Abstract
Sensorimotor gating, or the ability of a sensory event to suppress a motor response, can be measured operationally via prepulse inhibition (PPI) of the startle response. PPI is deficient in schizophrenia patients as well as other neuropsychiatric disorders, can be measured across species, and has been used widely as a translational tool in preclinical neuropharmacological and genetic research. First developed to assess drug effects in pharmacological and developmental models, PPI has become one of the standard behavioral measures in genetic models of schizophrenia and other neuropsychiatric disorders that exhibit PPI deficits. In this chapter we review the literature on genetic models of sensorimotor gating and discuss the utility of PPI as a tool in phenotyping mutant mouse models. We highlight the approaches to genetic mouse models of neuropsychiatric disease, discuss some of the important caveats to these approaches, and provide a comprehensive table covering the more recent genetic models that have evaluated PPI.
Keywords: Prepulse inhibition, Startle, Mouse models, Schizophrenia, Genetic, Mutant
1 Introduction: Definitions and Measures of Sensorimotor Gating
Sensorimotor gating refers to the regulation of sensory information as it is transmitted to motor output systems. When sensory information is processed centrally, it requires some degree of filtering or “gating” prior to accessing and impinging upon motor output. While this process has long been observed at multiple levels of biology, the synaptic and cellular mechanism of sensorimotor gating (Frost et al. 2003; Nusbaum and Contreras 2004; Rose and Scott 2003) as well as the functional implications (Braff 2010, 2011; Swerdlow et al. 2008) are starting to be elucidated. One form of sensorimotor gating that is widely studied in humans and animals is prepulse inhibition (PPI) of startle. PPI is a form of startle plasticity in which presentation of a weak stimulus (prepulse) preceding an intense startling stimulus (pulse) by 30–500 ms inhibits the startle response (Graham 1975; Hoffman and Ison 1980). The fundamental mechanism underlying this inhibition is thought to resemble the normal process of filtering incoming sensory stimuli (Geyer and Braff 1987). PPI levels may indicate the current integrity of sensorimotor gating mechanisms, providing an operational measure of sensorimotor gating. In humans, startle is measured from the eye blink response through electromyographic recordings of the orbicularis oculi muscle (Fridlund and Cacioppo 1986) and typically involves startle to acoustic stimuli, although tactile stimuli have been used as well (Braff et al. 1992; Kumari et al. 2003; Neuner et al. 2010; Swerdlow et al. 2001b). PPI deficits are observed in schizophrenia patients [for review see (Braff et al. 2001; Swerdlow et al. 2008)], their unaffected first degree relatives (Cadenhead et al. 2000), and patients with schizotypal personality disorder (Cadenhead et al. 1993). Additionally, several other neuropsychiatric disorders are associated with decreased PPI, including Obsessive–Compulsive Disorder (Swerdlow et al. 1993), Tourette’s syndrome (Swerdlow et al. 2001b), Huntington’s disease (Swerdlow et al. 1995), manic bipolar patients (Perry et al. 2001), Panic Disorder (Ludewig et al. 2002), Fragile X syndrome (Frankland et al. 2004; Hessl et al. 2009), and adults with autism (Perry et al. 2007). Although the core symptoms of these disorders are diverse, a feature common to all of them is deficient gating, with a gating deficit predominating in the cognitive sphere in some disorders and in the sensory or motor domains in others. Thus, deficient gating has been reported across a variety of neuropsychiatric disorders, with PPI deficits in schizophrenia patients being the best characterized and the most widely replicated (Braff et al. 2001; Kumari et al. 2008; Ludewig et al. 2003; Mackeprang et al. 2002; Swerdlow et al. 2008).
Sensorimotor gating abnormalities, as measured by PPI, are being used as an endophenotype in genetic studies of schizophrenia (Braff et al. 2007; Greenwood et al. 2011), and meet the criteria outlined for a viable endophenotype (Turetsky et al. 2007). With an increased focus on observable and measurable behaviors as rational approaches to genetic studies of heterogeneous neuropsychiatric diseases such as schizophrenia (Gottesman and Gould 2003), large-scale genetic studies are examining neurophysiological measures such as PPI, P50 auditory evoked suppression, antisaccade eye movement, mismatch negativity, and P300 event-related potential (Turetsky et al. 2007). Recent studies suggest that PPI is heritable (Hasenkamp et al. 2010) and associated with polymorphisms in the CHRNA3 gene (Petrovsky et al. 2010), neuregulin 1 (Roussos et al. 2011), and COMT (Giakoumaki et al. 2008; Quednow et al. 2008; Roussos et al. 2008). The use of PPI as an endophenotype in genetic studies of schizophrenia, combined with the observation that PPI has a strong genetic component in mice (Francis et al. 2003), indicates that PPI may be a useful behavioral phenotype to consider in genetic mouse models related to neuropsychiatric disease, particularly schizophrenia. While there are certainly many other symptoms, behavioral traits, and neurophysiological deficits observed across the heterogeneous group of patients with schizophrenia, PPI appears to be a viable endophenotype for human genetic studies and thus a reasonable approach to investigate in genetic animal models. Additionally, considering that PPI measures a fundamental component of information processing and is observable across many species, it is a useful endpoint with which to understand the more general impact of specific genes on neurobehavioral function and on the neural substrates underlying this function.
Mutant mouse models related to schizophrenia have been based primarily on the pathophysiology of schizophrenia, the known effects of antipsychotic drugs, and candidate genes for schizophrenia. In this review, we provide an overview of PPI in genetic mouse models, concentrating on the time period since the Swerdlow et al. (2008) review. We discuss the contribution and usefulness of PPI as a phenotype in the context of genetic mouse models and speculate about the significance of PPI deficits and future directions for the field. There have been previous reviews on candidate genes for schizophrenia (Arguello and Gogos 2010; O’Tuathaigh et al. 2007), and reviews focusing specifically on PPI, which summarized studies of strain differences, genetic mutants, and the pharmacology of PPI in mice (Geyer et al. 2002; Powell et al. 2009; Swerdlow et al. 2008). In this review, we highlight the approaches to genetic mouse models of neuropsychiatric disease, discuss some of the important caveats to these approaches, and provide a comprehensive table covering the genetic models that have evaluated PPI since the Swerdlow et al. (2008) review. To that end, we discuss PPI as a phenotype in genetic mouse models generated to address hypotheses regarding the pathophysiology of neuropsychiatric disease, candidate genes, and basic proteins involved in neural development or synaptic function. We also discuss the usefulness of PPI in phenotype-driven approaches in which a PPI phenotype could lead to “bottom up” approaches of identifying novel genes of relevance to PPI and/or neuropsychiatric disease [i.e. hypothesis-generating; or phenotype-genotype approaches (Jacobson and Cryan 2010)].
2 Utility of Prepulse Inhibition Measures in Genetic Models of Schizophrenia
Mutant mouse models of neuropsychiatric disease have targeted genes involved in the pathophysiology of the disease, candidate or susceptibility genes thought to be involved in the etiology of the disease, or genes involved in basic physiological processes. Because many of the target genes overlap at the functional level across different disorders (e.g. proteins involved in basic processes of neurodevelopment), manipulation of these genes may have relevance to many neuropsychiatric disorders. Thus, it is important to determine the relevant behavioral tests to best understand the underlying brain abnormalities resulting from genetic alterations of proteins. Second, it is critical to develop animal models to screen novel therapeutics for specific domains of function. In this context, PPI has emerged as a useful behavior with which to assess the integrity of basic neural circuits in genetic mouse models and to screen for pharmacological agents, particularly in the context of identifying treatments for schizophrenia.
2.1 Relationship of PPI to Psychiatric Symptoms, Neurocognitive Measures, and Overall Function
Attempts to relate PPI deficits to the positive, negative, and cognitive symptoms of schizophrenia have yielded mixed results (Thaker 2007). PPI negatively correlates with thought disorder (Meincke et al. 2004; Perry and Braff 1994; Perry et al. 1999) and distractibility (Karper et al. 1996) in schizophrenia. PPI increases S. B. Powell et al. observed with risperidone treatment correlated with improvements in PANSS general psychopathology subscale scores (Martinez-Gras et al. 2009) in schizophrenia patients. In a well-powered study with over 300 subjects, PPI did not correlate with cognitive measures using traditional “pen and paper” tests [i.e. Wisconsin card sorting task (WCST), California verbal learning task, etc.], but did correlate positively with global assessment of function (GAF) and Independent Living scales (Swerdlow et al. 2006). There have been some studies demonstrating a relationship between cognitive constructs and PPI levels. For example, converging evidence indicates that PPI is correlated with strategy formation and execution time in the Cambridge Neuropsychological Test Automated Battery (CANTAB) in healthy controls (Bitsios et al. 2006; Csomor et al. 2008; Giakoumaki et al. 2006), a finding that should be examined further in patient populations. The cognitive neuroscience treatment research to improve cognition in Schizophrenia (CNTRICS) program funded by the National institute of mental health considered PPI to provide a measure of the cognitive construct of “gain control” as a specific aspect of the perceptual abnormalities seen in patients with schizophrenia (Green et al. 2009). The series of CNTRICS workshops concluded that PPI may have utility as a biomarker for use in proof of concept studies of potential treatments for the cognitive deficits in schizophrenia that are not ameliorated by existing antipsychotic drugs. For the purpose of evaluating genetic mouse models of neuropsychiatric disease, the more useful comparison to make is not between PPI and specific symptoms of schizophrenia, but rather the relationship between a gene and the observable dependent measure, i.e. PPI. The approach of using endophenotypes in genetic studies has greatly strengthened the ability to conduct cross-species translational studies by providing specific observables for study in experimental animals [reviewed in (Geyer and Markou 2002; Gould and Gottesman 2006) Waddington, chapter in this book]. Useful endophenotypes in this context are measures that are observed in humans and can be measured in animals.
2.2 PPI as an Indicator of Neural Processes and a Pharmacological Screen
A PPI deficit in a genetic mutant could indicate that the gene may be involved in the neural circuitry known to modulate PPI [e.g. cortical, limbic, striatal (Swerdlow et al. 2001a)]; in other words it could function as a “surrogate measure for neural processes” as Swerdlow et al. (2008) suggest. While a PPI deficit per se is not indicative of altered striatal or limbic circuitry, the presence of the deficit may suggest that these brain regions are affected by the genetic manipulation and provide a reasonable starting place for further hypothesis testing regarding the neurobiological implications of the genetic manipulation. Additionally, mutant mouse models offer the opportunity to screen putative antipsychotics that may involve a novel target and avoid the problems of receptor tautology inherent in many pharmacological studies [e.g. dopamine agonist-induced disruption blocked by a dopamine antagonist; as discussed in Powell et al. (2009). Using mutant mice to screen for putative antipsychotics may provide a means to develop novel drug targets not achieved with current pharmacological disruptions of PPI (see Table 1 for examples).
Table 1.
References | Mouse strain, sex | Model description/background/ rationale |
Basal PPI | Effects of drugs or manipulations typically used to induce PPI-deficits |
Effects of (presumed) antipsychotics/other treatments |
---|---|---|---|---|---|
DISC1 | |||||
Hikida et al. (2007) | DN-DISC1 TG, M | DISC1 is a potential SZ susceptibility gene |
↓PPI in TG versus WT at lowest PP intensity only |
||
Ibi et al. (2010) | DN-DISC1 TG, combined with neonatal PolyI:C 5 mg/kg or saline (PND2-6), M & F |
Two-hit model of SZ, genetic susceptibility combined with environmental insult (immune activation) |
ØPPI & ØPA in TG versus WT |
||
Niwa et al. (2010) | DISC1 KD via in utero gene transfer (E14), M & F |
DISC1 plays important role in brain development. In utero gene transfer allows for analysis of DISC1 function during development and impact on adult brain function |
↓PPI & ØPA in KD versus WT at P56, ØPPI at P28, effect slightly less in F compared to M |
CLO: ↑PPI & ØPA in KD; ØPPI & ØPA in WT |
|
Lipina et al. (2010) | DISC1L100P point mutations |
Missense mutation in exon 2 of DISC1 at L100P in mice |
↓PPI & ↓PA in mutant versus WT |
AMP (0.5 mg/kg) ↓PPI in mutant but ØPPI in WT |
HAL: ↑PPI in mutant (↑PPI in WT at PP16); ↓PA in WT, ÆPA in mutant |
Lipina et al. (2011) | DISC1L100P point mutation (combined with GSK3 mutant) |
Missense mutation in exon 2 of DISC1 at L100P in mice; DISC1L100P mice crossed with Glycogen synthase kinase-3 (GSK-3), a serine/threonine protein kinase that interacts with the N-terminal region of DISC1 |
↓PPI in DISC1L100P mutant vs. WT; ØPPI in DISC1L100P × GSK-3α HET |
TDZD-8 (GSK-3 inhibitor): ↑PPI & ØPA in DISC1L100P mutant; ØPPI & ØPA in WT |
|
Reelin | |||||
Barr et al. (2008) | Reelin KO, M | Reelin levels in brains of SZ are reduced |
Ø(unimodal acoustic) PPI, but ↓crossmodal PPI at 100 ms PP intervals, PPP at PP intervals ≤ 60 ms, ↓PA habituation & ↑PA at 100–110 dB(A) intensity for +/− versus WT |
||
Neuregulin | |||||
Chen et al. (2008) | Type III NRG1 mutants, M | NRG1 may represent a SZ susceptibility gene. Involved in cell-cell interaction; signals via erbB TK |
↓PPI & ØPA in +/− versus WT |
Chronic NIC (↑PPI in +/−; ØPPI in WT) | |
Duffy et al. (2008) | EGF-like domain NRG1 mutants, M |
The EGF-like domain of NRG1 is sufficient for NRG’s activity |
ØPPI in −/− versus WT | MK801 (↓PPI in −/− versus WT); AMPH (↓PPI in −/−and WT; no significant differences between −/− & WT) |
|
Deakin et al. (2009) | NRG1Type 1-Tg | Type I NRG1 increased in Hpc and PFC of SZ patients |
↓PPI & ↑PA in Tg versus WT |
||
Barros et al. (2009) | ERBB2/B4-CNS-specific KO, M & F |
NRG1 and ERBB4 are candidate genes for SZ |
↓PPI in KO versus WT (M only; ØPPI in F); PA data not shown |
CLO ↑PPI in KO; ØPPI in WT CLO ↑ spine density |
|
Wen et al. (2010) | PV-Cre; ErbB4 KO | In adulthood NRG1 and ErbB4 may regulate neurotransmission. Evidence that ErbB4 is located at the postsynaptic density of excitatory neurons, likely on GABA interneurons |
↓PPI & ØPA in −/− versus WT |
DIAZ: ↑PPI in −/− mice; ØPPI in WT; PA data not shown |
|
Kato et al. (2010) | NRG1 TG5, TG7, M & F | Postmortem studies indicate increased NRG1 in SZ brain. Transgenic mutants to examine hyper NRG1 signaling |
ØPPI & ØPA in TG versus WT |
||
COMT | |||||
Papaleo et al. (2008) | COMT KO, COMT Val-Tg | Polymorphisms in COMT gene reported in SZ; mice overexpress human COMT-Val variant |
ØPPI in −/− or TG versus WT; ↑PA in −/− versus WT, ↓PA in TG versus WT |
||
Tammimäki et al. (2010) | S-COMT KO, M & F | Two isoforms of COMT, S-COMT and MB-COMT, differentially contribute to DA function in the frontal cortex |
ØPPI & ↑PA in −/− versus WT (although lack of ↑PA with ↑ dB in WT mice) |
||
G-alpha | |||||
Kelly et al. (2009) | Forebrain-specific Galpha OE, M, F |
↑mRNA G-protein subunit Galpha levels, genetically linked to SZ |
↓PPI & ØPA in +/− versus WT; developmental or adult expression ØPPI; ↓PPI correlates with ↓CTX & HPC cAMP levels |
HAL ↑PPI in OE; ØPPI in WT Rolipram (↑cAMP levels): ↑PPI in OE; ØPPI in WT |
|
G72/G30 | |||||
Otte et al. (2009) | G72Tg (G72/G30 “humanized” BAC transgenic) |
G72/G30 gene locus implicated in SZ, BD, and panic disorder |
↓PPI & ØPA in Tg versus WT |
HAL ↑PPI in Tg; ØPPI in WT (slight ↑PPI at low PP intensity in WT); PA data not reported |
|
SMARCA | |||||
Koga et al. (2009) | SMARCA2 KO, M & F | SMARCA encodes BRM, a chromatin remodeling multi- protein complex; highly expressed in differentiating neurons; SNP in gene found in SZ (this paper) |
↓PPI in −/− versus WT; PA data not shown |
||
Neurexin | |||||
Etherton et al. (2009) | Neurexin-1α KO, M & F | CNVs for neurexin-1 α identified in SZ & Autism. Neurexins bind to neuroligins and function as synaptic recognition molecules |
↓PPI in −/− versus WT; ↑PA to stimuli 80–100 dB in −/− versus WT |
||
TCF4 | |||||
Brzózka et al. (2010) | TCF4 Tg, M | TCF4, a helix-loop-helix transcription factor, identified in several GWAS studies as susceptibility gene for SZ |
↓PPI & ØPA in Tg versus WT |
||
AKT1 | |||||
Chen and Lai (2011) | AKT1 KO, M & F | AKT1 has been associated with schizophrenia in genetic studies as well as postmortem brain analyses. AKT1 signal transduction pathway important in diverse biological functions |
F: ↓PPI & ØPA in −/− versus WT; PA M: ØPPI & ØPA in −/− versus WT |
RAC: ØPPI & ØPA in −/− or WT (F only) CLOZ: ØPPI & ØPA in −/− or WT (F only) 8-OH-DPAT: ØPPI in −/− versus WT (attenuation weak; no genotype × drug interaction) SB216763: ØPPI in −/− versus WT (attenuation weak; no genotype × drug interaction) |
Abbreviations: 5-HT serotonin, AAV adeno-associated virus, Aβ amyloid β- peptide, ACE angiotensin converting enzyme, ACH acetylcholine (receptor), AD Alzheimer’s disease, ADX adrenalectomy, AIL advanced intercross line, AMP amphetamine, AMY amygdala, APD antipsychotic drug, APP amyloid precursor protein, ARIP aripiprazole, APO apomorphine, AT angiotensin, BAC bacterial artificial chromosome, BACE β-site APP cleaving enzyme, BD bipolar disorder, BG background, CaMKIV calcium-calmodulin-dependent protein kinease IV, cAMP cyclic adenosine monophosphate, Ckr chakragati, CLO clozapine, CNS central nervous system, COC cocaine, COMT catechol-O-methyltransferase, CORT corticosterone, CRF corticotropin releasing factor, CTR controls, CTX cortex, DA dopamine (receptor), DAT dopamine transporter, dB decibel, DCC deleted in colorectal cancer, DIAZ diazepam, DIZ dizocilpine, DN dominant-negative, DDO D-aspartate oxidase, E embryonic day, EE environmental enrichment, EGF epidermal growth factor, ENU N-ethyl-N-nitrosourea, f frontal, F female, FABP fatty acid binding protein, FGF fibroblast growth factor, Fmr1 fragile × mental retardation 1 gene, FMRP fragile × mental retardation protein, FXS fragile × Syndrome, GLAST glutamate and aspartate transporter, GLU glutamate, GluR glutamate receptor, GLUT glucose transporter, GR glucocorticoid receptor, GRIP glutamate receptor interacting protein, GSK glycogen synthase kinase, GWAS genome wide association studies, HAL haloperidol, HD Huntington’s disease, HPC hippocampus, IC imprinted cluster, IL interleukin, ISI interstimulus interval, KI knock-in, KO knock-out, M male, m metabotropic, MDB methyl-CpG binding protein, mo month, NIC nicotine, MPEP 2-methyl-6-(phenylethylyn)-pyridine hydrocholoride, METH metamphetamine, Mgat N-acetylglucosaminyltransferase, NAA N-acetyl-aspartate, NAAG N-acetylalpha L-aspartyl-L-glutamate, NAC nucleus accumbens, NCAM neural cell adhesion molecule, NIS nisoxetine, nNOS neuronal nitric oxide synthase, NO nitric oxide, NPS neuropeptide S (receptor), NPY neuropeptide Y, NR NMDA receptor subunit, NRG neuregulin, NRL neuroligin, ns not significant(ly), NSE neuron-specific enolase, NT neurotensin, OE overexpressor, OXO oxotremorine, PA magnitude of response to pulse alone, PACAP pituitary adenylate-cyclase-activating polypeptide, PD Parkinson’s disease, PDE phosphodiesterase, PET-1 plasmocytoma expressed transcript-1, PND postnatal day, PLC phospholipase C, PNS prenatal stress, poly I:C polyinosinic: polycytidylic acid, PP prepulse, PPI prepulse inhibition of startle, PPP prepulse potentiation, PS1 presinilin1, PWS Prader–Willi syndrome, QTL quantitative trait locus, QUET quetiapine, RAC raclopride, RAGE receptor for advanced glycation end-products, RasGAP Ras GTPase-activating protein, RE renin-enhancer, RIS risperidone, SCOP scopolamine, SI social isolation, SNP single nucleotide polymorphism, SOCS suppressor of cytokine signaling; SREB superconserved receptor expressed in brain; STR striatum, SYN synapsin, SynGAP synaptic GTP-ase-activating protein, SZ schizophrenia, TAAR trace amine-associated receptor, TG transgenic, TGF-β transforming growth factor beta, TK typosine kinase, TMS transcranial magnetic stimulation, V1b vasopressin receptor 1b, WT wild-type
↓ decreased, ↑ increased, Ø unchanged, −/− homozygous mice, +/− heterozygous mice
2.3 PPI as a Tool to Evaluate Gene–Environment Interactions
Studies of gene–environment interactions may be particularly informative for neuropsychiatric diseases, most of which likely involve a genetic susceptibility combined with environmental factors (e.g. stress) to observe the full manifestation of the disease (Gottesman 1991) (see also Sen and Karg, Gross and Carola chapters in this book). Three ways in which genetics and environmental manipulations have been utilized in genetic mouse models are: (1) using a mutant [e.g. knockout (KO)] to delineate the physiological mechanism of an environmental manipulation; (2) rescuing a phenotype in a mutant with an environmental manipulation; or (3) potentiating or unmasking a phenotype in a genetic mutant with an environmental manipulation. There are a few examples in which PPI has been a useful endpoint with which to assess gene–environment interactions in mouse models. For example, PPI deficits associated with maternal immune activation (MIA) with PolyI:C during mid-gestation, which typically leads to deficits in PPI in adult offspring (Meyer et al. 2005; Shi et al. 2003), are blocked in interleukin (IL)-6 KO dams (Smith et al. 2007). Thus, PPI in a genetic mutant (IL-6 KO mice) was used to determine the mechanism for the effects of an environmental manipulation (immune activation) on brain development. An example of a PPI phenotype being “rescued” in a KO mouse comes from studies in Phospholipase C –β 1 KO mice, in which PPI deficits and locomotor hyperactivity were attenuated in KO mice by environmental enrichment or clozapine (McOmish et al. 2008) (Table 1). More recently, and perhaps most important to etiological models of neuropsychiatric disease, there have been several studies examining the “two-hit” approach (Eells et al. 2006; Ibi et al. 2010). For example, nuclear receptor null Nurr1 heterozygous mice, which display reduced mesocortical and mesolimbic dopamine (Eells et al. 2002), showed reduced PPI after postnatal isolation rearing, an effect that was not observed with either isolation rearing or genotype alone (Eells et al. 2006). This study provides a good example of the utility of PPI in gene–environment models relevant to schizophrenia, specifically those designed to test the “two-hit” hypothesis for the etiology of schizophrenia. It should be kept in mind, however, that many studies assessing gene–environment effects are evaluating additive effects of two manipulations and must be interpreted with caution.
2.4 Evaluating the Role of a Susceptibility Gene in Pathology
When evaluating the role of a susceptibility gene implicated in any neuropsychiatric disease, it is important to consider what criteria should be placed on a genetic/etiological model. In the present context, it is relevant to consider whether or not deficient PPI is a necessary phenotype with which to evaluate the usefulness of a targeted gene deletion of potential relevance to the given neuropsychiatric condition under study (e.g. schizophrenia). Using schizophrenia as an example, the failure to see a PPI deficit in a mouse model may indicate a “false negative” particularly if other key behaviors relevant to schizophrenia are observed (e.g. deficient social interaction, disruptions in attentional set shifting). The lack of a PPI deficit in this case does not indicate that the genetic model is not of relevance to schizophrenia. There are several examples provided in Table 1 in which a PPI deficit was not present in a mutant mouse but other behavioral differences such as deficits in memory, social interaction, or set shifting were apparent in the mice. The likelihood of being able to reproduce all aspects of a heterogeneous disease in another species with a genetic mutation (most often a single gene deletion) is very rare if not impossible (see Jones et al. (2008); Powell et al. (2009) for discussion). For that matter, it is not the case that all aspects of schizophrenia are observed in each patient carrying the diagnosis. Rather, support for a model should be based on the convergence of data from multiple sources [e.g. many animal models, human genetic studies, etc. (Jones et al. 2008)]. Thus, no one phenotype should be considered as being either necessary or sufficient to support a model for a neuropsychiatric disease, particularly since the distributions of the behavioral measure often overlap between healthy volunteers and patients, as is the case with PPI and schizophrenia (Swerdlow et al. 2008). Thus an animal model should not be rejected based on “normal” PPI. For example, GLAST KO mice lacking the glutamate and aspartate transporter do not show any deficits in PPI but do show deficits in social approach, nest building, and pairwise discrimination learning (Karlsson et al. 2009). Along the same lines, there is the possibility that a PPI deficit in a mutant mouse model could represent a “false positive”, in which a PPI phenotype may be suggestive of an association between that gene or pathway and disease and no such association is found. As we have argued previously, the PPI phenotype should be interpreted as meaning that the given genetic manipulation may be involved in the regulation of PPI expression and caution that PPI phenotypes should not be automatically associated with a specific disease, e.g. schizophrenia (Powell et al. 2009). Interestingly, there are a few examples where KO or transgenic mice actually had increased PPI relative to wild-type controls. One such example is the FMR1 KO mice (Paylor et al. 2008; Thomas et al. 2011a, b; but see de Vrij et al. 2008; Table 2). It remains unclear how such findings should be interpreted, but the possibility that the loss of the gene may lead to gain of function should be considered.
