1
|
Ursini G, Di Carlo P, Mukherjee S, Chen Q, Han S, Kim J, Deyssenroth M, Marsit CJ, Chen J, Hao K, Punzi G, Weinberger DR. Prioritization of potential causative genes for schizophrenia in placenta. Nat Commun 2023; 14:2613. [PMID: 37188697 PMCID: PMC10185564 DOI: 10.1038/s41467-023-38140-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Our earlier work has shown that genomic risk for schizophrenia converges with early life complications in affecting risk for the disorder and sex-biased neurodevelopmental trajectories. Here, we identify specific genes and potential mechanisms that, in placenta, may mediate such outcomes. We performed TWAS in healthy term placentae (N = 147) to derive candidate placental causal genes that we confirmed with SMR; to search for placenta and schizophrenia-specific associations, we performed an analogous analysis in fetal brain (N = 166) and additional placenta TWAS for other disorders/traits. The analyses in the whole sample and stratifying by sex ultimately highlight 139 placenta and schizophrenia-specific risk genes, many being sex-biased; the candidate molecular mechanisms converge on the nutrient-sensing capabilities of placenta and trophoblast invasiveness. These genes also implicate the Coronavirus-pathogenesis pathway and showed increased expression in placentae from a small sample of SARS-CoV-2-positive pregnancies. Investigating placental risk genes for schizophrenia and candidate mechanisms may lead to opportunities for prevention that would not be suggested by study of the brain alone.
Collapse
Affiliation(s)
- Gianluca Ursini
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Pasquale Di Carlo
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
- Group of Psychiatric Neuroscience, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Sreya Mukherjee
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Qiang Chen
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Shizhong Han
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Jiyoung Kim
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Maya Deyssenroth
- Departments of Environmental Medicine and Public Health, Icahn School of Public Health at Mount Sinai, New York, NY, USA
| | - Carmen J Marsit
- Departments of Environmental Health and Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Jia Chen
- Departments of Environmental Medicine and Public Health, Icahn School of Public Health at Mount Sinai, New York, NY, USA
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giovanna Punzi
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Abstract
Most psychiatric illnesses, such as schizophrenia, show profound sex differences in incidence, clinical presentation, course, and outcome. Fortunately, more recently the literature on sex differences and (to a lesser extent) effects of sex steroid hormones is expanding, and in this review we have focused on such studies in psychosis, both from a clinical/epidemiological and preclinical/animal model perspective. We begin by briefly describing the clinical evidence for sex differences in schizophrenia epidemiology, symptomatology, and pathophysiology. We then detail sex differences and sex hormone effects in behavioral animal models of psychosis, specifically psychotropic drug-induced locomotor hyperactivity and disruption of prepulse inhibition. We expand on the preclinical data to include developmental and genetic models of psychosis, such as the maternal immune activation model and neuregulin transgenic animals, respectively. Finally, we suggest several recommendations for future studies, in order to facilitate a better understanding of sex differences in the development of psychosis.
Collapse
|
3
|
Zhang Z, Ye M, Li Q, You Y, Yu H, Ma Y, Mei L, Sun X, Wang L, Yue W, Li R, Li J, Zhang D. The Schizophrenia Susceptibility Gene OPCML Regulates Spine Maturation and Cognitive Behaviors through Eph-Cofilin Signaling. Cell Rep 2020; 29:49-61.e7. [PMID: 31577955 DOI: 10.1016/j.celrep.2019.08.091] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 07/09/2019] [Accepted: 08/27/2019] [Indexed: 12/30/2022] Open
Abstract
Previous genetic and biological evidence converge on the involvement of synaptic dysfunction in schizophrenia, and OPCML, encoding a synaptic membrane protein, is reported to be genetically associated with schizophrenia. However, its role in the pathophysiology of schizophrenia remains largely unknown. Here, we found that Opcml is strongly expressed in the mouse hippocampus; ablation of Opcml leads to reduced phosphorylated cofilin and dysregulated F-actin dynamics, which disturbs the spine maturation. Furthermore, Opcml interacts with EphB2 to control the stability of spines by regulating the ephrin-EphB2-cofilin signaling pathway. Opcml-deficient mice display impaired cognitive behaviors and abnormal sensorimotor gating, which are similar to features in neuropsychiatric disorders such as schizophrenia. Notably, the administration of aripiprazole partially restores the abnormal behaviors in Opcml-/- mice by increasing the phosphorylated cofilin level and facilitating spine maturation. We demonstrated a critical role of the schizophrenia-susceptible gene OPCML in spine maturation and cognitive behaviors via regulating the ephrin-EphB2-cofilin signaling pathway, providing further insights into the characteristics of schizophrenia.
Collapse
Affiliation(s)
- Zhengrong Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China; National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Maoqing Ye
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Cardiology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Qiongwei Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Yang You
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Hao Yu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Yuanlin Ma
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Liwei Mei
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Xiaqin Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Lifang Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Weihua Yue
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Rena Li
- National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Jun Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China.
| | - Dai Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| |
Collapse
|
4
|
Balmer LA, Whiting R, Rudnicka C, Gallo LA, Jandeleit KA, Chow Y, Chow Z, Richardson KL, Forbes JM, Morahan G. Genetic characterization of early renal changes in a novel mouse model of diabetic kidney disease. Kidney Int 2019; 96:918-926. [PMID: 31420193 DOI: 10.1016/j.kint.2019.04.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/09/2019] [Accepted: 04/22/2019] [Indexed: 01/13/2023]
Abstract
Genetic factors influence susceptibility to diabetic kidney disease. Here we mapped genes mediating renal hypertrophic changes in response to diabetes. A survey of 15 mouse strains identified variation in diabetic kidney hypertrophy. Strains with greater (FVB/N(FVB)) and lesser (C57BL/6 (B6)) responses were crossed and diabetic F2 progeny were characterized. Kidney weights of diabetic F2 mice were broadly distributed. Quantitative trait locus analyses revealed diabetic mice with kidney weights in the upper quartile shared alleles on chromosomes (chr) 6 and 12; these loci were designated as Diabetic kidney hypertrophy (Dkh)-1 and -2. To confirm these loci, reciprocal congenic mice were generated with defined FVB chromosome segments on the B6 strain background (B6.Dkh1/2f) or vice versa (FVB.Dkh1/2b). Diabetic mice of the B6.Dkh1/2f congenic strain developed diabetic kidney hypertrophy, while the reciprocal FVB.Dkh1/2b congenic strain was protected. The chr6 locus contained the candidate gene; Ark1b3, coding aldose reductase; the FVB allele has a missense mutation in this gene. Microarray analysis identified differentially expressed genes between diabetic B6 and FVB mice. Thus, since the two loci identified by quantitative trait locus mapping are syntenic with regions identified for human diabetic kidney disease, the congenic strains we describe provide a valuable new resource to study diabetic kidney disease and test agents that may prevent it.
Collapse
Affiliation(s)
- Lois A Balmer
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, the University of Western Australia, Perth, Western Australia, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup, Perth, Western Australia, Australia
| | - Rhiannon Whiting
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, the University of Western Australia, Perth, Western Australia, Australia
| | - Caroline Rudnicka
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, the University of Western Australia, Perth, Western Australia, Australia
| | - Linda A Gallo
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia; School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | | | - Yan Chow
- Glenferrie Private Hospital, Ramsay Health Care, Donvale, Victoria, Australia
| | - Zenia Chow
- ENT Doctors, Northpark Private Hospital, Bundoora, Victoria, Australia
| | - Kirsty L Richardson
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Western Australia, Australia
| | - Josephine M Forbes
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia; Mater Clinical School, University of Queensland, Brisbane, Queensland, Australia
| | - Grant Morahan
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, the University of Western Australia, Perth, Western Australia, Australia.
| |
Collapse
|
5
|
Hirjak D, Kubera KM, Thomann PA, Wolf RC. Motor dysfunction as an intermediate phenotype across schizophrenia and other psychotic disorders: Progress and perspectives. Schizophr Res 2018; 200:26-34. [PMID: 29074330 DOI: 10.1016/j.schres.2017.10.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 10/02/2017] [Accepted: 10/06/2017] [Indexed: 02/07/2023]
Abstract
Primary motor abnormalities (PMA), as found in patients with schizophrenia, are quantitatively and qualitatively distinct markers of motor system abnormalities. PMA have been often referred to phenomena that are present across schizophrenia-spectrum disorders. A dysfunction of frontoparietal and subcortical networks has been proposed as core pathophysiological mechanism underlying the expression of PMA. However, it is unclear at present if such mechanisms are a common within schizophrenia and other psychotic disorders. To address this question, we review recent neuroimaging studies investigating the neural substrates of PMA in schizophrenia and so-called "nonschizophrenic nonaffective psychoses" (NSNAP) such as schizophreniform, schizoaffective, brief psychotic, and other unspecified psychotic disorders. Although the extant data in patients with schizophrenia suggests that further investigation is warranted, MRI findings in NSNAP are less persuasive. It is unclear so far which PMA, if any, are characteristic features of NSNAP or, possibly even specific for these disorders. Preliminary data suggest a relationship between relapsing-remitting PMA in hyper-/hypokinetic cycloid syndromes and neurodegenerative disorders of the basal ganglia, likely reflecting the transnosological relevance of subcortical abnormalities. Despite this evidence, neural substrates and mechanisms underlying PMA that are common in schizophrenia and NSNAP cannot be clearly delineated at this stage of research. PMA and their underlying brain circuits could be promising intermediate phenotype candidates for psychotic disorders, but future multimodal neuroimaging studies in schizophrenia and NSNAP patients and their unaffected first-degree relatives are needed to answer fundamental transnosologic questions.
