1
|
Zhou Z, Yan Y, Gu H, Sun R, Liao Z, Xue K, Tang C. Dopamine in the prefrontal cortex plays multiple roles in the executive function of patients with Parkinson's disease. Neural Regen Res 2024; 19:1759-1767. [PMID: 38103242 PMCID: PMC10960281 DOI: 10.4103/1673-5374.389631] [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: 04/11/2023] [Revised: 08/05/2023] [Accepted: 10/10/2023] [Indexed: 12/18/2023] Open
Abstract
Parkinson's disease can affect not only motor functions but also cognitive abilities, leading to cognitive impairment. One common issue in Parkinson's disease with cognitive dysfunction is the difficulty in executive functioning. Executive functions help us plan, organize, and control our actions based on our goals. The brain area responsible for executive functions is called the prefrontal cortex. It acts as the command center for the brain, especially when it comes to regulating executive functions. The role of the prefrontal cortex in cognitive processes is influenced by a chemical messenger called dopamine. However, little is known about how dopamine affects the cognitive functions of patients with Parkinson's disease. In this article, the authors review the latest research on this topic. They start by looking at how the dopaminergic system, is altered in Parkinson's disease with executive dysfunction. Then, they explore how these changes in dopamine impact the synaptic structure, electrical activity, and connection components of the prefrontal cortex. The authors also summarize the relationship between Parkinson's disease and dopamine-related cognitive issues. This information may offer valuable insights and directions for further research and improvement in the clinical treatment of cognitive impairment in Parkinson's disease.
Collapse
Affiliation(s)
- Zihang Zhou
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yalong Yan
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Heng Gu
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Ruiao Sun
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Zihan Liao
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Ke Xue
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Chuanxi Tang
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| |
Collapse
|
2
|
Gao H, Zhang Y, Luo D, Xu J, Tan S, Li Y, Qi W, Zhai Q, Wang Q. Activation of the Hippocampal DRD2 Alleviates Neuroinflammation, Synaptic Plasticity Damage and Cognitive Impairment After Sleep Deprivation. Mol Neurobiol 2023; 60:7208-7221. [PMID: 37543530 DOI: 10.1007/s12035-023-03514-5] [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: 05/04/2023] [Accepted: 07/15/2023] [Indexed: 08/07/2023]
Abstract
Sleep loss is commonplace nowadays and profoundly impacts cognition. Dopamine receptor D2 (DRD2) makes a specific contribution to cognition, although the precise mechanism underlying how DRD2 affects the cognitive process after sleep deprivation remains unclear. Herein, we observed cognitive impairment and impaired synaptic plasticity, including downregulation of synaptophysin and PSD95, decreased postsynaptic density thickness, neuron complexity, and spine density in chronic sleep restriction (CSR) mice. We also observed downregulated hippocampal DRD2 and Cryab expression in the CSR mice. Meanwhile, NF-κB translocation from the cytoplasm to the nucleus occurred, indicating that neuroinflammation ensued. However, hippocampal delivery of the DRD2 agonist quinpirole effectively rescued these changes. In vitro, quinpirole treatment significantly decreased the release of proinflammatory cytokines in microglial supernatant, indicating a potential anti-neuroinflammatory effect of Drd2/Cryab/NF-κB in CSR mice. Our study provided the evidence that activation of the Drd2 may relieve neuroinflammation and improve sleep deprivation-induced cognitive deficits.
Collapse
Affiliation(s)
- Hui Gao
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Department of Anaesthesiology, Yan'an University Affiliated Hospital, Yan'an, 716000, China
| | - Yuxin Zhang
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200434, China
| | - Danlei Luo
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jing Xu
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shuwen Tan
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ying Li
- Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Wanling Qi
- Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Qian Zhai
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Qiang Wang
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
3
|
Seeman MV. Schizophrenia in Women: Clinical Considerations. Psychiatr Clin North Am 2023; 46:475-486. [PMID: 37500245 DOI: 10.1016/j.psc.2023.04.005] [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] [Indexed: 07/29/2023]
Abstract
Men and women, for biologic and sociocultural reasons, differ in the nature of their risks for schizophrenia and also in their care needs. Women with schizophrenia have several reproduction-associated risks and care needs that require special clinical consideration. They also have several specific risks related to antipsychotics and gender-associated needs not necessarily related to biology. These require clinicians' diagnostic acumen, treatment skills, cultural sensitivity, and advocacy know-how. Although this does not pertain to everyone, awareness on the part of clinicians is essential. This article addresses the current evidence for difference.
Collapse
Affiliation(s)
- Mary V Seeman
- Department of Psychiatry, University of Toronto, Toronto, Ontario M5P3L6, Canada.
| |
Collapse
|
4
|
Synaptic plasticity in Schizophrenia pathophysiology. IBRO Neurosci Rep 2023. [DOI: 10.1016/j.ibneur.2023.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
5
|
Male DAT Val559 Mice Exhibit Compulsive Behavior under Devalued Reward Conditions Accompanied by Cellular and Pharmacological Changes. Cells 2022; 11:cells11244059. [PMID: 36552823 PMCID: PMC9777203 DOI: 10.3390/cells11244059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Identified across multiple psychiatric disorders, the dopamine (DA) transporter (DAT) Ala559Val substitution triggers non-vesicular, anomalous DA efflux (ADE), perturbing DA neurotransmission and behavior. We have shown that DAT Val559 mice display a waiting impulsivity and changes in cognitive performance associated with enhanced reward motivation. Here, utilizing a within-subject, lever-pressing paradigm designed to bias the formation of goal-directed or habitual behavior, we demonstrate that DAT Val559 mice modulate their nose poke behavior appropriately to match context, but demonstrate a perseverative checking behavior. Although DAT Val559 mice display no issues with the cognitive flexibility required to acquire and re-learn a visual pairwise discrimination task, devaluation of reward evoked habitual reward seeking in DAT Val559 mutants in operant tasks regardless of reinforcement schedule. The direct DA agonist apomorphine also elicits locomotor stereotypies in DAT Val559, but not WT mice. Our observation that dendritic spine density is increased in the dorsal medial striatum (DMS) of DAT Val559 mice speaks to an imbalance in striatal circuitry that might underlie the propensity of DAT Val559 mutants to exhibit compulsive behaviors when reward is devalued. Thus, DAT Val559 mice represent a model for dissection of how altered DA signaling perturbs circuits that normally balance habitual and goal-directed behaviors.
Collapse
|
6
|
Zhu F, Xiao Y, Tao B, Gao Z, Gao X, Zhao Q, Zhang Q, Tang B, Zhang X, Zhao Y, Bishop JR, Sweeney JA, Lui S. Radiomic features of gray matter in never-treated first-episode schizophrenia. Cereb Cortex 2022; 33:5957-5967. [PMID: 36513368 DOI: 10.1093/cercor/bhac474] [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: 08/12/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 12/15/2022] Open
Abstract
Alterations of radiomic features (RFs) in gray matter are observed in schizophrenia, of which the results may be limited by small study samples and confounding effects of drug therapies. We tested for RFs alterations of gray matter in never-treated first-episode schizophrenia (NT-FES) patients and examined their associations with known gene expression profiles. RFs were examined in the first sample with 197 NT-FES and 178 healthy controls (HCs) and validated in the second independent sample (90 NT-FES and 74 HCs). One-year follow-up data were available from 87 patients to determine whether RFs were associated with treatment outcomes. Associations between identified RFs in NT-FES and gene expression profiles were evaluated. NT-FES exhibited alterations of 30 RFs, with the greatest involvement of microstructural heterogeneity followed by measures of brain region shape. The identified RFs were mainly located in the central executive network, frontal-temporal network, and limbic system. Two baseline RFs with the involvement of microstructural heterogeneity predicted treatment response with moderate accuracy (78% for the first sample, 70% for the second sample). Exploratory analyses indicated that RF alterations were spatially related to the expression of schizophrenia risk genes. In summary, the present findings link brain abnormalities in schizophrenia with molecular features and treatment response.
Collapse
Affiliation(s)
- Fei Zhu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yuan Xiao
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Bo Tao
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ziyang Gao
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xin Gao
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Qiannan Zhao
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Qi Zhang
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Biqiu Tang
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | | | - Yu Zhao
- Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jeffrey R Bishop
- Department of Experimental and Clinical Pharmacology and Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - John A Sweeney
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China.,Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH 45219, USA
| | - Su Lui
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
7
|
Zhang K, Liao P, Wen J, Hu Z. Synaptic plasticity in schizophrenia pathophysiology. IBRO Neurosci Rep 2022; 13:478-487. [PMID: 36590092 PMCID: PMC9795311 DOI: 10.1016/j.ibneur.2022.10.008] [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: 05/22/2022] [Accepted: 10/21/2022] [Indexed: 11/05/2022] Open
Abstract
Schizophrenia is a severe neuropsychiatric syndrome with psychotic behavioral abnormalities and marked cognitive deficits. It is widely accepted that genetic and environmental factors contribute to the onset of schizophrenia. However, the etiology and pathology of the disease remain largely unexplored. Recently, the synaptopathology and the dysregulated synaptic plasticity and function have emerging as intriguing and prominent biological mechanisms of schizophrenia pathogenesis. Synaptic plasticity is the ability of neurons to change the strength of their connections in response to internal or external stimuli, which is essential for brain development and function, learning and memory, and vast majority of behavior responses relevant to psychiatric diseases including schizophrenia. Here, we reviewed molecular and cellular mechanisms of the multiple forms synaptic plasticity, and the functional regulations of schizophrenia-risk factors including disease susceptible genes and environmental alterations on synaptic plasticity and animal behavior. Recent genome-wide association studies have provided fruitful findings of hundreds of risk gene variances associated with schizophrenia, thus further clarifying the role of these disease-risk genes in synaptic transmission and plasticity will be beneficial to advance our understanding of schizophrenia pathology, as well as the molecular mechanism of synaptic plasticity.
Collapse
Affiliation(s)
- Kexuan Zhang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410008, Hunan, PR China
| | - Panlin Liao
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Jin Wen
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Zhonghua Hu
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410008, Hunan, PR China,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha 410008, Hunan, PR China,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha 410008, Hunan, PR China,Correspondence to: Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, PR China.
| |
Collapse
|
8
|
Li C, Wu XJ, Li W. Neuropeptide S promotes maintenance of newly formed dendritic spines and performance improvement after motor learning in mice. Peptides 2022; 156:170860. [PMID: 35970276 DOI: 10.1016/j.peptides.2022.170860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/18/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
Neuropeptide S (NPS), an endogenous neuropeptide consisting of 20 amino acids, selectively binds and activates G protein-coupled receptor named neuropeptide S receptor (NPSR) to regulate a variety of physiological functions. NPS/NPSR system has been shown to play a pivotal role in regulating learning and memory in rodents. However, it remains unclear that how NPS/NPSR system affects neuronal functions and synaptic plasticity after learning. We found that intracerebroventricular (i.c.v.) injection of NPS promoted performance improvement and reduced sleep duration after motor learning, which could be blocked by pre-treatment with intraperitoneal (i.p.) injection of NPSR antagonist SHA 68. Using intravital two-photon imaging, we examined the effect of NPS on the postsynaptic dendritic spines of layer V pyramidal neurons in the mouse primary motor cortex after motor learning. We found that i.c.v. injection of NPS strengthened learning-induce new spines and facilitated their survival over time. Furthermore, i.c.v. injection of NPS increased calcium activity of apical dendrites and dendritic spines of layer V pyramidal neurons in the mouse primary motor cortex during the running period. These findings suggest that activation of NPSR by NPS increases synaptic calcium activity and learning-related synapse maintenance, thereby contributing to performance improvement after motor learning.