Table 2.
References | Mouse Strain, Sex | Model description/background/ rationale |
Basal PPI | Effects of drugs or manipulations typically used to induce PPI- deficits |
Effects of (presumed) antipsychotics/other treatments |
---|---|---|---|---|---|
A. DOPAMINE | |||||
DA receptors | |||||
Doherty et al. (2008) | DA D1, D2 & D3 KO, M, F |
Abnormalities in DA signaling in SZ patients in part have led to the DA hypothesis for SZ |
ØPPI in D1, D2 & D3 −/− versus WT; ↑PA in D1 −/− versus WT; ØPA in D2 (M only); ↓PA in D2 −/− (F only) & D3 −/− versus WT & in D3 (F) versus D3 (M) −/− |
COC (ØPPI in D1 −/−; ↓PPI in D2 −/−, D3 −/− & WT mice; but: attenuated ↓PPI in D2−/− versus WT and more pronounced ↓PPI in D3 −/− versus WT; ØPA in WT |
RAC; SCH23390 (a D1 antagonist) both ↓COC- induced PPI deficits in WT; ØPA in WT) |
Young et al. (2011b) | DA D4 KO, M | Alterations in D4R may contribute to cognitive and attentional deficits in neuropsychiatric disorders. |
ØPPI & ØPA in −/− or +/− versus WT |
||
DAT | |||||
Powell et al. (2008) | DAT KO, (F) M | A hyper-DA state and DAT abnormalities have been implicated in SZ |
M: ↓PPI & ØPA in −/− versus WT; F: inconsistent ↓PPI in −/− versus WT |
M: CLO (PPI in −/− normalized to WT levels, but ØPPI in −/− & WT versus VEH) ↓PA (main effect); QUET (↑PPI & ØPA in −/− & ØPPI & ↓PA in WT) |
|
Other Dopamine related | |||||
Grant et al. (2007) | DCC KO, M (PPI studies) |
DCC mice are netrin 1 receptor KO mice. Netrin 1 is strongly expressed on and may affect development of DA neurons |
↓PPI (yet ns) and ↑PA (trend only) in +/− versus +/+ |
ØAMP (ØPPI in +/−, but ↓PPI in +/+) |
|
B. GLUTAMATE | |||||
NR1 | |||||
Duncan et al. (2010) | NR1 hypomorph (NR1 neo/neo), |
Developmental effects of NR1 hypomorph on other Glu receptors could contribute to the SZ-like phenotype. Evidence for alterations in kainic acid receptors in SZ. |
↓PPI & ↑PA in hypomorph versus WT |
LY382884, kainic acid antagonist, ↓PA in hypomorphs & ØPA in WT; ↑PPI in hypomorphs & WT |
|
Belforte et al. (2010) | Early postnatal NR1 reduction in GABA interneurons (Ppp1r2- cre +/−; NR1loxP/loxP- line A [KO]), M |
NMDAR hypofunction, specifically in GABA interneurons, may contribute to SZ pathophysiology. NR1 is an obligatory subunit of NMDAR. |
Early postnatal NR1 reduction: ↓PPI & ØPA in KO versus Ctrl lines (Flox-A & Cre), in isolated & group-housed mice Adult NR1 reduction: ØPPI & ØPA |
||
AMPA | |||||
Sagata et al. (2010) | GluR4 KO, M & F | GluR4 subunit of AMPA receptor involved in synaptic plasticity; genetic association between GluR4 and SZ |
↓PPI & ↓PA in −/− versus WT | ||
mGLU | |||||
Gray et al. (2009) | mGlu5 KO, M, F | mGlu5 receptor interacts with NMDA receptor and has been implicated in behavioral endophenotypes of SZ |
↓PPI in −/− versus WT; ØPA in −/− versus WT |
Chronic (8 wks) CLO (↑PPI in −/− & ØPPI in WT); ↑NMDA receptors with CLO treatment |
|
Fendt et al. (2010) | mGlu8 KO, M | mGlu8 is a presynaptic receptor which regulates Glu release; may be important for neuropsychiatric disorders with altered Glu function (e.g. SZ). |
ØPPI and ↓PA in −/− versus WT |
PCP: ↓PPI in −/− and WT; ØPA in −/− or WT |
|
Chen et al. (2010) | mGlu5 KO, M & F | Evidence of NMDA hypofunction in mGlu5 KO mice; mGlu5 KO mice may be useful as novel screen of putative glutamatergic antipsychotics. |
↓PPI & ØPA in −/− versus WT | Sarcosine: ↑PPI in −/−; ØPPI in WT; ØPA in −/− or WT LY379268: ØPPI & ØPA in −/− or WT N-acetylcysteine: ↑PPI in −/− & WT; ↑PPI in −/− not blocked with LY341495 |
|
Vesicular glutamate transporter | |||||
Wallén-Mackenzie et al. (2009) | VGLUT2f/f;CKII/Cre (preadolescent forebrain- specific KO), M & F |
Forebrain glutamate plays an important role in many sensory and cognitive functions relevant to psychiatric disease; downstream changes in DA (this study) |
↓PPI & ↓PA in VGLUT2f/ f;CKII/Cre versus WT |
PCP: ↓PPI in WT; ØPPI in VGLUT2f/f;CKII/Cre (no drug versus post-PCP); PA (no data shown) |
ARIP (no-drug versus post-ARIP): ↑PPI in KO; ØPPI in WT; PA (no data shown) |
Glycine | |||||
Benneyworth et al. (2011) | SR KO, M GCP2 HET, M |
D-serine is an agonist and NAG an antagonist at the glycine modulatory site on NMDA receptors. Serine racemate (SR) and glutamic acid decarboxylase 2 (GCP2) regulate D-serine and NAG, respectively. |
ØPPI & ØPA in SR −/− versus WT; ØPPI & ØPA in GCP2 +/− versus WT |
PCP: ↓PPI in SR −/−, GCP2 +/−, & WT; inverted U-shaped dose response function on PA: high dose (6 mg/kg) ØPA in WT, ↑PA in SR −/−; 3 mg/kg dose ↑PA in WT, ØPA in GCP2 +/− AMPH: ↓PPI in Sr−/−, GCP2 +/−, & WT; U-shaped dose response function on PA; no difference in SR −/−, GCP2 +/−, & WT |
|
Other glutamate related | |||||
Karlsson et al. (2009) | GLAST (EAAT1) KO, M, F |
Glutamate dysfunction associated with SZ; glutamate transporter may be useful pharmacological target for SZ. |
ØPPI in −/− or +/− versus WT; ↓PA in −/− versus +/− & WT |
||
Guo et al. (2009) | SynGAP +/− gene deletion |
SynGAP is a RasGAP and NMDAR-associated protein; interacts with NMDA receptors |
↓PPI and ↑PA in +/− versus WT |
CLO: ØPPI in +/− or WT; ↓PA in +/− and WT | |
Han et al. (2009) | GCPII +/−, M & F | Glutamate carboxypeptidase II (GCPII) hydrolyzes NAAG into glutamate and NAA |
ØPPI & ØPA in +/− versus WT |
||
Wang et al. (2009) | Norbin KO | Norbin interacts with mGluR5 receptors and positively regulates mGluR5 signaling. |
↓PPI & ØPA in −/− versus WT | ||
C. SEROTONIN (5-HT) | |||||
Semenova et al. (2008) | 5-HT7 KO, M | Antipsychotics have high affinity for 5-HT7 receptors. |
ØPPI across PP intensities and ISI, ØPA |
PCP ↓PPI in WT, attenuated in −/−; Apo and Amp ↓PPI in WT & −/−; ØPA with Apo, Amp, PCP |
|
Shanahan et al. (2009) | 5-HTT KO, F | OCD patients have PPI deficits; gain of function 5-HTT alleles associated with OCD |
ØPPI in −/−, +/− versus WT | RU24969 (5HT1B agonist) ↓PPI in WT, ØPPI in −/−, intermediate effect in +/− |
|
Schaefer et al. (2009) | Pet-1 KO, M &F | Pet-1 is a transcription factor restricted to 5-HT neurons. Gene deletion results in loss of 5-HT. |
↑PA & ØPPI in −/− versus WT | ||
Hill et al. (2011) | 5HT2C KO | 5HT and BDNF interact; ↑BDNF levels 5HT2C KO mice (this study) |
ØPPI & ØPA in −/− versus WT |
||
Groenink et al. (2011) | 5HT1A KO, M (129S6, Swiss Webster [SW] backgrounds) |
5HT1A gene may interact with adverse life events. |
ØPPI & ØPA in −/− versus WT |
129S6: Maternal separation ↓PPI & ↑PA in −/− & WT SW: Maternal separation ØPPI & ØPA in −/− & WT |
|
van den Buuse et al. (2011a) | 5HT1A KO, M & F | 5HT1A receptor may mediate effects of MDMA on PPI |
ØPPI & ØPA in −/− versus WT |
MDMA: ↓PPI in −/− at lower doses than WT at 100 ms ISI (M only); ↓PA in −/− & WT |
|
van den Buuse et al. (2011b) | 5HT1A KO, M & F | 5HT1A receptor may interact with neurotransmitter systems involved in SZ |
ØPPI & ØPA in −/− versus WT |
APO: ↓PPI & ØPA in −/− & WT | |
D. ACETHYLCHOLINE (ACh) | |||||
Nicotinic | |||||
Darvas et al. (2009) | LYPD6 Tg | LYPD6 modulates nicotinic ACh receptors; LYPD6 Tg show enhanced nicotinic tone |
↑PPI in Tg versus WT; ØPA in Tg versus WT |
||
Young et al. (2011a) | α7-nAChR KO, M & F |
α7-nAChR is being investigated as a pro-cognitive target for SZ; comparison of KO mice across several cognitive tests |
ØPPI in −/−, +/−, and WT; ↑PA in −/− versus +/− & WT; |
||
Muscarinic | |||||
Turner et al. (2010) | M2, M4 KO, M | M2 and M4 muscarinic acetylcholine receptors differ in tissue distribution and physiology; may play a role in sleep and arousal |
ØPPI in M2 or M4 −/− versus C57BL/6 N mice; ØPA in M2 −/− versus M4 −/−; ↑PA in M2 −/− versus C57; ØPA in M4 −/− versus C57 (store bought mice; not littermates) |
||
Thomsen et al. (2010) | M1, M4 KO, F M1KO/ M4KO,M,F |
Muscarinic receptors may be related to pathophysiology of SZ, and muscarinic agonists have been proposed as targets for antipsychotics and cognition enhancers |
F: ↓PPI & ØPA in M1−/−/ M4−/− versus WT; ØPPI & ØPA in M1 −/− versus WT; ØPPI & ↑PA in M4 −/−WT; M: ØPPI & ØPA in M1−/−/M4−/− versus WT |
SCOP: ↓PPI in WT & M4−/− mice (F) & slight ↓PPI in M1−/−/ M4−/− (M); ØPPI in M1−/− (F) or M1−/−/M4−/− (F) ↑PA in all groups; M1−/−/ M4−/− (F) and M4−/− (F) more sensitive to ↑PA |
Xanomeline: blocked SCOP-induce ↓PPI in WT & M1−/−, but not in M4−/− mice; data in M1−/−/ M4−/− difficult to interpret because ØPPI with SCOP; blocked SCOP-induced ↑PA in M1−/− & M1−/−/M4−/−; ØPA in SCOP-treated M4−/− OXO (versus SCOP): blocked SCOP-induced ↓PPI in all genotypes (although SCOP effect not significant in M1−/−/M4−/−); trend for reversal of SCOP-induced ↑PA OXO (alone): ↓PPI in WT & ↑PPI in M1−/−/ M4−/−; ↓PA in WT & M1−/−M4−/− (which may lead to variability in PPI) CLO: ↑PPI in M1−/−/M4−/− at doses with ØPPI in WT; ↓PPI in WT & M1−/−; ØPPI in M4−/−; ↓PA in WT & M1−/−; ØPA in M1−/−/ M4−/− & M4−/− HAL: ↑PPI in WT & M1−/−/M4−/−; ØPA in WT or M1−/−/M4−/− |
Vesicular ACh Transporter (VAChT) |
|||||
Schmid et al. (2011) | VAChT KD, M | Cholinergic transmission associated with cognitive function and startle behavior. VAChT KD mice have reduced cholinergic tone. |
ØPPI & ØPA in −/− versus WT; ↓ between day HAB in −/− versus WT |
||
E. (NEURO)PEPTIDES | |||||
Angiotensin/Renin | |||||
Martin et al. (2008) | RE KO, NSE-AT1 TG, M |
AT/Renin may be involved in brain (DA) signaling. ACE levels were reported to be altered in SZ patients. REKO have reduced renin signaling; NSE-AT1 TG have over- expression of AT1 receptors. |
ØPPI (RE −/− & NSE-AT1 TG vs. WT); ↓PA (RE KO (trend)) & ØPA in NSE-AT1 versus WT |
APO, AMP & DIZ (all ↓PA (RE KO vs. WT; main effect across treatment conditions); ↓PPI in RE −/−, NSE-AT1 TG & WT (no genotype × drug interactions); APO (↓PA in RE −/−, NSE-AT1 TG & WT); AMP (↑PA in RE −/− & WT (trend), ØPA in NSE-AT1 TG & WT;) DIZ (↑PA in RE −/− & WT; more pronounced in RE −/−, ↑PA (trend) in NSE-AT1 TG & WT; |
|
Neurotensin | |||||
Feifel et al. (2010a) | NT1 KO, M & F | Neurotensin (NT) may play a role in SZ pathophysiology. NT levels decreased in CSF from SZ patients, and APDs increase NT levels. |
ØPPI in −/− versus WT; ØPA in −/− versus WT |
AMP ↓PPI & ØPA in −/− & WT; MK801 ↓PPI; MK801 ↑PA (low dose only; more pronounced in WT than −/−) |
PD149163, NT1 receptor agonist, ↑PPI in WT, ØPPI in −/−; |
Feifel et al. (2010b) | NT2 KO, M & F | NT2 receptors highly expressed in brain but not well studied |
↑PPI & ↓PA in −/− versus WT (both sexes) |
||
Oliveros et al. (2010) | NT1 KO & NT2 KO, M |
NT receptors may play a role in psychotomimetic and antipsychotic effects of drugs |
ØPPI & ØPA in NT1 −/−, NT2 −/−, & WT |
AMP: ↓PPI in NT2 −/− & WT, ØPPI in NT1 −/−; ↓PA in WT, ØPA in NT1 −/− or NT2 −/− DIZ: ↓PPI in NT2 −/− & WT, ØPPI in NT1 −/− |
CLO: blocks AMP-induced ↓PPI in WT & NT2 −/− ; blocks DIZ-induced ↓PPI in WT, does not block DIZ-induced ↓PPI in NT2 −/− NT69L: blocks AMP-induced ↓PPI in WT & NT2 −/−; blocks DIZ-induced ↓PPI in NT2 −/−, does not block DIZ-induced ↓PPI in WT; ↓PA in WT & NT2−/− |
CRF | |||||
Groenink et al. (2008) | CRF-OE, M | Stress physiology, particularly CRF and glucocorticoids, implicated in neuropsychiatric disorders |
↓PPI in CRF-OE versus WT at lower PP intensities; ØPA in CRF-OE versus WT |
CORT implant: ØPPI in CRF-OE or WT mice. |
CRF1 antagonists CP154,526 & DMP695 normalized PPI deficit in CRF-OE mice; GR antagonists and ADX did not; CP154,526 ↓PA |
Oxytocin | |||||
Caldwell et al. (2009) | Oxt KO, M, F | Oxytocin has shown antipsychotic potential in animal models of SZ and in some preliminary clinical trials |
ØPPI & ØPA in −/− versus WT |
PCP: Exacerbated ↓PPI in −/− versus WT, ØPA; Apo & Amp: ↓PPI in −/− & WT; ØPA |
|
Arginine Vasopressin | |||||
Egashira et al. (2009) | V1aR KO, V1bR KO |
Arginine vasopressin (AVP), a diuretic peptide with receptors widely expressed in the CNS, has been implicated in neuropsychiatric disorders. |
↓PPI in V1aR & V1bR −/− versus WT; ↑PA in V1aR & V1bR −/− versus WT |
||
Neuropeptide Y | |||||
Karl et al. (2010a) | Y2 KO, M & F | NPY levels altered in SZ patients. NPY Y2 receptor expressed in HPC and AMY |
M: ↑PPI & ØPA in −/− versus WT F: ØPPI in −/− versus WT |
MK801: ↓PPI & ↑PA in −/− & WT AMP: ↓PPI & ↑PA in −/− & WT |
|
Karl et al. (2010b) | Y1 KO, M | Decreased Y1 receptor expression in lymphocytes of SZ patients |
ØPPI, ↓PA, & ↓HAB in −/− versus WT |
MK801: ↓PPI & ↑PA in −/− & WT AMP: ↓PPI & ØPA in −/− & WT (effects are ISI & PP dependent) |
|
Neuropeptide S | |||||
Zhu et al. (2010) | NPSR1 KO, M & F | NPSR1 is a G protein coupled receptor located in brain regions involved in anxiety and learning and memory, whose endogenous ligand is NPS. NPSR1 SNPs identified in panic disorder. |
M: ↓PA & ØPPI in −/− versus WT; F: ØPA & ØPPI in −/− versus WT |
||
Fendt et al. (2011) | NPSR KO, M | NPS implicated in regulation of emotional behavior |
ØPPI in −/− or +/− versus WT; ↓PA in −/− & +/− versus WT |
HAL: ↑PPI & ↓PA in −/−, +/−, & WT | |
PACAP | |||||
Hashimoto et al. (2007) | PACAP KO, M | PACAP affects neurotransmission; potential SZ susceptibility gene |
↓PPI in −/− versus WT | RIS (↑PPI in −/−; Ø in WT) | |
Ishihama et al. (2010) | PACAP KO, M | Genetic susceptibility plus environmental factors may contribute to SZ |
↓PPI in −/− versus WT (PA data not shown) |
SI: ↓PPI & ØPA in −/− & WT | EE: ↓PPI in −/− & WT |
Relaxin-3 | |||||
Smith et al. (2009) | Relaxin-3 KO, M & F |
Relaxin-3 modulates feeding, stress response, exploratory behavior, and arousal |
ØPPI in −/− versus WT; PA data not reported |
||
F. ORPHAN RECEPTORS | |||||
Matsumoto et al. (2008) | SREB2 KO, SREB2 TG, M |
SREB (= GPR85) highly conserved GPCR; expressed in brain regions of high plasticity; overtransmission of 2 SNPs in SZ (this study) |
↓PPI & ØPA in TG versus WT; ØPPI & ØPA in −/− versus WT |
||
Logue et al. (2009) | GPR88 KO, M & F | GPR88 is highly expressed in brain regions implicated in SZ and regulates DA function (this paper). |
↓PPI in −/− versus WT; ØPA in −/− versus WT |
HAL: ↑PPI in −/−, ØPPI in WT; ↓PA in −/− & WT RISP: ↑PPI in −/− & WT; ↓PA in −/− & WT |
|
Kim et al. (2010) | TAK1 KO, M | TAK1 is a nuclear orphan receptor that regulates transcription; TAK1 deletion delays migration of cerebellar granular neurons |
↓PPI, ↓PA, ↓HAP in −/− versus WT |
||
G. TRACE AMINES | |||||
Karmacharya et al. (2011) | TAAR1 KO, M | Trace amines are very similar to biogenic amines and are implicated in neuropsychiatric disease; trace amine receptors may mediate effects of antipsychotic drugs |
ØPPI in −/− versus WT (PA data not shown) |
CLO: ↑PPI in WT, ØPPI in −/− |
Abbreviations: 5-HT serotonin, AAV adeno-associated virus, Aβ amyloid β- peptide, ACE angiotensin converting enzyme, ACH acetylcholine (receptor), AD Alzheimer’s disease, ADX adrenalectomy, AIL advanced intercross line, AMP amphetamine, AMY amygdala, APD antipsychotic drug, APP amyloid precursor protein, ARIP aripiprazole, APO apomorphine, AT angiotensin, BAC bacterial artificial chromosome, BACE β-site APP cleaving enzyme, BD bipolar disorder, BG background, CaMKIV calcium-calmodulin-dependent protein kinease IV, cAMP cyclic adenosine monophosphate, Ckr chakragati, CLO clozapine, CNS central nervous system, COC cocaine, COMT catechol-O-methyltransferase, CORT corticosterone, CRF corticotropin releasing factor, CTR controls, CTX cortex, DA dopamine (receptor), DAT dopamine transporter, dB decibel, DCC deleted in colorectal cancer, DIAZ diazepam, DIZ dizocilpine, DN dominant-negative, DDO D-aspartate oxidase, E embryonic day, EE environmental enrichment, EGF epidermal growth factor, ENU N-ethyl-N-nitrosourea, f frontal, F female, FABP fatty acid binding protein, FGF fibroblast growth factor, Fmr1 fragile × mental retardation 1 gene, FMRP fragile × mental retardation protein, FXS fragile × Syndrome, GLAST glutamate and aspartate transporter, GLU glutamate, GluR glutamate receptor, GLUT glucose transporter, GR glucocorticoid receptor, GRIP glutamate receptor interacting protein, GSK glycogen synthase kinase, GWAS genome wide association studies, HAL haloperidol, HD Huntington’s disease, HPC hippocampus, IC imprinted cluster, IL interleukin, ISI interstimulus interval, KI knock-in, KO knock-out, M male, m metabotropic, MDB methyl-CpG binding protein, mo month, NIC nicotine, MPEP 2-methyl-6-(phenylethylyn)-pyridine hydrocholoride, METH metamphetamine, Mgat N-acetylglucosaminyltransferase, NAA N-acetyl-aspartate, NAAG N-acetylalpha L-aspartyl-L-glutamate, NAC nucleus accumbens, NCAM neural cell adhesion molecule, NIS nisoxetine, nNOS neuronal nitric oxide synthase, NO nitric oxide, NPS neuropeptide S (receptor), NPY neuropeptide Y, NR NMDA receptor subunit, NRG neuregulin, NRL neuroligin, ns not significant(ly), NSE neuron-specific enolase, NT neurotensin, OE overexpressor, OXO oxotremorine, PA magnitude of response to pulse alone, PACAP pituitary adenylate-cyclase-activating polypeptide, PD Parkinson’s disease, PDE phosphodiesterase, PET-1 plasmocytoma expressed transcript-1, PND postnatal day, PLC phospholipase C, PNS prenatal stress, poly I:C polyinosinic: polycytidylic acid, PP prepulse, PPI prepulse inhibition of startle, PPP prepulse potentiation, PS1 presinilin1, PWS Prader–Willi syndrome, QTL quantitative trait locus, QUET quetiapine, RAC raclopride, RAGE receptor for advanced glycation end-products, RasGAP Ras GTPase-activating protein, RE renin-enhancer, RIS risperidone, SCOP scopolamine, SI social isolation, SNP single nucleotide polymorphism, SOCS suppressor of cytokine signaling; SREB superconserved receptor expressed in brain; STR striatum, SYN synapsin, SynGAP synaptic GTP-ase-activating protein, SZ schizophrenia, TAAR trace amine-associated receptor, TG transgenic, TGF-β transforming growth factor beta, TK typosine kinase, TMS transcranial magnetic stimulation, V1b vasopressin receptor 1b, WT wild-type
↓ decreased, ↑ increased, Ø unchanged, −/− homozygous mice, +/− heterozygous mice
2.5 Methodological Considerations with Specific Relevance to Genetic Models of Sensorimotor Gating
For a detailed description of the methods of acoustic startle and PPI in mice see Geyer and Dulawa (2003). When conducting an initial analysis of startle and PPI in a genetic mutant, several considerations should be made when evaluating PPI. Typically, startle sessions involve variable prepulse intensities (e.g. 3, 6, 12 dB above background), which should produce an incremental increase in PPI with increasing prepulse intensities. Of course any evaluation of a PPI phenotype should be considered in the context of a thorough assessment of physical and sensory abnormalities (e.g. hearing loss), as pointed out in Geyer et al. (2002). In a study examining strain differences in hearing and PPI, Willot et al. (1994) showed a relationship between PPI levels and hearing impairments. Hearing loss is also exacerbated by age, particularly in specific strains of mice such as C57s, which experience high frequency hearing loss with age (Willott et al. 1994). One way to avoid relying solely on auditory processing is to assess multimodal PPI using light as a prepulse and/or airpuffs as a startling stimulus (Brody et al. 2004; Young et al. 2010). This multisensory approach can avoid potential confounds of hearing loss and still enable the evaluation of PPI. It should be noted, however, that some degree of noise is often produced with the delivery of airpuff stimuli, therefore, data on airpuff-tactile startle should be interpreted with this consideration in mind. Other tactile stimuli, such as mild footshock could provide an alternative approach to this issue. Another way to provide a gross measure of hearing in genetic mutants is to incorporate a “startle threshold” block into the session, in which increasing decibels of acoustic stimuli are presented to measure startle magnitude across these intensities (Brody et al. 2004). The threshold at which the mouse begins to startle can then be evaluated. Additionally, differences in baseline startle magnitude can confound effects on PPI. Although dissociations between startle and PPI have been reported in mice (Brody et al. 2004) and rats (Sipes and Geyer 1994), large differences in baseline startle magnitude can complicate interpretations of changes in PPI. If large differences in baseline startle are observed, startle magnitude can be “matched” either by using a range of startle stimuli (e.g. 110, 120 dB) paired with the prepulse stimuli or doing a post hoc analysis of only those animals across genotypes that have comparable levels of startle. We have included results on startle magnitude [pulse alone (PA)] in Tables 1–5 when the data were provided and indicated studies that did not report data on startle magnitude. A good example of testing over a wide range of startle amplitudes can be found in Savonenko et al. (2009) in their studies of EP2 KO mice. The background strain on which genetic mutants are made should be considered as well. For a discussion of the issue surrounding background strain the reader is referred to Crawley (2007) and Geyer et al. (2002).
