1
|
Kim H, Jeon Y, Kim S, Guo Y, Kim D, Jang G, Brasch J, Um JW, Ko J. EphB2 receptor tyrosine kinase-mediated excitatory synaptic functions are negatively modulated by MDGA2. Prog Neurobiol 2025; 250:102772. [PMID: 40316130 DOI: 10.1016/j.pneurobio.2025.102772] [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: 01/03/2025] [Revised: 03/24/2025] [Accepted: 04/24/2025] [Indexed: 05/04/2025]
Abstract
MDGA2 is an excitatory synapse-specific suppressor that uses distinct extracellular mechanisms to negatively regulate various postsynaptic properties. Here, we identify EphB2, an excitatory synapse-specific receptor tyrosine kinase, as a new binding partner for MDGA2. The first three immunoglobulin domains of MDGA2 undergo cis-binding to the ligand-binding domain of EphB2, enabling MDGA2 to compete with Ephrin-B1 for binding to EphB2. Moreover, EphB2 forms complexes with MDGA2 and GluN2B-containing NMDA receptors (NMDARs) in mouse brains. MDGA2 deletion promotes formation of the EphB2/Ephrin-B1 complex but does not alter the surface expression levels and Ephrin-stimulated activation of EphB2 receptors and downstream GluN2B-containing NMDARs in cultured neurons. AlphaFold-based molecular replacement experiments reveal that MDGA2 must bind EphB2 to suppress spontaneous synaptic transmission and NMDAR-mediated, but not AMPAR-mediated, postsynaptic responses at excitatory synapses in cultured neurons. These results collectively suggest that MDGA2 is a versatile factor that suppresses distinct excitatory postsynaptic properties via different transsynaptic pathways.
Collapse
Affiliation(s)
- Hyeonho Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, South Korea; Center for Synapse Diversity and Specificity, DGIST, Daegu 42988, South Korea
| | - Younghyeon Jeon
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, South Korea; Center for Synapse Diversity and Specificity, DGIST, Daegu 42988, South Korea
| | - Seunghye Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, South Korea; Center for Synapse Diversity and Specificity, DGIST, Daegu 42988, South Korea
| | - Yuxuan Guo
- Department of Biochemistry, Spencer Fox Eccles School of Medicine, The University of Utah, Salt Lake City, UT 84112, USA
| | - Dongwook Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, South Korea; Center for Synapse Diversity and Specificity, DGIST, Daegu 42988, South Korea
| | - Gyubin Jang
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, South Korea; Center for Synapse Diversity and Specificity, DGIST, Daegu 42988, South Korea
| | - Julia Brasch
- Department of Biochemistry, Spencer Fox Eccles School of Medicine, The University of Utah, Salt Lake City, UT 84112, USA
| | - Ji Won Um
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, South Korea; Center for Synapse Diversity and Specificity, DGIST, Daegu 42988, South Korea.
| | - Jaewon Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, South Korea; Center for Synapse Diversity and Specificity, DGIST, Daegu 42988, South Korea.
| |
Collapse
|
2
|
Langlais VC, Mountadem S, Benazzouz I, Amadio A, Matos M, Jourdes A, Cannich A, Julio-Kalajzic F, Belluomo I, Matias I, Maitre M, Lesté-Lasserre T, Marais S, Avignone E, Marsicano G, Bellocchio L, Oliet SHR, Panatier A. Astrocytic EphB3 receptors regulate d-serine-gated synaptic plasticity and memory. Prog Neurobiol 2025; 248:102747. [PMID: 40081519 DOI: 10.1016/j.pneurobio.2025.102747] [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: 11/04/2024] [Revised: 02/04/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
The activation of classical NMDA receptors (NMDARs) requires the binding of a co-agonist in addition to glutamate. Whereas astrocytic-derived d-serine was shown to play such a role at CA3-CA1 hippocampal synapses, the exact mechanism by which neurons interact with neighboring astrocytes to regulate synaptic d-serine availability remains to be fully elucidated. Considering the close anatomical apposition of astrocytic and neuronal elements at synapses, the aforementioned process is likely to involve cells adhesion molecules. One very likely candidate could be the astrocytic EphB3 receptor and its neuronal partner, ephrinB3. Here, we first showed in acute hippocampal slices from adult mice that stimulation of EphB3 receptors with exogenous ephrinB3 increased d-serine availability at CA3-CA1 synapses, resulting in an increased NMDAR activity. Conversely, inhibiting endogenous EphB3 receptors caused an impairment of both synaptic NMDAR activity and NMDAR-dependent long-term synaptic potentiation (LTP), effects that could be rescued by exogenous d-serine. Most interestingly, knocking down EphB3 receptors specifically in astrocytes yielded a similar impairment in hippocampal plasticity and, most importantly, caused a deficit in novel object recognition memory. Altogether, our data thus indicate that EphB3 receptors in hippocampal astrocytes play a key role in regulating synaptic NMDAR function, activity-dependent plasticity and memory.
Collapse
Affiliation(s)
| | - Sarah Mountadem
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Ines Benazzouz
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Aurélie Amadio
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Marco Matos
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Aurélie Jourdes
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Astrid Cannich
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Francisca Julio-Kalajzic
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France; Univ. Bordeaux, CNRS, INSERM, BIC, US4, UAR 3420, Bordeaux F-33000, France; Lead contact, France
| | - Ilaria Belluomo
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Isabelle Matias
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Marlène Maitre
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | | | - Sébastien Marais
- Univ. Bordeaux, CNRS, INSERM, BIC, US4, UAR 3420, Bordeaux F-33000, France
| | - Elena Avignone
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Giovanni Marsicano
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Luigi Bellocchio
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | | | - Aude Panatier
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France; Lead contact, France.
| |
Collapse
|
3
|
Akter Y, Jones G, Daskivich GJ, Shifflett V, Vargas KJ, Hruska M. Combining nanobody labeling with STED microscopy reveals input-specific and layer-specific organization of neocortical synapses. PLoS Biol 2025; 23:e3002649. [PMID: 40184426 PMCID: PMC12002638 DOI: 10.1371/journal.pbio.3002649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 04/16/2025] [Accepted: 02/25/2025] [Indexed: 04/06/2025] Open
Abstract
The discovery of synaptic nanostructures revealed key insights into the molecular logic of synaptic function and plasticity. Yet, our understanding of how diverse synapses in the brain organize their nano-architecture remains elusive, largely due to the limitations of super-resolution imaging in complex brain tissue. Here, we characterized single-domain camelid nanobodies for the 3D quantitative multiplex imaging of synaptic nano-organization using tau-STED nanoscopy in cryosections from the mouse primary somatosensory cortex. We focused on thalamocortical (TC) and corticocortical (CC) synapses along the apical-basal axis of layer five pyramidal neurons as models of functionally diverse glutamatergic synapses in the brain. Spines receiving TC input were larger than those receiving CC input in all layers examined. However, the nano-architecture of TC synapses varied with dendritic location. TC afferents on apical dendrites frequently contacted spines with multiple aligned PSD-95/Bassoon nanomodules of constant size. In contrast, TC spines on basal dendrites predominantly contained a single aligned nanomodule, with PSD-95 nanocluster sizes scaling proportionally with spine volume. The nano-organization of CC synapses did not change across cortical layers and resembled modular architecture defined in vitro. These findings highlight the nanoscale diversity of synaptic architecture in the brain, that is, shaped by both the source of afferent input and the subcellular localization of individual synaptic contacts.
Collapse
Affiliation(s)
- Yeasmin Akter
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, United States of America
| | - Grace Jones
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, United States of America
| | - Grant J. Daskivich
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Victoria Shifflett
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, United States of America
| | - Karina J. Vargas
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Martin Hruska
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, United States of America
| |
Collapse
|
4
|
Zhou W, Munoz JR, Henry Ho HY, Hanamura K, Dalva MB. Specific neuroblast-derived signals control both cell migration and fate in the rostral migratory stream. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638163. [PMID: 40027825 PMCID: PMC11870606 DOI: 10.1101/2025.02.18.638163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Functional neuronal circuits require neuroblasts migrate to appropriate locations and then differentiate into neuronal subtypes. However, it remains unknown how neuroblasts in the subventricular zone (SVZ) are guided through the rostral migratory stream (RMS) to the olfactory bulb (OB). Here we define EphB2 as a neuroblast-derived cue that controls migration along the RMS and helps to determine cell fate. Within the RMS, EphB2 is expressed selectively in, kinase-active in, and required for the migration of neuroblasts. As neuroblasts enter the OB and differentiate, EphB kinase activity is down-regulated, and in the granule cell layer (GCL), EphB2 expression is down-regulated. Blocking EphB kinase activity or knocking down EphB2 results in defects in migration and premature cellular differentiation in the RMS. Unexpectedly, premature loss of EphB2 expression causes neuroblasts to stop migrating and differentiate into astrocyte-like cells. Thus, EphB2 kinase activity and expression are linked to migration and specification of neuroblast fate.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Neuroscience and the Jefferson Synaptic Biology Center, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
| | - James R. Munoz
- Present address: Department of Psychology and Neuroscience, Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, FL 33314, USA
| | - Hsin-Yi Henry Ho
- Department of Cell Biology and Human Anatomy, UC Davis School of Medicine, One Shields Avenue, Davis, CA 95616
| | - Kenji Hanamura
- Department of Neuroscience and the Jefferson Synaptic Biology Center, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
- Present address: Faculty of Medical Technology, Department of Radiological Technology, Niigata University of Health and Welfare, 1398 Shimami-cho, Kita-ku, Niigata-City, 950-3198, Japan
| | - Matthew B. Dalva
- Department of Neuroscience and the Jefferson Synaptic Biology Center, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
- Present Address: Tulane Brain Institute, Tulane University, 201 Flower Hall, 6823 St. Charles Avenue, New Orleans, LA 70118
| |
Collapse
|
5
|
Zhu X, Huang Y, Qiu J, Zhong Z, Peng Y, Liang X, Chen J, Zhou J, Liang X, Wang H, Xie W, Ding Y. Chaihu Guizhi Decoction prevents cognitive, memory impairments and sensorimotor gating deficit induced by N-methyl-d-aspartate receptor antibody in mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118806. [PMID: 39278296 DOI: 10.1016/j.jep.2024.118806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/18/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Anti-NMDAR encephalitis is one of the most common types of autoimmune encephalitis, primarily presenting with prodromal symptoms, such as fever and headache, followed by a range of neurological and psychiatric symptoms. Chaihu Guizhi Decoction (CGD), a traditional Chinese medicine formulated by Zhang Zhongjing in the Eastern Han Dynasty, has been effectively used in clinical practice to treat the symptoms of Taiyang and Shaoyang disorders, including fever, headache, and psychiatric disorders. AIM OF THE STUDY To demonstrate the protective effects of CGD in an animal model of anti-NMDAR encephalitis and explore the potential mechanisms involved. MATERIALS AND METHODS UHPLC-HRMS was used to identify CGD's chemical components and serum metabolomic profiles. Network pharmacology and molecular docking were performed to predict potential targets of CGD for the treatment of anti-NMDAR encephalitis. The effect of CGD on anti-NMDAR encephalitis was evaluated using a mouse model induced by patients' antibodies. Behavioral tests were performed to assess cognitive impairment and schizophrenia-like behaviors. The effect of CGD on the cell-surface NMDAR GluN1 subunit in cultured neurons treated with patient antibodies was detected by immunofluorescence. Golgi staining was used to observe morphological changes in hippocampal dendrites. The expression of NMDAR-interacting proteins and various neuroreceptors in the hippocampus were examined to validate the targets predicted using network pharmacology and molecular docking. RESULTS CGD alleviated cognitive, memory, and sensorimotor gating deficits in mice treated with anti-NMDAR encephalitis patients' antibodies. Further experiments demonstrated the effect of CGD in preventing NMDAR reduction both in vitro and in vivo. Meanwhile, CGD regulated NMDAR-interacting proteins and dopamine receptors but did not affect hippocampal dendritic morphology and synaptic density. Additionally, CGD modifies metabolic pathways associated with anti-NMDAR encephalitis and other neurological and psychiatric disorders. CONCLUSIONS CGD exhibited protective effects against anti-NMDAR encephalitis by mitigating the antibody-induced reduction in NMDAR and NMDAR-interacting proteins.
