1
|
Herrera G, Silvero C MJ, Becerra MC, Lasaga M, Scimonelli T. Modulatory role of α-MSH in hippocampal-dependent memory impairment, synaptic plasticity changes, oxidative stress, and astrocyte reactivity induced by short-term high-fat diet intake. Neuropharmacology 2023; 239:109688. [PMID: 37591460 DOI: 10.1016/j.neuropharm.2023.109688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 08/19/2023]
Abstract
High-fat diet (HFD) consumption is associated with cognitive deficits and neurodegenerative diseases. Since the hippocampus is extremely sensitive to pathophysiological changes, neuroinflammation and the concomitant oxidative stress induced by HFD can significantly interfere with hippocampal-dependent functions related to learning and memory. The neuropeptide alpha-melanocyte stimulating hormone (α-MSH) mediates neuroprotective actions in the central nervous system and can reverse the effects of neuroinflammation in cognitive functions that depend on the hippocampus. In this study, we used male Wistar rats to evaluate the effect of short-term HFD intake (5 days) plus a mild immune challenge, Lipopolysaccharide (LPS 10 μg/kg) on contextual fear, changes in structural plasticity, oxidative stress, and astrocyte reactivation in the hippocampus. We also determined the possible modulatory role of α-MSH. HFD consumption was associated with an increase in markers of oxidative stress (Advanced oxidation protein products and Malondialdehyde) in the dorsal hippocampus (DH). We also found changes in hippocampal structural synaptic plasticity, observing a decrease in total spine in the DH after HFD plus LPS. We observed astrocyte proliferation and a significant increase in the percentage of the area occupied by GFAP. Treatment with α-MSH (0.1 μg/0.25 μl) in the DH reversed the effect of short-term HFD plus LPS on contextual fear memory, oxidative stress, and spine density. α-MSH also reduced astrocyte proliferation. Our present results indicate that HFD consumption for a short period sensitizes the central nervous system (CNS) to a subsequent immune challenge and impairs contextual fear memory and that α-MSH could have a modulatory protective effect.
Collapse
Affiliation(s)
- Guadalupe Herrera
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET. Departamento de Farmacología Otto Orshinger, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.
| | - M Jazmín Silvero C
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica UNITEFA-CONICET. Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - M Cecilia Becerra
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica UNITEFA-CONICET. Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mercedes Lasaga
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Teresa Scimonelli
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET. Departamento de Farmacología Otto Orshinger, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
2
|
Xu L, Liu Y, Long J, He X, Xie F, Yin Q, Chen M, Long D, Chen Y. Loss of spines in the prelimbic cortex is detrimental to working memory in mice with early-life adversity. Mol Psychiatry 2023; 28:3444-3458. [PMID: 37500828 PMCID: PMC10618093 DOI: 10.1038/s41380-023-02197-7] [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: 11/22/2022] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023]
Abstract
Adverse experiences in early life can shape neuronal structures and synaptic function in multiple brain regions, leading to deficits of distinct cognitive functions later in life. Focusing on the pyramidal cells of the prelimbic cortex (PrL), a main subregion of the medial prefrontal cortex, the impact of early-life adversity (ELA) was investigated in a well-established animal model generated by changing the rearing environment during postnatal days 2 to 9 (P2-P9), a sensitive developmental period. ELA has enduring detrimental impacts on the dendritic spines of PrL pyramidal cells, which is most apparent in a spatially circumscribed region. Specifically, ELA affects both thin and mushroom-type spines, and ELA-provoked loss of spines is observed on selective dendritic segments of PrL pyramidal cells in layers II-III and V-VI. Reduced postsynaptic puncta represented by postsynaptic density protein-95 (PSD-95), but not synaptophysin-labelled presynaptic puncta, in ELA mice supports the selective loss of spines in the PrL. Correlation analysis indicates that loss of spines and postsynaptic puncta in the PrL contributes to the poor spatial working memory of ELA mice, and thin spines may play a major role in working memory performance. To further understand whether loss of spines affects glutamatergic transmission, AMPA- and NMDA-receptor-mediated synaptic currents (EPSCs) were recorded in a group of Thy1-expressing PrL pyramidal cells. ELA mice exhibited a depressed glutamatergic transmission, which is accompanied with a decreased expression of GluR1 and NR1 subunits in the PrL. Finally, upregulating the activation of Thy1-expressing PrL pyramidal cells via excitatory DREADDs can efficiently improve the working memory performance of ELA mice in a T-maze-based task, indicating the potential of a chemogenetic approach in restoring ELA-provoked memory deficits.
Collapse
Affiliation(s)
- Liping Xu
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Yue Liu
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Jingyi Long
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525GA, Nijmegen, the Netherlands
| | - Xiulan He
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Fanbing Xie
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Qiao Yin
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Michael Chen
- University of California, Los Angeles, CA, 90095, USA
| | - Dahong Long
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| | - Yuncai Chen
- Department of Pediatrics, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
3
|
Li L, Ru Q, Lu Y, Fang X, Chen G, Saifullah AB, Yao C, Tolias KF. Tiam1 coordinates synaptic structural and functional plasticity underpinning the pathophysiology of neuropathic pain. Neuron 2023; 111:2038-2050.e6. [PMID: 37146610 PMCID: PMC10330505 DOI: 10.1016/j.neuron.2023.04.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 09/27/2022] [Accepted: 04/10/2023] [Indexed: 05/07/2023]
Abstract
Neuropathic pain is a common, debilitating chronic pain condition caused by damage or a disease affecting the somatosensory nervous system. Understanding the pathophysiological mechanisms underlying neuropathic pain is critical for developing new therapeutic strategies to treat chronic pain effectively. Tiam1 is a Rac1 guanine nucleotide exchange factor (GEF) that promotes dendritic and synaptic growth during hippocampal development by inducing actin cytoskeletal remodeling. Here, using multiple neuropathic pain animal models, we show that Tiam1 coordinates synaptic structural and functional plasticity in the spinal dorsal horn via actin cytoskeleton reorganization and synaptic NMDAR stabilization and that these actions are essential for the initiation, transition, and maintenance of neuropathic pain. Furthermore, an antisense oligonucleotides (ASO) targeting spinal Tiam1 persistently alleviate neuropathic pain sensitivity. Our findings suggest that Tiam1-coordinated synaptic functional and structural plasticity underlies the pathophysiology of neuropathic pain and that intervention of Tiam1-mediated maladaptive synaptic plasticity has long-lasting consequences in neuropathic pain management.
Collapse
Affiliation(s)
- Lingyong Li
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35025, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Qin Ru
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Health and Kinesiology, School of Physical Education, Jianghan University, Wuhan 430056, China
| | - Yungang Lu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pain Medicine, Anesthesiology, Critical Care and Pain Medicine Division, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xing Fang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Guanxing Chen
- Department of Pain Medicine, Anesthesiology, Critical Care and Pain Medicine Division, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ali Bin Saifullah
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Changqun Yao
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35025, USA
| | - Kimberley F Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
4
|
Pompili M, Sarli G, Erbuto D, Manfredi G, Comparelli A. Clinical experiences with intranasal esketamine for major depressive disorder resistant to treatment and with a psychiatric emergency: case presentations. Int Clin Psychopharmacol 2023; 38:195-200. [PMID: 36630183 DOI: 10.1097/yic.0000000000000455] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Recently, esketamine became availableas an intranasal formulation, proposed for treatment-resistant depression (TRD). Three cases of TRD are presented, two with features of a psychiatric emergency. The first case is a 35-year-old man with MDD onset at the age of 27 years, with five previous failed therapies. The second patient is a middle-aged man with a 21-year MDD onset and six previous antidepressant treatments discontinued for poor therapeutic effects and tolerability. He also presented suicidal ideation with intent and a history of a failed suicide attempt by self-cutting his forearms. The third case is a 28-year-old female with a first MDD episode in 2020, treated first with amitriptyline and then with intravenous clomipramine. She had a history of a previous suicide attempt by self-cutting and, by her admission, showed active suicidal ideation with intent. In all three cases, a rapid reduction of depressive symptoms was observed with a subsequent complete resolution of suicidal ideation and intent in the two patients with such risk. Intranasal esketamine treatment was carried out with concomitant oral antidepressant therapy. The third patient reported the only recorded side effect: dissociation 20 min after every esketamine administration. Our preliminary experience proved esketamine's effectiveness on TRD symptoms and successful outcomes in psychiatric emergencies such as suicide risk.
