Vous êtes sur la page 1sur 30

Accepted Manuscript

Title: Animal Models of CNS Disorders Author: Paul McGonigle PII: DOI: Reference: To appear in: Received date: Accepted date: S0006-2952(13)00387-0 http://dx.doi.org/doi:10.1016/j.bcp.2013.06.016 BCP 11671 BCP 18-6-2013 18-6-2013

Please cite this article as: McGonigle P, Animal Models of CNS Disorders, Biochemical Pharmacology (2013), http://dx.doi.org/10.1016/j.bcp.2013.06.016 This is a PDF le of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its nal form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

McGonigle CNS Models FINAL 061713.docx

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

BCP Redefining Pharmacology Article #7d 6/16/13

Department of Pharmacology and Physiology Drexel University College of Medicine


th

245 North 15 Street, Philadelphia, PA 19102-1192

Running Title: Animal models of CNS disorders Key words:

Word count (abstract and text): 6578 98 References (word count: 2561) 2 Tables

* Correspondence: Paul McGonigle, Ph.D., E-mail: paul.mcgonigle@drexelmed.edu; Phone: 215-7628408; Fax: 215-762-2299

Ac ce p

te
1
Page 1 of 29

an

us

Paul McGonigle

cr

Animal Models of CNS Disorders

ip t

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

Abstract There is intense interest in the development and application of animal models of CNS disorders to explore pathology and molecular mechanisms, identify potential biomarkers, and to assess the therapeutic utility, estimate safety margins and establish pharmacodynamic and pharmacokinetic parameters of new chemical entities (NCEs). This is a daunting undertaking, due to the complex and heterogeneous nature of these disorders, the subjective and sometimes contradictory nature of the

Historically, these models have been invaluable in the discovery of therapeutics for a range of disorders including anxiety, depression, schizophrenia, and Parkinsons Disease. Recently, however, they have been increasing criticized in the wake of numerous clinical trial failures of NCEs with promising preclinical profiles. These failures have resulted from a number of factors including inherent limitations of the models, over-interpretation of preclinical results and the complex nature of clinical trials for CNS

commonly used models for psychiatric, neurodegenerative and neurological disorders as well as critical

to neurodegenerative disorder research. To date, transgenic animal models \ have not been the panacea

steady progress with the promise of eventual therapeutic breakthroughs.

Ac ce p

for drug discovery that many had hoped for. However continual refinement of these models is leading to

te

molecular genetics and the development of transgenic animals has fundamentally changed the approach

factors that affect the variability and reproducibility of these models. It also addresses how progress in

M
2

disorders. This review discusses the rationale, strengths, weaknesses and predictive validity of the most

an

us

cr

clinical endpoints and the paucity of information regarding underlying molecular mechanisms.

ip t

Page 2 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

1. Introduction Animal models are essential research tools that are used to: explore the underlying pathology and molecular mechanisms of disorders; evaluate the potential efficacy of therapeutic interventions; and provide an initial estimate of the safety margin and human dosing parameters of a drug candidate. There are numerous limitations and caveats to the use of such models, not the least of which is the inherent challenge associated with attempting to model complex and still poorly understood human disorders in a

the genetic and epigenetic origins and molecular mechanisms responsible for these disorders, the

endpoints that are used to describe their symptoms and severity. For example, the DSM IV criteria for depression include: large increases or decreases in appetite, insomnia or excessive sleeping and agitation or slowness of movement. The reader is left to ponder the challenge of trying to replicate such symptom clusters in an animal. A more basic technical challenge when using animal models of CNS disorders to assess mechanism of action, therapeutic potential or safety margin of candidate or tool compounds is surmounting the blood-brain barrier (BBB). This involves successfully penetrating the BBB to gain access to the intended target(s) and avoiding active transport out of the CNS by P-glycoprotein transporters. Considerable progress has been made in identifying the physicochemical properties of molecules that favor BBB penetration and developing vitro assays to assess P-glycoprotein transport liability but it is strongly recommended that brain levels of test compounds be measured to properly interpret behavioral results.

2. Animal models of neuropsychiatric disorders. Despite the challenges, significant progress has been made in the development and optimization of behavioral models for the majority of CNS disorders and these models have provided valuable insights regarding mechanism and treatment when used appropriately. These models are primarily focused on endophenotypes and designed to mimic one or more symptoms of the human disorder. Even this reductionist approach is complicated by the observation that DSM criteria for depression allow for

Ac ce p

te

M
3

an

heterogeneous nature of many of these conditions and the subjective and sometimes contradictory

us

lower species This task is particularly difficult for CNS disorders due to the paucity of information about

cr

ip t

Page 3 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

different symptom clusters, or endophenotypes, to lead to the same diagnosis [1]. A key criterion that is often used when assessing the utility of an animal model is validity. The most common types of validity that are considered are face validity which requires similar symptom manifestation to the clinical condition, construct validity which requires the model to have similar underlying biology and predictive validity which requires responsiveness to clinically effective therapeutic agents.

In general, animal models of CNS disorders have good predictive validity for compounds working

properties. In many cases, these models were established or refined to detect the therapeutic potential of prototypical or first in class molecules. For example, the Forced Swim Test (FST) gained acceptance based on its ability to detect the activity of tricyclic antidepressants and needed to be refined to detect the activity of Selective Serotonin Reuptake Inhibitors (SSRIs) [2,3]. CNS models exhibit varying degrees of face validity and one of the better examples is the test for pre-pulse inhibition (PPI) [4]. Schizophrenic

Very few CNS models exhibit construct validity but one good example is the Huntingtons disease (HD)

produces aberrant huntingtin protein and exhibits a rich phenotype that includes deficits in motor, mood

good correspondence in gene expression changes between the animal model and human patients [7,8]. There are no consensus gold standard models for any CNS disorder, only popular or common ly used models for which there is considerable historical data to aid in the interpretation of results. Perhaps the closest the CNS field comes to a gold standard are self -administration assays that have been shown to be highly reliable in predicting abuse potential [9]. With the evolution of molecular genetics and the widespread ability to create transgenic mouse models of several disorders, considerable effort has been invested in adapting tests established and validated in rats to work in mice. More recently, transgenic rats for disorders such as Amyotrophic Lateral Sclerosis, HD, and Alzheimers disease (AD) have been created, in part, to enable the use of better-validated and more robust behavioral assays [10,11,12].

Ac ce p

and cognition and reduced time of survival [5,6]. Interestingly, transcriptional profiling studies reveal a

te

resulting in the expression of aberrant huntingtin protein. The R6/2 knock-in transgenic mouse model

transgenic mouse [5]. HD is caused by a variable triplet repeat in the coding region of the huntingtin gene

M
4

patients exhibit deficits in PPI that can be mimicked by treatment with PCP or amphetamine in rodents.

an

us

cr

through established mechanisms assuming the compounds have appropriate pharmacokinetic

ip t

Page 4 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

While considerable progress has been made on this front, it is important to remember that a mouse is not a small rat and there are often critical procedural differences between the rat and mouse versions of these tests. The following sections describe some of the most popular models for common psychiatric and neurological disorders, their utility and limitations as well as some future trends. Table 1 provides a list of critical factors for different general categories of models and Table 2 provides a summary of models and recommended reviews for the disorders covered in this review. 2.1. Depression

or more symptoms of the disorder. One of the most popular and widely used models of depression is the

eventually adopt an immobile posture after initial attempts to escape [2,13]. This behavior is interpreted as a form of learned helplessness or behavioral despair and the time spent trying to escape can be increased by administration of antidepressants. Modification of the assay to recognize both swimming and climbing as escape behaviors was necessary for the assay to detect the antidepressant potential of the SSRIs [3]. The assay is quick and relatively easy to use, reliable across laboratories and is able to detect a broad range of antidepressants including the dissociative anesthetic, ketamine. There are distinct differences in the rat and mouse versions of the test, it is quite sensitive to the choice of mouse strain and compounds such as amphetamine that increase locomotion confound the interpretation of the results [14]. The major limitations of the test are that the state of behavioral despair is not sustained and unlike depressed patients, the animals respond to acute treatment. A conceptually similar assay,

the mouse tail suspension test that also measures the duration of escape-directed behavior is becoming increasingly popular and both the Forced Swim and Tail Suspension tests have been automated [15]. An open field test to assess locomotion is routinely performed when evaluating compounds in either of these tests to detect false positives resulting from a general increase in locomotor activity and false negatives resulting from a decrease in locomotor activity. In general, alterations in locomotor activity can confound the interpretation of several of the assays described in this review and routine assessment of locomotor activity can alert the investigator to this possibility as well as help establish the therapeutic index for behavioral testing. Both the FST and TST detect the therapeutic potential of all known classes of

