Abstract
Mania has long been recognized as aberrant behaviour indicative of mental illness. Manic states include a variety of complex and multifaceted symptoms that challenge clear clinical distinctions. Symptoms include over-activity, hypersexuality, irritability and reduced need for sleep, with cognitive deficits recently linked to functional outcome. Current treatments have arisen through serendipity or from other disorders. Hence, treatments are not efficacious for all patients, and there is an urgent need to develop targeted therapeutics. Part of the drug discovery process is the assessment of therapeutics in animal models. Here we review pharmacological, environmental and genetic manipulations developed to test the efficacy of therapeutics in animal models of mania. The merits of these models are discussed in terms of the manipulation used and the facet of mania measured. Moreover, the predictive validity of these models is discussed in the context of differentiating drugs that succeed or fail to meet criteria as approved mania treatments. The multifaceted symptomatology of mania has not been reflected in the majority of animal models, where locomotor activity remains the primary measure. This approach has resulted in numerous false positives for putative treatments. Recent work highlights the need to utilize multivariate strategies to enable comprehensive assessment of affective and cognitive dysfunction. Advances in therapeutic treatment may depend on novel models developed with an integrated approach that includes: (i) a comprehensive battery of tests for different aspects of mania, (ii) utilization of genetic information to establish aetiological validity and (iii) objective quantification of patient behaviour with translational cross-species paradigms.
LINKED ARTICLES
This article is part of a themed issue on Translational Neuropharmacology. To view the other articles in this issue visit http://dx.doi.org/10.1111/bph.2011.164.issue-4
Keywords: mania, animal models, activity, cognition, pharmacological, genetic, environmental, mice, rats, false positives
Mania: from clinical presentation to the challenges of validating animal models
Mania is typically defined as a distinct period characterized by elevated, expansive or irritable mood (APA, 1994). The word derives from the Greek term ‘µανία’, describing a madness or frenzy (Liddell and Scott, 1940), and literary descriptions of the phenomenon date back to antiquity (Jackson, 1986). Mania, as the very first word of Homer's epic saga, The Iliad, was used to describe the uncontained rage of Achilles against Agamemnon (Lattimore, 1961). This term was also defined as a mental illness by Hippocrates, the famed Hellenic physician and teacher (Luneberg, 1897). By the early 19th century, mania was considered to be part of the broad family of ‘insanities’ and was subcategorized into delusional and non-delusional states (Pinel, 1818). Other authors described mania as a class of behaviours resembling schizophrenia, with subtypes such as simplex (pure rage), estatica (insane), ecnoa (rage with folly) and catholica (common rage) (Berrios, 1988). Near the end of the century, the concept of mania was associated with depression within the Kraepelin's terminology of manic–depressive insanity as distinct from dementia praecox, initiating the basis of modern psychiatric nosology (Kraepelin, 1899). Several decades later, Leonhard introduced the concept of polarity in the classification of affective disorders, reflecting the opposite extremes of depression and euphoria (Leonhard, 1957). Unipolar patients experienced only depressive episodes, while a bipolar diagnosis required the presence of a manic episode, a framework preserved by current diagnostic guidelines (APA, 1994).
Despite extensive efforts to categorize and define specific criteria that describe mania, manic states may include a variety of complex and multifaceted symptoms that challenge clear clinical distinctions (Grunze et al., 2009). While mania may be precipitated by medical conditions that include viral infection, head trauma and neurological disorders (Arora and Daughton, 2007), this state is most commonly associated with bipolar disorder (BD), an illness with a worldwide lifetime prevalence of approximately 1% (Grunze et al., 2009). According to the Diagnostic and Statistical Manual of Mental Disorders IV (DSM-IV) (APA, 1994), a manic episode as required for a BD diagnosis involves a distinct period of euphoric or irritable mood lasting at least 1 week (or less if hospitalized), concurrent with several other symptoms that may include grandiosity, decreased need for sleep, pressured speech, racing thoughts, distractibility, increased goal-directed activity and pleasurable risky activities (e.g. promiscuity, overspending; Figure 1). Some studies indicate that irritation is the most frequently exhibited mood symptom during a manic episode (occurring in 71% of patients; (Goodwin and Jamison, 2007), while psychotic symptoms such as delusions are observed approximately half the time. Hyperactivity is present in nearly all acute manic states (90%), followed by a decreased need for sleep (83%) and hyperverbosity (89%), while symptoms such as hypersexuality and religiosity appear less common (40–50%) (Goodwin and Jamison, 2007). Recent factor analyses of manic signs and symptoms (Cassidy et al., 1998a,b) have identified between four and seven independent factors, including dysphoria (characterized by anxiety and depressed mood associated with mixed episodes), psychomotor acceleration (increased motor activity, pressured speech), psychosis (grandiosity, psychotic symptoms) and irritability/aggression. Despite the diversity of manic subgroups (irritable, euphoric, psychotic, depressed), psychomotor agitation appears to be a common denominator, leading some to hypothesize that increased activation or ‘over-activity’ represents the core underlying feature of mania (Depue et al., 1987; Bauer et al., 1991; Perry et al., 2010).
Figure 1.

Symptoms and assays of mania. The symptoms of bipolar mania as described by DSM-IV rating scales (green circle). Also the neurocognitive symptoms associated with bipolar mania (yellow circle). The putative measurements of these symptoms (green and yellow rectangles respectively) are also provided. BPM = Behavioural Pattern Monitor, 5C-CPT = 5-choice continuous performance test, PPI = prepulse inhibition, IGT = Iowa gambling task, ASST = attentional set-shifting task, DD task = delayed discounting task.
Although classically conceptualized as a mood disorder, a considerable body of research indicates that BD mania is also associated with significant neurocognitive impairment across a broad array of domains (Burdick et al., 2007; Goodwin et al., 2008); Figure 1). While executive dysfunction appears to be the most salient aspect of BD mania (Savitz et al., 2005), BD patients in manic states also exhibit deficits in vigilance, working memory, verbal fluency, inhibitory control and verbal recall and recognition (Sax et al., 1999; Borkowska and Rybakowski, 2001; Fleck et al., 2003; Martinez-Aran et al., 2004). While most clinical trials for mania treatments have focused exclusively on the amelioration of mood symptoms, relatively little work has been done to examine the efficacy of various therapies on cognitive dysfunction linked with the disorder (Torres et al., 2010b), despite close correlation of cognitive disruption with functional outcome (Green, 2006a; Torres et al., 2010a). Inhibitory abnormalities are also observed in other domains such as in sensorimotor gating quantified as deficits in prepulse inhibition (PPI) (Perry et al., 2001). PPI is a cross-species measure where a response to a startling stimulus (such as a loud noise) is inhibited by the prior presentation of a low intensity prepulse (Braff et al., 1978; Swerdlow et al., 2002; Geyer, 2006b). Thus, there are numerous behaviours beyond that of traditional mania rating scales that can be modelled in animals.
While animal models of human illness have proved to be a vital tool in the management and treatment of disease, developing cross-species paradigms of mania remains a challenging task. As with all animal models of psychiatric illness, it is difficult to infer affective states in rodents that mirror the clinical symptoms and cognitive deficits that characterize human psychopathology. Mania is a disorder marked by a variety of symptoms, including grandiosity and pressured speech, which are extremely difficult to derive from rodent behaviour. While increased motor activity remains the mainstay of animal models of BD, this measure has also been used as a proxy for other neuropsychiatric disorders such as schizophrenia and attention-deficit hyperactivity disorder (ADHD) (Young et al., 2007). In addition, the aetiology and pathophysiology of mania remain unclear, limiting confidence in the construct validity of various animal models in the context of clinical medications whose therapeutic mechanism remains unknown. Recent attempts to model mania in rodents have focused on assessing specific facets or individual symptoms associated with a manic episode, while a comprehensive multi-symptom model remains elusive. In this review, we will (i) outline clinical findings that support the efficacy of current treatments for BD mania; (ii) describe the pharmacological, environmental and genetic animal models of the illness; (iii) evaluate the utility of the available models in providing effective pharmacological predictive validity and developing treatment options for mania.
Pharmacological treatments for mania
While the number of drugs licensed for treatment of manic episodes has grown substantially over the past decade (Smith et al., 2007), there remains a dearth of consensus in current clinical practice regarding the optimal treatment for mania (Bourin et al., 2005). Some groups recommend monotherapy with a mood stabilizer or atypical antipsychotic as a first-choice treatment option for BD mania (Suppes et al., 1995; Bauer et al., 1999), while others advocate combination treatment with these two classes of drugs, especially in the case of severe manic episodes (APA, 2002). While older clinical trials for mania medications were conducted with less rigorous methodological requirements and did not necessarily satisfy current scientific criteria (comparative, randomized, double-blind design), more recent studies have validated the efficacy of a variety of anti-manic drugs (Grunze et al., 2009). It is relevant to note, however, that treatment efficacy is most commonly defined as a 50% improvement on subjective measures such as the Young Mania Rating Scale (YMRS) (Smith et al., 2007), while objective quantification of manic symptoms such as motor hyperactivity is rarely assessed (Henry et al., 2010). Recent investigations indicate that genetic variation in BD could also play a important role in treatment effects; the response to lithium therapy may relate to variants in genes that code for the serotonin transporter, glycogen synthase kinase 3 (GSK-3) and inositol monophosphatase (IMPase) proteins (Cruceanu et al., 2006). To provide a context for the challenges associated with designing optimal animal models and interpreting the preclinical effects of anti-manic agents, this section will outline a brief history of the clinical work supporting currently accepted treatments for BD mania (Table 1). Following a review of the clinical measures requisite for validation of animal models, we will describe and critique the preclinical assays utilized to assess potential therapeutics.
Table 1.
Drugs approved/ not approved for treatment of mania
| Drug | Measure | Outcome | Reference |
|---|---|---|---|
| US Food and Drug Administration approved agents for mania | |||
| Lithium | BPRS | Significant reduction in elevated mood, excitement, grandiosity and hostility | Prien et al., 1972 |
| IMPS | |||
| SADS | Half of lithium-treated subjects demonstrated a 50% reduction in mania scale ratings | Bowden et al., 1994 | |
| Valproate | BPRS | Reduction in conceptual disorganization, tension, hostility, excitement and motor activity; | Pope et al., 1991 |
| YMRS | |||
| SADS | Half of valproate-treated subjects demonstrated a 50% reduction in YMRS score | Bowden et al., 1994 | |
| Carbamazepine | BPRS | Pooled data from randomized controlled trials indicate that approximately 52% of mania patients show reduction in manic symptoms | McElroy and Keck, 2000 |
| YMRS | |||
| BRMS | |||
| Aripiprazole | YMRS | Significant reduction in YMRS scores compared with placebo | Keck et al. (2003) |
| Asenapine | YMRS | Significant reduction in YMRS scores compared with placebo | McIntyre et al., 2009 |
| Olanzapine | YMRS | Significant reduction in YMRS scores compared with placebo | Tohen et al., 1999 |
| Quetiapine | YMRS | Significant reduction in YMRS scores compared with placebo; similar efficacy to haloperidol | McIntyre et al., 2007 |
| Resperidone | YMRS | Significant reduction of manic symptoms and diminished excitement/hostility on the PANSS compared with placebo | Khanna et al. (2005) |
| CGI | |||
| PANSS | |||
| Ziprasidone | YMRS | Significant reduction of manic and psychotic symptoms compared with placebo on multiple rating scales | Keck et al. (2003) |
| CGI | |||
| PANSS | |||
| SADS | |||
| Potential mania agents not approved for treatment | |||
| Lamotrigine | YMRS | No significant difference between drug treatment and placebo for treatment of acute manic episodes | Goldsmith et al., 2003 |
| Oxcarbazepine | YMRS | Inconsistent effects on manic symptoms, may have little effect on severe psychotic mania | Hummel et al. (2002) |
| Mazza et al., 2007 | |||
| Tiagabine | YMRS | No significant effects of drug treatment reported in some studies; data are limited by lack of randomized controlled trials | Young et al., 2006 |
| BRMS | |||
| Gabapentin | YMRS | No significant difference between drug treatment and placebo for treatment of acute manic episodes | Pande et al. (2000) |
| Fullerton et al. (2010) | |||
| Topiramate | YMRS | No significant difference between drug treatment and placebo of acute manic episodes | Kushner et al., 2006 |
BPRS = Brief Psychiatric Rating Scale; CGI = Clinical Global Impressions; BRMS = Beach–Rafaelsen Mania Scale; IMPS = Inpatient Multidimensional Psychiatric Scale; PANSS = Positive and Negative Syndrome Scale; SADS = Schedule for Affective Disorder and Schizophrenia; YMRS = Young Mania Rating Scale.
Lithium
Long considered to be the gold standard of treatment for bipolar disorder, lithium is one of the oldest psychotropic agents prescribed today (Grandjean and Aubry, 2009). Originally discovered in 1817, the drug has been used since the mid-19th century to treat conditions such as gout and rheumatism (Atack, 2000). In 1949, the Australian physician John Cade, who was investigating the toxicity of urine samples from psychotic patients (stored on the top shelf of his kitchen refrigerator), observed that injections of lithium urate induced lethargic behaviour in guinea pigs (Cade, 1978). After injecting himself with lithium carbonate to assess the safety of the drug, Cade observed that the compound reduced or eliminated manic symptoms (such as being restless and euphoric) in a sample of 10 manic patients but did not ameliorate psychotic symptoms in a group of schizophrenia patients (Cade, 1949). No clear diagnostic or assessment criteria were reported in this study, which consisted of a series of very brief case histories.
Following Cade's pioneering study, several reports provided independent but poorly controlled observations supporting the efficacy of lithium in mania. Schou and colleagues examined the effect of two weeks of lithium treatment on 38 manic patients using a within-subjects design, evaluating clinical response with a three-point global severity scale (Schou et al., 1954). While 37% of patients showed a ‘positive response’ to the drug, another 47% exhibited a ‘possible response’, but these criteria were vague and specific symptom improvement remained undefined. A subsequent review of 45 manic patients treated with lithium indicated that the majority showed an ‘entirely satisfactory’ or ‘considerably improved’ condition but again failed to provide an adequate description of the assessment criteria (Hartigan, 1963). A concurrent study examined the effect of 2 weeks of lithium therapy or placebo on manic symptoms using the Wittenborn scale for Manic State and Schizophrenic Excitement (Maggs, 1963). Although the diagnostic criteria for patient selection were not clear and alterations in specific symptoms were not reported, the results indicated that the drug was superior to placebo in reducing the severity of manic episodes.
In the decade following these initial reports, a series of larger controlled studies confirmed the efficacy of lithium therapy for mania, culminating in Food and Drug Administration (FDA) approval for treatment of acute manic episodes (1970) and as a prophylactic agent (1974) (Bourin et al., 2005). Baastrup and colleagues observed the effect of chronic lithium treatment on 88 bipolar subjects over a 6 year duration, concluding that the drug significantly reduced the number of manic and depressive episodes (Baastrup and Schou, 1967). Goodwin et al. (1969) conducted a double-blind placebo-controlled study, demonstrating that lithium reduced manic symptomatology on the Bunney–Hamburg Scale, a 25-item rating instrument used to quantify mania and depression (Bunney et al., 1968; Goodwin et al., 1969). Several additional reports indicated that lithium exhibited an anti-manic effect comparable to the antipsychotic chlorpromazine as assessed by measures such as the Brief Psychiatric Rating Scale (BPRS) (Johnson et al., 1968; Spring et al., 1970; Johnson et al., 1971). One study observed that both drugs reduced sleep disturbance and grandiosity, but lithium was superior in controlling behaviours such as motor hyperactivity, flight of ideas, euphoria and pressured speech (Spring et al., 1970). In 1972, the Veterans Administration (VA) and National Institute of Mental Health (NIMH) conducted a collaborative 18-site study, reporting that 3 weeks of treatment with both drugs significantly reduced BPRS-rated symptoms such as excitement, elevated mood, grandiosity and hostility in manic and schizoaffective patients (Prien et al., 1972). Finally, more recent randomized, multicentre, double-blind studies (Bowden et al., 1994; Poolsup et al., 2000) have conclusively demonstrated that lithium is superior to placebo in reducing acute manic symptoms as assessed by multiple rating scales, including the BPRS, the YMRS and the Schedule for Affective Disorders and Schizophrenia (SADS-C).
Anticonvulsants
Anticonvulsant medications have been evaluated as mood stabilizers since the early 1970s, buttressed by the proposal that epilepsy and BD may share similar underlying features such as kindling, a phenomena involving repeated subthreshold neuronal stimulation (Post and Uhde, 1983; Brambilla and Soares, 2005). Substantial evidence supports the use of anticonvulsants such as valproate and carbamazepine in the treatment of mania, while more recent studies have examined the effectiveness of newer compounds such as lamotrigine, oxcarbazepine and topiramate (Singh et al., 2005). Valproate, originally developed as an organic solvent in 1881, was recognized serendipitously as an anticonvulsant in the 1960s and first observed to exert clinical benefit in BD in a 1966 French study (Lambert et al., 1966). Two pivotal reports in the early 1990s led to regulatory approval in the United States in 1995 (Bowden and Singh, 2006). Pope and colleagues conducted a double-blind placebo-controlled study of valproate treatment in 36 acute manic patients, reporting that the drug reduced YMRS-rated manic symptoms by an average of 54% and significantly mitigated BPRS-evaluated symptoms such as conceptual disorganization, tension, excitement and hostility relative to placebo (Pope et al., 1991). A subsequent randomized, multicentre study (Bowden et al., 1994) reported that valproate significantly improved manic symptoms as assessed by the SADS-C scale, decreasing elevated mood, motor hyperactivity and sleep disturbance compared with the placebo group. More recent studies indicated that valproate may be superior to lithium in treating mania characterized by co-morbid depressive symptoms, marked irritability or a higher number of lifetime manic episodes (Bowden and Singh, 2006).