Collapse
Affiliation(s)
- Dusan Hirjak
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University Mannheim, Germany.
| | - Katharina M Kubera
- Center for Psychosocial Medicine, Department of General Psychiatry, University of Heidelberg, Germany
| | - Philipp A Thomann
- Center for Psychosocial Medicine, Department of General Psychiatry, University of Heidelberg, Germany; Center for Mental Health, Odenwald District Healthcare Center, Erbach, Germany
| | - Robert C Wolf
- Center for Psychosocial Medicine, Department of General Psychiatry, University of Heidelberg, Germany
| |
Collapse
|
6
|
Electrophysiological alterations in a complex rat model of schizophrenia. Behav Brain Res 2016; 307:65-72. [DOI: 10.1016/j.bbr.2016.03.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/25/2016] [Accepted: 03/29/2016] [Indexed: 12/17/2022]
|
7
|
O'Tuathaigh CMP, Desbonnet L, Moran PM, Kirby BP, Waddington JL. Molecular genetic models related to schizophrenia and psychotic illness: heuristics and challenges. Curr Top Behav Neurosci 2016; 7:87-119. [PMID: 21298380 DOI: 10.1007/7854_2010_111] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Schizophrenia is a heritable disorder that may involve several common genes of small effect and/or rare copy number variation, with phenotypic heterogeneity across patients. Furthermore, any boundaries vis-à-vis other psychotic disorders are far from clear. Consequently, identification of informative animal models for this disorder, which typically relate to pharmacological and putative pathophysiological processes of uncertain validity, faces considerable challenges. In juxtaposition, the majority of mutant models for schizophrenia relate to the functional roles of a diverse set of genes associated with risk for the disorder or with such putative pathophysiological processes. This chapter seeks to outline the evidence from phenotypic studies in mutant models related to schizophrenia. These have commonly assessed the degree to which mutation of a schizophrenia-related gene is associated with the expression of several aspects of the schizophrenia phenotype or more circumscribed, schizophrenia-related endophenotypes; typically, they place specific emphasis on positive and negative symptoms and cognitive deficits, and extend to structural and other pathological features. We first consider the primary technological approaches to the generation of such mutants, to include their relative merits and demerits, and then highlight the diverse phenotypic approaches that have been developed for their assessment. The chapter then considers the application of mutant phenotypes to study pathobiological and pharmacological mechanisms thought to be relevant for schizophrenia, particularly in terms of dopaminergic and glutamatergic dysfunction, and to an increasing range of candidate susceptibility genes and copy number variants. Finally, we discuss several pertinent issues and challenges within the field which relate to both phenotypic evaluation and a growing appreciation of the functional genomics of schizophrenia and the involvement of gene × environment interactions.
Collapse
Affiliation(s)
- Colm M P O'Tuathaigh
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland,
| | | | | | | | | |
Collapse
|
8
|
Ayhan Y, McFarland R, Pletnikov MV. Animal models of gene-environment interaction in schizophrenia: A dimensional perspective. Prog Neurobiol 2015; 136:1-27. [PMID: 26510407 DOI: 10.1016/j.pneurobio.2015.10.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 09/07/2015] [Accepted: 10/22/2015] [Indexed: 12/12/2022]
Abstract
Schizophrenia has long been considered as a disorder with multifactorial origins. Recent discoveries have advanced our understanding of the genetic architecture of the disease. However, even with the increase of identified risk variants, heritability estimates suggest an important contribution of non-genetic factors. Various environmental risk factors have been proposed to play a role in the etiopathogenesis of schizophrenia. These include season of birth, maternal infections, obstetric complications, adverse events at early childhood, and drug abuse. Despite the progress in identification of genetic and environmental risk factors, we still have a limited understanding of the mechanisms whereby gene-environment interactions (G × E) operate in schizophrenia and psychoses at large. In this review we provide a critical analysis of current animal models of G × E relevant to psychotic disorders and propose that dimensional perspective will advance our understanding of the complex mechanisms of these disorders.
Collapse
Affiliation(s)
- Yavuz Ayhan
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA; Hacettepe University Faculty of Medicine, Department of Psychiatry, Turkey
| | - Ross McFarland
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, USA
| | - Mikhail V Pletnikov
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA; Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, USA; Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, USA.
| |
Collapse
|
9
|
Molecular underpinnings of prefrontal cortex development in rodents provide insights into the etiology of neurodevelopmental disorders. Mol Psychiatry 2015; 20:795-809. [PMID: 25450230 PMCID: PMC4486649 DOI: 10.1038/mp.2014.147] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/12/2014] [Accepted: 09/17/2014] [Indexed: 12/20/2022]
Abstract
The prefrontal cortex (PFC), seat of the highest-order cognitive functions, constitutes a conglomerate of highly specialized brain areas and has been implicated to have a role in the onset and installation of various neurodevelopmental disorders. The development of a properly functioning PFC is directed by transcription factors, guidance cues and other regulatory molecules and requires the intricate and temporal orchestration of a number of developmental processes. Disturbance or failure of any of these processes causing neurodevelopmental abnormalities within the PFC may contribute to several of the cognitive deficits seen in patients with neurodevelopmental disorders. In this review, we elaborate on the specific processes underlying prefrontal development, such as induction and patterning of the prefrontal area, proliferation, migration and axonal guidance of medial prefrontal progenitors, and their eventual efferent and afferent connections. We furthermore integrate for the first time the available knowledge from genome-wide studies that have revealed genes linked to neurodevelopmental disorders with experimental molecular evidence in rodents. The integrated data suggest that the pathogenic variants in the neurodevelopmental disorder-associated genes induce prefrontal cytoarchitectonical impairments. This enhances our understanding of the molecular mechanisms of prefrontal (mis)development underlying the four major neurodevelopmental disorders in humans, that is, intellectual disability, autism spectrum disorders, attention deficit hyperactivity disorder and schizophrenia, and may thus provide clues for the development of novel therapies.
Collapse
|
10
|
Zhu M, Ming Y, Swaim H, Swain MD, Myers MJ, Deaver C, Wu X, Jones YL, Yancy HF. Identification of potential biomarkers of P-glycoprotein substrate neurotoxicity in transgenic mice expressing the mutated canine ABCB1 gene. Am J Vet Res 2015; 75:1104-10. [PMID: 25419811 DOI: 10.2460/ajvr.75.12.1104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To identify biomarkers of P-glycoprotein (P-gp) substrate neurotoxicity in transgenic mice expressing the mutant canine ABCB1 gene (ABCB1-1Δ). ANIMALS 8 ABCB1 knock-in and knock-out transgenic mice expressing the ABCB1-1Δ gene and 8 control mice expressing the wild-type canine ABCB1 gene (ABCB1-WT). PROCEDURES Groups including 2 ABCB1-1Δ mutant mice and 2 ABCB1-WT mice were administered the P-gp substrates ivermectin (10 mg/kg, SC), doramectin (10 mg/kg, SC), moxidectin (10 mg/kg, PO), or digoxin (1.53 mg/kg, SC). A toxicogenomic approach based on DNA microarrays was used to examine whole-genome expression changes in mice administered P-gp substrates. RESULTS Compared with control ABCB1-WT mice, ABCB1-1Δ mutant mice developed neurotoxic signs including ataxia, lethargy, and tremors similar to those reported for dogs with the ABCB1-1Δ mutation. Microarray analysis revealed that gene expression was altered in ABCB1-1Δ mutant mice, compared with findings for ABCB1-WT mice, following administration of the same P-gp substrates. Gene pathway analysis revealed that genes with a ≥ 2-fold gene expression change were associated with behavior and nervous system development and function. Moreover, 34 genes were altered in the ABCB1-1Δ mutant mice in all 4 drug treatment groups. These genes were also associated with behavior, which was identified as the top-ranked gene network. CONCLUSIONS AND CLINICAL RELEVANCE These study data have facilitated understanding of the molecular mechanisms of neurotoxicosis in ABCB1-1Δ mutant mice following exposure to various P-gp substrates. Some genes appear to be potential biomarkers of P-gp substrate neurotoxicity that might be used to predict the safety of those drugs in dogs with the ABCB1-1Δ mutation.
Collapse
Affiliation(s)
- Min Zhu
- Office of Research, Center for Veterinary Medicine, US FDA, 8401 Muirkirk Rd, Laurel, MD 20708
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
Attentional set-shifting, as a measure of executive flexibility, has been a staple of investigations into human cognition for over six decades. Mediated by the frontal cortex in mammals, the cognitive processes involved in forming, maintaining and shifting an attentional set are vulnerable to dysfunction arising from a number of human neurodegenerative diseases (such as Alzheimer's, Parkinson's and Huntington's diseases) and other neurological disorders (such as schizophrenia, depression, and attention deficit/hyperactivity disorder). Our understanding of these diseases and disorders, and the cognitive impairments induced by them, continues to advance, in tandem with an increasing number of tools at our disposal. In this chapter, we review and compare commonly used attentional set-shifting tasks (the Wisconsin Card Sorting Task and Intradimensional/Extradimensional tasks) and their applicability across species. In addition to humans, attentional set-shifting has been observed in a number of other animals, with a substantial body of literature describing performance in monkeys and rodents. We consider the task designs used to investigate attentional set-shifting in these species and the methods used to model human diseases and disorders, and ultimately the comparisons and differences between species-specific tasks, and between performance across species.
Collapse
|
12
|
Crabtree GW, Gogos JA. Synaptic plasticity, neural circuits, and the emerging role of altered short-term information processing in schizophrenia. Front Synaptic Neurosci 2014; 6:28. [PMID: 25505409 PMCID: PMC4243504 DOI: 10.3389/fnsyn.2014.00028] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 10/22/2014] [Indexed: 01/01/2023] Open
Abstract
Synaptic plasticity alters the strength of information flow between presynaptic and postsynaptic neurons and thus modifies the likelihood that action potentials in a presynaptic neuron will lead to an action potential in a postsynaptic neuron. As such, synaptic plasticity and pathological changes in synaptic plasticity impact the synaptic computation which controls the information flow through the neural microcircuits responsible for the complex information processing necessary to drive adaptive behaviors. As current theories of neuropsychiatric disease suggest that distinct dysfunctions in neural circuit performance may critically underlie the unique symptoms of these diseases, pathological alterations in synaptic plasticity mechanisms may be fundamental to the disease process. Here we consider mechanisms of both short-term and long-term plasticity of synaptic transmission and their possible roles in information processing by neural microcircuits in both health and disease. As paradigms of neuropsychiatric diseases with strongly implicated risk genes, we discuss the findings in schizophrenia and autism and consider the alterations in synaptic plasticity and network function observed in both human studies and genetic mouse models of these diseases. Together these studies have begun to point toward a likely dominant role of short-term synaptic plasticity alterations in schizophrenia while dysfunction in autism spectrum disorders (ASDs) may be due to a combination of both short-term and long-term synaptic plasticity alterations.
