1
|
Yi W, Sylvester E, Lian J, Deng C. The effects of risperidone and voluntary exercise intervention on synaptic plasticity gene expressions in the hippocampus and prefrontal cortex of juvenile female rats. Physiol Behav 2025; 294:114879. [PMID: 40096936 DOI: 10.1016/j.physbeh.2025.114879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Psychiatric disorders and antipsychotics are associated with impaired neuroplasticity, while physical exercise has been reported to enhance neuroplasticity and improve cognitive and affective processes. Therefore, this study hypothesizes that voluntary exercise can enhance synaptic plasticity in juvenile rats disrupted by risperidone, a commonly prescribed antipsychotic for pediatric patients. METHODS Thirty-two juvenile female rats were randomly assigned to Vehicle+Sedentary, Risperidone (0.9mg/kg; b.i.d)+Sedentary, Vehicle+Exercise (three hours daily access to running wheels), and Risperidone+Exercise groups for four week treatment. Brains were collected for further analysis. RESULTS In the hippocampus, the mRNA expressions of Bdnf, Ntrk2, and Grin2b were increased by risperidone and exercise intervention. Exercise upregulated expression of Grin1 and Grin2a. Syn1 and Syp mRNA expression were enhanced by exercise in the risperidone-treated group. The expression of both Mfn1 and Drp1 mRNA were decreased by risperidone-only treatment. In the prefrontal cortex, Bdnf and Dlg4 expression was upregulated by exercise, while the Ntrk2 expression was reduced by risperidone and reversed by exercise. The Mfn1 mRNA expression was decreased by risperidone with or without voluntary exercise. The risperidone-decreased Ppargc1α gene expression was enhanced by exercise. CONCLUSION Risperidone affects synaptic plasticity through a complex mechanism in female juvenile rats: enhancing certain key genes in the hippocampus while inhibiting genes essential for mitochondrial function. In line with our hypothesis, voluntary exercise promotes genes beneficial for synaptic plasticity and enhances specific genes reduced by risperidone, in female juvenile rats.
Collapse
Affiliation(s)
- Weijie Yi
- School of Medical, Indigenous and Health Sciences, and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Emma Sylvester
- School of Medical, Indigenous and Health Sciences, and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Jiamei Lian
- School of Medical, Indigenous and Health Sciences, and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Chao Deng
- School of Medical, Indigenous and Health Sciences, and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia.
| |
Collapse
|
2
|
Banerjee TD, Zhang L, Monteiro A. Mapping Gene Expression in Whole Larval Brains of Bicyclus anynana Butterflies. Methods Protoc 2025; 8:31. [PMID: 40126249 PMCID: PMC11932290 DOI: 10.3390/mps8020031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
Butterfly larvae display intricate cognitive capacities and behaviors, but relatively little is known about how those behaviors alter their brains at the molecular level. Here, we optimized a hybridization chain reaction 3.0 (HCR v3.0) protocol to visualize the expression of multiple RNA molecules in fixed larval brains of the African butterfly Bicyclus anynana. We optimized the polyacrylamide gel mounting, fixation, and sample permeabilization steps, and mapped the expression domains of ten genes in whole larval brain tissue at single-cell resolution. The genes included optomotor blind (omb), yellow-like, zinc finger protein SNAI2-like (SNAI2), weary (wry), extradenticle (exd), Synapsin, Distal-less (Dll), bric-à-brac 1 (bab1), dachshund (dac), and acetyl coenzyme A acetyltransferase B (AcatB). This method can be used alongside single-cell sequencing to visualize the spatial location of brain cells that change in gene expression or splicing patterns in response to specific behaviors or cognitive experiences.
Collapse
Affiliation(s)
| | | | - Antónia Monteiro
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore;
| |
Collapse
|
3
|
Melrose J. Glycosaminoglycans, Instructive Biomolecules That Regulate Cellular Activity and Synaptic Neuronal Control of Specific Tissue Functional Properties. Int J Mol Sci 2025; 26:2554. [PMID: 40141196 PMCID: PMC11942259 DOI: 10.3390/ijms26062554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Glycosaminoglycans (GAGs) are a diverse family of ancient biomolecules that evolved over millennia as key components in the glycocalyx that surrounds all cells. GAGs have molecular recognition and cell instructive properties when attached to cell surface and extracellular matrix (ECM) proteoglycans (PGs), which act as effector molecules that regulate cellular behavior. The perception of mechanical cues which arise from perturbations in the ECM microenvironment allow the cell to undertake appropriate biosynthetic responses to maintain ECM composition and tissue function. ECM PGs substituted with GAGs provide structural support to weight-bearing tissues and an ability to withstand shear forces in some tissue contexts. This review outlines the structural complexity of GAGs and the diverse functional properties they convey to cellular and ECM PGs. PGs have important roles in cartilaginous weight-bearing tissues and fibrocartilages subject to tension and high shear forces and also have important roles in vascular and neural tissues. Specific PGs have roles in synaptic stabilization and convey specificity and plasticity in the regulation of neurophysiological responses in the CNS/PNS that control tissue function. A better understanding of GAG instructional roles over cellular behavior may be insightful for the development of GAG-based biotherapeutics designed to treat tissue dysfunction in disease processes and in novel tissue repair strategies following trauma. GAGs have a significant level of sophistication over the control of cellular behavior in many tissue contexts, which needs to be fully deciphered in order to achieve a useful therapeutic product. GAG biotherapeutics offers exciting opportunities in the modern glycomics arena.
Collapse
Affiliation(s)
- James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia;
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- Sydney Medical School, Northern, University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| |
Collapse
|
4
|
Lee VY, Nils AVM, Arruda BP, Xavier GF, Nogueira MI, Motta-Teixeira LC, Takada SH. Spontaneous running wheel exercise during pregnancy prevents later neonatal-anoxia-induced somatic and neurodevelopmental alterations. IBRO Neurosci Rep 2024; 17:263-279. [PMID: 39310269 PMCID: PMC11414703 DOI: 10.1016/j.ibneur.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction About 15-20 % of babies that suffer perinatal asphyxia die and around 25 % of the survivors exhibit permanent neural outcomes. Minimization of this global health problem has been warranted. This study investigated if the offspring of pregnant female rats allowed to spontaneously exercise on running wheels along a 11-day pregnancy period were protected for somatic and neurodevelopmental disturbs that usually follow neonatal anoxia. Methods spontaneous exercise was applied to female rats which were housed in cages allowing free access to running wheels along a 11-day pregnancy period. Their offspring were submitted to anoxia 24-36 h after birth. Somatic and sensory-motor development of the pups were recorded until postnatal day 21 (P21). Myelin basic protein (MBP)-stained areas of sensory and motor cortices were measured at P21. Neuronal nuclei (NeuN)-immunopositive cells and synapsin-I levels in hippocampal formation were estimated at P21 and P75. Results gestational exercise and / or neonatal anoxia increased the weight and the size of the pups. In addition, gestational exercise accelerated somatic and sensory-motor development of the pups and protected them against neonatal-anoxia-induced delay in development. Further, neonatal anoxia reduced MBP stained area in the secondary motor cortex and decreased hippocampal neuronal estimates and synapsin-I levels at P21; gestational exercise prevented these effects. Therefore, spontaneous exercise along pregnancy is a valuable strategy to prevent neonatal-anoxia-induced disturbs in the offspring. Conclusion spontaneous gestational running wheel exercise protects against neonatal anoxia-induced disturbs in the offspring, including (1) physical and neurobehavioral developmental impairments, and (2) hippocampal and cortical changes. Thus, spontaneous exercise during pregnancy may represent a valuable strategy to prevent disturbs which usually follow neonatal anoxia.
Collapse
Affiliation(s)
- Vitor Yonamine Lee
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 2415, Sao Paulo, SP 05508-900, Brazil
| | - Aline Vilar Machado Nils
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, R. do Matão, Travessa 14, 101, Sao Paulo 05508-900, Brazil
| | - Bruna Petrucelli Arruda
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Alameda da Universidade, s/n, Bloco Delta, São Bernardo do Campo, SP 09606-070, Brazil
| | - Gilberto Fernando Xavier
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, R. do Matão, Travessa 14, 101, Sao Paulo 05508-900, Brazil
| | - Maria Inês Nogueira
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 2415, Sao Paulo, SP 05508-900, Brazil
| | - Lívia Clemente Motta-Teixeira
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 2415, Sao Paulo, SP 05508-900, Brazil
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, R. do Matão, Travessa 14, 101, Sao Paulo 05508-900, Brazil
- Departamento de Ciências Fisiológicas, Faculdade de Ciências Médicas da Santa Casa de São Paulo, R. Jaguaribe, 155 - Vila Buarque, Sao Paulo, SP 01224-001, Brazil
| | - Silvia Honda Takada
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 2415, Sao Paulo, SP 05508-900, Brazil
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Alameda da Universidade, s/n, Bloco Delta, São Bernardo do Campo, SP 09606-070, Brazil
| |
Collapse
|
5
|
Karlström V, Sagredo E, Planells J, Welinder C, Jungfleisch J, Barrera-Conde A, Engfors L, Daniel C, Gebauer F, Visa N, Öhman M. ADAR3 modulates neuronal differentiation and regulates mRNA stability and translation. Nucleic Acids Res 2024; 52:12021-12038. [PMID: 39217468 PMCID: PMC11514483 DOI: 10.1093/nar/gkae753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
ADAR3 is a catalytically inactive member of the family of adenosine deaminases acting on RNA (ADARs). Here we have investigated its function in the context of the developing mouse brain. The expression of ADAR3 gradually increases throughout embryogenesis and drops after birth. Using primary cortical neurons, we show that ADAR3 is only expressed in a subpopulation of in vitro differentiated neurons, which suggests specific functions rather than being a general regulator of ADAR editing in the brain. The analysis of the ADAR3 interactome suggested a role in mRNA stability and translation, and we show that expression of ADAR3 in a neuronal cell line that is otherwise ADAR3-negative changes the expression and stability of a large number of mRNAs. Notably, we show that ADAR3 associates with polysomes and inhibits translation. We propose that ADAR3 binds to target mRNAs and stabilizes them in non-productive polysome complexes. Interestingly, the expression of ADAR3 downregulates genes related to neuronal differentiation and inhibits neurofilament outgrowth in vitro. In summary, we propose that ADAR3 negatively regulates neuronal differentiation, and that it does so by regulating mRNA stability and translation in an editing-independent manner.
Collapse
Affiliation(s)
- Victor Karlström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius väg 20C, Stockholm SE-106 91, Sweden
| | - Eduardo A Sagredo
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius väg 20C, Stockholm SE-106 91, Sweden
| | - Jordi Planells
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius väg 20C, Stockholm SE-106 91, Sweden
| | - Charlotte Welinder
- Mass Spectrometry, Clinical Sciences, Lund University, Lund SE-221 84, Sweden
| | - Jennifer Jungfleisch
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, ES-08003 Barcelona, Spain
| | - Andrea Barrera-Conde
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, ES-08003 Barcelona, Spain
| | - Linus Engfors
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius väg 20C, Stockholm SE-106 91, Sweden
| | - Chammiran Daniel
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius väg 20C, Stockholm SE-106 91, Sweden
| | - Fátima Gebauer
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, ES-08003 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), ES-08003 Barcelona, Spain
| | - Neus Visa
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius väg 20C, Stockholm SE-106 91, Sweden
| | - Marie Öhman
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius väg 20C, Stockholm SE-106 91, Sweden
| |
Collapse
|
6
|
Kim SH, Lee B, Lee SM, Kim Y. Restoring social deficits in IRSp53-deleted mice: chemogenetic inhibition of ventral dentate gyrus Emx1-expressing cells. Transl Psychiatry 2024; 14:425. [PMID: 39375329 PMCID: PMC11458854 DOI: 10.1038/s41398-024-03104-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/09/2024] Open
Abstract
IRSp53 is a synaptic scaffold protein reported to be involved in schizophrenia, autism spectrum disorders, and social deficits in knockout mice. Identifying critical brain regions and cells related to IRSp53 deletion is expected to be of great help in the treatment of psychiatric problems. In this study, we performed chemogenetic inhibition within the ventral dentate gyrus (vDG) of mice with IRSp53 deletion in Emx1-expressing cells (Emx1-Cre;IRSp53 flox/flox). We observed the recovery of social deficits after chemogenetic inhibition within vDG of Emx1-Cre;IRSp53 flox/flox mice. Additionally, chemogenetic activation induced social deficits in Emx1-Cre mice. CRHR1 expression increased in the hippocampus of Emx1-Cre;IRSp53 flox/flox mice, and CRHR1 was reduced by chemogenetic inhibition. Htd2, Ccn1, and Atp61l were decreased in bulk RNA sequencing, and Eya1 and Ecrg4 were decreased in single-cell RNA sequencing of the hippocampus in Emx1-Cre;IRSp53 flox/flox mice compared to control mice. This study determined that the vDG is a critical brain region for social deficits caused by IRSp53 deletion. Social deficits in Emx1-Cre;IRSp53 flox/flox mice were recovered through chemogenetic inhibition, providing clues for new treatment methods for psychiatric disorders accompanied by social deficits.
