1
|
Wang J, Sudhof T, Wernig M. Distinct mechanisms control the specific synaptic functions of Neuroligin 1 and Neuroligin 2. EMBO Rep 2025:10.1038/s44319-024-00286-4. [PMID: 39747663 DOI: 10.1038/s44319-024-00286-4] [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: 12/23/2023] [Revised: 09/16/2024] [Accepted: 09/26/2024] [Indexed: 01/04/2025] Open
Abstract
Neuroligins are postsynaptic cell-adhesion molecules that regulate synaptic function with a remarkable isoform specificity. Although Nlgn1 and Nlgn2 are highly homologous and biochemically interact with the same extra- and intracellular proteins, Nlgn1 selectively functions in excitatory synapses whereas Nlgn2 functions in inhibitory synapses. How this excitatory/inhibitory (E/I) specificity arises is unknown. Using a comprehensive structure-function analysis, we here expressed wild-type and mutant neuroligins in functional rescue experiments in cultured hippocampal neurons lacking all endogenous neuroligins. Electrophysiology confirmed that Nlgn1 and Nlgn2 selectively restored excitatory and inhibitory synaptic transmission, respectively, in neuroligin-deficient neurons, aligned with their synaptic localizations. Chimeric Nlgn1-Nlgn2 constructs reveal that the extracellular neuroligin domains confer synapse specificity, whereas their intracellular sequences are exchangeable. However, the cytoplasmic sequences of Nlgn2, including its Gephyrin-binding motif that is identically present in the Nlgn1, is essential for its synaptic function whereas they are dispensable for Nlgn1. These results demonstrate that although the excitatory vs. inhibitory synapse specificity of Nlgn1 and Nlgn2 are both determined by their extracellular sequences, these neuroligins enable normal synaptic connections via distinct intracellular mechanisms.
Collapse
Affiliation(s)
- Jinzhao Wang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Thomas Sudhof
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
2
|
Zahacy R, Ma Y, Winship IR, Jackson J, Chan AW. Claustrum modulation drives altered prefrontal cortex dynamics and connectivity. Commun Biol 2024; 7:1556. [PMID: 39578634 PMCID: PMC11584859 DOI: 10.1038/s42003-024-07256-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
This study delves into the claustrum's role in modulating spontaneous and sensory-evoked network activity across cortical regions. Using mesoscale calcium imaging and Gi and Gq DREADDs in anesthetized mice, we show that decreasing claustral activity enhances prefrontal cortical activity, while activation reduces prefrontal cortical activity. This claustrum modulation also caused changes to the brain's large-scale functional networks, emphasizing the claustrum's ability to influence long-range functional connectivity in the cortex. Claustrum inhibition increased the local coupling between frontal cortex areas, but reduced the correlation between anterior medial regions and lateral/posterior regions, while also enhancing sensory-evoked responses in the visual cortex. These findings indicate the claustrum can participate in orchestrating neural communication across cortical regions through modulation of prefrontal cortical activity. These insights deepen our understanding of the claustrum's impact on prefrontal connectivity, large-scale network dynamics, and sensory processing, positioning the claustrum as a key node modulating large-scale cortical dynamics.
Collapse
Affiliation(s)
- Ryan Zahacy
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Yonglie Ma
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| | - Ian R Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| | - Jesse Jackson
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
- Department of Physiology, University of Alberta, Edmonton, AB, Canada.
| | - Allen W Chan
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
3
|
Zeppillo T, Ali H, Ravichandran S, Ritter TC, Wenger S, López-Murcia FJ, Gideons E, Signorelli J, Schmeisser MJ, Wiltfang J, Rhee J, Brose N, Taschenberger H, Krueger-Burg D. Functional Neuroligin-2-MDGA1 interactions differentially regulate synaptic GABA ARs and cytosolic gephyrin aggregation. Commun Biol 2024; 7:1157. [PMID: 39284869 PMCID: PMC11405390 DOI: 10.1038/s42003-024-06789-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Neuroligin-2 (Nlgn2) is a key synaptic adhesion protein at virtually all GABAergic synapses, which recruits GABAARs by promoting assembly of the postsynaptic gephyrin scaffold. Intriguingly, loss of Nlgn2 differentially affects subsets of GABAergic synapses, indicating that synapse-specific interactors and redundancies define its function, but the nature of these interactions remain poorly understood. Here we investigated how Nlgn2 function in hippocampal area CA1 is modulated by two proposed interaction partners, MDGA1 and MDGA2. We show that loss of MDGA1 expression, but not heterozygous deletion of MDGA2, ameliorates the abnormal cytosolic gephyrin aggregation, the reduction in inhibitory synaptic transmission and the exacerbated anxiety-related behaviour characterizing Nlgn2 knockout (KO) mice. Additionally, combined Nlgn2 and MDGA1 deletion causes an exacerbated layer-specific loss of gephyrin puncta. Given that both Nlgn2 and the MDGA1 have been correlated with many psychiatric disorders, our data support the notion that cytosolic gephyrin aggregation may represent an interesting target for novel therapeutic strategies.
Collapse
Affiliation(s)
- Tommaso Zeppillo
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany
| | - Heba Ali
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
- Göttingen Graduate School for Neurosciences, Biophysics, and Molecular Biosciences (GGNB), Georg-August-University Göttingen, 37077, Göttingen, Germany
| | - Sowbarnika Ravichandran
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany
- Focus Program Translational Neurosciences (FTN), University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany
| | - Tamara C Ritter
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany
- Focus Program Translational Neurosciences (FTN), University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany
| | - Sally Wenger
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
| | - Francisco J López-Murcia
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
- Department of Pathology and Experimental Therapy, Institute of Neurosciences, University of Barcelona, and Bellvitge Biomedical Research Institute (IDIBELL), 08907, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Erinn Gideons
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
| | - Janetti Signorelli
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
- Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, 1240000, Antofagasta, Chile
| | - Michael J Schmeisser
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany
- Focus Program Translational Neurosciences (FTN), University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center of the Georg-August-University Göttingen Mainz, 37075, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), 37075, Goettingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - JeongSeop Rhee
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
| | - Holger Taschenberger
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
| | - Dilja Krueger-Burg
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany.
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany.
- Focus Program Translational Neurosciences (FTN), University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany.
- Department of Psychiatry and Psychotherapy, University Medical Center of the Georg-August-University Göttingen Mainz, 37075, Göttingen, Germany.
| |
Collapse
|
4
|
Ojima D, Tominaga Y, Kubota T, Tada A, Takahashi H, Kishimoto Y, Tominaga T, Yamamoto T. Impaired Hippocampal Long-Term Potentiation and Memory Deficits upon Haploinsufficiency of MDGA1 Can Be Rescued by Acute Administration of D-Cycloserine. Int J Mol Sci 2024; 25:9674. [PMID: 39273620 PMCID: PMC11394992 DOI: 10.3390/ijms25179674] [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: 08/08/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
The maintenance of proper brain function relies heavily on the balance of excitatory and inhibitory neural circuits, governed in part by synaptic adhesion molecules. Among these, MDGA1 (MAM domain-containing glycosylphosphatidylinositol anchor 1) acts as a suppressor of synapse formation by interfering with Neuroligin-mediated interactions, crucial for maintaining the excitatory-inhibitory (E/I) balance. Mdga1-/- mice exhibit selectively enhanced inhibitory synapse formation in their hippocampal pyramidal neurons, leading to impaired hippocampal long-term potentiation (LTP) and hippocampus-dependent learning and memory function; however, it has not been fully investigated yet if the reduction in MDGA1 protein levels would alter brain function. Here, we examined the behavioral and synaptic consequences of reduced MDGA1 protein levels in Mdga1+/- mice. As observed in Mdga1-/- mice, Mdga1+/- mice exhibited significant deficits in hippocampus-dependent learning and memory tasks, such as the Morris water maze and contextual fear-conditioning tests, along with a significant deficit in the long-term potentiation (LTP) in hippocampal Schaffer collateral CA1 synapses. The acute administration of D-cycloserine, a co-agonist of NMDAR (N-methyl-d-aspartate receptor), significantly ameliorated memory impairments and restored LTP deficits specifically in Mdga1+/- mice, while having no such effect on Mdga1-/- mice. These results highlight the critical role of MDGA1 in regulating inhibitory synapse formation and maintaining the E/I balance for proper cognitive function. These findings may also suggest potential therapeutic strategies targeting the E/I imbalance to alleviate cognitive deficits associated with neuropsychiatric disorders.
Collapse
Grants
- 16K08237, 19K07065, 19K07337, 16H06532, 24H01497, 23K18485, 23K21755, 21H03606, 23H03488, 23K28178, 23K21713, 22H05698, 24K18267, 21K15247, 19K07337, 22K06618 the Ministry of Education, Culture, Sports, Science and Technology, Japan
Collapse
Affiliation(s)
- Daiki Ojima
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho 761-0793, Kagawa, Japan (A.T.); (H.T.)
| | - Yoko Tominaga
- Institute of Neuroscience, Tokushima Bunri University, Sanuki 769-2193, Kagawa, Japan
| | - Takashi Kubota
- Department of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki 769-2193, Kagawa, Japan; (T.K.); (Y.K.)
| | - Atsushi Tada
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho 761-0793, Kagawa, Japan (A.T.); (H.T.)
| | - Hiroo Takahashi
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho 761-0793, Kagawa, Japan (A.T.); (H.T.)
| | - Yasushi Kishimoto
- Department of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki 769-2193, Kagawa, Japan; (T.K.); (Y.K.)
- Laboratory of Physical Chemistry, Faculty of Pharmaceutical Sciences, Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan
| | - Takashi Tominaga
- Institute of Neuroscience, Tokushima Bunri University, Sanuki 769-2193, Kagawa, Japan
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki 769-2193, Kagawa, Japan
| | - Tohru Yamamoto
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho 761-0793, Kagawa, Japan (A.T.); (H.T.)
| |
Collapse
|
5
|
Godavarthi SK, Li HQ, Pratelli M, Spitzer NC. Embryonic exposure to environmental factors drives transmitter switching in the neonatal mouse cortex causing autistic-like adult behavior. Proc Natl Acad Sci U S A 2024; 121:e2406928121. [PMID: 39178233 PMCID: PMC11363343 DOI: 10.1073/pnas.2406928121] [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: 04/06/2024] [Accepted: 07/23/2024] [Indexed: 08/25/2024] Open
Abstract
Autism spectrum disorders (ASD) can be caused by environmental factors. These factors act early in the development of the nervous system and induce stereotyped repetitive behaviors and diminished social interactions, among other outcomes. Little is known about how these behaviors are produced. In pregnant women, delivery of valproic acid (VPA) (to control seizure activity or stabilize mood) or immune activation by a virus increases the incidence of ASD in offspring. We found that either VPA or Poly Inosine:Cytosine (which mimics a viral infection), administered at mouse embryonic day 12.5, induced a neurotransmitter switch from GABA to glutamate in PV- and CCK-expressing interneurons in the medial prefrontal cortex by postnatal day 10. The switch was present for only a brief period during early postnatal development, observed in male and female mice at postnatal day 21 and reversed in both males and females by postnatal day 30. At postnatal day 90, male mice exhibited stereotyped repetitive behaviors and diminished social interaction while female mice exhibited only stereotyped repetitive behavior. Transfecting GAD1 in PV- and CCK-expressing interneurons at postnatal day 10, to reintroduce GABA expression, overrode the switch and prevented expression of autistic-like behavior. These findings point to an important role of neurotransmitter switching in mediating the environmental causes of autism.
Collapse
Affiliation(s)
- Swetha K. Godavarthi
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| | - Hui-quan Li
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| | - Marta Pratelli
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| | - Nicholas C. Spitzer
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| |
Collapse
|
6
|
Imamura I, Kiyama R. Potential involvement of KANK1 haploinsufficiency in centrosome aberrations. Biochim Biophys Acta Gen Subj 2024; 1868:130648. [PMID: 38830559 DOI: 10.1016/j.bbagen.2024.130648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/04/2024] [Accepted: 05/24/2024] [Indexed: 06/05/2024]
Abstract
KANK1 was found as a tumor suppressor gene based on frequent deletions in renal cell carcinoma and the inhibitory activity of tumor cell proliferation. Previously, we reported that knockdown of KANK1 induced centrosomal amplification, leading to abnormal cell division, through the hyperactivation of RhoA small GTPase. Here, we investigated the loss of KANK1 function by performing CRISPR/Cas9-based genome editing to knockout the gene. After several rounds of genome editing, however, there were no cell lines with complete loss of KANK1, and the less the wild-type KANK1 dosage, the greater the number of cells with abnormal numbers of centrosomes and rates of cell-doubling and apoptosis, suggesting the involvement of KANK1 haploinsufficiency in centrosome aberrations. The rescue of KANK1-knockdown cells with a KANK1-expressing plasmid restored the rates of cells exhibiting centrosomal amplification to the control level. RNA-sequencing analysis of the cells with reduced dosages of functional KANK1 revealed potential involvement of other cell proliferation-related genes, such as EGR1, MDGA2, and BMP3, which have been reported to show haploinsufficiency when they function. When EGR1 protein expression was reduced by siRNA technology, the number of cells exhibiting centrosomal amplification increased, along with the reduction of KANK1 protein expression, suggesting their functional relationship. Thus, KANK1 haploinsufficiency may contribute to centrosome aberrations through the network of haploinsufficiency-related genes.
