1
|
Park J, Shimbo H, Tamura S, Tomoda T, Hikida T, Okado H, Hirai S. Impact of feeding age on cognitive impairment in mice with Disrupted-In-Schizophrenia 1 (Disc1) mutation under a high sucrose diet. Behav Brain Res 2025; 476:115291. [PMID: 39401692 DOI: 10.1016/j.bbr.2024.115291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/17/2024]
Abstract
A combination of genetic predisposition and environmental factors contributes to the development of psychiatric disorders such as schizophrenia, bipolar disorder and major depressive disorder. Previous studies using mouse models suggested that prolonged high sucrose intake during puberty can serve as an environmental risk factor for the onset of psychiatric disorders. However, the impact of both the duration and timing of high sucrose consumption during different developmental stages on pathogenesis remains poorly defined. We therefore investigated the effects of a long-term high sucrose diet on cognitive deficit, a core symptom of psychiatric disorders, using Disrupted-in-Schizophrenia 1 locus-impairment heterozygous mutant (Disc1het) mice as a model for genetic predisposition. First, Disc1het mice and their littermate control (WT) were fed either a high sucrose diet or a control starch diet for nine weeks starting at weaning (postnatal day 24), and tested for cognitive performance in the object location test (OLT) and the novel object recognition test (NORT) (assessing spatial and recognition memory, respectively). Only Disc1het mice on a high sucrose diet displayed deficits in OLT (p < 0.0001), demonstrating impaired hippocampus-dependent spatial memory. This behavioral abnormality was accompanied by a decreased proportion of the high parvalbumin-expressing interneurons (High-PV neurons) in the ventral hippocampus, a cell type that regulates neural activity and a variety of learning and memory processes such as spatial and working memory. We further explored the critical developmental period for high sucrose intake to cause cognitive deficits in adulthood by comparing specific feeding periods during puberty (P24-P65) and post-puberty (P65-P90). Compared to those on a standard chow diet, high sucrose intake caused deficits in spatial memory in both WT and Disc1het mice, with more pronounced effects in Disc1het mice. In particular, Disc1het mice on a sucrose diet during adolescence showed more pronounced cognitive deficit than those fed after adolescence. Our results suggest that adolescence is particularly vulnerable to nutritional environmental risk factors, and that high sucrose consumption may cause hippocampus-dependent memory deficits via decreased High-PV interneuron function when combined with Disc1-related genetic predisposition.
Collapse
Affiliation(s)
- Jonghyuk Park
- Metabolic Regulation Group, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Japan
| | - Hiroko Shimbo
- Metabolic Regulation Group, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Japan; Clinical Research Institute, Kanagawa Children's Medical Center, Japan
| | - Shoko Tamura
- Metabolic Regulation Group, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Japan
| | - Toshifumi Tomoda
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, University of Toronto, Canada
| | - Takatoshi Hikida
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Japan
| | - Haruo Okado
- Metabolic Regulation Group, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Japan
| | - Shinobu Hirai
- Metabolic Regulation Group, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Japan.
| |
Collapse
|
2
|
Yang TT, Wei R, Jin FF, Yu W, Zhang F, Peng Y, Zhang SJ, Qi SH, Liu JR. Dexmedetomidine Alleviates the Long-Term Neurodevelopmental Toxicity Induced by Sevoflurane in the Developing Brain. Dev Neurosci 2024:1. [PMID: 39433029 DOI: 10.1159/000542114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
INTRODUCTION Sevoflurane is an extensively used anesthetic for pediatric patients; however, numerous studies showed that sevoflurane (SEVO) may cause long-term neurodevelopmental toxicity. Dexmedetomidine (DEX) has been shown to be protective against SEVO-induced neurotoxicity, but the mechanism remains unclear. The effects and mechanisms of different DEX administration routes on SEVO-induced neurotoxicity and long-term cognitive defects were determined and further investigated the role of sex in these processes. METHODS Male and female Sprague Dawley rats at postnatal day 7 (PND7) received an intraperitoneal injection of DEX (10 μg/kg) before or after exposure to 2.5% SEVO for 6 h, or before and after SEVO exposure. The respiratory and mortality rates of the pups were recorded during anesthesia. Neuroapoptosis was evaluated by TdT-mediated dUTP nick-end labeling staining. Immunohistochemistry and immunofluorescence were employed to detect the expression of caspase-3 in neuronal cells and neurons. The expression of GSK-3β and DISC1 was determined by Western blotting or RT-qPCR. Morris water maze (MWM) test was used to evaluate the learning and memory ability of rats until they were 3 weeks and 5 weeks old. RESULTS Compared with the control group, exposure to 2.5% SEVO resulted in increased neuroapoptosis and decreased the expression of DISC1 at levels of mRNA and protein and phosphorylated GSK-3β in the developing brain. SEVO exposure during critical neurodevelopmental periods could cause persistent cognitive defects in adolescent male and female rats and inhibited DISC1 and phosphorylated GSK-3β protein expression. The neurotoxic impacts of SEVO were lessened by the administration of DEX (10 μg/kg) before or after exposure. CONCLUSION Our findings suggest that DEX (10 μg/kg) mitigates the neurotoxic effects of SEVO on the developing rat brain as well as postnatal cognitive defects by regulating the DISC1/GSK-3β signaling.
Collapse
Affiliation(s)
- Ting-Ting Yang
- The Department of Clinical Laboratory, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China,
| | - Ran Wei
- The Department of Clinical Laboratory, Gaochun People's Hospital, Nanjing, China
| | - Fei-Fei Jin
- The Department of Clinical Laboratory, Yuhuangding Hospital, Yantai, China
| | - Wei Yu
- The Department of Anesthesiology, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fang Zhang
- The Center of Prenatal Diagnosis, Dongguan Maternal and Child Health Care Hospital, Dongguan, China
| | - Yu Peng
- The Department of Clinical Laboratory, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shu-Jun Zhang
- The Department of Pathology, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Si-Hua Qi
- The Department of Anesthesiology, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jia-Ren Liu
- The Department of Clinical Laboratory, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
3
|
Lv Y, Wen L, Hu WJ, Deng C, Ren HW, Bao YN, Su BW, Gao P, Man ZY, Luo YY, Li CJ, Xiang ZX, Wang B, Luan ZL. Schizophrenia in the genetic era: a review from development history, clinical features and genomic research approaches to insights of susceptibility genes. Metab Brain Dis 2024; 39:147-171. [PMID: 37542622 DOI: 10.1007/s11011-023-01271-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/27/2023] [Indexed: 08/07/2023]
Abstract
Schizophrenia is a devastating neuropsychiatric disorder affecting 1% of the world population and ranks as one of the disorders providing the most severe burden for society. Schizophrenia etiology remains obscure involving multi-risk factors, such as genetic, environmental, nutritional, and developmental factors. Complex interactions of genetic and environmental factors have been implicated in the etiology of schizophrenia. This review provides an overview of the historical origins, pathophysiological mechanisms, diagnosis, clinical symptoms and corresponding treatment of schizophrenia. In addition, as schizophrenia is a polygenic, genetic disorder caused by the combined action of multiple micro-effective genes, we further detail several approaches, such as candidate gene association study (CGAS) and genome-wide association study (GWAS), which are commonly used in schizophrenia genomics studies. A number of GWASs about schizophrenia have been performed with the hope to identify novel, consistent and influential risk genetic factors. Finally, some schizophrenia susceptibility genes have been identified and reported in recent years and their biological functions are also listed. This review may serve as a summary of past research on schizophrenia genomics and susceptibility genes (NRG1, DISC1, RELN, BDNF, MSI2), which may point the way to future schizophrenia genetics research. In addition, depending on the above discovery of susceptibility genes and their exact function, the development and application of antipsychotic drugs will be promoted in the future.
Collapse
Affiliation(s)
- Ye Lv
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Lin Wen
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Wen-Juan Hu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Chong Deng
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| | - Hui-Wen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ya-Nan Bao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Bo-Wei Su
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ping Gao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Zi-Yue Man
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Yi-Yang Luo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Cheng-Jie Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Zhi-Xin Xiang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Bing Wang
- Department of Endocrinology and Metabolism, The Central hospital of Dalian University of Technology, Dalian, 116000, China.
| | - Zhi-Lin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
4
|
Dysregulated Signaling at Postsynaptic Density: A Systematic Review and Translational Appraisal for the Pathophysiology, Clinics, and Antipsychotics' Treatment of Schizophrenia. Cells 2023; 12:cells12040574. [PMID: 36831241 PMCID: PMC9954794 DOI: 10.3390/cells12040574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Emerging evidence from genomics, post-mortem, and preclinical studies point to a potential dysregulation of molecular signaling at postsynaptic density (PSD) in schizophrenia pathophysiology. The PSD that identifies the archetypal asymmetric synapse is a structure of approximately 300 nm in diameter, localized behind the neuronal membrane in the glutamatergic synapse, and constituted by more than 1000 proteins, including receptors, adaptors, kinases, and scaffold proteins. Furthermore, using FASS (fluorescence-activated synaptosome sorting) techniques, glutamatergic synaptosomes were isolated at around 70 nm, where the receptors anchored to the PSD proteins can diffuse laterally along the PSD and were stabilized by scaffold proteins in nanodomains of 50-80 nm at a distance of 20-40 nm creating "nanocolumns" within the synaptic button. In this context, PSD was envisioned as a multimodal hub integrating multiple signaling-related intracellular functions. Dysfunctions of glutamate signaling have been postulated in schizophrenia, starting from the glutamate receptor's interaction with scaffolding proteins involved in the N-methyl-D-aspartate receptor (NMDAR). Despite the emerging role of PSD proteins in behavioral disorders, there is currently no systematic review that integrates preclinical and clinical findings addressing dysregulated PSD signaling and translational implications for antipsychotic treatment in the aberrant postsynaptic function context. Here we reviewed a critical appraisal of the role of dysregulated PSD proteins signaling in the pathophysiology of schizophrenia, discussing how antipsychotics may affect PSD structures and synaptic plasticity in brain regions relevant to psychosis.
Collapse
|
5
|
Kumar S, Mehan S, Narula AS. Therapeutic modulation of JAK-STAT, mTOR, and PPAR-γ signaling in neurological dysfunctions. J Mol Med (Berl) 2023; 101:9-49. [PMID: 36478124 DOI: 10.1007/s00109-022-02272-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/10/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
The cytokine-activated Janus kinase (JAK)-signal transducer and activator of transcription (STAT) cascade is a pleiotropic pathway that involves receptor subunit multimerization. The mammalian target of rapamycin (mTOR) is a ubiquitously expressed serine-threonine kinase that perceives and integrates a variety of intracellular and environmental stimuli to regulate essential activities such as cell development and metabolism. Peroxisome proliferator-activated receptor-gamma (PPARγ) is a prototypical metabolic nuclear receptor involved in neural differentiation and axon polarity. The JAK-STAT, mTOR, and PPARγ signaling pathways serve as a highly conserved signaling hub that coordinates neuronal activity and brain development. Additionally, overactivation of JAK/STAT, mTOR, and inhibition of PPARγ signaling have been linked to various neurocomplications, including neuroinflammation, apoptosis, and oxidative stress. Emerging research suggests that even minor disruptions in these cellular and molecular processes can have significant consequences manifested as neurological and neuropsychiatric diseases. Of interest, target modulators have been proven to alleviate neuronal complications associated with acute and chronic neurological deficits. This research-based review explores the therapeutic role of JAK-STAT, mTOR, and PPARγ signaling modulators in preventing neuronal dysfunctions in preclinical and clinical investigations.
Collapse
Affiliation(s)
- Sumit Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India.
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC, 27516, USA
| |
Collapse
|
6
|
de Sousa TR, Dt C, Novais F. Exploring the Hypothesis of a Schizophrenia and Bipolar Disorder Continuum: Biological, Genetic and Pharmacologic Data. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:161-171. [PMID: 34477537 DOI: 10.2174/1871527320666210902164235] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/19/2021] [Accepted: 08/08/2021] [Indexed: 12/16/2022]
Abstract
Present time nosology has its roots in Kraepelin's demarcation of schizophrenia and bipolar disorder. However, accumulating evidence has shed light on several commonalities between the two disorders, and some authors have advocated for the consideration of a disease continuum. Here, we review previous genetic, biological and pharmacological findings that provide the basis for this conceptualization. There is a cross-disease heritability, and they share single-nucleotide polymorphisms in some common genes. EEG and imaging patterns have a number of similarities, namely reduced white matter integrity and abnormal connectivity. Dopamine, serotonin, GABA and glutamate systems have dysfunctional features, some of which are identical among the disorders. Finally, cellular calcium regulation and mitochondrial function are, also, impaired in the two.
