1
|
Zhang T, Liu C, Zhong N, Wang Y, Huang Y, Zhang X. Advances in the Treatment of Cognitive Impairment in Schizophrenia: Targeting NMDA Receptor Pathways. Int J Mol Sci 2024; 25:10668. [PMID: 39408997 PMCID: PMC11477438 DOI: 10.3390/ijms251910668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Cognitive impairment is a core feature of schizophrenia, playing a pivotal role in the pathogenesis and prognosis of this disorder. Cognitive impairment in schizophrenia encompasses a wide range of domains, including processing speed, episodic memory, working memory, and executive function. These deficits persist throughout the course of the illness and significantly impact functional outcomes and quality of life. Therefore, it is imperative to identify the biological basis of cognitive deficits in schizophrenia and develop effective treatments. The role of N-methyl-D-aspartate (NMDA) receptors in synaptic transmission and plasticity has long been recognized, making them potential targets for schizophrenia treatment. This review will focus on emerging pharmacology targeting NMDA receptors, offering strategies for the prevention and treatment of cognitive deficits in schizophrenia.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaoqin Zhang
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo 315211, China; (T.Z.); (C.L.); (N.Z.); (Y.W.); (Y.H.)
| |
Collapse
|
2
|
Oh SJ, Kumari P, Auroni TT, Stone S, Pathak H, Elsharkawy A, Natekar JP, Shin OS, Kumar M. Upregulation of Neuroinflammation-Associated Genes in the Brain of SARS-CoV-2-Infected Mice. Pathogens 2024; 13:528. [PMID: 39057755 PMCID: PMC11280415 DOI: 10.3390/pathogens13070528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Neurological manifestations are a significant complication of coronavirus disease 2019 (COVID-19), but the underlying mechanisms are yet to be understood. Recently, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced neuroinvasion and encephalitis were observed in K18-hACE2 mice, leading to mortality. Our goal in this study was to gain insights into the molecular pathogenesis of neurological manifestations in this mouse model. To analyze differentially expressed genes (DEGs) in the brains of mice following SARS-CoV-2 infection, we performed NanoString gene expression analysis using three individual animal samples at 1, 3, and 6 days post-infection. We identified the DEGs by comparing them to animals that were not infected with the virus. We found that genes upregulated at day 6 post-infection were mainly associated with Toll-like receptor (TLR) signaling, RIG-I-like receptor (RLR) signaling, and cell death pathways. However, downregulated genes were associated with neurodegeneration and synaptic signaling pathways. In correlation with gene expression profiles, a multiplexed immunoassay showed the upregulation of multiple cytokines and chemokines involved in inflammation and cell death in SARS-CoV-2-infected brains. Furthermore, the pathway analysis of DEGs indicated a possible link between TLR2-mediated signaling pathways and neuroinflammation, as well as pyroptosis and necroptosis in the brain. In conclusion, our work demonstrates neuroinflammation-associated gene expression profiles, which can provide key insight into the severe disease observed in COVID-19 patients.
Collapse
Affiliation(s)
- Soo-Jin Oh
- BK21 Graduate Program, Department of Biomedical Sciences, College of Medicine, Korea University Guro Hospital, Seoul 08308, Republic of Korea;
| | - Pratima Kumari
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (P.K.); (T.T.A.); (S.S.); (H.P.); (A.E.); (J.P.N.)
| | - Tabassum Tasnim Auroni
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (P.K.); (T.T.A.); (S.S.); (H.P.); (A.E.); (J.P.N.)
| | - Shannon Stone
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (P.K.); (T.T.A.); (S.S.); (H.P.); (A.E.); (J.P.N.)
| | - Heather Pathak
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (P.K.); (T.T.A.); (S.S.); (H.P.); (A.E.); (J.P.N.)
| | - Amany Elsharkawy
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (P.K.); (T.T.A.); (S.S.); (H.P.); (A.E.); (J.P.N.)
| | - Janhavi Prasad Natekar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (P.K.); (T.T.A.); (S.S.); (H.P.); (A.E.); (J.P.N.)
| | - Ok Sarah Shin
- BK21 Graduate Program, Department of Biomedical Sciences, College of Medicine, Korea University Guro Hospital, Seoul 08308, Republic of Korea;
| | - Mukesh Kumar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (P.K.); (T.T.A.); (S.S.); (H.P.); (A.E.); (J.P.N.)
| |
Collapse
|
3
|
Eckmann S, Young EJ, Gjorgjieva J. Synapse-type-specific competitive Hebbian learning forms functional recurrent networks. Proc Natl Acad Sci U S A 2024; 121:e2305326121. [PMID: 38870059 PMCID: PMC11194505 DOI: 10.1073/pnas.2305326121] [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: 04/04/2023] [Accepted: 04/25/2024] [Indexed: 06/15/2024] Open
Abstract
Cortical networks exhibit complex stimulus-response patterns that are based on specific recurrent interactions between neurons. For example, the balance between excitatory and inhibitory currents has been identified as a central component of cortical computations. However, it remains unclear how the required synaptic connectivity can emerge in developing circuits where synapses between excitatory and inhibitory neurons are simultaneously plastic. Using theory and modeling, we propose that a wide range of cortical response properties can arise from a single plasticity paradigm that acts simultaneously at all excitatory and inhibitory connections-Hebbian learning that is stabilized by the synapse-type-specific competition for a limited supply of synaptic resources. In plastic recurrent circuits, this competition enables the formation and decorrelation of inhibition-balanced receptive fields. Networks develop an assembly structure with stronger synaptic connections between similarly tuned excitatory and inhibitory neurons and exhibit response normalization and orientation-specific center-surround suppression, reflecting the stimulus statistics during training. These results demonstrate how neurons can self-organize into functional networks and suggest an essential role for synapse-type-specific competitive learning in the development of cortical circuits.
Collapse
Affiliation(s)
- Samuel Eckmann
- Computation in Neural Circuits Group, Max Planck Institute for Brain Research, Frankfurt am Main60438, Germany
- Computational and Biological Learning Lab, Department of Engineering, University of Cambridge, CambridgeCB2 1PZ, United Kingdom
| | - Edward James Young
- Computational and Biological Learning Lab, Department of Engineering, University of Cambridge, CambridgeCB2 1PZ, United Kingdom
| | - Julijana Gjorgjieva
- Computation in Neural Circuits Group, Max Planck Institute for Brain Research, Frankfurt am Main60438, Germany
- School of Life Sciences, Technical University Munich, Freising85354, Germany
| |
Collapse
|
4
|
Pecori A, Luppieri V, Santin A, Spedicati B, Zampieri S, Cadenaro M, Girotto G, Concas MP. Clenching the Strings of Bruxism Etiopathogenesis: Association Analyses on Genetics and Environmental Risk Factors in a Deeply Characterized Italian Cohort. Biomedicines 2024; 12:304. [PMID: 38397906 PMCID: PMC10887134 DOI: 10.3390/biomedicines12020304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Bruxism is a worldwide oral health problem. Although there is a consensus about its multifactorial nature, its precise etiopathogenetic mechanisms are unclear. This study, taking advantage of a deeply characterized cohort of 769 individuals (aged 6-89 years) coming from Northern Italy's genetically isolated populations, aims to epidemiologically describe environmental risk factors for bruxism development and identify genes potentially involved through a Genome-Wide Association Study (GWAS) approach. Logistic mixed models adjusted for age and sex were performed to evaluate associations between bruxism and possible risk factors, e.g., anxiety, smoking, and alcohol and caffeine intake. A case-control GWAS (135 cases, 523 controls), adjusted for age, sex, and anxiety, was conducted to identify new candidate genes. The GTEx data analysis was performed to evaluate the identified gene expression in human body tissues. Statistical analyses determined anxiety as a bruxism risk factor (OR = 2.54; 95% CI: 1.20-5.38; p-value = 0.015), and GWAS highlighted three novel genes potentially associated with bruxism: NLGN1 (topSNP = rs2046718; p-value = 2.63 × 10-7), RIMBP2 (topSNP = rs571497947; p-value = 4.68 × 10-7), and LHFP (topSNP = rs2324342; p-value = 7.47 × 10-6). The GTEx data analysis showed their expression in brain tissues. Overall, this work provided a deeper understanding of bruxism etiopathogenesis with the long-term perspective of developing personalized therapeutic approaches for improving affected individuals' quality of life.
Collapse
Affiliation(s)
- Alessandro Pecori
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Via dell’Istria 65, 34137 Trieste, Italy; (A.P.); (V.L.); (B.S.); (S.Z.); (M.C.); (G.G.); (M.P.C.)
| | - Valentina Luppieri
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Via dell’Istria 65, 34137 Trieste, Italy; (A.P.); (V.L.); (B.S.); (S.Z.); (M.C.); (G.G.); (M.P.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149 Trieste, Italy
| | - Aurora Santin
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149 Trieste, Italy
| | - Beatrice Spedicati
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Via dell’Istria 65, 34137 Trieste, Italy; (A.P.); (V.L.); (B.S.); (S.Z.); (M.C.); (G.G.); (M.P.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149 Trieste, Italy
| | - Stefania Zampieri
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Via dell’Istria 65, 34137 Trieste, Italy; (A.P.); (V.L.); (B.S.); (S.Z.); (M.C.); (G.G.); (M.P.C.)
| | - Milena Cadenaro
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Via dell’Istria 65, 34137 Trieste, Italy; (A.P.); (V.L.); (B.S.); (S.Z.); (M.C.); (G.G.); (M.P.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149 Trieste, Italy
| | - Giorgia Girotto
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Via dell’Istria 65, 34137 Trieste, Italy; (A.P.); (V.L.); (B.S.); (S.Z.); (M.C.); (G.G.); (M.P.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149 Trieste, Italy
| | - Maria Pina Concas
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Via dell’Istria 65, 34137 Trieste, Italy; (A.P.); (V.L.); (B.S.); (S.Z.); (M.C.); (G.G.); (M.P.C.)
| |
Collapse
|
5
|
Sindi IA. Implications of Cell Adhesion Molecules in Autism Spectrum Disorder Pathogenesis. J Microsc Ultrastruct 2023; 11:199-205. [PMID: 38213654 PMCID: PMC10779445 DOI: 10.4103/jmau.jmau_15_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/23/2022] [Accepted: 05/09/2022] [Indexed: 11/04/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental illness that leads to repetitive behavior and debilitates social communication. Genetic changes such as susceptible genes and environmental factors promote ASD pathogenesis. Mutations in neuroligins (NLGNs) and neurexin (NRXNs) complex which encode cell adhesion molecules have a significant part in synapses formation, transcription, and excitatory-inhibitory balance. The ASD pathogenesis could partly, at the least, be related to synaptic dysfunction. Here, the NRXNs and NLGNs genes and signaling pathways involved in the synaptic malfunction that causes ASD have been reviewed. Besides, a new insight of NLGNs and NRXNs genes in ASD will be conferred.
Collapse
Affiliation(s)
- Ikhlas A. Sindi
- Department of Biology, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
6
|
Xiao X, Wang X, Zhu K, Li L, He Y, Zhang J, Li L, Hu H, Cui Y, Zhang J, Zheng Y. BACE1 in PV interneuron tunes hippocampal CA1 local circuits and resets priming of fear memory extinction. Mol Psychiatry 2023; 28:4151-4162. [PMID: 37452089 DOI: 10.1038/s41380-023-02176-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
BACE1 is the rate-limiting enzyme for β-amyloid (Aβ) production and therefore is considered a prime drug target for treating Alzheimer's disease (AD). Nevertheless, the BACE1 inhibitors failed in clinical trials, even exhibiting cognitive worsening, implying that BACE1 may function in regulating cognition-relevant neural circuits. Here, we found that parvalbumin-positive inhibitory interneurons (PV INs) in hippocampal CA1 express BACE1 at a high level. We designed and developed a mouse strain with conditional knockout of BACE1 in PV neurons. The CA1 fast-spiking PV INs with BACE1 deletion exhibited an enhanced response of postsynaptic N-methyl-D-aspartate (NMDA) receptors to local stimulation on CA1 oriens, with average intrinsic electrical properties and fidelity in synaptic integration. Intriguingly, the BACE1 deletion reorganized the CA1 recurrent inhibitory motif assembled by the heterogeneous pyramidal neurons (PNs) and the adjacent fast-spiking PV INs from the superficial to the deep layer. Moreover, the conditional BACE1 deletion impaired the AMPARs-mediated excitatory transmission of deep CA1 PNs. Further rescue experiments confirmed that these phenotypes require the enzymatic activity of BACE1. Above all, the BACE1 deletion resets the priming of the fear memory extinction. Our findings suggest a neuron-specific working model of BACE1 in regulating learning and memory circuits. The study may provide a potential path of targeting BACE1 and NMDAR together to circumvent cognitive worsening due to a single application of BACE1 inhibitor in AD patients.
