Effect of indoleamine dioxygenase-1 deficiency and kynurenine pathway inhibition on murine cerebral malaria

https://doi.org/10.1016/j.ijpara.2008.10.005Get rights and content

Abstract

Cerebral malaria (CM) can be a fatal manifestation of Plasmodium falciparum infection. In this study, two different approaches were used to examine the role of indoleamine 2,3-dioxygenase-1 (IDO-1) and its metabolites in the development of murine CM. Mice genetically deficient in IDO-1 were not protected against CM, but partial protection was observed in C57BL/6 mice treated with Ro 61-8048, an inhibitor of kynurenine-3-hydroxylase. This protection was associated with suppressed levels of picolinic acid (PA) within the brain, but not with changes in the levels of kynurenic acid (KA) or quinolinic acid (QA). These data suggest that although IDO-1 is not directly involved in the pathogenesis of CM in C57BL/6 mice, the production of the kynurenine pathway metabolite PA may contribute to the development of murine CM.

Introduction

Malaria remains an important cause of mortality and morbidity, particularly for children living in sub-Saharan Africa. Cerebral malaria (CM) can be a serious neurological complication arising from Plasmodium falciparum infection, with patients presenting with convulsions, impaired consciousness, coma and death (WHO, 2000). Murine models of malaria are important tools in studying the pathogenesis of malaria, with numerous clinical and histopathological similarities described between human and murine malaria (Hunt and Grau, 2003).

While the pathogenesis of CM is incompletely understood, indoleamine 2,3-dioxygenase (INDO or IDO-1), the initial and rate-limiting enzyme of the kynurenine pathway, is thought to be involved. IDO-1 is a redox-regulated enzyme (Thomas et al., 2001) that converts tryptophan to N-formylkynurenine, which is metabolised to kynurenine and a cascade of biologically active molecules. Recently we have identified and characterised a new isoform of IDO-1, indoleamine 2,3-dioxygenase-like protein 1 (INDOL1 or IDO-2) (Ball et al., 2007). The role of the kynurenine pathway has been examined previously in human (Dobbie et al., 2000, Medana et al., 2002, Medana et al., 2003) and murine (Sanni et al., 1998, Hansen et al., 2004) CM. IDO-1 activity is increased in the brain during CM in CBA mice (Sanni et al., 1998) and clear correlations have been drawn between CM and enhanced levels of kynurenine pathway metabolites, including the neuroprotectant kynurenic acid (KA), the neuroexcitotoxin quinolinic acid (QA) and the proinflammatory molecule picolinic acid (PA) (Sanni et al., 1998, Dobbie et al., 2000, Medana et al., 2002, Medana et al., 2003). IDO-2 also might contribute to the pathogenesis of CM, as it shares similarities with IDO-1, including the ability to catalyse the conversion of tryptophan to kynurenine (Ball et al., 2007).

The endothelium is the predominant site of IDO-1 expression during malaria (Hansen et al., 2004) and this is may be part of the host protective response to Plasmodium infection (Hunt et al., 2006), since IDO-1 activity and immunoreactivity, as well as increased levels of kynurenine metabolites, are also found in the brain during the late-stages of a non-cerebral form of malaria (NCM) (Sanni et al., 1998, Hansen et al., 2004). Metabolites that might have tissue protective effects include the neuroprotectant KA, the NAD needed to counter the failure of aerobic glucose metabolism seen in CM (Rae et al., 2004), and the antioxidant 3-hydroxyanthranilic acid (Christen et al., 1990) given that reactive oxygen species have been proposed to be involved in the pathogenesis of malaria (Clark et al., 1986). The source of such reactive oxygen species is not, however, NADPH oxidase (Potter et al., 2005). An imbalance in the ratio of QA:KA, with heightened increases in QA levels compared with KA, has been described in CM in the CBA strain of mice (Sanni et al., 1998). Moreover, chemical inhibition of kynurenine-3-hydroxylase can confer protection against murine CM through its ability to increase KA and decrease PA levels in the brain (Clark et al., 2005). Overall, these studies suggest an important role for IDO-1 in the pathogenesis of CM, and are reinforced by the relationship between IDO-1 and IFN-γ, which is both an important mediator of CM and an inducer of IDO-1 (Grau et al., 1989, Sanni et al., 1998).