Collapse
Affiliation(s)
- Gregg W. Crabtree
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia UniversityNew York, NY, USA
| | - Joseph A. Gogos
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia UniversityNew York, NY, USA
- Department of Neuroscience, College of Physicians and Surgeons, Columbia UniversityNew York, NY, USA
| |
Collapse
|
13
|
Brennand KJ, Landek-Salgado MA, Sawa A. Modeling heterogeneous patients with a clinical diagnosis of schizophrenia with induced pluripotent stem cells. Biol Psychiatry 2014; 75:936-44. [PMID: 24331955 PMCID: PMC4022707 DOI: 10.1016/j.biopsych.2013.10.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 10/29/2013] [Accepted: 10/30/2013] [Indexed: 12/28/2022]
Abstract
Schizophrenia (SZ) is a devastating complex genetic mental condition that is heterogeneous in terms of clinical etiologies, symptoms, and outcomes. Despite decades of postmortem, neuroimaging, pharmacological, and genetic studies of patients, in addition to animal models, much of the biological mechanisms that underlie the pathology of SZ remain unknown. The ability to reprogram adult somatic cells into human induced pluripotent stem cells (hiPSCs) provides a new tool that supplies live human neurons for modeling complex genetic conditions such as SZ. The purpose of this review is to discuss the technical and clinical constraints currently limiting hiPSC-based studies. We posit that reducing the clinical heterogeneity of hiPSC-based studies, by selecting subjects with common clinical manifestations or rare genetic variants, will help our ability to draw meaningful insights from the necessarily small patient cohorts that can be studied at this time.
Collapse
Affiliation(s)
- Kristen J Brennand
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.
| | | | - Akira Sawa
- Department of Psychiatry, John Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
14
|
Meyer F, Louilot A. Consequences at adulthood of transient inactivation of the parahippocampal and prefrontal regions during early development: new insights from a disconnection animal model for schizophrenia. Front Behav Neurosci 2014; 8:118. [PMID: 24778609 PMCID: PMC3985036 DOI: 10.3389/fnbeh.2014.00118] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 03/21/2014] [Indexed: 11/16/2022] Open
Abstract
The psychic disintegration characteristic of schizophrenia is thought to result from a defective connectivity, of neurodevelopmental origin, between several integrative brain regions. The parahippocampal region and the prefrontal cortex are described as the main regions affected in schizophrenia. Interestingly, latent inhibition (LI) has been found to be reduced in patients with schizophrenia, and the existence of a dopaminergic dysfunction is also generally well accepted in this disorder. In the present review, we have integrated behavioral and neurochemical data obtained in a LI protocol involving adult rats subjected to neonatal functional inactivation of the entorhinal cortex, the ventral subiculum or the prefrontal cortex. The data discussed suggest a subtle and transient functional blockade during early development of the aforementioned brain regions is sufficient to induce schizophrenia-related behavioral and dopaminergic abnormalities in adulthood. In summary, these results support the view that our conceptual and methodological approach, based on functional disconnections, is valid for modeling some aspects of the pathophysiology of schizophrenia from a neurodevelopmental perspective.
Collapse
Affiliation(s)
- Francisca Meyer
- 1Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Nijmegen, Netherlands
| | - Alain Louilot
- 2INSERM U 1114, Faculty of Medicine, FMTS, University of Strasbourg Strasbourg, France
| |
Collapse
|
15
|
Turner KM, Burne THJ. Comprehensive behavioural analysis of Long Evans and Sprague-Dawley rats reveals differential effects of housing conditions on tests relevant to neuropsychiatric disorders. PLoS One 2014; 9:e93411. [PMID: 24671152 PMCID: PMC3966872 DOI: 10.1371/journal.pone.0093411] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/05/2014] [Indexed: 01/07/2023] Open
Abstract
Genetic (G) and environmental (E) manipulations are known to alter behavioural outcomes in rodents, however many animal models of neuropsychiatric disorders only use a restricted selection of strain and housing conditions. The aim of this study was to examine GxE interactions comparing two outbred rat strains, which were housed in either standard or enriched cages. The strains selected were the albino Sprague-Dawley rat, commonly used for animal models, and the other was the pigmented Long Evans rat, which is frequently used in cognitive studies. Rats were assessed using a comprehensive behavioural test battery and included well-established tests frequently employed to examine animal models of neuropsychiatric diseases, measuring aspects of anxiety, exploration, sensorimotor gating and cognition. Selective strain and housing effects were observed on a number of tests. These included increased locomotion and reduced pre-pulse inhibition in Long Evans rats compared to Sprague Dawley rats; and rats housed in enriched cages had reduced anxiety-like behaviour compared to standard housed rats. Long Evans rats required fewer sessions than Sprague Dawley rats to learn operant tasks, including a signal detection task and reversal learning. Furthermore, Long Evans rats housed in enriched cages acquired simple operant tasks faster than standard housed Long Evans rats. Cognitive phenotypes in animal models of neuropsychiatric disorders would benefit from using strain and housing conditions where there is greater potential for both enhancement and deficits in performance.
Collapse
Affiliation(s)
- Karly M. Turner
- Queensland Brain Institute, The University of Queensland, St Lucia, Australia
| | - Thomas H. J. Burne
- Queensland Brain Institute, The University of Queensland, St Lucia, Australia
- Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, Australia
- * E-mail:
| |
Collapse
|
16
|
Reichelt AC, Lee JLC. Memory reconsolidation in aversive and appetitive settings. Front Behav Neurosci 2013; 7:118. [PMID: 24058336 PMCID: PMC3766793 DOI: 10.3389/fnbeh.2013.00118] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/20/2013] [Indexed: 11/16/2022] Open
Abstract
Memory reconsolidation has been observed across species and in a number of behavioral paradigms. The majority of memory reconsolidation studies have been carried out in Pavlovian fear conditioning and other aversive memory settings, with potential implications for the treatment of post-traumatic stress disorder. However, there is a growing literature on memory reconsolidation in appetitive reward-related memory paradigms, including translational models of drug addiction. While there appears to be substantial similarity in the basic phenomenon and underlying mechanisms of memory reconsolidation across unconditioned stimulus valence, there are also notable discrepancies. These arise both when comparing aversive to appetitive paradigms and also across different paradigms within the same valence of memory. We review the demonstration of memory reconsolidation across different aversive and appetitive memory paradigms, the commonalities and differences in underlying mechanisms and the conditions under which each memory undergoes reconsolidation. We focus particularly on whether principles derived from the aversive literature are applicable to appetitive settings, and also whether the expanding literature in appetitive paradigms is informative for fear memory reconsolidation.
Collapse
Affiliation(s)
- Amy C Reichelt
- School of Psychology, University of Birmingham Birmingham, UK
| | | |
Collapse
|
17
|
Turner KM, Burne THJ. Interaction of genotype and environment: effect of strain and housing conditions on cognitive behavior in rodent models of schizophrenia. Front Behav Neurosci 2013; 7:97. [PMID: 23914162 PMCID: PMC3728474 DOI: 10.3389/fnbeh.2013.00097] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 07/16/2013] [Indexed: 11/22/2022] Open
Abstract
Schizophrenia is associated with many genetic and environmental risk factors and there is growing evidence that the interactions between genetic and environmental "hits" are critical for disease onset. Animal models of schizophrenia have traditionally used specific strain and housing conditions to test potential risk factors. As the field moves towards testing gene (G) x environment (E) interactions the impact of these choices should be considered. Given the surge of research focused on cognitive deficits, we have examined studies of cognition in rodents from the perspective of GxE interactions, in which strain or housing manipulations have been varied. Behavior is clearly altered by these factors, yet few animal models of schizophrenia have investigated cognitive deficits using different strain and housing conditions. It is important to recognise the large variation in behavior observed when using different strain and housing combinations because GxE interactions may mask or exacerbate cognitive outcomes. Further consideration will improve our understanding of GxE interactions and the underlying neurobiology of cognitive impairments in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Karly M. Turner
- Queensland Brain Institute, The University of Queensland, St. Lucia, BrisbaneQLD, Australia
| | - Thomas H. J. Burne
- Queensland Brain Institute, The University of Queensland, St. Lucia, BrisbaneQLD, Australia
- Queensland Centre for Mental Health Research, The Park Centre for Mental Health, WacolQLD, Australia
| |
Collapse
|
18
|
Genes and environments in schizophrenia: The different pieces of a manifold puzzle. Neurosci Biobehav Rev 2013; 37:2424-37. [PMID: 23628741 DOI: 10.1016/j.neubiorev.2013.04.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 04/17/2013] [Indexed: 01/12/2023]
Abstract
Genetic research targeting schizophrenia has undergone tremendous development during recent years. Supported by recently developed high-throughput genotyping technologies, both rare and common genetic variants have been identified that show consistent association with schizophrenia. These results have been replicated by independent studies and refined in meta-analyses. The genetic variation uncovered consists of common alleles, i.e. single nucleotide polymorphisms (SNPs) conveying small effects (odds ratios below 1.1) on disease risk. The source of rare variants is copy number variations (CNVs), only detectable in a small proportion of patients (3-5% for all known CNVs) with schizophrenia, furthermore extremely rare de novo mutations captured by next generation sequencing, the most recent technological advancement in the field. Despite these findings, the search for the genetic architecture underlying schizophrenia continues since these variants explain only a small proportion of the overall phenotypic variance. Gene-environment interactions provide a compelling model for resolving this paradox and interpreting the risk factors of schizophrenia. Epidemiologically proven risk factors, such as prenatal infection, obstetric complications, urbanicity, cannabis, and trauma have been demonstrated to interact with genetic risk, giving rise to higher prevalence rates or more severe symptomatology in individuals with direct or indirect genetic predisposition for schizophrenia. Further research will have to explain how the different forms of genetic variation interact and how environmental factors modulate their effects. Moreover, the challenging question lying ahead of us is how genetic and environmental factors translate to molecular disease pathways. New approaches, including animal studies and in vitro disease modeling, as well as innovative real-world environment assessment methods, will help to understand the complex etiology of schizophrenia.