Collapse
Affiliation(s)
- Maurizio Pompili
- Department of Neurosciences, Mental Health and Sensory Organs, Suicide Prevention Centre, Sant'Andrea Hospital, Sapienza University of Rome
| | - Giuseppe Sarli
- Psychiatry Residency Training Programme, Faculty of Medicine and Psychology, Sapienza University of Rome
| | - Denise Erbuto
- Sant'Andrea Teaching Hospital, Sapienza University of Rome, Rome, Italy
| | - Giovanni Manfredi
- Sant'Andrea Teaching Hospital, Sapienza University of Rome, Rome, Italy
| | - Anna Comparelli
- Sant'Andrea Teaching Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
5
|
Fawcett JW, Fyhn M, Jendelova P, Kwok JCF, Ruzicka J, Sorg BA. The extracellular matrix and perineuronal nets in memory. Mol Psychiatry 2022; 27:3192-3203. [PMID: 35760878 PMCID: PMC9708575 DOI: 10.1038/s41380-022-01634-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/09/2022] [Accepted: 05/16/2022] [Indexed: 02/06/2023]
Abstract
All components of the CNS are surrounded by a diffuse extracellular matrix (ECM) containing chondroitin sulphate proteoglycans (CSPGs), heparan sulphate proteoglycans (HSPGs), hyaluronan, various glycoproteins including tenascins and thrombospondin, and many other molecules that are secreted into the ECM and bind to ECM components. In addition, some neurons, particularly inhibitory GABAergic parvalbumin-positive (PV) interneurons, are surrounded by a more condensed cartilage-like ECM called perineuronal nets (PNNs). PNNs surround the soma and proximal dendrites as net-like structures that surround the synapses. Attention has focused on the role of PNNs in the control of plasticity, but it is now clear that PNNs also play an important part in the modulation of memory. In this review we summarize the role of the ECM, particularly the PNNs, in the control of various types of memory and their participation in memory pathology. PNNs are now being considered as a target for the treatment of impaired memory. There are many potential treatment targets in PNNs, mainly through modulation of the sulphation, binding, and production of the various CSPGs that they contain or through digestion of their sulphated glycosaminoglycans.
Collapse
Affiliation(s)
- James W Fawcett
- John van Geest Centre for Brain Repair, Department Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK.
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic.
| | - Marianne Fyhn
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Pavla Jendelova
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic
| | - Jessica C F Kwok
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic
- School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Jiri Ruzicka
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic
| | - Barbara A Sorg
- Robert S. Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| |
Collapse
|
6
|
Vignoli B, Sansevero G, Sasi M, Rimondini R, Blum R, Bonaldo V, Biasini E, Santi S, Berardi N, Lu B, Canossa M. Astrocytic microdomains from mouse cortex gain molecular control over long-term information storage and memory retention. Commun Biol 2021; 4:1152. [PMID: 34611268 PMCID: PMC8492720 DOI: 10.1038/s42003-021-02678-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 09/15/2021] [Indexed: 12/23/2022] Open
Abstract
Memory consolidation requires astrocytic microdomains for protein recycling; but whether this lays a mechanistic foundation for long-term information storage remains enigmatic. Here we demonstrate that persistent synaptic strengthening invited astrocytic microdomains to convert initially internalized (pro)-brain-derived neurotrophic factor (proBDNF) into active prodomain (BDNFpro) and mature BDNF (mBDNF) for synaptic re-use. While mBDNF activates TrkB, we uncovered a previously unsuspected function for the cleaved BDNFpro, which increases TrkB/SorCS2 receptor complex at post-synaptic sites. Astrocytic BDNFpro release reinforced TrkB phosphorylation to sustain long-term synaptic potentiation and to retain memory in the novel object recognition behavioral test. Thus, the switch from one inactive state to a multi-functional one of the proBDNF provides post-synaptic changes that survive the initial activation. This molecular asset confines local information storage in astrocytic microdomains to selectively support memory circuits. Beatrice Vignoli et al. examine potential molecular mechanisms of long-term storage information in mice. Their results suggest that astrocytes may help convert neuronal BDNF precursor into active prodomain and mature forms to enhance post-synaptic signaling and memory, providing further insight into the development of memory circuits.
Collapse
Affiliation(s)
- Beatrice Vignoli
- Department of Physics, University of Trento, 38123, Povo (TN), Italy. .,Department of Cellular Computational and Integrative Biology (CIBIO), University of Trento, 38123, Povo (TN), Italy.
| | - Gabriele Sansevero
- Neuroscience Institute, National Research Council (IN-CNR), 56100, Pisa, Italy
| | - Manju Sasi
- Institute of Clinical Neurobiology and Department of Neurology, University Hospital Würzburg, 97078, Würzburg, Germany
| | - Roberto Rimondini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy
| | - Robert Blum
- Institute of Clinical Neurobiology and Department of Neurology, University Hospital Würzburg, 97078, Würzburg, Germany
| | - Valerio Bonaldo
- Department of Cellular Computational and Integrative Biology (CIBIO), University of Trento, 38123, Povo (TN), Italy
| | - Emiliano Biasini
- Department of Cellular Computational and Integrative Biology (CIBIO), University of Trento, 38123, Povo (TN), Italy
| | - Spartaco Santi
- Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", National Research Council of Italy, 40136, Bologna, Italy.,IRCCS, Istituto Ortopedico Rizzoli, 40136, Bologna, Italy
| | - Nicoletta Berardi
- Department of Neuroscience, Psychology, Drug Research, Child Health (NEUROFARBA), University of Florence, 50100, Florence, Italy
| | - Bai Lu
- School of Pharmaceutical Sciences, Tsinghua University, 100084, Beijing, China
| | - Marco Canossa
- Department of Cellular Computational and Integrative Biology (CIBIO), University of Trento, 38123, Povo (TN), Italy.
| |
Collapse
|
7
|
Integrated Bioinformatics Analysis to Identify Alternative Therapeutic Targets for Alzheimer's Disease: Insights from a Synaptic Machinery Perspective. J Mol Neurosci 2021; 72:273-286. [PMID: 34414562 DOI: 10.1007/s12031-021-01893-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD), the most common type of dementia, is a serious neurodegenerative disease that has no cure yet, but whose symptoms can be alleviated with available medications. Therefore, early and accurate diagnosis of the disease and elucidation of the molecular mechanisms involved in the progression of pathogenesis are critically important. This study aimed to identify dysregulated miRNAs and their target mRNAs through the integrated analysis of miRNA and mRNA expression profiling in AD patients versus unaffected controls. Expression profiles in postmortem brain samples from AD patients and healthy individuals were extracted from the Gene Expression Omnibus database and were analyzed using bioinformatics approaches to identify gene ontologies, pathways, and networks. Finally, the module analysis of the PPI network and hub gene selection was carried out. A total of five differentially expressed miRNAs were extracted from the miRNA dataset, and 4312 differentially expressed mRNAs were obtained from the mRNA dataset. By comparing the DEGs and the putative targets of the altered miRNAs, 116 (3 upregulated and 113 downregulated) coordinated genes were determined. Also, six hub genes (SNAP25, GRIN2A, GRIN2B, DLG2, ATP2B2, and SCN2A) were identified by constructing a PPI network. The results of the present study provide insight into mechanisms such as synaptic machinery and neuronal communication underlying AD pathogenesis, specifically concerning miRNAs.
Collapse
|
8
|
Zhao W, Hou Y, Song X, Wang L, Zhang F, Zhang H, Yu H, Zhou Y. Estrogen Deficiency Induces Mitochondrial Damage Prior to Emergence of Cognitive Deficits in a Postmenopausal Mouse Model. Front Aging Neurosci 2021; 13:713819. [PMID: 34335235 PMCID: PMC8319728 DOI: 10.3389/fnagi.2021.713819] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/28/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Estrogen deficiency contributes to the development of Alzheimer's disease (AD) in menopausal women. In the current study, we examined the impact of estrogen deficiency on mitochondrial function and cognition using a postmenopausal mouse model. Methods: Bilateral ovariectomy was conducted in adult females C57BL/6J. Cognitive function was examined using the Morris water maze (MWM) test at 2 weeks, 1, 2, and 3 months after ovariectomy. Neurodegeneration was assessed using an immunofluorescence assay of microtubule-associated protein 2 (MAP2) in the hippocampus and immunoblotting against postsynaptic density-95 (PSD95). Mitochondrial function in the hippocampus was assessed using immunoblotting for NDUFB8, SDHB, UQCRC2, MTCO1, and ATP5A1. Mitochondrial biogenesis was examined using immunoblotting for PGC-1α, NRF1, and mtTFA. Mitochondrion fission was assessed with immunoblotting for Drp1, whereas mitochondrion fusion was analyzed with immunoblotting for OPA1 and Mfn2. Mitophagy was examined with immunoblotting for PINK1 and LC3B. Mice receiving sham surgery were used as controls. Results: Ovariectomy resulted in significant learning and memory deficits in the MWM test at 3 months, but not at any earlier time points. At 2 weeks after ovariectomy, levels of Drp1 phosphorylated at Ser637 decreased in the hippocampus. At 1 month after ovariectomy, hippocampal levels of NDUFB8, SDHB, PGC-1α, mtTFA, OPA1, and Mfn2 were significantly reduced. At 2 months after ovariectomy, hippocampal levels of MAP2, PSD95, MTCO1, NRF1, and Pink1 were also reduced. At 3 months, levels of LC3B-II were reduced. Conclusions: The cognitive decline associated with estrogen deficiency is preceded by mitochondrial dysfunction, abnormal mitochondrial biogenesis, irregular mitochondrial dynamics, and decreased mitophagy. Thus, mitochondrial damage may contribute to cognitive impairment associated with estrogen deficiency.