Collapse
Affiliation(s)
- Cong Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Xu-Jun Wu
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Wei Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China; School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| |
Collapse
|
9
|
Zhang W, Lei M, Wen Q, Zhang D, Qin G, Zhou J, Chen L. Dopamine receptor D2 regulates GLUA1-containing AMPA receptor trafficking and central sensitization through the PI3K signaling pathway in a male rat model of chronic migraine. J Headache Pain 2022; 23:98. [PMID: 35948867 PMCID: PMC9364568 DOI: 10.1186/s10194-022-01469-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/30/2022] [Indexed: 11/10/2022] Open
Abstract
Background The pathogenesis of chronic migraine remains unresolved. Recent studies have affirmed the contribution of GLUA1-containing AMPA receptors to chronic migraine. The dopamine D2 receptor, a member of G protein-coupled receptor superfamily, has been proven to have an analgesic effect on pathological headaches. The present work investigated the exact role of the dopamine D2 receptor in chronic migraine and its effect on GLUA1-containing AMPA receptor trafficking. Methods A chronic migraine model was established by repeated inflammatory soup stimulation. Mechanical, periorbital, and thermal pain thresholds were assessed by the application of von Frey filaments and radiant heat. The mRNA and protein expression levels of the dopamine D2 receptor were analyzed by qRT‒PCR and western blotting. Colocalization of the dopamine D2 receptor and the GLUA1-containing AMPAR was observed by immunofluorescence. A dopamine D2 receptor agonist (quinpirole) and antagonist (sulpiride), a PI3K inhibitor (LY294002), a PI3K pathway agonist (740YP), and a GLUA1-containing AMPAR antagonist (NASPM) were administered to confirm the effects of the dopamine D2 receptor, the PI3K pathway and GULA1 on central sensitization and the GLUA1-containing AMPAR trafficking. Transmission electron microscopy and Golgi-Cox staining were applied to assess the impact of the dopamine D2 receptor and PI3K pathway on synaptic morphology. Fluo-4-AM was used to clarify the role of the dopamine D2 receptor and PI3K signaling on neuronal calcium influx. The Src family kinase (SFK) inhibitor PP2 was used to explore the effect of Src kinase on GLUA1-containing AMPAR trafficking and the PI3K signaling pathway. Results Inflammatory soup stimulation significantly reduced pain thresholds in rats, accompanied by an increase in PI3K-P110β subunit expression, loss of dopamine receptor D2 expression, and enhanced GLUA1-containing AMPA receptor trafficking in the trigeminal nucleus caudalis (TNC). The dopamine D2 receptor colocalized with the GLUA1-containing AMPA receptor in the TNC; quinpirole, LY294002, and NASPM alleviated pain hypersensitivity and reduced GLUA1-containing AMPA receptor trafficking in chronic migraine rats. Sulpiride aggravated pain hypersensitivity and enhanced GLUA1 trafficking in CM rats. Importantly, the anti-injury and central sensitization-mitigating effects of quinpirole were reversed by 740YP. Both quinpirole and LY294002 inhibited calcium influx to neurons and modulated the synaptic morphology in the TNC. Additional results suggested that DRD2 may regulate PI3K signaling through Src family kinases. Conclusion Modulation of GLUA1-containing AMPA receptor trafficking and central sensitization by the dopamine D2 receptor via the PI3K signaling pathway may contribute to the pathogenesis of chronic migraine in rats, and the dopamine D2 receptor could be a valuable candidate for chronic migraine treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s10194-022-01469-x.
Collapse
Affiliation(s)
- Wei Zhang
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, 1st You Yi Road, Yu Zhong District, Chongqing, 400016, China
| | - Ming Lei
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, 1st You Yi Road, Yu Zhong District, Chongqing, 400016, China
| | - Qianwen Wen
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, 1st You Yi Road, Yu Zhong District, Chongqing, 400016, China
| | - Dunke Zhang
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, 1st You Yi Road, Yu Zhong District, Chongqing, 400016, China
| | - Guangcheng Qin
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, 1st You Yi Road, Yu Zhong District, Chongqing, 400016, China
| | - Jiying Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lixue Chen
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, 1st You Yi Road, Yu Zhong District, Chongqing, 400016, China.
| |
Collapse
|
10
|
Glucocorticoid Receptor-Dependent Astrocytes Mediate Stress Vulnerability. Biol Psychiatry 2022; 92:204-215. [PMID: 35151464 DOI: 10.1016/j.biopsych.2021.11.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/04/2021] [Accepted: 11/28/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Major depressive disorder is a devastating psychiatric illness that affects approximately 17% of the population worldwide. Astrocyte dysfunction has been implicated in its pathophysiology. Traumatic experiences and stress contribute to the onset of major depressive disorder, but how astrocytes respond to stress is poorly understood. METHODS Using Western blotting analysis, we identified that stress vulnerability was associated with reduced astrocytic glucocorticoid receptor (GR) expression in mouse models of depression. We further investigated the functions of astrocytic GRs in regulating depression and the underlying mechanisms by using a combination of behavioral studies, fiber photometry, biochemical experiments, and RNA sequencing methods. RESULTS GRs in astrocytes were more sensitive to stress than those in neurons. GR absence in astrocytes induced depressive-like behaviors, whereas restoring astrocytic GR expression in the medial prefrontal cortex prevented the depressive-like phenotype. Furthermore, we found that GRs in the medial prefrontal cortex affected astrocytic Ca2+ activity and dynamic ATP (adenosine 5'-triphosphate) release in response to stress. RNA sequencing of astrocytes isolated from GR deletion mice identified the PI3K-Akt (phosphoinositide 3-kinase-Akt) signaling pathway, which was required for astrocytic GR-mediated ATP release. CONCLUSIONS These findings reveal that astrocytic GRs play an important role in stress response and that reduced astrocytic GR expression in the stressed subject decreases ATP release to mediate stress vulnerability.
Collapse
|
11
|
Extrastriatal dopamine D2/3 receptor binding, functional connectivity, and autism socio-communicational deficits: a PET and fMRI study. Mol Psychiatry 2022; 27:2106-2113. [PMID: 35181754 DOI: 10.1038/s41380-022-01464-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/12/2022] [Accepted: 01/26/2022] [Indexed: 11/08/2022]
Abstract
The social motivation hypothesis of autism proposes that social communication symptoms in autism-spectrum disorder (ASD) stem from atypical social attention and reward networks, where dopamine acts as a crucial mediator. However, despite evidence indicating that individuals with ASD show atypical activation in extrastriatal regions while processing reward and social stimuli, no previous studies have measured extrastriatal dopamine D2/3 receptor (D2/3R) availability in ASD. Here, we investigated extrastriatal D2/3R availability in individuals with ASD and its association with ASD social communication symptoms using positron emission tomography (PET). Moreover, we employed a whole-brain multivariate pattern analysis of resting-state functional magnetic resonance imaging (fMRI) to identify regions where functional connectivity atypically correlates with D2/3R availability depending on ASD diagnosis. Twenty-two psychotropic-free males with ASD and 24 age- and intelligence quotient-matched typically developing males underwent [11C]FLB457 PET, fMRI, and clinical symptom assessment. Participants with ASD showed lower D2/3R availability throughout the D2/3R-rich extrastriatal regions of the dopaminergic pathways. Among these, the posterior region of the thalamus, which primarily comprises the pulvinar, displayed the largest effect size for the lower D2/3R availability, which correlated with a higher score on the Social Affect domain of the Autism Diagnostic Observation Schedule-2 in participants with ASD. Moreover, lower D2/3R availability was correlated with lower functional connectivity of the thalamus-superior temporal sulcus and cerebellum-medial occipital cortex, specifically in individuals with ASD. The current findings provide novel molecular evidence for the social motivation theory of autism and offer a novel therapeutic target.
Collapse
|
12
|
Steinecke A, Bolton MM, Taniguchi H. Neuromodulatory control of inhibitory network arborization in the developing postnatal neocortex. SCIENCE ADVANCES 2022; 8:eabe7192. [PMID: 35263136 PMCID: PMC8906727 DOI: 10.1126/sciadv.abe7192] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/19/2022] [Indexed: 06/14/2023]
Abstract
Interregional neuronal communication is pivotal to instructing and adjusting cortical circuit assembly. Subcortical neuromodulatory systems project long-range axons to the cortex and affect cortical processing. However, their roles and signaling mechanisms in cortical wiring remain poorly understood. Here, we explored whether and how the cholinergic system regulates inhibitory axonal ramification of neocortical chandelier cells (ChCs), which control spike generation by innervating axon initial segments of pyramidal neurons. We found that acetylcholine (ACh) signaling through nicotinic ACh receptors (nAChRs) and downstream T-type voltage-dependent calcium (Ca2+) channels cell-autonomously controls axonal arborization in developing ChCs through regulating filopodia initiation. This signaling axis shapes the basal Ca2+ level range in varicosities where filopodia originate. Furthermore, the normal development of ChC axonal arbors requires proper levels of activity in subcortical cholinergic neurons. Thus, the cholinergic system regulates inhibitory network arborization in the developing neocortex and may tune cortical circuit properties depending on early-life experiences.
Collapse
Affiliation(s)
- André Steinecke
- Development and Function of Inhibitory Neural Circuits, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - McLean M. Bolton
- Disorders of Neural Circuit Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Hiroki Taniguchi
- Development and Function of Inhibitory Neural Circuits, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| |
Collapse
|
13
|
Fei E, Chen P, Zhang Q, Zhong Y, Zhou T. Protein kinase B/Akt1 phosphorylates dysbindin-1A at serine 10 to regulate neuronal development. Neuroscience 2022; 490:66-78. [DOI: 10.1016/j.neuroscience.2022.01.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 01/05/2023]
|
14
|
Caragea VM, Manahan-Vaughan D. Bidirectional Regulation of Hippocampal Synaptic Plasticity and Modulation of Cumulative Spatial Memory by Dopamine D2-Like Receptors. Front Behav Neurosci 2022; 15:803574. [PMID: 35095441 PMCID: PMC8789653 DOI: 10.3389/fnbeh.2021.803574] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/20/2021] [Indexed: 01/11/2023] Open
Abstract
Dopamine is a key factor in the enablement of cognition and hippocampal information processing. Its action in the hippocampus is mediated by D1/D5 and D2-like (D2, D3, D4) receptors. While D1/D5-receptors are well recognized as strong modulators of hippocampal synaptic plasticity and information storage, much less is known about the role of D2-like receptors (D2R) in these processes. Here, we explored to what extent D2R contribute to synaptic plasticity and cumulative spatial memory derived from semantic and episodic-like information storage. In freely behaving adult rats, we also assessed to what extent short and long-term forms of synaptic plasticity are influenced by pharmacological activation or blockade of D2R. Antagonism of D2R by means of intracerebral treatment with remoxipride, completely prevented the expression of both short-term (<1 h) and long-term potentiation (>4 h), as well as the expression of short-term depression (STD, <1 h) in the hippocampal CA1 region. Scrutiny of involvement of D2R in spatial learning revealed that D2R-antagonism prevented retention of a semantic spatial memory task, and also significantly impaired retention of recent spatiotemporal aspects of an episodic-like memory task. Taken together, these findings indicate that D2R are required for bidirectional synaptic plasticity in the hippocampal CA1 region. Furthermore, they are critically involved in enabling cumulative and episodic-like forms of spatial learning.
