Elsevier

Behavioural Brain Research

Volume 283, 15 April 2015, Pages 87-96
Behavioural Brain Research

Research report
Protective effects of cholecystokinin-8 on methamphetamine-induced behavioral changes and dopaminergic neurodegeneration in mice

https://doi.org/10.1016/j.bbr.2015.01.028Get rights and content

Highlights

  • Repeated administration of METH induced behavioral changes and dopaminergic neurotoxicity in mice.

  • CCK-8 attenuated METH-induced behavioral sensitization and stereotypic behaviors in mice.

  • CCK-8 inhibited hyperthermia and reduction of nigrostriatal TH and DAT in METH-treated mice.

Abstract

We investigated whether pretreatment with the neuropeptide cholecystokinin-8 affected methamphetamine (METH)-induced behavioral changes and dopaminergic neurodegeneration in male C57/BL6 mice. CCK-8 pretreatment alone had no effect on locomotion and stereotypic behavior and could not induce behavioral sensitization; however, it attenuated, in a dose-dependent manner, hyperlocomotion and behavioral sensitization induced by a low dose of METH (1 mg/kg). CCK-8 attenuated METH-induced stereotypic behavior at a dose of 3 mg/kg but not at 10 mg/kg. CCK-8 pretreatment attenuated METH (10 mg/kg)-induced hyperthermia, the decrease of tyrosine hydroxylase (TH) and dopamine transporter (DAT) in the striatum, and TH in the substantia nigra. CCK-8 alone had no effect on rectal temperature, TH and DAT expression in the nigrostriatal region. In conclusion, our study demonstrated that pretreatment with CCK-8 inhibited changes typically induced by repeated exposure to METH, such as hyperlocomotion, behavioral sensitization, stereotypic behavior, and dopaminergic neurotoxicity. These findings make CCK-8 a potential therapeutic agent for the treatment of multiple symptoms associated with METH abuse.

Introduction

Abuse of methamphetamine (METH) is an extremely serious and growing global problem. Long-term abuse of METH may result in memory loss, aggression, psychotic symptoms and potential heart and brain damage [1], [2], [3]. Rodents administered single or repeated METH treatments serve as an animal model for METH abuse in humans, as they present abnormal behaviors including hyperlocomotion and repetitive and compulsive behavior [4]. Systemic administration of METH to rodents induces increased locomotor activity and behavioral sensitization, and these are replaced by stereotypic behavior and hyperthermia at higher doses [5], [6], [7]. Once initiated, the stereotypic behavior induced by METH can persist for several hours, depending on the dose [8]. It was hypothesized that the action of METH on the dopamine (DA) neuronal system may contribute to the augmentation of psychostimulant use [9], [10]. Studies have demonstrated that METH is neurotoxic to striatal DA terminals, exemplified by a significant reduction in striatal DA and tyrosine hydroxylase (TH). In addition, dopamine transporter (DAT), an integral membrane protein that removes dopamine from the synaptic cleft and deposits it into surrounding cells, significantly decreased in both METH-treated animals and abuser [2], [7], [11].

However, the precise mechanisms by which METH elicits neurotoxic effects are still being elucidated. A number of studies have shown that METH-induced neurotoxicity involve the interaction of several neurotransmitters, neuropeptides, and neurotrophic factors. Cholecystokinin (CCK), a gut–brain peptide, exerts a wide range of biological activities in the gastrointestinal tract and central nervous system (CNS). It was initially isolated from the porcine duodenum as a 33 amino acid peptide [12] and acts via the CCK1 and CCK2 receptor subtypes. A number of biologically active molecular variants were subsequently described, and the most abundant peptide present in the brain was shown to be CCK-8: Asp-Tyr (SO3H)-Met-Gly-Trp-Met-Asp-Phe-NH2 [13], [14], [15]. CCK-8 is involved in the regulation of feeding, pain perception, and learning and memory and possibly in the pathogenesis of anxiety and psychosis [16], [17], [18], [19], [20]. It modulates the release of several neurotransmitters, such as DA and gamma-aminobutyric acid (GABA), and possibly acts as a neurotransmitter/modulator [21], [22]. A subpopulation of the dopaminergic neurons in the ventral tegmental area (VTA) projecting to the nucleus accumbens (NAc) contains CCK as a co-transmitter. Changes in the activity of the DA system have been observed in pharmacological studies conducted on CCK receptor-deficient mice. The targeted genetic suppression of CCK2 receptors has been shown to increase the sensitivity of pre- and post-synaptic D2 receptors. Loonam et al. found that CCK regulated neurochemical responses to METH in the striatum [23], [24]. Additionally, there are reports on the role of CCK in DA-mediated behaviors, with different subregions of the NAc involved in different effects on DA-mediated locomotor activity [25]. Moreover, our previous results have shown that CCK-8 has anti-oxidative stress and anti-inflammatory effects [26], [27]. In addition, it produced neuroprotective effects in neuronal injury models. These data show that CCK-8 exhibits a pharmaco-therapeutic potential for treating METH-induced neurotoxicity in CNS.

The present study aimed to evaluate the effects of CCK-8 on METH-induced behavioral changes, including hyperlocomotion, behavioral sensitization, and stereotypical behavior. Furthermore, the effects of CCK-8 on high doses of METH-induced hyperthermia and dopamine neurotoxicity were investigated.