Table 5.
References | Mouse strain, sex | Model description/ background/rationale |
Basal PPI | Effects of drugs or manipulations typically used to induce PPI-deficits |
Effects of (presumed) antipsychotics/other treatments |
---|---|---|---|---|---|
QTL approaches | |||||
Cook et al. (2007) | ENU-mutagenesis- induced variants |
Genome-wide screening for neurobehavioral phenotypes, including PPI |
2 PPI mutants with ↓PPI | ||
Watanabe et al. (2007) | F2 mice from C57BL/6 × C3H/HE crosses, M, & F |
Screening for PPI and startle phenotypes |
6 loci for PPI, 6 for startle and four for startle latency were identified, incl. FABP7 (linked to NMDAR function (↓PPI & ↓PA latency) |
||
Hitzemann et al. (2008) | C57BL/6J, DBA/2J, BALBc/J and LP/J crosses, M & F |
Selective breeding for high and low PPI |
ØPPI & PA in M versus F and no correlation between PPI & PA initially. Breeding line × generation effects on PPI (low vs. high PPI line) after five generations. No line × generation effects on PA, but ↑PA in low PPI line versus ↓PA in high PPI line (main effect only). QTL on chromosomes 11 & 16 in low PPI versus high PPI line |
METH (Line × dose interaction. ↓PPI in high PPI line, but ØPPI in low PPI line at highest METH dose). DIZ (↓PPI; main effect of dose only, no line × dose interaction) |
HAL(↑PPI; main effect of dose only, no line × dose interaction) |
Torkamanzehi et al. (2008) | A/J, C57Bl6/J and congenic crosses thereof, M |
Screening for PPI and PA phenotypes using light prepulses and tactile startle pulses |
Comparable PPI, but not PA in parental strains. Significant variation for congenic strains for PA for both BG and for PPI in C57Bl6 BG strains. Common markers with acoustic PA, but not acoustic PPI from previous study (Joober et al. 2002) |
||
Samocha et al. (2010) | LG/J × SM/J F2 cross & F34 advanced intercross line (AIL), M, F |
PPI and PA phenotyping followed by QTL in F2 generation and genotyped F34 line at 3000 SNP markers |
QTLs found at chromosome 12 for all PP intensities; QTLs for PP6 & PP12 on chromosome 7; QTLs on chromosomes 7 & 17 for startle response; QTLs on chromosome 4 for startle habituation; AIL mice reduced size of some QTLs to less than 5 cm |
||
Insertional mutagenesis | |||||
Verma et al. (2008) | Ckr mouse, M, F |
Ckr serendipidous mutation resulted in mouse with abnormal circling behavior |
↓PPI (modest) at all PP intensities in Ckr versus (+/− & WT); ØPA in Ckr versus −/+ or WT |
CLO (↑PPI in Ckr; ØPPI in WT; RIS (ØPPI in Ckr & WT), HAL (ØPPI in Ckr & WT); PA data not shown |
Abbreviations: 5-HT serotonin, AAV adeno-associated virus, Aβ amyloid β- peptide, ACE angiotensin converting enzyme, ACH acetylcholine (receptor), AD Alzheimer’s disease, ADX adrenalectomy, AIL advanced intercross line, AMP amphetamine, AMY amygdala, APD antipsychotic drug, APP amyloid precursor protein, ARIP aripiprazole, APO apomorphine, AT angiotensin, BAC bacterial artificial chromosome, BACE β-site APP cleaving enzyme, BD bipolar disorder, BG background, CaMKIV calcium-calmodulin-dependent protein kinease IV, cAMP cyclic adenosine monophosphate, Ckr chakragati, CLO clozapine, CNS central nervous system, COC cocaine, COMT catechol-O-methyltransferase, CORT corticosterone, CRF corticotropin releasing factor, CTR controls, CTX cortex, DA dopamine (receptor), DAT dopamine transporter, dB decibel, DCC deleted in colorectal cancer, DIAZ diazepam, DIZ dizocilpine, DN dominant-negative, DDO D-aspartate oxidase, E embryonic day, EE environmental enrichment, EGF epidermal growth factor, ENU N-ethyl-N-nitrosourea, f frontal, F female, FABP fatty acid binding protein, FGF fibroblast growth factor, Fmr1 fragile × mental retardation 1 gene, FMRP fragile × mental retardation protein, FXS fragile × Syndrome, GLAST glutamate and aspartate transporter, GLU glutamate, GluR glutamate receptor, GLUT glucose transporter, GR glucocorticoid receptor, GRIP glutamate receptor interacting protein, GSK glycogen synthase kinase, GWAS genome wide association studies, HAL haloperidol, HD Huntington’s disease, HPC hippocampus, IC imprinted cluster, IL interleukin, ISI interstimulus interval, KI knock-in, KO knockout, M male, m metabotropic, MDB methyl-CpG binding protein, mo month, NIC nicotine, MPEP 2-methyl-6-(phenylethylyn)-pyridine hydrocholoride, METH metamphetamine, Mgat N-acetylglucosaminyltransferase, NAA N-acetyl-aspartate, NAAG N-acetylalpha L-aspartyl-L-glutamate, NAC nucleus accumbens, NCAM neural cell adhesion molecule, NIS nisoxetine, nNOS neuronal nitric oxide synthase, NO nitric oxide, NPS neuropeptide S (receptor), NPY neuropeptide Y, NR NMDA receptor subunit, NRG neuregulin, NRL neuroligin, ns not significant(ly), NSE neuron-specific enolase, NT neurotensin, OE overexpressor, OXO oxotremorine, PA magnitude of response to pulse alone, PACAP pituitary adenylate-cyclase-activating polypeptide, PD Parkinson’s disease, PDE phosphodiesterase, PET-1 plasmocytoma expressed transcript-1, PND postnatal day, PLC phospholipase C, PNS prenatal stress, poly I:C polyinosinic: polycytidylic acid, PP prepulse, PPI prepulse inhibition of startle, PPP prepulse potentiation, PS1 presinilin1, PWS Prader–Willi syndrome, QTL quantitative trait locus, QUET quetiapine, RAC raclopride, RAGE receptor for advanced glycation end-products, RasGAP Ras GTPase-activating protein, RE renin-enhancer, RIS risperidone, SCOP scopolamine, SI social isolation, SNP single nucleotide polymorphism, SOCS suppressor of cytokine signaling; SREB superconserved receptor expressed in brain; STR striatum, SYN synapsin, SynGAP synaptic GTP-ase-activating protein, SZ schizophrenia, TAAR trace amine-associated receptor, TG transgenic, TGF-β transforming growth factor beta, TK typosine kinase, TMS transcranial magnetic stimulation, V1b vasopressin receptor 1b, WT wild-type
↓ decreased, ↑ increased, Ø unchanged, −/− homozygous mice, +/− heterozygous mice
3 Approaches to Genetic Models of Sensorimotor Gating
3.1 Overview of Molecular Genetic Techniques
Broadly defined, behavioral genetic approaches can be divided into two main approaches either beginning with the gene of interest or the behavior of interest (Jacobson and Cryan 2010). Gene-based or “reverse genetics” approaches begin with the targeted gene being manipulated in the animal and the resultant behavior evaluated. Phenotype-based or “forward genetics” approaches begin with the targeted behavioral phenotype (trait) and then involve subsequent genetic analysis of the trait. Most genetic studies of sensorimotor gating have focused on genebased approaches, and as such, these approaches are discussed in more detail than phenotype-based approaches, which are discussed more extensively in Tarantino and Eisener-Doman (2011). The goals for any genetic approach can be very different— some approaches seek to understand the more global role of a specific gene in a particular phenotype related to a disease, while others may be designed to elucidate the role of a particular gene in a cellular process, in neural circuit abnormalities, or in brain development. Studies representing all of these goals are well represented in the literature on genetic models of sensorimotor gating (see Tables 1–5). The techniques used to generate molecular genetic models using the gene-based (reverse genetic) approach include constitutive gene deletion (knockout), insertion of exogenous DNA into the genome (transgenic), and insertion of gene at a particular locus via homologous recombination (knockin) [reviewed in (Crawley 2007; Tecott and Wehner 2001)]. Conditional genetic manipulations, which restrict the expression of a targeted gene either temporally or regionally, are useful in avoiding complications of in utero lethality or compensatory brain changes, or when specific hypotheses exist regarding the developmental expression of a gene or the specific neuroanatomical function of the gene. Examples of conditional genetic manipulations include the Cre-LoxP system in which the targeted gene is floxed with lox-p sites. When combined with Cre recombinase, the floxed gene is expressed, giving regional, cell-specific, or temporal control over expression or deletion of targeted gene (van der Neut 1997; Wang 2009). Restriction to specific brain regions can be achieved through the use of specific promoters used to drive expression. For example, the CaMKIIα promoter restricts expression of a genetic mutation to the forebrain (Mayford et al. 1996). Additional examples of tools to achieve temporal control over gene function involve the Tet-On and Tet-Off systems. With these systems, the target gene is linked to either the tetracycline-controlled transactivator (tTA) or the reverse tTA (rtTA) and can be either turned “on” with doxycycline administered (tTA; Tet-On) or turned “off” in the presence of doxycycline (rtTA; Tet-Off) (Mansuy and Bujard 2000). Thus, by combining some of these approaches, conditional and inducible gene expression in mouse brain can be achieved. Additionally, insertion of large segments of DNA into the genome can be achieved through the use of bacterial artificial chromosomes (BAC transgenics) (Heintz 2001). The BAC transgenic technique is a useful approach when a large segment of DNA has been implicated in a disease (e.g. 22q11.2 microdeletion or microduplications). For example, BAC transgenic technology was used to generate two lines of transgenic mice expressing different genes located on the 22q11.2 segment of the chromosome. Interestingly, mice overexpressing Prodh and Vpreb2 showed increased PPI, while mice overexpressing Zdhhc8, Ranbp1, Htf9c, T10, Arvcf, COMT showed no differences in PPI (Stark et al. 2009). More recent techniques, such as in utero gene transfer, are being developed to modulate the expression of genes during specific stages of embryonic development (Niwa et al. 2010).
“Phenotype-based” approaches to behavioral genetics include “forward genetic screens” such as mutagenesis via radiation or chemical means [e.g. ENU mutagenesis; (Tarantino and Bucan 2000) and quantitative trait loci (QTL) studies on crosses of inbred mouse strains with distinct phenotypes or on mice or rats selectively bred for PPI levels (Hitzemann et al. 2008; Tarantino and Eisener-Doman 2011). Thus far, most of the PPI mutants identified through ENU mutagenesis screens have had some amount of hearing loss, and thus the specificity of the PPI “phenotype” was most likely confounded by deafness (Lisa Tarantino, personal communication). Other molecular genetic tools such as siRNA, in which double stranded RNA homologous to the targeted gene is made and then inserted into the cell to block expression of that gene, and viral transfection, in which genes are inserted into viral vectors and injected into the nucleus of cells, are additional techniques used in neuroscience research assessing sensorimotor gating, but will not be discussed in this chapter. Due to the increasingly large number of genetic models generated in which PPI was evaluated, we will provide some examples in the text of different genetic approaches undertaken to study sensorimotor gating, but the reader is referred to Tables 1–5 for a comprehensive review of the recent genetic models assessing PPI.
3.2 Models Based on Hypotheses Regarding the Pathophysiology of Disease
Genetic models of sensorimotor gating deficits were based initially on the hypothesized pathophysiology of schizophrenia or the known mechanism of action of both antipsychotic and psychotomimetic drugs [e.g. amphetamine, PCP]. Thus, early genetic mutants focused primarily on dopamine (Ralph et al. 2001) and glutamate (Brody and Geyer 2004; Duncan et al. 2004) neurotransmitters and receptors (e.g. dopamine-related genes, glutamate-related genes). Many of these observed effects [e.g. PPI deficits in dopamine transporter KO mice (Ralph et al. 2001)] were not surprising based on the extensive pharmacology of PPI deficits in rodents, but nevertheless the genetic models provided proof of concept that constitutive gene deletion could produce dramatic functional effects on sensorimotor gating in adult animals. PPI deficits in dopamine transporter knockout mice strengthened the converging evidence that dopamine hyperfunction played an important role in sensorimotor gating and potentially the pathophysiology of schizophrenia. Similarly, PPI deficits in metabotropic glutamate mutants (Brody et al. 2003; Brody and Geyer 2004; Gray et al. 2009; Kinney et al. 2003) have stimulated interest in these receptors as targets for drug development (Cleva and Olive 2011). Genetic approaches to pathophysiology of neuropsychiatric disease are becoming increasingly sophisticated and many of these models incorporate assessments of PPI as a relevant behavioral phenotype (Tables 3, 4). Jumping off from the original observation that N-methyl-D-aspartate (NMDA) NR1 hypomorphs show PPI deficits in addition to other behavioral abnormalities (Duncan et al. 2004), several genetic mutants have been made based on reduced NMDA receptor expression, including NMDA subtype-specific mutants (reviewed in Powell et al. (2009)) and downstream signaling proteins (Table 3).
Table 3.
References | Mouse strain, sex | Model description/background/rationale | Basal PPI | Effects of drugs or manipulations typically used to induce PPI-deficits |
Effects of (presumed) antipsychotics/other treatments |
---|---|---|---|---|---|
Second messengers | |||||
Siuciak et al.(2008) | PDE4B KO, M | PDE4 catalyzes the degradation of cAMP. Widely expressed in brain |
↓PPI & ↑PA in −/− versus WT (↓PPI not dependent on ↑PA) |
||
McOmish et al.(2008) | PLC-β1 KO | PLC-β1 is a rate-limiting enzyme involved in the intracellular signaling cascade linked to several metabotropic receptors implicated in SZ; PLC-β1 reduced in SZ brain |
↓PPI & PA (−/− vs. WT) |
EE (main effects): ↑PPI & ↓PA (trend) versus standard housing); EE × genotype × PP intensity effect on PPI (↑PPI in −/−); EE × genotoype effect on PA (↓PA in WT); HAL (no rescue of PPI deficit of −/− vs. VEH treated WT; ØPA); CLO (main effect ↑PPI; genotype × CLO × PP intensity effect: rescue of PPI deficit of −/− vs. VEH treated WT; main effect: ↓PA vs. VEH; genotype × CLO; more pronounced ↓PA in WT) |
|
Koh et al.(2008) | PLC-β1 KO | ↓PPI & trend for ↓PA (−/− vs. WT) |
HAL normalized PPI in −/−, no effect in WT; ØPA |
||
Growth factors | |||||
Ransome et al.(2008) | SOCS-2 TG | SOCS-2 affects growth hormone signaling & potentially hippocampal neurogenesis |
ØPPI in TG versus WT | ||
Scearce-Levie et al.(2008) | Fgf17 KO, M & F | Fgf17 is involved in region-specific development of the rodent fCTX |
ØPPI & ØPA in −/− versus +/− or WT |
||
Ohgake et al.(2009) | Mdk KO, M & F | Midkine involved in neurodevelopment and adult neuroplasticity; abnormal serum levels in SZ and AD; KO produces ↓DA and DAR in striatum (this paper) |
↓PPI in −/− versus WT, +/− intermediate (adult); ØPPI in −/− versus WT (4 wk old); ØPA in −/−, +/−, WT (adult, 4wk) |
HAL ↑PPI in −/−; ØPPI in WT CLO ↑PPI in −/−; ØPPI in WT Overall ↓PA with APD; CLO (3 mg/ kg) ↓PA in WT |
|
Sun et al.(2010) | Forebrain-specific SMAD4 KO, M |
SMAD4 is an intracellular transducer of TGF-β signaling; TGF-β signaling important for neurodevelopment and shown to be abnormal in SZ |
↓PPI & ØPA in −/− versus WT |
||
Nguyen et al.(2011) | PDGFR-β f/f;Nestin Cre+ KO, M (neuron- specific PDGFR KO) |
Platelet derived growth factor (PDGF) is important for embryogenesis and CNS development. PDFGR-β has been associated with SZ and ASD |
↓PPI & ØPA in −/− versus WT |
||
Protein kinase | |||||
Bersudsky et al.(2008) | GSK3β KO, M | GSK3β levels were reported to be reduced in SZ patients. Li inhibits GSK3β in vitro |
ØPPI & ØPA in +/− versus WT |
||
Kaidanovich-Beilin et al.(2009) | GSK3α KO, M & F | GSK3 implicated in several neuronal signaling pathways |
↑PPI & ØPA in −/− versus WT |
||
Takao et al.(2010) | CaMKIV KO, M | CaMKIV is a protein kinase that activates transcription factor CREB and thus may play a role in synaptic plasticity |
ØPPI & ØPA in −/− versus WT |
||
Siuta et al.(2010) | Rictor f/f;Nestin Cre+ KO (neuron- specific Rictor KO) |
Rictor is a component of mTOR2C, the kinase responsible for phosphorylation of Akt. Akt deficiencies associated with SZ |
↓PPI & ØPA in −/− versus WT |
NIS (↑PPI in −/−; no data in WT; PA data not shown) CLO (ØPPI in −/−; PA data not shown) |
|
Proteases | |||||
Horii et al.(2008) | Neuropsin KO, M | The serine protease neuropsin may be involved in neuronal plasticity/ degeneration |
ØPPI & PA in −/− versus WT |
||
Savonenko et al.(2008) | BACE1 KO, M | BACE1 is critical for APP cleavage/ amyloid β production. May also play a role in SZ via proteolysis of NRG & axon myelination |
↓PPI in −/− & ØPPI in +/− versus WT; ØPA in −/− & +/− versus WT; ↑PA latency in −/− versus +/− & WT |
CLO: (amelioration of PPI deficits in −/−; ØPPI in +/− & WT) |
|
Synaptic proteins | |||||
Dyck et al.(2007) | SYN II KO | SYN II, a vesicle-linked phosphoprotein plays a role in neuronal development & transmitter release; hypothesized to contribute to the etiology of SZ |
↓PPI (−/− relative to +/− & WT) |
||
Dyck et al.(2009) | SYN II KO, M & F | ↓SYNII in mPFC of SZ patients; ↑SYNII levels with chronic APD |
↓PPI in −/− versus +/− and WT; ↓PA in −/− versus +/− and WT; lack of HAB in −/− |
||
Savonenko et al.(2009) | EP2 KO, F | EP2 is a prostaglandin receptor involved in activity-dependent synaptic plasticity |
↓PPI in −/− versus WT; ØPA in −/− versus WT |
||
Oliver and Davies (2009) | SNAP-25 Bdr (Blind drunk mutant), SNAP-25 +/−, combined with prenatal stress (PNS), M |
SNARE protein SNAP-25 important for synaptic function and neurotransmitter release; SNAP-25 genomic region linked to SZ |
↓PPI & ØPA in Bdr mutant versus WT, ↓PPI exacerbated by PNS; ØPPI & ØPA in +/− versus WT, no effect of PNS in +/− |
CLO ↑PPI in BDR/NS & BDR/PNS; ØPPI in WT/NS or WT/PNS HAL ↑PPI in Bdr/PNS at 86dB PP only; ØPPI in WT/NS, Bdr/NS, or WT/ PNS |
|
Blundell et al.(2010) | Rim1α KO, M Rab3A KO, M Syt1R233Q (point mutation) KI, M&F Rim1αS413A KI, M |
Presynaptic proteins involved in neurotransmitter release and associated with SZ in genetic studies |
Rim1α: ↓PPI &ØPA in −/− versus WT Rab3A ↓PPI & ØPA in −/−versus WT Syt1R233Q ↓PPI & ØPA in KI versus WT Rim1αS413A ØPPI & ØPA in KI versus WT |
||
Calcium Signaling | |||||
Matsuo et al.(2009) | RyR3 KO | Ryanodine receptor 3 is an intracellular calcium release channel preferentially expressed in HPC and STR and involved in synaptic transmission and plasticity. RyR3 forms a signaling complex with calcineurin, a potential susceptibility gene for SZ and BD |
↓PPI in −/− versus WT at 78 dB PP + 110 dB startle pulse only; ØPA in −/− versus WT |
||
Nakagawasai et al.(2010) | Cav2.2 KO, M | N-type calcium channels are important for neurotransmitter release, brain development, and activity-depdendent plasticity |
↓PPI & ØPA in −/− versus WT |
||
Potassium channel signaling | |||||
Kapfhamer et al.(2010) | Ppp2r5δ KD, M & F GSK3β KD, M KCNQ2 KD, M & F KD created via gene trap (GT), reduced expression by 50% |
KCNQ2 gene encodes an M-type potassium channel subunit and may be a substrate for GSK3β; PP2A regulates GSK3β; GSK3β and M-type potassium channels may be linked to SZ |
Ppp2r5δGt: ↓PPI & ØPA in GT/+ versus WT GSK3β: ↓PPI & ØPA in +/− versus WT KCNQ2 ↓PPI & ØPA in +/− versus WT |
||
Immune activation | |||||
Smith et al.(2007) | IL6 KO | Maternal immune activation (here simulated via poly I:C injection) has been implied in the pathogenesis of SZ |
Maternal poly I:C injection. Effects in offspring: ØPPI in −/− but ↓PPI in WT |
||
Asp et al.(2010) | Tap1 KO mice | Prenatal influenza infection associated with increased SZ risk. Transporter associated with antigen processing 1 mice have reduced expression of MHC class I |
No direct comparisons between −/− and WT mice. C57BL6/J WT mice purchased from Jackson Laboratories |
Influenza infection on PND 3 or 4: ↓PPI in adult −/− mice (during ISI block), trend for ↑PA in infected mice in startle threshold block; ØPPI & ØPA in infected versus non-infected WT C57 mice |
|
López-Ramos et al.(2010) | TgIE96, M | Mice expressing pseudorabies virus immediate-early protein IE180 (TgIE96) have cerebellar abnormalities |
↓PPI & ØPA in Tg versus WT (C57BL/6J WT mice purchased from Jackson Laboratories) |
||
Glucose metabolism | |||||
Schmidt et al.(2008) | GLUT3 (Slc2a3) KO, M |
GLUT is widely expressed in brain. Role in neuronal glucose homeostasis and likely role in diverse behaviors |
↓PPI at lower PP intensities & ↑PA in +/− versus WT |
||
Neuromodulatory functions | |||||
Errico et al.(2008) | DDO KO, M | The enzyme DDO degrades D-aspartate, an amino acid with elusive function in the CNS. A neuromodulatory function at NMDA receptors has been implied |
ØPPI & ØPA in −/− versus WT |
AMP (ØPPI in DDO −/−; ↓PPI in WT) DIZ (↓PPI in −/− & WT, more pronounced in WT) |
Chronic D-aspartate (ØPPI & PA) antagonized AMP & DIZ-induced ↓PPI in WT (not tested in −/−) |
Tanda et al.(2009) | nNOS KO, M | nNOS synthesizes NO from L-arginine; involved in many intracellular signaling processes; ↓nNOS expression in STR and HPC of SZ; polymorphisms of nNOS associated with SZ |
ØPPI & ØPA in −/− versus WT |
SKF81297 (DA D1 agonist) ↓PPI & ØPA in −/−; ØPPI & ↓PA in WT |
|
Altered myelination | |||||
Tanaka et al.(2009) | PLP1(tg/−) | Mylein and oligodendrocyte dysfunction may contribute to schizophrenia pathogenesis. Myelin proteolipid protein (plp1) decreased in brains of SZ patients |
↓PPI in Tg versus WT at higher PP intensity (78dB), PA data not shown |
||
Spine morphogenesis | |||||
Cahill et al.(2009) | KALRN KO | Kalrinin, a guanine-nucleotide exchange factor (GEF) for Rac-like GTPases, regulates spine morphogenesis. KALRN −/− have decreased spine density (this paper) |
↓PPI in −/− versus WT; PA data not shown |
||
Oxidative stress | |||||
Benoit et al.(2010) | QR2 KO, M | QR2 enhances production of quinine and reactive oxygen species (ROS) and is upregulated in cognitively impaired aged rats; polymorphism in QR2 promoter region weakly associated with SZ |
ØPPI in −/− versus WT; PA data not shown |
||
Cole et al.(2011) | GCLM KO, M | GCLM is rate-limiting enzyme in glutathione (GSH) synthesis; GSH defends against oxidative stress |
ØPPI & ØPA in −/− versus WT |
||
Metallothionines | |||||
Koumura et al.(2009) | MT3 KO, M | Metallothionines (MTs) bind zinc and other metals and are neuroprotective in some models |
↓PPI & ØPA in −/− versus WT |
||
Neurodevelopmental genes (other) | |||||
Willi et al.(2010) | Nogo-A KO, M | Nogo-A inhibits growth during development and in adulthood and thus may be involved in neurodev and neural plasticity; some evidence of pathology in SZ |
↓PPI & ØPA in −/− versus WT |
||
Takata et al.(2010) | XBP1 KO, M | XBP1 gene encodes transcription factor in the ER unfolded protein response; involved in brain development and potentially pathogenesis of bipolar disorder and depression |
↑PPI & ØPA in +/− versus WT |
||
Glycoproteins | |||||
Sakatani et al.(2009) | RAGE KO, M | RAGE is a receptor for multiple ligands including Aβ, HMGB1, and S100B, which contribute to the pathology of AD, epilepsy, and ischemia |
↑PPI in −/− versus WT ↑PA in −/− versus WT at low sound pressure levels (70– 100 dB); ↓PA in −/− versus WT at 120 dB |
||
Fukuda et al.(2011) | Fut8 KO, M | α1,6 fucosyltransferase (Fut8)2 transfers fucose from a GDP-fucose to position 6 of N-acetylglucosamine to form N- linked oligosaccharides of glycoproteins. Fucosylated glycoproteins widely distributed in brain |
↓PPI & ØPA in −/− versus +/− & WT |
After restraint stress ↓PPI & ØPA in +/− versus WT |
|
Sex hormones | |||||
Chavez et al.(2009) | ArKO, M, F | Gender differences in the age of onset, severity, and response to treatment in SZ. Second increase in SZ incidence in older age for women when estrogen levels decline. Thus, estrogen may be neuroprotective against development of SZ. Aromatase (Ar) converts androgens to estrogens |
ØPPI in −/− versus WT | AMP ↓PPI in −/− & WT (↓PPI slightly attenuated in F −/−), ↓PA in −/− & WT Apo: ↓PPI in −/− & WT (↓PPI slightly attenuated in F −/−), ↓ PA in −/− & WT MK801: ↓PPI & ↑PA in −/− & WT |
Abbreviations 5-HT serotonin, AAV adeno-associated virus, Aβ amyloid β- peptide, ACE angiotensin converting enzyme, ACH acetylcholine (receptor), AD Alzheimer’s disease, ADX adrenalectomy, AIL advanced intercross line, AMP amphetamine, AMY amygdala, APD antipsychotic drug, APP amyloid precursor protein, ARIP aripiprazole, APO apomorphine, AT angiotensin, BAC bacterial artificial chromosome, BACE β-site APP cleaving enzyme, BD bipolar disorder, BG background, CaMKIV calcium-calmodulin-dependent protein kinease IV, cAMP cyclic adenosine monophosphate, Ckr chakragati, CLO clozapine, CNS central nervous system, COC cocaine, COMT catechol-O-methyltransferase, CORT corticosterone, CRF corticotropin releasing factor, CTR controls, CTX cortex, DA dopamine (receptor), DAT dopamine transporter, dB decibel, DCC deleted in colorectal cancer, DIAZ diazepam, DIZ dizocilpine, DN dominant-negative, DDO D-aspartate oxidase, E embryonic day, EE environmental enrichment, EGF epidermal growth factor, ENU N-ethyl-N-nitrosourea, f frontal, F female, FABP fatty acid binding protein, FGF fibroblast growth factor, Fmr1 fragile × mental retardation 1 gene, FMRP fragile × mental retardation protein, FXS fragile × Syndrome, GLAST glutamate and aspartate transporter, GLU glutamate, GluR glutamate receptor, GLUT glucose transporter, GR glucocorticoid receptor, GRIP glutamate receptor interacting protein, GSK glycogen synthase kinase, GWAS genome wide association studies, HAL haloperidol, HD Huntington’s disease, HPC hippocampus, IC imprinted cluster, IL interleukin, ISI interstimulus interval, KI knock-in, KO knock-out, M male, m metabotropic, MDB methyl-CpG binding protein, mo month, NIC nicotine, MPEP 2-methyl-6-(phenylethylyn)-pyridine hydrocholoride, METH metamphetamine, Mgat N-acetylglucosaminyltransferase, NAA N-acetyl-aspartate, NAAG N-acetylalpha L-aspartyl-L-glutamate, NAC nucleus accumbens, NCAM neural cell adhesion molecule, NIS nisoxetine, nNOS neuronal nitric oxide synthase, NO nitric oxide, NPS neuropeptide S (receptor), NPY neuropeptide Y, NR NMDA receptor subunit, NRG neuregulin, NRL neuroligin, ns not significant(ly), NSE neuron-specific enolase, NT neurotensin, OE overexpressor, OXO oxotremorine, PA magnitude of response to pulse alone, PACAP pituitary adenylate-cyclase-activating polypeptide, PD Parkinson’s disease, PDE phosphodiesterase, PET-1 plasmocytoma expressed transcript-1, PND postnatal day, PLC phospholipase C, PNS prenatal stress, poly I:C polyinosinic: polycytidylic acid, PP prepulse, PPI prepulse inhibition of startle, PPP prepulse potentiation, PS1 presinilin1, PWS Prader–Willi syndrome, QTL quantitative trait locus, QUET quetiapine, RAC raclopride, RAGE receptor for advanced glycation end-products, RasGAP Ras GTPase-activating protein, RE renin-enhancer, RIS risperidone, SCOP scopolamine, SI social isolation, SNP single nucleotide polymorphism, SOCS suppressor of cytokine signaling; SREB superconserved receptor expressed in brain; STR striatum, SYN synapsin, SynGAP synaptic GTP-ase-activating protein, SZ schizophrenia, TAAR trace amine-associated receptor, TG transgenic, TGF-β transforming growth factor beta, TK typosine kinase, TMS transcranial magnetic stimulation, V1b vasopressin receptor 1b, WT wild-type
↓ decreased, ↑ increased, Øunchanged, −/− homozygous mice, +/− heterozygous mice
Table 4.
