1
|
Carrasco M, Guzman L, Olloquequi J, Cano A, Fortuna A, Vazquez-Carrera M, Verdaguer E, Auladell C, Ettcheto M, Camins A. Licochalcone A prevents cognitive decline in a lipopolysaccharide-induced neuroinflammation mice model. Mol Med 2025; 31:54. [PMID: 39930360 PMCID: PMC11812219 DOI: 10.1186/s10020-025-01106-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
Inflammation plays a key role in the development of neurodegenerative disorders that are currently incurable. Licochalcone A (LCA) has been described as an emerging anti-inflammatory drug with multiple therapeutical properties that could potentially prevent neurodegeneration. However, its neuroprotective mechanism remains unclear. Here, we investigated if LCA prevents cognitive decline induced by Lipopolysaccharide (LPS) and elucidated its potential benefits. For that, 8-week-old C57BL6/J male mice were intraperitonially (i.p.) treated with saline solution or LCA (15 mg/kg/day, 3 times per week) for two weeks. The last day, a single i.p injection of LPS (1 mg/kg) or saline solution was administered 24 h before sacrifice. The results revealed a significant reduction in mRNA expression in genes involved in oxidative stress (Sod1, Cat, Pkm, Pdha1, Ndyfv1, Uqcrb1, Cycs and Cox4i1), metabolism (Slc2a1, Slc2a2, Prkaa1 and Gsk3b) and synapsis (Bdnf, Nrxn3 and Nlgn2) in LPS group compared to saline. These findings were linked to memory impairment and depressive-like behavior observed in this group. Interestingly, LCA protected against LPS alterations through its anti-inflammatory effect, reducing gliosis and regulating M1/M2 markers. Moreover, LCA-treated animals showed a significant improvement of antioxidant mechanisms, such as citrate synthase activity and SOD2. Additionally, LCA demonstrated protection against metabolic disturbances, downregulating GLUT4 and P-AKT, and enhanced the expression of synaptic-related proteins (P-CREB, BDNF, PSD95, DBN1 and NLG3), leading all together to dendritic spine preservation. In conclusion, our results demonstrate that LCA treatment prevents LPS-induced cognitive decline by reducing inflammation, enhancing the antioxidant response, protecting against metabolic disruptions and improving synapsis related mechanisms.
Collapse
Affiliation(s)
- Marina Carrasco
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Laura Guzman
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
| | - Jordi Olloquequi
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Talca, Chile
| | - Amanda Cano
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Ana Fortuna
- Laboratory of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, CIBIT/ICNAS, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Manuel Vazquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Networking Research Centre of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, 28031, Madrid, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), University of Barcelona, 08028, Barcelona, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950, Esplugues de Llobregat, Spain
| | - Ester Verdaguer
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Carme Auladell
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain.
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain.
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| |
Collapse
|
2
|
Xu L, Wang S, Wu L, Cao H, Fan Y, Wang X, Yu Z, Zhou M, Gao R, Wang J. Coprococcus eutactus screened from healthy adolescent attenuates chronic restraint stress-induced depression-like changes in adolescent mice: Potential roles in the microbiome and neurotransmitter modulation. J Affect Disord 2024; 356:737-752. [PMID: 38649105 DOI: 10.1016/j.jad.2024.04.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/20/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024]
Abstract
The onset of depression commonly occurs in adolescence; therefore, depressive prevention and intervention are pivotal during this period. It is becoming evident that neurotransmitter imbalance and gut microbiota dysbiosis are prominent causes of depression. However, the underlying links and mechanisms remain poorly understood. In this study, with 16S ribosomal RNA gene sequencing, genus Coprococcus markedly differentiated between the healthy and unmedicated depressive adolescents. Based on this, transplantation of Coprococcus eutactus (C.e.) was found to dramatically ameliorate the chronic restraint stress (CRS) induced depression-like changes and prevent synaptic loss and glial-stimulated neuroinflammation in mice. The Ultra-high performance liquid chromatography tandem mass spectrometry analysis (UHPLC-MS/MS) further showed that neurotoxic neurotransmitters in kynurenine pathway (KP) such as 3-hydroxykynurenine (3-HK) and 3-hydroxyanthranilic acid (3-HAA) decreased in mouse brains, mechanistically deciphering the transfer of the tryptophan metabolic pathway to serotonin metabolic signaling in the brain after C.e. treatment, which was also verified in the colon. Molecularly, blockage of KP activities mediated by C.e. was ascribed to the restraint of the limit-step enzymes responsible for kynurenine, 3-HK, and quinolinic acid generation. In the colon, C.e. treatment significantly recovered goblet cells and mucus secretion in CRS mice which may ascribe to the rebalance of the disordered gut microbiota, especially Akkermansia, Roseburia, Rikenella, Blautia, and Alloprevotella. Taken together, the current study reveals for the first time the beneficial effects and potential mechanisms of C.e. in ameliorating CRS-induced depression, unraveling the direct links between C.e. treatment and neurotransmitter rebalance, which may provide efficacious therapeutic avenues for adolescent depressive intervention.