Collapse
Affiliation(s)
- Su Hyun Kim
- Mental Health Research Institute, National Center for Mental Health, Seoul, South Korea
| | - Bomee Lee
- Mental Health Research Institute, National Center for Mental Health, Seoul, South Korea
| | - Seong Mi Lee
- Mental Health Research Institute, National Center for Mental Health, Seoul, South Korea
| | - Yangsik Kim
- Department of Psychiatry, Inha University Hospital, College of Medicine, Inha University, Incheon, South Korea.
| |
Collapse
|
7
|
Jones EJ, Skinner BM, Parker A, Baldwin LR, Greenman J, Carding SR, Funnell SGP. An in vitro multi-organ microphysiological system (MPS) to investigate the gut-to-brain translocation of neurotoxins. BIOMICROFLUIDICS 2024; 18:054105. [PMID: 39280192 PMCID: PMC11401645 DOI: 10.1063/5.0200459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024]
Abstract
The death of dopamine-producing neurons in the substantia nigra in the base of the brain is a defining pathological feature in the development of Parkinson's disease (PD). PD is, however, a multi-systemic disease, also affecting the peripheral nervous system and gastrointestinal tract (GIT) that interact via the gut-brain axis (GBA). Our dual-flow GIT-brain microphysiological system (MPS) was modified to investigate the gut-to-brain translocation of the neurotoxin trigger of PD, 1-methyl-4-phenylpyridinium (MPP+), and its impact on key GIT and brain cells that contribute to the GBA. The modular GIT-brain MPS in combination with quantitative and morphometric image analysis methods reproduces cell specific neurotoxin-induced dopaminergic cytotoxicity and mitochondria-toxicity with the drug having no detrimental impact on the viability or integrity of cellular membranes of GIT-derived colonic epithelial cells. Our findings demonstrate the utility and capability of the GIT-brain MPS for measuring neuronal responses and its suitability for identifying compounds or molecules produced in the GIT that can exacerbate or protect against neuronal inflammation and cell death.
Collapse
Affiliation(s)
- Emily J Jones
- Food, Microbiome and Health Research Programme, Quadram Institute, Norwich, United Kingdom
| | - Benjamin M Skinner
- School of Life Sciences, University of Essex, Colchester, United Kingdom
| | - Aimee Parker
- Food, Microbiome and Health Research Programme, Quadram Institute, Norwich, United Kingdom
| | - Lydia R Baldwin
- Centre of Biomedical Sciences, Hull York Medical School, University of Hull, Hull, United Kingdom
| | - John Greenman
- Centre of Biomedical Sciences, Hull York Medical School, University of Hull, Hull, United Kingdom
| | | | | |
Collapse
|
8
|
Falconieri A, Folino P, Da Palmata L, Raffa V. Nano-pulling stimulates axon regeneration in dorsal root ganglia by inducing stabilization of axonal microtubules and activation of local translation. Front Mol Neurosci 2024; 17:1340958. [PMID: 38633213 PMCID: PMC11022966 DOI: 10.3389/fnmol.2024.1340958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/11/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction Axonal plasticity is strongly related to neuronal development as well as regeneration. It was recently demonstrated that active mechanical tension, intended as an extrinsic factor, is a valid contribution to the modulation of axonal plasticity. Methods In previous publications, our team validated a the "nano-pulling" method used to apply mechanical forces to developing axons of isolated primary neurons using magnetic nanoparticles (MNP) actuated by static magnetic fields. This method was found to promote axon growth and synaptic maturation. Here, we explore the use of nano-pulling as an extrinsic factor to promote axon regeneration in a neuronal tissue explant. Results Whole dorsal root ganglia (DRG) were thus dissected from a mouse spinal cord, incubated with MNPs, and then stretched. We found that particles were able to penetrate the ganglion and thus become localised both in the somas and in sprouting axons. Our results highlight that nano-pulling doubles the regeneration rate, and this is accompanied by an increase in the arborizing capacity of axons, an accumulation of cellular organelles related to mass addition (endoplasmic reticulum and mitochondria) and pre-synaptic proteins with respect to spontaneous regeneration. In line with the previous results on isolated hippocampal neurons, we observed that this process is coupled to an increase in the density of stable microtubules and activation of local translation. Discussion Our data demonstrate that nano-pulling enhances axon regeneration in whole spinal ganglia exposed to MNPs and external magnetic fields. These preliminary data represent an encouraging starting point for proposing nano-pulling as a biophysical tool for the design of novel therapies based on the use of force as an extrinsic factor for promoting nerve regeneration.
Collapse
|
9
|
Melrose J. Keratan sulfate, an electrosensory neurosentient bioresponsive cell instructive glycosaminoglycan. Glycobiology 2024; 34:cwae014. [PMID: 38376199 PMCID: PMC10987296 DOI: 10.1093/glycob/cwae014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 02/21/2024] Open
Abstract
The roles of keratan sulfate (KS) as a proton detection glycosaminoglycan in neurosensory processes in the central and peripheral nervous systems is reviewed. The functional properties of the KS-proteoglycans aggrecan, phosphacan, podocalyxcin as components of perineuronal nets in neurosensory processes in neuronal plasticity, cognitive learning and memory are also discussed. KS-glycoconjugate neurosensory gels used in electrolocation in elasmobranch fish species and KS substituted mucin like conjugates in some tissue contexts in mammals need to be considered in sensory signalling. Parallels are drawn between KS's roles in elasmobranch fish neurosensory processes and its roles in mammalian electro mechanical transduction of acoustic liquid displacement signals in the cochlea by the tectorial membrane and stereocilia of sensory inner and outer hair cells into neural signals for sound interpretation. The sophisticated structural and functional proteins which maintain the unique high precision physical properties of stereocilia in the detection, transmittance and interpretation of acoustic signals in the hearing process are important. The maintenance of the material properties of stereocilia are essential in sound transmission processes. Specific, emerging roles for low sulfation KS in sensory bioregulation are contrasted with the properties of high charge density KS isoforms. Some speculations are made on how the molecular and electrical properties of KS may be of potential application in futuristic nanoelectronic, memristor technology in advanced ultrafast computing devices with low energy requirements in nanomachines, nanobots or molecular switches which could be potentially useful in artificial synapse development. Application of KS in such innovative areas in bioregulation are eagerly awaited.
Collapse
Affiliation(s)
- James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- Sydney Medical School, Northern, University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| |
Collapse
|
10
|
Jin X, Cheng H, Chen X, Cao X, Xiao C, Ding F, Qu H, Wang PG, Feng Y, Yang GY. A modular chemoenzymatic cascade strategy for the structure-customized assembly of ganglioside analogs. Commun Chem 2024; 7:17. [PMID: 38238524 PMCID: PMC10796935 DOI: 10.1038/s42004-024-01102-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 01/08/2024] [Indexed: 01/22/2024] Open
Abstract
Gangliosides play vital biological regulatory roles and are associated with neurological system diseases, malignancies, and immune deficiencies. They have received extensive attention in developing targeted drugs and diagnostic markers. However, it is difficult to obtain enough structurally defined gangliosides and analogs especially at an industrial-relevant scale, which prevent exploring structure-activity relationships and identifying drug ingredients. Here, we report a highly modular chemoenzymatic cascade assembly (MOCECA) strategy for customized and large-scale synthesis of ganglioside analogs with various glycan and ceramide epitopes. We typically accessed five gangliosides with therapeutic promising and systematically prepared ten GM1 analogs with diverse ceramides. Through further process amplification, we achieved industrial production of ganglioside GM1 in the form of modular assembly at hectogram scale. Using MOCECA-synthesized GM1 analogs, we found unique ceramide modifications on GM1 could enhance the ability to promote neurite outgrowth. By comparing the structures with synthetic analogs, we further resolved the problem of contradicting descriptions for GM1 components in different pharmaceutical documents by reinterpreting the exact two-component structures of commercialized GM1 drugs. Because of its applicability and stability, the MOCECA strategy can be extended to prepare other glycosphingolipid structures, which may pave the way for developing new glycolipid drugs.
Collapse
Affiliation(s)
- Xuefeng Jin
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Department of Clinical Pharmaceutics, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Hanchao Cheng
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, China
- Department of Pharmacology, Key University Laboratory of Metabolism and Health of Guangdong, School of Medicine, Southern University of Science and Technology, Guangdong, China
| | - Xiaohui Chen
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xuefeng Cao
- Glycogene LLC, 10th Floor, Building 3, Wuhan Precision Medicine Industrial Base, East Lake New Technology Development Zone, Wuhan, China
| | - Cong Xiao
- Glycogene LLC, 10th Floor, Building 3, Wuhan Precision Medicine Industrial Base, East Lake New Technology Development Zone, Wuhan, China
| | - Fengling Ding
- Glycogene LLC, 10th Floor, Building 3, Wuhan Precision Medicine Industrial Base, East Lake New Technology Development Zone, Wuhan, China
| | - Huirong Qu
- Glycogene LLC, 10th Floor, Building 3, Wuhan Precision Medicine Industrial Base, East Lake New Technology Development Zone, Wuhan, China
| | - Peng George Wang
- Department of Pharmacology, Key University Laboratory of Metabolism and Health of Guangdong, School of Medicine, Southern University of Science and Technology, Guangdong, China
| | - Yan Feng
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Guang-Yu Yang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
11
|
Yang AJT, Mohammad A, Finch MS, Tsiani E, Spencer G, Necakov A, MacPherson REK. Influence of metabolic stress and metformin on synaptic protein profile in SH-SY5Y-derived neurons. Physiol Rep 2023; 11:10.14814/phy2.15852. [PMID: 38010200 PMCID: PMC10680579 DOI: 10.14814/phy2.15852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 11/29/2023] Open
Abstract
Insulin resistance (IR) is associated with reductions in neuronal proteins often observed with Alzheimer's disease (AD), however, the mechanisms through which IR promotes neurodegeneration/AD pathogenesis are poorly understood. Metformin (MET), a potent activator of the metabolic regulator AMPK is used to treat IR but its effectiveness for AD is unclear. We have previously shown that chronic AMPK activation impairs neurite growth and protein synthesis in SH-SY5Y neurons, however, AMPK activation in IR was not explored. Therefore, we examined the effects of MET-driven AMPK activation with and without IR. Retinoic acid-differentiated SH-SY5Y neurons were treated with: (1) Ctl: 24 h vehicle followed by 24 h Vehicle; (2) HI: 100 nM insulin (24 h HI followed by 24 h HI); or (3) MET: 24 h vehicle followed by 24 h 2 mM metformin; (4) HI/MET: 24 h 100 nM insulin followed by 24 h 100 nM INS+2 mM MET. INS and INS/MET groups saw impairments in markers of insulin signaling (Akt S473, mTOR S2448, p70s6k T389, and IRS-1S636) demonstrating IR was not recovered with MET treatment. All treatment groups showed reductions in neuronal markers (post-synaptic marker HOMER1 mRNA content and synapse marker synaptophysin protein content). INS and MET treatments showed a reduction in the content of the mature neuronal marker NeuN that was prevented by INS/MET. Similarly, increases in cell size/area, neurite length/area observed with INS and MET, were prevented with INS/MET. These findings indicate that IR and MET impair neuronal markers through distinct pathways and suggest that MET is ineffective in treating IR-driven impairments in neurons.
Collapse
Affiliation(s)
- Alex J. T. Yang
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt CatharinesOntarioCanada
| | - Ahmad Mohammad
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt CatharinesOntarioCanada
| | - Michael S. Finch
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt CatharinesOntarioCanada
| | - Evangelia Tsiani
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt CatharinesOntarioCanada
| | - Gaynor Spencer
- Department of Biological SciencesBrock UniversitySt CatharinesOntarioCanada
- Centre for NeuroscienceBrock UniversitySt. CatharinesOntarioCanada
| | - Aleksandar Necakov
- Department of Biological SciencesBrock UniversitySt CatharinesOntarioCanada
- Centre for NeuroscienceBrock UniversitySt. CatharinesOntarioCanada
| | - Rebecca E. K. MacPherson
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt CatharinesOntarioCanada
- Centre for NeuroscienceBrock UniversitySt. CatharinesOntarioCanada
| |
Collapse
|
12
|
Bünger I, Talucci I, Kreye J, Höltje M, Makridis KL, Foverskov Rasmussen H, van Hoof S, Cordero-Gomez C, Ullrich T, Sedlin E, Kreissner KO, Hoffmann C, Milovanovic D, Turko P, Paul F, Meckies J, Verlohren S, Henrich W, Chaoui R, Maric HM, Kaindl AM, Prüss H. Synapsin autoantibodies during pregnancy are associated with fetal abnormalities. Brain Behav Immun Health 2023; 33:100678. [PMID: 37692096 PMCID: PMC10483408 DOI: 10.1016/j.bbih.2023.100678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/12/2023] Open
Abstract
Anti-neuronal autoantibodies can be transplacentally transferred during pregnancy and may cause detrimental effects on fetal development. It is unclear whether autoantibodies against synapsin-I, one of the most abundant synaptic proteins, are associated with developmental abnormalities in humans. We recruited a cohort of 263 pregnant women and detected serum synapsin-I IgG autoantibodies in 13.3% using cell-based assays. Seropositivity was strongly associated with abnormalities of fetal development including structural defects, intrauterine growth retardation, amniotic fluid disorders and neuropsychiatric developmental diseases in previous children (odds ratios of 3-6.5). Autoantibodies reached the fetal circulation and were mainly of IgG1/IgG3 subclasses. They bound to conformational and linear synapsin-I epitopes, five distinct epitopes were identified using peptide microarrays. The findings indicate that synapsin-I autoantibodies may be clinically useful biomarkers or even directly participate in the disease process of neurodevelopmental disorders, thus being potentially amenable to antibody-targeting interventional strategies in the future.