References | Mouse strain, sex | Model description/background/rationale | Basal PPI | Effects of drugs or manipulations typically used to induce PPI-deficits |
Effects of (presumed) antipsychotics/other treatments |
---|---|---|---|---|---|
Alzheimer’s disease | |||||
Dejaegere et al. (2008) | Alpha 1B/C γ- secretase KO |
γ-secretases have been implicated in the cleavage of membrane proteins incl. APP, & NRG (a SZ vulnerability gene) & in NOTCH signaling |
↓PPI (most pronounced at high PA intensities) & ØPA in −/− versus WT |
HAL: (↑PPI & ØPA in −/− & WT) CLO: (↑PPI in −/−; ØPPI in WT; ØPA in −/− & WT) |
|
Gruart et al. (2008) | APP, PS1, APP+PS1 TG, M |
Neuropathological changes in brain regions regulating PPI may occur in AD models & may affect PPI |
↓PPI in 18 mo old WT, APP KO, PS1 KO & APP+PS1 TG versus 3 mo old WT. Most pronounced ↓PPI in 18 mo APP+PS1 TG. PA data not shown |
||
Tsujimura et al. (2008) | KF-1 KO, M | KF-1 gene increased in AD patients; localized in hippocampus, cerebellum; modulates protein levels as a ubiquitin ligase; increased in frontal ctx after chronic antidepressant treatment, electroconvulsive therapy, and TMS |
↑PPI in −/− versus WT; ↓PA in −/− versus WT |
||
Takeuchi et al. (2011) | P301S Tg, M | Tau, a microtubule-associated protein, can cause cell death and is associated with AD and other neurodegenerative diseases. FTDP17 (frontotemporal dementia and PD linked to chromosome 17) is linked to a point mutation in tau (P301S) |
↑PPI & ↓PA at 120 dB only in Tg versus WT |
||
Autism | |||||
Allan et al. (2008) | MDB1 KO, M & F | Via DNA methylation, MDB mediate epigenetic gene regulation, which has been linked to neurodevelopmental disorders, including autism |
↓PPI & ØPA in −/− versus WT | ||
Chadman et al. (2008) | NL3 KI, M & F | Neuroligin-3 (NL3) is a cell adhesion molecule involved in synapse development & implicated in autism; “knock in” of point mutation identified in autism |
ØPPI in KI versus WT; ↓PA in KI versus WT |
||
Radyushkin et al. (2009) | NL3 KO, M | Point mutations in neuroligin-3 are associated with autism |
ØPPI & ØPA in −/− versus WT | ||
Moy et al. (2009) | NRCAM KO, M & F | NRCAM identified as a candidate gene for autism; involved in cell-cell interactions during brain development and thus affect axonal guidance and neural circuit development |
↓PPI in −/− versus WT (M only; ØPPI in F); ØPA in −/− versus WT |
||
DeLorey et al. (2011) | GABRB3 KO, M & F | GABRB3 gene associated with autism | ØPPI (F) & ↑PPI (M) in +/− versus WT; ↓PA (M & F) in +/− versus WT |
||
Mejias et al. (2011) | GRIP1/2 DKO (double KO, Grip1 flox/ KO, Grip2 KO/ KO) |
GRIP1 scaffolding protein that interacts with GLU2/3 via PDZ domains 4-6. SNP in PDZ4- 6 domain and 5 missense variants identified in ASD (this paper) |
↓PPI in −/− versus WT mice | ||
Fragile X-syndrome (FXS) | |||||
de Vrij et al. (2008) | FMR1 KO | FXS involves mental retardation & is caused by the absence of FMRP. PPI is reduced in FXS patients |
↓PPI at most PP intensities in −/− versus WT |
MPEP (mGluR5 antagonist; ↑PPI in −/− & WT) | |
Paylor et al. (2008) | FMR1 KO, FMR1 KO with a yeast artificial chromosome containing human FMR1 gene, M |
↑PPI & ↓PA (trend) in −/− versus WT |
Addition of human FMR1 gene normalized, i.e. ↓PPI & ↑PA in FMR1 KO to WT levels, & ↓PPI & ØPA in WT |
||
Baker et al. (2010) | FMR1 KO, M & F | Background strain may affect behavioral phenotype in FMR1 KO mice; bred to albino C57BL/6J-Tyr(c-Brd) background |
↑PPI & ↓PA in −/− versus WT (both sexes) |
||
Levenga et al. (2011) | FMR1 KO, M | FMRP involved in translation of mRNA at the synapse, downstream from mGlu5 signaling |
↓PPI in −/− versus WT; PA data not reported (startle measured via eyeblink) |
AFQ056, mGluR5 antagonist: ↑PPI in −/−; ØPPI in WT |
|
Veeraragavan et al. (2011) | FMR1 KO, M | Increased M1 muscarinic signaling in FMR1 KO mice |
ØPPI & ØPA in −/− versus WT | Dicyclomine: ØPPI & ØPA in −/− or WT | |
Thomas et al. (2011a) | mG1.FMR1 (mGlu1 HZ + FMR1 KO); mG5.FMR1 (mGlu5 HZ + FMR1 KO); FMR1 KO |
FMR1 may increase signaling through Group 1 mGluRs (mGlu1, mGlu5) |
↑PPI & ↓PA in FMR1−/− & mG5.FMR1 versus WT; ↑PPI & ↓PA in FMR1−/− & mG1.FMR1 versus WT |
||
Thomas et al. (2011b) | FMR1 KO, M & F | Group 1 mGluRs may be pharmacological target in Fragile X |
↓PA & ØPPI (trend for predicted ↑) in −/− versus WT |
JNJ (mGlu1 antagonist): ↓PPI in WT, ØPPI in −/−; ØPA in −/− & WT MPEP (mGlu5 antagonist): ØPPI & ØPA in −/− & WT |
|
Huntington’s disease | |||||
Menalled et al. (2009) | R6/2 TG, R6/2B TG, YAC128 TG, YAC128B TG, BACHD TG, HDHQ111 KI, M & F |
HD results from a known genetic CAG repeat mutation; ↓PPI in HD patients; HD mutant mice show varying degrees of neuropathology and behavioral abnormalities; comparison controlling for many factors including sex, background strain, husbandry, and handling |
R6/2: ↓PPI in TG versus WT beginning at 12 wks; ↓PA R6/2B: ↓PPI in TG versus WT BACHD: ↓PPI beginning at 24 wks; ØPA YAC128B: ↓PPI at 54 wks; ØPA YAC128: ØPPI; ØPA HDHQ111 KI: ØPPI; ØPA |
||
Brooks et al. (2010) | Hdh(CAG)150 KI, M & F | Longitudinal characterization of KI mouse carrying 150 CAG repeats on the mouse Htt locus |
↓PPI & ↓PA in KI versus WT | ||
Brooks et al. (2011) | R6/1 TG, M & F | R6/1 TG mouse contains ~ 115 CAG repeats on exon 1 of the HD gene. Characterization of R6/1 TG mice on C57BL/6 background is needed |
↓PPI & ↓PA in TG versus WT | ||
Pietropaolo et al. (2011) | R6/1 TG, M & F | Investigate neuropsychiatric symptoms in premotor stage of pathology |
ØPPI & ØPA in TG versus WT | ||
Hearing disorders | |||||
Allen et al. (2008) | Kv1.1 KO | PPI in rodents is a powerful means to identify hearing deficits. The role of Kv1.1 channels in PPI gap detection paradigms was assessed |
↓PPI (for long gaps only) | ||
22q11 deletion & duplication syndromes | |||||
Suzuki et al. (2009b) | SEPT5 (Septin5) KO, M |
22q11.2 microdeletions result in cognitive & behavioral abnormalities and are associated with SZ and autism; SEPT5 gene located on 200kb region spanning the deletion |
↑PPI & ØPA in −/− versus WT | ||
Stark et al. (2009) |
BAC Tg-1: Overexpress Prodh & Vpreb2 BAC Tg-2: overexpress Zdhhc8, Ranbp1, Htf9c, T10, Arvcf, & COMT |
22q11.2 microduplications are also associated with behavioral problems and learning disabilities; gain of function mutations can also be informative for the deletion syndrome |
Tg-1: ↑PPI & ØPA in Tg-1 versus WT Tg-2: ØPPI & ØPA in Tg-2 versus WT |
||
Suzuki et al. (2009a) |
BAC Tg, M: overexpress 190 kb segment of 22q11.2 including TXNRD2, COMT, ARVCF |
22q11.2 is a hotspot for CNVs. 97% of children with 22q11.2 duplications show cognitive deficits |
ØPPI & ØPA in Tg versus WT | ||
Down syndrome | |||||
Ortiz-Abalia et al. (2008) | TG DyrK1A overexpressors, M |
DyrK1A overexpression has been implicated in Down Syndrome |
↓PPI at higher PP intensities in TG versus WT (trend only); PA not shown |
Intrastriatal infusion of AAV iRNA against Dyrk1A: PP intensity-dependent ↑PPI in TG treated with functional versus scrambled viral vectors & pre versus post-treatment with functional vector; PA not shown |
|
Glycosilation IIa congenital disorders | |||||
Soleimani et al. (2008) | Mgat5 KO, M & F | Mgat5 is involed in glycosylation of cell surface glycoproteins, including receptors & transporters. Abundant in the CNS |
ØPPI & PA in −/− versus WT; ↑PA in M versus F (main effect) |
||
Prader-Willi syndrome | |||||
Relkovic et al. (2010) | PWS-IC (+/−), M & F | Prader-Willi Syndrome (PWS) neurodevelopmental disorder resulting from deletion of paternally expressed imprinted genes of chromosome 15q11–q13, a region also implicated in autism and SZ |
↓PPI & ↑PA in +/− versus WT in M & F (only at P105 in M) |
Abbreviations: 5-HT serotonin, AAV adeno-associated virus, Aβ amyloid β- peptide, ACE angiotensin converting enzyme, ACH acetylcholine (receptor), AD Alzheimer’s disease, ADX adrenalectomy, AIL advanced intercross line, AMP amphetamine, AMY amygdala, APD antipsychotic drug, APP amyloid precursor protein, ARIP aripiprazole, APO apomorphine, AT angiotensin, BAC bacterial artificial chromosome, BACE β-site APP cleaving enzyme, BD bipolar disorder, BG background, CaMKIV calcium-calmodulin-dependent protein kinease IV, cAMP cyclic adenosine monophosphate, Ckr chakragati, CLO clozapine, CNS central nervous system, COC cocaine, COMT catechol-O-methyltransferase, CORT corticosterone, CRF corticotropin releasing factor, CTR controls, CTX cortex, DA dopamine (receptor), DAT dopamine transporter, dB decibel, DCC deleted in colorectal cancer, DIAZ diazepam, DIZ dizocilpine, DN dominant-negative, DDO D-aspartate oxidase, E embryonic day, EE environmental enrichment, EGF epidermal growth factor, ENU N-ethyl-N-nitrosourea, f frontal, F female, FABP fatty acid binding protein, FGF fibroblast growth factor, Fmr1 fragile × mental retardation 1 gene, FMRP fragile × mental retardation protein, FXS fragile × Syndrome, GLAST glutamate and aspartate transporter, GLU glutamate, GluR glutamate receptor, GLUT glucose transporter, GR glucocorticoid receptor, GRIP glutamate receptor interacting protein, GSK glycogen synthase kinase, GWAS genome wide association studies, HAL haloperidol, HD Huntington’s disease, HPC hippocampus, IC imprinted cluster, IL interleukin, ISI interstimulus interval, KI knock-in, KO knock-out, M male, m metabotropic, MDB methyl-CpG binding protein, mo month, NIC nicotine, MPEP 2-methyl-6-(phenylethylyn)-pyridine hydrocholoride, METH metamphetamine, Mgat N-acetylglucosaminyltransferase, NAA N-acetyl-aspartate, NAAG N-acetylalpha L-aspartyl-L-glutamate, NAC nucleus accumbens, NCAM neural cell adhesion molecule, NIS nisoxetine, nNOS neuronal nitric oxide synthase, NO nitric oxide, NPS neuropeptide S (receptor), NPY neuropeptide Y, NR NMDA receptor subunit, NRG neuregulin, NRL neuroligin, ns not significant(ly), NSE neuron-specific enolase, NT neurotensin, OE overexpressor, OXO oxotremorine, PA magnitude of response to pulse alone, PACAP pituitary adenylate-cyclase-activating polypeptide, PD Parkinson’s disease, PDE phosphodiesterase, PET-1 plasmocytoma expressed transcript-1, PND postnatal day, PLC phospholipase C, PNS prenatal stress, poly I:C polyinosinic: polycytidylic acid, PP prepulse, PPI prepulse inhibition of startle, PPP prepulse potentiation, PS1 presinilin1, PWS Prader–Willi syndrome, QTL quantitative trait locus, QUET quetiapine, RAC raclopride, RAGE receptor for advanced glycation end-products, RasGAP Ras GTPase-activating protein, RE renin-enhancer, RIS risperidone, SCOP scopolamine, SI social isolation, SNP single nucleotide polymorphism, SOCS suppressor of cytokine signaling; SREB superconserved receptor expressed in brain; STR striatum, SYN synapsin, SynGAP synaptic GTP-ase-activating protein, SZ schizophrenia, TAAR trace amine-associated receptor, TG transgenic, TGF-β transforming growth factor beta, TK typosine kinase, TMS transcranial magnetic stimulation, V1b vasopressin receptor 1b, WT wild-type
↓ decreased, ↑ increased, Ø unchanged, −/− homozygous mice, +/− heterozygous mice
The converging GABA-glutamate theory of schizophrenia stems from two lines of evidence (Coyle 2004). First, the glutamate hypothesis of schizophrenia is derived from evidence that acute administration of phencyclidine (PCP), a noncompetitive NMDA antagonist, produces schizophrenia-like symptoms in healthy humans (Javitt 2004; Javitt and Zukin 1991). Extending such observations, several experimental studies have utilized another NMDA antagonist, ketamine, to induce a model psychosis in normal volunteers (Abel et al. 2003; Krystal et al. 1994; Oranje et al. 2002; van Berckel et al. 1998) and to exacerbate symptoms in patients with schizophrenia (Malhotra et al. 1997a, b; Krystal et al. 1994, 2003). Additionally, studies on postmortem brain tissue from schizophrenia patients show altered GABAergic interneuron function, particularly in parvalbumin (PV) interneurons (Benes and Berretta 2001; Lewis et al. 2005). Evidence that NMDA hypofunction specifically on GABA interneurons plays a role in the pathogenesis of schizophrenia comes from several lines of preclinical research. First, NMDA antagonists increase the firing rate of pyramidal cells and increase prefrontal glutamate release (Jackson et al. 2004; Moghaddam et al. 1997), suggesting that NMDA antagonists are preferentially blocking inhibitory interneurons. Second, preclinical studies suggest that GABA interneurons are more sensitive to NMDA antagonists than other neuronal subtypes such as pyramidal neurons (Grunze et al. 1996). Third, repeated exposure to NMDA antagonists such as ketamine decreases PV and GAD67 expression (Behrens et al. 2007). Based on these converging lines of evidence, Belforte et al. (2010) used the Cre-LoxP system to engineer mice with deletion of NR1 specifically in GABAergic interneurons (Table 1). Their elegant set of studies showed that reduction in NR1 early in postnatal development produced deficits in PPI, but that adult NR1 reduction had no effect on PPI. These studies corroborate previous studies reporting PPI deficits with constitutive NR1 reduction throughout the brain (Duncan et al. 2004). There have been several other genetic mutants created to test hypotheses regarding glutamate signaling (see Table 4). For example, mice with pre-adolescent forebrain-specific deletions of the vesicular glutamate transporter VGLUT2f/f;CKII/Cre show PPI deficits, which are attenuated with the antipsychotic drug aripiprazole (Wallén-Mackenzie et al. 2009). Genetic mutants of specific proteins involved in NMDA receptor signaling such as mice heterozygous for SynGAP gene deletion also show PPI deficits in addition to other behavioral abnormalities (Guo et al. 2009). For a review of other neurotransmitter-related genetic mutants [e.g. acetylcholine, serotonin, dopamine (see Table 4)].
In addition to classic neurotransmitters, the important role of neuropeptides in neuropsychiatric disease is becoming more widely appreciated. PPI has been assessed in genetic mutants for many neuropeptides including angiotensin, neurotensin, corticotropin releasing factor (CRF) overexpression, oxytocin, arginine vasopressin, neuropeptide Y, PACAP, and Relaxin-3 (Table 4). Stress physiology, particularly CRF and glucocorticoids, has been implicated in neuropsychiatric disease. Mice overexpressing a CRF transgene exhibit deficits in PPI (at low prepulse intensities), which are normalized by CRF1 antagonists, whereas glucocorticoid receptor antagonists did not reverse the PPI deficit (Groenink et al. 2008). Moreover, several genetic mutants have been created to determine the role of specific proteins in brain processes, including second messenger signaling, synaptic proteins, synaptic vesicles, protein kinases, etc. (Table 3). While the direct link for many of these proteins has not necessarily been established for specific neuropsychiatric diseases, a thorough behavioral evaluation of these mice including an assessment of PPI can help elucidate the functional implications of the target protein. For example, gene deletion of PDE4B, which catalyzes the degradation of cAMP and is widely expressed in brain, is associated with PPI deficits (Siuciak et al. 2008). Decreased presynaptic proteins such as synaptophysin, SNAP-25, and complexin II have been observed in postmortem brains of schizophrenia patients (Harrison and Weinberger 2005). These data, combined with evidence of problems with neuronal migration and abnormal neuronal processes, have led investigators to conceptualize schizophrenia as a disease of functional “dysconnectivity” (Friston and Frith 1995; McGlashan and Hoffman 2000; Weinberger et al. 1992) or a “disorder of the synapse” (Frankle et al. 2003; Mirnics et al. 2001) reviewed in (Harrison and Weinberger 2005). Thus many genetic mutants have been created for specific synaptic proteins (Table 3). For example, SYNII, a vesicle-linked phosphoprotein that plays a role in neuronal development and neurotransmitter release (Cesca et al. 2010), is genetically associated with schizophrenia, decreased in brain of schizophrenia patients, and increased with chronic antipsychotic drug treatment (Chen et al. 2004; Chong et al. 2002). SYNII KO mice exhibit decreased PPI, providing more evidence for the essential role of SYNII in synaptic function and behavior (Dyck et al. 2007, 2009). Similarly, a mutation of SNAP-25, a SNARE protein that is integral to synaptic function and neurotransmitter release, is associated with PPI deficits (Oliver and Davies 2009).
3.3 Genetic Mouse Models as Pharmacological Tools
Because many of the early molecular genetic approaches focused on neurotransmitter receptor genes, several of these mutants have also been used to delineate the receptor mechanisms of drugs that disrupt PPI in order to expand our understanding of the neurotransmitter systems and neural circuitry underlying deficient PPI (Doherty et al. 2008; Dulawa et al. 1997; Ralph et al. 1999; van den Buuse et al. 2011a) (Table 4). Since most of these genetic mutants were built on rat pharmacology, we must keep in mind several important species differences between the pharmacology of mouse versus rat PPI (e.g. differential effects of dopamine D1 and D2 receptors, 5-HT agonists, etc.; Powell et al. 2009). Pharmacological disruptions of PPI and their reversal with antipsychotics have been better characterized in rats than in mice, although the literature on PPI pharmacology in mice is rapidly increasing (Swerdlow et al. 2008). In order to use mutant mouse models to examine the receptor mechanisms for alterations in PPI, more complete dose response studies are warranted in mice. Nevertheless, drugs that both impair and improve PPI continue to be investigated in genetic mutants. This approach is particularly useful in determining the functional role of a receptor (e.g. does the gene deletion alter the response to a psychotomimetic drug) and in determining the receptor mechanisms for a drug effect when either receptor-selective drugs are not available or when novel drugs are being evaluated. For example, amphetamine disrupts PPI in WT mice, but not in mice with gene deletions of DDO, an enzyme that degrades D-aspartate (Errico et al. 2008), suggesting a functional role of DDO in PPI disruptions. Interestingly, oxytocin KO mice show an increased sensitivity to the PPI-disruptive effects of PCP (Caldwell et al. 2009), suggesting that endogenous oxytocin may protect against PCP-induced disruptions of PPI, a finding that supports the clinical data showing putative antipsychotic properties of oxytocin (Feifel et al. 2010a). A similar approach was used to show protective properties of nNOS, which synthesizes NO from l-arginine, against PPI-induced disruptions with the dopamine D1 agonist SKF81297 (Tanda et al. 2009).
