1
|
Fournier LA, Phadke RA, Salgado M, Brack A, Nocon JC, Bolshakova S, Grant JR, Padró Luna NM, Sen K, Cruz-Martín A. Overexpression of the schizophrenia risk gene C4 in PV cells drives sex-dependent behavioral deficits and circuit dysfunction. iScience 2024; 27:110800. [PMID: 39310747 PMCID: PMC11416532 DOI: 10.1016/j.isci.2024.110800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/09/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Fast-spiking parvalbumin (PV)-positive cells are key players in orchestrating pyramidal neuron activity, and their dysfunction is consistently observed in myriad brain diseases. To understand how immune complement pathway dysregulation in PV cells drives disease pathogenesis, we have developed a transgenic line that permits cell-type specific overexpression of the schizophrenia-associated C4 gene. We found that overexpression of mouse C4 (mC4) in PV cells causes sex-specific alterations in anxiety-like behavior and deficits in synaptic connectivity and excitability of PFC PV cells. Using a computational model, we demonstrated that these microcircuit deficits led to hyperactivity and disrupted neural communication. Finally, pan-neuronal overexpression of mC4 failed to evoke the same deficits in behavior as PV-specific mC4 overexpression, suggesting that perturbations of this neuroimmune gene in fast-spiking neurons are especially detrimental to circuits associated with anxiety-like behavior. Together, these results provide a causative link between C4 and the vulnerability of PV cells in brain disease.
Collapse
Affiliation(s)
- Luke A. Fournier
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Rhushikesh A. Phadke
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
| | - Maria Salgado
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Alison Brack
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
| | - Jian Carlo Nocon
- Neurophotonics Center, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
- Hearing Research Center, Boston University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sonia Bolshakova
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
- Bioinformatics MS Program, Boston University, Boston, MA, USA
| | - Jaylyn R. Grant
- Biological Sciences, Eastern Illinois University, Charleston, IL, USA
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, MA, USA
| | - Nicole M. Padró Luna
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, MA, USA
- Biology Department, College of Natural Sciences, University of Puerto Rico, Rio Piedras Campus, San Juan, PR, USA
| | - Kamal Sen
- Neurophotonics Center, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
- Hearing Research Center, Boston University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Alberto Cruz-Martín
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- NeuroTechnology Center (NTC), University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
2
|
Chen P, Chen W, Xu L, Luan L, Peng R, Zhang X, Yang H. Decreased serum VEGF and NRG1β1 levels in male patients with chronic schizophrenia: VEGF correlation with clinical symptoms and cognitive deficits. J Psychiatr Res 2024; 176:85-92. [PMID: 38850582 DOI: 10.1016/j.jpsychires.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/19/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) and neuregulin1 (NRG1) are multifunctional trophic factors reported to be dysregulated in schizophrenia. However, the relationships between serum concentrations and schizophrenia symptoms have differed markedly across studies, possibly because schizophrenia is a highly heterogenous disorder. The aim of this study was to investigate the associations of serum VEGF and NRG1 with clinical symptoms and cognitive deficits specifically in male patients with chronic schizophrenia. METHODS The study included 79 male patients with chronic schizophrenia and 79 matched healthy individuals. Serum VEGF, NRG1β1, S100B, S100A8, and neuropilin1 were measured using the Luminex liquid suspension chip detection method, psychopathological symptom severity using the Positive and Negative Symptom Scale (PANSS), and cognitive dysfunction using the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS). RESULTS Serum VEGF and NRG1β1 concentrations were significantly lower in male chronic schizophrenic patients than healthy controls (P < 0.05), while serum S100B, S100A8, and neuropilin1 concentrations did not differ between groups (P > 0.05). Serum VEGF concentration was negatively correlated with PANSS negative subscore (beta = -0.220, t = -2.07, P = 0.042), general psychopathology subscore (beta = -0.269, t = -2.55, P = 0.013), and total score (beta = -0.234, t = -2.12, P = 0.038), and positively correlated with RBANS language score (beta = 0.218, t = 2.03, P = 0.045). Alternatively, serum NRG1β1 concentration was not correlated with clinical symptoms or cognitive deficits (all P > 0.05). CONCLUSION Dysregulation of VEGF and NRG1β1 signaling may contribute to the pathogenesis of chronic schizophrenia in males. Moreover, abnormal VEGF signaling may contribute directly or through intermediary processes to neuropsychiatric and cognitive symptom expression.
Collapse
Affiliation(s)
- Peng Chen
- Institute of Mental Health, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, PR China.
| | - Wanming Chen
- Department of Psychiatry, The Fourth People's Hospital of Lianyungang, The Affiliated KangDa College of Nanjing Medical University, Lianyungang, 222003, PR China; Yangzhou University, Yangzhou, 225003, PR China.
| | - Li Xu
- Department of Psychiatry, The Fourth People's Hospital of Lianyungang, The Affiliated KangDa College of Nanjing Medical University, Lianyungang, 222003, PR China; Yangzhou University, Yangzhou, 225003, PR China.
| | - Lingshu Luan
- Department of Psychiatry, The Fourth People's Hospital of Lianyungang, The Affiliated KangDa College of Nanjing Medical University, Lianyungang, 222003, PR China.
| | - Ruijie Peng
- Institute of Mental Health, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, PR China.
| | - Xiaobin Zhang
- Institute of Mental Health, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, PR China.
| | - Haidong Yang
- Department of Psychiatry, The Fourth People's Hospital of Lianyungang, The Affiliated KangDa College of Nanjing Medical University, Lianyungang, 222003, PR China.
| |
Collapse
|
3
|
Fournier LA, Phadke RA, Salgado M, Brack A, Nocon JC, Bolshakova S, Grant JR, Padró Luna NM, Sen K, Cruz-Martín A. Overexpression of the schizophrenia risk gene C4 in PV cells drives sex-dependent behavioral deficits and circuit dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.27.575409. [PMID: 38328248 PMCID: PMC10849664 DOI: 10.1101/2024.01.27.575409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Fast-spiking parvalbumin (PV)-positive cells are key players in orchestrating pyramidal neuron activity, and their dysfunction is consistently observed in myriad brain diseases. To understand how immune complement dysregulation - a prevalent locus of brain disease etiology - in PV cells may drive disease pathogenesis, we have developed a transgenic mouse line that permits cell-type specific overexpression of the schizophrenia-associated complement component 4 (C4) gene. We found that overexpression of mouse C4 (mC4) in PV cells causes sex-specific behavioral alterations and concomitant deficits in synaptic connectivity and excitability of PV cells of the prefrontal cortex. Using a computational network, we demonstrated that these microcircuit deficits led to hyperactivity and disrupted neural communication. Finally, pan-neuronal overexpression of mC4 failed to evoke the same deficits in behavior as PV-specific mC4 overexpression, suggesting that C4 perturbations in fast-spiking neurons are more harmful to brain function than pan-neuronal alterations. Together, these results provide a causative link between C4 and the vulnerability of PV cells in brain disease.
Collapse
Affiliation(s)
- Luke A. Fournier
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, United States
| | - Rhushikesh A. Phadke
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, United States
| | - Maria Salgado
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, United States
| | - Alison Brack
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, United States
| | - Jian Carlo Nocon
- Neurophotonics Center, Boston University, Boston, Massachusetts, United States
- Center for Systems Neuroscience, Boston University, Boston, Massachusetts, United States
- Hearing Research Center, Boston University, Boston, Massachusetts, United States
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States
| | - Sonia Bolshakova
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, United States
- Bioinformatics MS Program, Boston University, Boston, MA, United States
| | - Jaylyn R. Grant
- Biological Sciences, Eastern Illinois University, Charleston, IL, United States
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, United States
| | - Nicole M. Padró Luna
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, United States
- Biology Department, College of Natural Sciences, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico
| | - Kamal Sen
- Neurophotonics Center, Boston University, Boston, Massachusetts, United States
- Center for Systems Neuroscience, Boston University, Boston, Massachusetts, United States
- Hearing Research Center, Boston University, Boston, Massachusetts, United States
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States
| | - Alberto Cruz-Martín
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, United States
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, United States
| |
Collapse
|
4
|
Heider J, Stahl A, Sperlich D, Hartmann SM, Vogel S, Breitmeyer R, Templin M, Volkmer H. Defined co-cultures of glutamatergic and GABAergic neurons with a mutation in DISC1 reveal aberrant phenotypes in GABAergic neurons. BMC Neurosci 2024; 25:12. [PMID: 38438989 PMCID: PMC10910844 DOI: 10.1186/s12868-024-00858-z] [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: 11/21/2023] [Accepted: 02/26/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Mutations in the gene DISC1 are associated with increased risk for schizophrenia, bipolar disorder and major depression. The study of mutated DISC1 represents a well-known and comprehensively characterized approach to understand neuropsychiatric disease mechanisms. However, previous studies have mainly used animal models or rather heterogeneous populations of iPSC-derived neurons, generated by undirected differentiation, to study the effects of DISC1 disruption. Since major hypotheses to explain neurodevelopmental, psychiatric disorders rely on altered neuronal connectivity observed in patients, an ideal iPSC-based model requires accurate representation of the structure and complexity of neuronal circuitries. In this study, we made use of an isogenic cell line with a mutation in DISC1 to study neuronal synaptic phenotypes in a culture system comprising a defined ratio of NGN2 and ASCL1/DLX2 (AD2)-transduced neurons, enriched for glutamatergic and GABAergic neurons, respectively, to mimic properties of the cortical microcircuitry. RESULTS In heterozygous DISC1 mutant neurons, we replicated the expected phenotypes including altered neural progenitor proliferation as well as neurite outgrowth, deregulated DISC1-associated signaling pathways, and reduced synaptic densities in cultures composed of glutamatergic neurons. Cultures comprising a defined ratio of NGN2 and AD2 neurons then revealed considerably increased GABAergic synapse densities, which have not been observed in any iPSC-derived model so far. Increased inhibitory synapse densities could be associated with an increased efficiency of GABAergic differentiation, which we observed in AD2-transduced cultures of mutant neurons. Additionally, we found increased neuronal activity in GABAergic neurons through calcium imaging while the activity pattern of glutamatergic neurons remained unchanged. CONCLUSIONS In conclusion, our results demonstrate phenotypic differences in a co-culture comprising a defined ratio of DISC1 mutant NGN2 and AD2 neurons, as compared to culture models comprising only one neuronal cell type. Altered synapse numbers and neuronal activity imply that DISC1 impacts the excitatory/inhibitory balance in NGN2/AD2 co-cultures, mainly through increased GABAergic input.
Collapse
Affiliation(s)
- Johanna Heider
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Aaron Stahl
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Denise Sperlich
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Sophia-Marie Hartmann
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Sabrina Vogel
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Ricarda Breitmeyer
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Markus Templin
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Hansjürgen Volkmer
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany.
| |
Collapse
|
5
|
Gawande DY, S Narasimhan KK, Shelkar GP, Pavuluri R, Stessman HAF, Dravid SM. GluN2D Subunit in Parvalbumin Interneurons Regulates Prefrontal Cortex Feedforward Inhibitory Circuit and Molecular Networks Relevant to Schizophrenia. Biol Psychiatry 2023; 94:297-309. [PMID: 37004850 PMCID: PMC10524289 DOI: 10.1016/j.biopsych.2023.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/01/2023] [Accepted: 03/21/2023] [Indexed: 04/04/2023]
Abstract
BACKGROUND Parvalbumin interneuron (PVI) activity synchronizes the medial prefrontal cortex circuit for normal cognitive function, and its impairment may contribute to schizophrenia (SZ). NMDA receptors in PVIs participate in these activities and form the basis for the NMDA receptor hypofunction hypothesis of SZ. However, the role of the GluN2D subunit, which is enriched in PVIs, in regulating molecular networks relevant to SZ is unknown. METHODS Using electrophysiology and a mouse model with conditional deletion of GluN2D from PVIs (PV-GluN2D knockout [KO]), we examined the cell excitability and neurotransmission in the medial prefrontal cortex. Histochemical, RNA sequencing analysis and immunoblotting were conducted to understand molecular mechanisms. Behavioral analysis was conducted to test cognitive function. RESULTS PVIs in the medial prefrontal cortex were found to express putative GluN1/2B/2D receptors. In a PV-GluN2D KO model, PVIs were hypoexcitable, whereas pyramidal neurons were hyperexcitable. Excitatory neurotransmission was higher in both cell types in PV-GluN2D KO, whereas inhibitory neurotransmission showed contrasting changes, which could be explained by reduced somatostatin interneuron projections and increased PVI projections. Genes associated with GABA (gamma-aminobutyric acid) synthesis, vesicular release, and uptake as well as those involved in formation of inhibitory synapses, specifically GluD1-Cbln4 and Nlgn2, and regulation of dopamine terminals were downregulated in PV-GluN2D KO. SZ susceptibility genes including Disc1, Nrg1, and ErbB4 and their downstream targets were also downregulated. Behaviorally, PV-GluN2D KO mice showed hyperactivity and anxiety behavior and deficits in short-term memory and cognitive flexibility. CONCLUSIONS These findings demonstrate that GluN2D in PVIs serves as a point of convergence of pathways involved in the regulation of GABAergic synapses relevant to SZ.
