1
|
Sun W, Yu H, Li X, Wan Y, Mei Y, Yang Y, An L. Subchronic cyanuric acid treatment impairs spatial flexible behavior in female adolescent rats through depressing GluN2B-dependent neuronal and synaptic function. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 289:117513. [PMID: 39662458 DOI: 10.1016/j.ecoenv.2024.117513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 09/13/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
Subchronic exposure to cyanuric acid (CA) and its structural analogue melamine induces long-term effects on brain and behavior in male rodents. To examine if this exposure induced negative effects on cognitive function in females, we examined the behavioral performance and further attempted to investigate synaptic and neuronal function. CA was intraperitoneal treated with 20 or 40 mg/kg/day to adolescent female rats for 4 consecutive weeks. Multiple behavioral tests were employed to assess spatial cognition, learning strategy, locomotion and motivation. Hippocampal synaptic function at Schaffer collaterals-CA1 synapses and excitatory postsynaptic currents (EPSCs) in CA1 pyramidal neurons was evaluated. Meanwhile, the glutamate transport inhibitor DL-threo-β-benzyloxyaspartate (DL-TBOA) was infused into hippocampal CA1 region to certify the underlying mechanism. We found that subchronic CA exposure impairs reversal learning ability with dose-dependent effects but did not affect spatial learning and memory, or learning strategy. The expression and phosphorylation of N-methyl-D-aspartate receptor (NMDAR) GluN2B subunits were simultaneously reduced in the hippocampus and the GluN2B-mediated synaptic function, including long-term depression (LTD) and paired-pulse facilitation (PPF), was suppressed. CA could also diminish postsynaptic density protein-95 (PSD-95) expression but did change the levels of α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionate receptor (AMPAR) GluA1 or NMDAR GluN2A subunit, or hippocampal spine density. Meanwhile, CA depressed frequency and amplitude of GluN2B-mediated EPSCs, indicating the presynaptic and postsynaptic actions of CA on neuronal activity. Furthermore, the DL-TBOA infusions could effectively mitigate the diminished GluN2B-LTD and GluN2B-EPSCs and the impairments in behavioral flexibility. Our findings provide the first evidence that CA can exert neurotoxic effects on females and certify that one of the potential mechanisms for neuronal and synaptic dysfunction is the GluN2B-mediated signaling pathway.
Collapse
Affiliation(s)
- Wei Sun
- Department of Pediatrics, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China
| | - Haiyang Yu
- Department of Pediatrics, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China
| | - Xiaoliang Li
- Department of Chinese Medicine, Jinan Geriatric/Rehabilitation Hospital, Jinan 250013, China
| | - Yiwen Wan
- Department of Rehabilitation Medicine, Shenzhen Bao'an Hospital Affiliated of Southern Medical University, Shenzhen, Guangdong 518100, China
| | - Yazi Mei
- Graduate School of Guangzhou University of Chinese Medicine; Guangzhou 510006, China
| | - Yang Yang
- Department of Pediatrics, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China
| | - Lei An
- Department of Pediatrics, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China; Department of Chinese Medicine, Jinan Geriatric/Rehabilitation Hospital, Jinan 250013, China; Graduate School of Guangzhou University of Chinese Medicine; Guangzhou 510006, China; Department of Proctology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China.
| |
Collapse
|
2
|
Giansante G, Mazzoleni S, Zippo AG, Ponzoni L, Ghilardi A, Maiellano G, Lewerissa E, van Hugte E, Nadif Kasri N, Francolini M, Sala M, Murru L, Bassani S, Passafaro M. Neuronal network activity and connectivity are impaired in a conditional knockout mouse model with PCDH19 mosaic expression. Mol Psychiatry 2024; 29:1710-1725. [PMID: 36997609 PMCID: PMC11371655 DOI: 10.1038/s41380-023-02022-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 04/01/2023]
Abstract
Mutations in PCDH19 gene, which encodes protocadherin-19 (PCDH19), cause Developmental and Epileptic Encephalopathy 9 (DEE9). Heterogeneous loss of PCDH19 expression in neurons is considered a key determinant of the disorder; however, how PCDH19 mosaic expression affects neuronal network activity and circuits is largely unclear. Here, we show that the hippocampus of Pcdh19 mosaic mice is characterized by structural and functional synaptic defects and by the presence of PCDH19-negative hyperexcitable neurons. Furthermore, global reduction of network firing rate and increased neuronal synchronization have been observed in different limbic system areas. Finally, network activity analysis in freely behaving mice revealed a decrease in excitatory/inhibitory ratio and functional hyperconnectivity within the limbic system of Pcdh19 mosaic mice. Altogether, these results indicate that altered PCDH19 expression profoundly affects circuit wiring and functioning, and provide new key to interpret DEE9 pathogenesis.
Collapse
Affiliation(s)
| | - Sara Mazzoleni
- Institute of Neuroscience, CNR, 20854, Vedano al Lambro, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20129, Milano, Italy
| | - Antonio G Zippo
- Institute of Neuroscience, CNR, 20854, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, 20126, Milano, Italy
| | - Luisa Ponzoni
- Institute of Neuroscience, CNR, 20854, Vedano al Lambro, Italy
| | - Anna Ghilardi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20129, Milano, Italy
| | - Greta Maiellano
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20129, Milano, Italy
| | - Elly Lewerissa
- Radboud University Nijmegen Medical Centre, Donders Institute for Brain, Cognition, and Behaviour, Department of Human Genetics, Department of Human Genetics Cognitive Neuroscience, Nijmegen, Netherlands
| | - Eline van Hugte
- Radboud University Nijmegen Medical Centre, Donders Institute for Brain, Cognition, and Behaviour, Department of Human Genetics, Department of Human Genetics Cognitive Neuroscience, Nijmegen, Netherlands
| | - Nael Nadif Kasri
- Radboud University Nijmegen Medical Centre, Donders Institute for Brain, Cognition, and Behaviour, Department of Human Genetics, Department of Human Genetics Cognitive Neuroscience, Nijmegen, Netherlands
| | - Maura Francolini
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20129, Milano, Italy
| | | | - Luca Murru
- Institute of Neuroscience, CNR, 20854, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, 20126, Milano, Italy
| | - Silvia Bassani
- Institute of Neuroscience, CNR, 20854, Vedano al Lambro, Italy.
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, 20126, Milano, Italy.
| | - Maria Passafaro
- Institute of Neuroscience, CNR, 20854, Vedano al Lambro, Italy.
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, 20126, Milano, Italy.
| |
Collapse
|
3
|
Zhang H, Wu LZ, Liu ZY, Jin ZB. Patient-derived induced pluripotent stem cells with a MERTK mutation exhibit cell junction abnormalities and aberrant cellular differentiation potential. World J Stem Cells 2024; 16:512-524. [PMID: 38817331 PMCID: PMC11135251 DOI: 10.4252/wjsc.v16.i5.512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/29/2024] [Accepted: 04/01/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND Human induced pluripotent stem cell (hiPSC) technology is a valuable tool for generating patient-specific stem cells, facilitating disease modeling, and investigating disease mechanisms. However, iPSCs carrying specific mutations may limit their clinical applications due to certain inherent characteristics. AIM To investigate the impact of MERTK mutations on hiPSCs and determine whether hiPSC-derived extracellular vesicles (EVs) influence anomalous cell junction and differentiation potential. METHODS We employed a non-integrating reprogramming technique to generate peripheral blood-derived hiPSCs with and hiPSCs without a MERTK mutation. Chromosomal karyotype analysis, flow cytometry, and immunofluorescent staining were utilized for hiPSC identification. Transcriptomics and proteomics were employed to elucidate the expression patterns associated with cell junction abnormalities and cellular differentiation potential. Additionally, EVs were isolated from the supernatant, and their RNA and protein cargos were examined to investigate the involvement of hiPSC-derived EVs in stem cell junction and differentiation. RESULTS The generated hiPSCs, both with and without a MERTK mutation, exhibited normal karyotype and expressed pluripotency markers; however, hiPSCs with a MERTK mutation demonstrated anomalous adhesion capability and differentiation potential, as confirmed by transcriptomic and proteomic profiling. Furthermore, hiPSC-derived EVs were involved in various biological processes, including cell junction and differentiation. CONCLUSION HiPSCs with a MERTK mutation displayed altered junction characteristics and aberrant differentiation potential. Furthermore, hiPSC-derived EVs played a regulatory role in various biological processes, including cell junction and differentiation.