Similar to valproate, the efficacy of carbamazepine treatment for mania has been demonstrated primarily through the use of rating scales (Bourin et al., 2005). Carbamazepine, the first anticonvulsant drug widely used for the treatment of BD, has been assessed as a therapy for mania in at least 16 controlled trials (Singh et al., 2005), although many of these studies were limited by small sample size, the absence of a placebo group and concurrent use of other anti-manic drugs (Keck et al., 2000). Pooled data from five studies with carbamazepine monotherapy indicates that the drug is superior to placebo and comparable with lithium and typical antipsychotics in treating BD mania (McElroy and Keck, 2000) and alleviating manic symptoms assessed by the BPRS, the Manic State Rating Scale and the Bunney–Hamburg Scale (Keck et al., 2000).
In contrast, a number of so-called ‘third-generation’ or atypical anticonvulsants have failed to effectively treat acute manic episodes of BD, including oxcarbazepine (Mazza et al., 2007), tiagabine, gabapentin, topiramate and lamotrigine (Yatham et al., 2002; Kushner et al., 2006). While a series of open-label and pilot studies reported that oxcarbazepine reduces manic symptoms on measures such as the YMRS and Bech–Rafaelson Mania Scale (Hirschfeld and Kasper, 2004; Popova et al., 2007), these results were not confirmed by subsequent controlled studies (Mazza et al., 2007). Administration of topiramate was indicated to decrease YMRS scores in BD patients as an adjunctive treatment or monotherapy (Yatham and Kusumaker, 2003), but more recent double-blind placebo-controlled trials did not observe significant effects of the drug (Kushner et al., 2006). While initial results indicated lamotrigine decreased manic symptoms as measured by the YMRS in a within subjects design (Bowden et al., 1999), these effects were not robust in two large placebo-controlled clinical trials (Bowden, 2002; Goldsmith et al., 2003; Singh et al., 2005; Ghaemi et al., 2008; Weisler et al., 2008). Despite these findings and lack of FDA approval, some of these drugs have been used as off-label treatment for BD mania (Grunze et al., 2009). In comparison, older drugs such as lithium have retained their clinical efficacy even when assessed in more recent and rigorous clinical trials (Bowden et al., 1994; Sachs et al., 2007). In addition, in the case of lamotrigine, inadequate description of the clinical findings (e.g. a dearth of details regarding drug efficacy on symptoms such as aggression, sleep disturbance or cognition) (Goldsmith et al., 2003) complicates interpretation of drug effects in animal models.
Antipsychotics
Antipsychotic medications have been utilized as a primary or adjunctive treatment for mania for over 50 years (Cookson, 2006). Clinical use of antipsychotic agents such as chlorpromazine began in the 1950s, predating widespread use of lithium (Tohen and Vieta, 2009). For decades, haloperidol was often the drug of first choice to rapidly tranquilize agitated or aggressive acutely manic patients, in contrast to the delayed response to lithium (Cookson, 2006). While typical antipsychotic drugs such as chlorpromazine and haloperidol were comparable with lithium in reducing manic symptomatology as assessed by the BPRS (Shopsin et al., 1975), more recent studies suggested that the atypical antipsychotic clozapine could have potential therapeutic efficacy in mania (Keck and McElroy, 2006). Over the past 10 years, a series of atypical antipsychotics have gained FDA approval to treat mania, including aripiprazole, asenapine, olanzapine, quetiapine, risperidone and ziprasidone (McIntyre et al., 2009; Tohen and Vieta, 2009). The efficacy of these drugs as monotherapy or adjunctive treatments to lithium or valproate has again been demonstrated primarily with the use of rating scales, including the YMRS, BPRS and Clinical Global Impression (CGI) (Cookson, 2006; Smith et al., 2007; Fountoulakis and Vieta, 2008; Grunze et al., 2009). While some reports include symptom-specific data (e.g. improvement in elevated mood with olanzapine, a reduction in hostility with risperidone and aripiprazole, or a general improvement across all the YMRS categories with quetiapine; (Tohen et al., 1999; Hirschfeld and Kasper, 2004; McIntyre et al., 2007; Keck et al., 2009b), data from many recent clinical trials are limited to assessing the overall change in the total YMRS score (Fountoulakis and Vieta, 2008). Although potential mechanisms of action remain unknown, this class of drugs does appear to exert a specific anti-manic effect independent of the presence of co-morbid psychosis or the degree of sedation induced by the agent (Keck and McElroy, 2006; McIntyre et al., 2007; Sachs and Gardner-Schuster, 2007; Tohen et al., 1999).
Investigational agents
Despite numerous anti-manic medications now available, identifying the neurobiological factors underlying manic episodes and the therapeutic targets of efficacious treatments remains a challenge. Numerous biochemical effects of lithium have been identified as potential mediators of the therapeutic response, including inhibition of IMPase, inhibition of GSK-3, inhibition of NMDA receptor–mediated signalling, alterations in cyclic adenosine monophosphate (cAMP) signalling and guanine nucleotide binding protein (G-protein) function, inhibition of protein kinase C (PKC) and augmentation of cholinergic activity (Manji et al., 2000; Chiu and Chuang, 2010). Anticonvulsants such as valproate may also exert anti-manic properties through a variety of mechanisms, including increased gamma-aminobutyric (GABA) release, inhibition of GSK-3 and PKC and activation of extracellular-related signal kinase (ERK) (Bowden and Singh, 2006). Both typical and atypical antipsychotic medications are hypothesized to not only reduce manic symptoms via dopamine D2 receptor blockade but to also impact multiple neurotransmitter systems crucial to regulating affect (Tohen and Vieta, 2009). Several recent clinical trials (NCT00026585, NCT00206544, NCT00411203 on http://www.clinicaltrials.gov) have focused specifically on PKC as a molecular target by assessing the effect of the PKC inhibitor Tamoxifen, a non-steroidal anti-oestrogen agent used in the treatment of breast cancer (DiazGranados and Zarate, 2008). PKC is an intracellular enzyme that coordinates the neuronal response to the activation of multiple neurotransmitter receptors, playing a key role in regulating pre- and post-synaptic transmission (Zarate et al., 2007). Data from four published studies indicate that tamoxifen reduces BD manic symptoms relative to placebo as indicated by decreased YMRS scores, including a reduction in elevated mood, motor hyperactivity and sexual interest (Bebchuk et al., 2000; Kulkarni et al., 2006; Zarate et al., 2007; Yildiz et al., 2008).
While almost all clinical studies focus on the psychiatric symptoms associated with mania, there is a growing awareness that treatments for disorders such as BD should also address the neurocognitive deficits coupled with the illness. Bipolar mania is characterized by significant impairment in several cognitive domains, including attention, memory and executive functioning (i.e. planning, abstract concept formation, set shifting, decision making and inhibitory control) (Doyle et al., 2005; Green, 2006b; Burdick et al., 2007; Bora et al., 2009). Current clinical trials are assessing the efficacy of several agents, including ashwaganhda (Withania somnifera), L-carnosine, memantine, methylene blue, intranasal insulin, pramipexole and galantamine on cognitive function in BD (see http://www.clinicaltrials.gov, trial identifiers NCT00761761, NCT00177463, NCT00586066, NCT00214877, NCT00314314, NCT00597896, NCT00741598). Outcome measures include performance in the domains of attention, executive function, working memory and verbal and visuospatial memory. Neuropsychological paradigms utilized to quantify cognition include Cogtest, the California Verbal Learning Task (CVLT), Trail Making Test A (insulin), Trail Making Test B and the Connors Continuous Performance Task (CPT). While much of this information has yet to be published, one recent study reported that 4 months of treatment with galantamine-ER improved attentional deficits (better CPT performance) and normalized lipid membrane metabolism in the left hippocampus of BD participants (Iosifescu et al., 2009). Finally, memantine is not only an agent used to treat Alzheimer's disease but is also being investigated as a potential treatment for BD mania (Zdanys and Tampi, 2008; Agarwal and Tripathi, 2009; Koukopoulos et al. 2010), despite a lack of preclinical testing with putative animal models of mania. Preliminary work suggests the drug may reduce BD manic symptoms (as assessed by YMRS) and improve cognitive function (Teng and Demetrio, 2006; Keck et al., 2009a). Memantine may also prevent amphetamine- or MDMA-induced neurotoxicity or cognitive impairment (Chipana et al., 2008a,b; Camarasa et al., 2010).
Assessing models of mania
It is evident that until recently, most clinical trials investigating anti-manic treatments have focused on reducing scores on BPRS and YMRS scales in patients. Widespread use of these measures has limited opportunities for assessing these agents in tasks with cross-species translational validity (Young et al., 2007). As discussed in the following sections, hyperactivity is frequently used as the primary outcome to assess the validity of many animal models of mania, while objective quantification of motor activity in clinical studies is rare. Recognition of the multi-faceted nature of mania has stimulated research in other domains (Gould and Einat 2007; Young et al., 2007), but the validation of models in these domains remain limited to date.
Pharmacological models of mania
Animal models of BD mania began with observational evidence. Psychostimulants (such as amphetamine) can produce mania-like symptoms in normal healthy subjects (Meyendorff et al., 1985; Peet and Peters, 1995; Hasler et al., 2006) and exacerbate symptoms or induce a manic episode in patients. Thus, the effects of pharmacological agents (stimulants) on behaviour have been widely used as an animal model of mania (Table 2; (Frey et al., 2006; Kato et al., 2007). Because psychomotor agitation is commonly observed during mania and locomotor activity is easily measured in rodents, activity level is most often the behavioural measure used in animal models of mania. Amphetamine-induced hyperactivity is considered to be the ‘gold-standard’ rodent model of mania (Davies et al., 1974; Berggren et al., 1978; Gould et al., 2001), despite detailed inspection of amphetamine-induced alterations of behaviour in humans revealing differences in behaviours (Silverstone et al., 1998). Amphetamine-induced hyperactivity has been reversed by acute lithium in some mouse strains but not others (Davies et al., 1974; Gould et al., 2001), although this effect is often confounded by lithium-induced reductions in activity alone (Berggren et al., 1978), especially when studies are done in habituated animals during their sleep cycle. Chronic lithium does not appear to reverse amphetamine-induced hyperactivity (Fessler et al., 1982; Cappeliez and Moore, 1990). The use of amphetamine as a model of BD mania does have a number of limitations, including the following: (i) amphetamine hyperactivity has been interpreted as a model for a number of distinct disorders besides BD (including schizophrenia, drug abuse and tardive dyskinesia); (ii) mania is characterized by a broad set of symptoms that may not always include motor hyperactivity; (iii) most models utilize acute doses, while mania is a chronic disease with long-term alterations in behaviour.
Table 2.
Pharmacological animal models of mania
| Manipulation | Spp. | Treatment | Outcome | Strengths | Weaknesses | Reference |
|---|---|---|---|---|---|---|
| Acute AMP-induced ↑ in activity | Mice | Acute lithium (150 and 300 mg·kg−1) | Attenuated AMP-induced hyperactivity | Approved mania treatment attenuated manipulation effects | Acute manipulation and treatments, and lithium decreased activity alone – additive effects? | Berggren et al. (1978) |
| Acute AMP + CDP–induced ↑ in head dips | Mice | Acute lithium (3 mEq·kg−1) | Reversed the AMP + CDP–induced ↑ in head dips | Approved mania treatment reversed manipulation effects | Acute manipulation & treatments. Full controls not assessed (see text) | Davies et al., (1974) |
| Acute AMP-induced ↑ in activity | Mice | Acute lithium (4 mEq·kg−1) | Attenuated AMP-induced hyperactivity in C57 but not C3H mice | Approved mania treatment attenuated manipulation effects, genetic differences between strains may yield interesting evidence for lithium effects | Acute manipulation and treatments, and lithium decreased activity alone – additive effects? | (Gould et al., 2001) |
| AMP-induced stereotypy | Rats | Chronic lithium mash (2.3 g·kg−1) for 14 days | No effect of AMP-induced stereotypy | Used chronic treatment of lithium, reduced weight of treated rats indicative of biological effect of treatment | No effect of treatment alone or on manipulation | (Fessler et al., 1982) |
| Chronic AMP-induced hyperactivity | Rats | Chronic lithium injections (1.5 mEq·kg−1) for 21 days | No effect on AMP-induced hyperactivity | Used chronic treatment of lithium, reduced weight of treated rats indicative of biological effect of treatment | No effect of treatment on behaviour during or after removal | Cappeliez and Moore, 1990 |
| Acute ouabain i.c.v.-induced hyperactivity | Rats | Chronic lithium 7 days (2.5 mEq·kg−1) | Lithium reversed ouabain-induced hyperactivity | Chronic approved mania treatment attenuated manipulation effects | Animals were habituated to environment so minimum activity levels may have masked lithium-induced decreases in activity | Li et al., 1997 |
| Acute quinpirole-induced hyperactivity | Rats | Chronic valproate (12 g·L−1), phenytoin (6 g·kg−1), carbemazepine (8 g·kg−1), & topiramate (30 mg·kg−1), each for 11 days | Quinpirole-induced hyperactivity was only attenuated by topiramate treatment | Chronic treatment examined | Approved mania treatments ineffective in paradigm, while agents not approved were effective | Shaldubina et al., 2002 |
| Acute AMP + CDP–induced hyperactivity | Rats and mice | Acute lithium, retigabine, lamotrigine and levetiracetam | All drugs were efficacious in reversing AMP + CDP effects | Approved mania treatment attenuated manipulation effects | Acute dosing only. Drugs not approved for mania were efficacious (lamotrigine and levetiracetam) | Dencker et al., 2008; Lamberty et al., 2001; Redrobe and Nielsen, 2009 |
| Repeated ouabain i.c.v. induced hyperactivity, measured at 0, 6 and 7 days post injection | Rats | Chronic olanzipine 7 days twice daily (1 or 6 mg·kg−1) or haloperidol twice at 21 mg·kg−1 last 7 days prior to testing | Haloperidol attenuated ouabain-induced hyperactivity, no effect of olanzapine | Approved mania treatment attenuated manipulation effects | Haloperidol decreased effects alone – additive effects? No differentiation between mania and schizophrenia, repeated testing may attenuate evidence of drug alone-induced reduction in activity | El-Mallakh et al., 2006 |
AMP = amphetamine, CDP = chlordiazepoxide
The adenosine triphosphotase inhibitor ouabain induced hyperactivity in the open field has also been proposed as a model of BD mania (el-Mallakh et al., 1995). The predictive validity for ouabain-induced increased activity has been investigated using antipsychotic medication approved for the treatment of BD mania. The typical antipsychotic haloperidol decreased ouabain-induced hyperactivity in rats, an effect confounded by haloperidol-induced reduction in activity alone (El-Mallakh et al., 2006). The atypical antipsychotic olanzapine did not significantly reduce activity alone, nor did it attenuate ouabain-induced hyperactivity (El-Mallakh et al., 2006). The effects of ouabain may be related to alterations in the phosphorylation of mitogen-activated protein kinase kinase1/2–ERK 1/2 pathway (Kim et al., 2008).
To assess the generalizability of ouabain-induced effects on activity, one study compared behaviour in an open field (86 × 86 cm and well lit) and an activity chamber (41.5 × 41.5 cm and dark) (Decker et al., 2000); the results indicated that ouabain increased activity only in the open-field test. Thus, the environment appears to be important for observing behavioural changes associated with mania, even in ‘simple’ hyperactivity. Environmental factors are also important in BD mania because complex environments can increase manic symptoms, while ‘quiet rooms’ can reduce symptoms (Fisher et al., 1991). Environmental novelty may disrupt a subject's circadian rhythm, resulting in manic episodes in predisposed subjects (Ehlers et al., 1988; Malkoff-Schwartz et al., 1998; 2000), while events that disrupt social rhythm can predict episodes of hypomania (Sylvia et al., 2009). Maintaining environmental consistencies and managing rhythm dysregulation may buffer against future manic episodes (Frank et al., 2005).
Environmental novelty can therefore be a critical factor when interpreting the effects of drugs on activity levels alone. For example, for a drug to treat symptoms, it should reverse a psychostimulant-induced effect (e.g. normalization of activity) rather than simply exerting an additive effect (e.g. reducing activity irrespective of prior treatment). The only way to assess whether a drug effect represents a reversal or additive finding is to include a drug-only group as a control (Figure 2). For example, haloperidol (El-Mallakh et al., 2006), valproate (Dencker and Husum, 2010), asenapine, olanzapine (Marston et al., 2009) and carbamazepine (Arban et al., 2005) all reduce activity alone, confounding the interpretation of their antagonism of amphetamine-induced hyperactivity. A second issue to consider is the degree of habituation to the testing chamber. Repeated exposure to an open-field chamber may induce a ‘floor effect’, where activity levels are so low that a drug effect alone could not lower them further. This condition may confuse data interpretation, suggesting a reversal effect when identification of additive effects is not possible. For example, Li et al. (1997) assessed the activity of rats for several days prior to quantifying ouabain-induced hyperactivity and acute lithium ‘reversal’ of effects. In this study, lithium failed to decrease activity levels that were already minimal due to environmental familiarity.
