Open access peer-reviewed chapter

Animal Models in Psychiatric Disorder Studies

Written By

João Victor Nani, Benjamín Rodríguez, Fabio Cardoso Cruz and Mirian Akemi Furuie Hayashi

Submitted: 28 May 2019 Reviewed: 06 August 2019 Published: 14 September 2019

DOI: 10.5772/intechopen.89034

From the Edited Volume

Animal Models in Medicine and Biology

Edited by Eva Tvrdá and Sarat Chandra Yenisetti

Chapter metrics overview

1,177 Chapter Downloads

View Full Metrics

Abstract

Among the diseases affecting the brain, special attention has been paid to psychiatric disorders (PDs) due to high prevalence and significant debilitating clinical features. Many difficulties need to be overcome to find good animal models for PDs, due to their multifactorial origins, high heterogeneity and symptoms, as for instance the hallucinations and delusions, which usually cannot be easily assessed employing ordinary experimental animal models. The use of animal models reproducing at least some specific traits that can be studied individually, known as endophenotypes, is often reported. However, since altered biological pathways are common to many of these disorders, each of these behaviors may also reflect different PDs. In this context, it is possible to perform several approaches, to elicit changes in the endophenotypes of interest, not only in vertebrate models like rodents, but also in invertebrate models which have important advantages due to high conservation of essential pathways, lower complexity, and shorter life cycle compared to mammals. Therefore, animal models are also helpful for elucidating the etiology underlying PDs, by allowing easier access to biological samples that are usually not accessible in clinical studies, as for instance, fresh brain samples, from embryos to adults.

Keywords

  • animal model
  • psychiatric disorders
  • neurodevelopment
  • biomarkers
  • CNS
  • endophenotypes

1. Introduction

According to the World Health Organization (WHO), psychiatric disorders (PDs) comprise a broad range of dysfunctions, with several and some common symptoms. PDs are generally characterized by the combination of symptoms as abnormal thoughts, emotions, behavior, and social interaction. The most common PDs include schizophrenia (SCZ), bipolar disorder (BD), major depression disorder (MDD), attention deficit hyperactivity disorder (ADHD), intellectual disabilities, drug abuse disorders, among others [1].

1.1 The need and the value of animal models for PD studies

There are several reasons to use animal models in the studies of disorders affecting the brain. The poor understanding of the etiopathogenesis and pathophysiology of PDs is clearly reflected by the unmet clinical need for better pharmacological treatments. Therefore, good models are clearly needed to clarify the neurobiology involved in PDs, as well as for the identification of biomarkers useful to assist diagnosis and/or for the development of novel therapies. It is also implausible to move forward in clinical trials with a novel drug tested only in a cell model, without any evidence about its efficacy in animal experiments. The value of animal models to drug development has been demonstrated empirically. For example, the first and the most efficacious drugs available for complex PDs such as SCZ (e.g., chlorpromazine and clozapine) was discovered observing the alterations in behaviors of experimental animals in response to each drug administration. In fact, in the last decades, most of the CNS drugs approved were discovered employing a phenotypic screening approach in animal models [2, 3].

1.2 Challenges to model PDs in animals

A reliable animal model must share several similarities with the studied target to allow a successful translation from the basic to the clinical research. However, several limitations need to be overcome. First, the heterogeneous behavioral symptom characteristics of PDs are in some grade uniquely expressed in humans, and they are certainly impossible to be reproduced authentically in animals as rodents, fishes or worms [4]. Second, there is a lack of an objective measure to unequivocally diagnose mental illness [5], which adds complexity to the modeling any mental disorder in experimental animals. Third, in order to develop meaningful animal models for PDs with potential translational power, the disease phenotypes must be represented in the experimental animals. The selection and update of these phenotypes, in agreement with the recent findings in clinical psychiatry and neuroscience, represents a challenge, as evidenced by the recognized gap between the clinical and basic scientific research [6]. In addition, a rising question is what are the specific traits or phenotypes that an animal model should express to be translatable to specific disorder? (Figure 1).

1.3 How to develop an animal model for PD studies

The traditional approach to establish an animal model in PDs is based on three classic constructs proposed by Willner in 1984: face validity, which determines how much a phenotype presented by a patient is represented by the animal model (corresponds to similarity between the model and the PDs assessed, that includes symptoms, signs, and pharmacological features); construct validity, which demonstrates whether it is possible to reproduce the pathological condition based on processes that are already known to be altered (correspondence between the physiological dysfunctions in the human population and in the animal model); predictive validity, which tries to evaluate if a pharmacological or non-pharmacological intervention is capable to reverse the pathological condition (in other words, if the treatment that is effective in reversing PDs in humans would reverse the changes seen in animals) [7, 8, 9, 10]. However, in practice, no animal models fully meet these three criteria of validity.

Many authors have proposed that instead of these three proposed criteria defining an external validation, in addition, the validity of an animal model should not be simply organisms that resemble human dysfunction, but they would also reproduce the processes by which animals and humans enter this state, and therefore, this could be better exploited by adding a new validation criteria [9]. For instance, the validity by homology, which proposes, for instance, an invertebrate model, such as Drosophila, may not be the ideal animal model for studying complex changes in a brain circuitry, but in turn, it may represent a great choice to study the genetic control of early embryonic development [11]. In fact, the nematode Caenorhabditis elegans is a reliable model with conserved neurobiological systems that has been helpful in the discovery of molecular mechanisms that underlie learning and memory, and, in addition, this animal model has a fully sequenced genome and other several molecular and genetic tools available for researchers [12, 13].

1.4 Symptoms versus endophenotypes in experimental model animals

There is a consensus about the low reliability of the diagnostic construct provided for the employment of Diagnostic and Statistical Manual of Mental Disorders or DMS (which is a manual that determines the criteria for the clinical diagnosis of PDs). The heterogeneity implicit in this classification system and the imprecise quantification of the symptoms make it impossible to deconstruct PDs within model organisms. In fact, an etiology-based nosology system has been advocate for psychiatry, and it has been proposed to identify the endophenotypes that occur in both healthy individuals and subjects with different psychopathologies [14]. Endophenotypes are basically quantitative trait-like deficits that are possible to assess by laboratory-based methods rather than by clinical observation. An endophenotype should be state-independent, heritable, occurring at a high rate in affected families, and in addition, it should be associated to genetic variants of the disorder, as it should be involved the same brain circuits associated with the symptoms of the illness in patients (Table 1).

