Original Contribution
Lipophilicity is a critical parameter that dominates the efficacy of metalloporphyrins in blocking the development of morphine antinociceptive tolerance through peroxynitrite-mediated pathways

https://doi.org/10.1016/j.freeradbiomed.2008.09.037Get rights and content

Abstract

Severe pain syndromes reduce the quality of life of patients with inflammatory and neoplastic diseases, partly because reduced analgesic effectiveness with chronic opiate therapy (i.e., tolerance) leads to escalating doses and distressing side effects. Peroxynitrite-mediated nitroxidative stress in the dorsal horn of the spinal cord plays a critical role in the induction and development of antinociceptive tolerance to morphine. This provides a valid pharmacological basis for developing peroxynitrite scavengers as potent adjuncts to opiates in the management of pain. The cationic Mn(III) ortho-N-alkylpyridylporphyrins MnTE-2-PyP5+ and MnTnHex-2-PyP5+ are among the most potent peroxynitrite scavengers, with nearly identical scavenging rate constants (~107 M 1 s 1). Yet, MnTnHex-2-PyP5+ is significantly more lipophilic and more bioavailable and, in turn, was 30-fold more effective in blocking the development of morphine antinociceptive tolerance than MnTE-2-PyP5+ using the hot-plate test in a well-characterized murine model. The hydrophilic MnTE-2-PyP5+ and the lipophilic MnTnHex-2-PyP5+ were 10- and 300-fold, respectively, more effective in inhibiting morphine tolerance than the hydrophilic Fe(III) porphyrin FeTM-4-PyP5+. Both Mn porphyrins decreased levels of TNF-α, IL-1β, and IL-6 to normal values. Neither of them affected acute morphine antinociceptive effects nor caused motor function impairment. Also neither was able to reverse already established morphine tolerance. We have recently shown that the anionic porphyrin Mn(III) tetrakis(4-carboxylatophenyl)porphyrin is selective in removing ONOO over O2radical dot, but at ∼ 2 orders of magnitude lower efficacy than MnTE-2-PyP5+ and MnTnHex-2-PyP5+, which in turn parallels up to 100-fold lower ability to reverse morphine tolerance. These data (1) support the role of peroxynitrite rather than superoxide as a major mechanism in blocking the development of morphine tolerance and (2) show that lipophilicity is a critical parameter in enhancing the potency of such novel peroxynitrite scavengers.

Section snippets

Mn porphyrins

MnTE-2-PyP5+ and MnTnHex-2-PyP5+ were synthesized and characterized as previously described [23], [46], [48].

Induction of morphine-induced antinociceptive tolerance in mice

Male CD-1 mice (24–30 g; Charles River) were housed and cared for in accordance with the guidelines of the Institutional Animal Care and Use Committee of the St. Louis University Health Science Center and in accordance with the National Institutes of Health guidelines on laboratory animals and the University of Messina, in compliance with Italian regulations on the protection of animals

The development of morphine-induced tolerance is blocked by the Mn porphyrin-based peroxynitrite decomposition catalysts MnTE-2-PyP5+ and MnTnHex-2-PyP5+

Compared to animals receiving an equivalent injection of vehicle (naïve group), acute injection of morphine (3 mg/kg) in animals that received saline over 4 days (vehicle group) produced a significant and near-maximal antinociceptive response (%MPE ranging between 90 and 95%) (Figs. 2A and 2B). In contrast, compared to the antinociceptive response to acute morphine in animals that received saline over 4 days, repeated administrations of morphine over the same time course (morphine group) led to

Discussion

Chronic administration of morphine promotes neuroimmune activation as evidenced by activation of spinal cord glial cells; production of proinflammatory cytokines such as TNF-α, IL-1β, and IL-6; and spinal sensitization [50], [51], [52]. Thus, inhibitors of glial cell metabolism and anti-cytokine approaches block morphine-induced antinociceptive tolerance and hyperalgesia [50], [51], [52]. The possible mechanisms for chronic morphine-induced glial cell activation are not known with certainty.

Concluding remarks

In summary, we have: (1) provided evidence that ONOO rather than O2radical dot is a predominant player in the development of morphine antinociceptive tolerance and (2) pointed to the lipophilicity as a critical parameter that determines the efficacy of the drug in blocking morphine tolerance and decreasing the levels of the inflammatory cytokines TNF-α, IL-1β, and IL-6. With high ability to eliminate ONOO along with enhanced lipophilicity, MnTnHex-2-PyP5+ (0.1 mg/kg/day) is 300-fold more effective in

Acknowledgments

Ines Batinić-Haberle and Júlio S. Rebouças are grateful for financial support from NIH U19 AI67798-01/Pilot Project and The Wallace H. Coulter Translational Partners Grant Program. Ivan Spasojević acknowledges NIH/NCI Duke Comprehensive Cancer Center Core Grant 5-P30-CA14236-29. Daniela Salvemini and Ines Batinić-Haberle acknowledge support from NIH 1 R01 DA24074-01A1.