Figure 2.

Hypothetical results of a putative treatment study in a pharmacological animal model of mania. The dependent measure of mania-like activity is on the y-axis and the seven treatment group by two pre-treatment groups are depicted on the x-axis. Veh = vehicle, Lam = lamotrigine (used here as a negative control), Li = lithium, Val = valproate (both used here as positive controls), Ds1 = dose 1, Ds2 = dose 2, Ds3 = dose 3 of the drug studied. Each of the treatment groups in the vehicle pretreatment arm would not affect ‘mania’-like behaviour. The ‘mania’-inducing pretreatment would increase the ‘mania’ symptoms of each treatment group in comparison with the corresponding treatment group in the vehicle pretreatment arm. Given that lamotrigine does not treat bipolar mania while lithium and valproate do, one would expect no improvement in mania with lamotrigine, but improvements are observed for lithium and valproate in the mania model. For the test therapeutic, a dose–response effect would be observed. Ideally, treatment effects would be assessed after a chronic 4 week plus treatment duration.
Antipsychotic drugs developed for the treatment of schizophrenia are typically assessed in preclinical studies that quantify their ability to reduce amphetamine-induced hyperactivity or stimulant-induced disruption in PPI, two paradigms also used to assess the effects of anti-manic drugs. Using a dopamine D2 receptor antagonist (the primary mechanism of action of antipsychotics is binding to the D2 receptor) to inhibit or block the effects of a dopamine agonist-induced change in behaviour has been referred to as receptor tautology (Geyer, 2006a). Although utilizing the same behaviour and stimulants that are used to model mania, these models appear primarily useful at identifying efficacious antipsychotics. The use of this model for developing antipsychotic treatment in schizophrenia has been covered elsewhere (Geyer and Moghaddam, 2002). A recent example, however, is the new atypical antipsychotic asenapine, initially approved for schizophrenia and now also approved for treating mania in BD (Minassian and Young, 2010). Early studies indicated that asenapine reversed the dopaminergic-induced hyperactivity model of schizophrenia, consistent with other antipsychotics (Ellenbroek, 1993; Moore et al., 1997; Geyer and Ellenbroek, 2003; Sun et al., 2009). The effects of asenapine alone were not presented (Costall et al., 1990); thus, it was unclear whether the asenapine reversal of dopamine-induced hyperactivity was simply due to a non-specific reduction in activity. Recently, it was demonstrated that systemic administration of asenapine did reduce spontaneous activity alone (Marston et al., 2009) – consistent with other antipsychotics. For example, the pattern of reduced activity in normal animals and amphetamine-induced hyperactivity has also been observed for aripiprazole (Mavrikaki et al., 2010). Asenapine also reversed apomorphine-induced disruption of prepulse inhibition (PPI; (Marston et al., 2009), consistent with other antipsychotics (Swerdlow et al., 1994) and an effect linked to dopamine D2 receptor binding levels. Topiramate, the failed putative anti-manic agent, was also efficacious in apomorphine-induced disruption in PPI (Frau et al., 2007). Ketamine- but not amphetamine-induced reduction in PPI in mice was attenuated by the failed putative anti-manic lamotrigine (Brody et al., 2003), suggesting that ketamine-induced disruption in PPI may not be an appropriate animal model of mania. Acute lithium attenuated amphetamine- but not ketamine-induced disruption in PPI in mice, but the opposite was true for carbemazapine, while valproate reversed neither effect (Ong et al., 2005). Other NMDA antagonist models of mania include phencyclidine-induced stereotypy and hyperactivity, with the former but not the latter being reversed by subchronic lithium treatment (Fessler et al., 1982). Phencyclidine-induced changes in behaviour are used more often as models of schizophrenia, however (Neill et al., 2010), and have not received much investigation in the context of treatments for BD mania.
Acute administration of the dopamine agonist quinpirole induces a pattern of hyperactivity also utilized as an animal model of mania. Shaldubina et al. (2002) observed that chronic administration of the anti-manic drugs valproate and carbamazepine successfully reversed quinpirole activation. The attenuation of quinpirole-induced hyperactivity was also observed however for topiramate (Shaldubina et al., 2002), which is not indicated for mania treatment (Kushner et al., 2006; Vasudev et al., 2006; Correll et al., 2010) and may in fact induce a manic episode in some patients (Jochum et al., 2002). It is relevant to note that topiramate was not assessed in animal models of mania prior to clinical studies (see above). Behavioural sensitization to psychostimulants that induce dopamine release (such as amphetamine) is also used as an animal model of mania (Cappeliez and Moore, 1990). Acute administration of lamotrigine, lithium, valproate, retigabine and carbamazepine all reversed the amphetamine-induced sensitization hyperactivity model of mania (Dencker and Husum, 2010), although valproate alone also reduced activity in vehicle-treated mice. These data suggest that false-positive effects are also observed for psychostimulant sensitization models of mania since lamotrigine has not proven effective to treat acute manic episodes (Bowden, 2002; Singh et al., 2005). In summary, the data from these studies indicate that the efficacy of putative mania treatments in preclinical models is not always maintained in clinical trials.
Another animal model of mania has sought to go beyond the simplicity and putative receptor tautology of single psychostimulant administration. The benzodiazepine derivative chlordiazepoxide, which has anxiolytic and sedative effects alone, has been used in combination with amphetamine as a rodent model of mania (Poitou et al., 1975; Arban et al., 2005). The most common behavioural output measure used is still activity level, as the amphetamine and chlordiazepoxide (AMP + CDP) model increases activity in both rats and mice. Consistent with amphetamine alone, AMP + CDP–induced hyperactivity is reversed by acute lithium, retigabine, lamotrigine and levetiracetam (Lamberty et al., 2001; Dencker et al., 2008; Redrobe and Nielsen, 2009). Similar to amphetamine alone, the predictive validity of the AMP + CDP model is questionable, based on evidence that both lamotrigine and levetiracetam have limited efficacy for treating mania (Goldberg and Burdick, 2002; Grunze et al., 2003; Kruger et al., 2008).
One of the major difficulties of the AMP + CDP model of mania is the number of control groups required for each study (vehicle alone, amphetamine alone, chlordiazepoxide alone, AMP + CDP in combination, AMP + CDP plus each treatment, amphetamine plus each treatment and chlordiazepoxide plus each treatment). Arban et al. (2005) conducted a full control comparison study utilizing the AMP + CPD model in conjunction with several anti-manic agents. Chlordiazepoxide alone often potentiated the activity-suppressant effects of anti-manic agents (Arban et al., 2005), a finding also observed for lamotrigine (Redrobe and Nielsen, 2009). It was unclear whether the combined model provided meaningful data on the action of anti-manic agents or whether alterations in activity simply represent a combination of additive drug effects. This work was supported recently by (Kelly et al., 2009), who describe a more complete dose–response interaction and highlight the difficulties in interpreting putative anti-manic responses based on this mixture of drug effects. Thus, the complexity of controls required in the AMP + CPD model of mania makes it difficult to interpret the responses to novel putative anti-manic agents. For example, Kozikowski et al. (2007) assessed the effects of a newly synthesized GSK-3β inhibitor in the AMP + CPD model of mania, observing that consistent with valproate, the novel inhibitor attenuated but did not reverse AMP + CDP–induced hyperactivity. The findings of the study were limited by several factors, including the use of a high dose of valproate (400 mg·kg−1); in addition, the authors did not assess the effects of valproate treatment alone or in combination with either amphetamine or chlordiazepoxide, nor did they provide statistics on the inhibitor effect compared to a vehicle treatment that itself appeared to reduce activity. In summary, the complexity and false positives observed for the AMP + CDP model of mania may limit its utility in the future.
The predictive validity of many of the models described above may be further limited given that efficacious treatment effects are often observed with acute treatment. Acute administration of anti-manic agents does not treat BD mania; in fact, in randomized controlled trials patients often still exhibit mania rating scores high enough to meet criteria for enrolling in the study even after 3 weeks of treatment (Sachs and Gardner-Schuster, 2007). Thus, it has been suggested that treatment predictive validity should be assessed in chronic not acute studies (Harrison-Read, 2009).
More recently, researchers have highlighted the need to develop animal models of mania that assess more than just levels of activity (Einat, 2006; Young et al., 2007; Einat, 2007b; Geyer, 2008). One technique would be to develop a battery of tests that putatively assess different facets of quantified mania symptoms, such as sleep deprivation, aggression (see below also), strain differences, reward-seeking behaviour or impaired cognition (Einat, 2007b; Geyer, 2008; Flaisher-Grinberg et al., 2009). This premise of using multiple techniques may filter out drugs with false positives in some assays in the hope of identifying effective agents that will treat mania (Einat, 2007b). One must be wary of focusing on ‘aspects of animal behaviour that bear a superficial resemblance to at least one of the clinical features of mania’ (authors' own italics) (Harrison-Read, 2009), or what is known as ‘face validity’. Therefore, another tactic has been to reverse translate the quantification of spontaneous exploration in rodents so that it can also be assessed in humans, thus producing behavioural profiles that can be modelled directly in animals (Young et al., 2007). This latter technique has led to the identification of a unique pattern of exploration in patients with mania that includes increased activity, more specific exploration and straighter line movement from location to location (Minassian et al., 2009; Perry et al., 2009; 2010). This technique also led to the development of novel animal models of mania including dopamine transporter (DAT) knockdown mice (discussed in greater detail below) and the effects of the selective DAT inhibitor GBR 12909 on spontaneous exploration. This methodology has also provided quantifiable evidence (Perry et al., 2009; Young et al., 2010a,b) to support self-report data in humans (Silverstone et al., 1998), questioning the validity of acute amphetamine as a model for BD mania. Extensive tests have yet to be completed on the predictive validity of these reverse-translational models, while assessment of cognitive performance should be carefully designed to account for potential impairment induced by anti-manic medications (Goldberg and Burdick, 2001; Gualtieri and Johnson, 2006).
Environmental models of BD mania
In contrast to pharmacological models of mania, some groups have attempted to use environmental manipulations to induce behaviour in animals that resembles human symptoms associated with the disorder. These environmental model challenges often use stimuli that can provoke a manic episode (e.g. sleep deprivation, stress – see below). Despite the use of such challenges, the animals being investigated in these models are not perturbed in any way, which equates to mania (non-mania prone subjects do not become manic under similar environmental challenges). Treatments identified using these models may not therefore be specific to mania but represent symptomatic treatment only (Table 3).
Table 3.
Environmental models of mania
| Manipulation | Spp. | Treatment | Outcome | Strengths | Weaknesses | Reference |
|---|---|---|---|---|---|---|
| Sleep deprivation (72 h) | Rats | Subchronic lithium (60 mmol·kg−1) 10 days | ↓ sleep latency and hyperactivity | Sleep disruption can induce manic episodes and ↓ sleep seen in mania | Performed in normal animals, other stressors also used may interfere with effects | Gessa et al., 1995 |
| Sleep deprivation (4 h in dark cycle) | Rats | Chronic lithium (2.4 g·kg−1) 4 weeks | ↓ forebrain PKC expression | ↓ PKC implicated in BD disorder affected by lithium and valproate | Performed in normal animals. Behavioural symptoms have not been assessed | Szabo et al., 2009 |
| Resident/intruder | Mice | Chronic lithium (2.4 g·kg−1) or valproate (20 g·kg−1) 4 weeks | ↓ resident aggressive attacks with lithium and valproate | Approved mania treatments ↓ mania-like behaviour (aggression) | Performed in normal animals, assay sensitive to non-mania treatments, antidepressants, anxiolytics etc | Einat, 2007a,b; |
| Food competition | Rats | Chronic daily lithium (100 mg·kg−1), carbemazepine (20 mg·kg−1) or valproate (30 mg·kg−1) 7 weeks | ↓ dominant behaviours | Approved mania treatments ↓ dominant-like behaviour, observed only with chronic treatment | Performed in normal animals, assay sensitive to non-mania treatments, antidepressants, anxiolytics etc, multiple injections problematic | Malatynska and Knapp, 2005; Malatynska et al., 2007 |
| Footshock stress | Rats | Subchronic lithium (20 mEq·L−1) 14 days | Lithium ↓ shock-induced aggression | Approved mania treatments ↓ aggression | Performed in normal animals, assay sensitive to non-mania treatments, antidepressants, psychstimulants etc, | Prasad and Sheard, 1982 |
PKC = protein kinase C
The rodent sleep deprivation model purports to examine a cluster of behavioural markers germane to manic episodes and also exhibits significant clinical relevance, because sleep disruption constitutes one of the most frequent prodromes of mania (Gessa et al., 1995; Mansell and Pedley, 2008). The paradigm involves placing a rat or mouse on a small platform (3–7 cm) surrounded by water for an extended period, typically 72 h (Gessa et al., 1995). Muscle relaxation associated with sleep results in the animal falling into the water, thereby waking the animal. When rodents are placed back into their home cage after the sleep deprivation period, they exhibit a series of mania-like behaviours that include insomnia, hyperactivity, aggressive actions towards other animals, hypersexuality (an increase in mounting behaviour) and stereotypy (sniffing and rearing) (Morden et al., 1967; Hicks et al., 1979; Fratta et al., 1987). This pattern of over-activity is maximal during a 30–40 min period immediately after the sleep deprivation treatment (Einat, 2007a). Gessa et al. (1995) reported that 10 days of lithium treatment (60 mmol·kg−1 of lithium salt added to food) significantly reduced motor hyperactivity and decreased the latency to fall asleep compared with vehicle in 72 h sleep-deprived rats (Gessa et al., 1995). Rats administered four injections of haloperidol in the last hour of the sleep deprivation period also demonstrated a decreased latency to fall asleep relative to saline-treated animals, which was interpreted as a reduction in mania-like insomnia. These data support the contention that this model has pharmacological predictive validity, but it is relevant to note that the paradigm includes several stressors aside from simple sleep deprivation, including isolation, immobilization and the experience of falling into water. A more recent study administered intervals of 24 h sleep deprivation to CD1 mice while also including a stress control group with a larger stable platform enabling sleep, but enforcing isolation and immobilization (Benedetti et al., 2008); only the sleep-deprived mice exhibited increased locomotion and aggressive behaviour relative to control, indicating that lack of sleep is key to the mania phenotype observed.
One report examined the effect of lithium and sleep deprivation on brain PKC activity, a pathway implicated in the motoric hyperactivity, risking taking and excessive hedonic drive associated with mania (Szabo et al., 2009). Male Wistar-Kyoto rats subjected to a relatively short period of sleep deprivation (4 h during the light cycle) exhibited evidence of increased PKC signalling as assessed by elevated PKC phosphorylation of AMPA receptors, neurogranin and myristoylated alanine-rich C kinase substrate (MARCKS) in frontal cortex relative to rested animals. In contrast, C57BL/6 mice treated with lithium in food for 4 weeks showed significantly reduced phosphorylation of these PKC molecular targets compared with animals fed a drug-free diet. While behavioural symptoms such as hyperactivity or aggression were not assessed in this paper, the data indicate that a sleep deprivation paradigm may have construct validity in representing the neurobiological alterations that may mediate manic episodes.
Manic individuals are frequently characterized by provocative, intrusive or aggressive actions, behaviours that are frequently modelled in animals using variants of the resident–intruder test (Einat, 2007a). This task typically consists of introducing a group-housed intruder rodent into the home cage of an isolated mouse or rat and quantifying the aggressive acts committed by the resident and the defensive acts and postures exhibited by the intruder (Miczek et al., 2001). Resident aggressive behaviours, including biting, threatening postures and tail rattling, can be augmented by prolonged isolation or exposure to acute stressors such as foot shock (Legrand and Fielder, 1973; Miczek and O'Donnell, 1978). While the resident–intruder test has been utilized as a model of depression by assessing social defeat characteristics demonstrated by the intruder, treatment with anti-manic medications such as lithium has also been demonstrated to reduce resident aggression in both rats and mice (O'Donnell and Gould, 2007). Lithium administration has consistently reduced aggression induced by mild electric foot shock in rats, as well as attenuate combative behaviour augmented by stimulants such as d-amphetamine (Eichelman et al., 1973; Mukherjee and Pradhan, 1976; Prasad and Sheard, 1982). The drug also diminishes conspecific isolation-induced fighting outside the home cage, as well as resident–intruder fighting within the home cage (Brain and Al-Maliki, 1979; Oehler et al., 1985). One recent study examined the effect of both lithium and valproate on aggression using a resident–intruder paradigm in C57BL/6 mice (Einat, 2007b). Resident mice treated with either mood-stabilizer for 4 weeks before the test exhibited a significant reduction in the number of attacks against intruder mice compared with control but did not show any difference in non-aggressive interactions such as rearing or sniffing. Finally, it is also worth noting that chronic treatment with a variety of antidepressant drugs increases aggression in the resident–intruder paradigm (Mitchell, 2005), suggesting that this may constitute a model of antidepressant-induced mania in BD individuals (Koszewska and Rybakowski, 2009).