Collapse
Affiliation(s)
- Xuansheng Xiao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Xiaotong Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Ke Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Lijuan Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Ying He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Jinglan Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Linying Li
- Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Hanning Hu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Yanqiu Cui
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jianliang Zhang
- Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yan Zheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
7
|
Zhou L, Sun X, Duan J. NMDARs regulate the excitatory-inhibitory balance within neural circuits. BRAIN SCIENCE ADVANCES 2023. [DOI: 10.26599/bsa.2022.9050020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Excitatory-inhibitory (E/I) balance is essential for normal neural development, behavior and cognition. E/I imbalance leads to a variety of neurological disorders, such as autism and schizophrenia. NMDA receptors (NMDARs) regulate AMPAR-mediated excitatory and GABAAR-mediated inhibitory synaptic transmission, suggesting that NMDARs play an important role in the establishment and maintenance of the E/I balance. In this review, we briefly introduced NMDARs, AMPARs and GABAARs, summarized the current studies on E/I balance mediated by NMDARs, and discussed the current advances in NMDAR-mediated AMPAR and GABAAR development. Specifically, we analyzed the role of NMDAR subunits in the establishment and maintenance of E/I balance, which may provide new therapeutic strategies for the recovery of E/I imbalance in neurological disorders.
Collapse
Affiliation(s)
- Liang Zhou
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Xiaohui Sun
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Jingjing Duan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| |
Collapse
|
8
|
Hong N, Kim HJ, Kang K, Park JO, Mun S, Kim HG, Kang BH, Chung PS, Lee MY, Ahn JC. Photobiomodulation improves the synapses and cognitive function and ameliorates epileptic seizure by inhibiting downregulation of Nlgn3. Cell Biosci 2023; 13:8. [PMID: 36635704 PMCID: PMC9837965 DOI: 10.1186/s13578-022-00949-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/21/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Temporal lobe epilepsy (TLE) remains one of the most drug-resistant focal epilepsies. Glutamate excitotoxicity and neuroinflammation which leads to loss of synaptic proteins and neuronal death appear to represent a pathogen that characterizes the neurobiology of TLE. Photobiomodulation (PBM) is a rapidly growing therapy for the attenuation of neuronal degeneration harboring non-invasiveness benefits. However, the detailed effects of PBM on excitotoxicity or neuroinflammation remain unclear. We investigated whether tPBM exerts neuroprotective effects on hippocampal neurons in epilepsy mouse model by regulating synapse and synapse-related genes. METHODS In an in vitro study, we performed imaging analysis and western blot in primary hippocampal neurons from embryonic (E17) rat pups. In an in vivo study, RNA sequencing was performed to identify the gene regulatory by PBM. Histological stain and immunohistochemistry analyses were used to assess synaptic connections, neuroinflammation and neuronal survival. Behavioral tests were used to evaluate the effects of PBM on cognitive functions. RESULTS PBM was upregulated synaptic connections in an in vitro. In addition, it was confirmed that transcranial PBM reduced synaptic degeneration, neuronal apoptosis, and neuroinflammation in an in vivo. These effects of PBM were supported by RNA sequencing results showing the relation of PBM with gene regulatory networks of neuronal functions. Specifically, Nlgn3 showed increase after PBM and silencing the Nlgn3 reversed the positive effect of PBM in in vitro. Lastly, behavioral alterations including hypoactivity, anxiety and impaired memory were recovered along with the reduction of seizure score in PBM-treated mice. CONCLUSIONS Our findings demonstrate that PBM attenuates epileptic excitotoxicity, neurodegeneration and cognitive decline induced by TLE through inhibition of the Nlgn3 gene decrease induced by excitotoxicity.
Collapse
Affiliation(s)
- Namgue Hong
- grid.411982.70000 0001 0705 4288Medical Laser Research Center, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Department of Biomedical Science, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Hee Jung Kim
- grid.411982.70000 0001 0705 4288Department of Physiology, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Keunsoo Kang
- grid.411982.70000 0001 0705 4288Department of Microbiology, College of Science & Technology, Dankook University, Cheonan, Republic of Korea
| | - Ji On Park
- grid.411982.70000 0001 0705 4288Department of Medicine, Graduate School of Dankook University, Dankook University, Cheonan, Republic of Korea
| | - Seyoung Mun
- grid.411982.70000 0001 0705 4288Department of Microbiology, College of Science & Technology, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Center for Bio-Medical Engineering Core Facility, Dankook University, Cheonan, Republic of Korea
| | - Hyung-Gun Kim
- grid.411982.70000 0001 0705 4288Department of Pharmacology, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Bong Hui Kang
- grid.411982.70000 0001 0705 4288Department of Neurology, Dankook University Hospital, Dankook University, Cheonan, Republic of Korea
| | - Phil-Sang Chung
- grid.411982.70000 0001 0705 4288Beckman Laser Institute Korea, Dankook University Hospital, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Department of Otolaryngology-Head & Neck Surgery, College of Medicine, Dankook University Hospital, Dankook University, Cheonan, Republic of Korea
| | - Min Young Lee
- grid.411982.70000 0001 0705 4288Medical Laser Research Center, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Department of Biomedical Science, College of Medicine, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Beckman Laser Institute Korea, Dankook University Hospital, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Department of Otolaryngology-Head & Neck Surgery, College of Medicine, Dankook University Hospital, Dankook University, Cheonan, Republic of Korea
| | - Jin-Chul Ahn
- grid.411982.70000 0001 0705 4288Medical Laser Research Center, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Department of Biomedical Science, College of Medicine, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Beckman Laser Institute Korea, Dankook University Hospital, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
9
|
Liu X, Hua F, Yang D, Lin Y, Zhang L, Ying J, Sheng H, Wang X. Roles of neuroligins in central nervous system development: focus on glial neuroligins and neuron neuroligins. Lab Invest 2022; 20:418. [PMID: 36088343 PMCID: PMC9463862 DOI: 10.1186/s12967-022-03625-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/01/2022] [Indexed: 11/10/2022]
Abstract
Neuroligins are postsynaptic cell adhesion molecules that are relevant to many neurodevelopmental disorders. They are differentially enriched at the postsynapse and interact with their presynaptic ligands, neurexins, whose differential binding to neuroligins has been shown to regulate synaptogenesis, transmission, and other synaptic properties. The proper functioning of functional networks in the brain depends on the proper connection between neuronal synapses. Impaired synaptogenesis or synaptic transmission results in synaptic dysfunction, and these synaptic pathologies are the basis for many neurodevelopmental disorders. Deletions or mutations in the neuroligins genes have been found in patients with both autism and schizophrenia. It is because of the important role of neuroligins in synaptic connectivity and synaptic dysfunction that studies on neuroligins in the past have mainly focused on their expression in neurons. As studies on the expression of genes specific to various cells of the central nervous system deepened, neuroligins were found to be expressed in non-neuronal cells as well. In the central nervous system, glial cells are the most representative non-neuronal cells, which can also express neuroligins in large amounts, especially astrocytes and oligodendrocytes, and they are involved in the regulation of synaptic function, as are neuronal neuroligins. This review examines the mechanisms of neuron neuroligins and non-neuronal neuroligins in the central nervous system and also discusses the important role of neuroligins in the development of the central nervous system and neurodevelopmental disorders from the perspective of neuronal neuroligins and glial neuroligins.
Collapse
|
10
|
Neuroligin-1 plays an important role in methamphetamine-induced hippocampal synaptic plasticity. Toxicol Lett 2022; 361:1-9. [PMID: 35331841 DOI: 10.1016/j.toxlet.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 02/02/2022] [Accepted: 03/17/2022] [Indexed: 11/21/2022]
Abstract
The neurotoxic effects of methamphetamine (METH) include not only neuronal apoptosis and autophagy, but also lead to substance use disorder and have become increasingly prominent. Studies suggest that synaptic plasticity may be the structural basis of METH-induced neurological impairment. Neuroligins are postsynaptic adhesion molecules involved in the regulation of synaptic organization and function. Animal studies have shown that neuroligin (NLG)- 1 is involved in memory formation; however, its role in METH-induced neurotoxicity is not clear. In the present study, we used 1 mM METH in vitro; mice in the acute and subacute exposure groups received intraperitoneal injections of 30 mg/kg METH (1 injection) or 15 mg/kg METH (8 separate injections at 12-h intervals). We found that the expression of NLG-1, Synapsin-1, and postsynaptic density-95 were increased after METH exposure. We further observed that METH-induced inhibition of long-term potentiation and spatial memory loss could be alleviated when mice were pretreated with NLG-1 small interfering RNA. Therefore, our study provides evidence that NLG-1 is involved in METH-induced hippocampal synaptic plasticity and may be a potential target for the treatment of METH-induced neurotoxicity.
Collapse
|
11
|
Bayat M, Khalili A, Bayat G, Akbari S, Yousefi Nejad A, Borhani Haghighi A, Haghani M. Effects of platelet-rich plasma on the memory impairment, apoptosis, and hippocampal synaptic plasticity in a rat model of hepatic encephalopathy. Brain Behav 2022; 12:e2447. [PMID: 34855284 PMCID: PMC8785608 DOI: 10.1002/brb3.2447] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/22/2021] [Accepted: 10/30/2021] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES In the present study, we aimed to determine whether intraperitoneal injection of platelet-rich plasma (PRP) could have a neuroprotective effect on learning, memory, and synaptic plasticity impairment as well as hippocampal apoptosis in rats with hepatic encephalopathy induced by bile duct ligated (BDL). METHODS The rats were divided into four groups: the control, sham, BDL+ V (vehicle), and BDL+ PRP. The BDL rats were treated with PRP immediately after the surgery, and the injection was done every 3 days for 30 days. The passive avoidance and Morris water maze tests were used for the evaluation of learning and memory. The long-term potentiation (LTP), basal-synaptic transmission, and paired-pulse ratio, as an index for measurement of neurotransmitter release probability, were evaluated by field-potential recording. After taking a blood sample for assessment of the liver enzymes, the animals were sacrificed and their hippocampus was removed for evaluation of cleaved caspase-3 by Western blot. RESULTS Serological assessment of the liver function showed that BDL severely impaired the liver function. Also, PRP treatment could partially improve the liver dysfunction along with recovery in fear memory and spatial learning memory performance, LTP, basal-synaptic transmission, and neurotransmitter release probability. PRP-treated rats also showed a significant reduction in neuronal apoptosis in the CA1 area. CONCLUSIONS The results of this study suggest that PRP improves cognitive performance and synaptic plasticity in BDL rats via direct neuroprotective property and/or indirectly by improvement of hepatic dysfunction.
Collapse
Affiliation(s)
- Mahnaz Bayat
- Clinical Neurology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Azadeh Khalili
- Department of Physiology-Pharmacology-Medical Physic, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Gholamreza Bayat
- Department of Physiology-Pharmacology-Medical Physic, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.,Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Somayeh Akbari
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amirhossein Yousefi Nejad
- Faculty of Veterinary Medicine, Department of Veterinary Medicine, Islamic Azad University of Kazeroon, Shiraz, Iran
| | | | - Masoud Haghani
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran.,Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Moussa AJ, Wester JC. Cell-type specific transcriptomic signatures of neocortical circuit organization and their relevance to autism. Front Neural Circuits 2022; 16:982721. [PMID: 36213201 PMCID: PMC9545608 DOI: 10.3389/fncir.2022.982721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
A prevailing challenge in neuroscience is understanding how diverse neuronal cell types select their synaptic partners to form circuits. In the neocortex, major classes of excitatory projection neurons and inhibitory interneurons are conserved across functionally distinct regions. There is evidence these classes form canonical circuit motifs that depend primarily on their identity; however, regional cues likely also influence their choice of synaptic partners. We mined the Allen Institute's single-cell RNA-sequencing database of mouse cortical neurons to study the expression of genes necessary for synaptic connectivity and physiology in two regions: the anterior lateral motor cortex (ALM) and the primary visual cortex (VISp). We used the Allen's metadata to parse cells by clusters representing major excitatory and inhibitory classes that are common to both ALM and VISp. We then performed two types of pairwise differential gene expression analysis: (1) between different neuronal classes within the same brain region (ALM or VISp), and (2) between the same neuronal class in ALM and VISp. We filtered our results for differentially expressed genes related to circuit connectivity and developed a novel bioinformatic approach to determine the sets uniquely enriched in each neuronal class in ALM, VISp, or both. This analysis provides an organized set of genes that may regulate synaptic connectivity and physiology in a cell-type-specific manner. Furthermore, it identifies candidate mechanisms for circuit organization that are conserved across functionally distinct cortical regions or that are region dependent. Finally, we used the SFARI Human Gene Module to identify genes from this analysis that are related to risk for autism spectrum disorder (ASD). Our analysis provides clear molecular targets for future studies to understand neocortical circuit organization and abnormalities that underlie autistic phenotypes.