While a causal link between CM and these neuroactive metabolites of the kynurenine pathway has been hypothesised, the recent generation of IDO-1−/− mice (Mellor et al., 2003) allows direct experimental evaluation of the role of IDO-1 in murine CM. Moreover, Ro 61-8048, a compound that is able to inhibit kynurenine-3-hydroxylase, offers further insight into the influence of kynurenine pathway metabolites on the pathogenesis of murine CM. Here we describe partial protection against CM in PbA-infected C57BL/6 mice that were administered Ro 61-8048 but not in mice genetically deficient in IDO-1. Significantly, increased levels of PA correlated with the development of murine CM in both experimental groups, and this occurred independently of compensatory increases in either Ido-2 or tryptophan dioxygenase (Tdo) mRNAs. Brain levels of KA and QA did not correlate with the development of murine CM. These results provide evidence for the involvement of peripheral sources of kynurenine pathway metabolites, for example PA, in the pathogenesis of CM in C57BL/6 mice.

Section snippets

Murine models of malaria

six- to eight- week-old female C57BL/6 (WT) and IDO-1−/− mice were housed in the Blackburn Animal House, University of Sydney and given food and water ad libitum. Inoculation of C57BL/6 mice (i.p.) with 1 × 106 Plasmodium berghei ANKA (PbA) parasitised red blood cells (courtesy of Prof. G. Grau, University of Sydney, Australia) is a well described model of murine CM, in which mice display neurological symptoms by days 6–7 p.i. (Ma et al., 1996). The parasite used has not been cloned and is

IDO-1−/− mice are not protected from murine CM

Susceptibility to murine CM was followed in C57BL/6 (WT) and IDO-1−/− mice after inoculation with PbA. All WT (n = 17) and IDO-1−/− (n = 15) mice developed murine CM by day 6 p.i., with similar levels of parasite burden and showing typical clinical signs of murine CM, including brain histopathological features (e.g. petechial haemorrhages, oedema and leukocyte adherence; data not shown). Uninfected mice did not show these features. Although Ido-1 mRNA is up-regulated in WT kidney and liver tissue

Discussion

The results from this study show that IDO-1 is not directly involved in the pathogenesis of CM in C57BL/6 mice, since genetic deficiency in IDO-1 did not confer any protection against CM, with mice developing similar clinical and histopathological features to their WT counterparts. Levels of KA were not found to increase during CM in these mice, in contrast to the CBA/T6 strain (Sanni et al., 1998), and were considerably reduced in IDO-1−/− mice. On the other hand, QA was found to increase

Acknowledgements

We thank Cacang Suarna for help with the KA assays. This work was supported by Grants from the National Health and Medical Research Council of Australia and Australian Research Council to NH. JM was supported by an Australian Postgraduate Award. HB was a Rolf Edgar Lake Research Fellow of the Faculty of Medicine, University of Sydney.

References (45)

  • G. Melillo et al.

    Regulation of nitric-oxide synthase mRNA expression by interferon-γ and picolinic acid

    J. Biol. Chem.

    (1994)
  • S.M. Potter et al.

    A role for Fas-Fas ligand interactions during the late-stage neuropathological processes of experimental cerebral malaria

    J. Neuroimmunol.

    (2006)
  • K. Saito et al.

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

    J. Biol. Chem.

    (1993)
  • G.A. Smythe et al.

    Concurrent quantification of quinolinic, picolinic, and nicotinic acids using electron-capture negative-ion gas chromatography-mass spectrometry

    Anal. Biochem.

    (2002)
  • R. Adam et al.

    Role of human brain microvascular endothelial cells during central nervous system infection. Significance of indoleamine 2, 3-dioxygenase in antimicrobial defence and immunoregulation

    Thromb. Haemost.

    (2005)
  • H.J. Ball et al.

    Cyclooxygenase-2 in the pathogenesis of murine cerebral malaria

    J. Infect. Dis.

    (2004)
  • M.C. Bosco et al.