Collapse
Affiliation(s)
- Isabel Bünger
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117, Berlin, Germany
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Ivan Talucci
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Jakob Kreye
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117, Berlin, Germany
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- Berlin Institute of Health (BIH), 10178, Berlin, Germany
- Department of Pediatric Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- Center for Chronically Sick Children, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Markus Höltje
- Institute of Integrative Neuroanatomy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Konstantin L. Makridis
- Department of Pediatric Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- Center for Chronically Sick Children, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- Institute of Cell Biology and Neurobiology, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Helle Foverskov Rasmussen
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117, Berlin, Germany
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Scott van Hoof
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117, Berlin, Germany
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - César Cordero-Gomez
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117, Berlin, Germany
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Tim Ullrich
- Department of Pediatric Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- Center for Chronically Sick Children, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Eva Sedlin
- Department of Pediatric Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- Center for Chronically Sick Children, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- Department of Neonatology, Helios Klinikum, Berlin-Buch, Germany
| | - Kai Oliver Kreissner
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Christian Hoffmann
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117, Berlin, Germany
| | - Dragomir Milovanovic
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117, Berlin, Germany
| | - Paul Turko
- Institute of Integrative Neuroanatomy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Friedemann Paul
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Jessica Meckies
- Gynecology Practice Frauenärztinnen am Schloß, 12163, Berlin, Germany
| | - Stefan Verlohren
- Department of Obstetrics, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Wolfgang Henrich
- Department of Obstetrics, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Rabih Chaoui
- Center for Prenatal Diagnosis and Human Genetics, 10719, Berlin, Germany
| | - Hans Michael Maric
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Angela M. Kaindl
- Department of Pediatric Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- Center for Chronically Sick Children, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- Institute of Cell Biology and Neurobiology, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Harald Prüss
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117, Berlin, Germany
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| |
Collapse
|
13
|
Kharlamova AS, Godovalova OS, Otlyga EG, Proshchina AE. Primary and secondary olfactory centres in human ontogeny. Neurosci Res 2023; 190:1-16. [PMID: 36521642 DOI: 10.1016/j.neures.2022.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/19/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
The olfactory centres are the evolutionary oldest and most conservative area of the telencephalon. Olfactory deficiencies are involved in a large spectrum of neurologic disorders and neurodegenerative diseases. The growing interest in human olfaction has been also been driven by COVID-19-induced transitional anosmia. Nevertheless, recent data on the human olfactory centres concerning normal histology and morphogenesis are rare. Published data in the field are mainly restricted to classic studies with non-uniform nomenclature and varied definitions of certain olfactory areas. While the olfactory system in model animals (rats, mice, and more rarely non-human primates) has been extensively investigated, the developmental timetable of olfactory centres in both human prenatal and postnatal ontogeny are poorly understood and unsystemised, which complicates the process of analysing human material, including medical researches. The main purpose of this review is to provide and discuss relevant morphological data on the normal ontogeny of the human olfactory centres, with a focus on the timetable of maturation and developmental cytoarchitecture, and with special reference to the definitions and terminology of certain olfactory areas.
Collapse
Affiliation(s)
- A S Kharlamova
- Avtsyn Research Institute of Human Morphology of FSBSI "Petrovsky National Research Centre of Surgery", Tsyurupy st., 3, 117418 Moscow, Russia.
| | - O S Godovalova
- Moscow Regional Research Institute of Obstetrics and Gynecology, Pokrovka St., 22A, 101000 Moscow, Russia
| | - E G Otlyga
- Avtsyn Research Institute of Human Morphology of FSBSI "Petrovsky National Research Centre of Surgery", Tsyurupy st., 3, 117418 Moscow, Russia
| | - A E Proshchina
- Avtsyn Research Institute of Human Morphology of FSBSI "Petrovsky National Research Centre of Surgery", Tsyurupy st., 3, 117418 Moscow, Russia
| |
Collapse
|
14
|
Schaefers C, Rothmiller S, Thiermann H, Rein T, Schmidt A. The Efficiency of Direct Maturation: the Comparison of Two hiPSC Differentiation Approaches into Motor Neurons. Stem Cells Int 2022; 2022:1320950. [PMID: 36530489 PMCID: PMC9757946 DOI: 10.1155/2022/1320950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 02/23/2025] Open
Abstract
Motor neurons (MNs) derived from human-induced pluripotent stem cells (hiPSC) hold great potential for the treatment of various motor neurodegenerative diseases as transplantations with a low-risk of rejection are made possible. There are many hiPSC differentiation protocols that pursue to imitate the multistep process of motor neurogenesis in vivo. However, these often apply viral vectors, feeder cells, or antibiotics to generate hiPSC and MNs, limiting their translational potential. In this study, a virus-, feeder-, and antibiotic-free method was used for reprogramming hiPSC, which were maintained in culture medium produced under clinical good manufacturing practice. Differentiation into MNs was performed with standardized, chemically defined, and antibiotic-free culture media. The identity of hiPSC, neuronal progenitors, and mature MNs was continuously verified by the detection of specific markers at the genetic and protein level via qRT-PCR, flow cytometry, Western Blot, and immunofluorescence. MNX1- and ChAT-positive motoneuronal progenitor cells were formed after neural induction via dual-SMAD inhibition and expansion. For maturation, an approach aiming to directly mature these progenitors was compared to an approach that included an additional differentiation step for further specification. Although both approaches generated mature MNs expressing characteristic postmitotic markers, the direct maturation approach appeared to be more efficient. These results provide new insights into the suitability of two standardized differentiation approaches for generating mature MNs, which might pave the way for future clinical applications.
Collapse
Affiliation(s)
- Catherine Schaefers
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80937 Munich, Germany
| | - Simone Rothmiller
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80937 Munich, Germany
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80937 Munich, Germany
| | - Theo Rein
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany
| | - Annette Schmidt
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80937 Munich, Germany
- Institute of Sport Science, University of the Bundeswehr Munich, Werner-Heisenberg-Weg 39, 85577 Neubiberg, Germany
| |
Collapse
|
15
|
Michetti C, Falace A, Benfenati F, Fassio A. Synaptic genes and neurodevelopmental disorders: From molecular mechanisms to developmental strategies of behavioral testing. Neurobiol Dis 2022; 173:105856. [PMID: 36070836 DOI: 10.1016/j.nbd.2022.105856] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 10/14/2022] Open
Abstract
Synaptopathies are a class of neurodevelopmental disorders caused by modification in genes coding for synaptic proteins. These proteins oversee the process of neurotransmission, mainly controlling the fusion and recycling of synaptic vesicles at the presynaptic terminal, the expression and localization of receptors at the postsynapse and the coupling between the pre- and the postsynaptic compartments. Murine models, with homozygous or heterozygous deletion for several synaptic genes or knock-in for specific pathogenic mutations, have been developed. They have proved to be extremely informative for understanding synaptic physiology, as well as for clarifying the patho-mechanisms leading to developmental delay, epilepsy and motor, cognitive and social impairments that are the most common clinical manifestations of neurodevelopmental disorders. However, the onset of these disorders emerges during infancy and adolescence while the behavioral phenotyping is often conducted in adult mice, missing important information about the impact of synaptic development and maturation on the manifestation of the behavioral phenotype. Here, we review the main achievements obtained by behavioral testing in murine models of synaptopathies and propose a battery of behavioral tests to improve classification, diagnosis and efficacy of potential therapeutic treatments. Our aim is to underlie the importance of studying behavioral development and better focusing on disease onset and phenotypes.
Collapse
Affiliation(s)
- Caterina Michetti
- Department of Experimental Medicine, University of Genoa, Genoa, Italy; Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, Genoa, Italy.
| | - Antonio Falace
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Children's Hospital A. Meyer-University of Florence, Florence, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Anna Fassio
- Department of Experimental Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
16
|
Liu R, Bai L, Liu M, Wang R, Wu Y, Li Q, Ba Y, Zhang H, Zhou G, Yu F, Huang H. Combined exposure of lead and high-fat diet enhanced cognitive decline via interacting with CREB-BDNF signaling in male rats. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 304:119200. [PMID: 35364187 DOI: 10.1016/j.envpol.2022.119200] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/03/2022] [Accepted: 03/21/2022] [Indexed: 06/14/2023]
Abstract
The health risks to populations induced by lead (Pb) and high-fat diets (HFD) have become a global public health problem. Pb and HFD often co-exist and are co-occurring risk factors for cognitive impairment. This study investigates effect of combined Pb and HFD on cognitive function, and explores the underlying mechanisms in terms of regulatory components of synaptic plasticity and insulin signaling pathway. We showed that the co-exposure of Pb and HFD further increased blood Pb levels, caused body weight loss and dyslipidemia. The results from Morris water maze (MWM) test and Nissl staining disclosed that Pb and HFD each contributed to cognitive deficits and neuronal damage and combined exposure enhanced this toxic injury. Pb and HFD decreased the levels of synapsin-1, GAP-43 and PSD-95 protein related to synaptic properties and SIRT1, NMDARs, phosphorylated CREB and BDNF related to synaptic plasticity regulatory, and these decreases was greater when combined exposure. Additionally, we revealed that Pb and HFD promoted IRS-1 phosphorylation and subsequently reduced downstream PI3K-Akt kinases phosphorylation in hippocampus and cortex of rats, and this process was aggravated when co-exposure. Collectively, our data suggested that combined exposure of Pb and HFD enhanced cognitive deficits, pointing to additive effects in rats than the individual stress effects related to multiple signaling pathways with CREB-BDNF signaling as the hub. This study emphasizes the need to evaluate the effects of mixed exposures on brain function in realistic environment and to better inform prevention of neurological disorders via modulating central pathway, such as CREB/BDNF signaling.
Collapse
Affiliation(s)
- Rundong Liu
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Lin Bai
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Mengchen Liu
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Ruike Wang
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yingying Wu
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Qiong Li
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yue Ba
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Huizhen Zhang
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Guoyu Zhou
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Fangfang Yu
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Hui Huang
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
17
|
McNaughton R, Huo Y, Li G, Ioschpe ADV, Yan L, Man HY, Zhang X. Regulatory Effects of Gradient Microtopographies on Synapse Formation and Neurite Growth in Hippocampal Neurons. JOURNAL OF MICROMECHANICS AND MICROENGINEERING : STRUCTURES, DEVICES, AND SYSTEMS 2022; 32:075005. [PMID: 35814808 PMCID: PMC9262107 DOI: 10.1088/1361-6439/ac73d7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Among approaches aiming toward functional nervous system restoration, those implementing microfabrication techniques allow the manufacture of platforms with distinct geometry where neurons can develop and be guided to form patterned connections in vitro. The interplay between neuronal development and the microenvironment, shaped by the physical limitations, remains largely unknown. Therefore, it is crucial to have an efficient way to quantify neuronal morphological changes induced by physical or contact guidance of the microenvironment. In this study, we first devise and assess a method to prepare anisotropic, gradient poly(dimethylsiloxane) micro-ridge/groove arrays featuring variable local pattern width. We then demonstrate the ability of this single substrate to simultaneously profile the morphologcial and synaptic connectivity changes of primary cultured hippocampal neurons reacting to variable physical conditons, throughout neurodevelopment, in vitro. The gradient microtopography enhanced adhesion within microgrooves, increasing soma density with decreasing pattern width. Decreasing pattern width also reduced dendritic arborization and increased preferential axon growth. Finally, decreasing pattern geometry inhibited presynaptic puncta architecture. Collectively, a method to examine structural development and connectivity in response to physical stimuli is established, and potentially provides insight into microfabricated geometries which promote neural regeneration and repair.
Collapse
Affiliation(s)
- Ryan McNaughton
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | - Yuda Huo
- Department of Biology, Boston University, Boston, MA, USA
| | - Guicai Li
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | | | - Lei Yan
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Xin Zhang
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
| |
Collapse
|
18
|
Coccurello R, Marrone MC, Maccarrone M. The Endocannabinoids-Microbiota Partnership in Gut-Brain Axis Homeostasis: Implications for Autism Spectrum Disorders. Front Pharmacol 2022; 13:869606. [PMID: 35721203 PMCID: PMC9204215 DOI: 10.3389/fphar.2022.869606] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/20/2022] [Indexed: 11/13/2022] Open
Abstract
The latest years have witnessed a growing interest towards the relationship between neuropsychiatric disease in children with autism spectrum disorders (ASD) and severe alterations in gut microbiota composition. In parallel, an increasing literature has focused the attention towards the association between derangement of the endocannabinoids machinery and some mechanisms and symptoms identified in ASD pathophysiology, such as alteration of neural development, immune system dysfunction, defective social interaction and stereotypic behavior. In this narrative review, we put together the vast ground of endocannabinoids and their partnership with gut microbiota, pursuing the hypothesis that the crosstalk between these two complex homeostatic systems (bioactive lipid mediators, receptors, biosynthetic and hydrolytic enzymes and the entire bacterial gut ecosystem, signaling molecules, metabolites and short chain fatty acids) may disclose new ideas and functional connections for the development of synergic treatments combining “gut-therapy,” nutritional intervention and pharmacological approaches. The two separate domains of the literature have been examined looking for all the plausible (and so far known) overlapping points, describing the mutual changes induced by acting either on the endocannabinoid system or on gut bacteria population and their relevance for the understanding of ASD pathophysiology. Both human pathology and symptoms relief in ASD subjects, as well as multiple ASD-like animal models, have been taken into consideration in order to provide evidence of the relevance of the endocannabinoids-microbiota crosstalk in this major neurodevelopmental disorder.