Collapse
Affiliation(s)
- Hang Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Ling-Zi Wu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Zhen-Yu Liu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China.
| |
Collapse
|
4
|
Wang M, Yu X. Experience-dependent structural plasticity of pyramidal neurons in the developing sensory cortices. Curr Opin Neurobiol 2023; 81:102724. [PMID: 37068383 DOI: 10.1016/j.conb.2023.102724] [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: 08/25/2022] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 04/19/2023]
Abstract
Sensory experience regulates the structural and functional wiring of neuronal circuits, during development and throughout adulthood. Here, we review current knowledge of how experience affects structural plasticity of pyramidal neurons in the sensory cortices. We discuss the pros and cons of existing labeling approaches, as well as what structural parameters are most plastic. We further discuss how recent advances in sparse labeling of specific neuronal subtypes, as well as development of techniques that allow fast, high resolution imaging in large fields, would enable future studies to address currently unanswered questions in the field of structural plasticity.
Collapse
Affiliation(s)
- Miao Wang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and PKU-IDG/McGovern Institute, Peking University, Beijing 100871, China.
| | - Xiang Yu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and PKU-IDG/McGovern Institute, Peking University, Beijing 100871, China; Autism Research Center of Peking University Health Science Center, Beijing 100191, China; Chinese Institute for Brain Research, Beijing 102206, China.
| |
Collapse
|
5
|
Li GY, Wu QZ, Song TJ, Zhen XC, Yu X. Dynamic regulation of excitatory and inhibitory synaptic transmission by growth hormone in the developing mouse brain. Acta Pharmacol Sin 2023; 44:1109-1121. [PMID: 36476808 PMCID: PMC10202927 DOI: 10.1038/s41401-022-01027-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/06/2022] [Indexed: 12/13/2022] Open
Abstract
Normal sensory and cognitive function of the brain relies on its intricate and complex neural network. Synaptogenesis and synaptic plasticity are critical to neural circuit formation and maintenance, which are regulated by coordinated intracellular and extracellular signaling. Growth hormone (GH) is the most abundant anterior pituitary hormone. Its deficiencies could alter brain development and impair learning and memory, while GH replacement therapy in human patients and animal models has been shown to ameliorate cognitive deficits caused by GH deficiency. However, the underlying mechanism remains largely unknown. In this study, we investigated the neuromodulatory function of GH in young (pre-weaning) mice at two developmental time points and in two different brain regions. Neonatal mice were subcutaneously injected with recombinant human growth hormone (rhGH) on postnatal day (P) 14 or 21. Excitatory and inhibitory synaptic transmission was measured using whole-cell recordings in acute cortical slices 2 h after the injection. We showed that injection of rhGH (2 mg/kg) in P14 mice significantly increased the frequency of mEPSCs, but not that of mIPSCs, in both hippocampal CA1 pyramidal neurons and L2/3 pyramidal neurons of the barrel field of the primary somatosensory cortex (S1BF). Injection of rhGH (2 mg/kg) in P21 mice significantly increased the frequency of mEPSCs and mIPSCs in both brain regions. Perfusion of rhGH (1 μM) onto acute brain slices in P14 mice had similar effects. Consistent with the electrophysiological results, the dendritic spine density of CA1 pyramidal neurons and S1BF L2/3 pyramidal neurons increased following in vivo injection of rhGH. Furthermore, NMDA receptors and postsynaptic calcium-dependent signaling contributed to rhGH-dependent regulation of both excitatory and inhibitory synaptic transmission. Together, these results demonstrate that regulation of excitatory and inhibitory synaptic transmission by rhGH occurs in a developmentally dynamic manner, and have important implication for identifying GH treatment strategies without disturbing excitation/inhibition balance.
Collapse
Affiliation(s)
- Guang-Ying Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
- School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and Peking University McGovern Institute, Peking University, Beijing, 100871, China.
| | - Qiu-Zi Wu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and Peking University McGovern Institute, Peking University, Beijing, 100871, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tian-Jia Song
- School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and Peking University McGovern Institute, Peking University, Beijing, 100871, China
| | - Xue-Chu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Xiang Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
- School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and Peking University McGovern Institute, Peking University, Beijing, 100871, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
6
|
Salemme V, Vedelago M, Sarcinella A, Moietta F, Piccolantonio A, Moiso E, Centonze G, Manco M, Guala A, Lamolinara A, Angelini C, Morellato A, Natalini D, Calogero R, Incarnato D, Oliviero S, Conti L, Iezzi M, Tosoni D, Bertalot G, Freddi S, Tucci FA, De Sanctis F, Frusteri C, Ugel S, Bronte V, Cavallo F, Provero P, Gai M, Taverna D, Turco E, Pece S, Defilippi P. p140Cap inhibits β-Catenin in the breast cancer stem cell compartment instructing a protective anti-tumor immune response. Nat Commun 2023; 14:2350. [PMID: 37169737 PMCID: PMC10175288 DOI: 10.1038/s41467-023-37824-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/03/2023] [Indexed: 05/13/2023] Open
Abstract
The p140Cap adaptor protein is a tumor suppressor in breast cancer associated with a favorable prognosis. Here we highlight a function of p140Cap in orchestrating local and systemic tumor-extrinsic events that eventually result in inhibition of the polymorphonuclear myeloid-derived suppressor cell function in creating an immunosuppressive tumor-promoting environment in the primary tumor, and premetastatic niches at distant sites. Integrative transcriptomic and preclinical studies unravel that p140Cap controls an epistatic axis where, through the upstream inhibition of β-Catenin, it restricts tumorigenicity and self-renewal of tumor-initiating cells limiting the release of the inflammatory cytokine G-CSF, required for polymorphonuclear myeloid-derived suppressor cells to exert their local and systemic tumor conducive function. Mechanistically, p140Cap inhibition of β-Catenin depends on its ability to localize in and stabilize the β-Catenin destruction complex, promoting enhanced β-Catenin inactivation. Clinical studies in women show that low p140Cap expression correlates with reduced presence of tumor-infiltrating lymphocytes and more aggressive tumor types in a large cohort of real-life female breast cancer patients, highlighting the potential of p140Cap as a biomarker for therapeutic intervention targeting the β-Catenin/ Tumor-initiating cells /G-CSF/ polymorphonuclear myeloid-derived suppressor cell axis to restore an efficient anti-tumor immune response.
Collapse
Affiliation(s)
- Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
| | - Mauro Vedelago
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alessandro Sarcinella
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Federico Moietta
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alessio Piccolantonio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
| | - Enrico Moiso
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
| | - Marta Manco
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Andrea Guala
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alessia Lamolinara
- Immuno-Oncology Laboratory, Center for Advanced Studies and Technology (CAST), Department of Neuroscience, Imaging and Clinical Sciences, G. d'Annunzio University of Chieti-Pescara, Chieti-Pescara, Italy
| | - Costanza Angelini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Raffaele Calogero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
| | - Danny Incarnato
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Groningen, the Netherlands
| | - Salvatore Oliviero
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
- Department of Life Sciences and Systems Biology, University of Turin, Torino, Italy and IIGM, Candiolo, Italy
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
| | - Manuela Iezzi
- Immuno-Oncology Laboratory, Center for Advanced Studies and Technology (CAST), Department of Neuroscience, Imaging and Clinical Sciences, G. d'Annunzio University of Chieti-Pescara, Chieti-Pescara, Italy
| | - Daniela Tosoni
- European Institute of Oncology IRCCS, 20141, Milan, Italy
| | | | - Stefano Freddi
- European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Francesco A Tucci
- European Institute of Oncology IRCCS, 20141, Milan, Italy
- School of Pathology, University of Milan, Milan, Italy
| | - Francesco De Sanctis
- Immunology Section, Department of Medicine, University of Verona, 37134, Verona, Italy
| | - Cristina Frusteri
- Immunology Section, Department of Medicine, University of Verona, 37134, Verona, Italy
| | - Stefano Ugel
- Immunology Section, Department of Medicine, University of Verona, 37134, Verona, Italy
| | - Vincenzo Bronte
- Immunology Section, Department of Medicine, University of Verona, 37134, Verona, Italy
- Istituto Oncologico Veneto, IRCCS, 35128, Padova, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
| | - Paolo Provero
- Neuroscience Department "Rita Levi Montalcini", University of Torino, Via Cherasco 15, 10126, Torino, Italy
| | - Marta Gai
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Daniela Taverna
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Salvatore Pece
- European Institute of Oncology IRCCS, 20141, Milan, Italy.
