throbber
Psychiatry Research 198 (2012) 521–526
`
`Contents lists available at SciVerse ScienceDirect
`
`Psychiatry Research
`
`j o ur n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / p s y c h r e s
`
`Effects of lithium and valproate on oxidative stress and behavioral changes induced
`by administration of m-AMPH
`Dayane D. da-Rosa a, Samira S. Valvassori a, Amanda V. Steckert a,b, Felipe Ornell a, Camila L. Ferreira a,
`Jéssica Lopes-Borges a, Roger B. Varela a, Felipe Dal-Pizzol b, Monica L. Andersen c, João Quevedo a,⁎
`a Laboratory of Neurosciences and National Institute for Translational Medicine (INCT-TM), Postgraduate Program in Health Sciences, Health Sciences Unit,
`University of Southern Santa Catarina, 88806-000 Criciúma, SC, Brazil
`b Laboratory of Experimental Pathophysiology and National Institute for Translational Medicine (INCT-TM), Postgraduate Program in Health Sciences,
`Health Sciences Unit, University of Southern Santa Catarina, 88806-000 Criciúma, SC, Brazil
`c Department of Psychobiology, Universidade Federal de São Paulo, 04024-002 São Paulo, SP, Brazil
`
`a r t i c l e
`
`i n f o
`
`a b s t r a c t
`
`Article history:
`Received 4 October 2011
`Received in revised form 12 January 2012
`Accepted 18 January 2012
`
`Keywords:
`Animal model of mania
`Lithium
`Valproate
`Methamphetamine
`Oxidative stress
`
`In the last years our research group has studied and validated the animal model of mania induced by dextro-
`amphetamine (D-AMPH). Considering the lack of animal models of mania reported in the literature; this
`study evaluated the possibilities to validate the animal model induced by methamphetamine (m-AMPH).
`Then, we evaluated the effects of lithium (Li), valproate (VPA) on the behavior and parameters of oxidative dam-
`age in rat brain after administration of m-AMPH. In the prevention treatment, Wistar rats were pretreated with
`Li, VPA or saline (Sal) for 14 days, and then, between days 8 and 14, rats were treated with m-AMPH (1, 0.5 or
`0.25 mg/kg) or Sal. In the reversal treatment, rats were first given m-AMPH (0.25 mg/kg) or Sal. Locomotor be-
`havior was assessed using the open-field task and parameters of oxidative damage were measured in brain struc-
`tures. Our results show that the hyperactivity was prevented and reverted by Li and VPA only when m-AMPH
`was administered in the dose of 0.25 mg/kg. In addition, the m-AMPH in all doses administrated induced oxida-
`tive damage in both structures tested in two models. Li and VPA reversed and prevented this impairment, how-
`ever in a way dependent of cerebral area, the dose of m-AMPH and technique.
`© 2012 Elsevier Ireland Ltd. All rights reserved.
`
`1. Introduction
`
`Bipolar disorder (BD) is one of the most severe psychiatric disor-
`ders, which is associated with morbidity and mortality and psychiat-
`ric comorbidity (Kupfer, 2005; McIntyre et al., 2007). However,
`despite the severity of the disorder, its pathophysiology remains
`largely unknown. Animal models are an important tool in gaining in-
`sights into the neural mechanisms underlying BD and in assessing po-
`tential therapeutic actions of new compounds in preclinical settings
`(Lipska and Weinberger, 2000; Boksa, 2007). Animal models of
`human diseases should meet three sets of criteria: face, construct,
`and predictive validities (Einat et al., 2003; Machado-Vieira et al.,
`2004). Face validity is the ability of the model to mimic the symptoms
`of the determinate human disorder, while construct validity is the ca-
`pacity of the model to mimic some pathophysiological change found
`in the human disorder. Finally, predictive validity refers to the ability
`of the conventional drugs used to treat the disorder, prevent and/or
`reverse the symptoms induced in the model (Machado-Vieira et al.,
`2004).
`
`⁎ Corresponding author at: Laboratório de Neurociências, PPGCS, UNASAU, Universidade
`do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil. Fax: +55 48 3443 4817.
`E-mail address: joq@unesc.net (J. Quevedo).
`
`0165-1781/$ – see front matter © 2012 Elsevier Ireland Ltd. All rights reserved.