References (86)

  • G. Ferrer-Sueta et al.

    Reactions of manganese porphyrins with peroxynitrite and carbonate radical anion

    J. Biol. Chem.

    (2003)
  • I. Batinić-Haberle et al.

    Pure MnTBAP selectively scavenges peroxynitrite over superoxide: comparison of pure and commercial MnTBAP samples to MnTE-2-PyP in two different models of oxidative stress injuries, SOD-specific E. coli model and carrageenan-induced pleurisy

    Free Radic. Biol. Med.

    (2009)
  • E. Michel et al.

    Kinetic properties of Cu,Zn-superoxide dismutases as a function of metal

    Arch. Biochem. Biophys.

    (2005)
  • S. Goldstein et al.

    Kinetic properties of Cu,Zn-superoxide dismutase as a function of metal content—order restored

    Free Radic. Biol. Med.

    (2006)
  • G. Ferrer-Sueta et al.

    Reduction of manganese porphyrins by flavoenzymes and submitochondrial particles and the catalytic redox cycle of peroxynitrite

    Free Radic. Biol. Med.

    (2006)
  • H. Tse et al.

    Mechanistic analysis of the immunomodulatory effects of a catalytic antioxidant on antigen-presenting cells: implication for their use in targeting oxidation/reduction reactions in innate immunity

    Free Radic. Biol. Med.

    (2004)
  • I. Batinić-Haberle et al.

    Tetrahydrobiopterin rapidly reduces SOD mimic, Mn(III) tetrakis(N-ethylpyridinium-2-yl)porphyrin

    Free Radic. Biol. Med.

    (2004)
  • I. Spasojević et al.

    New approach to the activation of anti-cancer pro-drug by metalloporphyrin-based cytochrome P450 mimics in all-aqueous biologically relevant system

    J. Inorg. Biochem.

    (2006)
  • A. Okado-Matsumoto et al.

    Complementation of SOD deficient Escherichia coli by manganese porphyrin mimics of superoxide dismutase

    Free Radic. Biol. Med.

    (2004)
  • M. Lodovici et al.

    Levels of 8-hydroxydeoxyguanosine as a marker of DNA damage in human leukocytes

    Free Radic. Biol. Med.

    (2000)
  • P. Song et al.

    The involvement of glial cells in the development of morphine tolerance

    Neurosci. Res.

    (2001)
  • L.R. Watkins et al.

    Glia: novel counter-regulators of opioid analgesia

    Trends Neurosci.

    (2005)
  • L.R. Watkins et al.

    Glia as the “bad guys”: implications for improving clinical pain control and the clinical utility of opioids

    Brain Behav. Immun.

    (2007)
  • P.K. Peterson et al.

    The opioid–cytokine connection

    J. Neuroimmunol.

    (1998)
  • D. Gius et al.

    Intracellular oxidation/reduction status in the regulation of transcription factors NF-kappaB and AP-1

    Toxicol. Lett.

    (1999)
  • B.M. Matata et al.

    Peroxynitrite is an essential component of cytokines production mechanism in human monocytes through modulation of nuclear factor-kappa B DNA binding activity

    J. Biol. Chem.

    (2002)
  • M.M. Khattab

    TEMPOL, a membrane-permeable radical scavenger, attenuates peroxynitrite- and superoxide anion-enhanced carrageenan-induced paw edema and hyperalgesia: a key role for superoxide anion

    Eur. J. Pharmacol.

    (2006)
  • C. Muscoli et al.

    Superoxide-mediated nitration of spinal manganese superoxide dismutase: a novel pathway in N-methyl-d-aspartate-mediated hyperalgesia

    Pain

    (2004)
  • E.S. Park et al.

    Levels of mitochondrial reactive oxygen species increase in rat neuropathic spinal dorsal horn neurons

    Neurosci. Lett.

    (2006)
  • E.S. Schwartz et al.

    Oxidative stress in the spinal cord is an important contributor in capsaicin-induced mechanical secondary hyperalgesia in mice

    Pain

    (2008)
  • H.K. Kim et al.

    Reactive oxygen species (ROS) play an important role in a rat model of neuropathic pain

    Pain

    (2004)
  • H.K. Kim et al.

    Analgesic effect of vitamin E is mediated by reducing central sensitization in neuropathic pain

    Pain

    (2006)
  • X. Gao et al.

    Reactive oxygen species (ROS) are involved in enhancement of NMDA-receptor phosphorylation in animal models of pain

    Pain

    (2007)
  • A. Viggiano et al.

    Trigeminal pain transmission requires reactive oxygen species production

    Brain Res.

    (2005)
  • G.R. Kirk et al.

    Combined antioxidant therapy reduces pain and improves quality of life in chronic pancreatitis

    J. Gastrointest. Surg.

    (2006)
  • I. Lee et al.

    The role of reactive oxygen species in capsaicin-induced mechanical hyperalgesia and in the activities of dorsal horn neurons

    Pain

    (2007)
  • I. Spasojević et al.

