You are on page 1of 18

Schizophrenia and Psychotic Disorders

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)

Insights from Proteomic Studies on


Schizophrenia Preclinical Models:
What Can We Learn for Drug Discovery?
Lorna A. Farrelly  · Melanie Föcking  · David R. Cotter 
Department of Psychiatry, Royal College of Surgeons in Ireland, Smurfit Education and Research Centre,
Beaumont Hospital, Dublin, Ireland

Abstract more they are central for the testing of new drugs. Ani-
Proteomics offers the ability to monitor and identify mal models in this aspect have the advantage that they
changes in protein expression and/or signalling path- enable the investigation of the neurobiology of the phe-
ways with high precision, accuracy and reproducibility. nomena of interest using invasive techniques that can-
Proteomic techniques, including bioinformatics soft- not be used in humans. Proteomics-based research of
ware, are steadily advancing, and technical improve- these preclinical models can greatly increase our under-
ments to the methods currently employed for protein standing of schizophrenia and shape the direction of fu-
separation and protein identification will dramatically ture research. Current proteomic studies have so far
increase the number of proteomics-based schizophre- been insightful and point to a role for mitochondrial
nia research studies. The advent of high-throughput dysfunction, alteration in cell signalling and cytoskeletal
techniques allows scientists to quantify complex biolog- functioning in preclinical models. Proteomic studies of
ical mixtures, such as postmortem brain, and to identify antipsychotics in preclinical models are an indispens-
and validate new biomarkers. Animal models have con- able tool for investigating underlying schizophrenia
tributed much to our understanding of disease mecha- neurobiology. Current investigations have found altera-
nisms and are often utilised in research because of their tions in core mitochondrial proteins and in protein syn-
abundant supply and ease of manipulation. Different thesis and cell signalling pathways in preclinical models
theories regarding the aetiology of schizophrenia make of schizophrenia undergoing antipsychotic treatment.
the design of a single schizophrenia animal model im- Research is encouraging, and advancement with pre-
possible. Therefore, there are many preclinical animal clinical models of schizophrenia can be made with more
models for schizophrenia research, each of which has reliable quantitative proteomic methods. Proteomic
relevance to different aspects of the neurobiology of the studies on preclinical models of schizophrenia are, to
disorder and its treatment. Preclinical animal models in date, somewhat sparse, but will hopefully multiply with
schizophrenia research are vital for biomarker discovery the establishment of better quantitative approaches.
and in the exploration of disease neurobiology. Further- © 2014 S. Karger AG, Basel
144.82.108.120 - 10/2/2016 9:33:56 PM
Downloaded by:
UCL
Introduction schizophrenia. Several candidates have been pro-
posed to explain what risk factor may be linked to
The definitions and boundaries of schizophrenia urbanicity, and among these, differences in sub-
have varied over the past century. This changing stance misuse, prenatal health, poverty and social
concept of schizophrenia has been influenced by stress have been proposed [5]. It was originally
available diagnostic tools and treatment, by related believed that the prevalence and risk of develop-
conditions from which it most needs to be distin- ing schizophrenia was similar among males and
guished and by scientific paradigms [1]. Further- females, but more recent studies have revealed
more there is significant heterogeneity in the aetio- that compared with females, males have a 4-fold
pathology, symptomatology and course of the dis- increased risk of the disease [6, 7]. Having an af-
ease. Additionally, the severity of positive, negative fected family member greatly increases the risk of
and cognitive symptoms varies across patients. Al- developing schizophrenia. This risk increases
though the therapeutic armamentarium for the substantially as the degree of genetic affinity with
treatment of schizophrenia has grown, current the affected family member increases, a concept
treatments for schizophrenia are inconsistent, with pioneered by Kallman in 1946 with his assess-
their success and their benefits varying from pa- ment of schizophrenic twins [8]. The genetic the-
tient to patient. Furthermore, certain side effects ory of schizophrenia was strengthened by investi-
can still have an impact on patient health and qual- gating adopted-away twins, where the risk of
ity of life, giving rise to relapses in medication and schizophrenia in the twin was related to the pres-
worsening of symptoms [2]. The hope for further ence of the disease in the biological parent, and
progress relies upon variation affecting drug and not in the adoptive [9, 10]. One of the most repli-
interindividual differences in pharmacoresponse cated findings in schizophrenia research is that
[3]. Current strategies, most often guided by vari- offspring born in the winter and spring months
ous disease hypotheses, often focus on the identifi- have a 10% increased risk of developing schizo-
cation of novel biomarkers to aid in the develop- phrenia compared with offspring born at other
ment of more effective therapies. The utilisation of times of the year [11–13] and maternal nutrition
preclinical models is pivotal in this endeavour. and mineral deficiency pose significant risks to
the developing fetus [14]. It is well established
that obstetric complications, those that occur pre-
Schizophrenia: Epidemiology and Clinical natally and perinatally, increase the risk of schizo-
Presentation phrenia in the offspring. Among these adversities
are included bleeding during pregnancy and
Schizophrenia is a debilitating neuropsychiatric emergency caesarean section [15, 16].
disease that disturbs perception, cognition, think-
ing and social behaviour. It is estimated that the
risk of developing the disease over a lifetime rang- Schizophrenia: Clinical Presentation
es from 0.3 to 2% with an average of 0.7% [4]. In-
cidence studies based on service provision may Schizophrenia is recognised by a diverse set of
grossly underestimate incidence rates as some af- signs and symptoms which ultimately include dis-
fected individuals may not seek professional tortion of thought and perception, avolition, apa-
treatment. However, numerous meta-analyses thy and cognitive impairments. As a result of these
have confirmed longstanding suggestions of a disabilities, patients experience increased feelings
strong link between urbanicity, migration and of isolation and day-to-day living becomes ex-
male gender with an increased risk of developing tremely difficult. These symptoms or abnormali-
144.82.108.120 - 10/2/2016 9:33:56 PM

Insights from Proteomic Studies on Schizophrenia Preclinical Models 57


Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
ties are classified into positive, negative and cogni- What Is a Preclinical Model?
tive disorganisation [17]. Delusion, hallucinations
and several other reality distortions comprise the Purpose
positive symptoms. The onset of positive symp- The term ‘preclinical’ is defined as the period of
toms generally occurs in late adolescence or early research prior to clinical trials in the general
adulthood, where reality distortion marks the for- population. This period of a drug trial is of ex-
mal onset of the disease [18]. Hyperactivity of the treme importance, as it is during this time that
dopaminergic mesolimbic pathway appears to un- iterative testing and drug safety data are collect-
derlie the positive symptoms, and these are the ed. A model is defined as any experimental prep-
most responsive to antipsychotics [19, 20]. Nega- aration developed for the purpose of studying a
tive symptoms involve a loss of affective and cona- disease.
tive functions. Patients exhibit a loss of motivation, The model is composed of both the indepen-
alogia, avolition (loss of initiative) and a general dent variable (i.e. inducing manipulation) and
loss of social drive [21, 22]. ‘Extrinsic’ factors such the dependent variable (i.e. the measure used to
as neuroleptic treatment and depression are com- assess the effects of the manipulation, e.g. be-
mon causes of so-called secondary negative symp- havioural testing) [28]. The choice of preclinical
toms [23]. The cognitive abnormalities associated model for a study depends on the aetiology of
with schizophrenia include deficits in episodic the disorder of interest, such as schizophrenia. If
memory, sustained attention, working memory, the aetiology of the disease is known, patholo-
processing speed and reasoning/problem solving gies with strong homology to the human case
[24]. Cognitive deficits persist over the long-term can be induced in animals more readily. In
course of schizophrenia and modest improve- schizophrenia, this remains challenging as there
ments are only observed with antipsychotic treat- are several hypotheses regarding the aetiology of
ment [25]. The therapeutic armamentarium com- the disease, and the pathology at the protein,
prises the earlier first-generation, so-called ‘typi- pathway and genetic levels is not clearly defined.
cal’, antipsychotics such as chlorpromazine and Preclinical models can involve either animal- or
haloperidol, and the second-generation, so-called human-derived preparations. The advantage
‘atypical’, antipsychotics such as olanzapine, que- with utilising non-human models is that it en-
tiapine and risperidone [26]. Although typical an- ables the investigation of the neurobiology of
tipsychotics are successful in tackling the positive the phenomena of interest using invasive tech-
symptoms of the disease, they are minimally effec- niques that cannot be used in humans [29]. If
tive against the negative and cognitive symptoms, standardised and used correctly, non-human
which are responsible for much of the disease-re- preclinical models can save costs hugely during
lated disability [1]. Although antipsychotics ame- drug development. However, as will be dis-
liorate a range of symptoms and prevent a risk of cussed later on, patient-derived human samples,
relapse, the extent to which they improve psycho- such as stem cells, are becoming increasingly
social function in patients is less clear [27]. The important.
molecular basis for the effects of antipsychotic
drugs is not fully understood, which makes the de- Criteria for a Preclinical Model
sign of further agents a challenging process [26]. In choosing and developing a preclinical research
There is an unmet need for a revolution in the de- model it is imperative to consider the purpose in-
velopment of pharmacological treatments which tended for the model. There are several criteria
can only be addressed with better knowledge of the required, as displayed in figure 1.
disease origin and pathophysiology.
144.82.108.120 - 10/2/2016 9:33:56 PM

58 Farrelly · Föcking · Cotter
Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
What disease
needs to be
modelled?

Predictive
validity: What symptoms
needs to be
Accurate drug mirrored?
response? Criteria
for a
preclinical
model

Construct Face validity:


validity: Does the model
Does the model mimic core
mimic disease- symptoms or
related deficits? biochemistry?
Fig. 1. Criteria to be considered
in choosing a preclinical model
[30–35].
 