`doi:10.1016/j.psychres.2012.01.019
`
`Dopamine (DA) is an important neurotransmitter that is involved
`in cognition, mood and motor functions of the brain (Sibley and
`Monsma, 1992; Missale et al., 1998). Changes in the dopamine signal-
`ing are implicated in numerous neuropsychiatric disorders, including
`schizophrenia and bipolar disorder (Greengard, 2001). Many of the
`signs and symptoms of BD can be reproduced in humans and animal
`models with dopaminergic stimulants such as cocaine and amphet-
`amine. For example, amphetamine (AMPH) administration in rats in-
`duces hyperlocomotion, insomnia and increased sexual drive (Fiorino
`and Phillips, 1999) — symptoms also found in bipolar patients. Addi-
`tionally, in previous studies, our research group demonstrated that
`lithium (Li) and valproate (VPA) – two classic drugs for the treatment
`of BD – reversed and prevented the dextroamphetamine-induced hy-
`peractivity in rats (Frey et al., 2006a,b,c; Andreazza et al., 2008;
`Valvassori et al., 2010).
`AMPHs are psychostimulant drugs of the phenethylamine class that
`act by increasing DA efflux, inhibit the uptake of DA and inhibit mono-
`amine oxidase (Fischer and Cho, 1979). It is well known that the dopami-
`nergic system and oxidation of DA play pivotal roles in the neurotoxicity
`produced by AMPHs, which increase the formation of reactive
`oxygen species (Chipana et al., 2008). Dextroamphetamine (D-AMPH)
`is chemically related to methamphetamine (m-AMPH), nevertheless,
`m-AMPH is considered to be a more potent central psychostimulant
`
`1 of 6
`
`Alkermes, Ex. 1051
`
`

`
`522
`
`D.D. da-Rosa et al. / Psychiatry Research 198 (2012) 521–526
`
`than D-AMPH (Mathias, 1998). However, behavioral and neurochemical
`differences between D-AMPH and m-AMPH remain poorly understood
`(Ricaurte et al., 1983; Kuczenski et al., 1995; Shoblock et al., 2003).
`Moreover, several lines of evidence show the oxidative damage to
`lipid and proteins as one of the possible mechanisms contributing to
`neuronal and glial impairment in BD (Kunz et al., 2008; Andreazza
`et al., 2010a,b; Steckert et al., 2010). Repeated injections of D-AMPH
`that induce hyperactivity in rodents is associated with increased in
`protein and lipid oxidative damage in brain (Gluck et al., 2001;
`Andreazza et al., 2008; Valvassori et al., 2011), supporting the view
`that oxidative stress is occurring during a manic-like state. In addi-
`tion, previous studies of our laboratory also demonstrated that Li
`and VPA prevented and reversed the D-AMPH-induced oxidative
`protein and lipid damage in the brain of rats (Frey et al., 2006c;
`Andreazza et al., 2008).
`Therapeutic development in bipolar is hampered by a lack of path-
`ophysiological model (Berk et al., 2007). In the last six years our re-
`search group has studied and validated the animal model of mania
`induced by D-AMPH, which has been well accepted in the literature
`(Frey et al., 2006c; Andreazza et al., 2008; Valvassori et al., 2010).
`Considering: 1) the lack of animal models of mania reported in the
`literature and 2) the lack of studies showing the difference between
`D-AMPH and m-AMPH; the present study aims to evaluate the possi-
`bilities to validate the animal model induced by m-AMPH. Then, we
`evaluated the effects of mood stabilizers on the behavior and parame-
`ters of oxidative damage in rat brain after administration of m-AMPH.
`
`2. Methods
`
`2.1. Animals
`
`The subjects were adult male Wistar rats (weighting 250–350 g) obtained from our
`breeding colony. Animals were housed five to a cage with food and water available ad libi-
`tum and were maintained on a 12-h light/dark cycle (lights on at 7:00 a.m.) at a temper-
`ature of 22±1 °C. All experimental procedures were performed in accordance with the
`approval of the local Ethics Committee in the use of animals at the Universidade do
`Extremo Sul Catarinense. All experiments were performed at the same time during the
`day to avoid circadian variations.
`
`2.2. Treatment protocols
`
`2.2.1. Prevention treatment
`In the prevention model, we simulated the maintenance phase of BD treatment.
`Animals were treated with Li (47.5 mg/kg i.p.), VPA (200 mg/kg i.p.) or saline twice a
`day for 14 days. Between the 8th and the 14th days, Li, VPA and saline-treated animals
`additionally received one daily intraperitoneal injection of either m-AMPH or saline.
`From this protocol 3 experiments were made . In experiment 1, a dose of 0.25 mg/kg
`of m-AMPH was administered and experimental groups were as follows: Sal+Sal, Li+
`Sal, VPA+Sal, Sal+m-AMPH 0.25 mg/kg, Li+m-AMPH 0.25 mg/kg, VPA+m-AMPH
`0.25 mg/kg (n=15 animals per group).