While the resident–intruder test has been in use for several decades (Miczek et al., 2001), a recent representation of dominant–submissive behaviour has also been proposed to model symptoms of both depression and mania (Malatynska and Knapp, 2005). This paradigm is based on the concept that subordinate animals, similar to depressed humans, show increased defensive behaviour and reduced activity; in contrast, dominant animals, similar to manic states, are characterized by self-confident, assertive and aggressive behaviour (Gardner, 1982). In this paradigm, two rats are placed in opposite chambers connected by a narrow tunnel allowing only one animal at a time access to a food source. When tested over 2 weeks in 5 min daily sessions, approximately half the animal pairs develop a dominant–submissive relationship, where one dominant animal monopolizes access to the food, defined as spending 40% more time at the feeder compared with the submissive rodent. When treated with antidepressant medications such as imipramine, desipramine and fluoxetine (daily dose of 10 mg·kg−1 i.p.), submissive rats become more assertive, resulting in a significantly greater access to the food after 2 to 4 weeks of treatment. Conversely, dominant rats injected with lithium (100 mg·kg−1), carbamazepine (20 mg·kg−1) or sodium valproate (30 mg·kg−1) become less aggressive and lose their dominant status (Malatynska and Knapp, 2005; Malatynska et al., 2007). In addition, the mood stabilizers exert an effect over a time course relatively similar to that observed in BD patients; lithium and carbamazepine treatment reduced rodent dominance only after 2–3 weeks of treatment. It is relevant to note, however, that resident–intruder paradigms are also sensitive to the effects of anxiolytic and anxiogenic drugs, suggesting that behavioural changes in this model could be interpreted as representing different behavioural constructs such as anxiety, limiting the predictive validity of this model for mania (Vassout, et al., 2000).
Genetic models
Genetic models of mania can assume any of several forms. One technique is to compare strains where the behaviour of one strain is akin to that of other models of mania, or is more susceptible to treatment-induced alterations in behaviour. Another technique is to examine the behaviour of mice with genetic mutations in genes that have been linked to BD mania. Finally, a third technique is the creation of mice with specific genetic alterations designed to recreate those observed in BD mania. Gould and Einat (2007) emphasize that greater understanding of the genes that contribute to BD mania will assist in the identification of endophenotypes of the disorder and increase the utility of mutant mice as useful models. Malkesman et al. (2009) describes the creation of genetic mouse models of BD that may provide greater etiological validity compared with models based on behavioural abnormalities observed in BD mania (for the predictive validity of such models, see Table 4).
Table 4.
Genetic models of mania
| Manipulation | Spp. | Treatment | Outcome | Strengths | Weaknesses | Reference |
|---|---|---|---|---|---|---|
| Assessing sweet preference of four strains | Mice | Subchronic lithium (200 mg·kg−1) or valproate (200 mg·kg−1) for 2–4 days | Both treatments reduced sweet preference of Black Swiss mice but not other strains | Approved mania treatment attenuated manipulation effects, genetic differences between strains may yield interesting evidence for lithium effects | Effects are on normal behaviour, thus additive and not specific to mania. Not all strains exhibited a sweet preference | Flaisher-Grinberg et al., 2009 |
| Reduced PPI of DBA vs. C57 mice | Mice | Acute lithium (30 mg·kg−1), carbemazepine (30–100 mg·kg−1), topiramate (100 and 300 mg·kg−1), valproate (178 and 316 mg·kg−1) and lamotrigine (3–30 mg·kg−1) | All treatments ↑ PPI in DBA mice | Approved mania treatments ↑ PPI except lithium | Effects are on normal behaviour, thus additive and not specific to mania. False positives of topiramate and lamotrigine limit the predictive validity of effect | (Flood et al., 2009 |
| Glutamate receptor 6 knockout mice hyperactivity, aggression and low anxiety | Mice | Chronic lithium (2.4 g·kg−1) | Lithium reversed the abnormal behaviour of the mice | Approved mania treatment reversed the mania-like behaviour without the need to antagonize an acute drug effect | Modest effects of lithium on wild-type mice, common polydipsea side effect not discussed, mania patients not null for glutamate receptor 6 | Shaltiel et al., 2008 |
| Hyperactivity in dopamine transporter knockdown mice | Mice | Acute valproate (200 mg·kg−1) | Lithium attenuated the hyperactivity of the mice | Approved mania treatment reversed the mania-like behaviour without the need to antagonize an acute drug effect, links of reduced DAT expression in mania patients | Mania-like behaviour reversed via acute not chronic treatment | Ralph-Williams et al., 2003 |
| D-box binding protein knockout mouse stress-induced hyperactivity | Mice | Subchronic lithium (0.6 g·L−1) for 10 days | Lithium attenuated some mania-like behaviours | Approved mania treatment reversed the mania-like behaviour without the need to antagonize an acute drug effect | Mania-like behaviour only induced via long-term stressors including isolation rearing | Roybal et al., 2007 |
Flaisher-Grinberg et al. (2009) compared the sweet solution preference of four mouse strains (Black Swiss, C57BL/6, CBA/J and AJ) as a putative method to model reward-seeking behaviour in mania. It was observed that Black Swiss mice exhibited the greatest preference for sweet solution, which was antagonized by twice daily injections of valproate (200 mg·kg−1) or lithium (200 mg·kg−1), but not the antidepressant imipramine (Flaisher-Grinberg et al., 2009). Further investigations with this model are warranted, especially because it is not clear why other strains did not exhibit a strong preference for sweet solutions, as well as the possibly confounding effect of polydipsia induced by these treatments. The Brattleboro rat, characterized by an inability to synthesize vasopressin, has been used commonly as an animal model of schizophrenia because they exhibit reduced PPI when compared with Long Evans rats (Feifel et al., 2004; Cilia et al., 2010). Because deficient PPI is observed in BD mania as well as in schizophrenia (Braff et al., 2001; Perry et al., 2001), this animal model may have relevance to mania. Treatments used for both mania and schizophrenia are effective in this model. Specifically, acute or subchronic clozapine, risperidone or haloperidol improved PPI in Brattleboro rats to levels comparable with Long Evans rats (Feifel et al., 2004; Feifel et al., 2007; Cilia et al., 2010). Brattleboro rats also exhibit increased activity in response to a novel test environment (Cilia et al., 2010). The effect of anti-manic medications has not yet been assessed in these animals, but such studies would prove useful to determine the specificity of this model for identifying novel treatments for the disorder. In mice exhibiting lower PPI than other strains (DBA mice), acute administration of lithium (30 mg·kg−1), carbamazepine (30–100 mg·kg−1), topiramate (100 and 300 mg·kg−1), valproate (178 and 316 mg·kg−1) and lamotrigine (3–30 mg·kg−1) increased PPI (Flood et al., 2009). These results demonstrate that PPI may be affected non-selectively by drugs both efficacious and ineffective in treating BD mania, limiting the specific applicability of this measure. Therefore, although using strain differences to investigate putative treatments for BD mania may prove useful, it is likely that any drug developed will not only specifically ‘treat’ the underlying causes but also exhibit subtractive effects on behaviour that would be observed in non-BD as well as BD patients (Harrison-Read, 2009). Models developed specifically to mimic neurobiological abnormalities observed in patients with BD mania are consequently more likely to enable BD mania-specific treatments.
There are several examples of genetically manipulated mice that could serve as models of BD mania, characterized by links to potential underlying mechanisms of the illness. Examples include genes that have been associated with BD, such as CLOCK, DAT, Glutamate 6 receptor (GluR6) and DISC1. GluR6 knockout (KO) mice exhibit hyperactivity, aggression and reduced anxiety, which were reversed with chronic lithium treatment (4 week 2.4 g·kg−1 chow), with modest effects in wild-type (WT) mice (Shaltiel et al., 2008). Given that lithium was so effective in these mice, but fails to effectively treat all patients with mania, other treatments should also be assessed, including antipsychotics, as well as failed therapeutics such as lamotrigine. The behaviours described in GluR6 KO mice differed from WT littermates, but the similarity to behaviours in BD mania is largely superficial.
As discussed above, one technique to generate models of mania based on the quantification of behaviour in patients with BD mania was to create a human exploratory paradigm that mimics a rodent exploratory paradigm, the behavioural pattern monitor (BPM) (Geyer et al., 1986; Young et al., 2007). This multivariate approach to assessing spontaneous exploration in the human BPM has resulted in the identification of a unique exploratory profile of patients with BD (increased motor activity, exaggerated specific exploration and more linear patterns of movement) that differed from control subjects, patients with schizophrenia and adult ADHD subjects (Paulus et al., 2007; Perry et al., 2009).
The BPM has been instrumental in the development of DAT KD mice as a model of BD mania. The DAT has been linked to BD mania across several genetic linkage studies (Kelsoe et al., 1996; Greenwood et al., 2001; Greenwood et al., 2006), with functional consequences of DAT mutation leading to reduced cell surface expression of the transporter (Horschitz et al., 2005). These findings support observations of lower levels of DAT reported in BD patients (Amsterdam and Newberg, 2007). DAT KO mice were reported as a putative model of mania because these mice exhibit increased motor activity (Giros et al., 1996) and impaired PPI (Ralph et al., 2001). Moreover, PPI deficits in these mice were reversed with acute treatment with the atypical antipsychotics clozapine and olanzapine (Powell et al., 2008). Given that the poor physical condition of these mice limits their usefulness in assessing novel therapeutics, a line of DAT KD mice was subsequently developed (Zhuang et al., 2001). Because DAT KD mice exhibit only 10% DAT compared with WT littermates, their spontaneous exploration was assessed in the mouse BPM to assess their possible utility as a model of mania. DAT KD mice exhibited a profile that is (i) consistent with that of patients with BD mania (Ralph-Williams et al., 2003; Perry et al., 2009); (ii) attenuated with environmental familiarity (Young et al., 2010c); (iii) reinstated with environmental novelty or stimulants at doses that do not affect WT littermates (Young et al., 2010c); and (iv) partially attenuated by chronic valproate treatment (Young et al., unpubl. obs.). Interestingly, in vitro studies suggest that valproate treatment may increase DAT expression (Wang et al., 2007), thus providing a putative mechanism for the valproate-induced treatment effects observed in these mice. Increased activity and straighter linear movement through the environment (as measured by reduced spatial d, also observed in patients with mania) are also observed in these mice when assessed in a smaller open-field test, both of which were attenuated by acute valproate treatment (200 mg·kg−1) (Ralph-Williams et al., 2003). DAT KD mice also exhibit hedonia-like behaviour in terms of food motivation behaviour (Cagniard et al., 2006), as well as increased risk preference in a mouse version of the IGT (Young et al., 2011). DAT KD mice therefore mimic several features that are observed in BD mania in terms of behaviour, cognition and putative neurobiological abnormalities.
Another gene putatively, although not consistently, linked to BD is GSK-3. Interest in this gene is motivated by the possibility that lithium and valproate may exert their effects via the pathway in which this gene acts. Heterozygous GSK-3 mutant mice have been created and exhibit reduced hole-poking behaviour and increased immobility in the forced swim test as well as other changes that resemble lithium-treated mice (O'Brien et al., 2004). Overexpression of GSK-3β in mice results in hyperactivity and reduced habituation, which may mimic some of the features of mania (Prickaerts et al., 2006). Therefore, GSK-3 mice may be useful as a screen for lithium-like compounds. Moreover, there appears to be greater evidence linking GSK-3 inhibition with antidepressant-like activity than with mania-reducing effects.
Finally, there are several circadian gene mutants that may prove to be useful animal models of mania. The circadian gene d-box binding protein (DBP) was identified as a putative candidate gene for BD (Le-Niculescu et al., 2008). DBP KO mice exhibit lower activity and a blunted response to stimulants, but after a chronic stress paradigm, these mice become hyperactive and sensitive to stimulant treatment, with sleep deprivation–induced hyperactivity that was attenuated by acute valproate (200 mg·kg−1) treatment (Le-Niculescu et al., 2008). The hyperactivity exhibited by these DBP KO mice only occurred after exposure to long-term stressors such as isolation housing, chronic unpredictable stress and foot shock testing over a 1 month period. If this stress manipulation is required in every study, these mice may not prove the most desirable as an animal model for assessing novel therapeutics. Polymorphisms in BMAL1, a binding partner of CLOCK, have been observed in BD patients, and mice with a mutation of the CLOCK gene were created (Roybal et al., 2007). These CLOCK mutant mice exhibited mania-like behaviours including reduced sleep, hyperactivity that wanes with time but is reinstated with novelty (consistent with DAT KD mice above), increased reward sensitivity to cocaine as well as sucrose preference, increased dopaminergic activity and subchronic lithium-induced (600 mg·L−1 for 10 days) reversal of some behaviours (McClung et al., 2005; Roybal et al., 2007). More studies are required for these CLOCK-related mutants, not only in other behaviours but also in investigating additional proven and ineffective treatments (see Figure 3).
Figure 3.

Hypothetical results of a putative treatment study in a mutant animal model of mania. The dependent measure of mania-like activity is on the y-axis, and the treatment groups are depicted on the x-axis. WT = wild-type littermate mice, Mt = genetically modified mouse model of mania. Veh = vehicle, Lam = lamotrigine (used here as a negative control), Li = lithium, Val = valproate (both used here as positive controls), Ds1 = dose 1, Ds2 = dose 2, Ds3 = dose 3 of the drug studied. The Mt mice would exhibit more ‘mania’ symptoms in comparison with WT mice. Given that lamotrigine does not treat Bipolar mania while lithium and valproate do, one would expect no improvement in mania with lamotrigine in either group, but improvements/normalization are observed for lithium and valproate treated mutant mice. For the test therapeutic, a dose–response effect would be observed in treating Mt mice. Ideally all treatments would be assessed using a chronic 4 week plus study.
Summary of findings
While manic states may include a diverse variety of multifacted symptoms that include euphoria, irritation, grandiosity and increased goal-directed activity, it is clear that the majority of the animal models of BD mania rely predominantly on motor activity levels as their primary behavioural outcome measure. Many factors can influence the effect of the inducing agent and/or treatment in this univariate approach to modelling BD mania, including the size of the chamber used, whether it is an open field or activity chamber, assessed in the dark or light and the familiarity of the testing environment. Moreover, treatment attenuation of stimulant-induced hyperactivity can be confounded by the selection of a hypoactive strain of animals, even in the absence of treatment effects on stimulant-naive subjects. These factors may contribute toward the large variety of effects of anti-manic agents and treatment regimens on these models. Acute stimulant-induced effects on behaviour reversed by drugs that act on the same receptors (e.g. amphetamine, an indirect dopamine agonist, and antipsychotics, dopamine D2 receptor antagonists) may be confounded by receptor tautological effects. Unfortunately, the combination of amphetamine and chlordiazepoxide produces so many possible interactions that the data produced can be unreliable unless all possibilities are addressed. The false-positive data generated by drugs that are effective in these models as well as in the ouabain-induced hyperactivity model, but not in BD mania, further highlight the limitation of these approaches for generating novel anti-manic treatments. Finally, many studies listed here examined the effects of acute administration of an ‘inducing’ agent as well as acute treatment-induced reversal of effects. While these studies are useful in behavioural pharmacological terms, mania is a long-term disease in which chronic treatment is required to alleviate symptoms. Studies that utilize chronic treatment are therefore more likely to exhibit predictive validity for treatment in patients. Care must be taken in chronic studies however, because the side effects of chronic lithium treatment in rodents may interact with mania-like behaviour in animals, limiting the predictive validity of the study.
Neither environmental models nor genetic models are limited by receptor tautological or by drug interaction confounds. Environmental models involve using manipulations such as sleep deprivation, competition or stressors to induce behavioural changes in an otherwise normal animal to recreate behaviours that may have relevance to BD mania. These models may mimic aspects of the disorder but care must be taken when assessing treatments in these models given their use in other diseases such as sleep disturbance and anxiety. Given that animals tested in these paradigms may not necessarily have links to the aetiology of BD mania, any treatments developed may have limited efficacy for specifically treating this disorder.
Genetic models appear under several guises, including assessing strain differences on behaviour. The concern arising from environmental models can also be levelled at strain difference models, because any treatment developed in such normal animals may not have specificity to BD mania. Genetic models that are created with reference to the aetiology of BD mania may provide greater validity for treatment in acute mania if that model is based on abnormalities observed in patients. Such investigations may offer the opportunity to develop pharmacogenomic treatments in psychiatry as has occurred for other disease populations (Moller and Rujescu, 2010; Serretti and Drago, 2010). There is only a limited number of treatment predictive validity studies performed on genetic models to date; thus, more are required to validate these models across treatments for a variety of psychiatric disorders, including mania and schizophrenia. Evaluation of a diverse group of drugs, including both efficacious medications and agents proven clinically ineffective, will enable improved development and validity of high-utility preclinical models.
Future directions
There are multiple treatments approved for BD mania, including lithium, valproate and carbamazepine, as well as several antipsychotics. A number of potential treatments have also failed to gain approval, including lamotrigine and gabapentin. Using these drugs as positive and negative controls would enable the pharmacological predictive validation of animal models of BD mania. One problem is the difficulty in identifying drugs that do not attain approval for the treatment of BD mania for further comparisons. Moreover, the lack of details in these failed studies also impedes interpretation of the effects of these drugs on animal models of BD and constrains future directions of research. Thus, greater detail should be provided from clinical studies to delineate specific aspects of mania impacted by novel agents under investigation.
The majority of approaches employed in modelling BD mania have also been limited in focusing on simple behaviours that may have only limited validity with respect to the self-report and observer-rated human behaviours they are supposed to model (Harrison-Read, 2009) (e.g. activity levels and psychomotor agitation, resident–intruder and aggression). Given the wide range of psychiatric conditions these paradigms are designed to model, their selectivity for identifying treatments for BD mania may be limited.
BD mania is characterized by extensive behavioural and cognitive dysfunction that could be modelled with a variety of preclinical paradigms (Einat, 2007b). Some abnormalities during manic episodes that can be detrimental to their overall functioning include attentional deficits, disinhibited responses and increased risk taking. These behaviours can be quantified in laboratory settings and assessed in rodents using cross-species tasks with translational validity, including measures of vigilance (Young et al., 2009a,b), stop-signal reaction time (Eagle and Baunez, 2010) and delay-discounting (Reynolds, 2006; Madden et al., 2007). Moreover, attempts have been made to objectively quantify the hyperactivity and goal-directed activity in these patients (Young et al., 2007; Minassian et al., 2009; Perry et al., 2009; 2010), resulting in novel animal models that may mimic some of the underlying behavioural abnormalities observed in BD mania (Perry et al., 2009; Young et al., 2010a,b). Combining the approaches of (i) using a comprehensive battery of tests for different aspects of mania (Einat, 2007b), (ii) utilizing genetic information on developing models with greater etiological validity (Gould and Einat 2007; Malkesman et al., 2009) and (iii) objective quantification of patient behaviour with cross-species paradigms (Young et al., 2007) will enable the development of models producing drug treatments that are specialized for treating BD mania. The validity of these models can be effectively assessed by testing a diverse group of drugs, including treatments approved for BD mania as positive controls, as well as using clinically ineffective agents as negative controls.
Acknowledgments
We thank Drs William Perry and Arpi Minassian for their support. This study was supported by NIH grants R01-DA002925, R21-MH085221, R01-MH071916 and F32-DA024524 as well as by the Veteran's Administration VISN 22 Mental Illness Research, Education, and Clinical Center.
Glossary
Abbreviations
- ADHD
attention deficit hyperactivity disorder
- AMP
amphetamine
- BPRS
Brief Psychiatric Rating Scale
- cAMP
cyclic adenosine monophosphate
- CDP
chlordiazepoxide
- CGI
Clinical Global Impression
- CPT
Continuous Performance Task
- CVLT
California Verbal Learning Task
- DAT
dopamine transporter
- DBP
d-box binding protein
- DSM-IV
Diagnostic and Statistical Manual of Mental Disorders IV
- ERK
extracellular-related signal kinase
- FDA
Food and Drug Administration
- GABA
gamma-aminobutyric acid
- GluR6
glutamate 6 receptor
- GSK-3
glycogen synthase kinase 3
- IGT
Iowa Gambling Task
- IMPase
inositol monophosphatase
- MARCKS
myristoylated alanine-rich C kinase substrate, MDMA, 3,4-methylenedioxymethamphetamine
- NMDA
N-methyl-D-aspartic acid
- PKC
protein kinase C, PPI, prepulse inhibition
- SADS-C
Schedule for Affective Disorders and Schizophrenia
- YMRS
Young Mania Rating Scale
Conflict of interest
The authors report no conflict of interest with the current manuscript.
References
- Amsterdam JD, Newberg AB. A preliminary study of dopamine transporter binding in bipolar and unipolar depressed patients and healthy controls. Neuropsychobiology. 2007;55:167–170. doi: 10.1159/000106476. [DOI] [PubMed] [Google Scholar]
- APA. Diagnostic and Statistical Manual of Mental Disorders. 4th edn. Washington, DC: American Psychiatric Association; 1994. [Google Scholar]
- APA. Practice guideline for the treatment of patients with bipolar disorder (revision) Am J Psychiatry. 2002;159:1–50. [PubMed] [Google Scholar]
- Arban R, Maraia G, Brackenborough K, Winyard L, Wilson A, Gerrard P, et al. Evaluation of the effects of lamotrigine, valproate and carbamazepine in a rodent model of mania. Behav Brain Res. 2005;158:123–132. doi: 10.1016/j.bbr.2004.08.015. [DOI] [PubMed] [Google Scholar]
- Agarwal V, Tripathi A. Memantine in the management of a clinically challenging case of bipolar disorder. Indian J Psychiatry. 2009;51:137–138. doi: 10.4103/0019-5545.49455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arora M, Daughton J. Mania in the medically ill. Curr Psychiatry Rep. 2007;9:232–235. doi: 10.1007/s11920-007-0024-8. [DOI] [PubMed] [Google Scholar]
- Atack J. Lithium, phosphatidylinositol signaling and bipolar disorder. In: Manji HK, Bowden CL, Belmaker RH, editors. Bipolar Medications: Mechanisms of Action. Washington DC: American Psychiatric Press; 2000. pp. 1–30. [Google Scholar]
- Baastrup PC, Schou M. Lithium as a prophylactic agents. Its effect against recurrent depressions and manic-depressive psychosis. Arch Gen Psychiatry. 1967;16:162–172. doi: 10.1001/archpsyc.1967.01730200030005. [DOI] [PubMed] [Google Scholar]
- Bauer MS, Crits-Christoph P, Ball WA, Dewees E, McAllister T, Alahi P, et al. Independent assessment of manic and depressive symptoms by self-rating. Scale characteristics and implications for the study of mania. Arch Gen Psychiatry. 1991;48:807–812. doi: 10.1001/archpsyc.1991.01810330031005. [DOI] [PubMed] [Google Scholar]
- Bauer MS, Callahan AM, Jampala C, Petty F, Sajatovic M, Schaefer V, et al. Clinical practice guidelines for bipolar disorder from the Department of Veterans Affairs. J Clin Psychiatry. 1999;60:9–21. doi: 10.4088/jcp.v60n0104. [DOI] [PubMed] [Google Scholar]
- Bebchuk JM, Arfken CL, Dolan-Manji S, Murphy J, Hasanat K, Manji HK. A preliminary investigation of a protein kinase C inhibitor in the treatment of acute mania. Arch Gen Psychiatry. 2000;57:95–97. doi: 10.1001/archpsyc.57.1.95. [DOI] [PubMed] [Google Scholar]
- Benedetti F, Fresi F, Maccioni P, Smeraldi E. Behavioural sensitization to repeated sleep deprivation in a mice model of mania. Behav Brain Res. 2008;187:221–227. doi: 10.1016/j.bbr.2007.09.012. [DOI] [PubMed] [Google Scholar]
- Berggren U, Tallstedt L, Ahlenius S, Engel J. The effect of lithium on amphetamine-induced locomotor stimulation. Psychopharmacology (Berl) 1978;59:41–45. doi: 10.1007/BF00428028. [DOI] [PubMed] [Google Scholar]
- Berrios GE. Depressive and manic states during the nineteenth century. In: Georgotas A, Cancro R, editors. Depression and Mania. New York: Elsevier; 1988. pp. 13–25. [Google Scholar]
- Bora E, Yucel M, Pantelis C. Cognitive endophenotypes of bipolar disorder: a meta-analysis of neuropsychological deficits in euthymic patients and their first-degree relatives. J Affect Disord. 2009;113:1–20. doi: 10.1016/j.jad.2008.06.009. [DOI] [PubMed] [Google Scholar]
- Borkowska A, Rybakowski JK. Neuropsychological frontal lobe tests indicate that bipolar depressed patients are more impaired than unipolar. Bipolar Disord. 2001;3:88–94. doi: 10.1034/j.1399-5618.2001.030207.x. [DOI] [PubMed] [Google Scholar]
- Bourin M, Lambert O, Guitton B. Treatment of acute mania–from clinical trials to recommendations for clinical practice. Hum Psychopharmacol. 2005;20:15–26. doi: 10.1002/hup.657. [DOI] [PubMed] [Google Scholar]
- Bowden CL. Lamotrigine in the treatment of bipolar disorder. Expert Opin Pharmacother. 2002;3:1513–1519. doi: 10.1517/14656566.3.10.1513. [DOI] [PubMed] [Google Scholar]
- Bowden CL, Brugger AM, Swann AC, Calabrese JR, Janicak PG, Petty F, et al. Efficacy of divalproex vs lithium and placebo in the treatment of mania. The Depakote Mania Study Group. JAMA. 1994;271:918–924. [PubMed] [Google Scholar]
- Bowden CL, Mitchell P, Suppes T. Lamotrigine in the treatment of bipolar depression. Eur Neuropsychopharmacol. 1999;9(Suppl. 4):S113–S117. doi: 10.1016/s0924-977x(99)00024-3. [DOI] [PubMed] [Google Scholar]
- Bowden C, Singh V. Valproate: clinical pharmacological profile. In: Akiskal H, Tohen M, editors. Bipolar Psychopharmacology. Chichester: John Wiley & Sons; 2006. pp. 27–42. [Google Scholar]
- Braff D, Stone C, Callaway E, Geyer M, Glick I, Bali L. Prestimulus effects on human startle reflex in normals and schizophrenics. Psychophysiology. 1978;15:339–343. doi: 10.1111/j.1469-8986.1978.tb01390.x. [DOI] [PubMed] [Google Scholar]
- Braff DL, Geyer MA, Swerdlow NR. Human studies of prepulse inhibition of startle: normal subjects, patient groups, and pharmacological studies. Psychopharmacology (Berl) 2001;156:234–258. doi: 10.1007/s002130100810. [DOI] [PubMed] [Google Scholar]
- Brain PF, Al-Maliki S. Effects of lithium chloride injections on rank-related fighting, maternal aggression and locust-killing responses in naive and experienced ‘TO’ strain mice. Pharmacol Biochem Behav. 1979;10:663–669. doi: 10.1016/0091-3057(79)90318-6. [DOI] [PubMed] [Google Scholar]
- Brambilla P, Soares J. The use of anticonvulsants in treatment of bipolar disorder. In: Kasper S, Hirschdelf RM, editors. Handbook of Bipolar Disorder. Boca Raton: Taylor & Francis; 2005. pp. 285–314. [Google Scholar]
- Brody SA, Geyer MA, Large CH. Lamotrigine prevents ketamine but not amphetamine-induced deficits in prepulse inhibition in mice. Psychopharmacology (Berl) 2003;169:240–246. doi: 10.1007/s00213-003-1421-2. [DOI] [PubMed] [Google Scholar]
- Bunney WE, Jr, Goodwin FK, Davis JM, Fawcett JA. A behavioral-biochemical study of lithium treatment. Am J Psychiatry. 1968;125:499–512. doi: 10.1176/ajp.125.4.499. [DOI] [PubMed] [Google Scholar]
- Burdick KE, Braga RJ, Goldberg JF, Malhotra AK. Cognitive dysfunction in bipolar disorder: future place of pharmacotherapy. CNS Drugs. 2007;21:971–981. doi: 10.2165/00023210-200721120-00002. [DOI] [PubMed] [Google Scholar]
- Cade JF. Lithium salts in the treatment of psychotic excitement. Med J Aust. 1949;2:349–352. doi: 10.1080/j.1440-1614.1999.06241.x. [DOI] [PubMed] [Google Scholar]
- Cade J. Lithium-past,present and future. In: Johnson FN, Johnson s, editors. Lithium in Medical Practice. Lancaster: MTP Press; 1978. pp. 1–16. [Google Scholar]
- Cagniard B, Balsam PD, Brunner D, Zhuang X. Mice with chronically elevated dopamine exhibit enhanced motivation, but not learning, for a food reward. Neuropsychopharmacology. 2006;31:1362–1370. doi: 10.1038/sj.npp.1300966. [DOI] [PubMed] [Google Scholar]
- Camarasa J, Rodrigo T, Pubill D, Escubedo E. Memantine is a useful drug to prevent the spatial and non-spatial memory deficits induced by methamphetamine in rats. Pharmacol Res. 2010;62:450–456. doi: 10.1016/j.phrs.2010.05.004. [DOI] [PubMed] [Google Scholar]
- Cappeliez P, Moore E. Effects of lithium on an amphetamine animal model of bipolar disorder. Prog Neuropsychopharmacol Biol Psychiatry. 1990;14:347–358. doi: 10.1016/0278-5846(90)90023-a. [DOI] [PubMed] [Google Scholar]
- Cassidy F, Forest K, Murry E, Carroll BJ. A factor analysis of the signs and symptoms of mania. Arch Gen Psychiatry. 1998a;55:27–32. doi: 10.1001/archpsyc.55.1.27. [DOI] [PubMed] [Google Scholar]
- Cassidy F, Murry E, Forest K, Carroll BJ. Signs and symptoms of mania in pure and mixed episodes. J Affect Disord. 1998b;50:187–201. doi: 10.1016/s0165-0327(98)00016-0. [DOI] [PubMed] [Google Scholar]
- Chipana C, Camarasa J, Pubill D, Escubedo E. Memantine prevents MDMA-induced neurotoxicity. Neurotoxicology. 2008a;29:179–183. doi: 10.1016/j.neuro.2007.09.005. [DOI] [PubMed] [Google Scholar]
- Chipana C, Torres I, Camarasa J, Pubill D, Escubedo E. Memantine protects against amphetamine derivatives-induced neurotoxic damage in rodents. Neuropharmacology. 2008b;54:1254–1263. doi: 10.1016/j.neuropharm.2008.04.003. [DOI] [PubMed] [Google Scholar]
- Chiu CT, Chuang DM. Molecular actions and therapeutic potential of lithium in preclinical and clinical studies of CNS disorders. Pharmacol Ther. 2010;128:281–304. doi: 10.1016/j.pharmthera.2010.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cilia J, Gartlon JE, Shilliam C, Dawson LA, Moore SH, Jones DN. Further neurochemical and behavioural investigation of Brattleboro rats as a putative model of schizophrenia. J Psychopharmacol. 2010;24:407–419. doi: 10.1177/0269881108098787. [DOI] [PubMed] [Google Scholar]
- Cookson J. Haloperidol and risperidone in mania. In: Akiskal H, Tohen M, editors. Bipolar Psychopharmacotherapy. Chichester: John Wiley & Sons; 2006. pp. 105–123. [Google Scholar]
- Correll CU, Sheridan EM, DelBello MP. Antipsychotic and mood stabilizer efficacy and tolerability in pediatric and adult patients with bipolar I mania: a comparative analysis of acute, randomized, placebo-controlled trials. Bipolar Disord. 2010;12:116–141. doi: 10.1111/j.1399-5618.2010.00798.x. [DOI] [PubMed] [Google Scholar]
- Costall B, Domeney AM, Kelly ME, Naylor RJ, Tomkins DM. Actions of ORG 5222 as a novel psychotropic agent. Pharmacol Biochem Behav. 1990;35:607–615. doi: 10.1016/0091-3057(90)90298-v. [DOI] [PubMed] [Google Scholar]
- Cruceanu C, Alda M, Rouleau G, Turecki G. Response to treatment in bipolar disorder. Curr Opin Psychiatry. 2006;24:24–28. doi: 10.1097/YCO.0b013e328341352c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davies JA, Jackson B, Redfern PH. The effect of amantadine, L-dopa, (plus)-amphetamine and apomorphine on the acquisition of the conditioned avoidance response. Neuropharmacology. 1974;13:199–204. doi: 10.1016/0028-3908(74)90107-5. [DOI] [PubMed] [Google Scholar]
- Decker S, Grider G, Cobb M, Li XP, Huff MO, El-Mallakh RS, et al. Open field is more sensitive than automated activity monitor in documenting ouabain-induced hyperlocomotion in the development of an animal model for bipolar illness. Prog Neuropsychopharmacol Biol Psychiatry. 2000;24:455–462. doi: 10.1016/s0278-5846(99)00111-6. [DOI] [PubMed] [Google Scholar]
- Dencker D, Husum H. Antimanic efficacy of retigabine in a proposed mouse model of bipolar disorder. Behav Brain Res. 2010;207:78–83. doi: 10.1016/j.bbr.2009.09.040. [DOI] [PubMed] [Google Scholar]
- Dencker D, Dias R, Pedersen ML, Husum H. Effect of the new antiepileptic drug retigabine in a rodent model of mania. Epilepsy Behav. 2008;12:49–53. doi: 10.1016/j.yebeh.2007.09.023. [DOI] [PubMed] [Google Scholar]
- Depue RA, Krauss SP, Spoont MR. A two-dimensional model of seasonal affective bipolar disorder. In: Magnusson D, Ohman A, editors. Psychopathology: An Interactionist Perspective. New York: Academic Press; 1987. pp. 95–123. [Google Scholar]
- DiazGranados N, Zarate CA., Jr A review of the preclinical and clinical evidence for protein kinase C as a target for drug development for bipolar disorder. Curr Psychiatry Rep. 2008;10:510–519. doi: 10.1007/s11920-008-0081-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doyle AE, Wilens TE, Kwon A, Seidman LJ, Faraone SV, Fried R, et al. Neuropsychological functioning in youth with bipolar disorder. Biol Psychiatry. 2005;58:540–548. doi: 10.1016/j.biopsych.2005.07.019. [DOI] [PubMed] [Google Scholar]