Preclinical Models of Schizophrenia vestigations have also identified dysregulation in


Preclinical models of schizophrenia are indispens- core cellular pathways responsible for cell growth
able tools for testing disease hypotheses which can- and differentiation [60]. Stem cells have the poten-
not be directly addressed in human subjects for tial to become a powerful tool for researchers en-
technical and ethical reasons. However, even gaged in the pathological study of neuronal disor-
though they are limited in their capability to mim- ders such as schizophrenia. This advent of human
ic the complete clinical presentation of a schizo- induced pluripotent stem cells makes it possible to
phrenic patient, they are effective in testing specific study schizophrenia-derived tissue in vitro, where
causative or mechanistic hypotheses regarding there is already a genetic predisposition towards the
schizophrenia [36]. Currently, available preclinical disease state [61]. A big advantage of utilising olfac-
models of schizophrenia, predominantly rodent, tory neural tissue is that it is accessible in human
fit into four different induction categories: (1) de- adults [62]. Induced pluripotent stem cells can re-
velopmental, (2) drug-induced, (3) lesion or (4) ge- capitulate in vitro aspects of patients’ neuronal de-
netic manipulation; they are displayed in figure 2. velopment and thus help to clarify early pathologi-
More recently, preclinical models of schizo- cal processes in the disease [58, 60, 61, 63].
phrenia have moved to an in vitro setting, utilising
patient-derived neural stem cells such as those from
the olfactory bulb [58]. An emerging report by Sawa Proteomics
and co-workers [59] shows olfactory cells to reflect
the developing brain in gene expression profiles Proteomics holds the promise of unravelling
and, more surprisingly, to have an expression pro- pathological mechanisms at the molecular level.
file closer to those of stem cells over blood cells. In- Mass spectrometry (MS) and gel-based approach-
144.82.108.120 - 10/2/2016 9:33:56 PM

Insights from Proteomic Studies on Schizophrenia Preclinical Models 59


Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
• Maternal infection
• Obstetric complications
Neurodevelopmental • Maternal malnutrition
• Maternal isolation
• Gestational methylazoxymethanol acetate

• PFC lesion
Pharmacological
• Hippocampal lesion
and
• Amphetamine model
lesion
• PCP model

Fig. 2. Summary of well-known • DISC1


• Reelin
­preclinical models of schizophrenia Genetic • Dysbindin 1
[14, 20, 32, 37–57]. PFC = Prefrontal • Neuregulin 1
cortex; PCP = phencyclidine; • ERBB4
DISC1 = disrupted in schizophrenia
1; ERBB4 = receptor tyrosine-protein
kinase erbB-4.

es such as two-dimensional difference gel electro- of samples [66]. Proteomic methods can be cate-
phoresis (2-D DIGE) allow in-depth analyses of gorised into those that are gel based and those
entire proteomes. Furthermore their use is be- which are not.
coming increasingly appreciated in the field of 2-D DIGE is a powerful technology for pro-
biomarker discovery [64]. MS-based proteomics tein abundance studies. Originally described by
is increasingly used to address basic and clinical O’Farrell, it is based on separation of the proteins
questions in biomedical research through studies according to their charge in the first dimension
of differential protein expression, protein-protein by isoelectric focusing and relative molecular
interactions and posttranslational modifications. mass in the second dimension by SDS-PAGE
The field of proteomics is undergoing rapid de- [67]. It is often coupled to MS for protein identi-
velopment in a number of different areas includ- fication, resulting in protein ‘maps’. Their reso-
ing improvements in mass spectrometric plat- lution, however, is not sufficient relative to the
forms, peptide identification algorithms and bio- enormous diversity of cellular proteins in terms
informatics. In particular, new and/or improved of size, and problems are especially prominent
approaches have established robust methods that for poorly water-soluble (hydrophobic) proteins
allow not only for in-depth and accurate peptide such as membrane and nuclear proteins [68].
and protein identification and modification, but Traditional gel-based proteomic quantitation
also for sensitive measurement of relative or ab- approaches are being overcome by the simplicity
solute quantitation [65]. In terms of biomarker of MS-based approaches, which address issues of
investigations, proteomic methods have been uti- reproducibility and allow for high precision over
lised in the clinical field and applied to a variety traditional methods [69, 70]. Non-gel-based
144.82.108.120 - 10/2/2016 9:33:56 PM

60 Farrelly · Föcking · Cotter
Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
methods are coming to the fore and their success iTRAQ literature, see Evans et al. [78]. Another
is owed to their ability to analyse more complex non-gel-based method for the multidimensional
samples and to generate data faster in a high- separation of proteins is the novel analytical
throughput manner [71]. These approaches technique referred to as ‘surface-enhanced laser
combine liquid chromatography with MS. In la- desorption/ionisation’. It is a high-throughput-
bel-free approaches, complex samples are digest- array-based technology and useful for detection
ed to peptides with an enzyme such as trypsin, of disease-associated proteins for biomarker dis-
separated by high-performance liquid chroma- covery [79]. Label-free ‘shotgun’ proteomics can
tography, and their mass analysed within the estimate protein abundance more accurately
mass spectrometer. Methods involving stable than gel-based methods, and can observe the dif-
isotope labelling have been advantageous in in- ferential abundance of proteins over a larger dy-
vestigating the differential quantitative expres- namic range than labelling techniques [80]. It is
sion of proteins in samples. In vivo labelling, for hoped that advances in instrumentation and bio-
example, employs biosynthetic incorporation of informatics software will overcome obstacles as-
isotopically labelled nutrients or amino acids sociated with quantitative reproducibility and
into proteins, and in vitro labelling is achieved will increase proteome coverage.
postbiosynthetically at a specific site in proteins
or peptides [72]. SILAC (stable isotope labelling
by amino acids in cell culture) is an example of Proteomics in Schizophrenia
such in vivo labelling. SILAC exhibits the capa-
bility to quantify both the proteome and its mod- The field of neuroproteomics is undergoing de-
ifications in response to stimuli and perturba- velopments in various areas such as improving
tions and has also made advances in quantitative mass spectrometric platforms, algorithm build-
phosphoproteomics (for a recent review see ing and bioinformatics for analysis of large data
Mann [73]). Protein labelling with isotope-coded sets from clinical samples. However, the area re-
affinity tags followed by tandem MS allows se- mains challenging due to the high degree of com-
quence identification and accurate quantifica- plexity, such as in the postmortem brain with its
tion of proteins in complex mixtures [74]. Pio- high heterogeneity of cell types. Therefore there is
neered by Gygi et al. [75], side chains of lysine a need for optimal sample preparation, sensitive
and cysteine are primarily used to incorporate an instrumentation and high-throughput validation
isotope-coded mass tag. Isotope-coded affinity [81]. Schizophrenia biomarker discovery heavily
tags can significantly reduce the complexity of relies on MS-based proteomics to unravel the
the peptide mixture, which can be advantageous complexity of biological samples such as brain tis-
when analysing highly complex samples [76]. sue, blood, urine, cerebrospinal fluid (CSF) and
Another stable isotope labelling technique uses plasma. The advancement in this area is made ev-
amine-specific isobaric tags (iTRAQ). The de- ident by the surge of research which is focusing
sign of the iTRAQ reagents allows quantitative on the application of MS in neuropsychiatry [82–
comparison of 4 (or more) protein pools simul- 86]. This move from gel-based methods is a result
taneously. The method is conceptually elegant as of improved MS instrument design, which allows
peptides are labelled at the N-terminus and at the in-depth analysis of tissues of high complexity
ε side chain of lysines. Furthermore, iTRAQ dif- such as brain.
fers by deriving quantification at the MS/MS Proteomic brain studies in schizophrenia
rather than MS level to result in a high signal/ have reported changes within both the white and
noise ratio [77]; for a recent review on the current grey matter of the brain for proteins such as al-
144.82.108.120 - 10/2/2016 9:33:56 PM