`In experiment 2, a dose of 0.5 mg/kg of m-AMPH was administered and experimental
`groups were as follows: Sal + Sal, Li + Sal, VPA + Sal, Sal + m-AMPH 0.5 mg/kg, Li +
`m-AMPH 0.5 mg/kg, VPA + m-AMPH 0.5 mg/kg (n = 15 animals per group).
`In experiment 3, a dose of 0.5 mg/kg of m-AMPH was administered and experi-
`mental groups were as follows: Sal+Sal, Li+Sal, VPA+Sal, Sal+m-AMPH 0.5 mg/kg,
`Li+m-AMPH 0.5 mg/kg, VPA+m-AMPH 0.5 mg/kg (n=15 animals per group).
`Locomotor activity was measured 2 h after the last injection. The rats were sacri-
`ficed by decapitation immediately after the open-field task and hippocampus, striatum
`and prefrontal were dissected, rapidly frozen and stored −70 °C until assayed.
`
`2.2.2. Reversal treatment
`In the reversal model, we reproduced the treatment of acute manic episode. The ani-
`mals received one daily intraperitoneal injection (i.p.) of m-AMPH 0.25 mg/kg or saline
`(Sal) for 14 days. On the 8th day of treatment, the animals in the saline and D-AMPH
`group were divided in three groups: 1) treatment with Li (47.5 mg/kg i.p.); 2) treatment
`with VPA (200 mg/kg i.p.) and 3) treatment with Sal for 7 days twice a day for all drugs.
`On the 15th day of treatment, the animals received a single injection of m-AMPH
`(0.25 mg/kg) or Sal and locomotor activity was assessed 2 h after the last injection. The ex-
`perimental groups were as follows: Sal+Sal, Sal+Li, Sal+VPA, m-AMPH 0.25 mg/kg+
`Sal, m-AMPH 0.25 mg/kg+Li, m-AMPH 0.25 mg/kg+VPA (n=12 animals per group).
`The rats were killed by decapitation immediately after the open-field task and pre-
`frontal, amygdale, hippocampus and striatum were dissected, rapidly frozen and
`stored at −70 °C until assayed.
`
`Note: The reversion protocol was made only with the dose of m-AMPH (0.25 mg/kg)
`in which Li and VPA could prevent the hyperactivity induced by m-AMPH.
`
`2.3. Locomotor activity
`
`Locomotor activity was assessed using the open-field task as previously described
`(Barros et al., 2002; Frey et al., 2006a,b,c). This task was performed in a 40 ×60 cm
`open field surrounded by 50 cm high walls, made of brown plywood, with the floor di-
`vided into 12 equal rectangles by black lines. The animals were gently placed on the
`left rear rectangle, and left free to explore the arena for 5 min. Crossings of the black
`lines (locomotor activity/horizontal activity) and rearings (exploratory activity/vertical
`activity) were counted.
`
`2.4. Measurement of oxidative damage markers
`
`Rats treated with m-AMPH or Saline as described above were sacrificed by decap-
`itation 2 h after the last injection and their brains were removed and dissected for eval-
`uation of oxidative damage levels in the prefrontal cortex, amygdala, hippocampus and
`striatum. Thiobarbituric acid reactive substances (TBARS) and protein carbonyl forma-
`tion were measured as previously described (Draper and Hadley, 1990; Levine et al.,
`1994).
`Note: For biochemical analysis n=5 animals per group, which were chosen ran-
`domly, were used.
`
`2.5. Thiobarbituric acid reactive substances (TBARS)
`
`The formation of TBARS during an acid-heating reaction was measured as an index
`of ROS production, which is widely adopted as a sensitive method for measurement of
`lipid peroxidation, as previously described (Draper and Hadley, 1990). Briefly, the sam-
`ples were mixed with 1 mL of trichloroacetic acid 10% (TCA) and 1 mL of thiobarbituric
`acid 0.67% (TBA), then heated in a boiling water bath for 15 min. TBARS were deter-
`mined by the absorbance at 535 nm. Results are expressed as MDA (malondialdehyde)
`equivalents (nmol/mg protein).
`
`2.6. Measurement of protein carbonyls
`
`The oxidative damage to proteins was assessed by the determination of carbonyl
`groups based on the reaction with dinitrophenylhidrazine (DNPH) as previously de-
`scribed (Levine et al., 1994). Briefly, proteins were precipitated by the addition of
`20% trichloroacetic acid and redissolved in DNPH and the absorbance read at 370 nm.