Collapse
Affiliation(s)
- Liuting Xu
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Hygienic Analysis and Detection, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Sizhe Wang
- Department of Hygienic Analysis and Detection, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Linlin Wu
- Department of Physical and Chemical Inspection, The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, China
| | - Hui Cao
- Department of Hygienic Analysis and Detection, Nanjing Qixia District Center for Disease Control and Prevention, Nanjing, China
| | - Yichun Fan
- Department of Hygienic Analysis and Detection, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xi Wang
- Department of Hygienic Analysis and Detection, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zheng Yu
- Department of Hygienic Analysis and Detection, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Manfei Zhou
- Department of Hygienic Analysis and Detection, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Rong Gao
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Hygienic Analysis and Detection, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Jun Wang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
3
|
Nunes M, Madeira N, Fonseca R. Cdc42 activation is necessary for heterosynaptic cooperation and competition. Mol Cell Neurosci 2024; 129:103921. [PMID: 38428552 DOI: 10.1016/j.mcn.2024.103921] [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: 09/26/2023] [Revised: 02/18/2024] [Accepted: 02/20/2024] [Indexed: 03/03/2024] Open
Abstract
Synapses change their weights in response to neuronal activity and in turn, neuronal networks alter their response properties and ultimately allow the brain to store information as memories. As for memories, not all events are maintained over time. Maintenance of synaptic plasticity depends on the interplay between functional changes at synapses and the synthesis of plasticity-related proteins that are involved in stabilizing the initial functional changes. Different forms of synaptic plasticity coexist in time and across the neuronal dendritic area. Thus, homosynaptic plasticity refers to activity-dependent synaptic modifications that are input-specific, whereas heterosynaptic plasticity relates to changes in non-activated synapses. Heterosynaptic forms of plasticity, such as synaptic cooperation and competition allow neurons to integrate events that occur separated by relatively large time windows, up to one hour. Here, we show that activation of Cdc42, a Rho GTPase that regulates actin cytoskeleton dynamics, is necessary for the maintenance of long-term potentiation (LTP) in a time-dependent manner. Inhibiting Cdc42 activation does not alter the time-course of LTP induction and its initial expression but blocks its late maintenance. We show that Cdc42 activation is involved in the phosphorylation of cofilin, a protein involved in modulating actin filaments and that weak and strong synaptic activation leads to similar levels on cofilin phosphorylation, despite different levels of LTP expression. We show that Cdc42 activation is required for synapses to interact by cooperation or competition, supporting the hypothesis that modulation of the actin cytoskeleton provides an activity-dependent and time-restricted permissive state of synapses allowing synaptic plasticity to occur. We found that under competition, the sequence in which synapses are activated determines the degree of LTP destabilization, demonstrating that competition is an active destabilization process. Taken together, we show that modulation of actin cytoskeleton by Cdc42 activation is necessary for the expression of homosynaptic and heterosynaptic forms of plasticity. Determining the temporal and spatial rules that determine whether synapses cooperate or compete will allow us to understand how memories are associated.
