Elsevier

Neuropharmacology

Volume 38, Issue 8, August 1999, Pages 1225-1233
Neuropharmacology

Quinolinic acid formation in immune-activated mice: studies with (m-nitrobenzoyl)-alanine (mNBA) and 3,4-dimethoxy-[-N-4-(-3-nitrophenyl) thiazol-2yl]-benzenesulfonamide (Ro 61-8048), two potent and selective inhibitors of kynurenine hydroxylase

https://doi.org/10.1016/S0028-3908(99)00048-9Get rights and content

Abstract

The role of kynurenine hydroxylase activity in the neo-formation of the excitotoxin quinolinic acid (QUIN) has been studied in mice by using (m-nitrobenzoyl)-alanine (mNBA) and 3,4-dimethoxy-[-N-4-(-3-nitrophenyl) thiazol-2yl]-benzenesulfonamide (Ro 61-8048), two potent and selective inhibitors of this enzyme. Immune-stimulation with pokeweed mitogen (PWM, 200 μg i.v., 12 h) induced a robust increase in kynurenine (KYN) and its metabolites kynurenic acid (KYNA) and QUIN in blood and brain. When incubated in a medium containing KYN but not tryptophan, spleen, lung and liver (but not brain) slices accumulated a measurable amount of QUIN in the supernatant. Slices obtained from PWM treated animals had a ten-fold increase in QUIN accumulation in spleen, no changes in lung and a 40% decrease in liver, suggesting that the spleen contributes to the increased QUIN levels found in the blood and brain of immune-stimulated mice. Large doses of kynurenine hydroxylase inhibitors increased KYN and KYNA, but unexpectedly did not decrease QUIN content in control blood and brain. When tested in organ slices obtained from either controls or immune-stimulated animals, mNBA (1–1000 μM) and Ro 61-8048 (0.1–100 μM) strongly reduced QUIN neo-formation, suggesting that, in vitro, kynurenine hydroxylase activity is required for QUIN neosynthesis. Indeed, after repeated doses of mNBA or Ro 61-8048, QUIN content in blood and brain of immune-stimulated animals significantly decreased. Our results suggest that, under basal conditions, sufficient QUIN synthesis may occur through kynurenine hydroxylase-independent pathways. In immune-stimulated animals, however, kynurenine hydroxylase inhibitors significantly reduce blood and brain accumulation of QUIN.

Introduction

Quinolinic acid (QUIN), a tryptophan (TRP) metabolite present in the brain (Moroni et al., 1984, Wolfensberger et al., 1984), is able to interact with glutamate receptors of the NMDA type and to cause convulsions or neurodegeneration (Stone and Perkins, 1981, Foster et al., 1983, Schwarcz et al., 1983). Its synthesis occurs along the so-called kynurenine metabolic pathway, which also leads to the formation of other neuroactive compounds such as 3-hydroxykynurenine (3OH-KYN), another metabolite able to cause neuronal damage (necrosis and apoptosis) in cell cultures (Eastman and Guilarte, 1989, Okuda et al., 1998) and kynurenic acid (KYNA), an antagonist of excitatory amino acid receptors (Perkins and Stone, 1982, Foster et al., 1984, Moroni et al., 1988).

The rate limiting step in the formation of the above mentioned neuroactive compounds is the oxidative cleavage of the pyrrole ring of TRP (Yoshida and Hayashi, 1987). This cleavage is catalyzed by two enzymes, tryptophan dioxygenase [E.C. 1.13.11.11], which is present only in liver, and indoleamine dioxygenase (IDO) [E.C. 1.13.11.17], which has a wider distribution and is also present in brain. The other enzymes required for the synthesis of QUIN are kynurenine hydroxylase [E.C. 1.14.13.9], kynureninase [E.C 7.3.1.3] and 3-hydroxyanthranilic acid dioxygenase [E.C. 1.13.11.6] (see Fig. 1). A large increase in the content of kynurenine (KYN) metabolites, including QUIN, has been repeatedly shown in blood and in central nervous system (CNS) of mammals affected by inflammatory neurological disorders (Heyes et al., 1991, Heyes et al., 1992a, Heyes et al., 1993, Moffet et al., 1993, Saito et al., 1993b) since these enzymes are strongly induced in immune-activated macrophages and microglia (Werner et al., 1988, Saito et al., 1992, Saito et al., 1993a). It has also been proposed that QUIN accumulation in the CNS may have neuropathological significance because, in the course of several encephalopathies, a direct correlation between its levels in cerebrospinal fluid (CSF) and the severity of the neurological deficits has been found (Heyes et al., 1991, Heyes et al., 1992a, Flanagan et al., 1995, Blight et al., 1997). We have previously studied the neuropharmacological and biochemical effects of a number of kynurenine hydroxylase and/or kynureninase inhibitors (Moroni et al., 1991, Russi et al., 1992, Carpenedo et al., 1994, Chiarugi et al., 1995, Chiarugi et al., 1996) since compounds able to inhibit the kynurenine pathway enzymes could be of therapeutic importance (Kerr et al., 1997). In particular we characterized m-nitrobenzoylalanine (mNBA; Fig. 1), a selective kynurenine hydroxylase inhibitor (IC50 1 μM) able to increase the convulsive threshold in mice, cause sedation and mild analgesia, and increase KYN and KYNA content in the rodent brain (Carpenedo et al., 1994). Recently, a new selective, specific and potent kynurenine hydroxylase inhibitor was described, 3,4-dimethoxy-[N-4-(-3-nitrophenyl)thiazol-2-yl]-benzenesulfonamide (Ro 61-8048; IC50 0.04 μM;) (Roever et al., 1997). We took advantage of the availability of this molecule to study, in vitro and in vivo, the importance of KYN hydroxylation in QUIN synthesis.