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20142, Milano, Italy.
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy.
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy.
| |
Collapse
|
7
|
Uchigashima M, Hayashi Y, Futai K. Regulation of Presynaptic Release Machinery by Cell Adhesion Molecules. ADVANCES IN NEUROBIOLOGY 2023; 33:333-356. [PMID: 37615873 DOI: 10.1007/978-3-031-34229-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
The synapse is a highly specialized asymmetric structure that transmits and stores information in the brain. The size of pre- and postsynaptic structures and function is well coordinated at the individual synapse level. For example, large postsynaptic dendritic spines have a larger postsynaptic density with higher α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) number on their surface, while juxtaposing presynaptic terminals have a larger active zone and higher release probability. This indicates that pre- and postsynaptic domains bidirectionally communicate to coordinate assembly of specific molecules on both sides of the synaptic cleft. Cell adhesion molecules (CAMs) that localize at synapses form transsynaptic protein interactions across the synaptic cleft and play important roles in synapse formation and regulation. The extracellular domain of CAMs is essential for specific synapse formation and function. In contrast, the intracellular domain is necessary for binding with synaptic molecules and signal transduction. Therefore, CAMs play an essential role on synapse function and structure. In fact, ample evidence indicates that transsynaptic CAMs instruct and modulate functions at presynaptic sites. This chapter focuses on transsynaptic protein interactions that regulate presynaptic functions emphasizing the role of neuronal CAMs and the intracellular mechanism of their regulation.
Collapse
Affiliation(s)
- Motokazu Uchigashima
- Department of Cellular Neuropathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yasunori Hayashi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kensuke Futai
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
8
|
Liu P, Dai S, Mi T, Tang G, Wang Z, Wang H, Du H, Tang Y, Teng Z, Liu C. Acetate supplementation restores cognitive deficits caused by ARID1A haploinsufficiency in excitatory neurons. EMBO Mol Med 2022; 14:e15795. [PMID: 36385502 PMCID: PMC9728054 DOI: 10.15252/emmm.202215795] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/18/2022] Open
Abstract
Mutations in AT-rich interactive domain-containing protein 1A (ARID1A) cause Coffin-Siris syndrome (CSS), a rare genetic disorder that results in mild to severe intellectual disabilities. However, the biological role of ARID1A in the brain remains unclear. In this study, we report that the haploinsufficiency of ARID1A in excitatory neurons causes cognitive impairment and defects in hippocampal synaptic transmission and dendritic morphology in mice. Similarly, human embryonic stem cell-derived excitatory neurons with deleted ARID1A exhibit fewer dendritic branches and spines, and abnormal electrophysiological activity. Importantly, supplementation of acetate, an epigenetic metabolite, can ameliorate the morphological and electrophysiological deficits observed in mice with Arid1a haploinsufficiency, as well as in ARID1A-null human excitatory neurons. Mechanistically, transcriptomic and ChIP-seq analyses demonstrate that acetate supplementation can increase the levels of H3K27 acetylation at the promoters of key regulatory genes associated with neural development and synaptic transmission. Collectively, these findings support the essential roles of ARID1A in the excitatory neurons and cognition and suggest that acetate supplementation could be a potential therapeutic intervention for CSS.
Collapse
Affiliation(s)
- Pei‐Pei Liu
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina,Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina,Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Shang‐Kun Dai
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina,School of Life Sciences and MedicineShandong University of TechnologyZiboChina
| | - Ting‐Wei Mi
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
| | - Gang‐Bin Tang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
| | - Zhuo Wang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina
| | - Hui Wang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina
| | - Hong‐Zhen Du
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina,Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina,Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Yi Tang
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Zhao‐Qian Teng
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina,Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina,Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Chang‐Mei Liu
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina,Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina,Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| |
Collapse
|
9
|
Learning and memory impairment and transcriptomic profile in hippocampus of offspring after maternal fructose exposure during gestation and lactation. Food Chem Toxicol 2022; 169:113394. [PMID: 36049592 DOI: 10.1016/j.fct.2022.113394] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/06/2022] [Accepted: 08/24/2022] [Indexed: 11/22/2022]
Abstract
Increased fructose intake is a global issue, especially in mothers. Maternal fructose exposure during gestation and lactation can affect learning and memory in offspring; however, the detailed mechanism is still unknown. The hippocampus is a mind locale liable for learning and memory. Here, we established a maternal high-fructose diet model by administering 13% and 40% fructose water, applied the Morris Water Maze test on postnatal day 60 offspring, and performed full-length RNA sequencing using the Oxford Nanopore Technologies platform to explore the changes in gene expression in the hippocampus. The results showed that learning and memory in offspring were negatively affected. Compared with the control group, 369 differentially expressed transcripts (DETs) were identified in the 13% fructose group, and 501 DETs were identified in the 40% fructose group. Gene Ontology enriched term and Kyoto Encyclopedia of Genes and Genomes enriched pathway analyses identified several terms and pathways related to brain development and cognitive function. Furthermore, we confirmed that the Wnt/β-catenin signaling pathway was down-regulated and neuron degeneration was enhanced. In summary, our results indicate that maternal fructose exposure during gestation and lactation can impair learning and memory in offspring and affect brain function at the transcriptome level.
Collapse
|
10
|
Xu Y, Wang ML, Tao H, Geng C, Guo F, Hu B, Wang R, Hou XY. ErbB4 in parvalbumin-positive interneurons mediates proactive interference in olfactory associative reversal learning. Neuropsychopharmacology 2022; 47:1292-1303. [PMID: 34707248 PMCID: PMC9117204 DOI: 10.1038/s41386-021-01205-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 09/04/2021] [Accepted: 10/02/2021] [Indexed: 11/09/2022]
Abstract
Consolidated memories influence later learning and cognitive processes when new information is overlapped with previous events. To reveal which cellular and molecular factors are associated with this proactive interference, we challenged mice with odor-reward associative learning followed by a reversal-learning task. The results showed that genetical ablation of ErbB4 in parvalbumin (PV)-positive interneurons improved performance in reversal-learning phase, with no alteration in learning phase, supporting that PV interneuron ErbB4 is required for proactive interference. Mechanistically, olfactory learning promoted PV interneuron excitatory synaptic plasticity and direct binding of ErbB4 with presynaptic Neurexin1β (NRXN1β) and postsynaptic scaffold PSD-95 in the prefrontal cortex. Interrupting ErbB4-NRXN1β interaction impaired network activity-driven excitatory inputs and excitatory synaptic transmission onto PV interneurons. Neuronal activity-induced ErbB4-PSD-95 association facilitated transsynaptic binding of ErbB4-NRXN1β and excitatory synapse formation in ErbB4-positive interneurons. Furthermore, ErbB4-NRXN1β binding was responsible for the activity-regulated activation of ErbB4 and extracellular signal-regulated kinase (ERK) 1/2 in PV interneurons, as well as synaptic plasticity-related expression of brain-derived neurotrophic factor (BDNF). Correlatedly, blocking ErbB4-NRXN1β coupling in the medial prefrontal cortex of adult mice facilitated reversal learning of an olfactory associative task. These findings provide novel insight into the physiological role of PV interneuron ErbB4 signaling in cognitive processes and reveal an associative learning-related transsynaptic NRXN1β-ErbB4-PSD-95 complex that affects the ERK1/2-BDNF pathway and underlies local inhibitory circuit plasticity and proactive interference.