Collapse
Affiliation(s)
- Anthony J Moussa
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Jason C Wester
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, OH, United States
| |
Collapse
|
13
|
Ciarpella F, Zamfir RG, Campanelli A, Ren E, Pedrotti G, Bottani E, Borioli A, Caron D, Di Chio M, Dolci S, Ahtiainen A, Malpeli G, Malerba G, Bardoni R, Fumagalli G, Hyttinen J, Bifari F, Palazzolo G, Panuccio G, Curia G, Decimo I. Murine cerebral organoids develop network of functional neurons and hippocampal brain region identity. iScience 2021; 24:103438. [PMID: 34901791 PMCID: PMC8640475 DOI: 10.1016/j.isci.2021.103438] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/13/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Brain organoids are in vitro three-dimensional (3D) self-organized neural structures, which can enable disease modeling and drug screening. However, their use for standardized large-scale drug screening studies is limited by their high batch-to-batch variability, long differentiation time (10-20 weeks), and high production costs. This is particularly relevant when brain organoids are obtained from human induced pluripotent stem cells (iPSCs). Here, we developed, for the first time, a highly standardized, reproducible, and fast (5 weeks) murine brain organoid model starting from embryonic neural stem cells. We obtained brain organoids, which progressively differentiated and self-organized into 3D networks of functional neurons with dorsal forebrain phenotype. Furthermore, by adding the morphogen WNT3a, we generated brain organoids with specific hippocampal region identity. Overall, our results showed the establishment of a fast, robust and reproducible murine 3D in vitro brain model that may represent a useful tool for high-throughput drug screening and disease modeling.
Collapse
Affiliation(s)
- Francesca Ciarpella
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Raluca Georgiana Zamfir
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Alessandra Campanelli
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Elisa Ren
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giulia Pedrotti
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Emanuela Bottani
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Andrea Borioli
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Davide Caron
- Department of Neuroscience and Brain Technologies (NBT), Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Marzia Di Chio
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Sissi Dolci
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Annika Ahtiainen
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Giorgio Malpeli
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, 37134 Verona, Italy
| | - Giovanni Malerba
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
| | - Rita Bardoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Guido Fumagalli
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Jari Hyttinen
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133 Milan, Italy
| | - Gemma Palazzolo
- Department of Neuroscience and Brain Technologies (NBT), Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Gabriella Panuccio
- Department of Neuroscience and Brain Technologies (NBT), Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Giulia Curia
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Ilaria Decimo
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| |
Collapse
|
14
|
Neuroligin-3 Regulates Excitatory Synaptic Transmission and EPSP-Spike Coupling in the Dentate Gyrus In Vivo. Mol Neurobiol 2021; 59:1098-1111. [PMID: 34845591 PMCID: PMC8857112 DOI: 10.1007/s12035-021-02663-9] [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] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 11/22/2021] [Indexed: 11/27/2022]
Abstract
Neuroligin-3 (Nlgn3), a neuronal adhesion protein implicated in autism spectrum disorder (ASD), is expressed at excitatory and inhibitory postsynapses and hence may regulate neuronal excitation/inhibition balance. To test this hypothesis, we recorded field excitatory postsynaptic potentials (fEPSPs) in the dentate gyrus of Nlgn3 knockout (KO) and wild-type mice. Synaptic transmission evoked by perforant path stimulation was reduced in KO mice, but coupling of the fEPSP to the population spike was increased, suggesting a compensatory change in granule cell excitability. These findings closely resemble those in neuroligin-1 (Nlgn1) KO mice and could be partially explained by the reduction in Nlgn1 levels we observed in hippocampal synaptosomes from Nlgn3 KO mice. However, unlike Nlgn1, Nlgn3 is not necessary for long-term potentiation. We conclude that while Nlgn1 and Nlgn3 have distinct functions, both are required for intact synaptic transmission in the mouse dentate gyrus. Our results indicate that interactions between neuroligins may play an important role in regulating synaptic transmission and that ASD-related neuroligin mutations may also affect the synaptic availability of other neuroligins.
Collapse
|
15
|
Durand N, Aguilar P, Demondion E, Bourgeois T, Bozzolan F, Debernard S. Neuroligin 1 expression is linked to plasticity of behavioral and neuronal responses to sex pheromone in the male moth Agrotis ipsilon. J Exp Biol 2021; 224:273481. [PMID: 34647597 DOI: 10.1242/jeb.243184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/07/2021] [Indexed: 11/20/2022]
Abstract
In the moth Agrotis ipsilon, the behavioral response of males to the female-emitted sex pheromone increases throughout adult life and following a prior exposure to sex pheromone, whereas it is temporally inhibited after the onset of mating. This behavioral flexibility is paralleled with changes in neuronal sensitivity to pheromone signal within the primary olfactory centers, the antennal lobes. In the present study, we tested the hypothesis that neuroligins, post-synaptic transmembrane proteins known to act as mediators of neuronal remodeling, are involved in the olfactory modulation in A. ipsilon males. We cloned a full-length cDNA encoding neuroligin 1, which is expressed predominantly in brain and especially in antennal lobes. The level of neuroligin 1 expression in antennal lobes gradually raised from day-2 until day-4 of adult life, as well as at 24 h, 48 h and 72 h following pre-exposure to sex pheromone, and the temporal dynamic of these changes correlated with increased sex pheromone responsiveness. By contrast, there was no significant variation in antennal lobe neuroligin 1 expression during the post-mating refractory period. Taken together, these results highlight that age- and odor experience-related increase in sex pheromone responsiveness is linked to the overexpression of neuroligin 1 in antennal lobes, thus suggesting a potential role played by this post-synaptic cell-adhesion molecule in mediating the plasticity of the central olfactory system in A. ipsilon.
Collapse
Affiliation(s)
- Nicolas Durand
- FRE CNRS 3498, Ecologie et Dynamique des Systèmes Anthropisés, Université de Picardie, Jules Verne, 80039 Amiens, France
| | - Paleo Aguilar
- Institute of Biology, Complutense University of Madrid, Pozuelo de Alarcon, 28223 Madrid, Spain
| | - Elodie Demondion
- Sorbonne Université, INRA, CNRS, UPEC, IRD, Univ. P7, Institute of Ecology and Environmental Sciences of Paris, 78026 Versailles, France
| | - Thomas Bourgeois
- Sorbonne Université, INRA, CNRS, UPEC, IRD, Univ. P7, Institute of Ecology and Environmental Sciences of Paris, 78026 Versailles, France
| | - Françoise Bozzolan
- Sorbonne Université, INRA, CNRS, UPEC, IRD, Univ. P7, Institute of Ecology and Environmental Sciences of Paris, 75005 Paris, France
| | - Stéphane Debernard
- Sorbonne Université, INRA, CNRS, UPEC, IRD, Univ. P7, Institute of Ecology and Environmental Sciences of Paris, 75005 Paris, France
| |
Collapse
|
16
|
Deciphering the mechanisms of regulation of an excitatory synapse via cyclooxygenase-2. A review. Biochem Pharmacol 2021; 192:114729. [PMID: 34400127 DOI: 10.1016/j.bcp.2021.114729] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/20/2022]
Abstract
Cyclooxygenase (COX) is a heme-containing enzyme that produces prostaglandins (PGs) via a pathway known as the arachidonic acid (AA) cascade. Two isoforms of COX enzyme (COX-1 and COX-2) and splice variant (COX-3) have been described so far. COX-2 is a neuronal enzyme that is intensively produced during activation of the synapse and glutamate (Glu) release. The end product of COX-2 action, prostaglandin E2 (PGE2), regulates Glu level in a retrograde manner. At the same time, the level of Glu, the primary excitatory neurotransmitter, is regulated in the excitatory synapse via Glu receptors, both ionotropic and metabotropic ones. Glu receptors are known modulators of behavior, engaged in cognition and mood. So far, the interaction between ionotropic N-methyl-D-aspartate (NMDA) receptors or metabotropic glutamate (mGluRs) receptors and COX-2 was found. Here, based on literature data and own research, a new mechanism of action of COX-2 in an excitatory synapse will be presented.
Collapse
|
17
|
Feng S, Huang H, Wang N, Wei Y, Liu Y, Qin D. Sleep Disorders in Children With Autism Spectrum Disorder: Insights From Animal Models, Especially Non-human Primate Model. Front Behav Neurosci 2021; 15:673372. [PMID: 34093147 PMCID: PMC8173056 DOI: 10.3389/fnbeh.2021.673372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/16/2021] [Indexed: 02/05/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a heterogeneous neurodevelopmental disorder with deficient social skills, communication deficits and repetitive behaviors. The prevalence of ASD has increased among children in recent years. Children with ASD experience more sleep problems, and sleep appears to be essential for the survival and integrity of most living organisms, especially for typical synaptic development and brain plasticity. Many methods have been used to assess sleep problems over past decades such as sleep diaries and parent-reported questionnaires, electroencephalography, actigraphy and videosomnography. A substantial number of rodent and non-human primate models of ASD have been generated. Many of these animal models exhibited sleep disorders at an early age. The aim of this review is to examine and discuss sleep disorders in children with ASD. Toward this aim, we evaluated the prevalence, clinical characteristics, phenotypic analyses, and pathophysiological brain mechanisms of ASD. We highlight the current state of animal models for ASD and explore their implications and prospects for investigating sleep disorders associated with ASD.
Collapse
Affiliation(s)
- Shufei Feng
- Department of Pediatric Rehabilitation Medicine, Kunming Children’s Hospital, Kunming, China
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Haoyu Huang
- Department of Pediatric Rehabilitation Medicine, Kunming Children’s Hospital, Kunming, China
| | - Na Wang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Yuanyuan Wei
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Yun Liu
- Department of Pediatric Rehabilitation Medicine, Kunming Children’s Hospital, Kunming, China
| | - Dongdong Qin
- Department of Pediatric Rehabilitation Medicine, Kunming Children’s Hospital, Kunming, China
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|
18
|
Qian KY, Zeng WX, Hao Y, Zeng XT, Liu H, Li L, Chen L, Tian FM, Chang C, Hall Q, Song CX, Gao S, Hu Z, Kaplan JM, Li Q, Tong XJ. Male pheromones modulate synaptic transmission at the C. elegans neuromuscular junction in a sexually dimorphic manner. eLife 2021; 10:e67170. [PMID: 33787493 PMCID: PMC8051947 DOI: 10.7554/elife.67170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/30/2021] [Indexed: 12/24/2022] Open
Abstract
The development of functional synapses in the nervous system is important for animal physiology and behaviors, and its disturbance has been linked with many neurodevelopmental disorders. The synaptic transmission efficacy can be modulated by the environment to accommodate external changes, which is crucial for animal reproduction and survival. However, the underlying plasticity of synaptic transmission remains poorly understood. Here we show that in Caenorhabditis elegans, the male environment increases the hermaphrodite cholinergic transmission at the neuromuscular junction (NMJ), which alters hermaphrodites' locomotion velocity and mating efficiency. We identify that the male-specific pheromones mediate this synaptic transmission modulation effect in a developmental stage-dependent manner. Dissection of the sensory circuits reveals that the AWB chemosensory neurons sense those male pheromones and further transduce the information to NMJ using cGMP signaling. Exposure of hermaphrodites to the male pheromones specifically increases the accumulation of presynaptic CaV2 calcium channels and clustering of postsynaptic acetylcholine receptors at cholinergic synapses of NMJ, which potentiates cholinergic synaptic transmission. Thus, our study demonstrates a circuit mechanism for synaptic modulation and behavioral flexibility by sexual dimorphic pheromones.
Collapse
Affiliation(s)
- Kang-Ying Qian
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of SciencesShanghaiChina
| | - Wan-Xin Zeng
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of SciencesShanghaiChina
| | - Yue Hao
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of SciencesShanghaiChina
| | - Xian-Ting Zeng
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
| | - Haowen Liu
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), The University of QueenslandBrisbaneAustralia
| | - Lei Li
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), The University of QueenslandBrisbaneAustralia
| | - Lili Chen
- College of Life Science and Technology, Huazhong University of Science and TechnologyWuhanChina
| | - Fu-min Tian
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Cindy Chang
- Department of Molecular Biology, Massachusetts General HospitalBostonUnited States
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Qi Hall
- Department of Molecular Biology, Massachusetts General HospitalBostonUnited States
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Chun-Xue Song
- Center for Brain Science, Shanghai Children's Medical CenterShanghaiChina
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shangbang Gao
- College of Life Science and Technology, Huazhong University of Science and TechnologyWuhanChina
| | - Zhitao Hu
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), The University of QueenslandBrisbaneAustralia
| | - Joshua M Kaplan
- Department of Molecular Biology, Massachusetts General HospitalBostonUnited States
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Qian Li
- Center for Brain Science, Shanghai Children's Medical CenterShanghaiChina
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Research Center for Brain Science and Brain-Inspired IntelligenceShanghaiChina
| | - Xia-Jing Tong
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
| |
Collapse
|
19
|
Kuhlmann N, Wagner Valladolid M, Quesada-Ramírez L, Farrer MJ, Milnerwood AJ. Chronic and Acute Manipulation of Cortical Glutamate Transmission Induces Structural and Synaptic Changes in Co-cultured Striatal Neurons. Front Cell Neurosci 2021; 15:569031. [PMID: 33679324 PMCID: PMC7930618 DOI: 10.3389/fncel.2021.569031] [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: 06/02/2020] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
In contrast to the prenatal topographic development of sensory cortices, striatal circuit organization is slow and requires the functional maturation of cortical and thalamic excitatory inputs throughout the first postnatal month. While mechanisms regulating synapse development and plasticity are quite well described at excitatory synapses of glutamatergic neurons in the neocortex, comparatively little is known of how this translates to glutamate synapses onto GABAergic neurons in the striatum. Here we investigate excitatory striatal synapse plasticity in an in vitro system, where glutamate can be studied in isolation from dopamine and other neuromodulators. We examined pre-and post-synaptic structural and functional plasticity in GABAergic striatal spiny projection neurons (SPNs), co-cultured with glutamatergic cortical neurons. After synapse formation, medium-term (24 h) TTX silencing increased the density of filopodia, and modestly decreased dendritic spine density, when assayed at 21 days in vitro (DIV). Spine reductions appeared to require residual spontaneous activation of ionotropic glutamate receptors. Conversely, chronic (14 days) TTX silencing markedly reduced spine density without any observed increase in filopodia density. Time-dependent, biphasic changes to the presynaptic marker Synapsin-1 were also observed, independent of residual spontaneous activity. Acute silencing (3 h) did not affect presynaptic markers or postsynaptic structures. To induce rapid, activity-dependent plasticity in striatal neurons, a chemical NMDA receptor-dependent “long-term potentiation (LTP)” paradigm was employed. Within 30 min, this increased spine and GluA1 cluster densities, and the percentage of spines containing GluA1 clusters, without altering the presynaptic signal. The results demonstrate that the growth and pruning of dendritic protrusions is an active process, requiring glutamate receptor activity in striatal projection neurons. Furthermore, NMDA receptor activation is sufficient to drive glutamatergic structural plasticity in SPNs, in the absence of dopamine or other neuromodulators.