Insights from Proteomic Studies on Schizophrenia Preclinical Models 61


Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
dolase C, creatine kinase, enolase, septin 3 and they may aid in the development of novel thera-
dynamin 1. These changes are related to meta- peutics. Below, we summarise the proteomic
bolic, cytoskeletal and synaptic function, respec- studies according to their model types, which are
tively, and have been identified in the prefrontal predominantly based on developmental stressors
cortex, anterior cingulate cortex, corpus callo- and genetic manipulation.
sum and insular cortex [87–92]. These findings
provide the support for a view of altered cytoskel- Developmental Stressors
etal and mitochondrial function as core compo- Phencyclidine
nents of the neuropathology of the disease [91, Phencyclidine (PCP) induces a broad range of
93, 94]. Myelin protein changes have also been schizophrenia-like symptoms, a finding that has
reported, further reinforcing the hypothesis of contributed to the hypoglutamatergic hypothesis
disrupted connectivity in schizophrenia [95, 96]. of schizophrenia [101]. In terms of its role as a
Clathrin-mediated endocytosis, a protein traf- preclinical model of schizophrenia, its chronic in-
ficking process, has recently been hypothesised termittent low dosage produces a pattern of met-
to be a core pathological feature of schizophrenia abolic and neurochemical changes in the rodent
[97]. Neuroproteomic studies of schizophrenia brain that mirrors those observed in the brains of
are not exclusive to brain tissue, and many have schizophrenic patients with remarkable preci-
utilised CSF and, in particular, blood serum to sion. This makes it an encouraging model in the
report disease-related protein changes [84, 85, search for newer effective antipsychotic drugs
98]. Ongoing proteomic studies reinforce the in- [102]. In 2012, Ernst et al. [103] analysed changes
terest in particular pathways and also encourage in brain and serum molecular profiles of PCP-
the search for new potential biomarkers [99, treated rats and compared these changes with
100]. There has yet to be a major assessment of those observed in first- and recent-onset antipsy-
current proteomic studies on preclinical models chotic-free schizophrenia patients. The authors
of schizophrenia, therefore this current review identified similar behavioural and molecular al-
tries to fill in that gap. Proteomics will lead to an terations in PCP in rat and human schizophrenia,
improved understanding of schizophrenia such as altered insulin signalling. Overall, this
pathophysiology and thus facilitate novel drug study proposes that the overlapping changes may
discovery. have potential as biomarkers to facilitate the de-
velopment of novel drugs.
Earlier in 2013, Pickering et al. [104] published
Proteomic Studies on Preclinical Models of a research article showing the effect of chronic
Schizophrenia PCP on synaptic proteins in the rodent prefrontal
cortex. Firstly, in validation of their model, PCP-
Using PubMed as a search tool for identifying ex- treated animals made more incorrect alternations
isting studies, it is clear that this area of research than did controls in the Y-maze test, indicating
is still in its infancy. At the time this chapter was and confirming cognitive dysfunction and, in
being put together (December 2013), with the other words, dysfunction at the level of the pre-
terms ‘proteomics’ and ‘schizophrenia’, 152 re- frontal cortex. In agreement with previous re-
sults were retrieved, with fewer than 20 studies views of PCP effects [105] suggesting that this
implementing the use of preclinical models. Some drug changes synaptic transmission and plasticity
of these studies will be discussed in terms of their and also affects glycolysis and gluconeogenesis,
findings, of how they may contribute to our fur- the authors, utilising 2-D DIGE, found dysregula-
ther understanding of the disease and also of how tion of proteins, mainly of those with synaptic,
144.82.108.120 - 10/2/2016 9:33:56 PM

62 Farrelly · Föcking · Cotter
Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
mitochondrial and cytoskeletal functions. In of schizophrenia, it will be included in this re-
sum, this study reported protein changes similar view. Proteins that were significantly changed
to those evident in human postmortem schizo- by maternal separation included molecular
phrenic brains, predominantly related to mito- chaperones, proteins related to energy metabo-
chondrial function. Not only does it support the lism, neuroplasticity, oxidative stress regulation
use of PCP as a valid preclinical model of schizo- and protein metabolism. Treatment with escita-
phrenia, but it also points to its role in transla- lopram, a selective serotonin reuptake inhibitor,
tional research in the search for new, more tar- induced changes in a different group of proteins,
geted antipsychotic drugs. A more recent study except for a few involved in energy metabolism
by Wesseling et al. or the Bahn group [106] com- and neuroprotective pathways. This model
bined a proteomic approach with metabonomics highly correlates with previous postmortem
to compare human schizophrenic brain patholo- proteomic studies of schizophrenia [88, 105,
gy with that of the frontal cortex of rodents ex- 112–114]. In 2012, Daniels et al. [115] performed
posed to PCP. With this approach they identified iTRAQ proteomic analyses of protein expres-
proteomic alterations in mitochondrial function, sion to determine whether exercise counteracts
glutamate-mediated calcium signalling and cyto- the detrimental effects of early-life adversity in
skeletal remodelling. Changes in the calcium sig- the form of maternal separation on protein ex-
nalling system and calcium-related protein kinas- pression in the brain. Proteins involved in neu-
es have previously been implicated in the aetiol- ronal structure, metabolism, signalling, antioxi-
ogy of schizophrenia [107, 108], reinforcing the dative stress and neurotransmission were affect-
dysregulation of cognition and long-term poten- ed by maternal separation. Remarkably, many of
tiation. Additionally, metabonomic profiling of these proteins were restored to normal by subse-
the PCP-treated animals revealed changes in lev- quent exposure to voluntary exercise in adoles-
els of pyruvate, glutamine, glycine and glutamate, cence.
which agrees with evidence of mitochondrial dys- Dimatelis et al. [116] quantified methamphet-
function and impaired brain energy metabolism amine-induced protein expression in the shell
in human schizophrenia [109]. Martins-de-Souza and core of the nucleus accumbens in animals ex-
et al. [110] established a selective reaction moni- posed to maternal separation using iTRAQ with
toring assay for multiplex analysis of enzymes of MS. The most revealing outcome was that, com-
the glycolysis pathway which are already known pared with maternal separation or methamphet-
to be affected in human schizophrenia and the amine alone, the maternal separation/metham-
PCP model. The results showed altered levels of phetamine combination altered more proteins
several core glycolytic enzymes in PCP rats com- involved in cellular growth and energy metabo-
pared with controls. This study not only supports lism. This study confirmed that both early-life
the PCP preclinical model, but also that selective stress and drug exposure behave in a ‘two-hit’
reaction monitoring MS could be used in the de- manner and result in more proteins being affect-
velopment of multiplex classification tools for ed than each by treatment/adversity alone [116].
schizophrenia. Exercise has been associated with improve-
ment in mood and anxiety in animals and in hu-
Maternal Separation man studies [117, 118], but the underlying mo-
A study by Marais et al. [111] in 2009 explicitly lecular mechanisms had yet to be elucidated. This
used the preclinical model of maternal separa- is a novel use of the preclinical model of maternal
tion for a study into depression, but considering separation utilising proteomics to convey a pos-
that depression is often a core negative symptom sible alternative to antipsychotic intervention in
144.82.108.120 - 10/2/2016 9:33:56 PM

Insights from Proteomic Studies on Schizophrenia Preclinical Models 63


Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
adolescence. Kitteringham et al. [119], with label- Prenatal Psychosocial Stress
free MS and multiple reaction monitoring, show It has been hypothesised that prenatal exposure
that maternal separation from early weaning to maternal stress increases the risk of subse-
leads to dysregulation of markers of mature oli- quently developing schizophrenia, so therefore
godendrocytes and genes involved in protein animal models of chronic stress may offer in-
translation and other categories, an apparent sights into pathways operating in the disease
downward biasing of translation, and to methyla- [123]. Proteomic studies in this regard are sparse,
tion changes in the promoter regions of selected and just three to date have utilised proteomic
dysregulated genes. These findings are likely to techniques to identify the protein changes in-
prove useful to our understanding of the mecha- duced by repeated exposure to psychosocial
nism by which early-life neglect affects brain stress. Investigations utilising the maternal re-
structure, cognition and behaviour. Additionally, straint stress paradigm have reported – by way of
a strength of this study was that multiple reaction 2-D DIGE and MS – changes in proteins involved
monitoring proteomics methods were used to in the regulation of signal transduction, synaptic
provide further confirmation of the effects of ma- vesicles, cytoskeleton dynamics and energy me-
ternal separation on myelin-related proteins tabolism in the offspring born to these mothers
[119]. Isolation rearing from weaning/maternal [Föcking et al., under review; 124]. An earlier
separation captures the neurobiological relation- study of the hippocampus employing a social de-
ship between environmental stress and schizo- feat model found changes in offspring relating to
phrenia [120]. It causes changes reminiscent of protein folding, signal transduction, synaptic
negative and cognitive impairments probably as plasticity, cytoskeleton regulation and energy
a consequence of alterations in frontal cortico- metabolism with 2-D DIGE and MS [125]. It is
limbic circuits. The advantage of this neurodevel- clear from these studies thus far that there is a
opmental preclinical model over pharmacologi- common effect of psychosocial stress on the hip-
cal models such as that of PCP is its ability to al- pocampus of offspring born to these mothers. Fu-
low proteomic investigations in the absence of ture proteomic investigations should further ex-
confounding drug interventions. Isolation- amine these particular pathways as they could
reared rodents show impaired prepulse inhibi- provide novel targets for drug discovery.
tion of acoustic startle compared with group-
housed controls, which is thought to reflect sen- Pharmacological/Lesion Models
sorimotor gating deficits. Proteomic studies Methylazoxymethanol acetate is a DNA-alkylat-
utilising this model, however, are sparse. Ronca- ing agent that, when administered during gesta-
da et al. [121], utilising 2-D DIGE and matrix- tion to the fetus, can produce behavioural and
assisted laser desorption/ionisation (MALDI)- anatomical brain abnormalities in adulthood
time-of-flight (TOF)-MS, correlated gating defi- which model some aspects of schizophrenia. Hra-
cits in isolation-reared rats with proteomic detzky et al. [126] reported hippocampus-specific
alterations in the nucleus accumbens. Notably, effects of methylazoxymethanol acetate in the rat,
α-synuclein and 14-3-3 proteins were increased using label-free MS. These proteomic alterations
in the isolation-reared rats. Considering the role mainly concerned those proteins related to syn-
of these proteins in synaptic trafficking, the find- aptic plasticity, such as AMPA1, a major player in
ings provide more clues to the foundations of the synaptic plasticity events [127]. This result in par-
psychosis-like behaviours in isolation-reared rats ticular agrees with previous literature implicating
[122]. This investigation possibly offers the pos- AMPA in the pathophysiology of schizophrenia
sibility of novel drug targets [121]. [128].
144.82.108.120 - 10/2/2016 9:33:56 PM