Collapse
Affiliation(s)
- Xiaoyu Zhu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yingyi Huang
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Jing Qiu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zheng Zhong
- Department of Chemistry and the Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Yu Peng
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Xiaoshan Liang
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jinyu Chen
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jieli Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Xiaotao Liang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Honghao Wang
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China.
| | - Wei Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Yuewen Ding
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
6
|
Wang H, Xie C, Deng B, Ding J, Li N, Kou Z, Jin M, He J, Wang Q, Wen H, Zhang J, Zhou Q, Chen S, Chen X, Yuan TF, Zhu S. Structural basis for antibody-mediated NMDA receptor clustering and endocytosis in autoimmune encephalitis. Nat Struct Mol Biol 2024; 31:1987-1996. [PMID: 39227720 PMCID: PMC11638077 DOI: 10.1038/s41594-024-01387-3] [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: 10/29/2023] [Accepted: 08/07/2024] [Indexed: 09/05/2024]
Abstract
Antibodies against N-methyl-D-aspartate receptors (NMDARs) are most frequently detected in persons with autoimmune encephalitis (AE) and used as diagnostic biomarkers. Elucidating the structural basis of monoclonal antibody (mAb) binding to NMDARs would facilitate the development of targeted therapy for AE. Here, we reconstructed nanodiscs containing green fluorescent protein-fused NMDARs to label and sort individual immune B cells from persons with AE and further cloned and identified mAbs against NMDARs. This allowed cryo-electron microscopy analysis of NMDAR-Fab complexes, revealing that autoantibodies bind to the R1 lobe of the N-terminal domain of the GluN1 subunit. Small-angle X-ray scattering studies demonstrated NMDAR-mAb stoichiometry of 2:1 or 1:2, structurally suitable for mAb-induced clustering and endocytosis of NMDARs. Importantly, these mAbs reduced the surface NMDARs and NMDAR-mediated currents, without tonically affecting NMDAR channel gating. These structural and functional findings imply that the design of neutralizing antibody binding to the R1 lobe of NMDARs represents a potential therapy for AE treatment.
Collapse
MESH Headings
- Humans
- Receptors, N-Methyl-D-Aspartate/chemistry
- Receptors, N-Methyl-D-Aspartate/metabolism
- Receptors, N-Methyl-D-Aspartate/immunology
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/metabolism
- Encephalitis/immunology
- Encephalitis/metabolism
- Encephalitis/pathology
- Endocytosis
- Autoantibodies/immunology
- Autoantibodies/metabolism
- Cryoelectron Microscopy
- Hashimoto Disease/immunology
- Hashimoto Disease/metabolism
- Models, Molecular
- HEK293 Cells
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/metabolism
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Han Wang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chun Xie
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Bo Deng
- Department of Neurology, Huashan Hospital and Institute of Neurology, National Center for Neurological Disorders, Fudan University, Shanghai, China
| | - Jinjun Ding
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine and School of Psychology, Shanghai, China
| | - Na Li
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Zengwei Kou
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Mengmeng Jin
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jie He
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | | | - Han Wen
- DP Technology, Beijing, China
| | - Jinbao Zhang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Qinming Zhou
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Sheng Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Xiangjun Chen
- Department of Neurology, Huashan Hospital and Institute of Neurology, National Center for Neurological Disorders, Fudan University, Shanghai, China.
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine and School of Psychology, Shanghai, China.
| | - Shujia Zhu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
7
|
Sutley-Koury SN, Taitano-Johnson C, Kulinich AO, Farooq N, Wagner VA, Robles M, Hickmott PW, Santhakumar V, Mimche PN, Ethell IM. EphB2 Signaling Is Implicated in Astrocyte-Mediated Parvalbumin Inhibitory Synapse Development. J Neurosci 2024; 44:e0154242024. [PMID: 39327008 PMCID: PMC11551896 DOI: 10.1523/jneurosci.0154-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 09/09/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
Impaired inhibitory synapse development is suggested to drive neuronal hyperactivity in autism spectrum disorders (ASD) and epilepsy. We propose a novel mechanism by which astrocytes control the development of parvalbumin (PV)-specific inhibitory synapses in the hippocampus, implicating ephrin-B/EphB signaling. Here, we utilize genetic approaches to assess functional and structural connectivity between PV and pyramidal cells (PCs) through whole-cell patch-clamp electrophysiology, optogenetics, immunohistochemical analysis, and behaviors in male and female mice. While inhibitory synapse development is adversely affected by PV-specific expression of EphB2, a strong candidate ASD risk gene, astrocytic ephrin-B1 facilitates PV→PC connectivity through a mechanism involving EphB signaling in PV boutons. In contrast, the loss of astrocytic ephrin-B1 reduces PV→PC connectivity and inhibition, resulting in increased seizure susceptibility and an ASD-like phenotype. Our findings underscore the crucial role of astrocytes in regulating inhibitory circuit development and discover a new role of EphB2 receptors in PV-specific inhibitory synapse development.
Collapse
Affiliation(s)
- Samantha N Sutley-Koury
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California 92521
| | - Christopher Taitano-Johnson
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California 92521
- Neuroscience Graduate Program, University of California Riverside, Riverside, California 92521
| | - Anna O Kulinich
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California 92521
| | - Nadia Farooq
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California 92521
| | - Victoria A Wagner
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California 92521
- Neuroscience Graduate Program, University of California Riverside, Riverside, California 92521
| | - Marissa Robles
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California 92521
| | - Peter W Hickmott
- Neuroscience Graduate Program, University of California Riverside, Riverside, California 92521
| | | | - Patrice N Mimche
- Department of Dermatology, and Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis Indiana 46202
| | - Iryna M Ethell
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California 92521
- Neuroscience Graduate Program, University of California Riverside, Riverside, California 92521
| |
Collapse
|
8
|
David ET, Yousuf MS, Mei HR, Jain A, Krishnagiri S, Elahi H, Venkatesan R, Srikanth KD, Dussor G, Dalva MB, Price TJ. ephrin-B2 promotes nociceptive plasticity and hyperalgesic priming through EphB2-MNK-eIF4E signaling in both mice and humans. Pharmacol Res 2024; 206:107284. [PMID: 38925462 DOI: 10.1016/j.phrs.2024.107284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 06/28/2024]
Abstract
Ephrin-B-EphB signaling can promote pain through ligand-receptor interactions between peripheral cells, like immune cells expressing ephrin-Bs, and EphB receptors expressed by DRG neurons. Previous studies have shown increased ephrin-B2 expression in peripheral tissues like synovium of rheumatoid and osteoarthritis patients, indicating the clinical significance of this signaling. The primary goal of this study was to understand how ephrin-B2 acts on mouse and human DRG neurons, which express EphB receptors, to promote pain and nociceptor plasticity. We hypothesized that ephrin-B2 would promote nociceptor plasticity and hyperalgesic priming through MNK-eIF4E signaling, a critical mechanism for nociceptive plasticity induced by growth factors, cytokines and nerve injury. Both male and female mice developed dose-dependent mechanical hypersensitivity in response to ephrin-B2, and both sexes showed hyperalgesic priming when challenged with PGE2 injection either to the paw or the cranial dura. Acute nociceptive behaviors and hyperalgesic priming were blocked in mice lacking MNK1 (Mknk1 knockout mice) and by eFT508, a specific MNK inhibitor. Sensory neuron-specific knockout of EphB2 using Pirt-Cre demonstrated that ephrin-B2 actions require this receptor. In Ca2+-imaging experiments on cultured DRG neurons, ephrin-B2 treatment enhanced Ca2+ transients in response to PGE2 and these effects were absent in DRG neurons from MNK1-/- and EphB2-PirtCre mice. In experiments on human DRG neurons, ephrin-B2 increased eIF4E phosphorylation and enhanced Ca2+ responses to PGE2 treatment, both blocked by eFT508. We conclude that ephrin-B2 acts directly on mouse and human sensory neurons to induce nociceptor plasticity via MNK-eIF4E signaling, offering new insight into how ephrin-B signaling promotes pain.
Collapse
Affiliation(s)
- Eric T David
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Muhammad Saad Yousuf
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Hao-Ruei Mei
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Ashita Jain
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Sharada Krishnagiri
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Hajira Elahi
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Rupali Venkatesan
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Kolluru D Srikanth
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA; Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70124, USA; Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70124, USA
| | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Matthew B Dalva
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA; Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70124, USA; Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70124, USA
| | - Theodore J Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA.
| |
Collapse
|
9
|
Jamet Z, Mergaux C, Meras M, Bouchet D, Villega F, Kreye J, Prüss H, Groc L. NMDA receptor autoantibodies primarily impair the extrasynaptic compartment. Brain 2024; 147:2745-2760. [PMID: 38758090 PMCID: PMC11292910 DOI: 10.1093/brain/awae163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/18/2024] Open
Abstract
Autoantibodies directed against the N-methyl-D-aspartate receptor (NMDAR-Ab) are pathogenic immunoglobulins detected in patients suffering from NMDAR encephalitis. NMDAR-Ab alter the receptor membrane trafficking, synaptic transmission and neuronal network properties, leading to neurological and psychiatric symptoms in patients. Patients often have very little neuronal damage but rapid and massive (treatment-responsive) brain dysfunctions related to an unknown early mechanism of NMDAR-Ab. Our understanding of this early molecular cascade remains surprisingly fragmented. Here, we used a combination of single molecule-based imaging of membrane proteins to unveil the spatiotemporal action of NMDAR-Ab on live hippocampal neurons. We first demonstrate that different clones of NMDAR-Ab primarily affect extrasynaptic (and not synaptic) NMDARs. In the first minutes, NMDAR-Ab increase extrasynaptic NMDAR membrane dynamics, declustering its surface interactome. NMDAR-Ab also rapidly reshuffle all membrane proteins located in the extrasynaptic compartment. Consistent with this alteration of multiple proteins, effects of NMDAR-Ab were not mediated through the sole interaction between the NMDAR and EphB2 receptor. In the long term, NMDAR-Ab reduce the NMDAR synaptic pool by slowing down receptor membrane dynamics in a cross-linking-independent manner. Remarkably, exposing only extrasynaptic NMDARs to NMDAR-Ab was sufficient to produce their full-blown effect on synaptic receptors. Collectively, we demonstrate that NMDAR-Ab initially impair extrasynaptic proteins, then the synaptic ones. These data thus shed new and unsuspected light on the mode of action of NMDAR-Ab and, probably, our understanding of (extra)synaptopathies.
Collapse
Affiliation(s)
- Zoe Jamet
- Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, University of Bordeaux, CNRS, F-33000 Bordeaux, France
| | - Camille Mergaux
- Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, University of Bordeaux, CNRS, F-33000 Bordeaux, France
| | - Morgane Meras
- Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, University of Bordeaux, CNRS, F-33000 Bordeaux, France
| | - Delphine Bouchet
- Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, University of Bordeaux, CNRS, F-33000 Bordeaux, France
| | - Frédéric Villega
- Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, University of Bordeaux, CNRS, F-33000 Bordeaux, France
- Department of Pediatric Neurology, CIC-0005, University Children's Hospital of Bordeaux, F-33000 Bordeaux, France
| | - Jakob Kreye
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Harald Prüss
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, 10117 Berlin, Germany
| | - Laurent Groc
- Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, University of Bordeaux, CNRS, F-33000 Bordeaux, France
| |
Collapse
|
10
|
Kakegawa W, Paternain AV, Matsuda K, Aller MI, Iida I, Miura E, Nozawa K, Yamasaki T, Sakimura K, Yuzaki M, Lerma J. Kainate receptors regulate synaptic integrity and plasticity by forming a complex with synaptic organizers in the cerebellum. Cell Rep 2024; 43:114427. [PMID: 38986610 DOI: 10.1016/j.celrep.2024.114427] [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: 10/12/2023] [Revised: 03/11/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024] Open
Abstract
Kainate (KA)-type glutamate receptors (KARs) are implicated in various neuropsychiatric and neurological disorders through their ionotropic and metabotropic actions. However, compared to AMPA- and NMDA-type receptor functions, many aspects of KAR biology remain incompletely understood. Our study demonstrates an important role of KARs in organizing climbing fiber (CF)-Purkinje cell (PC) synapses and synaptic plasticity in the cerebellum, independently of their ion channel or metabotropic functions. The amino-terminal domain (ATD) of the GluK4 KAR subunit binds to C1ql1, provided by CFs, and associates with Bai3, an adhesion-type G protein-coupled receptor expressed in PC dendrites. Mice lacking GluK4 exhibit no KAR-mediated responses, reduced C1ql1 and Bai3 levels, and fewer CF-PC synapses, along with impaired long-term depression and oculomotor learning. Remarkably, introduction of the ATD of GluK4 significantly improves all these phenotypes. These findings demonstrate that KARs act as synaptic scaffolds, orchestrating synapses by forming a KAR-C1ql1-Bai3 complex in the cerebellum.
Collapse
Affiliation(s)
- Wataru Kakegawa
- Department of Neurophysiology, Keio University School of Medicine, Tokyo 160-8582, Japan.
| | - Ana V Paternain
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain
| | - Keiko Matsuda
- Department of Neurophysiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - M Isabel Aller
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain
| | - Izumi Iida
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Meguro, Tokyo 153-8902, Japan; Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Eriko Miura
- Department of Neurophysiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kazuya Nozawa
- Department of Neurophysiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Tokiwa Yamasaki
- Department of Neurophysiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Michisuke Yuzaki
- Department of Neurophysiology, Keio University School of Medicine, Tokyo 160-8582, Japan.
| | - Juan Lerma
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain.
| |
Collapse
|
11
|
Jones G, Akter Y, Shifflett V, Hruska M. Nanoscale analysis of functionally diverse glutamatergic synapses in the neocortex reveals input and layer-specific organization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.592008. [PMID: 38746319 PMCID: PMC11092571 DOI: 10.1101/2024.05.01.592008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Discovery of synaptic nanostructures suggests a molecular logic for the flexibility of synaptic function. We still have little understanding of how functionally diverse synapses in the brain organize their nanoarchitecture due to challenges associated with super-resolution imaging in complex brain tissue. Here, we characterized single-domain camelid nanobodies for the 3D quantitative multiplex imaging of synaptic nano-organization in 6 µm brain cryosections using STED nanoscopy. We focused on thalamocortical (TC) and corticocortical (CC) synapses along the apical-basal axis of layer 5 pyramidal neurons as models of functionally diverse glutamatergic synapses in the brain. Spines receiving TC input were larger than CC spines in all layers examined. However, TC synapses on apical and basal dendrites conformed to different organizational principles. TC afferents on apical dendrites frequently contacted spines with multiple aligned PSD-95/Bassoon nanomodules, which are larger. TC spines on basal dendrites contained mostly one aligned PSD-95/Bassoon nanocluster. However, PSD-95 nanoclusters were larger and scaled with spine volume. The nano-organization of CC synapses did not change across cortical layers. These results highlight striking nanoscale diversity of functionally distinct glutamatergic synapses, relying on afferent input and sub-cellular localization of individual synaptic connections.