Collapse
Affiliation(s)
- Teresa Reynolds de Sousa
- Department of Neurosciences and Mental Health, Centro Hospitalar Universitário Lisboa Norte (CHULN), Hospital de Santa Maria, Lisbon, Portugal
| | - Correia Dt
- Department of Neurosciences and Mental Health, Centro Hospitalar Universitário Lisboa Norte (CHULN), Hospital de Santa Maria, Lisbon, Portugal
- Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
- ISAMB - Instituto de Saúde Ambiental, Lisboa, Portugal
| | - Filipa Novais
- Department of Neurosciences and Mental Health, Centro Hospitalar Universitário Lisboa Norte (CHULN), Hospital de Santa Maria, Lisbon, Portugal
- Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
- ISAMB - Instituto de Saúde Ambiental, Lisboa, Portugal
| |
Collapse
|
7
|
Teal LB, Ingram SM, Bubser M, McClure E, Jones CK. The Evolving Role of Animal Models in the Discovery and Development of Novel Treatments for Psychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2023; 30:37-99. [PMID: 36928846 DOI: 10.1007/978-3-031-21054-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Historically, animal models have been routinely used in the characterization of novel chemical entities (NCEs) for various psychiatric disorders. Animal models have been essential in the in vivo validation of novel drug targets, establishment of lead compound pharmacokinetic to pharmacodynamic relationships, optimization of lead compounds through preclinical candidate selection, and development of translational measures of target occupancy and functional target engagement. Yet, with decades of multiple NCE failures in Phase II and III efficacy trials for different psychiatric disorders, the utility and value of animal models in the drug discovery process have come under intense scrutiny along with the widespread withdrawal of the pharmaceutical industry from psychiatric drug discovery. More recently, the development and utilization of animal models for the discovery of psychiatric NCEs has undergone a dynamic evolution with the application of the Research Domain Criteria (RDoC) framework for better design of preclinical to clinical translational studies combined with innovative genetic, neural circuitry-based, and automated testing technologies. In this chapter, the authors will discuss this evolving role of animal models for improving the different stages of the discovery and development in the identification of next generation treatments for psychiatric disorders.
Collapse
Affiliation(s)
- Laura B Teal
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Shalonda M Ingram
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Michael Bubser
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Elliott McClure
- College of Pharmacy and Health Sciences, Lipscomb University, Nashville, TN, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
8
|
Combining fMRI and DISC1 gene haplotypes to understand working memory-related brain activity in schizophrenia. Sci Rep 2022; 12:7351. [PMID: 35513527 PMCID: PMC9072540 DOI: 10.1038/s41598-022-10660-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 04/11/2022] [Indexed: 11/30/2022] Open
Abstract
The DISC1 gene is one of the most relevant susceptibility genes for psychosis. However, the complex genetic landscape of this locus, which includes protective and risk variants in interaction, may have hindered consistent conclusions on how DISC1 contributes to schizophrenia (SZ) liability. Analysis from haplotype approaches and brain-based phenotypes can contribute to understanding DISC1 role in the neurobiology of this disorder. We assessed the brain correlates of DISC1 haplotypes associated with SZ through a functional neuroimaging genetics approach. First, we tested the association of two DISC1 haplotypes, the HEP1 (rs6675281-1000731-rs999710) and the HEP3 (rs151229-rs3738401), with the risk for SZ in a sample of 138 healthy subjects (HS) and 238 patients. This approach allowed the identification of three haplotypes associated with SZ (HEP1-CTG, HEP3-GA and HEP3-AA). Second, we explored whether these haplotypes exerted differential effects on n-back associated brain activity in a subsample of 70 HS compared to 70 patients (diagnosis × haplotype interaction effect). These analyses evidenced that HEP3-GA and HEP3-AA modulated working memory functional response conditional to the health/disease status in the cuneus, precuneus, middle cingulate cortex and the ventrolateral and dorsolateral prefrontal cortices. Our results are the first to show a diagnosis-based effect of DISC1 haplotypes on working memory-related brain activity, emphasising its role in SZ.
Collapse
|
9
|
Wang J, Su P, Yang J, Xu L, Yuan A, Li C, Zhang T, Dong F, Zhou J, Samsom J, Wong AH, Liu F. The D2R-DISC1 protein complex and associated proteins are altered in schizophrenia and normalized with antipsychotic treatment. J Psychiatry Neurosci 2022; 47:E134-E147. [PMID: 35361701 PMCID: PMC8979657 DOI: 10.1503/jpn.210145] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/30/2021] [Accepted: 01/24/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND For decades, the dopamine D2 receptor (D2R) has been known as the main target of antipsychotic medications, but the mechanism for antipsychotic effects beyond this pharmacological target remains unclear. Disrupted-in-schizophrenia 1 (DISC1) is a gene implicated in the etiology of schizophrenia, and we have found elevated levels of the D2R-DISC1 complex in the postmortem brain tissue of patients with schizophrenia. METHODS We used coimmunoprecipitation to measure D2R-DISC1 complex levels in peripheral blood samples from patients with schizophrenia and unaffected controls in 3 cohorts (including males and females) from different hospitals. We also used label-free mass spectrometry to conduct proteomic analysis of these samples. RESULTS Levels of the D2R-DISC1 complex were elevated in the peripheral blood samples of patients with schizophrenia from 3 independent cohorts, and were normalized with antipsychotic treatment. Proteomic analysis of the blood samples from patients with high D2R-DISC1 complex levels that were normalized with antipsychotic treatment revealed a number of altered proteins and pathways associated with D2R, DISC1 and the D2R-DISC1 complex. We identified additional proteins and pathways that were associated with antipsychotic treatment in schizophrenia, and that may also be novel targets for schizophrenia treatment. LIMITATIONS Sample sizes were relatively small, but were sufficient to detect associations between D2R-DISC1 levels, schizophrenia and treatment response. The relevance of leukocyte changes to the symptoms of schizophrenia is unknown. The coimmunoprecipitation lanes included several nonspecific bands. CONCLUSION Levels of the D2R-DISC1 complex were elevated in patients with schizophrenia and reduced with antipsychotic treatment. This finding reinforces the independent role of each protein in schizophrenia. Our results enhanced our understanding of the molecular pathways involved in schizophrenia and in antipsychotic medications, and identified novel potential molecular targets for treating schizophrenia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Fang Liu
- From the Shanghai Mental Health Centre, Shanghai Jiaotong University, School of Medicine, Shanghai, China (Wang, Xu, Yuan, Li, Zhang, Liu); the Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ont. (Su, Samsom, Wong, Liu); the National Clinical Research Centre for Mental Disorders, Beijing AnDing Hospital, Capital Medical University, Beijing, China (Yang, Dong, Zhou); the Department of Pharmacology, University of Toronto, Toronto, Ont. (Samsom, Wong); the Institute of Medical Science, University of Toronto, Toronto, Ont. (Wong, Liu); the Department of Psychiatry, University of Toronto, Toronto, Ont. (Wong, Liu); the Department of Physiology, University of Toronto, Toronto, Ont. (Liu)
| |
Collapse
|
10
|
Mutations in DISC1 alter IP 3R and voltage-gated Ca 2+ channel functioning, implications for major mental illness. Neuronal Signal 2021; 5:NS20180122. [PMID: 34956649 PMCID: PMC8663806 DOI: 10.1042/ns20180122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/26/2021] [Accepted: 11/08/2021] [Indexed: 12/16/2022] Open
Abstract
Disrupted in Schizophrenia 1 (DISC1) participates in a wide variety of
developmental processes of central neurons. It also serves critical roles that
underlie cognitive functioning in adult central neurons. Here we summarize
DISC1’s general properties and discuss its use as a model system for
understanding major mental illnesses (MMIs). We then discuss the cellular
actions of DISC1 that involve or regulate Ca2+ signaling in adult
central neurons. In particular, we focus on the tethering role DISC1 plays in
transporting RNA particles containing Ca2+ channel subunit RNAs,
including IP3R1, CACNA1C and CACNA2D1, and in transporting mitochondria into
dendritic and axonal processes. We also review DISC1’s role in modulating
IP3R1 activity within mitochondria-associated ER membrane (MAM).
Finally, we discuss DISC1-glycogen synthase kinase 3β (GSK3β)
signaling that regulates functional expression of voltage-gated Ca2+
channels (VGCCs) at central synapses. In each case, DISC1 regulates the movement
of molecules that impact Ca2+ signaling in neurons.
Collapse
|
11
|
Human iPSC-Derived Glia as a Tool for Neuropsychiatric Research and Drug Development. Int J Mol Sci 2021; 22:ijms221910254. [PMID: 34638595 PMCID: PMC8508580 DOI: 10.3390/ijms221910254] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 12/19/2022] Open
Abstract
Neuropsychiatric disorders such as schizophrenia or autism spectrum disorder represent a leading and growing burden on worldwide mental health. Fundamental lack in understanding the underlying pathobiology compromises efficient drug development despite the immense medical need. So far, antipsychotic drugs reduce symptom severity and enhance quality of life, but there is no cure available. On the molecular level, schizophrenia and autism spectrum disorders correlate with compromised neuronal phenotypes. There is increasing evidence that aberrant neuroinflammatory responses of glial cells account for synaptic pathologies through deregulated communication and reciprocal modulation. Consequently, microglia and astrocytes emerge as central targets for anti-inflammatory treatment to preserve organization and homeostasis of the central nervous system. Studying the impact of neuroinflammation in the context of neuropsychiatric disorders is, however, limited by the lack of relevant human cellular test systems that are able to represent the dynamic cellular processes and molecular changes observed in human tissue. Today, patient-derived induced pluripotent stem cells offer the opportunity to study neuroinflammatory mechanisms in vitro that comprise the genetic background of affected patients. In this review, we summarize the major findings of iPSC-based microglia and astrocyte research in the context of neuropsychiatric diseases and highlight the benefit of 2D and 3D co-culture models for the generation of efficient in vitro models for target screening and drug development.
Collapse
|
12
|
Priol AC, Denis L, Boulanger G, Thépaut M, Geoffray MM, Tordjman S. Detection of Morphological Abnormalities in Schizophrenia: An Important Step to Identify Associated Genetic Disorders or Etiologic Subtypes. Int J Mol Sci 2021; 22:ijms22179464. [PMID: 34502372 PMCID: PMC8430486 DOI: 10.3390/ijms22179464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/06/2021] [Indexed: 12/20/2022] Open
Abstract
Current research suggests that alterations in neurodevelopmental processes, involving gene X environment interactions during key stages of brain development (prenatal period and adolescence), are a major risk for schizophrenia. First, epidemiological studies supporting a genetic contribution to schizophrenia are presented in this article, including family, twin, and adoption studies. Then, an extensive literature review on genetic disorders associated with schizophrenia is reviewed. These epidemiological findings and clinical observations led researchers to conduct studies on genetic associations in schizophrenia, and more specifically on genomics (CNV: copy-number variant, and SNP: single nucleotide polymorphism). The main structural (CNV) and sequence (SNP) variants found in individuals with schizophrenia are reported here. Evidence of genetic contributions to schizophrenia and current knowledge on genetic syndromes associated with this psychiatric disorder highlight the importance of a clinical genetic examination to detect minor physical anomalies in individuals with ultra-high risk of schizophrenia. Several dysmorphic features have been described in schizophrenia, especially in early onset schizophrenia, and can be viewed as neurodevelopmental markers of vulnerability. Early detection of individuals with neurodevelopmental abnormalities is a fundamental issue to develop prevention and diagnostic strategies, therapeutic intervention and follow-up, and to ascertain better the underlying mechanisms involved in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Anne-Clémence Priol
- Pôle Hospitalo-Universitaire de Psychiatrie de l’Enfant et de l’Adolescent (PHUPEA), Centre Hospitalier Guillaume Régnier, University of Rennes 1, 35000 Rennes, France; (L.D.); (G.B.); (M.T.)
- Correspondence: (A.-C.P.); (S.T.); Tel.: +33-2-99-51-06-04 (A.-C.P. & S.T.); Fax: +33-2-99-32-46-98 (A.-C.P. & S.T.)
| | - Laure Denis
- Pôle Hospitalo-Universitaire de Psychiatrie de l’Enfant et de l’Adolescent (PHUPEA), Centre Hospitalier Guillaume Régnier, University of Rennes 1, 35000 Rennes, France; (L.D.); (G.B.); (M.T.)
| | - Gaella Boulanger
- Pôle Hospitalo-Universitaire de Psychiatrie de l’Enfant et de l’Adolescent (PHUPEA), Centre Hospitalier Guillaume Régnier, University of Rennes 1, 35000 Rennes, France; (L.D.); (G.B.); (M.T.)
| | - Mathieu Thépaut
- Pôle Hospitalo-Universitaire de Psychiatrie de l’Enfant et de l’Adolescent (PHUPEA), Centre Hospitalier Guillaume Régnier, University of Rennes 1, 35000 Rennes, France; (L.D.); (G.B.); (M.T.)
| | - Marie-Maude Geoffray
- Department of Child and Adolescent Psychiatry, Centre Hospitalier Le Vinatier, 69500 Bron, France;
| | - Sylvie Tordjman
- Pôle Hospitalo-Universitaire de Psychiatrie de l’Enfant et de l’Adolescent (PHUPEA), Centre Hospitalier Guillaume Régnier, University of Rennes 1, 35000 Rennes, France; (L.D.); (G.B.); (M.T.)
- CIC (Clinical Investigation Center) 1414 Inserm, Centre Hospitalier Universitaire (CHU) de Rennes, University of Rennes 1, 35033 Rennes, France
- Integrative Neuroscience and Cognition Center (INCC), CNRS UMR 8002, University of Paris, 75006 Paris, France
- Correspondence: (A.-C.P.); (S.T.); Tel.: +33-2-99-51-06-04 (A.-C.P. & S.T.); Fax: +33-2-99-32-46-98 (A.-C.P. & S.T.)
| |
Collapse
|
13
|
den Hoed J, Devaraju K, Fisher SE. Molecular networks of the FOXP2 transcription factor in the brain. EMBO Rep 2021; 22:e52803. [PMID: 34260143 PMCID: PMC8339667 DOI: 10.15252/embr.202152803] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/19/2021] [Accepted: 06/23/2021] [Indexed: 01/06/2023] Open
Abstract
The discovery of the FOXP2 transcription factor, and its implication in a rare severe human speech and language disorder, has led to two decades of empirical studies focused on uncovering its roles in the brain using a range of in vitro and in vivo methods. Here, we discuss what we have learned about the regulation of FOXP2, its downstream effectors, and its modes of action as a transcription factor in brain development and function, providing an integrated overview of what is currently known about the critical molecular networks.