64 Farrelly · Föcking · Cotter
Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
NMDA dysfunction is hypothesised to play a DIGE coupled with MS on the prefrontal cortex
role in schizophrenia aetiology, which can be of postpubertal, neonatally ventral hippocampal-
mimicked in the MK-801 rat model. MK-801 lesioned rats identifying dysregulation of core
can selectively block NMDA-mediated neuro- synaptic proteins such syntaxin-binding protein.
transmission in rodents, who then display be- This outcome provides vital clues as to what pro-
havioural abnormalities akin to human schizo- tein changes are behind the schizophrenia-like
phrenia [129]. The few studies which have ap- behavioural changes [137]. Additional proteomic
proached this model using proteomic methods studies with developmental lesion models will be
have identified alterations in the cortex and indispensable to identify early protein changes
thalamus of MK-801-treated rats relating to mi- that precede the schizophrenia phenotype, there-
tochondrial and skeletal function [130, 131]. Pro- fore aiding in the development of early interven-
teomics has been indispensable in the critical tion therapies.
analysis of MK-801 as a valid schizophrenia
model. Paulson et al. [132] used 2-D DIGE cou- Prenatal Nutritional Deficiency Models
pled with MS to reveal that different treatment In utero exposure to nutrient deficiency is a
times yield different biochemical results. There- well-known risk factor for developing schizo-
fore it is important to use the same treatment phrenia [138–142], and proteomic studies of this
time for comparative studies. model are underinvestigated, with only few
Smalla et al. [133] in 2008 utilised 2-D DIGE studies having been undertaken. A rat model of
with MS to compare the synaptic proteomes in prenatal vitamin D deficiency assessed protein
human schizophrenia with the cortex of ket- changes in the prefrontal cortex and hippocam-
amine-treated rodents, another pharmacological pus with 2-D DIGE coupled with MS. The au-
model of NMDA dysfunction [134]. Consistent thors reported changes in proteins relating to
changes were identified between the two, but the core metabolic pathways including oxidative
overlap was limited and only one protein was up- phosphorylation, synaptic plasticity and neuro-
regulated in both [133]. This study paved the way transmission, and, using computational analy-
for future studies to further examine the synapto- sis, discovered that the impaired synaptic net-
some in models of NMDA dysfunction. In order work may be a consequence of mitochondrial
to investigate the MK-801 model of NMDA dys- dysfunction, providing further insight into the
function at the synaptic level for contributing pathological effects of this nutrient deficiency
pathways, Zhou et al. [135] isolated the synapto- [143]. A study by McGrath et al. [144] in 2008
some from MK-801-treated rodents and, utilis- used the same model to show, via MALDI-TOF-
ing MALDI-TOF-MS, reported energy metabo- MS, disruptions in proteins related to calcium
lism and semaphorin signalling to be the most binding and mitochondrial function. These
affected pathways, further emphasising the im- protein changes may also underlie some of the
portance of synaptic function in schizophrenia. behavioural features associated with animal
An insult to the brain during a critical period models of developmental vitamin D deficiency
of development can produce subtle anatomical [46, 145], and even further studies can validate
changes sufficient to predate behavioural changes these putative pathways. Guest et al. [146] com-
in adulthood. Such a risk has been investigated in pleted molecular profiling of blood serum, the
the neonatal ventral hippocampal lesion model as frontal cortex and hypothalamus from adult
it mimics a spectrum of neurobiological and be- offspring prenatally exposed to a low-protein
havioural features of schizophrenia [136]. Ver- diet. This model influences impaired insulin
cauteren et al. [137] performed a study with 2-D signalling, which is found to be altered in first-
144.82.108.120 - 10/2/2016 9:33:56 PM

Insights from Proteomic Studies on Schizophrenia Preclinical Models 65


Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
onset schizophrenia [147]. The analysis revealed standing the causality of prenatal infection and
alterations in proteins relating to synaptic and may offer potential biomarkers and novel treat-
glutamatergic transmission and cytoskeletal ment targets for new treatments [156–158].
function, recapitulating pathophysiological at-
tributes seen in schizophrenia. Drug discovery Genetic Manipulation
can benefit from the results of this study in Proteomic characterisation of core molecular
identifying therapeutics that can modulate pathways in genetic preclinical models would be a
these systems. fundamental advancement in understanding
Patton et al. [148] utilised the iTRAQ method schizophrenia, and may have equally significant
to quantitatively assess proteins in the CSF of pre- therapeutic implications. Studies of this kind are
natal iron-deficient anaemic monkeys. Again, the few and insufficiently investigated. Bader et al.
proteins identified to be altered are involved in [159] identified disease-specific epitopes of col-
neurite outgrowth, migration or synapse forma- lapsin response mediator protein 1 in postmortem
tion. These aberrations may account for the be- brain samples of schizophrenia patients. Striking-
havioural deficits observed in iron-deficient chil- ly, further genetic association analysis found that
dren [149, 150]. collapsin response mediator protein 1 functionally
The latest report by English et al. [151] identi- interacts with disrupted in schizophrenia 1, sug-
fied the protein pathways disrupted as a conse- gesting it as a possible gene worth further investi-
quence of chronic ω–3 deficiency in the hippo- gating and incorporating it into a preclinical
campus of mice. The findings pointed towards knockout model of schizophrenia. Dysbindin, a
disturbed synaptic function, neuritogenesis and candidate schizophrenia gene, is involved in syn-
mitochondrial function, furthering our under- aptic trafficking, but its molecular function is
standing of the molecular mechanism by which largely unknown. To investigate this, Hikita et al.
ω–3 deficiency impairs normal brain function. [160] performed MALDI-TOF-MS and liquid
chromatography-MS/MS on isolated dysbindin-
Prenatal Maternal Infection interacting molecules from rat brain lysate. Pro-
Prenatal maternal infection has also been explored teins identified included Munc18-1, helping to
as a preclinical model by proteomic methods. Li- shed light on the molecular function of dysbindin,
popolysaccharides and polyinosinic:polycytidylic which had been largely unknown. Reelin, a glyco-
acid are both compounds which result in produc- protein secreted by GABAergic interneurons, is
tion of fever and induction of proinflammatory substantially decreased post mortem in schizo-
cytokines that affect neurodevelopment and pre- phrenia, making it a suitable candidate gene for the
date behavioural abnormalities akin to human disease [161]. Martins-de-Souza and co-workers
schizophrenia in animal models [43, 152–155]. [162] elegantly compared proteome changes caused
Proteomic studies utilising lipopolysaccharides in by reelin heterozygosis with those found in their
the brain have yet to be reported; however, earlier proteome analyses of schizophrenia human
polyinosinic:polycytidylic acid associated with vi- brain tissue and discovered similar findings in
ral infection has been relatively underinvestigated. terms of energy metabolism and cytoskeletal func-
To our knowledge, three studies have used pro- tion. These findings support the heterozygous ree-
teomics to investigate this preclinical model of lin mouse as a preclinical model, considering its
schizophrenia. So far, metabolic processes have strong overlap with the authors’ previous findings
been reported to be altered, such as the cellular on humans, and reveal new pathways involved in
immune response, glucose and lipid metabolism, reelin dysfunction, therefore providing new tar-
and together they have implications for under- gets for the development of drug therapies.
144.82.108.120 - 10/2/2016 9:33:56 PM

66 Farrelly · Föcking · Cotter
Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
Table 1. Summary of the 9 proteomic studies to date of rodent brain/cells under antipsychotic treatment

Study Drug Proteomic Findings


­method

Bro et al. [166], 2003* Lithium 2-D DIGE 1. 450 proteins identified in the yeast proteome
2. 27 were downregulated 2-fold with lithium, 21 proteins
were upregulated
3. Protein transcription and nucleotide metabolism were
downregulated
4. Stress response and monosaccharide metabolism were
upregulated
O’Brien et al. [167], Risperidone 2-D DIGE, 1. 30 protein spots related to drug treatment in the rat
2006 MALDI-TOF-MS ­striatum
2. Drug-related changes in cellular metabolism, cell
­signalling, transport, protein metabolism,
chaperone activity, DNA binding and cell cycle function
La et al. [168], 2006 Chlorpromazine, 2-D DIGE 1. Drug-related changes in malate dehydrogenase, peroxire-
Clozapine MALDI-TOF-MS doxin 3, ATP synthase subunit and MAPK1 in the rat hip-
pocampus
Chen and Chen [169], Clozapine 2-D DIGE 1. Drug related up-regulation of transthyretin in the rat
2007 ESI-MS/MS hippocampus
Kashem et al. [170], Haloperidol, 2-D DIGE 1. Drug-related protein changes related to the cytoskeleton,
2009 Risperidone calcium regulation, metabolism, signal transduction and
oxidative stress in rat neural stem cells
2. Risperidone expressed more proteins than haloperidol
Ji et al. [171], 2009 Chlorpromazine, 2-D DIGE 1. Drug-related protein changes in the respiratory electron
Clozapine, transport chain and oxidative
Quetiapine phosphorylation were found in rat mitochondria
Ma et al. [172], 2009 Haloperidol, LC-MS/MS 1. Haloperidol and olanzapine altered proteins associated
Olanzapine with cellular assembly/organization and nervous system
development/function in the rat frontal cortex
2. Olanzapine induced more protein changes related to
glycolysis/gluconeogenesis
Martins-de-Souza Clozapine 2-D DIGE, 1. Drug-related changes in cellular metabolism, cell signal-
et al. [173], 2011 LC-MS/MS ling, cell growth and chaperone activity were found in
cultured rat astrocytes
Ahmed et al. [174], Haloperidol, 2-D DIGE 1. Protein changes unique to haloperidol treatment related
2012 Risperidone MALDI-TOF-MS to the cytoskeleton, calcium regulation, oxidative stress
and apoptosis in rat neural stem cells
2. Protein changes unique to risperidone treatment related
to cellular metabolism