- Eagle DM, Baunez C. Is there an inhibitory-response-control system in the rat? Evidence from anatomical and pharmacological studies of behavioral inhibition. Neurosci Biobehav Rev. 2010;34:50–72. doi: 10.1016/j.neubiorev.2009.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ehlers CL, Frank E, Kupfer DJ. Social zeitgebers and biological rhythms. A unified approach to understanding the etiology of depression. Arch Gen Psychiatry. 1988;45:948–952. doi: 10.1001/archpsyc.1988.01800340076012. [DOI] [PubMed] [Google Scholar]
- Eichelman B, Thoa NB, Perez-Cruet J. Alkali metal cations: effects on aggression and adrenal enzymes. Pharmacol Biochem Behav. 1973;1:121–123. doi: 10.1016/0091-3057(73)90066-x. [DOI] [PubMed] [Google Scholar]
- Einat H. Modelling facets of mania–new directions related to the notion of endophenotypes. J Psychopharmacol. 2006;20:714–722. doi: 10.1177/0269881106060241. [DOI] [PubMed] [Google Scholar]
- Einat H. Different behaviors and different strains: potential new ways to model bipolar disorder. Neurosci Biobehav Rev. 2007a;31:850–857. doi: 10.1016/j.neubiorev.2006.12.001. [DOI] [PubMed] [Google Scholar]
- Einat H. Establishment of a battery of simple models for facets of bipolar disorder: a practical approach to achieve increased validity, better screening and possible insights into endophenotypes of disease. Behav Genet. 2007b;37:244–255. doi: 10.1007/s10519-006-9093-4. [DOI] [PubMed] [Google Scholar]
- Ellenbroek BA. Treatment of schizophrenia: a clinical and preclinical evaluation of neuroleptic drugs. Pharmacol Ther. 1993;57:1–78. doi: 10.1016/0163-7258(93)90036-d. [DOI] [PubMed] [Google Scholar]
- el-Mallakh RS, Harrison LT, Li R, Changaris DG, Levy RS. An animal model for mania: preliminary results. Prog Neuropsychopharmacol Biol Psychiatry. 1995;19:955–962. doi: 10.1016/0278-5846(95)00123-d. [DOI] [PubMed] [Google Scholar]
- El-Mallakh RS, Decker S, Morris M, Li XP, Huff MO, El-Masri MA, et al. Efficacy of olanzapine and haloperidol in an animal model of mania. Prog Neuropsychopharmacol Biol Psychiatry. 2006;30:1261–1264. doi: 10.1016/j.pnpbp.2006.04.003. [DOI] [PubMed] [Google Scholar]
- Feifel D, Melendez G, Shilling PD. Reversal of sensorimotor gating deficits in Brattleboro rats by acute administration of clozapine and a neurotensin agonist, but not haloperidol: a potential predictive model for novel antipsychotic effects. Neuropsychopharmacology. 2004;29:731–738. doi: 10.1038/sj.npp.1300378. [DOI] [PubMed] [Google Scholar]
- Feifel D, Melendez G, Priebe K, Shilling PD. The effects of chronic administration of established and putative antipsychotics on natural prepulse inhibition deficits in Brattleboro rats. Behav Brain Res. 2007;181:278–286. doi: 10.1016/j.bbr.2007.04.020. [DOI] [PubMed] [Google Scholar]
- Fessler RG, Sturgeon RD, London SF, Meltzer HY. Effects of lithium on behaviour induced by phencyclidine and amphetamine in rats. Psychopharmacology (Berl) 1982;78:373–376. doi: 10.1007/BF00433745. [DOI] [PubMed] [Google Scholar]
- Fisher G, Pelonero AL, Ferguson C. Mania precipitated by prednisone and bromocriptine. Gen Hosp Psychiatry. 1991;13:345–346. doi: 10.1016/0163-8343(91)90041-t. [DOI] [PubMed] [Google Scholar]
- Flaisher-Grinberg S, Overgaard S, Einat H. Attenuation of high sweet solution preference by mood stabilizers: a possible mouse model for the increased reward-seeking domain of mania. J Neurosci Methods. 2009;177:44–50. doi: 10.1016/j.jneumeth.2008.09.018. [DOI] [PubMed] [Google Scholar]
- Fleck DE, Shear PK, Zimmerman ME, Getz GE, Corey KB, Jak A, et al. Verbal memory in mania: effects of clinical state and task requirements. Bipolar Disord. 2003;5:375–380. doi: 10.1034/j.1399-5618.2003.00055.x. [DOI] [PubMed] [Google Scholar]
- Flood DG, Choinski M, Marino MJ, Gasior M. Mood stabilizers increase prepulse inhibition in DBA/2NCrl mice. Psychopharmacology (Berl) 2009;205:369–377. doi: 10.1007/s00213-009-1547-y. [DOI] [PubMed] [Google Scholar]
- Fountoulakis KN, Vieta E. Treatment of bipolar disorder: a systematic review of available data and clinical perspectives. Int J Neuropsychopharmacol. 2008;11:999–1029. doi: 10.1017/S1461145708009231. [DOI] [PubMed] [Google Scholar]
- Frank E, Kupfer DJ, Thase ME, Mallinger AG, Swartz HA, Fagiolini AM, et al. Two-year outcomes for interpersonal and social rhythm therapy in individuals with bipolar I disorder. Arch Gen Psychiatry. 2005;62:996–1004. doi: 10.1001/archpsyc.62.9.996. [DOI] [PubMed] [Google Scholar]
- Fratta W, Collu M, Martellotta MC, Pichiri M, Muntoni F, Gessa GL. Stress-induced insomnia: opioid-dopamine interactions. Eur J Pharmacol. 1987;142:437–440. doi: 10.1016/0014-2999(87)90084-7. [DOI] [PubMed] [Google Scholar]
- Frau R, Orru M, Fa M, Casti A, Manunta M, Fais N, et al. Effects of topiramate on the prepulse inhibition of the acoustic startle in rats. Neuropsychopharmacology. 2007;32:320–331. doi: 10.1038/sj.npp.1301115. [DOI] [PubMed] [Google Scholar]
- Frey BN, Valvassori SS, Reus GZ, Martins MR, Petronilho FC, Bardini K, et al. Effects of lithium and valproate on amphetamine-induced oxidative stress generation in an animal model of mania. J Psychiatry Neurosci. 2006;31:326–332. [PMC free article] [PubMed] [Google Scholar]
- Fullerton CA, Busch AB, Frank RG. The rise and fall of gabapentin for bipolar disorder: a case study on off-label pharmaceutical diffusion. Med Care. 2010;48:372–379. doi: 10.1097/MLR.0b013e3181ca404e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gardner R., Jr Mechanisms in manic-depressive disorder: an evolutionary model. Arch Gen Psychiatry. 1982;39:1436–1441. doi: 10.1001/archpsyc.1982.04290120066013. [DOI] [PubMed] [Google Scholar]
- Gessa GL, Pani L, Fadda P, Fratta W. Sleep deprivation in the rat: an animal model of mania. Eur Neuropsychopharmacol. 1995;5(Suppl.):89–93. doi: 10.1016/0924-977x(95)00023-i. [DOI] [PubMed] [Google Scholar]
- Geyer MA. Are cross-species measures of sensorimotor gating useful for the discovery of procognitive cotreatments for schizophrenia? Dialogues Clin Neurosci. 2006a;8:9–16. doi: 10.31887/DCNS.2006.8.1/mgeyer. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geyer MA. The family of sensorimotor gating disorders: comorbitities or diagnostic overlaps. Neurotox Res. 2006b;10:211–220. doi: 10.1007/BF03033358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geyer MA. Developing translational animal models for symptoms of schizophrenia or bipolar mania. Neurotox Res. 2008;14:71–78. doi: 10.1007/BF03033576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geyer MA, Ellenbroek B. Animal behavior models of the mechanisms underlying antipsychotic atypicality. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27:1071–1079. doi: 10.1016/j.pnpbp.2003.09.003. [DOI] [PubMed] [Google Scholar]
- Geyer MA, Moghaddam B. Animal models relevant to schizophrenia disorders. In: Davis KL, Charney DS, Coyle JT, Nemeroff C, editors. Neuropsychopharmacology: The Fifth Generation of Progress. Philadelphia: Lippincott Williams and Wilkins; 2002. pp. 689–701. [Google Scholar]
- Geyer MA, Russo PV, Masten VL. Multivariate assessment of locomotor behavior: pharmacological and behavioral analyses. Pharmacol Biochem Behav. 1986;25:277–288. doi: 10.1016/0091-3057(86)90266-2. [DOI] [PubMed] [Google Scholar]
- Ghaemi SN, Wingo AP, Filkowski MA, Baldessarini RJ. Long-term antidepressant treatment in bipolar disorder: meta-analyses of benefits and risks. Acta Psychiatr Scand. 2008;118:347–356. doi: 10.1111/j.1600-0447.2008.01257.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Giros B, Jaber M, Jones SR, Wightman RM, Caron MG. Hyperlocomotion and indifference to cocaine and amphetamine in mice lacking the dopamine transporter. Nature. 1996;379:606–612. doi: 10.1038/379606a0. [DOI] [PubMed] [Google Scholar]
- Goldberg JF, Burdick KE. Cognitive side effects of anticonvulsants. J Clin Psychiatry. 2001;62(Suppl. 14):27–33. [PubMed] [Google Scholar]
- Goldberg JF, Burdick KE. Levetiracetam for acute mania. Am J Psychiatry. 2002;159:148. doi: 10.1176/appi.ajp.159.1.148. [DOI] [PubMed] [Google Scholar]
- Goldsmith DR, Wagstaff AJ, Ibbotson T, Perry CM. Lamotrigine: a review of its use in bipolar disorder. Drugs. 2003;63:2029–2050. doi: 10.2165/00003495-200363190-00009. [DOI] [PubMed] [Google Scholar]
- Goodwin FK, Jamison KR. Manic-Depressive Illness. 2nd edn. New York: Oxford University Press; 2007. [Google Scholar]
- Goodwin FK, Murphy DL, Bunney WE., Jr Lithium-carbonate treatment in depression and mania. A longitudinal double-blind study. Arch Gen Psychiatry. 1969;21:486–496. doi: 10.1001/archpsyc.1969.01740220102012. [DOI] [PubMed] [Google Scholar]
- Goodwin GM, Martinez-Aran A, Glahn DC, Vieta E. Cognitive impairment in bipolar disorder: neurodevelopment or neurodegeneration? An ECNP expert meeting report. Eur Neuropsychopharmacol. 2008;18:787–793. doi: 10.1016/j.euroneuro.2008.07.005. [DOI] [PubMed] [Google Scholar]
- Gould TD, Einat H. Animal models of bipolar disorder and mood stabilizer efficacy: a critical need for improvement. Neurosci Biobehav Rev. 2007;31:825–831. doi: 10.1016/j.neubiorev.2007.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gould TJ, Keith RA, Bhat RV. Differential sensitivity to lithium's reversal of amphetamine-induced open-field activity in two inbred strains of mice. Behav Brain Res. 2001;118:95–105. doi: 10.1016/s0166-4328(00)00318-1. [DOI] [PubMed] [Google Scholar]
- Grandjean EM, Aubry JM. Lithium: updated human knowledge using an evidence-based approach: part I: clinical efficacy in bipolar disorder. CNS Drugs. 2009;23:225–240. doi: 10.2165/00023210-200923030-00004. [DOI] [PubMed] [Google Scholar]
- Green MF. Cognitive impairment and functional outcome in schizophrenia and bipolar disorder. J Clin Psychiatry. 2006a;67(Suppl. 9):3–8. discussion 36–42. [PubMed] [Google Scholar]
- Green MF. Cognitive impairment and functional outcome in schizophrenia and bipolar disorder. J Clin Psychiatry. 2006b;67:e12. [PubMed] [Google Scholar]
- Greenwood TA, Alexander M, Keck PE, McElroy S, Sadovnick AD, Remick RA, et al. Evidence for linkage disequilibrium between the dopamine transporter and bipolar disorder. Am J Med Genet. 2001;105:145–151. doi: 10.1002/1096-8628(2001)9999:9999<::aid-ajmg1161>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
- Greenwood TA, Schork NJ, Eskin E, Kelsoe JR. Identification of additional variants within the human dopamine transporter gene provides further evidence for an association with bipolar disorder in two independent samples. Mol Psychiatry. 2006;11:125–133. 115. doi: 10.1038/sj.mp.4001764. [DOI] [PubMed] [Google Scholar]
- Grunze H, Langosch J, Born C, Schaub G, Walden J. Levetiracetam in the treatment of acute mania: an open add-on study with an on-off-on design. J Clin Psychiatry. 2003;64:781–784. doi: 10.4088/jcp.v64n0707. [DOI] [PubMed] [Google Scholar]
- Grunze H, Vieta E, Goodwin GM, Bowden C, Licht RW, Moller HJ, et al. The World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the biological treatment of bipolar disorders: update 2009 on the treatment of acute mania. World J Biol Psychiatry. 2009;10:85–116. doi: 10.1080/15622970902823202. [DOI] [PubMed] [Google Scholar]
- Gualtieri CT, Johnson LG. Comparative neurocognitive effects of 5 psychotropic anticonvulsants and lithium. MedGenMed. 2006;8:46. [PMC free article] [PubMed] [Google Scholar]
- Harrison-Read PE. Models of mania and antimanic drug actions: progressing the endophenotype approach. J Psychopharmacol. 2009;23:334–337. doi: 10.1177/0269881108089840. [DOI] [PubMed] [Google Scholar]
- Hartigan GP. The use of lithium salts in affective disorders. Br J Psychiatry. 1963;109:810–814. doi: 10.1192/bjp.109.463.810. [DOI] [PubMed] [Google Scholar]
- Hasler G, Drevets WC, Gould TD, Gottesman II, Manji HK. Toward constructing an endophenotype strategy for bipolar disorders. Biol Psychiatry. 2006;60:93–105. doi: 10.1016/j.biopsych.2005.11.006. [DOI] [PubMed] [Google Scholar]
- Henry BL, Minassian A, Young JW, Paulus MP, Geyer MA, Perry W. Cross-species assessments of motor and exploratory behavior related to bipolar disorder. Neurosci Biobehav Rev. 2010;34:1296–1306. doi: 10.1016/j.neubiorev.2010.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hicks RA, Moore JD, Hayes C, Phillips N, Hawkins J. REM sleep deprivation increases aggressiveness in male rats. Physiol Behav. 1979;22:1097–1100. doi: 10.1016/0031-9384(79)90263-4. [DOI] [PubMed] [Google Scholar]
- Hirschfeld RM, Kasper S. A review of the evidence for carbamazepine and oxcarbazepine in the treatment of bipolar disorder. Int J Neuropsychopharmacol. 2004;7:507–522. doi: 10.1017/S1461145704004651. [DOI] [PubMed] [Google Scholar]
- Horschitz S, Hummerich R, Lau T, Rietschel M, Schloss P. A dopamine transporter mutation associated with bipolar affective disorder causes inhibition of transporter cell surface expression. Mol Psychiatry. 2005;10:1104–1109. doi: 10.1038/sj.mp.4001730. [DOI] [PubMed] [Google Scholar]
- Hummel B, Walden J, Stampfer R, Dittmann S, Amann B, Sterr A, Schaefer M, Frye MA, Grunze H. Acute antimanic efficacy and safety of oxcarbazepine in an open trial with an on-off-on design. Bipolar Disord. 2002;4:412–417. doi: 10.1034/j.1399-5618.2002.02228.x. [DOI] [PubMed] [Google Scholar]
- Iosifescu DV, Moore CM, Deckersbach T, Tilley CA, Ostacher MJ, Sachs GS, et al. Galantamine-ER for cognitive dysfunction in bipolar disorder and correlation with hippocampal neuronal viability: a proof-of-concept study. CNS Neurosci Ther. 2009;15:309–319. doi: 10.1111/j.1755-5949.2009.00090.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jackson S. Melancholia and Depression: From Hippocratic Times to Modern Times. New Haven, CT: Yale University Press; 1986. [Google Scholar]
- Jochum T, Bar KJ, Sauer H. Topiramate induced manic episode. J Neurol Neurosurg Psychiatry. 2002;73:208–209. doi: 10.1136/jnnp.73.2.208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson G, Gershon S, Hekimian LJ. Controlled evaluation of lithium and chlorpromazine in the treatment of manic states: an interim report. Compr Psychiatry. 1968;9:563–573. doi: 10.1016/s0010-440x(68)80053-7. [DOI] [PubMed] [Google Scholar]
- Johnson G, Gershon S, Burdock EI, Floyd A, Hekimian L. Comparative effects of lithium and chlorpromazine in the treatment of acute manic states. Br J Psychiatry. 1971;119:267–276. doi: 10.1192/bjp.119.550.267. [DOI] [PubMed] [Google Scholar]
- Kato T, Kubota M, Kasahara T. Animal models of bipolar disorder. Neurosci Biobehav Rev. 2007;31:832–842. doi: 10.1016/j.neubiorev.2007.03.003. [DOI] [PubMed] [Google Scholar]
- Keck PE, Jr, McElroy SL. A comparison of the ‘Second Generation Antipsychotics’ in the treatment for bipolar disorder: focus on Clozapine, Quetiapine, Ziprasidone, and Aripiprazole. In: Akiskal H, Tohen M, editors. Bipolar Psychopharmacotherapy. Chichester: John Wiley & Sons; 2006. pp. 125–134. [Google Scholar]