Ac ce p

te

M
5

an

rodent FST which is based on the observation that a rat placed in an inescapable cylinder of water will

us

Most animal models of depression involve an acute or chronic exposure to a stressor to elicit one

cr

ip t
Page 5 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

antidepressants including tricyclics, monoamine oxidase inhibitors, monoamine reuptake inhibitors, atypicals as well as some newer classes such as NMDA receptor inhibitors or mGluR allosteric modulators that are currently in clinical trials [16]. Moreover, neither anxiolytics nor antipsychotics are active in these assays [16]. The choice of models that require chronic administration of antidepressants to obtain efficacy is relatively limited. The most widely used and validated of these are the olfactory bulbectomy and chronic mild stress (CMS) models [15,17]. Both models involve procedures that result in sustained changes in

bulbectomy model, surgical removal of the olfactory bulbs results in the development of locomotor

rat version of this model has been extensively validated with a broad range of antidepressants but it has several caveats, including that antidepressants require several weeks to exhibit efficacy in the clinic but only days in the model, hyperactivity is not a common symptom of depression and the neuroanatomical basis for the response is poorly understood. The CMS model involves the repeated but unpredictable presentation of mild stressors such as temporary food and water deprivation, small temperature changes and housing changes over a period of 3-4 weeks [15,18,19]. This results in the appearance of several behaviors that are considered symptoms of depression, such as sustained decreases in sucrose consumption (a putative measure of anhedonia), grooming, sexual, aggressive and investigative behaviors, all of which can be reversed by chronic, but not acute, treatment with a broad range of antidepressants. The major drawbacks of the CMS model are the time (months) and labor intensive nature of the procedure and the low level of reliability across laboratories. The social defeat stress model also produces sustained behaviors that appear similar to symptoms of depression and these behaviors can be reversed by chronic treatment with antidepressant but like CMS, it is a complex procedure with limited reliability across laboratories. 2.2. Anxiety Animal models of anxiety can generally be classified into three major categories, ethological tests, conflict tests and cognitive-based tests [20]. Ethological tests typically combine a naturally motivated behavior,

Ac ce p

te

M
6

an

hyperactivity that can be reversed by chronic but not acute treatment with antidepressants [15,17]. The

us

behavior that can be reversed by chronic but not acute treatment with antidepressants. In the olfactory

cr

ip t

Page 6 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

such as exploration of the environment with a naturally aversive stimulus such as a brightly lit space to create conflict or anxiety. For example, in the elevated plus maze (EPM), the animal has a natural tendency to spend more time in the two enclosed arms rather than the two elevated open arms with no sides. Treatment with anxiolytic drugs, e.g., the benzodiazepines, increases the number of entries and time spent in the open arms [21]. While this test controls for some potential confounds, such as changes in locomotor activity, it is very sensitive to housing, environmental conditions such as ambient light, noise

zero maze, the light-dark box, the open field, ultrasonic vocalizations and defensive burying [20]. The defensive burying test, which measures the animals tendency to bury an aversive object, such as a shock probe or marbles, is noteworthy because it is the only test in this category that reliably detects the anxiolytic potential of SSRIs.

Conflict tests combine a motivated behavior, such as eating or drinking, with an aversive stimulus like a mild shock. This is typified by the Vogel Punished Drinking assay, in which water deprived rats are given access to water but drinking behavior is punished by a mild shock [23]. Anxiolytics such as benzodiazepines, produce an increase in water consumption. Other tests in this category include the Geller-Seifter test, Four-plate test, Novelty-suppressed feeding and Novelty-induced hypophagia [20]. The Vogel and Geller-Seifter tests are thought to have a low incidence of false positives when assessing anxiolytic potential but are insensitive to more recently discovered classes of anxiolytics such as the SSRIs. In contrast, the Four-plate test does seem to detect the anxiolytic potential of SSRIs. Cognitivebased tests use classical conditioning to train an animal to associate a neutral stimulus, such as a tone, with an unpleasant stimulus to change its natural behavior. For example, in Fear Conditioning, the animal is trained to associate a tone or place with a foot shock or aversive odor and increased levels of freezing behavior or enhanced startle response to the conditioned stimulus are indicative of a heightened level of anxiety [24]. Anxiolytics reduce the level of freezing behavior or startle and the test is sensitive to multiple classes of anxiolytics, including SSRIs. The potential drawbacks of this model are the time required to condition the animals and possible confounding effects of drugs that alter pain sensation, hearing or sense of smell and strain differences [25]. For a comprehensive review of models of anxiety and their sensitivity to compound treatment see [20].

Ac ce p

te

M
7

an

us

cr

and odor as well as different strains of mice [22]. Other popular tests in this category include the elevated

ip t

Page 7 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

2.3. Schizophrenia Animal models of schizophrenia can be fit into three general induction categories: pharmacological, developmental and genetic. Three distinct clusters of symptoms have been identified in schizophrenic patients: positive symptoms such as hallucinations and delusions, negative symptoms such as emotional withdrawal and anhedonia and cognitive dysfunction including impaired working memory and attention. Patients often present with heterogeneous combinations of symptoms making diagnosis, treatment and

or chronic treatment with amphetamine or phencyclidine (PCP) which produce increases in locomotor activity and deficits in PPI of startle [26]. These models are based on the observation that both drugs can produce hallucinations and delusions when administered in man and PCP can produce a sustained relapse in patients with schizophrenia following a single exposure [27]. All currently marketed antipsychotics are active in these models but the general consensus is that they only address the positive

neurodevelopmental disorder that is typically manifest after puberty. Treatment of pregnant rats with

deficits include reduction in the size of neocortical and limbic structures, enhanced locomotor response to

developmental model is post-weaning isolation which alters brain development including a selective reduction in prefrontal cortex volume and produces several behavioral abnormalities including hyperactivity, impaired PPI, cognitive deficits and aggression [27,29]. A third developmental model is the neonatal ventral hippocampal lesion model that is produced by injection of the excitotoxin, ibotinic acid on postnatal day 7. This lesion produces abnormalities in the architecture of the developing prefrontal cortex and nucleus accumbens as well as behavioral abnormalities that emerge after puberty [27,30]. By postnatal day 56, these animals exhibit locomotor hypersensitivity to stress, enhanced sensitivity to dopamine agonists and NMDA antagonists, deficits in PPI, enhanced sensitivity to drugs of abuse and marked impairment in several spatial working memory tasks [27]. The predictive validity of these developmental models remains to be established; moreover they are time consuming, labor intensive and costly. The

Ac ce p

amphetamine, increased dopamine release in the nucleus accumbens and impaired PPI [27,28]. Another

te

anatomical and behavioral deficits in the offspring that resemble many aspects of schizophrenia. These

methylazoxymethanol (MAM), a mitotic inhibitor that targets neuroblast proliferation produces long-lasting

M
8

symptoms and have limited effects on the other symptom clusters. Schizophrenia is considered to be a

an

us

cr

modeling extremely difficult. The most successful and widely used models of schizophrenia involve acute

ip t

Page 8 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

MAM model is very sensitive to the timing of MAM administration and the behavioral effects in the social isolation model are relatively fragile and can be reversed by handling. The mortality reported for the neonatal ventral hippocampal lesion model is about 15% and up to 30% exhibit hippocampal damage that fails to meet criteria. Clearly these models are not suitable for routine screening of drug candidates but can be used to explore the pathology and molecular mechanisms underlying this disorder.