Collapse
|
12
|
Srikanth KD, Elahi H, Chander P, Washburn HR, Hassler S, Mwirigi JM, Kume M, Loucks J, Arjarapu R, Hodge R, Shiers SI, Sankaranarayanan I, Erdjument-Bromage H, Neubert TA, Campbell ZT, Paik R, Price TJ, Dalva MB. VLK drives extracellular phosphorylation of EphB2 to govern the EphB2-NMDAR interaction and injury-induced pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585314. [PMID: 38562765 PMCID: PMC10983893 DOI: 10.1101/2024.03.18.585314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Phosphorylation of hundreds of protein extracellular domains is mediated by two kinase families, yet the significance of these kinases is underexplored. Here, we find that the presynaptic release of the tyrosine directed-ectokinase, Vertebrate Lonesome Kinase (VLK/Pkdcc), is necessary and sufficient for the direct extracellular interaction between EphB2 and GluN1 at synapses, for phosphorylation of the ectodomain of EphB2, and for injury-induced pain. Pkdcc is an essential gene in the nervous system, and VLK is found in synaptic vesicles, and is released from neurons in a SNARE-dependent fashion. VLK is expressed by nociceptive sensory neurons where presynaptic sensory neuron-specific knockout renders mice impervious to post-surgical pain, without changing proprioception. VLK defines an extracellular mechanism that regulates protein-protein interaction and non-opioid-dependent pain in response to injury.
Collapse
Affiliation(s)
- Kolluru D Srikanth
- Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University; New Orleans, LA 70118, USA
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Hajira Elahi
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Praveen Chander
- Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University; New Orleans, LA 70118, USA
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Halley R Washburn
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
- Department of Molecular Biology, Princeton University; Princeton, NJ 08544, USA
| | - Shayne Hassler
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- College of Medicine, University of Houston; Houston, TX 77004, USA
| | - Juliet M Mwirigi
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Moeno Kume
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Jessica Loucks
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
| | - Rohita Arjarapu
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
| | - Rachel Hodge
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Stephanie I Shiers
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Hediye Erdjument-Bromage
- Department of Neuroscience and Physiology and Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Thomas A Neubert
- Department of Neuroscience and Physiology and Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Zachary T Campbell
- Department of Anesthesiology, University of Wisconsin-Madison; Madison, WI 53792, USA
| | - Raehum Paik
- Department of Anesthesiology, University of Wisconsin-Madison; Madison, WI 53792, USA
- Department of Genetics, University of Texas Health Science Center at San Antonio; San Antonio, TX 78229, USA
| | - Theodore J Price
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Matthew B Dalva
- Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University; New Orleans, LA 70118, USA
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
13
|
Huang H, Chen L, Yuan J, Zhang H, Yang J, Xu Z, Chen Y. Role and mechanism of EphB3 in epileptic seizures and epileptogenesis through Kalirin. Mol Cell Neurosci 2024; 128:103915. [PMID: 38143048 DOI: 10.1016/j.mcn.2023.103915] [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: 10/22/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023] Open
Abstract
BACKGROUND The EphB receptor tyrosine kinase family participates in intricate signaling pathways that orchestrate neural networks, guide neuronal axon development, and modulate synaptic plasticity through interactions with surface-bound ephrinB ligands. Additionally, Kalirin, a Rho guanine nucleotide exchange factor, is notably expressed in the postsynaptic membrane of excitatory neurons and plays a role in synaptic morphogenesis. This study postulates that Kalirin may act as a downstream effector of EphB3 in epilepsy. This investigation focuses on understanding the link between EphB3 and epilepsy. MATERIALS AND METHODS Chronic seizure models using LiCl-pilocarpine (LiCl/Pilo) and pentylenetetrazol were developed in rats. Neuronal excitability was gauged through whole-cell patch clamp recordings on rat hippocampal slices. Real-time PCR determined Kalirin's mRNA expression, and Western blotting was employed to quantify EphB3 and Kalirin protein levels. Moreover, dendritic spine density in epileptic rats was evaluated using Golgi staining. RESULTS Modulation of EphB3 functionality influenced acute seizure severity, latency duration, and frequency of spontaneous recurrent seizures. Golgi staining disclosed an EphB3-driven alteration in dendritic spine density within the hippocampus of epileptic rats, underscoring its pivotal role in the reconfiguration of hippocampal neural circuits. Furthermore, our data propose Kalirin as a prospective downstream mediator of the EphB3 receptor. CONCLUSIONS Our findings elucidate that EphB3 impacts the action potential dynamics in isolated rat hippocampal slices and alters dendritic spine density in the inner molecular layer of epileptic rat hippocampi, likely through Kalirin-mediated pathways. This hints at EphB3's significant role in shaping excitatory circuit loops and recurrent seizure activity via Kalirin.
Collapse
Affiliation(s)
- Hao Huang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, No.74, Linjiang Road, Chongqing 400010, China; Department of Neurology, The Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Zunyi 563003, Guizhou Province, China
| | - Ling Chen
- Department of Neurology, The Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Zunyi 563003, Guizhou Province, China
| | - Jinxian Yuan
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, No.74, Linjiang Road, Chongqing 400010, China
| | - Haiqing Zhang
- Department of Neurology, The Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Zunyi 563003, Guizhou Province, China
| | - Juan Yang
- Department of Neurology, The Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Zunyi 563003, Guizhou Province, China
| | - Zucai Xu
- Department of Neurology, The Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Zunyi 563003, Guizhou Province, China.
| | - Yangmei Chen
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, No.74, Linjiang Road, Chongqing 400010, China.
| |
Collapse
|
14
|
Urban MW, Charsar BA, Heinsinger NM, Markandaiah SS, Sprimont L, Zhou W, Brown EV, Henderson NT, Thomas SJ, Ghosh B, Cain RE, Trotti D, Pasinelli P, Wright MC, Dalva MB, Lepore AC. EphrinB2 knockdown in cervical spinal cord preserves diaphragm innervation in a mutant SOD1 mouse model of ALS. eLife 2024; 12:RP89298. [PMID: 38224498 PMCID: PMC10945582 DOI: 10.7554/elife.89298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by motor neuron loss. Importantly, non-neuronal cell types such as astrocytes also play significant roles in disease pathogenesis. However, mechanisms of astrocyte contribution to ALS remain incompletely understood. Astrocyte involvement suggests that transcellular signaling may play a role in disease. We examined contribution of transmembrane signaling molecule ephrinB2 to ALS pathogenesis, in particular its role in driving motor neuron damage by spinal cord astrocytes. In symptomatic SOD1G93A mice (a well-established ALS model), ephrinB2 expression was dramatically increased in ventral horn astrocytes. Reducing ephrinB2 in the cervical spinal cord ventral horn via viral-mediated shRNA delivery reduced motor neuron loss and preserved respiratory function by maintaining phrenic motor neuron innervation of diaphragm. EphrinB2 expression was also elevated in human ALS spinal cord. These findings implicate ephrinB2 upregulation as both a transcellular signaling mechanism in mutant SOD1-associated ALS and a promising therapeutic target.
Collapse
Affiliation(s)
- Mark W Urban
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Brittany A Charsar
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Nicolette M Heinsinger
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Shashirekha S Markandaiah
- Jefferson Weinberg ALS Center, Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Lindsay Sprimont
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Wei Zhou
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Eric V Brown
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Nathan T Henderson
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Samantha J Thomas
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Biswarup Ghosh
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Rachel E Cain
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Davide Trotti
- Jefferson Weinberg ALS Center, Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Piera Pasinelli
- Jefferson Weinberg ALS Center, Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Megan C Wright
- Department of Biology, Arcadia UniversityGlensideUnited States
| | - Matthew B Dalva
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
- Department of Cell and Molecular Biology, Tulane Brain Institute, Tulane UniversityNew OrleansUnited States
| | - Angelo C Lepore
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| |
Collapse
|
15
|
Qiu P, Li D, Xiao C, Xu F, Chen X, Chang Y, Liu L, Zhang L, Zhao Q, Chen Y. The Eph/ephrin system symphony of gut inflammation. Pharmacol Res 2023; 197:106976. [PMID: 38032293 DOI: 10.1016/j.phrs.2023.106976] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/15/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023]
Abstract
The extent of gut inflammation depends largely on the gut barrier's integrity and enteric neuroimmune interactions. However, the factors and molecular mechanisms that regulate inflammation-related changes in the enteric nervous system (ENS) remain largely unexplored. Eph/ephrin signaling is critical for inflammatory response, neuronal activation, and synaptic plasticity in the brain, but its presence and function in the ENS have been largely unknown to date. This review discusses the critical role of Eph/ephrin in regulating gut homeostasis, inflammation, neuroimmune interactions, and pain pathways. Targeting the Eph/ephrin system offers innovative treatments for gut inflammation disorders, offering hope for enhanced patient prognosis, pain management, and overall quality of life.
Collapse
Affiliation(s)
- Peishan Qiu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China
| | - Daojiang Li
- Department of General Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Cong Xiao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China
| | - Fei Xu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China
| | - Xiaoyu Chen
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China
| | - Ying Chang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China
| | - Lan Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China
| | - Lei Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China.
| | - Yuhua Chen
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China.
| |
Collapse
|
16
|
Urban MW, Charsar BA, Heinsinger NM, Markandaiah SS, Sprimont L, Zhou W, Brown EV, Henderson NT, Thomas SJ, Ghosh B, Cain RE, Trotti D, Pasinelli P, Wright MC, Dalva MB, Lepore AC. EphrinB2 knockdown in cervical spinal cord preserves diaphragm innervation in a mutant SOD1 mouse model of ALS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.10.538887. [PMID: 37215009 PMCID: PMC10197713 DOI: 10.1101/2023.05.10.538887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by motor neuron loss. Importantly, non-neuronal cell types such as astrocytes also play significant roles in disease pathogenesis. However, mechanisms of astrocyte contribution to ALS remain incompletely understood. Astrocyte involvement suggests that transcellular signaling may play a role in disease. We examined contribution of transmembrane signaling molecule ephrinB2 to ALS pathogenesis, in particular its role in driving motor neuron damage by spinal cord astrocytes. In symptomatic SOD1-G93A mice (a well-established ALS model), ephrinB2 expression was dramatically increased in ventral horn astrocytes. Reducing ephrinB2 in the cervical spinal cord ventral horn via viral-mediated shRNA delivery reduced motor neuron loss and preserved respiratory function by maintaining phrenic motor neuron innervation of diaphragm. EphrinB2 expression was also elevated in human ALS spinal cord. These findings implicate ephrinB2 upregulation as both a transcellular signaling mechanism in mutant SOD1-associated ALS and a promising therapeutic target.
Collapse
|
17
|
Varley JA, Strippel C, Handel A, Irani SR. Autoimmune encephalitis: recent clinical and biological advances. J Neurol 2023; 270:4118-4131. [PMID: 37115360 PMCID: PMC10345035 DOI: 10.1007/s00415-023-11685-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023]
Abstract
In 2015, we wrote a review in The Journal of Neurology summarizing the field of autoantibody-associated neurological diseases. Now, in 2023, we present an update of the subject which reflects the rapid expansion and refinement of associated clinical phenotypes, further autoantibody discoveries, and a more detailed understanding of immunological and neurobiological pathophysiological pathways which mediate these diseases. Increasing awareness around distinctive aspects of their clinical phenotypes has been a key driver in providing clinicians with a better understanding as to how these diseases are best recognized. In clinical practice, this recognition supports the administration of often effective immunotherapies, making these diseases 'not to miss' conditions. In parallel, there is a need to accurately assess patient responses to these drugs, another area of growing interest. Feeding into clinical care are the basic biological underpinnings of the diseases, which offer clear pathways to improved therapies toward enhanced patient outcomes. In this update, we aim to integrate the clinical diagnostic pathway with advances in patient management and biology to provide a cohesive view on how to care for these patients in 2023, and the future.
Collapse
Affiliation(s)
- James A Varley
- Department of Brain Sciences, Charing Cross Hospital, Imperial College London, Fulham Palace Road, London, W6 8RF, UK
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Level 3, West Wing, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Christine Strippel
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Level 3, West Wing, John Radcliffe Hospital, Oxford, OX3 9DS, UK
- Department of Neurology, John Radcliffe Hospital, Oxford University Hospitals, Oxford, OX3 9DU, UK
| | - Adam Handel
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Level 3, West Wing, John Radcliffe Hospital, Oxford, OX3 9DS, UK
- Department of Neurology, John Radcliffe Hospital, Oxford University Hospitals, Oxford, OX3 9DU, UK
| | - Sarosh R Irani
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Level 3, West Wing, John Radcliffe Hospital, Oxford, OX3 9DS, UK.