Collapse
|
19
|
Petrovszki Z, Adam G, Tuboly G, Kekesi G, Benedek G, Keri S, Horvath G. Characterization of gene–environment interactions by behavioral profiling of selectively bred rats: The effect of NMDA receptor inhibition and social isolation. Behav Brain Res 2013. [DOI: 10.1016/j.bbr.2012.11.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
20
|
Nogo and Nogo receptor: relevance to schizophrenia? Neurobiol Dis 2013; 54:150-7. [PMID: 23369871 DOI: 10.1016/j.nbd.2013.01.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 01/14/2013] [Accepted: 01/17/2013] [Indexed: 12/14/2022] Open
Abstract
The membrane protein Nogo-A and its receptor NgR have been extensively characterized for their role in restricting axonal growth, regeneration, and plasticity in the central nervous system. Recent evidence suggests that Nogo and NgR might constitute candidate genes for schizophrenia susceptibility. In this article, we critically review the possibility that dysfunctions related to Nogo-A and NgR might contribute to increased risk for schizophrenia. To this end, we consider the most important insights that have emerged from human genetic and pathological studies and from experimental animal work. Furthermore, we discuss potential mechanisms of Nogo/NgR involvement in neural circuit development and stability, and how mutations or changes in expression levels of these proteins could be developmental risk factors contributing to schizophrenia.
Collapse
|
21
|
Drews E, Otte DM, Zimmer A. Involvement of the primate specific gene G72 in schizophrenia: From genetic studies to pathomechanisms. Neurosci Biobehav Rev 2012; 37:2410-7. [PMID: 23092656 DOI: 10.1016/j.neubiorev.2012.10.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 09/03/2012] [Accepted: 10/15/2012] [Indexed: 12/28/2022]
Abstract
Schizophrenia is a human mental disorder that affects an individual's thoughts, perception, affect and behavior, which is caused by a complex interaction of genetic and environmental factors. Genetic studies have implicated the evolutionary novel, anthropoid primate-specific gene locus G72/G30 in the etiology of schizophrenia and other psychiatric disorders. This gene encodes the protein LG72, which has been discussed as a modulator of the peroxisomal enzyme d-amino-acid-oxidase (DAO), or, alternatively as a mitochondrial protein. Recently, G72 transgenic (G72Tg) mice were generated that express the protein throughout the brain. These mice show several behavioral deficits that are related to schizophrenia. Further, G72Tg mice have a reduced activity of mitochondrial complex I, with a concomitantly increased production of reactive oxygen species, as well as deficits in short-term plasticity. Results from these studies demonstrate that expression of the human G72/G30 gene locus in mice produces behavioral phenotypes that are relevant to schizophrenia. They implicate LG72-induced mitochondrial and synaptic defects as a possible pathomechanism of this disease.
Collapse
Affiliation(s)
- Eva Drews
- Institute of Molecular Psychiatry, University of Bonn, Germany.
| | | | | |
Collapse
|
22
|
Hattori S, Takao K, Tanda K, Toyama K, Shintani N, Baba A, Hashimoto H, Miyakawa T. Comprehensive behavioral analysis of pituitary adenylate cyclase-activating polypeptide (PACAP) knockout mice. Front Behav Neurosci 2012; 6:58. [PMID: 23060763 PMCID: PMC3462416 DOI: 10.3389/fnbeh.2012.00058] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 08/22/2012] [Indexed: 12/05/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide acting as a neurotransmitter, neuromodulator, or neurotrophic factor. PACAP is widely expressed throughout the brain and exerts its functions through the PACAP-specific receptor (PAC(1)). Recent studies reveal that genetic variants of the PACAP and PAC(1) genes are associated with mental disorders, and several behavioral abnormalities of PACAP knockout (KO) mice are reported. However, an insufficient number of backcrosses was made using PACAP KO mice on the C57BL/6J background due to their postnatal mortality. To elucidate the effects of PACAP on neuropsychiatric function, the PACAP gene was knocked out in F1 hybrid mice (C57BL/6J × 129SvEv) for appropriate control of the genetic background. The PACAP KO mice were then subjected to a behavioral test battery. PACAP deficiency had no significant effects on neurological screen. As shown previously, the mice exhibited significantly increased locomotor activity in a novel environment and abnormal anxiety-like behavior, while no obvious differences between genotypes were shown in home cage (HC) activity. In contrast to previous reports, the PACAP KO mice showed normal prepulse inhibition (PPI) and slightly decreased depression-like behavior. Previous study demonstrates that the social interaction (SI) in a resident-intruder test was decreased in PACAP KO mice. On the other hand, we showed that PACAP KO mice exhibited increased SI in Crawley's three-chamber social approach test, although PACAP KO had no significant impact on SI in a HC. PACAP KO mice also exhibited mild performance deficit in working memory in an eight-arm radial maze (RM) and the T-maze (TM), while they did not show any significant abnormalities in the left-right discrimination task in the TM. These results suggest that PACAP has an important role in the regulation of locomotor activity, social behavior, anxiety-like behavior and, potentially, working memory.
Collapse
Affiliation(s)
- Satoko Hattori
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health UniversityToyoake, Aichi, Japan
- Japan Science and Technology Agency, Core Research for Evolutional Science and TechnologyKawaguchi, Saitama, Japan
| | - Keizo Takao
- Japan Science and Technology Agency, Core Research for Evolutional Science and TechnologyKawaguchi, Saitama, Japan
- Center for Genetic Analysis of Behavior, National Institute for Physiological SciencesOkazaki, Aichi, Japan
- Genetic Engineering and Functional Genomics Group, Frontier Technology Center, Graduate School of Medicine, Kyoto UniversityKyoto, Kyoto, Japan
| | - Koichi Tanda
- Genetic Engineering and Functional Genomics Group, Frontier Technology Center, Graduate School of Medicine, Kyoto UniversityKyoto, Kyoto, Japan
- Department of Pediatrics, Kyoto Prefectural University of MedicineKyoto, Kyoto, Japan
| | - Keiko Toyama
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health UniversityToyoake, Aichi, Japan
- Genetic Engineering and Functional Genomics Group, Frontier Technology Center, Graduate School of Medicine, Kyoto UniversityKyoto, Kyoto, Japan
| | - Norihito Shintani
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka UniversitySuita, Osaka, Japan
| | - Akemichi Baba
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka UniversitySuita, Osaka, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka UniversitySuita, Osaka, Japan
- United Graduate School of Child Development, Osaka University, Kanazawa University and Hamamatsu University School of MedicineSuita, Osaka, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health UniversityToyoake, Aichi, Japan
- Japan Science and Technology Agency, Core Research for Evolutional Science and TechnologyKawaguchi, Saitama, Japan
- Center for Genetic Analysis of Behavior, National Institute for Physiological SciencesOkazaki, Aichi, Japan
- Genetic Engineering and Functional Genomics Group, Frontier Technology Center, Graduate School of Medicine, Kyoto UniversityKyoto, Kyoto, Japan
| |
Collapse
|
23
|
Neuregulin 1-ErbB4-PI3K signaling in schizophrenia and phosphoinositide 3-kinase-p110δ inhibition as a potential therapeutic strategy. Proc Natl Acad Sci U S A 2012; 109:12165-70. [PMID: 22689948 DOI: 10.1073/pnas.1206118109] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Neuregulin 1 (NRG1) and ErbB4, critical neurodevelopmental genes, are implicated in schizophrenia, but the mediating mechanisms are unknown. Here we identify a genetically regulated, pharmacologically targetable, risk pathway associated with schizophrenia and with ErbB4 genetic variation involving increased expression of a PI3K-linked ErbB4 receptor (CYT-1) and the phosphoinositide 3-kinase subunit, p110δ (PIK3CD). In human lymphoblasts, NRG1-mediated phosphatidyl-inositol,3,4,5 triphosphate [PI(3,4,5)P3] signaling is predicted by schizophrenia-associated ErbB4 genotype and PIK3CD levels and is impaired in patients with schizophrenia. In human brain, the same ErbB4 genotype again predicts increased PIK3CD expression. Pharmacological inhibition of p110δ using the small molecule inhibitor, IC87114, blocks the effects of amphetamine in a mouse pharmacological model of psychosis and reverses schizophrenia-related phenotypes in a rat neonatal ventral hippocampal lesion model. Consistent with these antipsychotic-like properties, IC87114 increases AKT phosphorylation in brains of treated mice, implicating a mechanism of action. Finally, in two family-based genetic studies, PIK3CD shows evidence of association with schizophrenia. Our data provide insight into a mechanism of ErbB4 association with schizophrenia; reveal a previously unidentified biological and disease link between NRG1-ErbB4, p110δ, and AKT; and suggest that p110δ is a previously undescribed therapeutic target for the treatment of psychiatric disorders.
Collapse
|
24
|
McDannald MA, Whitt JP, Calhoon GG, Piantadosi PT, Karlsson RM, O’Donnell P, Schoenbaum G. Impaired reality testing in an animal model of schizophrenia. Biol Psychiatry 2011; 70:1122-6. [PMID: 21798517 PMCID: PMC3206179 DOI: 10.1016/j.biopsych.2011.06.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 06/10/2011] [Accepted: 06/16/2011] [Indexed: 01/11/2023]
Abstract
BACKGROUND Schizophrenia is a chronic and devastating brain disorder characterized by hallucinations and delusions, symptoms reflecting impaired reality testing. Although animal models have captured negative symptoms and cognitive deficits associated with schizophrenia, none have addressed these defining, positive symptoms. METHODS Here we tested the performance of adults given neonatal ventral hippocampal lesions (NVHL), a neurodevelopmental model of schizophrenia, in two taste aversion procedures. RESULTS Normal and NVHL rats formed aversions to a palatable food when the food was directly paired with nausea, but only NVHL rats formed a food aversion when the cue predicting that food was paired with nausea. The failure of NVHL rats to discriminate fully real from imagined food parallels the failure of people with schizophrenia to differentiate internal thoughts and beliefs from reality. CONCLUSIONS These results further validate the NVHL model of schizophrenia and provide a means to assess impaired reality testing in variety of animal models.