`
`2.7. Statistic analysis
`
`All analyses were performed with the statistical package for social sciences version
`19.0 (SPSS Inc. Chicago, IL, USA). All data are presented as mean+ S.E.M. To test differ-
`ences between groups, we used analysis of variance (ANOVA), followed by Tukey post-
`hoc tests. In all experiments, P values less than 0.05 were considered to indicate statistical
`significance.
`
`3. Results
`
`Administration of m-AMPH at 1 mg/kg increased locomotor activ-
`ity in rats, and both Li and VPA were not able to prevent the hyperac-
`tivity induced by m-AMPH (Fig. 1A). Administration of m-AMPH
`1 mg/kg significantly increased the carbonyl production (a direct
`index of cell protein peroxidation) in prefrontal, amygdala, hippo-
`campus and striatum of rats. In addition, Li and VPA were not able
`to prevent the m-AMPH-induced protein damage in the brain of rats
`(Fig. 1B). We also found that m-AMPH 1 mg/kg increased lipid perox-
`idation in all brain structure evaluated — as indicated by increased
`levels of TBARS. Pretreatment with Li increased and VPA decreased the
`lipid damage induced by m-AMPH 1 mg/kg in hippocampus. Converse-
`ly, Li prevent the m-AMPH-induced lipid damage in the striatum of rats.
`In prefrontal and amygdala the mood stabilizers were not able to protect
`against lipid damage induced by m-AMPH 1 mg/kg (Fig. 1C).
`Administration of m-AMPH at 0.5 mg/kg increased locomotor activity
`in rats, and both Li and VPA partially prevent the hyperactivity induced
`by m-AMPH (Fig. 2A). The administration of m-AMPH 0.5 mg/kg in rats
`increased the protein damage in all brain structures evaluated, and
`mood stabilizers were not able to prevent the m-AMPH-induced protein
`damage in the brain of rats (Fig. 2B). We verified also that administration
`of m-AMPH 0.5 mg/kg increased TBARS generation in all brain structures
`evaluated. The pretreatment with VPA significantly diminished AMPH-
`induced protein damage in all brain structures evaluated. Additionally,
`
`2 of 6
`
`Alkermes, Ex. 1051
`
`

`
`D.D. da-Rosa et al. / Psychiatry Research 198 (2012) 521–526
`
`523
`
`Fig. 1. (A) Number of crossings (n=12 for each group). (B) Protein carbonyl content
`(n=5 for each group). (C) TBARS content (n=5 for each group) in the prevention
`model. Rats were pretreated with Li (47.5 mg/kg) or VPA (200 mg/kg) for seven days
`and then and then were treated with plus amphetamine until the 14th day of the ex-
`periment. Li = lithium, VPA = valproate. Bars represent means; error bars represent
`standard error of the means (SEM). *Different to the saline group. */# Different to the
`m-AMPH (1 mg/kg) group.
`
`Fig. 2. (A) Number of crossings (n=12 for each group). (B) Protein carbonyl content
`(n=5 for each group). (C) TBARS content (n=5 for each group) in the prevention
`model. Rats were pretreated with Li (47.5 mg/kg) or VPA (200 mg/kg) for seven days
`and then and then were treated with plus amphetamine until the 14th day of the ex-
`periment. Li = lithium, VPA = valproate. Bars represent means; error bars represent
`standard error of the means (SEM). *Different to the saline group. */# Different to the
`m-AMPH (0.5 mg/kg) group.
`
`Li partially prevents the lipid damage induced by m-AMPH 0.5 mg/kg in
`the hippocampus and striatum (Fig. 2C).
`Administration of m-AMPH at 0.25 mg/kg increased locomotor ac-
`tivity in rats and both Li and VPA prevented the m-AMPH-induced hy-
`peractivity in rats (Fig. 3A). Besides, the administration of m-AMPH
`0.25 mg/kg increased carbonyl group formation in all brain structure
`evaluated, and both Li and VPA prevented the protein damage induced
`by m-AMPH 0.25 mg/kg (Fig. 3B). The administration of m-AMPH
`0.25 mg/kg increased lipid damage in all brain structures evaluated.
`Pretreatment with Li protects the brain against lipid damage induced
`by m-AMPH 0.25 mg/kg in the prefrontal and partially protects the
`amygdala and the hippocampus. In addition, pretreatment with VPA
`
`prevents the m-AMPH-induced lipid damage in the prefrontal, the
`amygdala and the hippocampus (Fig. 3C).