- Department of Neurology, John Radcliffe Hospital, Oxford University Hospitals, Oxford, OX3 9DU, UK.
| |
Collapse
|
18
|
González-González IM, Gray JA, Ferreira J, Conde-Dusman MJ, Bouchet D, Perez-Otaño I, Groc L. GluN3A subunit tunes NMDA receptor synaptic trafficking and content during postnatal brain development. Cell Rep 2023; 42:112477. [PMID: 37149869 PMCID: PMC11189104 DOI: 10.1016/j.celrep.2023.112477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/29/2023] [Accepted: 04/21/2023] [Indexed: 05/09/2023] Open
Abstract
Signaling via N-methyl-d-aspartate receptors (NMDARs) is critical for the maturation of glutamatergic synapses, partly through a developmental switch from immature synapses expressing primarily GluN2B- and GluN3A-containing subtypes to GluN2A-rich mature ones. This subunit switch is thought to underlie the synaptic stabilization of NMDARs necessary for neural network consolidation. However, the cellular mechanisms controlling the NMDAR exchange remain unclear. Using a combination of single-molecule and confocal imaging and biochemical and electrophysiological approaches, we show that surface GluN3A-NMDARs form a highly diffusive receptor pool that is loosely anchored to synapses. Remarkably, changes in GluN3A subunit expression selectively alter the surface diffusion and synaptic anchoring of GluN2A- but not GluN2B-NMDARs, possibly through altered interactions with cell surface receptors. The effects of GluN3A on NMDAR surface diffusion are restricted to an early time window of postnatal development in rodents, allowing GluN3A subunits to control the timing of NMDAR signaling maturation and neuronal network refinements.
Collapse
Affiliation(s)
- Inmaculada M González-González
- Cellular Neurobiology Laboratory, Centro de Investigación Médica Aplicada (CIMA) and Universidad de Navarra, Pamplona, Spain; Université de Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience (IINS), UMR 5297, 33000 Bordeaux, France
| | - John A Gray
- Department of Neurology, Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Joana Ferreira
- Université de Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience (IINS), UMR 5297, 33000 Bordeaux, France
| | - María Jose Conde-Dusman
- Cellular Neurobiology Laboratory, Centro de Investigación Médica Aplicada (CIMA) and Universidad de Navarra, Pamplona, Spain; Cellular and Systems Biology, Instituto de Neurociencias, CSIC-UMH, 03550 San Juan de Alicante, Spain
| | - Delphine Bouchet
- Université de Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience (IINS), UMR 5297, 33000 Bordeaux, France
| | - Isabel Perez-Otaño
- Cellular Neurobiology Laboratory, Centro de Investigación Médica Aplicada (CIMA) and Universidad de Navarra, Pamplona, Spain; Cellular and Systems Biology, Instituto de Neurociencias, CSIC-UMH, 03550 San Juan de Alicante, Spain.
| | - Laurent Groc
- Université de Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience (IINS), UMR 5297, 33000 Bordeaux, France.
| |
Collapse
|
19
|
Zhu D, Zhang J, Gao F, Hu M, Hashem J, Chen C. Augmentation of 2-arachidonoylglycerol signaling in astrocytes maintains synaptic functionality by regulation of miRNA-30b. Exp Neurol 2023; 361:114292. [PMID: 36481187 PMCID: PMC9892245 DOI: 10.1016/j.expneurol.2022.114292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/08/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
2-Arachidonoylglycerol (2-AG), the most abundant endocannabinoid, displays anti-inflammatory and neuroprotective properties. Inhibition of 2-AG degradation by inactivation of monoacylglycerol lipase (MAGL), a key enzyme degrading 2-AG in the brain, alleviates neuropathology and improves synaptic and cognitive functions in animal models of neurodegenerative diseases. In particular, global inactivation of MAGL by genetic deletion of mgll enhances hippocampal long-term potentiation (LTP) and hippocampus-dependent learning and memory. However, our understanding of the molecular mechanisms by which chronic inactivation of MAGL enhances synaptic activity is still limited. Here, we provide evidence that pharmacological inactivation of MAGL suppresses hippocampal expression of miR-30b, a small non-coding microRNA, and upregulates expression of its targets, including ephrin type-B receptor 2 (ephB2), sirtuin1 (sirt1), and glutamate ionotropic receptor AMPA type subunit 2 (GluA2). Importantly, suppression of miR-30b and increase of its targets by inactivation of MAGL result primarily from inhibition of 2-AG metabolism in astrocytes, rather than in neurons. Inactivation of MAGL in astrocytes prevents miR-30b overexpression-induced impairments in synaptic transmission and long-term potentiation (LTP) in the hippocampus. Suppression of miR-30b expression by inactivation of MAGL is apparently associated with augmentation of 2-AG signaling, as 2-AG induces a dose-dependent decrease in expression of miR-30b. 2-AG- or MAGL inactivation-suppressed expression of miR-30b is not mediated via CB1R, but by peroxisome proliferator-activated receptor γ (PPARγ). This is further supported by the results showing that MAGL inactivation-induced downregulation of miR-30b and upregulation of its targets are attenuated by antagonism of PPARγ, but mimicked by PPARγ agonists. In addition, we observed that 2-AG-induced reduction of miR-30b expression is mediated via NF-kB signaling. Our study provides evidence that 2-AG signaling in astrocytes plays an important role in maintaining the functional integrity of synapses in the hippocampus by regulation of miR-30b expression.
Collapse
Affiliation(s)
- Dexiao Zhu
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Jian Zhang
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Fei Gao
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Mei Hu
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Jack Hashem
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Chu Chen
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Biomedical Neuroscience, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
20
|
He LN, Chen S, Yang Q, Wu Z, Lao ZK, Tang CF, Song JJ, Liu XD, Lu J, Xu XH, Chen JJ, Xu TL, Sun S, Xu NJ. EphB2-dependent prefrontal cortex activation promotes long-range social approach and partner responsiveness. Proc Natl Acad Sci U S A 2023; 120:e2219952120. [PMID: 36802416 PMCID: PMC9992767 DOI: 10.1073/pnas.2219952120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/15/2023] [Indexed: 02/23/2023] Open
Abstract
Social behavior starts with dynamic approach prior to the final consummation. The flexible processes ensure mutual feedback across social brains to transmit signals. However, how the brain responds to the initial social stimuli precisely to elicit timed behaviors remains elusive. Here, by using real-time calcium recording, we identify the abnormalities of EphB2 mutant with autism-associated Q858X mutation in processing long-range approach and accurate activity of prefrontal cortex (dmPFC). The EphB2-dependent dmPFC activation precedes the behavioral onset and is actively associated with subsequent social action with the partner. Furthermore, we find that partner dmPFC activity is responsive coordinately to the approaching WT mouse rather than Q858X mutant mouse, and the social defects caused by the mutation are rescued by synchro-optogenetic activation in dmPFC of paired social partners. These results thus reveal that EphB2 sustains neuronal activation in the dmPFC that is essential for the proactive modulation of social approach to initial social interaction.
Collapse
Affiliation(s)
- Li-Na He
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai200062, China
- Songjiang Institute, Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai201699, China
| | - Si Chen
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai200062, China
- Songjiang Institute, Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai201699, China
| | - Qi Yang
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai200062, China
- Songjiang Institute, Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai201699, China
| | - Zheng Wu
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai200062, China
| | - Zheng-Kai Lao
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai200062, China
- Songjiang Institute, Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai201699, China
| | - Chang-Fei Tang
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai200062, China
- Songjiang Institute, Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai201699, China
| | - Jiao-Jiao Song
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai200062, China
| | - Xian-Dong Liu
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200025, China
| | - Jiangteng Lu
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai200062, China
- Songjiang Institute, Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai201699, China
| | - Xiao-Hong Xu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai200031, China
| | - Jin-Jin Chen
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai200062, China
| | - Tian-Le Xu
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai200062, China
- Songjiang Institute, Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai201699, China
| | - Suya Sun
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200025, China
| | - Nan-Jie Xu
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai200062, China
- Songjiang Institute, Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai201699, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai200025, China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai200025, China
| |
Collapse
|
21
|
Washburn HR, Chander P, Srikanth KD, Dalva MB. Transsynaptic Signaling of Ephs in Synaptic Development, Plasticity, and Disease. Neuroscience 2023; 508:137-152. [PMID: 36460219 DOI: 10.1016/j.neuroscience.2022.11.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022]
Abstract
Synapse formation between neurons is critical for proper circuit and brain function. Prior to activity-dependent refinement of connections between neurons, activity-independent cues regulate the contact and recognition of potential synaptic partners. Formation of a synapse results in molecular recognition events that initiate the process of synaptogenesis. Synaptogenesis requires contact between axon and dendrite, selection of correct and rejection of incorrect partners, and recruitment of appropriate pre- and postsynaptic proteins needed for the establishment of functional synaptic contact. Key regulators of these events are families of transsynaptic proteins, where one protein is found on the presynaptic neuron and the other is found on the postsynaptic neuron. Of these families, the EphBs and ephrin-Bs are required during each phase of synaptic development from target selection, recruitment of synaptic proteins, and formation of spines to regulation of synaptic plasticity at glutamatergic spine synapses in the mature brain. These roles also place EphBs and ephrin-Bs as important regulators of human neurological diseases. This review will focus on the role of EphBs and ephrin-Bs at synapses.
Collapse
Affiliation(s)
- Halley R Washburn
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA; Department of Neuroscience, Jefferson Synaptic Biology Center, Sidney Kimmel Medical College at Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA 19107, USA
| | - Praveen Chander
- Department of Neuroscience, Jefferson Synaptic Biology Center, Sidney Kimmel Medical College at Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA 19107, USA
| | - Kolluru D Srikanth
- Department of Neuroscience, Jefferson Synaptic Biology Center, Sidney Kimmel Medical College at Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA 19107, USA
| | - Matthew B Dalva
- Department of Neuroscience, Jefferson Synaptic Biology Center, Sidney Kimmel Medical College at Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA 19107, USA.
| |
Collapse
|
22
|
Dean CA, Metzbower SR, Dessain SK, Blanpied TA, Benavides DR. Regulation of NMDA Receptor Signaling at Single Synapses by Human Anti-NMDA Receptor Antibodies. Front Mol Neurosci 2022; 15:940005. [PMID: 35966009 PMCID: PMC9371948 DOI: 10.3389/fnmol.2022.940005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
The NMDA receptor (NMDAR) subunit GluN1 is critical for receptor function and plays a pivotal role in synaptic plasticity. Mounting evidence has shown that pathogenic autoantibody targeting of the GluN1 subunit of NMDARs, as in anti-NMDAR encephalitis, leads to altered NMDAR trafficking and synaptic localization. However, the underlying signaling pathways affected by antibodies targeting the NMDAR remain to be fully delineated. It remains unclear whether patient antibodies influence synaptic transmission via direct effects on NMDAR channel function. Here, we show using short-term incubation that GluN1 antibodies derived from patients with anti-NMDAR encephalitis label synapses in mature hippocampal primary neuron culture. Miniature spontaneous calcium transients (mSCaTs) mediated via NMDARs at synaptic spines are not altered in pathogenic GluN1 antibody exposed conditions. Unexpectedly, spine-based and cell-based analyses yielded distinct results. In addition, we show that calcium does not accumulate in neuronal spines following brief exposure to pathogenic GluN1 antibodies. Together, these findings show that pathogenic antibodies targeting NMDARs, under these specific conditions, do not alter synaptic calcium influx following neurotransmitter release. This represents a novel investigation of the molecular effects of anti-NMDAR antibodies associated with autoimmune encephalitis.
Collapse
Affiliation(s)
- Charles A. Dean
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Sarah R. Metzbower
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Scott K. Dessain
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | - Thomas A. Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - David R. Benavides
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States
- *Correspondence: David R. Benavides,
| |
Collapse
|
23
|
Shiosaka S. Kallikrein 8: A key sheddase to strengthen and stabilize neural plasticity. Neurosci Biobehav Rev 2022; 140:104774. [PMID: 35820483 DOI: 10.1016/j.neubiorev.2022.104774] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 11/19/2022]
Abstract
Neural networks are modified and reorganized throughout life, even in the matured brain. Synapses in the networks form, change, or disappear dynamically in the plasticity state. The pre- and postsynaptic signaling, transmission, and structural dynamics have been studied considerably well. However, not many studies have shed light on the events in the synaptic cleft and intercellular space. Neural activity-dependent protein shedding is a phenomenon in which (1) presynaptic excitation evokes secretion or activation of sheddases, (2) sheddases are involved not only in cleavage of membrane- or matrix-bound proteins but also in mechanical modulation of cell-to-cell connectivity, and (3) freed activity domains of protein factors play a role in receptor-mediated or non-mediated biological actions. Kallikrein 8/neuropsin (KLK8) is a kallikrein family serine protease rich in the mammalian limbic brain. Accumulated evidence has suggested that KLK8 is an important modulator of neural plasticity and consequently, cognition. Insufficiency, as well as excess of KLK8 may have detrimental effects on limbic functions.