3.4 Genetic Models of Candidate Genes for Neuropsychiatric Diseases
As the field of neuropsychiatric genetics has grown, several gene targets have been identified consistently for particular disorders and have thus been modified in mouse models through gene deletion, the addition of a transgene, etc. When evaluating candidate genes for mouse models, one should consider that many single nucleotide polymorphisms (SNPs) identified in genetic screens for neuropsychiatric disease involve mutations in introns of genes. Thus, it is very important to consider (1) whether or not functional mutations in the gene have been identified before embarking on mutant mouse models, and (2) the impact of species–specific alterations in gene function in the mutant mouse (Low and Hardy 2007). Table 1 summarizes genetic mouse models of vulnerability genes for schizophrenia, and Table 2 summarizes genetic models of other neuropsychiatric disorders in which PPI deficits have been observed. Two approaches in human studies have increased our understanding of the genetics of sensorimotor gating (1) assessing the relationship between polymorphisms in a target gene (e.g. NRG1, COMT) and PPI levels in healthy volunteers and (2) endophenotype-based genetic studies of schizophrenia (e.g. Consortium on the Genetics of Schizophrenia; COGS). For example, recent studies suggest that PPI is associated with polymorphisms in CHRNA3 (Petrovsky et al. 2010), neuregulin 1 (Roussos et al. 2011), and COMT (Giakoumaki et al. 2008; Quednow et al. 2008; Roussos et al. 2008) genes. Some of these same genes (e.g. NRG1, COMT) are also associated with prepulse inhibition in the COGS data set (Greenwood et al. 2011).
Several different mutants for neuregulin 1 isoforms have been created with varying effects on behavior and neuroanatomy (reviewed in Duffy et al. 2008; O’Tuathaigh et al. 2007). Overall, the PPI phenotype in NRG1 mutants has been inconsistent (O’Tuathaigh et al. 2011, 2009). The Type III NRG1 heterozygote mouse has shown abnormalities in multiple neuroanatomical and behavioral endpoints relevant to schizophrenia, including PPI deficits that are improved with chronic nicotine administration (Chen et al. 2008). CNS-specific ERBB2/B4 KO mice exhibit PPI deficits, which are attenuated by the antipsychotic drug clozapine (Barros et al. 2009). Conditional knockouts with better temporal and regional specificity have allowed for evaluation of more specific roles of NRG1 and its receptors. For example, deletion of ErbB4 selectively in PV interneurons produces PPI deficits, suggesting a functional interaction between NRG1 and the integrity of PV interneurons (Wen et al. 2010).
The COMT Val allele is associated with reduced P300 and P50 ERPs (Gallinat et al. 2003; Golimbet et al. 2006; Lu et al. 2007) and reduced PPI (Quednow et al. 2008; Roussos et al. 2008), presumably due to decreased dopamine function in the prefrontal cortex. There have been several mice created to examine the effects of COMT gene on schizophrenia relevant phenotypes, COMT KO mice (Gogos et al. 1998; Papaleo et al. 2008; Yavich et al. 2007), COMT Tg mice that overexpress the human COMT-Val polymorphism (Papaleo et al. 2008), and S-COMT isoform KO (Tammimäki et al. 2010) Thus far, none of the COMT mouse models have shown robust deficits in PPI (Table 1), although other behaviors probing prefrontal cortex such as the attentional set shifting task (ASST) are altered in the COMT-Val TG mice (Papaleo et al. 2008).
The disrupted in Schizophrenia 1 (DISC1) gene has shown association with schizophrenia and behavioral and neuroanatomical biomarkers for schizophrenia (for review see Mackie et al. 2007; Porteous et al. 2006). DISC1 is associated with schizophrenia neuropathology (Cannon et al. 2005; Callicott et al. 2005), abnormal P300 ERP (Blackwood et al. 2001), and neurocognitive function (e.g. learning and memory; Burdick et al. 2005; Cannon et al. 2005; Hennah et al. 2005). The data on the dominant negative DISC1 models have reported both decreased PPI (Hikida et al. 2007) and no change in PPI (Ibi et al. 2010); whereas, the DISC1L100P mutations have shown more consistent decreases in PPI (Lipina et al. 2011; Lipina et al. 2010). The new technology of in utero gene transfer, allows for the knockdown of DISC1 into the mouse brain during early embryonic development. Mice with in utero knockdown of DISC1 show reduced PPI when tested in adulthood (Niwa et al. 2010). As we have discussed previously, modeling the human DISC1 mutation in mice is difficult because most of the models have assumed that the mutation in neuropsychiatric disease is due to the formation of a truncated DISC1 protein (Powell et al. 2009). Another gene, Boymaw, is also disrupted on chromosome 11 in the Scottish family. Two fusion transcripts are generated between DISC1 and Boymaw genes in the translocation carriers in the Scottish schizophrenia family (Zhou et al. 2008). Thus, expressing the two fusion transcripts may be a better strategy for creating mutant DISC1 mice for the study of neuropsychiatric disorders. The increasing identification of copy number variants (CNVs) associated with schizophrenia and large-scale GWAS studies identifying novel candidate genes will lead to even greater potential to create new mouse models (see Tables for examples). Additionally, PPI is being used more widely to characterize mutant mouse models of candidate genes for other neuropsychiatric disorders such as autism and Huntington’s Disease (Table 2).
3.5 Phenotype-Based Models Revealing the Function of Genes
PPI deficits have also been used in phenotype-to-genotype approaches, or “forward genetic” screens such as ENU mutagenesis, QTL on strain crosses, and QTL with selective breeding. For example, using F2 mice from a C57BL/6 × C3h/HE cross, QTL identified 6 loci for PPI, including the gene FABP7, which has been linked to NMDA receptor function (Watanabe et al. 2007). In a study comparing A/J, C57Bl6/J, and congenic crosses thereof (recombinant congenic mouse strains) (Torkamanzehi et al. 2008) found some common markers for startle but not PPI compared to previous studies (Joober et al. 2002). Selective breeding for high and low levels of PPI has identified QTLs on chromosomes 11 and 16 in low PPI versus high PPI mouse lines (Hitzemann et al. 2008; Schwabe et al. 2007). An ENU mutagenesis screen produced 2 PPI mutants (Cook et al. 2007). One major problem with these approaches is that many of the PPI mutants, particularly with ENU mutagenesis, will likely be deaf or have some degree of hearing loss. In the (Torkamanzehi et al. 2008) study on crosses of A/J, C57Bl6/J and congenic crosses, light prepulses and tactile startle pulses were used to measure PPI to avoid this potential confound (but airpuff startle also has an acoustic component to it).
The field of molecular genetics continues to produce new mutant mouse models with unknown effects on the central nervous system. Many of these mutants have behavioral abnormalities that have been observed anecdotally. Phenotypic characterization of mice with mutations of genes heretofore not known to be relevant to CNS disease (some examples found in Tables 1–5) may reveal novel genes for further study of neuropsychiatric genetics. Although these approaches were gene-based, some have the capacity to identify novel genes for a particular disease, which is why we have included these examples here. Two examples of the way a novel gene of relevance to psychiatric conditions can be discovered through the creation of a mutant mouse are the SP4 and the SREB2 genes. SP4 is a member of the Sp1 family of transcription factors. Hypomorphic Sp4 mice showed vacuolization in the hippocampus, age-dependent decrease in neurotrophin-3 expression in the dentate granule cells, and robust deficits in both PPI and contextual memory (Zhou et al. 2004). These studies revealed a novel Sp4 pathway that is important for hippocampal development and essential to many behaviors, including PPI, relevant to schizophrenia (Zhou et al. 2004). Zhou et al. (2009) have gone on to examine the role of the human SP4 gene in schizophrenia and bipolar disorder. Several SNPs from the human SP4 gene are found to associate with both bipolar disorder and schizophrenia in both Caucasian and Chinese samples. This work represents an example in which a PPI phenotype, in combination with other behavioral abnormalities, suggested the association of this gene with neuropsychiatric disorders. A similar finding was also observed with SREB2 Tg mice, which display PPI deficits. Follow-up genetic studies in schizophrenia found a genetic association between SREB2 and schizophrenia (Matsumoto et al. 2008).
4 Discussion
Mutant mouse models of schizophrenia provide a unique way to assess the function of a susceptibility gene, test hypotheses about the pathophysiology of disease, address receptor mechanisms of drugs, and generate hypotheses about the function of relatively unknown genes. In this review, we provided a comprehensive overview of PPI deficits in genetic models since July, 2007 and elaborate on specific examples where appropriate. As illustrated in Tables 1–5, perhaps the most notable advances in genetic approaches to sensorimotor gating over the last few years have come from candidate genes for schizophrenia and other neuropsychiatric diseases exhibiting PPI deficits, genes involved in basic synaptic processes and receptor signaling, and conditional genetic approaches that help in understanding the dynamic function of specific genes both regionally and temporally. In particular, since our last reviews (Powell et al. 2009; Swerdlow et al. 2008), many genetic mutants of proteins involved in synaptic plasticity, second messenger systems, calcium signaling, etc. have been developed. In this case, PPI is used as a functional measure of sensorimotor processing in models of basic brain development. Whether or not these cellular processes turn out to be relevant for a specific disease such as schizophrenia remains to be determined. Thus far, most models are still in the “characterization” phase—testing multiple behavioral/cognitive constructs that are deficient across neuropsychiatric disorders (e.g. sensorimotor gating, attention, social interaction, learning, and memory, etc.). Few models, however, have shown any predictive power for drug development. Perhaps this new wave of mutants based on susceptibility genes, synaptic function, or brain development, as opposed to genetic manipulation based on mechanism of action of antipsychotics or psychotomimetics, will be better models to lead the field forward in medication development for mental illness. As Moore (2010) argues, too many models that are not based on either “etiologic or pathogenic” theories, may result in too many “false positives” in drug screens. Rather, refined models that more closely mimic the etiological risk factors and/or neuropathology of disease (e.g. schizophrenia) may generate more predictive models for drug development (Moore 2010). While this approach might be very useful if the goal is to use the mutant mice for drug development, we have illustrated the benefits of careful behavioral characterization in genetic models of basic brain processes as well. As mentioned above, it is unlikely that all aspects of a heterogeneous disease will be recapitulated in another species with a genetic mutation. Investigators must rely on convergence of behavioral and neuroanatomical or neurochemical data in a given mutant mouse to support clinical data on the link between the candidate gene and disease. No single phenotype such as PPI should be considered as being either necessary or sufficient to substantiate a model as having relevance to neuropsychiatric disease.
Animal studies addressing neuropsychiatric disease would benefit greatly from more neurobiologically based biomarkers for these disorders. Such an approach has been taken in the CNTRICS initiative, in an explicit attempt to incorporate more cognitive neuroscience-based testing into treatment trials of putative cognitive therapies for schizophrenia. Along the same lines, genetic studies of schizophrenia have focused on psychophysiological endophenotypes such as PPI instead of the broader, more heterogeneous diagnosis of schizophrenia. In fact, many laboratories are now using PPI as an endophenotype in genetic studies of schizophrenia (Braff et al. 2007; Greenwood et al. 2007; Greenwood et al. 2011; Hokyo et al. 2010). Some of these genetic studies have generated further support for a genetic contribution to PPI (Greenwood et al. 2007, 2011), and other studies have suggested that genetic variants in COMT (Quednow et al. 2008; Roussos et al. 2008), CHRNA3 (Petrovsky et al. 2010), and neuregulin 1 (Roussos et al. 2011) directly affect PPI levels (as reviewed above). Thus, as human studies materialize with more neurobiologically defined behavioral measures, the ability to translate these measures or “endophenotypes” into animal models should improve dramatically. PPI in genetic models offers a behavioral endpoint that has shown predictive validity in rat pharmacological models, cross-species homology with the same measure in humans, and alterations in response to genetic manipulations implicated in the pathogenesis of schizophrenia. While these mutant mouse models are not without shortcomings, they offer some of the best attempts at etiological models that are possible in rodents. Merging the genetic etiological models with a second hit approach may strengthen some of the mutant mouse models. Hence, consideration for other factors, such as the importance of environmental risk factors (e.g. prenatal infection) and the role of epigenetics (e.g. DNA methylation) in the etiology of schizophrenia, should also be incorporated with genetic models.
Acknowledgments
This work was supported by grants from the National Institute of Mental Health (R01MH042228, R01MH052885, R01MH091407) and by the Veterans Affairs VISN 22 Mental Illness Research, Education, and Clinical Center.
Contributor Information
Susan B. Powell, Email: sbpowell@ucsd.edu, Department of Psychiatry, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093-0804, USA; Research Service, VA San Diego Healthcare System, San Diego, CA, USA.
Martin Weber, Email: weber.martin@gene.com, Department of Neuroscience, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080-4990, USA.
Mark A. Geyer, Email: mgeyer@ucsd.edu, Department of Psychiatry, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093-0804, USA; Research Service, VA San Diego Healthcare System, San Diego, CA, USA.
References
- Abel KM, Allin MP, Hemsley DR, Geyer MA. Low dose ketamine increases prepulse inhibition in healthy men. Neuropharmacol. 2003;44:729–737. doi: 10.1016/s0028-3908(03)00073-x. [DOI] [PubMed] [Google Scholar]
- Allan AM, Liang X, Luo Y, Pak C, Li X, Szulwach KE, Chen D, Jin P, Zhao X. The loss of methyl-CpG binding protein 1 leads to autism-like behavioral deficits. Hum Mol Genet. 2008;17:2047–2057. doi: 10.1093/hmg/ddn102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Allen PD, Schmuck N, Ison JR, Walton JP. Kv1.1 channel subunits are not necessary for high temporal acuity in behavioral and electrophysiological gap detection. Hear Res. 2008;246:52–58. doi: 10.1016/j.heares.2008.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arguello PA, Gogos JA. Cognition in mouse models of schizophrenia susceptibility genes. Schizophr Bull. 2010;36:289–300. doi: 10.1093/schbul/sbp153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Asp L, Holtze M, Powell SB, Karlsson H, Erhardt S. Neonatal infection with neurotropic influenza A virus induces the kynurenine pathway in early life and disrupts sensorimotor gating in adult Tap −/− mice. Int J Neuropsychopharmacol. 2010;13:475–485. doi: 10.1017/S1461145709990253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baker KB, Wray SP, Ritter R, Mason S, Lanthorn TH, Savelieva KV. Male and female Fmr1 knockout mice on C57 albino background exhibit spatial learning and memory impairments. Genes Brain Behav. 2010;9:562–574. doi: 10.1111/j.1601-183X.2010.00585.x. [DOI] [PubMed] [Google Scholar]
- Barr AM, Fish KN, Markou A, Honer WG. Heterozygous reeler mice exhibit alterations in sensorimotor gating but not presynaptic proteins. Eur J Neurosci. 2008;27:2568–2574. doi: 10.1111/j.1460-9568.2008.06233.x. [DOI] [PubMed] [Google Scholar]
- Barros CS, Calabrese B, Chamero P, Roberts AJ, Korzus E, Lloyd K, Stowers L, Mayford M, Halpain S, Muller U. Impaired maturation of dendritic spines without disorganization of cortical cell layers in mice lacking NRG1/ErbB signaling in the central nervous system. Proc Natl Acad Sci U S A. 2009;106:4507–4512. doi: 10.1073/pnas.0900355106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Behrens MM, Ali SS, Dao DN, Lucero J, Shekhtman G, Quick KL, Dugan LL. Ketamine-induced loss of phenotype of fast-spiking interneurons is mediated by NADPH-oxidase. Science. 2007;318:1645–1647. doi: 10.1126/science.1148045. [DOI] [PubMed] [Google Scholar]
- Belforte JE, Zsiros V, Sklar ER, Jiang Z, Yu G, Li Y, Quinlan EM, Nakazawa K. Postnatal NMDA receptor ablation in corticolimbic interneurons confers schizophrenia-like phenotypes. Nat Neurosci. 2010;13:76–83. doi: 10.1038/nn.2447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benes FM, Berretta S. GABAergic interneurons: implications for understanding schizophrenia and bipolar disorder. Neuropsychopharmacology. 2001;25:1–27. doi: 10.1016/S0893-133X(01)00225-1. [DOI] [PubMed] [Google Scholar]
- Benneyworth MA, Basu AC, Coyle JT. Discordant behavioral effects of psychotomimetic drugs in mice with altered NMDA receptor function. Psychopharmacology (Berl) 2011;213:143–153. doi: 10.1007/s00213-010-2023-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benoit CE, Bastianetto S, Brouillette J, Tse Y, Boutin JA, Delagrange P, Wong T, Sarret P, Quirion R. Loss of quinone reductase 2 function selectively facilitates learning behaviors. J Neurosci. 2010;30:12690–12700. doi: 10.1523/JNEUROSCI.2808-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bersudsky Y, Shaldubina A, Kozlovsky N, Woodgett JR, Agam G, Belmaker RH. Glycogen synthase kinase-3beta heterozygote knockout mice as a model of findings in postmortem schizophrenia brain or as a model of behaviors mimicking lithium action: negative results. Behav Pharmacol. 2008;19:217–224. doi: 10.1097/FBP.0b013e3282feb099. [DOI] [PubMed] [Google Scholar]
- Bitsios P, Giakoumaki SG, Theou K, Frangou S. Increased prepulse inhibition of the acoustic startle response is associated with better strategy formation and execution times in healthy males. Neuropsychologia. 2006;44:2494–2499. doi: 10.1016/j.neuropsychologia.2006.04.001. [DOI] [PubMed] [Google Scholar]
- Blackwood DH, Fordyce A, Walker MT, Clair DM, Porteous DJ, Muir WJ. Schizophrenia and affective disorders—cosegregation with a translocation at chromosome 1q42 that directly disrupts brain-expressed genes: clinical and P300 findings in a family. Am J Hum Genet. 2001;69:428–433. doi: 10.1086/321969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blundell J, Kaeser PS, Sudhof TC, Powell CM. RIM1alpha and interacting proteins involved in presynaptic plasticity mediate prepulse inhibition and additional behaviors linked to schizophrenia. J Neurosci. 2010;30:5326–5333. doi: 10.1523/JNEUROSCI.0328-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Braff DL. Prepulse inhibition of the startle reflex: a window on the brain in schizophrenia. Curr Top Behav Neurosci. 2010;4:349–371. doi: 10.1007/7854_2010_61. [DOI] [PubMed] [Google Scholar]
- Braff DL. Gating in schizophrenia: from genes to cognition (to real world function?) Biol Psychiatry. 2011;69:395–396. doi: 10.1016/j.biopsych.2011.01.002. [DOI] [PubMed] [Google Scholar]
- Braff DL, Freedman R, Schork NJ, Gottesman II. Deconstructing schizophrenia: an overview of the use of endophenotypes in order to understand a complex disorder. Schizophr Bull. 2007;33:21–32. doi: 10.1093/schbul/sbl049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Braff DL, Geyer MA, Swerdlow NR. Human studies of prepulse inhibition of startle: normal subjects, patient groups, and pharmacological studies. Psychopharmacology (Berl) 2001;156:234–258. doi: 10.1007/s002130100810. [DOI] [PubMed] [Google Scholar]
- Braff DL, Grillon C, Geyer MA. Gating and habituation of the startle reflex in schizophrenic patients. Arch Gen Psychiatry. 1992;49:206–215. doi: 10.1001/archpsyc.1992.01820030038005. [DOI] [PubMed] [Google Scholar]
- Brody SA, Conquet F, Geyer MA. Disruption of prepulse inhibition in mice lacking mGluR1. Eur J Neurosci. 2003;18:3361–3366. doi: 10.1111/j.1460-9568.2003.03073.x. [DOI] [PubMed] [Google Scholar]
- Brody SA, Dulawa SC, Conquet F, Geyer MA. Assessment of a prepulse inhibition deficit in a mutant mouse lacking mGlu5 receptors. Mol Psychiatry. 2004;9:35–41. doi: 10.1038/sj.mp.4001404. [DOI] [PubMed] [Google Scholar]
- Brody SA, Geyer MA. Interactions of the mGluR5 gene with breeding and maternal factors on startle and prepulse inhibition in mice. Neurotoxic Res. 2004;6:79–90. doi: 10.1007/BF03033300. [DOI] [PubMed] [Google Scholar]
- Brooks S, Higgs G, Jones L, Dunnett SB. Longitudinal analysis of the behavioural phenotype in Hdh ((CAG)150) Huntington’s disease knock-in mice. Brain Res Bull. 2010 May 8; doi: 10.1016/j.brainresbull.2010.05.004. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
- Brooks SP, Janghra N, Workman VL, Bayram-Weston Z, Jones L, Dunnett SB. Longitudinal analysis of the behavioural phenotype in R6/1 (C57BL/6J) Huntington’s disease transgenic mice. Brain Res Bull. 2011 Jan 25; doi: 10.1016/j.brainresbull.2011.01.010. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
- Brzózka MM, Radyushkin K, Wichert SP, Ehrenreich H, Rossner MJ. Cognitive and sensorimotor gating impairments in transgenic mice overexpressing the schizophrenia susceptibility gene Tcf4 in the brain. Biol Psychiatry. 2010;68:33–40. doi: 10.1016/j.biopsych.2010.03.015. [DOI] [PubMed] [Google Scholar]
- Burdick KE, Hodgkinson CA, Szeszko PR, Lencz T, Ekholm JM, Kane JM, Goldman D, Malhotra AK. DISC1 and neurocognitive function in schizophrenia. Neuroreport. 2005;16:1399–1402. doi: 10.1097/01.wnr.0000175248.25535.f6. [DOI] [PubMed] [Google Scholar]
- Cadenhead KS, Geyer MA, Braff DL. Impaired startle prepulse inhibition and habituation in patients with schizotypal personality disorder. Am J Psychiatry. 1993;150:1862–1867. doi: 10.1176/ajp.150.12.1862. [DOI] [PubMed] [Google Scholar]
- Cadenhead KS, Swerdlow NR, Shafer KM, Diaz M, Braff DL. Modulation of the startle response and startle laterality in relatives of schizophrenic patients and in subjects with schizotypal personality disorder: evidence of inhibitory deficits. Am J Psychiatry. 2000;157:1660–1668. doi: 10.1176/appi.ajp.157.10.1660. [DOI] [PubMed] [Google Scholar]
- Cahill ME, Xie Z, Day M, Photowala H, Barbolina MV, Miller CA, Weiss C, Radulovic J, Sweatt JD, Disterhoft JF, Surmeier DJ, Penzes P. Kalirin regulates cortical spine morphogenesis and disease-related behavioral phenotypes. Proc Natl Acad Sci U S A. 2009;106:13058–13063. doi: 10.1073/pnas.0904636106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caldwell HK, Stephens SL, Young WS., 3rd Oxytocin as a natural antipsychotic: a study using oxytocin knockout mice. Mol Psychiatry. 2009;14:190–196. doi: 10.1038/sj.mp.4002150. [DOI] [PubMed] [Google Scholar]