`In the reversion protocol, the administration of m-AMPH 0.25 mg/kg
`increased locomotor activity in rats and Li and VPA reversed the hyper-
`activity induced by m-AMPH (Fig. 4A). Once more, the administration of
`m-AMPH 0.25 mg/kg increased the carbonyl group formation and Li re-
`versed m-AMPH-induced protein damage in all brain structure evaluat-
`ed. In addition, the treatment with VPA reversed the m-APH-induced
`protein damage (Fig. 4B) in the prefrontal and the hippocampus and
`partially reversed in the striatum. Besides, m-AMPH 0.25 mg/kg in-
`creased lipid damage in all brain structures evaluated. The treatment
`with Li diminished the lipid damage induced by m-AMPH 0.25 mg/kg
`
`3 of 6
`
`Alkermes, Ex. 1051
`
`

`
`524
`
`D.D. da-Rosa et al. / Psychiatry Research 198 (2012) 521–526
`
`Fig. 3. (A) Number of crossings (n=12 for each group). (B) Protein carbonyl content
`(n=5 for each group). (C) TBARS content (n=5 for each group) in the prevention
`model. Rats were pretreated with Li (47.5 mg/kg) or VPA (200 mg/kg) for seven days
`and then and then were treated with plus amphetamine between the 8th and 14th
`days. Li = lithium, VPA = valproate. Bars represent means; error bars represent standard
`error of the means (SEM). *Different to the saline group. */# Different to the m-AMPH
`(0.25 mg/kg) group.
`
`in the prefrontal. Moreover, treatment with VPA diminished the
`m-AMPH-induced lipid damage in the prefrontal and amygdala (Fig. 4C).
`
`4. Discussion
`
`In the present study we observed that all doses of m-AMPH (0.25,
`0.5, or 1 mg/kg) increased locomotor activity in rats. However, this
`hyperactivity was prevented and reverted by mood stabilizers – Li
`and VPA – only when m-AMPH was administered in the dose of
`0.25 mg/kg. In the high doses of m-AMPH, 1 and 0.5 mg/kg, Li and
`VPA were not able to prevent the hyperactivity induced in the rats.
`
`Fig. 4. (A) Number of crossings (n=12 for each group). (B) Protein carbonyl content
`(n=5 for each group). (C) TBARS content (n=5 for each group) in the reversal
`model. Rats were pretreated with m-amphetamine for seven days and then treated
`with amphetamine plus Li (47.5 mg/kg) or VPA (200 mg/kg) between the 8th and
`14th days. Li = lithium, VPA = valproate. Bars represent means; error bars represent
`standard error of the means (SEM). *Different to the saline group. # Different to the
`m-AMPH (0.25 mg/kg) group.
`
`According, Ellenbroek and Cools (1990) the validity of animal
`models in psychiatric disorders should demonstrate the face, con-
`struct and predictive validities. The clinical hallmark of BD is acute
`mania (Belmaker, 2004), showing symptoms such as irritable mood,
`psychomotor activation, reduced need for sleep, and excessive in-
`volvement in potentially problematic behavior (El-Mallakh et al.,
`2003). According to several guidelines or consensus statements, lith-
`ium, anticonvulsivants such as valproic acid and carbamazepine, and
`the second generation antipsychotics are recommended for the phar-
`macological treatment of BD. Here, we demonstrated that m-AMPH
`0.25 mg/kg administration increases locomotor activity of rats, and
`the usual mood stabilizers – Li and VPA – prevents and reverses this
`hyperactivity in the animals, presenting the face and predictive valid-
`ities of the model.
`
`4 of 6
`
`Alkermes, Ex. 1051
`
`

`
`D.D. da-Rosa et al. / Psychiatry Research 198 (2012) 521–526
`
`525
`
`It is known that m-AMPH and D-AMPH induces hyperactivity in
`animals by elevation in extracellular DA (Martinez et al., 2003).
`There are several studies showing the importance of dopaminergic
`transmission in the behavioral response to AMPHs. It is well de-
`scribed in the literature that amphetamine dose dependently in-
`creases extracellular levels of DA in the brain (Moghaddam and
`Bunney, 1989; Maisonneuve et al., 1990). Moreover, the blockade of
`D1 receptors in the medial prefrontal cortex attenuates AMPH- and
`m-AMPH-induced locomotor activity in the rats (Hall et al., 2009).
`In addition, intracerebral administration of 6-OHDA, which induces
`depletion of DA, prevents AMPH-induced hyperactivity (Dunnett et
`al., 1984; Banks and Gratton, 1995, King and Finlay, 1995) as well as
`the development of behavioral sensitization following repeated expo-
`sure to amphetamine (Bjijou et al., 2002).
