1
|
Cocito C, Xiang C, Huang M, Gongora T, Surana P, Davuluri R, Dahmane N, Greenfield JP. Immunoglobulin superfamily 3 (Igsf3) function is dispensable for brain development. Sci Rep 2025; 15:6526. [PMID: 39988603 PMCID: PMC11847924 DOI: 10.1038/s41598-024-79349-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 11/08/2024] [Indexed: 02/25/2025] Open
Abstract
The Immunoglobulin superfamily (IgSF) is a heterogeneous and conserved family of adhesion proteins crucial during the development of the central nervous system including neuronal migration and synaptogenesis. The Immunoglobulin superfamily member 3 (IGSF3) is expressed in the developing brain and has been suggested to play a role during morphological development of the granule cells neurites in the cerebellum. In addition, a role for IGSF3 in supporting glioma progression has been recently demonstrated. Remaining unexplored is the physiological role of IGSF3 in regulating brain development, including neocortical development. We generated an Igsf3 knockout (KO) mouse using a CRISPR/Cas9-based approach and explored the function of Igsf3 in regulating cortical development. We found that Igsf3 largely co-localizes with other IgSF proteins during cortical development and despite its expression being developmentally regulated in neuronal progenitors and in postmitotic neurons, Igsf3 is not essential for brain development, neuronal migration, or neuronal maturation.
Collapse
Affiliation(s)
- Carolina Cocito
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY, USA.
| | - Chaomei Xiang
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Meng Huang
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY, USA
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China
| | - Tatyana Gongora
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Pallavi Surana
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY, USA
| | - Ramana Davuluri
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY, USA
| | - Nadia Dahmane
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY, USA
| | | |
Collapse
|
2
|
McMillan HP, Lundy FT, Dunne OM, McLoughlin KJ, About I, Curtis TM, El Karim I. Immunological isolation and characterization of neuronal progenitors from human dental pulp: A laboratory-based investigation. Int Endod J 2024; 57:1136-1146. [PMID: 38713428 DOI: 10.1111/iej.14077] [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: 11/14/2023] [Revised: 02/21/2024] [Accepted: 04/14/2024] [Indexed: 05/08/2024]
Abstract
AIMS Dental pulp stem cells (DPSCs) contain a population of stem cells with a broad range of differentiation potentials, as well as more lineage-committed progenitors. Such heterogeneity is a significant obstacle to experimental and clinical applications. The aim of this study is to isolate and characterize a homogenous neuronal progenitor cell population from human DPSCs. METHODOLOGY Polysialylated-neural cell adhesion molecule (PSA-NCAM+) neural progenitors were isolated from the dental pulp of three independent donors using magnetic-activated cell sorting (MACS) technology. Immunofluorescent staining with a panel of neural and non-neural markers was used to characterize the magnetically isolated PSA-NCAM+ fraction. PSA-NCAM+ cells were then cultured in Neurobasal A supplemented with neurotrophic factors: dibutyryl cyclic-AMP, neurotrophin-3, B27 and N2 supplements to induce neuronal differentiation. Both PSA-NCAM+ and differentiated PSA-NCAM+ cells were used in Ca2+ imaging studies to assess the functionality of P2X3 receptors as well as membrane depolarization. RESULTS PSA-NCAM+ neural progenitors were isolated from a heterogeneous population of hDPSCs using magnetic-activated cell sorting and anti-PSA-NCAM MicroBeads. Flow cytometry analysis demonstrated that immunomagnetic sorting significantly increased the purity of PSA-NCAM+ cells. Immunofluorescent staining revealed expression of pan-neuronal and mature neuronal markers, PGP9.5 and MAP2, respectively, as well as weak expression of the mature sensory markers, peripherin and islet1. ATP-induced response was mediated predominately by P2X3 receptors in both undifferentiated and differentiated cells, with a greater magnitude observed in the latter. In addition, membrane depolarizations were also detected in cells before and after differentiation when loaded with fast-voltage-responding fluorescent molecule, FluoVolt™ in response to potassium chloride. Interestingly, only differentiated PSA-NCAM+ cells were capable of spontaneous membrane oscillations. CONCLUSIONS In summary, DPSCs contain a population of neuronal progenitors with enhanced neural differentiation and functional neural-like properties that can be effectively isolated with magnetic-activated cell sorting (MACS).
Collapse
Affiliation(s)
- Hayley P McMillan
- School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, UK
| | - Fionnuala T Lundy
- School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, UK
| | - Orla M Dunne
- School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, UK
| | - Kiran John McLoughlin
- School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, UK
| | - Imad About
- Aix Marseille University, CNRS, Institute of Movement Sciences, Marseille, France
| | - T M Curtis
- School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, UK
| | - Ikhlas El Karim
- School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, UK
| |
Collapse
|
3
|
Lin X, Ren P, Xue Z, Liu X, Cao Y, Li T, Miao H. Astrocytic GDNF ameliorates anesthesia and surgery-induced cognitive impairment by promoting hippocampal synaptic plasticity in aged mice. Neurochem Int 2024; 177:105765. [PMID: 38750960 DOI: 10.1016/j.neuint.2024.105765] [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: 01/06/2024] [Revised: 05/08/2024] [Accepted: 05/12/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Perioperative neurocognitive disorders (PND) are common complications after surgery in older patients. However, the specific mechanism of this condition remains unclear. Glial cell line-derived neurotrophic factor (GDNF) is an important neurotrophin that abundantly expressed throughout the brain. It can enhance synaptic plasticity and alleviate learning and memory impairments. Thus, the purpose of this study was to investigate the role of GDNF in PND and the mechanisms involved. METHODS The PND animal model was established by performing left tibial fracture surgery on 18-month-old C57BL/6 mice under sevoflurane anesthesia. Recombinant adeno-associated virus (rAAV)-GDNF or empty vectors were injected bilaterally into the hippocampal CA1 region of aged mice 3 weeks before anesthesia/surgery. The open field and fear conditioning test were used to assess the behavior changes. Golgi staining and electrophysiology were utilized to evaluate the morphological and functional alterations of neuronal synaptic plasticity. Western blot analysis was carried out to measure the proteins expression levels and immunofluorescence staining was performed to probe the cellular localization of GDNF. RESULTS Mice with surgery and anesthesia showed a significant decrease in hippocampus-dependent learning and memory, accompanied by a decline in hippocampal synaptic plasticity. Anesthesia/surgery induced a reduction of GDNF, which was colocalized with astrocytes. Overexpression of GDNF in astrocytes could ameliorate the decline in cognitive function by improving hippocampal synaptic plasticity, meanwhile astrocytic GDNF rescued the anesthesia/surgery-induced decrease in GFRα1 and NCAM. CONCLUSION The study concludes that astrocytic GDNF may improve anesthesia/surgery-induced cognitive impairment by promoting hippocampal synaptic plasticity in aged mice via the GFRα1/NCAM pathway.
Collapse
Affiliation(s)
- Xiaowan Lin
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Peng Ren
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Ziyi Xue
- Department of Anesthesiology, Peking University First Hospital, Beijing, China
| | - Xiao Liu
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Ying Cao
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Tianzuo Li
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
| | - Huihui Miao
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Sannes AC, Christensen JO, Nielsen MB, Gjerstad J. Stress-induced headache in the general working population is moderated by the NRCAM rs2300043 genotype. Scand J Pain 2022; 23:326-332. [PMID: 36181733 DOI: 10.1515/sjpain-2022-0094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/19/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Earlier findings suggest that social stress such as abusive supervision may promote pain. In the present study we examine the possible moderating role of genetic variability in the NRCAM gene in this process. METHODS The data were collected through a national survey drawn from the National Central Employee Register by Statistics Norway. A total of 1,205 individuals returned both the questionnaire and the saliva kit. Abusive supervision was scored by a 5-item version of the Tepper's 2,000 scale. Headache was measured on a four-category scale; 'not bothered,' 'a little bothered,' 'considerably bothered', 'seriously bothered'. Genotyping with regards to NRCAM rs2300043 was carried out using Taqman assay. Ordinal logistic regression was used to analyse the data. RESULTS For males exposed to abusive supervision, those carrying the rs2300043 CC genotype reported the highest levels of headache. Women showed a trend towards the opposite pattern. Women with the rs2300043 CC genotype seem to have a weaker effect of abusive supervision regarding reported headache than their male counterparts with the CC genotype when exposed to abusive supervision. CONCLUSIONS The present results indicated that the association between abusive supervision and headache in men with the NRCAM rs2300043 C allele was stronger than in other men. This suggests that the NRCAM genotype in men is important for the tolerance of social stress e.g., repeated negative acts from a superior. In contrast, a trend, though non-significant, towards the opposite pattern was observed in women. Our result suggests that the NRCAM genotype in men manifestly affects stress-induced pain such as headache.
Collapse
|
5
|
Luo W, Cruz-Ochoa NA, Seng C, Egger M, Lukacsovich D, Lukacsovich T, Földy C. Pcdh11x controls target specification of mossy fiber sprouting. Front Neurosci 2022; 16:888362. [PMID: 36117624 PMCID: PMC9475199 DOI: 10.3389/fnins.2022.888362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Circuit formation is a defining characteristic of the developing brain. However, multiple lines of evidence suggest that circuit formation can also take place in adults, the mechanisms of which remain poorly understood. Here, we investigated the epilepsy-associated mossy fiber (MF) sprouting in the adult hippocampus and asked which cell surface molecules define its target specificity. Using single-cell RNAseq data, we found lack and expression of Pcdh11x in non-sprouting and sprouting neurons respectively. Subsequently, we used CRISPR/Cas9 genome editing to disrupt the Pcdh11x gene and characterized its consequences on sprouting. Although MF sprouting still developed, its target specificity was altered. New synapses were frequently formed on granule cell somata in addition to dendrites. Our findings shed light onto a key molecular determinant of target specificity in MF sprouting and contribute to understanding the molecular mechanism of adult brain rewiring.