Collapse
Affiliation(s)
- Wei Zhao
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Yue Hou
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Xinxin Song
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Lei Wang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Fangfang Zhang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Hanting Zhang
- Departments of Neuroscience and Behavioral Medicine and Psychiatry, Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Haiyang Yu
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Yanmeng Zhou
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| |
Collapse
|
9
|
Graeve A, Ioannidou I, Reinhard J, Görl DM, Faissner A, Weiss LC. Brain volume increase and neuronal plasticity underly predator-induced morphological defense expression in Daphnia longicephala. Sci Rep 2021; 11:12612. [PMID: 34131219 PMCID: PMC8206331 DOI: 10.1038/s41598-021-92052-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/03/2021] [Indexed: 12/23/2022] Open
Abstract
Predator-induced phenotypic plasticity describes the ability of prey to respond to an increased predation risk by developing adaptive phenotypes. Upon the perception of chemical predator cues, the freshwater crustacean Daphnia longicephala develops defensive crests against its predator Notonecta spec. (Heteroptera). Chemical predator perception initiates a cascade of biological reactions that leads to the development of these morphological features. Neuronal signaling is a central component in this series, however how the nervous system perceives and integrates environmental signals is not well understood. As neuronal activity is often accompanied by functional and structural plasticity of the nervous system, we hypothesized that predator perception is associated with structural and functional changes of nervous tissues. We observe structural plasticity as a volume increase of the central brain, which is independent of the total number of brain cells. In addition, we find functional plasticity in form of an increased number of inhibitory post-synaptic sites during the initial stage of defense development. Our results indicate a structural rewiring of nerve-cell connections upon predator perception and provide important insights into how the nervous system of prey species interprets predator cues and develops cost-benefit optimized defenses.
Collapse
Affiliation(s)
- A Graeve
- Department of Animal Ecology, Evolution and Biodiversity, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - I Ioannidou
- Department of Animal Ecology, Evolution and Biodiversity, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - J Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - D M Görl
- Department of Animal Ecology, Evolution and Biodiversity, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - A Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - L C Weiss
- Department of Animal Ecology, Evolution and Biodiversity, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany.
| |
Collapse
|
10
|
Chemical Stimulation of Rodent and Human Cortical Synaptosomes: Implications in Neurodegeneration. Cells 2021; 10:cells10051174. [PMID: 34065927 PMCID: PMC8151714 DOI: 10.3390/cells10051174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/29/2021] [Accepted: 05/09/2021] [Indexed: 12/14/2022] Open
Abstract
Synaptic plasticity events, including long-term potentiation (LTP), are often regarded as correlates of brain functions of memory and cognition. One of the central players in these plasticity-related phenomena is the α-amino-3-hydroxy-5-methylisoxazole-4-propionate receptor (AMPAR). Increased levels of AMPARs on postsynaptic membranes thus constitute a biochemical measure of LTP. Isolated synaptic terminals (synaptosomes) are an excellent ex vivo tool to monitor synaptic physiology in healthy and diseased brains, particularly in human research. We herein describe three protocols for chemically-induced LTP (cLTP) in synaptosomes from both rodent and human brain tissues. Two of these chemical stimulation protocols are described for the first time in synaptosomes. A pharmacological block of synaptosomal actin dynamics confirmed the efficiency of the cLTP protocols. Furthermore, the study prototypically evaluated the deficiency of cLTP in cortical synaptosomes obtained from human cases of early-onset Alzheimer’s disease (EOAD) and frontotemporal lobar degeneration (FLTD), as well as an animal model that mimics FLTD.
Collapse
|
11
|
He JG, Zhou HY, Xue SG, Lu JJ, Xu JF, Zhou B, Hu ZL, Wu PF, Long LH, Ni L, Jin Y, Wang F, Chen JG. Transcription Factor TWIST1 Integrates Dendritic Remodeling and Chronic Stress to Promote Depressive-like Behaviors. Biol Psychiatry 2021; 89:615-626. [PMID: 33190845 DOI: 10.1016/j.biopsych.2020.09.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 08/20/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Deficiency in neuronal structural plasticity is involved in the development of major depressive disorder. TWIST1, a helix-loop-helix transcription factor that is essential for morphogenesis and organogenesis, is normally expressed at low levels in mature neurons. However, it is poorly understood what role TWIST1 plays in the brain and whether it is involved in the pathophysiology of depression. METHODS Depressive-like behaviors in C57BL/6J mice were developed by chronic social defeat stress. Genetic and pharmacological approaches were used to investigate the role of the TWIST1-miR-214-PPAR-δ signaling pathway in depressive-like behaviors. Molecular biological and morphological studies were performed to define the molecular mechanisms downstream of TWIST1. RESULTS The expression of TWIST1 was positively correlated with depressive behaviors in humans and mice. Chronic stress elevated TWIST1 expression in the medial prefrontal cortex of mice, which was reversed by fluoxetine treatment. While the overexpression of TWIST1 increased susceptibility to stress, the knockdown of TWIST1 prevented the defective morphogenesis of dendrites of pyramidal neurons in layer II/III of the medial prefrontal cortex and alleviated depressive-like behaviors. Mechanistically, this prodepressant property of TWIST1 was mediated, at least in part, through the repression of miR-214-PPAR-δ signaling and mitochondrial function, which was also mimicked by genetic and pharmacological inhibition of PPAR-δ. CONCLUSIONS These results suggest that TWIST1 in the medial prefrontal cortex mediates chronic stress-induced dendritic remodeling and facilitates the occurrence of depressive-like behavior, providing new information for developing drug targets for depression therapy.
Collapse
Affiliation(s)
- Jin-Gang He
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Hai-Yun Zhou
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Shi-Ge Xue
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jia-Jing Lu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jun-Feng Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Bin Zhou
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Zhuang-Li Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, People's Republic of China
| | - Peng-Fei Wu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, People's Republic of China
| | - Li-Hong Long
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, People's Republic of China
| | - Lan Ni
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - You Jin
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Laboratory of Neuropsychiatric Diseases, Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, People's Republic of China; Key Laboratory of Neurological Diseases, Ministry of Education of China, Wuhan, People's Republic of China.
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Laboratory of Neuropsychiatric Diseases, Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, People's Republic of China; Key Laboratory of Neurological Diseases, Ministry of Education of China, Wuhan, People's Republic of China.
| |
Collapse
|
12
|
Fan XY, Shi G, Zhao P. Neonatal Sevoflurane Exposure Impairs Learning and Memory by the Hypermethylation of Hippocampal Synaptic Genes. Mol Neurobiol 2021; 58:895-904. [PMID: 33052583 DOI: 10.1007/s12035-020-02161-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/04/2020] [Indexed: 12/20/2022]
Abstract
Sevoflurane anesthesia is widely used in pediatric patients. Clinical studies report memory impairment in those exposed to general anesthesia early in life. DNA methylation is essential for the modulation of synaptic plasticity through regulating the transcription of synaptic genes. Therefore, we tested whether neonatal sevoflurane exposure affects learning and memory underlying the hippocampal DNA methylation of synaptic genes. Male Sprague-Dawley rats were exposed to 3% sevoflurane or air for 2 h daily from postnatal day 7 (P7) to P9. 5-aza-2-deoxycytidine (5-AZA), an inhibitor of DNA methyltransferases (DNMTs), was intraperitoneally injected 30 min before sevoflurane or air exposure on P7-9. The rats were euthanized 6, 12, 24 h, and 28 days after the last sevoflurane exposure, followed by the determination of global and gene-specific DNA methylation. The expression of synaptic proteins and synaptic density and the transcription of Dnmts and ten eleven translocations (Tets) in the hippocampus were measured. The ability of learning and memory was assessed using Morris water maze, novel object recognition, and intruder tests. Repeated neonatal sevoflurane exposure impaired cognitive, social, and spatial memory. The memory impairment was associated with the increased Dnmt1, Dnmt3a, and 5-methylcytosine level and the decreased Tet1 and 5-hydromethylcytosine level. Sevoflurane subsequently induced hypermethylation of Shank2, Psd95, Syn1, and Syp gene and down-regulated the expression of synaptic proteins, which finally led to the decrease of synaptic density in a time-dependent manner. Notably, 5-AZA pretreatment ameliorated learning and memory in sevoflurane-treated rats. In conclusion, neonatal exposure to sevoflurane can impair learning and memory through DNA methylation of synaptic genes.