`Joyce et al. (1995) have found that higher urinary dopamine levels
`have been associated with the emergence of manic symptoms. In ad-
`dition, studies have demonstrated dopamine receptor changes in BD
`patients (Pantazopoulos et al., 2004; Vogel et al., 2004). These studies
`suggest that the dopaminergic system may play a role in the patho-
`physiology of BD. In addition, the mechanism of AMPH toxicity has
`been suggested that involves an increase in the formation of reactive
`oxygen species (Chipana et al., 2008). It has been widely demonstrat-
`ed that the generation of reactive oxygen species (ROS) plays a criti-
`cal role in the pathophysiology of BD (Kunz et al., 2008; Andreazza et
`al., 2010a,b; Steckert et al., 2010). In the present study we also found
`that m-AMPH in all doses administrated increased protein and lipid
`damage in the brain of rats. Finally, the increase in extracellular dopa-
`mine together with the oxidative damage induced by m-AMPH, both
`present in BD, characterizes the construct validity of the model.
`Another important result found here is that Li and VPA are not able
`to prevent protein damage induced by m-AMPH 1 and 0.5 mg/kg.
`However, when given m-AMPH at 0.25 mg/kg these mood stabilizers
`prevented and reversed the m-AMPH-induced protein damage. Fur-
`thermore, Li increased the lipid damage induced by m-AMPH 1 mg/kg
`in the prefrontal and VPA partially protects lipid damage only in the
`hippocampus and striatum. When m-AMPH was administered at
`0.5 mg/kg VPA diminished the m-AMPH-induced lipid damage in all
`brain structures evaluated. However, Li diminished the lipid damage in-
`duced by m-AMPH 0.5 mg/kg only in the hippocampus and striatum.
`Additionally, Li and VPA partially protect against lipid damage induced
`by m-AMPH 0.25 mg/kg in the prefrontal, amygdala and hippocampus,
`but not in the striatum. In the reversal protocol, the lipid damage in-
`duced by m-AMPH 0.25 mg/kg was diminished by VPA in the prefrontal
`and amygdala and by Li in the prefrontal. This discrepancy between the
`prevention and reversion protocols may be due to the fact that the time
`of treatment with mood stabilizers in the prevention protocol is greater.
`In previous studies of our laboratory we demonstrated that D-AMPH
`increased TBARS levels in the prefrontal cortex, and this effect was re-
`versed by both mood stabilizers — Li and VPA. It was also observed
`that the administration of Li and VPA increased TBARS formation in
`the hippocampus of rats pretreated with D-AMPH. In the same study,
`it has been demonstrated that the VPA pretreatment protects the pre-
`frontal and the hippocampus against D-AMPH-induced lipid damage.
`However, Li partially prevented D-AMPH-induced lipid peroxidation in
`the rat hippocampus but augmented in the prefrontal (Frey et al.,
`2006c). Together, the previous studies and the present study demon-
`strate that the effect of mood stabilizers depends on the experimental
`protocol and the brain area analyzed. Regions of the central nervous
`systems differentially respond to insults (Sullivan et al., 2004) and
`TBARS formation was evaluated from different brain regions, that in
`part represent different cell types. Furthermore, within a homogeneous
`population of cells, there is heterogeneity in terms of physiological and
`metabolic characteristics (Lai et al., 1977; Sims, 1991; Sonnewald et al.,
`1998).
`It is noteworthy that in the animal model of mania induced by
`D-AMPH protein damage was not observed (Frey et al., 2006c).
`
`Here we show that m-AMPH 0.25 mg/kg induces protein damage,
`which is reversed and prevented by administration of mood stabilizers.
`In a postmortem study, it was observed that protein oxidation and nitra-
`tion increased in the prefrontal cortex of bipolar patients (Andreazza et
`al., 2010a,b). In addition, Andreazza et al. (2009) found a significant in-
`crease in 3-nitrotyrosine (marker of protein damage) levels in peripher-
`al blood of patients in the early and late stages of bipolar disorder. From
`these evidences, we suggest that the animal model of mania induced by
`m-AMPH is more effective than D-AMPH in mimicking the oxidative
`brain damage seen in bipolar patients.
`
`5. Conclusions
`
`In conclusion, in the present study we demonstrate that the mood
`stabilizers Li and VPA reversed and prevented m-AMPH-induced hy-
`peractivity when m-AMPH was administered at 0.25 mg/kg. In addi-
`tion, the hyperactivity induced by m-AMPH was associated with
`lipid and protein damage, which Li and VPA were able to reverse
`and prevent depending on the experimental protocol and the brain
`region evaluated. Then, we propose a new model of mania induced
`by m-AMPH, being that this model has face, predictive and construct
`validities.