Collapse
Affiliation(s)
- Michael A. McDannald
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF-2 S251, Baltimore, MD 21201
| | - Joshua P. Whitt
- Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-2 S251, Baltimore, MD 21201
| | - Gwen G. Calhoon
- Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-2 S251, Baltimore, MD 21201
| | - Patrick T. Piantadosi
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF-2 S251, Baltimore, MD 21201
| | - Rose M. Karlsson
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF-2 S251, Baltimore, MD 21201
| | - Patricio O’Donnell
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF-2 S251, Baltimore, MD 21201, Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-2 S251, Baltimore, MD 21201, Department of Psychiatry, University of Maryland School of Medicine, 20 Penn St, HSF-2 S251, Baltimore, MD 21201
| | - Geoffrey Schoenbaum
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF-2 S251, Baltimore, MD 21201, Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-2 S251, Baltimore, MD 21201, Department of Psychiatry, University of Maryland School of Medicine, 20 Penn St, HSF-2 S251, Baltimore, MD 21201
| |
Collapse
|
25
|
Schizophrenia susceptibility gene dysbindin regulates glutamatergic and dopaminergic functions via distinctive mechanisms in Drosophila. Proc Natl Acad Sci U S A 2011; 108:18831-6. [PMID: 22049342 DOI: 10.1073/pnas.1114569108] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The dysfunction of multiple neurotransmitter systems is a striking pathophysiological feature of many mental disorders, schizophrenia in particular, but delineating the underlying mechanisms has been challenging. Here we show that manipulation of a single schizophrenia susceptibility gene, dysbindin, is capable of regulating both glutamatergic and dopaminergic functions through two independent mechanisms, consequently leading to two categories of clinically relevant behavioral phenotypes. Dysbindin has been reported to affect glutamatergic and dopaminergic functions as well as a range of clinically relevant behaviors in vertebrates and invertebrates but has been thought to have a mainly neuronal origin. We find that reduced expression of Drosophila dysbindin (Ddysb) in presynaptic neurons significantly suppresses glutamatergic synaptic transmission and that this glutamatergic defect is responsible for impaired memory. However, only the reduced expression of Ddysb in glial cells is the cause of hyperdopaminergic activities that lead to abnormal locomotion and altered mating orientation. This effect is attributable to the altered expression of a dopamine metabolic enzyme, Ebony, in glial cells. Thus, Ddysb regulates glutamatergic transmission through its neuronal function and regulates dopamine metabolism by regulating Ebony expression in glial cells.
Collapse
|
26
|
Lin CY, Sawa A, Jaaro-Peled H. Better understanding of mechanisms of schizophrenia and bipolar disorder: from human gene expression profiles to mouse models. Neurobiol Dis 2011; 45:48-56. [PMID: 21914480 DOI: 10.1016/j.nbd.2011.08.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 08/22/2011] [Accepted: 08/26/2011] [Indexed: 01/01/2023] Open
Abstract
The molecular mechanisms of major mental illnesses, such as schizophrenia and bipolar disorder, are unclear. To address this fundamental question, many groups have studied molecular expression profiles in postmortem brains and other tissues from patients compared with those from normal controls. Development of unbiased high-throughput approaches, such as microarray, RNA-seq, and proteomics, have supported and facilitated this endeavor. In addition to genes directly involved in neuron/glia signaling, especially those encoding for synaptic proteins, genes for metabolic cascades are differentially expressed in the brains of patients with schizophrenia and bipolar disorder, compared with those from normal controls in DNA microarray studies. Here we propose the importance and usefulness of genetic mouse models in which such differentially expressed molecules are modulated. These animal models allow us to dissect the mechanisms of how such molecular changes in patient brains may play a role in neuronal circuitries and overall behavioral phenotypes. We also point out that models in which the metabolic genes are modified are obviously untested from mental illness viewpoints, suggesting the potential to re-address these models with behavioral assays and neurochemical assessments.
Collapse
Affiliation(s)
- Chi-Ying Lin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | |
Collapse
|
27
|
Schijndel JEV, Martens GJM. Gene expression profiling in rodent models for schizophrenia. Curr Neuropharmacol 2011; 8:382-93. [PMID: 21629445 PMCID: PMC3080594 DOI: 10.2174/157015910793358132] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Revised: 04/15/2010] [Accepted: 04/30/2010] [Indexed: 12/12/2022] Open
Abstract
The complex neurodevelopmental disorder schizophrenia is thought to be induced by an interaction between predisposing genes and environmental stressors. In order to get a better insight into the aetiology of this complex disorder, animal models have been developed. In this review, we summarize mRNA expression profiling studies on neurodevelopmental, pharmacological and genetic animal models for schizophrenia. We discuss parallels and contradictions among these studies, and propose strategies for future research.
Collapse
Affiliation(s)
- Jessica E Van Schijndel
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Centre for Neuroscience & Nijmegen Centre for Molecular Life Sciences (NCMLS), Faculty of Science, Radboud University Nijmegen, 6525 GA, Nijmegen, The Netherlands
| | | |
Collapse
|
28
|
Lazar NL, Neufeld RWJ, Cain DP. Contribution of nonprimate animal models in understanding the etiology of schizophrenia. J Psychiatry Neurosci 2011; 36:E5-29. [PMID: 21247514 PMCID: PMC3120891 DOI: 10.1503/jpn.100054] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia is a severe psychiatric disorder that is characterized by positive and negative symptoms and cognitive impairments. The etiology of the disorder is complex, and it is thought to follow a multifactorial threshold model of inheritance with genetic and neurodevelop mental contributions to risk. Human studies are particularly useful in capturing the richness of the phenotype, but they are often limited to the use of correlational approaches. By assessing behavioural abnormalities in both humans and rodents, nonprimate animal models of schizophrenia provide unique insight into the etiology and mechanisms of the disorder. This review discusses the phenomenology and etiology of schizophrenia and the contribution of current nonprimate animal models with an emphasis on how research with models of neuro transmitter dysregulation, environmental risk factors, neurodevelopmental disruption and genetic risk factors can complement the literature on schizophrenia in humans.
Collapse
Affiliation(s)
- Noah L Lazar
- Department of Psychology, University of Western Ontario, London, Ont.
| | | | | |
Collapse
|
29
|
Taniguchi Y, Young-Pearse T, Sawa A, Kamiya A. In utero electroporation as a tool for genetic manipulation in vivo to study psychiatric disorders: from genes to circuits and behaviors. Neuroscientist 2011; 18:169-79. [PMID: 21551077 DOI: 10.1177/1073858411399925] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Many genetic risk factors for major mental disorders have key roles in brain development. Thus, exploring the roles for these genetic factors for brain development at the molecular, cellular, and neuronal circuit level is crucial for discovering how genetic disturbances affect high brain functions, which ultimately lead to disease pathologies. However, it is a tremendously difficult task, given that most mental disorders have genetic complexities in which many genetic risk factors have multiple roles in different cell types and brain regions over a time-course dependent manner. Furthermore, some genetic risk factors are likely to act epistatically in common molecular pathways. For this reason, a technique for spatial and temporal manipulation of multiple genes is necessary for understanding how genetic disturbances contribute to disease etiology. Here, the authors will review the said technique, in utero electroporation, which investigates the molecular disease pathways in rodent models for major mental disorders. This technique is also useful to examine the effect of genetic risks at the behavioral level. Furthermore, the authors will discuss the recent progress of this technology, such as inducible and cell type-specific targeting, as well as nonepisomal genetic manipulation, which provide further availability of this technique for research on major mental disorders.
Collapse
Affiliation(s)
- Yu Taniguchi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | | | | | | |
Collapse
|
30
|
Lu L, Mamiya T, Koseki T, Mouri A, Nabeshima T. Genetic Animal Models of Schizophrenia Related with the Hypothesis of Abnormal Neurodevelopment. Biol Pharm Bull 2011; 34:1358-63. [DOI: 10.1248/bpb.34.1358] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Lingling Lu
- The Academic Frontier Project for Private University, Comparative Cognitive Science Institute, Meijo University
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University
| | - Takayoshi Mamiya
- The Academic Frontier Project for Private University, Comparative Cognitive Science Institute, Meijo University
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University
- Japanese Drug Organization for Appropriate Use and Research
| | - Takenao Koseki
- The Academic Frontier Project for Private University, Comparative Cognitive Science Institute, Meijo University
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University
| | - Akihiro Mouri
- The Academic Frontier Project for Private University, Comparative Cognitive Science Institute, Meijo University
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University
- Japanese Drug Organization for Appropriate Use and Research
| | - Toshitaka Nabeshima
- The Academic Frontier Project for Private University, Comparative Cognitive Science Institute, Meijo University
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University
- Japanese Drug Organization for Appropriate Use and Research
| |
Collapse
|
31
|
TRP Channels and Psychiatric Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:987-1009. [DOI: 10.1007/978-94-007-0265-3_51] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
32
|
Manning EE, Ransome MI, Burrows EL, Hannan AJ. Increased adult hippocampal neurogenesis and abnormal migration of adult-born granule neurons is associated with hippocampal-specific cognitive deficits in phospholipase C-β1 knockout mice. Hippocampus 2010; 22:309-19. [PMID: 21080410 DOI: 10.1002/hipo.20900] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2010] [Indexed: 12/12/2022]
Abstract
Schizophrenia is a devastating psychiatric illness with a complex pathophysiology. We have recently documented schizophrenia-like endophenotypes in phospholipase C-β1 knockout (PLC-β1(-/-)) mice, including deficits in prepulse inhibition, hyperlocomotion, and cognitive impairments. PLC-β1 signals via multiple G-protein coupled receptor pathways implicated in neural cellular plasticity; however, adult neurogenesis has yet to be explored in this knockout model. In this study, we employed PLC-β1(-/-) mice to elucidate possible correlates between aberrant adult hippocampal neurogenesis (AHN) and schizophrenia-like behaviors. Using stereology and bromodeoxyuridine (BrdU) immunohistochemistry we demonstrated a significant increase in the density of adult-generated cells in the granule cell layer (GCL) of adult PLC-β1(-/-) mice compared with wild-type littermates. Cellular phenotype analysis using confocal microscopy revealed these cells to be mature granule neurons expressing NeuN and calbindin. Increased neuronal survival occurred concomitant with reduced caspase-3(+) cells in the GCL of PLC-β1(-/-) mice. Stereological analysis of Ki67(+) cells in the subgranular zone suggested that neural precursor proliferation is unchanged in PLC-β1(-/-) mice. We further showed aberrant migration of mature granule neurons within the GCL of adult PLC-β1(-/-) mice with excessive adult-generated mature neurons residing in the middle and outer GCL. PLC-β1(-/-) mice exhibited specific behavioral deficits in location recognition, a measure of hippocampal-dependent memory, but not novel object recognition. Overall, we have shown that PLC-β1(-/-) mice have a threefold increase in net AHN, and have provided further evidence to suggest a specific deficit in hippocampal-dependent cognition. We propose that abnormal cellular plasticity in these mice may contribute to their schizophrenia-like behavioral endophenotypes.