Collapse
Affiliation(s)
- Mariana Nunes
- Cellular and Systems Neurobiology, NOVA Medical Research, NOVA Medical School, Universidade NOVA de Lisboa, Portugal
| | - Natália Madeira
- Cellular and Systems Neurobiology, NOVA Medical Research, NOVA Medical School, Universidade NOVA de Lisboa, Portugal
| | - Rosalina Fonseca
- Cellular and Systems Neurobiology, NOVA Medical Research, NOVA Medical School, Universidade NOVA de Lisboa, Portugal.
| |
Collapse
|
4
|
McGowan SE, Gilfanov N, Chandurkar MK, Stiber JA, Han SJ. Drebrin is Required for Myosin-facilitated Actin Cytoskeletal Remodeling during Pulmonary Alveolar Development. Am J Respir Cell Mol Biol 2024; 70:308-321. [PMID: 38271699 DOI: 10.1165/rcmb.2023-0229oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 01/25/2024] [Indexed: 01/27/2024] Open
Abstract
Alveolar septation increases gas-exchange surface area and requires coordinated cytoskeletal rearrangement in lung fibroblasts (LFs) to balance the demands of contraction and cell migration. We hypothesized that DBN (drebrin), a modulator of the actin cytoskeleton in neuronal dendrites, regulates the remodeling of the LF cytoskeleton. Using mice bearing a transgelin-Cre-targeted deletion of Dbn in pulmonary fibroblasts and pericytes, we examined alterations in alveolar septal outgrowth, LF spreading and migration, and actomyosin function. The alveolar surface area and number of alveoli were reduced, whereas alveolar ducts were enlarged, in mice bearing the dbn deletion (DBNΔ) compared with their littermates bearing only one dbn-Flox allele (control). Cultured DBNΔ LFs were deficient in their responses to substrate rigidity and migrated more slowly. Drebrin was abundant in the actin cortex and lamella, and the actin fiber orientation was less uniform in lamella of DBNΔ LFs, which limited the development of traction forces and altered focal adhesion dynamics. Actin fiber orientation is regulated by contractile NM2 (nonmuscle myosin-2) motors, which help arrange actin stress fibers into thick ventral actin stress fibers. Using fluorescence anisotropy, we observed regional intracellular differences in myosin regulatory light chain phosphorylation in control LFs that were altered by dbn deletion. Using perturbations to induce and then release stalling of NM2 on actin in LFs from both genotypes, we made predictions explaining how DBN interacts with actin and NM2. These studies provide new insight for diseases such as emphysema and pulmonary fibrosis, in which fibroblasts inappropriately respond to mechanical cues in their environment.
Collapse
Affiliation(s)
- Stephen E McGowan
- Department of Veterans Affairs Medical Center, Iowa City, Iowa
- Department of Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | | | - Mohanish K Chandurkar
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan
| | - Jonathan A Stiber
- Department of Medicine, Duke University, Durham, North Carolina; and
| | - Sangyoon J Han
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan
| |
Collapse
|
5
|
Soundararajan A, Wang T, Pattabiraman PP. Proteomic analysis uncovers clusterin-mediated disruption of actin-based contractile machinery in the trabecular meshwork to lower intraocular pressure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.16.580757. [PMID: 38405803 PMCID: PMC10888873 DOI: 10.1101/2024.02.16.580757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Glaucoma, a major cause of blindness, is characterized by elevated intraocular pressure (IOP) due to improper drainage of aqueous humor via the trabecular meshwork (TM) outflow pathway. Our recent work identified that loss of clusterin resulted in elevated IOP. This study delves deeper to elucidate the role of clusterin in IOP regulation. Employing an ex vivo human anterior segment perfusion model, we established that constitutive expression and secretion as well as exogenous addition of clusterin can significantly lower IOP. Interestingly, clusterin significantly lowered transforming growth factor β2 (TGFβ2)-induced IOP elevation. This effect was linked to the suppression of extracellular matrix (ECM) deposition and, highlighting the crucial role of clusterin in maintaining ECM equilibrium. A comprehensive global proteomic approach revealed the broad impact of clusterin on TM cell structure and function by identifying alterations in protein expression related to cytoskeletal organization, protein processing, and cellular mechanics, following clusterin induction. These findings underscore the beneficial modulation of TM cell structure and functionality by clusterin. Specifically, clusterin influences the actin-cytoskeleton and focal adhesion dynamics, which are instrumental in cell contractility and adhesion processes. Additionally, it suppresses the activity of proteins critical in TGFβ2, G-protein, and JAK-STAT signaling pathways, which are vital for the regulation of ocular pressure. By delineating these targeted effects of clusterin within the TM outflow pathway, our findings pave the way for novel treatment strategies aimed at mitigating the progression of ocular hypertension and glaucoma through targeted molecular interventions.