Collapse
Affiliation(s)
- Joery den Hoed
- Language and Genetics DepartmentMax Planck Institute for PsycholinguisticsNijmegenThe Netherlands
- International Max Planck Research School for Language SciencesMax Planck Institute for PsycholinguisticsNijmegenThe Netherlands
| | - Karthikeyan Devaraju
- Language and Genetics DepartmentMax Planck Institute for PsycholinguisticsNijmegenThe Netherlands
| | - Simon E Fisher
- Language and Genetics DepartmentMax Planck Institute for PsycholinguisticsNijmegenThe Netherlands
- Donders Institute for Brain, Cognition and BehaviourRadboud UniversityNijmegenThe Netherlands
| |
Collapse
|
14
|
Kermath BA, Vanderplow AM, Cahill ME. Dysregulated Prefrontal Cortical RhoA Signal Transduction in Bipolar Disorder with Psychosis: New Implications for Disease Pathophysiology. Cereb Cortex 2021; 30:59-71. [PMID: 31220216 DOI: 10.1093/cercor/bhz070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
While research has identified alterations in dorsolateral prefrontal cortical function as a key factor to the etiology of bipolar disorder, few studies have uncovered robust changes in protein signal transduction pathways in this disorder. Given the direct relevance of protein-based expressional alterations to cellular functions and because many of the key regulatory mechanisms for the disease pathogenesis likely include alterations in protein activity rather than changes in expression alone, the identification of alterations in discrete signal transduction pathways in bipolar disorder would have broad implications for understanding the disease pathophysiology. As prior microarray data point to a previously unrecognized involvement of the RhoA network in bipolar disorder, here we investigate the protein expression and activity of key components of a RhoA signal transduction pathway in dorsolateral prefrontal cortical homogenates from subjects with bipolar disorder. The results of this investigation implicate overactivation of prefrontal cortical RhoA signaling in specific subtypes of bipolar disorder. The specificity of these findings is demonstrated by a lack of comparable changes in schizophrenia; however, our findings do identify convergence between both disorders at the level of activity-mediated actin cytoskeletal regulation. These findings have implications for understanding the altered cortical synaptic connectivity of bipolar disorder.
Collapse
Affiliation(s)
- Bailey A Kermath
- Department of Comparative Biosciences, University of Wisconsin at Madison, Madison, WI, USA
| | - Amanda M Vanderplow
- Department of Comparative Biosciences, University of Wisconsin at Madison, Madison, WI, USA
| | - Michael E Cahill
- Department of Comparative Biosciences, University of Wisconsin at Madison, Madison, WI, USA
| |
Collapse
|
15
|
Mapping Sex-Specific Neurodevelopmental Alterations in Neurite Density and Morphology in a Rat Genetic Model of Psychiatric Illness. eNeuro 2021; 8:ENEURO.0426-20.2020. [PMID: 33441401 PMCID: PMC7986540 DOI: 10.1523/eneuro.0426-20.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/19/2020] [Accepted: 12/28/2020] [Indexed: 11/21/2022] Open
Abstract
Neurite orientation dispersion and density imaging (NODDI) is an emerging magnetic resonance (MR) diffusion-weighted imaging (DWI) technique that permits non-invasive quantitative assessment of neurite density and morphology. NODDI has improved our ability to image neuronal microstructure over conventional techniques such as diffusion tensor imaging (DTI) and is particularly suited for studies of the developing brain as it can measure and characterize the dynamic changes occurring in dendrite cytoarchitecture that are critical to early brain development. Neurodevelopmental alterations to the diffusion tensor have been reported in psychiatric illness, but it remains unknown whether advanced DWI techniques such as NODDI are able to sensitively and specifically detect neurodevelopmental changes in brain microstructure beyond those provided by DTI. We show, in an extension of our previous work with a Disc1 svΔ2 rat genetic model of psychiatric illness, the enhanced sensitivity and specificity of NODDI to identify neurodevelopmental and sex-specific changes in brain microstructure that are otherwise difficult to observe with DTI and further corroborate observed changes in brain microstructure to differences in sex-specific systems-level animal behavior. Together, these findings inform the potential application and clinical translational utility of NODDI in studies of brain microstructure in psychiatric illness throughout neurodevelopment and further, the ability of advanced DWI methods such as NODDI to examine the role of biological sex and its influence on brain microstructure in psychiatric illness.
Collapse
|
16
|
Reiner BC, Doyle GA, Weller AE, Levinson RN, Rao AM, Davila Perea E, Namoglu E, Pigeon A, Arauco-Shapiro G, Weickert CS, Turecki G, Crist RC, Berrettini WH. Inherited L1 Retrotransposon Insertions Associated With Risk for Schizophrenia and Bipolar Disorder. SCHIZOPHRENIA BULLETIN OPEN 2021; 2:sgab031. [PMID: 34901866 PMCID: PMC8650070 DOI: 10.1093/schizbullopen/sgab031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Studies of the genetic heritability of schizophrenia and bipolar disorder examining single nucleotide polymorphisms (SNPs) and copy number variations have failed to explain a large portion of the genetic liability, resulting in substantial missing heritability. Long interspersed element 1 (L1) retrotransposons are a type of inherited polymorphic variant that may be associated with risk for schizophrenia and bipolar disorder. We performed REBELseq, a genome wide assay for L1 sequences, on DNA from male and female persons with schizophrenia and controls (n = 63 each) to identify inherited L1 insertions and validated priority insertions. L1 insertions of interest were genotyped in DNA from a replication cohort of persons with schizophrenia, bipolar disorder, and controls (n = 2268 each) to examine differences in carrier frequencies. We identified an inherited L1 insertion in ARHGAP24 and a quadallelic SNP (rs74169643) inside an L1 insertion in SNTG2 that are associated with risk for developing schizophrenia and bipolar disorder (all odds ratios ~1.2). Pathway analysis identified 15 gene ontologies that were differentially affected by L1 burden, including multiple ontologies related to glutamatergic signaling and immune function, which have been previously associated with schizophrenia. These findings provide further evidence supporting the role of inherited repetitive genetic elements in the heritability of psychiatric disorders.
Collapse
Affiliation(s)
- Benjamin C Reiner
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Glenn A Doyle
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew E Weller
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel N Levinson
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aditya M Rao
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emilie Davila Perea
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Esin Namoglu
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alicia Pigeon
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriella Arauco-Shapiro
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cyndi Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia & School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY, USA
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, Canada
| | - Richard C Crist
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wade H Berrettini
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
17
|
Trifu SC, Kohn B, Vlasie A, Patrichi BE. Genetics of schizophrenia (Review). Exp Ther Med 2020; 20:3462-3468. [PMID: 32905096 PMCID: PMC7465115 DOI: 10.3892/etm.2020.8973] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022] Open
Abstract
A comprehensive review of the body of genetic studies on schizophrenia seems even more daunting than the battle a psychiatrist wages daily in the office with her archenemy of a thousand faces. The following article reunites some genetic, epigenetic and environmental factors of schizophrenia from revered and vast studies in a chronological and progressive fashion. Twin studies set the basics of heritability and a particular study by Davis and Phelps considers the widely ignored influence of prenatal environment in the development of schizophrenia. Mostly ignited by linkage studies, candidate gene studies explore further by fine-mapping the hypothesized variants [mostly in the forms single nucleotide polymorphisms (SNPs) and less but with greater impact copy number variations (CNVs)] associated with the disease. Genome-wide association studies (GWAS) increase considerably the sample sizes and thus the validity of the results, while the next-generation sequencing (NGS) attain the highest yet unreplicated level of validity results.
Collapse
Affiliation(s)
- Simona Corina Trifu
- Department of Neurosciences, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Bianca Kohn
- Department of Psychiatry, ‘Prof. Dr. Alexandru Obregia’ Clinical Hospital of Psychiatry, 041914 Bucharest, Romania
| | - Andrei Vlasie
- Department of Psychiatry, ‘Prof. Dr. Alexandru Obregia’ Clinical Hospital of Psychiatry, 041914 Bucharest, Romania
| | - Bogdan-Eduard Patrichi
- Department of Psychiatry and Psychology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
18
|
Gong NJ, Dibb R, Pletnikov M, Benner E, Liu C. Imaging microstructure with diffusion and susceptibility MR: neuronal density correlation in Disrupted-in-Schizophrenia-1 mutant mice. NMR IN BIOMEDICINE 2020; 33:e4365. [PMID: 32627266 DOI: 10.1002/nbm.4365] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 05/23/2020] [Accepted: 06/09/2020] [Indexed: 06/11/2023]
Abstract
PURPOSE To probe cerebral microstructural abnormalities and assess changes of neuronal density in Disrupted-in-Schizophrenia-1 (DISC1) mice using non-Gaussian diffusion and quantitative susceptibility mapping (QSM). MATERIALS AND METHODS Brain specimens of transgenic DISC1 mice (n = 8) and control mice (n = 7) were scanned. Metrics of neurite orientation dispersion and density imaging (NODDI) and diffusion kurtosis imaging (DKI), as well as QSM, were acquired. Cell counting was performed on Nissl-stained sections. Group differences of imaging metrics and cell density were assessed. Pearson correlations between imaging metrics and cell densities were also examined. RESULTS Significant increases of neuronal density were observed in the hippocampus of DISC1 mice. DKI metrics such as mean kurtosis exhibited significant group differences in the caudate putamen (P = 0.015), cerebral cortex (P = 0.021), and hippocampus (P = 0.011). However, DKI metrics did not correlate with cell density. In contrast, significant positive correlation between density of neurons and the neurite density index of NODDI in the hippocampus was observed (r = 0.783, P = 0.007). Significant correlation between density of neurons and susceptibility (r = 0.657, P = 0.039), as well as between density of neuroglia and susceptibility (r = 0.750, P = 0.013), was also observed in the hippocampus. CONCLUSION The imaging metrics derived from DKI were not sensitive specifically to cell density, while NODDI could provide diffusion metrics sensitive to density of neurons. The magnetic susceptibility values derived from the QSM method can serve as a sensitive biomarker for quantifying neuronal density.
Collapse
Affiliation(s)
- Nan-Jie Gong
- Electrical Engineering and Computer Sciences, University of California, Berkeley, California, USA
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, China
| | - Russell Dibb
- Center for in vivo Microscopy, Duke University School of Medicine, Durham, North Carolina, USA
| | - Mikhail Pletnikov
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eric Benner
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Chunlei Liu
- Electrical Engineering and Computer Sciences, University of California, Berkeley, California, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, USA
- Brain Imaging and Analysis Center, Duke University School of Medicine, Durham, North Carolina, USA
- Radiology, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
19
|
Li W, Zhou FC, Zhang L, Ng CH, Ungvari GS, Li J, Xiang YT. Comparison of cognitive dysfunction between schizophrenia and bipolar disorder patients: A meta-analysis of comparative studies. J Affect Disord 2020; 274:652-661. [PMID: 32663999 DOI: 10.1016/j.jad.2020.04.051] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/19/2020] [Accepted: 04/27/2020] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Cognitive dysfunction is common in both schizophrenia and bipolar disorder. This is a meta-analysis of studies that compared cognitive dysfunction between schizophrenia and bipolar disorder. METHODS Both international and Chinese databases were systematically searched. Studies that compared cognitive function between schizophrenia and bipolar disorder with the MATRICS Consensus Cognitive Battery (MCCB) were analyzed using the random-effects model. RESULTS Twelve studies with 9,518 participants (4,411 schizophrenia and 5,107 bipolar patients) were included in the analyses. Schizophrenia patients performed significantly worse than bipolar patients on the MCCB total scores with a large effect size (SMD=-0.80, 95%CI: -1.21 to -0.39), as well as on all the 7 subscale scores; attention (SMD=-2.56, 95%CI: -3.55 to -1.57) and social cognition (SMD=-0.86, 95%CI: -1.13 to -0.58) with large effect sizes; and speed of processing (SMD=-0.75, 95%CI: -1.00 to -0.49), working memory (SMD=-0.68, 95%CI: -0.91 to -0.45), verbal learning (SMD=-0.78, 95%CI: -0.95 to -0.61), visual learning (SMD=-0.65, 95%CI: -0.83 to -0.48), and reasoning and problem solving (SMD=-0.61, 95%CI: -0.93 to -0.29) with medium effect sizes. CONCLUSION Compared to bipolar patients, patients with schizophrenia had more severe cognitive dysfunction in this meta-analysis, particularly in attention and social cognition. Timely assessment and treatment of cognitive dysfunction should be part of standard management protocols in both schizophrenia and bipolar disorder.