Collapse
Affiliation(s)
- Dinesh Y Gawande
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska
| | | | - Gajanan P Shelkar
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska
| | - Ratnamala Pavuluri
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska
| | - Holly A F Stessman
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska
| | - Shashank M Dravid
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska.
| |
Collapse
|
6
|
Sauer JF, Bartos M. Disrupted-in-schizophrenia-1 is required for normal pyramidal cell-interneuron communication and assembly dynamics in the prefrontal cortex. eLife 2022; 11:79471. [PMID: 36239988 PMCID: PMC9566853 DOI: 10.7554/elife.79471] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
We interrogated prefrontal circuit function in mice lacking Disrupted-in-schizophrenia-1 (Disc1-mutant mice), a risk factor for psychiatric disorders. Single-unit recordings in awake mice revealed reduced average firing rates of fast-spiking interneurons (INTs), including optogenetically identified parvalbumin-positive cells, and a lower proportion of INTs phase-coupled to ongoing gamma oscillations. Moreover, we observed decreased spike transmission efficacy at local pyramidal cell (PYR)-INT connections in vivo, suggesting a reduced excitatory effect of local glutamatergic inputs as a potential mechanism of lower INT rates. On the network level, impaired INT function resulted in altered activation of PYR assemblies: While assembly activations defined as coactivations within 25 ms were observed equally often, the expression strength of individual assembly patterns was significantly higher in Disc1-mutant mice. Our data, thus, reveal a role of Disc1 in shaping the properties of prefrontal assembly patterns by setting INT responsiveness to glutamatergic drive.
Collapse
Affiliation(s)
- Jonas-Frederic Sauer
- Institute for Physiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marlene Bartos
- Institute for Physiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
7
|
Elevated peripheral Neuregulin-1 protein levels in non-medicated focal epilepsy patients. J Clin Neurosci 2022; 102:1-4. [DOI: 10.1016/j.jocn.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 11/21/2022]
|
8
|
Hu L, Zhang L. Adult neural stem cells and schizophrenia. World J Stem Cells 2022; 14:219-230. [PMID: 35432739 PMCID: PMC8968214 DOI: 10.4252/wjsc.v14.i3.219] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/18/2021] [Accepted: 03/07/2022] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia (SCZ) is a devastating and complicated mental disorder accompanied by variable positive and negative symptoms and cognitive deficits. Although many genetic risk factors have been identified, SCZ is also considered as a neurodevelopmental disorder. Elucidation of the pathogenesis and the development of treatment is challenging because complex interactions occur between these genetic risk factors and environment in essential neurodevelopmental processes. Adult neural stem cells share a lot of similarities with embryonic neural stem cells and provide a promising model for studying neuronal development in adulthood. These adult neural stem cells also play an important role in cognitive functions including temporal and spatial memory encoding and context discrimination, which have been shown to be closely linked with many psychiatric disorders, such as SCZ. Here in this review, we focus on the SCZ risk genes and the key components in related signaling pathways in adult hippocampal neural stem cells and summarize their roles in adult neurogenesis and animal behaviors. We hope that this would be helpful for the understanding of the contribution of dysregulated adult neural stem cells in the pathogenesis of SCZ and for the identification of potential therapeutic targets, which could facilitate the development of novel medication and treatment.
Collapse
Affiliation(s)
- Ling Hu
- Department of Laboratory Animal Science and Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Lei Zhang
- Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center) and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| |
Collapse
|
9
|
Regulation of sensorimotor gating via Disc1/Huntingtin-mediated Bdnf transport in the cortico-striatal circuit. Mol Psychiatry 2022; 27:1805-1815. [PMID: 35165396 PMCID: PMC9272458 DOI: 10.1038/s41380-021-01389-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 10/15/2021] [Accepted: 11/03/2021] [Indexed: 11/30/2022]
Abstract
Sensorimotor information processing underlies normal cognitive and behavioral traits and has classically been evaluated through prepulse inhibition (PPI) of a startle reflex. PPI is a behavioral dimension deregulated in several neurological and psychiatric disorders, yet the mechanisms underlying the cross-diagnostic nature of PPI deficits across these conditions remain to be understood. To identify circuitry mechanisms for PPI, we performed circuitry recording over the prefrontal cortex and striatum, two brain regions previously implicated in PPI, using wild-type (WT) mice compared to Disc1-locus-impairment (LI) mice, a model representing neuropsychiatric conditions. We demonstrated that the corticostriatal projection regulates neurophysiological responses during the PPI testing in WT, whereas these circuitry responses were disrupted in Disc1-LI mice. Because our biochemical analyses revealed attenuated brain-derived neurotrophic factor (Bdnf) transport along the corticostriatal circuit in Disc1-LI mice, we investigated the potential role of Bdnf in this circuitry for regulation of PPI. Virus-mediated delivery of Bdnf into the striatum rescued PPI deficits in Disc1-LI mice. Pharmacologically augmenting Bdnf transport by chronic lithium administration, partly via phosphorylation of Huntingtin (Htt) serine-421 and its integration into the motor machinery, restored striatal Bdnf levels and rescued PPI deficits in Disc1-LI mice. Furthermore, reducing the cortical Bdnf expression negated this rescuing effect of lithium, confirming the key role of Bdnf in lithium-mediated PPI rescuing. Collectively, the data suggest that striatal Bdnf supply, collaboratively regulated by Htt and Disc1 along the corticostriatal circuit, is involved in sensorimotor gating, highlighting the utility of dimensional approach in investigating pathophysiological mechanisms across neuropsychiatric disorders.
Collapse
|
10
|
Mutations in DISC1 alter IP 3R and voltage-gated Ca 2+ channel functioning, implications for major mental illness. Neuronal Signal 2021; 5:NS20180122. [PMID: 34956649 PMCID: PMC8663806 DOI: 10.1042/ns20180122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/26/2021] [Accepted: 11/08/2021] [Indexed: 12/16/2022] Open
Abstract
Disrupted in Schizophrenia 1 (DISC1) participates in a wide variety of
developmental processes of central neurons. It also serves critical roles that
underlie cognitive functioning in adult central neurons. Here we summarize
DISC1’s general properties and discuss its use as a model system for
understanding major mental illnesses (MMIs). We then discuss the cellular
actions of DISC1 that involve or regulate Ca2+ signaling in adult
central neurons. In particular, we focus on the tethering role DISC1 plays in
transporting RNA particles containing Ca2+ channel subunit RNAs,
including IP3R1, CACNA1C and CACNA2D1, and in transporting mitochondria into
dendritic and axonal processes. We also review DISC1’s role in modulating
IP3R1 activity within mitochondria-associated ER membrane (MAM).
Finally, we discuss DISC1-glycogen synthase kinase 3β (GSK3β)
signaling that regulates functional expression of voltage-gated Ca2+
channels (VGCCs) at central synapses. In each case, DISC1 regulates the movement
of molecules that impact Ca2+ signaling in neurons.
Collapse
|
11
|
Wang X, Ye F, Wen Z, Guo Z, Yu C, Huang WK, Rojas Ringeling F, Su Y, Zheng W, Zhou G, Christian KM, Song H, Zhang M, Ming GL. Structural interaction between DISC1 and ATF4 underlying transcriptional and synaptic dysregulation in an iPSC model of mental disorders. Mol Psychiatry 2021; 26:1346-1360. [PMID: 31444471 PMCID: PMC8444148 DOI: 10.1038/s41380-019-0485-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 04/01/2019] [Accepted: 05/17/2019] [Indexed: 01/01/2023]
Abstract
Psychiatric disorders are a collection of heterogeneous mental disorders arising from a contribution of genetic and environmental insults, many of which molecularly converge on transcriptional dysregulation, resulting in altered synaptic functions. The underlying mechanisms linking the genetic lesion and functional phenotypes remain largely unknown. Patient iPSC-derived neurons with a rare frameshift DISC1 (Disrupted-in-schizophrenia 1) mutation have previously been shown to exhibit aberrant gene expression and deficits in synaptic functions. How DISC1 regulates gene expression is largely unknown. Here we show that Activating Transcription Factor 4 (ATF4), a DISC1 binding partner, is more abundant in the nucleus of DISC1 mutant human neurons and exhibits enhanced binding to a collection of dysregulated genes. Functionally, overexpressing ATF4 in control neurons recapitulates deficits seen in DISC1 mutant neurons, whereas transcriptional and synaptic deficits are rescued in DISC1 mutant neurons with CRISPR-mediated heterozygous ATF4 knockout. By solving the high-resolution atomic structure of the DISC1-ATF4 complex, we show that mechanistically, the mutation of DISC1 disrupts normal DISC1-ATF4 interaction, and results in excessive ATF4 binding to DNA targets and deregulated gene expression. Together, our study identifies the molecular and structural basis of an DISC1-ATF4 interaction underlying transcriptional and synaptic dysregulation in an iPSC model of mental disorders.
Collapse
Affiliation(s)
- Xinyuan Wang
- School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Fei Ye
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Center of Systems Biology and Human Health, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ziyuan Guo
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Chuan Yu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Wei-Kai Huang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Pathology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Francisca Rojas Ringeling
- The Human Genetics Pre-doctoral Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yijing Su
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Guomin Zhou
- School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China
| | - Kimberly M Christian
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Cell and Developmental Biology, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Regenerative Medicine, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- The Epigenetics Institute, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Mingjie Zhang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
- Center of Systems Biology and Human Health, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Cell and Developmental Biology, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Regenerative Medicine, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Psychiatry, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
12
|
Functional brain defects in a mouse model of a chromosomal t(1;11) translocation that disrupts DISC1 and confers increased risk of psychiatric illness. Transl Psychiatry 2021; 11:135. [PMID: 33608504 PMCID: PMC7895946 DOI: 10.1038/s41398-021-01256-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 11/24/2020] [Accepted: 01/22/2021] [Indexed: 12/17/2022] Open
Abstract
A balanced t(1;11) translocation that directly disrupts DISC1 is linked to schizophrenia and affective disorders. We previously showed that a mutant mouse, named Der1, recapitulates the effect of the translocation upon DISC1 expression. Here, RNAseq analysis of Der1 mouse brain tissue found enrichment for dysregulation of the same genes and molecular pathways as in neuron cultures generated previously from human t(1;11) translocation carriers via the induced pluripotent stem cell route. DISC1 disruption therefore apparently accounts for a substantial proportion of the effects of the t(1;11) translocation. RNAseq and pathway analysis of the mutant mouse predicts multiple Der1-induced alterations converging upon synapse function and plasticity. Synaptosome proteomics confirmed that the Der1 mutation impacts synapse composition, and electrophysiology found reduced AMPA:NMDA ratio in hippocampal neurons, indicating changed excitatory signalling. Moreover, hippocampal parvalbumin-positive interneuron density is increased, suggesting that the Der1 mutation affects inhibitory control of neuronal circuits. These phenotypes predict that neurotransmission is impacted at many levels by DISC1 disruption in human t(1;11) translocation carriers. Notably, genes implicated in schizophrenia, depression and bipolar disorder by large-scale genetic studies are enriched among the Der1-dysregulated genes, just as we previously observed for the t(1;11) translocation carrier-derived neurons. Furthermore, RNAseq analysis predicts that the Der1 mutation primarily targets a subset of cell types, pyramidal neurons and interneurons, previously shown to be vulnerable to the effects of common schizophrenia-associated genetic variants. In conclusion, DISC1 disruption by the t(1;11) translocation may contribute to the psychiatric disorders of translocation carriers through commonly affected pathways and processes in neurotransmission.