Collapse
Affiliation(s)
- Roberto Coccurello
- Institute for Complex Systems (ISC), National Council of Research (CNR), Rome, Italy
- European Center for Brain Research/Santa Lucia Foundation IRCCS, Rome, Italy
- *Correspondence: Roberto Coccurello, ; Mauro Maccarrone,
| | - Maria Cristina Marrone
- Ministry of University and Research, Mission Unity for Recovery and Resilience Plan, Rome, Italy
| | - Mauro Maccarrone
- European Center for Brain Research/Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Biotechnological and Applied Clinical and Sciences, University of L’Aquila, L’Aquila, Italy
- *Correspondence: Roberto Coccurello, ; Mauro Maccarrone,
| |
Collapse
|
19
|
Meng L, Zou L, Xiong M, Chen J, Zhang X, Yu T, Li Y, Liu C, Chen G, Wang Z, Ye K, Zhang Z. A synapsin Ⅰ cleavage fragment contributes to synaptic dysfunction in Alzheimer's disease. Aging Cell 2022; 21:e13619. [PMID: 35443102 PMCID: PMC9124304 DOI: 10.1111/acel.13619] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 02/24/2022] [Accepted: 04/04/2022] [Indexed: 12/02/2022] Open
Abstract
Synaptic dysfunction is a key feature of Alzheimer's disease (AD). However, the molecular mechanisms underlying synaptic dysfunction remain unclear. Here, we show that synapsin Ⅰ, one of the most important synaptic proteins, is fragmented by the cysteine proteinase asparagine endopeptidase (AEP). AEP cleaves synapsin at N82 in the brains of AD patients and generates the C‐terminal synapsin Ⅰ (83–705) fragment. This fragment is abnormally distributed in neurons and induces synaptic dysfunction. Overexpression of AEP in the hippocampus of wild‐type mice results in the production of the synapsin Ⅰ (83–705) fragment and induces synaptic dysfunction and cognitive deficits. Moreover, overexpression of the AEP‐generated synapsin Ⅰ (83–705) fragment in the hippocampus of tau P301S transgenic mice and wild‐type mice promotes synaptic dysfunction and cognitive deficits. These findings suggest a novel mechanism of synaptic dysfunction in AD.
Collapse
Affiliation(s)
- Lanxia Meng
- Department of Neurology Renmin Hospital of Wuhan University Wuhan China
| | - Li Zou
- Department of Neurology Renmin Hospital of Wuhan University Wuhan China
- Department of Neurology Zhongnan Hospital of Wuhan University Wuhan China
| | - Min Xiong
- Department of Neurology Renmin Hospital of Wuhan University Wuhan China
| | - Jiehui Chen
- Department of Neurology Renmin Hospital of Wuhan University Wuhan China
| | - Xingyu Zhang
- Department of Neurology Renmin Hospital of Wuhan University Wuhan China
| | - Ting Yu
- Department of Neurology Renmin Hospital of Wuhan University Wuhan China
| | - Yiming Li
- Department of Neurology Renmin Hospital of Wuhan University Wuhan China
| | - Congcong Liu
- Department of Neurology Renmin Hospital of Wuhan University Wuhan China
| | - Guiqin Chen
- Department of Neurology Renmin Hospital of Wuhan University Wuhan China
- Department of Pathology and Laboratory Medicine Emory University School of Medicine Atlanta Georgia USA
| | - Zhihao Wang
- Department of Neurology Renmin Hospital of Wuhan University Wuhan China
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine Emory University School of Medicine Atlanta Georgia USA
| | - Zhentao Zhang
- Department of Neurology Renmin Hospital of Wuhan University Wuhan China
| |
Collapse
|
20
|
Moschetta M, Ravasenga T, De Fusco A, Maragliano L, Aprile D, Orlando M, Sacchetti S, Casagrande S, Lignani G, Fassio A, Baldelli P, Benfenati F. Ca 2+ binding to synapsin I regulates resting Ca 2+ and recovery from synaptic depression in nerve terminals. Cell Mol Life Sci 2022; 79:600. [PMID: 36409372 PMCID: PMC9678998 DOI: 10.1007/s00018-022-04631-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/23/2022] [Accepted: 11/13/2022] [Indexed: 11/22/2022]
Abstract
Synapsin I (SynI) is a synaptic vesicle (SV)-associated phosphoprotein that modulates neurotransmission by controlling SV trafficking. The SynI C-domain contains a highly conserved ATP binding site mediating SynI oligomerization and SV clustering and an adjacent main Ca2+ binding site, whose physiological role is unexplored. Molecular dynamics simulations revealed that the E373K point mutation irreversibly deletes Ca2+ binding to SynI, still allowing ATP binding, but inducing a destabilization of the SynI oligomerization interface. Here, we analyzed the effects of this mutation on neurotransmitter release and short-term plasticity in excitatory and inhibitory synapses from primary hippocampal neurons. Patch-clamp recordings showed an increase in the frequency of miniature excitatory postsynaptic currents (EPSCs) that was totally occluded by exogenous Ca2+ chelators and associated with a constitutive increase in resting terminal Ca2+ concentrations. Evoked EPSC amplitude was also reduced, due to a decreased readily releasable pool (RRP) size. Moreover, in both excitatory and inhibitory synapses, we observed a marked impaired recovery from synaptic depression, associated with impaired RRP refilling and depletion of the recycling pool of SVs. Our study identifies SynI as a novel Ca2+ buffer in excitatory terminals. Blocking Ca2+ binding to SynI results in higher constitutive Ca2+ levels that increase the probability of spontaneous release and disperse SVs. This causes a decreased size of the RRP and an impaired recovery from depression due to the failure of SV reclustering after sustained high-frequency stimulation. The results indicate a physiological role of Ca2+ binding to SynI in the regulation of SV clustering and trafficking in nerve terminals.
Collapse
Affiliation(s)
- Matteo Moschetta
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy ,Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Tiziana Ravasenga
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy ,IRCCS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Antonio De Fusco
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy ,Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Luca Maragliano
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy ,Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Davide Aprile
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy ,Present Address: High-Definition Disease Modelling Lab, Campus IFOM-IEO, Milan, Italy
| | - Marta Orlando
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy ,Present Address: Charitè Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin Institute of Health, NeuroCure Cluster of Excellence, Berlin, Germany
| | - Silvio Sacchetti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Silvia Casagrande
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Gabriele Lignani
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy ,Present Address: Queens Square Institute of Neurology, University College London, London, UK
| | - Anna Fassio
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy ,IRCCS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Pietro Baldelli
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy ,IRCCS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy ,IRCCS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| |
Collapse
|
21
|
Rivera A, Suárez-Boomgaard D, Miguelez C, Valderrama-Carvajal A, Baufreton J, Shumilov K, Taupignon A, Gago B, Real MÁ. Dopamine D 4 Receptor Is a Regulator of Morphine-Induced Plasticity in the Rat Dorsal Striatum. Cells 2021; 11:31. [PMID: 35011592 PMCID: PMC8750869 DOI: 10.3390/cells11010031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 02/06/2023] Open
Abstract
Long-term exposition to morphine elicits structural and synaptic plasticity in reward-related regions of the brain, playing a critical role in addiction. However, morphine-induced neuroadaptations in the dorsal striatum have been poorly studied despite its key function in drug-related habit learning. Here, we show that prolonged treatment with morphine triggered the retraction of the dendritic arbor and the loss of dendritic spines in the dorsal striatal projection neurons (MSNs). In an attempt to extend previous findings, we also explored whether the dopamine D4 receptor (D4R) could modulate striatal morphine-induced plasticity. The combined treatment of morphine with the D4R agonist PD168,077 produced an expansion of the MSNs dendritic arbors and restored dendritic spine density. At the electrophysiological level, PD168,077 in combination with morphine altered the electrical properties of the MSNs and decreased their excitability. Finally, results from the sustantia nigra showed that PD168,077 counteracted morphine-induced upregulation of μ opioid receptors (MOR) in striatonigral projections and downregulation of G protein-gated inward rectifier K+ channels (GIRK1 and GIRK2) in dopaminergic cells. The present results highlight the key function of D4R modulating morphine-induced plasticity in the dorsal striatum. Thus, D4R could represent a valuable pharmacological target for the safety use of morphine in pain management.
Collapse
Affiliation(s)
- Alicia Rivera
- Facultad de Ciencias, Instituto de Investigación Biomédica, Universidad de Málaga, 29071 Málaga, Spain; (D.S.-B.); (A.V.-C.); (K.S.); (M.Á.R.)
| | - Diana Suárez-Boomgaard
- Facultad de Ciencias, Instituto de Investigación Biomédica, Universidad de Málaga, 29071 Málaga, Spain; (D.S.-B.); (A.V.-C.); (K.S.); (M.Á.R.)
| | - Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Alejandra Valderrama-Carvajal
- Facultad de Ciencias, Instituto de Investigación Biomédica, Universidad de Málaga, 29071 Málaga, Spain; (D.S.-B.); (A.V.-C.); (K.S.); (M.Á.R.)
| | - Jérôme Baufreton
- Institut des Maladies Neurodegeneratives, Université de Bordeaux, UMR 5293, 33000 Bordeaux, France; (J.B.); (A.T.)
- Institut des Maladies Neurodegeneratives, CNRS, UMR 5293, 33000 Bordeaux, France
| | - Kirill Shumilov
- Facultad de Ciencias, Instituto de Investigación Biomédica, Universidad de Málaga, 29071 Málaga, Spain; (D.S.-B.); (A.V.-C.); (K.S.); (M.Á.R.)
- School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Anne Taupignon
- Institut des Maladies Neurodegeneratives, Université de Bordeaux, UMR 5293, 33000 Bordeaux, France; (J.B.); (A.T.)
- Institut des Maladies Neurodegeneratives, CNRS, UMR 5293, 33000 Bordeaux, France
| | - Belén Gago
- Facultad de Medicina, Instituto de Investigación Biomédica, Universidad de Málaga, 29071 Málaga, Spain;
| | - M. Ángeles Real
- Facultad de Ciencias, Instituto de Investigación Biomédica, Universidad de Málaga, 29071 Málaga, Spain; (D.S.-B.); (A.V.-C.); (K.S.); (M.Á.R.)
| |
Collapse
|
22
|
Singh S, Singh TG, Rehni AK. An Insight into Molecular Mechanisms and Novel Therapeutic Approaches in Epileptogenesis. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 19:750-779. [PMID: 32914725 DOI: 10.2174/1871527319666200910153827] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 11/22/2022]
Abstract
Epilepsy is the second most common neurological disease with abnormal neural activity involving the activation of various intracellular signalling transduction mechanisms. The molecular and system biology mechanisms responsible for epileptogenesis are not well defined or understood. Neuroinflammation, neurodegeneration and Epigenetic modification elicit epileptogenesis. The excessive neuronal activities in the brain are associated with neurochemical changes underlying the deleterious consequences of excitotoxicity. The prolonged repetitive excessive neuronal activities extended to brain tissue injury by the activation of microglia regulating abnormal neuroglia remodelling and monocyte infiltration in response to brain lesions inducing axonal sprouting contributing to neurodegeneration. The alteration of various downstream transduction pathways resulted in intracellular stress responses associating endoplasmic reticulum, mitochondrial and lysosomal dysfunction, activation of nucleases, proteases mediated neuronal death. The recently novel pharmacological agents modulate various receptors like mTOR, COX-2, TRK, JAK-STAT, epigenetic modulators and neurosteroids are used for attenuation of epileptogenesis. Whereas the various molecular changes like the mutation of the cell surface, nuclear receptor and ion channels focusing on repetitive episodic seizures have been explored by preclinical and clinical studies. Despite effective pharmacotherapy for epilepsy, the inadequate understanding of precise mechanisms, drug resistance and therapeutic failure are the current fundamental problems in epilepsy. Therefore, the novel pharmacological approaches evaluated for efficacy on experimental models of epilepsy need to be identified and validated. In addition, we need to understand the downstream signalling pathways of new targets for the treatment of epilepsy. This review emphasizes on the current state of novel molecular targets as therapeutic approaches and future directions for the management of epileptogenesis. Novel pharmacological approaches and clinical exploration are essential to make new frontiers in curing epilepsy.
Collapse
Affiliation(s)
- Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | - Ashish Kumar Rehni
- Cerebral Vascular Disease Research Laboratories, Department of Neurology and Neuroscience Program, University of Miami School of Medicine, Miami, Florida 33101, United States
| |
Collapse
|
23
|
Adewole DO, Struzyna LA, Burrell JC, Harris JP, Nemes AD, Petrov D, Kraft RH, Chen HI, Serruya MD, Wolf JA, Cullen DK. Development of optically controlled "living electrodes" with long-projecting axon tracts for a synaptic brain-machine interface. SCIENCE ADVANCES 2021; 7:eaay5347. [PMID: 33523957 PMCID: PMC10670819 DOI: 10.1126/sciadv.aay5347] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 12/04/2020] [Indexed: 06/12/2023]
Abstract
For implantable neural interfaces, functional/clinical outcomes are challenged by limitations in specificity and stability of inorganic microelectrodes. A biological intermediary between microelectrical devices and the brain may improve specificity and longevity through (i) natural synaptic integration with deep neural circuitry, (ii) accessibility on the brain surface, and (iii) optogenetic manipulation for targeted, light-based readout/control. Accordingly, we have developed implantable "living electrodes," living cortical neurons, and axonal tracts protected within soft hydrogel cylinders, for optobiological monitoring/modulation of brain activity. Here, we demonstrate fabrication, rapid axonal outgrowth, reproducible cytoarchitecture, and simultaneous optical stimulation and recording of these tissue engineered constructs in vitro. We also present their transplantation, survival, integration, and optical recording in rat cortex as an in vivo proof of concept for this neural interface paradigm. The creation and characterization of these functional, optically controllable living electrodes are critical steps in developing a new class of optobiological tools for neural interfacing.