Collapse
Affiliation(s)
- Naila Kuhlmann
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada.,Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | | | - Lucía Quesada-Ramírez
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada
| | - Matthew J Farrer
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada.,McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Austen J Milnerwood
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada.,Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| |
Collapse
|
20
|
Gobert D, Schohl A, Kutsarova E, Ruthazer ES. TORC1 selectively regulates synaptic maturation and input convergence in the developing visual system. Dev Neurobiol 2020; 80:332-350. [DOI: 10.1002/dneu.22782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 08/07/2020] [Accepted: 09/16/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Delphine Gobert
- Montreal Neurological Institute‐Hospital McGill University Montreal QC Canada
| | - Anne Schohl
- Montreal Neurological Institute‐Hospital McGill University Montreal QC Canada
| | - Elena Kutsarova
- Montreal Neurological Institute‐Hospital McGill University Montreal QC Canada
| | - Edward S. Ruthazer
- Montreal Neurological Institute‐Hospital McGill University Montreal QC Canada
| |
Collapse
|
21
|
Ray PR, Wangzhou A, Ghneim N, Yousuf MS, Paige C, Tavares-Ferreira D, Mwirigi JM, Shiers S, Sankaranarayanan I, McFarland AJ, Neerukonda SV, Davidson S, Dussor G, Burton MD, Price TJ. A pharmacological interactome between COVID-19 patient samples and human sensory neurons reveals potential drivers of neurogenic pulmonary dysfunction. Brain Behav Immun 2020; 89:559-568. [PMID: 32497778 PMCID: PMC7263237 DOI: 10.1016/j.bbi.2020.05.078] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/21/2022] Open
Abstract
The SARS-CoV-2 virus infects cells of the airway and lungs in humans causing the disease COVID-19. This disease is characterized by cough, shortness of breath, and in severe cases causes pneumonia and acute respiratory distress syndrome (ARDS) which can be fatal. Bronchial alveolar lavage fluid (BALF) and plasma from mild and severe cases of COVID-19 have been profiled using protein measurements and bulk and single cell RNA sequencing. Onset of pneumonia and ARDS can be rapid in COVID-19, suggesting a potential neuronal involvement in pathology and mortality. We hypothesized that SARS-CoV-2 infection drives changes in immune cell-derived factors that then interact with receptors expressed by the sensory neuronal innervation of the lung to further promote important aspects of disease severity, including ARDS. We sought to quantify how immune cells might interact with sensory innervation of the lung in COVID-19 using published data from patients, existing RNA sequencing datasets from human dorsal root ganglion neurons and other sources, and a genome-wide ligand-receptor pair database curated for pharmacological interactions relevant for neuro-immune interactions. Our findings reveal a landscape of ligand-receptor interactions in the lung caused by SARS-CoV-2 viral infection and point to potential interventions to reduce the burden of neurogenic inflammation in COVID-19 pulmonary disease. In particular, our work highlights opportunities for clinical trials with existing or under development rheumatoid arthritis and other (e.g. CCL2, CCR5 or EGFR inhibitors) drugs to treat high risk or severe COVID-19 cases.
Collapse
Affiliation(s)
- Pradipta R Ray
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA.
| | - Andi Wangzhou
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Nizar Ghneim
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Muhammad S Yousuf
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Candler Paige
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Diana Tavares-Ferreira
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Juliet M Mwirigi
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Stephanie Shiers
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Ishwarya Sankaranarayanan
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Amelia J McFarland
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Sanjay V Neerukonda
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Steve Davidson
- University of Cincinnati, College of Medicine, Department of Anesthesiology, USA
| | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Michael D Burton
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Neuroimmunology and Behavior Research Group, USA
| | - Theodore J Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA.
| |
Collapse
|
22
|
Trobiani L, Meringolo M, Diamanti T, Bourne Y, Marchot P, Martella G, Dini L, Pisani A, De Jaco A, Bonsi P. The neuroligins and the synaptic pathway in Autism Spectrum Disorder. Neurosci Biobehav Rev 2020; 119:37-51. [PMID: 32991906 DOI: 10.1016/j.neubiorev.2020.09.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/11/2020] [Accepted: 09/19/2020] [Indexed: 12/13/2022]
Abstract
The genetics underlying autism spectrum disorder (ASD) is complex and heterogeneous, and de novo variants are found in genes converging in functional biological processes. Neuronal communication, including trans-synaptic signaling involving two families of cell-adhesion proteins, the presynaptic neurexins and the postsynaptic neuroligins, is one of the most recurrently affected pathways in ASD. Given the role of these proteins in determining synaptic function, abnormal synaptic plasticity and failure to establish proper synaptic contacts might represent mechanisms underlying risk of ASD. More than 30 mutations have been found in the neuroligin genes. Most of the resulting residue substitutions map in the extracellular, cholinesterase-like domain of the protein, and impair protein folding and trafficking. Conversely, the stalk and intracellular domains are less affected. Accordingly, several genetic animal models of ASD have been generated, showing behavioral and synaptic alterations. The aim of this review is to discuss the current knowledge on ASD-linked mutations in the neuroligin proteins and their effect on synaptic function, in various brain areas and circuits.
Collapse
Affiliation(s)
- Laura Trobiani
- Dept. Biology and Biotechnology, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Maria Meringolo
- Lab. Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy; Dept. Systems Medicine, University Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Tamara Diamanti
- Dept. Biology and Biotechnology, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Yves Bourne
- Lab. "Architecture et Fonction des Macromolécules Biologiques", CNRS/Aix Marseille Univ, Faculté des Sciences - Campus Luminy, 163 Avenue de Luminy, 13288 Marseille cedex 09, France
| | - Pascale Marchot
- Lab. "Architecture et Fonction des Macromolécules Biologiques", CNRS/Aix Marseille Univ, Faculté des Sciences - Campus Luminy, 163 Avenue de Luminy, 13288 Marseille cedex 09, France
| | - Giuseppina Martella
- Lab. Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy; Dept. Systems Medicine, University Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Luciana Dini
- Dept. Biology and Biotechnology, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Antonio Pisani
- Lab. Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy; Dept. Systems Medicine, University Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Antonella De Jaco
- Dept. Biology and Biotechnology, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.
| | - Paola Bonsi
- Lab. Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy.
| |
Collapse
|
23
|
Kuhlmann N, Milnerwood AJ. A Critical LRRK at the Synapse? The Neurobiological Function and Pathophysiological Dysfunction of LRRK2. Front Mol Neurosci 2020; 13:153. [PMID: 32973447 PMCID: PMC7482583 DOI: 10.3389/fnmol.2020.00153] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/22/2020] [Indexed: 12/25/2022] Open
Abstract
Since the discovery of LRRK2 mutations causal to Parkinson's disease (PD) in the early 2000s, the LRRK2 protein has been implicated in a plethora of cellular processes in which pathogenesis could occur, yet its physiological function remains elusive. The development of genetic models of LRRK2 PD has helped identify the etiological and pathophysiological underpinnings of the disease, and may identify early points of intervention. An important role for LRRK2 in synaptic function has emerged in recent years, which links LRRK2 to other genetic forms of PD, most notably those caused by mutations in the synaptic protein α-synuclein. This point of convergence may provide useful clues as to what drives dysfunction in the basal ganglia circuitry and eventual death of substantia nigra (SN) neurons. Here, we discuss the evolution and current state of the literature placing LRRK2 at the synapse, through the lens of knock-out, overexpression, and knock-in animal models. We hope that a deeper understanding of LRRK2 neurobiology, at the synapse and beyond, will aid the eventual development of neuroprotective interventions for PD, and the advancement of useful treatments in the interim.
Collapse
Affiliation(s)
- Naila Kuhlmann
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Austen J Milnerwood
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
24
|
Li X, Yu B, Wang B, Bao L, Zhang B, Li H, Yu Z, Zhang T, Yang Y, Huang R, Wu Y, Li M. Multi-terminal ionic-gated low-power silicon nanowire synaptic transistors with dendritic functions for neuromorphic systems. NANOSCALE 2020; 12:16348-16358. [PMID: 32725043 DOI: 10.1039/d0nr03141k] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Neuromorphic computing systems have shown powerful capability in tasks, such as recognition, learning, classification and decision-making, which are both challenging and inefficient in using the traditional computation architecture. The key elements including synapses and neurons, and their feasible hardware implementation are essential for practical neuromorphic computing. However, most existing synaptic devices used to emulate functions of a single synapse and the synapse-based networks are more energy intensive and less sustainable than their biological counterparts. The dendritic functions such as integration of spatiotemporal signals and spike-frequency coding characteristics have not been well implemented in a single synaptic device and thus play an imperative role in future practical hardware-based spiking neural networks. Moreover, most emerging synaptic transistors are fabricated by nanofabrication processes without CMOS compatibility for further wafer-scale integration. Herein, we demonstrate a novel ionic-gated silicon nanowire synaptic field-effect transistor (IGNWFET) with low power consumption (<400 fJ per switching event) based on the standard CMOS process platform. For the first time, the dendritic integration and dual-synaptic dendritic computations (such as "Add" and "Subtraction") could be realized by processing frequency coded spikes using a single device. Meanwhile, multi-functional characteristics of artificial synapses including the short-term and long-term synaptic plasticity, paired pulse facilitation and high-pass filtering were also successfully demonstrated based on 40 nm wide IGNWFETs. The migration of ions in polymer electrolyte and trapping in high-k dielectric were also experimentally studied in-depth to understand the short-term plasticity and long-term plasticity. Combined with statistical uniformity across a 4-inch wafer, the comprehensive performance of IGNWFET demonstrates its potential application in future biologically emulated neuromorphic systems.
Collapse
Affiliation(s)
- Xiaokang Li
- Key Laboratory of Microelectronic Devices and Circuits (MOE), Institute of Microelectronics, Peking University, Beijing 100871, China.
| | - Bocheng Yu
- Key Laboratory of Microelectronic Devices and Circuits (MOE), Institute of Microelectronics, Peking University, Beijing 100871, China.
| | - Bowen Wang
- Key Laboratory of Microelectronic Devices and Circuits (MOE), Institute of Microelectronics, Peking University, Beijing 100871, China.
| | - Lin Bao
- Key Laboratory of Microelectronic Devices and Circuits (MOE), Institute of Microelectronics, Peking University, Beijing 100871, China.
| | - Baotong Zhang
- Key Laboratory of Microelectronic Devices and Circuits (MOE), Institute of Microelectronics, Peking University, Beijing 100871, China.
| | - Haixia Li
- Key Laboratory of Microelectronic Devices and Circuits (MOE), Institute of Microelectronics, Peking University, Beijing 100871, China.
| | - Zhizhen Yu
- Key Laboratory of Microelectronic Devices and Circuits (MOE), Institute of Microelectronics, Peking University, Beijing 100871, China.
| | - Teng Zhang
- Key Laboratory of Microelectronic Devices and Circuits (MOE), Institute of Microelectronics, Peking University, Beijing 100871, China.
| | - Yuancheng Yang
- Key Laboratory of Microelectronic Devices and Circuits (MOE), Institute of Microelectronics, Peking University, Beijing 100871, China.
| | - Ru Huang
- Key Laboratory of Microelectronic Devices and Circuits (MOE), Institute of Microelectronics, Peking University, Beijing 100871, China.
| | - Yanqing Wu
- Key Laboratory of Microelectronic Devices and Circuits (MOE), Institute of Microelectronics, Peking University, Beijing 100871, China. and Frontiers Science Center for Nano-optoelectronics, Peking University, Beijing, 100871, China
| | - Ming Li
- Key Laboratory of Microelectronic Devices and Circuits (MOE), Institute of Microelectronics, Peking University, Beijing 100871, China. and Frontiers Science Center for Nano-optoelectronics, Peking University, Beijing, 100871, China
| |
Collapse
|
25
|
Uchigashima M, Leung M, Watanabe T, Cheung A, Le T, Pallat S, Dinis ALM, Watanabe M, Kawasawa YI, Futai K. Neuroligin3 splice isoforms shape inhibitory synaptic function in the mouse hippocampus. J Biol Chem 2020; 295:8589-8595. [PMID: 32381505 DOI: 10.1074/jbc.ac120.012571] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/01/2020] [Indexed: 01/15/2023] Open
Abstract
Synapse formation is a dynamic process essential for the development and maturation of the neuronal circuitry in the brain. At the synaptic cleft, trans-synaptic protein-protein interactions are major biological determinants of proper synapse efficacy. The balance of excitatory and inhibitory synaptic transmission (E-I balance) stabilizes synaptic activity, and dysregulation of the E-I balance has been implicated in neurodevelopmental disorders, including autism spectrum disorders. However, the molecular mechanisms underlying the E-I balance remain to be elucidated. Here, using single-cell transcriptomics, immunohistochemistry, and electrophysiology approaches to murine CA1 pyramidal neurons obtained from organotypic hippocampal slice cultures, we investigate neuroligin (Nlgn) genes that encode a family of postsynaptic adhesion molecules known to shape excitatory and inhibitory synaptic function. We demonstrate that the NLGN3 protein differentially regulates inhibitory synaptic transmission in a splice isoform-dependent manner at hippocampal CA1 synapses. We also found that distinct subcellular localizations of the NLGN3 isoforms contribute to the functional differences observed among these isoforms. Finally, results from single-cell RNA-Seq analyses revealed that Nlgn1 and Nlgn3 are the major murine Nlgn genes and that the expression levels of the Nlgn splice isoforms are highly diverse in CA1 pyramidal neurons. Our results delineate isoform-specific effects of Nlgn genes on the E-I balance in the murine hippocampus.