Collapse
Affiliation(s)
- Yan Xu
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Meng-Lin Wang
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Hui Tao
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China ,grid.254147.10000 0000 9776 7793State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198 China
| | - Chi Geng
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Feng Guo
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Bin Hu
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Ran Wang
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Xiao-Yu Hou
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China. .,State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
11
|
Sun W, Chen X, Mei Y, Yang Y, Li X, An L. Prelimbic proBDNF Facilitates Retrieval-Dependent Fear Memory Destabilization by Regulation of Synaptic and Neural Functions in Juvenile Rats. Mol Neurobiol 2022; 59:4179-4196. [PMID: 35501631 DOI: 10.1007/s12035-022-02849-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 04/23/2022] [Indexed: 10/18/2022]
Abstract
Fear regulation changes as a function of the early life is a key developmental period for the continued maturation of fear neural circuitry. The mechanisms of fear retrieval-induced reconsolidation have been investigated but remain poorly understood. The involvement of prelimbic proBDNF in fear memory extinction and its mediated signaling have been reported previously. Specifically, blocking the proBDNF/p75NTR pathway during the postnatal stage disrupts synaptic development and neuronal activity in adulthood. Given the inherent high expression of proBDNF during the juvenile period, we tested whether the prelimbic proBDNF regulated synaptic and neuronal functions allowing to influencing retrieval-dependent memory processing. By examining the freezing behavior of auditory fear-conditioned rats, we found the high level of the prelimbic proBDNF in juvenile rats enhanced the destabilization of the retrieval-dependent weak but not strong fear memory through activating p75NTR-GluN2B signaling. This modification of fear memory traces was attributed to the increment in the proportion of thin-type spine and promotion in synaptic function, as evidenced by the facilitation of NMDA-mediated EPSCs and GluN2B-dependent synaptic depression at the prelimbic projection. Furthermore, the strong prelimbic theta- and gamma-oscillation coupling predicted the suppressive effect of juvenile proBDNF on the recall of postretrieval memory. Our results critically emphasize the importance of developmental proBDNF for modification of retrieval-dependent memory and provide a potential critical targeting to inhibit threaten memories associated with neurodevelopment disorders.
Collapse
Affiliation(s)
- Wei Sun
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China.,Behavioral Neuroscience Laboratory, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| | - Xiao Chen
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China.,Department of Neurology, Jinan Geriatric/Rehabilitation Hospital, Jinan, 250013, China
| | - Yazi Mei
- Graduate School of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yang Yang
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| | - Xiaoliang Li
- Department of Neurology, Jinan Geriatric/Rehabilitation Hospital, Jinan, 250013, China
| | - Lei An
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China. .,Behavioral Neuroscience Laboratory, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China. .,Department of Neurology, Jinan Geriatric/Rehabilitation Hospital, Jinan, 250013, China. .,Graduate School of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
12
|
Camera M, Russo I, Zamboni V, Ammoni A, Rando S, Morellato A, Cimino I, Angelini C, Giacobini P, Oleari R, Amoruso F, Cariboni A, Franceschini I, Turco E, Defilippi P, Merlo GR. p140Cap Controls Female Fertility in Mice Acting via Glutamatergic Afference on Hypothalamic Gonadotropin-Releasing Hormone Neurons. Front Neurosci 2022; 16:744693. [PMID: 35237119 PMCID: PMC8884249 DOI: 10.3389/fnins.2022.744693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
p140Cap, encoded by the gene SRCIN1 (SRC kinase signaling inhibitor 1), is an adaptor/scaffold protein highly expressed in the mouse brain, participating in several pre- and post-synaptic mechanisms. p140Cap knock-out (KO) female mice show severe hypofertility, delayed puberty onset, altered estrus cycle, reduced ovulation, and defective production of luteinizing hormone and estradiol during proestrus. We investigated the role of p140Cap in the development and maturation of the hypothalamic gonadotropic system. During embryonic development, migration of Gonadotropin-Releasing Hormone (GnRH) neurons from the nasal placode to the forebrain in p140Cap KO mice appeared normal, and young p140Cap KO animals showed a normal number of GnRH-immunoreactive (-ir) neurons. In contrast, adult p140Cap KO mice showed a significant loss of GnRH-ir neurons and a decreased density of GnRH-ir projections in the median eminence, accompanied by reduced levels of GnRH and LH mRNAs in the hypothalamus and pituitary gland, respectively. We examined the number of kisspeptin (KP) neurons in the rostral periventricular region of the third ventricle, the number of KP-ir fibers in the arcuate nucleus, and the number of KP-ir punctae on GnRH neurons but we found no significant changes. Consistently, the responsiveness to exogenous KP in vivo was unchanged, excluding a cell-autonomous defect on the GnRH neurons at the level of KP receptor or its signal transduction. Since glutamatergic signaling in the hypothalamus is critical for both puberty onset and modulation of GnRH secretion, we examined the density of glutamatergic synapses in p140Cap KO mice and observed a significant reduction in the density of VGLUT-ir punctae both in the preoptic area and on GnRH neurons. Our data suggest that the glutamatergic circuitry in the hypothalamus is altered in the absence of p140Cap and is required for female fertility.
Collapse
Affiliation(s)
- Mattia Camera
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Isabella Russo
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Valentina Zamboni
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Alessandra Ammoni
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Simona Rando
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Irene Cimino
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, Inserm U1172, Lille, France
- Metabolic Research Laboratories, Wellcome Trust–Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Costanza Angelini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Paolo Giacobini
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, Inserm U1172, Lille, France
| | - Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Federica Amoruso
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Anna Cariboni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Isabelle Franceschini
- Physiologie de la Reproduction et des Comportements, French National Centre for Scientific Research, French Institute of the Horse and Riding, French National Research Institute for Agriculture, Food and Environment, Université de Tours, Nouzilly, France
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- *Correspondence: Paola Defilippi,
| | - Giorgio R. Merlo
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Giorgio R. Merlo,
| |
Collapse
|
13
|
Li Y, Ma C, Li S, Wang J, Li W, Yang Y, Li X, Liu J, Yang J, Liu Y, Li K, Li J, Huang D, Chen R, Lv L, Xiao X, Li M, Luo X. Regulatory Variant rs2535629 in ITIH3 Intron Confers Schizophrenia Risk By Regulating CTCF Binding and SFMBT1 Expression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104786. [PMID: 34978167 PMCID: PMC8867204 DOI: 10.1002/advs.202104786] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/16/2021] [Indexed: 06/14/2023]
Abstract
Genome-wide association studies have identified 3p21.1 as a robust risk locus for schizophrenia. However, the underlying molecular mechanisms remain elusive. Here a functional regulatory variant (rs2535629) is identified that disrupts CTCF binding at 3p21.1. It is confirmed that rs2535629 is also significantly associated with schizophrenia in Chinese population and the regulatory effect of rs2535629 is validated. Expression quantitative trait loci analysis indicates that rs2535629 is associated with the expression of three distal genes (GLT8D1, SFMBT1, and NEK4) in the human brain, and CRISPR-Cas9-mediated genome editing confirmed the regulatory effect of rs2535629 on GLT8D1, SFMBT1, and NEK4. Interestingly, differential expression analysis of GLT8D1, SFMBT1, and NEK4 suggested that rs2535629 may confer schizophrenia risk by regulating SFMBT1 expression. It is further demonstrated that Sfmbt1 regulates neurodevelopment and dendritic spine density, two key pathological characteristics of schizophrenia. Transcriptome analysis also support the potential role of Sfmbt1 in schizophrenia pathogenesis. The study identifies rs2535629 as a plausibly causal regulatory variant at the 3p21.1 risk locus and demonstrates the regulatory mechanism and biological effect of this functional variant, indicating that this functional variant confers schizophrenia risk by altering CTCF binding and regulating expression of SFMBT1, a distal gene which plays important roles in neurodevelopment and synaptic morphogenesis.
Collapse
Affiliation(s)
- Yifan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Changguo Ma
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Yunnan Key Laboratory for Basic Research on Bone and Joint Diseases & Yunnan Stem Cell Translational Research CenterKunming UniversityKunmingYunnan650214China
| | - Shiwu Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Junyang Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Wenqiang Li
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangHenan453002China
- Henan Key Lab of Biological PsychiatryInternational Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangHenan453002China
| | - Yongfeng Yang
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangHenan453002China
- Henan Key Lab of Biological PsychiatryInternational Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangHenan453002China
| | - Xiaoyan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of EducationInstitutes of Physical Science and Information TechnologyAnhui UniversityHefeiAnhui230601China
| | - Jiewei Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
| | - Jinfeng Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Yixing Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Kaiqin Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Jiao Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Di Huang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
| | - Rui Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Luxian Lv
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangHenan453002China
- Henan Key Lab of Biological PsychiatryInternational Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangHenan453002China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
| | - Xiong‐Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
- Center for Excellence in Animal Evolution and GeneticsChinese Academy of SciencesKunmingYunnan650204China
- KIZ‐CUHK Joint Laboratory of Bioresources and Molecular Research in Common DiseasesKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
| |
Collapse
|
14
|
Licheri V, Brigman JL. Altering Cell-Cell Interaction in Prenatal Alcohol Exposure Models: Insight on Cell-Adhesion Molecules During Brain Development. Front Mol Neurosci 2022; 14:753537. [PMID: 34975396 PMCID: PMC8715949 DOI: 10.3389/fnmol.2021.753537] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022] Open
Abstract
Alcohol exposure during pregnancy disrupts the development of the brain and produces long lasting behavioral and cognitive impairments collectively known as Fetal Alcohol Spectrum Disorders (FASDs). FASDs are characterized by alterations in learning, working memory, social behavior and executive function. A large body of literature using preclinical prenatal alcohol exposure models reports alcohol-induced changes in architecture and activity in specific brain regions affecting cognition. While multiple putative mechanisms of alcohol’s long-lasting effects on morphology and behavior have been investigated, an area that has received less attention is the effect of alcohol on cell adhesion molecules (CAMs). The embryo/fetal development represents a crucial period for Central Nervous System (CNS) development during which the cell-cell interaction plays an important role. CAMs play a critical role in neuronal migration and differentiation, synaptic organization and function which may be disrupted by alcohol. In this review, we summarize the physiological structure and role of CAMs involved in brain development, review the current literature on prenatal alcohol exposure effects on CAM function in different experimental models and pinpoint areas needed for future study to better understand how CAMs may mediate the morphological, sensory and behavioral outcomes in FASDs.