Collapse
Affiliation(s)
- Xin-Yu Fan
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, 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, Shenyang, 110004, China.
| |
Collapse
|
13
|
He Y, Xu B, Chen Y, Liu L, Xu L, Chen Y, Long D. Early-life adversity selectively interrupts the dendritic differentiation of dorsolateral striatal neurons in male mice. Brain Struct Funct 2021; 226:397-414. [PMID: 33386419 DOI: 10.1007/s00429-020-02183-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 11/24/2020] [Indexed: 01/04/2023]
Abstract
The effects of early-life adversity (ELA) on dendritic differentiation of striatal neurons were investigated in the dorsal striatum including the dorsomedial striatum and dorsolateral striatum (DMS and DLS, respectively). An animal model of ELA was created by changing the growth environment of newborn mouse pups by giving limited bedding and nesting materials from postnatal day 2 to day 9 (P2-P9). One week after the stress paradigm (P16), the dendritic branches and spines of striatal spiny neurons as well as the synapses represented by postsynaptic density protein-95 (PSD-95) in DMS and DLS were stereologically analyzed. Adverse experience in early life selectively affected the spiny neurons in DLS, leading to abundant proximal dendritic branches and an increased number of filopodia-like protrusions, but a reduced number of dendritic spines. The selective effects of stress on neurons in DLS were further identified by reduced expression of PSD-95, including a reduced optical density of PSD-95 immunoreactivity and fewer individual PSD-95 immunoreactive synapses in this region. Notably, stress in early life affected either D1 or D2 dopamine receptor-expressing DLS neurons. These findings suggest that adverse early-life experience delayed the maturation of dendritic spines on neurons in the dorsolateral striatum. Altered dendritic differentiation provoked by stress in early life may contribute critically to the formation of proper neuronal circuits in the dorsal striatum and, therefore, affect striatum-dependent habitual behavior and emotional function later in life.
Collapse
Affiliation(s)
- Yun He
- Department of Human Anatomy, School of Basic Medical Sciences, Yangtze University, Hubei, 434023, China
| | - Benke Xu
- Department of Human Anatomy, School of Basic Medical Sciences, Yangtze University, Hubei, 434023, China.,Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Hubei, 434020, China
| | - Yan Chen
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangdong, 510260, China
| | - Lian Liu
- Department of Medical Function, School of Basic Medical Sciences, Yangtze University, Hubei, 434023, China
| | - Liping Xu
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yuncai Chen
- Department of Pediatrics, University of California, Irvine, CA, 92697, USA.
| | - Dahong Long
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
14
|
Khan R, Kulasiri D, Samarasinghe S. Functional repertoire of protein kinases and phosphatases in synaptic plasticity and associated neurological disorders. Neural Regen Res 2021; 16:1150-1157. [PMID: 33269764 PMCID: PMC8224123 DOI: 10.4103/1673-5374.300331] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Protein phosphorylation and dephosphorylation are two essential and vital cellular mechanisms that regulate many receptors and enzymes through kinases and phosphatases. Ca2+- dependent kinases and phosphatases are responsible for controlling neuronal processing; balance is achieved through opposition. During molecular mechanisms of learning and memory, kinases generally modulate positively while phosphatases modulate negatively. This review outlines some of the critical physiological and structural aspects of kinases and phosphatases involved in maintaining postsynaptic structural plasticity. It also explores the link between neuronal disorders and the deregulation of phosphatases and kinases.
Collapse
Affiliation(s)
- Raheel Khan
- Centre for Advanced Computational Solutions (C-fACS), Lincoln University; Department of Molecular Biosciences, Lincoln University, Christchurch, New Zealand
| | - Don Kulasiri
- Centre for Advanced Computational Solutions (C-fACS), Lincoln University; Department of Molecular Biosciences, Lincoln University, Christchurch, New Zealand
| | - Sandhya Samarasinghe
- Centre for Advanced Computational Solutions (C-fACS), Lincoln University, Christchurch, New Zealand
| |
Collapse
|
15
|
Kang DE, Woo JA. Cofilin, a Master Node Regulating Cytoskeletal Pathogenesis in Alzheimer's Disease. J Alzheimers Dis 2020; 72:S131-S144. [PMID: 31594228 PMCID: PMC6971827 DOI: 10.3233/jad-190585] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The defining pathological hallmarks of Alzheimer’s disease (AD) are proteinopathies marked by the amyloid-β (Aβ) peptide and hyperphosphorylated tau. In addition, Hirano bodies and cofilin-actin rods are extensively found in AD brains, both of which are associated with the actin cytoskeleton. The actin-binding protein cofilin known for its actin filament severing, depolymerizing, nucleating, and bundling activities has emerged as a significant player in AD pathogenesis. In this review, we discuss the regulation of cofilin by multiple signaling events impinging on LIM kinase-1 (LIMK1) and/or Slingshot homolog-1 (SSH1) downstream of Aβ. Such pathophysiological signaling pathways impact actin dynamics to regulate synaptic integrity, mitochondrial translocation of cofilin to promote neurotoxicity, and formation of cofilin-actin pathology. Other intracellular signaling proteins, such as β-arrestin, RanBP9, Chronophin, PLD1, and 14-3-3 also impinge on the regulation of cofilin downstream of Aβ. Finally, we discuss the role of activated cofilin as a bridge between actin and microtubule dynamics by displacing tau from microtubules, thereby destabilizing tau-induced microtubule assembly, missorting tau, and promoting tauopathy.
Collapse
Affiliation(s)
- David E Kang
- Byrd Institute and Alzheimer's Center, USF Health Morsani College of Medicine, Tampa, FL, USA.,Department of Molecular Medicine, USF Health Morsani College of Medicine, Tampa, FL, USA.,Division of Research, James A. Haley VA Hospital, Tampa, FL, USA
| | - Jung A Woo
- Byrd Institute and Alzheimer's Center, USF Health Morsani College of Medicine, Tampa, FL, USA.,Department of Molecular Pharmacology and Physiology, USF Health Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
16
|
Khlghatyan J, Evstratova A, Bozoyan L, Chamberland S, Chatterjee D, Marakhovskaia A, Soares Silva T, Toth K, Mongrain V, Beaulieu J. Fxr1 regulates sleep and synaptic homeostasis. EMBO J 2020; 39:e103864. [PMID: 32893934 PMCID: PMC7604579 DOI: 10.15252/embj.2019103864] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 08/01/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022] Open
Abstract
The fragile X autosomal homolog 1 (Fxr1) is regulated by lithium and has been GWAS-associated with schizophrenia and insomnia. Homeostatic regulation of synaptic strength is essential for the maintenance of brain functions and involves both cell-autonomous and system-level processes such as sleep. We examined the contribution of Fxr1 to cell-autonomous homeostatic synaptic scaling and neuronal responses to sleep loss, using a combination of gene overexpression and Crispr/Cas9-mediated somatic knockouts to modulate gene expression. Our findings indicate that Fxr1 is downregulated during both scaling and sleep deprivation via a glycogen synthase kinase 3 beta (GSK3β)-dependent mechanism. In both conditions, downregulation of Fxr1 is essential for the homeostatic modulation of surface AMPA receptors and synaptic strength. Preventing the downregulation of Fxr1 during sleep deprivation results in altered EEG signatures. Furthermore, sequencing of neuronal translatomes revealed the contribution of Fxr1 to changes induced by sleep deprivation. These findings uncover a role of Fxr1 as a shared signaling hub between cell-autonomous homeostatic plasticity and system-level responses to sleep loss, with potential implications for neuropsychiatric illnesses and treatments.
Collapse
Affiliation(s)
- Jivan Khlghatyan
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
- Department of Psychiatry and NeuroscienceFaculty of MedicineUniversité LavalQuébec‐CityQCCanada
- Present address:
Department of NeuroscienceNovartis Institutes for Biomedical ResearchCambridgeMAUSA
| | - Alesya Evstratova
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
| | - Lusine Bozoyan
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
| | - Simon Chamberland
- Department of Psychiatry and NeuroscienceFaculty of MedicineUniversité LavalQuébec‐CityQCCanada
- Present address:
NYU Neuroscience InstituteLangone Medical CenterNew York UniversityNew YorkNYUSA
| | | | | | - Tiago Soares Silva
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
| | - Katalin Toth
- Department of Cellular and Molecular MedicineFaculty of MedicineUniversity of OttawaOttawaONCanada
| | - Valerie Mongrain
- Department of NeuroscienceUniversité de Montréal and Center for Advanced Research in Sleep MedicineHôpital du Sacré‐Coeur de Montréal (CIUSSS‐NIM)MontrealQCCanada
| | | |
Collapse
|
17
|
Grijalva LE, Miranda MI, Paredes RG. Differential changes in GAP-43 or synaptophysin during appetitive and aversive taste memory formation. Behav Brain Res 2020; 397:112937. [PMID: 32991926 DOI: 10.1016/j.bbr.2020.112937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 09/19/2020] [Accepted: 09/21/2020] [Indexed: 10/23/2022]
Abstract
Association between events in time and space is a major mechanism for all animals, including humans, which allows them to learn about the world and potentially change their behavior in the future to adapt to different environments. Conditioning taste aversion (CTA) is a single-trial learning paradigm where animals are trained to avoid a novel flavor which is associated with malaise. Many variables can be analyzed with this model and the circuits involved are well described. Thus, the amygdala and the gustatory cortex (GC) are some of the most relevant structures involved in CTA. In the present study we focused in plastic changes that occur during appetitive and/or aversive taste memory formation. Previous studies have demonstrated that memory consolidation, in hippocampal dependent paradigms, induces plastic changes like increase in the concentration of proteins considered as markers of neuronal plasticity, such as the growth associated protein 43 (GAP-43) and synaptophysin (SYN). In the present experiment in male rats we evaluated changes in GAP-43 and SYN expression, using immunofluorescence, induce by the formation of aversive and appetitive taste memory. We found that taste aversive memory formation can induce an increase in GAP-43 in the granular layer of the GC. Furthermore, we also found an increase in SYN expression in both layers of the GC, the basolateral amygdala (BLA) and the central amygdala (CeA). These results suggest that aversive memory representation induces a new circuitry (inferred from an increase in GAP 43). On the other hand, an appetitive taste learning increased SYN expression in the GC (both layers), the BLA and the CeA without any changes in GAP 43. Together these results indicate that aversive memory formation induces structural and synaptic changes, while appetitive memory formation induces synaptic changes; suggesting that aversive and appetitive memories require a different set of cortical and amygdala plastic changes.