    The tryptophan catabolite picolinic acid selectively induces the chemokines macrophage inflammatory protein-1α and -1β in macrophages

    J. Immunol.

    (2000)
  • S. Christen et al.

    Antioxidant activities of some tryptophan metabolites: possible implication for inflammatory diseases

    Proc. Natl. Acad. Sci. USA

    (1990)
  • C.J. Clark et al.

    Prolonged survival of a murine model of cerebral malaria by kynurenine pathway inhibition

    Infect. Immun.

    (2005)
  • A. Cozzi et al.

    Kynurenine hydroxylase inhibitors reduce ischemic brain damage: studies with (m-nitrobenzoyl)-alanine (mNBA) and 3, 4-dimethoxy-[-N-4-(nitrophenyl)thiazol-2yl]-benzenesulfonamide (Ro 61–8048) in models of focal or global brain ischemia

    J. Cereb. Blood Flow Metab.

    (1999)
  • J.H. Curfs et al.

    Immunological aspects of cerebral lesions in murine malaria

    Clin. Exp. Immunol.

    (1989)
  • M. Dobbie et al.

    Cerebrospinal fluid studies in children with cerebral malaria: an excitotoxic mechanism?

    Am. J. Trop. Med. Hyg.

    (2000)
  • Cited by (22)

    • Malaria systems immunology: Plasmodium vivax induces tolerance during primary infection through dysregulation of neutrophils and dendritic cells

      2018, Journal of Infection
      Citation Excerpt :

      IDO1 has been identified as an important immunoregulator inhibiting T-cell responses and promoting immune tolerance.46 In leishmaniasis47 and malaria,48 IDO mediated attenuation of adaptive immunity may facilitate parasite persistence and disease severity. The high expression levels of IDO1 are likely to be produced by DCs and in combination with the observed reduction in the HLA class II antigen presentation activity, may be responsible for induction of immunosuppressive responses.

    • The kynurenine pathway and parasitic infections that affect CNS function

      2017, Neuropharmacology
      Citation Excerpt :

      However, animals treated with the kynurenine-3-hydroxylase inhibitor Ro-61-8048 were protected against the development of CM (Clark et al., 2005). The latter observation was confirmed later, albeit the observed degree of protection was less (Miu et al., 2009). It is not clear how these apparently contradictory observations can be reconciled and interpreted.

    • Induction of depression-related behaviors by reactivation of chronic Toxoplasma gondii infection in mice

      2016, Behavioural Brain Research
      Citation Excerpt :

      First, a number of pro-inflammatory cytokines are known to stimulate IDO (e.g., IFN-γ and TNF-α) and precipitate depressive symptoms in mice injected with peripheral immune-stimulatory and inflammatory agents [5,6,43]. Reciprocally, Kyn pathway catabolites are involved in generation of the inflammatory reaction induced by pathogens such as Trypanosoma, malaria parasites, or experimental endotoxemia [49–52]. Second, following bacterial infection in mice, chronic IDO up-regulation by IFN-γ or TNF-α induces long-lasting depressive-like behavior [44,49].

    • Apicomplexan parasite, Eimeria falciformis, co-opts host tryptophan catabolism for life cycle progression in mouse

      2012, Journal of Biological Chemistry
      Citation Excerpt :

      KAT-II and Kyn-3OH are selectively inhibited by (S)-4-(ethylsulfonyl) benzoylalanine hydrochloride (21) and Ro61-8048 (22), respectively (supplemental Fig. S1A). The mouse can tolerate relatively high doses of (S)-4-(ethylsulfonyl) benzoylalanine hydrochloride (5 mm) (26) and Ro61-8048 (200 mg/kg body weight) (27) via different application routes without apparent signs of mortality and morbidity. We applied them orally at much lower doses ((S)-4-(ethylsulfonyl) benzoylalanine hydrochloride, 1 mm; Ro61-8048, 2 mg/kg) and monitored the oocyst output in comparison with the control carrier-treated animals (Fig. 4A).

    View all citing articles on Scopus
    1

    Present address: Department of Medicine, McGill University, Montreal, Que., Canada.

    View full text