Collapse
Affiliation(s)
- Violeta-Maria Caragea
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
- International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Denise Manahan-Vaughan
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
- International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
- *Correspondence: Denise Manahan-Vaughan
| |
Collapse
|
15
|
Gao WJ, Yang SS, Mack NR, Chamberlin LA. Aberrant maturation and connectivity of prefrontal cortex in schizophrenia-contribution of NMDA receptor development and hypofunction. Mol Psychiatry 2022; 27:731-743. [PMID: 34163013 PMCID: PMC8695640 DOI: 10.1038/s41380-021-01196-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023]
Abstract
The neurobiology of schizophrenia involves multiple facets of pathophysiology, ranging from its genetic basis over changes in neurochemistry and neurophysiology, to the systemic level of neural circuits. Although the precise mechanisms associated with the neuropathophysiology remain elusive, one essential aspect is the aberrant maturation and connectivity of the prefrontal cortex that leads to complex symptoms in various stages of the disease. Here, we focus on how early developmental dysfunction, especially N-methyl-D-aspartate receptor (NMDAR) development and hypofunction, may lead to the dysfunction of both local circuitry within the prefrontal cortex and its long-range connectivity. More specifically, we will focus on an "all roads lead to Rome" hypothesis, i.e., how NMDAR hypofunction during development acts as a convergence point and leads to local gamma-aminobutyric acid (GABA) deficits and input-output dysconnectivity in the prefrontal cortex, which eventually induce cognitive and social deficits. Many outstanding questions and hypothetical mechanisms are listed for future investigations of this intriguing hypothesis that may lead to a better understanding of the aberrant maturation and connectivity associated with the prefrontal cortex.
Collapse
Affiliation(s)
- Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| | - Sha-Sha Yang
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Nancy R Mack
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Linda A Chamberlin
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| |
Collapse
|
16
|
Jun R, Zhang W, Beacher NJ, Zhang Y, Li Y, Lin DT. Dysbindin-1, BDNF, and GABAergic Transmission in Schizophrenia. Front Psychiatry 2022; 13:876749. [PMID: 35815020 PMCID: PMC9258742 DOI: 10.3389/fpsyt.2022.876749] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Schizophrenia is a psychiatric disorder characterized by hallucinations, anhedonia, disordered thinking, and cognitive impairments. Both genetic and environmental factors contribute to schizophrenia. Dysbindin-1 (DTNBP1) and brain-derived neurotrophic factor (BDNF) are both genetic factors associated with schizophrenia. Mice lacking Dtnbp1 showed behavioral deficits similar to human patients suffering from schizophrenia. DTNBP1 plays important functions in synapse formation and maintenance, receptor trafficking, and neurotransmitter release. DTNBP1 is co-assembled with 7 other proteins into a large protein complex, known as the biogenesis of lysosome-related organelles complex-1 (BLOC-1). Large dense-core vesicles (LDCVs) are involved in the secretion of hormones and neuropeptides, including BDNF. BDNF plays important roles in neuronal development, survival, and synaptic plasticity. BDNF is also critical in maintaining GABAergic inhibitory transmission in the brain. Two studies independently showed that DTNBP1 mediated activity-dependent BDNF secretion to maintain inhibitory transmission. Imbalance of excitatory and inhibitory neural activities is thought to contribute to schizophrenia. In this mini-review, we will discuss a potential pathogenetic mechanism for schizophrenia involving DTNBP1, BDNF, and inhibitory transmission. We will also discuss how these processes are interrelated and associated with a higher risk of schizophrenia development.
Collapse
Affiliation(s)
- Rachel Jun
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Wen Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Nicholas J Beacher
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Yan Zhang
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Yun Li
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
| | - Da-Ting Lin
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
17
|
Zhang YQ, Lin WP, Huang LP, Zhao B, Zhang CC, Yin DM. Dopamine D2 receptor regulates cortical synaptic pruning in rodents. Nat Commun 2021; 12:6444. [PMID: 34750364 PMCID: PMC8576001 DOI: 10.1038/s41467-021-26769-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 10/22/2021] [Indexed: 12/22/2022] Open
Abstract
Synaptic pruning during adolescence is important for appropriate neurodevelopment and synaptic plasticity. Aberrant synaptic pruning may underlie a variety of brain disorders such as schizophrenia, autism and anxiety. Dopamine D2 receptor (Drd2) is associated with several neuropsychiatric diseases and is the target of some antipsychotic drugs. Here we generate self-reporting Drd2 heterozygous (SR-Drd2+/-) rats to simultaneously visualize Drd2-positive neurons and downregulate Drd2 expression. Time course studies on the developing anterior cingulate cortex (ACC) from control and SR-Drd2+/- rats reveal important roles of Drd2 in regulating synaptic pruning rather than synapse formation. Drd2 also regulates LTD, a form of synaptic plasticity which includes some similar cellular/biochemical processes as synaptic pruning. We further demonstrate that Drd2 regulates synaptic pruning via cell-autonomous mechanisms involving activation of mTOR signaling. Deficits of Drd2-mediated synaptic pruning in the ACC during adolescence lead to hyper-glutamatergic function and anxiety-like behaviors in adulthood. Taken together, our results demonstrate important roles of Drd2 in cortical synaptic pruning.
Collapse
Affiliation(s)
- Ya-Qiang Zhang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, 200062, Shanghai, China
| | - Wei-Peng Lin
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, 200062, Shanghai, China
- Joint Translational Science and Technology Research Institute, East China Normal University, 200062, Shanghai, China
| | - Li-Ping Huang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, 200062, Shanghai, China
| | - Bing Zhao
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, 200062, Shanghai, China
| | - Cheng-Cheng Zhang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, 200062, Shanghai, China
| | - Dong-Min Yin
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, 200062, Shanghai, China.
| |
Collapse
|
18
|
Dysbindin-1 regulates mitochondrial fission and gamma oscillations. Mol Psychiatry 2021; 26:4633-4651. [PMID: 33589740 PMCID: PMC8364574 DOI: 10.1038/s41380-021-01038-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 01/08/2021] [Accepted: 01/25/2021] [Indexed: 12/11/2022]
Abstract
Mitochondria are cellular ATP generators. They are dynamic structures undergoing fission and fusion. While much is known about the mitochondrial fission machinery, the mechanism of initiating fission and the significance of fission to neurophysiology are largely unclear. Gamma oscillations are synchronized neural activities that impose a great energy challenge to synapses. The cellular mechanism of fueling gamma oscillations has yet to be defined. Here, we show that dysbindin-1, a protein decreased in the brain of individuals with schizophrenia, is required for neural activity-induced fission by promoting Drp1 oligomerization. This process is engaged by gamma-frequency activities and in turn, supports gamma oscillations. Gamma oscillations and novel object recognition are impaired in dysbindin-1 null mice. These defects can be ameliorated by increasing mitochondrial fission. These findings identify a molecular mechanism for activity-induced mitochondrial fission, a role of mitochondrial fission in gamma oscillations, and mitochondrial fission as a potential target for improving cognitive functions.
Collapse
|
19
|
Zhang Z, Ye M, Li Q, You Y, Yu H, Ma Y, Mei L, Sun X, Wang L, Yue W, Li R, Li J, Zhang D. The Schizophrenia Susceptibility Gene OPCML Regulates Spine Maturation and Cognitive Behaviors through Eph-Cofilin Signaling. Cell Rep 2020; 29:49-61.e7. [PMID: 31577955 DOI: 10.1016/j.celrep.2019.08.091] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 07/09/2019] [Accepted: 08/27/2019] [Indexed: 12/30/2022] Open
Abstract
Previous genetic and biological evidence converge on the involvement of synaptic dysfunction in schizophrenia, and OPCML, encoding a synaptic membrane protein, is reported to be genetically associated with schizophrenia. However, its role in the pathophysiology of schizophrenia remains largely unknown. Here, we found that Opcml is strongly expressed in the mouse hippocampus; ablation of Opcml leads to reduced phosphorylated cofilin and dysregulated F-actin dynamics, which disturbs the spine maturation. Furthermore, Opcml interacts with EphB2 to control the stability of spines by regulating the ephrin-EphB2-cofilin signaling pathway. Opcml-deficient mice display impaired cognitive behaviors and abnormal sensorimotor gating, which are similar to features in neuropsychiatric disorders such as schizophrenia. Notably, the administration of aripiprazole partially restores the abnormal behaviors in Opcml-/- mice by increasing the phosphorylated cofilin level and facilitating spine maturation. We demonstrated a critical role of the schizophrenia-susceptible gene OPCML in spine maturation and cognitive behaviors via regulating the ephrin-EphB2-cofilin signaling pathway, providing further insights into the characteristics of schizophrenia.
Collapse
Affiliation(s)
- Zhengrong Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China; National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Maoqing Ye
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Cardiology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Qiongwei Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Yang You
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Hao Yu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Yuanlin Ma
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Liwei Mei
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Xiaqin Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Lifang Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Weihua Yue
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Rena Li
- National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Jun Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China.
| | - Dai Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| |
Collapse
|
20
|
Zheng P, Su QP, Jin D, Yu Y, Huang XF. Prevention of Neurite Spine Loss Induced by Dopamine D2 Receptor Overactivation in Striatal Neurons. Front Neurosci 2020; 14:642. [PMID: 32655360 PMCID: PMC7324769 DOI: 10.3389/fnins.2020.00642] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 05/25/2020] [Indexed: 11/22/2022] Open
Abstract
Psychosis has been considered a disorder of impaired neuronal connectivity. Evidence for excessive formation of dopamine D2 receptor (D2R) – disrupted in schizophrenia 1 (DISC1) complexes has led to a new perspective on molecular mechanisms involved in psychotic symptoms. Here, we investigated how excessive D2R–DISC1 complex formation induced by D2R agonist quinpirole affects neurite growth and dendritic spines in striatal neurons. Fluorescence resonance energy transfer (FRET), stochastic optical reconstruction microscopy (STORM), and cell penetrating-peptide delivery were used to study the cultured striatal neurons from mouse pups. Using these striatal neurons, our study showed that: (1) D2R interacted with DISC1 in dendritic spines, neurites and soma of cultured striatal neurons; (2) D2R and DISC1 complex accumulated in clusters in dendritic spines of striatal neurons and the number of the complex were reduced after application of TAT-D2pep; (3) uncoupling D2R–DISC1 complexes by TAT-D2pep protected neuronal morphology and dendritic spines; and (4) TAT-D2pep prevented neurite and dendritic spine loss, which was associated with restoration of expression levels of synaptophysin and PSD-95. In addition, we found that Neuropeptide Y (NPY) and GSK3β were involved in the protective effects of TAT-D2pep on the neurite spines of striatal spiny projection neurons. Thus, our results may offer a new strategy for precisely treating neurite spine deficits associated with schizophrenia.
Collapse
Affiliation(s)
- Peng Zheng
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Qian Peter Su
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Dayong Jin
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
21
|
Areal LB, Blakely RD. Neurobehavioral changes arising from early life dopamine signaling perturbations. Neurochem Int 2020; 137:104747. [PMID: 32325191 PMCID: PMC7261509 DOI: 10.1016/j.neuint.2020.104747] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/11/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022]
Abstract
Dopamine (DA) signaling is critical to the modulation of multiple brain functions including locomotion, reinforcement, attention and cognition. The literature provides strong evidence that altered DA availability and actions can impact normal neurodevelopment, with both early and enduring consequences on anatomy, physiology and behavior. An appreciation for the developmental contributions of DA signaling to brain development is needed to guide efforts to preclude and remedy neurobehavioral disorders, such as attention-deficit/hyperactivity disorder, addiction, bipolar disorder, schizophrenia and autism spectrum disorder, each of which exhibits links to DA via genetic, cellular and/or pharmacological findings. In this review, we highlight research pursued in preclinical models that use genetic and pharmacological approaches to manipulate DA signaling at sensitive developmental stages, leading to changes at molecular, circuit and/or behavioral levels. We discuss how these alterations can be aligned with traits displayed by neuropsychiatric diseases. Lastly, we review human studies that evaluate contributions of developmental perturbations of DA systems to increased risk for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Lorena B Areal
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Randy D Blakely
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, 33458, USA; Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA.
| |
Collapse
|
22
|
An in vivo brain-bacteria interface: the developing brain as a key regulator of innate immunity. NPJ Regen Med 2020; 5:2. [PMID: 32047653 PMCID: PMC7000827 DOI: 10.1038/s41536-020-0087-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 12/20/2019] [Indexed: 01/11/2023] Open
Abstract
Infections have numerous effects on the brain. However, possible roles of the brain in protecting against infection, and the developmental origin and role of brain signaling in immune response, are largely unknown. We exploited a unique Xenopus embryonic model to reveal control of innate immune response to pathogenic E. coli by the developing brain. Using survival assays, morphological analysis of innate immune cells and apoptosis, and RNA-seq, we analyzed combinations of infection, brain removal, and tail-regenerative response. Without a brain, survival of embryos injected with bacteria decreased significantly. The protective effect of the developing brain was mediated by decrease of the infection-induced damage and of apoptosis, and increase of macrophage migration, as well as suppression of the transcriptional consequences of the infection, all of which decrease susceptibility to pathogen. Functional and pharmacological assays implicated dopamine signaling in the bacteria–brain–immune crosstalk. Our data establish a model that reveals the very early brain to be a central player in innate immunity, identify the developmental origins of brain–immune interactions, and suggest several targets for immune therapies.