Collapse
Affiliation(s)
- Lucia E Grijalva
- Instituto de Neurobiología, UNAM, Campus Juriquilla, Querétaro, 76230, Mexico
| | - María I Miranda
- Instituto de Neurobiología, UNAM, Campus Juriquilla, Querétaro, 76230, Mexico
| | - Raúl G Paredes
- Instituto de Neurobiología, UNAM, Campus Juriquilla, Querétaro, 76230, Mexico; Escuela Nacional de Estudios Superiores, Unidad Juriquilla, UNAM, Querétaro, 76230 Mexico.
| |
Collapse
|
18
|
Conventional and Non-Conventional Roles of Non-Muscle Myosin II-Actin in Neuronal Development and Degeneration. Cells 2020; 9:cells9091926. [PMID: 32825197 PMCID: PMC7566000 DOI: 10.3390/cells9091926] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022] Open
Abstract
Myosins are motor proteins that use chemical energy to produce mechanical forces driving actin cytoskeletal dynamics. In the brain, the conventional non-muscle myosin II (NMII) regulates actin filament cytoskeletal assembly and contractile forces during structural remodeling of axons and dendrites, contributing to morphology, polarization, and migration of neurons during brain development. NMII isoforms also participate in neurotransmission and synaptic plasticity by driving actin cytoskeletal dynamics during synaptic vesicle release and retrieval, and formation, maturation, and remodeling of dendritic spines. NMIIs are expressed differentially in cerebral non-neuronal cells, such as microglia, astrocytes, and endothelial cells, wherein they play key functions in inflammation, myelination, and repair. Besides major efforts to understand the physiological functions and regulatory mechanisms of NMIIs in the nervous system, their contributions to brain pathologies are still largely unclear. Nonetheless, genetic mutations or deregulation of NMII and its regulatory effectors are linked to autism, schizophrenia, intellectual disability, and neurodegeneration, indicating non-conventional roles of NMIIs in cellular mechanisms underlying neurodevelopmental and neurodegenerative disorders. Here, we summarize the emerging biological roles of NMIIs in the brain, and discuss how actomyosin signaling contributes to dysfunction of neurons and glial cells in the context of neurological disorders. This knowledge is relevant for a deep understanding of NMIIs on the pathogenesis and therapeutics of neuropsychiatric and neurodegenerative diseases.
Collapse
|
19
|
Xu F, Zhang X, Wang J, Li X, He B, Xiao F, Yan T, Wu B, Jia Y, Wang Z. Spinosin protects N2a cells from H 2 O 2 -induced neurotoxicity through inactivation of p38MAPK. J Pharm Pharmacol 2020; 72:1607-1614. [PMID: 32667705 DOI: 10.1111/jphp.13334] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/14/2020] [Accepted: 06/21/2020] [Indexed: 01/26/2023]
Abstract
OBJECTIVES Previous studies have suggested that spinosin (SPI) exerted neuroprotective effects through inhibition of oxidative damage, but the underlying mechanisms are still unclear. Herein, the mechanisms underlying the protective effects of SPI against oxidative stress induced by hydrogen peroxide (H2 O2 ) were examined in neuro-2a (N2a) mouse neuroblastoma cells. METHODS N2a cells were pretreated with H2 O2 for 2 h, followed by a 24-h incubation with SPI. Intracellular reactive oxygen species (ROS) production was analysed by flow cytometry. Levels of Aβ1-42 production were determined by ELISA assay. Levels of expression of c-Jun N-terminal kinase (JNK), p-JNK, extracellular signal-regulated kinase (ERK), p-ERK, p38 mitogen-activated protein kinase (p38MAPK), p-p38MAPK, p-Tau (Ser199), p-Tau (Ser202), p-Tau (Ser396), synaptophysin (SYP) and postsynaptic scaffold postsynaptic density-95 (PSD-95) were detected by Western blot analysis. KEY FINDINGS Our results showed that H2 O2 treatment enhanced intracellular ROS production in N2a cells. SPI prevented H2 O2 -induced oxidative damage via inhibiting Aβ1-42 production, decreasing Tau phosphorylation and improving synaptic structural plasticity. Notably, H2 O2 -increased p38MAPK activation was attenuated by SPI administration, and p38MAPK inhibitor BIRB796 markedly reduced H2 O2 -induced oxidative damage in N2a cells. CONCLUSIONS Our findings suggest that SPI protects N2a cells from H2 O2 -induced oxidative damage through inactivation of p38MAPK.
Collapse
Affiliation(s)
- Fanxing Xu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China.,Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China.,State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Lianyungang, China
| | - Xiaoying Zhang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Jinyu Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Xu Li
- Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China.,State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Lianyungang, China
| | - Bosai He
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Feng Xiao
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Tingxu Yan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Bo Wu
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Ying Jia
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhenzhong Wang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China.,State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Lianyungang, China
| |
Collapse
|
20
|
Albanesi JP, Barylko B, DeMartino GN, Jameson DM. Palmitoylated Proteins in Dendritic Spine Remodeling. Front Synaptic Neurosci 2020; 12:22. [PMID: 32655390 PMCID: PMC7325885 DOI: 10.3389/fnsyn.2020.00022] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/12/2020] [Indexed: 12/14/2022] Open
Abstract
Activity-responsive changes in the actin cytoskeleton are required for the biogenesis, motility, and remodeling of dendritic spines. These changes are governed by proteins that regulate the polymerization, depolymerization, bundling, and branching of actin filaments. Thus, processes that have been extensively characterized in the context of non-neuronal cell shape change and migration are also critical for learning and memory. In this review article, we highlight actin regulatory proteins that associate, at least transiently, with the dendritic plasma membrane. All of these proteins have been shown, either in directed studies or in high-throughput screens, to undergo palmitoylation, a potentially reversible, and stimulus-dependent cysteine modification. Palmitoylation increases the affinity of peripheral proteins for the membrane bilayer and contributes to their subcellular localization and recruitment to cholesterol-rich membrane microdomains.
Collapse
Affiliation(s)
- Joseph P. Albanesi
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Barbara Barylko
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - George N. DeMartino
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - David M. Jameson
- Department of Cell and Molecular Biology, University of Hawaii, Honolulu, HI, United States
| |
Collapse
|
21
|
Seaton G, Hodges G, de Haan A, Grewal A, Pandey A, Kasai H, Fox K. Dual-Component Structural Plasticity Mediated by αCaMKII Autophosphorylation on Basal Dendrites of Cortical Layer 2/3 Neurones. J Neurosci 2020; 40:2228-2245. [PMID: 32001612 PMCID: PMC7083283 DOI: 10.1523/jneurosci.2297-19.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/30/2019] [Accepted: 01/03/2020] [Indexed: 11/21/2022] Open
Abstract
Sensory cortex exhibits receptive field plasticity throughout life in response to changes in sensory experience and offers the experimental possibility of aligning functional changes in receptive field properties with underpinning structural changes in synapses. We looked at the effects on structural plasticity of two different patterns of whisker deprivation in male and female mice: chessboard deprivation, which causes functional plasticity; and all deprived, which does not. Using 2-photon microscopy and chronic imaging through a cranial window over the barrel cortex, we found that layer 2/3 neurones exhibit robust structural plasticity, but only in response to whisker deprivation patterns that cause functional plasticity. Chessboard pattern deprivation caused dual-component plasticity in layer 2/3 by (1) increasing production of new spines that subsequently persisted for weeks and (2) enlarging spine head sizes in the preexisting stable spine population. Structural plasticity occurred on basal dendrites, but not apical dendrites. Both components of plasticity were absent in αCaMKII-T286A mutants that lack LTP and experience-dependent potentiation in barrel cortex, implying that αCaMKII autophosphorylation is not only important for stabilization and enlargement of spines, but also for new spine production. These studies therefore reveal the relationship between spared whisker potentiation in layer 2/3 neurones and the form and mechanisms of structural plasticity processes that underlie them.SIGNIFICANCE STATEMENT This study provides a missing link in a chain of reasoning that connects LTP to experience-dependent functional plasticity in vivo We found that increases in dendritic spine formation and spine enlargement (both of which are characteristic of LTP) only occurred in barrel cortex during sensory deprivation that produced potentiation of sensory responses. Furthermore, the dendritic spine plasticity did not occur during sensory deprivation in mice lacking LTP and experience-dependent potentiation (αCaMKII autophosphorylation mutants). We also found that the dual-component dendritic spine plasticity only occurred on basal dendrites and not on apical dendrites, thereby resolving a paradox in the literature suggesting that layer 2/3 neurones lack structural plasticity in response to sensory deprivation.