EndophenotypeDescriptionWhat can be evaluated
Locomotor activityDistance travelled, time spent, and frequency of the movements measured during or after a habituation period or after some stimuli (i.e. drug administration)Behavioral sensitization (BD; ADHD; SCZ); Depressive-like behaviors (MDD); etc…
Latent inhibitionLatent inhibition is the ability of a pre-exposed nonreinforced stimulus to inhibit later stimulus-response learningCognitive impairments (SCZ); etc…
Pre-pulse inhibition (PPI)Decrease of the startle reflex after exposure to a pre-pulse before the pulseCognitive impairments (SCZ); etc…
Working memory and learningDescribes short-term memory, in a olfactory domain and spatial domainCognitive impairments (PDs in general); etc…
Social interactionEvaluation of time spent on exploring a social stimulus.Anxiety-like behaviors; Depressive-like behaviors; etc…
RearingMeasure of activity, investigation and exploratory behavior induced by a drug or/and noveltyAnxiety-like behaviors; etc…
GroomingA maintenance behavior evaluated by the cleaning of the fur; is displayed as reaction to unexpected stimuli and in conflict situationsAnxiety-like behaviors; Depressive-like behaviors; etc…
AggressivenessEvaluation of attack and defensive behavior as reaction to a stimuli or other animalAnxiety-like behaviors; Depressive-like behaviors; etc…
Food intakeAmount of food ingested by the animalAnxiety-like behaviors; Depressive-like behaviors; etc…
Sucrose preference testAssesses the sensitivity to reward based on the rodent’s natural preference for sweets. This test measures the amount of a sweet-tasting solution that the animal ingestsDepressive-like behaviors; etc…
Fear conditioningClassical conditioning paradigm, in which an aversive stimulus is paired with some neutral stimuli. Used to assess associative fear learning and memory in rodents.Cognitive impairments (PDs in general); etc…
Forced swim testMeasures the scoring of swimming and climbing (active behavior), and immobility (passive behavior) when animals are placed in an inescapable cylinder filled with waterDepressive-like behaviors; etc…

Table 1.

Most common endophenotypes used to evaluate behaviors associated with psychiatric disorders (PDs).

Figure 1.

Different approaches to construct animal models for neuropsychiatric disorders studies.

The Research Domains Criteria (RDoC) framework was introduced as an alternative categorization system for psychopathological states [15, 16, 17]. This system provides a platform to improve the translatability of studies from animals to humans, since it supports the endophenotype-based comparison of animals and humans on an objective neurobiological basis across all behavioral domains. In fact, the endophenotypes have been reverse-translated into animal models successfully and allows the evaluation of the neural neurobiological substrates and their circuit dysfunctions [18]. Thus, it has been demonstrated that the modeling of neurobiological and behavioral endophenotypes to reproduce PDs in experimental animals is possible.

The ideal animal model should be derived from risk factors or the causative agent of the human disease. One of the strategies used during the construction of a model is focused on a specific factor that can reproduce the condition as a whole or an aspect of the disease [19]. The choice for the methodology used in establishing a model is fundamentally important to guide which aspect of the disease should be explored, and it is an essential component in the validation of a model known as construct validity.

Advertisement

2. PDs and animal models

In the following sections, selected examples of animal models used in the context of investigating PDs will be demonstrated, indicating which changes are observed in behavioral and molecular levels.

2.1 Animal models in schizophrenia (SCZ)

Schizophrenia (SCZ) is a severe brain disorder, characterized by a set of positive and negative symptoms and cognitive disorders, which are the basis for the clinical diagnosis of individuals who needs to present at least two or more of those symptoms, according to the DSM. SCZ is one of the most debilitating mental disorders, affecting about 21 million people worldwide. The antipsychotics used to treat SCZ patients can soften the development of the disorder, and this pharmacological treatment was the basis for the most accepted theory to explain the neurobiology of SCZ, as noticed by the alterations in the dopamine transmission. In addition, several other theories have been suggested soon after, as for instance, the serotoninergic, glutamatergic, GABAergic, and the neurodevelopmental susceptibility hypothesis, among others [20]. However, none of these theories had allowed the characterization of the etiology or the identification of strong biomarker for the diagnosis of SCZ. Many efforts are being made to characterize a model for SCZ, but there is a great difficulty in reproduce endophenotypes that frame all the groups of symptoms related to this disease, or which allow associating all risk factors that are already known. Below, we exemplify some of these models, and for a more detailed review of SCZ models can be found elsewhere [21].

Most of the models are based on the theory of neurotransmitter imbalance, and they are induced by the disruption of these pathways, other models explore changes in the levels of expression of candidate genes involved in the processes of SCZ susceptibility. It should be considered that SCZ is a multifactorial disorder, and thus, the genetic component should be evaluated in addition to changes in the environment, as in contrast to the models based on genetic alterations, there are those taking into account the environmental changes, such as the prenatal insults, which impose changes in the neurodevelopment processes. Some of these models are exemplified in Table 2.