Collapse
Affiliation(s)
- Valentina Licheri
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Jonathan L Brigman
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States.,New Mexico Alcohol Research Center, UNM Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
15
|
Dagar S, Teng Z, Gottmann K. Transsynaptic N-Cadherin Adhesion Complexes Control Presynaptic Vesicle and Bulk Endocytosis at Physiological Temperature. Front Cell Neurosci 2021; 15:713693. [PMID: 34759800 PMCID: PMC8573734 DOI: 10.3389/fncel.2021.713693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 09/13/2021] [Indexed: 11/30/2022] Open
Abstract
At mammalian glutamatergic synapses, most basic elements of synaptic transmission have been shown to be modulated by specific transsynaptic adhesion complexes. However, although crucial for synapse homeostasis, a physiological regulation of synaptic vesicle endocytosis by adhesion molecules has not been firmly established. The homophilic adhesion protein N-cadherin is localized at the peri-active zone, where the highly temperature-dependent endocytosis of vesicles occurs. Here, we demonstrate an important modulatory role of N-cadherin in endocytosis at near physiological temperature by synaptophysin-pHluorin imaging. Different modes of endocytosis including bulk endocytosis were dependent on N-cadherin expression and function. N-cadherin modulation might be mediated by actin filaments because actin polymerization ameliorated the knockout-induced endocytosis defect. Using super-resolution imaging, we found strong recruitment of N-cadherin to glutamatergic synapses upon massive vesicle release, which might in turn enhance vesicle endocytosis. This provides a novel, adhesion protein-mediated mechanism for efficient coupling of exo- and endocytosis.
Collapse
Affiliation(s)
- Sushma Dagar
- Institute of Neuro- and Sensory Physiology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Zenghui Teng
- Institute of Neuro- and Sensory Physiology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Kurt Gottmann
- Institute of Neuro- and Sensory Physiology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
16
|
Abstract
Interleukin-1 (IL-1) is an inflammatory cytokine that has been shown to modulate neuronal signaling in homeostasis and diseases. In homeostasis, IL-1 regulates sleep and memory formation, whereas in diseases, IL-1 impairs memory and alters affect. Interestingly, IL-1 can cause long-lasting changes in behavior, suggesting IL-1 can alter neuroplasticity. The neuroplastic effects of IL-1 are mediated via its cognate receptor, Interleukin-1 Type 1 Receptor (IL-1R1), and are dependent on the distribution and cell type(s) of IL-1R1 expression. Recent reports found that IL-1R1 expression is restricted to discrete subpopulations of neurons, astrocytes, and endothelial cells and suggest IL-1 can influence neural circuits directly through neuronal IL-1R1 or indirectly via non-neuronal IL-1R1. In this review, we analyzed multiple mechanisms by which IL-1/IL-1R1 signaling might impact neuroplasticity based upon the most up-to-date literature and provided potential explanations to clarify discrepant and confusing findings reported in the past.
Collapse
Affiliation(s)
- Daniel P. Nemeth
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| |
Collapse
|
17
|
Sun W, Cheng H, Yang Y, Tang D, Li X, An L. Requirements of Postnatal proBDNF in the Hippocampus for Spatial Memory Consolidation and Neural Function. Front Cell Dev Biol 2021; 9:678182. [PMID: 34336832 PMCID: PMC8319730 DOI: 10.3389/fcell.2021.678182] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/03/2021] [Indexed: 11/17/2022] Open
Abstract
Mature brain-derived neurotrophic factor (BDNF) and its downstream signaling pathways have been implicated in regulating postnatal development and functioning of rodent brain. However, the biological role of its precursor pro-brain-derived neurotrophic factor (proBDNF) in the postnatal brain remains unknown. The expression of hippocampal proBDNF was blocked in postnatal weeks, and multiple behavioral tests, Western blot and morphological techniques, and neural recordings were employed to investigate how proBDNF played a role in spatial cognition in adults. The peak expression and its crucial effects were found in the fourth but not in the second or eighth postnatal week. Blocking proBDNF expression disrupted spatial memory consolidation rather than learning or memory retrieval. Structurally, blocking proBDNF led to the reduction in spine density and proportion of mature spines. Although blocking proBDNF did not affect N-methyl-D-aspartate (NMDA) receptor (NMDAR) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunits, the learning-induced phosphorylation of the GluN2B subunit level declined significantly. Functionally, paired-pulse facilitation, post-low-frequency stimulation (LFS) transiently enhanced depression, and GluN2B-dependent short-lasting long-term depression in the Schaffer collateral-CA1 pathway were weakened. The firing rate of pyramidal neurons was significantly suppressed around the target region during the memory test. Furthermore, the activation of GluN2B-mediated signaling could effectively facilitate neural function and mitigate memory impairment. The findings were consistent with the hypothesis that postnatal proBDNF played an essential role in synaptic and cognitive functions.
Collapse
Affiliation(s)
- Wei Sun
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Hong Cheng
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Neurology, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yang Yang
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Dongxin Tang
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiaolian Li
- Department of Neurology, Jinan Geriatric Hospital, Jinan, China
| | - Lei An
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Neurology, Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Physiology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
18
|
Retinoid X Receptor α Regulates DHA-Dependent Spinogenesis and Functional Synapse Formation In Vivo. Cell Rep 2021; 31:107649. [PMID: 32433958 DOI: 10.1016/j.celrep.2020.107649] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 02/01/2020] [Accepted: 04/22/2020] [Indexed: 12/23/2022] Open
Abstract
Coordinated intracellular and extracellular signaling is critical to synapse development and functional neural circuit wiring. Here, we report that unesterified docosahexaenoic acid (DHA) regulates functional synapse formation in vivo via retinoid X receptor α (Rxra) signaling. Using Rxra conditional knockout (cKO) mice and virus-mediated transient gene expression, we show that endogenous Rxra plays important roles in regulating spinogenesis and excitatory synaptic transmission in cortical pyramidal neurons. We further show that the effects of RXRA are mediated through its DNA-binding domain in a cell-autonomous and reversible manner. Moreover, unesterified DHA increases spine formation and excitatory synaptic transmission in vivo in an Rxra-dependent fashion. Rxra cKO mice generally behave normally but show deficits in behavior tasks associated with social memory. Together, these results demonstrate that unesterified DHA signals through RXRA to regulate spinogenesis and functional synapse formation, providing insight into the mechanism through which DHA promotes brain development and cognitive function.
Collapse
|
19
|
Hoshina N, Johnson-Venkatesh EM, Hoshina M, Umemori H. Female-specific synaptic dysfunction and cognitive impairment in a mouse model of PCDH19 disorder. Science 2021; 372:372/6539/eaaz3893. [PMID: 33859005 PMCID: PMC9873198 DOI: 10.1126/science.aaz3893] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 09/25/2020] [Accepted: 03/01/2021] [Indexed: 01/26/2023]
Abstract
Protocadherin-19 (PCDH19) mutations cause early-onset seizures and cognitive impairment. The PCDH19 gene is on the X-chromosome. Unlike most X-linked disorders, PCDH19 mutations affect heterozygous females (PCDH19HET♀ ) but not hemizygous males (PCDH19HEMI♂ ); however, the reason why remains to be elucidated. We demonstrate that PCDH19, a cell-adhesion molecule, is enriched at hippocampal mossy fiber synapses. Pcdh19HET♀ but not Pcdh19HEMI♂ mice show impaired mossy fiber synaptic structure and physiology. Consistently, Pcdh19HET♀ but not Pcdh19HEMI♂ mice exhibit reduced pattern completion and separation abilities, which require mossy fiber synaptic function. Furthermore, PCDH19 appears to interact with N-cadherin at mossy fiber synapses. In Pcdh19HET♀ conditions, mismatch between PCDH19 and N-cadherin diminishes N-cadherin-dependent signaling and impairs mossy fiber synapse development; N-cadherin overexpression rescues Pcdh19HET♀ phenotypes. These results reveal previously unknown molecular and cellular mechanisms underlying the female-specific PCDH19 disorder phenotype.