- Callicott JH, Straub RE, Pezawas L, Egan MF, Mattay VS, Hariri AR, Verchinski BA, Meyer-Lindenberg A, Balkissoon R, Kolachana B, Goldberg TE, Weinberger DR. Variation in DISC1 affects hippocampal structure and function and increases risk for schizophrenia. Proc Natl Acad Sci U S A. 2005;102:8627–8632. doi: 10.1073/pnas.0500515102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cannon TD, Hennah W, van Erp TG, Thompson PM, Lonnqvist J, Huttunen M, Gasperoni T, Tuulio-Henriksson A, Pirkola T, Toga AW, Kaprio J, Mazziotta J, Peltonen L. Association of DISC1/TRAX haplotypes with schizophrenia, reduced prefrontal gray matter, and impaired short- and long-term memory. Arch Gen Psychiatry. 2005;62:1205–1213. doi: 10.1001/archpsyc.62.11.1205. [DOI] [PubMed] [Google Scholar]
- Cesca F, Baldelli P, Valtorta F, Benfenati F. The synapsins: key actors of synapse function and plasticity. Prog Neurobiol. 2010;91:313–348. doi: 10.1016/j.pneurobio.2010.04.006. [DOI] [PubMed] [Google Scholar]
- Chadman KK, Gong S, Scattoni ML, Boltuck SE, Gandhy SU, Heintz N, Crawley JN. Minimal aberrant behavioral phenotypes of neuroligin-3 R451C knockin mice. Autism Res. 2008;1:147–158. doi: 10.1002/aur.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chavez C, Gogos A, Jones ME, van den Buuse M. Psychotropic drug-induced locomotor hyperactivity and prepulse inhibition regulation in male and female aromatase knockout (ArKO) mice: role of dopamine D1 and D2 receptors and dopamine transporters. Psychopharmacology (Berl) 2009;206:267–279. doi: 10.1007/s00213-009-1604-6. [DOI] [PubMed] [Google Scholar]
- Chen HH, Stoker A, Markou A. The glutamatergic compounds sarcosine and N-acetylcysteine ameliorate prepulse inhibition deficits in metabotropic glutamate 5 receptor knockout mice. Psychopharmacology (Berl) 2010;209:343–350. doi: 10.1007/s00213-010-1802-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Q, He G, Qin W, Chen QY, Zhao XZ, Duan SW, Liu XM, Feng GY, Xu YF, Clair D, Li M, Wang JH, Xing YL, Shi JG, He L. Family-based association study of synapsin II and schizophrenia. Am J Hum Genet. 2004;75:873–877. doi: 10.1086/425588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y-JJ, Johnson MA, Lieberman MD, Goodchild RE, Schobel S, Lewandowski N, Rosoklija G, Liu R-C, Gingrich JA, Small S, Moore H, Dwork AJ, Talmage DA, Role LW. Type III Neuregulin-1 is required for normal sensorimotor gating, memory-related behaviors, and corticostriatal circuit components. J Neurosci. 2008;28:6872–6883. doi: 10.1523/JNEUROSCI.1815-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen YW, Lai WS. Behavioral phenotyping of v-akt murine thymoma viral oncogene homolog 1-deficient mice reveals a sex-specific prepulse inhibition deficit in females that can be partially alleviated by glycogen synthase kinase-3 inhibitors but not by antipsychotics. Neuroscience. 2011;174:178–189. doi: 10.1016/j.neuroscience.2010.09.056. [DOI] [PubMed] [Google Scholar]
- Chong VZ, Young LT, Mishra RK. cDNA array reveals differential gene expression following chronic neuroleptic administration: implications of synapsin II in haloperidol treatment. J Neurochem. 2002;82:1533–1539. doi: 10.1046/j.1471-4159.2002.01104.x. [DOI] [PubMed] [Google Scholar]
- Cleva RM, Olive MF. Positive allosteric modulators of type 5 metabotropic glutamate receptors (mGluR5) and their therapeutic potential for the treatment of CNS disorders. Molecules. 2011;16:2097–2106. doi: 10.3390/molecules16032097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cole TB, Giordano G, Co AL, Mohar I, Kavanagh TJ, Costa LG. Behavioral characterization of GCLM-knockout mice, a model for enhanced susceptibility to oxidative stress. J Toxicol. 2011;2011 doi: 10.1155/2011/157687. 157687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cook MN, Dunning JP, Wiley RG, Chesler EJ, Johnson DK, Miller DR, Goldowitz D. Neurobehavioral mutants identified in an ENU-mutagenesis project. Mamm Genome. 2007;18:559–572. doi: 10.1007/s00335-007-9035-3. [DOI] [PubMed] [Google Scholar]
- Coyle JT. The GABA-glutamate connection in schizophrenia: which is the proximate cause? Biochem Pharmacol. 2004;68:1507–1514. doi: 10.1016/j.bcp.2004.07.034. [DOI] [PubMed] [Google Scholar]
- Crawley JN. Behavioral phenotyping of transgenic and knockout mice. Wiley; 2007. What’s wrong with my mouse? [Google Scholar]
- Csomor PA, Stadler RR, Feldon J, Yee BK, Geyer MA, Vollenweider FX. Haloperidol differentially modulates prepulse inhibition and p50 suppression in healthy humans stratified for low and high gating levels. Neuropsychopharmacology. 2008;33:497–512. doi: 10.1038/sj.npp.1301421. [DOI] [PubMed] [Google Scholar]
- Darvas M, Morsch M, Racz I, Ahmadi S, Swandulla D, Zimmer A. Modulation of the Ca2+ conductance of nicotinic acetylcholine receptors by Lypd6. Eur Neuropsychopharmacol. 2009;19:670–681. doi: 10.1016/j.euroneuro.2009.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Vrij FM, Levenga J, van der Linde HC, Koekkoek SK, De Zeeuw CI, Nelson DL, Oostra BA, Willemsen R. Rescue of behavioral phenotype and neuronal protrusion morphology in Fmr1 KO mice. Neurobiol Dis. 2008;31:127–132. doi: 10.1016/j.nbd.2008.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deakin IH, Law AJ, Oliver PL, Schwab MH, Nave KA, Harrison PJ, Bannerman DM. Behavioural characterization of neuregulin 1 type I overexpressing transgenic mice. Neuroreport. 2009;20:1523–1528. doi: 10.1097/WNR.0b013e328330f6e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dejaegere T, Serneels L, Schafer MK, Van Biervliet J, Horre K, Depboylu C, Alvarez-Fischer D, Herreman A, Willem M, Haass C, Hoglinger GU, D’Hooge R, De Strooper B. Deficiency of Aph1B/C-gamma-secretase disturbs Nrg1 cleavage and sensorimotor gating that can be reversed with antipsychotic treatment. Proc Natl Acad Sci U S A. 2008;105:9775–9780. doi: 10.1073/pnas.0800507105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeLorey TM, Sahbaie P, Hashemi E, Li WW, Salehi A, Clark DJ. Somatosensory and sensorimotor consequences associated with the heterozygous disruption of the autism candidate gene, Gabrb3. Behav Brain Res. 2011;216(1):36–45. doi: 10.1016/j.bbr.2010.06.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doherty JM, Masten VL, Powell SB, Ralph RJ, Klamer D, Low MJ, Geyer MA. Contributions of dopamine D1, D2, and D3 receptor subtypes to the disruptive effects of cocaine on prepulse inhibition in mice. Neuropsychopharmacology. 2008;33:2648–2656. doi: 10.1038/sj.npp.1301657. [DOI] [PubMed] [Google Scholar]
- Duffy L, Cappas E, Scimone A, Schofield PR, Karl T. Behavioral profile of a heterozygous mutant mouse model for EGF-like domain neuregulin 1. Behav Neurosci. 2008;122:748–759. doi: 10.1037/0735-7044.122.4.748. [DOI] [PubMed] [Google Scholar]
- Dulawa SC, Hen R, Scearce-Levie K, Geyer MA. Serotonin1B receptor modulation of startle reactivity, habituation, and prepulse inhibition in wild-type and serotonin1B knockout mice. Psychopharmacology (Berl) 1997;132:125–134. doi: 10.1007/s002130050328. [DOI] [PubMed] [Google Scholar]
- Duncan GE, Inada K, Koller BH, Moy SS. Increased sensitivity to kainic acid in a genetic model of reduced NMDA receptor function. Brain Res. 2010;1307:166–176. doi: 10.1016/j.brainres.2009.10.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duncan GE, Moy SS, Perez A, Eddy DM, Zinzow WM, Lieberman JA, Snouwaert JN, Koller BH. Deficits in sensorimotor gating and tests of social behavior in a genetic model of reduced NMDA receptor function. Behav Brain Res. 2004;153:507–519. doi: 10.1016/j.bbr.2004.01.008. [DOI] [PubMed] [Google Scholar]
- Dyck BA, Skoblenick KJ, Castellano JM, Ki K, Thomas N, Mishra RK. Synapsin II knockout mice show sensorimotor gating and behavioural abnormalities similar to those in the phencyclidine-induced preclinical animal model of schizophrenia. Schizophr Res. 2007;97:292–293. doi: 10.1016/j.schres.2007.08.026. [DOI] [PubMed] [Google Scholar]
- Dyck BA, Skoblenick KJ, Castellano JM, Ki K, Thomas N, Mishra RK. Behavioral abnormalities in synapsin II knockout mice implicate a causal factor in schizophrenia. Synapse. 2009;63:662–672. doi: 10.1002/syn.20643. [DOI] [PubMed] [Google Scholar]
- Eells JB, Lipska BK, Yeung SK, Misler JA, Nikodem VM. Nurr1-null heterozygous mice have reduced mesolimbic and mesocortical dopamine levels and increased stress-induced locomotor activity. Behav Brain Res. 2002;136:267–275. doi: 10.1016/s0166-4328(02)00185-7. [DOI] [PubMed] [Google Scholar]
- Eells JB, Misler JA, Nikodem VM. Early postnatal isolation reduces dopamine levels, elevates dopamine turnover and specifically disrupts prepulse inhibition in Nurr1-null heterozygous mice. Neuroscience. 2006;140:1117–1126. doi: 10.1016/j.neuroscience.2005.12.065. [DOI] [PubMed] [Google Scholar]
- Egashira N, Mishima K, Iwasaki K, Oishi R, Fujiwara M. New topics in vasopressin receptors and approach to novel drugs: role of the vasopressin receptor in psychological and cognitive functions. J Pharmacol Sci. 2009;109:44–49. doi: 10.1254/jphs.08r14fm. [DOI] [PubMed] [Google Scholar]
- Errico F, Rossi S, Napolitano F, Catuogno V, Topo E, Fisone G, D’Aniello A, Centonze D, Usiello A. D-aspartate prevents corticostriatal long-term depression and attenuates schizophrenia-like symptoms induced by amphetamine and MK-801. J Neurosci. 2008;28:10404–10414. doi: 10.1523/JNEUROSCI.1618-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Etherton MR, Blaiss CA, Powell CM, Sudhof TC. Mouse neurexin-1alpha deletion causes correlated electrophysiological and behavioral changes consistent with cognitive impairments. Proc Natl Acad Sci U S A. 2009;106:17998–18003. doi: 10.1073/pnas.0910297106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feifel D, Macdonald K, Nguyen A, Cobb P, Warlan H, Galangue B, Minassian A, Becker O, Cooper J, Perry W, Lefebvre M, Gonzales J, Hadley A. Adjunctive intranasal oxytocin reduces symptoms in schizophrenia patients. Biol Psychiatry. 2010a;68:678–680. doi: 10.1016/j.biopsych.2010.04.039. [DOI] [PubMed] [Google Scholar]
- Feifel D, Pang Z, Shilling PD, Melendez G, Schreiber R, Button D. Effects of neurotensin-2 receptor deletion on sensorimotor gating and locomotor activity. Behav Brain Res. 2010b;212:174–178. doi: 10.1016/j.bbr.2010.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feifel D, Pang Z, Shilling PD, Melendez G, Schreiber R, Button D. Sensorimotor gating in neurotensin-1 receptor null mice. Neuropharmacology. 2010c;58:173–178. doi: 10.1016/j.neuropharm.2009.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fendt M, Buchi M, Burki H, Imobersteg S, Ricoux B, Suply T, Sailer AW. Neuropeptide S receptor deficiency modulates spontaneous locomotor activity and the acoustic startle response. Behav Brain Res. 2011;217:1–9. doi: 10.1016/j.bbr.2010.09.022. [DOI] [PubMed] [Google Scholar]
- Fendt M, Burki H, Imobersteg S, van der Putten H, McAllister K, Leslie JC, Shaw D, Holscher C. The effect of mGlu8 deficiency in animal models of psychiatric diseases. Genes Brain Behav. 2010;9:33–44. doi: 10.1111/j.1601-183X.2009.00532.x. [DOI] [PubMed] [Google Scholar]
- Francis DD, Szegda K, Campbell G, Martin WD, Insel TR. Epigenetic sources of behavioral differences in mice. Nat Neurosci. 2003;6:445–446. doi: 10.1038/nn1038. [DOI] [PubMed] [Google Scholar]
- Frankland PW, Wang Y, Rosner B, Shimizu T, Balleine BW, Dykens EM, Ornitz EM, Silva AJ. Sensorimotor gating abnormalities in young males with fragile X syndrome and Fmr1-knockout mice. Mol Psychiatry. 2004;9:417–425. doi: 10.1038/sj.mp.4001432. [DOI] [PubMed] [Google Scholar]
- Frankle WG, Lerma J, Laruelle M. The synaptic hypothesis of schizophrenia. Neuron. 2003;39:205–216. doi: 10.1016/s0896-6273(03)00423-9. [DOI] [PubMed] [Google Scholar]
- Fridlund AJ, Cacioppo JT. Guidelines for human electromyographic research. Psychophysiology. 1986;23(5):567–589. doi: 10.1111/j.1469-8986.1986.tb00676.x. [DOI] [PubMed] [Google Scholar]
- Friston KJ, Frith CD. Schizophrenia: a disconnection syndrome? Clin Neurosci. 1995;3:89–97. [PubMed] [Google Scholar]
- Frost WN, Tian LM, Hoppe TA, Mongeluzi DL, Wang J. A cellular mechanism for prepulse inhibition. Neuron. 2003;40:991–1001. doi: 10.1016/s0896-6273(03)00731-1. [DOI] [PubMed] [Google Scholar]
- Fukuda T, Hashimoto H, Okayasu N, Kameyama A, Onogi H, Nakagawasai O, Nakazawa T, Kurosawa T, Hao Y, Isaji T, Tadano T, Narimatsu H, Taniguchi N, Gu J. {alpha}1, 6-Fucosyltransferase-deficient mice exhibit multiple behavioral abnormalities associated with a schizophrenia-like phenotype: importance of the balance between the dopamine and serotonin systems. J Biol Chem. 2011;286:18434–18443. doi: 10.1074/jbc.M110.172536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Galici R, Boggs JD, Miller KL, Bonaventure P, Atack JR. Effects of SB-269970, a 5-HT7 receptor antagonist, in mouse models predictive of antipsychotic-like activity. Behav Pharmacol. 2008;19:153–159. doi: 10.1097/FBP.0b013e3282f62d8c. [DOI] [PubMed] [Google Scholar]
- Gallinat J, Bajbouj M, Sander T, Schlattmann P, Xu K, Ferro EF, Goldman D, Winterer G. Association of the G1947A COMT (Val(108/158)Met) gene polymorphism with prefrontal P300 during information processing. Biol Psychiatry. 2003;54:40–48. doi: 10.1016/s0006-3223(02)01973-x. [DOI] [PubMed] [Google Scholar]
- Geyer MA, Braff DL. Startle habituation and sensorimotor gating in schizophrenia and related animal models. Schizophr Bull. 1987;13:643–668. doi: 10.1093/schbul/13.4.643. [DOI] [PubMed] [Google Scholar]
- Geyer MA, Dulawa SC. Chapter 8 Assessment of murine startle reactivity, prepulse inhibition, and habituation. Curr Protoc Neurosci. 2003;(Unit 8):17. doi: 10.1002/0471142301.ns0817s24. [DOI] [PubMed] [Google Scholar]
- Geyer MA, Markou A. The role of preclinical models in the development of psychotropic drugs. In: Davis KL, Charney D, Coyle JT, Nemeroff CB, editors. Neuropsychopharmacology: the fifth generation of progress. Lippincott: Williams Wilkins; 2002. pp. 445–455. [Google Scholar]
- Geyer MA, McIlwain KL, Paylor R. Mouse genetic models for prepulse inhibition: an early review. Mol Psychiatry. 2002;7:1039–1053. doi: 10.1038/sj.mp.4001159. [DOI] [PubMed] [Google Scholar]
- Giakoumaki SG, Bitsios P, Frangou S. The level of prepulse inhibition in healthy individuals may index cortical modulation of early information processing. Brain Res. 2006;1078:168–170. doi: 10.1016/j.brainres.2006.01.056. [DOI] [PubMed] [Google Scholar]
- Giakoumaki SG, Roussos P, Bitsios P. Improvement of prepulse inhibition and executive function by the COMT inhibitor tolcapone depends on COMT Val158Met polymorphism. Neuropsychopharmacology. 2008;33:3058–3068. doi: 10.1038/npp.2008.82. [DOI] [PubMed] [Google Scholar]
- Gogos JA, Morgan M, Luine V, Santha M, Ogawa S, Pfaff D, Karayiorgou M. Catechol-O-methyltransferase-deficient mice exhibit sexually dimorphic changes in catecholamine levels and behavior. Proc Natl Acad Sci U S A. 1998;95:9991–9996. doi: 10.1073/pnas.95.17.9991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Golimbet V, Gritsenko I, Alfimova M, Lebedeva I, Lezheiko T, Abramova L, Kaleda V, Ebstein R. Association study of COMT gene Val158Met polymorphism with auditory P300 and performance on neurocognitive tests in patients with schizophrenia and their relatives. World J Biol Psychiatry. 2006;7:238–245. doi: 10.1080/15622970600670970. [DOI] [PubMed] [Google Scholar]
- Gottesman II, Gould TD. The endophenotype concept in psychiatry: etymology and strategic intentions. Am J Psychiatry. 2003;160:636–645. doi: 10.1176/appi.ajp.160.4.636. [DOI] [PubMed] [Google Scholar]
- Gottesman II. Schizophrenia Genesis. WH Freeman; 1991. [Google Scholar]
- Gould TD, Gottesman II. Psychiatric endophenotypes and the development of valid animal models. Genes Brain Behav. 2006;5:113–119. doi: 10.1111/j.1601-183X.2005.00186.x. [DOI] [PubMed] [Google Scholar]
- Graham FK. Presidential address, 1974. The more or less startling effects of weak prestimulation. Psychophysiology. 1975;12:238–248. doi: 10.1111/j.1469-8986.1975.tb01284.x. [DOI] [PubMed] [Google Scholar]
- Grant A, Hoops D, Labelle-Dumais C, Prevost M, Rajabi H, Kolb B, Stewart J, Arvanitogiannis A, Flores C. Netrin-1 receptor-deficient mice show enhanced mesocortical dopamine transmission and blunted behavioural responses to amphetamine. Eur J Neurosci. 2007;26:3215–3228. doi: 10.1111/j.1460-9568.2007.05888.x. [DOI] [PubMed] [Google Scholar]
- Gray L, van den Buuse M, Scarr E, Dean B, Hannan AJ. Clozapine reverses schizophrenia-related behaviours in the metabotropic glutamate receptor 5 knockout mouse: association with N-methyl-d-aspartic acid receptor up-regulation. Int J Neuropsychopharmacol. 2009;12:45–60. doi: 10.1017/S1461145708009085. [DOI] [PubMed] [Google Scholar]
- Green MF, Butler PD, Chen Y, Geyer MA, Silverstein S, Wynn JK, Yoon JH, Zemon V. Perception measurement in clinical trials of schizophrenia: promising paradigms from CNTRICS. Schizophr Bull. 2009;35:163–181. doi: 10.1093/schbul/sbn156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greenwood TA, Braff DL, Light GA, Cadenhead KS, Calkins ME, Dobie DJ, Freedman R, Green MF, Gur RE, Gur RC, Mintz J, Nuechterlein KH, Olincy A, Radant AD, Seidman LJ, Siever LJ, Silverman JM, Stone WS, Swerdlow NR, Tsuang DW, Tsuang MT, Turetsky BI, Schork NJ. Initial heritability analyses of endophenotypic measures for schizophrenia: the consortium on the genetics of schizophrenia. Arch Gen Psychiatry. 2007;64:1242–1250. doi: 10.1001/archpsyc.64.11.1242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greenwood TA, Lazzeroni LC, Murray SS, Cadenhead KS, Calkins ME, Dobie DJ, Green MF, Gur RE, Gur RC, Hardiman G, Kelsoe JR, Leonard S, Light GA, Nuechterlein KH, Olincy A, Radant AD, Schork NJ, Seidman LJ, Siever LJ, Silverman JM, Stone WS, Swerdlow NR, Tsuang DW, Tsuang MT, Turetsky BI, Freedman R, Braff DL. Analysis of 94 candidate genes and 12 endophenotypes for schizophrenia from the consortium on the genetics of schizophrenia. Am J Psychiatry. 2011 doi: 10.1176/appi.ajp.2011.10050723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Groenink L, Bijlsma EY, van Bogaert MJ, Oosting RS, Olivier B. Serotonin1A receptor deletion does not interact with maternal separation-induced increases in startle reactivity and prepulse inhibition deficits. Psychopharmacology (Berl) 2011;214:353–365. doi: 10.1007/s00213-010-1998-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Groenink L, Dirks A, Verdouw PM, de Graaff M, Peeters BW, Millan MJ, Olivier B. CRF1 not glucocorticoid receptors mediate prepulse inhibition deficits in mice overexpressing CRF. Biol Psychiatry. 2008;63:360–368. doi: 10.1016/j.biopsych.2007.06.002. [DOI] [PubMed] [Google Scholar]
- Gruart A, Lopez-Ramos JC, Munoz MD, Delgado-Garcia JM. Aged wild-type and APP, PS1, and APP+PS1 mice present similar deficits in associative learning and synaptic plasticity independent of amyloid load. Neurobiol Dis. 2008;30:439–450. doi: 10.1016/j.nbd.2008.03.001. [DOI] [PubMed] [Google Scholar]
- Grunze HC, Rainnie DG, Hasselmo ME, Barkai E, Hearn EF, McCarley RW, Greene RW. NMDA-dependent modulation of CA1 local circuit inhibition. J Neurosci. 1996;16:2034–2043. doi: 10.1523/JNEUROSCI.16-06-02034.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo X, Hamilton PJ, Reish NJ, Sweatt JD, Miller CA, Rumbaugh G. Reduced expression of the NMDA receptor-interacting protein SynGAP causes behavioral abnormalities that model symptoms of Schizophrenia. Neuropsychopharmacology. 2009;34:1659–1672. doi: 10.1038/npp.2008.223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han L, Picker JD, Schaevitz LR, Tsai G, Feng J, Jiang Z, Chu HC, Basu AC, Berger-Sweeney J, Coyle JT. Phenotypic characterization of mice heterozygous for a null mutation of glutamate carboxypeptidase II. Synapse. 2009;63:625–635. doi: 10.1002/syn.20649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harrison PJ, Weinberger DR. Schizophrenia genes, gene expression, and neuropathology: on the matter of their convergence. Mol Psychiatry. 2005;10:40–68. doi: 10.1038/sj.mp.4001558. image 5. [DOI] [PubMed] [Google Scholar]
- Hasenkamp W, Epstein MP, Green A, Wilcox L, Boshoven W, Lewison B, Duncan E. Heritability of acoustic startle magnitude, prepulse inhibition, and startle latency in schizophrenia and control families. Psychiatry Res. 2010;178:236–243. doi: 10.1016/j.psychres.2009.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hashimoto R, Hashimoto H, Shintani N, Chiba S, Hattori S, Okada T, Nakajima M, Tanaka K, Kawagishi N, Nemoto K, Mori T, Ohnishi T, Noguchi H, Hori H, Suzuki T, Iwata N, Ozaki N, Nakabayashi T, Saitoh O, Kosuga A, Tatsumi M, Kamijima K, Weinberger DR, Kunugi H, Baba A. Pituitary adenylate cyclase-activating polypeptide is associated with schizophrenia. Mol Psychiatry. 2007;12:1026–1032. doi: 10.1038/sj.mp.4001982. [DOI] [PubMed] [Google Scholar]
- Heintz N. BAC to the future: the use of BAC transgenic mice for neuroscience research. Nat Rev Neurosci. 2001;2:861–870. doi: 10.1038/35104049. [DOI] [PubMed] [Google Scholar]
- Hennah W, Tuulio-Henriksson A, Paunio T, Ekelund J, Varilo T, Partonen T, Cannon TD, Lonnqvist J, Peltonen L. A haplotype within the DISC1 gene is associated with visual memory functions in families with a high density of schizophrenia. Mol Psychiatry. 2005;10:1097–1103. doi: 10.1038/sj.mp.4001731. [DOI] [PubMed] [Google Scholar]
- Hessl D, Berry-Kravis E, Cordeiro L, Yuhas J, Ornitz EM, Campbell A, Chruscinski E, Hervey C, Long JM, Hagerman RJ. Prepulse inhibition in fragile X syndrome: feasibility, reliability, and implications for treatment. Am J Med Genet B Neuropsychiatr Genet. 2009;150B:545–553. doi: 10.1002/ajmg.b.30858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hikida T, Jaaro-Peled H, Seshadri S, Oishi K, Hookway C, Kong S, Wu D, Xue R, Andrade M, Tankou S, Mori S, Gallagher M, Ishizuka K, Pletnikov M, Kida S, Sawa A. Dominant-negative DISC1 transgenic mice display schizophrenia-associated phenotypes detected by measures translatable to humans. Proc Natl Acad Sci U S A. 2007;104:14501–14506. doi: 10.1073/pnas.0704774104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hill RA, Murray SS, Halley PG, Binder MD, Martin SJ, van den Buuse M. Brain-derived neurotrophic factor expression is increased in the hippocampus of 5-HT(2C) receptor knockout mice. Hippocampus. 2011;21:434–445. doi: 10.1002/hipo.20759. [DOI] [PubMed] [Google Scholar]
- Hitzemann R, Malmanger B, Belknap J, Darakjian P, McWeeney S. Short-term selective breeding for high and low prepulse inhibition of the acoustic startle response; pharmacological characterization and QTL mapping in the selected lines. Pharmacol Biochem Behav. 2008;90:525–533. doi: 10.1016/j.pbb.2008.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoffman HS, Ison JR. Reflex modification in the domain of startle: I. Some empirical findings and their implications for how the nervous system processes sensory input. Psychol Rev. 1980;87:175–189. [PubMed] [Google Scholar]