Collapse
Affiliation(s)
- Dayo O Adewole
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laura A Struzyna
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Justin C Burrell
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James P Harris
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| | - Ashley D Nemes
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| | - Dmitriy Petrov
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| | - Reuben H Kraft
- Computational Biomechanics Group, The Pennsylvania State University, University Park, PA 16802, USA
| | - H Isaac Chen
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| | - Mijail D Serruya
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Neurodelphus LLC, 3401 Grays Ferry Ave., Unit 6176, Philadelphia, PA 19146, USA
- Nuromo LLC, 405 Meadow Lane, Merion Station, PA 19066, USA
| | - John A Wolf
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| | - D Kacy Cullen
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
24
|
Synthesis, structural investigations, DFT studies, and neurotrophic activity of zinc complex with a multidentate ligand. MONATSHEFTE FUR CHEMIE 2020. [DOI: 10.1007/s00706-020-02696-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
25
|
Farouk FM, Ooi L, Law CSW, Yeong KY. Dual‐Target‐Directed Ligand Displaying Selective Butyrylcholinesterase Inhibitory and Neurite Promoting Activities as a Potential Therapeutic for Alzheimer's Disease. ChemistrySelect 2020. [DOI: 10.1002/slct.202001202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Fathima Manaal Farouk
- School of Science Monash University Malaysia Campus Jalan Lagoon Selatan, Bandar Sunway 47500 Selangor Malaysia
| | - Luyi Ooi
- School of Science Monash University Malaysia Campus Jalan Lagoon Selatan, Bandar Sunway 47500 Selangor Malaysia
| | - Christine Shing Wei Law
- School of Science Monash University Malaysia Campus Jalan Lagoon Selatan, Bandar Sunway 47500 Selangor Malaysia
| | - Keng Yoon Yeong
- School of Science Monash University Malaysia Campus Jalan Lagoon Selatan, Bandar Sunway 47500 Selangor Malaysia
- Tropical Medicine and Biology (TMB) multidisciplinary platform Monash University Malaysia 47500 Bandar Sunway Selangor Malaysia
| |
Collapse
|
26
|
Phenotypic and genotypic characterization of families with complex intellectual disability identified pathogenic genetic variations in known and novel disease genes. Sci Rep 2020; 10:968. [PMID: 31969655 PMCID: PMC6976666 DOI: 10.1038/s41598-020-57929-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/03/2020] [Indexed: 12/04/2022] Open
Abstract
Intellectual disability (ID), which presents itself during childhood, belongs to a group of neurodevelopmental disorders (NDDs) that are clinically widely heterogeneous and highly heritable, often being caused by single gene defects. Indeed, NDDs can be attributed to mutations at over 1000 loci, and all type of mutations, ranging from single nucleotide variations (SNVs) to large, complex copy number variations (CNVs), have been reported in patients with ID and other related NDDs. In this study, we recruited seven different recessive NDD families with comorbidities to perform a detailed clinical characterization and a complete genomic analysis that consisted of a combination of high throughput SNP-based genotyping and whole-genome sequencing (WGS). Different disease-associated loci and pathogenic gene mutations were identified in each family, including known (n = 4) and novel (n = 2) mutations in known genes (NAGLU, SLC5A2, POLR3B, VPS13A, SYN1, SPG11), and the identification of a novel disease gene (n = 1; NSL1). Functional analyses were additionally performed in a gene associated with autism-like symptoms and epileptic seizures for further proof of pathogenicity. Lastly, detailed genotype-phenotype correlations were carried out to assist with the diagnosis of prospective families and to determine genomic variation with clinical relevance. We concluded that the combination of linkage analyses and WGS to search for disease genes still remains a fruitful strategy for complex diseases with a variety of mutated genes and heterogeneous phenotypic manifestations, allowing for the identification of novel mutations, genes, and phenotypes, and leading to improvements in both diagnostic strategies and functional characterization of disease mechanisms.
Collapse
|
27
|
Hayes AJ, Melrose J. Keratan Sulphate in the Tumour Environment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1245:39-66. [PMID: 32266652 DOI: 10.1007/978-3-030-40146-7_2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Keratan sulphate (KS) is a bioactive glycosaminoglycan (GAG) of some complexity composed of the repeat disaccharide D-galactose β1→4 glycosidically linked to N-acetyl glucosamine. During the biosynthesis of KS, a family of glycosyltransferase and sulphotransferase enzymes act sequentially and in a coordinated fashion to add D-galactose (D-Gal) then N-acetyl glucosamine (GlcNAc) to a GlcNAc acceptor residue at the reducing terminus of a nascent KS chain to effect chain elongation. D-Gal and GlcNAc can both undergo sulphation at C6 but this occurs more frequently on GlcNAc than D-Gal. Sulphation along the developing KS chain is not uniform and contains regions of variable length where no sulphation occurs, regions which are monosulphated mainly on GlcNAc and further regions of high sulphation where both of the repeat disaccharides are sulphated. Each of these respective regions in the KS chain can be of variable length leading to KS complexity in terms of chain length and charge localization along the KS chain. Like other GAGs, it is these variably sulphated regions in KS which define its interactive properties with ligands such as growth factors, morphogens and cytokines and which determine the functional properties of tissues containing KS. Further adding to KS complexity is the identification of three different linkage structures in KS to asparagine (N-linked) or to threonine or serine residues (O-linked) in proteoglycan core proteins which has allowed the categorization of KS into three types, namely KS-I (corneal KS, N-linked), KS-II (skeletal KS, O-linked) or KS-III (brain KS, O-linked). KS-I to -III are also subject to variable addition of L-fucose and sialic acid groups. Furthermore, the GlcNAc residues of some members of the mucin-like glycoprotein family can also act as acceptor molecules for the addition of D-Gal and GlcNAc residues which can also be sulphated leading to small low sulphation glycoforms of KS. These differ from the more heavily sulphated KS chains found on proteoglycans. Like other GAGs, KS has evolved molecular recognition and information transfer properties over hundreds of millions of years of vertebrate and invertebrate evolution which equips them with cell mediatory properties in normal cellular processes and in aberrant pathological situations such as in tumourogenesis. Two KS-proteoglycans in particular, podocalyxin and lumican, are cell membrane, intracellular or stromal tissue-associated components with roles in the promotion or regulation of tumour development, mucin-like KS glycoproteins may also contribute to tumourogenesis. A greater understanding of the biology of KS may allow better methodology to be developed to more effectively combat tumourogenic processes.
Collapse
Affiliation(s)
- Anthony J Hayes
- Bioimaging Research Hub, Cardiff School of Biosciences, Cardiff University, Cardiff, Wales, UK
| | - James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia. .,Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards, NSW, Australia. .,Sydney Medical School, Northern, The University of Sydney, Faculty of Medicine and Health at Royal North Shore Hospital, St. Leonards, NSW, Australia.
| |
Collapse
|
28
|
Chen Y, Liu Y, Tian H, Chen Y, Lin S, Mao Q, Zheng N, Zhao J, Gu X, Wei H. Distribution of Pheromone Biosynthesis-Activating Neuropeptide in the Central Nervous System of Plutella xylostella (Lepidoptera: Plutellidae). JOURNAL OF ECONOMIC ENTOMOLOGY 2019; 112:2638-2648. [PMID: 31310309 DOI: 10.1093/jee/toz192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Indexed: 06/10/2023]
Abstract
Insect neuropeptides in the pyrokinin/pheromone biosynthesis-activating neuropeptide (PBAN) family are actively involved in many essential endocrinal functions and serve as potential targets in the search for novel insect control agents. Here, we dissect the nervous system of larval, pupal, and adult Plutella xylostella (L.) (Lepidoptera: Plutellidae) and describe the ganglion morphology and localization of PBAN during different insect developmental stages. Our results show that the central nervous system (CNS) of this species consists of four types of ganglia: cerebral ganglia (brain), subesophageal ganglion (SEG), thoracic ganglia, and abdominal ganglia. A two-lobed brain is connected to the reniform SEG with a nerve cord in larvae and prepupae, whereas in the late pupae and adults, the brain and SEG are fused, forming a brain-SEG complex. The larvae and prepupae have eight abdominal ganglia each, whereas the late pupae and adults each have four abdominal ganglia. Furthermore, all life stages of P. xylostella had similar patterns of PBAN immunoreactivity in the CNS, and the accumulation of PBAN was similar during all life stages except in adult males. PBAN immunoreactive signals were observed in the brain and SEG, and fluorescence signals originating in the SEG extended the entire length of the ventral nerve cord, ending in the terminal abdominal ganglia. Our results provide morphological data that inform the development and evolution of the CNS. In addition, they indicate that the nervous system contains PBAN, which could be used to control P. xylostella populations.
Collapse
Affiliation(s)
- Yong Chen
- Fujian Key Laboratory for Monitoring and Integrated Management of Crop Pests, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou, China
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian-Taiwan Joint Centre for Ecological Control of Crop Pests, Fujian Agriculture and Forestry University, Fuzhou, China
- Fuzhou Scientific Observing and Experimental Station of Crop Pests of Ministry of Agriculture, Fuzhou, China
| | - Yuyan Liu
- Fujian Key Laboratory for Monitoring and Integrated Management of Crop Pests, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou, China
| | - Houjun Tian
- Fujian Key Laboratory for Monitoring and Integrated Management of Crop Pests, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou, China
- Fuzhou Scientific Observing and Experimental Station of Crop Pests of Ministry of Agriculture, Fuzhou, China
| | - Yixin Chen
- Fujian Key Laboratory for Monitoring and Integrated Management of Crop Pests, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou, China
- Fuzhou Scientific Observing and Experimental Station of Crop Pests of Ministry of Agriculture, Fuzhou, China
| | - Shuo Lin
- Fujian Key Laboratory for Monitoring and Integrated Management of Crop Pests, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou, China
- Fuzhou Scientific Observing and Experimental Station of Crop Pests of Ministry of Agriculture, Fuzhou, China
| | - Qianzhuo Mao
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian-Taiwan Joint Centre for Ecological Control of Crop Pests, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Nan Zheng
- Fujian Key Laboratory for Monitoring and Integrated Management of Crop Pests, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou, China
| | - Jianwei Zhao
- Fujian Key Laboratory for Monitoring and Integrated Management of Crop Pests, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou, China
- Fuzhou Scientific Observing and Experimental Station of Crop Pests of Ministry of Agriculture, Fuzhou, China
| | - Xiaojun Gu
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian-Taiwan Joint Centre for Ecological Control of Crop Pests, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Hui Wei
- Fujian Key Laboratory for Monitoring and Integrated Management of Crop Pests, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou, China
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian-Taiwan Joint Centre for Ecological Control of Crop Pests, Fujian Agriculture and Forestry University, Fuzhou, China
- Fuzhou Scientific Observing and Experimental Station of Crop Pests of Ministry of Agriculture, Fuzhou, China
| |
Collapse
|
29
|
Fan XY, Shi G, Zhao P. Methylation in Syn and Psd95 genes underlie the inhibitory effect of oxytocin on oxycodone-induced conditioned place preference. Eur Neuropsychopharmacol 2019; 29:1464-1475. [PMID: 31735530 DOI: 10.1016/j.euroneuro.2019.10.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/02/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022]
Abstract
Oxycodone (Oxy) is one of the most effective analgesics in medicine, but is associated with the development of dependence. Recent studies demonstrating epigenetic changes in the brain after exposure to opiates have provided an insight into possible mechanisms underlying addiction. Oxytocin (OT), an endogenous neuropeptide well known for preventing drug abuse, is a promising pharmacotherapy to counteract addiction. Therefore, we explored the mechanism of Oxy addiction and the role of OT in Oxy-induced epigenetic alterations. In this study, drug-induced changes in conditioned place preference (CPP), i.e. the expression of synaptic proteins and synaptic density in the ventral tegmental area (VTA) were measured. We also sought to identify DNA methyltransferases (DNMTs), ten-eleven translocations (TETs), global 5-methylcytosine (5-mC), and DNA methylation of two genes implicated in plasticity (Synaptophysin, Syn; Post-synaptic density protein 95, Psd95). Oxy (3.0 mg/kg, i.p.) induced CPP acquisition in Sprague-Dawley rats. Oxy down-regulated DNMT1 and up-regulated TET1-3, leading to a decrease in global 5-mC levels and differential demethylation at exon 1 of Syn and exon 2 of Psd95. These changes in DNA methylation of Syn and Psd95 elevated the expression of synaptic proteins (SYN, PSD95) and synaptic density in the VTA. Pretreatment with OT (2.5 µg, i.c.v.) via its receptor specifically blocked Oxy CPP, normalized synaptic density, and regulated DNMT1 and TET2-3 causing reverse of DNA demethylation of Syn and Psd95. DNA methylation is an important gene regulation mechanism underlying Oxy CPP, and OT - via its receptor - could specifically inhibit Oxy addiction.
Collapse
Affiliation(s)
- Xin-Yu Fan
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, 110004, Shenyang, China
| | - Guang Shi
- Department of Neurology, Liaoning Provincial People's Hospital, Shenyang, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, 110004, Shenyang, China.
| |
Collapse
|
30
|
Rocchi A, Sacchetti S, De Fusco A, Giovedi S, Parisi B, Cesca F, Höltje M, Ruprecht K, Ahnert-Hilger G, Benfenati F. Autoantibodies to synapsin I sequestrate synapsin I and alter synaptic function. Cell Death Dis 2019; 10:864. [PMID: 31727880 PMCID: PMC6856194 DOI: 10.1038/s41419-019-2106-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/25/2019] [Accepted: 10/31/2019] [Indexed: 12/18/2022]
Abstract
Synapsin I is a phosphoprotein that coats the cytoplasmic side of synaptic vesicles and regulates their trafficking within nerve terminals. Autoantibodies against Syn I have been described in sera and cerebrospinal fluids of patients with numerous neurological diseases, including limbic encephalitis and clinically isolated syndrome; however, the effects and fate of autoantibodies in neurons are still unexplored. We found that in vitro exposure of primary hippocampal neurons to patient's autoantibodies to SynI decreased the density of excitatory and inhibitory synapses and impaired both glutamatergic and GABAergic synaptic transmission. These effects were reproduced with a purified SynI antibody and completely absent in SynI knockout neurons. Autoantibodies to SynI are internalized by FcγII/III-mediated endocytosis, interact with endogenous SynI, and promote its sequestration and intracellular aggregation. Neurons exposed to human autoantibodies to SynI display a reduced density of SVs, mimicking the SynI loss-of-function phenotype. Our data indicate that autoantibodies to intracellular antigens such as SynI can reach and inactivate their targets and suggest that an antibody-mediated synaptic dysfunction may contribute to the evolution and progression of autoimmune-mediated neurological diseases positive for SynI autoantibodies.