Collapse
Affiliation(s)
| | | | | | - Hisashi Umemori
- Corresponding author. Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Center for Life Sciences 13074, Boston, MA 02115,
| |
Collapse
|
20
|
Chen P, Jing H, Xiong M, Zhang Q, Lin D, Ren D, Wang S, Yin D, Chen Y, Zhou T, Li B, Fei E, Pan BX. Spine impairment in mice high-expressing neuregulin 1 due to LIMK1 activation. Cell Death Dis 2021; 12:403. [PMID: 33854034 PMCID: PMC8047019 DOI: 10.1038/s41419-021-03687-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 02/08/2023]
Abstract
The genes encoding for neuregulin1 (NRG1), a growth factor, and its receptor ErbB4 are both risk factors of major depression disorder and schizophrenia (SZ). They have been implicated in neural development and synaptic plasticity. However, exactly how NRG1 variations lead to SZ remains unclear. Indeed, NRG1 levels are increased in postmortem brain tissues of patients with brain disorders. Here, we studied the effects of high-level NRG1 on dendritic spine development and function. We showed that spine density in the prefrontal cortex and hippocampus was reduced in mice (ctoNrg1) that overexpressed NRG1 in neurons. The frequency of miniature excitatory postsynaptic currents (mEPSCs) was reduced in both brain regions of ctoNrg1 mice. High expression of NRG1 activated LIMK1 and increased cofilin phosphorylation in postsynaptic densities. Spine reduction was attenuated by inhibiting LIMK1 or blocking the NRG1–LIMK1 interaction, or by restoring NRG1 protein level. These results indicate that a normal NRG1 protein level is necessary for spine homeostasis and suggest a pathophysiological mechanism of abnormal spines in relevant brain disorders.
Collapse
Affiliation(s)
- Peng Chen
- School of Life Sciences, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Hongyang Jing
- School of Life Sciences, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Mingtao Xiong
- Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Qian Zhang
- School of Basic Medical Sciences, Nanchang University, Nanchang, 330031, China
| | - Dong Lin
- School of Life Sciences, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Dongyan Ren
- School of Life Sciences, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Shunqi Wang
- School of Life Sciences, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Dongmin Yin
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, 200062, China
| | - Yongjun Chen
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Tian Zhou
- School of Basic Medical Sciences, Nanchang University, Nanchang, 330031, China
| | - Baoming Li
- Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Erkang Fei
- School of Life Sciences, Nanchang University, Nanchang, 330031, China. .,Institute of Life Science, Nanchang University, Nanchang, 330031, China.
| | - Bing-Xing Pan
- School of Life Sciences, Nanchang University, Nanchang, 330031, China. .,Institute of Life Science, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
21
|
de Agustín-Durán D, Mateos-White I, Fabra-Beser J, Gil-Sanz C. Stick around: Cell-Cell Adhesion Molecules during Neocortical Development. Cells 2021; 10:118. [PMID: 33435191 PMCID: PMC7826847 DOI: 10.3390/cells10010118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/29/2020] [Accepted: 01/07/2021] [Indexed: 12/21/2022] Open
Abstract
The neocortex is an exquisitely organized structure achieved through complex cellular processes from the generation of neural cells to their integration into cortical circuits after complex migration processes. During this long journey, neural cells need to establish and release adhesive interactions through cell surface receptors known as cell adhesion molecules (CAMs). Several types of CAMs have been described regulating different aspects of neurodevelopment. Whereas some of them mediate interactions with the extracellular matrix, others allow contact with additional cells. In this review, we will focus on the role of two important families of cell-cell adhesion molecules (C-CAMs), classical cadherins and nectins, as well as in their effectors, in the control of fundamental processes related with corticogenesis, with special attention in the cooperative actions among the two families of C-CAMs.
Collapse
Affiliation(s)
| | | | | | - Cristina Gil-Sanz
- Neural Development Laboratory, Instituto Universitario de Biomedicina y Biotecnología (BIOTECMED) and Departamento de Biología Celular, Facultat de Biología, Universidad de Valencia, 46100 Burjassot, Spain; (D.d.A.-D.); (I.M.-W.); (J.F.-B.)
| |
Collapse
|
22
|
Coninx E, Chew YC, Yang X, Guo W, Coolkens A, Baatout S, Moons L, Verslegers M, Quintens R. Hippocampal and cortical tissue-specific epigenetic clocks indicate an increased epigenetic age in a mouse model for Alzheimer's disease. Aging (Albany NY) 2020; 12:20817-20834. [PMID: 33082299 PMCID: PMC7655172 DOI: 10.18632/aging.104056] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/27/2020] [Indexed: 04/17/2023]
Abstract
Epigenetic clocks are based on age-associated changes in DNA methylation of CpG-sites, which can accurately measure chronological age in different species. Recently, several studies have indicated that the difference between chronological and epigenetic age, defined as the age acceleration, could reflect biological age indicating functional decline and age-associated diseases. In humans, an epigenetic clock associated Alzheimer's disease (AD) pathology with an acceleration of the epigenetic age. In this study, we developed and validated two mouse brain region-specific epigenetic clocks from the C57BL/6J hippocampus and cerebral cortex. Both clocks, which could successfully estimate chronological age, were further validated in a widely used mouse model for AD, the triple transgenic AD (3xTg-AD) mouse. We observed an epigenetic age acceleration indicating an increased biological age for the 3xTg-AD mice compared to non-pathological C57BL/6J mice, which was more pronounced in the cortex as compared to the hippocampus. Genomic region enrichment analysis revealed that age-dependent CpGs were enriched in genes related to developmental, aging-related, neuronal and neurodegenerative functions. Due to the limited access of human brain tissues, these epigenetic clocks specific for mouse cortex and hippocampus might be important in further unravelling the role of epigenetic mechanisms underlying AD pathology or brain aging in general.
Collapse
Affiliation(s)
- Emma Coninx
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol 2400, Belgium
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven 3000, Belgium
| | - Yap Ching Chew
- Epigenetics Technologies, Zymo Research Corporation, Irvine, CA 92614, USA
| | - Xiaojing Yang
- Epigenetics Technologies, Zymo Research Corporation, Irvine, CA 92614, USA
| | - Wei Guo
- Epigenetics Technologies, Zymo Research Corporation, Irvine, CA 92614, USA
| | - Amelie Coolkens
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol 2400, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol 2400, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven 3000, Belgium
| | - Mieke Verslegers
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol 2400, Belgium
| | - Roel Quintens
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol 2400, Belgium
| |
Collapse
|
23
|
Salemme V, Angelini C, Chapelle J, Centonze G, Natalini D, Morellato A, Taverna D, Turco E, Ala U, Defilippi P. The p140Cap adaptor protein as a molecular hub to block cancer aggressiveness. Cell Mol Life Sci 2020; 78:1355-1367. [PMID: 33079227 PMCID: PMC7904710 DOI: 10.1007/s00018-020-03666-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/03/2020] [Accepted: 10/05/2020] [Indexed: 01/03/2023]
Abstract
The p140Cap adaptor protein is a scaffold molecule encoded by the SRCIN1 gene, which is physiologically expressed in several epithelial tissues and in the neurons. However, p140Cap is also strongly expressed in a significant subset of cancers including breast cancer and neuroblastoma. Notably, cancer patients with high p140Cap expression in their primary tumors have a lower probability of developing a distant event and ERBB2-positive breast cancer sufferers show better survival. In neuroblastoma patients, SRCIN1 mRNA levels represent an independent risk factor, which is inversely correlated to disease aggressiveness. Consistent with clinical data, SRCIN1 gain or loss of function mouse models demonstrated that p140Cap may affect tumor growth and metastasis formation by controlling the signaling pathways involved in tumorigenesis and metastatic features. This study reviews data showing the relevance of SRCIN1/p140Cap in cancer patients, the impact of SRCIN1 status on p140Cap expression, the specific mechanisms through which p140Cap can limit cancer progression, the molecular functions regulated by p140Cap, along with the p140Cap interactome, to unveil its key role for patient stratification in clinics.
