, M. The Serotonin-Immune Axis in Preeclampsia. Curr. Hypertens. Rep. 2021, 23, 37. [Google Scholar] [CrossRef]
  244. Ala, M. The Footprint of Kynurenine Pathway in Every Cancer: A New Target for Chemotherapy. Eur. J. Pharmacol. 2021, 896, 173921. [Google Scholar] [CrossRef] [PubMed]
  245. Castro-Portuguez, R.; Sutphin, G.L. Kynurenine Pathway, NAD+ Synthesis, and Mitochondrial Function: Targeting Tryptophan Metabolism to Promote Longevity and Healthspan. Exp. Gerontol. 2020, 132, 110841. [Google Scholar] [CrossRef]
  246. Savitz, J. The Kynurenine Pathway: A Finger in Every Pie. Mol. Psychiatry 2020, 25, 131–147. [Google Scholar] [CrossRef]
  247. Badawy, A.A.-B. Kynurenine Pathway and Human Systems. Exp. Gerontol. 2020, 129, 110770. [Google Scholar] [CrossRef]
  248. Lu, Y.; Shao, M.; Wu, T. Kynurenine-3-Monooxygenase: A New Direction for the Treatment in Different Diseases. Food Sci. Nutr. 2020, 8, 711–719. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Key enzymes regulating TRP metabolism. The pathway depicting key regulators and targets that play a role on the TRP metabolism with a connection to KYN and 5-HT. Figure created on biorender.com.
Figure 1. Key enzymes regulating TRP metabolism. The pathway depicting key regulators and targets that play a role on the TRP metabolism with a connection to KYN and 5-HT. Figure created on biorender.com.
Ijms 23 06300 g001
Figure 2. A review of the literature by (A) species and (B) physiological processes/pathways.
Figure 2. A review of the literature by (A) species and (B) physiological processes/pathways.
Ijms 23 06300 g002
Figure 3. A review of kynurenine pathway (A) enzymes and (B) metabolites detected across animal kingdoms. If both gene and protein data was available, it was considered detected, and labelled dark pink. If enzymatic and metabolite orthologs may be present (via. computational curation of databases) but have not yet been confirmed in gene or protein levels, it was labelled light pink. If no information could be found yet, it was considered unknown and labelled white.
Figure 3. A review of kynurenine pathway (A) enzymes and (B) metabolites detected across animal kingdoms. If both gene and protein data was available, it was considered detected, and labelled dark pink. If enzymatic and metabolite orthologs may be present (via. computational curation of databases) but have not yet been confirmed in gene or protein levels, it was labelled light pink. If no information could be found yet, it was considered unknown and labelled white.
Ijms 23 06300 g003
Figure 4. Across species, mammals, birds, and fish have similar responses to stress induced changes in TRP-KYN metabolism. Stress responses include increased GC and catecholamine signaling, which elevate TRP metabolism through the KYN pathway, as indicated by the levels of downstream metabolites. While it is generally accepted that this occurs through hepatic TDO induction in mammals, the molecular mechanism is not yet clear in other vertebrate species. Figure created on biorender.com.
Figure 4. Across species, mammals, birds, and fish have similar responses to stress induced changes in TRP-KYN metabolism. Stress responses include increased GC and catecholamine signaling, which elevate TRP metabolism through the KYN pathway, as indicated by the levels of downstream metabolites. While it is generally accepted that this occurs through hepatic TDO induction in mammals, the molecular mechanism is not yet clear in other vertebrate species. Figure created on biorender.com.
Ijms 23 06300 g004
Figure 5. TRP functions as a regulator of inflammatory signaling molecules that control the immune response and the antioxidant response to cell stress and tissue damage. Figure created on biorender.com.
Figure 5. TRP functions as a regulator of inflammatory signaling molecules that control the immune response and the antioxidant response to cell stress and tissue damage. Figure created on biorender.com.
Ijms 23 06300 g005
Figure 6. Alterations in TRP metabolism and KYN metabolites may be an indication of exposure to environmental contaminants. Many environmental contaminants (i.e., PACs) are known to act as AhR ligands and are also known to regulate IDO and TDO expression, thereby affecting the production of immunomodulatory KYN metabolites. Moreover, many of these metabolites can also act as AhR ligands and induce AhR target genes. Figure created on biorender.com.
Figure 6. Alterations in TRP metabolism and KYN metabolites may be an indication of exposure to environmental contaminants. Many environmental contaminants (i.e., PACs) are known to act as AhR ligands and are also known to regulate IDO and TDO expression, thereby affecting the production of immunomodulatory KYN metabolites. Moreover, many of these metabolites can also act as AhR ligands and induce AhR target genes. Figure created on biorender.com.
Ijms 23 06300 g006
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jamshed, L.; Debnath, A.; Jamshed, S.; Wish, J.V.; Raine, J.C.; Tomy, G.T.; Thomas, P.J.; Holloway, A.C. An Emerging Cross-Species Marker for Organismal Health: Tryptophan-Kynurenine Pathway. Int. J. Mol. Sci. 2022, 23, 6300. https://doi.org/10.3390/ijms23116300

AMA Style

Jamshed L, Debnath A, Jamshed S, Wish JV, Raine JC, Tomy GT, Thomas PJ, Holloway AC. An Emerging Cross-Species Marker for Organismal Health: Tryptophan-Kynurenine Pathway. International Journal of Molecular Sciences. 2022; 23(11):6300. https://doi.org/10.3390/ijms23116300

Chicago/Turabian Style

Jamshed, Laiba, Amrita Debnath, Shanza Jamshed, Jade V. Wish, Jason C. Raine, Gregg T. Tomy, Philippe J. Thomas, and Alison C. Holloway. 2022. "An Emerging Cross-Species Marker for Organismal Health: Tryptophan-Kynurenine Pathway" International Journal of Molecular Sciences 23, no. 11: 6300. https://doi.org/10.3390/ijms23116300

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop