Temporal relationship of peroxynitrite-induced oxidative damage, calpain-mediated cytoskeletal degradation and neurodegeneration after traumatic brain injury

https://doi.org/10.1016/j.expneurol.2007.01.023Get rights and content

Abstract

We assessed the temporal and spatial characteristics of PN-induced oxidative damage and its relationship to calpain-mediated cytoskeletal degradation and neurodegeneration in a severe unilateral controlled cortical impact (CCI) traumatic brain injury (TBI) model. Quantitative temporal time course studies were performed to measure two oxidative damage markers: 3-nitrotyrosine (3NT) and 4-hydroxynonenal (4HNE) at 30 min, 1, 3, 6, 12, 24, 48, 72 h and 7 days after injury in ipsilateral cortex of young adult male CF-1 mice. Secondly, the time course of Ca++-activated, calpain-mediated proteolysis was also analyzed using quantitative western-blot measurement of breakdown products of the cytoskeletal protein α-spectrin. Finally, the time course of neurodegeneration was examined using de Olmos silver staining. Both oxidative damage markers increased in cortical tissue immediately after injury (30 min) and elevated for the first 3–6 h before returning to baseline. In the immunostaining study, the PN-selective marker, 3NT, and the lipid peroxidation marker, 4HNE, were intense and overlapping in the injured cortical tissue. α-Spectrin breakdown products, which were used as biomarker for calpain-mediated cytoskeletal degradation, were also increased after injury, but the time course lagged behind the peak of oxidative damage and did not reach its maximum until 24 h post-injury. In turn, cytoskeletal degradation preceded the peak of neurodegeneration which occurred at 48 h post-injury. These studies have led us to the hypothesis that PN-mediated oxidative damage is an early event that contributes to a compromise of Ca++ homeostatic mechanisms which causes a massive Ca++ overload and calpain activation which is a final common pathway that results in post-traumatic neurodegeneration.

Introduction

There is compelling evidence supporting the role of oxidative damage in the delayed secondary neuronal cell death which is initiated by the primary traumatic brain injury (TBI). The first work in this regard, conducted by Kontos and his colleagues, demonstrated the formation of superoxide radicals in cerebral microvasculature in a cat fluid percussion TBI models (Kontos and Povlishock, 1986, Kontos and Wei, 1986). Using the salicylate trapping method, Hall and coworkers detected a rapid, but transient rise in brain hydroxyl radical (radical dotOH) in experimental head injury models (Hall et al., 1993, Hall et al., 1994, Smith et al., 1994). This was elegantly confirmed by others using brain microdialysis techniques to monitor salicylate-trapped radical dotOH over time in the same animals (Globus et al., 1995). The increase of radical dotOH is followed by an increase in lipid peroxidation products (Smith et al., 1994). The strongest support for a role of oxidative damage in the acute pathophysiology of TBI is derived from the fact that several antioxidant compounds have been shown to be neuroprotective in TBI models (Hall et al., 1994, Awasthi et al., 1997, Mori et al., 1998, Marklund et al., 2001).

The reactive oxygen species (ROS) peroxynitrite (PN), which can produce highly reactive and cytotoxic free radicals, has been suggested to be a key player in post-traumatic secondary brain oxidative damage (Hall et al., 1999, Hall et al., 2004). Peroxynitrite is formed by the chemical reaction of nitric oxide (radical dotNO), produced by nitric oxide synthase (NOS), with superoxide radical (O2radical dot) whenever the two are produced in close proximity (Saran et al., 1990). All three NOS isoforms (eNOS, iNOS, and nNOS), as a key element in production of PN, have been shown to be up-regulated in brain tissue following TBI (Cobbs et al., 1997, Wada et al., 1998, Gahm et al., 2000, Orihara et al., 2001). In addition, a novel isoform of Ca++-sensitive NOS (mtNOS), discovered within mitochondria (Bates et al., 1995, Lopez-Figueroa et al., 2000, Giulivi, 2003), has been shown to contribute to mitochondrial production of radical dotNO and PN (Radi et al., 2002). The PN-derived free radicals (radical dotNO2, radical dotOH, radical dotCO3) can induce extensive oxidative damage to cellular membranes, proteins and DNA (Beckman, 1996, Murphy et al., 1998, Radi, 1998). Each of the PN-derived free radicals can initiate lipid peroxidation (LP), or cause protein carbonylation by reaction with susceptible amino acids (e.g. lysine, cysteine, arginine). Moreover, aldehydic LP products (e.g. 4-hydroxynonenal) can bind to cellular proteins compromising their structural and functional integrity (Neely et al., 1999). Additionally, radical dotNO2 can nitrate 3 positions of tyrosine residues in proteins. As a result, 3-nitrotyrosine (3NT) is used as a specific footprint of PN-induced cellular damage (Hall et al., 1997).

In recent work we investigated the role of PN in a mouse model of diffuse closed head injury which demonstrated the spatial and temporal coincidence of PN-induced protein nitration and lipid peroxidation and that this oxidative damage precedes, and therefore may have a causal role in post-traumatic neurodegeneration (Hall et al., 2004). However, TBI is a complex disorder that is impossible to fully reproduce in a single model. For example, we have found significant differences between the time courses of neurodegeneration in mouse models of diffuse (Kupina et al., 2003) and focal (Hall et al., 2005a, Hall et al., 2005b) TBI. Hence, the magnitude and timing of post-traumatic secondary injury mechanisms probably vary across head injury models. Most importantly, if we are to accurately test the efficacy of novel neuroprotective pharmacological treatments, it is important to first understand the time course of the relevant secondary injury mechanisms in models of focal as well as diffuse TBI.

Thus, the present study was conducted to examine the temporal and spatial characteristics of PN-induced cortical oxidative damage in a model of severe controlled cortical impact (CCI) focal TBI, and its relationship to calpain-mediated cytoskeletal degradation and neurodegeneration. Markers for PN-induced protein nitration (3-nitrotyrosine, 3NT) and lipid peroxidation (4-hydroxynonenal, 4HNE) were measured using immuno-slotblotting and immuno-histochemistry. Secondly, we employed western-blotting methods to look at the time course of calpain-mediated cytoskeletal degradation in order to assess the temporal relationship of oxidative damage to Ca++−mediated proteolytic degradation. Finally, we used de Olmos silver staining to measure the evolution of post-traumatic neurodegeneration. The results demonstrate that the PN-mediated oxidative damage is an early event that probably contributes to an exacerbation of neuronal intracellular Ca++ overload, massive calpain activation and cytoskeletal degradation and neurodegeneration.

Section snippets

Materials and methods

All the surgical, injury and animal care protocols described below have been approved by the University of Kentucky Institutional Animal Care and Use Committee and are consistent with the animal care procedures set forth in the guidelines of the U.S. Public Health Service Policy on Humane Care and Use of Laboratory Animals.

Quantitative post-traumatic time course of protein nitration and lipid peroxidation

Fig. 1 displays the complete quantitative time course study for 3NT and 4HNE in the ipsilateral cortical samples taken from sham or CCI-injured mice. Fig. 1A indicates schematically the dorsal and coronal view of contusion site and peri-contusional cortical tissue that was collectively sampled for the current study. Fig. 1B shows the time course of changes in 3NT, a selective marker of PN-mediated damage. As noted in Materials and methods, two non-injured sham animals corresponding to each

Discussion

The present study in the context of the focal controlled cortical impact (CCI) mouse model has uniquely defined in a parallel fashion the time courses of oxidative damage, calpain-mediated cytoskeletal degradation and neurodegeneration. A careful analysis of these three secondary injury parameters has revealed clues concerning their mechanistic inter-relationships. The following discussion lays out the hypothesis that (1) the potent ROS PN is a key mediator of post-traumatic oxidative damage;

References (73)

  • S.L. Elfering et al.

    Biochemistry of mitochondrial nitric-oxide synthase

    J. Biol. Chem.

    (2002)
  • P. Ghafourifar et al.

    Nitric oxide synthase activity in mitochondria

    FEBS Lett.

    (1997)
  • P. Ghafourifar et al.