Collapse
|
6
|
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
|
7
|
Chen H, Carty RK, Bautista AC, Hayakawa KA, Lein PJ. Triiodothyronine or Antioxidants Block the Inhibitory Effects of BDE-47 and BDE-49 on Axonal Growth in Rat Hippocampal Neuron-Glia Co-Cultures. TOXICS 2022; 10:92. [PMID: 35202279 PMCID: PMC8879960 DOI: 10.3390/toxics10020092] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/02/2022] [Accepted: 02/02/2022] [Indexed: 12/31/2022]
Abstract
We previously demonstrated that polybrominated diphenyl ethers (PBDEs) inhibit the growth of axons in primary rat hippocampal neurons. Here, we test the hypothesis that PBDE effects on axonal morphogenesis are mediated by thyroid hormone and/or reactive oxygen species (ROS)-dependent mechanisms. Axonal growth and ROS were quantified in primary neuronal-glial co-cultures dissociated from neonatal rat hippocampi exposed to nM concentrations of BDE-47 or BDE-49 in the absence or presence of triiodothyronine (T3; 3-30 nM), N-acetyl-cysteine (NAC; 100 µM), or α-tocopherol (100 µM). Co-exposure to T3 or either antioxidant prevented inhibition of axonal growth in hippocampal cultures exposed to BDE-47 or BDE-49. T3 supplementation in cultures not exposed to PBDEs did not alter axonal growth. T3 did, however, prevent PBDE-induced ROS generation and alterations in mitochondrial metabolism. Collectively, our data indicate that PBDEs inhibit axonal growth via ROS-dependent mechanisms, and that T3 protects axonal growth by inhibiting PBDE-induced ROS. These observations suggest that co-exposure to endocrine disruptors that decrease TH signaling in the brain may increase vulnerability to the adverse effects of developmental PBDE exposure on axonal morphogenesis.
Collapse
Affiliation(s)
| | | | | | | | - Pamela J. Lein
- Department of Molecular Biosciences, University of California, Davis, CA 95616, USA; (H.C.); (R.K.C.); (A.C.B.); (K.A.H.)
| |
Collapse
|
8
|
Spatiotemporal processing of neural cell adhesion molecules 1 and 2 by BACE1 in vivo. J Biol Chem 2021; 296:100372. [PMID: 33548223 PMCID: PMC7949136 DOI: 10.1016/j.jbc.2021.100372] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/27/2021] [Accepted: 02/01/2021] [Indexed: 12/13/2022] Open
Abstract
Neural cell adhesion molecules 1 (NCAM1) and 2 (NCAM2) belong to the cell adhesion molecules of the immunoglobulin superfamily and have been shown to regulate formation, maturation, and maintenance of synapses. NCAM1 and NCAM2 undergo proteolysis, but the identity of all the proteases involved and how proteolysis is used to regulate their functions are not known. We report here that NCAM1 and NCAM2 are BACE1 substrates in vivo. NCAM1 and NCAM2 overexpressed in HEK cells were both cleaved by metalloproteinases or BACE1, and NCAM2 was also processed by γ-secretase. We identified the BACE1 cleavage site of NCAM1 (at Glu 671) and NCAM2 (at Glu 663) using mass spectrometry and site-directed mutagenesis. Next, we assessed BACE1-mediated processing of NCAM1 and NCAM2 in the mouse brain during aging. NCAM1 and NCAM2 were cleaved in the olfactory bulb of BACE1+/+ but not BACE1−/− mice at postnatal day 10 (P10), 4 and 12 months of age. In the hippocampus, a BACE1-specific soluble fragment of NCAM1 (sNCAM1β) was only detected at P10. However, we observed an accumulation of full-length NCAM1 in hippocampal synaptosomes in 4-month-old BACE1−/− mice. We also found that polysialylated NCAM1 (PSA-NCAM1) levels were increased in BACE1−/− mice at P10 and demonstrated that BACE1 cleaves both NCAM1 and PSA-NCAM1 in vitro. In contrast, we did not find evidence for BACE1-dependent NCAM2 processing in the hippocampus at any age analyzed. In summary, our data demonstrate that BACE1 differentially processes NCAM1 and NCAM2 depending on the region of brain, subcellular localization, and age in vivo.
Collapse
|
9
|
An H, Qin J, Fan H, Fan F, Tan S, Wang Z, Shi J, Yang F, Tan Y, Huang XF. Decreased serum NCAM is positively correlated with hippocampal volumes and negatively correlated with positive symptoms in first-episode schizophrenia patients. J Psychiatr Res 2020; 131:108-113. [PMID: 32950707 DOI: 10.1016/j.jpsychires.2020.09.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Neural cell adhesion molecule (NCAM) plays an important role in neurodevelopmental processes and regulates hippocampal plasticity. This study investigated the relationship between the serum NCAM concentrations and hippocampal volume and psychotic symptoms in first-episode drug naïve schizophrenia (FES) patients. METHODS Forty-four FES patients and forty-four healthy controls (HC) were recruited in this study. Serum concentrations of NCAM were measured by ELISA. Psychiatric symptoms were assessed by the positive and negative syndrome scale (PANSS). Brain structural images were obtained using a 3T MRI Scanner and obtained T1 images were processed in order to determine hippocampal grey matter volumes. RESULTS Schizophrenia patients revealed significantly decreased serum NCAM concentrations (p = 0.017), which were positively correlated with the left (r = 0.523, p < 0.001) and right (r = 0.449, p = 0.041) hippocampal volumes, but negatively correlated with the PANSS positive symptom scores (r = -0.522 p = 0.001). However, no such correlations existed in the HC group. CONCLUSIONS This is the first time to report that decreased serum NCAM concentrations were associated with hippocampal volumes and symptom severity in FES patients. Our data indicate that the low NCAM is possible neuropathology that is associated with the decreased hippocampus in FES patients.
Collapse
Affiliation(s)
- Huimei An
- Beijing HuiLongGuan Hospital, Peking University, Beijing, China
| | - Jun Qin
- Radiology Department, Civil Aviation General Hospital, Peking University, Beijing, China
| | - Hongzhen Fan
- Beijing HuiLongGuan Hospital, Peking University, Beijing, China
| | - Fengmei Fan
- Beijing HuiLongGuan Hospital, Peking University, Beijing, China
| | - Shuping Tan
- Beijing HuiLongGuan Hospital, Peking University, Beijing, China
| | - Zhiren Wang
- Beijing HuiLongGuan Hospital, Peking University, Beijing, China
| | - Jing Shi
- Beijing HuiLongGuan Hospital, Peking University, Beijing, China
| | - Fude Yang
- Beijing HuiLongGuan Hospital, Peking University, Beijing, China
| | - Yunlong Tan
- Beijing HuiLongGuan Hospital, Peking University, Beijing, China.
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute and School of Medicine, University of Wollongong, NSW, 2522, Australia.
| |
Collapse
|
10
|
The Role of Cell Adhesion Molecule Genes Regulating Neuroplasticity in Addiction. Neural Plast 2018; 2018:9803764. [PMID: 29675039 PMCID: PMC5838467 DOI: 10.1155/2018/9803764] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 12/10/2017] [Indexed: 01/06/2023] Open
Abstract
A variety of genetic approaches, including twin studies, linkage studies, and candidate gene studies, has established a firm genetic basis for addiction. However, there has been difficulty identifying the precise genes that underlie addiction liability using these approaches. This situation became especially clear in genome-wide association studies (GWAS) of addiction. Moreover, the results of GWAS brought into clarity many of the shortcomings of those early genetic approaches. GWAS studies stripped away those preconceived notions, examining genes that would not previously have been considered in the study of addiction, consequently creating a shift in our understanding. Most importantly, those studies implicated a class of genes that had not previously been considered in the study of addiction genetics: cell adhesion molecules (CAMs). Considering the well-documented evidence supporting a role for various CAMs in synaptic plasticity, axonal growth, and regeneration, it is not surprising that allelic variation in CAM genes might also play a role in addiction liability. This review focuses on the role of various cell adhesion molecules in neuroplasticity that might contribute to addictive processes and emphasizes the importance of ongoing research on CAM genes that have been implicated in addiction by GWAS.
Collapse
|
11
|
Kim DH, Kang M, Kim CH, Huh YH, Cho IH, Ryu HH, Chung KH, Park CS, Rhee S, Lee YS, Song WK. SPIN90 Modulates Long-Term Depression and Behavioral Flexibility in the Hippocampus. Front Mol Neurosci 2017; 10:295. [PMID: 28979184 PMCID: PMC5611360 DOI: 10.3389/fnmol.2017.00295] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 09/01/2017] [Indexed: 12/23/2022] Open
Abstract
The importance of actin-binding proteins (ABPs) in the regulation of synapse morphology and plasticity has been well established. SH3 protein interacting with Nck, 90 kDa (SPIN90), an Nck-interacting protein highly expressed in synapses, is essential for actin remodeling and dendritic spine morphology. Synaptic targeting of SPIN90 to spine heads or dendritic shafts depends on its phosphorylation state, leading to blockage of cofilin-mediated actin depolymerization and spine shrinkage. However, the physiological role of SPIN90 in long-term plasticity, learning and memory are largely unknown. In this study, we demonstrate that Spin90-knockout (KO) mice exhibit substantial deficits in synaptic plasticity and behavioral flexibility. We found that loss of SPIN90 disrupted dendritic spine density in CA1 neurons of the hippocampus and significantly impaired long-term depression (LTD), leaving basal synaptic transmission and long-term potentiation (LTP) intact. These impairments were due in part to deficits in AMPA receptor endocytosis and its pre-requisites, GluA1 dephosphorylation and postsynaptic density (PSD) 95 phosphorylation, but also by an intrinsic activation of Akt-GSK3β signaling as a result of Spin90-KO. In accordance with these defects, mice lacking SPIN90 were found to carry significant deficits in object-recognition and behavioral flexibility, while learning ability was largely unaffected. Collectively, these findings demonstrate a novel modulatory role for SPIN90 in hippocampal LTD and behavioral flexibility.
Collapse
Affiliation(s)
- Dae Hwan Kim
- Bio Imaging and Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and TechnologyGwangju, South Korea
| | - Minkyung Kang
- Department of Physiology, Department of Biomedical Sciences, Seoul National University College of MedicineSeoul, South Korea
| | - Chong-Hyun Kim
- Center for Neuroscience, Korea Institute of Science and Technology, Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and TechnologySeoul, South Korea
| | - Yun Hyun Huh
- Bio Imaging and Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and TechnologyGwangju, South Korea
| | - In Ha Cho
- Department of Biological Sciences, Dartmouth CollegeHanover, NH, United States
| | - Hyun-Hee Ryu
- Department of Physiology, Department of Biomedical Sciences, Seoul National University College of MedicineSeoul, South Korea.,Department of Life Science, Chung-Ang UniversitySeoul, South Korea
| | - Kyung Hwun Chung
- Electron Microscope Facility, Dental Research Institute, Seoul National UniversitySeoul, South Korea
| | - Chul-Seung Park
- School of Life Sciences, Gwangju Institute of Science and TechnologyGwangju, South Korea
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang UniversitySeoul, South Korea
| | - Yong-Seok Lee
- Department of Physiology, Department of Biomedical Sciences, Seoul National University College of MedicineSeoul, South Korea
| | - Woo Keun Song
- Bio Imaging and Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and TechnologyGwangju, South Korea
| |
Collapse
|
12
|
|
13
|
Salinas S, Junyent F, Coré N, Cremer H, Kremer EJ. What is CAR doing in the middle of the adult neurogenic road? NEUROGENESIS 2017; 4:e1304790. [PMID: 28516108 DOI: 10.1080/23262133.2017.1304790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/02/2017] [Accepted: 03/03/2017] [Indexed: 10/19/2022]
Abstract
The molecular and cellular basis of adult neurogenesis has attracted considerable attention for fundamental and clinical applications because neural stem cells and newborn neurons may, one day, be harnessed to replace neurons and allow cognitive improvement in the diseased brain. In rodents, neural progenitors are located in the dentate gyrus and the sub/periventricular zone. In the dentate gyrus the generation of newborn neurons is associated with plasticity, including regulation of memory. The role of subventricular zone neural precursors that migrate to the olfactory bulb is less characterized. Identifying factors that impact neural stem cell proliferation, migration and differentiation is therefore sine qua non before we can harness their potential. Here, we expand upon our recent results showing that CAR, the coxsackievirus and adenovirus receptor, is among the developing list of key players when it comes to the complex process of integrating newborn neurons into existing circuits in the mature brain.