`
`References
`
`Andreazza, A.C., Kauer-Sant'Anna, M., Frey, B.N., Stertz, L., Zanotto, C., Ribeiro, L.,
`Giasson, K., Valvassori, S.S., Réus, G.Z., Salvador, M., Quevedo, J., Gonçalves, C.A.,
`Kapczinski, F., 2008. Effects of mood stabilizers on DNA damage in an animal
`model of mania. Journal of Psychiatry & Neuroscience 33, 516–524.
`Andreazza, A.C., Kapczinski, F., Kauer-Sant'Anna, M., Walz, J.C., Bond, D.J., Gonçalves,
`C.A., Young, L.T., Yatham, L.N., 2009. 3-Nitrotyrosine and glutathione antioxidant
`system in patients in the early and late stages of bipolar disorder. Journal of Psychiatry
`& Neuroscience 34, 263–271.
`Andreazza, A.C., Shao, L., Wang, J.F., Young, L.T., 2010a. Mitochondrial complex I activity
`and oxidative damage to mitochondrial proteins in the prefrontal cortex of pa-
`tients with bipolar disorder. Archives of General Psychiatry 67, 360–368. Erratum
`in: Archives of General Psychiatry, 67, p. 1254.
`Andreazza, A.C., Shao, L., Wang, J.F., Young, L.T., 2010b. Mitochondrial complex I activ-
`ity and oxidative damage to mitochondrial proteins in the prefrontal cortex of
`patients with bipolar disorder. Archives of General Psychiatry 67, 360–368.
`Banks, K.E., Gratton, A., 1995. Possible involvement of medial prefrontal cortex in
`amphetamine-induced sensitization of mesolimbic dopamine function. European
`Journal of Pharmacology 282, 157–167.
`Barros, D.M., Izquierdo, L.A., Medina, J.H., Izquierdo, I., 2002. Bupropion and sertraline
`enhance retrieval of recent and remote long-term memory in rats. Behavioural
`Pharmacology 13, 215–220.
`Belmaker, R.H., 2004. Bipolar disorder. The New England Journal of Medicine 351,
`476–486.
`Berk, M., Dodd, S., Kauer-Sant'anna, M., Malhi, G.S., Bourin, M., Kapczinski, F., Norman, T.,
`2007. Dopamine dysregulation syndrome: implications for a dopamine hypothesis of
`bipolar disorder. Acta Psychiatrica Scandinavica. Supplementum 434, 41–49.
`Bjijou, Y., De Deurwaerdere, P., Spampinato, U., Stinus, L., Cador, M., 2002. D-amphetamine-
`induced behavioral sensitization: effect of lesioning dopaminergic terminals in the
`medial prefrontal cortex, the amygdala and the entorhinal cortex. Neuroscience 109,
`499–516.
`Boksa, P., 2007. Of rats and schizophrenia. Journal of Psychiatry & Neuroscience 32, 8–10.
`Chipana, C., Torres, I., Camarasa, J., Pubill, D., Escubedo, E., 2008. Memantine protects against
`amphetamine derivatives-induced neurotoxic damage in rodents. Neuropharmacology
`54, 1254–1263.
`Draper, H.H., Hadley, M., 1990. Malondialdehyde determination as index of lipid perox-
`idation. Methods in Enzymology 186, 421–431.
`Dunnett, S.B., Bunch, S.T., Gage, F.H., Björklund, A., 1984. Dopamine-rich transplants in
`rats with 6-OHDA lesions of the ventral tegmental area. I. Effects on spontaneous
`and drug-induced locomotor activity. Behavioural Brain Research 13, 71–82.
`Einat, H., Yuan, P., Gould, T.D., Li, J., Du, J., Manji, H.K., Chen, G., 2003. The role of the ex-
`tracellular signal-regulated kinase signaling pathway in mood regulation. The Journal
`of Neuroscience 23, 7311–7316.
`El-Mallakh, R.S., El-Masri, M.A., Huff, M.O., Li, X.P., Decker, S., Levy, R.S., 2003. Intracer-
`ebroventricular administration of ouabain as a model of mania in rats. Bipolar
`Disorders 5, 362–365.
`Ellenbroek, B.A., Cools, A.R., 1990. Animal models with construct validity for schizophrenia.
`Behavioural Pharmacology 6, 469–490.
`Fiorino, D.F., Phillips, A.G., 1999. Facilitation of sexual behavior and enhanced dopa-
`mine efflux in the nucleus accumbens of male rats after D-amphetamine-induced
`behavioral sensitization. The Journal of Neuroscience 1, 456–463.