Collapse
|
13
|
Lee SB, Park J, Kwak Y, Park YU, Nhung TTM, Suh BK, Woo Y, Suh Y, Cho E, Cho S, Park SK. Disrupted-in-schizophrenia 1 enhances the quality of circadian rhythm by stabilizing BMAL1. Transl Psychiatry 2021; 11:110. [PMID: 33542182 PMCID: PMC7862247 DOI: 10.1038/s41398-021-01212-1] [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: 06/30/2020] [Revised: 12/15/2020] [Accepted: 01/07/2021] [Indexed: 11/27/2022] Open
Abstract
Disrupted-in-schizophrenia 1 (DISC1) is a scaffold protein that has been implicated in multiple mental disorders. DISC1 is known to regulate neuronal proliferation, signaling, and intracellular calcium homeostasis, as well as neurodevelopment. Although DISC1 was linked to sleep-associated behaviors, whether DISC1 functions in the circadian rhythm has not been determined yet. In this work, we revealed that Disc1 expression exhibits daily oscillating pattern and is regulated by binding of circadian locomotor output cycles kaput (CLOCK) and Brain and muscle Arnt-like protein-1 (BMAL1) heterodimer to E-box sequences in its promoter. Interestingly, Disc1 deficiency increases the ubiquitination of BMAL1 and de-stabilizes it, thereby reducing its protein levels. DISC1 inhibits the activity of GSK3β, which promotes BMAL1 ubiquitination, suggesting that DISC1 regulates BMAL1 stability by inhibiting its ubiquitination. Moreover, Disc1-deficient cells and mice show reduced expression of other circadian genes. Finally, Disc1-LI (Disc1 knockout) mice exhibit damped circadian physiology and behaviors. Collectively, these findings demonstrate that the oscillation of DISC1 expression is under the control of CLOCK and BMAL1, and that DISC1 contributes to the core circadian system by regulating BMAL1 stability.
Collapse
Affiliation(s)
- Su Been Lee
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jihyun Park
- grid.289247.20000 0001 2171 7818Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Yongdo Kwak
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea ,grid.507563.2Present Address: SK biopharmaceuticals Ltd, Seongnam-Si, Republic of Korea
| | - Young-Un Park
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea ,grid.49606.3d0000 0001 1364 9317Present Address: Department of Pathology, College of Medicine, Hanyang University, Seoul, Korea
| | - Truong Thi My Nhung
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Bo Kyoung Suh
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Youngsik Woo
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Yeongjun Suh
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Eunbyul Cho
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Sehyung Cho
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea.
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea.
| |
Collapse
|
14
|
Yang JM, Shen CJ, Chen XJ, Kong Y, Liu YS, Li XW, Chen Z, Gao TM, Li XM. erbb4 Deficits in Chandelier Cells of the Medial Prefrontal Cortex Confer Cognitive Dysfunctions: Implications for Schizophrenia. Cereb Cortex 2020; 29:4334-4346. [PMID: 30590426 DOI: 10.1093/cercor/bhy316] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/18/2018] [Accepted: 11/21/2018] [Indexed: 12/20/2022] Open
Abstract
erbb4 is a known susceptibility gene for schizophrenia. Chandelier cells (ChCs, also known as axo-axonic cells) are a distinct GABAergic interneuron subtype that exclusively target the axonal initial segment, which is the site of pyramidal neuron action potential initiation. ChCs are a source of ErbB4 expression and alterations in ChC-pyramidal neuron connectivity occur in the medial prefrontal cortex (mPFC) of schizophrenic patients and animal models of schizophrenia. However, the contribution of ErbB4 in mPFC ChCs to the pathogenesis of schizophrenia remains unknown. By conditional deletion or knockdown of ErbB4 from mPFC ChCs, we demonstrated that ErbB4 deficits led to impaired ChC-pyramidal neuron connections and cognitive dysfunctions. Furthermore, the cognitive dysfunctions were normalized by L-838417, an agonist of GABAAα2 receptors enriched in the axonal initial segment. Given that cognitive dysfunctions are a core symptom of schizophrenia, our results may provide a new perspective for understanding the etiology of schizophrenia and suggest that GABAAα2 receptors may be potential pharmacological targets for its treatment.
Collapse
Affiliation(s)
- Jian-Ming Yang
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Chen-Jie Shen
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao-Juan Chen
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Kong
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yi-Si Liu
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao-Wen Li
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhong Chen
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tian-Ming Gao
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao-Ming Li
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
15
|
O'Brien K, Breyne K, Ughetto S, Laurent LC, Breakefield XO. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat Rev Mol Cell Biol 2020; 21:585-606. [PMID: 32457507 PMCID: PMC7249041 DOI: 10.1038/s41580-020-0251-y] [Citation(s) in RCA: 1032] [Impact Index Per Article: 258.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2020] [Indexed: 02/06/2023]
Abstract
The term 'extracellular vesicles' refers to a heterogeneous population of vesicular bodies of cellular origin that derive either from the endosomal compartment (exosomes) or as a result of shedding from the plasma membrane (microvesicles, oncosomes and apoptotic bodies). Extracellular vesicles carry a variety of cargo, including RNAs, proteins, lipids and DNA, which can be taken up by other cells, both in the direct vicinity of the source cell and at distant sites in the body via biofluids, and elicit a variety of phenotypic responses. Owing to their unique biology and roles in cell-cell communication, extracellular vesicles have attracted strong interest, which is further enhanced by their potential clinical utility. Because extracellular vesicles derive their cargo from the contents of the cells that produce them, they are attractive sources of biomarkers for a variety of diseases. Furthermore, studies demonstrating phenotypic effects of specific extracellular vesicle-associated cargo on target cells have stoked interest in extracellular vesicles as therapeutic vehicles. There is particularly strong evidence that the RNA cargo of extracellular vesicles can alter recipient cell gene expression and function. During the past decade, extracellular vesicles and their RNA cargo have become better defined, but many aspects of extracellular vesicle biology remain to be elucidated. These include selective cargo loading resulting in substantial differences between the composition of extracellular vesicles and source cells; heterogeneity in extracellular vesicle size and composition; and undefined mechanisms for the uptake of extracellular vesicles into recipient cells and the fates of their cargo. Further progress in unravelling the basic mechanisms of extracellular vesicle biogenesis, transport, and cargo delivery and function is needed for successful clinical implementation. This Review focuses on the current state of knowledge pertaining to packaging, transport and function of RNAs in extracellular vesicles and outlines the progress made thus far towards their clinical applications.
Collapse
Affiliation(s)
- Killian O'Brien
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Koen Breyne
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Stefano Ughetto
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Oncology, University of Turin, Candiolo, Italy
| | - Louise C Laurent
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, La Jolla, CA, USA.
- Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA.
| | - Xandra O Breakefield
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Yang X, Xu X, Cai X, He J, Lu P, Guo Q, Wang G, Zhu H, Wang H, Xue C. Gene set enrichment analysis and protein-protein interaction network analysis after sciatic nerve injury. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:988. [PMID: 32953788 PMCID: PMC7475449 DOI: 10.21037/atm-20-4958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background Peripheral nerves are able to regenerate spontaneously after injury. An increasing number of studies have investigated the mechanism of peripheral nerve regeneration and attempted to find potential therapeutic targets. The various bioinformatics analysis tools available, gene set enrichment analysis (GSEA) and protein-protein interaction (PPI) networks can effectively screen the crucial targets of neuroregeneration. Methods GSEA and PPI networks were constructed through ingenuity pathway analysis and sequential gene expression validation ex vitro to investigate the molecular processes at 1, 4, 7, and 14 days following sciatic nerve transection in rats. Results Immune response and the activation of related canonical pathways were classified as crucial biological events. Additionally, neural precursor cell expressed developmentally downregulated 4-like (NEDD4L), neuregulin 1 (NRG1), nuclear factor of activated T cells 2 (NFATC2), midline 1 (MID1), GLI family zinc finger 2 (GLI2), and ventral anterior homeobox 1 (VAX1), which were jointly involved in both immune response and axonal regeneration, were screened and their mRNA and protein expressions following nerve injury were validated. Among them, the expression of VAX1 continuously increased following nerve injury, and it was considered to be a potential therapeutic target. Conclusions The combined use of GSEA and PPI networks serves as a valuable way to identify potential therapeutic targets for neuroregeneration.
Collapse
Affiliation(s)
- Xiaoming Yang
- School of Biology and Basic Medical Sciences, Medical College of Soochow University, Suzhou, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xi Xu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaodong Cai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jin He
- School of Medicine, Nantong University, Nantong, China
| | - Panjian Lu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Qi Guo
- Department of Hand Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Gang Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Hui Zhu
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Hongkui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Chengbin Xue
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
17
|
Zhang Z, Li Y, He F, Cui Y, Zheng Y, Li R. Sex differences in circulating neuregulin1-β1 and β-secretase 1 expression in childhood-onset schizophrenia. Compr Psychiatry 2020; 100:152176. [PMID: 32430144 DOI: 10.1016/j.comppsych.2020.152176] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVE Early-onset schizophrenia is a severe and rare form of schizophrenia that is clinically and neurobiologically continuous with the adult form of schizophrenia. Neuregulin1 (NRG1)-mediated signaling is crucial for early neurodevelopment, which exerts its function by limited β-secretase 1 (BACE1) proteolysis processing. However, circulating neuregulin1-β1 (NRG1-β1), an isoform of NRG1, and its cleavage enzyme BACE1 have not been studied in early-onset patients with schizophrenia. METHODS In this study, we collected plasma and clinical information from 71 young patients (7 ≤ age years ≤20) with schizophrenia and 53 age- and sex-matched healthy controls. Immunoassay was used to test levels of circulating NRG1-β1 and BACE1 expression. We further analyzed the relationship of disease-onset age and gender with NRG1-β1 and BACE1 levels. RESULTS We found that circulating plasma levels of NRG1-β1 were significantly decreased in young patients with early-onset schizophrenia. In males with childhood onset schizophrenia (COS), NRG1-β1 was reduced and was inversely correlated with positive symptom of PANSS; moreover, these male patients with higher plasma BACE1 levels showed more severe general symptoms of PANSS and defective social functioning; whereas, no aforementioned results were found in adolescent-onset schizophrenia (AOS). Notably, young female patients with COS and AOS had no significant change in NRG1-β1 and BACE1, which demonstrated a sex-dependent effect in early-onset schizophrenia. CONCLUSION Our results suggest that decreased levels of NRG1-β1 and its cleavage enzyme BACE1 contribute to increased risk of etiology of schizophrenia. Synthetic biomarkers may have clinical applications for the early diagnosis of male COS.
Collapse
Affiliation(s)
- Zhengrong Zhang
- National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Yuhong Li
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Fan He
- National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Yonghua Cui
- National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Yi Zheng
- National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, 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.
| |
Collapse
|
18
|
Delevich K, Jaaro-Peled H, Penzo M, Sawa A, Li B. Parvalbumin Interneuron Dysfunction in a Thalamo-Prefrontal Cortical Circuit in Disc1 Locus Impairment Mice. eNeuro 2020; 7:ENEURO.0496-19.2020. [PMID: 32029441 PMCID: PMC7054897 DOI: 10.1523/eneuro.0496-19.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/22/2020] [Accepted: 01/27/2020] [Indexed: 01/22/2023] Open
Abstract
Altered cortical excitation-inhibition (E-I) balance resulting from abnormal parvalbumin interneuron (PV IN) function is a proposed pathophysiological mechanism of schizophrenia and other major psychiatric disorders. Preclinical studies have indicated that disrupted-in-schizophrenia-1 (Disc1) is a useful molecular lead to address the biology of prefrontal cortex (PFC)-dependent cognition and PV IN function. To date, PFC inhibitory circuit function has not been investigated in depth in Disc1 locus impairment (LI) mouse models. Therefore, we used a Disc1 LI mouse model to investigate E-I balance in medial PFC (mPFC) circuits. We found that inhibition onto layer 2/3 excitatory pyramidal neurons in the mPFC was significantly reduced in Disc1 LI mice. This reduced inhibition was accompanied by decreased GABA release from local PV, but not somatostatin (SOM) INs, and by impaired feedforward inhibition (FFI) in the mediodorsal thalamus (MD) to mPFC circuit. Our mechanistic findings of abnormal PV IN function in a Disc1 LI model provide insight into biology that may be relevant to neuropsychiatric disorders including schizophrenia.