Collapse
Affiliation(s)
- Sara Salinas
- Université de Montpellier, Montpellier, France.,Pathogenesis and Control of Chronic Infections, Inserm UMR 1058, Montpellier, France
| | - Felix Junyent
- Université de Montpellier, Montpellier, France.,Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier
| | - Nathalie Coré
- Institut de Biologie du Développement de Marseille, CNRS UMR 7288, Marseille, France.,Aix-Marseille Université, Marseille, France
| | - Harold Cremer
- Institut de Biologie du Développement de Marseille, CNRS UMR 7288, Marseille, France.,Aix-Marseille Université, Marseille, France
| | - Eric J Kremer
- Université de Montpellier, Montpellier, France.,Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier
| |
Collapse
|
14
|
Neural Cell Adhesion Molecules of the Immunoglobulin Superfamily Regulate Synapse Formation, Maintenance, and Function. Trends Neurosci 2017; 40:295-308. [PMID: 28359630 DOI: 10.1016/j.tins.2017.03.003] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 02/05/2023]
Abstract
Immunoglobulin superfamily adhesion molecules are among the most abundant proteins in vertebrate and invertebrate nervous systems. Prominent family members are the neural cell adhesion molecules NCAM and L1, which were the first to be shown to be essential not only in development but also in synaptic function and as key regulators of synapse formation, synaptic activity, plasticity, and synaptic vesicle recycling at distinct developmental and activity stages. In addition to interacting with each other, adhesion molecules interact with ion channels and cytokine and neurotransmitter receptors. Mutations in their genes are linked to neurological disorders associated with abnormal development and synaptic functioning. This review presents an overview of recent studies on these molecules and their crucial impact on neurological disorders.
Collapse
|
15
|
Gulisano W, Bizzoca A, Gennarini G, Palmeri A, Puzzo D. Role of the adhesion molecule F3/Contactin in synaptic plasticity and memory. Mol Cell Neurosci 2016; 81:64-71. [PMID: 28038945 DOI: 10.1016/j.mcn.2016.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 12/07/2016] [Accepted: 12/22/2016] [Indexed: 12/14/2022] Open
Abstract
Cell adhesion molecules (CAMs) have a pivotal role in building and maintaining synaptic structures during brain development participating in axonal elongation and pathfinding, glial guidance of neuronal migration, as well as myelination. CAMs expression persists in the adult brain particularly in structures undergoing postnatal neurogenesis and involved in synaptic plasticity and memory as the hippocampus. Among the neural CAMs, we have recently focused on F3/Contactin, a glycosylphosphatidyl inositol-anchored glycoprotein belonging to the immunoglobulin superfamily, involved in neuronal development, synaptic maintenance and organization of neuronal networks. Here, we discuss our recent data suggesting that F3/Contactin exerts a role in hippocampal synaptic plasticity and memory in adult and aged mice. In particular, we have studied long-term potentiation (LTP), spatial and object recognition memory, and phosphorylation of the transcription factor cAMP-Responsive-Element Binding protein (CREB) in a transgenic mouse model of F3/Contactin overexpression. We also investigated whether F3/Contactin might influence neuronal apoptosis and the production of amyloid-beta peptide (Aβ), known to be one of the main pathogenetic hallmarks of Alzheimer's disease (AD). In conclusion, a further understanding of F3/Contactin role in synaptic plasticity and memory might have interesting clinical outcomes in cognitive disorders, such as aging and AD, offering innovative therapeutic opportunities.
Collapse
Affiliation(s)
- Walter Gulisano
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Antonella Bizzoca
- Section of Physiology, Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, Bari, Italy
| | - Gianfranco Gennarini
- Section of Physiology, Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, Bari, Italy
| | - Agostino Palmeri
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| | - Daniela Puzzo
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
16
|
Regulation of extrasynaptic signaling by polysialylated NCAM: Impact for synaptic plasticity and cognitive functions. Mol Cell Neurosci 2016; 81:12-21. [PMID: 27865768 DOI: 10.1016/j.mcn.2016.11.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/10/2016] [Accepted: 11/11/2016] [Indexed: 01/24/2023] Open
Abstract
The activation of synaptic N-methyl-d-aspartate-receptors (NMDARs) is crucial for induction of synaptic plasticity and supports cell survival, whereas activation of extrasynaptic NMDARs inhibits long-term potentiation and triggers neurodegeneration. A soluble polysialylated form of the neural cell adhesion molecule (polySia-NCAM) suppresses signaling through peri-/extrasynaptic GluN2B-containing NMDARs. Genetic or enzymatic manipulations blocking this mechanism result in impaired synaptic plasticity and learning, which could be repaired by reintroduction of polySia, or inhibition of either GluN1/GluN2B receptors or downstream signaling through RasGRF1 and p38 MAP kinase. Ectodomain shedding of NCAM, and hence generation of soluble NCAM, is controlled by metalloproteases of a disintegrin and metalloprotease (ADAM) family. As polySia-NCAM is predominantly associated with GABAergic interneurons in the prefrontal cortex, it is noteworthy that EphrinA5/EphA3-induced ADAM10 activity promotes polySia-NCAM shedding in these neurons. Thus, in addition to the well-known regulation of synaptic NMDARs by the secreted molecule Reelin, shed polySia-NCAM may restrain activation of extrasynaptic NMDARs. These data support a concept that GABAergic interneuron-derived extracellular proteins control the balance in synaptic/extrasynaptic NMDAR-mediated signaling in principal cells. Strikingly, dysregulation of Reelin or polySia expression is linked to schizophrenia. Thus, targeting of the GABAergic interneuron-principle cell communication and restoring the balance in synaptic/extrasynaptic NMDARs represent promising strategies for treatment of psychiatric diseases.
Collapse
|
17
|
Aonurm-Helm A, Jaako K, Jürgenson M, Zharkovsky A. Pharmacological approach for targeting dysfunctional brain plasticity: Focus on neural cell adhesion molecule (NCAM). Pharmacol Res 2016; 113:731-738. [DOI: 10.1016/j.phrs.2016.04.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/29/2016] [Accepted: 04/08/2016] [Indexed: 11/26/2022]
|
18
|
Leshchyns'ka I, Sytnyk V. Intracellular transport and cell surface delivery of the neural cell adhesion molecule (NCAM). BIOARCHITECTURE 2016; 5:54-60. [PMID: 26605672 DOI: 10.1080/19490992.2015.1118194] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The neural cell adhesion molecule (NCAM) regulates differentiation and functioning of neurons by accumulating at the cell surface where it mediates the interactions of neurons with the extracellular environment. NCAM also induces a number of intracellular signaling cascades, which coordinate interactions at the cell surface with intracellular processes including changes in gene expression, transport and cytoskeleton remodeling. Since NCAM functions at the cell surface, its transport and delivery to the cell surface play a critical role. Here, we review recent advances in our understanding of the molecular mechanisms of the intracellular transport and cell surface delivery of NCAM. We also discuss the data suggesting a possibility of cross talk between activation of NCAM at the cell surface and the intracellular transport and cell surface delivery of NCAM.
Collapse
Affiliation(s)
- Iryna Leshchyns'ka
- a School of Biotechnology and Biomolecular Sciences ; The University of New South Wales ; Sydney , NSW , Australia
| | - Vladimir Sytnyk
- a School of Biotechnology and Biomolecular Sciences ; The University of New South Wales ; Sydney , NSW , Australia
| |
Collapse
|
19
|
Irala D, Bonafina A, Fontanet PA, Alsina FC, Paratcha G, Ledda F. The GDNF-GFRα1 complex promotes the development of hippocampal dendritic arbors and spines via NCAM. Development 2016; 143:4224-4235. [PMID: 27707798 DOI: 10.1242/dev.140350] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 09/28/2016] [Indexed: 12/26/2022]
Abstract
The formation of synaptic connections during nervous system development requires the precise control of dendrite growth and synapse formation. Although glial cell line-derived neurotrophic factor (GDNF) and its receptor GFRα1 are expressed in the forebrain, the role of this system in the hippocampus remains unclear. Here, we investigated the consequences of GFRα1 deficiency for the development of hippocampal connections. Analysis of conditional Gfra1 knockout mice shows a reduction in dendritic length and complexity, as well as a decrease in postsynaptic density specializations and in the synaptic localization of postsynaptic proteins in hippocampal neurons. Gain- and loss-of-function assays demonstrate that the GDNF-GFRα1 complex promotes dendritic growth and postsynaptic differentiation in cultured hippocampal neurons. Finally, in vitro assays revealed that GDNF-GFRα1-induced dendrite growth and spine formation are mediated by NCAM signaling. Taken together, our results indicate that the GDNF-GFRα1 complex is essential for proper hippocampal circuit development.