Collapse
Affiliation(s)
- Sadao Shiosaka
- Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka Prefectural Hospital Organization, Miyanosaka 3-16-21, Hirakata-shi, Osaka 573-0022, Japan.
| |
Collapse
|
24
|
Zhang L, Wang R, Chen Y, Yang P, Bai T, Song J, Hou X. EphrinB2/ephB2 activation facilitates colonic synaptic potentiation and plasticity contributing to long-term visceral hypersensitivity in irritable bowel syndrome. Life Sci 2022; 295:120419. [PMID: 35183555 DOI: 10.1016/j.lfs.2022.120419] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 12/11/2022]
Abstract
AIMS Sustained visceral hypersensitivity is a hallmark of irritable bowel syndrome (IBS) could be partially explained by enteric neural remodeling. Particularly, synaptic plasticity in the enteric nervous system, a form of enteric "memory", has been speculated as a participant in the pain maintenance in IBS. This study aimed to elucidate the role of ephrinB2/ephB2 in enteric synaptic plasticity and visceral pain in IBS. MATERIALS AND METHODS EphrinB2/ephB2 expression and synaptic plasticity were assessed in colonic tissues from IBS patients, and rats induced by Trichinella spiralis infection and those treated with ephB2-Fc (an ephB2 receptor blocker) or ifenprodil (a selective NR2B antagonist). Furthermore, abdominal withdrawal reflex scores to colorectal distention and mesenteric afferent firing were assessed. EphrinB2-Fc(an ephB2 receptor activator) induced enteric synaptic plasticity was further evaluated in longitudinal muscle-myenteric plexus(LMMP) cultures and primary cultured myenteric neurons. KEY FINDINGS EphrinB2/ephB2 was specifically expressed in colonic nerves and upregulated in IBS patients and rats, which was correlated with pain severity. The functional synaptic plasticity, visceral sensitivity to colorectal distention and colonic mesenteric afferent activity to mechanical and chemical stimulus were enhanced in IBS rats, and were blocked by ephB2-Fc or ifenprodil treatment. EphrinB2-Fc promoted the phosphorylation of NR2B in IBS rats and LMMP cultures, and could mediate sustained neural activation via increased [Ca2+]i and raised expression of synaptic plasticity-related early immediate genes, including c-fos and arc. SIGNIFICANCE EphrinB2/ephB2 facilitated NR2B-mediated synaptic potentiation in the enteric nervous system that may be a novel explanation and potential therapeutic target for sustained pain hypersensitivity in IBS.
Collapse
Affiliation(s)
- Lei Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ruiyun Wang
- Department of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuhua Chen
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Pengcheng Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Bai
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jun Song
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
25
|
Abstract
The EPH receptor tyrosine kinases and their signaling partners, the EPHRINS, comprise a large class of cell signaling molecules that plays diverse roles in development. As cell membrane-anchored signaling molecules, they regulate cellular organization by modulating the strength of cellular contacts, usually by impacting the actin cytoskeleton or cell adhesion programs. Through these cellular functions, EPH/EPHRIN signaling often regulates tissue shape. Indeed, recent evidence indicates that this signaling family is ancient and associated with the origin of multicellularity. Though extensively studied, our understanding of the signaling mechanisms employed by this large family of signaling proteins remains patchwork, and a truly "canonical" EPH/EPHRIN signal transduction pathway is not known and may not exist. Instead, several foundational evolutionarily conserved mechanisms are overlaid by a myriad of tissue -specific functions, though common themes emerge from these as well. Here, I review recent advances and the related contexts that have provided new understanding of the conserved and varied molecular and cellular mechanisms employed by EPH/EPHRIN signaling during development.
Collapse
Affiliation(s)
- Jeffrey O Bush
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, United States; Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, United States; Institute for Human Genetics, University of California San Francisco, San Francisco, CA, United States; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
26
|
Hruska M, Cain RE, Dalva MB. Nanoscale rules governing the organization of glutamate receptors in spine synapses are subunit specific. Nat Commun 2022; 13:920. [PMID: 35177616 PMCID: PMC8854560 DOI: 10.1038/s41467-022-28504-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 01/20/2022] [Indexed: 12/21/2022] Open
Abstract
Heterotetrameric glutamate receptors are essential for the development, function, and plasticity of spine synapses but how they are organized to achieve this is not known. Here we show that the nanoscale organization of glutamate receptors containing specific subunits define distinct subsynaptic features. Glutamate receptors containing GluA2 or GluN1 subunits establish nanomodular elements precisely positioned relative to Synaptotagmin-1 positive presynaptic release sites that scale with spine size. Glutamate receptors containing GluA1 or GluN2B specify features that exhibit flexibility: GluA1-subunit containing AMPARs are found in larger spines, while GluN2B-subunit containing NMDARs are enriched in the smallest spines with neither following a strict modular organization. Given that the precise positioning of distinct classes of glutamate receptors is linked to diverse events including cell death and synaptic plasticity, this unexpectedly robust synaptic nanoarchitecture provides a resilient system, where nanopositioned glutamate receptor heterotetramers define specific subsynaptic regions of individual spine synapses. Glutamate receptors comprise two obligate subunits and two subunits that confer distinct properties and functions to the specific tetramers, which also localize to distinct synaptic spines. Here, the authors use STimulated Emission Depletion nanoscopy (STED) to provide detailed insights into the spatial organization of glutamate receptor types.
Collapse
Affiliation(s)
- Martin Hruska
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Road, Morgantown, WV, 26506, USA
| | - Rachel E Cain
- Department of Neuroscience and Jefferson Synaptic Biology Center, Sidney Kimmel Medical College at Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA, 19107, USA
| | - Matthew B Dalva
- Department of Neuroscience and Jefferson Synaptic Biology Center, Sidney Kimmel Medical College at Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA, 19107, USA.
| |
Collapse
|
27
|
Chen S, Xu D, Fan L, Fang Z, Wang X, Li M. Roles of N-Methyl-D-Aspartate Receptors (NMDARs) in Epilepsy. Front Mol Neurosci 2022; 14:797253. [PMID: 35069111 PMCID: PMC8780133 DOI: 10.3389/fnmol.2021.797253] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Epilepsy is one of the most common neurological disorders characterized by recurrent seizures. The mechanism of epilepsy remains unclear and previous studies suggest that N-methyl-D-aspartate receptors (NMDARs) play an important role in abnormal discharges, nerve conduction, neuron injury and inflammation, thereby they may participate in epileptogenesis. NMDARs belong to a family of ionotropic glutamate receptors that play essential roles in excitatory neurotransmission and synaptic plasticity in the mammalian CNS. Despite numerous studies focusing on the role of NMDAR in epilepsy, the relationship appeared to be elusive. In this article, we reviewed the regulation of NMDAR and possible mechanisms of NMDAR in epilepsy and in respect of onset, development, and treatment, trying to provide more evidence for future studies.
Collapse
Affiliation(s)
| | | | | | | | | | - Man Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
Bodin R, Paillé V, Oullier T, Durand T, Aubert P, Le Berre-Scoul C, Hulin P, Neunlist M, Cissé M. The ephrin receptor EphB2 regulates the connectivity and activity of enteric neurons. J Biol Chem 2021; 297:101300. [PMID: 34648765 PMCID: PMC8569587 DOI: 10.1016/j.jbc.2021.101300] [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: 03/26/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 11/29/2022] Open
Abstract
Highly organized circuits of enteric neurons are required for the regulation of gastrointestinal functions, such as peristaltism or migrating motor complex. However, the factors and molecular mechanisms that regulate the connectivity of enteric neurons and their assembly into functional neuronal networks are largely unknown. A better understanding of the mechanisms by which neurotrophic factors regulate this enteric neuron circuitry is paramount to understanding enteric nervous system (ENS) physiology. EphB2, a receptor tyrosine kinase, is essential for neuronal connectivity and plasticity in the brain, but so far its presence and function in the ENS remain largely unexplored. Here we report that EphB2 is expressed preferentially by enteric neurons relative to glial cells throughout the gut in rats. We show that in primary enteric neurons, activation of EphB2 by its natural ligand ephrinB2 engages ERK signaling pathways. Long-term activation with ephrinB2 decreases EphB2 expression and reduces molecular and functional connectivity in enteric neurons without affecting neuronal density, ganglionic fiber bundles, or overall neuronal morphology. This is highlighted by a loss of neuronal plasticity markers such as synapsin I, PSD95, and synaptophysin, and a decrease of spontaneous miniature synaptic currents. Together, these data identify a critical role for EphB2 in the ENS and reveal a unique EphB2-mediated molecular program of synapse regulation in enteric neurons.
Collapse
Affiliation(s)
- Raphael Bodin
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Université de Nantes, Nantes, France
| | - Vincent Paillé
- UMR 1280 Physiologie des Adaptations Nutritionnelles, INRA, Institut des Maladies de l'Appareil Digestif, Université de Nantes, Nantes, France
| | - Thibauld Oullier
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Université de Nantes, Nantes, France
| | - Tony Durand
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Université de Nantes, Nantes, France
| | - Philippe Aubert
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Université de Nantes, Nantes, France
| | - Catherine Le Berre-Scoul
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Université de Nantes, Nantes, France
| | | | - Michel Neunlist
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Université de Nantes, Nantes, France
| | - Moustapha Cissé
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Université de Nantes, Nantes, France.
| |
Collapse
|
29
|
The Association of Ovarian Teratoma and Anti-N-Methyl-D-Aspartate Receptor Encephalitis: An Updated Integrative Review. Int J Mol Sci 2021; 22:ijms222010911. [PMID: 34681570 PMCID: PMC8535897 DOI: 10.3390/ijms222010911] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/26/2021] [Accepted: 10/07/2021] [Indexed: 01/09/2023] Open
Abstract
Ovarian teratomas are by far the most common ovarian germ cell tumor. Most teratomas are benign unless a somatic transformation occurs. The designation of teratoma refers to a neoplasm that differentiates toward somatic-type cell populations. Recent research shows a striking association between ovarian teratomas and anti-N-methyl-D-aspartate receptor (anti-NMDAR) encephalitis, a rare and understudied paraneoplastic neurological syndrome (PNS). Among teratomas, mature teratomas are thought to have a greater relevance with those neurological impairments. PNS is described as a neurologic deficit triggered by an underlying remote tumor, whereas anti-NMDAR encephalitis is characterized by a complex neuropsychiatric syndrome and the presence of autoantibodies in cerebral spinal fluid against the GluN1 subunit of the NMDAR. This review aims to summarize recent reports on the association between anti-NMDAR encephalitis and ovarian teratoma. In particular, the molecular pathway of pathogenesis and the updated mechanism and disease models would be discussed. We hope to provide an in-depth review of this issue and, therefore, to better understand its epidemiology, diagnostic approach, and treatment strategies.
Collapse
|
30
|
Singal CMS, Jaiswal P, Mehta A, Saleem K, Seth P. Role of EphrinA3 in HIV-1 Neuropathogenesis. ASN Neuro 2021; 13:17590914211044359. [PMID: 34618621 PMCID: PMC8504696 DOI: 10.1177/17590914211044359] [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] [Indexed: 11/30/2022] Open
Abstract
Glial cells perform important supporting functions for neurons through a dynamic crosstalk. Neuron–glia communication is the major phenomenon to sustain homeostatic functioning of the brain. Several interactive pathways between neurons and astrocytes are critical for the optimal functioning of neurons, and one such pathway is the ephrinA3–ephA4 signaling. The role of this pathway is essential in maintaining the levels of extracellular glutamate by regulating the excitatory amino acid transporters, EAAT1 and EAAT2 on astrocytes. Human immunodeficiency virus-1 (HIV-1) and its proteins cause glutamate excitotoxicity due to excess glutamate levels at sites of high synaptic activity. This study unravels the effects of HIV-1 transactivator of transcription (Tat) from clade B on ephrinA3 and its role in regulating glutamate levels in astrocyte–neuron co-cultures of human origin. It was observed that the expression of ephrinA3 increases in the presence of HIV-1 Tat B, while the expression of EAAT1 and EAAT2 was attenuated. This led to reduced glutamate uptake and therefore high neuronal death due to glutamate excitotoxicity. Knockdown of ephrinA3 using small interfering RNA, in the presence of HIV-1 Tat B reversed the neurotoxic effects of HIV-1 Tat B via increased expression of glutamate transporters that reduced the levels of extracellular glutamate. The in vitro findings were validated in autopsy brain sections from acquired immunodeficiency syndrome patients and we found ephrinA3 to be upregulated in the case of HIV-1-infected patients. This study offers valuable insights into astrocyte-mediated neuronal damage in HIV-1 neuropathogenesis.