Collapse
Affiliation(s)
- Elizabeth E Manning
- Howard Florey Institute, Florey Neuroscience Institutes, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | |
Collapse
|
33
|
Dawe GS, Nagarajah R, Albert R, Casey DE, Gross KW, Ratty AK. Antipsychotic drugs dose-dependently suppress the spontaneous hyperactivity of the chakragati mouse. Neuroscience 2010; 171:162-72. [PMID: 20816926 DOI: 10.1016/j.neuroscience.2010.08.061] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 08/18/2010] [Accepted: 08/30/2010] [Indexed: 12/22/2022]
Abstract
The chakragati (ckr) mouse has been proposed as a model of aspects of schizophrenia. The mice, created serendipitously as a result of a transgenic insertional mutation, exhibit spontaneous circling, hyperactivity, hypertone of the dopamine system, reduced social interactions, enlarged lateral ventricles, deficits in pre-pulse inhibition of acoustic startle and deficits in latent inhibition of conditioned learning. In this study, the dose-dependent effects of antipsychotic drugs (haloperidol, pimozide, risperidone, clozapine, olanzapine, ziprasidone, quetiapine and aripiprazole) on the spontaneous hyperactivity of the mice were investigated. All the antipsychotic drugs tested dose-dependently suppressed spontaneous hyperactivity. Aripriprazole, which is known to be a dopamine D2 receptor partial agonist, exhibited a tri-phasic dose-response, initially suppressing hyperactivity at low doses, having little effect on hyperactivity at intermediate doses, and suppressing activity again at high doses. These data suggest that the spontaneous circling and hyperactivity of the ckr mouse may allow screening of candidate antipsychotic compounds, distinguishing compounds with aripriprazole-like profiles.
Collapse
Affiliation(s)
- G S Dawe
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System and Neurobiology and Ageing Programme, Centre for Life Sciences, National University of Singapore, 28 Medical Drive, Singapore 117456.
| | | | | | | | | | | |
Collapse
|
34
|
Ben Abdallah NMB, Fuss J, Trusel M, Galsworthy MJ, Bobsin K, Colacicco G, Deacon RMJ, Riva MA, Kellendonk C, Sprengel R, Lipp HP, Gass P. The puzzle box as a simple and efficient behavioral test for exploring impairments of general cognition and executive functions in mouse models of schizophrenia. Exp Neurol 2010; 227:42-52. [PMID: 20851119 DOI: 10.1016/j.expneurol.2010.09.008] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 08/12/2010] [Accepted: 09/04/2010] [Indexed: 10/19/2022]
Abstract
Deficits in executive functions are key features of schizophrenia. Rodent behavioral paradigms used so far to find animal correlates of such deficits require extensive effort and time. The puzzle box is a problem-solving test in which mice are required to complete escape tasks of increasing difficulty within a limited amount of time. Previous data have indicated that it is a quick but highly reliable test of higher-order cognitive functioning. We evaluated the use of the puzzle box to explore executive functioning in five different mouse models of schizophrenia: mice with prefrontal cortex and hippocampus lesions, mice treated sub-chronically with the NMDA-receptor antagonist MK-801, mice constitutively lacking the GluA1 subunit of AMPA-receptors, and mice over-expressing dopamine D2 receptors in the striatum. All mice displayed altered executive functions in the puzzle box, although the nature and extent of the deficits varied between the different models. Deficits were strongest in hippocampus-lesioned and GluA1 knockout mice, while more subtle deficits but specific to problem solving were found in the medial prefrontal-lesioned mice, MK-801-treated mice, and in mice with striatal overexpression of D2 receptors. Data from this study demonstrate the utility of the puzzle box as an effective screening tool for executive functions in general and for schizophrenia mouse models in particular.
Collapse
Affiliation(s)
- Nada M-B Ben Abdallah
- Central Institute of Mental Health Mannheim (CIMH), RG Behavioral Biology, University of Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zhang X, Bearer EL, Boulat B, Hall FS, Uhl GR, Jacobs RE. Altered neurocircuitry in the dopamine transporter knockout mouse brain. PLoS One 2010; 5:e11506. [PMID: 20634895 PMCID: PMC2901340 DOI: 10.1371/journal.pone.0011506] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Accepted: 06/16/2010] [Indexed: 11/28/2022] Open
Abstract
The plasma membrane transporters for the monoamine neurotransmitters dopamine, serotonin, and norepinephrine modulate the dynamics of these monoamine neurotransmitters. Thus, activity of these transporters has significant consequences for monoamine activity throughout the brain and for a number of neurological and psychiatric disorders. Gene knockout (KO) mice that reduce or eliminate expression of each of these monoamine transporters have provided a wealth of new information about the function of these proteins at molecular, physiological and behavioral levels. In the present work we use the unique properties of magnetic resonance imaging (MRI) to probe the effects of altered dopaminergic dynamics on meso-scale neuronal circuitry and overall brain morphology, since changes at these levels of organization might help to account for some of the extensive pharmacological and behavioral differences observed in dopamine transporter (DAT) KO mice. Despite the smaller size of these animals, voxel-wise statistical comparison of high resolution structural MR images indicated little morphological change as a consequence of DAT KO. Likewise, proton magnetic resonance spectra recorded in the striatum indicated no significant changes in detectable metabolite concentrations between DAT KO and wild-type (WT) mice. In contrast, alterations in the circuitry from the prefrontal cortex to the mesocortical limbic system, an important brain component intimately tied to function of mesolimbic/mesocortical dopamine reward pathways, were revealed by manganese-enhanced MRI (MEMRI). Analysis of co-registered MEMRI images taken over the 26 hours after introduction of Mn2+ into the prefrontal cortex indicated that DAT KO mice have a truncated Mn2+ distribution within this circuitry with little accumulation beyond the thalamus or contralateral to the injection site. By contrast, WT littermates exhibit Mn2+ transport into more posterior midbrain nuclei and contralateral mesolimbic structures at 26 hr post-injection. Thus, DAT KO mice appear, at this level of anatomic resolution, to have preserved cortico-striatal-thalamic connectivity but diminished robustness of reward-modulating circuitry distal to the thalamus. This is in contradistinction to the state of this circuitry in serotonin transporter KO mice where we observed more robust connectivity in more posterior brain regions using methods identical to those employed here.
Collapse
Affiliation(s)
- Xiaowei Zhang
- Biological Imaging Center, Beckman Institute, California Institute of Technology, Pasadena, California, United States of America
| | - Elaine L. Bearer
- Biological Imaging Center, Beckman Institute, California Institute of Technology, Pasadena, California, United States of America
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Benoit Boulat
- Biological Imaging Center, Beckman Institute, California Institute of Technology, Pasadena, California, United States of America
| | - F. Scott Hall
- Molecular Neurobiology Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, United States of America
| | - George R. Uhl
- Molecular Neurobiology Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, United States of America
| | - Russell E. Jacobs
- Biological Imaging Center, Beckman Institute, California Institute of Technology, Pasadena, California, United States of America
- * E-mail:
| |
Collapse
|
36
|
Chow VW, Savonenko AV, Melnikova T, Kim H, Price DL, Li T, Wong PC. Modeling an anti-amyloid combination therapy for Alzheimer's disease. Sci Transl Med 2010; 2:13ra1. [PMID: 20371462 DOI: 10.1126/scitranslmed.3000337] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As only symptomatic treatments are now available for Alzheimer's disease (AD), safe and effective mechanism-based therapies remain a great unmet need for patients with this neurodegenerative disease. Although gamma-secretase and BACE1 [beta-site beta-amyloid (Abeta) precursor protein (APP) cleaving enzyme 1] are well-recognized therapeutic targets for AD, untoward side effects associated with strong inhibition or reductions in amounts of these aspartyl proteases have raised concerns regarding their therapeutic potential. Although moderate decreases of either gamma-secretase or BACE1 are not associated with mechanism-based toxicities, they provide only modest benefits in reducing Abeta in the brains of APPswe/PS1DeltaE9 mice. Because the processing of APP to generate Abeta requires both gamma-secretase and BACE1, it is possible that moderate reductions of both enzymes would provide additive and significant protection against Abeta amyloidosis. Here, we test this hypothesis and assess the value of this novel anti-amyloid combination therapy in mutant mice. We demonstrate that genetic reductions of both BACE1 and gamma-secretase additively attenuate the amyloid burden and ameliorate cognitive deficits occurring in aged APPswe/PS1DeltaE9 animals. No evidence of mechanism-based toxicities was associated with such decreases in amounts of both enzymes. Thus, we propose that targeting both gamma-secretase and BACE1 may be an effective and safe treatment strategy for AD.
Collapse
Affiliation(s)
- Vivian W Chow
- Department of Pathology, Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Niwa M, Kamiya A, Murai R, Kubo KI, 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; 65:480-9. [PMID: 20188653 DOI: 10.1016/j.neuron.2010.01.019] [Citation(s) in RCA: 241] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2010] [Indexed: 01/28/2023]
Abstract
Adult brain function and behavior are influenced by neuronal network formation during development. Genetic susceptibility factors for adult psychiatric illnesses, such as Neuregulin-1 and Disrupted-in-Schizophrenia-1 (DISC1), influence adult high brain functions, including cognition and information processing. These factors have roles during neurodevelopment and are likely to cooperate, forming pathways or "signalosomes." Here we report the potential to generate an animal model via in utero gene transfer in order to address an important question of how nonlethal deficits in early development may affect postnatal brain maturation and high brain functions in adulthood, which are impaired in various psychiatric illnesses such as schizophrenia. We show that transient knockdown of DISC1 in the pre- and perinatal stages, specifically in a lineage of pyramidal neurons mainly in the prefrontal cortex, leads to selective abnormalities in postnatal mesocortical dopaminergic maturation and behavioral abnormalities associated with disturbed cortical neurocircuitry after puberty.