Collapse
Affiliation(s)
- Gillian Seaton
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, United Kingdom, and
| | - Gladys Hodges
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, United Kingdom, and
| | - Annelies de Haan
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, United Kingdom, and
| | - Aneesha Grewal
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, United Kingdom, and
| | - Anurag Pandey
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, United Kingdom, and
| | - Haruo Kasai
- Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Kevin Fox
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, United Kingdom, and
| |
Collapse
|
22
|
Herrera-Morales WV, Herrera-Solís A, Núñez-Jaramillo L. Sexual Behavior and Synaptic Plasticity. ARCHIVES OF SEXUAL BEHAVIOR 2019; 48:2617-2631. [PMID: 31270644 DOI: 10.1007/s10508-019-01483-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 05/30/2019] [Accepted: 06/01/2019] [Indexed: 06/09/2023]
Abstract
Although sex drive is present in many animal species, sexual behavior is not static and, like many other behaviors, can be modified by experience. This modification relies on synaptic plasticity, a sophisticated mechanism through which neurons change how they process a given stimulus, and the neurophysiological basis of learning. This review addresses the main plastic effects of steroid sex hormones in the central nervous system (CNS) and the effects of sexual experience on the CNS, including effects on neurogenesis, intracellular signaling, gene expression, and changes in dendritic spines, as well as behavioral changes.
Collapse
Affiliation(s)
- Wendy Verónica Herrera-Morales
- División de Ciencias de la Salud, Universidad de Quintana Roo, Av. Erick Paolo Martínez S/N esquina Av 4 de marzo. Colonia Magisterial, 77039, Chetumal, Quintana Roo, Mexico
| | - Andrea Herrera-Solís
- Laboratorio Efectos Terapéuticos de los Canabinoides, Subdirección de Investigación Biomédica, Hospital General Dr. Manuel Gea González, Ciudad de México, Mexico
| | - Luis Núñez-Jaramillo
- División de Ciencias de la Salud, Universidad de Quintana Roo, Av. Erick Paolo Martínez S/N esquina Av 4 de marzo. Colonia Magisterial, 77039, Chetumal, Quintana Roo, Mexico.
| |
Collapse
|
23
|
Sevoflurane Impairs Short-Term Memory by Affecting PSD-95 and AMPA Receptor in the Hippocampus of a Mouse Model. Behav Neurol 2019; 2019:1068260. [PMID: 31772680 PMCID: PMC6854262 DOI: 10.1155/2019/1068260] [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: 03/20/2019] [Revised: 07/23/2019] [Accepted: 09/10/2019] [Indexed: 11/18/2022] Open
Abstract
Objective To explore the effects of sevoflurane on the latency and error times of the passive avoidance and levels of PSD-95 and AMPA receptors in the hippocampus. We evaluated the effects of sevoflurane on short-term memory in adult mice and explored the possible mechanism. Methods 144 Kunming mice (2-3 months, 30-35 g) were randomly divided into two groups A (n = 64) and B (n = 80) and received the dark-avoidance (DA) and step-down avoidance (SA) tests, respectively. The groups DA and SA were further divided into control (inhaled 40% O2 2 h) and sevoflurane (3.3% sevoflurane and 40% O2 2 h) subgroups. Before inhalation intervention, all mice were trained to be familiar with the Morris water maze (MWM). According to the test points of behavioral indicators, 8 mice were randomly selected from each subgroup at point 12 h (T1), 24 h (T2), 48 h (T3), and 72 h (T4) after inhalation intervention. The step-through latency and error times were measured in 5 min. After the behavioral test, the mice were killed and the tissues of the hippocampus were taken for hematoxylin and eosin (H&E) staining. The expression level of PSD-95 and AMPA receptors in the hippocampus was detected by immunohistochemistry and Western Blot. The changes of synaptic transmission were measured via electrophysiology analysis of hippocampal slices. Results The mice in the control subgroups found the platform in a shorter pathway than those in the sevoflurane subgroups during an MWM test. The step-through latency of T1 and T2 in the sevoflurane subgroup was shorter than baseline time, and the error times were increased in 5 min and higher than baseline time when compared with the control subgroup (P < 0.05) in the A and B groups. Compared with the control subgroup, the expression level of PSD-95 and AMPA receptors in the hippocampus was decreased at T1 and T2 in the sevoflurane subgroup (P < 0.05). The nerve cells were partially swelling. Electrophysiology analysis showed that the levels of PSD-95 and AMPA receptor expression were associated with synaptic transmission. Conclusion Sevoflurane impaired short-term memory in adult mice by inhibiting the expression of PSD-95 and AMPA receptors in the hippocampus, which led to the decrease in synaptic transmission.
Collapse
|
24
|
Joshi VV, Patel ND, Rehan MA, Kuppa A. Mysterious Mechanisms of Memory Formation: Are the Answers Hidden in Synapses? Cureus 2019; 11:e5795. [PMID: 31728242 PMCID: PMC6827877 DOI: 10.7759/cureus.5795] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 09/28/2019] [Indexed: 12/18/2022] Open
Abstract
After decades of research on memory formation and retention, we are still searching for the definite concept and process behind neuroplasticity. This review article will address the relationship between synapses, memory formation, and memory retention and their genetic correlations. In the last six decades, there have been enormous improvements in the neurochemistry domain, especially in the area of neural plasticity. In the central nervous system, the complexity of the synapses between neurons allows communication among them. It is believed that each time certain types of sensory signals pass through sequences of synapses, these synapses can transmit the same signals more efficiently the following time. The concept of Hebb synapse has provided revolutionary thinking about the nature of neural mechanisms of learning and memory formation. To improve the local circuitry for memory formation and behavioral change and stabilization in the mammalian central nervous system, long-term potentiation and long-term depression are the crucial components of Hebbian plasticity. In this review, we will be discussing the role of glutamatergic synapses, engram cells, cytokines, neuropeptides, neurosteroids and many aspects, covering the synaptic basis of memory. Lastly, we have tried to cover the etiology of neurodegenerative disorders due to synaptic dysfunction. To enhance pharmacological interventions for neurodegenerative diseases, we need more research in this direction. With the help of technology, and a better understanding of the disease etiology, not only can we identify the missing pieces of synaptic functions, but we might also cure or even prevent serious neurodegenerative diseases like Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Viraj V Joshi
- Neuropsychiatry, California Instititute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Nishita D Patel
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Muhammad Awais Rehan
- Miscellenous, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Annapurna Kuppa
- Internal Medicine and Gastroenterology, University of Michigan, Ann Arbor, USA
| |
Collapse
|
25
|
Amyloid β oligomers suppress excitatory transmitter release via presynaptic depletion of phosphatidylinositol-4,5-bisphosphate. Nat Commun 2019; 10:1193. [PMID: 30867420 PMCID: PMC6416269 DOI: 10.1038/s41467-019-09114-z] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 02/21/2019] [Indexed: 12/21/2022] Open
Abstract
Amyloid β (Aβ) oligomer-induced aberrant neurotransmitter release is proposed to be a crucial early event leading to synapse dysfunction in Alzheimer's disease (AD). In the present study, we report that the release probability (Pr) at the synapse between the Schaffer collateral (SC) and CA1 pyramidal neurons is significantly reduced at an early stage in mouse models of AD with elevated Aβ production. High nanomolar synthetic oligomeric Aβ42 also suppresses Pr at the SC-CA1 synapse in wild-type mice. This Aβ-induced suppression of Pr is mainly due to an mGluR5-mediated depletion of phosphatidylinositol-4,5-bisphosphate (PIP2) in axons. Selectively inhibiting Aβ-induced PIP2 hydrolysis in the CA3 region of the hippocampus strongly prevents oligomeric Aβ-induced suppression of Pr at the SC-CA1 synapse and rescues synaptic and spatial learning and memory deficits in APP/PS1 mice. These results first reveal the presynaptic mGluR5-PIP2 pathway whereby oligomeric Aβ induces early synaptic deficits in AD.
Collapse
|
26
|
Chen Y, Fu AKY, Ip NY. Synaptic dysfunction in Alzheimer's disease: Mechanisms and therapeutic strategies. Pharmacol Ther 2018; 195:186-198. [PMID: 30439458 DOI: 10.1016/j.pharmthera.2018.11.006] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD), the most prevalent neurodegenerative disease in the elderly population, is characterized by progressive cognitive decline and pathological hallmarks of amyloid plaques and neurofibrillary tangles. However, its pathophysiological mechanisms are poorly understood, and diagnostic tools and interventions are limited. Here, we review recent research on the amyloid hypothesis and beta-amyloid-induced dysfunction of neuronal synapses through distinct cell surface receptors. We also review how tau protein leads to synaptotoxicity through pathological modification, localization, and propagation. Finally, we discuss experimental therapeutics for AD and propose potential applications of disease-modifying strategies targeting synaptic failure for improved treatment of AD.