Collapse
Affiliation(s)
- Anna Rocchi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genova, Italy
- IRCSS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genova, Italy
| | - Silvio Sacchetti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genova, Italy
| | - Antonio De Fusco
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genova, Italy
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132, Genova, Italy
| | - Silvia Giovedi
- IRCSS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genova, Italy
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132, Genova, Italy
| | - Barbara Parisi
- IRCSS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genova, Italy
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132, Genova, Italy
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genova, Italy
- Department of Life Science, University of Trieste, via Giorgieri, 5, 34127, Trieste, Italy
| | - Markus Höltje
- Institute of Integrative Neuroanatomy, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Klemens Ruprecht
- Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Gudrun Ahnert-Hilger
- Institute of Integrative Neuroanatomy, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genova, Italy.
- IRCSS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genova, Italy.
| |
Collapse
|
31
|
Tellone E, Galtieri A, Ficarra S. Reviewing Biochemical Implications of Normal and Mutated Huntingtin in Huntington's Disease. Curr Med Chem 2019; 27:5137-5158. [PMID: 31223078 DOI: 10.2174/0929867326666190621101909] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/08/2019] [Accepted: 05/22/2019] [Indexed: 12/17/2022]
Abstract
Huntingtin (Htt) is a multi-function protein of the brain. Normal Htt shows a common alpha-helical structure but conformational changes in the form with beta strands are the principal cause of Huntington's disease. Huntington's disease is a genetic neurological disorder caused by a repeated expansion of the CAG trinucleotide, causing instability in the N-terminal of the gene coding for the Huntingtin protein. The mutation leads to the abnormal expansion of the production of the polyglutamine tract (polyQ) resulting in the form of an unstable Huntingtin protein commonly referred to as mutant Huntingtin. Mutant Huntingtin is the cause of the complex neurological metabolic alteration of Huntington's disease, resulting in both the loss of all the functions of normal Huntingtin and the genesis of abnormal interactions due to the presence of this mutation. One of the problems arising from the misfolded Huntingtin is the increase in oxidative stress, which is common in many neurological diseases such as Alzheimer's, Parkinson's, Amyotrophic Lateral Sclerosis and Creutzfeldt-Jakob disease. In the last few years, the use of antioxidants had a strong incentive to find valid therapies for defence against neurodegenerations. Although further studies are needed, the use of antioxidant mixtures to counteract neuronal damages seems promising.
Collapse
Affiliation(s)
- Ester Tellone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, V.le Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Antonio Galtieri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, V.le Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Silvana Ficarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, V.le Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
32
|
Fu T, Wang J, Ding Y, Zhang Y, Han S, Li J. Modulation of cPKCγ on Synapsin-Ia/b-Specific Phosphorylation Sites in the Developing Visual Cortex of Mice. Invest Ophthalmol Vis Sci 2019; 60:2676-2684. [PMID: 31242289 DOI: 10.1167/iovs.19-26675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To explore the role of synapsin-Ia/b in visual cortical plasticity, the dynamic changes in total protein expression (T-) and conventional protein kinase C (cPKC)γ-modulated phosphorylation (P-) levels of synapsin-Ia/b were observed in the developing visual cortex of mice. Methods The Western blot analysis was used to determine the levels of T- and P-synapsin-Ia/b at site of Ser9, 549, and 603; the cPKCγ gene wild-type (cPKCγ+/+) and knockout (cPKCγ-/-) mice were applied to explore the modulation of cPKCγ on synapsin-Ia/b phosphorylation status in visual cortex of mice at postnatal 7 to 60 days (P7-P60, n = 6 per group). Results The results showed that T-synapsin-Ia/b protein levels significantly increased at P14 to P35 and peaked at P42 to 60 (P < 0.001) in visual cortex when compared with that of P7 cPKCγ+/+ mice, and cPKCγ-/- did not affect this pattern of T-synapsin-Ia/b protein expressions. For synapsin-Ia/b phosphorylation status, the levels of P-Ser9 and 603 synapsin-Ia/b significantly elevated at P21 to P28 (P < 0.05 or 0.001), and then went down and maintained at lower levels at P35 to P60 (P < 0.05 or 0.001) compared with P7 cPKCγ+/+ mice. In addition, the cPKCγ gene knockout could significantly (P < 0.001) inhibit both the increase and decrease of P-Ser9 and 603 synapsin-Ia/b levels when compared with cPKCγ+/+ mice at P7 to P60. However, there were no significant changes of P-Ser549 synapsin-Ia/b in the developing visual cortex of both cPKCγ+/+ and cPKCγ-/- mice at P7 to P60. Conclusions These results suggested that both protein expression levels and cPKCγ-modulated phosphorylation status at Ser9 and 603 of synapsin-Ia/b may play important role in developing visual cortex of mice.
Collapse
Affiliation(s)
- Tao Fu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Jing Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Yichao Ding
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Yunxia Zhang
- Department of Neurobiology and Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Song Han
- Department of Neurobiology and Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Junfa Li
- Department of Neurobiology and Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
33
|
Lugarà E, De Fusco A, Lignani G, Benfenati F, Humeau Y. Synapsin I Controls Synaptic Maturation of Long-Range Projections in the Lateral Amygdala in a Targeted Selective Fashion. Front Cell Neurosci 2019; 13:220. [PMID: 31164805 PMCID: PMC6536628 DOI: 10.3389/fncel.2019.00220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 05/01/2019] [Indexed: 01/11/2023] Open
Abstract
The amygdala, and more precisely its lateral nucleus, is thought to attribute emotional valence to external stimuli by generating long-term plasticity changes at long-range projections to principal cells. Aversive experience has also been shown to modify pre- and post-synaptic markers in the amygdala, suggesting their possible role in the structural organization of adult amygdala networks. Here, we focused on how the maturation of cortical and thalamic long-range projections occurs on principal neurons and interneurons in the lateral amygdala (LA). We performed dual electrophysiological recordings of identified cells in juvenile and adult GAD67-GFP mice after independent stimulation of cortical and thalamic afferent systems. The results demonstrate that synaptic strengthening occurs during development at synapses projecting to LA principal neurons, but not interneurons. As synaptic strengthening underlies fear conditioning which depends, in turn, on presence and increasing expression of synapsin I, we tested if synapsin I contributes to synaptic strengthening during development. Interestingly, the physiological synaptic strengthening of cortical and thalamic synapses projecting to LA principal neurons was virtually abolished in synapsin I knockout mice, but not differences were observed in the excitatory projections to interneurons. Immunohistochemistry analysis showed that the presence of synapsin I is restricted to excitatory contacts projecting to principal neurons in LA of adult mice. These results indicate that synapsin I is a key regulator of the maturation of synaptic connectivity in this brain region and that is expression is dependent on postsynaptic identity.
Collapse
Affiliation(s)
- Eleonora Lugarà
- Department of Experimental Medicine, Section of Human Physiology, University of Genova, Genoa, Italy.,Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Antonio De Fusco
- Department of Experimental Medicine, Section of Human Physiology, University of Genova, Genoa, Italy.,Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Gabriele Lignani
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Yann Humeau
- Team Synapse in Cognition, Institut Interdisciplinaire de Neuroscience, Centre National de la Recherche Scientifique CNRS UMR5297, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
34
|
Xu N, Li AD, Ji LL, Ye Y, Wang ZY, Tong L. miR-132 regulates the expression of synaptic proteins in APP/PS1 transgenic mice through C1q. Eur J Histochem 2019; 63. [PMID: 31060348 PMCID: PMC6511887 DOI: 10.4081/ejh.2019.3008] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 04/01/2019] [Indexed: 02/02/2023] Open
Abstract
Cognitive impairment in Alzheimer’s disease (AD) is usually accompanied by synaptic loss in both the hippocampus and neocortex. In the early stage of AD, amyloid β-induced synapse changes is the main reason, while in the later stage, the accumulation of Tau protein promotes synapse degeneration as the key factor leading to dementia. MicroRNA (miRNA) is closely related to the expression changes of many AD-related genes. One of the most abundant brain-enriched miRNAs is miR-132, which has been shown to regulate both neuron morphogenesis and plasticity. It has been reported that miR-132 is significantly reduced in the brains of Alzheimer’s patients. Genetic deletion of miR-132 in mice promotes Aβ deposition, leading to impaired memory and enhanced Tau pathology, but how the miRNA-mediated gene expression dysregulation contributes to AD pathology remains unclear. Here we found the possible downstream target of miR-132 by in silico analysis, namely C1q. C1q is the primary protein of classical complement cascade, which is highly expressed in the synaptic regions of the central nervous system in Alzheimer’s patients. However, it is not clear whether miR-132 plays a role in AD through regulating C1q. To address this question, the APP/PS1 transgenic mice were transfected with miR-132 and given C1 inhibitors. Behavior tests were conducted to assess memory and cognitive abilities seven days after administration. In addition, we analyzed the expression of PSD95, Synapsin-1 and phosphorylated (p)- Synapsin. We found that the expression levels of the synaptic proteins treated with miR-132 or C1INH were significantly increased compared with the AD group. Further RT-qPCR result suggested that miR-132 might regulate C1q expression in AD.
Collapse
Affiliation(s)
- Nan Xu
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang.
| | | | | | | | | | | |
Collapse
|
35
|
Structural and biological evaluation of a platinum complex as a potential anti-neurodegenerative agent. Inorganica Chim Acta 2019. [DOI: 10.1016/j.ica.2019.03.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
36
|
Cheng W, Han F, Shi Y. Neonatal isolation modulates glucocorticoid-receptor function and synaptic plasticity of hippocampal and amygdala neurons in a rat model of single prolonged stress. J Affect Disord 2019; 246:682-694. [PMID: 30611912 DOI: 10.1016/j.jad.2018.12.084] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/23/2018] [Accepted: 12/24/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Early life and stressful experiences affect hippocampal and amygdala structure and function. They also increase the incidence of mental and nervous system disorders in adults. However, prospective studies have yet to show if early-life experiences affect the risk/severity of post-traumatic stress disorder (PTSD). METHODS We applied neonatal isolation (NI) alone, single prolonged stress (SPS) alone and NI + SPS to rats. We evaluated anxiety-like behavior and spatial memory of behavior using open field, elevated plus maze, and Morris water maze tests. Then, we measured expression of glucocorticoid receptors (GRs) and synaptic-related proteins by immunofluorescence, immunohistochemistry and western blotting in the hippocampus and amygdala. RESULTS NI + SPS exacerbated the increased anxiety levels and impaired spatial memory induced by NI alone or SPS alone. NI alone or SPS alone induced varying degrees of change in expression of GRs and synaptic proteins (synapsin I and postsynaptic density protein-95) in the hippocampus and amygdala. There were opposite changes in GR expression in the hippocampal dentate gyrus and basolateral amygdala. The degree of such change was exacerbated considerably by NI + SPS. In addition, neuroligin (NLG)-1 and NLG-2 were distributed in postsynaptic sites of excitatory and inhibitory synapses, respectively. NI, SPS, and NI + SPS altered the patterns of NLG-1 and NLG-2 colocalization as well as their intensity. NI + SPS strengthened the increased ratio of NLG-1/NLG-2 in the hippocampus, but decreased this ratio in the amygdala. CONCLUSIONS NI and SPS together induced greater degrees of change in anxiety and spatial memory, as well as GR and synaptic protein levels, in the hippocampus and amygdala than the changes induced by NI alone or SPS alone.
Collapse
Affiliation(s)
- Wei Cheng
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, 77, Puhe Road, Shenbei New District, 110001 Shenyang, China; Neonatal Department, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Fan Han
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, 77, Puhe Road, Shenbei New District, 110001 Shenyang, China
| | - Yuxiu Shi
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, 77, Puhe Road, Shenbei New District, 110001 Shenyang, China.
| |
Collapse
|
37
|
Cabana J, Gilbert G, Létourneau‐Guillon L, Safi D, Rouleau I, Cossette P, Nguyen DK. Effects of SYN1 Q555X mutation on cortical gray matter microstructure. Hum Brain Mapp 2018; 39:3428-3448. [PMID: 29671924 PMCID: PMC6866302 DOI: 10.1002/hbm.24186] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 04/08/2018] [Accepted: 04/09/2018] [Indexed: 01/16/2023] Open
Abstract
A new Q555X mutation on the SYN1 gene was recently found in several members of a family segregating dyslexia, epilepsy, and autism spectrum disorder. To describe the effects of this mutation on cortical gray matter microstructure, we performed a surface-based group study using novel diffusion and quantitative multiparametric imaging on 13 SYN1Q555X mutation carriers and 13 age- and sex-matched controls. Specifically, diffusion kurtosis imaging (DKI) and neurite orientation and dispersion and density imaging (NODDI) were used to analyze multi-shell diffusion data and obtain parametric maps sensitive to tissue structure, while quantitative metrics sensitive to tissue composition (T1, T2* and relative proton density [PD]) were obtained from a multi-echo variable flip angle FLASH acquisition. Results showed significant microstructural alterations in several regions usually involved in oral and written language as well as dyslexia. The most significant changes in these regions were lowered mean diffusivity and increased fractional anisotropy. This study is, to our knowledge, the first to successfully use diffusion imaging and multiparametric mapping to detect cortical anomalies in a group of subjects with a well-defined genotype linked to language impairments, epilepsy and autism spectrum disorder (ASD).