Collapse
Affiliation(s)
- Ikumi Imamura
- Faculty of Life Science, Kyushu Sangyo University, Japan
| | - Ryoiti Kiyama
- Faculty of Life Science, Kyushu Sangyo University, Japan.
| |
Collapse
|
7
|
Xu QW, Larosa A, Wong TP. Roles of AMPA receptors in social behaviors. Front Synaptic Neurosci 2024; 16:1405510. [PMID: 39056071 PMCID: PMC11269240 DOI: 10.3389/fnsyn.2024.1405510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
As a crucial player in excitatory synaptic transmission, AMPA receptors (AMPARs) contribute to the formation, regulation, and expression of social behaviors. AMPAR modifications have been associated with naturalistic social behaviors, such as aggression, sociability, and social memory, but are also noted in brain diseases featuring impaired social behavior. Understanding the role of AMPARs in social behaviors is timely to reveal therapeutic targets for treating social impairment in disorders, such as autism spectrum disorder and schizophrenia. In this review, we will discuss the contribution of the molecular composition, function, and plasticity of AMPARs to social behaviors. The impact of targeting AMPARs in treating brain disorders will also be discussed.
Collapse
Affiliation(s)
- Qi Wei Xu
- Douglas Hospital Research Centre, Montreal, QC, Canada
| | - Amanda Larosa
- Douglas Hospital Research Centre, Montreal, QC, Canada
| | - Tak Pan Wong
- Douglas Hospital Research Centre, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
8
|
Wang X, Lin D, Jiang J, Liu Y, Dong X, Fan J, Gong L, Shen W, Zeng L, Xu T, Jiang K, Connor SA, Xie Y. MDGA2 Constrains Glutamatergic Inputs Selectively onto CA1 Pyramidal Neurons to Optimize Neural Circuits for Plasticity, Memory, and Social Behavior. Neurosci Bull 2024; 40:887-904. [PMID: 38321347 PMCID: PMC11250762 DOI: 10.1007/s12264-023-01171-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/08/2023] [Indexed: 02/08/2024] Open
Abstract
Synapse organizers are essential for the development, transmission, and plasticity of synapses. Acting as rare synapse suppressors, the MAM domain containing glycosylphosphatidylinositol anchor (MDGA) proteins contributes to synapse organization by inhibiting the formation of the synaptogenic neuroligin-neurexin complex. A previous analysis of MDGA2 mice lacking a single copy of Mdga2 revealed upregulated glutamatergic synapses and behaviors consistent with autism. However, MDGA2 is expressed in diverse cell types and is localized to both excitatory and inhibitory synapses. Differentiating the network versus cell-specific effects of MDGA2 loss-of-function requires a cell-type and brain region-selective strategy. To address this, we generated mice harboring a conditional knockout of Mdga2 restricted to CA1 pyramidal neurons. Here we report that MDGA2 suppresses the density and function of excitatory synapses selectively on pyramidal neurons in the mature hippocampus. Conditional deletion of Mdga2 in CA1 pyramidal neurons of adult mice upregulated miniature and spontaneous excitatory postsynaptic potentials, vesicular glutamate transporter 1 intensity, and neuronal excitability. These effects were limited to glutamatergic synapses as no changes were detected in miniature and spontaneous inhibitory postsynaptic potential properties or vesicular GABA transporter intensity. Functionally, evoked basal synaptic transmission and AMPAR receptor currents were enhanced at glutamatergic inputs. At a behavioral level, memory appeared to be compromised in Mdga2 cKO mice as both novel object recognition and contextual fear conditioning performance were impaired, consistent with deficits in long-term potentiation in the CA3-CA1 pathway. Social affiliation, a behavioral analog of social deficits in autism, was similarly compromised. These results demonstrate that MDGA2 confines the properties of excitatory synapses to CA1 neurons in mature hippocampal circuits, thereby optimizing this network for plasticity, cognition, and social behaviors.
Collapse
Affiliation(s)
- Xuehui Wang
- School of Life Sciences, Nanchang University, Nanchang, 330031, China
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Donghui Lin
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Jie Jiang
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China
- Department of Biology, York University, 4700 Keele Street, Toronto, ON, M3J 1P3, Canada
| | - Yuhua Liu
- School of Life Sciences, Nanchang University, Nanchang, 330031, China
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Xinyan Dong
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Jianchen Fan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Lifen Gong
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Weida Shen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Linghui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Tonghui Xu
- School of Life Sciences, Nanchang University, Nanchang, 330031, China
| | - Kewen Jiang
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China.
| | - Steven A Connor
- Department of Biology, York University, 4700 Keele Street, Toronto, ON, M3J 1P3, Canada.
| | - Yicheng Xie
- School of Life Sciences, Nanchang University, Nanchang, 330031, China.
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China.
| |
Collapse
|
9
|
Kim S, Jang G, Kim H, Lim D, Han KA, Um JW, Ko J. MDGAs perform activity-dependent synapse type-specific suppression via distinct extracellular mechanisms. Proc Natl Acad Sci U S A 2024; 121:e2322978121. [PMID: 38900791 PMCID: PMC11214077 DOI: 10.1073/pnas.2322978121] [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: 12/29/2023] [Accepted: 05/15/2024] [Indexed: 06/22/2024] Open
Abstract
MDGA (MAM domain containing glycosylphosphatidylinositol anchor) family proteins were previously identified as synaptic suppressive factors. However, various genetic manipulations have yielded often irreconcilable results, precluding precise evaluation of MDGA functions. Here, we found that, in cultured hippocampal neurons, conditional deletion of MDGA1 and MDGA2 causes specific alterations in synapse numbers, basal synaptic transmission, and synaptic strength at GABAergic and glutamatergic synapses, respectively. Moreover, MDGA2 deletion enhanced both N-methyl-D-aspartate (NMDA) receptor- and α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor-mediated postsynaptic responses. Strikingly, ablation of both MDGA1 and MDGA2 abolished the effect of deleting individual MDGAs that is abrogated by chronic blockade of synaptic activity. Molecular replacement experiments further showed that MDGA1 requires the meprin/A5 protein/PTPmu (MAM) domain, whereas MDGA2 acts via neuroligin-dependent and/or MAM domain-dependent pathways to regulate distinct postsynaptic properties. Together, our data demonstrate that MDGA paralogs act as unique negative regulators of activity-dependent postsynaptic organization at distinct synapse types, and cooperatively contribute to adjustment of excitation-inhibition balance.
Collapse
Affiliation(s)
- Seungjoon Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Gyubin Jang
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Hyeonho Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Dongseok Lim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Kyung Ah Han
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Ji Won Um
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Jaewon Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| |
Collapse
|
10
|
Luo W, Egger M, Cruz-Ochoa N, Tse A, Maloveczky G, Tamás B, Lukacsovich D, Seng C, Amrein I, Lukacsovich T, Wolfer D, Földy C. Activation of feedforward wiring in adult hippocampal neurons by the basic-helix-loop-helix transcription factor Ascl4. PNAS NEXUS 2024; 3:pgae174. [PMID: 38711810 PMCID: PMC11071515 DOI: 10.1093/pnasnexus/pgae174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/17/2024] [Indexed: 05/08/2024]
Abstract
Although evidence indicates that the adult brain retains a considerable capacity for circuit formation, adult wiring has not been broadly considered and remains poorly understood. In this study, we investigate wiring activation in adult neurons. We show that the basic-helix-loop-helix transcription factor Ascl4 can induce wiring in different types of hippocampal neurons of adult mice. The new axons are mainly feedforward and reconfigure synaptic weights in the circuit. Mice with the Ascl4-induced circuits do not display signs of pathology and solve spatial problems equally well as controls. Our results demonstrate reprogrammed connectivity by a single transcriptional factor and provide insights into the regulation of brain wiring in adults.
Collapse
Affiliation(s)
- Wenshu Luo
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - Matteo Egger
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, Zürich 8057, Switzerland
| | - Natalia Cruz-Ochoa
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, Zürich 8057, Switzerland
| | - Alice Tse
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - Gyula Maloveczky
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - Bálint Tamás
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - David Lukacsovich
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - Charlotte Seng
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - Irmgard Amrein
- Institute of Anatomy, Faculty of Medicine, University of Zürich, Zürich 8057, Switzerland
| | - Tamás Lukacsovich
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - David Wolfer
- Institute of Anatomy, Faculty of Medicine, University of Zürich, Zürich 8057, Switzerland
- Institute of Human Movement Sciences and Sport, D-HEST, ETH Zürich, Zürich 8057, Switzerland
| | - Csaba Földy
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, Zürich 8057, Switzerland
| |
Collapse
|
11
|
Connor SA, Siddiqui TJ. Synapse organizers as molecular codes for synaptic plasticity. Trends Neurosci 2023; 46:971-985. [PMID: 37652840 DOI: 10.1016/j.tins.2023.08.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/13/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023]
Abstract
Synapse organizing proteins are multifaceted molecules that coordinate the complex processes of brain development and plasticity at the level of individual synapses. Their importance is demonstrated by the major brain disorders that emerge when their function is compromised. The mechanisms whereby the various families of organizers govern synapses are diverse, but converge on the structure, function, and plasticity of synapses. Therefore, synapse organizers regulate how synapses adapt to ongoing activity, a process central for determining the developmental trajectory of the brain and critical to all forms of cognition. Here, we explore how synapse organizers set the conditions for synaptic plasticity and the associated molecular events, which eventually link to behavioral features of neurodevelopmental and neuropsychiatric disorders. We also propose central questions on how synapse organizers influence network function through integrating nanoscale and circuit-level organization of the brain.
Collapse
Affiliation(s)
- Steven A Connor
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
| | - Tabrez J Siddiqui
- PrairieNeuro Research Centre, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, MB R3E 0Z3, Canada; Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; The Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada; Program in Biomedical Engineering, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
12
|
Fertan E, Wong AA, Montbrun TSGD, Purdon MK, Roddick KM, Yamamoto T, Brown RE. Early postnatal development of the MDGA2 +/- mouse model of synaptic dysfunction. Behav Brain Res 2023; 452:114590. [PMID: 37499910 DOI: 10.1016/j.bbr.2023.114590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/13/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023]
Abstract
Synaptic dysfunction underlies many neurodevelopmental disorders (NDDs). The membrane-associated mucin domain-containing glycosylphosphatidylinositol anchor proteins (MDGAs) regulate synaptic development by modulating neurexin-neuroligin complex formation. Since understanding the neurodevelopmental profile and the sex-based differences in the manifestation of the symptoms of NDDs is important for their early diagnosis, we tested a mouse model haploinsufficient for MDGA2 (MDGA2+/-) on a neurodevelopmental test battery, containing sensory, motor, and cognitive measures, as well as ultrasonic vocalizations. When male and female MDGA2+/- and wildtype (WT) C57BL/6 J mice were examined from 2 to 23 days of age using this test battery, genotype and sex differences in body weight, sensory-motor processes, and ultrasonic vocalizations were observed. The auditory startle reflex appeared earlier in the MDGA2+/- than in WT mice and the MDGA2+/- mice produced fewer ultrasonic vocalizations. The MDGA2+/- mice showed reduced locomotion and rearing than WT mice in the open field after 17 days of age and spent less time investigating a novel object than WT mice at 21 days of age. Female MDGA2+/- mice weighed less than WT females and showed lower grip strength, indicating a delay in sensory-motor development in MDGA2+/- mice, which appears to be more pronounced in females than males. The behavioural phenotypes resulting from MDGA2 haploinsufficiency suggests that it shows delayed development of motor behaviour, grip strength and exploratory behaviour, non-social phenotypes of NDDs.
Collapse
Affiliation(s)
- Emre Fertan
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Aimée A Wong
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | | | - Michaela K Purdon
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Kyle M Roddick
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Tohru Yamamoto
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kagawa 761-0793, Japan
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
13
|
Bemben MA, Sandoval M, Le AA, Won S, Chau VN, Lauterborn JC, Incontro S, Li KH, Burlingame AL, Roche KW, Gall CM, Nicoll RA, Diaz-Alonso J. Contrastsing synaptic roles of MDGA1 and MDGA2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.25.542333. [PMID: 37720016 PMCID: PMC10503827 DOI: 10.1101/2023.05.25.542333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Neurodevelopmental disorders are frequently linked to mutations in synaptic organizing molecules. MAM domain containing glycosylphosphatidylinositol anchor 1 and 2 (MDGA1 and MDGA2) are a family of synaptic organizers suggested to play an unusual role as synaptic repressors, but studies offer conflicting evidence for their localization. Using epitope-tagged MDGA1 and MDGA2 knock-in mice, we found that native MDGAs are expressed throughout the brain, peaking early in postnatal development. Surprisingly, endogenous MDGA1 was enriched at excitatory, but not inhibitory, synapses. Both shRNA knockdown and CRISPR/Cas9 knockout of MDGA1 resulted in cell-autonomous, specific impairment of AMPA receptor-mediated synaptic transmission, without affecting GABAergic transmission. Conversely, MDGA2 knockdown/knockout selectively depressed NMDA receptor-mediated transmission but enhanced inhibitory transmission. Our results establish that MDGA2 acts as a synaptic repressor, but only at inhibitory synapses, whereas both MDGAs are required for excitatory transmission. This nonoverlapping division of labor between two highly conserved synaptic proteins is unprecedented.