Section snippets

Animals

Male C57BL/6 mice, initially weighing 18–20 g, were purchased from Beijing Vital River Laboratory Animal Technology Co. Ltd., China. The animals were housed in a climate-controlled environment. Constant temperature (21 ± 2 °C), humidity (approximately 60%) and a 12 h light/dark cycle (lights on at 7:00 am) were maintained. Food and water were available ad libitum. All experiments were conducted according to the guidelines of the National Institutes of Health Guide for the Care and Use of Laboratory

Animal exclusion

The group sizes were unequal due to the loss of the cannula, injection leakage, or poor locomotor activity. A total of 18 out of 235 mice were not included in the statistical analyses due to the mislocation of the cannulae. A total of 41 mice were excluded because of cannula surgery failure or poor general activity. In total, 270 animals were evaluable.

Effects of CCK-8 on single METH administration induced hyper-locomotion in mice

As shown in Fig. 2, a dose of 1 mg/kg (i.p.) METH before testing produced a significant hyper-locomotion compared with the baseline locomotion in

Discussions

In all METH-treated groups, significant hypo-locomotion, behavioral sensitization, hyperthermia and stereotypic behavior were established. The present study shows that CCK-8 has protective effects against behavioral and histological changes induced by low and high doses of METH in mice. CCK-8 significantly attenuated the hypo-locomotion induced by a single low dose (1 mg/kg) of METH. Pre-treatment with CCK-8 significantly inhibited the development and expression of METH-induced behavioral

Acknowledgments

All authors contributed to the writing of the manuscript and approved the final version. This study was financially supported in part by the National Natural Science Foundation of China (nos. 81302618,81471822), the Training funds for Talent Project in Hebei Province (no. LJRC005), and the Natural Science Foundation of Hebei Province (nos. H2013206157, H2014206275).

References (59)

  • K.A. Helm et al.

    Cholecystokinin combined with serotonin in the hypothalamus limits accumbens dopamine release while increasing acetylcholine: a possible satiation mechanism

    Brain Res

    (2003)
  • Z. He et al.

    Chicago sky blue 6B, a vesicular glutamate transporters inhibitor, attenuates methamphetamine-induced hyperactivity and behavioral sensitization in mice

    Behav Brain Res

    (2013)
  • B.D. Jung et al.

    Potentiation of methamphetamine neurotoxicity by intrastriatal lipopolysaccharide administration

    Neurochem Int

    (2010)
  • H.A. Boger et al.

    Minocycline restores striatal tyrosine hydroxylase in GDNF heterozygous mice but not in methamphetamine-treated mice

    Neurobiol Dis

    (2009)
  • T. Kawasaki et al.

    Protective effect of the radical scavenger edaravone against methamphetamine-induced dopaminergic neurotoxicity in mouse striatum

    Eur J Pharmacol

    (2006)
  • D. Wen et al.

    The effects of exogenous CCK-8 on the acquisition and expression of morphine-induced CPP

    Neurosci Lett

    (2012)
  • Y. Hirosue et al.

    Effects of CCK antagonists on CCK-induced suppression of locomotor activity in mice

    Peptides

    (1992)
  • D. Wen et al.

    Effects of CCK-8 on the reinstatement of morphine-induced CPP and expression of behavioral sensitization in rats

    Neuroscience

    (2013)
  • A. Bjorklund et al.

    Dopamine neuron systems in the brain: an update

    Trends Neurosci

    (2007)
  • Y. Takeda et al.

    Effect of injection of CCK-8 into the nucleus caudatus on the behavior of rats

    Jpn J Pharmacol

    (1986)
  • Y.L. Hurd et al.

    Amphetamine regulation of mesolimbic dopamine/cholecystokinin neurotransmission

    Brain Res

    (1992)
  • L. Záborszky et al.

    Cholecystokinin innervation of the ventral striatum: a morphological and radioimmunological study

    Neuroscience

    (1985)
  • F. Weiss et al.

    Opposite actions of CCK-8 on amphetamine-induced hyperlocomotion and stereotypy following intracerebroventricular and intra-accumbens injections in rats

    Pharmacol Biochem Behav

    (1988)
  • P. Nicotera et al.

    Mechanisms for nitric oxide-induced cell death: involvement of apoptosis

    Adv Neuroimmunol

    (1995)
  • L.E. Halpin et al.

    Neurotoxicity of methamphetamine and 3,4-methylenedioxymethamphetamine

    Life Sci

    (2014)
  • D.M. Thomas et al.

    MK-801 and dextromethorphan block microglial activation and protect against methamphetamine-induced neurotoxicity

    Brain Res

    (2005)
  • V. Chaparro-Huerta et al.

    Proinflammatory cytokines and apoptosis following glutamate-induced excitotoxicity mediated by p38 MAPK in the hippocampus of neonatal rats

    J Neuroimmunol

    (2005)
  • M.S. Wyeth et al.

    Increased cholecystokinin labeling in the hippocampus of a mouse model of epilepsy maps to spines and glutamatergic terminals

    Neuroscience

    (2012)
  • B.K. Yamamoto et al.

    Amphetamine toxicities: classical and emerging mechanisms

    Ann NY Acad Sci

    (2010)
  • Cited by (0)

    1

    These authors contributed equally to this work.

    View full text