Collapse
Affiliation(s)
- Motokazu Uchigashima
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA.,Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Ming Leung
- Department of Biochemistry and Molecular Biology and Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Takuya Watanabe
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Amy Cheung
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Timmy Le
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Sabine Pallat
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Alexandre Luis Marques Dinis
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Yuka Imamura Kawasawa
- Department of Biochemistry and Molecular Biology and Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA .,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Kensuke Futai
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
26
|
Ray PR, Wangzhou A, Ghneim N, Yousuf MS, Paige C, Tavares-Ferreira D, Mwirigi JM, Shiers S, Sankaranarayanan I, McFarland AJ, Neerukonda SV, Davidson S, Dussor G, Burton MD, Price TJ. A pharmacological interactome between COVID-19 patient samples and human sensory neurons reveals potential drivers of neurogenic pulmonary dysfunction. SSRN 2020:3581446. [PMID: 32714114 PMCID: PMC7366818 DOI: 10.2139/ssrn.3581446] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/04/2020] [Indexed: 11/15/2022]
Abstract
The SARS-CoV-2 virus infects cells of the airway and lungs in humans causing the disease COVID-19. This disease is characterized by cough, shortness of breath, and in severe cases causes pneumonia and acute respiratory distress syndrome (ARDS) which can be fatal. Bronchial alveolar lavage fluid (BALF) and plasma from mild and severe cases of COVID-19 have been profiled using protein measurements and bulk and single cell RNA sequencing. Onset of pneumonia and ARDS can be rapid in COVID-19, suggesting a potential neuronal involvement in pathology and mortality. We sought to quantify how immune cells might interact with sensory innervation of the lung in COVID-19 using published data from patients, existing RNA sequencing datasets from human dorsal root ganglion neurons and other sources, and a genome-wide ligand-receptor pair database curated for pharmacological interactions relevant for neuro-immune interactions. Our findings reveal a landscape of ligand-receptor interactions in the lung caused by SARS-CoV-2 viral infection and point to potential interventions to reduce the burden of neurogenic inflammation in COVID-19 disease. In particular, our work highlights opportunities for clinical trials with existing or under development rheumatoid arthritis and other (e.g. CCL2, CCR5 or EGFR inhibitors) drugs to treat high risk or severe COVID-19 cases.
Collapse
Affiliation(s)
- Pradipta R. Ray
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Andi Wangzhou
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Nizar Ghneim
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Muhammad S. Yousuf
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Candler Paige
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Diana Tavares-Ferreira
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Juliet M. Mwirigi
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Stephanie Shiers
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Ishwarya Sankaranarayanan
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Amelia J. McFarland
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Sanjay V. Neerukonda
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Steve Davidson
- University of Cincinnati, College of Medicine, Department of Anesthesiology
| | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Michael D. Burton
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Theodore J. Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| |
Collapse
|
27
|
Effect of Neuroligin1 and Neurexin1 on the Colonic Motility in a Mouse Model of Neuronal Intestinal Dysplasia. Gastroenterol Res Pract 2020; 2020:9818652. [PMID: 32184818 PMCID: PMC7059090 DOI: 10.1155/2020/9818652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 12/21/2019] [Accepted: 12/23/2019] [Indexed: 01/16/2023] Open
Abstract
Aim To investigate the expressions of neuroligin1 (NL1) and neurexin1 (NX1) in a mouse model of neuronal intestinal dysplasia (Tlx2−/− mice) and to explore their effects on colonic motility. Methods Immunohistochemistry staining was employed to explore the histological appearances of NL1, NX1, the presynaptic marker of glutamatergic synapses VGLUT1, and the subunit of NMDA receptors of NR1 in the colon of mice with or without Tlx2 mutation. Western blotting and qRT-PCR were performed to detect their relative expressions in the colon. Colonic motility was measured by a glass bead technique. Then, the Tlx2−/− mice were intervened by Huperzine A. Variations on expressions of NL1, NX1, VGLUT1, and NR1 and variations on colonic motility were measured. Additionally, serum concentrations of Glu were measured by ELISA. Results Immunohistochemistry staining reveals that NL1, NX1, VGLUT1, and NR1 were mainly concentrated in the myenteric plexus of ENS. Compared to those in WT and Tlx2+/- mice, expressions of NL1 and NX1 in colon of Tlx2−/− mice were upregulated with increased VGLUT1 and NR1 abundances and impaired colonic motility (P < 0.05). After intervention, the upregulated expressions of NL1 and NX1 were decreased with a correlated reduce of VGLUT1 and NR1 and a recovery of the impaired colonic motility (P < 0.05). After intervention, the upregulated expressions of NL1 and NX1 were decreased with a correlated reduce of VGLUT1 and NR1 and a recovery of the impaired colonic motility (P < 0.05). After intervention, the upregulated expressions of NL1 and NX1 were decreased with a correlated reduce of VGLUT1 and NR1 and a recovery of the impaired colonic motility ( Conclusion NL1 and NX1 are closely related to the colonic motility through their effects of targeting the formation of glutamatergic synapses and may be involved in the pathogenesis of NID. The variations of serum Glu seem to be a potential and less painful auxiliary measure for colonic motility and NID.
Collapse
|
28
|
Bhandari R, Paliwal JK, Kuhad A. Neuropsychopathology of Autism Spectrum Disorder: Complex Interplay of Genetic, Epigenetic, and Environmental Factors. ADVANCES IN NEUROBIOLOGY 2020; 24:97-141. [PMID: 32006358 DOI: 10.1007/978-3-030-30402-7_4] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Autism spectrum disorder (ASD) is a complex heterogeneous consortium of pervasive development disorders (PDD) which ranges from atypical autism, autism, and Asperger syndrome affecting brain in the developmental stage. This debilitating neurodevelopmental disorder results in both core as well as associated symptoms. Core symptoms observed in autistic patients are lack of social interaction, pervasive, stereotyped, and restricted behavior while the associated symptoms include irritability, anxiety, aggression, and several comorbid disorders.ASD is a polygenic disorder and is multifactorial in origin. Copy number variations (CNVs) of several genes that regulate the synaptogenesis and signaling pathways are one of the major factors responsible for the pathogenesis of autism. The complex integration of various CNVs cause mutations in the genes which code for molecules involved in cell adhesion, voltage-gated ion-channels, scaffolding proteins as well as signaling pathways (PTEN and mTOR pathways). These mutated genes are responsible for affecting synaptic transmission by causing plasticity dysfunction responsible, in turn, for the expression of ASD.Epigenetic modifications affecting DNA transcription and various pre-natal and post-natal exposure to a variety of environmental factors are also precipitating factors for the occurrence of ASD. All of these together cause dysregulation of glutamatergic signaling as well as imbalance in excitatory: inhibitory pathways resulting in glial cell activation and release of inflammatory mediators responsible for the aberrant social behavior which is observed in autistic patients.In this chapter we review and provide insight into the intricate integration of various genetic, epigenetic, and environmental factors which play a major role in the pathogenesis of this disorder and the mechanistic approach behind this integration.
Collapse
Affiliation(s)
- Ranjana Bhandari
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India
| | - Jyoti K Paliwal
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India
| | - Anurag Kuhad
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India.
| |
Collapse
|
29
|
Gomez G, Escande MV, Suarez LM, Rela L, Belforte JE, Moratalla R, Murer MG, Gershanik OS, Taravini IRE. Changes in Dendritic Spine Density and Inhibitory Perisomatic Connectivity onto Medium Spiny Neurons in L-Dopa-Induced Dyskinesia. Mol Neurobiol 2019; 56:6261-6275. [PMID: 30746639 DOI: 10.1007/s12035-019-1515-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/28/2019] [Indexed: 11/26/2022]
Abstract
Using bacterial artificial chromosome-double transgenic mice expressing tdTomato in D1 receptor-medium spiny neurons (MSNs) and enhanced green fluorescent protein in D2 receptor-MSNs, we have studied changes in spine density and perisomatic GABAergic boutons density in MSNs of both the D1R and D2R pathways, in an experimental model of parkinsonism (mouse injected with 6-hydroxydopamine in the medial forebrain bundle), both in the parkinsonian and dyskinetic condition induced by L-DOPA treatment. To assess changes in perisomatic GABAergic connectivity onto MSNs, we measured the number of contacts originated from parvalbumin (PV)-containing striatal "fast-spiking" interneurons (FSIs), the major component of a feed-forward inhibition mechanism that regulates spike timing in MSNs, in both cell types as well as the number of vesicular GABA transporter (VGAT) contacts. Furthermore, we determined changes in PV-immunoreactive cell density by PV immunolabeling combined with Wisteria floribunda agglutinin (WFA) labeling to detect FSI in a PV-independent manner. We also explored the differential expression of striatal activity-regulated cytoskeleton-associated protein (Arc) and c-Fos in both types of MSNs as a measure of neuronal activation. Our results confirm previous findings of major structural changes in dendritic spine density after nigrostriatal denervation, which are further modified in the dyskinetic condition. Moreover, the finding of differential modifications in perisomatic GABAergic connectivity and neuronal activation in MSNs suggests an attempt by the system to regain homeostasis after denervation and an imbalance between excitation and inhibition leading to the development of dyskinesia after exposure to L-DOPA.
Collapse
Affiliation(s)
- G Gomez
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto de Investigaciones Farmacológicas (ININFA), Laboratorio de Parkinson Experimental, CONICET - Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - M V Escande
- Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - L M Suarez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - L Rela
- Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - J E Belforte
- Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - R Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - M G Murer
- Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - O S Gershanik
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto de Investigaciones Farmacológicas (ININFA), Laboratorio de Parkinson Experimental, CONICET - Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - I R E Taravini
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina.
- Instituto de Investigaciones Farmacológicas (ININFA), Laboratorio de Parkinson Experimental, CONICET - Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina.
