A catalyst of peroxynitrite decomposition inhibits murine experimental autoimmune encephalomyelitis

https://doi.org/10.1016/S0165-5728(00)00242-3Get rights and content

Abstract

Peroxynitrite (PN), the product of nitric oxide (NO) reacted with superoxide, is generated at sites of inflammation. Nitrotyrosine (NT), a marker of PN formation, is abundant in lesions of acute experimental autoimmune encephalomyelitis (EAE), and in active multiple sclerosis (MS) plaques. To determine whether PN plays a role in EAE pathogenesis, mice induced to develop EAE were treated with a catalyst specific for the decomposition of PN. Because this catalyst has no effect upon NO, using it allowed differentiation of PN-mediated effects from NO-mediated effects. Mice receiving the PN decomposition catalyst displayed less severe clinical disease, and less inflammation and demyelination than control mice. Encephalitogenic T cells could be recovered from catalyst-treated mice, indicating that the PN decomposition catalyst blocked the pathogenic action of PN at the effector stage of EAE, but was not directly toxic to encephalitogenic T cells. PN plays an important role distinct from that of NO in the pathogenesis of EAE, a major model for MS.

Introduction

It is now recognized that the free radical gas, nitric oxide (NO) can be generated in large quantity by several mammalian cell types via the inducible isoform of NO synthase (iNOS) (Moncada et al., 1991, Xie and Nathan, 1994). Excess NO has been implicated in tissue damage in several diseases (Nathan and Hibbs, 1991). We and others have shown that inhibition of the production of excess NO in experimental autoimmune encephalomyelitis (EAE) by inhibiting iNOS ameliorates EAE induced by adoptive transfer of myelin-reactive T cells, a passive EAE induction paradigm (Cross et al., 1994, Zhao et al., 1996, Brenner et al., 1997, Ding et al., 1998). In contrast to this effector function of NO, studies of EAE induced by active immunization with myelin components in mice carrying a disrupted iNOS gene or rodents treated with NOS inhibitors have demonstrated a regulatory role for NO in EAE (Ruuls et al., 1996, Cowden et al., 1998, Fenyk-Meoldy et al., 1998). These studies suggest a complex, multifactorial relationship of NO with EAE pathogenesis. However, any assessment of the role of NO in pathophysiology requires consideration of potentially toxic metabolites formed from its reaction with other free radical species, such as superoxide radical (SOxR).

The powerful oxidant, peroxynitrite (PN, ONOO–), is one such product of NO and SOxR (Pryor and Squadrito, 1995). PN has recently been implicated as a mediator of tissue injury associated with inflammatory processes that generate excess NO. Nitrotyrosine (NT) immunoreactivity, a surrogate marker for PN formation (Beckman et al., 1994, Haddad et al., 1994a), has been found to be abundantly present within the central nervous system (CNS) of mice with acute EAE and in human CNS tissues affected by MS, however the role of PN in these diseases was unknown (Cross et al., 1997, Cross et al., 1998, Hooper et al., 1997, Van der Veen et al., 1997).

Recently, catalysts for the decomposition of PN have been developed which specifically catalyze the isomerization of PN to nitrate, its innocuous oxidation product (Stern et al., 1996). These catalysts have no effect upon NO, thereby enabling the pharmacological differentiation of PN-mediated versus NO-mediated effects (Misko et al., 1998, Salvemini et al., 1998). To define and differentiate the role of PN in EAE from the actions of NO, we used a PN decomposition catalyst to examine the effects of PN upon the induction and course of adoptively transferred murine EAE. Our findings showed that the clinical and histological features of EAE were inhibited in mice receiving the PN decomposition catalyst. These data indicate an important role for PN, distinct from that for NO, in the pathogenesis of murine EAE. Since recent data from our laboratories and others indicate that PN is formed in active MS lesions, the present data also suggest a potential role of PN in the pathogenesis of MS, and a therapeutic potential in MS for inhibitors of PN-mediated pathology.

Section snippets

Mice

Pathogen-free naı̈ve, female SJL and (PL×SJL)F1 mice were purchased from Jackson Laboratories, Bar Harbor ME. Mice were caged in microisolator cages. Sentinels were periodically tested for infection, with no infections being detected.

Chemicals

Bovine myelin basic protein (MBP), and concanavalin A were purchased from Sigma. The decomposition catalyst 5,10,15,20-tetrakis (2,4,6-trimethyl-3,5-sulfonatophenyl) porphyrinatoiron (III) (FeTMPS) and its inactive analog, TMPS, were synthesized as published (Stern

EAE incidence and severity are reduced during treatment with peroxynitrite decomposition catalyst

In five separate experiments, the PN decomposition catalyst FeTMPS was administered to mice induced to develop EAE by adoptive transfer. Drug was dosed at 10 mg/kg i.v. twice daily (20 mg/kg per day) in three experiments, and 5 mg/kg i.v. twice daily (10 mg/kg per day) in two experiments. An inactive compound (TMPS) with the same porphyrin structure but lacking Fe was administered to control mice. Clinical scoring was performed in blinded fashion.