Collapse
Affiliation(s)
- Michael A. Bemben
- Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Matthew Sandoval
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Aliza A. Le
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Sehoon Won
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Vivian N. Chau
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Julie C. Lauterborn
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Salvatore Incontro
- Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, CA 94158, USA
- Present address: Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR1072, INSERM, Aix-Marseille University, Marseille, 13015, France
| | - Kathy H. Li
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Alma L. Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Katherine W. Roche
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Christine M. Gall
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Roger A. Nicoll
- Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, CA 94158, USA
- Department of Physiology, University of California at San Francisco, San Francisco, CA, 94158, USA
| | - Javier Diaz-Alonso
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| |
Collapse
|
14
|
Lee H, Chofflet N, Liu J, Fan S, Lu Z, Resua Rojas M, Penndorf P, Bailey AO, Russell WK, Machius M, Ren G, Takahashi H, Rudenko G. Designer molecules of the synaptic organizer MDGA1 reveal 3D conformational control of biological function. J Biol Chem 2023; 299:104586. [PMID: 36889589 PMCID: PMC10131064 DOI: 10.1016/j.jbc.2023.104586] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
MDGAs (MAM domain-containing glycosylphosphatidylinositol anchors) are synaptic cell surface molecules that regulate the formation of trans-synaptic bridges between neurexins (NRXNs) and neuroligins (NLGNs), which promote synaptic development. Mutations in MDGAs are implicated in various neuropsychiatric diseases. MDGAs bind NLGNs in cis on the postsynaptic membrane and physically block NLGNs from binding to NRXNs. In crystal structures, the six immunoglobulin (Ig) and single fibronectin III domains of MDGA1 reveal a striking compact, triangular shape, both alone and in complex with NLGNs. Whether this unusual domain arrangement is required for biological function or other arrangements occur with different functional outcomes is unknown. Here, we show that WT MDGA1 can adopt both compact and extended 3D conformations that bind NLGN2. Designer mutants targeting strategic molecular elbows in MDGA1 alter the distribution of 3D conformations while leaving the binding affinity between soluble ectodomains of MDGA1 and NLGN2 intact. In contrast, in a cellular context, these mutants result in unique combinations of functional consequences, including altered binding to NLGN2, decreased capacity to conceal NLGN2 from NRXN1β, and/or suppressed NLGN2-mediated inhibitory presynaptic differentiation, despite the mutations being located far from the MDGA1-NLGN2 interaction site. Thus, the 3D conformation of the entire MDGA1 ectodomain appears critical for its function, and its NLGN-binding site on Ig1-Ig2 is not independent of the rest of the molecule. As a result, global 3D conformational changes to the MDGA1 ectodomain via strategic elbows may form a molecular mechanism to regulate MDGA1 action within the synaptic cleft.
Collapse
Affiliation(s)
- Hubert Lee
- Deptartment of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA
| | - Nicolas Chofflet
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Jianfang Liu
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Shanghua Fan
- Deptartment of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA
| | - Zhuoyang Lu
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Martin Resua Rojas
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, Quebec, Canada
| | - Patrick Penndorf
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, Quebec, Canada
| | - Aaron O Bailey
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - William K Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mischa Machius
- Deptartment of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA
| | - Gang Ren
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| | - Hideto Takahashi
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montréal, Quebec, Canada; Division of Experimental Medicine, McGill University, Montréal, Quebec, Canada.
| | - Gabby Rudenko
- Deptartment of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
15
|
Anderson AG, Rogers BB, Loupe JM, Rodriguez-Nunez I, Roberts SC, White LM, Brazell JN, Bunney WE, Bunney BG, Watson SJ, Cochran JN, Myers RM, Rizzardi LF. Single nucleus multiomics identifies ZEB1 and MAFB as candidate regulators of Alzheimer's disease-specific cis-regulatory elements. CELL GENOMICS 2023; 3:100263. [PMID: 36950385 PMCID: PMC10025452 DOI: 10.1016/j.xgen.2023.100263] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/06/2022] [Accepted: 01/12/2023] [Indexed: 02/05/2023]
Abstract
Cell type-specific transcriptional differences between brain tissues from donors with Alzheimer's disease (AD) and unaffected controls have been well documented, but few studies have rigorously interrogated the regulatory mechanisms responsible for these alterations. We performed single nucleus multiomics (snRNA-seq plus snATAC-seq) on 105,332 nuclei isolated from cortical tissues from 7 AD and 8 unaffected donors to identify candidate cis-regulatory elements (CREs) involved in AD-associated transcriptional changes. We detected 319,861 significant correlations, or links, between gene expression and cell type-specific transposase accessible regions enriched for active CREs. Among these, 40,831 were unique to AD tissues. Validation experiments confirmed the activity of many regions, including several candidate regulators of APP expression. We identified ZEB1 and MAFB as candidate transcription factors playing important roles in AD-specific gene regulation in neurons and microglia, respectively. Microglia links were globally enriched for heritability of AD risk and previously identified active regulatory regions.
Collapse
Affiliation(s)
| | - Brianne B. Rogers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jacob M. Loupe
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | | | | | - Lauren M. White
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | | | - William E. Bunney
- Department of Psychiatry and Human Behavior, College of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Blynn G. Bunney
- Department of Psychiatry and Human Behavior, College of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Stanley J. Watson
- Mental Health Research Institute, University of Michigan, Ann Arbor, MI, USA
| | | | | | | |
Collapse
|
16
|
Li M, Usui N, Shimada S. Prenatal Sex Hormone Exposure Is Associated with the Development of Autism Spectrum Disorder. Int J Mol Sci 2023; 24:ijms24032203. [PMID: 36768521 PMCID: PMC9916422 DOI: 10.3390/ijms24032203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/19/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Sexual differentiation is a major developmental process. Sex differences resulting from sexual differentiation have attracted the attention of researchers. Unraveling what contributes to and underlies sex differences will provide valuable insights into the development of neurodevelopmental disorders that exhibit sex biases. Autism spectrum disorder (ASD) is a neurodevelopmental disorder that affects an individual's social interaction and communication abilities, and its male preponderance has been consistently reported in clinical studies. The etiology of male preponderance remains unclear, but progress has been made in studying prenatal sex hormone exposure. The present review examined studies that focused on the association between prenatal testosterone exposure and ASD development, as well as sex-specific behaviors in individuals with ASD. This review also included studies on maternal immune activation-induced developmental abnormalities that also showed striking sex differences in offspring and discussed its possible interacting roles in ASD so as to present a potential approach for future studies on sex biases in ASD.
Collapse
Affiliation(s)
- Mengwei Li
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
| | - Noriyoshi Usui
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
- United Graduate School of Child Development, Osaka University, Suita 565-0871, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Suita 565-0871, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
- Correspondence: ; Tel.: +81-6-6879-3124
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
- United Graduate School of Child Development, Osaka University, Suita 565-0871, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Suita 565-0871, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
| |
Collapse
|
17
|
Li H, Guo R, Guan Y, Li J, Wang Y. Modulation of Trans-Synaptic Neurexin-Neuroligin Interaction in Pathological Pain. Cells 2022; 11:cells11121940. [PMID: 35741069 PMCID: PMC9222181 DOI: 10.3390/cells11121940] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 11/16/2022] Open
Abstract
Synapses serve as the interface for the transmission of information between neurons in the central nervous system. The structural and functional characteristics of synapses are highly dynamic, exhibiting extensive plasticity that is shaped by neural activity and regulated primarily by trans-synaptic cell-adhesion molecules (CAMs). Prototypical trans-synaptic CAMs, such as neurexins (Nrxs) and neuroligins (Nlgs), directly regulate the assembly of presynaptic and postsynaptic molecules, including synaptic vesicles, active zone proteins, and receptors. Therefore, the trans-synaptic adhesion mechanisms mediated by Nrx-Nlg interaction can contribute to a range of synaptopathies in the context of pathological pain and other neurological disorders. The present review provides an overview of the current understanding of the roles of Nrx-Nlg interaction in the regulation of trans-synaptic connections, with a specific focus on Nrx and Nlg structures, the dynamic shaping of synaptic function, and the dysregulation of Nrx-Nlg in pathological pain. Additionally, we discuss a range of proteins capable of modulating Nrx-Nlg interactions at the synaptic cleft, with the objective of providing a foundation to guide the future development of novel therapeutic agents for managing pathological pain.
Collapse
Affiliation(s)
- Huili Li
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China;
| | - Ruijuan Guo
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100030, China;
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | - Junfa Li
- Department of Neurobiology, Capital Medical University, Beijing 100069, China;
| | - Yun Wang
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China;
- Correspondence: ; Tel.: +86-10-85231463
| |
Collapse
|
18
|
Toledo A, Letellier M, Bimbi G, Tessier B, Daburon S, Favereaux A, Chamma I, Vennekens K, Vanderlinden J, Sainlos M, de Wit J, Choquet D, Thoumine O. MDGAs are fast-diffusing molecules that delay excitatory synapse development by altering neuroligin behavior. eLife 2022; 11:75233. [PMID: 35532105 PMCID: PMC9084894 DOI: 10.7554/elife.75233] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/11/2022] [Indexed: 12/28/2022] Open
Abstract
MDGA molecules can bind neuroligins and interfere with trans-synaptic interactions to neurexins, thereby impairing synapse development. However, the subcellular localization and dynamics of MDGAs, or their specific action mode in neurons remain unclear. Here, surface immunostaining of endogenous MDGAs and single molecule tracking of recombinant MDGAs in dissociated hippocampal neurons reveal that MDGAs are homogeneously distributed and exhibit fast membrane diffusion, with a small reduction in mobility across neuronal maturation. Knocking-down/out MDGAs using shRNAs and CRISPR/Cas9 strategies increases the density of excitatory synapses, the membrane confinement of neuroligin-1, and the phosphotyrosine level of neuroligins associated with excitatory post-synaptic differentiation. Finally, MDGA silencing reduces the mobility of AMPA receptors, increases the frequency of miniature EPSCs (but not IPSCs), and selectively enhances evoked AMPA-receptor-mediated EPSCs in CA1 pyramidal neurons. Overall, our results support a mechanism by which interactions between MDGAs and neuroligin-1 delays the assembly of functional excitatory synapses containing AMPA receptors.
Collapse
Affiliation(s)
- Andrea Toledo
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Mathieu Letellier
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Giorgia Bimbi
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Béatrice Tessier
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Sophie Daburon
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Alexandre Favereaux
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Ingrid Chamma
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Kristel Vennekens
- VIB Center for Brain & Disease Research and KU Leuven, Department of Neurosciences, Leuven Brain Institute
| | - Jeroen Vanderlinden
- VIB Center for Brain & Disease Research and KU Leuven, Department of Neurosciences, Leuven Brain Institute
| | - Matthieu Sainlos
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Joris de Wit
- VIB Center for Brain & Disease Research and KU Leuven, Department of Neurosciences, Leuven Brain Institute
| | - Daniel Choquet
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
- University of Bordeaux, CNRS UAR 3420, INSERM, Bordeaux Imaging Center
| | - Olivier Thoumine
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| |
Collapse
|
19
|
Zhang Z, Hou M, Ou H, Wang D, Li Z, Zhang H, Lu J. Expression and structural analysis of human neuroligin 2 and neuroligin 3 implicated in autism spectrum disorders. Front Endocrinol (Lausanne) 2022; 13:1067529. [PMID: 36479216 PMCID: PMC9719943 DOI: 10.3389/fendo.2022.1067529] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 10/31/2022] [Indexed: 11/22/2022] Open
Abstract
The development of autism spectrum disorders (ASDs) involves both environmental factors such as maternal diabetes and genetic factors such as neuroligins (NLGNs). NLGN2 and NLGN3 are two members of NLGNs with distinct distributions and functions in synapse development and plasticity. The relationship between maternal diabetes and NLGNs, and the distinct working mechanisms of different NLGNs currently remain unclear. Here, we first analyzed the expression levels of NLGN2 and NLGN3 in a streptozotocin-induced ASD mouse model and different brain regions to reveal their differences and similarities. Then, cryogenic electron microscopy (cryo-EM) structures of human NLGN2 and NLGN3 were determined. The overall structures are similar to their homologs in previous reports. However, structural comparisons revealed the relative rotations of two protomers in the homodimers of NLGN2 and NLGN3. Taken together with the previously reported NLGN2-MDGA1 complex, we speculate that the distinct assembly adopted by NLGN2 and NLGN3 may affect their interactions with MDGAs. Our results provide structural insights into the potential distinct mechanisms of NLGN2 and NLGN3 implicated in the development of ASD.