* First proteomic paper to utilise a schizophrenia-related medication. Lithium is commonly used as an adjunct to antipsychotics
[163]. ESI = Electrospray ionisation; LC = liquid chromatography; MAPK1 = mitogen-activated protein kinase 1.
144.82.108.120 - 10/2/2016 9:33:56 PM

Insights from Proteomic Studies on Schizophrenia Preclinical Models 67


Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
Proteomic Studies of Preclinical Animal Models der. As with all animal research, the three R’s un-
Utilising Antipsychotics derpinning their humane use must be mentioned:
Preclinical models are vital to understand the mo- upon the discovery of an adequate preclinical
lecular mechanisms behind antipsychotics in or- model, experiments must be ‘refined’, numbers
der to advance the development of more novel must be kept to a minimum for studies, or ‘re-
therapeutics. As the precise mechanisms of action duced’, and, ultimately, animals must be ‘replaced’
of antipsychotic medications are not fully known, [164]. This last principle is becoming more prom-
these quantitative proteomic studies of the effects ising with the use of human-derived neural stem
of antipsychotic drugs on the brain are warrant- cells [58, 165], and further research utilising this
ed. The obstacle to our discovery of novel treat- avenue will hopefully obviate the necessity for an-
ments lies in our lack of understanding of the dis- imals in schizophrenia research to a good extent.
ease-related biological mechanisms. Few studies Although research involving rodent models is rel-
have been undertaken utilising antipsychotics atively economical, and an abundance of informa-
with proteomic studies in preclinical schizophre- tion exists on their genomes, the aetiology of
nia models. Using the search terms ‘antipsychot- schizophrenia contains a large genetic compo-
ics and proteomics’ in PubMed, 45 studies were nent, and patient-derived cell models have the su-
reported. However, of these, only 9 were per- periority in that they do not require genetic ma-
formed on rodent brain or stem cells. Consider- nipulations to mirror this aspect of the disease
ing the focus of this review, only these studies [58].
have been summarised in t­able 1. These current There is no doubt that current proteomic stud-
proteomic studies of preclinical models utilising ies of preclinical animal models have advanced our
antipsychotics have predominantly employed the understanding of schizophrenia neurobiology by
less sensitive gel-based methods but have thus far providing candidates that, once observed to be dif-
been insightful in relation to drug-related protein ferentially expressed in the brain, can now be tested
changes. From the proteomic studies identified in the serum of schizophrenia patients [English et
(table 1), antipsychotics are seen to act through a al., in press]. They have also accelerated the search
variety of pathways, primarily relating to cellular for reliable biomarkers, and progress in proteomic
metabolism, cytoskeletal function and oxidative methods, such as more quantitative approaches of
stress, mechanisms previously found to be altered validation and targeted methods, will help in this
post mortem in schizophrenia [105]. These types respect. Until then, we must remain cautious in the
of investigation are essential for elucidating the interpretation of results, pick the best validated/
mechanism of antipsychotics and are crucial for predictive biomarkers for drug efficacy studies
novel therapeutic developments. and, most importantly, report contradicting results
from current proteomic studies. Without a doubt,
research in this area will have a striking impact on
Challenges Currently Faced and Concluding the future of schizophrenia treatment.
Remarks

Clearly, many preclinical animal models exist for


research into schizophrenia neuropathology and Acknowledgment
drug development. The variety arises because an
This study was supported by the Science Foundation
ideal preclinical animal model of schizophrenia ­Ireland, Research Frontiers Programme 10/RFP/NES2744,
does not exist and each animal model focuses on and by a Health Research Board Clinician Scientist Award
a particular spectrum of the schizophrenia disor- (CSA/2012/8) to DRC.
144.82.108.120 - 10/2/2016 9:33:56 PM

68 Farrelly · Föcking · Cotter
Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
References
  1 Tandon R, Nasrallah HA, Keshavan MS: 15 Cannon M, Jones PB, Murray RM: Ob- 29 Nestler EJ, et al: Preclinical models: sta-
Schizophrenia, ‘just the facts’. 4. Clinical stetric complications and schizophrenia: tus of basic research in depression. Biol
features and conceptualization. historical and meta-analytic review. Am Psychiatry 2002;52:503–528.
Schizophr Res 2009;110:1–23. J Psychiatry 2002;159:1080–1092. 30 Matthysse S: Animal models in psychi-
  2 Eaton WW, et al: Case identification in 16 Clarke MC, et al: Predicting risk and the atric research. Prog Brain Res 1986;65:
psychiatric epidemiology: a review. Int emergence of schizophrenia. Psychiatr 259–270.
Rev Psychiatry 2007;19:497–507. Clin North Am 2012;35:585–612. 31 Willner P: The validity of animal models
  3 Cichon S, et al: Pharmacogenetics of 17 van der Does AJ, et al: A dimensional of depression. Psychopharmacology
schizophrenia. Am J Med Genet 2000; and categorical approach to the symp- 1984;83:1–16.
97:98–106. tomatology of recent-onset schizo- 32 Jones CA, Watson DJ, Fone KC: Animal
  4 Saha S, et al: A systematic review of the phrenia. J Nerv Ment Dis 1993; 181: models of schizophrenia. Br J Pharmacol
prevalence of schizophrenia. PLoS Med- 744–749. 2011;164:1162–1194.
icine 2005;2:e141. 18 Keshavan MS, et al: Schizophrenia, ‘just 33 van der Staay FJ: Animal models of be-
  5 Tandon R, Keshavan MS, Nasrallah HA: the facts’: what we know in 2008. 3. havioral dysfunctions: basic concepts
Schizophrenia, ‘just the facts’ what we Neurobiology. Schizophr Res 2008;106: and classifications, and an evaluation
know in 2008. 2. Epidemiology and eti- 89–107. strategy. Brain Res Rev 2006;52:131–159.
ology. Schizophr Res 2008;102:1–18. 19 Snyder EM, Murphy MR: Schizophrenia 34 Nestler EJ, Hyman SE: Animal models of
  6 Aleman A, Kahn RS, Selten JP: Sex dif- therapy: beyond atypical antipsychotics. neuropsychiatric disorders. Nat Neuro-
ferences in the risk of schizophrenia: Nat Rev Drug Discov 2008;7:471–472. sci 2010;13:1161–1169.
evidence from meta-analysis. Arch Gen 20 Harrison PJ, Weinberger DR: Schizo- 35 Cronbach LJ, Meehl PE: Construct valid-
Psychiatry 2003;60:565–571. phrenia genes, gene expression, and ity in psychological tests. Psychol Bull
  7 McGrath J, et al: A systematic review of neuropathology: on the matter of their 1955;52:281–302.
the incidence of schizophrenia: the dis- convergence. Mol Psychiatry 2005;10: 36 Marcotte ER, Pearson DM, Srivastava
tribution of rates and the influence of 40–68, image 5. LK: Animal models of schizophrenia: a
sex, urbanicity, migrant status and 21 Crow TJ: Molecular pathology of schizo- critical review. J Psychiatry Neurosci
methodology. BMC Med 2004;2:13. phrenia: more than one disease process? 2001;26:395–410.
  8 Kallmann FJ: The genetic theory of Br Med J 1980;280:66–68. 37 Brown AS, Derkits EJ: Prenatal infection
schizophrenia: an analysis of 691 schizo- 22 Kirkpatrick B, et al: The NIMH-­ and schizophrenia: a review of epidemi-
phrenic twin index families. Am J Psy- MATRICS consensus statement on ologic and translational studies. Am J
chiatry 1946;103:309–322. ­negative symptoms. Schizophr Bull Psychiatry 2010;167:261–280.
  9 Sullivan PF, Kendler KS, Neale MC: 2006;32:214–219. 38 Cotter D, et al: Does prenatal exposure
Schizophrenia as a complex trait: evi- 23 Carpenter WT Jr: Clinical constructs to influenza in mice induce pyramidal
dence from a meta-analysis of twin and therapeutic discovery. Schizophr cell disarray in the dorsal hippocampus?
studies. Arch Gen Psychiatry 2003;60: Res 2004;72:69–73. Schizophr Res 1995;16:233–241.
1187–1192. 24 Dickinson D, et al: General and specific 39 Fatemi SH, et al: Prenatal viral infection
10 Gottesman II, McGuffin P, Farmer AE: cognitive deficits in schizophrenia: Goli- in mouse causes differential expression
Clinical genetics as clues to the ‘real’ ath defeats David? Biol Psychiatry 2008; of genes in brains of mouse progeny: a
genetics of schizophrenia (a decade of 64:823–827. potential animal model for schizophre-
modest gains while playing for time). 25 Keefe RS, et al: Neurocognitive effects of nia and autism. Synapse 2005;57:91–99.
Schizophr Bull 1987;13:23–47. antipsychotic medications in patients 40 Meyer U, Feldon J: To poly(I:C) or not
11 Torrey EF, et al: Seasonality of births in with chronic schizophrenia in the to poly(I:C): advancing preclinical
schizophrenia and bipolar disorder: a ­CATIE Trial. Arch Gen Psychiatry 2007; schizophrenia research through the use
review of the literature. Schizophr Res 64:633–647. of prenatal immune activation models.
1997;28:1–38. 26 Tandon R, Nasrallah HA, Keshavan MS: Neuropharmacology 2012;62:1308–
12 Bradbury TN, Miller GA: Season of birth Schizophrenia, ‘just the facts’. 5. Treat- 1321.
in schizophrenia: a review of evidence, ment and prevention: past, present, and 41 Baharnoori M, Bhardwaj SK, Srivastava
methodology, and etiology. Psychol Bull future. Schizophr Res 2010;122:1–23. LK: Neonatal behavioral changes in rats
1985;98:569–594. 27 Lehman AF, et al: Practice guideline for with gestational exposure to lipopoly-
13 Schwartz PJ: Season of birth in schizo- the treatment of patients with schizo- saccharide: a prenatal infection model
phrenia: a maternal-fetal chronobiologi- phrenia, second edition. Am J Psychia- for developmental neuropsychiatric dis-
cal hypothesis. Med Hypotheses 2011; try 2004;161(suppl):1–56. orders. Schizophr Bull 2012;38:444–456.
76:785–793. 28 Davis KL, et al (eds): Neuropsychophar- 42 Ashdown H, et al: The role of cytokines
14 Meli G, et al: Prenatal and perinatal risk macology: The Fifth Generation of Prog- in mediating effects of prenatal infection
factors of schizophrenia. J Matern Fetal ress. An Official Publication of the on the fetus: implications for schizo-
Neonatal Med 2012;25:2559–2563. American College of Neuropsychophar- phrenia. Mol Psychiatry 2006;11:47–55.
macology. Philadelphia, Lippincott Wil-
liams & Wilkins, 2002.
144.82.108.120 - 10/2/2016 9:33:56 PM