Collapse
Affiliation(s)
- Kristen Delevich
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Hanna Jaaro-Peled
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Mario Penzo
- National Institute of Mental Health, Bethesda, MD 20892
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Bo Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
19
|
Barnett BR, Anderson JM, Torres-Velázquez M, Yi SY, Rowley PA, Yu JPJ. Exercise ameliorates deficits in neural microstructure in a Disc1 model of psychiatric illness. Magn Reson Imaging 2019; 61:90-96. [PMID: 31103832 PMCID: PMC6663582 DOI: 10.1016/j.mri.2019.05.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 01/06/2023]
Abstract
Recent studies have investigated the effectiveness of aerobic exercise to improve physical and mental health outcomes in schizophrenia; however, few have explicitly explored the impact of aerobic exercise on neural microstructure, which is hypothesized to mediate the behavioral changes observed. Neural microstructure is influenced by numerous genetic factors including DISC1, which is a major molecular scaffold protein that interacts with partners like GSK3β, NDEL1, and PDE4. DISC1 has been shown to play a role in neurogenesis, neuronal migration, neuronal maturation, and synaptic signaling. As with other genetic variants that present an increased risk for disease, mutations of the DISC1 gene have been implicated in the molecular intersection of schizophrenia and numerous other major psychiatric illnesses. This study investigated whether short-term exercise recovers deficits in neural microstructure in a novel genetic Disc1 svΔ2 rat model. Disc1 svΔ2 animals and age- and sex-matched controls were subjected to a treadmill exercise protocol. Subsequent ex-vivo diffusion tensor imaging (DTI) and neurite orientation dispersion and density imaging (NODDI) compared neural microstructure in regions of interest (ROI) between sedentary and exercise wild-type animals and between sedentary and exercise Disc1 svΔ2 animals. Short-term exercise uncovered no significant differences in neural microstructure between sedentary and exercise control animals but did lead to significant differences between sedentary and exercise Disc1 svΔ2 animals in neocortex, basal ganglia, corpus callosum, and external capsule, suggesting a positive benefit derived from a short-term exercise regimen. Our findings suggest that Disc1 svΔ2 animals are more sensitive to the effects of short-term exercise and highlight the ameliorating potential of positive treatment interventions such as exercise on neural microstructure in genetic backgrounds of psychiatric disease susceptibility.
Collapse
Affiliation(s)
- Brian R Barnett
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jacqueline M Anderson
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Maribel Torres-Velázquez
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sue Y Yi
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Paul A Rowley
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - John-Paul J Yu
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA.
| |
Collapse
|
20
|
Kang E, Song J, Lin Y, Park J, Lee JH, Hussani Q, Gu Y, Ge S, Li W, Hsu KS, Berninger B, Christian KM, Song H, Ming GL. Interplay between a Mental Disorder Risk Gene and Developmental Polarity Switch of GABA Action Leads to Excitation-Inhibition Imbalance. Cell Rep 2019; 28:1419-1428.e3. [PMID: 31390557 PMCID: PMC6690484 DOI: 10.1016/j.celrep.2019.07.024] [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: 06/19/2018] [Revised: 05/29/2019] [Accepted: 07/10/2019] [Indexed: 12/17/2022] Open
Abstract
Excitation-inhibition (E-I) imbalance is considered a hallmark of various neurodevelopmental disorders, including schizophrenia and autism. How genetic risk factors disrupt coordinated glutamatergic and GABAergic synapse formation to cause an E-I imbalance is not well understood. Here, we show that knockdown of Disrupted-in-schizophrenia 1 (DISC1), a risk gene for major mental disorders, leads to E-I imbalance in mature dentate granule neurons. We found that excessive GABAergic inputs from parvalbumin-, but not somatostatin-, expressing interneurons enhance the formation of both glutamatergic and GABAergic synapses in immature mutant neurons. Following the switch in GABAergic signaling polarity from depolarizing to hyperpolarizing during neuronal maturation, heightened inhibition from excessive parvalbumin+ GABAergic inputs causes loss of excitatory glutamatergic synapses in mature mutant neurons, resulting in an E-I imbalance. Our findings provide insights into the developmental role of depolarizing GABA in establishing E-I balance and how it can be influenced by genetic risk factors for mental disorders.
Collapse
Affiliation(s)
- Eunchai Kang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Juan Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Neuroscience Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yuting Lin
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City 701, Taiwan
| | - Jaesuk Park
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jennifer H Lee
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Qassim Hussani
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yan Gu
- Department of Neurobiology and Behavior, State University of New York at Stony Brook, Stony Brook, NY 11794, USA
| | - Shaoyu Ge
- Department of Neurobiology and Behavior, State University of New York at Stony Brook, Stony Brook, NY 11794, USA
| | - Weidong Li
- Bio-X Institute, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
| | - Kuei-Sen Hsu
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City 701, Taiwan
| | - Benedikt Berninger
- Center for Developmental Neurobiology, King's College London, London SE1UL, UK
| | - Kimberly M Christian
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
21
|
Yang K, Kondo MA, Jaaro-Peled H, Cash-Padgett T, Kano SI, Ishizuka K, Pevsner J, Tomoda T, Sawa A, Niwa M. The transcriptome landscape associated with Disrupted-in-Schizophrenia-1 locus impairment in early development and adulthood. Schizophr Res 2019; 210:149-156. [PMID: 31204062 PMCID: PMC8050833 DOI: 10.1016/j.schres.2019.05.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/21/2019] [Accepted: 05/26/2019] [Indexed: 01/08/2023]
Abstract
DISC1 was originally expected to be a genetic risk factor for schizophrenia, but the genome wide association studies have not supported this idea. In contrast, neurobiological studies of DISC1 in cell and animal models have demonstrated that direct perturbation of DISC1 protein elicits neurobiological and behavioral abnormalities relevant to a wide range of psychiatric conditions, in particular psychosis. Thus, the utility of DISC1 as a biological lead for psychosis research is clear. In the present study, we aimed to capture changes in the molecular landscape in the prefrontal cortex upon perturbation of DISC1, using the Disc1 locus impairment (Disc1-LI) model in which the majority of Disc1 isoforms have been depleted, and to explore potential molecular mediators relevant to psychiatric conditions. We observed a robust change in gene expression profile elicited by Disc1-LI in which the stronger effects on molecular networks were observed in early stage compared with those in adulthood. Significant alterations were found in specific pathways relevant to psychiatric conditions, such as pathways of signaling by G protein-coupled receptor, neurotransmitter release cycle, and voltage gated potassium channels. The differentially expressed genes (DEGs) between Disc1-LI and wild-type mice are significantly enriched not only in neurons, but also in astrocytes and oligodendrocyte precursor cells. The brain-disorder-associated genes at the mRNA and protein levels rather than those at the genomic levels are enriched in the DEGs. Together, our present study supports the utility of Disc1-LI mice in biological research for psychiatric disorder-associated molecular networks.
Collapse
Affiliation(s)
- Kun Yang
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mari A Kondo
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hanna Jaaro-Peled
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Tyler Cash-Padgett
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Shin-Ichi Kano
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Koko Ishizuka
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jonathan Pevsner
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Toshifumi Tomoda
- Medical Innovation Center, Kyoto University, Kyoto 606-8397, Japan
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Mental Health, Johns Hopkins University Bloomberg School of Medicine, Baltimore, MD 21205, USA.
| | - Minae Niwa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
22
|
Ooms M, Tsujikawa T, Lohith TG, Mabins SN, Zoghbi SS, Sumitomo A, Jaaro-Peled H, Kimura Y, Telu S, Pike VW, Tomoda T, Innis RB, Sawa A, Fujita M. [ 11C]( R)-Rolipram positron emission tomography detects DISC1 inhibition of phosphodiesterase type 4 in live Disc1 locus-impaired mice. J Cereb Blood Flow Metab 2019; 39:1306-1313. [PMID: 29430995 PMCID: PMC6668514 DOI: 10.1177/0271678x18758997] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Although still a matter of controversy, disrupted in schizophrenia protein 1 (DISC1) was suggested as a potential inhibitor of phosphodiesterase 4 (PDE4). We used Disc1 locus impairment (LI) mice to investigate the interaction between PDE4 and DISC 1 in vivo and in vitro. [11C](R)-Rolipram binding was measured by PET in LI (n = 11) and C57BL/6 wild-type (WT, n = 9) mice. [11C](R)-Rolipram total distribution volumes (VT) were calculated and corrected for plasma-free fraction (fP) measured in a separate group of LI (n = 6) and WT (n = 7) mice. PDE4 enzyme activity was measured using in vitro samples of cerebral cortices from groups of LI (n = 4), heterozygote (n = 4), and WT (n = 4) mice. Disc1 LI mice showed a 41% increase in VT (18 ± 6 vs. 13±4 mL/cm3, P = 0.04) compared to WT mice. VT/fP showed a 73% significant increase (90 ± 31 vs. 52 ± 15 mL/cm3, P = 0.004) in Disc1 LI compared to WT mice. PDE4 enzymatic activity assay confirmed in vivo findings showing significant group differences (p < 0.0001). In conclusion, PDE4 activity was increased in the absence of critical DISC1 protein isoforms both in vivo and in vitro. Additionally, [11C](R)-Rolipram PET was sensitive enough to assess altered PDE4 activity caused by PDE4-DISC1 interaction.
Collapse
Affiliation(s)
- Maarten Ooms
- 1 Molecular Imaging Branch, NIMH, Bethesda, MD, USA
| | | | | | | | | | - Akiko Sumitomo
- 2 Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hanna Jaaro-Peled
- 3 Department of Psychiatry, Johns Hopkins University School of Medicine, MD, USA
| | | | - Sanjay Telu
- 1 Molecular Imaging Branch, NIMH, Bethesda, MD, USA
| | | | - Toshifumi Tomoda
- 2 Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Akira Sawa
- 3 Department of Psychiatry, Johns Hopkins University School of Medicine, MD, USA
| | | |
Collapse
|
23
|
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: 2.6] [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
|
24
|
Liu CM, Liu YL, Hwu HG, Fann CSJ, Yang UC, Hsu PC, Chang CC, Chen WJ, Hwang TJ, Hsieh MH, Liu CC, Chien YL, Lin YT, Tsuang MT. Genetic associations and expression of extra-short isoforms of disrupted-in-schizophrenia 1 in a neurocognitive subgroup of schizophrenia. J Hum Genet 2019; 64:653-663. [PMID: 30976040 DOI: 10.1038/s10038-019-0597-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 03/20/2019] [Accepted: 03/21/2019] [Indexed: 01/20/2023]
Abstract
Disrupted-in-schizophrenia 1 (DISC1) was reported to be associated with schizophrenia. In a previous study, we found significant association with schizophrenia patients with deficient sustained attention assessed by continuous performance test (CPT). This study aimed to identify risk polymorphisms in this specific neurocognitive subgroup and investigate the expression of different isoforms of DISC1. A total of 83 genetic variants were identified through direct sequencing in 50 controls and 100 schizophrenia patients. Fourteen variants were genotyped in 600 controls and 912 patients. Patients were subgrouped by familial loading (multiplex or simplex) and performance on CPT. The frequency of AA genotype of rs11122324 at the 3'-UTR of Es and Esv1 isoforms and of rs2793091 at intron 4 were significantly higher in multiplex schizophrenia patients than those in controls (corrected p < 0.05). In further subgrouping, the frequency of AA genotype of the two SNPs were significantly higher in multiplex schizophrenia patients with deficient sustained attention than those in controls (corrected p < 0.005). The mRNA expression levels of two extra-short isoforms (Es and Esv1) in the EBV-transformed lymphocytes of schizophrenia were significantly higher than those of controls. Luciferase reporter assays demonstrated that the A-allele of rs11122324 significantly upregulated DISC1 extra-short isoforms transcription compared with the G-allele. We found two SNPs (rs11122324 and rs2793091) of DISC1 may be specifically associated with multiplex schizophrenia patients with deficient sustained attention. The SNP rs11122324 may be a risk polymorphism, which may have functional influence on the transcription of Es and Esv1 through increasing their expression.
Collapse
Affiliation(s)
- Chih-Min Liu
- Department of Psychiatry, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan. .,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan.
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Hai-Gwo Hwu
- Department of Psychiatry, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | | | - Ueng-Cheng Yang
- Institute of Bioinformatics, National Yang-Ming University, Taipei, Taiwan
| | - Pei-Chun Hsu
- Institute of Bioinformatics, National Yang-Ming University, Taipei, Taiwan
| | | | - Wei J Chen
- Institute of Epidemiology, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Tzung-Jeng Hwang
- Department of Psychiatry, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan
| | - Ming H Hsieh
- Department of Psychiatry, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chen-Chung Liu
- Department of Psychiatry, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yi-Ling Chien
- Department of Psychiatry, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yi-Tin Lin
- Department of Psychiatry, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ming T Tsuang
- Center for Behavioral Genomics, Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA.,Harvard Departments of Epidemiology and Psychiatry, Harvard Institute of Psychiatric Epidemiology and Genetics, Boston, MA, USA.,Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
25
|
Suh Y, Noh SJ, Lee S, Suh BK, Lee SB, Choi J, Jeong J, Kim S, Park SK. Dopamine D1 Receptor (D1R) Expression Is Controlled by a Transcriptional Repressor Complex Containing DISC1. Mol Neurobiol 2019; 56:6725-6735. [PMID: 30915712 PMCID: PMC6728282 DOI: 10.1007/s12035-019-1566-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 03/13/2019] [Indexed: 11/26/2022]
Abstract
Disrupted-in-Schizophrenia 1 (DISC1) is a scaffold protein implicated in various psychiatric diseases. Dysregulation of the dopamine system has been associated with DISC1 deficiency, while the molecular mechanism is unclear. In this study, we propose a novel molecular mechanism underlying the transcriptional regulation of the dopamine D1 receptor (D1R) in the striatum via DISC1. We verified the increase in D1R at the transcriptional level in the striatum of DISC1-deficient mouse models and altered histone acetylation status at the D1r locus. We identified a functional interaction between DISC1 and Krüppel-like factor 16 (KLF16). KLF16 translocates DISC1 into the nucleus and forms a regulatory complex by recruiting SIN3A corepressor complexes to the D1r locus. Moreover, DISC1-deficient mice have altered D1R-mediated signaling in the striatum and exhibit hyperlocomotion in response to cocaine; the blockade of D1R suppresses these effects. Taken together, our results suggest that nuclear DISC1 plays a critical role in the transcriptional regulation of D1R in the striatal neuron, providing a mechanistic link between DISC1 and dopamine-related psychiatric symptoms.