Collapse
Affiliation(s)
- Zhenzhen Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Mengzhuo Hou
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Huaxing Ou
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Daping Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhifang Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
- *Correspondence: Jianping Lu, ; Huawei Zhang, ; Zhifang Li,
| | - Huawei Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
- *Correspondence: Jianping Lu, ; Huawei Zhang, ; Zhifang Li,
| | - Jianping Lu
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, China
- *Correspondence: Jianping Lu, ; Huawei Zhang, ; Zhifang Li,
| |
Collapse
|
20
|
Hageter J, Waalkes M, Starkey J, Copeland H, Price H, Bays L, Showman C, Laverty S, Bergeron SA, Horstick EJ. Environmental and Molecular Modulation of Motor Individuality in Larval Zebrafish. Front Behav Neurosci 2021; 15:777778. [PMID: 34938167 PMCID: PMC8685292 DOI: 10.3389/fnbeh.2021.777778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/17/2021] [Indexed: 11/21/2022] Open
Abstract
Innate behavioral biases such as human handedness are a ubiquitous form of inter-individual variation that are not strictly hardwired into the genome and are influenced by diverse internal and external cues. Yet, genetic and environmental factors modulating behavioral variation remain poorly understood, especially in vertebrates. To identify genetic and environmental factors that influence behavioral variation, we take advantage of larval zebrafish light-search behavior. During light-search, individuals preferentially turn in leftward or rightward loops, in which directional bias is sustained and non-heritable. Our previous work has shown that bias is maintained by a habenula-rostral PT circuit and genes associated with Notch signaling. Here we use a medium-throughput recording strategy and unbiased analysis to show that significant individual to individual variation exists in wildtype larval zebrafish turning preference. We classify stable left, right, and unbiased turning types, with most individuals exhibiting a directional preference. We show unbiased behavior is not due to a loss of photo-responsiveness but reduced persistence in same-direction turning. Raising larvae at elevated temperature selectively reduces the leftward turning type and impacts rostral PT neurons, specifically. Exposure to conspecifics, variable salinity, environmental enrichment, and physical disturbance does not significantly impact inter-individual turning bias. Pharmacological manipulation of Notch signaling disrupts habenula development and turn bias individuality in a dose dependent manner, establishing a direct role of Notch signaling. Last, a mutant allele of a known Notch pathway affecter gene, gsx2, disrupts turn bias individuality, implicating that brain regions independent of the previously established habenula-rostral PT likely contribute to inter-individual variation. These results establish that larval zebrafish is a powerful vertebrate model for inter-individual variation with established neural targets showing sensitivity to specific environmental and gene signaling disruptions. Our results provide new insight into how variation is generated in the vertebrate nervous system.
Collapse
Affiliation(s)
- John Hageter
- Department of Biology, West Virginia University, Morgantown, WV, United States
| | - Matthew Waalkes
- Department of Biology, West Virginia University, Morgantown, WV, United States
| | - Jacob Starkey
- Department of Biology, West Virginia University, Morgantown, WV, United States
| | - Haylee Copeland
- Department of Biology, West Virginia University, Morgantown, WV, United States
| | - Heather Price
- Department of Biology, West Virginia University, Morgantown, WV, United States
| | - Logan Bays
- Department of Biology, West Virginia University, Morgantown, WV, United States
| | - Casey Showman
- Department of Biology, West Virginia University, Morgantown, WV, United States
| | - Sean Laverty
- Department of Mathematics and Statistics, University of Central Oklahoma, Edmond, OK, United States
| | - Sadie A. Bergeron
- Department of Biology, West Virginia University, Morgantown, WV, United States
- Department of Neuroscience, West Virginia University, Morgantown, WV, United States
| | - Eric J. Horstick
- Department of Biology, West Virginia University, Morgantown, WV, United States
- Department of Neuroscience, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
21
|
Uchigashima M, Cheung A, Futai K. Neuroligin-3: A Circuit-Specific Synapse Organizer That Shapes Normal Function and Autism Spectrum Disorder-Associated Dysfunction. Front Mol Neurosci 2021; 14:749164. [PMID: 34690695 PMCID: PMC8526735 DOI: 10.3389/fnmol.2021.749164] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/06/2021] [Indexed: 01/02/2023] Open
Abstract
Chemical synapses provide a vital foundation for neuron-neuron communication and overall brain function. By tethering closely apposed molecular machinery for presynaptic neurotransmitter release and postsynaptic signal transduction, circuit- and context- specific synaptic properties can drive neuronal computations for animal behavior. Trans-synaptic signaling via synaptic cell adhesion molecules (CAMs) serves as a promising mechanism to generate the molecular diversity of chemical synapses. Neuroligins (Nlgns) were discovered as postsynaptic CAMs that can bind to presynaptic CAMs like Neurexins (Nrxns) at the synaptic cleft. Among the four (Nlgn1-4) or five (Nlgn1-3, Nlgn4X, and Nlgn4Y) isoforms in rodents or humans, respectively, Nlgn3 has a heterogeneous expression and function at particular subsets of chemical synapses and strong association with non-syndromic autism spectrum disorder (ASD). Several lines of evidence have suggested that the unique expression and function of Nlgn3 protein underlie circuit-specific dysfunction characteristic of non-syndromic ASD caused by the disruption of Nlgn3 gene. Furthermore, recent studies have uncovered the molecular mechanism underlying input cell-dependent expression of Nlgn3 protein at hippocampal inhibitory synapses, in which trans-synaptic signaling of specific alternatively spliced isoforms of Nlgn3 and Nrxn plays a critical role. In this review article, we overview the molecular, anatomical, and physiological knowledge about Nlgn3, focusing on the circuit-specific function of mammalian Nlgn3 and its underlying molecular mechanism. This will provide not only new insight into specific Nlgn3-mediated trans-synaptic interactions as molecular codes for synapse specification but also a better understanding of the pathophysiological basis for non-syndromic ASD associated with functional impairment in Nlgn3 gene.
Collapse
Affiliation(s)
- Motokazu Uchigashima
- Department of Cellular Neuropathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Amy Cheung
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, United States
| | - Kensuke Futai
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
22
|
Drulis-Fajdasz D, Gostomska-Pampuch K, Duda P, Wiśniewski JR, Rakus D. Quantitative Proteomics Reveals Significant Differences between Mouse Brain Formations in Expression of Proteins Involved in Neuronal Plasticity during Aging. Cells 2021; 10:2021. [PMID: 34440790 PMCID: PMC8393337 DOI: 10.3390/cells10082021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/30/2021] [Accepted: 08/05/2021] [Indexed: 12/22/2022] Open
Abstract
Aging is associated with a general decline in cognitive functions, which appears to be due to alterations in the amounts of proteins involved in the regulation of synaptic plasticity. Here, we present a quantitative analysis of proteins involved in neurotransmission in three brain regions, namely, the hippocampus, the cerebral cortex and the cerebellum, in mice aged 1 and 22 months, using the total protein approach technique. We demonstrate that although the titer of some proteins involved in neurotransmission and synaptic plasticity is affected by aging in a similar manner in all the studied brain formations, in fact, each of the formations represents its own mode of aging. Generally, the hippocampal and cortical proteomes are much more unstable during the lifetime than the cerebellar proteome. The data presented here provide a general picture of the effect of physiological aging on synaptic plasticity and might suggest potential drug targets for anti-aging therapies.
Collapse
Affiliation(s)
- Dominika Drulis-Fajdasz
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (D.D.-F.); (P.D.)
| | - Kinga Gostomska-Pampuch
- Biochemical Proteomics Group, Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; (K.G.-P.); (J.R.W.)
- Department of Biochemistry and Immunochemistry, Wrocław Medical University, Chałubińskiego 10, 50-368 Wrocław, Poland
| | - Przemysław Duda
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (D.D.-F.); (P.D.)
| | - Jacek Roman Wiśniewski
- Biochemical Proteomics Group, Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; (K.G.-P.); (J.R.W.)
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (D.D.-F.); (P.D.)
| |
Collapse
|
23
|
Steffen DM, Ferri SL, Marcucci CG, Blocklinger KL, Molumby MJ, Abel T, Weiner JA. The γ-Protocadherins Interact Physically and Functionally with Neuroligin-2 to Negatively Regulate Inhibitory Synapse Density and Are Required for Normal Social Interaction. Mol Neurobiol 2021; 58:2574-2589. [PMID: 33471287 PMCID: PMC8137559 DOI: 10.1007/s12035-020-02263-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 12/15/2020] [Indexed: 12/16/2022]
Abstract
Cell adhesion molecules (CAMs) are key players in the formation of neural circuits during development. The γ-protocadherins (γ-Pcdhs), a family of 22 CAMs encoded by the Pcdhg gene cluster, are known to play important roles in dendrite arborization, axon targeting, and synapse development. We showed previously that multiple γ-Pcdhs interact physically with the autism-associated CAM neuroligin-1, and inhibit the latter's ability to promote excitatory synapse maturation. Here, we show that γ-Pcdhs can also interact physically with the related neuroligin-2, and inhibit this CAM's ability to promote inhibitory synapse development. In an artificial synapse assay, γ-Pcdhs co-expressed with neuroligin-2 in non-neuronal cells reduce inhibitory presynaptic maturation in contacting hippocampal axons. Mice lacking the γ-Pcdhs from the forebrain (including the cortex, the hippocampus, and portions of the amygdala) exhibit increased inhibitory synapse density and increased co-localization of neuroligin-2 with inhibitory postsynaptic markers in vivo. These Pcdhg mutants also exhibit defective social affiliation and an anxiety-like phenotype in behavioral assays. Together, these results suggest that γ-Pcdhs negatively regulate neuroligins to limit synapse density in a manner that is important for normal behavior.
Collapse
Affiliation(s)
- David M Steffen
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Department of Biology, The University of Iowa, Iowa City, IA, 52242, USA
| | - Sarah L Ferri
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, The University of Iowa, Iowa City, IA, 52242, USA
| | - Charles G Marcucci
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Department of Biology, The University of Iowa, Iowa City, IA, 52242, USA
| | - Kelsey L Blocklinger
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, The University of Iowa, Iowa City, IA, 52242, USA
| | - Michael J Molumby
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Department of Biology, The University of Iowa, Iowa City, IA, 52242, USA
| | - Ted Abel
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, The University of Iowa, Iowa City, IA, 52242, USA
| | - Joshua A Weiner
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA.
- Department of Biology, The University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
24
|
Li S, Liang X, Liang Y, Li L, Gan J, Cao L, Zou Y. Identification of the transcription factor, AFF4, as a new target of miR-203 in CNS. Int J Biol Macromol 2021; 181:919-927. [PMID: 33878354 DOI: 10.1016/j.ijbiomac.2021.04.089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 12/18/2022]
Abstract
MiR-203 was identified as a hub of a potential regulatory miRNA network in central nervous system. Overexpressing of miR-203 in the frontal cortex of C57BL/6J wild type mouse induced neurodegeneration by increasing the apoptotic pathway and neuron death. AFF4, a transcription factor, was identified as a new bona fida protein target of miR-203 in CNS. The miRNA:mRNA interaction of miR-203 and AFF4 was verified using Dural-luciferase assay. Down-regulated expression of AFF4 was induced by overexpressing miR-203 both in vitro and in vivo. Open field test, Y maze and Morris water maze test were conducted for the behavioral assessment of the mice with stereotactic injection of lentiviral vector overexpressing miR-203 in the hippocampus. No anxiety-like behavior or impaired cognition was noticed in these mice. Consistent with the results of the behavioral assessment, the electron micrograph and Nissl staining revealed no significant change in the synaptic density and no neuron injuries in the hippocampus of mice overexpressing miR-203, respectively. Our results indicated that instead of promoting neurodegenerative phenotype, a more profound function should be ascribed to miR-203 in regulating neuron behavioral activities and cognition. Neuron-type specific functions of miR-203 are likely to be executed via its various downstream protein interactors.
Collapse
Affiliation(s)
- Shufang Li
- The Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China
| | - Xiaosheng Liang
- The Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China
| | - Yaohui Liang
- The Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China
| | - Linpeng Li
- The Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China
| | - Jia Gan
- The Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China
| | - Lin Cao
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Yi Zou
- The Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China.
| |
Collapse
|
25
|
Yagishita-Kyo N, Ikai Y, Uekita T, Shinohara A, Koshimoto C, Yoshikawa K, Maruyama K, Yagishita S. Testosterone interrupts binding of Neurexin and Neuroligin that are expressed in a highly socialized rodent, Octodon degus. Biochem Biophys Res Commun 2021; 551:54-62. [PMID: 33721831 DOI: 10.1016/j.bbrc.2021.03.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/03/2021] [Indexed: 12/24/2022]
Abstract
Octodon degus is said to be one of the most human-like rodents because of its improved cognitive function. Focusing on its high sociality, we cloned and characterized some sociality-related genes of degus, in order to establish degus as a highly socialized animal model in molecular biology. We cloned degus Neurexin and Neuroligin as sociality-related genes, which are genetically related to autism spectrum disorder in human. According to our results, amino acid sequences of Neurexin and Neuroligin expressed in degus brain, are highly conserved to that of human sequences. Most notably, degus Neuroligin4 is highly similar to human Neuroligin4X, which is one of the most important autism-related genes, whereas mouse Neuroligin4 is known to be poorly similar to human Neuroligin4X. Furthermore, our work also indicated that testosterone directly binds to degus Neurexin and intercepts intercellular Neurexin-Neuroligin binding. Moreover, it is of high interest that testosterone is another key molecule of the higher incidence of autism in male. These results indicated that degus has the potential for animal model of sociality, and furthermore may promote understanding toward the pathogenic mechanism of autism.