Collapse
Affiliation(s)
- Jean‐François Cabana
- Centre Hospitalier de l'Université de Montréal (CHUM)MontréalQuébec
- Université de Montréal
| | - Guillaume Gilbert
- Centre Hospitalier de l'Université de Montréal (CHUM)MontréalQuébec
- Université de Montréal
- Philips Healthcare CanadaMarkhamQuébec
| | - Laurent Létourneau‐Guillon
- Centre Hospitalier de l'Université de Montréal (CHUM)MontréalQuébec
- Centre de Recherche du CHUM (CRCHUM)MontréalQuébec
| | - Dima Safi
- Université du Québec à Trois‐Rivières (UQTR), Trois‐RivièresQuébec
- Groupe de recherche CogNAC (UQTR), Trois‐RivièresQuébec
| | - Isabelle Rouleau
- Centre de Recherche du CHUM (CRCHUM)MontréalQuébec
- Université du Québec à Montréal (UQAM), MontréalQuébec
| | - Patrick Cossette
- Centre Hospitalier de l'Université de Montréal (CHUM)MontréalQuébec
- Université de Montréal
- Centre de Recherche du CHUM (CRCHUM)MontréalQuébec
| | - Dang Khoa Nguyen
- Centre Hospitalier de l'Université de Montréal (CHUM)MontréalQuébec
- Université de Montréal
- Centre de Recherche du CHUM (CRCHUM)MontréalQuébec
| |
Collapse
|
38
|
Guarnieri FC, Pozzi D, Raimondi A, Fesce R, Valente MM, Delvecchio VS, Van Esch H, Matteoli M, Benfenati F, D'Adamo P, Valtorta F. A novel SYN1 missense mutation in non-syndromic X-linked intellectual disability affects synaptic vesicle life cycle, clustering and mobility. Hum Mol Genet 2018; 26:4699-4714. [PMID: 28973667 DOI: 10.1093/hmg/ddx352] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 08/29/2017] [Indexed: 01/04/2023] Open
Abstract
Intellectual Disability is a common and heterogeneous disorder characterized by limitations in intellectual functioning and adaptive behaviour, whose molecular mechanisms remain largely unknown. Among the numerous genes found to be involved in the pathogenesis of intellectual disability, 10% are located on the X-chromosome. We identified a missense mutation (c.236 C > G; p.S79W) in the SYN1 gene coding for synapsin I in the MRX50 family, affected by non-syndromic X-linked intellectual disability. Synapsin I is a neuronal phosphoprotein involved in the regulation of neurotransmitter release and neuronal development. Several mutations in SYN1 have been identified in patients affected by epilepsy and/or autism. The S79W mutation segregates with the disease in the MRX50 family and all affected members display intellectual disability as sole clinical manifestation. At the protein level, the S79W Synapsin I mutation is located in the region of the B-domain involved in recognition of highly curved membranes. Expression of human S79W Synapsin I in Syn1 knockout hippocampal neurons causes aberrant accumulation of small clear vesicles in the soma, increased clustering of synaptic vesicles at presynaptic terminals and increased frequency of excitatory spontaneous release events. In addition, the presence of S79W Synapsin I strongly reduces the mobility of synaptic vesicles, with possible implications for the regulation of neurotransmitter release and synaptic plasticity. These results implicate SYN1 in the pathogenesis of non-syndromic intellectual disability, showing that alterations of synaptic vesicle trafficking are one possible cause of this disease, and suggest that distinct mutations in SYN1 may lead to distinct brain pathologies.
Collapse
Affiliation(s)
- Fabrizia C Guarnieri
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy.,San Raffaele Vita-Salute University, 20132 Milan, Italy
| | - Davide Pozzi
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Andrea Raimondi
- Experimental Imaging Center, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Riccardo Fesce
- Centre of Neuroscience and DISTA, University of Insubria, 21100 Varese, Italy
| | - Maria M Valente
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Hilde Van Esch
- Center for Human Genetics, University Hospitals Leuven, B3000 Leuven, Belgium
| | - Michela Matteoli
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy.,CNR Institute of Neuroscience, Milan, Italy
| | - Fabio Benfenati
- Department of Experimental Medicine, University of Genova, 16132 Genova, Italy.,Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy
| | - Patrizia D'Adamo
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Flavia Valtorta
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy.,San Raffaele Vita-Salute University, 20132 Milan, Italy
| |
Collapse
|
39
|
Keren-Aviram G, Dachet F, Bagla S, Balan K, Loeb JA, Dratz EA. Proteomic analysis of human epileptic neocortex predicts vascular and glial changes in epileptic regions. PLoS One 2018; 13:e0195639. [PMID: 29634780 PMCID: PMC5892923 DOI: 10.1371/journal.pone.0195639] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 03/26/2018] [Indexed: 01/21/2023] Open
Abstract
Epilepsy is a common neurological disorder, which is not well understood at the molecular level. Exactly why some brain regions produce epileptic discharges and others do not is not known. Patients who fail to respond to antiseizure medication (refractory epilepsy) can benefit from surgical removal of brain regions to reduce seizure frequency. The tissue removed in these surgeries offers an invaluable resource to uncover the molecular and cellular basis of human epilepsy. Here, we report a proteomic study to determine whether there are common proteomic patterns in human brain regions that produce epileptic discharges. We analyzed human brain samples, as part of the Systems Biology of Epilepsy Project (SBEP). These brain pieces are in vivo electrophysiologically characterized human brain samples withdrawn from the neocortex of six patients with refractory epilepsy. This study is unique in that for each of these six patients the comparison of protein expression was made within the same patient: a more epileptic region was compared to a less epileptic brain region. The amount of epileptic activity was defined for each patient as the frequency of their interictal spikes (electric activity between seizures that is a parameter strongly linked to epilepsy). Proteins were resolved from three subcellular fractions, using a 2D differential gel electrophoresis (2D-DIGE), revealing 31 identified protein spots that changed significantly. Interestingly, glial fibrillary acidic protein (GFAP) was found to be consistently down regulated in high spiking brain tissue and showed a strong negative correlation with spike frequency. We also developed a two-step analysis method to select for protein species that changed frequently among the patients and identified these proteins. A total of 397 protein spots of interest (SOI) were clustered by protein expression patterns across all samples. These clusters were used as markers and this analysis predicted proteomic changes due to both histological differences and molecular pathways, revealed by examination of gene ontology clusters. Our experimental design and proteomic data analysis predicts novel glial changes, increased angiogenesis, and changes in cytoskeleton and neuronal projections between high and low interictal spiking regions. Quantitative histological staining of these same tissues for both the vascular and glial changes confirmed these findings, which provide new insights into the structural and functional basis of neocortical epilepsy.
Collapse
Affiliation(s)
- Gal Keren-Aviram
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, Montana, United States of America
| | - Fabien Dachet
- The Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Shruti Bagla
- The Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Karina Balan
- The Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Jeffrey A. Loeb
- The Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| | - Edward A. Dratz
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, Montana, United States of America
| |
Collapse
|
40
|
Barbieri R, Contestabile A, Ciardo MG, Forte N, Marte A, Baldelli P, Benfenati F, Onofri F. Synapsin I and Synapsin II regulate neurogenesis in the dentate gyrus of adult mice. Oncotarget 2018; 9:18760-18774. [PMID: 29721159 PMCID: PMC5922353 DOI: 10.18632/oncotarget.24655] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 02/25/2018] [Indexed: 01/23/2023] Open
Abstract
Adult neurogenesis is emerging as an important player in brain functions and homeostasis, while impaired or altered adult neurogenesis has been associated with a number of neuropsychiatric diseases, such as depression and epilepsy. Here we investigated the possibility that synapsins (Syns) I and II, beyond their known functions in developing and mature neurons, also play a role in adult neurogenesis. We performed a systematic evaluation of the distinct stages of neurogenesis in the hippocampal dentate gyrus of Syn I and Syn II knockout (KO) mice, before (2-months-old) and after (6-months-old) the appearance of the epileptic phenotype. We found that Syns I and II play an important role in the regulation of adult neurogenesis. In juvenile mice, Syn II deletion was associated with a specific decrease in the proliferation of neuronal progenitors, whereas Syn I deletion impaired the survival of newborn neurons. These defects were reverted after the appearance of the epileptic phenotype, with Syn I KO and Syn II KO mice exhibiting significant increases in survival and proliferation, respectively. Interestingly, long-term potentiation dependent on newborn neurons was present in both juvenile Syn mutants while, at later ages, it was only preserved in Syn II KO mice that also displayed an increased expression of brain-derived neurotrophic factor. This study suggests that Syns I and II play a role in adult neurogenesis and the defects in neurogenesis associated with Syn deletion may contribute to the alterations of cognitive functions observed in Syn-deficient mice.
Collapse
Affiliation(s)
- Raffaella Barbieri
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy
| | - Andrea Contestabile
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 16163, Genova, Italy
| | - Maria Grazia Ciardo
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy
| | - Nicola Forte
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132, Genova, Italy
| | - Antonella Marte
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy
| | - Pietro Baldelli
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy.,Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132, Genova, Italy
| | - Fabio Benfenati
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy.,Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132, Genova, Italy
| | - Franco Onofri
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy
| |
Collapse
|
41
|
Caterson B, Melrose J. Keratan sulfate, a complex glycosaminoglycan with unique functional capability. Glycobiology 2018; 28:182-206. [PMID: 29340594 PMCID: PMC5993099 DOI: 10.1093/glycob/cwy003] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 12/20/2017] [Accepted: 01/08/2018] [Indexed: 12/19/2022] Open
Abstract
From an evolutionary perspective keratan sulfate (KS) is the newest glycosaminoglycan (GAG) but the least understood. KS is a sophisticated molecule with a diverse structure, and unique functional roles continue to be uncovered for this GAG. The cornea is the richest tissue source of KS in the human body but the central and peripheral nervous systems also contain significant levels of KS and a diverse range of KS-proteoglycans with essential functional roles. KS also displays important cell regulatory properties in epithelial and mesenchymal tissues and in bone and in tumor development of diagnostic and prognostic utility. Corneal KS-I displays variable degrees of sulfation along the KS chain ranging from non-sulfated polylactosamine, mono-sulfated and disulfated disaccharide regions. Skeletal KS-II is almost completely sulfated consisting of disulfated disaccharides interrupted by occasional mono-sulfated N-acetyllactosamine residues. KS-III also contains highly sulfated KS disaccharides but differs from KS-I and KS-II through 2-O-mannose linkage to serine or threonine core protein residues on proteoglycans such as phosphacan and abakan in brain tissue. Historically, the major emphasis on the biology of KS has focused on its sulfated regions for good reason. The sulfation motifs on KS convey important molecular recognition information and direct cell behavior through a number of interactive proteins. Emerging evidence also suggest functional roles for the poly-N-acetyllactosamine regions of KS requiring further investigation. Thus further research is warranted to better understand the complexities of KS.
Collapse
Affiliation(s)
- Bruce Caterson
- Connective Tissue Biology Laboratories, School of Biosciences, College of Biological & Life Sciences, Cardiff University, Cardiff, Wales, UK
| | - James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute of Medical Research, Northern Sydney Local Health District, St. Leonards, NSW, Australia
- Sydney Medical School, Northern, The University of Sydney, Royal North Shore Hospital, St. Leonards, NSW, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
42
|
Calahorro F, Izquierdo PG. The presynaptic machinery at the synapse of C. elegans. INVERTEBRATE NEUROSCIENCE : IN 2018; 18:4. [PMID: 29532181 PMCID: PMC5851683 DOI: 10.1007/s10158-018-0207-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/22/2018] [Indexed: 11/17/2022]
Abstract
Synapses are specialized contact sites that mediate information flow between neurons and their targets. Important physical interactions across the synapse are mediated by synaptic adhesion molecules. These adhesions regulate formation of synapses during development and play a role during mature synaptic function. Importantly, genes regulating synaptogenesis and axon regeneration are conserved across the animal phyla. Genetic screens in the nematode Caenorhabditis elegans have identified a number of molecules required for synapse patterning and assembly. C. elegans is able to survive even with its neuronal function severely compromised. This is in comparison with Drosophila and mice where increased complexity makes them less tolerant to impaired function. Although this fact may reflect differences in the function of the homologous proteins in the synapses between these organisms, the most likely interpretation is that many of these components are equally important, but not absolutely essential, for synaptic transmission to support the relatively undemanding life style of laboratory maintained C. elegans. Here, we review research on the major group of synaptic proteins, involved in the presynaptic machinery in C. elegans, showing a strong conservation between higher organisms and highlight how C. elegans can be used as an informative tool for dissecting synaptic components, based on a simple nervous system organization.
Collapse
Affiliation(s)
- Fernando Calahorro
- Biological Sciences, University of Southampton, Life Sciences Building 85, Southampton, SO17 1BJ, UK.
| | - Patricia G Izquierdo
- Biological Sciences, University of Southampton, Life Sciences Building 85, Southampton, SO17 1BJ, UK
| |
Collapse
|
43
|
Hu Y, Pan S, Zhang HT. Interaction of Cdk5 and cAMP/PKA Signaling in the Mediation of Neuropsychiatric and Neurodegenerative Diseases. ADVANCES IN NEUROBIOLOGY 2018; 17:45-61. [PMID: 28956329 DOI: 10.1007/978-3-319-58811-7_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Both cyclin-dependent kinase 5 (Cdk5) and cyclic AMP (cAMP)/protein kinase A (PKA) regulate fundamental central nervous system (CNS) functions including neuronal survival, neurite and axonal outgrowth, neuron development and cognition. Cdk5, a serine/threonine kinase, is activated by p35 or p39 and phosphorylates multiple signaling components of various pathways, including cAMP/PKA signaling. Here, we review the recent literature on the interaction between Cdk5 and cAMP/PKA signaling and their role in the mediation of CNS functions and neuropsychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- Yafang Hu
- Department of Neurology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong, 510515, China.