Insights from Proteomic Studies on Schizophrenia Preclinical Models 69


Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
43 Meyer U: Prenatal poly(i:C) exposure 55 Sesack SR, Pickel VM: In the rat medial 70 Wasinger VC, Zeng M, Yau Y: Current
and other developmental immune acti- nucleus accumbens, hippocampal and status and advances in quantitative pro-
vation models in rodent systems. Biol catecholaminergic terminals converge on teomic mass spectrometry. Int J Pro-
Psychiatry 2014;75:307–315. spiny neurons and are in apposition to teomics 2013;2013:180605.
44 Zuckerman L, Weiner I: Post-pubertal each other. Brain Res 1990;527:266–279. 71 Gygi SP, Aebersold R: Mass spectrom-
emergence of disrupted latent inhibition 56 Chen XS, et al: Neonatal ventral hippo- etry and proteomics. Curr Opin Chem
following prenatal immune activation. campal lesion as a valid model of schizo- Biol 2000;4:489–494.
Psychopharmacology 2003;169:308– phrenia: evidence from sensory gating 72 Julka S, Regnier FE: Recent advance-
313. study. Chin Med J (Engl) 2012;125: ments in differential proteomics based
45 Ozawa K, et al: Immune activation dur- 2752–2754. on stable isotope coding. Brief Funct
ing pregnancy in mice leads to dopami- 57 Chambers RA, et al: Cognitive effects of Genomic Proteomic 2005;4:158–177.
nergic hyperfunction and cognitive im- neonatal hippocampal lesions in a rat 73 Mann M: Functional and quantitative
pairment in the offspring: a model of schizophrenia. Neuropsycho- proteomics using SILAC. Nat Rev Mol
neurodevelopmental animal model of pharmacology 1996;15:587–594. Cell Biol 2006;7:952–958.
schizophrenia. Biol Psychiatry 2006;59: 58 Matigian N, et al: Disease-specific, neu- 74 Shiio Y, Aebersold R: Quantitative pro-
546–554. rosphere-derived cells as models for teome analysis using isotope-coded af-
46 Becker A, et al: Transient prenatal vita- brain disorders. Dis Model Mech 2010; finity tags and mass spectrometry. Nat
min D deficiency is associated with sub- 3:785–798. Protoc 2006;1:139–145.
tle alterations in learning and memory 59 Horiuchi Y, et al: Olfactory cells via na- 75 Gygi SP, et al: Quantitative analysis of
functions in adult rats. Behav Brain Res sal biopsy reflect the developing brain in complex protein mixtures using isotope-
2005;161:306–312. gene expression profiles: utility and lim- coded affinity tags. Nat Biotechnol 1999;
47 McGrath JJ, et al: Developmental vita- itation of the surrogate tissues in re- 17:994–999.
min D deficiency and risk of schizophre- search for brain disorders. Neurosci Res 76 Bantscheff M, et al: Quantitative mass
nia: a 10-year update. Schizophr Bull 2013;77:247–250. spectrometry in proteomics: a critical
2010;36:1073–1078. 60 Brennand KJ, et al: Modelling schizo- review. Anal Bioanal Chem 2007;389:
48 Aguilar-Valles A, Flores C, Luheshi GN: phrenia using human induced pluripo- 1017–1031.
Prenatal inflammation-induced hypo- tent stem cells. Nature 2011;473:221– 77 Aggarwal K, Choe LH, Lee KH: Shotgun
ferremia alters dopamine function in the 225. proteomics using the iTRAQ isobaric
adult offspring in rat: relevance for 61 Brennand KJ, Gage FH: Concise review: tags. Brief Funct Genomic Proteomic
schizophrenia. PLoS One 2010;5:e10967. the promise of human induced pluripo- 2006;5:112–120.
49 Beard J: Iron deficiency alters brain de- tent stem cell-based studies of schizo- 78 Evans C, et al: An insight into iTRAQ:
velopment and functioning. J Nutr 2003; phrenia. Stem Cells 2011;29:1915–1922. where do we stand now? Anal Bioanal
133(suppl 1):1468S–1472S. 62 Girard SD, et al: Isolating nasal olfactory Chem 2012;404:1011–1027.
50 Erikson KM, et al: Iron deficiency de- stem cells from rodents or humans. J Vis 79 Merchant M, Weinberger SR: Recent
creases dopamine D1 and D2 receptors Exp 2011;54:2762. advancements in surface-enhanced laser
in rat brain. Pharmacol Biochem Behav 63 Pedrosa E, et al: Development of pa- desorption/ionization-time of flight-
2001;69:409–418. tient-specific neurons in schizophrenia mass spectrometry. Electrophoresis
51 Matsumoto H, Higa HH: Studies on using induced pluripotent stem cells. J 2000;21:1164–1177.
methylazoxymethanol, the aglycone of Neurogenet 2011;25:88–103. 80 Neilson KA, et al: Less label, more free:
cycasin: methylation of nucleic acids in 64 Wilkins MR, et al: Guidelines for the approaches in label-free quantitative
vitro. Biochem J 1966;98:20C–22C. next 10 years of proteomics. Proteomics mass spectrometry. Proteomics 2011;11:
52 Lodge DJ: The MAM rodent model of 2006;6:4–8. 535–553.
schizophrenia. Curr Protoc Neurosci 65 Craft GE, Chen A, Nairn AC: Recent 81 Craft GE, Chen A, Nairn AC: Recent
2013;chapt 9:unit 9.43. advances in quantitative neuropro- advances in quantitative neuropro-
53 Du Y, Grace AA: Peripubertal diazepam teomics. Methods 2013;61:186–218. teomics. Methods 2013;61:186–218.
administration prevents the emergence 66 Aldred S, Grant MM, Griffiths HR: The 82 Lakhan SE, Kramer A: Schizophrenia
of dopamine system hyperresponsivity use of proteomics for the assessment of genomics and proteomics: are we any
in the MAM developmental disruption clinical samples in research. Clin Bio- closer to biomarker discovery? Behav
model of schizophrenia. Neuropsycho- chem 2004;37:943–952. Brain Funct 2009;5:2.
pharmacology 2013;38:1881–1888. 67 O’Farrell PH: High resolution two-di- 83 He Y, et al: Schizophrenia shows a
54 Jaskiw GE, et al: Effect of ibotenic acid mensional electrophoresis of proteins. J unique metabolomics signature in plas-
lesions of the medial prefrontal cortex Biol Chem 1975;250:4007–4021. ma. Transl Psychiatry 2012;2:e149.
on amphetamine-induced locomotion 68 Gygi SP, et al: Evaluation of two-dimen- 84 Cai HL, et al: Metabolomic analysis of
and regional brain catecholamine con- sional gel electrophoresis-based pro- biochemical changes in the plasma and
centrations in the rat. Brain Res 1990; teome analysis technology. Proc Natl urine of first-episode neuroleptic-naive
534:263–272. Acad Sci USA 2000;97:9390–9395. schizophrenia patients after treatment
69 Yates JR, Ruse CI, Nakorchevsky A: Pro- with risperidone. J Proteome Res 2012;
teomics by mass spectrometry: ap- 11:4338–4350.
proaches, advances, and applications.
Annu Rev Biomed Eng 2009;11:49–79.
144.82.108.120 - 10/2/2016 9:33:56 PM