Collapse
Affiliation(s)
- Nan Yagishita-Kyo
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan.
| | - Yuki Ikai
- Division of Bio-Resources, Department of Biotechnology, Frontier Science Research Center, University of Miyazaki, 5200 Kihara, Kiyotake-cho, Miyazaki-shi, Miyazaki, 889-1692, Japan
| | - Tomoko Uekita
- Department of Psychology, Faculty of Health Sciences, Kyoto Tachibana University, 34 Yamada-cho, Oyake, Yamashina-ku, Kyoto, 607-8175, Japan
| | - Akio Shinohara
- Division of Bio-Resources, Department of Biotechnology, Frontier Science Research Center, University of Miyazaki, 5200 Kihara, Kiyotake-cho, Miyazaki-shi, Miyazaki, 889-1692, Japan
| | - Chihiro Koshimoto
- Division of Bio-Resources, Department of Biotechnology, Frontier Science Research Center, University of Miyazaki, 5200 Kihara, Kiyotake-cho, Miyazaki-shi, Miyazaki, 889-1692, Japan
| | - Keisuke Yoshikawa
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan
| | - Kei Maruyama
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan
| | - Sosuke Yagishita
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan
| |
Collapse
|
26
|
Interplay between hevin, SPARC, and MDGAs: Modulators of neurexin-neuroligin transsynaptic bridges. Structure 2021; 29:664-678.e6. [PMID: 33535026 DOI: 10.1016/j.str.2021.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/20/2020] [Accepted: 01/08/2021] [Indexed: 12/18/2022]
Abstract
Hevin is secreted by astrocytes and its synaptogenic effects are antagonized by the related protein, SPARC. Hevin stabilizes neurexin-neuroligin transsynaptic bridges in vivo. A third protein, membrane-tethered MDGA, blocks these bridges. Here, we reveal the molecular underpinnings of a regulatory network formed by this trio of proteins. The hevin FS-EC structure differs from SPARC, in that the EC domain appears rearranged around a conserved core. The FS domain is structurally conserved and it houses nanomolar affinity binding sites for neurexin and neuroligin. SPARC also binds neurexin and neuroligin, competing with hevin, so its antagonist action is rooted in its shortened N-terminal region. Strikingly, the hevin FS domain competes with MDGA for an overlapping binding site on neuroligin, while the hevin EC domain binds the extracellular matrix protein collagen (like SPARC), so that this trio of proteins can regulate neurexin-neuroligin transsynaptic bridges and also extracellular matrix interactions, impacting synapse formation and ultimately neural circuits.
Collapse
|
27
|
Ali H, Marth L, Krueger-Burg D. Neuroligin-2 as a central organizer of inhibitory synapses in health and disease. Sci Signal 2020; 13:13/663/eabd8379. [DOI: 10.1126/scisignal.abd8379] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Postsynaptic organizational protein complexes play central roles both in orchestrating synapse formation and in defining the functional properties of synaptic transmission that together shape the flow of information through neuronal networks. A key component of these organizational protein complexes is the family of synaptic adhesion proteins called neuroligins. Neuroligins form transsynaptic bridges with presynaptic neurexins to regulate various aspects of excitatory and inhibitory synaptic transmission. Neuroligin-2 (NLGN2) is the only member that acts exclusively at GABAergic inhibitory synapses. Altered expression and mutations in NLGN2 and several of its interacting partners are linked to cognitive and psychiatric disorders, including schizophrenia, autism, and anxiety. Research on NLGN2 has fundamentally shaped our understanding of the molecular architecture of inhibitory synapses. Here, we discuss the current knowledge on the molecular and cellular functions of mammalian NLGN2 and its role in the neuronal circuitry that regulates behavior in rodents and humans.
Collapse
Affiliation(s)
- Heba Ali
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
- Göttingen Graduate School for Neurosciences, Biophysics, and Molecular Biosciences, University of Göttingen, 37077 Göttingen, Germany
| | - Lena Marth
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Dilja Krueger-Burg
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
28
|
Kishimoto Y, Hirono M, Atarashi R, Sakaguchi S, Yoshioka T, Katamine S, Kirino Y. Impairment of cerebellar long-term depression and GABAergic transmission in prion protein deficient mice ectopically expressing PrPLP/Dpl. Sci Rep 2020; 10:15900. [PMID: 32985542 PMCID: PMC7522223 DOI: 10.1038/s41598-020-72753-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/27/2020] [Indexed: 12/22/2022] Open
Abstract
Prion protein (PrPC) knockout mice, named as the “Ngsk” strain (Ngsk Prnp0/0 mice), show late-onset cerebellar Purkinje cell (PC) degeneration because of ectopic overexpression of PrPC-like protein (PrPLP/Dpl). Our previous study indicated that the mutant mice also exhibited alterations in cerebellum-dependent delay eyeblink conditioning, even at a young age (16 weeks of age) when neurological changes had not occurred. Thus, this electrophysiological study was designed to examine the synaptic function of the cerebellar cortex in juvenile Ngsk Prnp0/0 mice. We showed that Ngsk Prnp0/0 mice exhibited normal paired-pulse facilitation but impaired long-term depression of excitatory synaptic transmission at synapses between parallel fibres and PCs. GABAA-mediated inhibitory postsynaptic currents recorded from PCs were also weakened in Ngsk Prnp0/0 mice. Furthermore, we confirmed that Ngsk Prnp0/0 mice (7–8-week-old) exhibited abnormalities in delay eyeblink conditioning. Our findings suggest that these alterations in both excitatory and inhibitory synaptic transmission to PCs caused deficits in delay eyeblink conditioning of Ngsk Prnp0/0 mice. Therefore, the Ngsk Prnp0/0 mouse model can contribute to study underlying mechanisms for impairments of synaptic transmission and neural plasticity, and cognitive deficits in the central nervous system.
Collapse
Affiliation(s)
- Yasushi Kishimoto
- Laboratory of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Kagawa, 769-2193, Japan.
| | - Moritoshi Hirono
- Department of Physiology, Faculty of Medicine, Wakayama Medical University School of Medicine, Wakayama, 641-8509, Japan.
| | - Ryuichiro Atarashi
- Division of Microbiology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Suehiro Sakaguchi
- Division of Molecular Neurobiology, Institute for Enzyme Research (KOSOKEN), Tokushima University, Tokushima, 770-8501, Japan
| | - Tohru Yoshioka
- Center of Excellence for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Shigeru Katamine
- Center for International Collaborative Research, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Yutaka Kirino
- Laboratory of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Kagawa, 769-2193, Japan
| |
Collapse
|
29
|
Platsaki S, Zhou X, Pinan-Lucarré B, Delauzun V, Tu H, Mansuelle P, Fourquet P, Bourne Y, Bessereau JL, Marchot P. The Ig-like domain of Punctin/MADD-4 is the primary determinant for interaction with the ectodomain of neuroligin NLG-1. J Biol Chem 2020; 295:16267-16279. [PMID: 32928959 DOI: 10.1074/jbc.ra120.014591] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/02/2020] [Indexed: 02/01/2023] Open
Abstract
Punctin/MADD-4, a member of the ADAMTSL extracellular matrix protein family, was identified as an anterograde synaptic organizer in the nematode Caenorhabditis elegans. At GABAergic neuromuscular junctions, the short isoform MADD-4B binds the ectodomain of neuroligin NLG-1, itself a postsynaptic organizer of inhibitory synapses. To identify the molecular bases of their partnership, we generated recombinant forms of the two proteins and carried out a comprehensive biochemical and biophysical study of their interaction, complemented by an in vivo localization study. We show that spontaneous proteolysis of MADD-4B first generates a shorter N-MADD-4B form, which comprises four thrombospondin (TSP) domains and one Ig-like domain and binds NLG-1. A second processing event eliminates the C-terminal Ig-like domain along with the ability of N-MADD-4B to bind NLG-1. These data identify the Ig-like domain as the primary determinant for N-MADD-4B interaction with NLG-1 in vitro We further demonstrate in vivo that this Ig-like domain is essential, albeit not sufficient per se, for efficient recruitment of GABAA receptors at GABAergic synapses in C. elegans The interaction of N-MADD-4B with NLG-1 is also disrupted by heparin, used as a surrogate for the extracellular matrix component, heparan sulfate. High-affinity binding of heparin/heparan sulfate to the Ig-like domain may proceed from surface charge complementarity, as suggested by homology three-dimensional modeling. These data point to N-MADD-4B processing and cell-surface proteoglycan binding as two possible mechanisms to regulate the interaction between MADD-4B and NLG-1 at GABAergic synapses.
Collapse
Affiliation(s)
- Semeli Platsaki
- CNRS/Aix-Marseille Univ, Laboratory "Architecture et Fonction des Macromolécules Biologiques" (AFMB), Marseille, France
| | - Xin Zhou
- Univ Lyon/Univ Claude Bernard Lyon 1/CNRS/INSERM, Institut NeuroMyoGène (INMG), Lyon, France
| | - Bérangère Pinan-Lucarré
- Univ Lyon/Univ Claude Bernard Lyon 1/CNRS/INSERM, Institut NeuroMyoGène (INMG), Lyon, France
| | - Vincent Delauzun
- CNRS/Aix-Marseille Univ, Laboratory "Architecture et Fonction des Macromolécules Biologiques" (AFMB), Marseille, France
| | - Haijun Tu
- Univ Lyon/Univ Claude Bernard Lyon 1/CNRS/INSERM, Institut NeuroMyoGène (INMG), Lyon, France
| | - Pascal Mansuelle
- CNRS/Aix-Marseille Univ, Institut de Microbiologie de la Méditerranée (IMM), Marseille Proteomics (MaP), Marseille, France
| | - Patrick Fourquet
- Aix-Marseille Univ/INSERM/CNRS, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), Marseille Proteomics (MaP), Marseille, France
| | - Yves Bourne
- CNRS/Aix-Marseille Univ, Laboratory "Architecture et Fonction des Macromolécules Biologiques" (AFMB), Marseille, France
| | - Jean-Louis Bessereau
- Univ Lyon/Univ Claude Bernard Lyon 1/CNRS/INSERM, Institut NeuroMyoGène (INMG), Lyon, France
| | - Pascale Marchot
- CNRS/Aix-Marseille Univ, Laboratory "Architecture et Fonction des Macromolécules Biologiques" (AFMB), Marseille, France.
| |
Collapse
|
30
|
Fertan E, Wong AA, Purdon MK, Weaver ICG, Brown RE. The effect of background strain on the behavioral phenotypes of the MDGA2 +/- mouse model of autism spectrum disorder. GENES BRAIN AND BEHAVIOR 2020; 20:e12696. [PMID: 32808443 DOI: 10.1111/gbb.12696] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/27/2020] [Accepted: 08/14/2020] [Indexed: 12/26/2022]
Abstract
The membrane-associated mucin (MAM) domain containing glycosylphosphatidylinositol anchor 2 protein single knock-out mice (MDGA2+/- ) are models of ASD. We examined the behavioral phenotypes of male and female MDGA2+/- and wildtype mice on C57BL6/NJ and C57BL6/N backgrounds at 2 months of age and measured MDGA2, neuroligin 1 and neuroligin 2 levels at 7 months. Mice on the C57BL6/NJ background performed better than those on the C57BL6/N background in visual ability and in learning and memory performance in the Morris water maze and differed in measures of motor behavior and anxiety. Mice with the MDGA2+/- genotype differed from WT mice in motor, social and repetitive behavior and anxiety, but most of these effects involved interactions between MDGA2+/- genotype and background strain. The background strain also influenced MDGA2 levels and NLGN2 association in MDGA2+/- mice. Our findings emphasize the importance of the background strain used in studies of genetically modified mice.
Collapse
Affiliation(s)
- Emre Fertan
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Aimée A Wong
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michaela K Purdon
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ian C G Weaver
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.,Brain Repair Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Brain Repair Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
31
|
Qin L, Guo S, Han Y, Wang X, Zhang B. Functional mosaic organization of neuroligins in neuronal circuits. Cell Mol Life Sci 2020; 77:3117-3127. [PMID: 32077971 PMCID: PMC11104838 DOI: 10.1007/s00018-020-03478-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 11/30/2022]
Abstract
Complex brain circuitry with feedforward and feedback systems regulates neuronal activity, enabling neural networks to process and drive the entire spectrum of cognitive, behavioral, sensory, and motor functions. Simultaneous orchestration of distinct cells and interconnected neural circuits is underpinned by hundreds of synaptic adhesion molecules that span synaptic junctions. Dysfunction of a single molecule or molecular interaction at synapses can lead to disrupted circuit activity and brain disorders. Neuroligins, a family of cell adhesion molecules, were first identified as postsynaptic-binding partners of presynaptic neurexins and are essential for synapse specification and maturation. Here, we review recent advances in our understanding of how this family of adhesion molecules controls neuronal circuit assembly by acting in a synapse-specific manner.