- Laboratorio de Neurobiología Experimental (LNE), CONICET - Facultad de Bromatología, Universidad Nacional de Entre Ríos, Gualeguaychú, Entre Ríos, Argentina.
| |
Collapse
|
30
|
Hannou L, Roy P, Ballester Roig MN, Mongrain V. Transcriptional control of synaptic components by the clock machinery. Eur J Neurosci 2019; 51:241-267. [DOI: 10.1111/ejn.14294] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/01/2018] [Accepted: 11/27/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Lydia Hannou
- Center for Advanced Research in Sleep Medicine and Research CenterHôpital du Sacré‐Cœur de Montréal (CIUSSS‐NIM) Montreal Quebec Canada
- Department of PsychiatryUniversité de Montréal Montreal Quebec Canada
| | - Pierre‐Gabriel Roy
- Center for Advanced Research in Sleep Medicine and Research CenterHôpital du Sacré‐Cœur de Montréal (CIUSSS‐NIM) Montreal Quebec Canada
- Department of NeuroscienceUniversité de Montréal Montreal Quebec Canada
| | - Maria Neus Ballester Roig
- Center for Advanced Research in Sleep Medicine and Research CenterHôpital du Sacré‐Cœur de Montréal (CIUSSS‐NIM) Montreal Quebec Canada
- Department of NeuroscienceUniversité de Montréal Montreal Quebec Canada
| | - Valérie Mongrain
- Center for Advanced Research in Sleep Medicine and Research CenterHôpital du Sacré‐Cœur de Montréal (CIUSSS‐NIM) Montreal Quebec Canada
- Department of NeuroscienceUniversité de Montréal Montreal Quebec Canada
| |
Collapse
|
31
|
Hirano T. Visualization of Exo- and Endocytosis of AMPA Receptors During Hippocampal Synaptic Plasticity Around Postsynaptic-Like Membrane Formed on Glass Surface. Front Cell Neurosci 2018; 12:442. [PMID: 30519162 PMCID: PMC6258823 DOI: 10.3389/fncel.2018.00442] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 11/05/2018] [Indexed: 11/13/2022] Open
Abstract
Regulation of exo- and endocytosis of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptor (AMPAR) plays a critical role in the expression of synaptic plasticity such as long-term potentiation (LTP) and long-term depression (LTD) at excitatory central synapses. Enhanced AMPAR exocytosis or endocytosis has been suggested to contribute to LTP or LTD, respectively. However, several unsettled fundamental questions have remained about AMPAR exo- and endocytosis in the basal condition and during synaptic plasticity: (1) Does the size of each exo- or endocytosis event, and/or do the frequencies of these events change during LTP or LTD? If they change, what are the time courses of the respective changes? (2) Where does the exo- or endocytosis preferentially occur in each condition: inside or in the vicinity of postsynaptic membrane, or in the extrasynaptic membrane? (3) Do different types of AMPAR, such as GluA1 homo-tetramer, GluA1/2 hetero-tetramer and GluA2/3 hetero-tetramer, show distinct exo- and endocytosis changes? To address these questions, we developed new methods to observe individual events of AMPAR exo- or endocytosis with a high signal to noise (SN) ratio in a culture preparation using total internal reflection fluorescence microscopy (TIRFM). In these studies, hippocampal neurons were cultured on a neurexin (NRX)-coated glass coverslip, which induced formation of postsynaptic-like membrane (PSLM) directly on the glass surface. Then, a super-ecliptic pHluorin (SEP)-tagged AMPAR subunit such as GluA1 (GluA1-SEP) was expressed in neurons and its fluorescence changes during LTP induced by high frequency electrical field stimulation were observed with TIRFM, which showed different time courses of exocytosis changes of GluA1-, GluA2-, or GluA3-SEP in and around PSLM. In addition, a new method to detect individual endocytosis events of AMPAR was developed by combining TIFRM observation of GluA-SEP around PSLM with a rapid extracellular pH exchange method using a U-tube. Recent results on exo- and endocytosis changes of GluA-SEP during N-methyl-D-aspartate (NMDA)-induced LTD suggested that suppression of AMPAR exocytosis rather than enhancement of AMPAR endocytosis primarily contributes to LTD expression, although the NMDA application transiently enhances clathrin-dependent endocytosis of GluA1-containing AMPAR.
Collapse
Affiliation(s)
- Tomoo Hirano
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan
| |
Collapse
|
32
|
Gorlewicz A, Kaczmarek L. Pathophysiology of Trans-Synaptic Adhesion Molecules: Implications for Epilepsy. Front Cell Dev Biol 2018; 6:119. [PMID: 30298130 PMCID: PMC6160742 DOI: 10.3389/fcell.2018.00119] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 08/30/2018] [Indexed: 12/31/2022] Open
Abstract
Chemical synapses are specialized interfaces between neurons in the brain that transmit and modulate information, thereby integrating cells into multiplicity of interacting neural circuits. Cell adhesion molecules (CAMs) might form trans-synaptic complexes that are crucial for the appropriate identification of synaptic partners and further for the establishment, properties, and dynamics of synapses. When affected, trans-synaptic adhesion mechanisms play a role in synaptopathies in a variety of neuropsychiatric disorders including epilepsy. This review recapitulates current understanding of trans-synaptic interactions in pathophysiology of interneuronal connections. In particular, we discuss here the possible implications of trans-synaptic adhesion dysfunction for epilepsy.
Collapse
Affiliation(s)
- Adam Gorlewicz
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
33
|
Ali Rodriguez R, Joya C, Hines RM. Common Ribs of Inhibitory Synaptic Dysfunction in the Umbrella of Neurodevelopmental Disorders. Front Mol Neurosci 2018; 11:132. [PMID: 29740280 PMCID: PMC5928253 DOI: 10.3389/fnmol.2018.00132] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/03/2018] [Indexed: 01/06/2023] Open
Abstract
The term neurodevelopmental disorder (NDD) is an umbrella term used to group together a heterogeneous class of disorders characterized by disruption in cognition, emotion, and behavior, early in the developmental timescale. These disorders are heterogeneous, yet they share common behavioral symptomatology as well as overlapping genetic contributors, including proteins involved in the formation, specialization, and function of synaptic connections. Advances may arise from bridging the current knowledge on synapse related factors indicated from both human studies in NDD populations, and in animal models. Mounting evidence has shown a link to inhibitory synapse formation, specialization, and function among Autism, Angelman, Rett and Dravet syndromes. Inhibitory signaling is diverse, with numerous subtypes of inhibitory interneurons, phasic and tonic modes of inhibition, and the molecular and subcellular diversity of GABAA receptors. We discuss common ribs of inhibitory synapse dysfunction in the umbrella of NDD, highlighting alterations in the developmental switch to inhibitory GABA, dysregulation of neuronal activity patterns by parvalbumin-positive interneurons, and impaired tonic inhibition. Increasing our basic understanding of inhibitory synapses, and their role in NDDs is likely to produce significant therapeutic advances in behavioral symptom alleviation for interrelated NDDs.
Collapse
Affiliation(s)
- Rachel Ali Rodriguez
- Neuroscience Emphasis, Department of Psychology, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Christina Joya
- Neuroscience Emphasis, Department of Psychology, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Rochelle M Hines
- Neuroscience Emphasis, Department of Psychology, University of Nevada, Las Vegas, Las Vegas, NV, United States
| |
Collapse
|
34
|
Zhao ZH, Zheng G, Wang T, Du KJ, Han X, Luo WJ, Shen XF, Chen JY. Low-level Gestational Lead Exposure Alters Dendritic Spine Plasticity in the Hippocampus and Reduces Learning and Memory in Rats. Sci Rep 2018; 8:3533. [PMID: 29476096 PMCID: PMC5824819 DOI: 10.1038/s41598-018-21521-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 02/05/2018] [Indexed: 11/24/2022] Open
Abstract
Lead (Pb) is known to impair children's cognitive function. It has been previously shown that developmental Pb exposure alters dendritic spine formation in hippocampal pyramidal neurons. However, the underlying mechanism has not yet been defined. In this study, a low-level gestational Pb exposure (GLE) rat model was employed to investigate the impact of Pb on the spine density of the hippocampal pyramidal neurons and its regulatory mechanism. Pb exposure resulted in impaired performance of the rats in the Morris water maze tasks, and in decreased EPSC amplitudes in hippocampal CA3-CA1 regions. With a 3D reconstruction by the Imaris software, the results from Golgi staining showed that the spine density in the CA1 region was reduced in the Pb-exposed rats in a dose-dependent manner. Decreased spine density was also observed in cultured hippocampal neurons following the Pb treatment. Furthermore, the expression level of NLGN1, a postsynaptic protein that mediates synaptogenesis, was significantly decreased following the Pb exposure both in vivo and in vitro. Up-regulation of NLGN1 in cultured primary neurons partially attenuated the impact of Pb on the spine density. Taken together, our resultssuggest that Pb exposure alters spine plasticity in the developing hippocampus by down-regulating NLGN1 protein levels.
Collapse
Affiliation(s)
- Zai-Hua Zhao
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, No 169 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Gang Zheng
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, No 169 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Tao Wang
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, No 169 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Ke-Jun Du
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, No 169 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Xiao Han
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, No 169 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Wen-Jing Luo
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, No 169 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Xue-Feng Shen
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, No 169 of West Changle Road, Xi'an, Shaanxi, 710032, China.
| | - Jing-Yuan Chen
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, No 169 of West Changle Road, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
35
|
Jaramillo TC, Escamilla CO, Liu S, Peca L, Birnbaum SG, Powell CM. Genetic background effects in Neuroligin-3 mutant mice: Minimal behavioral abnormalities on C57 background. Autism Res 2017; 11:234-244. [PMID: 29028156 DOI: 10.1002/aur.1857] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/09/2017] [Indexed: 11/11/2022]
Abstract
Neuroligin-3 (NLGN3) is a postsynaptic cell adhesion protein that interacts with presynaptic ligands including neurexin-1 (NRXN1) [Ichtchenko et al., Journal of Biological Chemistry, 271, 2676-2682, 1996]. Mice harboring a mutation in the NLGN3 gene (NL3R451C) mimicking a mutation found in two brothers with autism spectrum disorder (ASD) were previously generated and behaviorally phenotyped for autism-related behaviors. In these NL3R451C mice generated and tested on a hybrid C57BL6J/129S2/SvPasCrl background, we observed enhanced spatial memory and reduced social interaction [Tabuchi et al., Science, 318, 71-76, 2007]. Curiously, an independently generated second line of mice harboring the same mutation on a C57BL6J background exhibited minimal aberrant behavior, thereby providing apparently discrepant results. To investigate the origin of the discrepancy, we previously replicated the original findings of Tabuchi et al. by studying the same NL3R451C mutation on a pure 129S2/SvPasCrl genetic background. Here we complete the behavioral characterization of the NL3R451C mutation on a pure C57BL6J genetic background to determine if background genetics play a role in the discrepant behavioral outcomes involving NL3R451C mice. NL3R451C mutant mice on a pure C57BL6J background did not display spatial memory enhancements or social interaction deficits. We only observed a decreased startle response and mildly increased locomotor activity in these mice suggesting that background genetics influences behavioral outcomes involving the NL3R451C mutation. Autism Res 2018, 11: 234-244. © 2017 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY Behavioral symptoms of autism can be highly variable, even in cases that involve identical genetic mutations. Previous studies in mice with a mutation of the Neuroligin-3 gene showed enhanced learning and social deficits. We replicated these findings on the same and different genetic backgrounds. In this study, however, the same mutation in mice on a different genetic background did not reproduce our previous findings. Our results suggest that genetic background influences behavioral symptoms of this autism-associated mutation.
Collapse
Affiliation(s)
- Thomas C Jaramillo
- Department of Neurology & Neurotherapeutics, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8813
| | - Christine Ochoa Escamilla
- Department of Neurology & Neurotherapeutics, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8813.,Neuroscience Graduate Program, The University of Texas Southwestern Medical Center, Dallas, TX, 75390
| | - Shunan Liu
- Department of Neurology & Neurotherapeutics, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8813
| | - Lauren Peca
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, 75390-9170
| | - Shari G Birnbaum
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, 75390-9170
| | - Craig M Powell
- Department of Neurology & Neurotherapeutics, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8813.,Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, 75390-9170.,Neuroscience Graduate Program, The University of Texas Southwestern Medical Center, Dallas, TX, 75390.,Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX, 75390
| |
Collapse
|
36
|
Unbalance between Excitation and Inhibition in Phenylketonuria, a Genetic Metabolic Disease Associated with Autism. Int J Mol Sci 2017; 18:ijms18050941. [PMID: 28468253 PMCID: PMC5454854 DOI: 10.3390/ijms18050941] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/20/2017] [Accepted: 04/23/2017] [Indexed: 02/06/2023] Open
Abstract
Phenylketonuria (PKU) is the most common genetic metabolic disease with a well-documented association with autism spectrum disorders. It is characterized by the deficiency of the phenylalanine hydroxylase activity, causing plasmatic hyperphenylalaninemia and variable neurological and cognitive impairments. Among the potential pathophysiological mechanisms implicated in autism spectrum disorders is the excitation/inhibition (E/I) imbalance which might result from alterations in excitatory/inhibitory synapse development, synaptic transmission and plasticity, downstream signalling pathways, and intrinsic neuronal excitability. Here, we investigated functional and molecular alterations in the prefrontal cortex (pFC) of BTBR-Pahenu2 (ENU2) mice, the animal model of PKU. Our data show higher frequency of inhibitory transmissions and significant reduced frequency of excitatory transmissions in the PKU-affected mice in comparison to wild type. Moreover, in the pFC of ENU2 mice, we reported higher levels of the post-synaptic cell-adhesion proteins neuroligin1 and 2. Altogether, our data point toward an imbalance in the E/I neurotransmission favouring inhibition in the pFC of ENU2 mice, along with alterations of the molecular components involved in the organization of cortical synapse. In addition to being the first evidence of E/I imbalance within cortical areas of a mouse model of PKU, our study provides further evidence of E/I imbalance in animal models of pathology associated with autism spectrum disorders.
Collapse
|
37
|
Neural plasticity and network remodeling: From concepts to pathology. Neuroscience 2017; 344:326-345. [PMID: 28069532 DOI: 10.1016/j.neuroscience.2016.12.048] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/24/2016] [Accepted: 12/27/2016] [Indexed: 11/22/2022]
Abstract
Neuroplasticity has been subject to a great deal of research in the last century. Recently, significant emphasis has been placed on the global effect of localized plastic changes throughout the central nervous system, and on how these changes integrate in a pathological context. Specifically, alterations of network functionality have been described in various pathological contexts to which corresponding structural alterations have been proposed. However, considering the amount of literature and the different pathological contexts, an integration of this information is still lacking. In this paper we will review the concepts of neural plasticity as well as their repercussions on network remodeling and provide a possible explanation to how these two concepts relate to each other. We will further examine how alterations in different pathological contexts may relate to each other and will discuss the concept of plasticity diseases, its models and implications.