Collapse
|
23
|
Petrelli F, Dallérac G, Pucci L, Calì C, Zehnder T, Sultan S, Lecca S, Chicca A, Ivanov A, Asensio CS, Gundersen V, Toni N, Knott GW, Magara F, Gertsch J, Kirchhoff F, Déglon N, Giros B, Edwards RH, Mothet JP, Bezzi P. Dysfunction of homeostatic control of dopamine by astrocytes in the developing prefrontal cortex leads to cognitive impairments. Mol Psychiatry 2020; 25:732-749. [PMID: 30127471 PMCID: PMC7156348 DOI: 10.1038/s41380-018-0226-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 06/28/2018] [Accepted: 07/18/2018] [Indexed: 01/07/2023]
Abstract
Astrocytes orchestrate neural development by powerfully coordinating synapse formation and function and, as such, may be critically involved in the pathogenesis of neurodevelopmental abnormalities and cognitive deficits commonly observed in psychiatric disorders. Here, we report the identification of a subset of cortical astrocytes that are competent for regulating dopamine (DA) homeostasis during postnatal development of the prefrontal cortex (PFC), allowing for optimal DA-mediated maturation of excitatory circuits. Such control of DA homeostasis occurs through the coordinated activity of astroglial vesicular monoamine transporter 2 (VMAT2) together with organic cation transporter 3 and monoamine oxidase type B, two key proteins for DA uptake and metabolism. Conditional deletion of VMAT2 in astrocytes postnatally produces loss of PFC DA homeostasis, leading to defective synaptic transmission and plasticity as well as impaired executive functions. Our findings show a novel role for PFC astrocytes in the DA modulation of cognitive performances with relevance to psychiatric disorders.
Collapse
Affiliation(s)
- Francesco Petrelli
- 0000 0001 2165 4204grid.9851.5Department of Fundamental Neurosciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Glenn Dallérac
- 0000 0001 2176 4817grid.5399.6Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Aix-Marseille Université UMR7286 CNRS, 13344 Marseille, Cedex 15 France
| | - Luca Pucci
- 0000 0001 2165 4204grid.9851.5Department of Fundamental Neurosciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Corrado Calì
- 0000 0001 2165 4204grid.9851.5Department of Fundamental Neurosciences, University of Lausanne, CH-1005 Lausanne, Switzerland ,0000 0001 1926 5090grid.45672.32BESE division, King Abdullah University of Science and Technology, 23955-69000 Thuwal, Saudi Arabia
| | - Tamara Zehnder
- 0000 0001 2165 4204grid.9851.5Department of Fundamental Neurosciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Sébastien Sultan
- 0000 0001 2165 4204grid.9851.5Department of Fundamental Neurosciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Salvatore Lecca
- 0000 0001 2165 4204grid.9851.5Department of Fundamental Neurosciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Andrea Chicca
- 0000 0001 0726 5157grid.5734.5Institute of Biochemistry and Molecular Medicine (IBMM), University of Bern, Buehlstrasse, 28 3012 Bern, Switzerland
| | - Andrei Ivanov
- “Biophotonics and Synapse Physiopathology” Team, UMR9188 CNRS – ENS Paris Saclay, 91405 Orsay, France
| | - Cédric S. Asensio
- 0000 0001 2297 6811grid.266102.1Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA 94158 USA
| | - Vidar Gundersen
- 0000 0004 1936 8921grid.5510.1CMBN, Rikshospitalet, University of Oslo, Oslo, Norway
| | - Nicolas Toni
- 0000 0001 2165 4204grid.9851.5Department of Fundamental Neurosciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Graham William Knott
- 0000000121839049grid.5333.6BioEM Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Fulvio Magara
- 0000 0001 2165 4204grid.9851.5Centre for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital Center, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Jürg Gertsch
- 0000 0001 0726 5157grid.5734.5Institute of Biochemistry and Molecular Medicine (IBMM), University of Bern, Buehlstrasse, 28 3012 Bern, Switzerland
| | - Frank Kirchhoff
- 0000 0001 2167 7588grid.11749.3aDepartment of Molecular Physiology, University of Saarland, D-66421 Homburg, Germany
| | - Nicole Déglon
- 0000 0001 0423 4662grid.8515.9Department of Clinical Neurosciences, Lausanne University Hospital, Lausanne, Switzerland ,0000 0001 0423 4662grid.8515.9Neuroscience Research Center, Lausanne University Hospital, CH-1011 Lausanne, Switzerland
| | - Bruno Giros
- 0000 0004 1936 8649grid.14709.3bDepartment of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec H4H1R3 Canada ,0000 0001 2112 9282grid.4444.0INSERM, UMRS 1130; CNRS, UMR 8246; Sorbonne University UPMC, Neuroscience Paris-Seine, F-75005 Paris, France
| | - Robert H. Edwards
- 0000 0001 2297 6811grid.266102.1Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA 94158 USA
| | - Jean-Pierre Mothet
- Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Aix-Marseille Université UMR7286 CNRS, 13344, Marseille, Cedex 15, France. .,"Biophotonics and Synapse Physiopathology" Team, UMR9188 CNRS - ENS Paris Saclay, 91405, Orsay, France.
| | - Paola Bezzi
- Department of Fundamental Neurosciences, University of Lausanne, CH-1005, Lausanne, Switzerland.
| |
Collapse
|
24
|
|
25
|
Aripiprazole and haloperidol protect neurite lesions via reducing excessive D2R-DISC1 complex formation. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:59-69. [PMID: 30597182 DOI: 10.1016/j.pnpbp.2018.12.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 12/11/2018] [Accepted: 12/14/2018] [Indexed: 12/15/2022]
Abstract
Dopamine D2 receptor (D2R) hyperactivity causes altered brain development and later produces onset of symptoms mimicking schizophrenia. It is known that D2R interacts with disrupted in schizophrenia 1 (DISC1); however, the effect of D2R-DISC1 interaction in intracellular signalling and neurite growth has not been studied. This study investigated the effect of D2R over-activation on Akt-GSK3β signalling and neurite morphology in cortical neurons. Over-activation of D2Rs caused neurite lesions, which were associated with decreased protein kinase B (Akt) and glycogen synthase kinase 3 beta (GSK3β) phosphorylation in cortical neurons. The antipsychotic drug aripiprazole was more effective in the prevention of neurite lesions than haloperidol. Unlike haloperidol, aripiprazole prevented downregulation of phospho (p) Akt-pGSK3β induced by D2R hyperactivity, indicating involvement of different pathways. D2Rs were hyperactive in cortical neurons of mice with DISC1 mutation, which caused more severe neurite lesions in cortical neurons treated with quinpirole. Immunofluorescent staining for Ca2+/calmodulin-dependent protein kinase II (CaMKII) confirmed that cortical pyramidal neurons were involved in the D2R hyperactivity-induced neurite lesions. Using the fluorescence resonance energy transfer (FRET) technique, we provide direct evidence that D2R hyperactivity led to D2R-DISC1 complex formation, which altered pGSK3β signalling. This study showed that D2R hyperactivity-induced D2R-DISC1 complex formation is associated with decreased pAkt-pGSK3β signalling and in turn, caused neurite impairment. Aripiprazole and haloperidol prevented the impairment of neurite growth but appeared to do so via different intracellular signalling pathways.
Collapse
|
26
|
Jin X, Chen Q, Song Y, Zheng J, Xiao K, Shao S, Fu Z, Yi M, Yang Y, Huang Z. Dopamine D2 receptors regulate the action potential threshold by modulating T‐type calcium channels in stellate cells of the medial entorhinal cortex. J Physiol 2019; 597:3363-3387. [PMID: 31049961 DOI: 10.1113/jp277976] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 04/24/2019] [Indexed: 11/08/2022] Open
Affiliation(s)
- Xueqin Jin
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking University Health Science Centre Beijing 100191 China
| | - Qian Chen
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking University Health Science Centre Beijing 100191 China
| | - Yan Song
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking University Health Science Centre Beijing 100191 China
| | - Jie Zheng
- Neuroscience Research InstitutePeking University Health Science Center Beijing 100191 China
| | - Kuo Xiao
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking University Health Science Centre Beijing 100191 China
| | - Shan Shao
- Neuroscience Research InstitutePeking University Health Science Center Beijing 100191 China
| | - Zibing Fu
- Neuroscience Research InstitutePeking University Health Science Center Beijing 100191 China
| | - Ming Yi
- Neuroscience Research InstitutePeking University Health Science Center Beijing 100191 China
- Key Laboratory for NeuroscienceMinistry of Education/National Health and Family Planning CommissionPeking University Beijing 100191 China
| | - Yang Yang
- Department of Medicinal Chemistry and Molecular PharmacologyCollege of Pharmacy, Purdue University West Lafayette IN 47907 USA
- Purdue Institute for Integrative Neuroscience 575 Stadium Mall Drive West Lafayette IN 47907 USA
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking University Health Science Centre Beijing 100191 China
- Key Laboratory for NeuroscienceMinistry of Education/National Health and Family Planning CommissionPeking University Beijing 100191 China
- Department of Molecular and Cellular PharmacologyPeking University Health Science Center 38 Xue Yuan Road Beijing 100191 China
| |
Collapse
|
27
|
Ye J, Ji F, Jiang D, Lin X, Chen G, Zhang W, Shan P, Zhang L, Zhuo C. Polymorphisms in Dopaminergic Genes in Schizophrenia and Their Implications in Motor Deficits and Antipsychotic Treatment. Front Neurosci 2019; 13:355. [PMID: 31057354 PMCID: PMC6479209 DOI: 10.3389/fnins.2019.00355] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/28/2019] [Indexed: 12/14/2022] Open
Abstract
Dopaminergic system dysfunction is involved in schizophrenia (SCZ) pathogenesis and can mediate SCZ-related motor disorders. Recent studies have gradually revealed that SCZ susceptibility and the associated motor symptoms can be mediated by genetic factors, including dopaminergic genes. More importantly, polymorphisms in these genes are associated with both antipsychotic drug sensitivity and adverse effects. The study of genetic polymorphisms in the dopaminergic system may help to optimize individualized drug strategies for SCZ patients. This review summarizes the current progress about the involvement of the dopamine system in SCZ-associated motor disorders and the motor-related adverse effects after antipsychotic treatment, with a special focus on polymorphisms in dopaminergic genes. We hypothesize that the genetic profile of the dopaminergic system mediates both SCZ-associated motor deficits associated and antipsychotic drug-related adverse effects. The study of dopaminergic gene polymorphisms may help to predict drug efficacy and decrease adverse effects, thereby optimizing treatment strategies.