Collapse
Affiliation(s)
- Vincenzo Salemme
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Costanza Angelini
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Jennifer Chapelle
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Giorgia Centonze
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Daniela Taverna
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Ugo Ala
- Department of Veterinary Sciences, Università degli Studi di Torino, Largo Paolo Braccini 2, 10095, Grugliasco, TO, Italy.
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy.
| |
Collapse
|
24
|
Ligon C, Seong E, Schroeder EJ, DeKorver NW, Yuan L, Chaudoin TR, Cai Y, Buch S, Bonasera SJ, Arikkath J. δ-Catenin engages the autophagy pathway to sculpt the developing dendritic arbor. J Biol Chem 2020; 295:10988-11001. [PMID: 32554807 DOI: 10.1074/jbc.ra120.013058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/14/2020] [Indexed: 01/21/2023] Open
Abstract
The development of the dendritic arbor in pyramidal neurons is critical for neural circuit function. Here, we uncovered a pathway in which δ-catenin, a component of the cadherin-catenin cell adhesion complex, promotes coordination of growth among individual dendrites and engages the autophagy mechanism to sculpt the developing dendritic arbor. Using a rat primary neuron model, time-lapse imaging, immunohistochemistry, and confocal microscopy, we found that apical and basolateral dendrites are coordinately sculpted during development. Loss or knockdown of δ-catenin uncoupled this coordination, leading to retraction of the apical dendrite without altering basolateral dendrite dynamics. Autophagy is a key cellular pathway that allows degradation of cellular components. We observed that the impairment of the dendritic arbor resulting from δ-catenin knockdown could be reversed by knockdown of autophagy-related 7 (ATG7), a component of the autophagy machinery. We propose that δ-catenin regulates the dendritic arbor by coordinating the dynamics of individual dendrites and that the autophagy mechanism may be leveraged by δ-catenin and other effectors to sculpt the developing dendritic arbor. Our findings have implications for the management of neurological disorders, such as autism and intellectual disability, that are characterized by dendritic aberrations.
Collapse
Affiliation(s)
- Cheryl Ligon
- Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Eunju Seong
- Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Ethan J Schroeder
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Nicholas W DeKorver
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Li Yuan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Tammy R Chaudoin
- Division of Geriatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Yu Cai
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Stephen J Bonasera
- Division of Geriatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jyothi Arikkath
- Department of Anatomy, Howard University, Washington, D. C., USA
| |
Collapse
|
25
|
Wang M, Yu Z, Li G, Yu X. Multiple Morphological Factors Underlie Experience-Dependent Cross-Modal Plasticity in the Developing Sensory Cortices. Cereb Cortex 2020; 30:2418-2433. [PMID: 31828301 DOI: 10.1093/cercor/bhz248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 08/21/2019] [Accepted: 09/18/2019] [Indexed: 11/14/2022] Open
Abstract
Sensory experience regulates the structural and functional wiring of sensory cortices. In previous work, we showed that whisker deprivation (WD) from birth not only reduced excitatory synaptic transmission of layer (L) 2/3 pyramidal neurons of the correspondent barrel cortex in mice, but also cross-modally reduced synaptic transmission of L2/3 pyramidal neurons in other sensory cortices. Here, we used in utero electroporation, in combination with optical clearing, to examine the main morphological components regulating neural circuit wiring, namely presynaptic bouton density, spine density, as well as dendrite and axon arbor lengths. We found that WD from P0 to P14 reduced presynaptic bouton density in both L4 and L2/3 inputs to L2/3 pyramidal neurons, as well as spine density across the dendritic tree of L2/3 pyramidal neurons, in the barrel field of the primary somatosensory cortex. The cross-modal effects in the primary auditory cortex were manifested mostly as reduced dendrite and axon arbor size, as well as reduced bouton density of L2/3 inputs. Increasing sensory experience by rearing mice in an enriched environment rescued the effects of WD. Together, these results demonstrate that multiple morphological factors contribute to experience-dependent structural plasticity during early wiring of the sensory cortices.
Collapse
Affiliation(s)
- Miao Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zixian Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guangying Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiang Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,School of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
26
|
Cardozo PL, de Lima IBQ, Maciel EMA, Silva NC, Dobransky T, Ribeiro FM. Synaptic Elimination in Neurological Disorders. Curr Neuropharmacol 2020; 17:1071-1095. [PMID: 31161981 PMCID: PMC7052824 DOI: 10.2174/1570159x17666190603170511] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/23/2019] [Accepted: 05/31/2019] [Indexed: 12/12/2022] Open
Abstract
Synapses are well known as the main structures responsible for transmitting information through the release and recognition of neurotransmitters by pre- and post-synaptic neurons. These structures are widely formed and eliminated throughout the whole lifespan via processes termed synaptogenesis and synaptic pruning, respectively. Whilst the first pro-cess is needed for ensuring proper connectivity between brain regions and also with the periphery, the second phenomenon is important for their refinement by eliminating weaker and unnecessary synapses and, at the same time, maintaining and fa-voring the stronger ones, thus ensuring proper synaptic transmission. It is well-known that synaptic elimination is modulated by neuronal activity. However, only recently the role of the classical complement cascade in promoting this phenomenon has been demonstrated. Specifically, microglial cells recognize activated complement component 3 (C3) bound to synapses tar-geted for elimination, triggering their engulfment. As this is a highly relevant process for adequate neuronal functioning, dis-ruptions or exacerbations in synaptic pruning could lead to severe circuitry alterations that could underlie neuropathological alterations typical of neurological and neuropsychiatric disorders. In this review, we focus on discussing the possible in-volvement of excessive synaptic elimination in Alzheimer’s disease, as it has already been reported dendritic spine loss in post-synaptic neurons, increased association of complement proteins with its synapses and, hence, augmented microglia-mediated pruning in animal models of this disorder. In addition, we briefly discuss how this phenomenon could be related to other neurological disorders, including multiple sclerosis and schizophrenia.
Collapse
Affiliation(s)
- Pablo L Cardozo
- Laboratório de Neurobioquímica, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabella B Q de Lima
- Laboratório de Neurobioquímica, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Esther M A Maciel
- Laboratório de Neurobioquímica, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Nathália C Silva
- Laboratório de Neurobioquímica, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Fabíola M Ribeiro
- Laboratório de Neurobioquímica, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
27
|
Chapelle J, Sorokina O, McLean C, Salemme V, Alfieri A, Angelini C, Morellato A, Adrait A, Menna E, Matteoli M, Couté Y, Ala U, Turco E, Defilippi P, Armstrong JD. Dissecting the Shared and Context-Dependent Pathways Mediated by the p140Cap Adaptor Protein in Cancer and in Neurons. Front Cell Dev Biol 2019; 7:222. [PMID: 31681758 PMCID: PMC6803390 DOI: 10.3389/fcell.2019.00222] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/19/2019] [Indexed: 12/26/2022] Open
Abstract
The p140Cap adaptor protein is a scaffold molecule physiologically expressed in few epithelial tissues, such as the mammary gland, and in differentiated neurons. While the role of p140Cap in mammary gland epithelia is not still understood, we already know that a significant subset of breast cancers express p140Cap. In the subgroup of ERBB2-amplified breast cancers, a high p140Cap status predicts a significantly lower probability of developing a distant event and a clear difference in survival. p140Cap is causal in dampening ERBB2-positive tumor cell progression, impairing tumor onset and growth, and counteracting epithelial mesenchymal transition, resulting in decreased metastasis formation. Since only a few p140Cap interacting proteins have been identified in breast cancer and the molecular complexes and pathways underlying the cancer function of p140Cap are largely unknown, we generated a p140Cap interactome from ERBB2-positive breast cancer cells, identifying cancer specific components and those shared with the synaptic interactome. We identified 373 interacting proteins in cancer cells, including those with functions relevant to cell adhesion, protein homeostasis, regulation of cell cycle and apoptosis, which are frequently deregulated in cancer. Within the interactome, we identified 15 communities (clusters) with topology-functional relationships. In neurons, where p140Cap is key in regulating synaptogenesis, synaptic transmission and synaptic plasticity, it establishes an extensive interactome with proteins that cluster to sub complexes located in the postsynaptic density. p140Cap interactors converge on key synaptic processes, including synaptic transmission, actin cytoskeleton remodeling and cell-cell junction organization. Comparing the breast cancer to the synaptic interactome, we found 39 overlapping proteins, a relatively small overlap. However, cell adhesion and remodeling of actin cytoskeleton clearly emerge as common terms in the shared subset. Thus, the functional signature of the two interactomes is primarily determined by organ/tissue and functional specificity, while the overlap provides a list of shared functional terms, which might be linked to both cancer and neurological functions.