Collapse
|
38
|
Nie J, Yang X. Modulation of Synaptic Plasticity by Exercise Training as a Basis for Ischemic Stroke Rehabilitation. Cell Mol Neurobiol 2017; 37:5-16. [PMID: 26910247 DOI: 10.1007/s10571-016-0348-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/11/2016] [Indexed: 12/23/2022]
Abstract
In recent years, rehabilitation of ischemic stroke draws more and more attention in the world, and has been linked to changes of synaptic plasticity. Exercise training improves motor function of ischemia as well as cognition which is associated with formation of learning and memory. The molecular basis of learning and memory might be synaptic plasticity. Research has therefore been conducted in an attempt to relate effects of exercise training to neuroprotection and neurogenesis adjacent to the ischemic injury brain. The present paper reviews the current literature addressing this question and discusses the possible mechanisms involved in modulation of synaptic plasticity by exercise training. This review shows the pathological process of synaptic dysfunction in ischemic roughly and then discusses the effects of exercise training on scaffold proteins and regulatory protein expression. The expression of scaffold proteins generally increased after training, but the effects on regulatory proteins were mixed. Moreover, the compositions of postsynaptic receptors were changed and the strength of synaptic transmission was enhanced after training. Finally, the recovery of cognition is critically associated with synaptic remodeling in an injured brain, and the remodeling occurs through a number of local regulations including mRNA translation, remodeling of cytoskeleton, and receptor trafficking into and out of the synapse. We do provide a comprehensive knowledge of synaptic plasticity enhancement obtained by exercise training in this review.
Collapse
Affiliation(s)
- Jingjing Nie
- Department of Neurology, Xiang Ya Hospital, Central South University, Xiang Ya Road 87, Changsha, 410008, Hunan, China
| | - Xiaosu Yang
- Department of Neurology, Xiang Ya Hospital, Central South University, Xiang Ya Road 87, Changsha, 410008, Hunan, China.
| |
Collapse
|
39
|
Gonzalez-Lozano MA, Klemmer P, Gebuis T, Hassan C, van Nierop P, van Kesteren RE, Smit AB, Li KW. Dynamics of the mouse brain cortical synaptic proteome during postnatal brain development. Sci Rep 2016; 6:35456. [PMID: 27748445 PMCID: PMC5066275 DOI: 10.1038/srep35456] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 09/28/2016] [Indexed: 01/04/2023] Open
Abstract
Development of the brain involves the formation and maturation of numerous synapses. This process requires prominent changes of the synaptic proteome and potentially involves thousands of different proteins at every synapse. To date the proteome analysis of synapse development has been studied sparsely. Here, we analyzed the cortical synaptic membrane proteome of juvenile postnatal days 9 (P9), P15, P21, P27, adolescent (P35) and different adult ages P70, P140 and P280 of C57Bl6/J mice. Using a quantitative proteomics workflow we quantified 1560 proteins of which 696 showed statistically significant differences over time. Synaptic proteins generally showed increased levels during maturation, whereas proteins involved in protein synthesis generally decreased in abundance. In several cases, proteins from a single functional molecular entity, e.g., subunits of the NMDA receptor, showed differences in their temporal regulation, which may reflect specific synaptic development features of connectivity, strength and plasticity. SNARE proteins, Snap 29/47 and Stx 7/8/12, showed higher expression in immature animals. Finally, we evaluated the function of Cxadr that showed high expression levels at P9 and a fast decline in expression during neuronal development. Knock down of the expression of Cxadr in cultured primary mouse neurons revealed a significant decrease in synapse density.
Collapse
Affiliation(s)
- Miguel A Gonzalez-Lozano
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Patricia Klemmer
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Titia Gebuis
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Chopie Hassan
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Pim van Nierop
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Ronald E van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Genome-wide gene-based analysis suggests an association between Neuroligin 1 (NLGN1) and post-traumatic stress disorder. Transl Psychiatry 2016; 6:e820. [PMID: 27219346 PMCID: PMC5070067 DOI: 10.1038/tp.2016.69] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 03/13/2016] [Accepted: 03/20/2016] [Indexed: 01/20/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) develops in only some people following trauma exposure, but the mechanisms differentially explaining risk versus resilience remain largely unknown. PTSD is heritable but candidate gene studies and genome-wide association studies (GWAS) have identified only a modest number of genes that reliably contribute to PTSD. New gene-based methods may help identify additional genes that increase risk for PTSD development or severity. We applied gene-based testing to GWAS data from the Grady Trauma Project (GTP), a primarily African American cohort, and identified two genes (NLGN1 and ZNRD1-AS1) that associate with PTSD after multiple test correction. Although the top SNP from NLGN1 did not replicate, we observed gene-based replication of NLGN1 with PTSD in the Drakenstein Child Health Study (DCHS) cohort from Cape Town. NLGN1 has previously been associated with autism, and it encodes neuroligin 1, a protein involved in synaptogenesis, learning, and memory. Within the GTP dataset, a single nucleotide polymorphism (SNP), rs6779753, underlying the gene-based association, associated with the intermediate phenotypes of higher startle response and greater functional magnetic resonance imaging activation of the amygdala, orbitofrontal cortex, right thalamus and right fusiform gyrus in response to fearful faces. These findings support a contribution of the NLGN1 gene pathway to the neurobiological underpinnings of PTSD.
Collapse
|
41
|
Neuroligin-2 Expression in the Prefrontal Cortex is Involved in Attention Deficits Induced by Peripubertal Stress. Neuropsychopharmacology 2016; 41:751-61. [PMID: 26152839 PMCID: PMC4707821 DOI: 10.1038/npp.2015.200] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 06/28/2015] [Accepted: 06/28/2015] [Indexed: 12/11/2022]
Abstract
Emerging evidence indicates that attention deficits, which are frequently observed as core symptoms of neuropsychiatric disorders, may be elicited by early life stress. However, the mechanisms mediating these stress effects remain unknown. The prefrontal cortex (PFC) has been implicated in the regulation of attention, including dysfunctions in GABAergic transmission, and it is highly sensitive to stress. Here, we investigated the involvement of neuroligin-2 (NLGN-2), a synaptic cell adhesion molecule involved in the stabilization and maturation of GABAergic synapses, in the PFC in the link between stress and attention deficits. First, we established that exposure of rats to stress during the peripubertal period impairs attention in the five-choice serial reaction time task and results in reductions in the GABA-synthesizing enzyme glutamic acid decarboxylase in different PFC subregions (ie, prelimbic (PL), infralimbic, and medial and ventral orbitofrontal (OFC) cortex) and in NLGN-2 in the PL cortex. In peripubertally stressed animals, NLGN-2 expression in the PL and OFC cortex correlated with attention measurements. Subsequently, we found that adeno-associated virus-induced rescue of NLGN-2 in the PFC reverses the stress-induced attention deficits regarding omitted trials. Therefore, our findings highlight peripuberty as a period that is highly vulnerable to stress, leading to the development of attention deficits and a dysfunction in the PFC GABAergic system and NLGN-2 expression. Furthermore, NLGN-2 is underscored as a promising target to treat stress-induced cognitive alterations, and in particular attentional deficits as manifested by augmented omissions in a continuous performance task.
Collapse
|
42
|
Przekwas A, Somayaji MR, Gupta RK. Synaptic Mechanisms of Blast-Induced Brain Injury. Front Neurol 2016; 7:2. [PMID: 26834697 PMCID: PMC4720734 DOI: 10.3389/fneur.2016.00002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 01/04/2016] [Indexed: 01/08/2023] Open
Abstract
Blast wave-induced traumatic brain injury (TBI) is one of the most common injuries to military personnel. Brain tissue compression/tension due to blast-induced cranial deformations and shear waves due to head rotation may generate diffuse micro-damage to neuro-axonal structures and trigger a cascade of neurobiological events culminating in cognitive and neurodegenerative disorders. Although diffuse axonal injury is regarded as a signature wound of mild TBI (mTBI), blast loads may also cause synaptic injury wherein neuronal synapses are stretched and sheared. This synaptic injury may result in temporary disconnect of the neural circuitry and transient loss in neuronal communication. We hypothesize that mTBI symptoms such as loss of consciousness or dizziness, which start immediately after the insult, could be attributed to synaptic injury. Although empirical evidence is beginning to emerge; the detailed mechanisms underlying synaptic injury are still elusive. Coordinated in vitro-in vivo experiments and mathematical modeling studies can shed light into the synaptic injury mechanisms and their role in the potentiation of mTBI symptoms.
Collapse
Affiliation(s)
- Andrzej Przekwas
- Computational Medicine and Biology Division, CFD Research Corporation, Huntsville, AL, USA
| | | | - Raj K. Gupta
- Department of Defense Blast Injury Research Program Coordinating Office, U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, USA
| |
Collapse
|
43
|
Speed HE, Masiulis I, Gibson JR, Powell CM. Increased Cortical Inhibition in Autism-Linked Neuroligin-3R451C Mice Is Due in Part to Loss of Endocannabinoid Signaling. PLoS One 2015; 10:e0140638. [PMID: 26469287 PMCID: PMC4607423 DOI: 10.1371/journal.pone.0140638] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 09/29/2015] [Indexed: 11/19/2022] Open
Abstract
A single, maternally inherited, X-linked point mutation leading to an arginine to cysteine substitution at amino acid 451 (R451C) of Neuroligin 3 (NLGN3R451C) is a likely cause of autism in two brothers. Knockin mice expressing the Nlgn3R451C mutation in place of wild-type Nlgn3 demonstrate increased inhibitory synaptic strength in somatosensory cortex, resulting in an excitatory/inhibitory (E/I) imbalance that is potentially relevant for autism-associated behavioral deficits characteristic of these mice. We have replicated the increase in evoked inhibitory postsynaptic currents (eIPSCs) onto layer II/III cortical pyramidal neurons. We also find that increased frequency of spontaneous mIPSCs in Nlgn3R451C mice occurs in the absence of action potential-driven transmission. This suggests the E/I imbalance is due to changes at the synapse level, as opposed to the network level. Next, we use paired whole-cell recordings in an attempt to identify specific interneuron subtypes affected by the Nlgn3R451C mutation. Curiously, we observe no change in the amplitude of cell-to-cell, unitary IPSCs (uIPSCs) from parvalbumin-positive (PV) or somatostatin-positive (SOM) interneurons onto pyramidal neurons. We also observe no change in the number or density of PV and SOM interneurons in LII/III of somatosensory cortex. This effectively rules out a role for these particular interneurons in the increased inhibitory synaptic transmission, pointing to perhaps alternative interneuron subtypes. Lastly, impaired endocannabinoid signaling has been implicated in hippocampal synaptic dysfunction in Nlgn3R451C mice, but has not been investigated at cortical synapses. We find that bath application of the CB1 antagonist, AM 251 in WT mice eliminates the Nlgn3R451C increase in eIPSC amplitude and mIPSC frequency, indicating that increased inhibitory transmission in mutant mice is due, at least in part, to a loss of endocannabinoid signaling through CB1 receptors likely acting at interneurons other than PV or SOM.
Collapse
Affiliation(s)
- Haley E. Speed
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Irene Masiulis
- Quantitative Morphology Core, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Jay R. Gibson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Craig M. Powell
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Psychiatry and Neuroscience Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail:
| |
Collapse
|
44
|
Wang J, Du H, Mou YR, Niu JY, Zhang WT, Yang HC, Li AW. Abundance and significance of neuroligin-1 and glutamate in Hirschsprung’s disease. World J Gastroenterol 2015; 21:7172-7180. [PMID: 26109803 PMCID: PMC4476878 DOI: 10.3748/wjg.v21.i23.7172] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 02/20/2015] [Accepted: 04/09/2015] [Indexed: 02/07/2023] Open
Abstract
AIM: To investigate the abundance and potential diagnostic significance of neuroligin-1 and glutamate (Glu) in Hirschsprung’s disease (HSCR).
METHODS: Ninety children with HSCR and 50 children without HSCR matched for similar nutritional status, age and basal metabolic index were studied. The expression and localization of neuroligin-1 and Glu were assessed using double-labeling immunofluorescence staining of longitudinal muscles with adherent myenteric plexus from the surgically excised colon of children with HSCR. Western blot analysis, quantitative real-time PCR (qRT-PCR) and immunohistochemistry were performed to evaluate the abundance of neuroligin-1 and Glu in different HSCR-affected segments (ganglionic, transitional, and aganglionic segments). Enzyme-linked immunosorbent assay (ELISA) was used to detect and compare serum Glu levels in the long-segment HSCR, short-segment HSCR and non-HSCR samples.