Collapse
Affiliation(s)
- Jiaen Ye
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Feng Ji
- Department of Psychiatry, College of Mental Health, Jining Medical University, Jining, China
| | - Deguo Jiang
- Department of Psychiatric-Neuroimaging-Genetics and Morbidity Laboratory (PNGC-Lab), Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| | - Xiaodong Lin
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Guangdong Chen
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Wei Zhang
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Peiwei Shan
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Li Zhang
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Chuanjun Zhuo
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China.,Department of Psychiatry, College of Mental Health, Jining Medical University, Jining, China.,Department of Psychiatric-Neuroimaging-Genetics and Morbidity Laboratory (PNGC-Lab), Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
28
|
Genetic labeling reveals temporal and spatial expression pattern of D2 dopamine receptor in rat forebrain. Brain Struct Funct 2019; 224:1035-1049. [PMID: 30604007 PMCID: PMC6499762 DOI: 10.1007/s00429-018-01824-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 12/20/2018] [Indexed: 01/11/2023]
Abstract
The D2 dopamine receptor (Drd2) is implicated in several brain disorders such as schizophrenia, Parkinson’s disease, and drug addiction. Drd2 is also the primary target of both antipsychotics and Parkinson’s disease medications. Although the expression pattern of Drd2 is relatively well known in mouse brain, the temporal and spatial distribution of Drd2 is lesser clear in rat brain due to the lack of Drd2 reporter rat lines. Here, we used CRISPR/Cas9 techniques to generate two knockin rat lines: Drd2::Cre and Rosa26::loxp-stop-loxp-tdTomato. By crossing these two lines, we produced Drd2 reporter rats expressing the fluorescence protein tdTomato under the control of the endogenous Drd2 promoter. Using fluorescence imaging and unbiased stereology, we revealed the cellular expression pattern of Drd2 in adult and postnatal rat forebrain. Strikingly, the Drd2 expression pattern differs between Drd2 reporter rats and Drd2 reporter mice generated by BAC transgene in prefrontal cortex and hippocampus. These results provide fundamental information needed for the study of Drd2 function in rat forebrain. The Drd2::Cre rats generated here may represent a useful tool to study the function of neuronal populations expressing Drd2.
Collapse
|
29
|
Cortical dendritic spine development and plasticity: insights from in vivo imaging. Curr Opin Neurobiol 2018; 53:76-82. [DOI: 10.1016/j.conb.2018.06.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 01/14/2023]
|
30
|
Dysbindin-1 contributes to prefrontal cortical dendritic arbor pathology in schizophrenia. Schizophr Res 2018; 201:270-277. [PMID: 29759351 PMCID: PMC6230503 DOI: 10.1016/j.schres.2018.04.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 04/20/2018] [Accepted: 04/29/2018] [Indexed: 01/22/2023]
Abstract
Deep layer III pyramidal cells in the dorsolateral prefrontal cortex (DLPFC) from subjects with schizophrenia and bipolar disorder previously were shown to exhibit dendritic arbor pathology. This study sought to determine whether MARCKS, its regulatory protein dysbindin-1, and two proteins, identified using microarray data, CDC42BPA and ARHGEF6, were associated with dendritic arbor pathology in the DLPFC from schizophrenia and bipolar disorder subjects. Using western blotting, relative protein expression was assessed in the DLPFC (BA 46) grey matter from subjects with schizophrenia (n = 19), bipolar disorder (n = 17) and unaffected control subjects (n = 19). Protein expression data were then correlated with dendritic parameter data obtained previously. MARCKS and dysbindin-1a expression levels did not differ among the three groups. Dysbindin-1b expression was 26% higher in schizophrenia subjects (p = 0.01) and correlated inversely with basilar dendrite length (r = -0.31, p = 0.048) and the number of spines per basilar dendrite (r = -0.31, p = 0.048), but not with dendritic spine density (r = -0.16, p = 0.32). The protein expression of CDC42BPA was 33% higher in schizophrenia subjects (p = 0.03) but, did not correlate with any dendritic parameter (p > 0.05). ARHGEF6 87 kDa isoform expression did not differ among the groups. CDC42BPA expression was not altered in frontal cortex from rats chronically administered haloperidol or clozapine. Dysbindin-1b appears to play a role in dendritic arbor pathology observed previously in the DLPFC in schizophrenia.
Collapse
|
31
|
Hu M, Zheng P, Xie Y, Boz Z, Yu Y, Tang R, Jones A, Zheng K, Huang XF. Propionate Protects Haloperidol-Induced Neurite Lesions Mediated by Neuropeptide Y. Front Neurosci 2018; 12:743. [PMID: 30374288 PMCID: PMC6196753 DOI: 10.3389/fnins.2018.00743] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 09/26/2018] [Indexed: 01/22/2023] Open
Abstract
Haloperidol is a commonly used antipsychotic drug for treating schizophrenia. Clinical imaging studies have found that haloperidol can cause volume loss of human brain tissue, which is supported by animal studies showing that haloperidol reduces the number of synaptic spines. The mechanism remains unknown. Gut microbiota metabolites, short chain fatty acids including propionate, are reported to have neuroprotective effect and influence gene expression. This study aims to investigate the effect and mechanism of propionate in the protection of neurite lesion induced by haloperidol. This study showed that 10 μM haloperidol (clinical relevant dose) impaired neurite length in human blastoma SH-SY5Y cells, which were confirmed by using primary mouse striatal spiny neurons. We found that haloperidol impaired neurite length were accompanied by a decreased neuropeptide Y (NPY) expression, but no effect on GSK3β signaling. Importantly, this project research found that propionate was capable of protecting against haloperidol-induced neurite lesions and preventing NPY reduction. To confirm this finding, we used specific siRNAs targeting NPY which blocked the protective effect of propionate on haloperidol-induced neurite lesions. Furthermore, since NPY is regulated by the nuclear transcription factor CREB, we measured pCREB that was decreased by haloperidol and was normalized by propionate. Therefore, propionate has a protective effect against pCREB-NPY mediated haloperidol-induced neurite lesions.
Collapse
Affiliation(s)
- Minmin Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Jiangsu, China.,Illawarra Health and Medical Research Institute, School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Peng Zheng
- Illawarra Health and Medical Research Institute, School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Yuanyi Xie
- Illawarra Health and Medical Research Institute, School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Zehra Boz
- Illawarra Health and Medical Research Institute, School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Jiangsu, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Jiangsu, China
| | - Alison Jones
- Illawarra Health and Medical Research Institute, School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Jiangsu, China
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute, School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
32
|
Mansilla A, Jordán-Álvarez S, Santana E, Jarabo P, Casas-Tintó S, Ferrús A. Molecular mechanisms that change synapse number. J Neurogenet 2018; 32:155-170. [DOI: 10.1080/01677063.2018.1506781] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
33
|
Striatal dopamine 2 receptor upregulation during development predisposes to diet-induced obesity by reducing energy output in mice. Proc Natl Acad Sci U S A 2018; 115:10493-10498. [PMID: 30254156 DOI: 10.1073/pnas.1800171115] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dopaminergic signaling in the striatum, particularly at dopamine 2 receptors (D2R), has been a topic of active investigation in obesity research in the past decades. However, it still remains unclear whether variations in striatal D2Rs modulate the risk for obesity and if so in which direction. Human studies have yielded contradictory findings that likely reflect a complex nonlinear relationship, possibly involving a combination of causal effects and compensatory changes. Animal work indicates that although chronic obesogenic diets reduce striatal D2R function, striatal D2R down-regulation does not lead to obesity. In this study, we evaluated the consequences of striatal D2R up-regulation on body-weight gain susceptibility and energy balance in mice. We used a mouse model of D2R overexpression (D2R-OE) in which D2Rs were selectively up-regulated in striatal medium spiny neurons. We uncover a pathological mechanism by which striatal D2R-OE leads to reduced brown adipose tissue thermogenesis, reduced energy expenditure, and accelerated obesity despite reduced eating. We also show that D2R-OE restricted to development is sufficient to promote obesity and to induce energy-balance deficits. Together, our findings indicate that striatal D2R-OE during development persistently increases the propensity for obesity by reducing energy output in mice. This suggests that early alterations in the striatal dopamine system could represent a key predisposition factor toward obesity.
Collapse
|
34
|
Xie Y, Huang XF. Commentary: GLYX-13 Ameliorates Schizophrenia-Like Phenotype Induced by MK-801 in Mice: Role of Hippocampal NR2B and DISC1. Front Mol Neurosci 2018; 11:315. [PMID: 30233316 PMCID: PMC6134048 DOI: 10.3389/fnmol.2018.00315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/16/2018] [Indexed: 11/30/2022] Open
Affiliation(s)
- Yuanyi Xie
- Illawarra Health and Medical Research Institute and School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute and School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
35
|
Schwarz AP, Rotov AY, Chuprina OI, Krytskaya DU, Trofimov AN, Kosheverova VV, Ischenko AM, Zubareva OE. Developmental prefrontal mRNA expression of D2 dopamine receptor splice variants and working memory impairments in rats after early life Interleukin-1β elevation. Neurobiol Learn Mem 2018; 155:231-238. [PMID: 30092312 DOI: 10.1016/j.nlm.2018.08.008] [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: 03/24/2018] [Revised: 07/13/2018] [Accepted: 08/03/2018] [Indexed: 11/29/2022]
Abstract
Long (D2L) and Short (D2S) isoforms of D2 dopamine receptor differ in their biochemical and physiological properties, which could affect functioning of prefrontal cortex. Contribution of distinct D2 dopamine receptor isoforms to cognitive dysfunctions and its developmental regulation are currently not fully elucidated. In the present study, we evaluated developmental mRNA expression of D2S/D2L dopamine receptor isoforms within the rat medial prefrontal cortex (mPFC) in the model of neurodevelopmental cognitive dysfunction. Working memory performance (Y-maze spontaneous alternations) and D2S/D2L mRNA expression in the mPFC (by qRT-PCR) were evaluated in juvenile (P27), adolescent (P42-47) and adult (P75-90) rats after chronic early life treatment with proinflammatory cytokine interleukin (IL)-1β (1 µg/kg i.p. daily P15-21). It was shown that IL-1β elevation during the 3rd week of life leads to working memory deficit originating in juvenile animals and persisting into adulthood. D2S mRNA expression was strongly downregulated during adolescence, and such downregulation was exaggerated in animals injected with IL-1β during P15-21. Early life IL-1β administrations influenced developmental changes in the D2S/D2L mRNA ratio. This measure was found to be decreased in adolescent and adult control (intact and vehicle-treated) rats compared to juvenile control, while in the case of IL-1β-treated animals, the decrease in D2S/D2L ratio was observed only in adulthood but not in adolescence compared to juvenile rats. During the adolescence, D2S mRNA expression was downregulated and D2S/D2L ratio was upregulated in the mPFC of rats treated with IL-1β during the 3rd week of life compared to controls. Based on these data we conclude that changes in the developmental expression of D2 dopamine receptor splice variants within mPFC may underlie long-lasting cognitive deficit associated with neonatal pathology.