The incidence of EAE was decreased among mice

Discussion

Considerable work in the last decade has established the EAE model to be a prototypic T-cell mediated autoimmune disease in which CD4+ T cells of the Th1 phenotype initiate disease (Zamvil et al., 1985, Ando et al., 1989). Due to a number of similarities which include the presence of primary CNS demyelination, the phenotypes of the inflammatory cells within the CNS lesions, and a relapsing-remitting clinical course, murine EAE is often used as a model to better elucidate the disease process in

Acknowledgements

We thank Drs Jeri A. Lyons, Grace Ku and John L. Trotter for discussion, and Mr Michael Ramsbottom for technical assistance. These studies were funded by grants from the National Multiple Sclerosis Society (RG-2934) and the National Institutes of Health (RO1-NS37037).

References (41)

  • Y. Okuda et al.

    Nitric oxide via an inducible isoform of nitric oxide synthase is a possible factor to eliminate inflammatory cells from the central nervous system of mice with experimental autoimmune encephalomyelitis

    J. Neuroimmunol.

    (1997)
  • D. Rachmilewitz et al.

    Peroxynitrite-induced rat colitis — a new model of colonic inflammation

    Gastroenterology

    (1993)
  • R.C. Van der Veen et al.

    Extensive peroxynitrite activity during progressive stages of central nervous system inflammation

    J. Neuroimmunol.

    (1997)
  • W. Zhao et al.

    Experimental allergic encephalomyelitis in the rat is inhibited by aminoguanidine, an inhibitor of inducible nitric oxide synthase

    J. Neuroimmunol.

    (1996)
  • J. Zielasek et al.

    Administration of nitric oxide synthase inhibitors in experimental autoimmune neuritis and experimental autoimmune encephalomyelitis

    J. Neuroimmunol.

    (1995)
  • E.C. Alvord et al.

    Experimental allergic encephalomyelitis: a useful model for multiple sclerosis

    Prog. Clin. Biol. Res.

    (1984)
  • O. Bagasra et al.

    Activation of the inducible form of nitric oxide synthase in the brains of patients with multiple sclerosis

    Proc. Natl. Acad. Sci. USA

    (1995)
  • J.S. Beckman et al.

    Pathological implication of nitric oxide, superoxide and peroxynitrite formation

    Biochem. Soc. Trans.

    (1993)
  • J.S. Beckman et al.

    Apparent hydroxyl radical production by peroxynitrite: implications for endothelial injury from nitric oxide and superoxide

    Proc. Natl. Acad. Sci. USA

    (1990)
  • J.S. Beckman et al.

    Extensive nitration of protein tyrosines in human atherosclerosis detected by immunohistochemistry

    Biol. Chem. Hoppe-Seyler

    (1994)
  • Cited by (39)

    • Matrine promotes NT3 expression in CNS cells in experimental autoimmune encephalomyelitis

      2017, Neuroscience Letters
      Citation Excerpt :

      NO, which is also considered to be a tissue-damaging free radical, could react with superoxide anion to produce peroxynitrite, which has the capacity to promote lipid peroxidation as well as nitration of tyrosine residues on proteins, and eventually induces neuronal damage in acute and chronic CNS degenerative diseases [25]. A decomposition catalyst of peroxynitrite could ameliorate clinical EAE and inhibit demyelination in mice [7,16]. Our results show a switch of macrophages/microglia from M1 to M2 phenotype upon MAT treatment and, although the total numbers of M1 cells were reduced and M2 cells were increased, this treatment enhanced NT3 production in both types of macrophages/microglia.

    • Arachidonyl trifluoromethyl ketone ameliorates experimental autoimmune encephalomyelitis via blocking peroxynitrite formation in mouse spinal cord white matter

      2011, Experimental Neurology
      Citation Excerpt :

      Peroxynitrite, the highly toxic reaction product of nitric oxide and superoxide (Beckman and Koppenol, 1996), has been shown to contribute to the pathology found in EAE/MS (Hooper et al., 1997; Liu et al., 2001; Spitsin et al., 2001). To support this notion, scavengers or the decomposition catalysts of peroxynitrite are found to be efficacious in the treatment of EAE (Bolton et al., 2008; Cross et al., 2000; Hooper et al., 1998, 2000). Here we tested whether the protective effects of AACOCF3 can be, at least in part, due to its inhibition of peroxynitrite formation.

    View all citing articles on Scopus
    View full text