Collapse
Affiliation(s)
| | - Paritosh Jaiswal
- Cellular and Molecular Neuroscience, 29050National Brain Research Centre, Manesar, Gurgaon, India
| | - Anuradha Mehta
- Cellular and Molecular Neuroscience, 29050National Brain Research Centre, Manesar, Gurgaon, India
| | - Kanza Saleem
- Cellular and Molecular Neuroscience, 29050National Brain Research Centre, Manesar, Gurgaon, India
| | - Pankaj Seth
- Cellular and Molecular Neuroscience, 29050National Brain Research Centre, Manesar, Gurgaon, India
| |
Collapse
|
31
|
Motz CT, Kabat V, Saxena T, Bellamkonda RV, Zhu C. Neuromechanobiology: An Expanding Field Driven by the Force of Greater Focus. Adv Healthc Mater 2021; 10:e2100102. [PMID: 34342167 PMCID: PMC8497434 DOI: 10.1002/adhm.202100102] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 07/06/2021] [Indexed: 12/14/2022]
Abstract
The brain processes information by transmitting signals through highly connected and dynamic networks of neurons. Neurons use specific cellular structures, including axons, dendrites and synapses, and specific molecules, including cell adhesion molecules, ion channels and chemical receptors to form, maintain and communicate among cells in the networks. These cellular and molecular processes take place in environments rich of mechanical cues, thus offering ample opportunities for mechanical regulation of neural development and function. Recent studies have suggested the importance of mechanical cues and their potential regulatory roles in the development and maintenance of these neuronal structures. Also suggested are the importance of mechanical cues and their potential regulatory roles in the interaction and function of molecules mediating the interneuronal communications. In this review, the current understanding is integrated and promising future directions of neuromechanobiology are suggested at the cellular and molecular levels. Several neuronal processes where mechanics likely plays a role are examined and how forces affect ligand binding, conformational change, and signal induction of molecules key to these neuronal processes are indicated, especially at the synapse. The disease relevance of neuromechanobiology as well as therapies and engineering solutions to neurological disorders stemmed from this emergent field of study are also discussed.
Collapse
Affiliation(s)
- Cara T Motz
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| | - Victoria Kabat
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| | - Tarun Saxena
- Department of Biomedical Engineering, Duke University, Durham, NC, 27709, USA
| | - Ravi V Bellamkonda
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| |
Collapse
|
32
|
Pontifex MG, Martinsen A, Saleh RNM, Harden G, Tejera N, Müller M, Fox C, Vauzour D, Minihane AM. APOE4 genotype exacerbates the impact of menopause on cognition and synaptic plasticity in APOE-TR mice. FASEB J 2021; 35:e21583. [PMID: 33891334 DOI: 10.1096/fj.202002621rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 01/15/2023]
Abstract
The impact of sex and menopausal status in Alzheimer's disease remains understudied despite increasing evidence of greater female risk, particularly in APOE4 carriers. Utilizing female APOE-TR mice maintained on a high-fat diet background we induced ovarian failure through repeated VCD injections, to mimic human menopause. At 12 months of age, recognition memory and spatial memory were assessed using object recognition, Y-maze spontaneous alternation, and Barnes maze. A VCD*genotype interaction reduced the recognition memory (P < .05), with APOE4 VCD-treated animals unable to distinguish between novel and familiar objects. APOE4 mice displayed an additional 37% and 12% reduction in Barnes (P < .01) and Y-maze (P < .01) performance, indicative of genotype-specific spatial memory impairment. Molecular analysis indicated both VCD and genotype-related deficits in synaptic plasticity with BDNF, Akt, mTOR, and ERK signaling compromised. Subsequent reductions in the transcription factors Creb1 and Atf4 were also evident. Furthermore, the VCD*genotype interaction specifically diminished Ephb2 expression, while Fos, and Cnr1 expression reduced as a consequence of APOE4 genotype. Brain DHA levels were 13% lower in VCD-treated animals independent of genotype. Consistent with this, we detected alterations in the expression of the DHA transporters Acsl6 and Fatp4. Our results indicate that the combination of ovarian failure and APOE4 leads to an exacerbation of cognitive and neurological deficits.
Collapse
Affiliation(s)
| | | | | | - Glenn Harden
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Noemi Tejera
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Michael Müller
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Chris Fox
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - David Vauzour
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | |
Collapse
|
33
|
Petit-Pedrol M, Groc L. Regulation of membrane NMDA receptors by dynamics and protein interactions. J Cell Biol 2021; 220:211609. [PMID: 33337489 PMCID: PMC7754687 DOI: 10.1083/jcb.202006101] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/16/2022] Open
Abstract
Understanding neurotransmitter system crosstalk in the brain is a major challenge in neurobiology. Several intracellular and genomic cascades have been identified in this crosstalk. However, the discovery that neurotransmitter receptors are highly diffusive in the plasma membrane of neurons, where they form heterocomplexes with other proteins, has profoundly changed our view of neurotransmitter signaling. Here, we review new insights into neurotransmitter crosstalk at the plasma membrane. We focus on the membrane organization and interactome of the ionotropic glutamate N-methyl-D-aspartate receptor (NMDAR) that plays a central role in excitatory synaptic and network physiology and is involved in the etiology of several major neuropsychiatric disorders. The nanoscale organization and dynamics of NMDAR is a key regulatory process for glutamate synapse transmission, plasticity, and crosstalk with other neurotransmitter systems, such as the monoaminergic ones. The plasma membrane appears to be a prime regulatory compartment for spatial and temporal crosstalk between neurotransmitter systems in the healthy and diseased brain. Understanding the molecular mechanisms regulating membrane neurotransmitter receptor crosstalk will likely open research avenues for innovative therapeutical strategies.
Collapse
Affiliation(s)
- Mar Petit-Pedrol
- Université de Bordeaux, Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, Unité Mixte de Recherche 5297, Bordeaux, France
| | - Laurent Groc
- Université de Bordeaux, Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, Unité Mixte de Recherche 5297, Bordeaux, France
| |
Collapse
|
34
|
Ma X, Cheng O, Jiang Q, Yang J, Xiao H, Qiu H. Activation of ephrinb1/EPHB2/MAP-2/NMDAR Mediates Hippocampal Neurogenesis Promoted by Transcranial Direct Current Stimulation in Cerebral-Ischemic Mice. Neuromolecular Med 2021; 23:521-530. [PMID: 33782855 DOI: 10.1007/s12017-021-08654-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 03/16/2021] [Indexed: 11/28/2022]
Abstract
tDCS, a new, safe, non-invasive physical therapy method, is often used in motor dysfunction rehabilitation. However, the effects and underlying mechanisms of tDCS on hippocampal neurogenesis after cerebral ischemia (CI) are still unclear. This study aimed to investigate the promotive effect and mechanism of repetitive anodal-tDCS on hippocampal neurogenesis after CI in mice. The CI model in mice was established using bilateral common carotid artery occlusion (BCCAO). The pathological changes in the hippocampal CA1 region and cognitive function were assessed by hematoxylin and eosin staining and Morris water maze test, respectively. Hippocampal neurogenesis was observed by immunofluorescence staining. The levels of expression of ephrinb1, EPHB2, MAP-2, and NMDAR in the hippocampi were analyzed by qRT-PCR and Western blotting. Compared with the sham mice, the model mice showed significant neuronal damage in the hippocampal CA1 region (P < 0.01), cognitive dysfunction (P < 0.01), and endogenous hippocampal neurogenesis (P < 0.01). These results suggested that the CI model was successfully established, and that CI could promote endogenous hippocampal neurogenesis, but this hippocampal neurogenesis was unable to recover cognitive dysfunction. Compared with the model mice, the tDCS mice had ameliorated pathological damage in the CA1 region (P < 0.01), improved cognitive function (P < 0.01), increased hippocampal neurogenesis (P < 0.01), and increased mRNA and protein expression of ephrinb1, EPHB2, MAP-2, and NMDAR (P < 0.05). Repetitive anodal-tDCS can promote hippocampal neurogenesis and improve cognitive function in CI mice. The effect may be related to the activation of the ephrinb1/EPHB2/MAP-2/NMDAR signaling pathway.
Collapse
Affiliation(s)
- Xiaojiao Ma
- Department of Pharmacology, College of Pharmacy, Chongqing Key Lab of Biochemistry and Molecular Pharmacology, Chongqing Key Lab Drug Metabolism, Chongqing Medical University, No. 1 Yixueyuan Road, Chongqing, 400016, China
| | - Oumei Cheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qingsong Jiang
- Department of Pharmacology, College of Pharmacy, Chongqing Key Lab of Biochemistry and Molecular Pharmacology, Chongqing Key Lab Drug Metabolism, Chongqing Medical University, No. 1 Yixueyuan Road, Chongqing, 400016, China
| | - Junxia Yang
- Department of Pharmacology, College of Pharmacy, Chongqing Key Lab of Biochemistry and Molecular Pharmacology, Chongqing Key Lab Drug Metabolism, Chongqing Medical University, No. 1 Yixueyuan Road, Chongqing, 400016, China
| | - Huan Xiao
- Department of Pharmacology, College of Pharmacy, Chongqing Key Lab of Biochemistry and Molecular Pharmacology, Chongqing Key Lab Drug Metabolism, Chongqing Medical University, No. 1 Yixueyuan Road, Chongqing, 400016, China
| | - Hongmei Qiu
- Department of Pharmacology, College of Pharmacy, Chongqing Key Lab of Biochemistry and Molecular Pharmacology, Chongqing Key Lab Drug Metabolism, Chongqing Medical University, No. 1 Yixueyuan Road, Chongqing, 400016, China.
| |
Collapse
|
35
|
Identification of tetracycline combinations as EphB1 tyrosine kinase inhibitors for treatment of neuropathic pain. Proc Natl Acad Sci U S A 2021; 118:2016265118. [PMID: 33627480 DOI: 10.1073/pnas.2016265118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Previous studies have demonstrated that the synaptic EphB1 receptor tyrosine kinase is a major mediator of neuropathic pain, suggesting that targeting the activity of this receptor might be a viable therapeutic option. Therefore, we set out to determine if any FDA-approved drugs can act as inhibitors of the EphB1 intracellular catalytic domain. An in silico screen was first used to identify a number of tetracycline antibiotics which demonstrated potential docking to the ATP-binding catalytic domain of EphB1. Kinase assays showed that demeclocycline, chlortetracycline, and minocycline inhibit EphB1 kinase activity at low micromolar concentrations. In addition, we cocrystallized chlortetracycline and EphB1 receptor, which confirmed its binding to the ATP-binding domain. Finally, in vivo administration of the three-tetracycline combination inhibited the phosphorylation of EphB1 in the brain, spinal cord, and dorsal root ganglion (DRG) and effectively blocked neuropathic pain in mice. These results indicate that demeclocycline, chlortetracycline, and minocycline can be repurposed for treatment of neuropathic pain and potentially for other indications that would benefit from inhibition of EphB1 receptor kinase activity.
Collapse
|
36
|
Zhou J, Lin Y, Huynh T, Noguchi H, Bush JO, Pleasure SJ. NMDA receptors control development of somatosensory callosal axonal projections. eLife 2021; 10:59612. [PMID: 33661095 PMCID: PMC7959694 DOI: 10.7554/elife.59612] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 02/22/2021] [Indexed: 11/25/2022] Open
Abstract
Callosal projections from primary somatosensory cortex (S1) are key for processing somatosensory inputs and integrating sensory-motor information. How the callosal innervation pattern in S1 is formed during early postnatal development is not clear. We found that the normal termination pattern of these callosal projections is disrupted in cortex specific NMDAR mutants. Rather than projecting selectively to the primary/secondary somatosensory cortex (S1/S2) border, axons were uniformly distributed throughout S1. In addition, the density of this projection increased over postnatal life until the mice died by P30. By combining genetic and antibody-mediated loss of function, we demonstrated that it is GluN2B-containing NMDA receptors in target S1 that mediate this guidance phenotype, thus playing a central role in interhemispheric connectivity. Furthermore, we found that this function of NMDA receptors in callosal circuit formation is independent of ion channel function and works with the EPHRIN-B/EPHB system. Thus, NMDAR in target S1 cortex regulates the formation callosal circuits perhaps by modulating EPH-dependent repulsion.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Yong Lin
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Department of Neurological Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Trung Huynh
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Hirofumi Noguchi
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Jeffrey O Bush
- Department of Cell and Tissue Biology, Program in Craniofacial Biology and Institute for Human Genetics, University of California, San Francisco, San Francisco, United States.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, United States
| | - Samuel J Pleasure
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States.,Programs in Neuroscience and Developmental Stem Cell Biology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Kavli Institute for Fundamental Neuroscience, San Francisco, United States
| |
Collapse
|
37
|
Nourbakhsh K, Yadav S. Kinase Signaling in Dendritic Development and Disease. Front Cell Neurosci 2021; 15:624648. [PMID: 33642997 PMCID: PMC7902504 DOI: 10.3389/fncel.2021.624648] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/06/2021] [Indexed: 01/19/2023] Open
Abstract
Dendrites undergo extensive growth and remodeling during their lifetime. Specification of neurites into dendrites is followed by their arborization, maturation, and functional integration into synaptic networks. Each of these distinct developmental processes is spatially and temporally controlled in an exquisite fashion. Protein kinases through their highly specific substrate phosphorylation regulate dendritic growth and plasticity. Perturbation of kinase function results in aberrant dendritic growth and synaptic function. Not surprisingly, kinase dysfunction is strongly associated with neurodevelopmental and psychiatric disorders. Herein, we review, (a) key kinase pathways that regulate dendrite structure, function and plasticity, (b) how aberrant kinase signaling contributes to dendritic dysfunction in neurological disorders and (c) emergent technologies that can be applied to dissect the role of protein kinases in dendritic structure and function.