ModelEndophenotypeMolecular alterationsReferences
Drug-induced models
Amphetamine model of SCZAcute:
↓ Latent inhibition; ↑ locomotion
Chronic:
Same as acute but with ↓ PPI
↑ Mesolimbic dopamine response;
↑ Acetilcholine in PFc
[22, 23, 24, 25, 26]
Glutamatergic manipulation (Phencyclidine; MK-801; Ketamine)↑ Locomotion; ↓ working memory;
↓ Reversal learning performance;
↓ Social interaction; ↓ PPI
↓ PV-immunoreactive neurons in PFc and hippocampus[27, 28, 29]
Genetic manipulation
DISC-1 mutations
Missense mutations models↓ PPI; ↓ latent inhibition;
↑ Depressive-like phenotype
↓ Brain volume;
↓ PDE4B activity and binding to DISC1;
↓ PV-immunoreactive;
↓ Dendritic density
[30, 31, 32]
Dominant-negative isoforms of DISC1↑ Amphetamine sensibility; ↓ working memory↓ Dopamine, DOPAC;
↓ PV-immunoreactive
[33, 34]
Knockdown↑ Amphetamine sensibility; ↓ PPI; ↓ working memory↓ Dopamine; ↓ PV-immunoreactive[35]
Overexpression↑ Amphetamine sensibility; ↑ rearing behavior; ↑ locomotion; ↓ learning in rotarod task↑ Increase in high-affinity D2R;
↑ Translocation of dopamine transporter;
↑ Dopamine inflow
[36]
Neuregulin1, ErbB4, and dysbindin
Knock-out↑ Amphetamine sensibility; ↑ locomotion; ↓ PPI;
↓ Working memory; ↓ social interaction
Neuregulin1; ErbB4:
↓ Hippocampal spine density;
↑ Lateral ventricles;
Dysbindin:
↑ HVA/DA ratio;
↑ Excitability of PFc pyramidal neurones
[37, 38, 39]
Developmental models
Neonatal excitotoxic hippocampal lesion↓ PPI; ↓ Working memory; ↓ Social interaction; ↑ Amphetamine sensibility; ↑ MK-801/PCP sensibility; ↑ locomotion↑ Mesolimbic dopamine response;
↑ Acetilcholine in PFc
[40, 41]
Methylazomethanol (MAM) and polyinosinic-polycytidylic acid (poly I:C)↑ Locomotion;↑ Amphetamine sensibility;↑ MK-801/PCP sensibility; ↓ Social interaction; ↓ PPI; ↓ Working memory↓ PV-immunoreactive neurons in PFc and hippocampus[42, 43, 44]

Table 2.

Some examples of SCZ models induced by drugs, genetic manipulation, and prenatal insults.

All of these models show behavioral and molecular changes that can be associated with SCZ.

PPI = prepulse inhibition; PFc = prefrontal cortex; PV = parvalbumin; PDE4B = cAMP-specific 3",5"-cyclic phosphodiesterase 4B; DISC1 = disrupted-in-schizophrenia 1; DOPAC = dihydroxyphenylacetic acid; HVA = homovanillic acid; DA = dopamine; poly I:C = Polyinosinic:polycytidylic acid.

All of these models show behavioral and molecular changes that can be associated with SCZ.

2.2 Animal models in major depressive disorder (MDD)

Major depressive disorder (MDD) is a common, complex, and heterogeneous mental disorder, characterized by persistent sadness and loss of interest in general activities, affecting about 10% of the population worldwide, and which is caused by multifactorial mechanisms not fully understood yet, characterizing MDD as a disorder with many variations in clinical features among the patients, imposing a consequent high variability in the diagnosis, time course of response and remission [45], which is one of the main reasons justifying the intensive search for animal models and biomarkers, aiming for advances in MDD diagnosis [46]. In addition, these advances could be helpful for a better classification for depressive spectrum, and thereby for improving the treatment [47]. The animal models of depression have been developed based on acute or chronic stress exposure, exogenous administration of glucocorticoids, injuries in brain regions and/or genetic manipulations [48, 49, 50]. There is a great variation in the number of protocols that can be used to induce these changes, in which the stressor, time of exposure to the stimulus, and other parameters may vary. For more detailed review of MDD models, see also [51] (Table 3).

ModelEndophenotypeMolecular alterationsReferences
Stress-induced models
Learned Helplessness↓ Locomotion; ↑ aggressiveness
↓ Grooming; ↓ response to rewards
↑ Sleep disturbance
↓ Norepinephrine ; ↑ BDNF; aberrant miRNA brain- region specific expression[52, 53, 54, 55, 56, 57]
Unpredictable chronic mild stress↓ Food intake; ↓ growth rate;
↓ Locomotion; ↑ aggressiveness;
↓ Response to rewards
↑ Corticosterone; ↓ glucocorticoid receptor expression; ↓ endogenous ATP[58, 59, 60, 61]
Chronic restraint stress model↑ Aggressiveness; ↑ fear conditioning; ↓ locomotion; ↓ food intake↑ CA3 dendritic atrophy and damage; ↓ neurogenesis in dentate gyrus; ↑ apoptotic cell death; ↑ corticosteroid[62, 63, 64]
Social defeat↓ Locomotion; ↓ exploratory activity;
↓ Aggression; ↓ sexual behavior;
↑ Anhedonia; ↑ sleep disturbance ;
↓ Growth rate
↓ Volume and cell proliferation in hippocampus and PFc; ↑ corticosteroid; ↓ serotonin; ↓ BDNF[65, 66, 67]
Early life stress model↑ Anxiety-like behavior;
↑ Depression-like behavior; ↑ Novelty responsivity
↑ BDNF expression PFC and hippocampus[68, 69]
Brain lesion model
Olfactory bulbectomy↑ Locomotion; ↓ working memory; ↓ response to rewards; ↓ food intake; ↑ sleep disturbance; ↑ responsivity to stressorsDysfucntion in HPA and neuro-immune axis; ↓ neurotransmitters; ↑ neuronal degeneration; ↑ BDNF; ↓ neuropeptides[70, 71]
Selective inbreeding
Wistar-Kyoto↓ Locomotion; ↑ immobility in forced swim test; ↑ social avoidance;
↑ freezing to context
↑ Adrenal glands; ↑ corticosterone[72, 73]
Flinders Sensitive Line rat↓ Activity in enclosed arena;
↑ immobility in forced swim test;
↓ sucrose intake under stress
↓ Serotonin synthesis; dysfunction in dopaminergic and noradrenergic systems[74, 75, 76, 77]

Table 3.

Examples of models for MDD induced by stressors, injuries in brain regions, and by selective inbreeding.

BDNF = brain-derived neurotrophic factor; miRNA = microRNA; ATP = adenosine triphosphate; PFc = prefrontal cortex; HPA = hypothalamic–pituitary–adrenal axis.

2.3 Animal models in bipolar disorder (BD)

Bipolar disorder (BD) is a chronic mood disorder, characterized by fluctuations between mania and depressive episodes, which affects approximately 1% of the global population irrespective of nationality, ethnic origin, or socioeconomic status [78]. Due to the complex mood alterations, misdiagnosis in BD is very common, as other mental illnesses as depression and SCZ share several common symptoms, in addition to the specific and common endophenotypes and brain structural changes [79, 80]. The search for advances in diagnosis is important for these disorders, since early diagnosis would be essential to foster earlier suited pharmacological treatment in BD, which was proved to be beneficial to prevent the cognitive deficits and disabilities in these BD patients [81], as also demonstrated for SCZ patients [82]. The major limitation in evaluating a model for BD is the difficulty in reproducing the phases of mania and depression observed in the clinic. Many of these models present only one of these parameters, and they are often developed by genetic alterations in genes known to be involved in this disorder or stressors, mainly involved in the circadian cycle as also demonstrated for other PDs. Another interesting approach used for the development of animal models for BD is the one induced by psychostimulant sensitization (which causes mania-like behavior), as withdrawal from psychostimulants is accompanied by depressive-like behavior, which together leads to changes and compulsory behaviors. Some of these models are exemplified in Table 4. A more detailed review of BD models can be found elsewhere [93].