- Keck PE, Jr, Mendlwicz J, Calabrese JR, Fawcett J, Suppes T, Vestergaard PA, et al. A review of randomized, controlled clinical trials in acute mania. J Affect Disord. 2000;59(Suppl. 1):S31–S37. doi: 10.1016/s0165-0327(00)00177-4. [DOI] [PubMed] [Google Scholar]
- Keck PE, Jr, Marcus R, Tourkodimitris S, Ali M, Liebeskind A, Saha A, Ingenito G. A placebo-controlled, double-blind study of the efficacy and safety of aripiprazole in patients with acute bipolar mania. Am J Psychiatry. 2003;160:1651–1684. doi: 10.1176/appi.ajp.160.9.1651. [DOI] [PubMed] [Google Scholar]
- Keck PE, Jr, Hsu HA, Papadakis K, Russo J., Jr Memantine efficacy and safety in patients with acute mania associated with bipolar I disorder: a pilot evaluation. Clin Neuropharmacol. 2009a;32:199–204. doi: 10.1097/WNF.0b013e318184fae2. [DOI] [PubMed] [Google Scholar]
- Keck PE, Orsulak PJ, Cutler AJ, Sanchez R, Torbeyns A, Marcus RN, et al. Aripiprazole monotherapy in the treatment of acute bipolar I mania: a randomized, double-blind, placebo- and lithium-controlled study. J Affect Disord. 2009b;112:36–49. doi: 10.1016/j.jad.2008.05.014. [DOI] [PubMed] [Google Scholar]
- Kelly MP, Logue SF, Dwyer JM, Beyer CE, Majchrowski H, Cai Z, et al. The supra-additive hyperactivity caused by an amphetamine-chlordiazepoxide mixture exhibits an inverted-U dose-response: negative implications for the use of a model in screening for mood stabilizers. Pharmacol Biochem Behav. 2009;92:649–654. doi: 10.1016/j.pbb.2009.03.003. [DOI] [PubMed] [Google Scholar]
- Kelsoe JR, Sadovnick AD, Kristbjarnarson H, Bergesch P, Mroczkowski-Parker Z, Drennan M, et al. Possible locus for bipolar disorder near the dopamine transporter on chromosome 5. Am J Med Genet. 1996;67:533–540. doi: 10.1002/(SICI)1096-8628(19961122)67:6<533::AID-AJMG4>3.0.CO;2-I. [DOI] [PubMed] [Google Scholar]
- Khanna S, Vieta E, Lyons B, Grossman F, Eerdekens M, Kramer M. Risperidone in the treatment of acute mania: double-blind, placebo-controlled study. Br J Psychiatry. 2005;187:229–234. doi: 10.1192/bjp.187.3.229. [DOI] [PubMed] [Google Scholar]
- Kim SH, Yu HS, Park HG, Jeon WJ, Song JY, Kang UG, et al. Dose-dependent effect of intracerebroventricular injection of ouabain on the phosphorylation of the MEK1/2-ERK1/2-p90RSK pathway in the rat brain related to locomotor activity. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32:1637–1642. doi: 10.1016/j.pnpbp.2008.05.027. [DOI] [PubMed] [Google Scholar]
- Koszewska I, Rybakowski JK. Antidepressant-induced mood conversions in bipolar disorder: a retrospective study of tricyclic versus non-tricyclic antidepressant drugs. Neuropsychobiology. 2009;59:12–16. doi: 10.1159/000202824. [DOI] [PubMed] [Google Scholar]
- Koukopoulos A, Reginaldi D, Serra G, Koukopoulos A, Sani G, Serra G. Antimanic and mood-stabilizing effect of memantine as an augmenting agent in treatment-resistant bipolar disorder. Bipolar Disord. 2010;12:348–349. doi: 10.1111/j.1399-5618.2010.00803.x. [DOI] [PubMed] [Google Scholar]
- Kozikowski AP, Gaisina IN, Yuan H, Petukhov PA, Blond SY, Fedolak A, et al. Structure-based design leads to the identification of lithium mimetics that block mania-like effects in rodents. possible new GSK-3beta therapies for bipolar disorders. J Am Chem Soc. 2007;129:8328–8332. doi: 10.1021/ja068969w. [DOI] [PubMed] [Google Scholar]
- Kraepelin E. Psychiatrie. Ein Lehrbuch Für Studirende Und Aerzte. Sechste, Vollständig Umgearbeitete Auflage, 2 Vols. Barth: Leipzig; 1899. [Google Scholar]
- Kruger S, Sarkar R, Pietsch R, Hasenclever D, Braunig P. Levetiracetam as monotherapy or add-on to valproate in the treatment of acute mania-a randomized open-label study. Psychopharmacology (Berl) 2008;198:297–299. doi: 10.1007/s00213-008-1109-8. [DOI] [PubMed] [Google Scholar]
- Kulkarni J, Garland KA, Scaffidi A, Headey B, Anderson R, de Castella A, et al. A pilot study of hormone modulation as a new treatment for mania in women with bipolar affective disorder. Psychoneuroendocrinology. 2006;31:543–547. doi: 10.1016/j.psyneuen.2005.11.001. [DOI] [PubMed] [Google Scholar]
- Kushner SF, Khan A, Lane R, Olson WH. Topiramate monotherapy in the management of acute mania: results of four double-blind placebo-controlled trials. Bipolar Disord. 2006;8:15–27. doi: 10.1111/j.1399-5618.2006.00276.x. [DOI] [PubMed] [Google Scholar]
- Lambert PA, Carraz G, Borselli S, Carbel S. [Neuropsychotropic action of a new anti-epileptic agent: depamide] Ann Med Psychol (Paris) 1966;124:707–710. [PubMed] [Google Scholar]
- Lamberty Y, Margineanu DG, Klitgaard H. Effect of the new antiepileptic drug levetiracetam in an animal model of mania. Epilepsy Behav. 2001;2:454–459. doi: 10.1006/ebeh.2001.0254. [DOI] [PubMed] [Google Scholar]
- Lattimore R. The Iliad of Homer (Transl.) Chicago: University of Chicago Press; 1961. [Google Scholar]
- Legrand R, Fielder R. Role of dominance-submission relationships in shock-induced fighting of mice. J Comp Physiol Psychol. 1973;82:501–506. doi: 10.1037/h0034127. [DOI] [PubMed] [Google Scholar]
- Le-Niculescu H, McFarland MJ, Ogden CA, Balaraman Y, Patel S, Tan J, et al. Phenomic, convergent functional genomic, and biomarker studies in a stress-reactive genetic animal model of bipolar disorder and co-morbid alcoholism. Am J Med Genet B Neuropsychiatr Genet. 2008;147B:134–166. doi: 10.1002/ajmg.b.30707. [DOI] [PubMed] [Google Scholar]
- Leonhard K. Aufteiling Der Endogenen Psychosen Und Ihre Differenzierte Aetiologie. Berlin: Akademie-Verlag; 1957. [Google Scholar]
- Li R, el-Mallakh RS, Harrison L, Changaris DG, Levy RS. Lithium prevents ouabain-induced behavioral changes. Toward an animal model for manic depression. Mol Chem Neuropathol. 1997;31:65–72. doi: 10.1007/BF02815161. [DOI] [PubMed] [Google Scholar]
- Liddell HG, Scott R. A Greek-English Lexicon. Oxford: Clarendon Press; 1940. [Google Scholar]
- Luneberg H. Hippocrates. München: Sämtliche werke; 1897. [Google Scholar]
- Madden GJ, Ewan EE, Lagorio CH. Toward an animal model of gambling: delay discounting and the allure of unpredictable outcomes. J Gambl Stud. 2007;23:63–83. doi: 10.1007/s10899-006-9041-5. [DOI] [PubMed] [Google Scholar]
- Maggs R. Treatment of manic illness with lithium carbonate. Br J Psychiatry. 1963;109:56–65. [Google Scholar]
- Malatynska E, Knapp RJ. Dominant-submissive behavior as models of mania and depression. Neurosci Biobehav Rev. 2005;29:715–737. doi: 10.1016/j.neubiorev.2005.03.014. [DOI] [PubMed] [Google Scholar]
- Malatynska E, Pinhasov A, Crooke JJ, Smith-Swintosky VL, Brenneman DE. Reduction of dominant or submissive behaviors as models for antimanic or antidepressant drug testing: technical considerations. J Neurosci Methods. 2007;165:175–182. doi: 10.1016/j.jneumeth.2007.05.035. [DOI] [PubMed] [Google Scholar]
- Malkesman O, Austin DR, Chen G, Manji HK. Reverse translational strategies for developing animal models of bipolar disorder. Dis Model Mech. 2009;2:238–245. doi: 10.1242/dmm.001628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malkoff-Schwartz S, Frank E, Anderson BP, Hlastala SA, Luther JF, Sherrill JT, et al. Social rhythm disruption and stressful life events in the onset of bipolar and unipolar episodes. Psychol Med. 2000;30:1005–1016. doi: 10.1017/s0033291799002706. [DOI] [PubMed] [Google Scholar]
- Malkoff-Schwartz S, Frank E, Anderson B, Sherrill JT, Siegel L, Patterson D, et al. Stressful life events and social rhythm disruption in the onset of manic and depressive bipolar episodes: a preliminary investigation. Arch Gen Psychiatry. 1998;55:702–707. doi: 10.1001/archpsyc.55.8.702. [DOI] [PubMed] [Google Scholar]
- Manji HK, Bowden CL, Belmaker RH. Bipolar Medications: Mechanisms of Action. Washington DC: American Psychiatric Press; 2000. [Google Scholar]
- Mansell W, Pedley R. The ascent into mania: a review of psychological processes associated with the development of manic symptoms. Clin Psychol Rev. 2008;28:494–520. doi: 10.1016/j.cpr.2007.07.010. [DOI] [PubMed] [Google Scholar]
- Marston HM, Young JW, Martin FD, Serpa KA, Moore CL, Wong EH, et al. Asenapine effects in animal models of psychosis and cognitive function. Psychopharmacology (Berl) 2009;206:699–714. doi: 10.1007/s00213-009-1570-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martinez-Aran A, Vieta E, Colom F, Torrent C, Sanchez-Moreno J, Reinares M, et al. Cognitive impairment in euthymic bipolar patients: implications for clinical and functional outcome. Bipolar Disord. 2004;6:224–232. doi: 10.1111/j.1399-5618.2004.00111.x. [DOI] [PubMed] [Google Scholar]
- Mavrikaki M, Nomikos GG, Panagis G. Efficacy of the atypical antipsychotic aripiprazole in d-amphetamine-based preclinical models of mania. Int J Neuropsychopharmacol. 2010;13:541–548. doi: 10.1017/S1461145709991143. [DOI] [PubMed] [Google Scholar]
- Mazza M, Bria P, Mazza S. Efficacy and safety of oxcarbazepine in bipolar disorder. Can J Psychiatry. 2007;52:272. doi: 10.1177/070674370705200414. author reply 272–273. [DOI] [PubMed] [Google Scholar]
- McClung CA, Sidiropoulou K, Vitaterna M, Takahashi JS, White FJ, Cooper DC, et al. Regulation of dopaminergic transmission and cocaine reward by the clock gene. Proc Natl Acad Sci U S A. 2005;102:9377–9381. doi: 10.1073/pnas.0503584102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McElroy SL, Keck PE., Jr Pharmacologic agents for the treatment of acute bipolar mania. Biol Psychiatry. 2000;48:539–557. doi: 10.1016/s0006-3223(00)00961-6. [DOI] [PubMed] [Google Scholar]
- McIntyre RS, Cohen M, Zhao J, Alphs L, Macek TA, Panagides J. Asenapine versus olanzapine in acute mania: a double-blind extension study. Bipolar Disord. 2009;11:815–826. doi: 10.1111/j.1399-5618.2009.00749.x. [DOI] [PubMed] [Google Scholar]
- McIntyre RS, Konarski JZ, Jones M, Paulsson B. Quetiapine in the treatment of acute bipolar mania: efficacy across a broad range of symptoms. J Affect Disord. 2007;100(Suppl. 1):S5–14. doi: 10.1016/j.jad.2007.02.007. [DOI] [PubMed] [Google Scholar]
- Meyendorff E, Lerer B, Moore NC, Bow J, Gershon S. Methylphenidate infusion in euthymic bipolars: effect of carbamazepine pretreatment. Psychiatry Res. 1985;16:303–308. doi: 10.1016/0165-1781(85)90121-0. [DOI] [PubMed] [Google Scholar]
- Miczek KA, O'Donnell JM. Intruder-evoked aggression in isolated and nonisolated mice: effects of psychomotor stimulants and L-dopa. Psychopharmacology (Berl) 1978;57:47–55. doi: 10.1007/BF00426957. [DOI] [PubMed] [Google Scholar]
- Miczek KA, Maxson SC, Fish EW, Faccidomo S. Aggressive behavioral phenotypes in mice. Behav Brain Res. 2001;125:167–181. doi: 10.1016/s0166-4328(01)00298-4. [DOI] [PubMed] [Google Scholar]
- Minassian A, Young JW. Evaluation of the clinical efficacy of asenapine in schizophrenia. Expert Opin Pharmacother. 2010;11:2107–2115. doi: 10.1517/14656566.2010.506188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Minassian A, Henry BL, Geyer MA, Paulus MP, Young JW, Perry W. The quantitative assessment of motor activity in mania and schizophrenia. J Affect Disord. 2009;120:200–206. doi: 10.1016/j.jad.2009.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mitchell PJ. Antidepressant treatment and rodent aggressive behaviour. Eur J Pharmacol. 2005;526:147–162. doi: 10.1016/j.ejphar.2005.09.029. [DOI] [PubMed] [Google Scholar]
- Moller HJ, Rujescu D. Pharmacogenetics–genomics and personalized psychiatry. Eur Psychiatry. 2010;25:291–293. doi: 10.1016/j.eurpsy.2009.12.015. [DOI] [PubMed] [Google Scholar]
- Moore NA, Leander JD, Benvenga MJ, Gleason SD, Shannon H. Behavioral pharmacology of olanzapine: a novel antipsychotic drug. J Clin Psychiatry. 1997;58(Suppl. 10):37–44. [PubMed] [Google Scholar]
- Morden B, Mitchell G, Dement W. Selective REM sleep deprivation and compensation phenomena in the rat. Brain Res. 1967;5:339–349. doi: 10.1016/0006-8993(67)90042-x. [DOI] [PubMed] [Google Scholar]
- Mukherjee BP, Pradhan SN. Effects of lithium on foot shock-induced aggressive behavior in rats. Arch Int Pharmacodyn Ther. 1976;222:125–131. [PubMed] [Google Scholar]
- Neill JC, Barnes S, Cook S, Grayson B, Idris NF, McLean SL, et al. Animal models of cognitive dysfunction and negative symptoms of schizophrenia: focus on NMDA receptor antagonism. Pharmacol Ther. 2010;128:419–432. doi: 10.1016/j.pharmthera.2010.07.004. [DOI] [PubMed] [Google Scholar]
- O'Donnell KC, Gould TD. The behavioral actions of lithium in rodent models: leads to develop novel therapeutics. Neurosci Biobehav Rev. 2007;31:932–962. doi: 10.1016/j.neubiorev.2007.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O'Brien WT, Harper AD, Jove F, Woodgett JR, Maretto S, Piccolo S, et al. Glycogen synthase kinase-3beta haploinsufficiency mimics the behavioral and molecular effects of lithium. J Neurosci. 2004;24:6791–6798. doi: 10.1523/JNEUROSCI.4753-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oehler J, Jahkel M, Schmidt J. [Effect of lithium, carbamazepine, ca-valproate and diazepam on changes in social isolation-induced behavior in mice] Biomed Biochim Acta. 1985;44:1523–1530. [PubMed] [Google Scholar]
- Ong JC, Brody SA, Large CH, Geyer MA. An investigation of the efficacy of mood stabilizers in rodent models of prepulse inhibition. J Pharmacol Exp Ther. 2005;315:1163–1171. doi: 10.1124/jpet.105.090845. [DOI] [PubMed] [Google Scholar]
- Pande AC, Crockatt JG, Janney CA, Werth JL, Tsaroucha G. Gabapentin in bipolar disorder: a placebo-controlled trial of adjunctive therapy. Gabapentin Bipolar Disorder Study Group. Bipolar Disord. 2000;2:249–255. doi: 10.1034/j.1399-5618.2000.20305.x. [DOI] [PubMed] [Google Scholar]
- Paulus M, Minassian A, Masten V, Feifel D, Geyer M, Perry W. Human Behavioral Pattern Monitor differentiates activity patterns of bipolar manic and attention deficit hyperactivity subjects. Biological Psychiatry. 2007;61:227S. [Google Scholar]
- Peet M, Peters S. Drug-induced mania. Drug Saf. 1995;12:146–153. doi: 10.2165/00002018-199512020-00007. [DOI] [PubMed] [Google Scholar]
- Perry W, Minassian A, Feifel D, Braff DL. Sensorimotor gating deficits in bipolar disorder patients with acute psychotic mania. Biol Psychiatry. 2001;50:418–424. doi: 10.1016/s0006-3223(01)01184-2. [DOI] [PubMed] [Google Scholar]
- Perry W, Minassian A, Paulus MP, Young JW, Kincaid MJ, Ferguson EJ, et al. A reverse-translational study of dysfunctional exploration in psychiatric disorders: from mice to men. Arch Gen Psychiatry. 2009;66:1072–1080. doi: 10.1001/archgenpsychiatry.2009.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perry W, Minassian A, Henry B, Kincaid M, Young JW, Geyer MA. Quantifying over-activity in bipolar and schizophrenia patients in a human open field paradigm. Psychiatry Res. 2010;178:84–91. doi: 10.1016/j.psychres.2010.04.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pinel P. Nosographie Philosphique Ou La Methode De L'analyse Appliquee A Lamedecine. 6th edn. Paris: Brosson; 1818. [Google Scholar]
