Elsevier

Brain Research

Volume 989, Issue 1, 31 October 2003, Pages 58-66
Brain Research

Research report
Deleterious poly(ADP-ribose)polymerase-1 pathway activation in traumatic brain injury in rat

https://doi.org/10.1016/S0006-8993(03)03362-6Get rights and content

Abstract

Traumatic brain injury produces nitric oxide and reactive oxygen species. Peroxynitrite, resulting from the combination of nitric oxide and superoxide anions, triggers DNA strand breaks, leading to the activation of poly(ADP-ribose)polymerase-1. As excessive activation of this enzyme induces cell death, we examined the production of nitrosative stress, the activation of poly(ADP-ribose)polymerase-1, and the role of this enzyme in the outcomes of traumatic brain injury produced by fluid percussion in rats. Immunohistochemistry showed that 3-nitrotyrosine, an indicator of nitrosative stress, and poly(ADP-ribose), a marker of poly(ADP-ribose)polymerase-1 activation, were present as early as 30 min post-injury, and that persisted for 72 h. The poly(ADP-ribose)polymerase inhibitor, 3-aminobenzamide, at 10 and 30 mg/kg, significantly improved the neurological deficit, with a 60% reduction in the brain lesion volume and inhibition of poly(ADP-ribose)polymerase-1 activation. Thus, poly(ADP-ribose)polymerase-1 is involved in the neurological consequences of traumatic brain injury and may be a promising therapeutic target in clinical treatment of acute brain trauma.

Introduction

Reactive free radicals, including reactive oxygen species and reactive nitrogen species are implicated in the pathogenesis of central nervous system injuries such as traumatic brain injury (TBI), spinal cord injury, ischemia and chronic neurodegenerative diseases [13], [18], [25]. These highly reactive radicals and oxidants may indiscriminately attack proteins, lipids and DNA, causing oxidative modification and strand breakage [8], [9], [27]. The DNA strand breaks activate the constitutive nuclear enzyme poly(ADP-ribose)polymerase 1 (PARP-1, EC 2.4.2.30), which is implicated in such physiological processes as DNA repair [10], genomic stability [5] and apoptosis [20]. Moreover, PARP-1 has been shown to mediate necrotic cell death in response to excessive DNA damage under pathological conditions [20]. Activated PARP-1 catalyses the addition of long branched chains of poly(ADP-ribose) from its substrate NAD to a set of nuclear proteins including DNA polymerase I and II, Ca2+–Mg2+-endonuclease, histones, several chromatin-binding proteins and PARP-1 itself [10]. There is now evidence that excess active PARP-1 is a crucial factor in oxidative and excitotoxic cell death [33], [42]. PARP-1 activation leads to NAD depletion, resulting in a loss of ATP as it is used to synthesize new NAD, and finally to cell death [33]. In cultured cells exposed to hydrogen peroxide or peroxynitrite, DNA strand breaks are increased, PARP-1 is activated and energy resources are depleted as indicated by marked drops in NAD and ATP concentrations. These cellular disturbances can be prevented by PARP inhibitors [7], [37], [38], [41]. Multiple and strong experimental evidences have implicated PARP in cerebral ischemia and reperfusion in vivo. Indeed, cerebral ischemia–reperfusion activates PARP [16], [17], and PARP-1 mediates ischemic/reperfusion brain injury in vivo, since PARP-1 null mice and normal rodents treated with various PARP inhibitors, present a reduced infarct size [1], [12], [16], [17], [26], [43], [52]. The findings that both PARP-1 gene disruption and PARP inhibition greatly attenuate ischemic neuronal death in vivo indicate that PARP-1 activation is important in ischemic brain injury. By contrast, only few studies have demonstrated the implication of PARP in traumatic brain injury. PARP inhibition protects hippocampal slices against percussion-induced loss of CA1 pyramidal cells evoked response in vitro [48]. Whalen and co-workers [49], [50] have shown that the motor and cognitive deficits of mice submitted to TBI are less severe when the PARP-1 gene is inactivated. Lastly, it has recently been demonstrated that GPI 6150 (1,11b-dihydro-[2H]benzopyrano[4,3,2-de]isoquinolin-3-one), a novel PARP inhibitor, reduces the lesion area evaluated 24 h after TBI in rats [24]. To date, no published data have shown the effect of the pharmacological inhibition of PARP on the outcomes in the late phase after traumatic brain injury.

The present study was therefore carried out to explore the implication of the peroxynitrite–PARP-1 pathway in the post-traumatic outcomes of focal TBI caused by fluid percussion on rats. We first determined whether TBI triggered the production of peroxynitrite and the activation of PARP-1. We then assessed the role of PARP activation on post-traumatic events by examining the effect of the well-established and commonly used PARP inhibitor 3-aminobenzamide [45], on the neurological deficit and brain lesion volume 7 days after TBI. Lastly, we studied the effect of 3-aminobenzamide on the activation of PARP-1 following TBI.

Section snippets

Materials and methods

Animal care complied with the French regulations covering the protection of animals used for experimental and other scientific purposes (D2001-486), and with the European Community regulations (Official Journal of European Community L358 12/18/1986).

Experiment 1: kinetics of immunohistochemistry of 3-nitrotyrosine and poly(ADP-ribose)

The corpus callosum contained immunostaining for 3-nitrotyrosine as early as 30 min and 2 h after TBI. Large stained areas with an equivalent density were present in the cortex ipsilateral to injury at 4 h, which persisted until 72 h after TBI (Fig. 1). Some points of poly(ADP-ribose) immunostaining were also found in the cortex ipsilateral to injury 30 min and 2 h after TBI. By contrast, there was considerable staining, mainly in the nuclei with faint staining in the cytoplasm, 4 h after TBI,

Discussion

We find that TBI produced by fluid percussion leads to the production of peroxynitrite and/or other nitrosylating agents, and the activation of PARP-1. The PARP inhibitor, 3-aminobenzamide, improves the neurological deficit and reduces the brain lesion volume caused by TBI. This neuroprotection is associated with a decrease in the formation of (ADP-ribose) polymers, demonstrating PARP-1 inhibition. Thus, PARP-1 seems to be implicated in the pathogenesis of TBI.

TBI caused the formation of

Acknowledgements

We thank Owen Parkes for checking the English text.

References (52)

  • C. Szabó et al.

    Role of poly(ADP-ribose)synthetase in inflammation and ischaemia–reperfusion

    Trends Pharmacol. Sci.

    (1998)
  • K. Takahashi et al.

    Post-treatment with an inhibitor of poly(ADP-ribose)polymerase attenuates cerebral damage in focal ischemia

    Brain Res.

    (1999)
  • S. Toulmond et al.

    Biochemical and histological alterations induced by fluid percussion brain injury in the rat

    Brain Res.

    (1993)
  • F. Wahl et al.

    Riluzole reduces brain lesions and improves neurological function in rats after a traumatic brain injury

    Brain Res.

    (1997)
  • R.A. Wallis et al.

    Traumatic neuroprotection with inhibitors of nitric oxide and ADP-ribosylation

    Brain Res.

    (1996)
  • J. Yang et al.

    Nicotinamide therapy protects against both necrosis and apoptosis in a stroke model

    Pharmacol. Biochem. Behav.

    (2002)
  • G.E. Abdelkarim et al.

    Protective effects of PJ34, a novel, potent inhibitor of poly(ADP-ribose) polymerase (PARP) in in vitro and in vivo models of stroke

    Int. J. Mol. Med.

    (2001)
  • I.A. Ayoub et al.

    Therapeutic window for nicotinamide following transient focal cerebral ischemia

    Neuroreport

    (2002)
  • J.S. Beckman

    Oxidative damage and tyrosine nitration from peroxynitrite

    Chem. Res. Toxicol.

    (1996)
  • A. Burkle

    Physiology and pathophysiology of poly(ADP-ribosyl)ation

    BioEssays

    (2001)
  • T.M. Carlos et al.

    Expression of endothelial adhesion molecules and recruitment of neutrophils after traumatic brain injury in rats

    J. Leukoc. Biol.

    (1997)
  • M.R. Cookson et al.

    Peroxynitrite and hydrogen peroxide induced cell death in the NSC34 neuroblastoma×spinal cord cell line: role of poly(ADP-ribose)polymerase

    J. Neurochem.

    (1998)
  • M.R. Cookson et al.

    Oxidative stress and motor neurone disease

    Brain Pathol.

    (1999)
  • S. Cuzzocrea et al.

    Antioxidant therapy: a new pharmacological approach in shock, inflammation, and ischemia/reperfusion injury

    Pharmacol. Rev.

    (2001)
  • D. D’Amours et al.

    Poly(ADP-ribosyl)ation reactions in the regulation of nuclear functions

    Biochem. J.

    (1999)
  • W. Droge

    Free radicals in the physiological control of cell function

    Physiol. Rev.

    (2002)
  • Cited by (69)

    • Effects of nicotinamide on spatial memory and inflammation after juvenile traumatic brain injury

      2019, Behavioural Brain Research
      Citation Excerpt :

      NAM acts as an antioxidant and acts on the poly (ADP-ribose) polymerase (PARP) system, which utilizes NAD + to repair DNA fragmentation. DNA fragmentation is largely due to free radicals, which are increased following TBI [35,36]. In older animals, free radicals are scavenged and rendered inert by an antioxidant system.

    • Alterations in nitric oxide homeostasis during traumatic brain injury

      2017, Biochimica et Biophysica Acta - Molecular Basis of Disease
    View all citing articles on Scopus
    View full text