ModelEndophenotypeMolecular alterationsReferences
Genetic manipulation
BDNF haploinsufficient↑ Locomotion; ↑ agressive behavior; ↑ food intake↓ Brain volume; ↓ BDNF; ↓ dopamine[83, 84]
ERK1 Knock-out↑ Amphetamine sensibility; ↓ learing in fear conditioning; ↑ locomotion; ↓ immobility in forced swim↓ Phospho-RSK1/3 in PFC and striatum;
shift of activity rhythm
[85, 86]
DAT Knock-down↑ Locomotion; ↓ anxiety; ↑ rearing↑ Dopamine[87, 88, 89]
Environmental stress
Sleep deprivation↑ Locomotion; ↑ agressive behavior; ↑ exploratory behavior[90, 91]
Photoperiod lenghts↑ Anxiety; ↑ helplessnessSwitch in dopamine neurotransmission to somatostatin[92]
Sensitization model
Chronic amphetamine administration followed by withdrawal↑ Locomotion; ↑ anxiety; ↑ anhedonia; ↓ motivation; ↓ working memory↓ Dopamine responsiveness
↑ serotonin sensitivty
[94, 95, 96]

Table 4.

Examples of models for BD induced by genetic manipulation, environmental stressors, and induced by sensitization, which lead to some aspects of molecular and behavioral changes related to BD.

BDNF = brain-derived neurotrophic factor; ERK1 = Extracellular signal-regulated kinase 1; DAT = dopamine transporter.

BDNF = brain-derived neurotrophic factor; ERK1 = Extracellular signal-regulated kinase 1; DAT = dopamine transporter.

2.4 Animal models in attention-deficit/hyperactivity disorder (ADHD)

Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder, affecting approximately 2.2–2.8% of worldwide, with multifactorial inducement, as reflected by the heterogeneity found in this disorder, and as indicated by the diversity in its psychiatric comorbidities [97]. This disorder is defined by inappropriate levels of attention deficits and/or hyperactivity behavior, which directly interfere with the normal life and functioning of an individual [98]. While there is no cure for ADHD, currently available treatments can help reducing the symptoms and improving the general functioning, although with a peculiar wide variability due to the clinically and scientifically difficulties to exactly determine the specificity and the origin of the symptoms [99]. As for other PDs, due to the high heritability, animal models for ADHD are mostly derived from genetic alterations or breeding selection or from neonatal insults that can lead to neurodevelopmental changes. Models related to dopaminergic neurotransmission are also important to evaluate ADHD, as also listed in Table 2, and which includes the administration of psychostimulants as amphetamine. A more detailed review on ADHD animal models can be found elsewhere [100] (Table 5).

ModelEndophenotypeMolecular alterationsReferences
Genetic manipulation
Spontaneously hypertensive rats↓ Attention; ↑ motor impulsiveness
↑ Locomotion; ↑ exploratory behavior
↑ Dopamine
↓ Dopamine transporter 1 expression
↓ Brain volume
[101, 102, 103, 104]
Coloboma mouse mutant↑ Locomotion; ↑ exploratory behavior; ↑ amphetamine sensibility↑ Noraedrenergic function
↓ Dopamine
↓ DOPAC and HVA
[105, 106, 107, 108]
Neonatal insults
6-hydroxydopamine↓ Working memory; ↑ locomotion;
↑ Exploratory behavior
↓ Dopamine
↑ Dopamine receptor 4
↓ Serotonin transporter binding in striatum
[109, 110, 111]
Neonatal anoxia↑ Locomotion; ↑ exploratory behavior; ↓ spatial memoryTransient changes in neurotransmitters
↑ Dopamine turnover
↓ Noraepinephrine and 5-HIAA
↓ CA1 cell density
[112–114]

Table 5.

Examples of models for ADHD induced by genetic manipulation in susceptibility genes and selective inbreeding and by prenatal insults.

DOPAC = 3,4-Dihydroxyphenylacetic acid; HVA = Homovanillic acid; 5-HIIA = 5-Hydroxyindoleacetic acid.

Advertisement

3. Conclusion

There is a consensus about the critical role of animal models for the advance and understanding the functioning of brain and brain disorders, as well as for the development of new treatments. However, it is important to use them judiciously and avoid the over interpretations derived for the findings, as it is noticeable that the results obtained on experimental animals are not necessarily confirmed in clinical studies. As it has been shown, there are several approaches to obtain an animal model for studies in psychiatry, but there is still a limitation in reproducing all the conditions involved in the pathophysiology of the disorder, and it is extremely crucial to recognize this limitation. An alternative that has proved to be efficient is to direct the study to a specific symptom domain that can answer at least in part, the significance of these findings to concretely improve the knowledge in PDs, and thereby bring advances in treatment. The crisis of the classification system is evidenced in the diagnostic inflation in psychiatry, which adds complexity to the preclinical research and complicates the modeling of PDs within the available experimental laboratory animals. The recent and alternative approaches as the RDoC to study the brain and behavior are in a relative infancy, but promises bringing new perspectives in how models that can be improved to become indeed helpful to benefit the quality of life of patients with PDs.

Advertisement

Acknowledgments

This work was supported by the São Paulo Research Foundation (Fundação de Amparo à Pesquisa do Estado de São Paulo) (FAPESP No. 2013/13392-4 and 2017/02413-1 for M.A.F.H) and the National Council of Technological and Scientific Development (Conselho Nacional de Desenvolvimento Científico e Tecnológico - CNPq) (477760/2010-4, 557753/2010-4; 508113/2010-5; 311815/2012-0; 475739/2013-2; 311815/2012-0 and 309337/2016-0 for M.A.F.H). Both João V. Nani and Benjamín Rodríguez receive fellowship from CAPES. This study was financed in part by the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), Brazil - Finance Code 001.