Collapse
Affiliation(s)
- Dolores Irala
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Buenos Aires 1121, Argentina
| | - Antonela Bonafina
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Buenos Aires 1121, Argentina
| | - Paula Aldana Fontanet
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Buenos Aires 1121, Argentina
| | - Fernando Cruz Alsina
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Buenos Aires 1121, Argentina
| | - Gustavo Paratcha
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Buenos Aires 1121, Argentina
| | - Fernanda Ledda
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Buenos Aires 1121, Argentina
| |
Collapse
|
20
|
Jaako K, Waniek A, Parik K, Klimaviciusa L, Aonurm-Helm A, Noortoots A, Anier K, Van Elzen R, Gérard M, Lambeir AM, Roßner S, Morawski M, Zharkovsky A. Prolyl endopeptidase is involved in the degradation of neural cell adhesion molecules in vitro. J Cell Sci 2016; 129:3792-3802. [PMID: 27566163 DOI: 10.1242/jcs.181891] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 08/18/2016] [Indexed: 12/14/2022] Open
Abstract
Membrane-associated glycoprotein neural cell adhesion molecule (NCAM) and its polysialylated form (PSA-NCAM) play an important role in brain plasticity by regulating cell-cell interactions. Here, we demonstrate that the cytosolic serine protease prolyl endopeptidase (PREP) is able to regulate NCAM and PSA-NCAM. Using a SH-SY5Y neuroblastoma cell line with stable overexpression of PREP, we found a remarkable loss of PSA-NCAM, reduced levels of NCAM180 and NCAM140 protein species, and a significant increase in the NCAM immunoreactive band migrating at an apparent molecular weight of 120 kDa in PREP-overexpressing cells. Moreover, increased levels of NCAM fragments were found in the concentrated medium derived from PREP-overexpressing cells. PREP overexpression selectively induced an activation of matrix metalloproteinase-9 (MMP-9), which could be involved in the observed degradation of NCAM, as MMP-9 neutralization reduced the levels of NCAM fragments in cell culture medium. We propose that increased PREP levels promote epidermal growth factor receptor (EGFR) signaling, which in turn activates MMP-9. In conclusion, our findings provide evidence for newly-discovered roles for PREP in mechanisms regulating cellular plasticity through NCAM and PSA-NCAM.
Collapse
Affiliation(s)
- Külli Jaako
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu 50411, Estonia
| | - Alexander Waniek
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig 04103, Germany
| | - Keiti Parik
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu 50411, Estonia
| | - Linda Klimaviciusa
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu 50411, Estonia
| | - Anu Aonurm-Helm
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu 50411, Estonia
| | - Aveli Noortoots
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu 50411, Estonia
| | - Kaili Anier
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu 50411, Estonia
| | - Roos Van Elzen
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp B-2610, Belgium
| | - Melanie Gérard
- Interdisciplinary Research Centre KU Leuven-Kortrijk, Kortrijk B-8500, Belgium
| | - Anne-Marie Lambeir
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp B-2610, Belgium
| | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig 04103, Germany
| | - Markus Morawski
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig 04103, Germany
| | - Alexander Zharkovsky
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu 50411, Estonia
| |
Collapse
|
21
|
The anticonvulsant action of the galanin receptor agonist NAX-5055 involves modulation of both excitatory- and inhibitory neurotransmission. Epilepsy Res 2016; 121:55-63. [PMID: 26894875 DOI: 10.1016/j.eplepsyres.2016.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 01/11/2016] [Accepted: 01/24/2016] [Indexed: 11/23/2022]
Abstract
The endogenous neuropeptide galanin is ubiquitously expressed throughout the mammalian brain. Through the galanin receptors GalR1-3, galanin has been demonstrated to modulate both glutamatergic and GABAergic neurotransmission, and this appears to be important in epilepsy and seizure activity. Accordingly, galanin analogues are likely to provide a new approach to seizure management. However, since peptides are generally poor candidates for therapeutic agents due to their poor metabolic stability and low brain bioavailability, a search for alternative strategies for the development of galanin-based anti-convulsant drugs was prompted. Based on this, a rationally designed GalR1 preferring galanin analogue, NAX-5055, was synthesized. This compound demonstrates anti-convulsant actions in several animal models of epilepsy. However, the alterations at the cellular level leading to this anti-convulsant action of NAX-5055 are not known. Here we investigate the action of NAX-5055 at the cellular level by determining its effects on excitatory and inhibitory neurotransmission, i.e. vesicular release of glutamate and GABA, respectively, in cerebellar, neocortical and hippocampal preparations. In addition, its effects on cell viability and neurotransmitter transporter capacity were examined to evaluate potential cell toxicity mediated by NAX-5055. It was found that vesicular release of glutamate was reduced concentration-dependently by NAX-5055 in the range from 0.1 to 1000 nM. Moreover, exposure to 1 μM NAX-5055 led to a reduction in the extracellular level of glutamate and an elevation of the extracellular level of GABA. Altogether these findings may at least partly explain the anti-convulsant effect of NAX-5055 observed in vivo.
Collapse
|
22
|
Kuhn PH, Colombo AV, Schusser B, Dreymueller D, Wetzel S, Schepers U, Herber J, Ludwig A, Kremmer E, Montag D, Müller U, Schweizer M, Saftig P, Bräse S, Lichtenthaler SF. Systematic substrate identification indicates a central role for the metalloprotease ADAM10 in axon targeting and synapse function. eLife 2016; 5. [PMID: 26802628 PMCID: PMC4786429 DOI: 10.7554/elife.12748] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/22/2016] [Indexed: 12/11/2022] Open
Abstract
Metzincin metalloproteases have major roles in intercellular communication by modulating the function of membrane proteins. One of the proteases is the a-disintegrin-and-metalloprotease 10 (ADAM10) which acts as alpha-secretase of the Alzheimer's disease amyloid precursor protein. ADAM10 is also required for neuronal network functions in murine brain, but neuronal ADAM10 substrates are only partly known. With a proteomic analysis of Adam10-deficient neurons we identified 91, mostly novel ADAM10 substrate candidates, making ADAM10 a major protease for membrane proteins in the nervous system. Several novel substrates, including the neuronal cell adhesion protein NrCAM, are involved in brain development. Indeed, we detected mistargeted axons in the olfactory bulb of conditional ADAM10-/- mice, which correlate with reduced cleavage of NrCAM, NCAM and other ADAM10 substrates. In summary, the novel ADAM10 substrates provide a molecular basis for neuronal network dysfunctions in conditional ADAM10-/- mice and demonstrate a fundamental function of ADAM10 in the brain.
Collapse
Affiliation(s)
- Peer-Hendrik Kuhn
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Institut für Pathologie und Pathologische Anatomie, Technische Universität München, Munich, Germany.,Institute for Advanced Study, Technische Universität München, Munich, Germany
| | - Alessio Vittorio Colombo
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany
| | - Benjamin Schusser
- Department of Animal Science, Institute for Animal Physiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Daniela Dreymueller
- Institute of Pharmacology and Toxicology, Uniklinik RWTH Aachen, Aachen, Germany
| | - Sebastian Wetzel
- Institute of Biochemistry, Christian-Albrechts Universität zu Kiel, Kiel, Germany
| | - Ute Schepers
- Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Julia Herber
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany
| | - Andreas Ludwig
- Institute of Pharmacology and Toxicology, Uniklinik RWTH Aachen, Aachen, Germany
| | - Elisabeth Kremmer
- German Research Center for Environmental Health, Institute of Molecular Tumor immunology, Helmholtz Zentrum München, Munich, Germany
| | - Dirk Montag
- Neurogenetics, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Ulrike Müller
- Department of Functional Genomics, Institute for Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Michaela Schweizer
- Service-Gruppe für Elektronenmikroskopie, Zentrum für Molekulare Neurobiologie, Hamburg, Germany
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts Universität zu Kiel, Kiel, Germany
| | - Stefan Bräse
- Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Stefan F Lichtenthaler
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Institute for Advanced Study, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany.,Munich Cluster for Systems Neurology, Munich, Germany
| |
Collapse
|
23
|
Aonurm-Helm A, Anier K, Zharkovsky T, Castrén E, Rantamäki T, Stepanov V, Järv J, Zharkovsky A. NCAM-deficient mice show prominent abnormalities in serotonergic and BDNF systems in brain - Restoration by chronic amitriptyline. Eur Neuropsychopharmacol 2015; 25:2394-403. [PMID: 26499173 DOI: 10.1016/j.euroneuro.2015.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 08/13/2015] [Accepted: 10/05/2015] [Indexed: 11/19/2022]
Abstract
Mood disorders are associated with alterations in serotonergic system, deficient BDNF (brain-derived neurotrophic factor) signaling and abnormal synaptic plasticity. Increased degradation and reduced functions of NCAM (neural cell adhesion molecule) have recently been associated with depression and NCAM deficient mice show depression-related behavior and impaired learning. The aim of the present study was to investigate potential changes in serotonergic and BDNF systems in NCAM knock-out mice. Serotonergic nerve fiber density and SERT (serotonin transporter) protein levels were robustly reduced in the hippocampus, prefrontal cortex and basolateral amygdala of adult NCAM(-)(/-) mice. This SERT reduction was already evident during early postnatal development. [(3)H]MADAM binding experiments further demonstrated reduced availability of SERT in cell membranes of NCAM(-)(/-) mice. Moreover, the levels of serotonin and its major metabolite 5-HIAA were down regulated in the brains of NCAM(-)(/-) mice. NCAM(-)(/-) mice also showed a dramatic reduction in the BDNF protein levels in the hippocampus and prefrontal cortex. This BDNF deficiency was associated with reduced phosphorylation of its receptor TrkB. Importantly, chronic administration of antidepressant amitriptyline partially or completely restored these changes in serotonergic and BDNF systems, respectively. In conclusion, NCAM deficiency lead to prominent and persistent abnormalities in brain serotonergic and BDNF systems, which likely contributes to the behavioral and neurobiological phenotype of NCAM(-/-) mice.
Collapse
Affiliation(s)
- Anu Aonurm-Helm
- Institute of Biomedicine and Translational Medicine, Department of Pharmacology, University of Tartu, 50411 Tartu, Estonia.
| | - Kaili Anier
- Institute of Biomedicine and Translational Medicine, Department of Pharmacology, University of Tartu, 50411 Tartu, Estonia
| | - Tamara Zharkovsky
- Institute of Biomedicine and Translational Medicine, Department of Pharmacology, University of Tartu, 50411 Tartu, Estonia
| | - Eero Castrén
- Neuroscience Center, University of Helsinki, P.O. Box 56, Helsinki, Finland
| | - Tomi Rantamäki
- Neuroscience Center, University of Helsinki, P.O. Box 56, Helsinki, Finland
| | | | - Jaak Järv
- Institute of Chemistry, University of Tartu, 50411 Tartu, Estonia
| | - Alexander Zharkovsky
- Institute of Biomedicine and Translational Medicine, Department of Pharmacology, University of Tartu, 50411 Tartu, Estonia
| |
Collapse
|
24
|
Definition of a Bidirectional Activity-Dependent Pathway Involving BDNF and Narp. Cell Rep 2015; 13:1747-56. [PMID: 26655895 DOI: 10.1016/j.celrep.2015.10.064] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/15/2015] [Accepted: 10/21/2015] [Indexed: 11/23/2022] Open
Abstract
One of the cardinal features of neural development and adult plasticity is the contribution of activity-dependent signaling pathways. However, the interrelationships between different activity-dependent genes are not well understood. The immediate early gene neuronal-activity-regulated pentraxin (NPTX2 or Narp) encodes a protein that has been associated with excitatory synaptogenesis, AMPA receptor aggregation, and the onset of critical periods. Here, we show that Narp is a direct transcriptional target of brain-derived neurotrophic factor (BDNF), another highly regulated activity-dependent gene involved in synaptic plasticity. Unexpectedly, Narp is bidirectionally regulated by BDNF. Acute BDNF withdrawal results in downregulation of Narp, whereas transcription of Narp is greatly enhanced by BDNF. Furthermore, our results show that BDNF directly regulates Narp to mediate glutamatergic transmission and mossy fiber plasticity. Hence, Narp serves as a significant epistatic target of BDNF to regulate synaptic plasticity during periods of dynamic activity.
Collapse
|
25
|
Wiera G, Mozrzymas JW. Extracellular proteolysis in structural and functional plasticity of mossy fiber synapses in hippocampus. Front Cell Neurosci 2015; 9:427. [PMID: 26582976 PMCID: PMC4631828 DOI: 10.3389/fncel.2015.00427] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/09/2015] [Indexed: 02/04/2023] Open
Abstract
Brain is continuously altered in response to experience and environmental changes. One of the underlying mechanisms is synaptic plasticity, which is manifested by modification of synapse structure and function. It is becoming clear that regulated extracellular proteolysis plays a pivotal role in the structural and functional remodeling of synapses during brain development, learning and memory formation. Clearly, plasticity mechanisms may substantially differ between projections. Mossy fiber synapses onto CA3 pyramidal cells display several unique functional features, including pronounced short-term facilitation, a presynaptically expressed long-term potentiation (LTP) that is independent of NMDAR activation, and NMDA-dependent metaplasticity. Moreover, structural plasticity at mossy fiber synapses ranges from the reorganization of projection topology after hippocampus-dependent learning, through intrinsically different dynamic properties of synaptic boutons to pre- and postsynaptic structural changes accompanying LTP induction. Although concomitant functional and structural plasticity in this pathway strongly suggests a role of extracellular proteolysis, its impact only starts to be investigated in this projection. In the present report, we review the role of extracellular proteolysis in various aspects of synaptic plasticity in hippocampal mossy fiber synapses. A growing body of evidence demonstrates that among perisynaptic proteases, tissue plasminogen activator (tPA)/plasmin system, β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) and metalloproteinases play a crucial role in shaping plastic changes in this projection. We discuss recent advances and emerging hypotheses on the roles of proteases in mechanisms underlying mossy fiber target specific synaptic plasticity and memory formation.
Collapse
Affiliation(s)
- Grzegorz Wiera
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University Wroclaw, Poland ; Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University Wroclaw, Poland
| | - Jerzy W Mozrzymas
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University Wroclaw, Poland ; Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University Wroclaw, Poland
| |
Collapse
|
26
|
Conant K, Allen M, Lim ST. Activity dependent CAM cleavage and neurotransmission. Front Cell Neurosci 2015; 9:305. [PMID: 26321910 PMCID: PMC4531370 DOI: 10.3389/fncel.2015.00305] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/27/2015] [Indexed: 12/13/2022] Open
Abstract
Spatially localized proteolysis represents an elegant means by which neuronal activity dependent changes in synaptic structure, and thus experience dependent learning and memory, can be achieved. In vitro and in vivo studies suggest that matrix metalloproteinase and adamalysin activity is concentrated at the cell surface, and emerging evidence suggests that increased peri-synaptic expression, release and/or activation of these proteinases occurs with enhanced excitatory neurotransmission. Synaptically expressed cell adhesion molecules (CAMs) could therefore represent important targets for neuronal activity-dependent proteolysis. Several CAM subtypes are expressed at the synapse, and their cleavage can influence the efficacy of synaptic transmission through a variety of non-mutually exclusive mechanisms. In the following review, we discuss mechanisms that regulate neuronal activity-dependent synaptic CAM shedding, including those that may be calcium dependent. We also highlight CAM targets of activity-dependent proteolysis including neuroligin and intercellular adhesion molecule-5 (ICAM-5). We include discussion focused on potential consequences of synaptic CAM shedding, with an emphasis on interactions between soluble CAM cleavage products and specific pre- and post-synaptic receptors.
Collapse
Affiliation(s)
- Katherine Conant
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center Washington, DC, USA
| | - Megan Allen
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center Washington, DC, USA
| | - Seung T Lim
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center Washington, DC, USA
| |
Collapse
|
27
|
Colley KJ, Kitajima K, Sato C. Polysialic acid: biosynthesis, novel functions and applications. Crit Rev Biochem Mol Biol 2014; 49:498-532. [PMID: 25373518 DOI: 10.3109/10409238.2014.976606] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
As an anti-adhesive, a reservoir for key biological molecules, and a modulator of signaling, polysialic acid (polySia) is critical for nervous system development and maintenance, promotes cancer metastasis, tissue regeneration and repair, and is implicated in psychiatric diseases. In this review, we focus on the biosynthesis and functions of mammalian polySia, and the use of polySia in therapeutic applications. PolySia modifies a small subset of mammalian glycoproteins, with the neural cell adhesion molecule, NCAM, serving as its major carrier. Studies show that mammalian polysialyltransferases employ a unique recognition mechanism to limit the addition of polySia to a select group of proteins. PolySia has long been considered an anti-adhesive molecule, and its impact on cell adhesion and signaling attributed directly to this property. However, recent studies have shown that polySia specifically binds neurotrophins, growth factors, and neurotransmitters and that this binding depends on chain length. This work highlights the importance of considering polySia quality and quantity, and not simply its presence or absence, as its various roles are explored. The capsular polySia of neuroinvasive bacteria allows these organisms to evade the host immune response. While this "stealth" characteristic has made meningitis vaccine development difficult, it has also made polySia a worthy replacement for polyetheylene glycol in the generation of therapeutic proteins with low immunogenicity and improved circulating half-lives. Bacterial polysialyltransferases are more promiscuous than the protein-specific mammalian enzymes, and new studies suggest that these enzymes have tremendous therapeutic potential, especially for strategies aimed at neural regeneration and tissue repair.
Collapse
Affiliation(s)
- Karen J Colley
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago , Chicago, IL , USA and
| | | | | |
Collapse
|
28
|
Podestá MF, Yam P, Codagnone MG, Uccelli NA, Colman D, Reinés A. Distinctive PSA-NCAM and NCAM hallmarks in glutamate-induced dendritic atrophy and synaptic disassembly. PLoS One 2014; 9:e108921. [PMID: 25279838 PMCID: PMC4184824 DOI: 10.1371/journal.pone.0108921] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 09/05/2014] [Indexed: 12/13/2022] Open
Abstract
Dendritic and synapse remodeling are forms of structural plasticity that play a critical role in normal hippocampal function. Neural cell adhesion molecule (NCAM) and its polysialylated form (PSA-NCAM) participate in neurite outgrowth and synapse formation and plasticity. However, it remains unclear whether they contribute to dendritic retraction and synaptic disassembly. Cultured hippocampal neurons exposed to glutamate (5 µM) showed a reduced MAP-2 (+) area in the absence of neuronal death 24 h after the insult. Concomitantly, synapse loss, revealed by decreased synaptophysin and post-synaptic density-95 cluster number and area, together with changes in NCAM and PSA-NCAM levels were found. Dendritic atrophy and PSA-NCAM reduction proved NMDA-receptor dependent. Live-imaging experiments evidenced dendritic atrophy 4 h after the insult; this effect was preceded by smaller NCAM clusters (1 h) and decreased surface and total PSA-NCAM levels (3 h). Simultaneously, total NCAM cluster number and area remained unchanged. The subsequent synapse disassembly (6 h) was accompanied by reductions in total NCAM cluster number and area. A PSA mimetic peptide prevented both the dendritic atrophy and the subsequent synaptic changes (6 h) but had no effect on the earliest synaptic remodeling (3 h). Thus, NCAM-synaptic reorganization and PSA-NCAM level decrease precede glutamate-induced dendritic atrophy, whereas the NCAM level reduction is a delayed event related to synapse loss. Consequently, distinctive stages in PSA-NCAM/NCAM balance seem to accompany glutamate-induced dendritic atrophy and synapse loss.
Collapse
Affiliation(s)
- María Fernanda Podestá
- Instituto de Investigaciones Farmacológicas (ININFA, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
- Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto de Biología Celular y Neurociencias “Prof. E. De Robertis” (IBCN, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Patricia Yam
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| | - Martín Gabriel Codagnone
- Instituto de Investigaciones Farmacológicas (ININFA, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
- Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto de Biología Celular y Neurociencias “Prof. E. De Robertis” (IBCN, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Nonthué Alejandra Uccelli
- Instituto de Investigaciones Farmacológicas (ININFA, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
- Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto de Biología Celular y Neurociencias “Prof. E. De Robertis” (IBCN, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
| | - David Colman
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| | - Analía Reinés
- Instituto de Investigaciones Farmacológicas (ININFA, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
- Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto de Biología Celular y Neurociencias “Prof. E. De Robertis” (IBCN, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
29
|
Beesley PW, Herrera-Molina R, Smalla KH, Seidenbecher C. The Neuroplastin adhesion molecules: key regulators of neuronal plasticity and synaptic function. J Neurochem 2014; 131:268-83. [PMID: 25040546 DOI: 10.1111/jnc.12816] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 07/02/2014] [Accepted: 07/03/2014] [Indexed: 01/21/2023]
Abstract
The Neuroplastins Np65 and Np55 are neuronal and synapse-enriched immunoglobulin superfamily molecules that play important roles in a number of key neuronal and synaptic functions including, for Np65, cell adhesion. In this review we focus on the physiological roles of the Neuroplastins in promoting neurite outgrowth, regulating the structure and function of both inhibitory and excitatory synapses in brain, and in neuronal and synaptic plasticity. We discuss the underlying molecular and cellular mechanisms by which the Neuroplastins exert their physiological effects and how these are dependent upon the structural features of Np65 and Np55, which enable them to bind to a diverse range of protein partners. In turn this enables the Neuroplastins to interact with a number of key neuronal signalling cascades. These include: binding to and activation of the fibroblast growth factor receptor; Np65 trans-homophilic binding leading to activation of p38 MAPK and internalization of glutamate (GluR1) receptor subunits; acting as accessory proteins for monocarboxylate transporters, thus affecting neuronal energy supply, and binding to GABAA α1, 2 and 5 subunits, thus regulating the composition and localization of GABAA receptors. An emerging theme is the role of the Neuroplastins in regulating the trafficking and subcellular localization of specific binding partners. We also discuss the involvement of Neuroplastins in a number of pathophysiological conditions, including ischaemia, schizophrenia and breast cancer and the role of a single nucleotide polymorphism in the human Neuroplastin (NPTN) gene locus in impairment of cortical development and cognitive functions. Neuroplastins are neuronal cell adhesion molecules, which induce neurite outgrowth and play important roles in synaptic maturation and plasticity. This review summarizes the functional implications of Neuroplastins for correct synaptic membrane protein localization, neuronal energy supply, expression of LTP and LTD, animal and human behaviour, and pathophysiology and disease. It focuses particularly on Neuroplastin binding partners and signalling mechanisms, and proposes perspectives for future research on these important immunoglobulin superfamily members.
Collapse
Affiliation(s)
- Philip W Beesley
- School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | | | | | | |
Collapse
|
30
|
Van Battum EY, Gunput RAF, Lemstra S, Groen EJN, Yu KL, Adolfs Y, Zhou Y, Hoogenraad CC, Yoshida Y, Schachner M, Akhmanova A, Pasterkamp RJ. The intracellular redox protein MICAL-1 regulates the development of hippocampal mossy fibre connections. Nat Commun 2014; 5:4317. [PMID: 25007825 DOI: 10.1038/ncomms5317] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 06/05/2014] [Indexed: 02/05/2023] Open
Abstract
Mical is a reduction-oxidation (redox) enzyme that functions as an unusual F-actin disassembly factor during Drosophila development. Although three Molecule interacting with CasL (MICAL) proteins exist in vertebrate species, their mechanism of action remains poorly defined and their role in vivo unknown. Here, we report that vertebrate MICAL-1 regulates the targeting of secretory vesicles containing immunoglobulin superfamily cell adhesion molecules (IgCAMs) to the neuronal growth cone membrane through its ability to control the actin cytoskeleton using redox chemistry, thereby maintaining appropriate IgCAM cell surface levels. This precise regulation of IgCAMs by MICAL-1 is essential for the lamina-specific targeting of mossy fibre axons onto CA3 pyramidal neurons in the developing mouse hippocampus in vivo. These findings reveal the first in vivo role for a vertebrate MICAL protein, expand the repertoire of cellular functions controlled through MICAL-mediated effects on the cytoskeleton, and provide insights into the poorly characterized mechanisms underlying neuronal protein cell surface expression and lamina-specific axonal targeting.
Collapse
Affiliation(s)
- Eljo Y Van Battum
- 1] Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands [2]
| | - Rou-Afza F Gunput
- 1] Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands [2] [3]
| | - Suzanne Lemstra
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Ewout J N Groen
- 1] Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands [2] Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3508 GA, Utrecht, The Netherlands
| | - Ka Lou Yu
- Cell Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Yeping Zhou
- 1] Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands [2]
| | - Casper C Hoogenraad
- Cell Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Yukata Yoshida
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Anna Akhmanova
- Cell Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
31
|
Brandewiede J, Stork O, Schachner M. NCAM deficiency in the mouse forebrain impairs innate and learned avoidance behaviours. GENES, BRAIN, AND BEHAVIOR 2014; 13:468-77. [PMID: 24751161 DOI: 10.1111/gbb.12138] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 01/04/2014] [Accepted: 04/16/2014] [Indexed: 02/05/2023]
Abstract
The neural cell adhesion molecule (NCAM) has been implicated in the development and plasticity of neural circuits and the control of hippocampus- and amygdala-dependent learning and behaviour. Previous studies in constitutive NCAM null mutants identified emotional behaviour deficits related to disturbances of hippocampal and amygdala functions. Here, we studied these behaviours in mice conditionally deficient in NCAM in the postmigratory forebrain neurons. We report deficits in both innate and learned avoidance behaviours, as observed in elevated plus maze and passive avoidance tasks. In contrast, general locomotor activity, trait anxiety or neophobia were unaffected by the mutation. Altered avoidance behaviour of the conditional NCAM mutants was associated with a deficit in serotonergic signalling, as indicated by their reduced responsiveness to (±)-8-hydroxy-2-(dipropylamino)-tetralin-induced hypothermia. Another serotonin-dependent behaviour, namely intermale aggression that is massively increased in constitutively NCAM-deficient mice, was not affected in the forebrain-specific mutants. Our data suggest that genetically or environmentally induced changes of NCAM expression in the late postnatal and mature forebrain determine avoidance behaviour and serotonin (5-HT)1A receptor signalling.
Collapse
Affiliation(s)
- J Brandewiede
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Universität Hamburg, Hamburg
| | - O Stork
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University
- Center for Behavioural Brain Sciences, Magdeburg, Germany
| | - M Schachner
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Universität Hamburg, Hamburg
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| |
Collapse
|
32
|
Li L, Cao J, Zhang S, Wang C, Wang J, Song G, Wang H, Zhang L. NCAM signaling mediates the effects of GDNF on chronic morphine-induced neuroadaptations. J Mol Neurosci 2014; 53:580-9. [PMID: 24399412 DOI: 10.1007/s12031-013-0224-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 12/25/2013] [Indexed: 10/25/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a potent neurotrophic factor for midbrain dopamine (DA) neurons, while the DA neurons in the ventral tegmental area (VTA) is a crucial part of the neural circuits associated with drug addiction. Recently, more and more evidence suggests that GDNF plays an important role in negatively regulating the neuroadaptations induced by chronic exposure to drugs, which was thought to be the neurobiological basis of drug addiction, but the underlying mechanism is still unknown. More recently, the neural cell adhesion molecule (NCAM), which plays an important role in the process of neural plasticity, has been identified as an alternative signaling receptor for GDNF. The purpose of this study was to investigate whether NCAM was involved in the effects of GDNF on the neuroadaptations induced by chronic morphine exposure. Immunostaining results showed that NCAM was widely expressed in the VTA of rats, including all the DA neurons. The results also showed that the phosphorylation of NCAM-associated FAK, but not the total NCAM, was upregulated by GDNF, and this upregulation was inhibited by pre-treatment with the NCAM function-blocking antibody. Moreover, pre-treatment with the antibody could antagonize the effect of GDNF on inhibiting the neuroadaptations induced by chronic morphine exposure, including the decreases of the number and length of neurites and the size of cell bodies of VTA dopamine neurons, as well as the increase of tyrosine hydroxylase in the VTA dopamine neurons. These results suggest that NCAM signaling is involved in the negative regulatory effects of GDNF on chronic morphine-induced neuroadaptations.
Collapse
Affiliation(s)
- Li Li
- Jiangsu Key Laboratory of Anesthesiology & Jiangsu Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical College, No. 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Beesley P, Kraus M, Parolaro N. The neuroplastins: multifunctional neuronal adhesion molecules--involvement in behaviour and disease. ADVANCES IN NEUROBIOLOGY 2014; 8:61-89. [PMID: 25300133 DOI: 10.1007/978-1-4614-8090-7_4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The neuroplastins np65 and np55 are neuronal and synapse-enriched immunoglobulin (Ig) superfamily cell adhesion molecules that contain 3 and 2 Ig domains, respectively. Np65 is neuron specific whereas np55 is expressed in many tissues. They are multifunctional proteins whose physiological roles are defined by the partner proteins they bind to and the signalling pathways they activate. The neuroplastins are implicated in activity-dependent long-term synaptic plasticity. Thus neuroplastin-specific antibodies and a recombinant peptide inhibit long-term potentiation in hippocampal neurones. This is mediated by activation of the p38MAP kinase signalling pathway, resulting in the downregulation of the surface expression of GluR1 receptors. Np65, but not np55, exhibits trans-homophilic binding. Both np65 and np55 induce neurite outgrowth and both activate the FGF receptor and associated downstream signalling pathways. Np65 binds to and colocalises with GABA(A) receptor subtypes and may play a role in anchoring them to specific synaptic and extrasynaptic sites. Most recently the neuroplastins have been shown to chaperone and support the monocarboxylate transporter MCT2 in transporting lactate across the neuronal plasma membrane. Thus the neuroplastins are multifunctional adhesion molecules which support neurite outgrowth, modulate long-term activity-dependent synaptic plasticity, regulate surface expression of GluR1 receptors, modulate GABA(A) receptor localisation, and play a key role in delivery of monocarboxylate energy substrates both to the synapse and to extrasynaptic sites. The diverse functions and range of signalling pathways activated by the neuroplastins suggest that they are important in modulating behaviour and in relation to human disease.
Collapse
|
34
|
Wakade CG, Mehta SH, Maeda M, Webb RC, Chiu FC. Axonal fasciculation and the role of polysialic acid-neural cell adhesion molecule in rat cortical neurons. J Neurosci Res 2013; 91:1408-18. [PMID: 23963795 DOI: 10.1002/jnr.23268] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 05/20/2013] [Accepted: 05/23/2013] [Indexed: 12/28/2022]
Abstract
Axonal fasciculation is a mechanism deployed by growing axons to reach their targets during development of the nervous system. Published data have suggested the involvement of neuronal cell adhesion molecules (NCAM) in axonal fasciculation. We have characterized the formation of axonal fascicles in serum-free, primary cultures of cortical neurons from embryonic rat brains. Unlike the published data, axonal fascicles in our system have a unique morphology: they are waveform, are rarely thicker than 20 μm, and can reach up to several millimeters in length. We observed an age and time dependence in the formation of fascicles. They formed only in cultures from embryonic day 15-17 brain and only between 4 days in vitro (DIV) and 11 DIV. Electron microscopy showed that the fascicles consisted of mostly axonal processes. Immunocytochemical staining confirmed that the fascicles were positive for the 66-kDa neurofilament protein, NF66, but they contained few, if any, microtubule-associated protein-2-positive or glial fibrillary acidic protein-positive processes. Polysialic acids appeared to be critical in the formation of fascicles. Neuraminidase treatment prevented the formation of fascicles when added before 5 DIV. Addition of a specific inhibitor blocked the effect of neuraminidase. The cortical neurons in our model shared several important features with axon fasciculation in vivo and may provide a unique system for studying the molecular mechanisms involved in the formation of axonal tracts in the brain.
Collapse
Affiliation(s)
- Chandramohan G Wakade
- Department of Physical Therapy, Georgia Regents University, Augusta, Georgia; Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, Georgia
| | | | | | | | | |
Collapse
|
35
|
Neural cell adhesion molecule-mediated Fyn activation promotes GABAergic synapse maturation in postnatal mouse cortex. J Neurosci 2013; 33:5957-68. [PMID: 23554477 DOI: 10.1523/jneurosci.1306-12.2013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
GABAergic basket interneurons form perisomatic synapses, which are essential for regulating neural networks, and their alterations are linked to various cognitive dysfunction. Maturation of basket synapses in postnatal cortex is activity dependent. In particular, activity-dependent downregulation of polysialiac acid carried by the neural cell adhesion molecule (NCAM) regulates the timing of their maturation. Whether and how NCAM per se affects GABAergic synapse development is unknown. Using single-cell genetics to knock out NCAM in individual basket interneurons in mouse cortical slice cultures, at specific developmental time periods, we found that NCAM loss during perisomatic synapse formation impairs the process of basket cell axonal branching and bouton formation. However, loss of NCAM once the synapses are already formed did not show any effect. We further show that NCAM120 and NCAM140, but not the NCAM180 isoform, rescue the phenotype. Finally, we demonstrate that a dominant-negative form of Fyn kinase mimics, whereas a constitutively active form of Fyn kinase rescues, the effects of NCAM knockdown. Altogether, our data suggest that NCAM120/NCAM140-mediated Fyn activation promotes GABAergic synapse maturation in postnatal cortex.
Collapse
|
36
|
Theis T, Mishra B, von der Ohe M, Loers G, Prondzynski M, Pless O, Blackshear PJ, Schachner M, Kleene R. Functional role of the interaction between polysialic acid and myristoylated alanine-rich C kinase substrate at the plasma membrane. J Biol Chem 2013; 288:6726-42. [PMID: 23329829 PMCID: PMC3585110 DOI: 10.1074/jbc.m112.444034] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Indexed: 12/14/2022] Open
Abstract
Polysialic acid (PSA) is a homopolymeric glycan that plays crucial roles in the developing and adult nervous system. So far only a few PSA-binding proteins have been identified. Here, we identify myristoylated alanine-rich C kinase substrate (MARCKS) as novel PSA binding partner. Binding assays showed a direct interaction between PSA and a peptide comprising the effector domain of MARCKS (MARCKS-ED). Co-immunoprecipitation of PSA-carrying neural cell adhesion molecule (PSA-NCAM) with MARCKS and co-immunostaining of MARCKS and PSA at the cell membrane of hippocampal neurons confirm the interaction between PSA and MARCKS. Co-localization and an intimate interaction of PSA and MARCKS at the cell surface was seen by confocal microscopy and fluorescence resonance energy transfer (FRET) analysis after the addition of fluorescently labeled PSA or PSA-NCAM to live CHO cells or hippocampal neurons expressing MARCKS as a fusion protein with green fluorescent protein (GFP). Cross-linking experiments showed that extracellularly applied PSA or PSA-NCAM and intracellularly expressed MARCKS-GFP are in close contact, suggesting that PSA and MARCKS interact with each other at the plasma membrane from opposite sides. Insertion of PSA and MARCKS-ED peptide into lipid bilayers from opposite sides alters the electric properties of the bilayer confirming the notion that PSA and the effector domain of MARCKS interact at and/or within the plane of the membrane. The MARCKS-ED peptide abolished PSA-induced enhancement of neurite outgrowth from cultured hippocampal neurons indicating an important functional role for the interaction between MARCKS and PSA in the developing and adult nervous system.
Collapse
Affiliation(s)
- Thomas Theis
- From the Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Bibhudatta Mishra
- From the Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maren von der Ohe
- From the Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Gabriele Loers
- From the Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | | | - Ole Pless
- European Screening Port GmbH, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Perry J. Blackshear
- the Departments of Medicine and Biochemistry, Duke University, Durham, North Carolina 27709
- Laboratory of Signal Transduction, NIEHS, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and
| | - Melitta Schachner
- From the Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou 515041, China
| | - Ralf Kleene
- From the Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
37
|
|
38
|
Efficient neuronal in vitro and in vivo differentiation after immunomagnetic purification of mESC derived neuronal precursors. Stem Cell Res 2012; 10:133-46. [PMID: 23237958 DOI: 10.1016/j.scr.2012.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 10/22/2012] [Accepted: 10/25/2012] [Indexed: 02/06/2023] Open
Abstract
The cellular heterogeneity that is generated during the differentiation of pluripotent stem cells into specific neural subpopulations represents a major obstacle for experimental and clinical progress. To address this problem we developed an optimized strategy for magnetic isolation of PSA-NCAM positive neuronal precursors from embryonic stem cells (ESCs) derived neuronal cultures. PSA-NCAM enrichment at an early step of the in vitro differentiation process increased the number of ES cell derived neurons and reduced cellular diversity. Gene expression analysis revealed that mainly genes involved in neuronal activity were over-represented after purification. In vitro derived PSA-NCAM(+) enriched precursors were characterized in vivo through grafting into the forebrain of adult mice. While unsorted control cells 40 days post graft gave rise to a mixed population composed of immature precursors, early postmitotic neurons and glial cells, PSA-NCAM(+) enriched cells differentiated predominantly into NeuN positive cells. Furthermore, PSA-NCAM enriched population showed efficient migration towards the olfactory bulb after transplantation into the rostral migratory stream of the forebrain neurogenic system. Thus, enrichment of neuronal precursors based on PSA-NCAM expression represents a general and straightforward approach to narrow cellular heterogeneity during neuronal differentiation of pluripotent cells.
Collapse
|
39
|
Singh R, Lakhanpal D, Kumar S, Sharma S, Kataria H, Kaur M, Kaur G. Late-onset intermittent fasting dietary restriction as a potential intervention to retard age-associated brain function impairments in male rats. AGE (DORDRECHT, NETHERLANDS) 2012; 34:917-33. [PMID: 21861096 PMCID: PMC3682068 DOI: 10.1007/s11357-011-9289-2] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 07/08/2011] [Indexed: 05/19/2023]
Abstract
Lifelong dietary restriction (DR) is known to have many potential beneficial effects on brain function as well as delaying the onset of neurological diseases. In the present investigation, the effect of late-onset short-term intermittent fasting dietary restriction (IF-DR) regimen was studied on motor coordination and cognitive ability of ageing male rats. These animals were further used to estimate protein carbonyl content and mitochondrial complex I-IV activity in different regions of brain and peripheral organs, and the degree of age-related impairment and reversion by late-onset short-term IF-DR was compared with their levels in 3-month-old young rats. The results of improvement in motor coordination by rotarod test and cognitive skills by Morris water maze in IF-DR rats were found to be positively correlated with the decline in the oxidative molecular damage to proteins and enhanced mitochondrial complex IV activity in different regions of ageing brain as well as peripheral organs. The work was further extended to study the expression of synaptic plasticity-related proteins, such as synaptophysin, calcineurin and CaM kinase II to explore the molecular basis of IF-DR regimen to improve cognitive function. These results suggest that even late-onset short-term IF-DR regimen have the potential to retard age-associated detrimental effects, such as cognitive and motor performance as well as oxidative molecular damage to proteins.
Collapse
Affiliation(s)
- Rumani Singh
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| | - Dinesh Lakhanpal
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| | - Sushil Kumar
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| | - Sandeep Sharma
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| | - Hardeep Kataria
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| | - Manpreet Kaur
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| | - Gurcharan Kaur
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| |
Collapse
|
40
|
Synaptic functions of invertebrate varicosities: what molecular mechanisms lie beneath. Neural Plast 2012; 2012:670821. [PMID: 22655209 PMCID: PMC3359714 DOI: 10.1155/2012/670821] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 02/27/2012] [Indexed: 11/26/2022] Open
Abstract
In mammalian brain, the cellular and molecular events occurring in both synapse formation and plasticity are difficult to study due to the large number of factors involved in these processes and because the contribution of each component is not well defined. Invertebrates, such as Drosophila, Aplysia, Helix, Lymnaea, and Helisoma, have proven to be useful models for studying synaptic assembly and elementary forms of learning. Simple nervous system, cellular accessibility, and genetic simplicity are some examples of the invertebrate advantages that allowed to improve our knowledge about evolutionary neuronal conserved mechanisms. In this paper, we present an overview of progresses that elucidates cellular and molecular mechanisms underlying synaptogenesis and synapse plasticity in invertebrate varicosities and their validation in vertebrates. In particular, the role of invertebrate synapsin in the formation of presynaptic terminals and the cell-to-cell interactions that induce specific structural and functional changes in their respective targets will be analyzed.
Collapse
|
41
|
Vogt J, Glumm R, Schlüter L, Schmitz D, Rost BR, Streu N, Rister B, Suman Bharathi B, Gagiannis D, Hildebrandt H, Weinhold B, Mühlenhoff M, Naumann T, Savaskan NE, Brauer AU, Reutter W, Heimrich B, Nitsch R, Horstkorte R. Homeostatic regulation of NCAM polysialylation is critical for correct synaptic targeting. Cell Mol Life Sci 2012; 69:1179-91. [PMID: 22068610 PMCID: PMC11115167 DOI: 10.1007/s00018-011-0868-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 10/05/2011] [Accepted: 10/18/2011] [Indexed: 02/05/2023]
Abstract
During development, axonal projections have a remarkable ability to innervate correct dendritic subcompartments of their target neurons and to form regular neuronal circuits. Altered axonal targeting with formation of synapses on inappropriate neurons may result in neurodevelopmental sequelae, leading to psychiatric disorders. Here we show that altering the expression level of the polysialic acid moiety, which is a developmentally regulated, posttranslational modification of the neural cell adhesion molecule NCAM, critically affects correct circuit formation. Using a chemically modified sialic acid precursor (N-propyl-D: -mannosamine), we inhibited the polysialyltransferase ST8SiaII, the principal enzyme involved in polysialylation during development, at selected developmental time-points. This treatment altered NCAM polysialylation while NCAM expression was not affected. Altered polysialylation resulted in an aberrant mossy fiber projection that formed glutamatergic terminals on pyramidal neurons of the CA1 region in organotypic slice cultures and in vivo. Electrophysiological recordings revealed that the ectopic terminals on CA1 pyramids were functional and displayed characteristics of mossy fiber synapses. Moreover, ultrastructural examination indicated a "mossy fiber synapse"-like morphology. We thus conclude that homeostatic regulation of the amount of synthesized polysialic acid at specific developmental stages is essential for correct synaptic targeting and circuit formation during hippocampal development.