Collapse
Affiliation(s)
- Yu Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China.
| | - Amy K Y Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China
| | - Nancy Y Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China.
| |
Collapse
|
27
|
Sadigh-Eteghad S, Geranmayeh MH, Majdi A, Salehpour F, Mahmoudi J, Farhoudi M. Intranasal cerebrolysin improves cognitive function and structural synaptic plasticity in photothrombotic mouse model of medial prefrontal cortex ischemia. Neuropeptides 2018; 71:61-69. [PMID: 30054019 DOI: 10.1016/j.npep.2018.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/07/2018] [Accepted: 07/08/2018] [Indexed: 12/12/2022]
Abstract
Medial prefrontal cortex (mPFC) ischemia affects post-stroke cognitive outcomes. We aimed to investigate the effects of different doses and routes of cerebrolysin (CBL) on the structural synaptic plasticity and cognitive function after mPFC ischemia in mice. Thence, CBL (1, 2.5 ml/kg/i.p./daily) or (1 ml/kg/i.n./daily), were administrated in photothrombotic mouse model of mPFC ischemia for two weeks. Episodic and spatial memories were assessed by the What-Where-Which (WWWhich) and Barnes tasks. Growth-associated protein 43 (GAP-43), postsynaptic density-95 (PSD-95), and synaptophysin (SYN) levels were measured in the lesioned area using western blot analysis. Dendritic arbors, spine densities, and morphology were assessed via Golgi-Cox staining. Treatment with 2.5 ml/kg/i.p. and 1 ml/kg/i.n. doses attenuated mPFC ischemia-induced episodic and spatial memories impairment. Results showed an obvious increase in the GAP-43, PSD-95 and SYN levels and improvement in the structural synaptic indexes in lesioned area induced by the same doses and routes of CBL. In conclusion, we found that specific doses/routes of CBL have positive effects on the structural synaptic plasticity and cognitive outcomes after mPFC ischemia.
Collapse
Affiliation(s)
- Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical sciences, Tabriz, Iran
| | - Mohammad Hossein Geranmayeh
- Neurosciences Research Center, Tabriz University of Medical sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Science, Tabriz, Iran
| | - Alireza Majdi
- Neurosciences Research Center, Tabriz University of Medical sciences, Tabriz, Iran
| | - Farzad Salehpour
- Neurosciences Research Center, Tabriz University of Medical sciences, Tabriz, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical sciences, Tabriz, Iran
| | - Mehdi Farhoudi
- Neurosciences Research Center, Tabriz University of Medical sciences, Tabriz, Iran.
| |
Collapse
|
28
|
Grunwald Kadow IC. State-dependent plasticity of innate behavior in fruit flies. Curr Opin Neurobiol 2018; 54:60-65. [PMID: 30219668 DOI: 10.1016/j.conb.2018.08.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/28/2018] [Indexed: 01/27/2023]
Abstract
Behaviors are often categorized into innate or learned. Innate behaviors are thought to be genetically encoded and hardwired into the brain, while learned behavior is a product of the interaction between experience and the plasticity of synapses and neurons. Recent work in different models show that innate behavior, too, is plastic and depends on the current behavioral context and the internal state of an animal. Furthermore, these studies suggest that the neural circuits underpinning innate and learned behavior interact and even overlap. For instance, hunger modulates several innate behaviors relying in part on neural circuits required for learning and memory such as the mushroom body in the fruit fly. These new findings suggest that state-dependent innate behavior and learning rely on functionally and anatomically overlapping and shared neural circuits indicating a common evolutionary history.
Collapse
Affiliation(s)
- Ilona C Grunwald Kadow
- Technical University of Munich, School of Life Sciences, ZIEL, Liesel-Beckmann-Str. 4, 85354, Freising, Germany.
| |
Collapse
|
29
|
Harris KP, Littleton JT, Stewart BA. Postsynaptic Syntaxin 4 negatively regulates the efficiency of neurotransmitter release. J Neurogenet 2018; 32:221-229. [PMID: 30175640 PMCID: PMC6317344 DOI: 10.1080/01677063.2018.1501372] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/13/2018] [Indexed: 12/12/2022]
Abstract
Signaling from the postsynaptic compartment regulates multiple aspects of synaptic development and function. Syntaxin 4 (Syx4) is a plasma membrane t-SNARE that promotes the growth and plasticity of Drosophila neuromuscular junctions (NMJs) by regulating the localization of key synaptic proteins in the postsynaptic compartment. Here, we describe electrophysiological analyses and report that loss of Syx4 leads to enhanced neurotransmitter release, despite a decrease in the number of active zones. We describe a requirement for postsynaptic Syx4 in regulating several presynaptic parameters, including Ca2+ cooperativity and the abundance of the presynaptic calcium channel Cacophony (Cac) at active zones. These findings indicate Syx4 negatively regulates presynaptic neurotransmitter release through a retrograde signaling mechanism from the postsynaptic compartment.
Collapse
Affiliation(s)
- Kathryn P Harris
- a Department of Biology , University of Toronto Mississauga , Mississauga , ON, Canada
- b Department of Cell and Systems Biology , University of Toronto , Toronto , ON, Canada
| | - J Troy Littleton
- c The Picower Institute for Learning and Memory , Massachusetts Institute of Technology , Cambridge , MA , USA
- d Department of Biology , Massachusetts Institute of Technology , Cambridge , MA , USA
- e Department of Brain and Cognitive Sciences , Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Bryan A Stewart
- a Department of Biology , University of Toronto Mississauga , Mississauga , ON, Canada
- b Department of Cell and Systems Biology , University of Toronto , Toronto , ON, Canada
| |
Collapse
|
30
|
Borin M, Saraceno C, Catania M, Lorenzetto E, Pontelli V, Paterlini A, Fostinelli S, Avesani A, Di Fede G, Zanusso G, Benussi L, Binetti G, Zorzan S, Ghidoni R, Buffelli M, Bolognin S. Rac1 activation links tau hyperphosphorylation and Aβ dysmetabolism in Alzheimer's disease. Acta Neuropathol Commun 2018; 6:61. [PMID: 30005699 PMCID: PMC6045891 DOI: 10.1186/s40478-018-0567-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/03/2018] [Indexed: 12/22/2022] Open
Abstract
One of the earliest pathological features characterizing Alzheimer’s disease (AD) is the loss of dendritic spines. Among the many factors potentially mediating this loss of neuronal connectivity, the contribution of Rho-GTPases is of particular interest. This family of proteins has been known for years as a key regulator of actin cytoskeleton remodeling. More recent insights have indicated how its complex signaling might be triggered also in pathological conditions. Here, we showed that the Rho-GTPase family member Rac1 levels decreased in the frontal cortex of AD patients compared to non-demented controls. Also, Rac1 increased in plasma samples of AD patients with Mini-Mental State Examination < 18 compared to age-matched non demented controls. The use of different constitutively active peptides allowed us to investigate in vitro Rac1 specific signaling. Its activation increased the processing of amyloid precursor protein and induced the translocation of SET from the nucleus to the cytoplasm, resulting in tau hyperphosphorylation at residue pT181. Notably, Rac1 was abnormally activated in the hippocampus of 6-week-old 3xTg-AD mice. However, the total protein levels decreased at 7-months. A rescue strategy based on the intranasal administration of Rac1 active peptide at 6.5 months prevented dendritic spine loss. This data suggests the intriguing possibility of a dual role of Rac1 according to the different stages of the pathology. In an initial stage, Rac1 deregulation might represent a triggering co-factor due to the direct effect on Aβ and tau. However, at a later stage of the pathology, it might represent a potential therapeutic target due to the beneficial effect on spine dynamics.
Collapse
|
31
|
Sugie A, Marchetti G, Tavosanis G. Structural aspects of plasticity in the nervous system of Drosophila. Neural Dev 2018; 13:14. [PMID: 29960596 PMCID: PMC6026517 DOI: 10.1186/s13064-018-0111-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 06/12/2018] [Indexed: 12/15/2022] Open
Abstract
Neurons extend and retract dynamically their neurites during development to form complex morphologies and to reach out to their appropriate synaptic partners. Their capacity to undergo structural rearrangements is in part maintained during adult life when it supports the animal's ability to adapt to a changing environment or to form lasting memories. Nonetheless, the signals triggering structural plasticity and the mechanisms that support it are not yet fully understood at the molecular level. Here, we focus on the nervous system of the fruit fly to ask to which extent activity modulates neuronal morphology and connectivity during development. Further, we summarize the evidence indicating that the adult nervous system of flies retains some capacity for structural plasticity at the synaptic or circuit level. For simplicity, we selected examples mostly derived from studies on the visual system and on the mushroom body, two regions of the fly brain with extensively studied neuroanatomy.