Collapse
Affiliation(s)
- Minae Niwa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
O'Tuathaigh CMP, Kirby BP, Moran PM, Waddington JL. Mutant mouse models: genotype-phenotype relationships to negative symptoms in schizophrenia. Schizophr Bull 2010; 36:271-88. [PMID: 19934211 PMCID: PMC2833123 DOI: 10.1093/schbul/sbp125] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Negative symptoms encompass diminution in emotional expression and motivation, some of which relate to human attributes that may not be accessible readily in animals. Additionally, their refractoriness to treatment precludes therapeutic validation of putative models. This review considers critically the application of mutant mouse models to the study of the pathobiology of negative symptoms. It focuses on 4 main approaches: genes related to the pathobiology of schizophrenia, genes associated with risk for schizophrenia, neurodevelopmental-synaptic genes, and variant approaches from other areas of neurobiology. Despite rapid advances over the past several years, it is clear that we continue to face substantive challenges in applying mutant models to better understand the pathobiology of negative symptoms: the majority of evidence relates to impairments in social behavior, with only limited data relating to anhedonia and negligible data concerning avolition and other features; even for the most widely examined feature, social behavior, studies have used diverse assessments thereof; modelling must proceed in cognizance of increasing evidence that genes and pathobiologies implicated in schizophrenia overlap with other psychotic disorders, particularly bipolar disorder. Despite the caveats and challenges, several mutant lines evidence a phenotype for at least one index of social behavior. Though this may suggest superficially some shared relationship to negative symptoms, it is not yet possible to specify either the scope or the pathobiology of that relationship for any given gene. The breadth and depth of ongoing studies in mutants hold the prospect of addressing these shortcomings.
Collapse
Affiliation(s)
- Colm M. P. O'Tuathaigh
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, St Stephen's Green, Dublin 2, Ireland,To whom correspondence should be addressed; tel: +353-1-402-2377, fax: +353-1-402-2453, e-mail:
| | - Brian P. Kirby
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Paula M. Moran
- School of Psychology, University of Nottingham, Nottingham, UK
| | - John L. Waddington
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, St Stephen's Green, Dublin 2, Ireland
| |
Collapse
|
39
|
van den Buuse M. Modeling the positive symptoms of schizophrenia in genetically modified mice: pharmacology and methodology aspects. Schizophr Bull 2010; 36:246-70. [PMID: 19900963 PMCID: PMC2833124 DOI: 10.1093/schbul/sbp132] [Citation(s) in RCA: 283] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In recent years, there have been huge advances in the use of genetically modified mice to study pathophysiological mechanisms involved in schizophrenia. This has allowed rapid progress in our understanding of the role of several proposed gene mechanisms in schizophrenia, and yet this research has also revealed how much still remains unresolved. Behavioral studies in genetically modified mice are reviewed with special emphasis on modeling psychotic-like behavior. I will particularly focus on observations on locomotor hyperactivity and disruptions of prepulse inhibition (PPI). Recommendations are included to address pharmacological and methodological aspects in future studies. Mouse models of dopaminergic and glutamatergic dysfunction are then discussed, reflecting the most important and widely studied neurotransmitter systems in schizophrenia. Subsequently, psychosis-like behavior in mice with modifications in the most widely studied schizophrenia susceptibility genes is reviewed. Taken together, the available studies reveal a wealth of available data which have already provided crucial new insight and mechanistic clues which could lead to new treatments or even prevention strategies for schizophrenia.
Collapse
Affiliation(s)
- Maarten van den Buuse
- Mental Health Research Institute of Victoria, Parkville, Melbourne, Victoria 3052, Australia.
| |
Collapse
|
40
|
Wallén-Mackenzie A, Wootz H, Englund H. Genetic inactivation of the vesicular glutamate transporter 2 (VGLUT2) in the mouse: what have we learnt about functional glutamatergic neurotransmission? Ups J Med Sci 2010; 115:11-20. [PMID: 20187846 PMCID: PMC2853350 DOI: 10.3109/03009730903572073] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
During the past decade, three proteins that possess the capability of packaging glutamate into presynaptic vesicles have been identified and characterized. These three vesicular glutamate transporters, VGLUT1-3, are encoded by solute carrier genes Slc17a6-8. VGLUT1 (Slc17a7) and VGLUT2 (Slc17a6) are expressed in glutamatergic neurons, while VGLUT3 (Slc17a8) is expressed in neurons classically defined by their use of another transmitter, such as acetylcholine and serotonin. As glutamate is both a ubiquitous amino acid and the most abundant neurotransmitter in the adult central nervous system, the discovery of the VGLUTs made it possible for the first time to identify and specifically target glutamatergic neurons. By molecular cloning techniques, different VGLUT isoforms have been genetically targeted in mice, creating models with alterations in their glutamatergic signalling. Glutamate signalling is essential for life, and its excitatory function is involved in almost every neuronal circuit. The importance of glutamatergic signalling was very obvious when studying full knockout models of both VGLUT1 and VGLUT2, none of which were compatible with normal life. While VGLUT1 full knockout mice die after weaning, VGLUT2 full knockout mice die immediately after birth. Many neurological diseases have been associated with altered glutamatergic signalling in different brain regions, which is why conditional knockout mice with abolished VGLUT-mediated signalling only in specific circuits may prove helpful in understanding molecular mechanisms behind such pathologies. We review the recent studies in which mouse genetics have been used to characterize the functional role of VGLUT2 in the central nervous system.
Collapse
Affiliation(s)
- Asa Wallén-Mackenzie
- Department of Neuroscience, Unit of Developmental Genetics, Biomedical Center, Box 593, Uppsala University, S-751 24 Uppsala, Sweden.
| | | | | |
Collapse
|
41
|
Constitutive genetic deletion of the growth regulator Nogo-A induces schizophrenia-related endophenotypes. J Neurosci 2010; 30:556-67. [PMID: 20071518 DOI: 10.1523/jneurosci.4393-09.2010] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The membrane protein Nogo-A, which is predominantly expressed by oligodendrocytes in the adult CNS and by neurons mainly during development, is well known for limiting neurite outgrowth and regeneration in the injured mammalian CNS. In addition, it has recently been proposed that abnormal Nogo-A expression or Nogo receptor (NgR) mutations may confer genetic risks for neuropsychiatric disorders of presumed neurodevelopmental origin, such as schizophrenia. We therefore evaluated whether Nogo-A deletion may lead to schizophrenia-like abnormalities in a mouse model of genetic Nogo-A deficiency. Here, we show that systemic, lifelong knock-out of the Nogo-A gene can lead to specific behavioral abnormalities resembling schizophrenia-related endophenotypes: deficient sensorimotor gating, disrupted latent inhibition, perseverative behavior, and increased sensitivity to the locomotor stimulating effects of amphetamine. These behavioral phenotypes were accompanied by altered monoaminergic transmitter levels in specific striatal and limbic structures, as well as changes in dopamine D2 receptor expression in the same brain regions. Nogo-A deletion was further associated with elevated expression of growth-related markers. In contrast, acute antibody-mediated Nogo-A neutralization in adult wild-type mice failed to produce such phenotypes, suggesting that the phenotypes observed in the knock-out mice might be of developmental origin, and that Nogo-A normally subserves critical functions in neurodevelopment. This study provides the first experimental demonstration that Nogo-A bears neuropsychiatric relevance, and alterations in its expression may be one etiological factor in schizophrenia and related disorders.
Collapse
|
42
|
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; 20:1523-8. [PMID: 19829162 PMCID: PMC2880453 DOI: 10.1097/wnr.0b013e328330f6e7] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Neuregulin 1 (NRG1) is a pleiotropic growth factor involved in diverse aspects of brain development and function. In schizophrenia, expression of the NRG1 type I isoform is selectively increased. However, virtually nothing is known about the roles of this isoform in brain. We have studied transgenic mice overexpressing type I NRG1(NRG1type 1-tg) using a series of behavioural tests. NRG1(type 1-tg) mice have a tremor, are impaired on the accelerating rotarod, and have reduced prepulse inhibition in the context of an increased baseline startle response. There is no overall anxiety or activity phenotype, although female NRG(1type 1-tg) mice show mild increases in anxiety on some measures. The pattern of results shows both similarities and differences to those reported in hypomorphic NRG1 mice, and may be relevant for interpreting the increased NRG1 type I expression observed in schizophrenia.
Collapse
Affiliation(s)
- Inga H. Deakin
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford OX3 7JX, UK
| | - Amanda J. Law
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford OX3 7JX, UK
- Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Peter L. Oliver
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Markus H. Schwab
- Department of Neurogenetics, Max-Planck Institute of Experimental Medicine, Göttingen 37075, Germany
| | - Klaus Armin Nave
- Department of Neurogenetics, Max-Planck Institute of Experimental Medicine, Göttingen 37075, Germany
| | - Paul J. Harrison
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford OX3 7JX, UK
| | | |
Collapse
|
43
|
Abstract
Schizophrenia is a debilitating mental illness that affects 1% of the population worldwide. Although its molecular etiology remains unclear, recent advances in human psychiatric genetics have identified a large number of candidate genetic risk factors involved in schizophrenia. Modeling the disease in genetically tractable animals is thus a challenging but increasingly important task. In this review, I discuss the potential problems and perspectives associated with modeling schizophrenia in fruit flies, and briefly review the recent studies analyzing the molecular and cellular functions of Disrupted-In-Schizophrenia-1 (DISC1) in transgenic flies.
Collapse
Affiliation(s)
- Katsuo Furukubo-Tokunaga
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tennodai, Tsukuba, Japan.
| |
Collapse
|
44
|
Gaisler-Salomon I, Schobel SA, Small SA, Rayport S. How high-resolution basal-state functional imaging can guide the development of new pharmacotherapies for schizophrenia. Schizophr Bull 2009; 35:1037-44. [PMID: 19828591 PMCID: PMC2762634 DOI: 10.1093/schbul/sbp114] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We describe here a coordinated brain imaging and animal models approach in which we have shown that the hippocampal CA1 region is a principal node in schizophrenia pathogenesis and have identified a novel treatment approach to the disorder based on inhibition of glutamate release. To identify biomarkers, we have focused on the putative prodromal period, typically lasting a few years, preceding the first onset of psychosis. About one-third of a high-risk cohort followed prospectively for 2.5 years will progress to threshold psychosis, making it possible to perform a relatively short prospective study. We have utilized a technological development in functional imaging techniques in which we measure cerebral blood volume (CBV), which allows for interrogation of subregions of the brain in the basal state at submillimeter resolution. Measurements of CBV in schizophrenia as well as in high-risk or prodromal stages can then pinpoint brain subregions differentially targeted during the earliest stages of the disorder. Our data suggest that the CA1 subfield of the hippocampal formation is most consistently implicated across disease stages, identifying a putative biomarker suitable for guiding drug development. Our studies in transgenic mice mutant in the glutamate synthetic enzyme glutaminase support the hypothesis that CA1 hyperfunction is due to altered glutamatergic neurotransmission. As a proof of principle, the glutaminase-deficient mice suggest that pharmacotherapies that reduce glutamatergic neurotransmission in the CA1 subfield may be a uniquely effective therapeutic strategy in schizophrenia and preventative in prodromal stages of the disorder.