Collapse
Affiliation(s)
- Jennifer Chapelle
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Turin, Italy
| | - Oksana Sorokina
- Simons Initiative for the Developing Brain, School of Informatics, The University of Edinburgh, Edinburgh, United Kingdom
| | - Colin McLean
- Simons Initiative for the Developing Brain, School of Informatics, The University of Edinburgh, Edinburgh, United Kingdom
| | - Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Turin, Italy
| | - Annalisa Alfieri
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Turin, Italy
| | - Costanza Angelini
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Turin, Italy
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Turin, Italy
| | - Annie Adrait
- Univ. Grenoble Alpes, CEA, INSERM, IRIG, BGE, Grenoble, France
| | - Elisabetta Menna
- Institute of Neuroscience, CNR, Milan, Italy
- Istituto Clinico Humanitas, IRCCS, Rozzano, Italy
| | - Michela Matteoli
- Institute of Neuroscience, CNR, Milan, Italy
- Istituto Clinico Humanitas, IRCCS, Rozzano, Italy
| | - Yohann Couté
- Univ. Grenoble Alpes, CEA, INSERM, IRIG, BGE, Grenoble, France
| | - Ugo Ala
- Department of Veterinary Sciences, Università degli Studi di Torino, Turin, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Turin, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Turin, Italy
| | - J. Douglas Armstrong
- Simons Initiative for the Developing Brain, School of Informatics, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
28
|
Petrov AM, Mast N, Li Y, Pikuleva IA. The key genes, phosphoproteins, processes, and pathways affected by efavirenz-activated CYP46A1 in the amyloid-decreasing paradigm of efavirenz treatment. FASEB J 2019; 33:8782-8798. [PMID: 31063705 DOI: 10.1096/fj.201900092r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Efavirenz (EFV) is an anti-HIV drug, and cytochrome P450 46A1 (CYP46A1) is the major brain cholesterol hydroxylase. Previously, we discovered that EFV activates CYP46A1 and improves behavioral performance in 5XFAD mice, an Alzheimer's disease model. Herein, the unbiased omics and other approaches were used to study 5XFAD mice in the amyloid-decreasing paradigm of CYP46A1 activation by EFV. These approaches revealed increases in the brain levels of postsynaptic density protein 95, gephyrin, synaptophysin, synapsin, glial fibrillary acidic protein, and CYP46A1 and documented altered expression and phosphorylation of 66 genes and 77 proteins, respectively. The data obtained pointed to EFV effects at the synaptic level, plasmin-depended amyloid clearance, inflammation and microglia phenotype, oxidative stress and cellular hypoxia, autophagy and ubiquitin-proteasome systems as well as apoptosis. These effects could be realized in part via changes in the Ca2+-, small GTPase, and catenin signaling. A model is proposed, in which CYP46A1-dependent lipid raft rearrangement and subsequent decrease of protein phosphorylation are central in EFV effects and explain behavioral improvements in EFV-treated 5XFAD mice.-Petrov, A. M., Mast, N., Li, Y., Pikuleva, I. A. The key genes, phosphoproteins, processes, and pathways affected by efavirenz-activated CYP46A1 in the amyloid-decreasing paradigm of efavirenz treatment.
Collapse
Affiliation(s)
- Alexey M Petrov
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yong Li
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
29
|
Han KA, Um JW, Ko J. Intracellular protein complexes involved in synapse assembly in presynaptic neurons. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 116:347-373. [PMID: 31036296 DOI: 10.1016/bs.apcsb.2018.11.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The presynaptic active zone, composed of evolutionarily conserved protein complexes, is a specialized area that serves to orchestrate precise and efficient neurotransmitter release by organizing various presynaptic proteins involved in mediating docking and priming of synaptic vesicles, recruiting voltage-gated calcium channels, and modulating presynaptic nerve terminals with aligned postsynaptic structures. Among membrane proteins localized to active zone, presynaptic neurexins and LAR-RPTPs (leukocyte common antigen-related receptor tyrosine phosphatase) have emerged as hubs that orchestrate both shared and distinct extracellular synaptic adhesion pathways. In this chapter, we discuss intracellular signaling cascades involved in recruiting various intracellular proteins at both excitatory and inhibitory synaptic sites. In particular, we highlight recent studies on key active zone proteins that physically and functionally link these cascades with neurexins and LAR-RPTPs in both vertebrate and invertebrate model systems. These studies allow us to build a general, universal view of how presynaptic active zones operate together with postsynaptic structures in neural circuits.
Collapse
Affiliation(s)
- Kyung Ah Han
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea
| | - Ji Won Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea.
| |
Collapse
|
30
|
Liu XD, Zhu XN, Halford MM, Xu TL, Henkemeyer M, Xu NJ. Retrograde regulation of mossy fiber axon targeting and terminal maturation via postsynaptic Lnx1. J Cell Biol 2018; 217:4007-4024. [PMID: 30185604 PMCID: PMC6219728 DOI: 10.1083/jcb.201803105] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/25/2018] [Accepted: 08/14/2018] [Indexed: 11/22/2022] Open
Abstract
Synapse formation relies on the coordination of dynamic pre- and postsynaptic structures during brain development. Liu et al. reveal that presynaptic terminal maturation of mossy fiber axons is retrogradely regulated by postsynaptic scaffold protein Lnx1 via stabilizing EphB receptor kinases. Neuronal connections are initiated by axon targeting to form synapses. However, how the maturation of axon terminals is modulated through interacting with postsynaptic elements remains elusive. In this study, we find that ligand of Numb protein X 1 (Lnx1), a postsynaptic PDZ protein expressed in hippocampal CA3 pyramidal neurons, is essential for mossy fiber (MF) axon targeting during the postnatal period. Lnx1 deletion causes defective synaptic arrangement that leads to aberrant presynaptic terminals. We further identify EphB receptors as novel Lnx1-binding proteins to form a multiprotein complex that is stabilized on the CA3 neuron membrane through preventing proteasome activity. EphB1 and EphB2 are independently required to transduce distinct signals controlling MF pruning and targeting for precise DG-CA3 synapse formation. Furthermore, constitutively active EphB2 kinase rescues structure of the wired MF terminals in Lnx1 mutant mice. Our data thus define a retrograde trans-synaptic regulation required for integration of post- and presynaptic structure that participates in building hippocampal neural circuits during the adolescence period.
Collapse
Affiliation(s)
- Xian-Dong Liu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Na Zhu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Michael M Halford
- Department of Neuroscience, Kent Waldrep Center for Basic Research on Nerve Growth and Regeneration, University of Texas Southwestern Medical Center, Dallas, TX
| | - Tian-Le Xu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mark Henkemeyer
- Department of Neuroscience, Kent Waldrep Center for Basic Research on Nerve Growth and Regeneration, University of Texas Southwestern Medical Center, Dallas, TX
| | - Nan-Jie Xu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China .,Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai China
| |
Collapse
|
31
|
Chevée M, Brown SP. The development of local circuits in the neocortex: recent lessons from the mouse visual cortex. Curr Opin Neurobiol 2018; 53:103-109. [PMID: 30053693 DOI: 10.1016/j.conb.2018.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/14/2018] [Accepted: 06/14/2018] [Indexed: 12/26/2022]
Abstract
Precise synaptic connections among neurons in the neocortex generate the circuits that underlie a broad repertoire of cortical functions including perception, learning and memory, and complex problem solving. The specific patterns and properties of these synaptic connections are fundamental to the computations cortical neurons perform. How such specificity arises in cortical circuits has remained elusive. Here, we first consider the cell-type, subcellular and synaptic specificity required for generating mature patterns of cortical connectivity and responses. Next, we focus on recent progress in understanding how the synaptic connections among excitatory cortical projection neurons are established during development using the primary visual cortex of the mouse as a model.
Collapse
Affiliation(s)
- Maxime Chevée
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Solange P Brown
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|