Collapse
Affiliation(s)
- Liming Qin
- School of Chemical Biology and Biotechnology, Shenzhen Bay Laboratory, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Sile Guo
- School of Chemical Biology and Biotechnology, Shenzhen Bay Laboratory, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Ying Han
- School of Chemical Biology and Biotechnology, Shenzhen Bay Laboratory, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Xiankun Wang
- School of Chemical Biology and Biotechnology, Shenzhen Bay Laboratory, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Bo Zhang
- School of Chemical Biology and Biotechnology, Shenzhen Bay Laboratory, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| |
Collapse
|
32
|
Janickova L, Rechberger KF, Wey L, Schwaller B. Absence of parvalbumin increases mitochondria volume and branching of dendrites in inhibitory Pvalb neurons in vivo: a point of convergence of autism spectrum disorder (ASD) risk gene phenotypes. Mol Autism 2020; 11:47. [PMID: 32517751 PMCID: PMC7285523 DOI: 10.1186/s13229-020-00323-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/27/2020] [Indexed: 01/01/2023] Open
Abstract
Background In fast firing, parvalbumin (PV)-expressing (Pvalb) interneurons, PV acts as an intracellular Ca2+ signal modulator with slow-onset kinetics. In Purkinje cells of PV−/− mice, adaptive/homeostatic mechanisms lead to an increase in mitochondria, organelles equally capable of delayed Ca2+ sequestering/buffering. An inverse regulation of PV and mitochondria likewise operates in cell model systems in vitro including myotubes, epithelial cells, and oligodendrocyte-like cells overexpressing PV. Whether such opposite regulation pertains to all Pvalb neurons is currently unknown. In oligodendrocyte-like cells, PV additionally decreases growth and branching of processes in a cell-autonomous manner. Methods The in vivo effects of absence of PV were investigated in inhibitory Pvalb neurons expressing EGFP, present in the somatosensory and medial prefrontal cortex, striatum, thalamic reticular nucleus, hippocampal regions DG, CA3, and CA1 and cerebellum of mice either wild-type or knockout (PV−/−) for the Pvalb gene. Changes in Pvalb neuron morphology and PV concentrations were determined using immunofluorescence, followed by 3D-reconstruction and quantitative image analyses. Results PV deficiency led to an increase in mitochondria volume and density in the soma; the magnitude of the effect was positively correlated with the estimated PV concentrations in the various Pvalb neuron subpopulations in wild-type neurons. The increase in dendrite length and branching, as well as thickness of proximal dendrites of selected PV−/− Pvalb neurons is likely the result of the observed increased density and length of mitochondria in these PV−/− Pvalb neuron dendrites. The increased branching and soma size directly linked to the absence of PV is assumed to contribute to the increased volume of the neocortex present in juvenile PV−/− mice. The extended dendritic branching is in line with the hypothesis of local hyperconnectivity in autism spectrum disorder (ASD) and ASD mouse models including PV−/− mice, which display all ASD core symptoms and several comorbidities including cortical macrocephaly at juvenile age. Conclusion PV is involved in most proposed mechanisms implicated in ASD etiology: alterations in Ca2+ signaling affecting E/I balance, changes in mitochondria structure/function, and increased dendritic length and branching, possibly resulting in local hyperconnectivity, all in a likely cell autonomous way.
Collapse
Affiliation(s)
- Lucia Janickova
- Anatomy, Section of Medicine, University of Fribourg, Route Albert-Gockel 1, 1700, Fribourg, Switzerland
| | - Karin Farah Rechberger
- Anatomy, Section of Medicine, University of Fribourg, Route Albert-Gockel 1, 1700, Fribourg, Switzerland
| | - Lucas Wey
- Anatomy, Section of Medicine, University of Fribourg, Route Albert-Gockel 1, 1700, Fribourg, Switzerland
| | - Beat Schwaller
- Anatomy, Section of Medicine, University of Fribourg, Route Albert-Gockel 1, 1700, Fribourg, Switzerland.
| |
Collapse
|
33
|
Zhang L, Qin Z, Ricke KM, Cruz SA, Stewart AFR, Chen HH. Hyperactivated PTP1B phosphatase in parvalbumin neurons alters anterior cingulate inhibitory circuits and induces autism-like behaviors. Nat Commun 2020; 11:1017. [PMID: 32094367 PMCID: PMC7039907 DOI: 10.1038/s41467-020-14813-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 02/05/2020] [Indexed: 01/05/2023] Open
Abstract
Individuals with autism spectrum disorder (ASD) have social interaction deficits and difficulty filtering information. Inhibitory interneurons filter information at pyramidal neurons of the anterior cingulate cortex (ACC), an integration hub for higher-order thalamic inputs important for social interaction. Humans with deletions including LMO4, an endogenous inhibitor of PTP1B, display intellectual disabilities and occasionally autism. PV-Lmo4KO mice ablate Lmo4 in PV interneurons and display ASD-like repetitive behaviors and social interaction deficits. Surprisingly, increased PV neuron-mediated peri-somatic feedforward inhibition to the pyramidal neurons causes a compensatory reduction in (somatostatin neuron-mediated) dendritic inhibition. These homeostatic changes increase filtering of mediodorsal-thalamocortical inputs but reduce filtering of cortico-cortical inputs and narrow the range of stimuli ACC pyramidal neurons can distinguish. Simultaneous ablation of PTP1B in PV-Lmo4KO neurons prevents these deficits, indicating that PTP1B activation in PV interneurons contributes to ASD-like characteristics and homeostatic maladaptation of inhibitory circuits may contribute to deficient information filtering in ASD.
Collapse
Affiliation(s)
- Li Zhang
- Ottawa Hospital Research Institute, Neuroscience, Ottawa, Canada. .,University of Ottawa Brain and Mind Institute, Ottawa, Canada.
| | - Zhaohong Qin
- Ottawa Hospital Research Institute, Neuroscience, Ottawa, Canada.,University of Ottawa Brain and Mind Institute, Ottawa, Canada
| | - Konrad M Ricke
- Ottawa Hospital Research Institute, Neuroscience, Ottawa, Canada.,University of Ottawa Brain and Mind Institute, Ottawa, Canada.,University of Ottawa Heart Institute, Ottawa, Canada.,Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | - Shelly A Cruz
- Ottawa Hospital Research Institute, Neuroscience, Ottawa, Canada.,University of Ottawa Brain and Mind Institute, Ottawa, Canada
| | - Alexandre F R Stewart
- University of Ottawa Heart Institute, Ottawa, Canada. .,Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada. .,Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Canada.
| | - Hsiao-Huei Chen
- Ottawa Hospital Research Institute, Neuroscience, Ottawa, Canada. .,University of Ottawa Brain and Mind Institute, Ottawa, Canada. .,Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Canada. .,Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada. .,Medicine, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
34
|
Hossain MR, Jamal M, Tanoue Y, Ojima D, Takahashi H, Kubota T, Ansary TM, Ito A, Tanaka N, Kinoshita H, Kishimoto Y, Yamamoto T. MDGA1-deficiency attenuates prepulse inhibition with alterations of dopamine and serotonin metabolism: An ex vivo HPLC-ECD analysis. Neurosci Lett 2020; 716:134677. [PMID: 31812551 DOI: 10.1016/j.neulet.2019.134677] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 10/25/2022]
Abstract
MDGA1 (MAM domain-containing glycosylphosphatidylinositol anchor) has recently been linked to schizophrenia and bipolar disorder. Dysregulation of dopamine (DA) and serotonin (5-HT) systems has long been associated with schizophrenia and other neuropsychiatric disorders. Here, we measured prepulse inhibition (PPI) of the startle response and ex vivo tissue content of monoamines and their metabolites in the frontal cortex, striatum and hippocampus of Mdga1 homozygous (Mdga1-KO), Mdga1 heterozygous (Mdga1-HT) and wild-type (WT) male mice. We found that Mdga1-KO mice exhibited statistically significant impairment of PPI, and had higher levels of homovanillic acid in all three brain regions studied compared with Mdga1-HT and WT mice (P < 0.05), while levels of norepinephrine, DA and its metabolites 3,4-dihydroxyphenylacetic acid and 3-methoxytyramine remained unchanged. Mdga1-KO mice also had a lower 5-hydroxyindoleacetic acid level in the striatum (P < 0.05) compared with WT mice. 5-HT levels remained unchanged with the exception of a significant increase in the level in the cortex. These data are the first evidence suggesting that MDGA1 deficiency leads to a pronounced deficit in PPI and plays an important role in perturbation of DA and 5-HT metabolism in mouse brain; such changes may contribute to a range of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Md Razib Hossain
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Mostofa Jamal
- Department of Forensic Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan.
| | - Yu Tanoue
- Department of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa, Japan
| | - Daiki Ojima
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Hiroo Takahashi
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takashi Kubota
- Department of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa, Japan
| | - Tuba M Ansary
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Asuka Ito
- Department of Forensic Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Naoko Tanaka
- Department of Forensic Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Hiroshi Kinoshita
- Department of Forensic Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yasushi Kishimoto
- Department of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa, Japan
| | - Tohru Yamamoto
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan.
| |
Collapse
|
35
|
Du Y, Du Y, Zhang Y, Huang Z, Fu M, Li J, Pang Y, Lei P, Wang YT, Song W, He G, Dong Z. MKP-1 reduces Aβ generation and alleviates cognitive impairments in Alzheimer's disease models. Signal Transduct Target Ther 2019; 4:58. [PMID: 31840000 PMCID: PMC6895219 DOI: 10.1038/s41392-019-0091-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/01/2019] [Accepted: 11/01/2019] [Indexed: 12/23/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) phosphatase 1 (MKP-1) is an essential negative regulator of MAPKs by dephosphorylating MAPKs at both tyrosine and threonine residues. Dysregulation of the MAPK signaling pathway has been associated with Alzheimer's disease (AD). However, the role of MKP-1 in AD pathogenesis remains elusive. Here, we report that MKP-1 levels were decreased in the brain tissues of patients with AD and an AD mouse model. The reduction in MKP-1 gene expression appeared to be a result of transcriptional inhibition via transcription factor specificity protein 1 (Sp1) cis-acting binding elements in the MKP-1 gene promoter. Amyloid-β (Aβ)-induced Sp1 activation decreased MKP-1 expression. However, upregulation of MKP-1 inhibited the expression of both Aβ precursor protein (APP) and β-site APP-cleaving enzyme 1 by inactivating the extracellular signal-regulated kinase 1/2 (ERK)/MAPK signaling pathway. Furthermore, upregulation of MKP-1 reduced Aβ production and plaque formation and improved hippocampal long-term potentiation (LTP) and cognitive deficits in APP/PS1 transgenic mice. Our results demonstrate that MKP-1 impairment facilitates the pathogenesis of AD, whereas upregulation of MKP-1 plays a neuroprotective role to reduce Alzheimer-related phenotypes. Thus, this study suggests that MKP-1 is a novel molecule for AD treatment.
Collapse
Affiliation(s)
- Yehong Du
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
| | - Yexiang Du
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, 400016 PR China
| | - Yun Zhang
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Zhilin Huang
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
| | - Min Fu
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
| | - Junjie Li
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
| | - Yayan Pang
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
| | - Peng Lei
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041 Sichuan China
| | - Yu Tian Wang
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
- Brain Research Centre, The University of British Columbia, Vancouver, BC V6T 2B5 Canada
| | - Weihong Song
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Guiqiong He
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, 400016 PR China
| | - Zhifang Dong
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
| |
Collapse
|
36
|
A negative regulator of synaptic development: MDGA and its links to neurodevelopmental disorders. World J Pediatr 2019; 15:415-421. [PMID: 30997654 DOI: 10.1007/s12519-019-00253-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 03/28/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Formation of protein complexes across synapses is a critical process in neurodevelopment, having direct implications on brain function and animal behavior. Here, we present the understanding, importance, and potential impact of a newly found regulator of such a key interaction. DATA SOURCES A systematic search of the literature was conducted on PubMed (Medline), Embase, and Central-Cochrane Database. RESULTS Membrane-associated mucin domain-containing glycosylphosphatidylinositol anchor proteins (MDGAs) were recently discovered to regulate synaptic development and transmission via suppression of neurexins-neuroligins trans-synaptic complex formation. MDGAs also regulate axonal migration and outgrowth. In the context of their physiological role, we begin to consider the potential links to the etiology of certain neurodevelopmental disorders. We present the gene expression and protein structure of MDGAs and discuss recent progress in our understanding of the neurobiological role of MDGAs to explore its potential as a therapeutic target. CONCLUSION MDGAs play a key role in neuron migration, axon guidance and synapse development, as well as in regulating brain excitation and inhibition balance.