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong, 510515, China
| | - Han-Ting Zhang
- Department of Behavioral Medicine and Psychiatry, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506, USA.,Department of Physiology and Pharmacology, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506, USA.,Institute of Pharmacology, Taishan Medical University, Taian, 271016, China
| |
Collapse
|
44
|
Zhu L, Tang T, Fan R, Luo JK, Cui HJ, Zhang CH, Peng WJ, Sun P, Xiong XG, Wang Y. Xuefu Zhuyu decoction improves neurological dysfunction by increasing synapsin expression after traumatic brain injury. Neural Regen Res 2018; 13:1417-1424. [PMID: 30106054 PMCID: PMC6108199 DOI: 10.4103/1673-5374.235297] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Xuefu Zhuyu decoction has been used for treating traumatic brain injury and improving post-traumatic dysfunction, but its mechanism of action needs further investigation. This study established rat models of traumatic brain injury by controlled cortical impact. Rat models were intragastrically administered 9 and 18 g/kg Xuefu Zhuyu decoction once a day for 14 or 21 days. Changes in neurological function were assessed by modified neurological severity scores and the Morris water maze. Immunohistochemistry, western blot assay, and reverse-transcription polymerase chain reaction were used to analyze synapsin protein and mRNA expression at the injury site of rats. Our results showed that Xuefu Zhuyu decoction visibly improved neurological function of rats with traumatic brain injury. These changes were accompanied by increased expression of synaptophysin, synapsin I, and postsynaptic density protein-95 protein and mRNA in a dose-dependent manner. These findings indicate that Xuefu Zhuyu decoction increases synapsin expression and improves neurological deficits after traumatic brain injury.
Collapse
Affiliation(s)
- Lin Zhu
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Tao Tang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Rong Fan
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jie-Kun Luo
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Han-Jin Cui
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Chun-Hu Zhang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Wei-Jun Peng
- Department of Integrated Traditional Chinese and Western Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Peng Sun
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Xin-Gui Xiong
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yang Wang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
45
|
Mirza FJ, Zahid S. The Role of Synapsins in Neurological Disorders. Neurosci Bull 2017; 34:349-358. [PMID: 29282612 DOI: 10.1007/s12264-017-0201-7] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 10/27/2017] [Indexed: 12/12/2022] Open
Abstract
Synapsins serve as flagships among the presynaptic proteins due to their abundance on synaptic vesicles and contribution to synaptic communication. Several studies have emphasized the importance of this multi-gene family of neuron-specific phosphoproteins in maintaining brain physiology. In the recent times, increasing evidence has established the relevance of alterations in synapsins as a major determinant in many neurological disorders. Here, we give a comprehensive description of the diverse roles of the synapsin family and the underlying molecular mechanisms that contribute to several neurological disorders. These physiologically important roles of synapsins associated with neurological disorders are just beginning to be understood. A detailed understanding of the diversified expression of synapsins may serve to strategize novel therapeutic approaches for these debilitating neurological disorders.
Collapse
Affiliation(s)
- Fatima Javed Mirza
- Neurobiology Research Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Saadia Zahid
- Neurobiology Research Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan.
| |
Collapse
|
46
|
Richard AE, Scheffer IE, Wilson SJ. Features of the broader autism phenotype in people with epilepsy support shared mechanisms between epilepsy and autism spectrum disorder. Neurosci Biobehav Rev 2017; 75:203-233. [DOI: 10.1016/j.neubiorev.2016.12.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 12/15/2016] [Accepted: 12/20/2016] [Indexed: 12/29/2022]
|
47
|
Wang S, Zhang P, Liu R, Li Y, Liu C, Liao X. A DEHP plasticizer alters synaptic proteins via peroxidation. Toxicol Res (Camb) 2016; 6:89-97. [PMID: 30090480 DOI: 10.1039/c6tx00361c] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 11/01/2016] [Indexed: 12/24/2022] Open
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is a widely used commercial plasticizer. DEHP exposure has a negative impact on brain development and cognition, but the mechanisms responsible for DEHP-induced neurotoxicity are not well understood. Here we showed that DEHP exposure increased maleic dialdehyde and reactive oxygen species contents and decreased endogenous superoxide dismutase activity in a mouse neuroblastoma cell line (N2a cell line). DEHP exposure not only induced reduction of neurite outgrowth, but also led to microtubule-associated protein tau hyperphosphorylation and dissociation from microtubules. Furthermore, DEHP exposure decreased the levels of synapsin-1 and postsynaptic density protein 95 (PSD95), which play critical roles in synaptic function. Antioxidant vitamin E pretreatment prevented DEHP-induced abnormalities in the cells. These results indicate that DEHP exposure could induce abnormal action of proteins including tau, synapsin-1 and PSD95, which play critical roles in the synaptic structure and function, and that these alterations might be mediated by peroxidative damage.
Collapse
Affiliation(s)
- Shaohui Wang
- School of Life Sciences , Central China Normal University , Hubei Key Lab of Genetic Regulation and Integrative Biology , Wuhan , P.R. China . ; ; Tel: +86 027 67867229
| | - Pengyan Zhang
- School of Life Sciences , Central China Normal University , Hubei Key Lab of Genetic Regulation and Integrative Biology , Wuhan , P.R. China . ; ; Tel: +86 027 67867229
| | - Ruifang Liu
- School of Life Sciences , Central China Normal University , Hubei Key Lab of Genetic Regulation and Integrative Biology , Wuhan , P.R. China . ; ; Tel: +86 027 67867229
| | - Yuan Li
- School of Life Sciences , Central China Normal University , Hubei Key Lab of Genetic Regulation and Integrative Biology , Wuhan , P.R. China . ; ; Tel: +86 027 67867229
| | - Chao Liu
- School of Life Sciences , Central China Normal University , Hubei Key Lab of Genetic Regulation and Integrative Biology , Wuhan , P.R. China . ; ; Tel: +86 027 67867229
| | - Xiaomei Liao
- School of Life Sciences , Central China Normal University , Hubei Key Lab of Genetic Regulation and Integrative Biology , Wuhan , P.R. China . ; ; Tel: +86 027 67867229
| |
Collapse
|
48
|
Sphingosine-1-Phosphate (S1P) Impacts Presynaptic Functions by Regulating Synapsin I Localization in the Presynaptic Compartment. J Neurosci 2016; 36:4624-34. [PMID: 27098703 DOI: 10.1523/jneurosci.3588-15.2016] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 03/16/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Growing evidence indicates that sphingosine-1-P (S1P) upregulates glutamate secretion in hippocampal neurons. However, the molecular mechanisms through which S1P enhances excitatory activity remain largely undefined. The aim of this study was to identify presynaptic targets of S1P action controlling exocytosis. Confocal analysis of rat hippocampal neurons showed that S1P applied at nanomolar concentration alters the distribution of Synapsin I (SynI), a presynaptic phosphoprotein that controls the availability of synaptic vesicles for exocytosis. S1P induced SynI relocation to extrasynaptic regions of mature neurons, as well as SynI dispersion from synaptic vesicle clusters present at axonal growth cones of developing neurons. S1P-induced SynI relocation occurred in a Ca(2+)-independent but ERK-dependent manner, likely through the activation of S1P3 receptors, as it was prevented by the S1P3 receptor selective antagonist CAY1044 and in neurons in which S1P3 receptor was silenced. Our recent evidence indicates that microvesicles (MVs) released by microglia enhance the metabolism of endogenous sphingolipids in neurons and stimulate excitatory transmission. We therefore investigated whether MVs affect SynI distribution and whether endogenous S1P could be involved in the process. Analysis of SynI immunoreactivity showed that exposure to microglial MVs induces SynI mobilization at presynaptic sites and growth cones, whereas the use of inhibitors of sphingolipid cascade identified S1P as the sphingolipid mediating SynI redistribution. Our data represent the first demonstration that S1P induces SynI mobilization from synapses, thereby indicating the phosphoprotein as a novel target through which S1P controls exocytosis. SIGNIFICANCE STATEMENT Growing evidence indicates that the bioactive lipid sphingosine and its metabolite sphingosine-1-P (S1P) stimulate excitatory transmission. While it has been recently clarified that sphingosine influences directly the exocytotic machinery by activating the synaptic vesicle protein VAMP2 to form SNARE fusion complexes, the molecular mechanism by which S1P promotes neurotransmission remained largely undefined. In this study, we identify Synapsin I, a presynaptic phosphoprotein involved in the control of availability of synaptic vesicles for exocytosis, as the key target of S1P action. In addition, we provide evidence that S1P can be produced at mature axon terminals as well as at immature growth cones in response to microglia-derived signals, which may be important to stabilize nascent synapses and to restore or potentiate transmission.
Collapse
|
49
|
Beining M, Jungenitz T, Radic T, Deller T, Cuntz H, Jedlicka P, Schwarzacher SW. Adult-born dentate granule cells show a critical period of dendritic reorganization and are distinct from developmentally born cells. Brain Struct Funct 2016; 222:1427-1446. [PMID: 27514866 DOI: 10.1007/s00429-016-1285-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/02/2016] [Indexed: 02/05/2023]
Abstract
Adult-born dentate granule cells (abGCs) exhibit a critical developmental phase during function integration. The time window of this phase is debated and whether abGCs become indistinguishable from developmentally born mature granule cells (mGCs) is uncertain. We analyzed complete dendritic reconstructions from abGCs and mGCs using viral labeling. AbGCs from 21-77 days post intrahippocampal injection (dpi) exhibited comparable dendritic arbors, suggesting that structural maturation precedes functional integration. In contrast, significant structural differences were found compared to mGCs: AbGCs had more curved dendrites, more short terminal segments, a different branching pattern, and more proximal terminal branches. Morphological modeling attributed these differences to developmental dendritic pruning and postnatal growth of the dentate gyrus. We further correlated GC morphologies with the responsiveness to unilateral medial perforant path stimulation using the immediate-early gene Arc as a marker of synaptic activation. Only abGCs at 28 and 35 dpi but neither old abGCs nor mGCs responded to stimulation with a remodeling of their dendritic arbor. Summarized, abGCs stay distinct from mGCs and their dendritic arbor can be shaped by afferent activity during a narrow critical time window.
Collapse
Affiliation(s)
- Marcel Beining
- Institute of Clinical Neuroanatomy, Goethe University, 60528, Frankfurt am Main, Germany. .,Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, Deutschordenstr. 46, 60528, Frankfurt am Main, Germany. .,Frankfurt Institute for Advanced Studies (FIAS), 60438, Frankfurt am Main, Germany.
| | - Tassilo Jungenitz
- Institute of Clinical Neuroanatomy, Goethe University, 60528, Frankfurt am Main, Germany
| | - Tijana Radic
- Institute of Clinical Neuroanatomy, Goethe University, 60528, Frankfurt am Main, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Goethe University, 60528, Frankfurt am Main, Germany
| | - Hermann Cuntz
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, Deutschordenstr. 46, 60528, Frankfurt am Main, Germany.,Frankfurt Institute for Advanced Studies (FIAS), 60438, Frankfurt am Main, Germany
| | - Peter Jedlicka
- Institute of Clinical Neuroanatomy, Goethe University, 60528, Frankfurt am Main, Germany
| | | |
Collapse
|
50
|
Quaranta MT, Spinello I, Paolillo R, Macchia G, Boe A, Ceccarini M, Labbaye C, Macioce P. Identification of β-Dystrobrevin as a Direct Target of miR-143: Involvement in Early Stages of Neural Differentiation. PLoS One 2016; 11:e0156325. [PMID: 27223470 PMCID: PMC4880309 DOI: 10.1371/journal.pone.0156325] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 05/12/2016] [Indexed: 11/25/2022] Open
Abstract
Duchenne Muscular Dystrophy, a genetic disorder that results in a gradual breakdown of muscle, is associated to mild to severe cognitive impairment in about one-third of dystrophic patients. The brain dysfunction is independent of the muscular pathology, occurs early, and is most likely due to defects in the assembly of the Dystrophin-associated Protein Complex (DPC) during embryogenesis. We have recently described the interaction of the DPC component β-dystrobrevin with members of complexes that regulate chromatin dynamics, and suggested that β-dystrobrevin may play a role in the initiation of neuronal differentiation. Since oxygen concentrations and miRNAs appear as well to be involved in the cellular processes related to neuronal development, we have studied how these factors act on β-dystrobrevin and investigated the possibility of their functional interplay using the NTera-2 cell line, a well-established model for studying neurogenesis. We followed the pattern of expression and regulation of β-dystrobrevin during the early stages of neuronal differentiation induced by exposure to retinoic acid (RA) under hypoxia as compared with normoxia, and found that β-dystrobrevin expression is regulated during RA-induced differentiation of NTera-2 cells. We also found that β-dystrobrevin pattern is delayed under hypoxic conditions, together with a delay in the differentiation and an increase in the proliferation rate of cells. We identified miRNA-143 as a direct regulator of β-dystrobrevin expression, demonstrated that β-dystrobrevin is expressed in the nucleus and showed that, in line with our previous in vitro results, β-dystrobrevin is a repressor of synapsin I in live cells. Altogether the newly identified regulatory pathway miR-143/β-dystrobrevin/synapsin I provides novel insights into the functions of β-dystrobrevin and opens up new perspectives for elucidating the molecular mechanisms underlying the neuronal involvement in muscular dystrophy.
Collapse
Affiliation(s)
- Maria Teresa Quaranta
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Isabella Spinello
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Rosa Paolillo
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Gianfranco Macchia
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Boe
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Marina Ceccarini
- National Centre for Rare Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Catherine Labbaye
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Pompeo Macioce
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
- * E-mail:
| |
Collapse
|