References

  1. 1. Sansone RA, Sansone LA. Psychiatric disorders: A global look at facts and figures. Psychiatry. 2010;7(12):16-19
  2. 2. Swinney DC, Anthony J. How were new medicines discovered? Nature Reviews Drug Discovery. 2011;10(7):507-519
  3. 3. Alexandrov V, Brunner D, Hanania T, Leahy E. High-throughput analysis of behavior for drug discovery. European Journal of Pharmacology. 2015;750(5):82-89
  4. 4. Salgado JV, Sandner G. A critical overview of animal models of psychiatric disorders: Challenges and perspectives. Revista Brasileira de Psiquiatria. 2013;35(2):77-81
  5. 5. Lema YY, Gamo NJ, Yang K, Ishizuka K. Trait and state biomarkers for psychiatric disorders: Importance of infrastructure to bridge the gap between basic and clinical research and industry. Psychiatry and Clinical Neurosciences. 2018;72(7):482-489
  6. 6. Kesby JP, Eyles DW, McGrath JJ, Scott JG. Dopamine, psychosis and schizophrenia: The widening gap between basic and clinical neuroscience. Translational Psychiatry. 2018;8:30
  7. 7. McKinney WT, Bunney WE. Animal model of depression. I. Review of evidence: Implications for research. Archives of General Psychiatry. 1969;21(2):240-248
  8. 8. Willner P. The validity of animal models of depression. Psychopharmacology. 1984;83(1):1-16
  9. 9. Belzung C, Lemoine M. Criteria of validity for animal models of psychiatric disorders: Focus on anxiety disorders and depression. Biology of Mood & Anxiety Disorders. 2011;1(1):9
  10. 10. Vervliet B, Raes F. Criteria of validity in experimental psychopathology: Application to models of anxiety and depression. Psychological Medicine. 2012;43(11):2241-2244
  11. 11. Lewis E. A gene complex controlling segmentation in drosophila. Nature. 1978;276(5688):565-570
  12. 12. Kandel ER. The molecular biology of memory storage: A dialogue between genes and synapses. Science. 2001;294(5544):1030-1038
  13. 13. Hulme SE, Whitesides GM. Chemistry and the worm: Caenorhabditis elegans as a platform for integrating chemical and biological research. Angewandte Chemie International Edition in English. 2011;50(21):4774-4807
  14. 14. Surís A, Holliday R, North CS. The evolution of the classification of psychiatric disorders. Behavioral Science. 2016;6(1):5
  15. 15. Insel T, Cuthbert B, Garvey M, Heinssen R, Pine DS, Quinn K, et al. Research domain criteria (RDoC): Toward a new classification framework for research on mental disorders. American Journal of Psychiatry. 2010;167(7):748-751
  16. 16. Cuthbert BN, Insel TR. Toward new approaches to psychotic disorders: The NIMH research domain criteria project. Schizophrenia Bulletin. 2010;36(6):1061-1062
  17. 17. Cuthbert BN. The RDoC framework: Facilitating transition from ICD/DSM to dimensional approaches that integrate neuroscience and psychopathology. World Psychiatry. 2014;3(1):28-35
  18. 18. Greenwood TA, Shutes-David A, Tsuang DW. Endophenotypes in schizophrenia: Digging deeper to identify genetic mechanisms. Journal of Psychiatry and Brain Science. 2019;4(2):e190005
  19. 19. Nestler EJ, Hyman SE. Animal models of neuropsychiatric disorders. Nature Neuroscience. 2010;13(10):1161-1169
  20. 20. Owen M, Sawa A, Mortensen P. Schizophrenia. The Lancet. 2016;388(10039):86-97
  21. 21. Jones CA, Watson DJ, Fone KC. Animal models of schizophrenia. British Journal of Pharmacology. 2011;164(4):1162-1194
  22. 22. Murphy C, Fend M, Russig H, Feldon J. Latent inhibition, but not prepulse inhibition, is reduced during withdrawal from an escalating dosage schedule of amphetamine. Behavioral Neuroscience. 2001;115(6):1247-1256
  23. 23. Peleg-Raibstein D, Sydekum E, Russig H, Feldon J. Withdrawal from repeated amphetamine administration leads to disruption of prepulse inhibition but not to disruption of latent inhibition. Journal of Neural Transmission. 2005;113(9):1323-1336
  24. 24. Martinez V, Parikh V, Sarter M. Sensitized Attentional performance and Fos-Immunoreactive cholinergic neurons in the basal forebrain of amphetamine-pretreated rats. Biological Psychiatry. 2005;57(10):1138-1146
  25. 25. Tenn C, Fletcher P, Kapur S. Amphetamine-sensitized animals show a sensorimotor gating and neurochemical abnormality similar to that of schizophrenia. Schizophrenia Research. 2003;64(2-3):103-114
  26. 26. Turner K, Burne T. Improvement of attention with amphetamine in low- and high-performing rats. Psychopharmacology. 2016;233(18):3383-3394
  27. 27. Castañé A, Santana N, Artigas F. PCP-based mice models of schizophrenia: Differential behavioral, neurochemical and cellular effects of acute and subchronic treatments. Psychopharmacology. 2015;232(21-22):4085-4097
  28. 28. Mouri A, Noda Y, Enomoto T, Nabeshima T. Phencyclidine animal models of schizophrenia: Approaches from abnormality of glutamatergic neurotransmission and neurodevelopment. Neurochemistry International. 2007;51(2-4):173-184
  29. 29. Neill J, Barnes S, Cook S, Grayson B, Idris N, McLean S, et al. Animal models of cognitive dysfunction and negative symptoms of schizophrenia: Focus on NMDA receptor antagonism. Pharmacology & Therapeutics. 2010;128(3):419-432
  30. 30. Jaaro-Peled H. Gene models of schizophrenia: DISC1 mouse models. Progress in Brain Research. 2009;179:75-86
  31. 31. Clapcote S, Lipina T, Millar J, Mackie S, Christie S, Ogawa F, et al. Behavioral phenotypes of disc1 missense mutations in mice. Neuron. 2007;54(3):387-402
  32. 32. Lee F, Fadel M, Preston-Maher K, Cordes S, Clapcote S, Price D, et al. Disc1 point mutations in mice affect development of the cerebral cortex. Journal of Neuroscience. 2011;31(9):3197-3206