Collapse
Affiliation(s)
- Wen Li
- Unit of Psychiatry, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macao SAR, China; Center for Cognition and Brain Sciences, University of Macau, Macao SAR, China
| | - Fu-Chun Zhou
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital & the Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Ling Zhang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital & the Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Chee H Ng
- Department of Psychiatry, The Melbourne Clinic and St Vincent's Hospital, University of Melbourne, Richmond, Victoria, Australia
| | - Gabor S Ungvari
- Division of Psychiatry, School of Medicine, University of Western Australia, Perth, Australia; University of Notre Dame Australia, Fremantle, Australia
| | - Jun Li
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, China
| | - Yu-Tao Xiang
- Unit of Psychiatry, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macao SAR, China; Center for Cognition and Brain Sciences, University of Macau, Macao SAR, China.
| |
Collapse
|
20
|
Onozato M, Umino M, Shoji A, Ichiba H, Tsujino N, Funatogawa T, Tagata H, Nemoto T, Mizuno M, Fukushima T. Serum d- and l-lactate, pyruvate and glucose levels in individuals with at-risk mental state and correlations with clinical symptoms. Early Interv Psychiatry 2020; 14:410-417. [PMID: 31402579 DOI: 10.1111/eip.12866] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/31/2019] [Accepted: 07/14/2019] [Indexed: 11/26/2022]
Abstract
AIM Little information exists on the peripheral metabolite levels in individuals with at-risk mental state who meet the criteria for a high-risk state of psychosis. Here, we aimed to investigate serum levels of glucose, pyruvate and d- and l-lactate, which may act as a signalling molecule for learning and memory in neuronal cells. METHODS High performance liquid chromatography or commercial kits were used to assess serum metabolites in individuals with attenuated psychosis symptoms of at-risk mental state (n = 24, men = 12) who were not receiving antipsychotics. The metabolite levels of these individuals were compared with those of age- and sex-matched healthy individuals (controls, n = 23, men = 11). Correlations between the metabolites and clinical symptoms of at-risk mental state were also examined. RESULTS Individuals with at-risk mental state had higher serum glucose levels than did controls (P = 2.18 × 10-3 ), while no significant difference in pyruvate levels were observed between the groups. Individuals with at-risk mental state had significantly lower serum l-lactate levels than did controls (P = 6.31 × 10-5 ), while no differences in d-lactate levels were observed. Furthermore, a negative correlation was identified between serum l-lactate levels and Positive and Negative Syndrome Scale negative symptoms scores (r = -0.5651, P = 4.01 × 10-3 ) in individuals with at-risk mental state. CONCLUSIONS We found that, compared with controls, individuals with at-risk mental state have reduced serum l-lactate levels, which may predate psychosis onset, and may be involved in the related negative symptoms.
Collapse
Affiliation(s)
- Mayu Onozato
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences, Toho University, Chiba, Japan
| | - Maho Umino
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences, Toho University, Chiba, Japan
| | - Ayako Shoji
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences, Toho University, Chiba, Japan
| | - Hideaki Ichiba
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences, Toho University, Chiba, Japan
| | | | - Tomoyuki Funatogawa
- Department of Neuropsychiatry, Toho University School of Medicine, Tokyo, Japan
| | - Hiromi Tagata
- Department of Neuropsychiatry, Toho University School of Medicine, Tokyo, Japan
| | - Takahiro Nemoto
- Department of Neuropsychiatry, Toho University School of Medicine, Tokyo, Japan
| | - Masafumi Mizuno
- Department of Neuropsychiatry, Toho University School of Medicine, Tokyo, Japan
| | - Takeshi Fukushima
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences, Toho University, Chiba, Japan
| |
Collapse
|
21
|
Barnett BR, Casey CP, Torres-Velázquez M, Rowley PA, Yu JPJ. Convergent brain microstructure across multiple genetic models of schizophrenia and autism spectrum disorder: A feasibility study. Magn Reson Imaging 2020; 70:36-42. [PMID: 32298718 PMCID: PMC7685399 DOI: 10.1016/j.mri.2020.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/27/2020] [Accepted: 04/08/2020] [Indexed: 11/17/2022]
Abstract
Neuroimaging studies of psychiatric illness have revealed a broad spectrum of structural and functional perturbations that have been attributed in part to the complex genetic heterogeneity underpinning these disorders. These perturbations have been identified in both preclinical genetic models and in patients when compared to control populations, but recent work has also demonstrated strong evidence for genetic, molecular, and structural convergence of several psychiatric diseases. We explored potential similarities in neural microstructure in preclinical genetic models of ASD (Fmr1, Nrxn1, Pten) and schizophrenia (Disc1 svΔ2) and in age- and sex-matched control animals with diffusion tensor imaging (DTI) and neurite orientation dispersion and density imaging (NODDI). Our findings demonstrate a convergence in brain microstructure across these four genetic models with both tract-based and region-of-interest based analyses, which continues to buttress an emerging understanding of converging neural microstructure in psychiatric disease.
Collapse
Affiliation(s)
- Brian R Barnett
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Cameron P Casey
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Maribel Torres-Velázquez
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Paul A Rowley
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - John-Paul J Yu
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA.
| |
Collapse
|
22
|
The black sheep of the family- whole-exome sequencing in family of lithium response discordant bipolar monozygotic twins. Eur Neuropsychopharmacol 2020; 34:19-27. [PMID: 32305265 DOI: 10.1016/j.euroneuro.2020.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 03/06/2020] [Indexed: 01/20/2023]
Abstract
Twin studies are among the most promising strategies for studying heritable disorders, including bipolar disorder (BD). The aim of the present study was to identify distinguishing genes between monozygotic (MZ) twins with different BD phenotype and compare them to their non-affected siblings. Whole-exome sequencing (WES) can identify rare and structural variants that could detect the polygenetic burden of complex disorders. WES was performed on a family composed of two MZ twins with BD, their unaffected brother and unaffected parents. The twins have a discordant response to lithium and distinct course of illness. Following WES, six genes of particular interest emerged: Neurofibromin type 1 (NF1), Biorientation of chromosomes in cell division 1 (BOD1), Golgi-associated gamma adaptin ear-containing ARF binding protein 3 (GGA3), Disrupted in schizophrenia 1 (DISC1), Neuromedin U receptor 2 (NMUR2), and Huntingtin interacting protein 1-related (HIP1R). Interestingly, many of these influence glutamatergic pathways and thus the findings may have therapeutical implications. These results may provide important insights to unveil genetic underpinnings of BD and the response to lithium.
Collapse
|
23
|
Barnett BR, Fathi F, Falco Cobra P, Yi SY, Anderson JM, Eghbalnia HR, Markley JL, Yu JPJ. Metabolic Changes in Synaptosomes in an Animal Model of Schizophrenia Revealed by 1H and 1H, 13C NMR Spectroscopy. Metabolites 2020; 10:E79. [PMID: 32102223 PMCID: PMC7074231 DOI: 10.3390/metabo10020079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/31/2020] [Accepted: 02/22/2020] [Indexed: 12/15/2022] Open
Abstract
Synaptosomes are isolated nerve terminals that contain synaptic components, including neurotransmitters, metabolites, adhesion/fusion proteins, and nerve terminal receptors. The essential role of synaptosomes in neurotransmission has stimulated keen interest in understanding both their proteomic and metabolic composition. Mass spectrometric (MS) quantification of synaptosomes has illuminated their proteomic composition, but the determination of the metabolic composition by MS has been met with limited success. In this study, we report a proof-of-concept application of one- and two-dimensional nuclear magnetic resonance (NMR) spectroscopy for analyzing the metabolic composition of synaptosomes. We utilize this approach to compare the metabolic composition synaptosomes from a wild-type rat with that from a newly generated genetic rat model (Disc1 svΔ2), which qualitatively recapitulates clinically observed early DISC1 truncations associated with schizophrenia. This study demonstrates the feasibility of using NMR spectroscopy to identify and quantify metabolites within synaptosomal fractions.
Collapse
Affiliation(s)
- Brian R. Barnett
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin–Madison, Madison, WI 53705, USA; (B.R.B.); (S.Y.Y.)
| | - Fariba Fathi
- Biochemistry Department, University of Wisconsin–Madison, Madison, WI 53706, USA; (F.F.); (P.F.C.); (H.R.E.); (J.L.M.)
| | - Paulo Falco Cobra
- Biochemistry Department, University of Wisconsin–Madison, Madison, WI 53706, USA; (F.F.); (P.F.C.); (H.R.E.); (J.L.M.)
| | - Sue Y. Yi
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin–Madison, Madison, WI 53705, USA; (B.R.B.); (S.Y.Y.)
| | - Jacqueline M. Anderson
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| | - Hamid R. Eghbalnia
- Biochemistry Department, University of Wisconsin–Madison, Madison, WI 53706, USA; (F.F.); (P.F.C.); (H.R.E.); (J.L.M.)
| | - John L. Markley
- Biochemistry Department, University of Wisconsin–Madison, Madison, WI 53706, USA; (F.F.); (P.F.C.); (H.R.E.); (J.L.M.)
| | - John-Paul J. Yu
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin–Madison, Madison, WI 53705, USA; (B.R.B.); (S.Y.Y.)
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin–Madison, Madison, WI 53706, USA
- Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
24
|
Ward J, Lyall LM, Bethlehem RAI, Ferguson A, Strawbridge RJ, Lyall DM, Cullen B, Graham N, Johnston KJA, Bailey MES, Murray GK, Smith DJ. Novel genome-wide associations for anhedonia, genetic correlation with psychiatric disorders, and polygenic association with brain structure. Transl Psychiatry 2019; 9:327. [PMID: 31797917 PMCID: PMC6892870 DOI: 10.1038/s41398-019-0635-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/18/2019] [Accepted: 10/20/2019] [Indexed: 12/20/2022] Open
Abstract
Anhedonia is a core symptom of several psychiatric disorders but its biological underpinnings are poorly understood. We performed a genome-wide association study of state anhedonia in 375,275 UK Biobank participants and assessed for genetic correlation between anhedonia and neuropsychiatric conditions (major depressive disorder, schizophrenia, bipolar disorder, obsessive compulsive disorder and Parkinson's Disease). We then used a polygenic risk score approach to test for association between genetic loading for anhedonia and both brain structure and brain function. This included: magnetic resonance imaging (MRI) assessments of total grey matter volume, white matter volume, cerebrospinal fluid volume, and 15 cortical/subcortical regions of interest; diffusion tensor imaging (DTI) measures of white matter tract integrity; and functional MRI activity during an emotion processing task. We identified 11 novel loci associated at genome-wide significance with anhedonia, with a SNP heritability estimate (h2SNP) of 5.6%. Strong positive genetic correlations were found between anhedonia and major depressive disorder, schizophrenia and bipolar disorder; but not with obsessive compulsive disorder or Parkinson's Disease. Polygenic risk for anhedonia was associated with poorer brain white matter integrity, smaller total grey matter volume, and smaller volumes of brain regions linked to reward and pleasure processing, including orbito-frontal cortex. In summary, the identification of novel anhedonia-associated loci substantially expands our current understanding of the biological basis of state anhedonia and genetic correlations with several psychiatric disorders confirm the utility of this phenotype as a transdiagnostic marker of vulnerability to mental illness. We also provide the first evidence that genetic risk for state anhedonia influences brain structure, including in regions associated with reward and pleasure processing.
Collapse
Affiliation(s)
- Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Laura M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | | | - Amy Ferguson
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Rona J Strawbridge
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
- Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Donald M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Breda Cullen
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Nicholas Graham
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | | | - Mark E S Bailey
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Graham K Murray
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Daniel J Smith
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK.
| |
Collapse
|
25
|
Translating preclinical findings in clinically relevant new antipsychotic targets: focus on the glutamatergic postsynaptic density. Implications for treatment resistant schizophrenia. Neurosci Biobehav Rev 2019; 107:795-827. [DOI: 10.1016/j.neubiorev.2019.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 07/20/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023]
|
26
|
Dahoun T, Nour MM, Adams RA, Trossbach S, Lee SH, Patel H, Curtis C, Korth C, Howes OD. Disrupted-in-schizophrenia 1 functional polymorphisms and D 2 /D 3 receptor availability: A [ 11 C]-(+)-PHNO imaging study. GENES BRAIN AND BEHAVIOR 2019; 18:e12596. [PMID: 31264367 DOI: 10.1111/gbb.12596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 06/17/2019] [Accepted: 06/17/2019] [Indexed: 11/28/2022]
Abstract
The disrupted-in-schizophrenia 1 (DISC1) protein has been implicated in a range of biological mechanisms underlying chronic mental disorders such as schizophrenia. Schizophrenia is associated with abnormal striatal dopamine signalling, and all antipsychotic drugs block striatal dopamine 2/3 receptors (D2/3 Rs). Importantly, the DISC1 protein directly interacts and forms a protein complex with the dopamine D2 receptor (D2 R) that inhibits agonist-induced D2 R internalisation. Moreover, animal studies have found large striatal increases in the proportion of D2 R receptors in a high affinity state (D2 high R) in DISC1 rodent models. Here, we investigated the relationship between the three most common polymorphisms altering the amino-acid sequence of the DISC1 protein (Ser704Cys (rs821616), Leu607Phe (rs6675281) and Arg264Gln (rs3738401)) and striatal D2/3 R availability in 41 healthy human volunteers, using [11 C]-(+)-PHNO positron emission tomography. We found no association between DISC1 polymorphisms and D2/3 R availability in the striatum and D2 R availability in the caudate and putamen. Therefore, despite a direct interaction between DISC1 and the D2 R, none of its main functional polymorphisms impact striatal D2/3 R binding potential, suggesting DISC1 variants act through other mechanisms.