- Hokyo A, Kanazawa T, Uenishi H, Tsutsumi A, Kawashige S, Kikuyama H, Glatt SJ, Koh J, Nishimoto Y, Matsumura H, Motomura N, Yoneda H. Habituation in prepulse inhibition is affected by a polymorphism on the NMDA receptor 2B subunit gene (GRIN2B) Psychiatr Genet. 2010;20:191–198. doi: 10.1097/YPG.0b013e32833a201d. [DOI] [PubMed] [Google Scholar]
- Horii Y, Yamasaki N, Miyakawa T, Shiosaka S. Increased anxiety-like behavior in neuropsin (kallikrein-related peptidase 8) gene-deficient mice. Behav Neurosci. 2008;122(3):498–504. doi: 10.1037/0735-7044.122.3.498. [DOI] [PubMed] [Google Scholar]
- Ibi D, Nagai T, Koike H, Kitahara Y, Mizoguchi H, Niwa M, Jaaro-Peled H, Nitta A, Yoneda Y, Nabeshima T, Sawa A, Yamada K. Combined effect of neonatal immune activation and mutant DISC1 on phenotypic changes in adulthood. Behav Brain Res. 2010;206:32–37. doi: 10.1016/j.bbr.2009.08.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishihama T, Ago Y, Shintani N, Hashimoto H, Baba A, Takuma K, Matsuda T. Environmental factors during early developmental period influence psychobehavioral abnormalities in adult PACAP-deficient mice. Behav Brain Res. 2010;209:274–280. doi: 10.1016/j.bbr.2010.02.009. [DOI] [PubMed] [Google Scholar]
- Jackson ME, Homayoun H, Moghaddam B. NMDA receptor hypofunction produces concomitant firing rate potentiation and burst activity reduction in the prefrontal cortex. Proc Natl Acad Sci U S A. 2004;101:8467–8472. doi: 10.1073/pnas.0308455101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobson LH, Cryan JF. Genetic approaches to modeling anxiety in animals. Curr Top Behav Neurosci. 2010;2:161–201. doi: 10.1007/7854_2009_31. [DOI] [PubMed] [Google Scholar]
- Javitt DC. Glutamate as a therapeutic target in psychiatric disorders. Mol Psychiatry. 2004;9:984–997. doi: 10.1038/sj.mp.4001551. [DOI] [PubMed] [Google Scholar]
- Javitt DC, Zukin SR. Recent advances in the phencyclidine model of schizophrenia. Am J Psychiatry. 1991;148:1301–1308. doi: 10.1176/ajp.148.10.1301. [DOI] [PubMed] [Google Scholar]
- Jones DNC, Gartlon JE, Minassian A, Perry W, Geyer MA. Developing new drugs for schizophrenia: from animals to the clinic. In: Borsini R, McArthur R, editors. Animal and translational models for CNS drug discovery. New York: Psychiatric Disorders Elsevier; 2008. pp. 199–262. [Google Scholar]
- Joober R, Zarate JM, Rouleau GA, Skamene E, Boksa P. Provisional mapping of quantitative trait loci modulating the acoustic startle response and prepulse inhibition of acoustic startle. Neuropsychopharmacol. 2002;27:765–781. doi: 10.1016/S0893-133X(02)00333-0. [DOI] [PubMed] [Google Scholar]
- Kaidanovich-Beilin O, Lipina TV, Takao K, van Eede M, Hattori S, Laliberte C, Khan M, Okamoto K, Chambers JW, Fletcher PJ, Macaulay K, Doble BW, Henkelman M, Miyakawa T, Roder J, Woodgett JR. Abnormalities in brain structure and behavior in GSK-3alpha mutant mice. Mol Brain. 2009;2:35. doi: 10.1186/1756-6606-2-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kapfhamer D, Berger KH, Hopf FW, Seif T, Kharazia V, Bonci A, Heberlein U. Protein Phosphatase 2a and glycogen synthase kinase 3 signaling modulate prepulse inhibition of the acoustic startle response by altering cortical M-Type potassium channel activity. J Neurosci. 2010;30:8830–8840. doi: 10.1523/JNEUROSCI.1292-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karl T, Chesworth R, Duffy L, Herzog H. Acoustic startle response and sensorimotor gating in a genetic mouse model for the Y1 receptor. Neuropeptides. 2010a;44:233–239. doi: 10.1016/j.npep.2009.12.008. [DOI] [PubMed] [Google Scholar]
- Karl T, Chesworth R, Duffy L, Herzog H. Schizophrenia-relevant behaviours in a genetic mouse model for Y2 deficiency. Behav Brain Res. 2010b;207:434–440. doi: 10.1016/j.bbr.2009.10.029. [DOI] [PubMed] [Google Scholar]
- Karlsson RM, Tanaka K, Saksida LM, Bussey TJ, Heilig M, Holmes A. Assessment of glutamate transporter GLAST (EAAT1)-deficient mice for phenotypes relevant to the negative and executive/cognitive symptoms of schizophrenia. Neuropsychopharmacol. 2009;34:1578–1589. doi: 10.1038/npp.2008.215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karmacharya R, Lynn SK, Demarco S, Ortiz A, Wang X, Lundy MY, Xie Z, Cohen BM, Miller GM, Buttner EA. Behavioral effects of clozapine: involvement of trace amine pathways in C. elegans and M. musculus. Brain Res. 2011;1393:91–99. doi: 10.1016/j.brainres.2011.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karper LP, Freeman GK, Grillon C, Morgan CA, 3rd, Charney DS, Krystal JH. Preliminary evidence of an association between sensorimotor gating and distractibility in psychosis. J Neuropsychiatry Clin Neurosci. 1996;8:60–66. doi: 10.1176/jnp.8.1.60. [DOI] [PubMed] [Google Scholar]
- Kato T, Kasai A, Mizuno M, Fengyi L, Shintani N, Maeda S, Yokoyama M, Ozaki M, Nawa H. Phenotypic characterization of transgenic mice overexpressing neuregulin-1. PLoS One. 2010;5(12):e14185. doi: 10.1371/journal.pone.0014185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelly MP, Stein JM, Vecsey CG, Favilla C, Yang X, Bizily SF, Esposito MF, Wand G, Kanes SJ, Abel T. Developmental etiology for neuroanatomical and cognitive deficits in mice overexpressing Galphas, a G-protein subunit genetically linked to schizophrenia. Mol Psychiatry. 2009;14(398–415):347. doi: 10.1038/mp.2008.124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim YS, Harry GJ, Kang HS, Goulding D, Wine RN, Kissling GE, Liao G, Jetten AM. Altered cerebellar development in nuclear receptor TAK1/TR4 null mice is associated with deficits in GLAST(+) glia, alterations in social behavior, motor learning, startle reactivity, and microglia. Cerebellum. 2010 doi: 10.1007/s12311-010-0163-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kinney GG, Burno M, Campbell UC, Hernandez LM, Rodriguez D, Bristow LJ, Conn PJ. Metabotropic glutamate subtype 5 receptors modulate locomotor activity and sensorimotor gating in rodents. J Pharmacol Exp Ther. 2003;306:116–123. doi: 10.1124/jpet.103.048702. [DOI] [PubMed] [Google Scholar]
- Koga M, Ishiguro H, Yazaki S, Horiuchi Y, Arai M, Niizato K, Iritani S, Itokawa M, Inada T, Iwata N, Ozaki N, Ujike H, Kunugi H, Sasaki T, Takahashi M, Watanabe Y, Someya T, Kakita A, Takahashi H, Nawa H, Muchardt C, Yaniv M, Arinami T. Involvement of SMARCA2/BRM in the SWI/SNF chromatin-remodeling complex in schizophrenia. Hum Mol Genet. 2009;18:2483–2494. doi: 10.1093/hmg/ddp166. [DOI] [PubMed] [Google Scholar]
- Koh HY, Kim D, Lee J, Lee S, Shin HS. Deficits in social behavior and sensorimotor gating in mice lacking phospholipase Cbeta1. Genes Brain Behav. 2008;7:120–128. doi: 10.1111/j.1601-183X.2007.00351.x. [DOI] [PubMed] [Google Scholar]
- Koumura A, Kakefuda K, Honda A, Ito Y, Tsuruma K, Shimazawa M, Uchida Y, Hozumi I, Satoh M, Inuzuka T, Hara H. Metallothionein-3 deficient mice exhibit abnormalities of psychological behaviors. Neurosci Lett. 2009;467:11–14. doi: 10.1016/j.neulet.2009.09.051. [DOI] [PubMed] [Google Scholar]
- Krystal J, D’Souza DC, Mathalon D, Perry E, Belger A, Hoffman R. NMDA receptor antagonist effects, cortical glutamatergic function, and schizophrenia: toward a paradigm shift in medication development. Psychopharmacol. 2003;169:215–233. doi: 10.1007/s00213-003-1582-z. [DOI] [PubMed] [Google Scholar]
- Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, Heninger GR, Bowers MB, Jr, Charney DS. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry. 1994;51:199–214. doi: 10.1001/archpsyc.1994.03950030035004. [DOI] [PubMed] [Google Scholar]
- Kumari V, Fannon D, Geyer MA, Premkumar P, Antonova E, Simmons A, Kuipers E. Cortical grey matter volume and sensorimotor gating in schizophrenia. Cortex. 2008;44:1206–1214. doi: 10.1016/j.cortex.2007.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumari V, Gray JA, Geyer MA, ffytche D, Soni W, Mitterschiffthaler MT, Vythelingum GN, Simmons A, Williams SC, Sharma T. Neural correlates of tactile prepulse inhibition: a functional MRI study in normal and schizophrenic subjects. Psychiatry Res. 2003;122:99–113. doi: 10.1016/s0925-4927(02)00123-3. [DOI] [PubMed] [Google Scholar]
- Levenga J, Hayashi S, de Vrij FM, Koekkoek SK, van der Linde HC, Nieuwenhuizen I, Song C, Buijsen RA, Pop AS, Gomezmancilla B, Nelson DL, Willemsen R, Gasparini F, Oostra BA. AFQ056, a new mGluR5 antagonist for treatment of fragile X syndrome. Neurobiol Dis. 2011;42(3):311–317. doi: 10.1016/j.nbd.2011.01.022. [DOI] [PubMed] [Google Scholar]
- Lewis DA, Hashimoto T, Volk DW. Cortical inhibitory neurons and schizophrenia. Nat Rev Neurosci. 2005;6:312–324. doi: 10.1038/nrn1648. [DOI] [PubMed] [Google Scholar]
- Lipina TV, Kaidanovich-Beilin O, Patel S, Wang M, Clapcote SJ, Liu F, Woodgett JR, Roder JC. Genetic and pharmacological evidence for schizophrenia-related DISC1 interaction with GSK-3. Synapse. 2011;65:234–248. doi: 10.1002/syn.20839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lipina TV, Niwa M, Jaaro-Peled H, Fletcher PJ, Seeman P, Sawa A, Roder JC. Enhanced dopamine function in DISC1-L100P mutant mice: implications for schizophrenia. Genes Brain Behav. 2010;9:777–789. doi: 10.1111/j.1601-183X.2010.00615.x. [DOI] [PubMed] [Google Scholar]
- Logue SF, Grauer SM, Paulsen J, Graf R, Taylor N, Sung MA, Zhang L, Hughes Z, Pulito VL, Liu F, Rosenzweig-Lipson S, Brandon NJ, Marquis KL, Bates B, Pausch M. The orphan GPCR, GPR88, modulates function of the striatal dopamine system: a possible therapeutic target for psychiatric disorders? Mol Cell Neurosci. 2009;42:438–447. doi: 10.1016/j.mcn.2009.09.007. [DOI] [PubMed] [Google Scholar]
- López-Ramos JC, Tomioka Y, Morimatsu M, Yamamoto S, Ozaki K, Ono E, Delgado-García JM. Motor-coordination-dependent learning, more than others, is impaired in transgenic mice expressing pseudorabies virus immediate-early protein IE180. PLoS One. 2010;5(8):e12123. doi: 10.1371/journal.pone.0012123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Low NC, Hardy J. What is a schizophrenic mouse? Neuron. 2007;54:348–349. doi: 10.1016/j.neuron.2007.04.014. [DOI] [PubMed] [Google Scholar]
- Lu BY, Martin KE, Edgar JC, Smith AK, Lewis SF, Escamilla MA, Miller GA, Canive JM. Effect of catechol O-methyltransferase val(158)met polymorphism on the p50 gating endophenotype in schizophrenia. Biol Psychiatry. 2007;62:822–825. doi: 10.1016/j.biopsych.2006.11.030. [DOI] [PubMed] [Google Scholar]
- Ludewig K, Geyer MA, Vollenweider FX. Deficits in prepulse inhibition and habituation in never-medicated, first-episode schizophrenia. Biol Psychiatry. 2003;54:121–128. doi: 10.1016/s0006-3223(02)01925-x. [DOI] [PubMed] [Google Scholar]
- Ludewig S, Ludewig K, Geyer MA, Hell D, Vollenweider FX. Prepulse inhibition deficits in patients with panic disorder. Depress Anxiety. 2002;15:55–60. doi: 10.1002/da.10026. [DOI] [PubMed] [Google Scholar]
- Mackeprang T, Kristiansen KT, Glenthoj BY. Effects of antipsychotics on prepulse inhibition of the startle response in drug-naive schizophrenic patients. Biol Psychiatry. 2002;52:863–873. doi: 10.1016/s0006-3223(02)01409-9. [DOI] [PubMed] [Google Scholar]
- Mackie S, Millar JK, Porteous DJ. Role of DISC1 in neural development and schizophrenia. Curr Opin Neurobiol. 2007;17:95–102. doi: 10.1016/j.conb.2007.01.007. [DOI] [PubMed] [Google Scholar]
- Malhotra AK, Adler CM, Kennison SD, Elman I, Pickar D, Breier A. Clozapine blunts N-methyl-D-aspartate antagonist-induced psychosis: a study with ketamine. Biol Psychiatry. 1997a;42:664–668. doi: 10.1016/s0006-3223(96)00546-x. [DOI] [PubMed] [Google Scholar]
- Malhotra AK, Pinals DA, Adler CM, Elman I, Clifton A, Pickar D, Breier A. Ketamine-induced exacerbation of psychotic symptoms and cognitive impairment in neuroleptic-free schizophrenics. Neuropsychopharmacol. 1997b;17:141–150. doi: 10.1016/S0893-133X(97)00036-5. [DOI] [PubMed] [Google Scholar]
- Mansuy IM, Bujard H. Tetracycline-regulated gene expression in the brain. Curr Opin Neurobiol. 2000;10:593–596. doi: 10.1016/s0959-4388(00)00127-6. [DOI] [PubMed] [Google Scholar]
- Martin S, Markus MA, Morris BJ, Davisson RL, Lawrence AJ, van den Buuse M. Does angiotensin interact with dopaminergic mechanisms in the brain to modulate prepulse inhibition in mice? Neuropharmacol. 2008;54:399–404. doi: 10.1016/j.neuropharm.2007.10.008. [DOI] [PubMed] [Google Scholar]
- Martinez-Gras I, Rubio G, del Manzano BA, Rodriguez-Jimenez R, Garcia-Sanchez F, Bagney A, Leza JC, Borrell J. The relationship between prepulse inhibition and general psychopathology in patients with schizophrenia treated with long-acting risperidone. Schizophr Res. 2009;115(2–3):215–221. doi: 10.1016/j.schres.2009.09.035. [DOI] [PubMed] [Google Scholar]
- Matsumoto M, Straub RE, Marenco S, Nicodemus KK, Matsumoto S, Fujikawa A, Miyoshi S, Shobo M, Takahashi S, Yarimizu J, Yuri M, Hiramoto M, Morita S, Yokota H, Sasayama T, Terai K, Yoshino M, Miyake A, Callicott JH, Egan MF, Meyer-Lindenberg A, Kempf L, Honea R, Vakkalanka RK, Takasaki J, Kamohara M, Soga T, Hiyama H, Ishii H, Matsuo A, Nishimura S, Matsuoka N, Kobori M, Matsushime H, Katoh M, Furuichi K, Weinberger DR. The evolutionarily conserved G protein-coupled receptor SREB2/GPR85 influences brain size, behavior, and vulnerability to schizophrenia. Proc Natl Acad Sci U S A. 2008;105:6133–6138. doi: 10.1073/pnas.0710717105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matsuo N, Tanda K, Nakanishi K, Yamasaki N, Toyama K, Takao K, Takeshima H, Miyakawa T. Comprehensive behavioral phenotyping of ryanodine receptor type 3 (RyR3) knockout mice: decreased social contact duration in two social interaction tests. Front Behav Neurosci. 2009;3:3. doi: 10.3389/neuro.08.003.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mayford M, Bach ME, Huang YY, Wang L, Hawkins RD, Kandel ER. Control of memory formation through regulated expression of a CaMKII transgene. Science. 1996;274:1678–1683. doi: 10.1126/science.274.5293.1678. [DOI] [PubMed] [Google Scholar]
- McGlashan TH, Hoffman RE. Schizophrenia as a disorder of developmentally reduced synaptic connectivity. Arch Gen Psychiatry. 2000;57:637–648. doi: 10.1001/archpsyc.57.7.637. [DOI] [PubMed] [Google Scholar]
- McOmish CE, Burrows E, Howard M, Scarr E, Kim D, Shin HS, Dean B, van den Buuse M, Hannan AJ. Phospholipase C-beta1 knockout mice exhibit endophenotypes modeling schizophrenia which are rescued by environmental enrichment and clozapine administration. Mol Psychiatry. 2008;13:661–672. doi: 10.1038/sj.mp.4002046. [DOI] [PubMed] [Google Scholar]
- Meincke U, Morth D, Voss T, Thelen B, Geyer MA, Gouzoulis-Mayfrank E. Prepulse inhibition of the acoustically evoked startle reflex in patients with an acute schizophrenic psychosis–a longitudinal study. Eur Arch Psychiatry Clin Neurosci. 2004;254:415–421. doi: 10.1007/s00406-004-0523-0. [DOI] [PubMed] [Google Scholar]
- Mejias R, Adamczyk A, Anggono V, Niranjan T, Thomas GM, Sharma K, Skinner C, Schwartz CE, Stevenson RE, Fallin MD, Kaufmann W, Pletnikov M, Valle D, Huganir RL, Wang T. Gain-of-function glutamate receptor interacting protein 1 variants alter GluA2 recycling and surface distribution in patients with autism. Proc Natl Acad Sci U S A. 2011;108:4920–4925. doi: 10.1073/pnas.1102233108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Menalled L, El-Khodor BF, Patry M, Suarez-Farinas M, Orenstein SJ, Zahasky B, Leahy C, Wheeler V, Yang XW, MacDonald M, Morton AJ, Bates G, Leeds J, Park L, Howland D, Signer E, Tobin A, Brunner D. Systematic behavioral evaluation of Huntington’s disease transgenic and knock-in mouse models. Neurobiol Dis. 2009;35:319–336. doi: 10.1016/j.nbd.2009.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer U, Feldon J, Schedlowski M, Yee BK. Towards an immuno-precipitated neurodevelopmental animal model of schizophrenia. Neurosci Biobehav Rev. 2005;29:913–947. doi: 10.1016/j.neubiorev.2004.10.012. [DOI] [PubMed] [Google Scholar]
- Mirnics K, Middleton FA, Lewis DA, Levitt P. Analysis of complex brain disorders with gene expression microarrays: schizophrenia as a disease of the synapse. Trends Neurosci. 2001;24:479–486. doi: 10.1016/s0166-2236(00)01862-2. [DOI] [PubMed] [Google Scholar]
- Moghaddam B, Adams B, Verma A, Daly D. Activation of glutamatergic neurotransmission by ketamine: a novel step in the pathway from NMDA receptor blockade to dopaminergic and cognitive disruptions associated with the prefrontal cortex. J Neurosci. 1997;17:2921–2927. doi: 10.1523/JNEUROSCI.17-08-02921.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moore H. The role of rodent models in the discovery of new treatments for schizophrenia: updating our strategy. Schizophr Bull. 2010;36:1066–1072. doi: 10.1093/schbul/sbq106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moy SS, Nonneman RJ, Young NB, Demyanenko GP, Maness PF. Impaired sociability and cognitive function in Nrcam-null mice. Behav Brain Res. 2009;205:123–131. doi: 10.1016/j.bbr.2009.06.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakagawasai O, Onogi H, Mitazaki S, Sato A, Watanabe K, Saito H, Murai S, Nakaya K, Murakami M, Takahashi E, Tan-No K, Tadano T. Behavioral and neurochemical characterization of mice deficient in the N-type Ca2+ channel alpha1B subunit. Behav Brain Res. 2010;208:224–230. doi: 10.1016/j.bbr.2009.11.042. [DOI] [PubMed] [Google Scholar]
- Neuner I, Stocker T, Kellermann T, Ermer V, Wegener HP, Eickhoff SB, Schneider F, Shah NJ. Electrophysiology meets fMRI: neural correlates of the startle reflex assessed by simultaneous EMG-fMRI data acquisition. Hum Brain Mapp. 2010;31:1675–1685. doi: 10.1002/hbm.20965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nguyen PT, Nakamura T, Hori E, Urakawa S, Uwano T, Zhao J, Li R, Bac ND, Hamashima T, Ishii Y, Matsushima T, Ono T, Sasahara M, Nishijo H. Cognitive and socio-emotional deficits in platelet-derived growth factor receptor-beta gene knockout mice. PLoS One. 2011;6:e18004. doi: 10.1371/journal.pone.0018004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niwa M, Kamiya A, Murai R, Kubo K, Gruber AJ, Tomita K, Lu L, Tomisato S, Jaaro-Peled H, Seshadri S, Hiyama H, Huang B, Kohda K, Noda Y, O’Donnell P, Nakajima K, Sawa A, Nabeshima T. Knockdown of DISC1 by in utero gene transfer disturbs postnatal dopaminergic maturation in the frontal cortex and leads to adult behavioral deficits. Neuron. 2010;65:480–489. doi: 10.1016/j.neuron.2010.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nusbaum MP, Contreras D. Sensorimotor gating: startle submits to presynaptic inhibition. Curr Biol. 2004;14:R247–R249. doi: 10.1016/j.cub.2004.02.059. [DOI] [PubMed] [Google Scholar]
- O’Tuathaigh CM, Babovic D, O’Meara G, Clifford JJ, Croke DT, Waddington JL. Susceptibility genes for schizophrenia: characterisation of mutant mouse models at the level of phenotypic behaviour. Neurosci Biobehav Rev. 2007;31:60–78. doi: 10.1016/j.neubiorev.2006.04.002. [DOI] [PubMed] [Google Scholar]
- O’Tuathaigh CM, Desbonnet L, Moran PM, Kirby BP, Waddington JL. Molecular genetic models related to schizophrenia and psychotic illness: heuristics and challenges. Curr Top Behav Neurosci. 2011;7:87–119. doi: 10.1007/7854_2010_111. [DOI] [PubMed] [Google Scholar]
- O’Tuathaigh CM, Desbonnet L, Waddington JL. Neuregulin-1 signaling in schizophrenia: ‘Jack of all trades’ or master of some? Expert Rev Neurother. 2009;9:1–3. doi: 10.1586/14737175.9.1.1. [DOI] [PubMed] [Google Scholar]
- Ohgake S, Shimizu E, Hashimoto K, Okamura N, Koike K, Koizumi H, Fujisaki M, Kanahara N, Matsuda S, Sutoh C, Matsuzawa D, Muramatsu H, Muramatsu T, Iyo M. Dopaminergic hypofunctions and prepulse inhibition deficits in mice lacking midkine. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33:541–546. doi: 10.1016/j.pnpbp.2009.02.005. [DOI] [PubMed] [Google Scholar]
- Oliver PL, Davies KE. Interaction between environmental and genetic factors modulates schizophrenic endophenotypes in the Snap-25 mouse mutant blind-drunk. Hum Mol Genet. 2009;18:4576–4589. doi: 10.1093/hmg/ddp425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oliveros A, Heckman MG, Corena-McLeod DPM, Williams K, Boules M, Richelson E. Sensorimotor gating in NTS1 and NTS2 null mice: effects of d-amphetamine, dizocilpine, clozapine and NT69L. J Exp Biol. 2010;213:4232–4239. doi: 10.1242/jeb.046318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oranje B, Gispen-de Wied CC, Verbaten MN, Kahn RS. Modulating sensory gating in healthy volunteers: the effects of ketamine and haloperidol. Biol Psychiatry. 2002;52:887–895. doi: 10.1016/s0006-3223(02)01377-x. [DOI] [PubMed] [Google Scholar]
- Ortiz-Abalia J, Sahun I, Altafaj X, Andreu N, Estivill X, Dierssen M, Fillat C. Targeting Dyrk1A with AAVshRNA attenuates motor alterations in TgDyrk1A, a mouse model of down syndrome. Am J Hum Genet. 2008;83:479–488. doi: 10.1016/j.ajhg.2008.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Otte DM, Bilkei-Gorzo A, Filiou MD, Turck CW, Yilmaz O, Holst MI, Schilling K, Abou-Jamra R, Schumacher J, Benzel I, Kunz WS, Beck H, Zimmer A. Behavioral changes in G72/G30 transgenic mice. Eur Neuropsychopharmacol. 2009;19:339–348. doi: 10.1016/j.euroneuro.2008.12.009. [DOI] [PubMed] [Google Scholar]
- Papaleo F, Crawley JN, Song J, Lipska BK, Pickel J, Weinberger DR, Chen J. Genetic dissection of the role of catechol-O-methyltransferase in cognition and stress reactivity in mice. J Neurosci. 2008;28:8709–8723. doi: 10.1523/JNEUROSCI.2077-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paylor R, Yuva-Paylor LA, Nelson DL, Spencer CM. Reversal of sensorimotor gating abnormalities in Fmr1 knockout mice carrying a human Fmr1 transgene. Behav Neurosci. 2008;122:1371–1377. doi: 10.1037/a0013047. [DOI] [PubMed] [Google Scholar]
- Perry W, Braff DL. Information-processing deficits and thought disorder in schizophrenia. Am J Psychiatry. 1994;151:363–367. doi: 10.1176/ajp.151.3.363. [DOI] [PubMed] [Google Scholar]
- Perry W, Geyer MA, Braff DL. Sensorimotor gating and thought disturbance measured in close temporal proximity in schizophrenic patients. Arch Gen Psychiatry. 1999;56:277–281. doi: 10.1001/archpsyc.56.3.277. [DOI] [PubMed] [Google Scholar]
- Perry W, Minassian A, Feifel D, Braff DL. Sensorimotor gating deficits in bipolar disorder patients with acute psychotic mania. Biol Psychiatry. 2001;50:418–424. doi: 10.1016/s0006-3223(01)01184-2. [DOI] [PubMed] [Google Scholar]
- Perry W, Minassian A, Lopez B, Maron L, Lincoln A. Sensorimotor gating deficits in adults with autism. Biol Psychiatry. 2007;61:482–486. doi: 10.1016/j.biopsych.2005.09.025. [DOI] [PubMed] [Google Scholar]
- Petrovsky N, Quednow BB, Ettinger U, Schmechtig A, Mossner R, Collier DA, Kuhn KU, Maier W, Wagner M, Kumari V. Sensorimotor gating is associated with CHRNA3 polymorphisms in schizophrenia and healthy volunteers. Neuropsychopharmacology. 2010;35:1429–1439. doi: 10.1038/npp.2010.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pietropaolo S, Delage P, Cayzac S, Crusio WE, Cho YH. Sex-dependent changes in social behaviors in motor pre-symptomatic r6/1 mice. PLoS One. 2011;6:e19965. doi: 10.1371/journal.pone.0019965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Porteous DJ, Thomson P, Brandon NJ, Millar JK. The genetics and biology of Disc1—an emerging role in psychosis and cognition. Biol Psychiatry. 2006;60:123–131. doi: 10.1016/j.biopsych.2006.04.008. [DOI] [PubMed] [Google Scholar]