  33. 33. Pletnikov M, Ayhan Y, Nikolskaia O, Xu Y, Ovanesov M, Huang H, et al. Inducible expression of mutant human DISC1 in mice is associated with brain and behavioral abnormalities reminiscent of schizophrenia. Molecular Psychiatry. 2008;13(2):173-186
  34. 34. Hikida T, Jaaro-Peled H, Seshadri S, Oishi K, Hookway C, Kong S, et al. Dominant-negative DISC1 transgenic mice display schizophrenia-associated phenotypes detected by measures translatable to humans. Proceedings of the National Academy of Sciences. 2007;104(36):14501-14506
  35. 35. Niwa M, Kamiya A, Murai R, Kubo K, Gruber A, Tomita K, et al. Knockdown of DISC1 by in utero gene transfer disturbs postnatal dopaminergic maturation in the frontal cortex and leads to adult behavioral deficits. Neuron. 2010;65(4):480-489
  36. 36. Trossbach S, Bader V, Hecher L, Pum M, Masoud S, Prikulis I, et al. Misassembly of full-length disrupted-in-schizophrenia 1 protein is linked to altered dopamine homeostasis and behavioral deficits. Molecular Psychiatry. 2016;21(11):1561-1572
  37. 37. Mei L, Xiong W. Neuregulin 1 in neural development, synaptic plasticity and schizophrenia. Nature Reviews Neuroscience. 2008;9(6):437-452
  38. 38. Papaleo F, Yang F, Garcia S, Chen J, Lu B, Crawley J, et al. Dysbindin-1 modulates prefrontal cortical activity and schizophrenia-like behaviors via dopamine/D2 pathways. Molecular Psychiatry. 2010;17(1):85-98
  39. 39. Karlsgodt K, Robleto K, Trantham-Davidson H, Jairl C, Cannon T, Lavin A, et al. Reduced dysbindin expression mediates n-methyl-d-aspartate receptor hypofunction and impaired working memory performance. Biological Psychiatry. 2011;69(1):28-34
  40. 40. Sams-Dodd F, Lipska B, Weinberger D. Neonatal lesions of the rat ventral hippocampus result in hyperlocomotion and deficits in social behaviour in adulthood. Psychopharmacology. 1997;132(3):303-310
  41. 41. Lipska B. Using animal models to test a neurodevelopmental hypothesis of schizophrenia. Journal of Psychiatry & Neuroscience. 2004;29(4):282-286
  42. 42. Moore H, Jentsch J, Ghajarnia M, Geyer M, Grace A. A neurobehavioral systems analysis of adult rats exposed to methylazoxymethanol acetate on E17: Implications for the neuropathology of schizophrenia. Biological Psychiatry. 2006;60(3):253-264
  43. 43. Meyer U. Prenatal poly(I:C) exposure and other developmental immune activation models in rodent systems. Biological Psychiatry. 2014;75(4):307-315
  44. 44. Winship I, Dursun S, Baker G, Balista P, Kandratavicius L, Maia-de-Oliveira J, et al. An overview of animal models related to schizophrenia. The Canadian Journal of Psychiatry. 2018;64(1):5-17
  45. 45. Belmaker RH, Agam G. Major Depressive Disorder. The New England Journal of Medicine. 2008;358:55-68
  46. 46. Redei EE, Mehta NS. The promise of biomarkers in diagnosing major depression in primary care: The present and future. Current Psychiatry Reports. 2015;17(8):601
  47. 47. Woods AG, Iosifescu DV, Darie CC. Biomarkers in major depressive disorder: The role of mass spectrometry. Advances in Experimental Medicine and Biology. 2014;806:545-560
  48. 48. Caspi A, Moffitt TE. Gene-environment interactions in psychiatry: Joining forces with neuroscience. Nature Reviews Neuroscience. 2006;7(7):583-590
  49. 49. McGonagle KA, Kessler RC. Chronic stress, acute stress, and depressive symptoms. American Journal of Community Psychology. 1990;18(5):681-706
  50. 50. Uher R, McGuffin P. The moderation by the serotonin transporter gene of environmental adversity in the etiology of depression: 2009 update. Molecular Psychiatry. 2010;15(1):18-22
  51. 51. Wang Q , Timberlake MA 2nd, Prall K, Dwivedi Y. The recent progress in animal models of depression. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2017;77:99-109
  52. 52. Weiss J, Simson PG, Ambrose M, Webster A, Hoffman L. Neurochemical basis of behavioral depression. Advances in Behavioral Medicine. 1985;1:233-275
  53. 53. Weiss J, Bailey WH, Pohorecky LA, Korzeniowski D, Grillione G. Stress-induced depression of motor activity correlates with regional changes in brain norepinephrine but not in dopamine. Neurochemical Research. 1980;5(1):9-22
  54. 54. Zacharko R, Bowers W, Kokkinidis L, Anisma H. Region-specific reductions of intracranial self-stimulation after uncontrollable stress: Possible effects on reward processes. Behavioural Brain Research. 1983;9(2):129-141
  55. 55. Corum R, Thurmond J. Effects of acute exposure to stress on subsequent aggression and locomotion performance. Psychosomatic Medicine. 1977;39(6):436-443
  56. 56. Dwivedi Y, Mondal A, Shukla P, Rizavi H, Lyons J. Altered protein kinase a in brain of learned helpless rats: Effects of acute and repeated stress. Biological Psychiatry. 2004;56(1):30-40
  57. 57. Aznar S, Klein A, Santini M, Knudsen G, Henn F, Gass P, et al. Aging and depression vulnerability interaction results in decreased serotonin innervation associated with reduced BDNF levels in hippocampus of rats bred for learned helplessness. Synapse. 2010;64(7):561-565
  58. 58. Katz R, Roth K, Carroll B. Acute and chronic stress effects on open field activity in the rat: Implications for a model of depression. Neuroscience & Biobehavioral Reviews. 1981;5(2):247-251
  59. 59. Willner P, Muscat R, Papp M. Chronic mild stress-induced anhedonia: A realistic animal model of depression. Neuroscience & Biobehavioral Reviews. 1992;16(4):525-534
  60. 60. Boyle M, Brewer J, Funatsu M, Wozniak D, Tsien J, Izumi Y, et al. Acquired deficit of forebrain glucocorticoid receptor produces depression-like changes in adrenal axis regulation and behavior. Proceedings of the National Academy of Sciences. 2005;102(2):473-478
