Elsevier

Neuropharmacology

Volume 43, Issue 3, September 2002, Pages 427-433
Neuropharmacology

The 2-oxopyrrolidinacetamide piracetam reduces infarct brain volume induced by permanent middle cerebral artery occlusion in male rats

https://doi.org/10.1016/S0028-3908(02)00093-XGet rights and content

Abstract

In this study, the temporal development of focal cerebral infarction induced by permanent middle cerebral artery occlusion (pMCAO) and the effects of piracetam, a derivative of γ-aminobutyric acid widely used in clinical practice as a nootropic agent, on infarct area and volume were investigated. pMCAO caused a cerebral infarct whose size progressively increased after 3, 6, 9, and 24 h. Piracetam (125 mg/kg i.p.), administered 6, 9, and 22 h after pMCAO, did not reduce pMCAO-induced brain infarct area size detected at the 24th hour. By contrast, when this agent was administered at the doses of 250 and 500 mg/kg, it caused a marked reduction of the infarct area size. This reduction was observed in almost every brain slice affected by pMCAO, although statistical differences (p <0.05) were detected in slices located at 3–5.5 mm posterior to the anterior pole in animals treated with 250 mg/kg piracetam and in slices located at 3.5–5 mm in those receiving 500 mg/kg. When the mean total volumes of brain infarct resulting from pMCAO were calculated, it was observed that in animals which had received piracetam (250 or 500 mg/kg) infarction volume was markedly (≈50%) and significantly (p <0.05) reduced in comparison with saline injected rats. Finally, piracetam (250 mg/kg administered i.p. 6, 9, and 22 h after the ischemic insult) significantly reduced brain infarct area evaluated 48 h and 7 days after pMCAO.

Introduction

Piracetam (2-oxopyrrolidinacetamide), a low-molecular-weight derivative of γ-aminobutyric acid, is widely used in clinical practice as a nootropic agent. In fact, it has been reported that piracetam is able to improve higher cerebral integrative functions including those involved in cognitive processes such as learning and memory (Giurgea, 1976).

In addition to its nootropic properties, piracetam has been also shown to exert a protective effect on brain function against hypoxic insults both in experimental animals and humans. In fact, it has been reported that this agent is able to reduce the amnesic effect of hypoxia in rats (Sara and Lefevre, 1972) and the brain recovery time after readmission of air to hypoxic rabbits (Giurgea et al., 1970). Furthermore, more recently it has been shown that piracetam restores dopamine release impaired by hypoxia (Wustmann et al., 1982) and improves cortical neuronal responsiveness to cutaneous stimulation after focal brain injury (Coq and Xerry, 1999).

In regard to studies performed in humans, it has been reported that piracetam increases compromised regional cerebral blood flow (Platt et al., 1992) and exerts a beneficial effect on neuronal metabolism, increasing the impaired glucose and oxygen cerebral metabolic rate when studied by positron emission tomography scan in the infarcted and adjacent brain tissue in acute stroke patients (Heiss et al., 1983, Depresseux et al., 1986). Furthermore, a neuroprotective effect of piracetam in the early therapy of acute hemispheric stroke has been reported (De Deyn et al., 1997. Finally, it has been demonstrated that piracetam can also improve neurologic functions impaired by stroke such as aphasia (Enderby et al., 1994, Huber, 1999) and palatal myoclonus (Karacostas et al., 1999).

In the present study, the possibility that piracetam could directly influence the extent of the infarcted brain region following acute stroke was investigated. For this purpose, the permanent middle cerebral artery occlusion (pMCAO) model was used in male rats. Brain infarct area was evaluated by 2,3,5-triphenyl tetrazolium chloride (TTC) method staining (Bederson et al., 1986). Piracetam (125, 250, and 500 mg/kg) was administered intraperitoneally three times at 6, 9, and 22 h after pMCAO. This time schedule was chosen on the basis of the results showing that the infarct size, detected 24 h after pMCAO, was significantly larger than that measured after 6 and 9 h respectively. This means that at these times ischemic damage is not yet complete and therefore a possible protective agent administered 6 and 9 h after pMCAO still has the possibility to induce a reduction of brain infarct. In addition, to ensure that piracetam treatment really prevented and not simply delayed neuronal damage after pMCAO, the drug effect on the extent of brain infarct volume was evaluated also 48 h and 7 days after the ischemic insult.

Section snippets

Experimental groups

Sprague–Dawley male rats (Charles River, Italy) weighing 250–270 g were housed under diurnal lighting conditions (12 h darkness and 12 h light) and fasted overnight but allowed free access to water before the experiment. Each experimental group consisted of 5–8 animals. Experiments were performed according to international guidelines for animal research and the experimental protocol was approved by Animal Care Committee of the University of Naples.