Collapse
Affiliation(s)
- Tarik Dahoun
- Psychiatric Imaging Group, Robert Steiner MRI Unit, MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK.,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK.,Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - Matthew M Nour
- Psychiatric Imaging Group, Robert Steiner MRI Unit, MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK.,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK.,Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King's College London, London, UK.,Max Planck UCL Centre for Computational Psychiatry and Ageing Research, University College London, Russell Square House, London, UK.,Wellcome Centre for Human Neuroimaging (WCHN), University College London, London, UK
| | - Rick A Adams
- Psychiatric Imaging Group, Robert Steiner MRI Unit, MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK.,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK.,Division of Psychiatry, University College London, London, UK.,Institute of Cognitive Neuroscience, University College London, London, UK
| | - Svenja Trossbach
- Department Neuropathology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sang H Lee
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM) & Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK.,Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Hamel Patel
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM) & Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK.,Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Charles Curtis
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK.,Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM) & Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Carsten Korth
- Department Neuropathology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Oliver D Howes
- Psychiatric Imaging Group, Robert Steiner MRI Unit, MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK.,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK.,Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King's College London, London, UK
| |
Collapse
|
27
|
Sultana R, Ogundele OM, Lee CC. Contrasting characteristic behaviours among common laboratory mouse strains. ROYAL SOCIETY OPEN SCIENCE 2019; 6:190574. [PMID: 31312505 PMCID: PMC6599779 DOI: 10.1098/rsos.190574] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/14/2019] [Indexed: 05/15/2023]
Abstract
Mice are widely used to model wide-ranging human neurological disorders, from development to degenerative pathophysiology. Behavioural and molecular characteristics of these mouse models are influenced by the genetic background of each strain. Among the most commonly used strains, the inbred C57BL/6J, BALB/c, CBA and 129SvEv lines and the CD1 outbred line are particularly predominant. Despite their prevalence, comparative performance of these strains on many standard behavioural tests commonly used to assess neurological conditions remains diffusely and indirectly accessible in the literature. Given that independent studies may be conducted with mice of differing genetic backgrounds, any variation in characteristic behavioural responses of specific strains should be delineated in order to properly interpret results among studies. Thus, in the present study, we aimed to characterize these commonly used mice strains through several standard behavioural tests. Here, we found that animals from different genetic background strains exhibited varying behavioural patterns when assessed for sociability/novelty, memory function, and negative behaviours like despair and stress calls. These results suggest that genetic variation among strains may be responsible-in part-for strain-specific behavioural phenotypes and potential predisposition to some neurological disorders.
Collapse
Affiliation(s)
- Razia Sultana
- Neural Systems Laboratory, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
- Synapse Biology Laboratory, Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
- Author for correspondence: Razia Sultana e-mail:
| | - Olalekan M. Ogundele
- Synapse Biology Laboratory, Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - Charles C. Lee
- Neural Systems Laboratory, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| |
Collapse
|
28
|
Kakuda K, Niwa A, Honda R, Yamaguchi KI, Tomita H, Nojebuzzaman M, Hara A, Goto Y, Osawa M, Kuwata K. A DISC1 point mutation promotes oligomerization and impairs information processing in a mouse model of schizophrenia. J Biochem 2019; 165:369-378. [PMID: 30561706 DOI: 10.1093/jb/mvy116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 12/14/2018] [Indexed: 12/15/2022] Open
Abstract
Disrupted-in-schizophrenia 1 (DISC1) is strongly associated with schizophrenia, but it remains elusive how the modification of the intermolecular interaction of DISC1 affects the information processing in brain. We show that a DISC1 point mutation alters intermolecular cohesiveness promoting the phase separation, and disrupts sensorimotor gating monitored by the prepulse inhibition in a mouse model of schizophrenia. Although the conformation of DISC1 partial peptide with the schizophrenia-related mutation L607F in human or the corresponding L604F in mouse was essentially indistinguishable from the wild type (WT) as long as monitored by fluorescence, circular dichroism, ultracentrifugation, dynamic light scattering and nuclear magnetic resonance, the atomic force microscopy was able to detect their morphological distinctions. The WT peptides were round and well dispersed, while mutants were inhomogeneous and disrupted to form dimer to trimer that aligned along one direction without apparent aggregate formation. Homozygous L604F mutant mice created by CRISPR exhibited the significant decrease in DISC1 level in the immunohistopathology at the hippocampal region compared to the WTs. The ratio of prepulse inhibition of the homozygous mutant mice was significantly impaired compared to WTs. Altered DISC1 distribution or function caused by aberrant intermolecular interactions may contribute to information processing characteristics in schizophrenia.
Collapse
Affiliation(s)
- Kyosuke Kakuda
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu
| | - Ayumi Niwa
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu
| | - Ryo Honda
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu
| | - Kei-Ichi Yamaguchi
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu
| | - Md Nojebuzzaman
- Division of Regeneration Technology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu
| | - Yuji Goto
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka
| | - Masatake Osawa
- Division of Regeneration Technology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Kazuo Kuwata
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu
| |
Collapse
|
29
|
Segal-Gavish H, Barzilay R, Rimoni O, Offen D. Voluntary exercise improves cognitive deficits in female dominant-negative DISC1 transgenic mouse model of neuropsychiatric disorders. World J Biol Psychiatry 2019; 20:243-252. [PMID: 28593819 DOI: 10.1080/15622975.2017.1323118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Physical exercise has gained increasing interest as a treatment modality that improves prognosis in psychiatric patients. The disrupted in schizophrenia 1 (DISC1) gene is a candidate gene for major mental illness. In this study, we aimed to determine whether voluntary wheel running can improve cognitive deficits of dominant-negative DISC1 transgenic mice (DN-DISC1). METHODS DN-DISC1 and control mice (10-week-old male and female) were placed for 14 days in a cage with or without access to a running wheel. Two weeks later, mice underwent behavioural tests evaluating cognition and social approach and recognition. RESULTS Voluntary exercise improved performance in the novel object recognition test, restored the impairment in spatial memory in the Y maze, and reversed the deficit in social recognition memory in DN-DISC1 females. DN-DISC1 males did not exhibit behavioural deficits at baseline. Tissue analysis revealed that exercise induced a significant increase in hippocampal expression of doublecortin (DCX), brain-derived neurotrophic factor (BDNF) and cannabinoid receptor type 1 (CB1R) only in DN-DISC1 females. CONCLUSIONS Voluntary exercise is beneficial in attenuating cognitive deficits observed in a rodent model relevant for neuropsychiatric disorders. The data add a preclinical aspect to the accumulating clinical data supporting the incorporation of physical exercise to patients' care.
Collapse
Affiliation(s)
- Hadar Segal-Gavish
- a Laboratory of Neuroscience , Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv , Israel
| | - Ran Barzilay
- a Laboratory of Neuroscience , Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv , Israel.,b Research Unit , Geha Mental Health Center , Petach Tikva , Israel
| | - Ofri Rimoni
- a Laboratory of Neuroscience , Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv , Israel
| | - Daniel Offen
- a Laboratory of Neuroscience , Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv , Israel
| |
Collapse
|
30
|
Barnett BR, Torres-Velázquez M, Yi SY, Rowley PA, Sawin EA, Rubinstein CD, Krentz K, Anderson JM, Bakshi VP, Yu JPJ. Sex-specific deficits in neurite density and white matter integrity are associated with targeted disruption of exon 2 of the Disc1 gene in the rat. Transl Psychiatry 2019; 9:82. [PMID: 30745562 PMCID: PMC6370885 DOI: 10.1038/s41398-019-0429-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 01/24/2019] [Accepted: 01/26/2019] [Indexed: 02/06/2023] Open
Abstract
Diffusion tensor imaging (DTI) has provided remarkable insight into our understanding of white matter microstructure and brain connectivity across a broad spectrum of psychiatric disease. While DTI and other diffusion weighted magnetic resonance imaging (MRI) methods have clarified the axonal contribution to the disconnectivity seen in numerous psychiatric diseases, absent from these studies are quantitative indices of neurite density and orientation that are especially important features in regions of high synaptic density that would capture the synaptic contribution to the psychiatric disease state. Here we report the application of neurite orientation dispersion and density imaging (NODDI), an emerging microstructure imaging technique, to a novel Disc1 svΔ2 rat model of psychiatric illness and demonstrate the complementary and more specific indices of tissue microstructure found in NODDI than those reported by DTI. Our results demonstrate global and sex-specific changes in white matter microstructural integrity and deficits in neurite density as a consequence of the Disc1 svΔ2 genetic variation and highlight the application of NODDI and quantitative measures of neurite density and neurite dispersion in psychiatric disease.
Collapse
Affiliation(s)
- Brian R Barnett
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Maribel Torres-Velázquez
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Sue Y Yi
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Paul A Rowley
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Emily A Sawin
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - C Dustin Rubinstein
- Biotechnology Center, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Kathleen Krentz
- Biotechnology Center, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jacqueline M Anderson
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Vaishali P Bakshi
- Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - John-Paul J Yu
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.
- Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.
| |
Collapse
|
31
|
Nedic Erjavec G, Svob Strac D, Tudor L, Konjevod M, Sagud M, Pivac N. Genetic Markers in Psychiatry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1192:53-93. [PMID: 31705490 DOI: 10.1007/978-981-32-9721-0_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Psychiatric disorders such as addiction (substance use and addictive disorders), depression, eating disorders, schizophrenia, and post-traumatic stress disorder (PTSD) are severe, complex, multifactorial mental disorders that carry a high social impact, enormous public health costs, and various comorbidities as well as premature morbidity. Their neurobiological foundation is still not clear. Therefore, it is difficult to uncover new set of genes and possible genetic markers of these disorders since the understanding of the molecular imbalance leading to these disorders is not complete. The integrative approach is needed which will combine genomics and epigenomics; evaluate epigenetic influence on genes and their influence on neuropeptides, neurotransmitters, and hormones; examine gene × gene and gene × environment interplay; and identify abnormalities contributing to development of these disorders. Therefore, novel genetic approaches based on systems biology focused on improvement of the identification of the biological underpinnings might offer genetic markers of addiction, depression, eating disorders, schizophrenia, and PTSD. These markers might be used for early prediction, detection of the risk to develop these disorders, novel subtypes of the diseases and tailored, personalized approach to therapy.
Collapse
Affiliation(s)
- Gordana Nedic Erjavec
- Division of Molecular Medicine, Rudjer Boskovic Institute, Bijenicka 54, HR-10000, Zagreb, Croatia
| | - Dubravka Svob Strac
- Division of Molecular Medicine, Rudjer Boskovic Institute, Bijenicka 54, HR-10000, Zagreb, Croatia
| | - Lucija Tudor
- Division of Molecular Medicine, Rudjer Boskovic Institute, Bijenicka 54, HR-10000, Zagreb, Croatia
| | - Marcela Konjevod
- Division of Molecular Medicine, Rudjer Boskovic Institute, Bijenicka 54, HR-10000, Zagreb, Croatia
| | - Marina Sagud
- School of Medicine, University of Zagreb, Salata 2, HR-10000, Zagreb, Croatia
- Department of Psychiatry, University Hospital Centre Zagreb, Kispaticeva 12, HR-10000, Zagreb, Croatia
| | - Nela Pivac
- Division of Molecular Medicine, Rudjer Boskovic Institute, Bijenicka 54, HR-10000, Zagreb, Croatia.
| |
Collapse
|
32
|
Dobyns WB, Aldinger KA, Ishak GE, Mirzaa GM, Timms AE, Grout ME, Dremmen MH, Schot R, Vandervore L, van Slegtenhorst MA, Wilke M, Kasteleijn E, Lee AS, Barry BJ, Chao KR, Szczałuba K, Kobori J, Hanson-Kahn A, Bernstein JA, Carr L, D’Arco F, Miyana K, Okazaki T, Saito Y, Sasaki M, Das S, Wheeler MM, Bamshad MJ, Nickerson DA, Engle EC, Verheijen FW, Doherty D, Mancini GM, Doherty D, Mancini GMS. MACF1 Mutations Encoding Highly Conserved Zinc-Binding Residues of the GAR Domain Cause Defects in Neuronal Migration and Axon Guidance. Am J Hum Genet 2018; 103:1009-1021. [PMID: 30471716 DOI: 10.1016/j.ajhg.2018.10.019] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 10/22/2018] [Indexed: 01/08/2023] Open
Abstract
To date, mutations in 15 actin- or microtubule-associated genes have been associated with the cortical malformation lissencephaly and variable brainstem hypoplasia. During a multicenter review, we recognized a rare lissencephaly variant with a complex brainstem malformation in three unrelated children. We searched our large brain-malformation databases and found another five children with this malformation (as well as one with a less severe variant), analyzed available whole-exome or -genome sequencing data, and tested ciliogenesis in two affected individuals. The brain malformation comprised posterior predominant lissencephaly and midline crossing defects consisting of absent anterior commissure and a striking W-shaped brainstem malformation caused by small or absent pontine crossing fibers. We discovered heterozygous de novo missense variants or an in-frame deletion involving highly conserved zinc-binding residues within the GAR domain of MACF1 in the first eight subjects. We studied cilium formation and found a higher proportion of mutant cells with short cilia than of control cells with short cilia. A ninth child had similar lissencephaly but only subtle brainstem dysplasia associated with a heterozygous de novo missense variant in the spectrin repeat domain of MACF1. Thus, we report variants of the microtubule-binding GAR domain of MACF1 as the cause of a distinctive and most likely pathognomonic brain malformation. A gain-of-function or dominant-negative mechanism appears likely given that many heterozygous mutations leading to protein truncation are included in the ExAC Browser. However, three de novo variants in MACF1 have been observed in large schizophrenia cohorts.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Dan Doherty
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Grazia M S Mancini
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam 3015 CN, the Netherlands.
| |
Collapse
|
33
|
Ma JH, Sun XY, Guo TJ, Barot E, Wang DF, Yan LL, Ni DW, Huang NH, Xie Q, Zeng J, Ou-Yang L, Liu YQ, Lu QB. Association on DISC1 SNPs with schizophrenia risk: A meta-analysis. Psychiatry Res 2018; 270:306-309. [PMID: 30286368 DOI: 10.1016/j.psychres.2018.09.056] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 09/17/2018] [Accepted: 09/22/2018] [Indexed: 01/09/2023]
Abstract
Schizophrenia is a major psychiatric disorder with complex genetic, environmental, and psychological etiologies. Although DISC1 gene has been shown as a risk factor for schizophrenia in some reports, there is a lack of a consensus. We therefore performed separate meta-analyses aiming to assess the associations between DISC1 SNPs and schizophrenia risk. We found that SNP rs821597 is significantly associated with schizophrenia risk in terms of both allelic and genotypic distribution, while SNP rs821616 is associated with schizophrenia in terms of genotypic distribution, especially in cases above 40 years old.