There is compelling evidence that schizophrenia is a genetic disorder with heritability around 80% but there is no single genetic mutation that is responsible for this heterogeneous disorder [31,32]. Rather,

primarily involved in neuronal plasticity, dopaminergic or glutamatergic function and synaptogenesis and

genes. Genes of interest include DISC-1 (disrupted-in-schizophrenia 1), NRG1 (neuregulin-1), and Reelin which are all involved in synaptogenesis and synaptic plasticity and dysbindin which is involved in exocytosis and receptor trafficking [27]. Additional studies involving 18 GWAS studies and over 1 million SNPs have implicated a wide variety of genes including a number involved in T-cell related immune function [31]. Transgenic mice with constitutive knockout, inducible knockout, reduced expression or mutations of these genes have been created and each exhibit unique and complex phenotypes that include characteristics associated with schizophrenia [33,34]. Further study is required however before the utility and predictive validity for the treatment of negative and cognitive symptoms of one or more of these models are definitively established. The pressing need for medication to treat the cognitive impairment associated with schizophrenia led to the MATRICS (Measurement and Treatment Research to Improve Cognition in Schizophrenia) initiative, a collaboration between the NIMH, the FDA and academic partners to more accurately measure cognitive deficits associated with schizophrenia and improve the translational fidelity between animals models and human patients. The MATRICS initiative identified seven core domains of cognition that are impaired in schizophrenia: working memory, attention/vigilance, reasoning and problem solving, processing speed, visual learning and memory, verbal learning and memory and social cognition. There are numerous preclinical tests of learning and memory that have been designed to assess each of these domains (see [35]). A major focus of the MATRICS initiative has been the evaluation of the predictive value and clinical relevance of these preclinical

Ac ce p

te

M
9

an

there is a significant effort underway to create transgenic animal models based on manipulation of these

us

several candidate genes have been associated with increased risk of schizophrenia. These genes are

cr

ip t

Page 9 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

cognitive tests and there are several excellent reviews on this topic [36,37,38]. This is an ongoing effort that will no doubt affect the development of animal models of schizophrenia for the foreseeable future. Many recently completed clinical studies of cognitive impairment associated with schizophrenia followed the MATRICS guidelines but in general were underpowered or too short to detect modest effects. Larger ongoing studies that are following these guidelines are more likely to provide useful insight into the predictive validity of the recommended preclinical tests [39,40]. 3. Neurodegenerative diseases 3.1. Alzheimers Disease

Over the last 15 years, there has been a tremendous effort focused on the development and

prevalence of this disease based on the rapid growth and increased longevity of an aging population, and the recognition that there are presently no disease-modifying therapies for this disease. Moreover, progress in human genetics has identified multiple genes linked to specific forms of the disease, at last count in excess of 130 [41].

Animal models of AD fall into 3 general categories, pharmacological, lesioned and transgenic. The first two categories are based on the observation that cholinergic transmission is impaired in AD and

pharmacological model is the scopolamine-induced amnesia model which uses blockade of cholinergic receptors to produce cognitive impairment [42,43]. Lesion models include: surgical procedures, e.g., transection of the fimbria/fornix leading to cholinergic denervation of the hippocampus; electrolytic lesioning of discrete cholinergic nuclei, such as the nucleus basalis magnocellularis or the septal nuclei, or microinjection of neurotoxins such as quinolinic, acid, ibotinic acid or the cholinotoxin, AF64 [44]. Lesion models are notoriously susceptible to variability due in part to strain differences, location and extent of the lesion, choice and concentration of neurotoxin and even housing conditions. While these two categories of models effectively mimic some of the cognitive deficits associated with AD and have been instrumental in evaluating the symptomatic and limited efficacy of currently marketed medications such as the cholinesterase inhibitors, they do not approximate the complex molecular and cellular

Ac ce p

cholinergic neurons preferentially degenerate as the disease progresses. The most commonly used

te

M
10

an

characterization of animal models for AD. This has been fueled by the anticipated tidal wave in

us

cr
Page 10 of 29

ip t

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

pathology that is characteristic of this disease [45,46]. Consequently, recent efforts have been devoted primarily to the development of transgenic models of AD. Mutations in three genes, amyloid precursor protein (APP), presenilin-1 (PSEN1) and presenilin-2 (PSEN2) cause autosomal dominant AD and mutated forms of these genes are the basis of the majority of current transgenic models. These autosomal dominant forms make up only a small fraction of AD cases, but their pathology and symptoms are thought to be similar to the more prevalent sporadic form of AD. Mutations in each of these genes alter the processing of amyloid (A) by APP and result in

gene and are named for the geographic location of their discovery. Thus, the Swedish mutation

Arctic mutation increases fibrillogenesis. The most widely used mouse models of AD involve transgenic expression of mutated human APP [47]. Several APP lines exist and they all develop amyloid pathology, exhibit synaptic toxicity and memory deficits but do not exhibit degeneration or loss of neurons. The lines differ in terms of the promoters used, which drive levels and spatial patterns of expression, the isoforms (APP695, APP751, or APP770), the mutation or combination of mutations which influences the severity and onset of the phenotype and the background strain which can modulate the phenotype [47]. One of the earliest transgenic lines, the Tg2576 as well as most currently used mouse lines use the Swedish mutation [48]. Transgenic animals incorporating mutated forms of presenilin-1 have also been created. Mice that only have a mutated form of PSEN-1, such as the PS1 line, produce increased levels of A42 but do not develop AD pathology or cognitive deficits. This is most likely due to key amino acid differences in mouse and human APP in the A region. Consequently, this PS1 line was crossed with a hAPP line to produce the PSAPP line which develops A plaques earlier than the Tg2576 line, has increased A42 levels and exhibits cognitive deficits [49]. To produce a line with even more rapidly progressing pathology, the 5XFAD line was created by combining 3 mutations in hAPP and 2 mutations in PS1. This line expresses high levels of A42, exhibits amyloid pathology and cognitive deficits by 4 months and unlike most other lines it also exhibits neuronal loss [50]. It should be noted that aged

Ac ce p

te

M
11

an

increases Aproduction, the London and Indiana mutations increase the proportion of A42 and the

us

increased levels of the toxic A42 form [47]. Several distinct mutations have been identified in the APP

cr

ip t

Page 11 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

rodents and primates both exhibit accumulation of A, cognitive impairment and development of neurodegeneration but are technically challenging to use on a routine basis [51]. None of the models described so far produce the neurofibrillary tau pathology that is characteristic of AD. No mutations of tau have been directly linked to AD but some have been shown to cause frontotemporal dementia (FTD). To produce lines that exhibit both the plaque and neurofibrillary tangle pathology of AD, lines containing mutant tau have been crossed with lines containing mutant hAPP and/or PS1. The most notable of these is the 3xTg line which combines hAPP with the Swedish mutation

just as observed in humans [52]. It is anticipated that lines of transgenic rodents will be further refined in

a transgenic rat line has been produced using hAPP with the Swedish mutation, and PS1 with the delta 9 mutation in the Fischer 344 strain. The animals exhibit both amyloid plaque and tau neurofibrillary tangle pathology, cognitive deficits, neurodegeneration and neuronal loss [12]. This model seems to have a high level of fidelity with regard to current concepts of AD pathology and will provide the opportunity to evaluate the behavioral phenotype using some of the more robust rat tests for cognition and mood. One important caveat is that although mild cognitive deficits are detectable at 6 months in this model, robust pathology and cognitive deficits are not observed until 15 months [34]. Despite considerable progress in the development of transgenic models of AD, none of the disease-modifying therapeutics that exhibited apparent efficacy in one or more of these models has produced significant improvement in clinical trials. The disappointing clinical results with vaccination against A, humanized anti-A antibodies and gammasecretase inhibitors have led to legitimate concerns about the predictive utility of the transgenic models [41, 51]. It would seem to be premature however, to conclude that these models have no utility for the identification and evaluation of potential therapeutics given the complex nature of AD, the number of available transgenic models and the technical challenges associated with clinical trials for this indication. This is clearly a rapidly evolving area and it is likely that clarity regarding the choice and utility of animal models for therapeutic development will take a while to establish. 3.2. Huntingtons Disease

Ac ce p

te

M
12

an

an effort to more faithfully recapitulate the pathology and behavioral phenotype of AD. Along these lines,

us

with single mutation forms of tau and PS1. This line develops plaque pathology before tangle pathology

cr

ip t

Page 12 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

Huntingtons Disease (HD) is an autosomal dominant neurodegenerative disorder that is associated with numerous motor, cognitive, behavioral and psychological impairments. The mutation consists of an expansion within the poly-CAG repeat region in exon 1 of the huntingtin gene that results in production of aberrant huntingtin protein with an expanded stretch of polyglutamine near the N-terminus [53,54]. The physiological function of huntingtin remains unknown but a longer stretch of polyglutamine repeats is associated with an earlier age of onset and more severe symptoms. Prior to the identification of the

These neurotoxins destroy discrete populations of neurons, with quinolinic acid lesions exhibiting greater fidelity to the human pathology and resulted in animals with behavioral and neurochemical abnormalities. Quinolinic acid lesions in the monkey have been used to create a primate model that exhibits many of the neuropathological, neurochemical and clinical features of HD [56,57]. The transgenic mouse models of HD can be grouped into three general categories: mice that express the N-terminal fragment of the

repeats inserted into the existing CAG expansion in the murine gene; and mice that express the full-

fragment of huntingtin with approximately 144-150 CAG repeats and exhibits a progressive behavioral

early as 5-6 weeks and survival ranges from 14 -18 weeks [6]. Knock-in lines, such as the HdhQ111, [36,40] exhibit a mild and protracted behavioral and neuropathological phenotype as well as a normal life span. Similarly, the full-length human lines, such as the YAC128, display a gradual development of behavioral and neuropathological abnormalities including motor deficits by 6 -12 months as well as a normal lifespan [36,40]. While the latter two models may more closely mimic the progression of HD, they have more limited utility for evaluating therapeutic interventions. It is important to note that there are significant similarities between the transcriptional profiles of all three categories of transgenic mice and patients with HD when the lines are aged appropriately [7,8]. Moreover, there is some preliminary evidence of predictive validity with reports that tetrabenazine, a VMAT (Vesicular monoamine transporter) -inhibitor approved for the chorea associated with HD reduces motor symptoms in both mice and patients

Ac ce p

and neuropathological phenotype that closely resembles human HD [59]. Motor deficits are observed as

te

model of HD and it is the most thoroughly characterized and widely used. It expresses an N-terminal

length human huntingtin gene along with the murine form [55,58]. The R6/2 line was the first transgenic

M
13

human huntingtin gene containing the polyglutatamine mutations; knock-in mice with additional CAG

an

us

cr

mutation in 1993 [53], HD was modeled by injection of kainic or quinolinic acid into the striatum [55].

ip t

Page 13 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

[60,61]. The current focus of work in this area is likely to be on the evaluation of potential therapeutics in existing transgenic models. 3.3. Parkinsons Disease Parkinsons Disease (PD) is the second most common age-related neurodegenerative disease after AD and is characterized by resting tremor, slowness of movement, postural instability and freezing. It is caused by a progressive degeneration of dopaminergic neurons that are primarily located in the

substantia nigra pars compacta but the molecular basis of the disorder remains unknown [62,63]. The

involved the central administration the toxin, 6-hydroxydopamine (6-OHDA) into dopaminergic cell or

transporter where it accumulates and produces toxic reactive oxygen species (ROS) that ultimately kill the neuron. The toxin does not cross the blood-brain barrier and unilateral administration results in asymmetric circling behavior that is particularly suitable for the evaluation of therapeutic interventions [62,64,65]. 6-OHDA does not produce the same cellular pathology that is observed in PD and does not appear to work via the same molecular mechanism. The current gold standard animal model of PD is the MPTP primate model [62]. MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) is a neurotoxin that was inadvertently produced during the synthesis of an analog of Demerol for recreational use. Ingestion of the compound by young drug abusers resulted in the production of motor symptoms that were indistinguishable from PD [62]. MPTP has subsequently been shown to be converted to MPP in the brain which is preferentially taken up into dopaminergic neurons by the dopamine transporter (DAT). Once inside the neuron, it blocks the electron transport chain in the mitochondria, decreasing cellular ATP levels and leading to the formation of toxic ROS. In monkeys, there is preferential loss of dopaminergic neurons in the putamen and substantia nigra pars compacta (SNpc) and induction of an irreversible Parkinsonian syndrome that includes all of the features of PD [62]. Despite the remarkable similarity in the behavioral phenotype, the MPTP-treated monkey does not exhibit loss of monoamine neurons in the locus coeruleus or the appearance of Lewy bodies, two characteristic features of PD. It should be noted that MPTP-treated mice also exhibit preferential loss of cells in the SNpc and exhibit motor impairments
+

Ac ce p

te

M
14

an

terminal regions. 6-OHDA is preferentially transported into dopaminergic neurons by the dopamine

us

two categories of PD models are toxin-based models and transgenic mice. The first animal model of PD

cr

ip t

Page 14 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

[62]. Unfortunately, the translational utility of these models for new mechanistic approaches focused on neuroprotection in PD has been disappointing [66]. There are relatively rare forms of PD that have been linked to genetic mutations. The two autosomal dominant genes are -synuclein and leucine rich repeat kinase 2 (LRRK2). -synuclein is an abundant presynaptic phosphoprotein that is a major structural component of Lewy bodies, a hallmark pathologic feature of PD and point mutations or duplications are sufficient to cause PD. Several -

synuclein transgenic lines have been created and their phenotype heavily depends on the choice of

responsive functional abnormalities, there is no progressive loss of dopaminergic neurons. Only

observed in humans [68]. LRRK2 is a large multi-domain containing protein that is localized to membrane structures and mutations linked to PD are concentrated in the GTPase and kinase domains [69]. Transgenic LRRK2 mice have abnormalities in the nigrostriatal system and behavioral deficits that are dopamine-responsive. However, they display a very mild phenotype with minimal evidence of neurodegeneration and are clearly not robust models of PD [70]. There are several autosomal recessive genes that have been linked to PD and the best characterized of these are parkin, DJ-1 and PINK1. Knockout mice have been created for each of these genes but none of them exhibit nigrostriatal pathology, hence their greatest utility is for the exploration of early, pre-neurodegenerative changes that occur in PD [70]. 4. Pain

Chronic pain affects over 25% of the general population and medications to treat it have significant limitations in terms of efficacy and safety [71]. Current animal models, of which there are many [72] have been instrumental in the identification and development of currently used analgesics but have been much less predictive for new generations of potential analgesics acting via non-classical (e.g., opioid and NSAID) mechanisms in terms of both efficacy and dose-limiting side effects. Efforts to develop more robust and predictive models face numerous challenges. Pain is a subjective experience that is heterogeneous in nature and involves sensory, emotional and cognitive components as well as multiple

Ac ce p

te

M
15

an

transgenic mice with the prion promotor (mPrP) exhibit the full range of -synuclein pathology that is

us

promotor [67,68]. None of the models accurately represent PD. While there are some dopamine-

cr

ip t

Page 15 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

sensory modalities which makes interpretation of an animals behavioral response to pain particularly difficult. There are acute pain models as well as chronic models of arthritis, cancer and neuropathyrelated pain. Tests that are most commonly used to evaluate different sensory modalities of pain include the von Frey and Randal-Sellito tests for mechanical sensation; tail flick, hot plate and Hargreaves tests for heat sensation; and the acetone, cold plate and cold water tests for cold sensation (see [73]). Acute models of pain include subcutaneous injection of formalin into the plantar tissue of the paw, i.p.

general, these models are sensitive to analgesics of different classes. In recent years, the major focus in this field has been on the development and improvement in the assessment of models of chronic pain [72]. The two most commonly used models of neuropathic pain are the chronic constriction injury (CCI) and spinal nerve ligation (SNL) models [75,76]. The CCI model utilizes loosely constrictive ligatures around the sciatic nerve at mid-thigh level to produce long-lasting changes in posture, gait and

dorsal root ganglia to produce increased sensitivity to noxious heat and mechanical stimuli as well as

important to note that surgical skill, variations in procedures and differences in genetic strains can all

The most widely used chemical-induced model of pain is the paclitaxel-induced model of chemotherapy-induced neuropathy which, unlike earlier chemotherapeutic agents, produces a pain syndrome without systemic toxicity [80]. Repeated administration of a low dose of paclitaxel over alternate days produces a neuropathy characterized by cold and mechanical allodynia but no heat hyperalgesia [80]. The most common model of diabetic neuropathy-induced pain involves administration of streptozotocin that selectively destroys pancreatic cells and produces hyperalgesia within 2-3 weeks [75,81]. Unfortunately, these animals develop other metabolic and physical abnormalities consistent with the induction of a diabetic state including ketosis, reduced growth and motor activity, and lethargy which complicate their use in measuring nociception. Genetic models of diabetes such as the Goto-Kakizaki

Ac ce p

influence the performance of these models [74].

te

mechanical hypersensitivity whereas the CCI model was associated with more behavioral signs [79]. It is

licking of the affected paw [78]. When directly compared, the SNL model exhibited more pronounced

M
16

spontaneous paw-lifting [77]. The SNL involves tight ligation of spinal nerves L5 and L6 distal to the

an

us

cr

administration of an irritant such as acetone and surgical incisions in multiple parts of the body [74]. In

ip t

Page 16 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

and Zucker Diabetic Fatty rats and ob/ob mice all exhibit symptoms of diabetes and develop progressive peripheral neuropathies (see [75]). The most common models of cancer pain involve injection of sarcoma or carcinoma cells into various bones such as the femur, calcaneus and humeral bones where they induce bone destruction leading to spontaneous and evoked flinching [82,83,84]. Similarly, models of arthritis pain involve the injection of toxic substances directly into the knee or ankle joint to produce a local inflammation. For

example, injection of monosodium acetate leads to sustained hypersensitivity that is sensitive to NSAIDs

recently been questioned following numerous failures in clinical trials of compounds that were robustly

several clinical trials despite a large body of positive data in animal models [86]. On the other hand, the synthetic -conotoxin, ziconotide, is an example of an analgesic working by a novel mechanism whose efficacy was correctly predicted by the CCI, SNL and formalin models [72]. Moreover, a comparison of pharmacodynamic and pharmacokinetic data from clinical and preclinical studies revealed that the SNL model was relatively predictive for celecoxib, indomethacin, gabapentin, lamotrigine and carbamazepine but not the classical tricyclic antidepressants, e.g. amitriptyline, which are effective in neuropathic pain [87]. Based on concerns that recent failures in clinical trials may be related to the observation that preclinical tests measure evoked rather than the more relevant spontaneous aspects of human pain, several laboratories have developed models which use conditioned place preference or selfadministration to more accurately assess the more subjective components of pain [72,88,89]. The predictive validity of these models remains to be determined. 5. Stroke

Perhaps the most challenging neurological disorder to model is stroke, due to the enormous variability in both the causes and consequences and the extremely limited ability to perform well controlled clinical trials of therapeutics with a homogeneous group of subjects that typically present long after the cerebral trauma . The most commonly used models of stroke involve occlusion of the middle cerebral artery (MCA). This is done by either ligating or placing a clip on the MCA which can be loosened

Ac ce p

te

M
17

an

active in a variety of preclinical models. Of particular note is the clinical failure of NK-1 antagonists in

us

[85]. The predictive validity of several of these models, particularly the neuropathic pain models, has

cr

ip t

Page 17 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

after a set period of time to make the occlusion transient rather than permanent [90]. The disadvantage of both of these approaches is that they involve opening the skull and require significant surgical skill [91]. Alternatively, stroke may be produced by a procedure termed thread occlusion in which a thread or balloon catheter is inserted into a large peripheral artery and advanced to the origin of the MCA [92]. These methods do not require a craniotomy and the thread or catheter can be removed at any time to permit controlled reperfusion but they also require considerable surgical skill. Unfortunately, the vessel

from thromboembolism [91,93]. To address this limitation, embolic models have been developed, in which a suspension of small clot fragments is injected into the common carotid artery [94]. Acute mortality associated with this approach is low but, not surprisingly, the foci of infarction are widely distributed and mortality can be as high as 30-40% within 24 hours [91]. There have been numerous clinical trials with compounds that were efficacious in one or more of these models but to date only tPA

neuroprotective activity in preclinical stoke models (average of 25%) have advanced to clinical trials with

certainly possible that greater than 25% neuroprotection is required to obtain efficacy in stroke patients

of minimal relevance to the real world situation. 6. Conclusion

Several excellent review articles describing the merits and limitations of each of the models discussed as well as descriptions of additional models that are beyond the scope of this review are noted in Table 2. For a comprehensive review of the use of animal models for CNS drug discovery, see [96]. There has been an enormous investment of time and resources in the development of these models and while they have significantly advanced understanding and treatment of CNS disorders they have significant limitations and the field is constantly evolving with refinements of existing models and development of new ones.

Ac ce p

and that administration of candidates before or concomitantly with the stroke insult in preclinical studies is

te

the clinical trials, but in either case, the predictive validity of these models remains to be determined. It is

no evidence of significant efficacy [95]. It is still not clear whether this is due to limitations of the models or

M
18

(tissue plasminogen activator) is approved for the treatment of stroke. Some 114 compounds with

an

us

cr

occlusion approaches do not model the actual mechanism of occlusion since most large infarcts result

ip t

Page 18 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

References [1] Nestler EJ, Hyman SE. Animal models of neuropsychiatric disorders. Nat Neurosci. 2010; 13:11611169. [2] Porsolt RD, LePichon M, Jalfre M. Depression: a new animal model sensitive to antidepressant treatment. Nature. 1977; 266:730-732. [3] Lucki I. The forced swimming test as a model for core and component behavioral effects of antidepressant drugs. Behav. Pharmacol. 1997; 8:523-532.

[7] Kuhn, A., Goldstein, D.R., Hodges, A., Strand, A.D., Sengstag, T., Kooperberg, C., Becanovic, K., Pouladi, M.A., Sathasivam, K., Cha, J.-H.J., et al. Mutant huntingtins effects on striatal gene expression in mice recapitulate changes observed in h uman Huntingtons disease brain and do not differ with mutant huntingtin length or wild-type huntingtin dosage. Hum. Mol. Genet. 2007; 16, 18451861. [8] Strand, A.D., Baquet, Z.C., Aragaki, A.K., Holmans, P., Yang, L., Cleren, C., Beal, M.F., Jones, L., Kooperberg, C., Olson, J.M., and Jones, K.R. Expression profiling of Huntingtons disease models suggests that brain-derived neurotrophic factor depletion plays a major role in striatal degeneration. J. Neurosci. 2007: 27, 1175811768. [9] Fernando ABP, Robbins TW. Animal models of neuropsychiatric disorders. Ann Rev Clin Psychol. 2011; 7:39-61. [10] Zhou H, Huang C, Chen H, Wang D, Landel CP, Xia PY, Bowser R, Liu YJ, Xia XG. Transgenic rat model of neurodegeneration caused by mutation in the TDP gene. PLoS Genet. 2010; 6:e1000887. [11] von Horsten S, Schmitt I, Nguyen HP, Holzmann C, Schmidt T, et al. Transgenic rat model of Huntingtons disease. Hum Mol Genet. 2003; 12:617-624. [12] Cohen RM, Rezai-Zadeh K, Weitz TM, Rentsendorj A, Gate D, Spivak I, Bholat Y, Vasilevko V, Glabe CG, Breunig JJ, Rakic P, Davtyan H, Agadjanyan MG, Kepe V, Barrio JR, Bannykh S, Szekely CA, Pechnick RN, Town T. A Transgenic Alzheimer Rat with Plaques, Tau Pathology, Behavioral Impairment, Oligomeric A, and Frank Neuronal Loss. J Neurosci. 2013; 33:6245-6256. [13] Cryan JF, Markou A, Lucki I. Assessing antidepressant activity in rodents: recent developments and future needs. Trends Pharmacol Sci. 2002; 23(5):238-45. [14] Lucki, I. A prescription to resist proscriptions for murine models of depression. Psychopharm. 2001; 153:395-398.

Ac ce p

te

M
19

an

[6] Mangiarini, L., Sathasivam, K., Seller, M., Cozens, B., Harper, A., Hetherington, C., Lawton, M., Trottier, Y., Lehrach, H., Davies, S.W., and Bates, G.P. Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell 1996; 87, 493506.

us

[5] Menalled L, Cesselet M-F. Mouse models of Huntingtons disease. Trends in Pharmacol Sci. 2002; 23:32-39.

cr

[4] Powell SB, Zhou X, Geyer MA. Prepulse inhibition and genetic mouse models of schizophrenia. Behav Brain Res. 2009; 204:282-94.

ip t

Page 19 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

[15] Cryan JF, Mombereau C. In search of a depressed mouse: utility of models for studying depressionrelated behavior in genetically modified mice. Mol Psychiatry. 2004; 9:326-57. [16] Cryan JF, Holmes A. The ascent of mouse: advances in modelling human depression and anxiety. Nat Rev Drug Disc 2005; 4:775-790. [17] Kelly JP, Wrynn AS, Leonard BE. The olfactrory bulbectomized rat as a model of depression: an update. Pharmacol Ther. 1997; 74:299-316. [18] Willner P. Validity, reliability and utility of the chronic mild stress model of depression: a 10-year review and evaluation. Psychopharm. 1997; 134:319-29.

[19] Katz RJ. Animal model of depression: pharmacological sensitivity of a hedonic deficit. 1982; 16:965968.

[21] Rodgers RJ. Animal models of anxiety: where next? Behav Pharmacol. 1997; 8:477496. [22] Griebel G, Belzung C, Perrault G, Sanger DJ. Differences in anxiety-related behaviours and in sensitivity to diazepam in inbred and outbred strains of mice. Psychopharmacology 2000; 148: 164 170. [23] Rodgers R. Animal tests for anxiety. In: Koob GF, Le Moal M, Thompson RF (eds). Encyclopedia of Behavioral Neuroscience. Academic Press: Oxford, 2010; pp. 90100. [24] Borsini F, Podhorna J, Marazziti D. Do animal models of anxiety predict anxiolytic-like effects of antidepressants? Psychopharmacology (Berl), 2002; 163:121 141.

[27] Jones CA, Watson DJ, Fone KC. Animal models of schizophrenia. Br J Pharmacol. 2011; 164(4):1162-94 [28] Moore H, Jentsch JD, Ghajarnia M, Geyer MA, Grace AA. A neurobehavioral systems analysis of adult rats exposed to methylazoxymethanol acetate on E17: implications for the neuropathology of schizophrenia. Biol Psychiatry 2006; 60: 253 264. [29] Fone KCF, Porkess MV. Behavioural and neurochemical effects of post-weaning social isolation in rodents relevance to developmental neuropsychiatric disorders. Neurosci Biobehav Rev. 2008; 32:10871102. [30] Lipska BK, Weinberger DR (1993). Delayed-effects of neonatal hippocampal damage on haloperidolinduced catalepsy and apomorphine-induced stereotypic behaviours in the rat. Brain Res Dev Brain Res 1993; 75: 213222. [31] Aberg KA, Liu Y, Bukszr J, et al. A Comprehensive Family-Based Replication Study of Schizophrenia Genes. JAMA Psychiatry, 2013; 2013:1-9.

Ac ce p

[26] Mouri A, Noda Y, Enomoto T, Nabeshima T. Phencyclidine animal models of schizophrenia: approaches from abnormality of glutamatergic neurotransmission and neurodevelopment. Neurochem Int. 2007; 51:173184.

te

[25] Balogh SA, Wehner JM. Inbred mouse strain differences in the establishment of long-term fear memory. Behav Brain Res. 2003; 140(1-2):97-106.

M
20

an

us

[20] Cryan JF, Sweeney FF. The age of anxiety: role of animal models of anxiolytic action in drug discovery. Br J Pharmacol. 2011; 164:1129-61

cr

ip t
Page 20 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

[32] Sun J, Jia P, Fanous AH, van den Oord E, Chen X, et al. (2010) Schizophrenia Gene Networks and Pathways and Their Applications for Novel Candidate Gene Selection. PLoS ONE 2010; 5:e11351. [33] Jaaro-Peled H, Ayhan Y, Pletnikov M, Sawa A. Review of pathological hallmarks of schizophrenia: comparison of genetic models with patients and nongenetic models. Schizophr Bull 2010; 36: 301 313.

[35] Keeler JF, Robbins TW. Translating cognition from animals to humans. Biochem Pharm. 2011; 81:1356-1366.

[36] Hagan JJ, Jones DNC. Predicting drug efficacy for cognitive deficits in schizophrenia. Schizophr Bull 2005; 31: 830853. [37] Floresco SB, Geyer MA, Gold LH, Grace AA. Developing predictive animal models and establishing a preclinical trials network for assessing treatment effects on cognition in schizophrenia. Schizophr Bull. 2005; 31:888-894. [38] 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 2009; 122:150202. [39] Keefe RS, Buchanan RW, Marder SR, Schooler NR, Dugar A, Zivkov M, Stewart M. Clinical trials of potential cognitive-enhancing drugs in schizophrenia: what have we learned so far? Schizophr Bull. 2013; 39:417-35.

[41] Mullane K, Williams M. Alzheimers Therapeutics: Continued clinical failures question the validity of the amyloid hypothesis but what lies beyond? Biochem. Pharmacol. 2013; 85:289-305. [42] Sunderland T, Tariot PN, Weingartner H, Murphy DL, Newhouse PA, Mueller EA, Cohen RM. Pharmacologic modelling of Alzheimers disease. Prog Neuropsychopharmacol Biol Psychiatry 1986; 10: 599610. [43] Ebert U, Kirch W. Scopolamine models of dementia: electroencephalogram findings and cognitive performance. Eur J Clin Invest. 1998; 28:944-9. [44] Toledana A, lvarez MI. Lesion-induced vertebrate models of Alzheimer dementia. In: De Deyn PP, Van Dam D (eds). Animal Models of Dementia. Springer Science + Business Media: New York, 2010; pp. 295345. [45] Birks J. Cholinesterase inhibitors for Alzheimers disease. Cochrane Database 1391 System Rev 2006;1:CD005593. [46] Van Dam D, De Deyn PP. Animal models in the drug discovery pipeline for Alzheimer's disease. Br J Pharmacol. 2011; 164:1285-300. [47] Hall AM, Roberson ED. Mouse models of Alzheimer's disease. Brain Res Bull. 2012; 88:3-12. [48] Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S, Yang F, Cole G. Correlative memory deficits, A elevation, and amyloid plaques in transgenic mice. Science 1996; 274: 99 102.

Ac ce p

te

[40] Kirkpatrick B, Fenton WS, Carpenter WT Jr, Marder SR. The NIMH-MATRICS consensus statement on negative symptoms. Schizophr Bull. 2006; 32:214-9.

M
21

an

us

cr

ip t
Page 21 of 29

[34] OTuathaigh C, Kirby B, Moran P, Waddington J. Mutant mouse models: genotypephenotype relationships to negative symptoms in schizophrenia. Schizophr Bull 2010; 36: 271288.

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

[49] Holcomb L, Gordon MN, McGowan E, Yu X, Benkovic S, Jantzen P, Wright K, Saad I, Mueller R, Morgan D, Sanders S, Zehr C, OCampo K, Hardy J, Prada CM, Eckman C, Younkin S, Hsiao K, Duff K. Accelerated Alzheimer-type phenotype in transgenic mice carrying both mutant amyloid precursor protein and presenilin 1 transgenes. Nat Med. 1998; 4:97100. [50] Oakley H, Cole SL, Logan S, Maus E, Shao P, Craft J, Guillozet-Bobgaarts A, Ohno M, Disterhoft J, Van Eldik L, Berry R, Vassar. Intraneuronal beta-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimers disease mutations: potential factors in amyloid plaque formation. J Neurosci. 2006; 26:10129-40. [51] Sabbagh JJ, Kinney JW, Cummings JL. Animal systems in the development of treatments for Alzheimer's disease: challenges, methods, and implications. Neurobiol Aging. 2013; 34:169-83.

[53] Huntington's Disease Collaborative Research Group. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington's disease chromosomes, Cell 1993; 72:971983. [54] Zuccato C, Valenza M, Cattaneo C. Molecular mechanisms and potential therapeutic targets in Huntington's disease. Physiol Rev 2010; 90:905981.

[59] Menalled L, El-Khodor BF, Patry M, Surez-Farias M, Orenstein SJ, Zahasky B, Leahy C, Wheeler V, Yang XW, MacDonald M, Morton AJ, Bates G, Leeds J, Park L, Howland D, Signer E, Tobin A, Brunner D. Systematic behavioral evaluation of Huntingtons disease transgenic and knock -in mouse models. Neurobiol. Dis. 2009; 35, 319336. [60] Frank, S.; Huntington Study Group/TETRA-HD Investigators. Tetrabenazine as anti-chorea therapy in Huntington disease: An open-label continuation study. BMC Neurol. 2009; 9, 62. [61] Wang, H., Chen, X., Li, Y., Tang, T.-S., and Bezprozvanny, I. Tetrabenazine is neuroprotective in Huntingtons disease mice. Mol. Neurodegener. 2010; 5, 18. [62] Dauer W, Przedborski S. Parkinsons disease: mechanisms and models. Neuron. 2003; 39:889 -909. [63] Blesa J, Phani S, Jackson-Lewis V, Przedborski S. Classic and new animal models of Parkinson's disease. J Biomed Biotechnol. 2012; 2012:845618. [64] Tolwani RJ, Jakowec MW, Petzinger GM, Green S, Waggie K. Experimental models of Parkinsons disease: insights from many models. Lab Anim Sci. 1999; 49:363-371.

Ac ce p

[58] Crook ZR, Housman D. Huntingtons Disease: Can mice lead the way to treatment. Neuron. 2011; 69: 423-435.

te

[57] Beal MF, Ferrante RJ, Swartz KJ, Kowal NW. Chronic quinolinic acid lesions in rats closely resemble Huntingtons disease. J. Neurosci. 11:1649-1659.

[56] Ferrante, RJ, Kowall NW, Cipolloni PB, Storey E, Beal MF. Excitotoxin lesions in primates as a model for Huntingtons disease: histopathologic and neurochemical characterization. Exp Neurol. 1993; 119:46-71.

M
22

[55] Ferrante RJ. Mouse models of Huntington's disease and methodological considerations for therapeutic trials. Biochim Biophys Acta. 2009; 1792:506-20.

an

us

[52] Oddo S, Caccamo A, Shepherd JD, Murphy MP, Golde TE, Kayed R, Metherate R, Mattson MP, Akbari Y, LaFerla FM. Triple-transgenic model of Alzheimers disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron 2003; 39: 409 421.

cr

ip t

Page 22 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

[65] Duty S, Jenner P. Animal models of Parkinson's disease: a source of novel treatments and clues to the cause of the disease. Br J Pharmacol. 2011; 164:1357-91. [66] Waldmeier P, Bozyczko-Coyne D, Williams M, Vaught JL. Recent clinical failures in Parkinson's disease with apoptosis inhibitors underline the need for a paradigm shift in drug discovery for neurodegenerative diseases. Biochem Pharmacol. 2006; 72:11971206.

[68] Chesselet MF, Fleming S, Mortazavi F, Meurers B. Strengths and limitations of genetic mouse models of Parkinsons disease. Parkinsonism Relat Disord. 2008; 14:S84-S87.

[70] Dawson TM, Ko HS, Dawson VL. Genetic animal models of Parkinson's disease. Neuron. 2010; 66(5):646-61 [71] Johannes CB, Le TK, Zhou X, Johnston JA, Dworkin RH. The prevalence of chronic pain in United States adults: results of an internet-based survey. J Pain. 2010; 11:12301239. [72] Mogil JS. Animal models of pain: progress and challenges. Nature Rev Neurosci. 2009; 10:283-294. [73] Sandkuhler J. Models and Mechanisms of Hyperalgesia and Allodynia. Physiol Rev. 2009; 89:707758.

[75] Jaggi AS, Jain V, Singh, N. Animal models of neuropathic pain. Fund. Clin. Pharm. 2011; 25:1-28.

[77] Bennett GJ, Xie YK. A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain. 1988; 33:87107. [78] Kim SH, Chung JM. An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat. Pain. 1992; 50:355 363. [79] Kim KJ, Yoon YW, Chung JM. Comparison of three rodent neuropathic pain models. Exp Brain Res. 1997; 113:200206. [80] Polomanoa RC, Mannes AJ, Clark US, Bennett GJ. A painful peripheral neuropathy in the rat produced by the chemotherapeutic drug, paclitaxel. Pain. 2001; 94:293-304. [81] Courteix C, Eschalier A, Lavarenne J. Streptozocin-induced diabetic rats: behavioural evidence for a model of chronic pain. Pain 1993; 53:81-88. [82] Schwei MJ, Honore P, Rogers SD, Salak-Johnson JL, Finke MP, Ramnaraine ML, Clohisy DR, Mantyh PW. Neurochemical and cellular reorganization of the spinal cord in a murine model of bone cancer pain. J. Neurosci. 1999; 19:10886-10897.

Ac ce p

[76] Dowdall T, Robinson I, Meert TF. Comparison of five different rat models of peripheral nerve injury. Pharmacol Biochem Behav. 2005; 80:93-108.

te

[74] Berge O-G. Predictive validity of behavioral animal models for chronic pain. Brit. J Pharm. 2011; 164:1195-1206.

M
23

an

us

[69] Biskup S, Moore DJ, Celsi F, Higashi S, West AB, Andrabi SA, Kurkinen K, Yu SW, Savitt JM, Waldvogel HJ, Faull RL, Emson PC, Torp R. Otterson OP, Dawson TM, Dawson VL. Localization of LRRK2 to membranous and vesicular structures in mammalian brain. Ann Neurol. 2006; 60:557-569.

cr

ip t

[67] Lee Y, Dawson VL, Dawson TM. Animal models of Parkinson's disease: vertebrate genetics. Cold Spring Harb Perspect Med. 2012; 2:1-12

Page 23 of 29

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

[83] Bove SE, Flatters SJ, Inglis JJ, Mantyh PW. New advances in musculoskeletal pain. Brain Res Rev 2009; 60:187-201. [84] Pacharinsak C, Beitz A. Animal Models of Cancer Pain. Comp Med. 2008; 58:220-233. [85] Kalbhen DA. Chemical model of osteoarthritis a pharmacological evaluation. J Rheumatol 1987; 14:130131. [86] Hill R. NK1 (substance P) receptor antagonists why are they not analgesic in humans? Trends Pharmacol Sci 2000; 21:244246. [87] Whiteside GT, Adedoyin A, Leventhal L. Predictive validity of animal pain models? A comparison of the pharmacokinetic-pharmacodynamic relationship for pain drugs in rats and humans. Neuropharmacol 2008; 54:767775.

[90] Crowell RM, Marcoux FW, DeGirolami U. Variability and reversibility of focal ischemia in unanesthetized monkeys. Neurology. 1981; 31:1295-1302. [91] Howells DW, Poritt MJ, Rewell SSJ, OCollins V, Sena ES, van der Worp HB, Traystman RJ, MacLeod MR. Different strokes for different folks: the rich diversity of animal models of focal cerebral ischemia. J Cereb Blood Flow and Metab. 2010; 30:1412-1431. [92] Koizumi J, Yoshida Y, Nakazawa T, Ooneda G. Experimental studies of ischemic brain edema. 1. A new experimental model of experimental embolism in rats in which recirculation can be reintroduced in the ischemic area. Jpn J Stroke. 1986; 8:1-8.

[94] Kudo M, Aoyama A, Ichimori S, Fukunaga N. An animal model of cerebral infarction. Homologous blood clot emboli in rats. Stroke. 1982; 13:505-508. [95] O'Collins VE, Macleod MR, Donnan GA, Horky LL, van der Worp BH, Howells DW. 1,026 experimental treatments in acute stroke. Ann Neurol. 2006; 59::467-77. [96] Borsini and MacArthur RA, Borsini F. Animal and Translational Models for CNS Drug Discovery. 2008; Elsevier Inc. [97] Yan HC, Cao X, Das M, Zhu XH, Gao TM. Behavioral animal models of depression. Neurosci Bull. 2010; 26(4):327-37. [98] Cryan JF, Mombereau C, Vassout A. The tail suspension test as a model for assessing antidepressant activity: Review of pharmacological and genetic studies in mice. Neurosci Behav Rev. 2005; 29:571-625.

Ac ce p

[93] Donnan GA, Fisher M, MacLeod M, Davis SM. Stroke. Lancet. 2008; 371:1612-1623.

te

M
24

an

[89] King T, Vera-Portocarrero L, Gutierrez T, Vanderah TW, Dussor G, Lai J, Fields HL, Porreca F. Unmasking the tonic-aversive state in neuropathic pain. Nat. Neurosci. 2009; 12:1364-1366.

us

[88] Martin TJ, Kim, SA, Buechler NL, Porreca F, Eisenach JC. Opioid self-administration in the nerveinjured rat: Relevance of antiallodynic effects to drug consumption and effects of intrathecal analgesics. Anesthesiol. 2007; 106:312322.

cr

ip t

Page 24 of 29

McGonigle CNS Models - Tables-Revised 061713.docx

Ac

ce pt

ed

M an

All Models Sex Strain Age Housing Handling Environment Confounding Behavior

Transgenic Models Gene construct Penetrance Constitutive/Inducible Choice of Promotor Pathological Phenotype Behavioral Phenotype Rare or common mutation

Lesion Models Surgical skill Choice of toxin Dose and speed of delivery Extent of damage Recovery environment & Time Infections Post-op anesthetic

us

cr

Table 1. Critical Factors Affecting Variability and Reproducibility

ip t

Page 25 of 29

Disorder Depression

Category Acute

Chronic

Chronic Mild Stress, Olfactory Bulbectomy, Social Defeat

Anxiety

Ethological

Conflict

ce pt

Elevated Plus Maze, Light/Dark Box, Open Field, Defensive Burying

Cognitive

Vogel Punished Drinking, GellerSeifter, 4-Plate, Novelty Suppressed Feeding Fear Conditioning, Fear Potentiated Startle

Ac

Acute stress produces form of behavioral despair Chronic stress produces long lasting behavioral changes that resemble symptoms of depression Conflict between naturally occurring behavior and naturally aversive situation Conflict between motivated behavior and aversive stimulus

us

Representative Tests Forced Swim Tail Suspension

Rationale

Strengths Fast, reproducible, widely used

cr

Table 2. Summary of animal models of CNS Disorders

ip t
Multiple symptoms that can be reversed by chronic but not acute drug treatment Predictive Validity Acute effect not Good for SSRIs, consistent with SNRIs, TCAs and delayed onset of Ketamine ADs in clinic Time consumGood for SSRIs, ing, labor inten- SNRIs, TCAs, ECS sive, costly, and Ketamine limited reproducibility across labs Weaknesses Reviews [13], [15] [97], [98]

M an

[13], [15] [18], [97]

ed

Fast, reproducible, sensitive, widely used

Very sensitive to environmental factors, strain differences, False positives

Good for benzodiazepines, 5-HT1A agonists, Defensive Burying also good for SSRIs Good for benzodiazepines, 4-plate and NSF detect SSRIs, SNRIs, TCAs Good for benzodiazepines, 5-HT1A agonists

[21]

Fewer false positives

Conditioned to associate neutral stimulus with aversive properties

Assesses the learning component of fear and anxiety

May require operant conditioning, less sensitive to novel mechanisms Time consuming, requires conditioning

[20], [23]

[9], [20] [24]

Page 26 of 29

Genetic

M an

Developmental

MAM treatment, Post-weaning Isolation, Ventral Hippocampal Lesion DISC1, NRG1, Dysbindin

us

cr

Schizophrenia Pharmacological PCP, Amphetamine

Compounds produce psychotic symptoms in man

Alzheimers Disease

Pharmacological Scopolamine

Lesion

Transgenics

Ac

Transection of Fimbria Fornix, Electrolytic or chemical lesion of cholinergic nuclei Tg2576, hAPP, PSAPP, 5XFAD, 3XTg, PSAPP rat

Fast, reproducible, reliable across labs, good for positive symptoms Produce Mimics time developmental course and some abnormalities developmental and post-puberty aspects of deficits disorder Based on genetic Produce distinct linkage, neurological and associated with behavioral increased phenotype susceptibility Based on Produces degeneration of cognitive cholinergic impairment, system in AD mimics degeneration of cholinergic neurons Based on Produces Degeneration of cognitive cholinergic impairment and system in AD degeneration of cholinergic neurons Genetic Recapitulate mutations linked multiple aspects to rare forms of of the pathology, AD that exhibit exhibit cognitive symptoms of deficits, show

ip t
Does not reproduce pathology or progressive nature of disorder

Acute model, no developmental pathology, does not model negative symptoms Time consuming, low throughput, not suitable for screening Mild phenotypes

Good for typical and atypical antipsychotics

[26]

Not sufficiently validated

[27], [29]

Not sufficiently validated

[4], [27] [34]

ed

ce pt

Good for symptomatic therapies such as AchE inhibitors

[43]

Does not reproduce the pathology or progressive nature of the disorder Majority exhibit only subset of pathology, based on rare forms

Good for symptomatic therapies such as AchE inhibitors

[44], [51]

Good for AChE inhibitors, disappointing to date for novel therapies

[46], [47] [51]

Page 27 of 29

Huntingtons Disease

Lesion

Kainic Acid, Quinolinic Acid

Transgenics

R6/2, HdhQ111, YAC128

M an

sporadic AD Mimic loss of discrete populations of neurons in caudate putamen Based on autosomal dominant mutation responsible for disorder

us

progression Produce many behavioral deficits associated with HD

cr

ip t
Do not mimic pathology, progressive nature or reduced survival Most faithful mutations produce mild phenotype, limited progression, normal survival Some aspects of disorder not observed, such as Lewy Bodies and Locus Coeruleus involvement Mild or no phenotypes and no dopaminergic degeneration Dont model more common chronic or pathological conditions Can be technically challenging, Limited validation [56], [57] Not sufficiently validated but tetrabenazine is active in YAC128 mice and patients Good for dopaminergic mechanisms, not for diseasemodifying agents [5], [54] [55], [58] [62], [63] [64], [65] No information [63], [67] [70] Good for several established classes e.g., opiates and NSAIDS Good for established classes of [72], [74] [72], [74] [75], [76]

Transgenics

ce pt
-synuclein, LRRK2, Parkin, DJ1, Pink-2 Formalin Injection, Writhing, Surgical Incision Spinal Nerve Ligation, Chronic Constriction

ed

Parkinsons Disease

Toxins

6-OHDA, MPTP

Based on wellestablished degeneration of dopaminergic neurons

Complex phenotypes, mimic pathology, progressive deficits and reduced survival for R6/2 Robust motor deficits, MPTP produces Parkinsonism in man and primates Reproduce some of the pathology associated with the disorder Fast, reproducible and sensitive to many mechanisms Chronic condition with robust

Pain

Acute

Ac

Based on genetic mutations linked to rare forms of disorder Introduction of irritant or production of tissue damage Based on production of nerve damage

Neuropathic

Page 28 of 29

Cancer

Bone tumors

us

Stroke

Occlusion

Ligature, clips, cauterize

Emboli

Ac

ce pt

ed

Injection of emboli

Mimic focal ischemia by restricting blood flow Mimic the cause of major infarcts

Produces robust symptoms and mimics physiological response Produces ischemic damage to discrete regions Produces significant ischemic damage

cr

Injury, Streptozotocin, Paclitaxel

M an

using surgical, chemical or metabolic approaches Mimic bone metastases to produce pain

symptoms, reproducible across labs

ip t
Difficult to control and reverse Difficult to control, not transient

time consuming, subject to false positives Technically difficult and time consuming

analgesics but several false positives recently Good for established classes, not yet validated for new mechanisms Poor, based on failure of multiple stroke trials Good for tPA, so far not predictive for any other mechanisms [83], [84]

[91]

[91]

Page 29 of 29

Vous aimerez peut-être aussi