Collapse
|
37
|
Connor SA, Ammendrup-Johnsen I, Kishimoto Y, Karimi Tari P, Cvetkovska V, Harada T, Ojima D, Yamamoto T, Wang YT, Craig AM. Loss of Synapse Repressor MDGA1 Enhances Perisomatic Inhibition, Confers Resistance to Network Excitation, and Impairs Cognitive Function. Cell Rep 2019; 21:3637-3645. [PMID: 29281813 DOI: 10.1016/j.celrep.2017.11.109] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 10/10/2017] [Accepted: 11/29/2017] [Indexed: 01/27/2023] Open
Abstract
Synaptopathies contributing to neurodevelopmental disorders are linked to mutations in synaptic organizing molecules, including postsynaptic neuroligins, presynaptic neurexins, and MDGAs, which regulate their interaction. The role of MDGA1 in suppressing inhibitory versus excitatory synapses is controversial based on in vitro studies. We show that genetic deletion of MDGA1 in vivo elevates hippocampal CA1 inhibitory, but not excitatory, synapse density and transmission. Furthermore, MDGA1 is selectively expressed by pyramidal neurons and regulates perisomatic, but not distal dendritic, inhibitory synapses. Mdga1-/- hippocampal networks demonstrate muted responses to neural excitation, and Mdga1-/- mice are resistant to induced seizures. Mdga1-/- mice further demonstrate compromised hippocampal long-term potentiation, consistent with observed deficits in spatial and context-dependent learning and memory. These results suggest that mutations in MDGA1 may contribute to cognitive deficits through altered synaptic transmission and plasticity by loss of suppression of inhibitory synapse development in a subcellular domain- and cell-type-selective manner.
Collapse
Affiliation(s)
- Steven A Connor
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, BC V6T 2B5, Canada; Djavad Mowafaghian Centre for Brain Health and Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Ina Ammendrup-Johnsen
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Yasushi Kishimoto
- Department of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Kagawa 769-2101, Japan
| | - Parisa Karimi Tari
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Vedrana Cvetkovska
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Takashi Harada
- Department of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Kagawa 769-2101, Japan
| | - Daiki Ojima
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa 761-0793, Japan
| | - Tohru Yamamoto
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa 761-0793, Japan
| | - Yu Tian Wang
- Djavad Mowafaghian Centre for Brain Health and Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Ann Marie Craig
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, BC V6T 2B5, Canada.
| |
Collapse
|
38
|
Traumatic Brain Injury by Weight-Drop Method Causes Transient Amyloid- β Deposition and Acute Cognitive Deficits in Mice. Behav Neurol 2019; 2019:3248519. [PMID: 30944661 PMCID: PMC6421814 DOI: 10.1155/2019/3248519] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/11/2018] [Accepted: 01/09/2019] [Indexed: 11/17/2022] Open
Abstract
There has been growing awareness of the correlation between an episode of traumatic brain injury (TBI) and the development of Alzheimer's disease (AD) later in life. It has been reported that TBI accelerated amyloid-β (Aβ) pathology and cognitive decline in the several lines of AD model mice. However, the short-term and long-term effects of TBI by the weight-drop method on amyloid-β pathology and cognitive performance are unclear in wild-type (WT) mice. Hence, we examined AD-related histopathological changes and cognitive impairment after TBI in wild-type C57BL6J mice. Five- to seven-month-old WT mice were subjected to either TBI by the weight-drop method or a sham treatment. Seven days after TBI, the WT mice exhibited significantly lower spatial learning than the sham-treated WT mice. However, 28 days after TBI, the cognitive impairment in the TBI-treated WT mice recovered. Correspondingly, while significant amyloid-β (Aβ) plaques and amyloid precursor protein (APP) accumulation were observed in the TBI-treated mouse hippocampus 7 days after TBI, the Aβ deposition was no longer apparent 28 days after TBI. Thus, TBI induced transient amyloid-β deposition and acute cognitive impairments in the WT mice. The present study suggests that the TBI could be a risk factor for acute cognitive impairment even when genetic and hereditary predispositions are not involved. The system might be useful for evaluating and developing a pharmacological treatment for the acute cognitive deficits.
Collapse
|
39
|
Chamma I, Sainlos M, Thoumine O. Biophysical mechanisms underlying the membrane trafficking of synaptic adhesion molecules. Neuropharmacology 2019; 169:107555. [PMID: 30831159 DOI: 10.1016/j.neuropharm.2019.02.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 02/14/2019] [Accepted: 02/27/2019] [Indexed: 01/13/2023]
Abstract
Adhesion proteins play crucial roles at synapses, not only by providing a physical trans-synaptic linkage between axonal and dendritic membranes, but also by connecting to functional elements including the pre-synaptic neurotransmitter release machinery and post-synaptic receptors. To mediate these functions, adhesion proteins must be organized on the neuronal surface in a precise and controlled manner. Recent studies have started to describe the mobility, nanoscale organization, and turnover rate of key synaptic adhesion molecules including cadherins, neurexins, neuroligins, SynCAMs, and LRRTMs, and show that some of these proteins are highly mobile in the plasma membrane while others are confined at sub-synaptic compartments, providing evidence for different regulatory pathways. In this review article, we provide a biophysical view of the diffusional trapping of adhesion molecules at synapses, involving both extracellular and intracellular protein interactions. We review the methodology underlying these measurements, including biomimetic systems with purified adhesion proteins, means to perturb protein expression or function, single molecule imaging in cultured neurons, and analytical models to interpret the data. This article is part of the special issue entitled 'Mobility and trafficking of neuronal membrane proteins'.
Collapse
Affiliation(s)
- Ingrid Chamma
- Univ. Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France
| | - Matthieu Sainlos
- Univ. Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France
| | - Olivier Thoumine
- Univ. Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France.
| |
Collapse
|
40
|
Connor SA, Elegheert J, Xie Y, Craig AM. Pumping the brakes: suppression of synapse development by MDGA-neuroligin interactions. Curr Opin Neurobiol 2019; 57:71-80. [PMID: 30771697 DOI: 10.1016/j.conb.2019.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/03/2019] [Accepted: 01/04/2019] [Indexed: 12/22/2022]
Abstract
Synapse development depends on a dynamic balance between synapse promoters and suppressors. MDGAs, immunoglobulin superfamily proteins, negatively regulate synapse development through blocking neuroligin-neurexin interactions. Recent analyses of MDGA-neuroligin complexes revealed the structural basis of this activity and indicate that MDGAs interact with all neuroligins with differential affinities. Surprisingly, analyses of mouse mutants revealed a functional divergence, with targeted mutation of Mdga1 and Mdga2 elevating inhibitory and excitatory synapses, respectively, on hippocampal pyramidal neurons. Further research is needed to determine the synapse-specific organizing properties of MDGAs in neural circuits, which may depend on relative levels and subcellular distributions of each MDGA, neuroligin and neurexin. Behavioral deficits in Mdga mutant mice support genetic links to schizophrenia and autism spectrum disorders and raise the possibility of harnessing these interactions for therapeutic purposes.
Collapse
Affiliation(s)
- Steven A Connor
- Department of Biology, York University, 4700 Keele Street, Toronto, ON, M3J 1P3, Canada.
| | - Jonathan Elegheert
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297 and University of Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Yicheng Xie
- The Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Ann Marie Craig
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada.
| |
Collapse
|
41
|
Su S, Li H, Du F, Zhang C, Li X, Jing X, Liu L, Li Z, Yang X, Xu P, Yuan X, Zhu J, Bouzoualegh R. Combined QTL and Genome Scan Analyses With the Help of 2b-RAD Identify Growth-Associated Genetic Markers in a New Fast-Growing Carp Strain. Front Genet 2018; 9:592. [PMID: 30581452 PMCID: PMC6293859 DOI: 10.3389/fgene.2018.00592] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 11/15/2018] [Indexed: 11/17/2022] Open
Abstract
Common carp is one of the oldest and most popular cultured freshwater fish species both globally and in China. In a previous study, we used a carp strain with a long breeding tradition in China, named Huanghe, to create a new fast-growing strain by selection for fast growth for 6 years. The growth performance at 8 months of age has been improved by 20.84%. To achieve this, we combined the best linear unbiased prediction with marker-assisted selection techniques. Recent progress in genome-wide association studies and genomic selection in livestock breeding inspired common carp breeders to consider genome-based breeding approaches. In this study, we developed a 2b-RAD sequence assay as a means of investigating the quantitative trait loci in common carp. A total of 4,953,017,786 clean reads were generated for 250 specimens (average reads/specimen = 19,812,071) with BsaXI Restriction Enzyme. From these, 56,663 SNPs were identified, covering 50 chromosomes and 3,377 scaffolds. Principal component analysis indicated that selection and control groups are relatively clearly distinct. Top 1% of Fst values was selected as the threshold signature of artificial selection. Among the 244 identified loci, genes associated with sex-related factors and nutritional metabolism (especially fat metabolism) were annotated. Eighteen QTL were associated with growth parameters. Body length at 3 months of age and body weight (both at 3 and 8 months) were controlled by polygenic effects, but body size (length, depth, width) at 8 months of age was controlled mainly by several loci with major effects. Importantly, a single shared QTL (IGF2 gene) partially controlled the body length, depth, and width. By merging the above results, we concluded that mainly the genes related to neural pathways, sex and fatty acid metabolism contributed to the improved growth performance of the new Huanghe carp strain. These findings are one of the first investigations into the potential use of genomic selection in the breeding of common carp. Moreover, our results show that combining the Fst, QTL mapping and CRISPR–Cas9 methods can be an effective way to identify important novel candidate molecular markers in economic breeding programs.
Collapse
Affiliation(s)
- Shengyan Su
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China.,Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Hengde Li
- Ministry of Agriculture Key Laboratory of Aquatic Genomics, CAFS Key Laboratory of Aquatic Genomics and Beijing Key Laboratory of Fishery Biotechnology, Center for Applied Aquatic Genomics, Chinese Academy of Fishery Sciences, Beijing, China
| | - Fukuan Du
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Chengfeng Zhang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China.,Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Xinyuan Li
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Xiaojun Jing
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China.,Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Liyue Liu
- China Zebrafish Resource Center, Wuhan, China
| | - Zhixun Li
- Henan Academy of Fishery Sciences, Zhengzhou, China
| | - Xingli Yang
- Henan Academy of Fishery Sciences, Zhengzhou, China
| | - Pao Xu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China.,Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Xinhua Yuan
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China.,Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Jian Zhu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China.,Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Raouf Bouzoualegh
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| |
Collapse
|
42
|
Vevera J, Zarrei M, Hartmannová H, Jedličková I, Mušálková D, Přistoupilová A, Oliveriusová P, Trešlová H, Nosková L, Hodaňová K, Stránecký V, Jiřička V, Preiss M, Příhodová K, Šaligová J, Wei J, Woodbury-Smith M, Bleyer AJ, Scherer SW, Kmoch S. Rare copy number variation in extremely impulsively violent males. GENES BRAIN AND BEHAVIOR 2018; 18:e12536. [DOI: 10.1111/gbb.12536] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Jan Vevera
- Department of Psychiatry; Faculty of Medicine and University Hospital in Pilsen, Charles University; Prague Czech Republic
- Department of Psychiatry, First Faculty of Medicine; Charles University and General University Hospital in Prague; Prague Czech Republic
- Institute for Postgraduate Medical Education; Prague Czech Republic
- Psychology Department; National Institute of Mental Health; Klecany Czech Republic
| | - Mehdi Zarrei
- The Centre for Applied Genomics and Program in Genetics and Genome Biology; The Hospital for Sick Children; Toronto Ontario Canada
| | - Hana Hartmannová
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine; First Faculty of Medicine, Charles University; Prague Czech Republic
| | - Ivana Jedličková
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine; First Faculty of Medicine, Charles University; Prague Czech Republic
| | - Dita Mušálková
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine; First Faculty of Medicine, Charles University; Prague Czech Republic
| | - Anna Přistoupilová
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine; First Faculty of Medicine, Charles University; Prague Czech Republic
| | - Petra Oliveriusová
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine; First Faculty of Medicine, Charles University; Prague Czech Republic
| | - Helena Trešlová
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine; First Faculty of Medicine, Charles University; Prague Czech Republic
| | - Lenka Nosková
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine; First Faculty of Medicine, Charles University; Prague Czech Republic
| | - Kateřina Hodaňová
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine; First Faculty of Medicine, Charles University; Prague Czech Republic
| | - Viktor Stránecký
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine; First Faculty of Medicine, Charles University; Prague Czech Republic
| | - Václav Jiřička
- Prison Service of the Czech Republic, Directorate General; Department of Psychology; Prague Czech Republic
| | - Marek Preiss
- Psychology Department; National Institute of Mental Health; Klecany Czech Republic
- Psychology Department; University of New York in Prague; Prague Czech Republic
| | - Kateřina Příhodová
- Psychology Department; National Institute of Mental Health; Klecany Czech Republic
| | - Jana Šaligová
- Children's Faculty Hospital; Department of Pediatrics and Adolescent Medicine; Kosice Slovakia
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine of Pavel Jozef Šafárik University Kosice; Kosice Slovakia
| | - John Wei
- The Centre for Applied Genomics and Program in Genetics and Genome Biology; The Hospital for Sick Children; Toronto Ontario Canada
| | - Marc Woodbury-Smith
- The Centre for Applied Genomics and Program in Genetics and Genome Biology; The Hospital for Sick Children; Toronto Ontario Canada
- Institute of Neuroscience, Newcastle University, Sir James Spence Institute, Royal Victoria Infirmary; Newcastle upon Tyne UK
| | - Anthony J. Bleyer
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine; First Faculty of Medicine, Charles University; Prague Czech Republic
- Section on Nephrology, Wake Forest School of Medicine; Medical Center Blvd.; Winston-Salem North Carolina USA
| | - Stephen W. Scherer
- The Centre for Applied Genomics and Program in Genetics and Genome Biology; The Hospital for Sick Children; Toronto Ontario Canada
- Department of Molecular Genetics and McLaughlin Centre; University of Toronto; Toronto Ontario Canada
| | - Stanislav Kmoch
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine; First Faculty of Medicine, Charles University; Prague Czech Republic
| |
Collapse
|
43
|
Abstract
Synapse formation is mediated by a surprisingly large number and wide variety of genes encoding many different protein classes. One of the families increasingly implicated in synapse wiring is the immunoglobulin superfamily (IgSF). IgSF molecules are by definition any protein containing at least one Ig-like domain, making this family one of the most common protein classes encoded by the genome. Here, we review the emerging roles for IgSF molecules in synapse formation specifically in the vertebrate brain, focusing on examples from three classes of IgSF members: ( a) cell adhesion molecules, ( b) signaling molecules, and ( c) immune molecules expressed in the brain. The critical roles for IgSF members in regulating synapse formation may explain their extensive involvement in neuropsychiatric and neurodevelopmental disorders. Solving the IgSF code for synapse formation may reveal multiple new targets for rescuing IgSF-mediated deficits in synapse formation and, eventually, new treatments for psychiatric disorders caused by altered IgSF-induced synapse wiring.