Collapse
Affiliation(s)
- Jia-Hui Ma
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, No. 38 Xue-Yuan Road, Haidian District, Beijing 100191, PR China.
| | - Xiao-Yu Sun
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, No. 38 Xue-Yuan Road, Haidian District, Beijing 100191, PR China.
| | - Tong-Jun Guo
- Graduate School of Peking Union Medical College, Beijing 100730, China.
| | - Emily Barot
- Bioinformatics Center, University of Copenhagen, Copenhagen 2200, Denmark.
| | | | - Lai-Lai Yan
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, No. 38 Xue-Yuan Road, Haidian District, Beijing 100191, PR China.
| | - Da-Wei Ni
- Central Hospital of Fengxian, Shanghai 201499, China
| | - Ning-Hua Huang
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, No. 38 Xue-Yuan Road, Haidian District, Beijing 100191, PR China.
| | - Qing Xie
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, No. 38 Xue-Yuan Road, Haidian District, Beijing 100191, PR China.
| | - Jing Zeng
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, No. 38 Xue-Yuan Road, Haidian District, Beijing 100191, PR China.
| | - Li Ou-Yang
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, No. 38 Xue-Yuan Road, Haidian District, Beijing 100191, PR China.
| | - Ya-Qiong Liu
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, No. 38 Xue-Yuan Road, Haidian District, Beijing 100191, PR China.
| | - Qing-Bin Lu
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, No. 38 Xue-Yuan Road, Haidian District, Beijing 100191, PR China.
| |
Collapse
|
34
|
Shen X, Yeung HT, Lai KO. Application of Human-Induced Pluripotent Stem Cells (hiPSCs) to Study Synaptopathy of Neurodevelopmental Disorders. Dev Neurobiol 2018; 79:20-35. [PMID: 30304570 DOI: 10.1002/dneu.22644] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/27/2018] [Accepted: 10/04/2018] [Indexed: 12/15/2022]
Abstract
Synapses are the basic structural and functional units for information processing and storage in the brain. Their diverse properties and functions ultimately underlie the complexity of human behavior. Proper development and maintenance of synapses are essential for normal functioning of the nervous system. Disruption in synaptogenesis and the consequent alteration in synaptic function have been strongly implicated to cause neurodevelopmental disorders such as autism spectrum disorders (ASDs) and schizophrenia (SCZ). The introduction of human-induced pluripotent stem cells (hiPSCs) provides a new path to elucidate disease mechanisms and potential therapies. In this review, we will discuss the advantages and limitations of using hiPSC-derived neurons to study synaptic disorders. Many mutations in genes encoding for proteins that regulate synaptogenesis have been identified in patients with ASDs and SCZ. We use Methyl-CpG binding protein 2 (MECP2), SH3 and multiple ankyrin repeat domains 3 (SHANK3) and Disrupted in schizophrenia 1 (DISC1) as examples to illustrate the promise of using hiPSCs as cellular models to elucidate the mechanisms underlying disease-related synaptopathy.
Collapse
Affiliation(s)
- Xuting Shen
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | - Hoi Ting Yeung
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | - Kwok-On Lai
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| |
Collapse
|
35
|
Kirchner SK, Roeh A, Nolden J, Hasan A. Diagnosis and treatment of schizotypal personality disorder: evidence from a systematic review. NPJ SCHIZOPHRENIA 2018; 4:20. [PMID: 30282970 PMCID: PMC6170383 DOI: 10.1038/s41537-018-0062-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 08/15/2018] [Accepted: 08/21/2018] [Indexed: 02/08/2023]
Abstract
The main objective of this review was to evaluate studies on the diagnosis, treatment, and course of schizotypal personality disorder and to provide a clinical guidance on the basis of that evaluation. A systematic search in the PubMed/MEDLINE databases was conducted. Two independent reviewers extracted and assessed the quality of the data. A total of 54 studies were eligible for inclusion: 18 were on diagnostic instruments; 22, on pharmacological treatment; 3, on psychotherapy; and 13, on the longitudinal course of the disease. We identified several suitable and reliable questionnaires for screening (PDQ-4+ and SPQ) and diagnosing (SIDP, SIDP-R, and SCID-II) schizotypal personality disorder. Second-generation antipsychotics (mainly risperidone) were the most often studied drug class and were described as beneficial. Studies on the longitudinal course described a moderate remission rate and possible conversion rates to other schizophrenia spectrum disorders. Because of the heterogeneity of the studies and the small sample sizes, it is not yet possible to make evidence-based recommendations for treatment. This is a systematic evaluation of diagnostic instruments and treatment studies in schizotypal personality disorder. We conclude that there is currently only limited evidence on which to base treatment decisions in this disorder. Larger interventional trials are needed to provide the data for evidence-based recommendations.
Collapse
Affiliation(s)
- Sophie K Kirchner
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University, Munich, Germany.
| | - Astrid Roeh
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University, Munich, Germany
| | - Jana Nolden
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University, Munich, Germany
| | - Alkomiet Hasan
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
36
|
Gene polymorphisms of DISC1 is associated with schizophrenia: Evidence from a meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry 2018; 81:64-73. [PMID: 29031911 DOI: 10.1016/j.pnpbp.2017.10.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 10/08/2017] [Accepted: 10/08/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Previous studies suggest an association between Disrupted in schizophrenia 1 (DISC1) polymorphisms and schizophrenia (SCZ). However, the available data are often inconsistent, regarding the difference in sample size, ethnicity, genotyping method, etc. Thus, we carried out a meta-analysis to determine whether DISC1 polymorphisms contributed susceptibility to SCZ. METHODS A methodical literature review was operated using the English and Chinese core electronic databases. Odds ratios (ORs) with 95% confidence intervals (CIs) were applied to determine the correlation between DISC1 gene polymorphisms and SCZ susceptibility. Subgroup analyses were carried out by stratification of ethnicity. P values were Bonferroni adjusted to account for multiple testing. Publication bias was evaluated by funnel plots, Egger's test and the trim and fill method. RESULTS Meta-analyses results suggested that DISC1 polymorphisms (rs821616 and rs821597) increased SCZ risk in overall populations. In subgroups of ethnicity, DISC1 polymorphisms (rs821616 and rs821597) was associated with susceptibility to SCZ among the Chinese population (for rs821616: TT+AT vs. AA: OR=1.338, 95% CI=1.124-1.592, P=0.001; T vs. A: OR=1.300, 95% CI=1.124-1.504, P<0.000; for rs821597: AA+AG vs. GG: OR=1.508, 95% CI=1.268-1.794, P<0.001; A vs. G: OR=1.345, 95% CI=1.184-1.527, P<0.001). A positive correlation was also observed between the single marker rs821616 and SCZ among the Japanese population in the recessive model (TT vs. AT+AA: OR=1.524, 95% CI=1.185-1.959, P=0.001). There was no significant relationship between other DISC1 polymorphisms (rs3738401, rs2273890, rs3738398, rs3738402, rs2492367, rs843979, rs3737597, rs4658971, rs1538979, rs1000731 and rs3738399) and SCZ. CONCLUSIONS DISC1 polymorphisms increased a risk of SCZ, especially in the Chinese population. In order to further corroborate our findings, large well-designed epidemiological studies are needed.
Collapse
|
37
|
Xu Y, Ren J, Ye H. Association between variations in the disrupted in schizophrenia 1 gene and schizophrenia: A meta-analysis. Gene 2018; 651:94-99. [PMID: 29410289 DOI: 10.1016/j.gene.2018.01.069] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 11/17/2017] [Accepted: 01/20/2018] [Indexed: 10/18/2022]
Abstract
Schizophrenia is a severe psychiatric disorder. Genetic and functional studies have strongly implicated the disrupted in schizophrenia 1 gene (DISC1) as a candidate susceptibility gene for schizophrenia. Moreover, recent association studies have indicated that several DISC1 single nucleotide polymorphisms (SNPs) are associated with schizophrenia. However, the association is hardly replicate in different ethnic group. Here, we performed a meta-analysis of the association between DISC1 SNPs and schizophrenia in which the samples were divided into subgroups according to ethnicity. Both rs3738401 and rs821616 showed not significantly association with schizophrenia in the Caucasian, Asian, Japanese or Han Chinese populations.
Collapse
Affiliation(s)
- Yiliang Xu
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders, Center of Schizophrenia, Capital Medical University, Beijing 100069, China.
| | - Jun Ren
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders, Center of Schizophrenia, Capital Medical University, Beijing 100069, China
| | - Haihong Ye
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders, Center of Schizophrenia, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
38
|
Association of DISC1, BDNF, and COMT polymorphisms with exploratory eye movement of schizophrenia in a Chinese Han population. Psychiatr Genet 2017; 26:258-265. [PMID: 27285059 DOI: 10.1097/ypg.0000000000000138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Previous studies suggested that exploratory eye movement (EEM) dysfunction appears to be a biological marker specific to schizophrenia, with an unknown molecular mechanism. Genetic studies indicate that disrupted-in-schizophrenia-1 (DISC1), brain-derived neurotrophic factor (BDNF), and catechol-O-methyl transferase (COMT) genes might be implicated in the etiology of schizophrenia, but not in all populations. OBJECTIVES The present study aimed to explore associations between these candidate genes and EEM endophenotypes for schizophrenia in a Chinese Han population. METHODS EEM recordings were examined in 139 patients with schizophrenia and 143 healthy control participants. RESULTS All five EEM parameters, responsive search score, cognitive search score, number of eye fixations, total eye scanning length, and mean eye scanning length, of schizophrenic patients differed significantly from those of healthy controls (P<0.001). The DISC1 SerCys, BDNF ValMet, and COMT ValMet were genotyped in a total sample of 818 schizophrenic patients and 827 healthy control participants, including the above EEM samples. We found that DISC1 Cys and BDNF Met were associated with an increased risk of developing schizophrenia (P<0.001). Furthermore, responsive search score scores of BDNF Met/Met carriers were significantly lower than those of Val allele carriers (P=0.022), which remained modest after Bonferroni correction. CONCLUSION The BDNF MetMet polymorphism might be associated with the EEM dysfunction of schizophrenia.
Collapse
|
39
|
Lee A, Shen M, Qiu A. Psychiatric polygenic risk associates with cortical morphology and functional organization in aging. Transl Psychiatry 2017; 7:1276. [PMID: 29225336 PMCID: PMC5802582 DOI: 10.1038/s41398-017-0036-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/04/2017] [Accepted: 09/07/2017] [Indexed: 01/23/2023] Open
Abstract
Common brain abnormalities in cortical morphology and functional organization are observed in psychiatric disorders and aging, reflecting shared genetic influences. This preliminary study aimed to examine the contribution of a polygenetic risk for psychiatric disorders (PRScross) to aging brain and to identify molecular mechanisms through the use of multimodal brain images, genotypes, and transcriptome data. We showed age-related cortical thinning in bilateral inferior frontal cortex (IFC) and superior temporal gyrus and alterations in the functional connectivity between bilateral IFC and between right IFC and right inferior parietal lobe as a function of PRScross. Interestingly, the genes in PRScross, that contributed most to aging neurodegeneration, were expressed in the functioanlly connected cortical regions. Especially, genes identified through the genotype-functional connectivity association analysis were commonly expressed in both cortical regions and formed strong gene networks with biological processes related to neural plasticity and synaptogenesis, regulated by glutamatergic and GABAergic transmission, neurotrophin signaling, and metabolism. This study suggested integrating genotype and transcriptome with neuroimage data sheds new light on the mechanisms of aging brain.