RESULTS: Neuroligin-1 and Glu were co-expressed highest to lowest in the ganglionic, transitional and aganglionic segments based on Western blot (neuroligin-1: 0.177 ± 0.008 vs 0.101 ± 0.006, 0.177 ± 0.008 vs 0.035 ± 0.005, and 0.101 ± 0.006 vs 0.035 ± 0.005, P < 0.005; Glu: 0.198 ± 0.006 vs 0.115 ± 0.008, 0.198 ± 0.006 vs 0.040 ± 0.003, and 0.115 ± 0.008 vs 0.040 ± 0.003, P < 0.005) and qRT-PCR (neuroligin-1: 9.58 × 10-5± 9.94 × 10-6vs 2.49 × 10-5± 1.38 × 10-6, 9.58 × 10-5± 9.94 × 10-6vs 7.17 × 10-6 ± 1.12 × 10-6, and 2.49 × 10-5± 1.38 × 10-6vs 7.17 × 10-6± 1.12 × 10-6, P < 0.005). Serum Glu level was the highest to lowest in the non-HSCR, short-type HSCR and long-type HSCR samples based on ELISA (in nmol/μL, 0.93 ± 0.31 vs 0.57 ± 0.25, 0.93 ± 0.31 vs 0.23 ± 0.16, and 0.57 ± 0.25 vs 0.23 ± 0.16, P < 0.005).
CONCLUSION: Neuroligin-1 and Glu may represent new markers of ganglion cells, whose expression may correlate with the pathogenesis, diagnosis, differential diagnosis or classification of HSCR.
Collapse
|
45
|
High B, Cole AA, Chen X, Reese TS. Electron microscopic tomography reveals discrete transcleft elements at excitatory and inhibitory synapses. Front Synaptic Neurosci 2015; 7:9. [PMID: 26113817 PMCID: PMC4461817 DOI: 10.3389/fnsyn.2015.00009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/28/2015] [Indexed: 12/24/2022] Open
Abstract
Electron microscopy has revealed an abundance of material in the clefts of synapses in the mammalian brain, and the biochemical and functional characteristics of proteins occupying synaptic clefts are well documented. However, the detailed spatial organization of the proteins in the synaptic clefts remains unclear. Electron microscope tomography provides a way to delineate and map the proteins spanning the synaptic cleft because freeze substitution preserves molecular details with sufficient contrast to visualize individual cleft proteins. Segmentation and rendering of electron dense material connected across the cleft reveals discrete structural elements that are readily classified into five types at excitatory synapses and four types at inhibitory synapses. Some transcleft elements resemble shapes and sizes of known proteins and could represent single dimers traversing the cleft. Some of the types of cleft elements at inhibitory synapses roughly matched the structure and proportional frequency of cleft elements at excitatory synapses, but the patterns of deployments in the cleft are quite different. Transcleft elements at excitatory synapses were often evenly dispersed in clefts of uniform (18 nm) width but some types show preference for the center or edges of the cleft. Transcleft elements at inhibitory synapses typically were confined to a peripheral region of the cleft where it narrowed to only 6 nm wide. Transcleft elements in both excitatory and inhibitory synapses typically avoid places where synaptic vesicles attach to the presynaptic membrane. These results illustrate that elements spanning synaptic clefts at excitatory and inhibitory synapses consist of distinct structures arranged by type in a specific but different manner at excitatory and inhibitory synapses.
Collapse
Affiliation(s)
- Brigit High
- Laboratory of Neurobiology, National Institute of Neurological Disorders and Strokes, National Institutes of Health Bethesda, MD, USA
| | - Andy A Cole
- Laboratory of Neurobiology, National Institute of Neurological Disorders and Strokes, National Institutes of Health Bethesda, MD, USA ; Department of Cell and Molecular Biology, Northwestern University Chicago, IL, USA
| | - Xiaobing Chen
- Laboratory of Neurobiology, National Institute of Neurological Disorders and Strokes, National Institutes of Health Bethesda, MD, USA
| | - Thomas S Reese
- Laboratory of Neurobiology, National Institute of Neurological Disorders and Strokes, National Institutes of Health Bethesda, MD, USA
| |
Collapse
|
46
|
Kohl C, Wang XD, Grosse J, Fournier C, Harbich D, Westerholz S, Li JT, Bacq A, Sippel C, Hausch F, Sandi C, Schmidt MV. Hippocampal neuroligin-2 links early-life stress with impaired social recognition and increased aggression in adult mice. Psychoneuroendocrinology 2015; 55:128-43. [PMID: 25765754 DOI: 10.1016/j.psyneuen.2015.02.016] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 02/19/2015] [Accepted: 02/19/2015] [Indexed: 11/17/2022]
Abstract
Early-life stress is a key risk factor for the development of neuropsychiatric disorders later in life. Neuronal cell adhesion molecules have been strongly implicated in the pathophysiology of psychiatric disorders and in modulating social behaviors associated with these diseases. Neuroligin-2 is a synaptic cell adhesion molecule, located at the postsynaptic membrane of inhibitory GABAergic synapses, and is involved in synaptic stabilization and maturation. Alterations in neuroligin-2 expression have previously been associated with changes in social behavior linked to psychiatric disorders, including schizophrenia and autism. In this study, we show that early-life stress, induced by limited nesting and bedding material, leads to impaired social recognition and increased aggression in adult mice, accompanied by increased expression levels of hippocampal neuroligin-2. Viral overexpression of hippocampal neuroligin-2 in adulthood mimics early-life stress-induced alterations in social behavior and social cognition. Moreover, viral knockdown of neuroligin-2 in the adult hippocampus attenuates the early-life stress-induced behavioral changes. Our results highlight the importance of neuroligin-2 in mediating early-life stress effects on social behavior and social cognition and its promising role as a novel therapeutic target for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Christine Kohl
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, EPFL, 1015 Lausanne, Switzerland; Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, 80804 Munich, Germany
| | - Xiao-Dong Wang
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, 80804 Munich, Germany; Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, 310058 Hangzhou, China
| | - Jocelyn Grosse
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, EPFL, 1015 Lausanne, Switzerland
| | - Céline Fournier
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, EPFL, 1015 Lausanne, Switzerland
| | - Daniela Harbich
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, 80804 Munich, Germany
| | - Sören Westerholz
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, 80804 Munich, Germany
| | - Ji-Tao Li
- Institute of Mental Health, Peking University, No. 51 Hua Yuan Bei Road, 100191 Beijing, China
| | - Alexandre Bacq
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, EPFL, 1015 Lausanne, Switzerland
| | - Claudia Sippel
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, 80804 Munich, Germany
| | - Felix Hausch
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, 80804 Munich, Germany
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, EPFL, 1015 Lausanne, Switzerland
| | - Mathias V Schmidt
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, 80804 Munich, Germany.
| |
Collapse
|
47
|
Park SW, Lee JG, Seo MK, Cho HY, Lee CH, Lee JH, Lee BJ, Baek JH, Seol W, Kim YH. Effects of mood-stabilizing drugs on dendritic outgrowth and synaptic protein levels in primary hippocampal neurons. Bipolar Disord 2015; 17:278-90. [PMID: 25307211 DOI: 10.1111/bdi.12262] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 07/23/2014] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Mood-stabilizing drugs, such as lithium (Li) and valproate (VPA), are widely used for the treatment of bipolar disorder, a disease marked by recurrent episodes of mania and depression. Growing evidence suggests that Li exerts neurotrophic and neuroprotective effects, leading to an increase in neural plasticity. The present study investigated whether other mood-stabilizing drugs produce similar effects in primary hippocampal neurons. METHODS The effects of the mood-stabilizing drugs Li, VPA, carbamazepine (CBZ), and lamotrigine (LTG) on hippocampal dendritic outgrowth were examined. Western blotting analysis was used to measure the expression of synaptic proteins - that is, brain-derived neurotrophic factor (BDNF), postsynaptic density protein-95 (PSD-95), neuroligin 1 (NLG1), β-neurexin, and synaptophysin (SYP). To determine neuroprotective effects, we used a B27-deprivation cytotoxicity model which causes hippocampal cell death upon removal of B27 from the culture medium. RESULTS Li (0.5-2.0 mM), VPA (0.5-2.0 mM), CBZ (0.01-0.10 mM), and LTG (0.01-0.10 mM) significantly increased dendritic outgrowth. The neurotrophic effect of Li and VPA was blocked by inhibition of phosphatidylinositol 3-kinase, extracellular signal-regulated kinase, and protein kinase A signaling; the effects of CBZ and LTG were not affected by inhibition of these signaling pathways. Li, VPA, and CBZ prevented B27 deprivation-induced decreases in BDNF, PSD-95, NLG1, β-neurexin, and SYP levels, whereas LTG did not. CONCLUSIONS These results suggest that Li, VPA, CBZ, and LTG exert neurotrophic effects by promoting dendritic outgrowth; however, the mechanism of action differs. Furthermore, certain mood-stabilizing drugs may exert neuroprotective effects by enhancing synaptic protein levels against cytotoxicity in hippocampal cultures.
Collapse
Affiliation(s)
- Sung Woo Park
- Paik Institute for Clinical Research, Inje University, Busan, Korea; Department of Health Science and Technology, Graduate School of Inje University, Busan, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Calahorro F, Holden-Dye L, O'Connor V. Analysis of splice variants for the C. elegans orthologue of human neuroligin reveals a developmentally regulated transcript. Gene Expr Patterns 2015; 17:69-78. [PMID: 25726726 DOI: 10.1016/j.gep.2015.02.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 02/17/2015] [Accepted: 02/19/2015] [Indexed: 11/20/2022]
Abstract
Neuroligins are synaptic adhesion molecules and important determinants of synaptic function. They are expressed at postsynaptic sites and involved in synaptic organization through key extracellular and intracellular protein interactions. They undergo trans-synaptic interaction with presynaptic neurexins. Distinct neuroligins use differences in their intracellular domains to selectively recruit synaptic scaffolds and this plays an important role in how they encode specialization of synaptic function. Several levels of regulation including gene expression, splicing, protein translation and processing regulate the expression of neuroligin function. We have used in silico and cDNA analyses to investigate the mRNA splicing of the Caenorhabditis elegans orthologue nlg-1. Transcript analysis highlights the potential for gene regulation with respect to both temporal expression and splicing. We found nlg-1 splice variants with all the predicted exons are a minor species relative to major splice variants lacking exons 13 and 14, or 14 alone. These major alternatively spliced variants change the intracellular domain of the gene product NLG-1. Interestingly, exon 14 encodes a cassette with two distinct potential functional domains. One is a polyproline SH3 binding domain and the other has homology to a region encoding the binding site for the scaffolding protein gephyrin in mammalian neuroligins. This suggests differential splicing impacts on NLG-1 competence to recruit intracellular binding partners. This may have developmental relevance as nlg-1 exon 14 containing transcripts are selectively expressed in L2-L3 larvae. These results highlight a developmental regulation of C. elegans nlg-1 that could play a key role in the assembly of synaptic protein complexes during the early stages of nervous system development.
Collapse
Affiliation(s)
- Fernando Calahorro
- Centre for Biological Sciences, Life Sciences Building 85, University of Southampton, Southampton SO17 1BJ, UK.
| | - Lindy Holden-Dye
- Centre for Biological Sciences, Life Sciences Building 85, University of Southampton, Southampton SO17 1BJ, UK
| | - Vincent O'Connor
- Centre for Biological Sciences, Life Sciences Building 85, University of Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|
49
|
Neuroligins, synapse balance and neuropsychiatric disorders. Pharmacol Rep 2014; 66:830-5. [DOI: 10.1016/j.pharep.2014.04.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 04/17/2014] [Accepted: 04/22/2014] [Indexed: 11/19/2022]
|
50
|
Yang H, Niu J, Wang J, Zhang F, Zhang Q, Zhang W, Li A. The down-regulation of neuroligin-2 and the correlative clinical significance of serum GABA over-expression in Hirschsprung's disease. Neurochem Res 2014; 39:1451-7. [PMID: 24842555 DOI: 10.1007/s11064-014-1334-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 04/17/2014] [Accepted: 05/12/2014] [Indexed: 01/15/2023]
Abstract
The goal of this study was to investigate the expression level of neuroligin-2 in different colon tissue segments of children with Hirschsprung's disease (HSCR) and the correlative clinical significance of serum Gamma-Aminobutyric Acid (serum GABA) in HSCR. Neuroligin-2 was assessed by Immunohistochemistry staining method on routine paraffin section from different colon tissue segments of HSCR (ganglionic colonic segment, transitional colonic segment and aganglionic colonic segment). Western-blot analysis and real-time fluorescence quantitative PCR(qRT-PCR) were applied to compare and evaluate the expression levels of neuroligin-2 from three segments of HSCR, and we used Enzyme-linked Immunosorbent Assay (ELISA) method to detect and compare the serum GABA between HSCR and non-HSCR. Immunohistochemistry staining demonstrated that intensive neuroligin-2 staining was detected in the ganglion cells in the ganglionic colonic and transitional colonic segments from the HSCR children; however, neuroligin-2 staining was down-regulated significantly in the aganglionic colonic segments. The expression levels of neuroligin-2 mRNA and protein in the aganglionic colonic segment were decreased compared to the ganglionic colonic segment and transitional colonic segment (P < 0.05). And the level of serum GABA was significantly higher in HSCR than that in non-HSCR. The expression of neuroligin-2 varies from different segments of HSCR. The down-regulation of neuroligin-2 in aganglionic colonic segments may be correlated with the excessive intestine contraction and further result in HSCR. The over-expression of serum GABA may be considered as a new diagnostic method of HSCR.
Collapse
Affiliation(s)
- Hongchao Yang
- Department of Pediatric Surgery, Qilu Hospital, Shandong University, 44#, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|