Collapse
Affiliation(s)
- Inna Gaisler-Salomon
- Department of Psychiatry, Columbia University, New York, NY 10032,Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, NY 10032
| | - Scott A. Schobel
- Department of Psychiatry, Columbia University, New York, NY 10032,Department of Translational Imaging, NYS Psychiatric Institute, New York, NY 10032
| | - Scott A. Small
- Department of Neurology, Columbia University, New York, NY 10032
| | - Stephen Rayport
- Department of Psychiatry, Columbia University, New York, NY 10032,Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, NY 10032,To whom correspondence should be addressed; tel: 212-543-5641, fax: 212-504-3135, e-mail:
| |
Collapse
|
45
|
Kirby BP, Waddington JL, O'Tuathaigh CMP. Advancing a functional genomics for schizophrenia: psychopathological and cognitive phenotypes in mutants with gene disruption. Brain Res Bull 2009; 83:162-76. [PMID: 19800398 DOI: 10.1016/j.brainresbull.2009.09.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2009] [Revised: 08/18/2009] [Accepted: 09/21/2009] [Indexed: 12/19/2022]
Abstract
Schizophrenia is a complex, heritable psychotic disorder in which numerous genes and environmental adversities appear to interact in determining disease phenotype. In addition to genes regulating putative pathophysiological mechanisms, a new generation of molecular studies has indicated numerous candidate genes to be associated with risk for schizophrenia. The present review focuses on studies in mice mutant for genes associated with putative pathophysiological mechanisms and candidate risk genes for the disorder. It seeks to evaluate the extent to which each mutation of a schizophrenia-related gene accurately models multiple aspects of the schizophrenia phenotype or more circumscribed, distinct endophenotypes in terms of psychopathology and pathobiology; in doing so, it places particular emphasis on positive symptoms, negative symptoms and cognitive dysfunction. To further this goal, it juxtaposes continually evolving mutant genomics with emergent clinical genomic studies. Opportunities and challenges associated with the use of such mutants, including diagnostic specificity and the translational barrier associated with modelling schizophrenia, are discussed. The potential value of genetic models for exploring gene-gene and gene-environment interactions relating to schizophrenia is highlighted. Elucidation of the contribution of genetic variation to specific symptom clusters and underlying aspects of pathobiology will have important implications for identifying treatments that target distinct domains of psychopathology and dysfunction on an individual patient basis.
Collapse
Affiliation(s)
- Brian P Kirby
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | | | | |
Collapse
|
46
|
Desbonnet L, Waddington JL, Tuathaigh CMPO. Mice mutant for genes associated with schizophrenia: common phenotype or distinct endophenotypes? Behav Brain Res 2009; 204:258-73. [PMID: 19728400 DOI: 10.1016/j.bbr.2009.04.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Schizophrenia is a complex neuropsychiatric disorder whose etiology involves a mixture of genetic and environmental factors. By virtue of this complexity, schizophrenia is a field of research in which a number of key technologies converge: in particular, identification of putative susceptibility genes through association studies in clinical populations leads to investigation of the behavioural roles of these genes by targeted manipulation in mice and their phenotypic characterisation ('gene-driven' approach); in a complementary manner, identification of putative pathophysiological processes and therapeutic pathways leads to investigation of behavioural phenotype in mice mutant for genes regulating such processes and pathways ('phenotype-driven' approach). As several susceptibility genes for schizophrenia and numerous genes implicated in the pathophysiology of schizophrenia have now been genetically manipulated in mice, it is timely to consider the roles of these genes in abnormal brain development and the ontogeny of putative schizophrenia-like phenotypes. The aim of this review is to outline existing knowledge from mutant studies concerning the contribution of these genes to the development of a common schizophrenia phenotype vis-à-vis discrete schizophrenia endophenotypes. Emphasis is also placed on the importance of studying gene x environment and gene x gene interactions, as well as addressing methodological issues related to genetic modelling and phenotyping strategies.
Collapse
Affiliation(s)
- Lieve Desbonnet
- Molecular & Cellular Therapeutics, Royal College of Surgeons in Ireland, St. Stephen's Green, Dublin 2, Ireland
| | | | | |
Collapse
|
47
|
Takao K, Miyakawa T. Intrauterine environment-genome interaction and children's development (4): Brain-behavior phenotypying of genetically-engineered mice using a comprehensive behavioral test battery on research of neuropsychiatric disorders. J Toxicol Sci 2009; 34 Suppl 2:SP293-305. [PMID: 19571483 DOI: 10.2131/jts.34.sp293] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Despite massive research efforts, the exact pathogenesis and pathophysiology of psychiatric disorders, such as schizophrenia and bipolar disorder, remain largely unknown. Animal models can serve as essential tools for investigating the etiology and treatment of such disorders. Some mutant mouse strains were found to exhibit behavioral abnormalities reminiscent of human psychiatric disorders. Here we outline our unique approach of extrapolating findings in mice to humans, and present studies on alpha-CaMKII heterozygous knockout (alpha-CaMKII+/-) mice as examples. Alpha-CaMKII+/- mice have profoundly dysregulated behavior and impaired neuronal development in the dentate gyrus (DG). The behavioral abnormalities include a severe working memory deficit and an exaggerated infradian rhythm, which are similar to symptoms seen in schizophrenia, bipolar mood disorder and other psychiatric disorders. By conducting a series of experiments, we discovered that almost all the neurons in the mutant DG were very similar to the immature DG neurons of normal rodents. In other words, alpha-CaMKII+/- mice have an "immature DG". We proposed that an "immature DG" in adulthood might induce alterations in behavior and serve as a promising candidate endophenotype of schizophrenia and other human psychiatric disorders. The impact of a large-scale mouse phenotyping on studies of psychiatric disorders and the potential utility of an "animal-model-array" of psychiatric disorders for the development of suitable therapeutic agents is also discussed.
Collapse
Affiliation(s)
- Keizo Takao
- Division of Systems Medicine, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | | |
Collapse
|
48
|
Abstract
Maternal infection during pregnancy increases the risk of schizophrenia and other brain disorders of neurodevelopmental origin in the offspring. A multitude of infectious agents seem to be involved in this association. Therefore, it has been proposed that factors common to the immune response to a wide variety of bacterial and viral pathogens may be the critical link between prenatal infection and postnatal brain and behavioral pathology. More specifically, it has been suggested that the maternal induction of pro-inflammatory cytokines may mediate the neurodevelopmental effects of maternal infections. Here, we review recent findings from in vitro and in vivo investigations supporting this hypothesis and further emphasize the influence of enhanced anti-inflammatory cytokine signaling on early brain development. Disruption of the fetal brain balance between pro- and anti-inflammatory cytokine signaling may thus represent a key mechanism involved in the precipitation of schizophrenia-related pathology following prenatal maternal infection and innate immune imbalances.
Collapse
Affiliation(s)
| | - Joram Feldon
- To whom correspondence should be addressed; tel: +41 44 655 7448, fax: 41 44 655 7203, e-mail:
| | | |
Collapse
|
49
|
Inta D, Monyer H, Sprengel R, Meyer-Lindenberg A, Gass P. Mice with genetically altered glutamate receptors as models of schizophrenia: a comprehensive review. Neurosci Biobehav Rev 2009; 34:285-94. [PMID: 19651155 DOI: 10.1016/j.neubiorev.2009.07.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2009] [Revised: 07/24/2009] [Accepted: 07/24/2009] [Indexed: 01/28/2023]
Abstract
Recent clinical evidence for the effectiveness of new antipsychotic drugs that specifically target glutamate receptors has rekindled interest in the glutamatergic system regarding pathophysiology and treatment of schizophrenia. The glutamatergic hypothesis of schizophrenia was triggered by the clinical/behavioural observation that NMDA receptor antagonists can induce psychosis in humans and abnormal behaviour with schizophrenia-like symptoms in animals. Initial models focused on NMDA receptor hypofunction as a potential pathogenetic mechanism. More recent genetic and pharmacological studies revealed that malfunction of other components of the glutamatergic system might also be relevant in explaining specific symptoms of this complex disease. Here, we review mutant mouse models with relevance for schizophrenia. These rodent models, in which specific glutamate receptor subtypes or various components of their intracellular transduction machinery are genetically altered, permit a detailed dissection of the contribution of different components of the glutamate system in inducing schizophrenia-like behaviours. They may provide insight into the pathophysiology of schizophrenia and prove useful in the development of new therapeutics.
Collapse
Affiliation(s)
- Dragos Inta
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, J5, 68159 Mannheim, Germany.
| | | | | | | | | |
Collapse
|
50
|
Kalirin regulates cortical spine morphogenesis and disease-related behavioral phenotypes. Proc Natl Acad Sci U S A 2009; 106:13058-63. [PMID: 19625617 DOI: 10.1073/pnas.0904636106] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dendritic spine morphogenesis contributes to brain function, cognition, and behavior, and is altered in psychiatric disorders. Kalirin is a brain-specific guanine-nucleotide exchange factor (GEF) for Rac-like GTPases and is a key regulator of spine morphogenesis. Here, we show that KALRN-knockout mice have specific reductions in cortical, but not hippocampal, Rac1 signaling and spine density, and exhibit reduced cortical glutamatergic transmission. These mice exhibit robust deficits in working memory, sociability, and prepulse inhibition, paralleled by locomotor hyperactivity reversible by clozapine in a kalirin-dependent manner. Several of these deficits are delayed and age-dependent. Our study thus links spine morphogenic signaling with age-dependent, delayed, disease-related phenotypes, including cognitive dysfunction.
Collapse
|