Collapse
Affiliation(s)
- Annie Lee
- 0000 0001 2180 6431grid.4280.eDepartment of Biomedical Engineering, National University of Singapore, Singapore, 117576 Singapore
| | - Mojun Shen
- 0000 0004 0637 0221grid.185448.4Singapore Institute for Clinical Sciences, The Agency for Science, Technology and Research, Singapore, 117609 Singapore
| | - Anqi Qiu
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore. .,Singapore Institute for Clinical Sciences, The Agency for Science, Technology and Research, Singapore, 117609, Singapore. .,Clinical Imaging Research Center, National University of Singapore, Singapore, 117456, Singapore.
| |
Collapse
|
40
|
Matsumoto Y, Niwa M, Mouri A, Noda Y, Fukushima T, Ozaki N, Nabeshima T. Adolescent stress leads to glutamatergic disturbance through dopaminergic abnormalities in the prefrontal cortex of genetically vulnerable mice. Psychopharmacology (Berl) 2017; 234:3055-3074. [PMID: 28756461 PMCID: PMC8034555 DOI: 10.1007/s00213-017-4704-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 07/04/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Stress during the adolescent period influences postnatal maturation and behavioral patterns in adulthood. Adolescent stress-induced molecular and functional changes in neurons are the key clinical features of psychiatric disorders including schizophrenia. OBJECTIVE In the present study, we exposed genetically vulnerable mice to isolation stress to examine the molecular changes in the glutamatergic system involving N-methyl-d-aspartate (NMDA) receptors via dopaminergic disturbance in the prefrontal cortex (PFc). RESULTS We report that late adolescent stress in combination with Disrupted-in-Schizophrenia 1 (DISC1) genetic risk elicited alterations in glutamatergic neurons in the PFc, such as increased expression of glutamate transporters, decreased extracellular levels of glutamate, decreased concentration of d-serine, and impaired activation of NMDA-Ca2+/calmodulin kinase II signaling. These changes resulted in behavioral deficits in locomotor activity, forced swim, social interaction, and novelty preference tests. The glutamatergic alterations in the PFc were prevented if the animals were treated with an atypical antipsychotic drug clozapine and a dopamine D1 agonist SKF81297, which suggests that the activation of dopaminergic neurons is involved in the regulation of the glutamatergic system. CONCLUSION Our results suggest that adolescent stress combined with dopaminergic abnormalities in the PFc of genetically vulnerable mice induces glutamatergic disturbances, which leads to behavioral deficits in the young adult stage.
Collapse
Affiliation(s)
- Yurie Matsumoto
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University, Nagoya, 468-8503, Japan
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, 466-8560, Japan
| | - Minae Niwa
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University, Nagoya, 468-8503, Japan.
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, 466-8560, Japan.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Akihiro Mouri
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University, Nagoya, 468-8503, Japan
- Advanced Diagnostic System Research Laboratory, Fujita Health University, Graduate School of Health Science, Toyoake, 470-1192, Japan
- NPO Japanese Drug Organization of Appropriate Use and Research, Nagoya, 468-0069, Japan
| | - Yukihiro Noda
- NPO Japanese Drug Organization of Appropriate Use and Research, Nagoya, 468-0069, Japan
- Division of Clinical Sciences and Neuropsychopharmacology, Graduate School of Pharmacy, Meijo University, Nagoya, 468-8503, Japan
| | - Takeshi Fukushima
- Department of Analytical Chemistry, Faculty of Pharmaceutical Science, Toho University, Chiba, 274-8510, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, 466-8560, Japan
| | - Toshitaka Nabeshima
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University, Nagoya, 468-8503, Japan.
- Advanced Diagnostic System Research Laboratory, Fujita Health University, Graduate School of Health Science, Toyoake, 470-1192, Japan.
- NPO Japanese Drug Organization of Appropriate Use and Research, Nagoya, 468-0069, Japan.
- Aino University, Ibaragi, Osaka, 567-0012, Japan.
| |
Collapse
|
41
|
Accumulation of minor alleles and risk prediction in schizophrenia. Sci Rep 2017; 7:11661. [PMID: 28916820 PMCID: PMC5601945 DOI: 10.1038/s41598-017-12104-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/01/2017] [Indexed: 12/15/2022] Open
Abstract
Schizophrenia is a common neuropsychiatric disorder with a lifetime risk of 1%. Accumulation of common polygenic variations has been found to be an important risk factor. Recent studies showed a role for the enrichment of minor alleles (MAs) of SNPs in complex diseases such as Parkinson’s disease. Here we similarly studied the role of genome wide MAs in schizophrenia using public datasets. Relative to matched controls, schizophrenia cases showed higher average values in minor allele content (MAC) or the average amount of MAs per subject. By risk prediction analysis based on weighted genetic risk score (wGRS) of MAs, we identified an optimal MA set consisting of 23 238 variants that could be used to predict 3.14% of schizophrenia cases, which is comparable to using 22q11 deletion to detect schizophrenia cases. Pathway enrichment analysis of these SNPs identified 30 pathways with false discovery rate (FDR) <0.02 and of significant P-value, most of which are known to be linked with schizophrenia and other neurological disorders. These results suggest that MAs accumulation may be a risk factor to schizophrenia and provide a method to genetically screen for this disease.
Collapse
|
42
|
Norkett R, Modi S, Kittler JT. Mitochondrial roles of the psychiatric disease risk factor DISC1. Schizophr Res 2017; 187:47-54. [PMID: 28087269 DOI: 10.1016/j.schres.2016.12.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 12/17/2016] [Accepted: 12/22/2016] [Indexed: 12/31/2022]
Abstract
Ion transport during neuronal signalling utilizes the majority of the brain's energy supply. Mitochondria are key sites for energy provision through ATP synthesis and play other important roles including calcium buffering. Thus, tightly regulated distribution and function of these organelles throughout the intricate architecture of the neuron is essential for normal synaptic communication. Therefore, delineating mechanisms coordinating mitochondrial transport and function is essential for understanding nervous system physiology and pathology. While aberrant mitochondrial transport and dynamics have long been associated with neurodegenerative disease, they have also more recently been linked to major mental illness including schizophrenia, autism and depression. However, the underlying mechanisms have yet to be elucidated, due to an incomplete understanding of the combinations of genetic and environmental factors contributing to these conditions. Consequently, the DISC1 gene has undergone intense study since its discovery at the site of a balanced chromosomal translocation, segregating with mental illness in a Scottish pedigree. The precise molecular functions of DISC1 remain elusive. Reported functions of DISC1 include regulation of intracellular signalling pathways, neuronal migration and dendritic development. Intriguingly, a role for DISC1 in mitochondrial homeostasis and transport is fast emerging. Therefore, a major function of DISC1 in regulating mitochondrial distribution, ATP synthesis and calcium buffering may be disrupted in psychiatric disease. In this review, we discuss the links between DISC1 and mitochondria, considering both trafficking of these organelles and their function, and how, via these processes, DISC1 may contribute to the regulation of neuronal behavior in normal and psychiatric disease states.
Collapse
Affiliation(s)
- R Norkett
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| | - S Modi
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| | - J T Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK.
| |
Collapse
|
43
|
Tang J, Fan Y, Li H, Xiang Q, Zhang DF, Li Z, He Y, Liao Y, Wang Y, He F, Zhang F, Shugart YY, Liu C, Tang Y, Chan RCK, Wang CY, Yao YG, Chen X. Whole-genome sequencing of monozygotic twins discordant for schizophrenia indicates multiple genetic risk factors for schizophrenia. J Genet Genomics 2017; 44:295-306. [PMID: 28645778 DOI: 10.1016/j.jgg.2017.05.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/29/2017] [Accepted: 05/09/2017] [Indexed: 12/18/2022]
Abstract
Schizophrenia is a common disorder with a high heritability, but its genetic architecture is still elusive. We implemented whole-genome sequencing (WGS) analysis of 8 families with monozygotic (MZ) twin pairs discordant for schizophrenia to assess potential association of de novo mutations (DNMs) or inherited variants with susceptibility to schizophrenia. Eight non-synonymous DNMs (including one splicing site) were identified and shared by twins, which were either located in previously reported schizophrenia risk genes (p.V24689I mutation in TTN, p.S2506T mutation in GCN1L1, IVS3+1G > T in DOCK1) or had a benign to damaging effect according to in silico prediction analysis. By searching the inherited rare damaging or loss-of-function (LOF) variants and common susceptible alleles from three classes of schizophrenia candidate genes, we were able to distill genetic alterations in several schizophrenia risk genes, including GAD1, PLXNA2, RELN and FEZ1. Four inherited copy number variations (CNVs; including a large deletion at 16p13.11) implicated for schizophrenia were identified in four families, respectively. Most of families carried both missense DNMs and inherited risk variants, which might suggest that DNMs, inherited rare damaging variants and common risk alleles together conferred to schizophrenia susceptibility. Our results support that schizophrenia is caused by a combination of multiple genetic factors, with each DNM/variant showing a relatively small effect size.
Collapse
Affiliation(s)
- Jinsong Tang
- Institute of Mental Health, National Clinical Research Center for Mental Health Disorders and National Technology Institute of Psychiatry, and Key Laboratory of Psychiatry and Mental Health of Hunan Province, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yu Fan
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Hong Li
- Institute of Mental Health, National Clinical Research Center for Mental Health Disorders and National Technology Institute of Psychiatry, and Key Laboratory of Psychiatry and Mental Health of Hunan Province, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qun Xiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Deng-Feng Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Zongchang Li
- Institute of Mental Health, National Clinical Research Center for Mental Health Disorders and National Technology Institute of Psychiatry, and Key Laboratory of Psychiatry and Mental Health of Hunan Province, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Ying He
- Institute of Mental Health, National Clinical Research Center for Mental Health Disorders and National Technology Institute of Psychiatry, and Key Laboratory of Psychiatry and Mental Health of Hunan Province, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yanhui Liao
- Institute of Mental Health, National Clinical Research Center for Mental Health Disorders and National Technology Institute of Psychiatry, and Key Laboratory of Psychiatry and Mental Health of Hunan Province, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Ya Wang
- Neuropsychology and Applied Cognitive Neuroscience Laboratory, and CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China
| | - Fan He
- Beijing Key Laboratory of Mental Disorders, Department of Psychiatry, Beijing Anding Hospital, and Center of Schizophrenia, Beijing Institute for Brain Disorders and Laboratory of Brain Disorders of the Ministry of Science and Technology, Capital Medical University, Beijing 100088, China
| | - Fengyu Zhang
- Institute of Mental Health, National Clinical Research Center for Mental Health Disorders and National Technology Institute of Psychiatry, and Key Laboratory of Psychiatry and Mental Health of Hunan Province, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yin Yao Shugart
- Unit on Statistical Genomics, Intramural Research Programs, National Institute of Mental Health, NIH, Bethesda 20892, USA
| | - Chunyu Liu
- Institute of Human Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Yanqing Tang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110122, China.
| | - Raymond C K Chan
- Neuropsychology and Applied Cognitive Neuroscience Laboratory, and CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Chuan-Yue Wang
- Beijing Key Laboratory of Mental Disorders, Department of Psychiatry, Beijing Anding Hospital, and Center of Schizophrenia, Beijing Institute for Brain Disorders and Laboratory of Brain Disorders of the Ministry of Science and Technology, Capital Medical University, Beijing 100088, China.
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Xiaogang Chen
- Institute of Mental Health, National Clinical Research Center for Mental Health Disorders and National Technology Institute of Psychiatry, and Key Laboratory of Psychiatry and Mental Health of Hunan Province, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| |
Collapse
|
44
|
Chlorpromazine Increases the Expression of Polysialic Acid (PolySia) in Human Neuroblastoma Cells and Mouse Prefrontal Cortex. Int J Mol Sci 2017; 18:ijms18061123. [PMID: 28538701 PMCID: PMC5485947 DOI: 10.3390/ijms18061123] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 05/17/2017] [Accepted: 05/18/2017] [Indexed: 01/11/2023] Open
Abstract
The neural cell adhesion molecule (NCAM) is modified by polysialic acid (polySia or PSA) in embryonic brains. In adult brains, polySia modification of NCAM is only observed in restricted areas where neural plasticity, remodeling of neural connections, or neural generation is ongoing although the amount of NCAM remains unchanged. Impairments of the polySia-expression and several single nucleotide polymorphisms (SNPs) of the polysialyltransferase (polyST) ST8SIA2 gene are reported to be associated with schizophrenia and bipolar disorder. Chlorpromazine (CPZ) is well-known as an agent for treating schizophrenia, and our hypothesis is that CPZ may affect the polySia expression or the gene expression of polySTs or NCAM. To test this hypothesis, we analyzed the effects of CPZ on the expression of polySia-NCAM on human neuroblastoma cell line, IMR-32 cells, by immunochemical and chemical methods. Interestingly, the cell surface expression of polySia, especially those with lower chain lengths, was significantly increased on the CPZ-treated cells, while mRNAs for polySTs and NCAM, and the amounts of total polySia-NCAM remained unchanged. The addition of brefeldin A, an inhibitor of endocytosis, suppressed the CPZ-induced cell surface polySia expression. In addition, polySia-NCAM was also observed in the vesicle compartment inside the cell. All these data suggest that the level of cell surface expression of polySia in IMR-32 is highly regulated and that CPZ changes the rate of the recycling of polySia-NCAM, leading to the up-regulation of polySia-NCAM on the cell surface. We also analyzed the effect of CPZ on polySia-expression in various brain regions in adult mice and found that CPZ only influenced the total amounts of polySia-NCAM in prefrontal cortex. These results suggest a brain-region-specific effect of CPZ on the expression of total polySia in mouse brain. Collectively, anti-schizophrenia agent CPZ consistently up-regulates the expression polySia at both cellular and animal levels.