Collapse
|
6
|
Yamazaki H, Koganezawa N, Yokoo H, Sekino Y, Shirao T. Super-resolution imaging reveals the relationship between CaMKIIβ and drebrin within dendritic spines. Neurosci Res 2024; 199:30-35. [PMID: 37659612 DOI: 10.1016/j.neures.2023.08.002] [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: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/04/2023]
Abstract
Dendritic spines are unique postsynaptic structures that emerge from the dendrites of neurons. They undergo activity-dependent morphological changes known as structural plasticity. The changes involve actin cytoskeletal remodeling, which is regulated by actin-binding proteins. CaMKII is a crucial molecule in synaptic plasticity. Notably, CaMKIIβ subtype is known to bind to filamentous-actin and is closely involved in structural plasticity. We have shown that CaMKIIβ binds to drebrin, and is localized in spines as both drebrin-dependent and drebrin-independent pools. However, the nanoscale relationship between drebrin and CaMKIIβ within dendritic spines has not been clarified. In this study, we used stochastic optical reconstruction microscopy (STORM) to examine the detailed localization of these proteins. STORM imaging showed that CaMKIIβ co-localized with drebrin in the core region of spines, and localized in the submembrane region of spines without drebrin. Interestingly, the dissociation of CaMKIIβ and drebrin in the core region was induced by NMDA receptor activation. In drebrin knockdown neurons, CaMKIIβ was decreased in the core region but not in the submembrane region. Together it indicates that the clustering of CaMKIIβ in the spine core region is dependent on drebrin. These findings suggest that drebrin-dependent CaMKIIβ is in a standby state before its activation.
Collapse
Affiliation(s)
- Hiroyuki Yamazaki
- Faculty of Social Welfare, Gunma University of Health and Welfare, 191-1 Kawamagari-cho, Maebashi 371-0823, Japan; Department of Pharmacology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi 371-8511, Gunma, Japan.
| | - Noriko Koganezawa
- Department of Pharmacology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi 371-8511, Gunma, Japan
| | - Hideaki Yokoo
- Department of Human Pathology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi 371-8511, Gunma, Japan
| | - Yuko Sekino
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan; Institute for Drug Discovery Innovation, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tomoaki Shirao
- AlzMed, Inc, UT South building Entrepreneurs Lab, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8485, Japan
| |
Collapse
|
7
|
Peppercorn K, Kleffmann T, Hughes SM, Tate WP. Secreted Amyloid Precursor Protein Alpha (sAPPα) Regulates the Cellular Proteome and Secretome of Mouse Primary Astrocytes. Int J Mol Sci 2023; 24:ijms24087165. [PMID: 37108327 PMCID: PMC10138557 DOI: 10.3390/ijms24087165] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/23/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Secreted amyloid precursor protein alpha (sAPPα), processed from a parent mammalian brain protein, amyloid precursor protein, can modulate learning and memory. Recently it has been shown to modulate the transcriptome and proteome of human neurons, including proteins with neurological functions. Here, we analysed whether the acute administration of sAPPα facilitated changes in the proteome and secretome of mouse primary astrocytes in culture. Astrocytes contribute to the neuronal processes of neurogenesis, synaptogenesis and synaptic plasticity. Cortical mouse astrocytes in culture were exposed to 1 nM sAPPα, and changes in both the whole-cell proteome (2 h) and the secretome (6 h) were identified with Sequential Window Acquisition of All Theoretical Fragment Ion Spectra-Mass Spectrometry (SWATH-MS). Differentially regulated proteins were identified in both the cellular proteome and secretome that are involved with neurologically related functions of the normal physiology of the brain and central nervous system. Groups of proteins have a relationship to APP and have roles in the modulation of cell morphology, vesicle dynamics and the myelin sheath. Some are related to pathways containing proteins whose genes have been previously implicated in Alzheimer's disease (AD). The secretome is also enriched in proteins related to Insulin Growth Factor 2 (IGF2) signaling and the extracellular matrix (ECM). There is the promise that a more specific investigation of these proteins will help to understand the mechanisms of how sAPPα signaling affects memory formation.