Collapse
Affiliation(s)
- Johannes Vogt
- Institute for Microanatomy and Neurobiology, University Medical Center of the J. Gutenberg, University Mainz, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Brennaman LH, Zhang X, Guan H, Triplett JW, Brown A, Demyanenko GP, Manis PB, Landmesser L, Maness PF. Polysialylated NCAM and ephrinA/EphA regulate synaptic development of GABAergic interneurons in prefrontal cortex. ACTA ACUST UNITED AC 2012; 23:162-77. [PMID: 22275477 DOI: 10.1093/cercor/bhr392] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A novel function for the neural cell adhesion molecule (NCAM) was identified in ephrinA/EphA-mediated repulsion as an important regulatory mechanism for development of GABAergic inhibitory synaptic connections in mouse prefrontal cortex. Deletion of NCAM, EphA3, or ephrinA2/3/5 in null mutant mice increased the numbers and size of perisomatic synapses between GABAergic basket interneurons and pyramidal cells in the developing cingulate cortex (layers II/III). A functional consequence of NCAM loss was increased amplitudes and faster kinetics of miniature inhibitory postsynaptic currents in NCAM null cingulate cortex. NCAM and EphA3 formed a molecular complex and colocalized with the inhibitory presynaptic marker vesicular GABA transporter (VGAT) in perisomatic puncta and neuropil in the cingulate cortex. EphrinA5 treatment promoted axon remodeling of enhanced green fluorescent protein-labeled basket interneurons in cortical slice cultures and induced growth cone collapse in wild-type but not NCAM null mutant neurons. NCAM modified with polysialic acid (PSA) was required to promote ephrinA5-induced axon remodeling of basket interneurons in cortical slices, likely by providing a permissive environment for ephrinA5/EphA3 signaling. These results reveal a new mechanism in which NCAM and ephrinAs/EphA3 coordinate to constrain GABAergic interneuronal arborization and perisomatic innervation, potentially contributing to excitatory/inhibitory balance in prefrontal cortical circuitry.
Collapse
Affiliation(s)
- Leann H Brennaman
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Shiosaka S, Ishikawa Y. Neuropsin—A possible modulator of synaptic plasticity. J Chem Neuroanat 2011; 42:24-9. [DOI: 10.1016/j.jchemneu.2011.05.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 05/30/2011] [Accepted: 05/30/2011] [Indexed: 01/20/2023]
|
44
|
Shichi K, Fujita-Hamabe W, Harada S, Mizoguchi H, Yamada K, Nabeshima T, Tokuyama S. Involvement of matrix metalloproteinase-mediated proteolysis of neural cell adhesion molecule in the development of cerebral ischemic neuronal damage. J Pharmacol Exp Ther 2011; 338:701-10. [PMID: 21602423 DOI: 10.1124/jpet.110.178079] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Neural cell adhesion molecule (NCAM) is a membrane protein abundantly expressed in the central nervous system. Recently, it has been reported that dysfunction of NCAM is linked to human brain disorders. Furthermore, NCAM is one of the proteolysis targets of matrix metalloproteinase (MMP), whose activation is implicated in neuronal damage. The aim of this study was to elucidate the involvement of MMP-mediated proteolysis of NCAM in the development of ischemic neuronal damage. Male ddY and MMP-9 knockout (KO) C57BL/6J mice were subjected to 2 h of middle cerebral artery occlusion (MCAO). In MCAO model mice, development of infarction and behavioral abnormality were clearly observed on days 1 and 3 after MCAO. Protein levels of MMP-2 and MMP-9 were significantly increased on days 1 and 3 after MCAO. In addition, full-length NCAM (180 kDa) was significantly decreased, but its metabolite levels increased on day 1 by ischemic stress per se. NCAM small interfering RNA significantly increased the neuronal damage induced by MCAO. MMP inhibition or MMP-9 gene KO attenuated the infarction, behavioral abnormalities, and decrease of NCAM (180 kDa) observed after MCAO in mice. The present findings clearly suggest that MMP-2/MMP-9-mediated NCAM proteolysis is implicated in the exacerbation of ischemic neuronal damage.
Collapse
Affiliation(s)
- Kanako Shichi
- Department of Clinical Pharmacy, Kobe Gakuin University School of Pharmaceutical Sciences, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan
| | | | | | | | | | | | | |
Collapse
|
45
|
Tereshchenko Y, Morellini F, Dityatev A, Schachner M, Irintchev A. Neural cell adhesion molecule ablation in mice causes hippocampal dysplasia and loss of septal cholinergic neurons. J Comp Neurol 2011; 519:2475-92. [DOI: 10.1002/cne.22636] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
46
|
Yang D, Lauridsen H, Buels K, Chi LH, La Du J, Bruun DA, Olson JR, Tanguay RL, Lein PJ. Chlorpyrifos-oxon disrupts zebrafish axonal growth and motor behavior. Toxicol Sci 2011; 121:146-59. [PMID: 21346248 DOI: 10.1093/toxsci/kfr028] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Axonal morphology is a critical determinant of neuronal connectivity, and perturbation of the rate or extent of axonal growth during development has been linked to neurobehavioral deficits in animal models and humans. We previously demonstrated that the organophosphorus pesticide (OP) chlorpyrifos (CPF) inhibits axonal growth in cultured neurons. In this study, we used a zebrafish model to determine whether CPF, its oxon metabolite (CPFO), or the excreted metabolite trichloro-2-pyridinol (TCPy) alter spatiotemporal patterns of axonal growth in vivo. Static waterborne exposure to CPFO, but not CPF or TCPy, at concentrations ≥ 0.03 μM from 24- to 72-h post fertilization significantly inhibited acetylcholinesterase, and high-performance liquid chromatography detected significantly more TCPy in zebrafish exposed to 0.1 μM CPFO versus 1.0 μM CPF. These data suggest that zebrafish lack the metabolic enzymes to activate CPF during these early developmental stages. Consistent with this, CPFO, but not CPF, significantly inhibited axonal growth of sensory neurons, primary motoneurons, and secondary motoneurons at concentrations ≥ 0.1 μM. Secondary motoneurons were the most sensitive to axonal growth inhibition by CPFO, which was observed at concentrations that did not cause mortality, gross developmental defects, or aberrant somatic muscle differentiation. CPFO effects on axonal growth correlated with adverse effects on touch-induced swimming behavior, suggesting the functional relevance of these structural changes. These data suggest that altered patterns of neuronal connectivity contribute to the developmental neurotoxicity of CPF and demonstrate the relevance of zebrafish as a model for studying OP developmental neurotoxicity.
Collapse
Affiliation(s)
- Dongren Yang
- Department of Molecular Biosciences, University of California Davis School of Veterinary Medicine, Davis, California 95616, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Synaptic junctions are generated by adhesion proteins that bridge the synaptic cleft to firmly anchor pre- and postsynaptic membranes. Several cell adhesion molecule (CAM) families localize to synapses, but it is not yet completely understood how each synaptic CAM family contributes to synapse formation and/or structure, and whether or how smaller groups of CAMs serve as minimal, functionally cooperative adhesive units upon which structure is based. Synapse structure and function evolve over the course of development, and in mature animals, synapses are composed of a greater number of proteins, surrounded by a stabilizing extracellular matrix, and often contacted by astrocytic processes. Thus, in mature networks undergoing plasticity, persistent changes in synapse strength, morphology, or number must be accompanied by selective and regulated remodeling of the neuropil. Recent work indicates that regulated, extracellular proteolysis may be essential for this, and rather than simply acting permissively to enable synapse plasticity, is more likely playing a proactive role in driving coordinated synaptic structural and functional modifications that underlie persistent changes in network activity.
Collapse
Affiliation(s)
- Deanna L Benson
- Fishberg Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, New York, New York 10029, USA.
| | | |
Collapse
|
48
|
Guiraudie-Capraz G, Chaillan FA, Truchet B, Franc JL, Mourre C, Roman FS. Increase in polysialyltransferase gene expression following LTP in adult rat dentate gyrus. Hippocampus 2010; 21:1180-9. [PMID: 20665595 DOI: 10.1002/hipo.20835] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2010] [Indexed: 11/10/2022]
Abstract
Neural cell adhesion molecule (NCAM) is frequently associated with polysialic acid (PSA), and its function is highly dependent on this polysialylation. PSA-NCAM plays an important role in synaptic plasticity in the hippocampus. STX and PST are the enzymes responsible for NCAM polysialylation. We investigated whether unilateral long-term potentiation (LTP) induction in vivo, in adult rat dentate gyrus (DG), triggered NCAM polysialylation by STX and PST produced in the hippocampus. We found that levels of STX and PST mRNA increased strongly since the early stage of hippocampal LTP and remained high during the maintenance of DG-LTP for 4 h. This rapid increase in polysialyltransferase gene expression occurred in both the hippocampi, probably resulting from bilateral LTP induction by strong unilateral HFS. Thus, LTP triggers interhemispheric molecular changes in the hippocampal network. This study is the first to describe the effects of LTP induction and maintenance on polysialyl-transferases in vivo. Our findings suggest that hippocampal synaptic remodeling requires NCAM polysialylation.
Collapse
Affiliation(s)
- G Guiraudie-Capraz
- Laboratoire Neurobiologie des Processus Mnésiques, Marseille Cedex, France.
| | | | | | | | | | | |
Collapse
|
49
|
Role of the neural cell adhesion molecule (NCAM) in amygdalo-hippocampal interactions and salience determination of contextual fear memory. Int J Neuropsychopharmacol 2010; 13:661-74. [PMID: 20003620 DOI: 10.1017/s1461145709991106] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Evidence suggests that the neural cell adhesion molecule (NCAM) is an important molecular constituent of adaptive and maladaptive circuit (re-)organization in the central nervous system. Here, we further investigate its putative involvement in amygdala and hippocampus functions during context fear memory formation. Using laser capture microdissection and quantitative RT-PCR, we show high NCAM mRNA expression levels in the lateral and basolateral subnuclei of the amygdala, as well as their training intensity- and context-dependent regulation during fear memory consolidation. Moreover, we demonstrate that deficits of NCAM-/- mice in context fear memory can be overcome through contextual pre-exposure, i.e. by reducing the modulatory influence of the amygdala on this hippocampus-dependent memory. On the contrary, NCAM-/- mice failed to increase contextual fear memory after salient overtraining, although they adequately increased their response to auditory-cued fear stimuli. Finally, we demonstrate a reduction of amygdalo-hippocampal theta synchronization in NCAM-/- mice during fear memory retrieval. Together, these results suggest an involvement of NCAM-mediated cell recognition processes in information processing of the amygdalo-hippocampal system and in the amygdala-mediated modulation of context fear memory according to stimulus salience.
Collapse
|
50
|
Neuritogenic and neuroprotective properties of peptide agonists of the fibroblast growth factor receptor. Int J Mol Sci 2010; 11:2291-305. [PMID: 20640153 PMCID: PMC2904917 DOI: 10.3390/ijms11062291] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Accepted: 05/21/2010] [Indexed: 11/17/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) interact with their cognate ligands, FGFs, and with a number of cell adhesion molecules (CAMs), such as the neural cell adhesion molecule (NCAM), mediating a wide range of events during the development and maintenance of the nervous system. Determination of protein structure, in silico modeling and biological studies have recently resulted in the identification of FGFR binding peptides derived from various FGFs and NCAM mimicking the effects of these molecules with regard to their neuritogenic and neuroprotective properties. This review focuses on recently developed functional peptide agonists of FGFR with possible therapeutic potential.
Collapse
|