Collapse
Affiliation(s)
- Scott Cameron
- Center for Neuroscience, University of California, Davis, California 95618, USA; ,
| | | |
Collapse
|
44
|
Dynamics, nanoscale organization, and function of synaptic adhesion molecules. Mol Cell Neurosci 2018; 91:95-107. [DOI: 10.1016/j.mcn.2018.04.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 12/13/2022] Open
|
45
|
Zerbi V, Ielacqua GD, Markicevic M, Haberl MG, Ellisman MH, A-Bhaskaran A, Frick A, Rudin M, Wenderoth N. Dysfunctional Autism Risk Genes Cause Circuit-Specific Connectivity Deficits With Distinct Developmental Trajectories. Cereb Cortex 2018; 28:2495-2506. [PMID: 29901787 PMCID: PMC5998961 DOI: 10.1093/cercor/bhy046] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/16/2018] [Accepted: 02/12/2018] [Indexed: 12/22/2022] Open
Abstract
Autism spectrum disorders (ASD) are a set of complex neurodevelopmental disorders for which there is currently no targeted therapeutic approach. It is thought that alterations of genes regulating migration and synapse formation during development affect neural circuit formation and result in aberrant connectivity within distinct circuits that underlie abnormal behaviors. However, it is unknown whether deviant developmental trajectories are circuit-specific for a given autism risk-gene. We used MRI to probe changes in functional and structural connectivity from childhood to adulthood in Fragile-X (Fmr1-/y) and contactin-associated (CNTNAP2-/-) knockout mice. Young Fmr1-/y mice (30 days postnatal) presented with a robust hypoconnectivity phenotype in corticocortico and corticostriatal circuits in areas associated with sensory information processing, which was maintained until adulthood. Conversely, only small differences in hippocampal and striatal areas were present during early postnatal development in CNTNAP2-/- mice, while major connectivity deficits in prefrontal and limbic pathways developed between adolescence and adulthood. These findings are supported by viral tracing and electron micrograph approaches and define 2 clearly distinct connectivity endophenotypes within the autism spectrum. We conclude that the genetic background of ASD strongly influences which circuits are most affected, the nature of the phenotype, and the developmental time course of the associated changes.
Collapse
Affiliation(s)
- Valerio Zerbi
- Neural Control of Movement Lab, HEST, ETH Zürich, Winterthurerstrasse 190, Zurich, Switzerland
| | - Giovanna D Ielacqua
- Institute for Biomedical Engineering, University and ETH Zurich, Wolfgang-Pauli-Str. 27, Zurich, Switzerland
| | - Marija Markicevic
- Neural Control of Movement Lab, HEST, ETH Zürich, Winterthurerstrasse 190, Zurich, Switzerland
| | - Matthias Georg Haberl
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Arjun A-Bhaskaran
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, Bordeaux, France
| | - Andreas Frick
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, Bordeaux, France
| | - Markus Rudin
- Institute for Biomedical Engineering, University and ETH Zurich, Wolfgang-Pauli-Str. 27, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Winterthurerstrasse 190, Zurich, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, Zurich, Switzerland
| | - Nicole Wenderoth
- Neural Control of Movement Lab, HEST, ETH Zürich, Winterthurerstrasse 190, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Winterthurerstrasse 190, Zurich, Switzerland
| |
Collapse
|
46
|
Nakai N, Takumi T, Nakai J, Sato M. Common Defects of Spine Dynamics and Circuit Function in Neurodevelopmental Disorders: A Systematic Review of Findings From in Vivo Optical Imaging of Mouse Models. Front Neurosci 2018; 12:412. [PMID: 29970983 PMCID: PMC6018076 DOI: 10.3389/fnins.2018.00412] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/29/2018] [Indexed: 11/18/2022] Open
Abstract
In vivo optical imaging is a powerful tool for revealing brain structure and function at both the circuit and cellular levels. Here, we provide a systematic review of findings obtained from in vivo imaging studies of mouse models of neurodevelopmental disorders, including the monogenic disorders fragile X syndrome, Rett syndrome, and Angelman syndrome, which are caused by genetic abnormalities of FMR1, MECP2, and UBE3A, as well as disorders caused by copy number variations (15q11-13 duplication and 22q11.2 deletion) and BTBR mice as an inbred strain model of autism spectrum disorder (ASD). Most studies visualize the structural and functional responsiveness of cerebral cortical neurons to sensory stimuli and the developmental and experience-dependent changes in these responses as a model of brain functions affected by these disorders. The optical imaging techniques include two-photon microscopy of fluorescently labeled dendritic spines or neurons loaded with fluorescent calcium indicators and macroscopic imaging of cortical activity using calcium indicators, voltage-sensitive dyes or intrinsic optical signals. Studies have revealed alterations in the density, stability, and turnover of dendritic spines, aberrant cortical sensory responses, impaired inhibitory function, and concomitant failure of circuit maturation as common causes for neurological deficits. Mechanistic hypotheses derived from in vivo imaging also provide new directions for therapeutic interventions. For instance, it was recently demonstrated that early postnatal administration of a selective serotonin reuptake inhibitor (SSRI) restores impaired cortical inhibitory function and ameliorates the aberrant social behaviors in a mouse model of ASD. We discuss the potential use of SSRIs for treating ASDs in light of these findings.
Collapse
Affiliation(s)
| | | | - Junichi Nakai
- RIKEN Center for Brain Science, Wako, Japan
- RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Graduate School of Science and Engineering, Saitama University, Saitama, Japan
- Brain and Body System Science Institute, Saitama University, Saitama, Japan
| | - Masaaki Sato
- RIKEN Center for Brain Science, Wako, Japan
- Graduate School of Science and Engineering, Saitama University, Saitama, Japan
- Brain and Body System Science Institute, Saitama University, Saitama, Japan
| |
Collapse
|
47
|
Jiang DY, Wu Z, Forsyth CT, Hu Y, Yee SP, Chen G. GABAergic deficits and schizophrenia-like behaviors in a mouse model carrying patient-derived neuroligin-2 R215H mutation. Mol Brain 2018; 11:31. [PMID: 29859117 PMCID: PMC5984814 DOI: 10.1186/s13041-018-0375-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/24/2018] [Indexed: 01/03/2023] Open
Abstract
Schizophrenia (SCZ) is a severe mental disorder characterized by delusion, hallucination, and cognitive deficits. We have previously identified from schizophrenia patients a loss-of-function mutation Arg215→His215 (R215H) of neuroligin 2 (NLGN2) gene, which encodes a cell adhesion molecule critical for GABAergic synapse formation and function. Here, we generated a novel transgenic mouse line with neuroligin-2 (NL2) R215H mutation. The single point mutation caused a significant loss of NL2 protein in vivo, reduced GABAergic transmission, and impaired hippocampal activation. Importantly, R215H KI mice displayed anxiety-like behavior, impaired pre-pulse inhibition (PPI), cognition deficits and abnormal stress responses, recapitulating several key aspects of schizophrenia-like behaviors. Our results demonstrate a significant impact of a single point mutation NL2 R215H on brain functions, providing a novel animal model for the study of schizophrenia and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Dong-Yun Jiang
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802 USA
| | - Zheng Wu
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802 USA
| | - Cody Tieu Forsyth
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802 USA
| | - Yi Hu
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802 USA
| | - Siu-Pok Yee
- Department of Cell Biology, University of Connecticut Health center, Farmington, CT 06030 USA
| | - Gong Chen
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802 USA
| |
Collapse
|
48
|
Um SM, Ha S, Lee H, Kim J, Kim K, Shin W, Cho YS, Roh JD, Kang J, Yoo T, Noh YW, Choi Y, Bae YC, Kim E. NGL-2 Deletion Leads to Autistic-like Behaviors Responsive to NMDAR Modulation. Cell Rep 2018; 23:3839-3851. [DOI: 10.1016/j.celrep.2018.05.087] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/13/2018] [Accepted: 05/25/2018] [Indexed: 01/01/2023] Open
|
49
|
Chen L, Huang Z, Du Y, Fu M, Han H, Wang Y, Dong Z. Capsaicin Attenuates Amyloid-β-Induced Synapse Loss and Cognitive Impairments in Mice. J Alzheimers Dis 2018; 59:683-694. [PMID: 28671132 DOI: 10.3233/jad-170337] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of progressive cognitive impairment in the aged. The aggregation of the amyloid β-protein (Aβ) is a hallmark of AD and is linked to synapse loss and cognitive impairment. Capsaicin, a specific agonist of the transient receptor potential vanilloid 1 (TRPV1), has been proven to ameliorate stress-induced AD-like pathological and cognitive impairments, but it is unclear whether TRPV1 activation can affect cognitive and synaptic functions in Aβ-induced mouse model of AD. In this study, we investigated the effects of TRPV1 activation on spatial memory and synaptic plasticity in mice treated with Aβ. To induce AD-like pathological and cognitive impairments, adult C57Bl/6 mice were microinjected with Aβ42 (100 μM, 2.5 μl/mouse, i.c.v.). Two weeks after Aβ42 microinjection, spatial learning and memory as well as hippocampal long-term potentiation (LTP) were examined. The results showed that Aβ42 microinjection significantly impaired spatial learning and memory in the Morris water maze and novel object recognition tests compared with controls. These behavioral changes were accompanied by synapse loss and impaired LTP in the CA1 area of hippocampus. More importantly, daily capsaicin (1 mg/kg, i.p.) treatment throughout the experiment dramatically improved spatial learning and memory and synaptic function, as reflected by enhanced hippocampal LTP and reduced synapse loss, whereas the TRPV1 antagonist capsazepine (1 mg/kg, i.p.) treatment had no effects on cognitive and synaptic function in Aβ42-treated mice. These results indicate that TRPV1 activation by capsaicin rescues cognitive deficit in the Aβ42-induced mouse model of AD both structurely and functionally.
Collapse
Affiliation(s)
- Long Chen
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Zhilin Huang
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yehong Du
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Min Fu
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Huili Han
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yutian Wang
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Brain Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Zhifang Dong
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| |
Collapse
|
50
|
Lin L, Murphy JG, Karlsson RM, Petralia RS, Gutzmann JJ, Abebe D, Wang YX, Cameron HA, Hoffman DA. DPP6 Loss Impacts Hippocampal Synaptic Development and Induces Behavioral Impairments in Recognition, Learning and Memory. Front Cell Neurosci 2018; 12:84. [PMID: 29651237 PMCID: PMC5884885 DOI: 10.3389/fncel.2018.00084] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/08/2018] [Indexed: 11/13/2022] Open
Abstract
DPP6 is well known as an auxiliary subunit of Kv4-containing, A-type K+ channels which regulate dendritic excitability in hippocampal CA1 pyramidal neurons. We have recently reported, however, a novel role for DPP6 in regulating dendritic filopodia formation and stability, affecting synaptic development and function. These results are notable considering recent clinical findings associating DPP6 with neurodevelopmental and intellectual disorders. Here we assessed the behavioral consequences of DPP6 loss. We found that DPP6 knockout (DPP6-KO) mice are impaired in hippocampus-dependent learning and memory. Results from the Morris water maze and T-maze tasks showed that DPP6-KO mice exhibit slower learning and reduced memory performance. DPP6 mouse brain weight is reduced throughout development compared with WT, and in vitro imaging results indicated that DPP6 loss affects synaptic structure and motility. Taken together, these results show impaired synaptic development along with spatial learning and memory deficiencies in DPP6-KO mice.
Collapse
Affiliation(s)
- Lin Lin
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, United States
| | - Jonathan G Murphy
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, United States
| | - Rose-Marie Karlsson
- Section on Neuroplasticity, National Institute of Mental Health, Bethesda, MD, United States
| | - Ronald S Petralia
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Jakob J Gutzmann
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, United States
| | - Daniel Abebe
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, United States
| | - Ya-Xian Wang
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Heather A Cameron
- Section on Neuroplasticity, National Institute of Mental Health, Bethesda, MD, United States
| | - Dax A Hoffman
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, United States
| |
Collapse
|