- Poitou P, Boulu R, Bohuon C. Effect of lithium and other drugs on the amphetamine chlordiazeposice hyperactivity in mice. Experientia. 1975;31:99–101. doi: 10.1007/BF01924701. [DOI] [PubMed] [Google Scholar]
- Poolsup N, Li Wan Po A, de Oliveira IR. Systematic overview of lithium treatment in acute mania. J Clin Pharm Ther. 2000;25:139–156. doi: 10.1046/j.1365-2710.2000.00278.x. [DOI] [PubMed] [Google Scholar]
- Pope HG, Jr, McElroy SL, Keck PE, Jr, Hudson JI. Valproate in the treatment of acute mania. A placebo-controlled study. Arch Gen Psychiatry. 1991;48:62–68. doi: 10.1001/archpsyc.1991.01810250064008. [DOI] [PubMed] [Google Scholar]
- Popova E, Leighton C, Bernabarre A, Bernardo M, Vieta E. Oxcarbazepine in the treatment of bipolar and schizoaffective disorders. Expert Rev Neurother. 2007;7:617–626. doi: 10.1586/14737175.7.6.617. [DOI] [PubMed] [Google Scholar]
- Post RM, Uhde TW. Treatment of mood disorders with antiepileptic medications: clinical and theoretical implications. Epilepsia. 1983;24(Suppl. 2):S97–108. doi: 10.1111/j.1528-1157.1983.tb04652.x. [DOI] [PubMed] [Google Scholar]
- Powell SB, Young JW, Ong JC, Caron MG, Geyer MA. Atypical antipsychotics clozapine and quetiapine attenuate prepulse inhibition deficits in dopamine transporter knockout mice. Behav Pharmacol. 2008;19:562–565. doi: 10.1097/FBP.0b013e32830dc110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prasad V, Sheard MH. Effect of lithium upon desipramine enhanced shock-elicited fighting in rats. Pharmacol Biochem Behav. 1982;17:377–378. doi: 10.1016/0091-3057(82)90097-1. [DOI] [PubMed] [Google Scholar]
- Prickaerts J, Moechars D, Cryns K, Lenaerts I, van Craenendonck H, Goris I, et al. Transgenic mice overexpressing glycogen synthase kinase 3beta: a putative model of hyperactivity and mania. J Neurosci. 2006;26:9022–9029. doi: 10.1523/JNEUROSCI.5216-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prien RF, Caffey EM, Jr, Klett CJ. Comparison of lithium carbonate and chlorpromazine in the treatment of mania. Report of the Veterans Administration and National Institute of Mental Health Collaborative Study Group. Arch Gen Psychiatry. 1972;26:146–153. doi: 10.1001/archpsyc.1972.01750200050011. [DOI] [PubMed] [Google Scholar]
- Ralph RJ, Paulus MP, Fumagalli F, Caron MG, Geyer MA. Prepulse inhibition deficits and perseverative motor patterns in dopamine transporter knock-out mice: differential effects of D1 and D2 receptor antagonists. J Neurosci. 2001;21:305–313. doi: 10.1523/JNEUROSCI.21-01-00305.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ralph-Williams RJ, Paulus MP, Zhuang X, Hen R, Geyer MA. Valproate attenuates hyperactive and perseverative behaviors in mutant mice with a dysregulated dopamine system. Biol Psychiatry. 2003;53:352–359. doi: 10.1016/s0006-3223(02)01489-0. [DOI] [PubMed] [Google Scholar]
- Redrobe JP, Nielsen AN. Effects of neuronal Kv7 potassium channel activators on hyperactivity in a rodent model of mania. Behav Brain Res. 2009;198:481–485. doi: 10.1016/j.bbr.2008.12.027. [DOI] [PubMed] [Google Scholar]
- Reynolds B. A review of delay-discounting research with humans: relations to drug use and gambling. Behav Pharmacol. 2006;17:651–667. doi: 10.1097/FBP.0b013e3280115f99. [DOI] [PubMed] [Google Scholar]
- Roybal K, Theobold D, Graham A, DiNieri JA, Russo SJ, Krishnan V, et al. Mania-like behavior induced by disruption of CLOCK. Proc Natl Acad Sci U S A. 2007;104:6406–6411. doi: 10.1073/pnas.0609625104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sachs GS, Gardner-Schuster EE. Adjunctive treatment of acute mania: a clinical overview. Acta Psychiatr Scand Suppl. 2007;s434:27–34. doi: 10.1111/j.1600-0447.2007.01056.x. [DOI] [PubMed] [Google Scholar]
- Sachs GS, Nierenberg AA, Calabrese JR, Marangell LB, Wisniewski SR, Gyulai L, et al. Effectiveness of adjunctive antidepressant treatment for bipolar depression. N Engl J Med. 2007;356:1711–1722. doi: 10.1056/NEJMoa064135. [DOI] [PubMed] [Google Scholar]
- Savitz J, Solms M, Ramesar R. Neuropsychological dysfunction in bipolar affective disorder: a critical opinion. Bipolar Disord. 2005;7:216–235. doi: 10.1111/j.1399-5618.2005.00203.x. [DOI] [PubMed] [Google Scholar]
- Sax KW, Strakowski SM, Zimmerman ME, DelBello MP, Keck PE, Jr, Hawkins JM. Frontosubcortical neuroanatomy and the continuous performance test in mania. Am J Psychiatry. 1999;156:139–141. doi: 10.1176/ajp.156.1.139. [DOI] [PubMed] [Google Scholar]
- Schou M, Juel-Nielsen N, Stromgren E, Voldby H. The treatment of manic psychoses by the administration of lithium salts. J Neurol Neurosurg Psychiatry. 1954;17:250–260. doi: 10.1136/jnnp.17.4.250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Serretti A, Drago A. Pharmacogenetics of lithium long-term treatment: focus on initiation and adaptation mechanisms. Neuropsychobiology. 2010;62:61–71. doi: 10.1159/000314311. [DOI] [PubMed] [Google Scholar]
- Shaldubina A, Einat H, Szechtman H, Shimon H, Belmaker RH. Preliminary evaluation of oral anticonvulsant treatment in the quinpirole model of bipolar disorder. J Neural Transm. 2002;109:433–440. doi: 10.1007/s007020200035. [DOI] [PubMed] [Google Scholar]
- Shaltiel G, Maeng S, Malkesman O, Pearson B, Schloesser RJ, Tragon T, et al. Evidence for the involvement of the kainate receptor subunit GluR6 (GRIK2) in mediating behavioral displays related to behavioral symptoms of mania. Mol Psychiatry. 2008;13:858–872. doi: 10.1038/mp.2008.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shopsin B, Gershon S, Thompson H, Collins P. Psychoactive drugs in mania. A controlled comparison of lithium carbonate, chlorpromazine, and haloperidol. Arch Gen Psychiatry. 1975;32:34–42. doi: 10.1001/archpsyc.1975.01760190036004. [DOI] [PubMed] [Google Scholar]
- Silverstone PH, Pukhovsky A, Rotzinger S. Lithium does not attenuate the effects of d-amphetamine in healthy volunteers. Psychiatry Res. 1998;79:219–226. doi: 10.1016/s0165-1781(98)00037-7. [DOI] [PubMed] [Google Scholar]
- Singh V, Muzina DJ, Calabrese JR. Anticonvulsants in bipolar disorder. Psychiatr Clin North Am. 2005;28:301–323. doi: 10.1016/j.psc.2005.02.004. [DOI] [PubMed] [Google Scholar]
- Smith LA, Cornelius V, Warnock A, Tacchi MJ, Taylor D. Pharmacological interventions for acute bipolar mania: a systematic review of randomized placebo-controlled trials. Bipolar Disord. 2007;9:551–560. doi: 10.1111/j.1399-5618.2007.00468.x. [DOI] [PubMed] [Google Scholar]
- Spring G, Schweid D, Gray C, Steinberg J, Horwitz M. A double-blind comparison of lithium and chlorpromazine in the treatment of manic states. Am J Psychiatry. 1970;126:1306–1310. doi: 10.1176/ajp.126.9.1306. [DOI] [PubMed] [Google Scholar]
- Sun T, Hu G, Li M. Repeated antipsychotic treatment progressively potentiates inhibition on phencyclidine-induced hyperlocomotion, but attenuates inhibition on amphetamine-induced hyperlocomotion: relevance to animal models of antipsychotic drugs. Eur J Pharmacol. 2009;602:334–342. doi: 10.1016/j.ejphar.2008.11.036. [DOI] [PubMed] [Google Scholar]
- Suppes T, Calabrese JR, Mitchell PB, Pazzaglia PJ, Potter WZ, Zarin DA. Algorithms for the treatment of bipolar manic-depressive illness. Psychopharmacol Bull. 1995;31:469–474. [PubMed] [Google Scholar]
- Swerdlow NR, Braff DL, Taaid N, Geyer MA. Assessing the validity of an animal model of deficient sensorimotor gating in schizophrenic patients. Arch Gen Psychiatry. 1994;51:139–154. doi: 10.1001/archpsyc.1994.03950020063007. [DOI] [PubMed] [Google Scholar]
- Swerdlow NR, Shoemaker JM, Stephany N, Wasserman L, Ro HJ, Geyer MA. Prestimulus effects on startle magnitude: sensory or motor? Behav Neurosci. 2002;116:672–681. doi: 10.1037//0735-7044.116.4.672. [DOI] [PubMed] [Google Scholar]
- Sylvia LG, Alloy LB, Hafner JA, Gauger MC, Verdon K, Abramson LY. Life events and social rhythms in bipolar spectrum disorders: a prospective study. Behav Ther. 2009;40:131–141. doi: 10.1016/j.beth.2008.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Szabo ST, Machado-Vieira R, Yuan P, Wang Y, Wei Y, Falke C, et al. Glutamate receptors as targets of protein kinase C in the pathophysiology and treatment of animal models of mania. Neuropharmacology. 2009;56:47–55. doi: 10.1016/j.neuropharm.2008.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Teng CT, Demetrio FN. Memantine may acutely improve cognition and have a mood stabilizing effect in treatment-resistant bipolar disorder. Rev Bras Psiquiatr. 2006;28:252–254. doi: 10.1590/s1516-44462006000300020. [DOI] [PubMed] [Google Scholar]
- Tohen M, Vieta E. Antipsychotic agents in the treatment of bipolar mania. Bipolar Disord. 2009;11(Suppl. 2):45–54. doi: 10.1111/j.1399-5618.2009.00710.x. [DOI] [PubMed] [Google Scholar]
- Tohen M, Sanger TM, McElroy SL, Tollefson GD, Chengappa KN, Daniel DG, et al. Olanzapine versus placebo in the treatment of acute mania. Olanzapine HGEH Study Group. Am J Psychiatry. 1999;156:702–709. doi: 10.1176/ajp.156.5.702. [DOI] [PubMed] [Google Scholar]
- Torres IJ, Defreitas CM, Defreitas VG, Bond DJ, Kunz M, Honer WG, et al. Relationship between cognitive functioning and 6-month clinical and functional outcome in patients with first manic episode bipolar I disorder. Psychol Med. 2010a;41:1–12. doi: 10.1017/S0033291710001613. [DOI] [PubMed] [Google Scholar]
- Torres IJ, DeFreitas VG, DeFreitas CM, Kauer-Sant'Anna M, Bond DJ, Honer WG, et al. Neurocognitive functioning in patients with bipolar I disorder recently recovered from a first manic episode. J Clin Psychiatry. 2010b;71:1234–1242. doi: 10.4088/JCP.08m04997yel. [DOI] [PubMed] [Google Scholar]
- Vassout A, Veenstra S, Hauser K, Ofner S, Brugger F, Schilling W, Gentsch C. NKP608: a selective NK-1 receptor antagonist with anxiolytic-like effects in the social interaction and social exploration test in rats. Regul Pept. 2000;96:7–16. doi: 10.1016/s0167-0115(00)00194-4. [DOI] [PubMed] [Google Scholar]
- Vasudev K, Macritchie K, Geddes J, Watson S, Young A. Topiramate for acute affective episodes in bipolar disorder. Cochrane Database Syst Rev. 2006;(1):CD003384. doi: 10.1002/14651858.CD003384.pub2. [DOI] [PubMed] [Google Scholar]
- Wang J, Michelhaugh SK, Bannon MJ. Valproate robustly increases Sp transcription factor-mediated expression of the dopamine transporter gene within dopamine cells. Eur J Neurosci. 2007;25:1982–1986. doi: 10.1111/j.1460-9568.2007.05460.x. [DOI] [PubMed] [Google Scholar]
- Weisler RH, Calabrese JR, Bowden CL, Ascher JA, DeVeaugh-Geiss J, Evoniuk G. Discovery and development of lamotrigine for bipolar disorder: a story of serendipity, clinical observations, risk taking, and persistence. J Affect Disord. 2008;108:1–9. doi: 10.1016/j.jad.2007.09.012. [DOI] [PubMed] [Google Scholar]
- Yatham LN, Kusumaker V. Anticonvulsants in treatment of bipolar disorder. In: Yatham LN, Kusumaker V, Kuthcer SP, editors. Bipolar Disorder: A Clinician's Guide to Biological Treatments. New York: Brunner-Routledge; 2003. pp. 201–240. [Google Scholar]
- Yatham LN, Kusumakar V, Calabrese JR, Rao R, Scarrow G, Kroeker G. Third generation anticonvulsants in bipolar disorder: a review of efficacy and summary of clinical recommendations. J Clin Psychiatry. 2002;63:275–283. doi: 10.4088/jcp.v63n0402. [DOI] [PubMed] [Google Scholar]
- Yildiz A, Guleryuz S, Ankerst DP, Ongur D, Renshaw PF. Protein kinase C inhibition in the treatment of mania: a double-blind, placebo-controlled trial of tamoxifen. Arch Gen Psychiatry. 2008;65:255–263. doi: 10.1001/archgenpsychiatry.2007.43. [DOI] [PubMed] [Google Scholar]
- Young AH, Geddes JR, Macritchie K, Rao SN, Watson S, Vasudev A. Tiagabine in the treatment of acute affective episodes in bipolar disorder: efficacy and acceptability. Cochrane Database Syst Rev. 2006;3 doi: 10.1002/14651858.CD004694.pub2. CD004694. [DOI] [PubMed] [Google Scholar]
- Young JW, Minassian A, Paulus MP, Geyer MA, Perry W. A reverse-translational approach to bipolar disorder: rodent and human studies in the Behavioral Pattern Monitor. Neurosci Biobehav Rev. 2007;31:882–896. doi: 10.1016/j.neubiorev.2007.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young JW, Light GA, Marston HM, Sharp R, Geyer MA. The 5-choice continuous performance test: evidence for a translational test of vigilance for mice. PLoS ONE. 2009a;4:e4227. doi: 10.1371/journal.pone.0004227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young JW, Powell SB, Risbrough V, Marston HM, Geyer MA. Using the MATRICS to guide development of a preclinical cognitive test battery for research in schizophrenia. Pharmacol Ther. 2009b;122:150–202. doi: 10.1016/j.pharmthera.2009.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young JW, Goey AK, Minassian A, Perry W, Paulus MP, Geyer MA. GBR 12909 administration as a mouse model of bipolar disorder mania: mimicking quantitative assessment of manic behavior. Psychopharmacology (Berl) 2010a;208:443–454. doi: 10.1007/s00213-009-1744-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young JW, Goey AK, Minassian A, Perry W, Paulus MP, Geyer MA. The mania-like exploratory profile in genetic dopamine transporter mouse models is diminished in a familiar environment and reinstated by subthreshold psychostimulant administration. Pharmacol Biochem Behav. 2010b;96:7–15. doi: 10.1016/j.pbb.2010.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young JW, Goey AK, Minassian A, Perry W, Paulus MP, Geyer MA. The mania-like exploratory profile in genetic dopamine transporter mouse models is diminished in a familiar environment and reinstated by subthreshold psychostimulant administration. Pharmacology Biochem Behavior. 2010c;96:7–15. doi: 10.1016/j.pbb.2010.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young JW, van Enkhuizen JE, Winstanley C, Geyer MA. Increased gambling behaviour in a mouse model of mania. J Psychopharmacol. 2011 doi: 10.1177/0269881111400646. DOI: 10.1177/0269881111400646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zarate CA, Jr, Singh JB, Carlson PJ, Quiroz J, Jolkovsky L, Luckenbaugh DA, et al. Efficacy of a protein kinase C inhibitor (tamoxifen) in the treatment of acute mania: a pilot study. Bipolar Disord. 2007;9:561–570. doi: 10.1111/j.1399-5618.2007.00530.x. [DOI] [PubMed] [Google Scholar]
- Zdanys K, Tampi RR. A systematic review of off-label uses of memantine for psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32:1362–1374. doi: 10.1016/j.pnpbp.2008.01.008. [DOI] [PubMed] [Google Scholar]
- Zhuang X, Oosting RS, Jones SR, Gainetdinov RR, Miller GW, Caron MG, et al. Hyperactivity and impaired response habituation in hyperdopaminergic mice. Proc Natl Acad Sci U S A. 2001;98:1982–1987. doi: 10.1073/pnas.98.4.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