Collapse
Affiliation(s)
- Yeongjun Suh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Su-Jin Noh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Saebom Lee
- The Russell H. Morgan Department of Radiology and Radiological Sciences, The Johns Hopkins University of School of Medicine, Baltimore, MD, USA
- The Center for Nanomedicine at Wilmer Eye Institute, The Johns Hopkins University of School of Medicine, Baltimore, MD, USA
| | - Bo Kyoung Suh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Su Been Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Jinhyuk Choi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Jaehoon Jeong
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Sangjune Kim
- Neurodegeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University of School of Medicine, Baltimore, MD, USA
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.
| |
Collapse
|
26
|
Kano SI, Choi EY, Dohi E, Agarwal S, Chang DJ, Wilson AM, Lo BD, Rose IVL, Gonzalez S, Imai T, Sawa A. Glutathione S-transferases promote proinflammatory astrocyte-microglia communication during brain inflammation. Sci Signal 2019; 12:12/569/eaar2124. [PMID: 30783009 DOI: 10.1126/scisignal.aar2124] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Astrocytes and microglia play critical roles in brain inflammation. Here, we report that glutathione S-transferases (GSTs), particularly GSTM1, promote proinflammatory signaling in astrocytes and contribute to astrocyte-mediated microglia activation during brain inflammation. In vivo, astrocyte-specific knockdown of GSTM1 in the prefrontal cortex attenuated microglia activation in brain inflammation induced by systemic injection of lipopolysaccharides (LPS). Knocking down GSTM1 in astrocytes also attenuated LPS-induced production of the proinflammatory cytokine tumor necrosis factor-α (TNF-α) by microglia when the two cell types were cocultured. In astrocytes, GSTM1 was required for the activation of nuclear factor κB (NF-κB) and the production of proinflammatory mediators, such as granulocyte-macrophage colony-stimulating factor (GM-CSF) and C-C motif chemokine ligand 2 (CCL2), both of which enhance microglia activation. Our study suggests that GSTs play a proinflammatory role in priming astrocytes and enhancing microglia activation in a microglia-astrocyte positive feedback loop during brain inflammation.
Collapse
Affiliation(s)
- Shin-Ichi Kano
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Eric Y Choi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Eisuke Dohi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Swati Agarwal
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Daniel J Chang
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ashley M Wilson
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Brian D Lo
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Indigo V L Rose
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Santiago Gonzalez
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Takashi Imai
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8510, Japan
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA. .,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
27
|
Barnett BR, Torres-Velázquez M, Yi SY, Rowley PA, Sawin EA, Rubinstein CD, Krentz K, Anderson JM, Bakshi VP, Yu JPJ. Sex-specific deficits in neurite density and white matter integrity are associated with targeted disruption of exon 2 of the Disc1 gene in the rat. Transl Psychiatry 2019; 9:82. [PMID: 30745562 PMCID: PMC6370885 DOI: 10.1038/s41398-019-0429-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 01/24/2019] [Accepted: 01/26/2019] [Indexed: 02/06/2023] Open
Abstract
Diffusion tensor imaging (DTI) has provided remarkable insight into our understanding of white matter microstructure and brain connectivity across a broad spectrum of psychiatric disease. While DTI and other diffusion weighted magnetic resonance imaging (MRI) methods have clarified the axonal contribution to the disconnectivity seen in numerous psychiatric diseases, absent from these studies are quantitative indices of neurite density and orientation that are especially important features in regions of high synaptic density that would capture the synaptic contribution to the psychiatric disease state. Here we report the application of neurite orientation dispersion and density imaging (NODDI), an emerging microstructure imaging technique, to a novel Disc1 svΔ2 rat model of psychiatric illness and demonstrate the complementary and more specific indices of tissue microstructure found in NODDI than those reported by DTI. Our results demonstrate global and sex-specific changes in white matter microstructural integrity and deficits in neurite density as a consequence of the Disc1 svΔ2 genetic variation and highlight the application of NODDI and quantitative measures of neurite density and neurite dispersion in psychiatric disease.
Collapse
Affiliation(s)
- Brian R Barnett
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Maribel Torres-Velázquez
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Sue Y Yi
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Paul A Rowley
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Emily A Sawin
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - C Dustin Rubinstein
- Biotechnology Center, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Kathleen Krentz
- Biotechnology Center, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jacqueline M Anderson
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Vaishali P Bakshi
- Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - John-Paul J Yu
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.
- Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.
| |
Collapse
|
28
|
Kimoto S, Makinodan M, Kishimoto T. Neurobiology and treatment of social cognition in schizophrenia: Bridging the bed-bench gap. Neurobiol Dis 2018; 131:104315. [PMID: 30391541 DOI: 10.1016/j.nbd.2018.10.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 10/01/2018] [Accepted: 10/31/2018] [Indexed: 01/15/2023] Open
Abstract
Social cognition refers to the psychological processes involved in the perception, encoding, storage, retrieval, and regulation of information about others and ourselves. This process is essential for survival and reproduction in complex social environments. Recent evidence suggests that impairments in social cognition frequently occur in schizophrenia, mainly contributing to poor functional outcomes, including the inability to engage in meaningful work and maintain satisfying interpersonal relationships. With the ambiguous definition of social cognition, the neurobiology underlying impaired social cognition remains unknown, and the effectiveness of currently available intervention strategies in schizophrenia remain limited. Considering the advances and challenges of translational research for schizophrenia, social cognition has been considered a high-priority domain for treatment development. Here, we describe the current state of the framework, clinical concerns, and intervention approaches for social cognition in schizophrenia. Next, we introduce translatable rodent models associated with schizophrenia that allow the evaluation of different components of social behaviors, providing deeper insights into the neural substrates of social cognition in schizophrenia. Our review presents a valuable perspective that indicates the necessity of building bridges between basic and clinical science researchers for the development of novel therapeutic approaches in impaired social cognition in schizophrenia.
Collapse
Affiliation(s)
- Sohei Kimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Japan.
| | - Manabu Makinodan
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Japan
| | - Toshifumi Kishimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Japan
| |
Collapse
|
29
|
Neuronal serine racemase associates with Disrupted-In-Schizophrenia-1 and DISC1 agglomerates: Implications for schizophrenia. Neurosci Lett 2018; 692:107-114. [PMID: 30391323 DOI: 10.1016/j.neulet.2018.10.055] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/18/2018] [Accepted: 10/29/2018] [Indexed: 12/21/2022]
Abstract
D-Serine, an endogenous coagonist of N-methyl-d-aspartate receptors (NMDARs) at the glycine binding site, is synthesized by serine racemase (SR) through conversion of l-Serine. Dysregulation of SR/D-Serine and Disrupted-In-Schizophrenia-1 (DISC1) contributes to the pathogenesis of schizophrenia at converging pathways, as perturbation of SR-DISC1 binding in astrocytes elicits schizophrenia-like behaviors in mice. However, an association of neuronal SR with DISC1 remains elusive. Here we report that SR associates with DISC1 and its agglomerates in cortical neurons, which can be modulated by NMDAR activity. Endogenous SR colocalizes with DISC1 large agglomerates in the soma and with smaller puncta in the nucleus and dendrites of cortical neurons. Co-immunoprecipitation assays demonstrate SR interaction with DISC1 in cortical neuronal lysates, suggesting the physiological presence of functional SR-DISC1 complexes in neurons. Moreover, exogenous d-Serine application significantly increases the interaction of SR with DISC1, the number of DISC1-SR large agglomerates and the levels of DISC1 agglomerated form along with SR in the triton-insoluble pellet fraction, whereas application of glycine with a glycine transporter inhibitor fails to increase their interactions, abundance of DISC1-SR large agglomerates and levels of DISC1 agglomerated form. This increase by d-Serine application is blocked by 7-chlorokynurenic acid, a specific antagonist at the glycine site of NMDARs, suggesting mediation through NMDARs. Our findings thus demonstrate neuronal SR association with DISC1 and its agglomerates, which can be modulated by d-Serine, thereby validating a novel neuronal SR-DISC1 complex responsive to NMDAR activation and providing a molecular mechanism by which pathways implicated in schizophrenia converge.
Collapse
|
30
|
Murphy E, Benítez-Burraco A. Toward the Language Oscillogenome. Front Psychol 2018; 9:1999. [PMID: 30405489 PMCID: PMC6206218 DOI: 10.3389/fpsyg.2018.01999] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 09/28/2018] [Indexed: 12/20/2022] Open
Abstract
Language has been argued to arise, both ontogenetically and phylogenetically, from specific patterns of brain wiring. We argue that it can further be shown that core features of language processing emerge from particular phasal and cross-frequency coupling properties of neural oscillations; what has been referred to as the language ‘oscillome.’ It is expected that basic aspects of the language oscillome result from genetic guidance, what we will here call the language ‘oscillogenome,’ for which we will put forward a list of candidate genes. We have considered genes for altered brain rhythmicity in conditions involving language deficits: autism spectrum disorders, schizophrenia, specific language impairment and dyslexia. These selected genes map on to aspects of brain function, particularly on to neurotransmitter function. We stress that caution should be adopted in the construction of any oscillogenome, given the range of potential roles particular localized frequency bands have in cognition. Our aim is to propose a set of genome-to-language linking hypotheses that, given testing, would grant explanatory power to brain rhythms with respect to language processing and evolution.
Collapse
Affiliation(s)
- Elliot Murphy
- Division of Psychology and Language Sciences, University College London, London, United Kingdom.,Department of Psychology, University of Westminster, London, United Kingdom
| | - Antonio Benítez-Burraco
- Department of Spanish Language, Linguistics and Literary Theory, University of Seville, Seville, Spain
| |
Collapse
|
31
|
Endo R, Takashima N, Nekooki-Machida Y, Komi Y, Hui KKW, Takao M, Akatsu H, Murayama S, Sawa A, Tanaka M. TAR DNA-Binding Protein 43 and Disrupted in Schizophrenia 1 Coaggregation Disrupts Dendritic Local Translation and Mental Function in Frontotemporal Lobar Degeneration. Biol Psychiatry 2018; 84:509-521. [PMID: 29752072 PMCID: PMC6123275 DOI: 10.1016/j.biopsych.2018.03.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 02/06/2018] [Accepted: 03/07/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Neurodegenerative diseases involving protein aggregation often accompany psychiatric symptoms. Frontotemporal lobar degeneration (FTLD) associated with TAR DNA-binding protein 43 (TDP-43) aggregation is characterized by progressive neuronal atrophy in frontal and temporal lobes of cerebral cortex. Furthermore, patients with FTLD display mental dysfunction in multiple behavioral dimensions. Nevertheless, their molecular origin for psychiatric symptoms remains unclear. METHODS In FTLD neurons and mouse models with TDP-43 aggregates, we examined coaggregation between TDP-43 and disrupted in schizophrenia 1 (DISC1), a key player in the pathology of mental conditions and its effects on local translation in dendrites and psychiatric behaviors. The protein coaggregation and the expression level of synaptic proteins were also investigated with postmortem brains from patients with FTLD (n = 6). RESULTS We found cytosolic TDP-43/DISC1 coaggregates in brains of both FTLD mouse model and patients with FTLD. At the mechanistic levels, the TDP-43/DISC1 coaggregates disrupted the activity-dependent dendritic local translation through impairment of translation initiation and, in turn, reduced synaptic protein expression. Behavioral deficits detected in FTLD model mice were ameliorated by exogenous DISC1 expression. CONCLUSIONS Our findings reveal a novel role of the aggregate-prone TDP-43/DISC1 protein complex in regulating local translation, which affects aberrant behaviors relevant to multiple psychiatric dimensions.