Collapse
Affiliation(s)
- Atsushi Sugie
- Center for Transdisciplinary Research, Niigata University, Niigata, 951-8585 Japan
- Brain Research Institute, Niigata University, Niigata, 951-8585 Japan
| | | | - Gaia Tavosanis
- Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| |
Collapse
|
32
|
Evidence for altered dendritic spine compartmentalization in Alzheimer's disease and functional effects in a mouse model. Acta Neuropathol 2018; 135:839-854. [PMID: 29696365 DOI: 10.1007/s00401-018-1847-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/30/2018] [Accepted: 04/05/2018] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is associated with a progressive loss of synapses and neurons. Studies in animal models indicate that morphological alterations of dendritic spines precede synapse loss, increasing the proportion of large and short ("stubby") spines. Whether similar alterations occur in human patients, and what their functional consequences could be, is not known. We analyzed biopsies from AD patients and APP x presenilin 1 knock-in mice that were previously shown to present a loss of pyramidal neurons in the CA1 area of the hippocampus. We observed that the proportion of stubby spines and the width of spine necks are inversely correlated with synapse density in frontal cortical biopsies from non-AD and AD patients. In mice, the reduction in the density of synapses in the stratum radiatum was preceded by an alteration of spine morphology, with a reduction of their length and an enlargement of their neck. Serial sectioning examined with electron microscopy allowed us to precisely measure spine parameters. Mathematical modeling indicated that the shortening and widening of the necks should alter the electrical compartmentalization of the spines, leading to reduced postsynaptic potentials in spine heads, but not in soma. Accordingly, there was no alteration in basal synaptic transmission, but long-term potentiation and spatial memory were impaired. These results indicate that an alteration of spine morphology could be involved in the early cognitive deficits associated with AD.
Collapse
|
33
|
McLachlan IG, Beets I, de Bono M, Heiman MG. A neuronal MAP kinase constrains growth of a Caenorhabditis elegans sensory dendrite throughout the life of the organism. PLoS Genet 2018; 14:e1007435. [PMID: 29879119 PMCID: PMC6007932 DOI: 10.1371/journal.pgen.1007435] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 06/19/2018] [Accepted: 05/18/2018] [Indexed: 02/07/2023] Open
Abstract
Neurons develop elaborate morphologies that provide a model for understanding cellular architecture. By studying C. elegans sensory dendrites, we previously identified genes that act to promote the extension of ciliated sensory dendrites during embryogenesis. Interestingly, the nonciliated dendrite of the oxygen-sensing neuron URX is not affected by these genes, suggesting it develops through a distinct mechanism. Here, we use a visual forward genetic screen to identify mutants that affect URX dendrite morphogenesis. We find that disruption of the MAP kinase MAPK-15 or the βH-spectrin SMA-1 causes a phenotype opposite to what we had seen before: dendrites extend normally during embryogenesis but begin to overgrow as the animals reach adulthood, ultimately extending up to 150% of their normal length. SMA-1 is broadly expressed and acts non-cell-autonomously, while MAPK-15 is expressed in many sensory neurons including URX and acts cell-autonomously. MAPK-15 acts at the time of overgrowth, localizes at the dendrite ending, and requires its kinase activity, suggesting it acts locally in time and space to constrain dendrite growth. Finally, we find that the oxygen-sensing guanylate cyclase GCY-35, which normally localizes at the dendrite ending, is localized throughout the overgrown region, and that overgrowth can be suppressed by overexpressing GCY-35 or by genetically mimicking elevated cGMP signaling. These results suggest that overgrowth may correspond to expansion of a sensory compartment at the dendrite ending, reminiscent of the remodeling of sensory cilia or dendritic spines. Thus, in contrast to established pathways that promote dendrite growth during early development, our results reveal a distinct mechanism that constrains dendrite growth throughout the life of the animal, possibly by controlling the size of a sensory compartment at the dendrite ending.
Collapse
Affiliation(s)
- Ian G McLachlan
- Department of Genetics, Harvard Medical School and Boston Children's Hospital, Boston MA, United States of America
| | - Isabel Beets
- Division of Cell Biology, Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Mario de Bono
- Division of Cell Biology, Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Maxwell G Heiman
- Department of Genetics, Harvard Medical School and Boston Children's Hospital, Boston MA, United States of America
| |
Collapse
|
34
|
Russell TA, Grubisha MJ, Remmers CL, Kang SK, Forrest MP, Smith KR, Kopeikina KJ, Gao R, Sweet RA, Penzes P. A Schizophrenia-Linked KALRN Coding Variant Alters Neuron Morphology, Protein Function, and Transcript Stability. Biol Psychiatry 2018; 83:499-508. [PMID: 29241584 PMCID: PMC5809265 DOI: 10.1016/j.biopsych.2017.10.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/12/2017] [Accepted: 10/10/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND Large-scale genetic studies have revealed that rare sequence variants, including single nucleotide variants (SNVs), in glutamatergic synaptic genes are enriched in schizophrenia patients. However, the majority are too rare to show any association with disease and have not been examined functionally. One such SNV, KALRN-P2255T, displays a penetrance that greatly exceeds that of previously identified schizophrenia-associated SNVs. Therefore, we sought to characterize its effects on the function of kalirin (Kal)-9, a dual Ras-related C3 botulinum toxin substrate 1 and Ras homologue gene family, member A (RhoA) guanine nucleotide exchange factor, upregulated in human schizophrenia brain tissue. METHODS Kal9 was overexpressed in primary rat cortical neurons or human embryonic kidney 293 (HEK293) cells. The effects of the P2255T variant on dendritic branching, dendritic spine morphology, protein and messenger RNA stability, and catalytic activity were examined. RESULTS Kal9-P2255T leads to diminished basal dendritic branching and dendritic spine size, compared with wild-type Kal9. The P2255T SNV directly affected Kal9 protein function, causing increased RhoA activation in HEK293 cells, but had no effect on Ras-related C3 botulinum toxin substrate 1 activation. Consistent with human postmortem findings, we found that Kal9-P2255T protein levels were higher than those of wild-type Kal9 in neurons. Increased messenger RNA stability was detected in HEK293 cells, indicating that this was the cause of the higher protein levels. When analyzed together, increased intrinsic RhoA guanine nucleotide exchange factor catalytic activity combined with increased messenger RNA expression led to net enhancement of RhoA activation, known to negatively impact neuronal morphology. CONCLUSIONS Taken together, our data reveal a novel mechanism for disease-associated SNVs and provide a platform for modeling morphological changes in mental disorders.
Collapse
Affiliation(s)
- Theron A. Russell
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Melanie J. Grubisha
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Christine L. Remmers
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Seok Kyu Kang
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Marc P. Forrest
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Katharine R. Smith
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Katherine J. Kopeikina
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Ruoqi Gao
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Robert A. Sweet
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA,VISN 4 Mental Illness Research, Education and Clinical Center (MIRECC), VA Pittsburgh Healthcare System, Pittsburgh, PA
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
35
|
Alexandrov PN, Zhao Y, Jaber V, Cong L, Lukiw WJ. Deficits in the Proline-Rich Synapse-Associated Shank3 Protein in Multiple Neuropsychiatric Disorders. Front Neurol 2017; 8:670. [PMID: 29321759 PMCID: PMC5732231 DOI: 10.3389/fneur.2017.00670] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 11/27/2017] [Indexed: 12/18/2022] Open
Abstract
Signaling between neurons in the human central nervous system (CNS) is accomplished through a highly interconnected network of presynaptic and postsynaptic elements essential in the conveyance of electrical and neurochemical information. One recently characterized core postsynaptic element essential to the efficient operation of this complex network is a relatively abundant ~184.7 kDa proline-rich synapse-associated cytoskeletal protein known as Shank3 (SH3-ankyrin repeat domain; encoded at human chr 22q13.33). In this “Perspectives” article, we review and comment on current advances in Shank3 research and include some original data that show common Shank3 deficits in a number of seemingly unrelated human neurological disorders that include sporadic Alzheimer’s disease (AD), autism spectrum disorder (ASD), bipolar disorder (BD), Phelan–McDermid syndrome (PMS; 22q13.3 deletion syndrome), and schizophrenia (SZ). Shank3 was also found to be downregulated in the CNS of the transgenic AD (TgAD) 5x familial Alzheimer’s disease murine model engineered to overexpress the 42 amino acid amyloid-beta (Aβ42) peptide. Interestingly, the application of known pro-inflammatory stressors, such as the Aβ42 peptide and the metal-neurotoxin aluminum sulfate, to human neuronal–glial cells in primary culture resulted in a significant decrease in the expression of Shank3. These data indicate that deficits in Shank3-expression may be one common denominator linking a wide-range of human neurological disorders that exhibit a progressive or developmental synaptic disorganization that is temporally associated with cognitive decline.
Collapse
Affiliation(s)
| | - Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States.,Department of Anatomy and Cell Biology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Vivian Jaber
- LSU Neuroscience Center, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States
| | - Lin Cong
- LSU Neuroscience Center, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States.,Department of Neurology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Walter J Lukiw
- Russian Academy of Medical Sciences, Moscow, Russia.,LSU Neuroscience Center, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States.,Department of Neurology, Shengjing Hospital, China Medical University, Shenyang, China.,Department of Ophthalmology, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States.,Department of Neurology, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States
| |
Collapse
|