Collapse
Affiliation(s)
- Alexander P Schwarz
- Laboratory of Neurobiology of the Brain Integrative Functions, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Akademika Pavlova street 12, 197376 St. Petersburg, Russia.
| | - Alexander Yu Rotov
- Laboratory of Evolution of the Sensory Organs, I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Thorez avenue 44, 199223 St. Petersburg, Russia
| | - Olga I Chuprina
- Laboratory of Neurobiology of the Brain Integrative Functions, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Akademika Pavlova street 12, 197376 St. Petersburg, Russia
| | - Darya U Krytskaya
- Laboratory of Neurobiology of the Brain Integrative Functions, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Akademika Pavlova street 12, 197376 St. Petersburg, Russia
| | - Alexander N Trofimov
- Laboratory of Neurobiology of the Brain Integrative Functions, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Akademika Pavlova street 12, 197376 St. Petersburg, Russia
| | - Vera V Kosheverova
- Laboratory of Intracellular Membranes Dynamics, Department of the Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky avenue 4, 194064 St. Petersburg, Russia
| | - Alexander M Ischenko
- Laboratory of Protein Biochemistry, Research Institute of Highly Pure Biopreparations, Pudozhskaya street 7, 197110 St. Petersburg, Russia
| | - Olga E Zubareva
- Laboratory of Neurobiology of the Brain Integrative Functions, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Akademika Pavlova street 12, 197376 St. Petersburg, Russia; Laboratory of Molecular Mechanisms of Neuronal Interactions, I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Thorez avenue 44, 199223 St. Petersburg, Russia
| |
Collapse
|
36
|
Huang XF, Song X. Effects of antipsychotic drugs on neurites relevant to schizophrenia treatment. Med Res Rev 2018; 39:386-403. [PMID: 29785841 DOI: 10.1002/med.21512] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 04/06/2018] [Accepted: 04/30/2018] [Indexed: 12/31/2022]
Abstract
Although antipsychotic drugs are mainly used for treating schizophrenia, they are widely used for treating various psychiatric diseases in adults, the elderly, adolescents and even children. Today, about 1.2% of the worldwide population suffers from psychosis and related disorders, which translates to about 7.5 million subjects potentially targeted by antipsychotic drugs. Neurites project from the cell body of neurons and connect neurons to each other to form neural networks. Deficits in neurite outgrowth and integrity are implicated in psychiatric diseases including schizophrenia. Neurite deficits contribute to altered brain development, neural networking and connectivity as well as symptoms including psychosis and altered cognitive function. This review revealed that (1) antipsychotic drugs could have profound effects on neurites, synaptic spines and synapse, by which they may influence and regulate neural networking and plasticity; (2) antipsychotic drugs target not only neurotransmitter receptors but also intracellular signaling molecules regulating the signaling pathways responsible for neurite outgrowth and maintenance; (3) high doses and chronic administration of antipsychotic drugs may cause some loss of neurites, synaptic spines, or synapsis in the cortical structures. In addition, confounding effects causing neurite deficits may include elevated inflammatory cytokines and antipsychotic drug-induced metabolic side effects in patients on chronic antipsychotic therapy. Unraveling how antipsychotic drugs affect neurites and neural connectivity is essential for improving therapeutic outcomes and preventing aversive effects for patients on antipsychotic drug treatment.
Collapse
Affiliation(s)
- Xu-Feng Huang
- Henan Medical Key Laboratory of Translational Research on Psychiatric Diseases, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China.,The Illawarra Health and Medical Research Institute and School of Medicine, University of Wollongong, Wollongong, Australia
| | - Xueqin Song
- Henan Medical Key Laboratory of Translational Research on Psychiatric Diseases, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
37
|
Deletion of dopamine D 2 receptors from parvalbumin interneurons in mouse causes schizophrenia-like phenotypes. Proc Natl Acad Sci U S A 2018. [PMID: 29531031 DOI: 10.1073/pnas.1719897115] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Excessive dopamine neurotransmission underlies psychotic episodes as observed in patients with some types of bipolar disorder and schizophrenia. The dopaminergic hypothesis was postulated after the finding that antipsychotics were effective to halt increased dopamine tone. However, there is little evidence for dysfunction within the dopaminergic system itself. Alternatively, it has been proposed that excessive afferent activity onto ventral tegmental area dopaminergic neurons, particularly from the ventral hippocampus, increase dopamine neurotransmission, leading to psychosis. Here, we show that selective dopamine D2 receptor deletion from parvalbumin interneurons in mouse causes an impaired inhibitory activity in the ventral hippocampus and a dysregulated dopaminergic system. Conditional mutant animals show adult onset of schizophrenia-like behaviors and molecular, cellular, and physiological endophenotypes as previously described from postmortem brain studies of patients with schizophrenia. Our findings show that dopamine D2 receptor expression on parvalbumin interneurons is required to modulate and limit pyramidal neuron activity, which may prevent the dysregulation of the dopaminergic system.
Collapse
|
38
|
Yoshikawa A, Nishimura F, Inai A, Eriguchi Y, Nishioka M, Takaya A, Tochigi M, Kawamura Y, Umekage T, Kato K, Sasaki T, Kasai K, Kakiuchi C. Novel rare variations in genes that regulate developmental change in N-methyl-d-aspartate receptor in patients with schizophrenia. Hum Genome Var 2018; 5:17056. [PMID: 29423241 PMCID: PMC5794673 DOI: 10.1038/hgv.2017.56] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/28/2017] [Accepted: 10/31/2017] [Indexed: 11/29/2022] Open
Abstract
The mechanism underlying the vulnerability to developing schizophrenia (SCZ) during adolescence remains elusive. Hypofunction of N-methyl-d-aspartate receptors (NMDARs) has been implicated in the pathophysiology of SCZ. During development, the composition of synaptic NMDARs dramatically changes from NR2B-containing NMDARs to NR2A-containing NMDARs through the phosphorylation of NR2B S1480 or Y1472 by CDK5, CSNK2A1, and EphB2, which plays a pivotal role in the maturation of neural circuits. We hypothesized that the dysregulation of developmental change in NMDARs could be involved in the onset of SCZ. Using next-generation sequencing, we re-sequenced all the coding regions and splice sites of CDK5, CSNK2A1, and EphB2 in 474 patients with SCZ and 475 healthy controls. Variants on the database for human control subjects of Japanese origin were removed and all the nonsynonymous and nonsense variants were validated using Sanger sequencing. Four novel variants in CDK5 were observed in patients with SCZ but were not observed in controls. The total number of variants, however, was not significantly different between the SCZ and control groups (P=0.062). In silico analyses predicted P271T to be damaging. Further genetic research using a larger sample is required to examine whether CDK5 is involved in the pathophysiology of SCZ.
Collapse
Affiliation(s)
- Akane Yoshikawa
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Fumichika Nishimura
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Aya Inai
- Department of Child Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yosuke Eriguchi
- Department of Child Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaki Nishioka
- Division for Counseling and Support, Office for Mental Health Support, The University of Tokyo, Tokyo, Japan
| | - Atsuhiko Takaya
- Department of Psychiatry, Fukui Memorial Hospital, Kanagawa, Japan
| | - Mamoru Tochigi
- Department of Neuropsychiatry, Teikyo University School of Medicine, Tokyo, Japan
| | - Yoshiya Kawamura
- Department of Psychiatry, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Tadashi Umekage
- Division for Environment, Health and Safety, University of Tokyo, Tokyo, Japan
| | - Kayoko Kato
- Department of Health Education, Graduate School of Education, The University of Tokyo, Tokyo, Japan
| | - Tsukasa Sasaki
- Department of Health Education, Graduate School of Education, The University of Tokyo, Tokyo, Japan
| | - Kiyoto Kasai
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Chihiro Kakiuchi
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Disability Services Office, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
39
|
Rozman J, Rathkolb B, Oestereicher MA, Schütt C, Ravindranath AC, Leuchtenberger S, Sharma S, Kistler M, Willershäuser M, Brommage R, Meehan TF, Mason J, Haselimashhadi H, Hough T, Mallon AM, Wells S, Santos L, Lelliott CJ, White JK, Sorg T, Champy MF, Bower LR, Reynolds CL, Flenniken AM, Murray SA, Nutter LMJ, Svenson KL, West D, Tocchini-Valentini GP, Beaudet AL, Bosch F, Braun RB, Dobbie MS, Gao X, Herault Y, Moshiri A, Moore BA, Kent Lloyd KC, McKerlie C, Masuya H, Tanaka N, Flicek P, Parkinson HE, Sedlacek R, Seong JK, Wang CKL, Moore M, Brown SD, Tschöp MH, Wurst W, Klingenspor M, Wolf E, Beckers J, Machicao F, Peter A, Staiger H, Häring HU, Grallert H, Campillos M, Maier H, Fuchs H, Gailus-Durner V, Werner T, Hrabe de Angelis M. Identification of genetic elements in metabolism by high-throughput mouse phenotyping. Nat Commun 2018; 9:288. [PMID: 29348434 PMCID: PMC5773596 DOI: 10.1038/s41467-017-01995-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 10/30/2017] [Indexed: 12/20/2022] Open
Abstract
Metabolic diseases are a worldwide problem but the underlying genetic factors and their relevance to metabolic disease remain incompletely understood. Genome-wide research is needed to characterize so-far unannotated mammalian metabolic genes. Here, we generate and analyze metabolic phenotypic data of 2016 knockout mouse strains under the aegis of the International Mouse Phenotyping Consortium (IMPC) and find 974 gene knockouts with strong metabolic phenotypes. 429 of those had no previous link to metabolism and 51 genes remain functionally completely unannotated. We compared human orthologues of these uncharacterized genes in five GWAS consortia and indeed 23 candidate genes are associated with metabolic disease. We further identify common regulatory elements in promoters of candidate genes. As each regulatory element is composed of several transcription factor binding sites, our data reveal an extensive metabolic phenotype-associated network of co-regulated genes. Our systematic mouse phenotype analysis thus paves the way for full functional annotation of the genome.
Collapse
Affiliation(s)
- Jan Rozman
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Birgit Rathkolb
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Ludwig-Maximilians-Universität München, Gene Center, Institute of Molecular Animal Breeding and Biotechnology, Feodor-Lynen Strasse 25, 81377, Munich, Germany
| | - Manuela A Oestereicher
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Christine Schütt
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Aakash Chavan Ravindranath
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Stefanie Leuchtenberger
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Sapna Sharma
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Martin Kistler
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Monja Willershäuser
- Chair of Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences Weihenstephan, 85354, Freising, Germany
- EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, 85354, Freising, Germany
- ZIEL - Institute for Food & Health, Technical University of Munich, 85354, Freising, Germany
| | - Robert Brommage
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Terrence F Meehan
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Jeremy Mason
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Hamed Haselimashhadi
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Tertius Hough
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Oxfordshire, OX11 0RD, UK
| | - Ann-Marie Mallon
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Oxfordshire, OX11 0RD, UK
| | - Sara Wells
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Oxfordshire, OX11 0RD, UK
| | - Luis Santos
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Oxfordshire, OX11 0RD, UK
| | - Christopher J Lelliott
- The Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Jacqueline K White
- The Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Tania Sorg
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), 1 Rue Laurent Fries, 67404, Illkirch-Graffenstaden, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Parc d'innovation, 1 Rue Laurent Fries - BP 10142, 67404, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, 67404, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, 67404, Illkirch, France
- Université de Strasbourg, 67404, Illkirch, France
| | - Marie-France Champy
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), 1 Rue Laurent Fries, 67404, Illkirch-Graffenstaden, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Parc d'innovation, 1 Rue Laurent Fries - BP 10142, 67404, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, 67404, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, 67404, Illkirch, France
- Université de Strasbourg, 67404, Illkirch, France
| | - Lynette R Bower
- Mouse Biology Program, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Corey L Reynolds
- Department of Molecular and Human Genetics, Baylor College of Medicine, 7702 Main St, Houston Medical Center, Houston, TX, 77030-4406, USA
| | - Ann M Flenniken
- The Centre for Phenogenomics, 25 Orde St, Toronto, M5T 3H7, ON, Canada
- The Hospital for Sick Children, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Joseph and Wolf Lebovic Health Complex, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - Stephen A Murray
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Lauryl M J Nutter
- The Centre for Phenogenomics, 25 Orde St, Toronto, M5T 3H7, ON, Canada
- The Hospital for Sick Children, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - Karen L Svenson
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - David West
- Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr. Way, Oakland, CA, 94609, USA
| | - Glauco P Tocchini-Valentini
- Monterotondo Mouse Clinic, Italian National Research Council (CNR), Institute of Cell Biology and Neurobiology, Adriano Buzzati-Traverso Campus, Via E. Ramarini 32, Monterotondo Scalo, RM, 00015, Italy
| | - Arthur L Beaudet
- The Centre for Phenogenomics, 25 Orde St, Toronto, M5T 3H7, ON, Canada
- The Hospital for Sick Children, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Robert B Braun
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Michael S Dobbie
- Australian Phenomics Network, John Curtin School of Medical Research, Australian National University, 131 Garran Road, Canberra, ACT, 2601, Australia
| | - Xiang Gao
- SKL of Pharmaceutical Biotechnology and Model Animal Research Center, Collaborative Innovation Center for Genetics and Development, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061, China
| | - Yann Herault
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), 1 Rue Laurent Fries, 67404, Illkirch-Graffenstaden, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Parc d'innovation, 1 Rue Laurent Fries - BP 10142, 67404, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, 67404, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, 67404, Illkirch, France
- Université de Strasbourg, 67404, Illkirch, France
| | - Ala Moshiri
- Department of Ophthalmology & Vision Science, School of Medicine, U.C. Davis, 77 Cadillac Drive, Sacramento, 95825, CA, USA
| | - Bret A Moore
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, U.C. Davis, One Shields Avenue, Davis, 95616, CA, USA
| | - K C Kent Lloyd
- Mouse Biology Program, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Colin McKerlie
- The Centre for Phenogenomics, 25 Orde St, Toronto, M5T 3H7, ON, Canada
- The Hospital for Sick Children, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - Hiroshi Masuya
- RIKEN BioResource Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Nobuhiko Tanaka
- RIKEN BioResource Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Helen E Parkinson
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Je Kyung Seong
- Korea Mouse Phenotyping Consortium (KMPC) and BK21 Program for Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 599 Gwanangno, Gwanak-gu, Seoul, 151-742, South Korea
| | - Chi-Kuang Leo Wang
- National Laboratory Animal Center, National Applied Research Laboratories (NARLabs), 128 Yen-Chiou-Yuan Rd., Sec. 2, Nankang, Taipei, 11529, Taiwan
| | | | - Steve D Brown
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Oxfordshire, OX11 0RD, UK
| | - Matthias H Tschöp
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333, Munich, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Chair of Developmental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Deutsches Institut für Neurodegenerative Erkrankungen (DZNE) Site Munich, Feodor-Lynen-Str. 17, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 17, 81377, Munich, Germany
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences Weihenstephan, 85354, Freising, Germany
- EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, 85354, Freising, Germany
- ZIEL - Institute for Food & Health, Technical University of Munich, 85354, Freising, Germany
| | - Eckhard Wolf
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Ludwig-Maximilians-Universität München, Gene Center, Institute of Molecular Animal Breeding and Biotechnology, Feodor-Lynen Strasse 25, 81377, Munich, Germany
| | - Johannes Beckers
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Alte Akademie 8, 85354, Freising, Germany
| | - Fausto Machicao
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, University of Tübingen, 72076, Tübingen, Germany
| | - Andreas Peter
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, University of Tübingen, 72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard-Karls-University of Tuebingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
| | - Harald Staiger
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard-Karls-University of Tuebingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Hans-Ulrich Häring
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, University of Tübingen, 72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard-Karls-University of Tuebingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
| | - Harald Grallert
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München and Ludwig-Maximilians Universität München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Monica Campillos
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Holger Maier
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Valerie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Thomas Werner
- Internal Medicine Nephrology and Center for Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Martin Hrabe de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany.
- Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Alte Akademie 8, 85354, Freising, Germany.
| |
Collapse
|
40
|
Lee Y, Kim H, Kim JE, Park JY, Choi J, Lee JE, Lee EH, Han PL. Excessive D1 Dopamine Receptor Activation in the Dorsal Striatum Promotes Autistic-Like Behaviors. Mol Neurobiol 2017; 55:5658-5671. [PMID: 29027111 DOI: 10.1007/s12035-017-0770-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/11/2017] [Indexed: 12/27/2022]
Abstract
The dopamine system has been characterized in motor function, goal-directed behaviors, and rewards. Recent studies recognize various dopamine system genes as being associated with autism spectrum disorder (ASD). However, how dopamine system dysfunction induces ASD pathophysiology remains unknown. In the present study, we demonstrated that mice with increased dopamine functions in the dorsal striatum via the suppression of dopamine transporter expression in substantia nigra neurons or the optogenetic stimulation of the nigro-striatal circuitry exhibited sociability deficits and repetitive behaviors relevant to ASD pathology in animal models, while these behavioral changes were blocked by a D1 receptor antagonist. Pharmacological activation of D1 dopamine receptors in normal mice or the genetic knockout (KO) of D2 dopamine receptors also produced typical autistic-like behaviors. Moreover, the siRNA-mediated inhibition of D2 dopamine receptors in the dorsal striatum was sufficient to replicate autistic-like phenotypes in D2 KO mice. Intervention of D1 dopamine receptor functions or the signaling pathways-related D1 receptors in D2 KO mice produced anti-autistic effects. Together, our results indicate that increased dopamine function in the dorsal striatum promotes autistic-like behaviors and that the dorsal striatum is the neural correlate of ASD core symptoms.
Collapse
Affiliation(s)
- Yunjin Lee
- Department of Brain and Cognitive Sciences, Ewha Womans University, 11-1 Daehyun-Dong, Seodaemoon-Gu, Seoul, 120-750, Republic of Korea
| | - Hannah Kim
- Department of Brain and Cognitive Sciences, Ewha Womans University, 11-1 Daehyun-Dong, Seodaemoon-Gu, Seoul, 120-750, Republic of Korea
| | - Ji-Eun Kim
- Department of Brain and Cognitive Sciences, Ewha Womans University, 11-1 Daehyun-Dong, Seodaemoon-Gu, Seoul, 120-750, Republic of Korea
| | - Jin-Young Park
- Department of Brain and Cognitive Sciences, Ewha Womans University, 11-1 Daehyun-Dong, Seodaemoon-Gu, Seoul, 120-750, Republic of Korea
| | - Juli Choi
- Department of Brain and Cognitive Sciences, Ewha Womans University, 11-1 Daehyun-Dong, Seodaemoon-Gu, Seoul, 120-750, Republic of Korea
| | - Jung-Eun Lee
- Department of Brain and Cognitive Sciences, Ewha Womans University, 11-1 Daehyun-Dong, Seodaemoon-Gu, Seoul, 120-750, Republic of Korea
| | - Eun-Hwa Lee
- Department of Brain and Cognitive Sciences, Ewha Womans University, 11-1 Daehyun-Dong, Seodaemoon-Gu, Seoul, 120-750, Republic of Korea
| | - Pyung-Lim Han
- Department of Brain and Cognitive Sciences, Ewha Womans University, 11-1 Daehyun-Dong, Seodaemoon-Gu, Seoul, 120-750, Republic of Korea. .,Brain Disease Research Institute, Ewha Womans University, Seoul, Republic of Korea. .,Department of Chemistry and Nano Science, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
41
|
Role of G Protein-Coupled Receptors in the Regulation of Structural Plasticity and Cognitive Function. Molecules 2017; 22:molecules22071239. [PMID: 28737723 PMCID: PMC6152405 DOI: 10.3390/molecules22071239] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
Cognition and other higher brain functions are known to be intricately associated with the capacity of neural circuits to undergo structural reorganization. Structural remodelling of neural circuits, or structural plasticity, in the hippocampus plays a major role in learning and memory. Dynamic modifications of neuronal connectivity in the form of dendritic spine morphology alteration, as well as synapse formation and elimination, often result in the strengthening or weakening of specific neural circuits that determine synaptic plasticity. Changes in dendritic complexity and synapse number are mediated by cellular processes that are regulated by extracellular signals such as neurotransmitters and neurotrophic factors. As many neurotransmitters act on G protein-coupled receptors (GPCRs), it has become increasingly apparent that GPCRs can regulate structural plasticity through a myriad of G protein-dependent pathways and non-canonical signals. A thorough understanding of how GPCRs exert their regulatory influence on dendritic spine morphogenesis may provide new insights for treating cognitive impairment and decline in various age-related diseases. In this article, we review the evidence of GPCR-mediated regulation of structural plasticity, with a special emphasis on the involvement of common as well as distinct signalling pathways that are regulated by major neurotransmitters.
Collapse
|
42
|
McQueen J, Ryan TJ, McKay S, Marwick K, Baxter P, Carpanini SM, Wishart TM, Gillingwater TH, Manson JC, Wyllie DJA, Grant SGN, McColl BW, Komiyama NH, Hardingham GE. Pro-death NMDA receptor signaling is promoted by the GluN2B C-terminus independently of Dapk1. eLife 2017; 6:e17161. [PMID: 28731405 PMCID: PMC5544426 DOI: 10.7554/elife.17161] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/17/2017] [Indexed: 01/09/2023] Open
Abstract
Aberrant NMDA receptor (NMDAR) activity contributes to several neurological disorders, but direct antagonism is poorly tolerated therapeutically. The GluN2B cytoplasmic C-terminal domain (CTD) represents an alternative therapeutic target since it potentiates excitotoxic signaling. The key GluN2B CTD-centred event in excitotoxicity is proposed to involve its phosphorylation at Ser-1303 by Dapk1, that is blocked by a neuroprotective cell-permeable peptide mimetic of the region. Contrary to this model, we find that excitotoxicity can proceed without increased Ser-1303 phosphorylation, and is unaffected by Dapk1 deficiency in vitro or following ischemia in vivo. Pharmacological analysis of the aforementioned neuroprotective peptide revealed that it acts in a sequence-independent manner as an open-channel NMDAR antagonist at or near the Mg2+ site, due to its high net positive charge. Thus, GluN2B-driven excitotoxic signaling can proceed independently of Dapk1 or altered Ser-1303 phosphorylation.
Collapse
Affiliation(s)
- Jamie McQueen
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Tomás J Ryan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia
| | - Sean McKay
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Katie Marwick
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Paul Baxter
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Sarah M Carpanini
- The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
- nPAD MRC Mouse consortium, University of Edinburgh, Edinburgh, United Kingdom
| | - Thomas M Wishart
- The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
- nPAD MRC Mouse consortium, University of Edinburgh, Edinburgh, United Kingdom
| | - Thomas H Gillingwater
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
- nPAD MRC Mouse consortium, University of Edinburgh, Edinburgh, United Kingdom
| | - Jean C Manson
- The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
- nPAD MRC Mouse consortium, University of Edinburgh, Edinburgh, United Kingdom
| | - David J A Wyllie
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Seth G N Grant
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, United Kingdom
| | - Barry W McColl
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
- The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Noboru H Komiyama
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, United Kingdom
| | - Giles E Hardingham
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- nPAD MRC Mouse consortium, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
43
|
Chen YT, Lin CH, Huang CH, Liang WM, Lane HY. PICK1 Genetic Variation and Cognitive Function in Patients with Schizophrenia. Sci Rep 2017; 7:1889. [PMID: 28507309 PMCID: PMC5432511 DOI: 10.1038/s41598-017-01975-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 04/04/2017] [Indexed: 11/09/2022] Open
Abstract
The gene of protein interacting with C kinase 1 alpha (PICK1) has been implicated in schizophrenia, nevertheless, conflicting results existed. However, its role in cognitive function remains unclear. Besides, cognitive deficits impair the long-term outcome. We explored whether the polymorphisms of PICK1 (rs2076369, rs3952) affected cognitive functions in schizophrenic patients. We analyzed 302 patients and tested the differences of cognitive functions, clinical symptoms between genetic groups. We also used general linear model to analyze the effect of PICK1 genetic polymorphisms on cognitive functions. After adjustment for gender, age, education, the patients with rs2076369 G/T genotype showed better performance than T/T homozygotes in the summary score, global composite score, neurocognitive composite score, category fluency subtest, WAIS-III-Digit Symbol Coding subtest, working memory, WMS-III-Spatial Span (backward) subtest, MSCEIT-managing emotions branch (p = 0.038, 0.025, 0.046, 0.036, 0.025, 0.027, 0.035, 0.028, respectively). G/G homozygotes performed better than T/T in category fluency subtest (p = 0.049). A/A homozygotes of rs3952 performed better than G/G in trail making A subtest (p = 0.048). To our knowledge, this is the first study to indicate that PICK1 polymorphisms may associate with cognitive functions in schizophrenic patients. Further replication studies in healthy controls or other ethnic groups are warranted.