    Mitochondrial nitric-oxide synthase stimulation causes cytochrome c release from isolated mitochondria. Evidence for intramitochondrial peroxynitrite formation

    J. Biol. Chem.

    (1999)
  • C. Giulivi

    Characterization and function of mitochondrial nitric-oxide synthase

    Free Radical Biol. Med.

    (2003)
  • R.P. Guttmann et al.

    Oxidative stress inhibits calpain activity in situ

    J. Biol. Chem.

    (1998)
  • R.P. Guttmann et al.

    Oxidation inhibits substrate proteolysis by calpain I but not autolysis

    J. Biol. Chem.

    (1997)
  • E.D. Hall et al.

    Immunocytochemical method for investigating in vivo neuronal oxygen radical-induced lipid peroxidation

    J. Neurosci. Methods

    (1997)
  • N.C. Kupina et al.

    Cytoskeletal protein degradation and neurodegeneration evolves differently in males and females following experimental head injury

    Exp. Neurol.

    (2003)
  • Z. Lacza et al.

    Mitochondrial nitric oxide synthase is constitutively active and is functionally upregulated in hypoxia

    Free Radic. Biol. Med.

    (2001)
  • M.O. Lopez-Figueroa et al.

    Direct evidence of nitric oxide presence within mitochondria

    Biochem. Biophys. Res. Commun.

    (2000)
  • C. Mesenge et al.

    Reduction of tyrosine nitration after N(omega)-nitro-l-arginine-methylester treatment of mice with traumatic brain injury

    Eur. J. Pharmacol.

    (1998)
  • K.P. Mohanakumar et al.

    Neuroprotection by nitric oxide against hydroxyl radical-induced nigral neurotoxicity

    J. Chem. Neuroanat.

    (1998)
  • M.P. Murphy et al.

    Peroxynitrite: a biologically significant oxidant

    Gen. Pharmacol.

    (1998)
  • R.W. Neumar et al.

    Calpain activity in the rat brain after transient forebrain ischemia

    Exp. Neurol.

    (2001)
  • Y. Orihara et al.

    Induction of nitric oxide synthase by traumatic brain injury

    Forensic Sci. Int.

    (2001)
  • R. Radi et al.

    Inhibition of mitochondrial electron transport by peroxynitrite

    Arch. Biochem. Biophys.

    (1994)
  • R. Radi et al.

    Peroxynitrite reactions and formation in mitochondria

    Free Radic. Biol. Med.

    (2002)
  • A. Roveri et al.

    Effect of hydrogen peroxide on calcium homeostasis in smooth muscle cells

    Arch. Biochem. Biophys.

    (1992)
  • P.G. Sullivan et al.

    Cyclosporin A attenuates acute mitochondrial dysfunction following traumatic brain injury

    Exp. Neurol.

    (1999)
  • S.N. Thompson et al.

    Relationship of calpain-mediated proteolysis to the expression of axonal and synaptic plasticity markers following traumatic brain injury in mice

    Exp. Neurol.

    (2006)
  • K.K. Wang

    Calpain and caspase: can you tell the difference?

    Trends Neurosci.

    (2000)
  • F. Zhou et al.

    Neuronal free Ca(2+)and BBB permeability and ultrastructure in head injury with secondary insult

    J. Clin. Neurosci.

    (2001)
  • E. Arrigoni et al.

    Calcium-activated neutral protease activities in brain trauma

    Neurochem. Res.

    (1991)
  • R.T. Bartus et al.

    Calpain as a novel target for treating acute neurodegenerative disorders

    Neurol. Res.

    (1995)
  • J.S. Beckman

    Oxidative damage and tyrosine nitration from peroxynitrite

    Chem. Res. Toxicol.

    (1996)
  • G. Bicker

    Nitric oxide: an unconventional messenger in the nervous system of an orthopteroid insect

    Arch. Insect. Biochem. Physiol.

    (2001)
  • Cited by (125)

    • Seizures in traumatic brain injury: A focus on cellular aspects

      2022, Cellular, Molecular, Physiological, and Behavioral Aspects of Traumatic Brain Injury
    View all citing articles on Scopus
    View full text