Collapse
Affiliation(s)
- Ryo Endo
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Japan
| | - Noriko Takashima
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Japan
| | - Yoko Nekooki-Machida
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Japan
| | - Yusuke Komi
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Japan
| | - Kelvin Kai-Wan Hui
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Japan
| | - Masaki Takao
- Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Japan,Department of Neurology, Saitama Medical University, Japan
| | - Hiroyasu Akatsu
- Choju Medical Institute, Fukushimura Hospital, Japan,Department of Medicine for Aging in Place and Community-Based Medical Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Shigeo Murayama
- Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Japan
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD.
| | - Motomasa Tanaka
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Wako, Japan.
| |
Collapse
|
32
|
Malavasi ELV, Economides KD, Grünewald E, Makedonopoulou P, Gautier P, Mackie S, Murphy LC, Murdoch H, Crummie D, Ogawa F, McCartney DL, O'Sullivan ST, Burr K, Torrance HS, Phillips J, Bonneau M, Anderson SM, Perry P, Pearson M, Constantinides C, Davidson-Smith H, Kabiri M, Duff B, Johnstone M, Polites HG, Lawrie SM, Blackwood DH, Semple CA, Evans KL, Didier M, Chandran S, McIntosh AM, Price DJ, Houslay MD, Porteous DJ, Millar JK. DISC1 regulates N-methyl-D-aspartate receptor dynamics: abnormalities induced by a Disc1 mutation modelling a translocation linked to major mental illness. Transl Psychiatry 2018; 8:184. [PMID: 30190480 PMCID: PMC6127284 DOI: 10.1038/s41398-018-0228-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 07/16/2018] [Indexed: 01/01/2023] Open
Abstract
The neuromodulatory gene DISC1 is disrupted by a t(1;11) translocation that is highly penetrant for schizophrenia and affective disorders, but how this translocation affects DISC1 function is incompletely understood. N-methyl-D-aspartate receptors (NMDAR) play a central role in synaptic plasticity and cognition, and are implicated in the pathophysiology of schizophrenia through genetic and functional studies. We show that the NMDAR subunit GluN2B complexes with DISC1-associated trafficking factor TRAK1, while DISC1 interacts with the GluN1 subunit and regulates dendritic NMDAR motility in cultured mouse neurons. Moreover, in the first mutant mouse that models DISC1 disruption by the translocation, the pool of NMDAR transport vesicles and surface/synaptic NMDAR expression are increased. Since NMDAR cell surface/synaptic expression is tightly regulated to ensure correct function, these changes in the mutant mouse are likely to affect NMDAR signalling and synaptic plasticity. Consistent with these observations, RNASeq analysis of the translocation carrier-derived human neurons indicates abnormalities of excitatory synapses and vesicle dynamics. RNASeq analysis of the human neurons also identifies many differentially expressed genes previously highlighted as putative schizophrenia and/or depression risk factors through large-scale genome-wide association and copy number variant studies, indicating that the translocation triggers common disease pathways that are shared with unrelated psychiatric patients. Altogether, our findings suggest that translocation-induced disease mechanisms are likely to be relevant to mental illness in general, and that such disease mechanisms include altered NMDAR dynamics and excitatory synapse function. This could contribute to the cognitive disorders displayed by translocation carriers.
Collapse
Affiliation(s)
- Elise L V Malavasi
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | | | - Ellen Grünewald
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Paraskevi Makedonopoulou
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Philippe Gautier
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Shaun Mackie
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Laura C Murphy
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Hannah Murdoch
- Molecular Pharmacology Group, Wolfson Building, Institute of Neuroscience and Psychology, The University of Glasgow, University Avenue, Glasgow, UK
| | - Darragh Crummie
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Fumiaki Ogawa
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Daniel L McCartney
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Shane T O'Sullivan
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Karen Burr
- Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, UK
| | - Helen S Torrance
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Jonathan Phillips
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Marion Bonneau
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Susan M Anderson
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Paul Perry
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Matthew Pearson
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Costas Constantinides
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Hazel Davidson-Smith
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Mostafa Kabiri
- Translational In Vivo Models at Sanofi, Frankfurt, Germany
| | - Barbara Duff
- Division of Psychiatry, The University of Edinburgh, Edinburgh, UK
| | - Mandy Johnstone
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
- Division of Psychiatry, The University of Edinburgh, Edinburgh, UK
| | | | - Stephen M Lawrie
- Division of Psychiatry, The University of Edinburgh, Edinburgh, UK
| | | | - Colin A Semple
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Kathryn L Evans
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Michel Didier
- Translational Sciences at Sanofi, Chilly-Mazarin, France
| | | | | | - David J Price
- Centre for Integrative Physiology, The University of Edinburgh, Edinburgh, UK
| | - Miles D Houslay
- School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - J Kirsty Millar
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
33
|
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: 4.5] [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
|
34
|
Wulaer B, Nagai T, Sobue A, Itoh N, Kuroda K, Kaibuchi K, Nabeshima T, Yamada K. Repetitive and compulsive-like behaviors lead to cognitive dysfunction in Disc1Δ2-3/Δ2-3mice. GENES BRAIN AND BEHAVIOR 2018; 17:e12478. [DOI: 10.1111/gbb.12478] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/03/2018] [Accepted: 04/03/2018] [Indexed: 01/19/2023]
Affiliation(s)
- B. Wulaer
- Department of Neuropsychopharmacology and Hospital Pharmacy; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - T. Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - A. Sobue
- Department of Neuropsychopharmacology and Hospital Pharmacy; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - N. Itoh
- Department of Neuropsychopharmacology and Hospital Pharmacy; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - K. Kuroda
- Department of Cell Pharmacology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - K. Kaibuchi
- Department of Cell Pharmacology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - T. Nabeshima
- Advanced Diagnostic System Research Laboratory; Fujita Health University, Graduate School of Health Sciences; Toyoake Japan
- Aino University; Ibaragi Japan
| | - K. Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy; Nagoya University Graduate School of Medicine; Nagoya Japan
| |
Collapse
|
35
|
Monday HR, Younts TJ, Castillo PE. Long-Term Plasticity of Neurotransmitter Release: Emerging Mechanisms and Contributions to Brain Function and Disease. Annu Rev Neurosci 2018; 41:299-322. [PMID: 29709205 DOI: 10.1146/annurev-neuro-080317-062155] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Long-lasting changes of brain function in response to experience rely on diverse forms of activity-dependent synaptic plasticity. Chief among them are long-term potentiation and long-term depression of neurotransmitter release, which are widely expressed by excitatory and inhibitory synapses throughout the central nervous system and can dynamically regulate information flow in neural circuits. This review article explores recent advances in presynaptic long-term plasticity mechanisms and contributions to circuit function. Growing evidence indicates that presynaptic plasticity may involve structural changes, presynaptic protein synthesis, and transsynaptic signaling. Presynaptic long-term plasticity can alter the short-term dynamics of neurotransmitter release, thereby contributing to circuit computations such as novelty detection, modifications of the excitatory/inhibitory balance, and sensory adaptation. In addition, presynaptic long-term plasticity underlies forms of learning and its dysregulation participates in several neuropsychiatric conditions, including schizophrenia, autism, intellectual disabilities, neurodegenerative diseases, and drug abuse.
Collapse
Affiliation(s)
- Hannah R Monday
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461, USA;
| | - Thomas J Younts
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461, USA;
| |
Collapse
|
36
|
Selten M, van Bokhoven H, Nadif Kasri N. Inhibitory control of the excitatory/inhibitory balance in psychiatric disorders. F1000Res 2018; 7:23. [PMID: 29375819 PMCID: PMC5760969 DOI: 10.12688/f1000research.12155.1] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/21/2017] [Indexed: 12/21/2022] Open
Abstract
Neuronal networks consist of different types of neurons that all play their own role in order to maintain proper network function. The two main types of neurons segregate in excitatory and inhibitory neurons, which together regulate the flow of information through the network. It has been proposed that changes in the relative strength in these two opposing forces underlie the symptoms observed in psychiatric disorders, including autism and schizophrenia. Here, we review the role of alterations to the function of the inhibitory system as a cause of psychiatric disorders. First, we explore both patient and post-mortem evidence of inhibitory deficiency. We then discuss the function of different interneuron subtypes in the network and focus on the central role of a specific class of inhibitory neurons, parvalbumin-positive interneurons. Finally, we discuss genes known to be affected in different disorders and the effects that mutations in these genes have on the inhibitory system in cortex and hippocampus. We conclude that alterations to the inhibitory system are consistently identified in animal models of psychiatric disorders and, more specifically, that mutations affecting the function of parvalbumin-positive interneurons seem to play a central role in the symptoms observed in these disorders.
Collapse
Affiliation(s)
- Martijn Selten
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, New Hunt's House, Guy's Campus, King's College London, London, SE1 1UL, UK.,MRC Centre for Neurodevelopmental Disorders, New Hunt's House, Guy's Campus, King's College London, London, SE1 1UL, UK.,Department of Human Genetics & Department of Cognitive Neuroscience, Radboudumc, Geert Grooteplein 10, Box 9101, 6500 HB Nijmegen, Netherlands.,Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ Nijmegen, Netherlands
| | - Hans van Bokhoven
- Department of Human Genetics & Department of Cognitive Neuroscience, Radboudumc, Geert Grooteplein 10, Box 9101, 6500 HB Nijmegen, Netherlands.,Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ Nijmegen, Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics & Department of Cognitive Neuroscience, Radboudumc, Geert Grooteplein 10, Box 9101, 6500 HB Nijmegen, Netherlands.,Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ Nijmegen, Netherlands
| |
Collapse
|
37
|
Skirzewski M, Karavanova I, Shamir A, Erben L, Garcia-Olivares J, Shin JH, Vullhorst D, Alvarez VA, Amara SG, Buonanno A. ErbB4 signaling in dopaminergic axonal projections increases extracellular dopamine levels and regulates spatial/working memory behaviors. Mol Psychiatry 2018; 23:2227-2237. [PMID: 28727685 PMCID: PMC5775946 DOI: 10.1038/mp.2017.132] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 03/13/2017] [Accepted: 04/04/2017] [Indexed: 02/07/2023]
Abstract
Genetic variants of Neuregulin 1 (NRG1) and its neuronal tyrosine kinase receptor ErbB4 are associated with risk for schizophrenia, a neurodevelopmental disorder characterized by excitatory/inhibitory imbalance and dopamine (DA) dysfunction. To date, most ErbB4 studies have focused on GABAergic interneurons in the hippocampus and neocortex, particularly fast-spiking parvalbumin-positive (PV+) basket cells. However, NRG has also been shown to modulate DA levels, suggesting a role for ErbB4 signaling in dopaminergic neuron function. Here we report that ErbB4 in midbrain DAergic axonal projections regulates extracellular DA levels and relevant behaviors. Mice lacking ErbB4 in tyrosine hydroxylase-positive (TH+) neurons, but not in PV+ GABAergic interneurons, exhibit different regional imbalances of basal DA levels and fail to increase DA in response to local NRG1 infusion into the dorsal hippocampus, medial prefrontal cortex and dorsal striatum measured by reverse microdialysis. Using Lund Human Mesencephalic (LUHMES) cells, we show that NRG/ErbB signaling increases extracellular DA levels, at least in part, by reducing DA transporter (DAT)-dependent uptake. Interestingly, TH-Cre;ErbB4f/f mice manifest deficits in learning, spatial and working memory-related behaviors, but not in numerous other behaviors altered in PV-Cre;ErbB4f/f mice. Importantly, microinjection of a Cre-inducible ErbB4 virus (AAV-ErbB4.DIO) into the mesencephalon of TH-Cre;ErbB4f/f mice, which selectively restores ErbB4 expression in DAergic neurons, rescues DA dysfunction and ameliorates behavioral deficits. Our results indicate that direct NRG/ErbB4 signaling in DAergic axonal projections modulates DA homeostasis, and that NRG/ErbB4 signaling in both GABAergic interneurons and DA neurons contribute to the modulation of behaviors relevant to psychiatric disorders.