- Powell SB, Young JW, Ong JC, Caron MG, Geyer MA. Atypical antipsychotics clozapine and quetiapine attenuate prepulse inhibition deficits in dopamine transporter knockout mice. Behav Pharmacol. 2008;19:562–565. doi: 10.1097/FBP.0b013e32830dc110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Powell SB, Zhou X, Geyer MA. Prepulse inhibition and genetic mouse models of schizophrenia. Behav Brain Res. 2009;204:282–294. doi: 10.1016/j.bbr.2009.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quednow BB, Wagner M, Mossner R, Maier W, Kuhn K-U. Sensorimotor gating of schizophrenia patients depends on catechol O-Methyltransferase Val158Met polymorphism. Schizophr Bull: sbn088. 2008 doi: 10.1093/schbul/sbn088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Radyushkin K, Hammerschmidt K, Boretius S, Varoqueaux F, El-Kordi A, Ronnenberg A, Winter D, Frahm J, Fischer J, Brose N, Ehrenreich H. Neuroligin-3-deficient mice: model of a monogenic heritable form of autism with an olfactory deficit. Genes Brain Behav. 2009;8:416–425. doi: 10.1111/j.1601-183X.2009.00487.x. [DOI] [PubMed] [Google Scholar]
- Ralph RJ, Paulus MP, Fumagalli F, Caron MG, Geyer MA. Prepulse inhibition deficits and perseverative motor patterns in dopamine transporter knock-out mice: differential effects of D1 and D2 receptor antagonists. J Neurosci. 2001;21:305–313. doi: 10.1523/JNEUROSCI.21-01-00305.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ralph RJ, Varty GB, Kelly MA, Wang YM, Caron MG, Rubinstein M, Grandy DK, Low MJ, Geyer MA. The dopamine D2, but not D3 or D4, receptor subtype is essential for the disruption of prepulse inhibition produced by amphetamine in mice. J Neurosci. 1999;19:4627–4633. doi: 10.1523/JNEUROSCI.19-11-04627.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ransome MI, Turnley AM. Growth hormone signaling and hippocampal neurogenesis: insights from genetic models. Hippocampus. 2008;18:1034–1050. doi: 10.1002/hipo.20463. [DOI] [PubMed] [Google Scholar]
- Relkovic D, Doe CM, Humby T, Johnstone KA, Resnick JL, Holland AJ, Hagan JJ, Wilkinson LS, Isles AR. Behavioural and cognitive abnormalities in an imprinting centre deletion mouse model for Prader–Willi syndrome. Eur J Neurosci. 2010;31:156–164. doi: 10.1111/j.1460-9568.2009.07048.x. [DOI] [PubMed] [Google Scholar]
- Rose PK, Scott SH. Sensory-motor control: a long-awaited behavioral correlate of presynaptic inhibition. Nat Neurosci. 2003;6:1243–1245. doi: 10.1038/nn1203-1243. [DOI] [PubMed] [Google Scholar]
- Roussos P, Giakoumaki SG, Adamaki E, Bitsios P. The influence of schizophrenia-related neuregulin-1 polymorphisms on sensorimotor gating in healthy males. Biol Psychiatry. 2011;69:479–486. doi: 10.1016/j.biopsych.2010.09.009. [DOI] [PubMed] [Google Scholar]
- Roussos P, Giakoumaki SG, Rogdaki M, Pavlakis S, Frangou S, Bitsios P. Prepulse inhibition of the startle reflex depends on the catechol O-methyltransferase Val158Met gene polymorphism. Psychol Med. 2008:1–8. doi: 10.1017/S0033291708002912. [DOI] [PubMed] [Google Scholar]
- Sagata NA, Iwaki A, Aramaki T, Takao K, Kura S, Tsuzuki T, Kawakami R, Ito I, Kitamura T, Sugiyama H, Miyakawa T, Fukumaki Y. Comprehensive behavioral study of GluR4 knockout mice, implication in cognitive function. Genes Brain Behav. 2010 doi: 10.1111/j.1601-183X.2010.00629.x. [DOI] [PubMed] [Google Scholar]
- Sakatani S, Yamada K, Homma C, Munesue S, Yamamoto Y, Yamamoto H, Hirase H. Deletion of RAGE causes hyperactivity and increased sensitivity to auditory stimuli in mice. PLoS One. 2009;4:e8309. doi: 10.1371/journal.pone.0008309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samocha KE, Lim JE, Cheng R, Sokoloff G, Palmer AA. Fine mapping of QTL for prepulse inhibition in LG/J and SM/J mice using F2 and advanced intercross lines. Genes Brain Behav. 2010;9:759–767. doi: 10.1111/j.1601-183X.2010.00613.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Savonenko A, Munoz P, Melnikova T, Wang Q, Liang X, Breyer RM, Montine TJ, Kirkwood A, Andreasson K. Impaired cognition, sensorimotor gating, and hippocampal long-term depression in mice lacking the prostaglandin E2 EP2 receptor. Exp Neurol. 2009;217:63–73. doi: 10.1016/j.expneurol.2009.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Savonenko AV, Melnikova T, Laird FM, Stewart KA, Price DL, Wong PC. Alteration of BACE1-dependent NRG1/ErbB4 signaling and schizophrenia-like phenotypes in BACE1-null mice. Proc Natl Acad Sci U S A. 2008;105:5585–5590. doi: 10.1073/pnas.0710373105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scearce-Levie K, Roberson ED, Gerstein H, Cholfin JA, Mandiyan VS, Shah NM, Rubenstein JL, Mucke L. Abnormal social behaviors in mice lacking Fgf17. Genes Brain Behav. 2008;7:344–354. doi: 10.1111/j.1601-183X.2007.00357.x. [DOI] [PubMed] [Google Scholar]
- Schaefer TL, Vorhees CV, Williams MT. Mouse plasmacytoma-expressed transcript 1 knock out induced 5-HT disruption results in a lack of cognitive deficits and an anxiety phenotype complicated by hypoactivity and defensiveness. Neuroscience. 2009;164:1431–1443. doi: 10.1016/j.neuroscience.2009.09.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmid S, Azzopardi E, De Jaeger X, Prado MA, Prado VF. VAChT knock-down mice show normal prepulse inhibition but disrupted long-term habituation. Genes Brain Behav. 2011 doi: 10.1111/j.1601-183X.2011.00686.x. [DOI] [PubMed] [Google Scholar]
- Schmidt S, Richter M, Montag D, Sartorius T, Gawlik V, Hennige AM, Scherneck S, Himmelbauer H, Lutz SZ, Augustin R, Kluge R, Ruth P, Joost HG, Schürmann A. Neuronal functions, feeding behavior, and energy balance in Slc2a3+/− mice. Am J Physiol Endocrinol Metab. 2008;295(5):E1084–E1094. doi: 10.1152/ajpendo.90491.2008. [DOI] [PubMed] [Google Scholar]
- Schneider M, Spanagel R, Zhang SJ, Bading H, Klugmann M. Adeno-associated virus (AAV)-mediated suppression of Ca2+/calmodulin kinase IV activity in the nucleus accumbens modulates emotional behaviour in mice. BMC Neurosci. 2007;8:105. doi: 10.1186/1471-2202-8-105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schwabe K, Freudenberg F, Koch M. Selective breeding of reduced sensorimotor gating in wistar rats. Behav Genet. 2007;37:706–712. doi: 10.1007/s10519-007-9166-z. [DOI] [PubMed] [Google Scholar]
- Semenova S, Geyer MA, Sutcliffe JG, Markou A, Hedlund PB. Inactivation of the 5-HT(7) receptor partially blocks phencyclidine-induced disruption of prepulse inhibition. Biol Psychiatry. 2008;63:98–105. doi: 10.1016/j.biopsych.2006.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shanahan NA, Holick Pierz KA, Masten VL, Waeber C, Ansorge M, Gingrich JA, Geyer MA, Hen R, Dulawa SC. Chronic reductions in serotonin transporter function prevent 5-HT1B-induced behavioral effects in mice. Biol Psychiatry. 2009;65:401–408. doi: 10.1016/j.biopsych.2008.09.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi L, Fatemi SH, Sidwell RW, Patterson PH. Maternal influenza infection causes marked behavioral and pharmacological changes in the offspring. J Neurosci. 2003;23:297–302. doi: 10.1523/JNEUROSCI.23-01-00297.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sipes TA, Geyer MA. Multiple serotonin receptor subtypes modulate prepulse inhibition of the startle response in rats. Neuropharmacology. 1994;33:441–448. doi: 10.1016/0028-3908(94)90074-4. [DOI] [PubMed] [Google Scholar]
- Siuciak JA, McCarthy SA, Chapin DS, Martin AN. Behavioral and neurochemical characterization of mice deficient in the phosphodiesterase-4B (PDE4B) enzyme. Psychopharmacology (Berl) 2008;197:115–126. doi: 10.1007/s00213-007-1014-6. [DOI] [PubMed] [Google Scholar]
- Siuta MA, Robertson SD, Kocalis H, Saunders C, Gresch PJ, Khatri V, Shiota C, Kennedy JP, Lindsley CW, Daws LC, Polley DB, Veenstra-Vanderweele J, Stanwood GD, Magnuson MA, Niswender KD, Galli A. Dysregulation of the norepinephrine transporter sustains cortical hypodopaminergia and schizophrenia-like behaviors in neuronal rictor null mice. PLoS Biol. 2010;8 doi: 10.1371/journal.pbio.1000393. e1000393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith CM, Lawrence AJ, Sutton SW, Gundlach AL. Behavioral phenotyping of mixed background (129S5:B6) relaxin-3 knockout mice. Ann N Y Acad Sci. 2009;1160:236–241. doi: 10.1111/j.1749-6632.2009.03953.x. [DOI] [PubMed] [Google Scholar]
- Smith SE, Li J, Garbett K, Mirnics K, Patterson PH. Maternal immune activation alters fetal brain development through interleukin-6. J Neurosci. 2007;27:10695–10702. doi: 10.1523/JNEUROSCI.2178-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soleimani L, Roder JC, Dennis JW, Lipina T. Beta N-acetylglucosaminyltransferase V (Mgat5) deficiency reduces the depression-like phenotype in mice. Genes Brain Behav. 2008;7:334–343. doi: 10.1111/j.1601-183X.2007.00358.x. [DOI] [PubMed] [Google Scholar]
- Stark KL, Burt RA, Gogos JA, Karayiorgou M. Analysis of prepulse inhibition in mouse lines overexpressing 22q11.2 orthologues. Int J Neuropsychopharmacol. 2009:1–7. doi: 10.1017/S1461145709000492. [DOI] [PubMed] [Google Scholar]
- Sun M, Gewirtz JC, Bofenkamp L, Wickham RJ, Ge H, O’Connor MB. Canonical TGF-beta signaling is required for the balance of excitatory/inhibitory transmission within the hippocampus and prepulse inhibition of acoustic startle. J Neurosci. 2010;30:6025–6035. doi: 10.1523/JNEUROSCI.0789-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suzuki G, Harper KM, Hiramoto T, Sawamura T, Lee M, Kang G, Tanigaki K, Buell M, Geyer MA, Trimble WS, Agatsuma S, Hiroi N. Sept5 deficiency exerts pleiotropic influence on affective behaviors and cognitive functions in mice. Hum Mol Genet. 2009a;18:1652–1660. doi: 10.1093/hmg/ddp086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suzuki G, Harper KM, Hiramoto T, Funke B, Lee M, Kang G, Buell M, Geyer MA, Kucherlapati R, Morrow B, Mannisto PT, Agatsuma S, Hiroi N. Over-expression of a human chromosome 22q11.2 segment including TXNRD2, COMT and ARVCF developmentally affects incentive learning and working memory in mice. Hum Mol Genet. 2009b;18:3914–3925. doi: 10.1093/hmg/ddp334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swerdlow NR, Benbow CH, Zisook S, Geyer MA, Braff DL. A preliminary assessment of sensorimotor gating in patients with obsessive compulsive disorder. Biol Psychiatry. 1993;33:298–301. doi: 10.1016/0006-3223(93)90300-3. [DOI] [PubMed] [Google Scholar]
- Swerdlow NR, Geyer MA, Braff DL. Neural circuit regulation of prepulse inhibition of startle in the rat: current knowledge and future challenges. Psychopharmacology (Berl) 2001a;156:194–215. doi: 10.1007/s002130100799. [DOI] [PubMed] [Google Scholar]
- Swerdlow NR, Karban B, Ploum Y, Sharp R, Geyer MA, Eastvold A. Tactile prepuff inhibition of startle in children with Tourette’s syndrome: in search of an “fMRI-friendly” startle paradigm. Biol Psychiatry. 2001b;50:578–585. doi: 10.1016/s0006-3223(01)01164-7. [DOI] [PubMed] [Google Scholar]
- Swerdlow NR, Light GA, Cadenhead KS, Sprock J, Hsieh MH, Braff DL. Startle gating deficits in a large cohort of patients with schizophrenia: relationship to medications, symptoms, neurocognition, and level of function. Arch Gen Psychiatry. 2006;63:1325–1335. doi: 10.1001/archpsyc.63.12.1325. [DOI] [PubMed] [Google Scholar]
- Swerdlow NR, Paulsen J, Braff DL, Butters N, Geyer MA, Swenson MR. Impaired prepulse inhibition of acoustic and tactile startle response in patients with Huntington’s disease. J Neurol Neurosurg Psychiatry. 1995;58:192–200. doi: 10.1136/jnnp.58.2.192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swerdlow NR, Weber M, Qu Y, Light GA, Braff DL. Realistic expectations of prepulse inhibition in translational models for schizophrenia research. Psychopharmacology (Berl) 2008;199:331–388. doi: 10.1007/s00213-008-1072-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takao K, Tanda K, Nakamura K, Kasahara J, Nakao K, Katsuki M, Nakanishi K, Yamasaki N, Toyama K, Adachi M, Umeda M, Araki T, Fukunaga K, Kondo H, Sakagami H, Miyakawa T. Comprehensive behavioral analysis of calcium/calmodulin-dependent protein kinase IV knockout mice. PLoS One. 2010;5:e9460. doi: 10.1371/journal.pone.0009460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takata A, Kakiuchi C, Ishiwata M, Kanba S, Kato T. Behavioral and gene expression analyses in heterozygous XBP1 knockout mice: possible contribution of chromosome 11qA1 locus to prepulse inhibition. Neurosci Res. 2010 doi: 10.1016/j.neures.2010.07.2042. [DOI] [PubMed] [Google Scholar]
- Takeuchi H, Iba M, Inoue H, Higuchi M, Takao K, Tsukita K, Karatsu Y, Iwamoto Y, Miyakawa T, Suhara T, Trojanowski JQ, Lee VM, Takahashi R. P301S mutant human tau transgenic mice manifest early symptoms of human tauopathies with dementia and altered sensorimotor gating. PLoS One. 2011;6:e21050. doi: 10.1371/journal.pone.0021050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tammimäki A, Kaenmaki M, Kambur O, Kulesskaya N, Keisala T, Karvonen E, Garcia-Horsman JA, Rauvala H, Mannisto PT. Effect of S-COMT deficiency on behavior and extracellular brain dopamine concentrations in mice. Psychopharmacology (Berl) 2010;211:389–401. doi: 10.1007/s00213-010-1944-2. [DOI] [PubMed] [Google Scholar]
- Tanaka H, Ma J, Tanaka KF, Takao K, Komada M, Tanda K, Suzuki A, Ishibashi T, Baba H, Isa T, Shigemoto R, Ono K, Miyakawa T, Ikenaka K. Mice with altered myelin proteolipid protein gene expression display cognitive deficits accompanied by abnormal neuron-glia interactions and decreased conduction velocities. J Neurosci. 2009;29:8363–8371. doi: 10.1523/JNEUROSCI.3216-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanda K, Nishi A, Matsuo N, Nakanishi K, Yamasaki N, Sugimoto T, Toyama K, Takao K, Miyakawa T. Abnormal social behavior, hyperactivity, impaired remote spatial memory, and increased D1-mediated dopaminergic signaling in neuronal nitric oxide synthase knockout mice. Mol Brain. 2009;2:19. doi: 10.1186/1756-6606-2-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tarantino M, Eisener-Dorman AF. Forward genetic approachesto understanding complex behaviors. 2011 doi: 10.1007/7854_2011_189. (in this volume) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tarantino LM, Bucan M. Dissection of behavior and psychiatric disorders using the mouse as a model. Hum Mol Genet. 2000;9:953–965. doi: 10.1093/hmg/9.6.953. [DOI] [PubMed] [Google Scholar]
- Tecott LH, Wehner JM. Mouse molecular genetic technologies: promise for psychiatric research. Arch Gen Psychiatry. 2001;58:995–1004. doi: 10.1001/archpsyc.58.11.995. [DOI] [PubMed] [Google Scholar]
- Thaker GK. Schizophrenia endophenotypes as treatment targets. Expert Opin Ther Targets. 2007;11:1189–1206. doi: 10.1517/14728222.11.9.1189. [DOI] [PubMed] [Google Scholar]
- Thomas AM, Bui N, Graham D, Perkins JR, Yuva-Paylor LA, Paylor R. Genetic reduction of group 1 metabotropic glutamate receptors alters select behaviors in a mouse model for fragile X syndrome. Behav Brain Res. 2011a;223:310–321. doi: 10.1016/j.bbr.2011.04.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thomas AM, Bui N, Perkins JR, Yuva-Paylor LA, Paylor R. Group I metabotropic glutamate receptor antagonists alter select behaviors in a mouse model for fragile X syndrome. Psychopharmacology (Berl) 2011b doi: 10.1007/s00213-011-2375-4. [DOI] [PubMed] [Google Scholar]
- Thomsen M, Wess J, Fulton BS, Fink-Jensen A, Caine SB. Modulation of prepulse inhibition through both M(1) and M (4) muscarinic receptors in mice. Psychopharmacology (Berl) 2010;208:401–416. doi: 10.1007/s00213-009-1740-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Torkamanzehi A, Boksa P, Joober R. Prepulse inhibition (PPI) of tactile startle response in recombinant congenic strains of mice: QTL mapping and comparison with acoustic PPI. J Genet Genomics. 2008;35:139–151. doi: 10.1016/S1673-8527(08)60020-X. [DOI] [PubMed] [Google Scholar]
- Tsujimura A, Matsuki M, Takao K, Yamanishi K, Miyakawa T, Hashimoto-Gotoh T. Mice lacking the kf-1 gene exhibit increased anxiety-but not despair-like behavior. Front Behav Neurosci. 2008;2:4. doi: 10.3389/neuro.08.004.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turetsky BI, Calkins ME, Light GA, Olincy A, Radant AD, Swerdlow NR. Neurophysiological endophenotypes of schizophrenia: the viability of selected candidate measures. Schizophr Bull. 2007;33:69–94. doi: 10.1093/schbul/sbl060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turner J, Hughes LF, Toth LA. Sleep, activity, temperature and arousal responses of mice deficient for muscarinic receptor M2 or M4. Life Sci. 2010;86:158–169. doi: 10.1016/j.lfs.2009.11.019. [DOI] [PubMed] [Google Scholar]
- van Berckel BN, Oranje B, van Ree JM, Verbaten MN, Kahn RS. The effects of low dose ketamine on sensory gating, neuroendocrine secretion and behavior in healthy human subjects. Psychopharmacology (Berl) 1998;137:271–281. doi: 10.1007/s002130050620. [DOI] [PubMed] [Google Scholar]
- van den Buuse M, Becker T, Kwek P, Martin S, Ruimschotel E, Risbrough V. Disruption of prepulse inhibition by 3,4-methylenedioxymethamphetamine (MDMA): comparison between male and female wild-type and 5-HT1A receptor knockout mice. Int J Neuropsychopharmacol. 2011a:1–6. doi: 10.1017/S1461145711000101. [DOI] [PubMed] [Google Scholar]
- van den Buuse M, Ruimschotel E, Martin S, Risbrough VB, Halberstadt AL. Enhanced effects of amphetamine but reduced effects of the hallucinogen-5-MeO-DMT, on locomotor activity in 5-HT(1A) receptor knockout mice: implications for schizophrenia. Neuropharmacology. 2011b doi: 10.1016/j.neuropharm.2011.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van der Neut R. Targeted gene disruption: applications in neurobiology. J Neurosci Methods. 1997;71:19–27. doi: 10.1016/s0165-0270(96)00123-9. [DOI] [PubMed] [Google Scholar]
- Veeraragavan S, Bui N, Perkins JR, Yuva-Paylor LA, Carpenter RL, Paylor R. Modulation of behavioral phenotypes by a muscarinic M1 antagonist in a mouse model of fragile X syndrome. Psychopharmacology (Berl) 2011 doi: 10.1007/s00213-011-2276-6. [DOI] [PubMed] [Google Scholar]
- Verma V, Tan CH, Ong WY, Grigoryan GA, Jones CA, Stolzberg D, Salvi R, Gross KW, Ratty AK, Dawe GS. The chakragati mouse shows deficits in prepulse inhibition of acoustic startle and latent inhibition. Neurosci Res. 2008;60:281–288. doi: 10.1016/j.neures.2007.11.007. [DOI] [PubMed] [Google Scholar]
- Wallén-Mackenzie A, Nordenankar K, Fejgin K, Lagerstrom MC, Emilsson L, Fredriksson R, Wass C, Andersson D, Egecioglu E, Andersson M, Strandberg J, Lindhe O, Schioth HB, Chergui K, Hanse E, Langstrom B, Fredriksson A, Svensson L, Roman E, Kullander K. Restricted cortical and amygdaloid removal of vesicular glutamate transporter 2 in preadolescent mice impacts dopaminergic activity and neuronal circuitry of higher brain function. J Neurosci. 2009;29:2238–2251. doi: 10.1523/JNEUROSCI.5851-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H, Westin L, Nong Y, Birnbaum S, Bendor J, Brismar H, Nestler E, Aperia A, Flajolet M, Greengard P. Norbin is an endogenous regulator of metabotropic glutamate receptor 5 signaling. Science. 2009;326:1554–1557. doi: 10.1126/science.1178496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang X. Cre transgenic mouse lines. Methods Mol Biol. 2009;561:265–273. doi: 10.1007/978-1-60327-019-9_17. [DOI] [PubMed] [Google Scholar]
- Watanabe A, Toyota T, Owada Y, Hayashi T, Iwayama Y, Matsumata M, Ishitsuka Y, Nakaya A, Maekawa M, Ohnishi T, Arai R, Sakurai K, Yamada K, Kondo H, Hashimoto K, Osumi N, Yoshikawa T. Fabp7 maps to a quantitative trait locus for a schizophrenia endophenotype. PLoS Biol. 2007;5:e297. doi: 10.1371/journal.pbio.0050297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weinberger DR, Berman KF, Suddath R, Torrey EF. Evidence of dysfunction of a prefrontal-limbic network in schizophrenia: a magnetic resonance imaging and regional cerebral blood flow study of discordant monozygotic twins. Am J Psychiatry. 1992;149:890–897. doi: 10.1176/ajp.149.7.890. [DOI] [PubMed] [Google Scholar]
- Wen L, Lu YS, Zhu XH, Li XM, Woo RS, Chen YJ, Yin DM, Lai C, Terry AV, Jr, Vazdarjanova A, Xiong WC, Mei L. Neuregulin 1 regulates pyramidal neuron activity via ErbB4 in parvalbumin-positive interneurons. Proc Natl Acad Sci U S A. 2010;107:1211–1216. doi: 10.1073/pnas.0910302107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Willi R, Weinmann O, Winter C, Klein J, Sohr R, Schnell L, Yee BK, Feldon J, Schwab ME. Constitutive genetic deletion of the growth regulator Nogo-A induces schizophrenia-related endophenotypes. J Neurosci. 2010;30:556–567. doi: 10.1523/JNEUROSCI.4393-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Willott JF, Carlson S, Chen H. Prepulse inhibition of the startle response in mice: relationship to hearing loss and auditory system plasticity. Behav Neurosci. 1994;108:6. doi: 10.1037//0735-7044.108.4.703. [DOI] [PubMed] [Google Scholar]
- Yavich L, Forsberg MM, Karayiorgou M, Gogos JA, Mannisto PT. Site-specific role of catechol-O-methyltransferase in dopamine overflow within prefrontal cortex and dorsal striatum. J Neurosci. 2007;27:10196–10209. doi: 10.1523/JNEUROSCI.0665-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young JW, Meves JM, Tarantino IS, Caldwell S, Geyer MA. Delayed procedural learning in alpha7-nicotinic acetylcholine receptor knockout mice. Genes Brain Behav. 2011a doi: 10.1111/j.1601-183X.2011.00711.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young JW, Powell SB, Scott CN, Zhou X, Geyer MA. The effect of reduced dopamine D4 receptor expression in the 5-choice continuous performance task: Separating response inhibition from premature responding. Behav Brain Res. 2011b;222:183–192. doi: 10.1016/j.bbr.2011.03.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young JW, Wallace CK, Geyer MA, Risbrough VB. Age-associated improvements in cross-modal prepulse inhibition in mice. Behav Neurosci. 2010;124:133–140. doi: 10.1037/a0018462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou X, Geyer MA, Kelsoe JR. Does disrupted-in-schizophrenia (DISC1) generate fusion transcripts? Mol Psychiatry. 2008;13:361–363. doi: 10.1038/sj.mp.4002125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou X, Long JM, Geyer MA, Masliah E, Kelsoe JR, Wynshaw-Boris A, Chien KR. Reduced expression of the Sp4 gene in mice causes deficits in sensorimotor gating and memory associated with hippocampal vacuolization. Mol Psychiatry. 2004;10:393–406. doi: 10.1038/sj.mp.4001621. [DOI] [PubMed] [Google Scholar]
- Zhou X, Tang W, Greenwood TA, Guo S, He3 L, Geyer MA, Kelsoe JR. Transcription factor SP4 is a susceptibility gene for bipolar disorder. PlosOne. 2009 doi: 10.1371/journal.pone.0005196. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu H, Mingler MK, McBride ML, Murphy AJ, Valenzuela DM, Yancopoulos GD, Williams MT, Vorhees CV, Rothenberg ME. Abnormal response to stress and impaired NPS-induced hyperlocomotion, anxiolytic effect and corticosterone increase in mice lacking NPSR1. Psychoneuroendocrinology. 2010;35:1119–1132. doi: 10.1016/j.psyneuen.2010.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]