    Mn porphyrin-based SOD mimic MnTE-2-PyP5+ targets mouse heart mitochondria

    Free Radic. Biol. Med.

    (2007)
  • T. Ohse et al.

    Cell death by reactive oxygen species generated from water-soluble cationic metalloporphyrins as superoxide dismutase mimics

    J. Inorg. Biochem.

    (2001)
  • I. Batinić-Haberle et al.

    The ortho effect makes manganese(III) meso-tetrakis(N-methylpyridinium-2-yl)porphyrin (MnTM-2-PyP) a powerful and potentially useful superoxide dismutase mimic

    J. Biol. Chem.

    (1998)
  • S. Renfrey et al.

    The painful reality

    Nat. Rev. Drug Discovery

    (2003)
  • S. Arner et al.

    Clinical experience of long-term treatment with epidural and intrathecal opioids—a nationwide survey

    Acta Anaesthesiol. Scand.

    (1988)
  • M.H. Ossipov et al.

    Antinociceptive and nociceptive actions of opioids

    J. Neurobiol.

    (2004)
  • K.M. Foley

    Misconceptions and controversies regarding the use of opioids in cancer pain

    Anticancer Drugs

    (1995)
  • Cited by (42)

    • Nitroxidative Signaling Mechanisms in Pathological Pain

      2016, Trends in Neurosciences
      Citation Excerpt :

      NOX activity is elevated by morphine, and genetic or pharmacological disruption of these enzymes attenuates tolerance and hyperalgesia [171–173]. Superoxide and peroxynitrite have been implicated as downstream mediators because decomposition catalysts also attenuate tolerance and hyperalgesia [174–176]. It remains unclear how morphine engages these enzymes, but it may be mediated by classical μ-opioid receptors and/or TLR4 [168].

    • NADPH-oxidase 2 activation promotes opioid-induced antinociceptive tolerance in mice

      2013, Neuroscience
      Citation Excerpt :

      We have reported that in the spinal cord, peroxynitrite (PN) formation, the diffusion-limited product of superoxide and nitric oxide (NO) (Beckman et al., 1990), is central to these processes (Dang and Christie, 2012). Systemic delivery of PN decomposition catalysts (PNDCs) in mice and rats not only blocks morphine-induced antinociceptive tolerance (Muscoli et al., 2007; Batinic-Haberle et al., 2009), but does so by attenuating the activation of redox-sensitive transcription factors (NFκB) and MAPK kinases (ERK, p38 kinase) and the increased formation of various proinflammatory cytokines (Muscoli et al., 2007). PNDCs also block post-translational nitration and inactivation of glutamate transporters, GLAST and GLT-1, and glutamine synthetase (Salvemini and Neumann, 2009).

    • Thermal stability of the prototypical Mn porphyrin-based superoxide dismutase mimic and potent oxidative-stress redox modulator Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin chloride, MnTE-2-PyP<sup>5+</sup>

      2013, Journal of Pharmaceutical and Biomedical Analysis
      Citation Excerpt :

      These therapeutics, initially developed as catalytic antioxidant agents, are now regarded as potent modulators that are able to redox-cycle with reactive oxygen and nitrogen species (e.g., superoxide and peroxynitrite) and modulate several redox-dependent transcription factors (e.g., NF-κB, HIF-1α, VEGF, AP-1, and p53) [1–4], affecting, thus, a variety of inflammatory, angiogenic, and oncogenic pathways [3,6]. The prototypical cationic Mn porphyrin most studied in vivo, Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin chloride (MnTE-2-PyPCl5, Fig. 1) has been efficacious in many animal models of oxidative stress-related diseases and injuries [1–4], such as diabetes [7] and radiation injury [8], and as an adjunct in cancer pain management (reversal of morphine tolerance) [9] as well as cancer therapy [4,6]. Master drug file for MnTE-2-PyPCl5 has been filed with the U.S. Food and Drug Administration (FDA) and the toxicity studies needed for Investigational New Drug filing with FDA are finalized [3].

    • A new SOD mimic, Mn(III) ortho N-butoxyethylpyridylporphyrin, combines superb potency and lipophilicity with low toxicity

      2012, Free Radical Biology and Medicine
      Citation Excerpt :

      Among the lipophilic analogs, MnTnHex-2-PyP5+ has been the most frequently studied porphyrin [1,3,6]. The remarkable efficacy of MnTE-2-PyP5+ and MnTnHex-2-PyP5+ in numerous animal models of oxidative stress diseases [1,3,8–10] may be at least in part attributed to their ability to accumulate in mitochondria, mimicking therefore the site and the action of critical mitochondrial enzyme, MnSOD [7]. Mitochondrial accumulation of lipophilic MnTnHex-2-PyP5+ was significantly enhanced when compared to a more hydrophilic MnTE-2-PyP5+ [7].

    View all citing articles on Scopus
    1

    Present address: VA Medical Center, 915 N. Grand Boulevard, St. Louis, MO 63106, USA.

    View full text