Collapse
Affiliation(s)
- M Skirzewski
- 0000 0001 2297 5165grid.94365.3dSection on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - I Karavanova
- 0000 0001 2297 5165grid.94365.3dSection on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - A Shamir
- 0000 0001 2297 5165grid.94365.3dSection on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - L Erben
- 0000 0001 2297 5165grid.94365.3dSection on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA ,0000 0001 2240 3300grid.10388.32Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | - J Garcia-Olivares
- 0000 0001 2297 5165grid.94365.3dLaboratory of Molecular and Cellular Neurobiology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD USA
| | - J H Shin
- 0000 0001 2297 5165grid.94365.3dLaboratory for Integrative Neuroscience, Section on Neuronal Structure, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD USA
| | - D Vullhorst
- 0000 0001 2297 5165grid.94365.3dSection on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - V A Alvarez
- 0000 0001 2297 5165grid.94365.3dLaboratory for Integrative Neuroscience, Section on Neuronal Structure, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD USA
| | - S G Amara
- 0000 0001 2297 5165grid.94365.3dLaboratory of Molecular and Cellular Neurobiology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD USA
| | - A Buonanno
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
38
|
DISC1 Modulates Neuronal Stress Responses by Gate-Keeping ER-Mitochondria Ca 2+ Transfer through the MAM. Cell Rep 2017; 21:2748-2759. [DOI: 10.1016/j.celrep.2017.11.043] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/21/2017] [Accepted: 11/12/2017] [Indexed: 12/21/2022] Open
|
39
|
Matsumoto Y, Niwa M, Mouri A, Noda Y, Fukushima T, Ozaki N, Nabeshima T. Adolescent stress leads to glutamatergic disturbance through dopaminergic abnormalities in the prefrontal cortex of genetically vulnerable mice. Psychopharmacology (Berl) 2017; 234:3055-3074. [PMID: 28756461 PMCID: PMC8034555 DOI: 10.1007/s00213-017-4704-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 07/04/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Stress during the adolescent period influences postnatal maturation and behavioral patterns in adulthood. Adolescent stress-induced molecular and functional changes in neurons are the key clinical features of psychiatric disorders including schizophrenia. OBJECTIVE In the present study, we exposed genetically vulnerable mice to isolation stress to examine the molecular changes in the glutamatergic system involving N-methyl-d-aspartate (NMDA) receptors via dopaminergic disturbance in the prefrontal cortex (PFc). RESULTS We report that late adolescent stress in combination with Disrupted-in-Schizophrenia 1 (DISC1) genetic risk elicited alterations in glutamatergic neurons in the PFc, such as increased expression of glutamate transporters, decreased extracellular levels of glutamate, decreased concentration of d-serine, and impaired activation of NMDA-Ca2+/calmodulin kinase II signaling. These changes resulted in behavioral deficits in locomotor activity, forced swim, social interaction, and novelty preference tests. The glutamatergic alterations in the PFc were prevented if the animals were treated with an atypical antipsychotic drug clozapine and a dopamine D1 agonist SKF81297, which suggests that the activation of dopaminergic neurons is involved in the regulation of the glutamatergic system. CONCLUSION Our results suggest that adolescent stress combined with dopaminergic abnormalities in the PFc of genetically vulnerable mice induces glutamatergic disturbances, which leads to behavioral deficits in the young adult stage.
Collapse
Affiliation(s)
- Yurie Matsumoto
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University, Nagoya, 468-8503, Japan
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, 466-8560, Japan
| | - Minae Niwa
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University, Nagoya, 468-8503, Japan.
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, 466-8560, Japan.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Akihiro Mouri
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University, Nagoya, 468-8503, Japan
- Advanced Diagnostic System Research Laboratory, Fujita Health University, Graduate School of Health Science, Toyoake, 470-1192, Japan
- NPO Japanese Drug Organization of Appropriate Use and Research, Nagoya, 468-0069, Japan
| | - Yukihiro Noda
- NPO Japanese Drug Organization of Appropriate Use and Research, Nagoya, 468-0069, Japan
- Division of Clinical Sciences and Neuropsychopharmacology, Graduate School of Pharmacy, Meijo University, Nagoya, 468-8503, Japan
| | - Takeshi Fukushima
- Department of Analytical Chemistry, Faculty of Pharmaceutical Science, Toho University, Chiba, 274-8510, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, 466-8560, Japan
| | - Toshitaka Nabeshima
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University, Nagoya, 468-8503, Japan.
- Advanced Diagnostic System Research Laboratory, Fujita Health University, Graduate School of Health Science, Toyoake, 470-1192, Japan.
- NPO Japanese Drug Organization of Appropriate Use and Research, Nagoya, 468-0069, Japan.
- Aino University, Ibaragi, Osaka, 567-0012, Japan.
| |
Collapse
|
40
|
Tomoda T, Hikida T, Sakurai T. Role of DISC1 in Neuronal Trafficking and its Implication in Neuropsychiatric Manifestation and Neurotherapeutics. Neurotherapeutics 2017; 14:623-629. [PMID: 28664299 PMCID: PMC5509643 DOI: 10.1007/s13311-017-0556-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Disrupted-in-schizophrenia 1 (DISC1) was initially identified as a gene disrupted by a translocation mutation co-segregating with a variety of psychotic and mood disorders in a Scottish pedigree. In agreement with this original finding, mouse models that perturb Disc1 display deficits of behaviors in specific dimensions, such as cognition and emotion, but not a motor dimension. Although DISC1 is not a risk gene for sporadic cases of specific psychiatric disorders defined by categorical diagnostic criteria (e.g., schizophrenia and major depressive disorder), DISC1 is now regarded as an important molecular lead to decipher molecular pathology for specific dimensions relevant to major mental illnesses. Emerging evidence points to the role of DISC1 in the regulation of intracellular trafficking of a wide range of neuronal cargoes. We will review recent progress in this aspect of DISC1 biology and discuss how we could utilize this body of knowledge to better understand the pathophysiology of mental illnesses.
Collapse
Affiliation(s)
- Toshifumi Tomoda
- Centre for Addiction and Mental Health, University of Toronto, Toronto, Canada.
| | - Takatoshi Hikida
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Takeshi Sakurai
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
41
|
Erbb4 Deletion from Medium Spiny Neurons of the Nucleus Accumbens Core Induces Schizophrenia-Like Behaviors via Elevated GABA A Receptor α1 Subunit Expression. J Neurosci 2017; 37:7450-7464. [PMID: 28667174 DOI: 10.1523/jneurosci.3948-16.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 06/03/2017] [Accepted: 06/19/2017] [Indexed: 12/21/2022] Open
Abstract
Medium spiny neurons (MSNs), the major GABAergic projection neurons in the striatum, are implicated in many neuropsychiatric diseases such as schizophrenia, but the underlying mechanisms remain unclear. We found that a deficiency in Erbb4, a schizophrenia risk gene, in MSNs of the nucleus accumbens (NAc) core, but not the dorsomedial striatum, markedly induced schizophrenia-like behaviors such as hyperactivity, abnormal marble-burying behavior, damaged social novelty recognition, and impaired sensorimotor gating function in male mice. Using immunohistochemistry, Western blot, RNA interference, electrophysiology, and behavior test studies, we found that these phenomena were mediated by increased GABAA receptor α1 subunit (GABAAR α1) expression, which enhanced inhibitory synaptic transmission on MSNs. These results suggest that Erbb4 in MSNs of the NAc core may contribute to the pathogenesis of schizophrenia by regulating GABAergic transmission and raise the possibility that GABAAR α1 may therefore serve as a new therapeutic target for schizophrenia.SIGNIFICANCE STATEMENT Although ErbB4 is highly expressed in striatal medium spiny neurons (MSNs), its role in this type of neuron has not been reported previously. The present study demonstrates that Erbb4 deletion in nucleus accumbens (NAc) core MSNs can induce schizophrenia-like behaviors via elevated GABAA receptor α1 subunit (GABAAR α1) expression. To our knowledge, this is the first evidence that ErbB4 signaling in the MSNs is involved in the pathology of schizophrenia. Furthermore, restoration of GABAAR α1 in the NAc core, but not the dorsal medium striatum, alleviated the abnormal behaviors. Here, we highlight the role of the NAc core in the pathogenesis of schizophrenia and suggest that GABAAR α1 may be a potential pharmacological target for its treatment.
Collapse
|
42
|
Tanaka M, Ishizuka K, Nekooki-Machida Y, Endo R, Takashima N, Sasaki H, Komi Y, Gathercole A, Huston E, Ishii K, Hui KKW, Kurosawa M, Kim SH, Nukina N, Takimoto E, Houslay MD, Sawa A. Aggregation of scaffolding protein DISC1 dysregulates phosphodiesterase 4 in Huntington's disease. J Clin Invest 2017; 127:1438-1450. [PMID: 28263187 DOI: 10.1172/jci85594] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 01/11/2017] [Indexed: 01/19/2023] Open
Abstract
Huntington's disease (HD) is a polyglutamine (polyQ) disease caused by aberrant expansion of the polyQ tract in Huntingtin (HTT). While motor impairment mediated by polyQ-expanded HTT has been intensively studied, molecular mechanisms for nonmotor symptoms in HD, such as psychiatric manifestations, remain elusive. Here we have demonstrated that HTT forms a ternary protein complex with the scaffolding protein DISC1 and cAMP-degrading phosphodiesterase 4 (PDE4) to regulate PDE4 activity. We observed pathological cross-seeding between DISC1 and mutant HTT aggregates in the brains of HD patients as well as in a murine model that recapitulates the polyQ pathology of HD (R6/2 mice). In R6/2 mice, consequent reductions in soluble DISC1 led to dysregulation of DISC1-PDE4 complexes, aberrantly increasing the activity of PDE4. Importantly, exogenous expression of a modified DISC1, which binds to PDE4 but not mutant HTT, normalized PDE4 activity and ameliorated anhedonia in the R6/2 mice. We propose that cross-seeding of mutant HTT and DISC1 and the resultant changes in PDE4 activity may underlie the pathology of a specific subset of mental manifestations of HD, which may provide an insight into molecular signaling in mental illness in general.
Collapse
|
43
|
Zhang Q, Esrafilzadeh D, Crook JM, Kapsa R, Stewart EM, Tomaskovic-Crook E, Wallace GG, Huang XF. Electrical Stimulation Using Conductive Polymer Polypyrrole Counters Reduced Neurite Outgrowth of Primary Prefrontal Cortical Neurons from NRG1-KO and DISC1-LI Mice. Sci Rep 2017; 7:42525. [PMID: 28198409 PMCID: PMC5309772 DOI: 10.1038/srep42525] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/11/2017] [Indexed: 01/07/2023] Open
Abstract
Deficits in neurite outgrowth, possibly involving dysregulation of risk genes neuregulin-1 (NRG1) and disrupted in schizophrenia 1 (DISC1) have been implicated in psychiatric disorders including schizophrenia. Electrical stimulation using conductive polymers has been shown to stimulate neurite outgrowth of differentiating human neural stem cells. This study investigated the use of the electroactive conductive polymer polypyrrole (Ppy) to counter impaired neurite outgrowth of primary pre-frontal cortical (PFC) neurons from NRG1-knock out (NRG1-KO) and DISC1-locus impairment (DISC1-LI) mice. Whereas NRG1-KO and DISC1-LI exhibited reduced neurite length and number of neurite branches compared to wild-type controls, this was not apparent for cultures on electroactive Ppy. Additionally, the use of the Ppy substrate normalised the synaptophysin and PSD95 protein and mRNA expression whereas both are usually reduced by NRG1-KO or DISC1-LI. Our findings support the utility of Ppy mediated electrical stimulation to prevent the reduction of neurite outgrowth and related synaptic protein expression in the primary PFC neurons from NRG1-KO and DISC1-LI mice, providing proof-of-concept for treating neurodevelopmental diseases including schizophrenia.
Collapse
Affiliation(s)
- Qingsheng Zhang
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia.,ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Fairy Meadow, NSW 2519, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Dorna Esrafilzadeh
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Fairy Meadow, NSW 2519, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Jeremy M Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Fairy Meadow, NSW 2519, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.,Departments of Surgery, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Robert Kapsa
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Fairy Meadow, NSW 2519, Australia.,Departments of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Elise M Stewart
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Fairy Meadow, NSW 2519, Australia
| | - Eva Tomaskovic-Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Fairy Meadow, NSW 2519, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Gordon G Wallace
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Fairy Meadow, NSW 2519, Australia
| | - Xu-Feng Huang
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.,Schizophrenia Research Institute, 384 Victoria Street, Darlinghurst, NSW 2010, Australia
| |
Collapse
|
44
|
Dahoun T, Trossbach SV, Brandon NJ, Korth C, Howes OD. The impact of Disrupted-in-Schizophrenia 1 (DISC1) on the dopaminergic system: a systematic review. Transl Psychiatry 2017; 7:e1015. [PMID: 28140405 PMCID: PMC5299392 DOI: 10.1038/tp.2016.282] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/16/2016] [Accepted: 11/27/2016] [Indexed: 12/21/2022] Open
Abstract
Disrupted-in-Schizophrenia 1 (DISC1) is a gene known as a risk factor for mental illnesses possibly associated with dopamine impairments. DISC1 is a scaffold protein interacting with proteins involved in the dopamine system. Here we summarise the impact of DISC1 disruption on the dopamine system in animal models, considering its effects on presynaptic dopaminergic function (tyrosine hydroxylase levels, dopamine transporter levels, dopamine levels at baseline and after amphetamine administration) and postsynaptic dopaminergic function (dopamine D1 and D2 receptor levels, dopamine receptor-binding potential and locomotor activity after amphetamine administration). Our findings show that many but not all DISC1 models display (1) increased locomotion after amphetamine administration, (2) increased dopamine levels after amphetamine administration in the nucleus accumbens, and (3) inconsistent basal dopamine levels, dopamine receptor levels and binding potentials. There is also limited evidence for decreased tyrosine hydroxylase levels in the frontal cortex and increased dopamine transporter levels in the striatum but not nucleus accumbens, but these conclusions warrant further replication. The main dopaminergic findings are seen across different DISC1 models, providing convergent evidence that DISC1 has a role in regulating dopaminergic function. These results implicate dopaminergic dysregulation as a mechanism underlying the increased rate of schizophrenia seen in DISC1 variant carriers, and provide insights into how DISC1, and potentially DISC1-interacting proteins such as AKT and GSK-3, could be used as novel therapeutic targets for schizophrenia.