`Fischer, J.F., Cho, A.K., 1979. Chemical release of dopamine from striatal homogenates:
`evidence for an exchange diffusion model. The Journal of Pharmacology and Exper-
`imental Therapeutics 208, 203–209.
`
`5 of 6
`
`Alkermes, Ex. 1051
`
`

`
`526
`
`D.D. da-Rosa et al. / Psychiatry Research 198 (2012) 521–526
`
`Frey, B.N., Martins, M.R., Petronilho, F.C., Dal-Pizzol, F., Quevedo, J., Kapczinski, F.,
`2006a. Increased oxidative stress after repeated amphetamine exposure: possible
`relevance as a model of mania. Bipolar Disorders 8, 275–280.
`Frey, B.N., Valvassori, S.S., Gomes, K.M., Martins, M.R., Dal-Pizzol, F., Kapczinski, F.,
`Quevedo, J., 2006b. Increased oxidative stress in submitochondrial particles after
`chronic amphetamine exposure. Brain Research 1097, 224–229.
`Frey, B.N., Valvassori, S.S., Réus, G.Z., Martins, M.R., Petronilho, F.C., Bardini, K., Dal-
`Pizzol, F., Kapczinski, F., Quevedo, J., 2006c. Effects of lithium and valproate on
`amphetamine-induced oxidative stress generation in an animal model of mania.
`Journal of Psychiatry & Neuroscience 31, 326–332.
`Gluck, M.R., Moy, L.Y., Jayatilleke, E., Hogan, K.A., Manzino, L., Sonsalla, P.K., 2001. Par-
`allel increases in lipid and protein oxidative markers in several mouse brain re-
`gions after methamphetamine treatment. Journal of Neurochemistry 79, 152–160.
`Greengard, P., 2001. The neurobiology of slow synaptic transmission. Science 294,
`1024–1030.
`Hall, D.A., Powers, J.P., Gulley, J.M., 2009. Blockade of D1 dopamine receptors in the me-
`dial prefrontal cortex attenuates amphetamine- and methamphetamine-induced
`locomotor activity in the rat. Brain Research 1300, 51–57.
`Joyce, P.R., Fergusson, D.M., Woollard, G., Abbott, R.M., Horwood, L.J., Upton, J., 1995.
`Urinary catecholamines and plasma hormones predict mood state in rapid cycling
`bipolar affective disorder. Journal of Affective Disorders 33, 233–243.
`King, D., Finlay, J.M., 1995. Effects of selective dopamine depletion in medial prefrontal
`cortex on basal and evoked extracellular dopamine in neostriatum. Brain Research
`685, 117–128.
`Kuczenski, R., Segal, D.S., Cho, A.K., Melega, W., 1995. Hippocampus norepinephrine,
`caudate dopamine and serotonin, and behavioral responses to the stereoisomers
`of amphetamine and methamphetamine. Journal of Neuroscience 15, 1308–1317.
`Kunz, M., Gama, C.S., Andreazza, A.C., Salvador, M., Ceresér, K.M., Gomes, F.A.,
`Belmonte-de-Abreu, P.S., Berk, M., Kapczinski, F., 2008. Elevated serum superoxide
`dismutase and thiobarbituric acid reactive substances in different phases of bipolar
`disorder and in schizophrenia. Progress in Neuro-psychopharmacology and Biological
`Psychiatry 32, 1677–1681.
`Kupfer, D.J., 2005. The increasing medical burden in bipolar disorder. JAMA : The Journal of
`the American Medical Association 293, 2528–2530.
`Lai, Y.L., Rodarte, J.R., Hyatt, R.E., 1977. Effect of body position on lung emptying in
`recumbent anesthetized dogs. Journal of Applied Physiology 43, 983–987.
`Levine, R.L., Williams, J.A., Stadtman, E.R., Shacter, E., 1994. Carbonyl assays for deter-
`mination of oxidatively modified proteins. Methods in Enzymology 233,
`346–357.
`Lipska, B.K., Weinberger, D.R., 2000. To model a psychiatric disorder in animals: schizo-
`phrenia as a reality test. Neuropsychopharmacology 23, 223–239.
`Machado-Vieira, R., Schmidt, A.P., Avila, T.T., Kapczinski, F., Soares, J.C., Souza, D.O.,
`Portela, L.V., 2004. Increased cerebrospinal fluid levels of S100B protein in rat
`model of mania induced by ouabain. Life Sciences 76, 805–811.
`Maisonneuve, I.M., Keller, R.W., Glick, S.D., 1990. Similar effects of D-amphetamine and
`coc

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket