Animal Models of CNS Disorders

Embed Size (px)

Citation preview

  • 7/29/2019 Animal Models of CNS Disorders

    1/30

    Accepted Manuscript

    Title: Animal Models of CNS Disorders

    Author: Paul McGonigle

    PII: S0006-2952(13)00387-0DOI: http://dx.doi.org/doi:10.1016/j.bcp.2013.06.016

    Reference: BCP 11671

    To appear in: BCP

    Received date: 18-6-2013

    Accepted date: 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 file 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 final form. Please note that during the production process

    errors may be discovered which could affect the content, and all legal disclaimers thatapply to the journal pertain.

    http://dx.doi.org/doi:10.1016/j.bcp.2013.06.016http://dx.doi.org/10.1016/j.bcp.2013.06.016http://dx.doi.org/10.1016/j.bcp.2013.06.016http://dx.doi.org/doi:10.1016/j.bcp.2013.06.016
  • 7/29/2019 Animal Models of CNS Disorders

    2/30

    pted

    Manuscri

    pt

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    BCP

    Redefining Pharmacology

    Article #7d

    6/16/13

    Animal Models of CNS Disorders

    Paul McGonigle

    Department of Pharmacology and Physiology

    Drexel University College of Medicine

    245 North 15th

    Street, Philadelphia, PA 19102-1192

    Running Title: Animal models of CNS disorders

    Key words:

    Word count (abstract and text): 6578

    98 R f ( d t 2561)

    McGonigle CNS Models FINAL 061713.docx

    http://ees.elsevier.com/bcp/viewRCResults.aspx?pdf=1&docID=14187&rev=0&fileID=312318&msid={B4FC665A-DCEB-4E31-A2A7-2CE9BCFF81ED}
  • 7/29/2019 Animal Models of CNS Disorders

    3/30

    pted

    Manuscri

    pt

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    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

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

    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

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

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

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

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

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

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

  • 7/29/2019 Animal Models of CNS Disorders

    4/30

    pted

    Manuscri

    pt

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    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

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

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

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

    endpoints that are used to describe their symptoms and severity. For example, the DSM IV criteria fordepression 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

  • 7/29/2019 Animal Models of CNS Disorders

    5/30

    pted

    Manuscri

    pt

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    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 validitywhich requires similar symptom manifestation to the clinical

    condition, construct validitywhich requires the model to have similar underlying biology andpredictive

    validitywhich requires responsiveness to clinically effective therapeutic agents.

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

    through established mechanisms assuming the compounds have appropriate pharmacokinetic

    properties. In many cases, these models were established or refined to detect the therapeutic potential of

    prototypical orfirst 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

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

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

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

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

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

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

  • 7/29/2019 Animal Models of CNS Disorders

    6/30

    pted

    Manuscri

    pt

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    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

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

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

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

    eventually adopt an immobile posture after initial attempts to escape [2,13]. This behavior is interpretedas 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

  • 7/29/2019 Animal Models of CNS Disorders

    7/30pted

    Manuscri

    pt

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    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

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

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

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

    rat version of this model has been extensively validated with a broad range of antidepressants but it hasseveral 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

  • 7/29/2019 Animal Models of CNS Disorders

    8/30pted

    Manuscri

    pt

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    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

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

    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 a n 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

  • 7/29/2019 Animal Models of CNS Disorders

    9/30pted

    Manuscri

    pt

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    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

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

    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

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

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

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

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

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

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

  • 7/29/2019 Animal Models of CNS Disorders

    10/30pted

    Man

    uscript

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    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,

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

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

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

    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

  • 7/29/2019 Animal Models of CNS Disorders

    11/30pted

    Man

    uscript

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    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

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

    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

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

  • 7/29/2019 Animal Models of CNS Disorders

    12/30pted

    Man

    uscript

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    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 ofamyloid (A) by APP and result in

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

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

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

    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

  • 7/29/2019 Animal Models of CNS Disorders

    13/30pted

    Man

    uscript

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    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

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

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

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

    a transgenic rat line has been produced using hAPP with the Swedish mutation, and PS1 with the delta 9mutation 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

  • 7/29/2019 Animal Models of CNS Disorders

    14/30pted

    Man

    uscript

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    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

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

    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

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

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

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

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

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

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

  • 7/29/2019 Animal Models of CNS Disorders

    15/30pted

    Man

    uscript

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    [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

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

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

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

    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

  • 7/29/2019 Animal Models of CNS Disorders

    16/30pted

    Man

    uscript

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    [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

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

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

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

    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

  • 7/29/2019 Animal Models of CNS Disorders

    17/30pted

    Man

    uscript

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    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 neuropathy-

    related 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.

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

    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

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

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

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

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

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

    influence the performance of these models [74].

  • 7/29/2019 Animal Models of CNS Disorders

    18/30

  • 7/29/2019 Animal Models of CNS Disorders

    19/30pted

    Man

    uscript

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    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

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

    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

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

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

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

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

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

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

  • 7/29/2019 Animal Models of CNS Disorders

    20/30pted

    Man

    uscript

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    References

    [1] Nestler EJ, Hyman SE. Animal models of neuropsychiatric disorders. Nat Neurosci. 2010; 13:1161-1169.

    [2] Porsolt RD, LePichon M, Jalfre M. Depression: a new animal model sensitive to antidepressanttreatment. Nature. 1977; 266:730-732.

    [3] Lucki I. The forced swimming test as a model for core and component behavioral effects ofantidepressant drugs. Behav. Pharmacol. 1997; 8:523-532.

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

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

    [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 expandedCAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell

    1996; 87, 493506.[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 geneexpression in mice recapitulate changes observed in human Huntingtons disease brain and do notdiffer 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

  • 7/29/2019 Animal Models of CNS Disorders

    21/30pted

    Man

    uscript

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    [15] Cryan JF, Mombereau C. In search of a depressed mouse: utility of models for studying depression-related 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: anupdate. Pharmacol Ther. 1997; 74:299-316.

    [18] Willner P. Validity, reliability and utility of the chronic mild stress model of depression: a 10-yearreview and evaluation. Psychopharm. 1997; 134:319-29.

    [19] Katz RJ. Animal model of depression: pharmacological sensitivity of a hedonic deficit. 1982; 16:965-968.

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

    [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: 164170.

    [23] Rodgers R. Animal tests for anxiety. In: Koob GF, Le Moal M, Thompson RF (eds). Encyclopedia ofBehavioral Neuroscience. Academic Press: Oxford, 2010; pp. 90100.

    [24] Borsini F, Podhorna J, Marazziti D. Do animal models of anxiety predict anxiolytic-like effects ofantidepressants? Psychopharmacology (Berl), 2002; 163:121141.

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

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

    http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/14743184http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/14743184http://www.nature.com/nrd/journal/v4/n9/full/nrd1825.htmlhttp://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/21545412http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/21545412http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed?term=Balogh%20SA%5BAuthor%5D&cauthor=true&cauthor_uid=12644283http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed?term=Wehner%20JM%5BAuthor%5D&cauthor=true&cauthor_uid=12644283http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/12644283http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/12644283http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed?term=Wehner%20JM%5BAuthor%5D&cauthor=true&cauthor_uid=12644283http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed?term=Balogh%20SA%5BAuthor%5D&cauthor=true&cauthor_uid=12644283http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/21545412http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/21545412http://www.nature.com/nrd/journal/v4/n9/full/nrd1825.htmlhttp://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/14743184http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/14743184
  • 7/29/2019 Animal Models of CNS Disorders

    22/30pted

    Man

    uscript

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    [32] Sun J, Jia P, Fanous AH, van den Oord E, Chen X, et al. (2010) Schizophrenia Gene Networks andPathways 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: 301313.

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

    [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 Bull2005; 31: 830853.

    [37] Floresco SB, Geyer MA, Gold LH, Grace AA. Developing predictive animal models and establishing apreclinical trials network for assessing treatment effects on cognition in schizophrenia. SchizophrBull. 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 Ther2009; 122:150202.

    [39] Keefe RS, Buchanan RW, Marder SR, Schooler NR, Dugar A, Zivkov M, Stewart M. Clinical trials ofpotential cognitive-enhancing drugs in schizophrenia: what have we learned so far? Schizophr Bull.2013; 39:417-35.

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

    [41] Mullane K, Williams M.Alzheimers Therapeutics: Continued clinical failures question the validity ofthe 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

    http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/16481659http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/16481659http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/16481659http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/16481659
  • 7/29/2019 Animal Models of CNS Disorders

    23/30pted

    Man

    uscript

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    [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, andneuron loss in transgenic mice with five familial Alzheimers disease mutations: potential factors inamyloid plaque formation. J Neurosci. 2006; 26:10129-40.

    [51] Sabbagh JJ, Kinney JW, Cummings JL. Animal systems in the development of treatments forAlzheimer's disease: challenges, methods, and implications. Neurobiol Aging. 2013; 34:169-83.

    [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: 409421.

    [53] Huntington's Disease Collaborative Research Group. A novel gene containing a trinucleotide repeatthat is expanded and unstable on Huntington's disease chromosomes, Cell 1993; 72:971983.

    [54]ZuccatoC, ValenzaM, CattaneoC. Molecular mechanisms and potential therapeutic targets in

    Huntington's disease. Physiol Rev 2010; 90:905981.

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

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

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

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

    http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed?term=Morton%20AJ%5BAuthor%5D&cauthor=true&cauthor_uid=19464370http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/22464953http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/22464953http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/22464953http://physrev.physiology.org/search?author1=Chiara+Zuccato&sortspec=date&submit=Submithttp://physrev.physiology.org/search?author1=Chiara+Zuccato&sortspec=date&submit=Submithttp://physrev.physiology.org/search?author1=Chiara+Zuccato&sortspec=date&submit=Submithttp://physrev.physiology.org/search?author1=Marta+Valenza&sortspec=date&submit=Submithttp://physrev.physiology.org/search?author1=Marta+Valenza&sortspec=date&submit=Submithttp://physrev.physiology.org/search?author1=Marta+Valenza&sortspec=date&submit=Submithttp://physrev.physiology.org/search?author1=Elena+Cattaneo&sortspec=date&submit=Submithttp://physrev.physiology.org/search?author1=Elena+Cattaneo&sortspec=date&submit=Submithttp://physrev.physiology.org/search?author1=Elena+Cattaneo&sortspec=date&submit=Submithttp://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed?term=Ferrante%20RJ%5BAuthor%5D&cauthor=true&cauthor_uid=19362590http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/?term=(ferrante+rj%5BAuthor+-+First%5D)+AND+(%222009%22%5BDate+-+Publication%5D+%3A+%222009%22%5BDate+-+Publication%5D)http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/?term=(ferrante+rj%5BAuthor+-+First%5D)+AND+(%222009%22%5BDate+-+Publication%5D+%3A+%222009%22%5BDate+-+Publication%5D)http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed?term=Ferrante%20RJ%5BAuthor%5D&cauthor=true&cauthor_uid=19362590http://physrev.physiology.org/search?author1=Elena+Cattaneo&sortspec=date&submit=Submithttp://physrev.physiology.org/search?author1=Marta+Valenza&sortspec=date&submit=Submithttp://physrev.physiology.org/search?author1=Chiara+Zuccato&sortspec=date&submit=Submithttp://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/22464953http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/22464953
  • 7/29/2019 Animal Models of CNS Disorders

    24/30pted

    Man

    uscript

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    [65] Duty S, Jenner P. Animal models of Parkinson's disease: a source of novel treatments and clues tothe 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'sdisease with apoptosis inhibitors underline the need for a paradigm shift in drug discovery forneurodegenerative diseases. Biochem Pharmacol. 2006; 72:11971206.

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

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

    [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 ofLRRK2 to membranous and vesicular structures in mammalian brain. Ann Neurol. 2006; 60:557-569.

    [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; 89:707-758.

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

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

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

    http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/22960626http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/20547124http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/20547124http://www-ncbi-nlm-nih-gov.ezproxy2.library.drexel.edu/pubmed/22960626
  • 7/29/2019 Animal Models of CNS Disorders

    25/30pted

    Man

    uscript

    1

    2

    3

    4

    5

    6

    78

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    2627

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    [83] Bove SE, Flatters SJ, Inglis JJ, Mantyh PW. New advances in musculoskeletal pain. Brain Res Rev2009; 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? TrendsPharmacol 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.

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

    [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.

    [90] Crowell RM, Marcoux FW, DeGirolami U. Variability and reversibility of focal ischemia inunanesthetized 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 cerebralischemia. 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. Anew experimental model of experimental embolism in rats in which recirculation can be reintroducedin the ischemic area. Jpn J Stroke. 1986; 8:1-8.

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

    McGonigle CNS Models - Tables-Revised 061713.docx

  • 7/29/2019 Animal Models of CNS Disorders

    26/30

    Page 25 of 29

    Accepted

    Manus

    criptTable 1. Critical Factors Affecting Variability and Reproducibility

    All Models Transgenic Models Lesion Models

    Sex Gene construct Surgical skill

    Strain Penetrance Choice of toxin

    Age Constitutive/Inducible Dose and speed of delivery

    Housing Choice of Promotor Extent of damage

    Handling Pathological Phenotype Recovery environment & Time

    Environment Behavioral Phenotype Infections

    Confounding Behavior Rare or common mutation Post-op anesthetic

  • 7/29/2019 Animal Models of CNS Disorders

    27/30

    Page 26 of 29

    Accepted

    Manus

    criptTable 2.Summary of animal models of CNS Disorders

    Disorder Category Representative

    Tests

    Rationale Strengths Weaknesses Predictive

    Validity

    Reviews

    Depression Acute Forced Swim

    Tail Suspension

    Acute stress

    produces form ofbehavioral

    despair

    Fast,

    reproducible,widely used

    Acute effect not

    consistent withdelayed onset of

    ADs in clinic

    Good for SSRIs,

    SNRIs, TCAs andKetamine

    [13], [15]

    [97], [98]

    Chronic Chronic Mild

    Stress, Olfactory

    Bulbectomy,

    Social Defeat

    Chronic stress

    produces long

    lasting

    behavioral

    changes that

    resemble

    symptoms of

    depression

    Multiple

    symptoms that

    can be reversed

    by chronic but

    not acute drug

    treatment

    Time consum-

    ing, labor inten-

    sive, costly,

    limited

    reproducibility

    across labs

    Good for SSRIs,

    SNRIs, TCAs, ECS

    and Ketamine

    [13], [15]

    [18], [97]

    Anxiety Ethological Elevated Plus

    Maze, Light/Dark

    Box, Open Field,

    Defensive Burying

    Conflict between

    naturally

    occurring

    behavior and

    naturally

    aversive

    situation

    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

    [21]

    Conflict Vogel Punished

    Drinking, Geller-

    Seifter, 4-Plate,

    Novelty

    Suppressed

    Feeding

    Conflict between

    motivated

    behavior and

    aversive stimulus

    Fewer false

    positives

    May require

    operant

    conditioning,

    less sensitive to

    novel

    mechanisms

    Good for

    benzodiazepines,

    4-plate and NSF

    detect SSRIs,

    SNRIs, TCAs

    [20], [23]

    Cognitive Fear Conditioning,

    Fear Potentiated

    Startle

    Conditioned to

    associate neutral

    stimulus with

    aversive

    properties

    Assesses the

    learning

    component of

    fear and anxiety

    Time

    consuming,

    requires

    conditioning

    Good for

    benzodiazepines,

    5-HT1A agonists

    [9], [20]

    [24]

  • 7/29/2019 Animal Models of CNS Disorders

    28/30

    Page 27 of 29

    Accepted

    Manus

    criptSchizophrenia Pharmacological PCP,

    Amphetamine

    Compounds

    produce

    psychotic

    symptoms in

    man

    Fast,

    reproducible,

    reliable across

    labs, good for

    positive

    symptoms

    Acute model, no

    developmental

    pathology, does

    not model

    negative

    symptoms

    Good for typical

    and atypical

    antipsychotics

    [26]

    Developmental MAM treatment,

    Post-weaning

    Isolation, Ventral

    Hippocampal

    Lesion

    Produce

    developmental

    abnormalities

    and post-puberty

    deficits

    Mimics time

    course and some

    developmental

    aspects of

    disorder

    Time

    consuming, low

    throughput, not

    suitable for

    screening

    Not sufficiently

    validated

    [27], [29]

    Genetic DISC1, NRG1,

    Dysbindin

    Based on genetic

    linkage,

    associated with

    increased

    susceptibility

    Produce distinct

    neurological and

    behavioral

    phenotype

    Mild

    phenotypes

    Not sufficiently

    validated

    [4], [27]

    [34]

    Alzheimers

    Disease

    Pharmacological Scopolamine Based on

    degeneration of

    cholinergic

    system in AD

    Produces

    cognitive

    impairment,

    mimics

    degeneration of

    cholinergic

    neurons

    Does not

    reproduce

    pathology or

    progressive

    nature of

    disorder

    Good for

    symptomatic

    therapies such as

    AchE inhibitors

    [43]

    Lesion Transection of

    Fimbria Fornix,

    Electrolytic orchemical lesion of

    cholinergic nuclei

    Based on

    Degeneration of

    cholinergicsystem in AD

    Produces

    cognitive

    impairment anddegeneration of

    cholinergic

    neurons

    Does not

    reproduce the

    pathology orprogressive

    nature of the

    disorder

    Good for

    symptomatic

    therapies such asAchE inhibitors

    [44], [51]

    Transgenics Tg2576, hAPP,

    PSAPP, 5XFAD,

    3XTg, PSAPP rat

    Genetic

    mutations linked

    to rare forms of

    AD that exhibit

    symptoms of

    Recapitulate

    multiple aspects

    of the pathology,

    exhibit cognitive

    deficits, show

    Majority exhibit

    only subset of

    pathology,

    based on rare

    forms

    Good for AChE

    inhibitors,

    disappointing to

    date for novel

    therapies

    [46], [47]

    [51]

  • 7/29/2019 Animal Models of CNS Disorders

    29/30

    Page 28 of 29

    Accepted

    Manus

    criptsporadic AD progression

    Huntingtons

    Disease

    Lesion Kainic Acid,

    Quinolinic Acid

    Mimic loss of

    discrete

    populations of

    neurons in

    caudateputamen

    Produce many

    behavioral

    deficits

    associated with

    HD

    Do not mimic

    pathology,

    progressive

    nature or

    reduced survival

    Limited

    validation

    [56], [57]

    Transgenics R6/2, HdhQ111,

    YAC128

    Based on

    autosomal

    dominant

    mutation

    responsible for

    disorder

    Complex

    phenotypes,

    mimic pathology,

    progressive

    deficits and

    reduced survival

    for R6/2

    Most faithful

    mutations

    produce mild

    phenotype,

    limited

    progression,

    normal survival

    Not sufficiently

    validated but

    tetrabenazine is

    active in YAC128

    mice and

    patients

    [5], [54]

    [55], [58]

    Parkinsons

    Disease

    Toxins 6-OHDA, MPTP Based on well-

    establisheddegeneration of

    dopaminergic

    neurons

    Robust motor

    deficits, MPTPproduces

    Parkinsonism

    in man and

    primates

    Some aspects of

    disorder notobserved, such

    as Lewy Bodies

    and Locus

    Coeruleus

    involvement

    Good for

    dopaminergicmechanisms, not

    for disease-

    modifying agents

    [62], [63]

    [64], [65]

    Transgenics -synuclein,

    LRRK2, Parkin, DJ-

    1, Pink-2

    Based on genetic

    mutations linked

    to rare forms of

    disorder

    Reproduce some

    of the pathology

    associated with

    the disorder

    Mild or no

    phenotypes and

    no

    dopaminergic

    degeneration

    No information [63], [67]

    [70]

    Pain Acute Formalin

    Injection,

    Writhing, Surgical

    Incision

    Introduction of

    irritant or

    production of

    tissue damage

    Fast,

    reproducible and

    sensitive to

    many

    mechanisms

    Dont model

    more common

    chronic or

    pathological

    conditions

    Good for several

    established

    classes e.g.,

    opiates and

    NSAIDS

    [72], [74]

    Neuropathic Spinal Nerve

    Ligation, Chronic

    Constriction

    Based on

    production of

    nerve damage

    Chronic

    condition with

    robust

    Can be

    technically

    challenging,

    Good for

    established

    classes of

    [72], [74]

    [75], [76]

  • 7/29/2019 Animal Models of CNS Disorders

    30/30

    Page 29 of 29

    Accepted

    Manus

    criptInjury,

    Streptozotocin,

    Paclitaxel

    using surgical,

    chemical or

    metabolic

    approaches

    symptoms,

    reproducible

    across labs

    time consuming,

    subject to false

    positives

    analgesics but

    several false

    positives recently

    Cancer Bone tumors Mimic bone

    metastases toproduce pain

    Produces robust

    symptoms andmimics

    physiological

    response

    Technically

    difficult andtime consuming

    Good for

    establishedclasses, not yet

    validated for new

    mechanisms

    [83], [84]

    Stroke Occlusion Ligature, clips,

    cauterize

    Mimic focal

    ischemia by

    restricting blood

    flow

    Produces

    ischemic damage

    to discrete

    regions

    Difficult to

    control and

    reverse

    Poor, based on

    failure of

    multiple stroke

    trials

    [91]

    Emboli Injection of

    emboli

    Mimic the cause

    of major infarcts

    Produces

    significant

    ischemic damage

    Difficult to

    control, not

    transient

    Good for tPA, so

    far not predictive

    for any othermechanisms

    [91]