Collapse
Affiliation(s)
- T Dahoun
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London, UK
- Department of the Institute of Clinical Sciences, Psychiatric Imaging Group, MRC London Institute of Medical Sciences (LMS), Imperial College-Hammersmith Hospital Campus, London, UK
| | - S V Trossbach
- Department of Neuropathology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - N J Brandon
- AstraZeneca Neuroscience, Innovative Medicines and Early Development Biotech Unit, R&D Boston, Waltham, MA, USA
| | - C Korth
- Department of Neuropathology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - O D Howes
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London, UK
- Department of the Institute of Clinical Sciences, Psychiatric Imaging Group, MRC London Institute of Medical Sciences (LMS), Imperial College-Hammersmith Hospital Campus, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London, UK
| |
Collapse
|
45
|
Developmental pruning of excitatory synaptic inputs to parvalbumin interneurons in monkey prefrontal cortex. Proc Natl Acad Sci U S A 2017; 114:E629-E637. [PMID: 28074037 DOI: 10.1073/pnas.1610077114] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Working memory requires efficient excitatory drive to parvalbumin-positive (PV) interneurons in the primate dorsolateral prefrontal cortex (DLPFC). Developmental pruning eliminates superfluous excitatory inputs, suggesting that working memory maturation during adolescence requires pruning of excitatory inputs to PV interneurons. Therefore, we tested the hypothesis that excitatory synapses on PV interneurons are pruned during adolescence. The density of excitatory synapses, defined by overlapping vesicular glutamate transporter 1-positive (VGlut1+) and postsynaptic density 95-positive (PSD95+) puncta, on PV interneurons was lower in postpubertal relative to prepubertal monkeys. In contrast, puncta levels of VGlut1 and PSD95 proteins were higher in postpubertal monkeys and positively predicted activity-dependent PV levels, suggesting a greater strength of the remaining synapses after pruning. Because excitatory synapse number on PV interneurons is regulated by erb-b2 receptor tyrosine kinase 4 (ErbB4), whose function is influenced by alternative splicing, we tested the hypothesis that pruning of excitatory synapses on PV interneurons is associated with developmental shifts in ErbB4 expression and/or splicing. Pan-ErbB4 expression did not change, whereas the minor-to-major splice variant ratios increased with age. In cell culture, the major, but not the minor, variant increased excitatory synapse number on PV interneurons and displayed greater kinase activity than the minor variant, suggesting that the effect of ErbB4 signaling in PV interneurons is mediated by alternative splicing. Supporting this interpretation, in monkey DLPFC, higher minor-to-major variant ratios predicted lower PSD95+ puncta density on PV interneurons. Together, our findings suggest that ErbB4 splicing may regulate the pruning of excitatory synapses on PV interneurons during adolescence.
Collapse
|
46
|
O’Tuathaigh CMP, Fumagalli F, Desbonnet L, Perez-Branguli F, Moloney G, Loftus S, O’Leary C, Petit E, Cox R, Tighe O, Clarke G, Lai D, Harvey RP, Cryan JF, Mitchell KJ, Dinan TG, Riva MA, Waddington JL. Epistatic and Independent Effects on Schizophrenia-Related Phenotypes Following Co-disruption of the Risk Factors Neuregulin-1 × DISC1. Schizophr Bull 2017; 43:214-225. [PMID: 27613806 PMCID: PMC5216856 DOI: 10.1093/schbul/sbw120] [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] [Indexed: 01/03/2023]
Abstract
Few studies have addressed likely gene × gene (ie, epistatic) interactions in mediating risk for schizophrenia. Using a preclinical genetic approach, we investigated whether simultaneous disruption of the risk factors Neuregulin-1 (NRG1) and Disrupted-in-schizophrenia 1 (DISC1) would produce a disease-relevant phenotypic profile different from that observed following disruption to either gene alone. NRG1 heterozygotes exhibited hyperactivity and disruption to prepulse inhibition, both reversed by antipsychotic treatment, and accompanied by reduced striatal dopamine D2 receptor protein expression, impaired social cognition, and altered glutamatergic synaptic protein expression in selected brain areas. Single gene DISC1 mutants demonstrated a disruption in social cognition and nest-building, altered brain 5-hydroxytryptamine levels and hippocampal ErbB4 expression, and decreased cortical expression of the schizophrenia-associated microRNA miR-29b. Co-disruption of DISC1 and NRG1, indicative of epistasis, evoked an impairment in sociability and enhanced self-grooming, accompanied by changes in hypothalamic oxytocin/vasopressin gene expression. The findings indicate specific behavioral correlates and underlying cellular pathways downstream of main effects of DNA variation in the schizophrenia-associated genes NRG1 and DISC1.
Collapse
Affiliation(s)
- Colm M. P. O’Tuathaigh
- School of Medicine, Brookfield Health Sciences Complex, University College Cork, Cork, Ireland;,Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2,Ireland;,*To whom correspondence should be addressed; School of Medicine, Brookfield Health Sciences Complex, University College Cork, Cork T12 YN60, Ireland; tel: +353-(0)21-420-5303, fax: +353-(0)21-490-1594, e-mail:
| | - Fabio Fumagalli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Universita` degli Studi di Milano, Milan,
Italy
| | - Lieve Desbonnet
- Neurogastroenterology Laboratory, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland
| | - Francesc Perez-Branguli
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland;,IZKF Junior Research Group and BMBF Research Group Neuroscience, IZKF, Friedrich-Alexander-Universitaet Erlangen-Nuernberg, Erlangen, Germany
| | - Gerard Moloney
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Samim Loftus
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Claire O’Leary
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2,Ireland
| | - Emilie Petit
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2,Ireland
| | - Rachel Cox
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2,Ireland
| | - Orna Tighe
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2,Ireland
| | - Gerard Clarke
- Neurogastroenterology Laboratory, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland;,Department of Psychiatry, University College Cork, Cork, Ireland
| | - Donna Lai
- Victor Chang Cardiac Research Institute, Sydney, Australia
| | | | - John F. Cryan
- Neurogastroenterology Laboratory, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland;,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Kevin J. Mitchell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Timothy G. Dinan
- Neurogastroenterology Laboratory, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland;,Department of Psychiatry, University College Cork, Cork, Ireland
| | - Marco A. Riva
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Universita` degli Studi di Milano, Milan,
Italy
| | - John L. Waddington
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2,Ireland;,Jiangsu Key Laboratory of Translational Research & Therapy for Neuro-Psychiatric-Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
47
|
Kwan V, Unda BK, Singh KK. Wnt signaling networks in autism spectrum disorder and intellectual disability. J Neurodev Disord 2016; 8:45. [PMID: 27980692 PMCID: PMC5137220 DOI: 10.1186/s11689-016-9176-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 11/07/2016] [Indexed: 12/20/2022] Open
Abstract
Background Genetic factors play a major role in the risk for neurodevelopmental disorders such as autism spectrum disorders (ASDs) and intellectual disability (ID). The underlying genetic factors have become better understood in recent years due to advancements in next generation sequencing. These studies have uncovered a vast number of genes that are impacted by different types of mutations (e.g., de novo, missense, truncation, copy number variations). Abstract Given the large volume of genetic data, analyzing each gene on its own is not a feasible approach and will take years to complete, let alone attempt to use the information to develop novel therapeutics. To make sense of independent genomic data, one approach is to determine whether multiple risk genes function in common signaling pathways that identify signaling “hubs” where risk genes converge. This approach has led to multiple pathways being implicated, such as synaptic signaling, chromatin remodeling, alternative splicing, and protein translation, among many others. In this review, we analyze recent and historical evidence indicating that multiple risk genes, including genes denoted as high-confidence and likely causal, are part of the Wingless (Wnt signaling) pathway. In the brain, Wnt signaling is an evolutionarily conserved pathway that plays an instrumental role in developing neural circuits and adult brain function. Conclusions We will also review evidence that pharmacological therapies and genetic mouse models further identify abnormal Wnt signaling, particularly at the synapse, as being disrupted in ASDs and contributing to disease pathology.
Collapse
Affiliation(s)
- Vickie Kwan
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8S 4K1 Canada
| | - Brianna K Unda
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8S 4K1 Canada
| | - Karun K Singh
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8S 4K1 Canada
| |
Collapse
|
48
|
Niwa M, Cash-Padgett T, Kubo KI, Saito A, Ishii K, Sumitomo A, Taniguchi Y, Ishizuka K, Jaaro-Peled H, Tomoda T, Nakajima K, Sawa A, Kamiya A. DISC1 a key molecular lead in psychiatry and neurodevelopment: No-More Disrupted-in-Schizophrenia 1. Mol Psychiatry 2016; 21:1488-1489. [PMID: 27595595 PMCID: PMC5472474 DOI: 10.1038/mp.2016.154] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Minae Niwa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Tyler Cash-Padgett
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Atsushi Saito
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kazuhiro Ishii
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Akiko Sumitomo
- Medical Innovation Center, Kyoto University, Kyoto 606-8507, Japan
| | - Yu Taniguchi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Koko Ishizuka
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hanna Jaaro-Peled
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Toshifumi Tomoda
- Medical Innovation Center, Kyoto University, Kyoto 606-8507, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Atsushi Kamiya
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
49
|
Neuregulin-1 Regulates Cortical Inhibitory Neuron Dendrite and Synapse Growth through DISC1. Neural Plast 2016; 2016:7694385. [PMID: 27847649 PMCID: PMC5099462 DOI: 10.1155/2016/7694385] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 07/05/2016] [Accepted: 09/05/2016] [Indexed: 11/20/2022] Open
Abstract
Cortical inhibitory neurons play crucial roles in regulating excitatory synaptic networks and cognitive function and aberrant development of these cells have been linked to neurodevelopmental disorders. The secreted neurotrophic factor Neuregulin-1 (NRG1) and its receptor ErbB4 are established regulators of inhibitory neuron connectivity, but the developmental signalling mechanisms regulating this process remain poorly understood. Here, we provide evidence that NRG1-ErbB4 signalling functions through the multifunctional scaffold protein, Disrupted in Schizophrenia 1 (DISC1), to regulate the development of cortical inhibitory interneuron dendrite and synaptic growth. We found that NRG1 increases inhibitory neuron dendrite complexity and glutamatergic synapse formation onto inhibitory neurons and that this effect is blocked by expression of a dominant negative DISC1 mutant, or DISC1 knockdown. We also discovered that NRG1 treatment increases DISC1 expression and its localization to glutamatergic synapses being made onto cortical inhibitory neurons. Mechanistically, we determined that DISC1 binds ErbB4 within cortical inhibitory neurons. Collectively, these data suggest that a NRG1-ErbB4-DISC1 signalling pathway regulates the development of cortical inhibitory neuron dendrite and synaptic growth. Given that NRG1, ErbB4, and DISC1 are schizophrenia-linked genes, these findings shed light on how independent risk factors may signal in a common developmental pathway that contributes to neural connectivity defects and disease pathogenesis.
Collapse
|
50
|
Mostaid MS, Lloyd D, Liberg B, Sundram S, Pereira A, Pantelis C, Karl T, Weickert CS, Everall IP, Bousman CA. Neuregulin-1 and schizophrenia in the genome-wide association study era. Neurosci Biobehav Rev 2016; 68:387-409. [DOI: 10.1016/j.neubiorev.2016.06.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 05/30/2016] [Accepted: 06/03/2016] [Indexed: 12/22/2022]
|