Collapse
|
45
|
Huang X, Luo YL, Mao YS, Ji JL. The link between long noncoding RNAs and depression. Prog Neuropsychopharmacol Biol Psychiatry 2017; 73:73-78. [PMID: 27318257 DOI: 10.1016/j.pnpbp.2016.06.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 06/07/2016] [Accepted: 06/13/2016] [Indexed: 12/28/2022]
Abstract
The major depressive disorder (MDD) is a relatively common mental disorder from which that hundreds of million people have suffered, leading to displeasing life quality, which is characterized by health damage and even suicidal thoughts. The complicated development and functioning of MDD is still under exploration. Long noncoding RNA (lncRNAs) are highly expressed in the brain, could affect neural stem cell maintenance, neurogenesis and gliogenesis, brain patterning, synaptic and stress responses, and neural plasticity. The dysregulation of certain lncRNAs induces in neurodevelopmental, neurodegenerative and neuroimmunological disorders, primary brain tumors, and psychiatric diseases. Although advances have been made, no fully satisfactory treatments for major depression are available, further investigation is requested. And recently data showed that the expression level of the majority of lncRNAs demonstrated a clear tendency of upregulation, and the certain dysregulated miRNAs and lncRNAs in the MDD have been proved to have a co-synergism mechanism, that is why we speculate lncRNA might get the capability to regulate MDD. Few identified lncRNAs have been deeply studied in detailed experiments up until now, little predictions of their function have been raised, and further researches is calling for discover their signal pathway and related regulatory networks.
Collapse
Affiliation(s)
- Xiao Huang
- Department of Psychological Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan-Li Luo
- Department of Psychiatry, Tongji Hospital of Tongji University, Shanghai 200065, China
| | - Yue-Shi Mao
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jian-Lin Ji
- Department of Psychological Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
46
|
Trulioff A, Ermakov A, Malashichev Y. Primary Cilia as a Possible Link between Left-Right Asymmetry and Neurodevelopmental Diseases. Genes (Basel) 2017; 8:genes8020048. [PMID: 28125008 PMCID: PMC5333037 DOI: 10.3390/genes8020048] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/21/2016] [Accepted: 01/19/2017] [Indexed: 12/11/2022] Open
Abstract
Cilia have multiple functions in the development of the entire organism, and participate in the development and functioning of the central nervous system. In the last decade, studies have shown that they are implicated in the development of the visceral left-right asymmetry in different vertebrates. At the same time, some neuropsychiatric disorders, such as schizophrenia, autism, bipolar disorder, and dyslexia, are known to be associated with lateralization failure. In this review, we consider possible links in the mechanisms of determination of visceral asymmetry and brain lateralization, through cilia. We review the functions of seven genes associated with both cilia, and with neurodevelopmental diseases, keeping in mind their possible role in the establishment of the left-right brain asymmetry.
Collapse
Affiliation(s)
- Andrey Trulioff
- Department of Vertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Universitetskaya nab., 7/9, Saint Petersburg 199034, Russia.
| | - Alexander Ermakov
- Department of Vertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Universitetskaya nab., 7/9, Saint Petersburg 199034, Russia.
- Laboratory of Molecular Neurobiology, Department of Ecological Physiology, Institute of Experimental Medicine, ul. Akad. Pavlov, 12, Saint Petersburg 197376, Russia.
| | - Yegor Malashichev
- Department of Vertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Universitetskaya nab., 7/9, Saint Petersburg 199034, Russia.
- Laboratory of Molecular Neurobiology, Department of Ecological Physiology, Institute of Experimental Medicine, ul. Akad. Pavlov, 12, Saint Petersburg 197376, Russia.
| |
Collapse
|
47
|
Mulligan KA, Cheyette BNR. Neurodevelopmental Perspectives on Wnt Signaling in Psychiatry. MOLECULAR NEUROPSYCHIATRY 2017; 2:219-246. [PMID: 28277568 DOI: 10.1159/000453266] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mounting evidence indicates that Wnt signaling is relevant to pathophysiology of diverse mental illnesses including schizophrenia, bipolar disorder, and autism spectrum disorder. In the 35 years since Wnt ligands were first described, animal studies have richly explored how downstream Wnt signaling pathways affect an array of neurodevelopmental processes and how their disruption can lead to both neurological and behavioral phenotypes. Recently, human induced pluripotent stem cell (hiPSC) models have begun to contribute to this literature while pushing it in increasingly translational directions. Simultaneously, large-scale human genomic studies are providing evidence that sequence variation in Wnt signal pathway genes contributes to pathogenesis in several psychiatric disorders. This article reviews neurodevelopmental and postneurodevelopmental functions of Wnt signaling, highlighting mechanisms, whereby its disruption might contribute to psychiatric illness, and then reviews the most reliable recent genetic evidence supporting that mutations in Wnt pathway genes contribute to psychiatric illness. We are proponents of the notion that studies in animal and hiPSC models informed by the human genetic data combined with the deep knowledge base and tool kits generated over the last several decades of basic neurodevelopmental research will yield near-term tangible advances in neuropsychiatry.
Collapse
Affiliation(s)
- Kimberly A Mulligan
- Department of Biological Sciences, California State University, Sacramento, CA, USA
| | - Benjamin N R Cheyette
- Department of Psychiatry, Kavli Institute for Fundamental Neuroscience, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
48
|
Kassan A, Egawa J, Zhang Z, Almenar-Queralt A, Nguyen QM, Lajevardi Y, Kim K, Posadas E, Jeste DV, Roth DM, Patel PM, Patel HH, Head BP. Caveolin-1 regulation of disrupted-in-schizophrenia-1 as a potential therapeutic target for schizophrenia. J Neurophysiol 2017; 117:436-444. [PMID: 27832597 PMCID: PMC5253400 DOI: 10.1152/jn.00481.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 10/31/2016] [Indexed: 02/08/2023] Open
Abstract
Schizophrenia is a debilitating psychiatric disorder manifested in early adulthood. Disrupted-in-schizophrenia-1 (DISC1) is a susceptible gene for schizophrenia (Hodgkinson et al. 2004; Millar et al. 2000; St Clair et al. 1990) implicated in neuronal development, brain maturation, and neuroplasticity (Brandon and Sawa 2011; Chubb et al. 2008). Therefore, DISC1 is a promising candidate gene for schizophrenia, but the molecular mechanisms underlying its role in the pathogenesis of the disease are still poorly understood. Interestingly, caveolin-1 (Cav-1), a cholesterol binding and scaffolding protein, regulates neuronal signal transduction and promotes neuroplasticity. In this study we examined the role of Cav-1 in mediating DISC1 expression in neurons in vitro and the hippocampus in vivo. Overexpressing Cav-1 specifically in neurons using a neuron-specific synapsin promoter (SynCav1) increased expression of DISC1 and proteins involved in synaptic plasticity (PSD95, synaptobrevin, synaptophysin, neurexin, and syntaxin 1). Similarly, SynCav1-transfected differentiated human neurons derived from induced pluripotent stem cells (hiPSCs) exhibited increased expression of DISC1 and markers of synaptic plasticity. Conversely, hippocampi from Cav-1 knockout (KO) exhibited decreased expression of DISC1 and proteins involved in synaptic plasticity. Finally, SynCav1 delivery to the hippocampus of Cav-1 KO mice and Cav-1 KO neurons in culture restored expression of DISC1 and markers of synaptic plasticity. Furthermore, we found that Cav-1 coimmunoprecipitated with DISC1 in brain tissue. These findings suggest an important role by which neuron-targeted Cav-1 regulates DISC1 neurobiology with implications for synaptic plasticity. Therefore, SynCav1 might be a potential therapeutic target for restoring neuronal function in schizophrenia. NEW & NOTEWORTHY The present study is the first to demonstrate that caveolin-1 can regulate DISC1 expression in neuronal models. Furthermore, the findings are consistent across three separate neuronal models that include rodent neurons (in vitro and in vivo) and human differentiated neurons derived from induced pluripotent stem cells. These findings justify further investigation regarding the modulatory role by caveolin on synaptic function and as a potential therapeutic target for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Adam Kassan
- Department of Anesthesiology, University of California San Diego, La Jolla, California
- VA San Diego Healthcare System, San Diego, California
- Department of Psychiatry and the Sam and Rose Stein Institute for Research on Aging, University of California, San Diego, La Jolla, California
| | - Junji Egawa
- VA San Diego Healthcare System, San Diego, California
| | - Zheng Zhang
- VA San Diego Healthcare System, San Diego, California
| | - Angels Almenar-Queralt
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California; and
| | | | | | - Kaitlyn Kim
- VA San Diego Healthcare System, San Diego, California
| | | | - Dilip V Jeste
- Department of Psychiatry and the Sam and Rose Stein Institute for Research on Aging, University of California, San Diego, La Jolla, California
| | - David M Roth
- Department of Anesthesiology, University of California San Diego, La Jolla, California
- VA San Diego Healthcare System, San Diego, California
| | - Piyush M Patel
- Department of Anesthesiology, University of California San Diego, La Jolla, California
- VA San Diego Healthcare System, San Diego, California
| | - Hemal H Patel
- Department of Anesthesiology, University of California San Diego, La Jolla, California
- VA San Diego Healthcare System, San Diego, California
| | - Brian P Head
- Department of Anesthesiology, University of California San Diego, La Jolla, California;
- VA San Diego Healthcare System, San Diego, California
- Sanford Consortium for Regenerative Medicine, La Jolla, California
| |
Collapse
|
49
|
Powers A, Almli L, Smith A, Lori A, Leveille J, Ressler KJ, Jovanovic T, Bradley B. A genome-wide association study of emotion dysregulation: Evidence for interleukin 2 receptor alpha. J Psychiatr Res 2016; 83:195-202. [PMID: 27643478 PMCID: PMC5896292 DOI: 10.1016/j.jpsychires.2016.09.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 07/01/2016] [Accepted: 09/08/2016] [Indexed: 11/26/2022]
Abstract
Emotion dysregulation has been implicated as a risk factor for many psychiatric conditions. Therefore, examining genetic risk associated with emotion dysregulation could help inform cross-disorder risk more generally. A genome-wide association study (GWAS) of emotion dysregulation using single nucleotide polymorphism (SNP) array technology was conducted in a highly traumatized, minority, urban sample (N = 2600, males = 774). Post-hoc analyses examined associations between SNPs identified in the GWAS and current depression, posttraumatic stress disorder (PTSD), and history of suicide attempt. Methylation quantitative trait loci were identified and gene set enrichment analyses were used to broadly determine biological processes involved with these SNPs. Among males, SNP rs6602398, located within the interleukin receptor 2A gene, IL2RA, was significantly associated with emotion dysregulation (p = 1.1 × 10-8). Logistic regression analyses revealed this SNP was significantly associated with depression (Exp(B) = 2.67, p < 0.001) and PTSD (Exp(B) = 2.07, p < 0.01). This SNP was associated with differential DNA methylation (p < 0.05) suggesting it may be functionally active. Finally, through gene set enrichment analyses, ten psychiatric disease pathways (adjusted p < 0.01) and the calcium signaling pathway (adjusted p = 0.008) were significantly associated with emotion dysregulation. We found initial evidence for an association between emotion dysregulation and genetic risk loci that have already been implicated in medical disorders that have high comorbidity with psychiatric disorders. Our results provide further evidence that emotion dysregulation can be understood as a potential psychiatric cross-disorder risk factor, and that sex differences across these phenotypes may be critical. Continued research into genetic and biological risk associated with emotion dysregulation is needed.
Collapse
Affiliation(s)
- Abigail Powers
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, United States.
| | - Lynn Almli
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine
| | - Alicia Smith
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine
| | - Adriana Lori
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine
| | - Jen Leveille
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine
| | - Kerry J. Ressler
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine,McLean Hospital, Harvard Medical School
| | - Tanja Jovanovic
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine
| | - Bekh Bradley
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine,Atlanta VA Medical Center
| |
Collapse
|
50
|
Neuregulin-1 Regulates Cortical Inhibitory Neuron Dendrite and Synapse Growth through DISC1. Neural Plast 2016; 2016:7694385. [PMID: 27847649 PMCID: PMC5099462 DOI: 10.1155/2016/7694385] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 07/05/2016] [Accepted: 09/05/2016] [Indexed: 11/20/2022] Open
Abstract
Cortical inhibitory neurons play crucial roles in regulating excitatory synaptic networks and cognitive function and aberrant development of these cells have been linked to neurodevelopmental disorders. The secreted neurotrophic factor Neuregulin-1 (NRG1) and its receptor ErbB4 are established regulators of inhibitory neuron connectivity, but the developmental signalling mechanisms regulating this process remain poorly understood. Here, we provide evidence that NRG1-ErbB4 signalling functions through the multifunctional scaffold protein, Disrupted in Schizophrenia 1 (DISC1), to regulate the development of cortical inhibitory interneuron dendrite and synaptic growth. We found that NRG1 increases inhibitory neuron dendrite complexity and glutamatergic synapse formation onto inhibitory neurons and that this effect is blocked by expression of a dominant negative DISC1 mutant, or DISC1 knockdown. We also discovered that NRG1 treatment increases DISC1 expression and its localization to glutamatergic synapses being made onto cortical inhibitory neurons. Mechanistically, we determined that DISC1 binds ErbB4 within cortical inhibitory neurons. Collectively, these data suggest that a NRG1-ErbB4-DISC1 signalling pathway regulates the development of cortical inhibitory neuron dendrite and synaptic growth. Given that NRG1, ErbB4, and DISC1 are schizophrenia-linked genes, these findings shed light on how independent risk factors may signal in a common developmental pathway that contributes to neural connectivity defects and disease pathogenesis.
Collapse
|