70 Farrelly · Föcking · Cotter
Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
85 Jaros JA, et al: Protein phosphorylation   97 Schubert KO, et al: Hypothesis review: 108 Novak G, Seeman P, Tallerico T: In-
patterns in serum from schizophrenia are clathrin-mediated endocytosis and creased expression of calcium/calmod-
patients and healthy controls. J Pro- clathrin-dependent membrane and ulin-dependent protein kinase IIβ in
teomics 2012;76(spec No):43–55. protein trafficking core pathophysi- frontal cortex in schizophrenia and
86 Chan MK, et al: Evidence for disease and ological processes in schizophrenia depression. Synapse 2006;59:61–68.
antipsychotic medication effects in post- and bipolar disorder? Mol Psychiatry 109 Manji H, et al: Impaired mitochondrial
mortem brain from schizophrenia pa- 2012;17:669–681. function in psychiatric disorders. Nat
tients. Mol Psychiatry 2011;16:1189–   98 Herberth M, et al: Impaired glycolytic Rev Neurosci 2012;13:293–307.
1202. response in peripheral blood mono- 110 Martins-de-Souza D, et al: The applica-
87 Johnston-Wilson NL, et al: Disease-spe- nuclear cells of first-onset antipsychot- tion of selective reaction monitoring
cific alterations in frontal cortex brain ic-naive schizophrenia patients. Mol confirms dysregulation of glycolysis in
proteins in schizophrenia, bipolar disor- Psychiatry 2011;16:848–859. a preclinical model of schizophrenia.
der, and major depressive disorder. The   99 Filiou MD, Turck CW, Martins-de- BMC Res Notes 2012;5:146.
Stanley Neuropathology Consortium. Souza D: Quantitative proteomics for 111 Marais L, et al: A proteomic analysis of
Mol Psychiatry 2000;5:142–149. investigating psychiatric disorders. the ventral hippocampus of rats sub-
88 Prabakaran S, et al: Mitochondrial dys- Proteomics Clin Appl 2011;5:38–49. jected to maternal separation and esci-
function in schizophrenia: evidence for 100 Martins-de-Souza D: Proteome and talopram treatment. Metab Brain Dis
compromised brain metabolism and transcriptome analysis suggests oligo- 2009;24:569–586.
oxidative stress. Mol Psychiatry 2004;9: dendrocyte dysfunction in schizophre- 112 McCullumsmith RE, Clinton SM,
684–697, 643. nia. J Psychiatr Res 2010;44:149–156. Meador-Woodruff JH: Schizophrenia
89 Beasley CL, et al: Proteomic analysis of 101 Jentsch JD, Roth RH: The neuropsy- as a disorder of neuroplasticity. Int Rev
the anterior cingulate cortex in the ma- chopharmacology of phencyclidine: Neurobiol 2004;59:19–45.
jor psychiatric disorders: evidence for from NMDA receptor hypofunction to 113 Bitanihirwe BK, Woo TU: Oxidative
disease-associated changes. Proteomics the dopamine hypothesis of schizo- stress in schizophrenia: an integrated
2006;6:3414–3425. phrenia. Neuropsychopharmacology approach. Neurosci Biobehav Rev
90 Clark D, et al: Altered proteins of the 1999;20:201–225. 2011;35:878–893.
anterior cingulate cortex white matter 102 Morris BJ, Cochran SM, Pratt JA: PCP: 114 Martins-de-Souza D, et al: The role of
proteome in schizophrenia. Proteomics from pharmacology to modelling energy metabolism dysfunction and
Clin Appl 2007;1:157–166. schizophrenia. Curr Opin Pharmacol oxidative stress in schizophrenia re-
91 Pennington K, et al: Prominent synaptic 2005;5:101–106. vealed by proteomics. Antioxid Redox
and metabolic abnormalities revealed by 103 Ernst A, et al: Molecular validation of Signal 2011;15:2067–2079.
proteomic analysis of the dorsolateral the acute phencyclidine rat model for 115 Daniels WM, et al: Exercise normalizes
prefrontal cortex in schizophrenia and schizophrenia: identification of trans- altered expression of proteins in the
bipolar disorder. Mol Psychiatry 2008; lational changes in energy metabolism ventral hippocampus of rats subjected
13:1102–1117. and neurotransmission. J Proteome to maternal separation. Exp Physiol
92 Sivagnanasundaram S, et al: Abnormal Res 2012;11:3704–3714. 2012;97:239–247.
pathways in the genu of the corpus cal- 104 Pickering C, Ericson M, Söderpalm B: 116 Dimatelis JJ, et al: Effects of maternal
losum in schizophrenia pathogenesis: a Chronic phencyclidine increases syn- separation and methamphetamine
proteome study. Proteomics Clin Appl apsin-1 and synaptic adaptation pro- exposure on protein expression in the
2007;1:1291–1305. teins in the medial prefrontal cortex. nucleus accumbens shell and core.
93 English JA, et al: 2-D DIGE analysis im- ISRN Psychiatry 2013;2013:620361. Metab Brain Dis 2012;27:363–375.
plicates cytoskeletal abnormalities in 105 English JA, et al: The neuroproteomics 117 Duman CH, et al: Voluntary exercise
psychiatric disease. Proteomics 2009;9: of schizophrenia. Biol Psychiatry 2011; produces antidepressant and anxiolytic
3368–3382. 69:163–172. behavioral effects in mice. Brain Res
94 Föcking M, et al: Common proteomic 106 Wesseling H, et al: A combined meta- 2008;1199:148–158.
changes in the hippocampus in schizo- bonomic and proteomic approach 118 Ströhle A: Physical activity, exercise,
phrenia and bipolar disorder and par- identifies frontal cortex changes in a depression and anxiety disorders. J
ticular evidence for involvement of cor- chronic phencyclidine rat model in Neural Transm 2009;116:777–784.
nu ammonis regions 2 and 3. Arch Gen relation to human schizophrenia brain 119 Kitteringham NR, et al: Multiple reac-
Psychiatry 2011;68:477–488. pathology. Neuropsychopharmacology tion monitoring for quantitative bio-
95 Martins-de-Souza D, et al: Proteomic 2013;38:2532–2544. marker analysis in proteomics and
analysis of dorsolateral prefrontal cortex 107 Braunewell KH: The darker side of Ca2+ metabolomics. J Chromatogr B Analyt
indicates the involvement of cytoskeleton, signaling by neuronal Ca2+-sensor pro- Technol Biomed Life Sci 2009;877:
oligodendrocyte, energy metabolism and teins: from Alzheimer’s disease to can- 1229–1239.
new potential markers in schizophrenia. J cer. Trends Pharmacol Sci 2005;26: 120 Fone KC, Porkess MV: Behavioural
Psychiatr Res 2009;43:978–986. 345–351. and neurochemical effects of post-
96 Davis KL, et al: White matter changes in weaning social isolation in rodents:
schizophrenia: evidence for myelin-re- relevance to developmental neuropsy-
lated dysfunction. Arch Gen Psychiatry chiatric disorders. Neurosci Biobehav
2003;60:443–456. Rev 2008;32:1087–1102.
144.82.108.120 - 10/2/2016 9:33:56 PM