Section snippets

Materials

mNBA and Ro 61-8048 were kindly provided by Drs S. Roever and A. Cesura (Hoffman-La Roche, Basel, CH). [137C]-QUIN was provided by Dr J.F. Reinhard Jr. (Research Triangle Park, North Carolina, USA).The TRP-depleted medium was from GIBCO Life Technologies Inc. (Gaithersburg, MD, USA). l-KYN, KYNA, QUIN and pokeweed mitogen (PWM) were from Sigma (St. Louis, MO, USA). Hexafluor-2-propanol and trifluoroacetyl-imidazole were from Aldrich (Milan, Italy). All remaining compounds were from Merck

Effects of mNBA and Ro 61-8048 on the content of KYN metabolites in brain and blood of PWM-treated mice

Kynurenine hydroxylase inhibitor administration has been previously shown to increase KYN and KYNA content in rat and gerbil brains (Carpenedo et al., 1994, Speciale et al., 1996, Roever et al., 1997). Table 1 shows that the administration of mNBA (400 mg/kg, i.p.) or Ro 61-8048 (60 mg/kg, i.p.) increased brain KYN content by two or three fold and brain KYNA content by nine or 15-fold, respectively in mice as well. To our surprise, brain QUIN content did not change after administration of the

Discussion

The most interesting finding of the present study is the demonstration that repeated treatments with large doses of mNBA or Ro 61-8048, two potent and selective kynurenine hydroxylase inhibitors, diminish the PWM-induced increase of QUIN content in blood and brain of mice (see Fig. 3). We also showed that, mNBA or Ro 61-8048 do not modify basal QUIN content, but significantly increase KYN and KYNA content in the blood and brain of saline pretreated animals (see Table 1). This observation could

Acknowledgements

This work was supported by: University of Florence; MURST: CNR, European Union Grants No B104-CT96-0049 and B104-CT960752 and Research Project on Multiple Sclerosis of the Istituto Superiore di Sanità, Rome, Italy.

References (40)

  • K. Saito et al.

    Effects of immune activation on quinolinic acid and kynurenine pathway metabolism in the mouse

    Neuroscience

    (1992)
  • K. Saito et al.

    A mechanism for increased quinolinic acid formation following acute systemic immune stimulation

    J. Biol. Chem.

    (1993)
  • T.W. Stone et al.

    Quinolinic acid: a potent, endogenous excitant at amino acid receptors in CNS

    Eur. J. Pharmacol.

    (1981)
  • O. Takikawa et al.

    Tryptophan degradation in mice initiated by indoleamine 2,3-dioxygenase

    J. Biol. Chem.

    (1986)
  • R. Yoshida et al.

    Indoleamine 2,3-dioxygenase

    Methods Enzymol.

    (1987)
  • H. Baran et al.

    Presence of 3-hydroxyanthranilic acid in rat tissues and evidences for its synthesis from anthranilic acid in the brain

    J. Neurochem.

    (1990)
  • B. Beutler et al.

    Cachectin and tumor necrosis factor as two sides of the same biological coin

    Nature

    (1986)
  • A.R. Blight et al.

    Quinolinic acid accumulation in injured spinal cord: time course, distribution and species differences between rat and guinea pig

    J. Neurotrauma

    (1997)
  • A. Chiarugi et al.

    Comparison of the neurochemical and behavioral effects resulting from the inhibition of kynurenine hydroxylase and/or kynureninase

    J. Neurochem.

    (1995)
  • A. Chiarugi et al.