Collapse
Affiliation(s)
- Yi-Ting Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Psychiatry & Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Chieh-Hsin Lin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Psychiatry, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Center for General Education, Cheng Shiu University, Kaohsiung, Taiwan
| | - Chiung-Hsien Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Psychiatry & Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Miin Liang
- Graduate Institute of Biostatistics, School of Public Health, China Medical University, Taichung, Taiwan
| | - Hsien-Yuan Lane
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
- Department of Psychiatry & Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
44
|
Cognitive performance of juvenile monkeys after chronic fluoxetine treatment. Dev Cogn Neurosci 2017; 26:52-61. [PMID: 28521247 PMCID: PMC5557667 DOI: 10.1016/j.dcn.2017.04.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 02/01/2023] Open
Abstract
Potential long term effects on brain development are a concern when drugs are used to treat depression and anxiety in childhood. In this study, male juvenile rhesus monkeys (three-four years of age) were dosed with fluoxetine or vehicle (N=16/group) for two years. Histomorphometric examination of cortical dendritic spines conducted after euthanasia at one year postdosing (N=8/group) suggested a trend toward greater dendritic spine synapse density in prefrontal cortex of the fluoxetine-treated monkeys. During dosing, subjects were trained for automated cognitive testing, and evaluated with a test of sustained attention. After dosing was discontinued, sustained attention, recognition memory and cognitive flexibility were evaluated. Sustained attention was affected by fluoxetine, both during and after dosing, as indexed by omission errors. Response accuracy was not affected by fluoxetine in post-dosing recognition memory and cognitive flexibility tests, but formerly fluoxetine-treated monkeys compared to vehicle controls had more missed trial initiations and choices during testing. Drug treatment also interacted with genetic and environmental variables: MAOA genotype (high- and low transcription rate polymorphisms) and testing location (upper or lower tier of cages). Altered development of top-down cortical regulation of effortful attention may be relevant to this pattern of cognitive test performance after juvenile fluoxetine treatment.
Collapse
|
45
|
Lee SA, Suh Y, Lee S, Jeong J, Kim SJ, Kim SJ, Park SK. Functional expression of dopamine D2 receptor is regulated by tetraspanin 7-mediated postendocytic trafficking. FASEB J 2017; 31:2301-2313. [PMID: 28223337 DOI: 10.1096/fj.201600755rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 01/30/2017] [Indexed: 11/11/2022]
Abstract
The dopaminergic system plays an essential role in various functions of the brain, including locomotion, memory, and reward, and the deregulation of dopaminergic signaling as a result of altered functionality of dopamine D2 receptor (DRD2) is implicated in multiple neurologic and psychiatric disorders. Tetraspanin-7 (TSPAN7) is expressed to variable degrees in different tissues, with the highest level in the brain, and multiple mutations in TSPAN7 have been implicated in intellectual disability. Here, we tested the hypothesis that TSPAN7 may be a binding partner of DRD2 that is involved in the regulation of its functional activity. Our results showed that TSPAN7 was associated with DRD2 and reduced its surface expression by enhancing DRD2 internalization. Immunocytochemical analysis revealed that TSPAN7 that resides in the plasma membrane and early and late endosomes promoted internalization of DRD2 and its localization to endosomal compartments of the endocytic pathway. Furthermore, we observed that TSPAN7 deficiency increased surface localization of DRD2 concurrent with the decrease of its endocytosis, regardless of dopamine treatment. Finally, TSPAN7 negatively affects DRD2-mediated signaling. These results disclosed a previously uncharacterized role of TSPAN7 in the regulation of the expression and functional activity of DRD2 by postendocytic trafficking.-Lee, S.-A., Suh, Y., Lee, S., Jeong, J., Kim, S. J., Kim, S. J., Park, S. K. Functional expression of dopamine D2 receptor is regulated by tetraspanin 7-mediated postendocytic trafficking.
Collapse
Affiliation(s)
- Seol-Ae Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Yeongjun Suh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Saebom Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Jaehoon Jeong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Soo Jeong Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - So Jung Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| |
Collapse
|
46
|
Ham S, Kim TK, Chung S, Im HI. Drug Abuse and Psychosis: New Insights into Drug-induced Psychosis. Exp Neurobiol 2017; 26:11-24. [PMID: 28243163 PMCID: PMC5326711 DOI: 10.5607/en.2017.26.1.11] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 01/26/2017] [Accepted: 01/30/2017] [Indexed: 01/20/2023] Open
Abstract
Addictive drug use or prescribed medicine abuse can cause psychosis. Some representative symptoms frequently elicited by patients with psychosis are hallucination, anhedonia, and disrupted executive functions. These psychoses are categorized into three classifications of symptoms: positive, negative, and cognitive. The symptoms of DIP are not different from the symptoms of schizophrenia, and it is difficult to distinguish between them. Due to this ambiguity of distinction between the DIP and schizophrenia, the DIP animal model has been frequently used as the schizophrenia animal model. However, although the symptoms may be the same, its causes are clearly different in that DIP is acquired and schizophrenia is heritable. Therefore, in this review, we cover several DIP models such as of amphetamine, PCP/ketamine, scopolamine, and LSD, and then we also address three schizophrenia models through a genetic approach with a new perspective that distinguishes DIP from schizophrenia.
Collapse
Affiliation(s)
- Suji Ham
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.; Department of Neuroscience, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - Tae Kyoo Kim
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.; Department of Biology, Boston University, Boston 02215, USA
| | - Sooyoung Chung
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Heh-In Im
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.; Department of Neuroscience, Korea University of Science and Technology (UST), Daejeon 34113, Korea.; Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| |
Collapse
|
47
|
Tada H, Miyazaki T, Takemoto K, Takase K, Jitsuki S, Nakajima W, Koide M, Yamamoto N, Komiya K, Suyama K, Sano A, Taguchi A, Takahashi T. Neonatal isolation augments social dominance by altering actin dynamics in the medial prefrontal cortex. Proc Natl Acad Sci U S A 2016; 113:E7097-E7105. [PMID: 27791080 PMCID: PMC5111648 DOI: 10.1073/pnas.1606351113] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Social separation early in life can lead to the development of impaired interpersonal relationships and profound social disorders. However, the underlying cellular and molecular mechanisms involved are largely unknown. Here, we found that isolation of neonatal rats induced glucocorticoid-dependent social dominance over nonisolated control rats in juveniles from the same litter. Furthermore, neonatal isolation inactivated the actin-depolymerizing factor (ADF)/cofilin in the juvenile medial prefrontal cortex (mPFC). Isolation-induced inactivation of ADF/cofilin increased stable actin fractions at dendritic spines in the juvenile mPFC, decreasing glutamate synaptic AMPA receptors. Expression of constitutively active ADF/cofilin in the mPFC rescued the effect of isolation on social dominance. Thus, neonatal isolation affects spines in the mPFC by reducing actin dynamics, leading to altered social behavior later in life.
Collapse
Affiliation(s)
- Hirobumi Tada
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Tomoyuki Miyazaki
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Kiwamu Takemoto
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
- Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Kenkichi Takase
- Laboratory of Psychology, Jichi Medical University, Tochigi 329-0498, Japan
| | - Susumu Jitsuki
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Waki Nakajima
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Mayu Koide
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Naoko Yamamoto
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Kasane Komiya
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Kumiko Suyama
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Akane Sano
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Akiko Taguchi
- Department of Integrative Aging Neuroscience, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan
| | - Takuya Takahashi
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan;
| |
Collapse
|
48
|
Kim Y, Simon NW, Wood J, Moghaddam B. Reward Anticipation Is Encoded Differently by Adolescent Ventral Tegmental Area Neurons. Biol Psychiatry 2016; 79:878-86. [PMID: 26067679 PMCID: PMC4636980 DOI: 10.1016/j.biopsych.2015.04.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/23/2015] [Accepted: 04/28/2015] [Indexed: 11/23/2022]
Abstract
BACKGROUND Elucidating the neurobiology of the adolescent brain is fundamental to our understanding of the etiology of psychiatric disorders such as schizophrenia and addiction, the symptoms of which often manifest during this developmental period. Dopamine neurons in the ventral tegmental area (VTA) are strongly implicated in adolescent behavioral and psychiatric vulnerabilities, but little is known about how adolescent VTA neurons encode information during motivated behavior. METHODS We recorded daily from VTA neurons in adolescent and adult rats during learning and maintenance of a cued, reward-motivated instrumental task and extinction from this task. RESULTS During performance of the same motivated behavior, identical events were encoded differently by adult and adolescent VTA neurons. Adolescent VTA neurons with dopamine-like characteristics lacked a reward anticipation signal and showed a smaller response to reward delivery compared with adults. After extinction, however, these neurons maintained a strong phasic response to cues formerly predictive of reward opportunity. CONCLUSIONS Anticipatory neuronal activity in the VTA supports preparatory attention and is implicated in error prediction signaling. Absence of this activity, combined with persistent representations of previously rewarded experiences, may provide a mechanism for rash decision making in adolescents.
Collapse
Affiliation(s)
- Yunbok Kim
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nicholas W Simon
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jesse Wood
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bita Moghaddam
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania..
| |
Collapse
|
49
|
Abstract
Despite a lack of recent progress in the treatment of schizophrenia, our understanding of its genetic and environmental causes has considerably improved, and their relationship to aberrant patterns of neurodevelopment has become clearer. This raises the possibility that 'disease-modifying' strategies could alter the course to - and of - this debilitating disorder, rather than simply alleviating symptoms. A promising window for course-altering intervention is around the time of the first episode of psychosis, especially in young people at risk of transition to schizophrenia. Indeed, studies performed in both individuals at risk of developing schizophrenia and rodent models for schizophrenia suggest that pre-diagnostic pharmacotherapy and psychosocial or cognitive-behavioural interventions can delay or moderate the emergence of psychosis. Of particular interest are 'hybrid' strategies that both relieve presenting symptoms and reduce the risk of transition to schizophrenia or another psychiatric disorder. This Review aims to provide a broad-based consideration of the challenges and opportunities inherent in efforts to alter the course of schizophrenia.
Collapse
|
50
|
Methyl-Arginine Profile of Brain from Aged PINK1-KO+A53T-SNCA Mice Suggests Altered Mitochondrial Biogenesis. PARKINSONS DISEASE 2016; 2016:4686185. [PMID: 27034888 PMCID: PMC4791501 DOI: 10.1155/2016/4686185] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 11/29/2022]
Abstract
Hereditary Parkinson's disease can be triggered by an autosomal dominant overdose of alpha-Synuclein (SNCA) or the autosomal recessive deficiency of PINK1. We recently showed that the combination of PINK1-knockout with overexpression of A53T-SNCA in double mutant (DM) mice potentiates phenotypes and reduces survival. Now we studied brain hemispheres of DM mice at age of 18 months in a hypothesis-free approach, employing a quantitative label-free global proteomic mass spectrometry scan of posttranslational modifications focusing on methyl-arginine. The strongest effects were documented for the adhesion modulator CMAS, the mRNA decapping/deadenylation factor PATL1, and the synaptic plasticity mediator CRTC1/TORC1. In addition, an intriguing effect was observed for the splicing factor PSF/SFPQ, known to interact with the dopaminergic differentiation factor NURR1 as well as with DJ-1, the protein responsible for the autosomal recessive PARK7 variant of PD. CRTC1, PSF, and DJ-1 are modulators of PGC1alpha and of mitochondrial biogenesis. This pathway was further stressed by dysregulations of oxygen sensor EGLN3 and of nuclear TMPO. PSF and TMPO cooperate with dopaminergic differentiation factors LMX1B and NURR1. Further dysregulations concerned PRR18, TRIO, HNRNPA1, DMWD, WAVE1, ILDR2, DBNDD1, and NFM. Thus, we report selective novel endogenous stress responses in brain, which highlight early dysregulations of mitochondrial homeostasis and midbrain vulnerability.
Collapse
|