Insights from Proteomic Studies on Schizophrenia Preclinical Models 71


Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
121 Roncada P, et al: Gating deficits in iso- 133 Smalla KH, et al: A comparison of the 145 Burne TH, et al: Transient prenatal
lation-reared rats are correlated with synaptic proteome in human chronic vitamin D deficiency is associated with
alterations in protein expression in schizophrenia and rat ketamine psy- hyperlocomotion in adult rats. Behav
nucleus accumbens. J Neurochem chosis suggest that prohibitin is in- Brain Res 2004;154:549–555.
2009;108:611–620. volved in the synaptic pathology of 146 Guest PC, et al: Proteomic analysis of
122 Aitken A: Functional specificity in 14- schizophrenia. Mol Psychiatry 2008; the maternal protein restriction rat
3–3 isoform interactions through di- 13:878–896. model for schizophrenia: identification
mer formation and phosphorylation: 134 Kapur S, Seeman P: NMDA receptor of translational changes in hormonal
chromosome location of mammalian antagonists ketamine and PCP have signaling pathways and glutamate neu-
isoforms and variants. Plant Mol Biol direct effects on the dopamine D2 and rotransmission. Proteomics 2012;12:
2002;50:993–1010. serotonin 5-HT2 receptors: implica- 3580–3589.
123 van Os J, Selten JP: Prenatal exposure tions for models of schizophrenia. Mol 147 Guest PC, et al: Altered levels of circu-
to maternal stress and subsequent Psychiatry 2002;7:837–844. lating insulin and other neuroendo-
schizophrenia: the May 1940 invasion 135 Zhou K, et al: NMDA receptor hypo- crine hormones associated with the
of The Netherlands. Br J Psychiatry function induces dysfunctions of en- onset of schizophrenia. Psychoneuro-
1998;172:324–326. ergy metabolism and semaphorin sig- endocrinology 2011;36:1092–1096.
124 Mairesse J, et al: Proteomic character- naling in rats: a synaptic proteome 148 Patton SM, et al: Quantitative pro-
ization in the hippocampus of prena- study. Schizophr Bull 2012;38:579– teomic analyses of cerebrospinal fluid
tally stressed rats. J Proteomics 2012; 591. using iTRAQ in a primate model of
75:1764–1770. 136 Lipska BK: Using animal models to test iron deficiency anemia. Dev Neurosci
125 Carboni L, et al: Proteomic analysis of a neurodevelopmental hypothesis of 2012;34:354–365.
rat hippocampus after repeated psy- schizophrenia. J Psychiatry Neurosci 149 Lozoff B, et al: Functional significance
chosocial stress. Neuroscience 2006; 2004;29:282–286. of early-life iron deficiency: outcomes
137:1237–1246. 137 Vercauteren FG, et al: An organelle at 25 years. J Pediatr 2013;163:1260–
126 Hradetzky E, et al: The methylazoxy- proteomic method to study neuro- 1266.
methanol acetate (MAM-E17) rat transmission-related proteins, applied 150 Lozoff B, et al: Long-lasting neural and
model: molecular and functional ef- to a neurodevelopmental model of behavioral effects of iron deficiency in
fects in the hippocampus. Neuropsy- schizophrenia. Proteomics 2007;7: infancy. Nutr Rev 2006;64(pt 2):S34–
chopharmacology 2012;37:364–377. 3569–3579. S43, discussion S72–S91.
127 Lee HK, et al: Phosphorylation of the 138 Brown AS, Susser ES: Prenatal nutri- 151 English JA, et al: Omega-3 fatty acid
AMPA receptor GluR1 subunit is re- tional deficiency and risk of adult deficiency disrupts endocytosis, neuri-
quired for synaptic plasticity and reten- schizophrenia. Schizophr Bull 2008;34: togenesis, and mitochondrial protein
tion of spatial memory. Cell 2003;112: 1054–1063. pathways in the mouse hippocampus.
631–643. 139 Susser E, et al: Schizophrenia after pre- Front Genet 2013;4:208.
128 Hammond JC, et al: Evidence for ab- natal famine: further evidence. Arch 152 Meyer U, Feldon J: Neural basis of psy-
normal forward trafficking of AMPA Gen Psychiatry 1996;53:25–31. chosis-related behaviour in the infec-
receptors in frontal cortex of elderly 140 Xu MQ, et al: Prenatal malnutrition tion model of schizophrenia. Behav
patients with schizophrenia. Neuro- and adult schizophrenia: further evi- Brain Res 2009;204:322–334.
psychopharmacology 2010;35:2110– dence from the 1959–1961 Chinese 153 Li Q, et al: Prenatal immune challenge
2119. famine. Schizophr Bull 2009;35:568– is an environmental risk factor for
129 Mohn AR, et al: Mice with reduced 576. brain and behavior change relevant to
NMDA receptor expression display 141 Insel BJ, et al: Maternal iron deficiency schizophrenia: evidence from MRI in a
behaviors related to schizophrenia. and the risk of schizophrenia in off- mouse model. PLoS One 2009;4:e6354.
Cell 1999;98:427–436. spring. Arch Gen Psychiatry 2008;65: 154 Boksa P: Effects of prenatal infection
130 Paulson L, et al: Comparative pro- 1136–1144. on brain development and behavior: a
teome analysis of thalamus in MK- 142 Harvey L, Boksa P: Do prenatal im- review of findings from animal mod-
801-treated rats. Proteomics 2004;4: mune activation and maternal iron els. Brain Behav Immun 2010;24:881–
819–825. deficiency interact to affect neurode- 897.
131 Paulson L, et al: Comparative genome velopment and early behavior in rat 155 Smith SE, et al: Maternal immune acti-
and proteome analysis of cerebral cor- offspring? Brain Behav Immun 2013; vation alters fetal brain development
tex from MK-801-treated rats. J Neu- 35:144–154. through interleukin-6. J Neurosci 2007;
rosci Res 2003;71:526–533. 143 Almeras L, et al: Developmental vita- 27:10695–10702.
132 Paulson L, et al: Effects on rat thalamic min D deficiency alters brain protein 156 Matijevic T, Pavelic J: Poly(I:C) treat-
proteome by acute and subchronic expression in the adult rat: implica- ment influences the expression of cal-
MK-801-treatment. Eur J Pharmacol tions for neuropsychiatric disorders. reticulin and profilin-1 in a human
2004;505:103–109. Proteomics 2007;7:769–780. HNSCC cell line: a proteomic study.
144 McGrath J, et al: Protein expression in Tumour Biol 2012;33:1201–1208.
the nucleus accumbens of rats exposed
to developmental vitamin D deficiency.
PLoS One 2008;3:e2383.
144.82.108.120 - 10/2/2016 9:33:56 PM

72 Farrelly · Föcking · Cotter
Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)
157 Grunert T, et al: A comparative pro- 163 Leucht S, Kissling W, McGrath J: Lithi- 170 Kashem MA, et al: Effects of typical
teome analysis links tyrosine kinase 2 um for schizophrenia revisited: a sys- (haloperidol) and atypical (risperi-
(Tyk2) to the regulation of cellular tematic review and meta-analysis of done) antipsychotic agents on protein
glucose and lipid metabolism in re- randomized controlled trials. J Clin expression in rat neural stem cells.
sponse to poly(I:C). J Proteomics 2011; Psychiatry 2004;65:177–186. Neurochem Int 2009; 55: 558–565.
74:2866–2880. 164 Schechtman LM: Implementation of 171 Ji B, et al: A comparative proteomics
158 Deng MY, et al: Frontal-subcortical the 3Rs (refinement, reduction, and analysis of rat mitochondria from the
protein expression following prenatal replacement): validation and regula- cerebral cortex and hippocampus in
exposure to maternal inflammation. tory acceptance considerations for al- response to antipsychotic medica-
PLoS One 2011;6:e16638. ternative toxicological test methods. tions. J Proteome Res 2009; 8: 3633–
159 Bader V, et al: Proteomic, genomic and ILAR J 2002;43(suppl):S85–S94. 3641.
translational approaches identify 165 Fan Y, et al: Altered cell cycle dynam- 172 Ma D, et al: Antipsychotic treatment
CRMP1 for a role in schizophrenia and ics in schizophrenia. Biol Psychiatry alters protein expression associated
its underlying traits. Hum Mol Genet 2012;71:129–135. with presynaptic function and nervous
2012;21:4406–4418. 166 Bro C, et al: Transcriptional, pro- system development in rat frontal cor-
160 Hikita T, et al: Proteomic analysis teomic, and metabolic responses to tex. J Proteome Res 2009; 8: 3284–
reveals novel binding partners of lithium in galactose-grown yeast cells. 3297.
dysbindin, a schizophrenia-related J Biol Chem 2003;278:32141–32149. 173 Martins-de-Souza D, et al: Proteome
protein. J Neurochem 2009; 110: 167 O’Brien E, et al: Effects of chronic ris- analyses of cultured astrocytes treated
1567–1574. peridone treatment on the striatal pro- with MK-801 and clozapine: similari-
161 Costa E, et al: REELIN and schizophre- tein profiles in rats. Brain Res 2006; ties with schizophrenia. Eur Arch Psy-
nia: a disease at the interface of the 1113:24–32. chiatry Clin Neurosci 2011;261:217–
genome and the epigenome. Mol In- 168 La Y, et al: Hippocampus protein profil- 228.
terv 2002;2:47–57. ing reveals aberration of malate dehy- 174 Ahmed EU, et al: Antipsychotic in-
162 Schmitt A, et al: Proteomic similarities drogenase in chlorpromazine/clozapine duced alteration of growth and pro-
between heterozygous reeler mice and treated rats. Neurosci Lett 2006;408: teome of rat neural stem cells. Neuro-
schizophrenia. Biol Psychiatry 2013; 29–34. chem Res 2012;37:1649–1659.
74:e5–e10. 169 Chen ML, Chen CH: Comparative pro-
teome analysis revealed up-regulation
of transthyretin in rat brain under
chronic clozapine treatment. J Psychi-
atr Res 2007;41:63–68.

Lorna A. Farrelly, PhD


Department of Psychiatry, Royal College of Surgeons in Ireland
Smurfit Education and Research Centre
Beaumont Hospital, Beaumont, Dublin 9 (Ireland)
E-Mail lornafarrelly@gmail.com
144.82.108.120 - 10/2/2016 9:33:56 PM

Insights from Proteomic Studies on Schizophrenia Preclinical Models 73


Downloaded by:

Martins-de-Souza D (ed): Proteomics and Metabolomics in Psychiatry.


UCL

Adv Biol Psychiatry. Basel, Karger, 2014, vol 29, pp 56–73 (DOI: 10.1159/000358028)

You might also like