Permanent middle cerebral artery occlusion

Animals were anesthetized intraperitoneally

Time course of the development of brain infarct after pMCAO in Sprague–Dawley male rats

Infarct area enlarged progressively and in a time-dependent way after 3, 6, 9, and 24 h of ischemia. In particular, infarct size was small 3 h after pMCAO and significantly increased after 6 h. At the 9th hour the size was not significantly different from that detected at 6 h, whereas at the 24th hour it doubled the value detected at 9 h (Fig. 1).

Effects of increasing doses of piracetam on infarct area size resulting from pMCAO

In rats receiving vehicle, at the 24th h, pMCAO caused large brain infarcts involving predominantly striatum and fronto-temporoparietal cortex. The

Discussion

The results of the present study showed that piracetam produced a marked reduction of brain infarct volume induced by pMCAO in male rats. Piracetam was administered 6, 9, and 22 h after pMCAO since the study of the time course of the development of cerebral infarct after pMCAO showed that the infarct size obtained 24 h after pMCAO was significantly larger than that evaluated 6 and 9 h after pMCAO. The rationale for administering piracetam 6 h after pMCAO and for continuing its administration 22

Acknowledgements

This work was supported by the following grants: CNR 99.02371 CT04; MURST COFIN 1999 and 2000 to LA; Ministero della Sanità “Programma per la ricerca finalizzata 1998” to SA. In addition this work was partially supported by UCB-Pharma Italia and Belgique.

References (26)

  • P.P. De Deyn et al.

    for Members of the piracetam in acute stroke study (PASS) group. Treatment of acute ischemic stroke with piracetam

    Stroke

    (1997)
  • Depresseux, J.C., Salmon, E., Sadzot, B., Cornette, M., Franck, G., 1986. Evaluation of the effects of piracetam on CBF...
  • P. Enderby et al.

    Effect of piracetam on recovery and rehabilitation after stroke: a double-blind, placebo-controlled study

    Clinical Neuropharmacology

    (1994)
  • Cited by (20)

    • Neurochemical changes in unilateral cerebral hemisphere during the subacute stage of focal cerebral ischemia-reperfusion in rats: An ex vivo <sup>1</sup> H magnetic resonance spectroscopy study

      2018, Brain Research
      Citation Excerpt :

      The body temperature was continuously monitored and maintained at 37 ± 0.5 °C until the awakening. Transient cerebral focal ischemia (tMCAO) was produced by MCAO as reported previously (Aarts et al., 2002; Bederson et al., 1986; Tortiglione et al., 2002). 120 min post ischemia, nylon suture was removed under anesthesia again with 10% chloral hydrate (1 mL/kg) to achieve reperfusion.

    • Amelioration of cognitive, motor and endogenous defense functions with silymarin, piracetam and protocatechuic acid in the cerebral global ischemic rat model

      2013, Life Sciences
      Citation Excerpt :

      The brain homogenates were prepared in normal saline, centrifuged (3000 rpm for 10 min) and the cloudy supernatant (0.5 ml) was used for the assay of malondialdehyde (MDA) levels. MDA formation was measured at 535 nM (Tortiglione et al., 2002) and expressed as nM of MDA/g of wet tissue. Global ischemic/reperfusion rats (6) were sacrificed by cervical dislocation 24 h after the last dose of the drug treatment.

    • Silymarin improves the behavioural, biochemical and histoarchitecture alterations in focal ischemic rats: A comparative evaluation with piracetam and protocatachuic acid

      2012, Pharmacology Biochemistry and Behavior
      Citation Excerpt :

      A decrease in ischemic damage relates to the fact, that it is able to decrease the glutamate content in neocortex and augment calcium efflux. Events described earlier might trigger reduction of intracellular calcium accumulation, resulting overstimulation of N-methyl-D-aspartate (NMDA) receptors by released glutamate, is found to be responsible for post-ischemic neuronal death (Tortiglione et al., 2002). In the present study, improvement in ischemic damage too, may be related to decrease NMDA stimulation causing reduced intracellular calcium accumulation.

    • Synaptic plasticity during recovery from permanent occlusion of the middle cerebral artery

      2007, Neurobiology of Disease
      Citation Excerpt :

      Thus, cells which are dead or which have impaired oxidative metabolism do not stain or stain less well than healthy cells. The technique has been fully validated as a method for assessment of impaired brain cell metabolic function in our model of cerebral ischemia (Tortiglione et al., 2002; Picconi et al., 2006). Unstained areas on each brain slice, defined as infarction area, were then measured by using an image analyzer (Image-Pro plus 4.5, Media Cybernetics) and the volume of infarction was calculated by integration of the lesion areas.

    View all citing articles on Scopus
    View full text