  61. 61. Crema L, Schlabitz M, Tagliari B, Cunha A, Simão F, Krolow R, et al. Na+, K+ ATPase activity is reduced in amygdala of rats with chronic stress-induced anxiety-like behavior. Neurochemical Research. 2010;35(11):1787-1795
  62. 62. Conrad C, Magariños A, LeDoux J, McEwen B. Repeated restraint stress facilitates fear conditioning independently of causing hippocampal CA3 dendritic atrophy. Behavioral Neuroscience. 1999;113(5):902-913
  63. 63. Wood G, Young L, Reagan L, McEwen B. Acute and chronic restraint stress alter the incidence of social conflict in male rats. Hormones and Behavior. 2003;43(1):205-213
  64. 64. Zhang L, Luo J, Zhang M, Yao W, Ma X, Yu S. Effects of curcumin on chronic, unpredictable, mild, stress-induced depressive-like behaviour and structural plasticity in the lateral amygdala of rats. The International Journal of Neuropsychopharmacology. 2014;17(05):793-806
  65. 65. Koolhaas J, Meerlo P, De Boer S, Strubbe J, Bohus B. The temporal dynamics of the stress response. Neuroscience & Biobehavioral Reviews. 1997;21(6):775-782
  66. 66. Crawford L, Rahman S, Beck S. Social stress alters inhibitory synaptic input to distinct subpopulations of raphe serotonin neurons. ACS Chemical Neuroscience. 2013;4(1):200-209
  67. 67. Hollis F, Kabbaj M. Social defeat as an animal model for depression. ILAR Journal. 2014;55(2):221-232
  68. 68. Liu H, Atrooz F, Salvi A, Salim S. Behavioral and cognitive impact of early life stress: Insights from an animal model. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2017;78:88-95
  69. 69. Boulle F, Pawluski JL, Homberg JR, Machiels B, Kroeze Y, Kumar N, et al. Prenatal stress and early-life exposure to fluoxetine have enduring effects on anxiety and hippocampal BDNF gene expression in adult male offspring. Developmental Psychobiology. 2016;58(4):427-438
  70. 70. Harkin A, Kelly J, Leonard B. A review of the relevance and validity of olfactory bulbectomy as a model of depression. Clinical Neuroscience Research. 2003;3(4-5):253-262
  71. 71. Hellweg R, Zueger M, Fink K, Hörtnagl H, Gass P. Olfactory bulbectomy in mice leads to increased BDNF levels and decreased serotonin turnover in depression-related brain areas. Neurobiology of Disease. 2007;25(1):1-7
  72. 72. Nam H, Clinton S, Jackson N, Kerman I. Learned helplessness and social avoidance in the Wistar-Kyoto rat. Frontiers in Behavioral Neuroscience. 2014;8:109
  73. 73. Will C, Aird F, Redei E. Selectively bred Wistar–Kyoto rats: An animal model of depression and hyper-responsiveness to antidepressants. Molecular Psychiatry. 2003;8(11):925-932
  74. 74. Zangen A, Overstreet D, Yadid G. High serotonin and 5-hydroxyindoleacetic acid levels in limbic brain regions in a rat model of depression; normalization by chronic antidepressant treatment. Journal of Neurochemistry. 2002;69(6):2477-2483
  75. 75. Overstreet D, Russell R. Selective breeding for diisopropyl fluorophosphate-sensitivity: Behavioural effects of cholinergic agonists and antagonists. Psychopharmacology. 1982;78(2):150-155
  76. 76. Overstreet D, Friedman E, Mathé A, Yadid G. The Flinders sensitive line rat: A selectively bred putative animal model of depression. Neuroscience & Biobehavioral Reviews. 2005;29(4-5):739-759
  77. 77. Nishi K, Kanemaru K, Hasegawa S, Watanabe A, Diksic M. Both acute and chronic buspirone treatments have different effects on regional 5-HT synthesis in Flinders sensitive line rats (a rat model of depression) than in control rats. Neurochemistry International. 2009;54(3-4):205-214
  78. 78. Merikangas KR, Jin R, He JP, Kessler RC, Lee S, Sampson NA, et al. Prevalence and correlates of bipolar spectrum disorder in the world mental health survey initiative. Archives of General Psychiatry. 2011;68(3):241-251
  79. 79. Grande I, Berk M, Birmaher B, Vieta E. Bipolar disorder. Lancet. 2016;387(10027):1561-1572
  80. 80. Johansson V, Hultman CM, Kizling I, Martinsson L, Borg J, Hedman A, et al. The schizophrenia and bipolar twin study in Sweden (STAR). Schizophrenia Research. 2019;204:183-192
  81. 81. Sanchez-Moreno J, Martinez-Aran A, Vieta E. Treatment of functional impairment in patients with bipolar disorder. Current Psychiatry Reports. 2017;19(1):3
  82. 82. Lewandowski KE, Whitton AE, Pizzagalli DA, Norris LA, Ongur D, Hall MH. Reward learning, neurocognition, social cognition, and symptomatology in psychosis. Frontiers in Psychiatry. 2016;7:100
  83. 83. Beyer D, Freund N. Animal models for bipolar disorder: From bedside to the cage. International Journal of Bipolar Disorders. 2017;5(1):35
  84. 84. Magariños A, Li C, Gal Toth J, Bath K, Jing D, Lee F, et al. Effect of brain-derived neurotrophic factor haploinsufficiency on stress-induced remodeling of hippocampal neurons. Hippocampus. 2011;21(3):253-264
  85. 85. Kernie S. BDNF regulates eating behavior and locomotor activity in mice. The EMBO Journal. 2000;19(6):1290-1300
  86. 86. Engel S, Creson T, Hao Y, Shen Y, Maeng S, Nekrasova T, et al. The extracellular signal-regulated kinase pathway contributes to the control of behavioral excitement. Molecular Psychiatry. 2008;14(4):448-461
  87. 87. Young JW, Cope ZA, Romoli B, Schurs E, Joosen A, Enkhuzien J, et al. Mice with reduced DAT levels recreate seasonal-induced switching between states in bipolar disorder. Neuropsychopharmacology. 2018;43(8):1732-1731