    Kynurenine disposition in blood and brain of mice: effects of selective inhibitors of kynurenine hydroxylase and of kynureninase

    J. Neurochem.

    (1996)
  • Cited by (36)

    • Kynurenine emerges from the shadows – Current knowledge on its fate and function

      2021, Pharmacology and Therapeutics
      Citation Excerpt :

      These findings clearly indicate that excess of KYN resulting from KMO inhibition is redirected at least in part to other branches of the kynurenine pathway, thus leading to an increase of kynurenine metabolites such as kynurenic acid and anthranilic acid. In contrast, acute administration of the KYNU inhibitor, oMBA, produced only moderate increase of KYN in plasma (up to 151% of its control level), as well as elevation of kynurenic acid (ranging 183–217%) and 3-HK (up to 172%) (Chiarugi et al., 1995; Chiarugi & Moroni, 1999; Moroni, Carpenedo, & Chiarugi, 1996). Moreover, in the brain, content of KYN, kynurenic acid and 3-HK was elevated up to 112%, 167% and 487% of its control level, respectively (Chiarugi et al., 1995 and 1996).

    • Addiction and the kynurenine pathway: A new dancing couple?

      2021, Pharmacology and Therapeutics
      Citation Excerpt :

      A low brain penetrability could be advantageous by maximizing clinical benefit and managing toxicity in combination therapies (Reinhart & Kelly, 2011). Although there is concern that the increase in brain KYN, in the absence of brain KMO inhibition, might lead to an increased conversion to QA (Muneer, 2020) this does not appear to be the case since QA levels in the brains of mice were unaltered following treatment with Ro 61–8048 (Chiarugi & Moroni, 1999) or JM6 (a prodrug of Ro 61–8048; Zwilling et al., 2011). Thus, peripheral inhibition of KMO appears to be a valuable mechanism for increasing levels of KYN and KYNA in the CNS.

    • Characterization of the Effects of L-4-Chlorokynurenine on Nociception in Rodents

      2017, Journal of Pain
      Citation Excerpt :

      This is because 4-Cl-KYN is also metabolized to 4-Cl-3-hydroxyanthranilic acid,21 a highly potent, selective inhibitor of 3-hydroxyanthranilic acid oxygenase (IC50: approximately 6 nM), the immediate biosynthetic enzyme of the excitotoxic NMDA receptor agonist QUIN.50,55 Elevated QUIN levels may be causally involved in various neurodegenerative processes and diseases58,60-64 and are believed to be especially significant when the immune system—and consequently the neosynthesis of QUIN—is activated.2,10,20,31,51,53 The transformation of systemically applied 4-Cl-KYN to 4-Cl-3-hydroxyanthranilic acid may therefore play an especially relevant role in the antinociceptive effects seen in pain models that are associated with an inflammation-related accumulation of QUIN in the spinal cord.25,68

    • Inhibition of the kynurenine pathway protects against reactive microglial-associated reductions in the complexity of primary cortical neurons

      2017, European Journal of Pharmacology
      Citation Excerpt :

      As such it was used in this experiment as an inhibitor of IDO gene induction as opposed to inhibiting enzyme activity as is the case with 1-MT (L). Ro 61–8048 is a widely used KMO inhibitor and has been shown to reduce the levels of quinolinic acid in the blood and brain of mice in the presence of strong immune stimulation (Chiarugi and Moroni, 1999). MK-801 is a potent, non-competitive antagonist of the NMDA receptor, and is highly selective for this receptor over other glutamatergic receptors (Wong et al., 1986).

    • Modified neocortical and cerebellar protein expression and morphology in adult rats following prenatal inhibition of the kynurenine pathway

      2014, Brain Research
      Citation Excerpt :

      We have shown previously that when given during gestation this compound produces a substantial elevation of kynurenic acid concentration in the embryo after 5 h. Levels are increased up to 100-fold, similar to findings in adult, non-pregnant animals (Chiarugi et al., 1995; Speciale et al., 1996; Rover et al., 1997; Clark et al., 2005) and would block glutamate receptors throughout the CNS (Perkins and Stone, 1982, Stone and Darlington, 2002; Stone et al., 2013). Endogenous concentrations of quinolinic acid are not normally affected, as reported in previous studies using this inhibitor (Chiarugi and Moroni, 1999; Clark et al., 2005) or after the genetic deletion of KMO (Giorgini et al., 2013). The reason for this remains an enigma and may suggest the existence of an alternative pathway for quinolinic acid synthesis such as the kynureninase shunt via anthranilic acid proposed by Baran and Schwarcz (1990).

    View all citing articles on Scopus
    View full text