Collapse
Affiliation(s)
| | - Smita Yadav
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| |
Collapse
|
38
|
Engin AB, Engin A. Alzheimer's Disease and Protein Kinases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1275:285-321. [PMID: 33539020 DOI: 10.1007/978-3-030-49844-3_11] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and accounts for more than 60-80% of all cases of dementia. Loss of pyramidal neurons, extracellular amyloid beta (Abeta) accumulated senile plaques, and neurofibrillary tangles that contain hyperphosphorylated tau constitute the main pathological alterations in AD.Synaptic dysfunction and extrasynaptic N-methyl-D-aspartate receptor (NMDAR) hyperactivation contributes to excitotoxicity in patients with AD. Amyloid precursor protein (APP) and Abeta promoted neurodegeneration develop through the activation of protein kinase signaling cascade in AD. Furthermore, ultimate neuronal death in AD is under control of protein kinases-related signaling pathways. In this chapter, critical check-points within the cross-talk between neuron and protein kinases have been defined regarding the initiation and progression of AD. In this context, amyloid cascade hypothesis, neuroinflammation, oxidative stress, granulovacuolar degeneration, loss of Wnt signaling, Abeta-related synaptic alterations, prolonged calcium ions overload and NMDAR-related synaptotoxicity, damage signals hypothesis and type-3 diabetes are discussed briefly.In addition to clinical perspective of AD pathology, recommendations that might be effective in the treatment of AD patients have been reviewed.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey.
| | - Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
39
|
Sell J, Haselmann H, Hallermann S, Hust M, Geis C. Autoimmune encephalitis: novel therapeutic targets at the preclinical level. Expert Opin Ther Targets 2020; 25:37-47. [PMID: 33233983 DOI: 10.1080/14728222.2021.1856370] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Antibody-mediated encephalitides (AE) with pathogenic autoantibodies (aAB) against neuronal surface antigens are a growing group of diseases characterized by antineuronal autoimmunity in the brain. AE patients typically present with rapidly progressive encephalitis and characteristic disease symptoms dependent on the target antigen. Current treatment consists of an escalating immunotherapy strategy including plasma exchange, steroid application, and B cell depletion. AREAS COVERED For this review, we searched Medline database and google scholar with inclusive dates from 2000. We summarize current treatment strategies and present novel therapeutic approaches of target-specific interventions at the pre-clinical level as well as immunotherapy directed at antibody-induced pathology. Treatment options include modulation of target proteins, intervention with downstream pathways, antibody modification, and depletion of antibody-secreting cells. EXPERT OPINION Although current therapies in AE are effective in many patients, recovery is often prolonged and relapses as well as persistent deficits can occur. Specific immunotherapy together with supportive target-specific therapy may provide faster control of severe symptoms, shorten the disease course, and lead to long-lasting disease stability. Among the various novel therapeutic approaches, modulation of targeted receptors by small molecules crossing the blood-brain barrier as well as prevention of aAB binding is of particular interest.
Collapse
Affiliation(s)
- Josefine Sell
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital , Jena, Germany
| | - Holger Haselmann
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital , Jena, Germany
| | - Stefan Hallermann
- Carl-Ludwig-Institute for Physiology, Medical Faculty, Leipzig University , Leipzig, Germany
| | - Michael Hust
- Department Biotechnology, Technische Universität Braunschweig, Institute for Biochemistry, Biotechnology and Bioinformatics , Braunschweig, Germany
| | - Christian Geis
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital , Jena, Germany
| |
Collapse
|
40
|
Hong J, Won M, Ro H. The Molecular and Pathophysiological Functions of Members of the LNX/PDZRN E3 Ubiquitin Ligase Family. Molecules 2020; 25:E5938. [PMID: 33333989 PMCID: PMC7765395 DOI: 10.3390/molecules25245938] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/10/2020] [Accepted: 12/10/2020] [Indexed: 12/27/2022] Open
Abstract
The ligand of Numb protein-X (LNX) family, also known as the PDZRN family, is composed of four discrete RING-type E3 ubiquitin ligases (LNX1, LNX2, LNX3, and LNX4), and LNX5 which may not act as an E3 ubiquitin ligase owing to the lack of the RING domain. As the name implies, LNX1 and LNX2 were initially studied for exerting E3 ubiquitin ligase activity on their substrate Numb protein, whose stability was negatively regulated by LNX1 and LNX2 via the ubiquitin-proteasome pathway. LNX proteins may have versatile molecular, cellular, and developmental functions, considering the fact that besides these proteins, none of the E3 ubiquitin ligases have multiple PDZ (PSD95, DLGA, ZO-1) domains, which are regarded as important protein-interacting modules. Thus far, various proteins have been isolated as LNX-interacting proteins. Evidence from studies performed over the last two decades have suggested that members of the LNX family play various pathophysiological roles primarily by modulating the function of substrate proteins involved in several different intracellular or intercellular signaling cascades. As the binding partners of RING-type E3s, a large number of substrates of LNX proteins undergo degradation through ubiquitin-proteasome system (UPS) dependent or lysosomal pathways, potentially altering key signaling pathways. In this review, we highlight recent and relevant findings on the molecular and cellular functions of the members of the LNX family and discuss the role of the erroneous regulation of these proteins in disease progression.
Collapse
Affiliation(s)
- Jeongkwan Hong
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 305-764, Korea;
| | - Minho Won
- Biotechnology Process Engineering Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 30 Yeongudanji-ro, Cheongwon-gu, Cheongju 28116, Korea
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 305-764, Korea;
| |
Collapse
|
41
|
Gorski K, Spoljaric A, Nyman TA, Kaila K, Battersby BJ, Lehesjoki AE. Quantitative Changes in the Mitochondrial Proteome of Cerebellar Synaptosomes From Preclinical Cystatin B-Deficient Mice. Front Mol Neurosci 2020; 13:570640. [PMID: 33281550 PMCID: PMC7691638 DOI: 10.3389/fnmol.2020.570640] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 10/21/2020] [Indexed: 12/04/2022] Open
Abstract
Progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1) is a neurodegenerative disorder caused by loss-of-function mutations in the cystatin B (CSTB) gene. Progression of the clinical symptoms in EPM1 patients, including stimulus-sensitive myoclonus, tonic-clonic seizures, and ataxia, are well described. However, the cellular dysfunction during the presymptomatic phase that precedes the disease onset is not understood. CSTB deficiency leads to alterations in GABAergic signaling, and causes early neuroinflammation followed by progressive neurodegeneration in brains of a mouse model, manifesting as progressive myoclonus and ataxia. Here, we report the first proteome atlas from cerebellar synaptosomes of presymptomatic Cstb-deficient mice, and propose that early mitochondrial dysfunction is important to the pathogenesis of altered synaptic function in EPM1. A decreased sodium- and chloride dependent GABA transporter 1 (GAT-1) abundance was noted in synaptosomes with CSTB deficiency, but no functional difference was seen between the two genotypes in electrophysiological experiments with pharmacological block of GAT-1. Collectively, our findings provide novel insights into the early onset and pathogenesis of CSTB deficiency, and reveal greater complexity to the molecular pathogenesis of EPM1.
Collapse
Affiliation(s)
- Katarin Gorski
- Folkhälsan Research Center, Helsinki, Finland.,Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Albert Spoljaric
- Molecular and Integrative Biosciences, and Neuroscience Center (HiLIFE), Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Tuula A Nyman
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Kai Kaila
- Molecular and Integrative Biosciences, and Neuroscience Center (HiLIFE), Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | | | - Anna-Elina Lehesjoki
- Folkhälsan Research Center, Helsinki, Finland.,Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| |
Collapse
|
42
|
Zeneyedpour L, Sten-van `t Hoff J, Luider T. Using phosphoproteomics and next generation sequencing to discover novel therapeutic targets in patient antibodies. Expert Rev Proteomics 2020; 17:675-684. [DOI: 10.1080/14789450.2020.1845147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Lona Zeneyedpour
- Department of Neurology, Erasmus MC, Laboratory of Neuro-Oncology/Clinical & Cancer Proteomics, Rotterdam, The Netherlands
| | - Jenny Sten-van `t Hoff
- Department of Neurology, Erasmus MC, Laboratory of Neuro-Oncology/Clinical & Cancer Proteomics, Rotterdam, The Netherlands
| | - Theo Luider
- Department of Neurology, Erasmus MC, Laboratory of Neuro-Oncology/Clinical & Cancer Proteomics, Rotterdam, The Netherlands
| |
Collapse
|
43
|
Wu XR, Zhang Y, Liu XD, Han WB, Xu NJ, Sun S. EphB2 mediates social isolation-induced memory forgetting. Transl Psychiatry 2020; 10:389. [PMID: 33168800 PMCID: PMC7653962 DOI: 10.1038/s41398-020-01051-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 09/16/2020] [Accepted: 10/06/2020] [Indexed: 01/05/2023] Open
Abstract
Social isolation in adolescence leads to lasting deficits, including emotional and cognitive dysregulation. It remains unclear, however, how social isolation affects certain processes of memory and what molecular mechanisms are involved. In this study, we found that social isolation during the post-weaning period resulted in forgetting of the long-term fear memory, which was attributable to the downregulation of synaptic function in the hippocampal CA1 region mediated by EphB2, a receptor tyrosine kinase which involves in the glutamate receptor multiprotein complex. Viral-mediated EphB2 knockdown in CA1 mimicked the memory defects in group-housed mice, whereas restoration of EphB2 by either viral overexpression or resocialization reversed the memory decline in isolated mice. Taken together, our finding indicates that social isolation gives rise to memory forgetting by disrupting EphB2-mediated synaptic plasticity, which may provide a potential target for preventing memory loss caused by social isolation or loneliness.
Collapse
Affiliation(s)
- Xin-Rong Wu
- grid.16821.3c0000 0004 0368 8293Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Yu Zhang
- grid.16821.3c0000 0004 0368 8293Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Xian-Dong Liu
- grid.16821.3c0000 0004 0368 8293Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China ,grid.16821.3c0000 0004 0368 8293Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Wu-Bo Han
- grid.16821.3c0000 0004 0368 8293Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Nan-Jie Xu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China. .,Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China. .,Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Suya Sun
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| |
Collapse
|
44
|
Astrocytic Ephrin-B1 Controls Excitatory-Inhibitory Balance in Developing Hippocampus. J Neurosci 2020; 40:6854-6871. [PMID: 32801156 DOI: 10.1523/jneurosci.0413-20.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/08/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
Astrocytes are implicated in synapse formation and elimination, which are associated with developmental refinements of neuronal circuits. Astrocyte dysfunctions are also linked to synapse pathologies associated with neurodevelopmental disorders and neurodegenerative diseases. Although several astrocyte-derived secreted factors are implicated in synaptogenesis, the role of contact-mediated glial-neuronal interactions in synapse formation and elimination during development is still unknown. In this study, we examined whether the loss or overexpression of the membrane-bound ephrin-B1 in astrocytes during postnatal day (P) 14-28 period would affect synapse formation and maturation in the developing hippocampus. We found enhanced excitation of CA1 pyramidal neurons in astrocyte-specific ephrin-B1 KO male mice, which coincided with a greater vGlut1/PSD95 colocalization, higher dendritic spine density, and enhanced evoked AMPAR and NMDAR EPSCs. In contrast, EPSCs were reduced in CA1 neurons neighboring ephrin-B1-overexpressing astrocytes. Overexpression of ephrin-B1 in astrocytes during P14-28 developmental period also facilitated evoked IPSCs in CA1 neurons, while evoked IPSCs and miniature IPSC amplitude were reduced following astrocytic ephrin-B1 loss. Lower numbers of parvalbumin-expressing cells and a reduction in the inhibitory VGAT/gephyrin-positive synaptic sites on CA1 neurons in the stratum pyramidale and stratum oriens layers of KO hippocampus may contribute to reduced inhibition and higher excitation. Finally, dysregulation of excitatory/inhibitory balance in KO male mice is most likely responsible for impaired sociability observed in these mice. The ability of astrocytic ephrin-B1 to influence both excitatory and inhibitory synapses during development can potentially contribute to developmental refinement of neuronal circuits.SIGNIFICANCE STATEMENT This report establishes a link between astrocytes and the development of excitatory and inhibitory balance in the mouse hippocampus during early postnatal development. We provide new evidence that astrocytic ephrin-B1 differentially regulates development of excitatory and inhibitory circuits in the hippocampus during early postnatal development using a multidisciplinary approach. The ability of astrocytic ephrin-B1 to influence both excitatory and inhibitory synapses during development can potentially contribute to developmental refinement of neuronal circuits and associated behaviors. Given widespread and growing interest in the astrocyte-mediated mechanisms that regulate synapse development, and the role of EphB receptors in neurodevelopmental disorders, these findings establish a foundation for future studies of astrocytes in clinically relevant conditions.
Collapse
|
45
|
Young D. The NMDA Receptor Antibody Paradox: A Possible Approach to Developing Immunotherapies Targeting the NMDA Receptor. Front Neurol 2020; 11:635. [PMID: 32719654 PMCID: PMC7347966 DOI: 10.3389/fneur.2020.00635] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 05/28/2020] [Indexed: 12/29/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDAR) play a key role in brain development and function, including contributing to the pathogenesis of many neurological disorders. Immunization against the GluN1 subunit of the NMDAR and the production of GluN1 antibodies is associated with neuroprotective and seizure-protective effects in rodent models of stroke and epilepsy, respectively. Whilst these data suggest the potential for the development of GluN1 antibody therapy, paradoxically GluN1 autoantibodies in humans are associated with the pathogenesis of the autoimmune disease anti-NMDA receptor encephalitis. This review discusses possible reasons for the differential effects of GluN1 antibodies on NMDAR physiology that could contribute to these phenotypes.