  88. 88. van Enkhuizen J, Henry B, Minassian A, Perry W, Milienne-Petiot M, Higa K, et al. Reduced dopamine transporter functioning induces high-reward risk-preference consistent with bipolar disorder. Neuropsychopharmacology. 2014;39(13):3112-3122
  89. 89. Giros B, Jaber M, Jones S, Wightman R, Caron M. Hyperlocomotion and indifference to cocaine and amphetamine in mice lacking the dopamine transporter. Nature. 1996;379(6566):606-612
  90. 90. Benedetti F, Fresi F, Maccioni P, Smeraldi E. Behavioural sensitization to repeated sleep deprivation in a mice model of mania. Behavioural Brain Research. 2008;187(2):221-227
  91. 91. Gessa G, Pani L, Fadda P, Fratta W. Sleep deprivation in the rat: An animal model of mania. European Neuropsychopharmacology. 1995;5:89-93
  92. 92. Dulcis D, Jamshidi P, Leutgeb S, Spitzer N. Neurotransmitter switching in the adult brain regulates behavior. Science. 2013;340(6131):449-453
  93. 93. Paulson P, Camp D, Robinson T. Time course of transient behavioral depression and persistent behavioral sensitization in relation to regional brain monoamine concentrations during amphetamine withdrawal in rats. Psychopharmacology. 1991;103(4):480-492
  94. 94. Barr A, Fiorino D, Phillips A. Effects of withdrawal from an escalating dose schedule of d-amphetamine on sexual behavior in the male rat. Pharmacology Biochemistry and Behavior. 1999;64(3):597-604
  95. 95. Barr A, Phillips A. Increased successive negative contrast in rats withdrawn from an escalating-dose schedule of d-amphetamine. Pharmacology Biochemistry and Behavior. 2002;71(1-2):293-299
  96. 96. Marszalek-Grabska M, Gibula-Bruzda E, Jenda M, Gawel K, Kotlinska J. Memantine improves memory impairment and depressive-like behavior induced by amphetamine withdrawal in rats. Brain Research. 1642;2016:389-396
  97. 97. Luo Y, Weibman D, Halperin JM, Li X. A review of heterogeneity in attention deficit/hyperactivity disorder (ADHD). Frontiers in Human Neuroscience. 2019;13:42
  98. 98. Franke B, Michelini G, Asherson P, Banaschewski T, Bilbow A, Buitelaar JK, et al. Live fast, die young? A review on the developmental trajectories of ADHD across the lifespan. European Neuropsychopharmacology. 2018;28(10):1059-1088
  99. 99. Nigg JT. Attention-deficit/hyperactivity disorder and adverse health outcomes. Clinical Psychology Review. 2013;33(2):215-228
  100. 100. Russell VA, Sagvolden T, Johansen E. Animal models of attention-deficit hyperactivity disorder. Behavioral and Brain Functions. 2005;1:9
  101. 101. Sagvolden T, Russell V, Aase H, Johansen E, Farshbaf M. Rodent models of attention-deficit/hyperactivity disorder. Biological Psychiatry. 2005;57(11):1239-1247
  102. 102. Sagvolden T. Behavioral validation of the spontaneously hypertensive rat (SHR) as an animal model of attention-deficit/hyperactivity disorder (AD/HD). Neuroscience & Biobehavioral Reviews. 2000;24(1):31-39
  103. 103. Carboni E, Silvagni A, Valentini V, Di Chiara G. Effect of amphetamine, cocaine and depolarization by high potassium on extracellular dopamine in the nucleus accumbens shell of SHR rats. An in vivo microdyalisis study. Neuroscience & Biobehavioral Reviews. 2003;27(7):653-659
  104. 104. Linthorst A, van Giersbergen P, Gras M, Versteeg D, de Jong W. The nigrostriatal dopamine system: Role in the development of hypertension in spontaneously hypertensive rats. Brain Research. 1994;639(2):261-268
  105. 105. Wilson M. Coloboma mouse mutant as an animal model of hyperkinesis and attention deficit hyperactivity disorder. Neuroscience & Biobehavioral Reviews. 2000;24(1):51-57
  106. 106. Jones M, Williams M, Hess E. Expression of catecholaminergic mRNAs in the hyperactive mouse mutant coloboma. Molecular Brain Research. 2001;96(1-2):114-121
  107. 107. Raber J, Mehta P, Kreifeldt M, Parsons L, Weiss F, Bloom F, et al. Coloboma hyperactive mutant mice exhibit regional and transmitter-specific deficits in neurotransmission. Journal of Neurochemistry. 2002;68(1):176-186
  108. 108. Jones M, Hess E. Norepinephrine regulates locomotor hyperactivity in the mouse mutant coloboma. Pharmacology Biochemistry and Behavior. 2003;75(1):209-216
  109. 109. Luthman J, Fredriksson A, Lewander T, Jonsson G, Archer T. Effects ofd-amphetamine and methylphenidate on hyperactivity produced by neonatal 6-hydroxydopamine treatment. Psychopharmacology. 1989;99(4):550-557
  110. 110. Zhang K. Role of dopamine D4 receptors in motor hyperactivity induced by neonatal 6-hydroxydopamine lesions in rats. Neuropsychopharmacology. 2001;25(5):624-632
  111. 111. Zhang K, Davids E, Tarazi F, Baldessarini R. Serotonin transporter binding increases in caudate-putamen and nucleus accumbens after neonatal 6-hydroxydopamine lesions in rats: Implications for motor hyperactivity. Developmental Brain Research. 2002;137(2):135-138
  112. 112. Dell’Anna M. Neonatal anoxia induces transitory hyperactivity, permanent spatial memory deficits and CA1 cell density reduction in developing rats. Behavioural Brain Research. 1999;45:125-134
  113. 113. Dell’Anna M, Calzolari S, Molinari M, Iuvone L, Calimici R. Neonatal anoxia induces transitory hyperactivity, permanent spatial memory deficits and CA1 cell density reduction in developing rats. Behavioural Brain Research. 1991;45(2):125-134
  114. 114. Iuvone L, Geloso M, Dell’Anna E. Changes in open field behavior, spatial memory, and hippocampal parvalbumin immunoreactivity following enrichment in rats exposed to neonatal anoxia. Experimental Neurology. 1996;139(1):25-33

Written By

João Victor Nani, Benjamín Rodríguez, Fabio Cardoso Cruz and Mirian Akemi Furuie Hayashi

Submitted: 28 May 2019 Reviewed: 06 August 2019 Published: 14 September 2019