Collapse
Affiliation(s)
- Deborah Young
- Molecular Neurotherapeutics Laboratory, Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
46
|
Gao R, Chen C, Zhao Q, Li M, Wang Q, Zhou L, Chen E, Chen H, Zhang Y, Cai X, Liu C, Cheng X, Zhang S, Mao X, Qiu Y, Gan L, Yu H, Liu J, Zhu T. Identification of the Potential Key Circular RNAs in Elderly Patients With Postoperative Cognitive Dysfunction. Front Aging Neurosci 2020; 12:165. [PMID: 32655392 PMCID: PMC7324535 DOI: 10.3389/fnagi.2020.00165] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 05/13/2020] [Indexed: 02/05/2023] Open
Abstract
Background Postoperative cognitive dysfunction (POCD) is one of the severe complications after surgery, inducing low life quality and high mortality, especially in elderly patients. However, the underlying molecular mechanism of POCD remains largely unknown, and the ideal biomarker for clinical diagnosis and prognosis is lacking. Circular RNAs (circRNAs), as a unique class of non-coding RNAs, were characterized by its stability and conservativeness, serving as novel biomarkers in various diseases. Nevertheless, the role of circRNAs in the occurrence of POCD remains elusive. Methods To investigate the differentially expressed circRNAs in the serum of POCD patients and its potential role in the development of POCD, we performed a circRNA microarray to screen the differentially expressed circRNAs in the serum samples from three patients of the POCD group and three paired patients of the non-POCD group. Subsequently, quantitative real-time polymerase chain reaction analysis (qRT-PCR) was utilized to verify the microarray data with the serum samples from 10 paired patients. Cytoscape software was used to construct the circRNA–miRNA–mRNA network for circRNAs with different expression levels as well as the target genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses showed the biological functions of the differentially expressed circRNAs target genes. Results In total, we have analyzed 10,198 circRNAs through the microarray. Compared with the non-POCD patient group, there were 210 differentially expressed circRNAs with 133 upregulated and 77 downregulated in the POCD group (≥2-fold differential expression, P ≤ 0.05). The qRT-PCR confirmed 10 circRNAs with different expressed levels, and the results were consistent with the microarray findings. Among them, hsa_circRNA_001145, hsa_circRNA_101138, and hsa_circRNA_061570 had the highest magnitude of change. The GO analysis showed that the differentially expressed circRNAs were associated with the regulation of the developmental process, cell-to-cell adhesion, and nervous system development. The KEGG analysis showed that the target genes of circRNAs were enriched in the MAPK signaling pathway and RAS signaling pathway. According to the targetscan7.1 and mirdbV5 databases, the circRNA–miRNA–mRNA network was constructed, and these results provided a vital landscape of circRNA expression profile in POCD. Conclusions Our study provides an essential perspective for the differential expression of circRNAs in POCD patients. Further studies need to be performed to explore their potential therapeutic roles in the development of POCD.
Collapse
Affiliation(s)
- Rui Gao
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chan Chen
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qi Zhao
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Li
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qiao Wang
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Zhou
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Erya Chen
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hai Chen
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Zhang
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xingwei Cai
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Changliang Liu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xu Cheng
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shu Zhang
- Department of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaobo Mao
- Department of Neurology, Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Yanhua Qiu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Gan
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hai Yu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Liu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Zhu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
47
|
Keable R, Leshchyns'ka I, Sytnyk V. Trafficking and Activity of Glutamate and GABA Receptors: Regulation by Cell Adhesion Molecules. Neuroscientist 2020; 26:415-437. [PMID: 32449484 DOI: 10.1177/1073858420921117] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The efficient targeting of ionotropic receptors to postsynaptic sites is essential for the function of chemical excitatory and inhibitory synapses, constituting the majority of synapses in the brain. A growing body of evidence indicates that cell adhesion molecules (CAMs), which accumulate at synapses at the earliest stages of synaptogenesis, are critical for this process. A diverse variety of CAMs assemble into complexes with glutamate and GABA receptors and regulate the targeting of these receptors to the cell surface and synapses. Presynaptically localized CAMs provide an additional level of regulation, sending a trans-synaptic signal that can regulate synaptic strength at the level of receptor trafficking. Apart from controlling the numbers of receptors present at postsynaptic sites, CAMs can also influence synaptic strength by modulating the conductivity of single receptor channels. CAMs thus act to maintain basal synaptic transmission and are essential for many forms of activity dependent synaptic plasticity. These activities of CAMs may underlie the association between CAM gene mutations and synaptic pathology and represent fundamental mechanisms by which synaptic strength is dynamically tuned at both excitatory and inhibitory synapses.
Collapse
Affiliation(s)
- Ryan Keable
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Iryna Leshchyns'ka
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
48
|
Perdigão C, Barata MA, Araújo MN, Mirfakhar FS, Castanheira J, Guimas Almeida C. Intracellular Trafficking Mechanisms of Synaptic Dysfunction in Alzheimer's Disease. Front Cell Neurosci 2020; 14:72. [PMID: 32362813 PMCID: PMC7180223 DOI: 10.3389/fncel.2020.00072] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 03/12/2020] [Indexed: 12/15/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disease characterized by progressive memory loss. Although AD neuropathological hallmarks are extracellular amyloid plaques and intracellular tau tangles, the best correlate of disease progression is synapse loss. What causes synapse loss has been the focus of several researchers in the AD field. Synapses become dysfunctional before plaques and tangles form. Studies based on early-onset familial AD (eFAD) models have supported that synaptic transmission is depressed by β-amyloid (Aβ) triggered mechanisms. Since eFAD is rare, affecting only 1% of patients, research has shifted to the study of the most common late-onset AD (LOAD). Intracellular trafficking has emerged as one of the pathways of LOAD genes. Few studies have assessed the impact of trafficking LOAD genes on synapse dysfunction. Since endocytic traffic is essential for synaptic function, we reviewed Aβ-dependent and independent mechanisms of the earliest synaptic dysfunction in AD. We have focused on the role of intraneuronal and secreted Aβ oligomers, highlighting the dysfunction of endocytic trafficking as an Aβ-dependent mechanism of synapse dysfunction in AD. Here, we reviewed the LOAD trafficking genes APOE4, ABCA7, BIN1, CD2AP, PICALM, EPH1A, and SORL1, for which there is a synaptic link. We conclude that in eFAD and LOAD, the earliest synaptic dysfunctions are characterized by disruptions of the presynaptic vesicle exo- and endocytosis and of postsynaptic glutamate receptor endocytosis. While in eFAD synapse dysfunction seems to be triggered by Aβ, in LOAD, there might be a direct synaptic disruption by LOAD trafficking genes. To identify promising therapeutic targets and biomarkers of the earliest synaptic dysfunction in AD, it will be necessary to join efforts in further dissecting the mechanisms used by Aβ and by LOAD genes to disrupt synapses.
Collapse
Affiliation(s)
- Catarina Perdigão
- Laboratory Neuronal Trafficking in Aging, CEDOC Chronic Diseases Research Center, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Mariana A Barata
- Laboratory Neuronal Trafficking in Aging, CEDOC Chronic Diseases Research Center, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Margarida N Araújo
- Laboratory Neuronal Trafficking in Aging, CEDOC Chronic Diseases Research Center, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Farzaneh S Mirfakhar
- Laboratory Neuronal Trafficking in Aging, CEDOC Chronic Diseases Research Center, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Jorge Castanheira
- Laboratory Neuronal Trafficking in Aging, CEDOC Chronic Diseases Research Center, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Cláudia Guimas Almeida
- Laboratory Neuronal Trafficking in Aging, CEDOC Chronic Diseases Research Center, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
49
|
Theofanous SA, Florens MV, Appeltans I, Denadai Souza A, Wood JN, Wouters MM, Boeckxstaens GE. Ephrin-B2 signaling in the spinal cord as a player in post-inflammatory and stress-induced visceral hypersensitivity. Neurogastroenterol Motil 2020; 32:e13782. [PMID: 32004400 DOI: 10.1111/nmo.13782] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 11/07/2019] [Accepted: 11/27/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Ephrin-B2/EphB receptor signaling contributes to persistent pain states such as postinflammatory and neuropathic pain. Visceral hypersensitivity (VHS) is a major mechanism underlying abdominal pain in patients with irritable bowel syndrome (IBS) and inflammatory bowel diseases (IBD) in remission, but the underlying pathophysiology remains unclear. Here, we evaluated the spinal ephrin-B2/EphB pathway in VHS in 2 murine models of VHS, that is, postinflammatory TNBS colitis and maternal separation (MS). METHODS Wild-type (WT) mice and mice lacking ephrin-B2 in Nav 1.8 nociceptive neurons (cKO) were studied. VHS was induced by: 1. intracolonic instillation of TNBS or 2. water avoidance stress (WAS) in mice that underwent maternal separation (MS). VHS was assessed by quantifying the visceromotor response (VMRs) during colorectal distention. Colonic tissue and spinal cord were collected for histology, gene, and protein expression evaluation. KEY RESULTS In WT mice, but not cKO mice, TNBS induced VHS at day 14 after instillation, which returned to baseline perception from day 28 onwards. In MS WT mice, WAS induced VHS for up to 4 weeks. In cKO however, visceral pain perception returned to basal level by week 4. The development of VHS in WT mice was associated with significant upregulation of spinal ephrin-B2 and EphB1 mRNA expression or protein levels in the TNBS model and upregulation of spinal ephrin-B2 protein in the MS model. No changes were observed in cKO mice. VHS was not associated with persistent intestinal inflammation. CONCLUSIONS AND INFERENCES Overall, our data indicate that the ephrin-B2/EphB1 spinal signaling pathway is involved in VHS and may represent a novel therapeutic target.
Collapse
Affiliation(s)
| | - Morgane V Florens
- Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Iris Appeltans
- Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | | | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research (WIBR), University College London (UCL), London, UK
| | - Mira M Wouters
- Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Guy E Boeckxstaens
- Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| |
Collapse
|
50
|
Nguyen AQ, Koeppen J, Woodruff S, Mina K, Figueroa Z, Ethell IM. Astrocytic Ephrin-B1 Controls Synapse Formation in the Hippocampus During Learning and Memory. Front Synaptic Neurosci 2020; 12:10. [PMID: 32256333 PMCID: PMC7092624 DOI: 10.3389/fnsyn.2020.00010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/24/2020] [Indexed: 01/20/2023] Open
Abstract
Astrocytes play a fundamental role in synapse formation, pruning, and plasticity, which are associated with learning and memory. However, the role of astrocytes in learning and memory is still largely unknown. Our previous study showed that astrocyte-specific ephrin-B1 knock-out (KO) enhanced but ephrin-B1 overexpression (OE) in hippocampal astrocytes impaired contextual memory recall following fear conditioning. The goal of this study was to understand the mechanism by which astrocytic ephrin-B1 influences learning; specifically, learning-induced remodeling of synapses and dendritic spines in CA1 hippocampus using fear-conditioning paradigm. While we found a higher dendritic spine density and clustering on c-Fos-positive (+) neurons activated during contextual memory recall in both wild-type (WT) and KO mice, overall spine density and mEPSC amplitude were increased in CA1 neurons of KO compared to WT. In contrast, ephrin-B1 OE in hippocampal astrocytes impaired dendritic spine formation and clustering, specifically on c-Fos(+) neurons, coinciding with an overall decrease in vGlut1/PSD95 co-localization. Although astrocytic ephrin-B1 influenced learning-induced spine formation, the changes in astrocytic ephrin-B1 levels did not affect spine enlargement as no genotype differences in spine volume were observed between trained WT, KO, and OE groups. Our results suggest that a reduced formation of new spines rather than spine maturation in activated CA1 hippocampal neurons is most likely responsible for impaired contextual learning in OE mice due to abundantly high ephrin-B1 levels in astrocytes. The ability of astrocytic ephrin-B1 to negatively influence new spine formation during learning can potentially regulate new synapse formation at specific dendritic domains and underlie memory encoding.
Collapse
Affiliation(s)
- Amanda Q. Nguyen
- Division of Biomedical Sciences, University of California Riverside School of Medicine, Riverside, CA, United States
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States
| | - Jordan Koeppen
- Division of Biomedical Sciences, University of California Riverside School of Medicine, Riverside, CA, United States
- Cell, Molecular, and Developmental Biology Graduate Program, University of California, Riverside, Riverside, CA, United States
| | - Simone Woodruff
- Division of Biomedical Sciences, University of California Riverside School of Medicine, Riverside, CA, United States
| | - Karen Mina
- Division of Biomedical Sciences, University of California Riverside School of Medicine, Riverside, CA, United States
| | - Zoe Figueroa
- Division of Biomedical Sciences, University of California Riverside School of Medicine, Riverside, CA, United States
| | - Iryna M. Ethell
- Division of Biomedical Sciences, University of California Riverside School of Medicine, Riverside, CA, United States
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States
- Cell, Molecular, and Developmental Biology Graduate Program, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|