Collapse
Affiliation(s)
- Katie Peppercorn
- Department of Biochemistry, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin 9016, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| | - Torsten Kleffmann
- Research Infrastructure Centre, Division of Health Sciences, University of Otago, Dunedin 9016, New Zealand
| | - Stephanie M Hughes
- Department of Biochemistry, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin 9016, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
- Genetics Otago, University of Otago, Dunedin 9016, New Zealand
| | - Warren P Tate
- Department of Biochemistry, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin 9016, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
8
|
Espinosa-Jiménez T, Cano A, Sánchez-López E, Olloquequi J, Folch J, Bulló M, Verdaguer E, Auladell C, Pont C, Muñoz-Torrero D, Parcerisas A, Camins A, Ettcheto M. A novel rhein-huprine hybrid ameliorates disease-modifying properties in preclinical mice model of Alzheimer's disease exacerbated with high fat diet. Cell Biosci 2023; 13:52. [PMID: 36895036 PMCID: PMC9999531 DOI: 10.1186/s13578-023-01000-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 02/28/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by a polyetiological origin. Despite the global burden of AD and the advances made in AD drug research and development, the cure of the disease remains elusive, since any developed drug has demonstrated effectiveness to cure AD. Strikingly, an increasing number of studies indicate a linkage between AD and type 2 diabetes mellitus (T2DM), as both diseases share some common pathophysiological features. In fact, β-secretase (BACE1) and acetylcholinesterase (AChE), two enzymes involved in both conditions, have been considered promising targets for both pathologies. In this regard, due to the multifactorial origin of these diseases, current research efforts are focusing on the development of multi-target drugs as a very promising option to derive effective treatments for both conditions. In the present study, we evaluated the effect of rhein-huprine hybrid (RHE-HUP), a synthesized BACE1 and AChE inhibitor, both considered key factors not only in AD but also in metabolic pathologies. Thus, the aim of this study is to evaluate the effects of this compound in APP/PS1 female mice, a well-established familial AD mouse model, challenged by high-fat diet (HFD) consumption to concomitantly simulate a T2DM-like condition. RESULTS Intraperitoneal treatment with RHE-HUP in APP/PS1 mice for 4 weeks reduced the main hallmarks of AD, including Tau hyperphosphorylation, Aβ42 peptide levels and plaque formation. Moreover, we found a decreased inflammatory response together with an increase in different synaptic proteins, such as drebrin 1 (DBN1) or synaptophysin, and in neurotrophic factors, especially in BDNF levels, correlated with a recovery in the number of dendritic spines, which resulted in memory improvement. Notably, the improvement observed in this model can be attributed directly to a protein regulation at central level, since no peripheral modification of those alterations induced by HFD consumption was observed. CONCLUSIONS Our results suggest that RHE-HUP could be a new candidate for the treatment of AD, even for individuals with high risk due to peripheral metabolic disturbances, given its multi-target profile which allows for the improvement of some of the most important hallmarks of the disease.
Collapse
Affiliation(s)
- Triana Espinosa-Jiménez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain.,Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Amanda Cano
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain.,Ace Alzheimer Center Barcelona-International University of Catalunya (UIC), Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, Barcelona, Spain
| | - Elena Sánchez-López
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, Barcelona, Spain.,Unit of Synthesis and Biomedical Applications of Peptides, IQAC-CSIC, 08034, Barcelona, Spain
| | - Jordi Olloquequi
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain.,Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Talca, Chile
| | - Jaume Folch
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institut d'Investigació Sanitària Pere Virgili (IISPV), 43201, Reus, Spain.,Nutrition and Metabolic Health Research Group, Institute of Health Pere Virgili-IISPV, 43201, Reus, Spain
| | - Mònica Bulló
- Institut d'Investigació Sanitària Pere Virgili (IISPV), 43201, Reus, Spain.,Nutrition and Metabolic Health Research Group, Institute of Health Pere Virgili-IISPV, 43201, Reus, Spain.,CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, 28029, Madrid, Spain
| | - Ester Verdaguer
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Carme Auladell
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Caterina Pont
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Diego Muñoz-Torrero
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain.,Institute of Biomedicine (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Antoni Parcerisas
- Department of Basic Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain.,Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain. .,Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain. .,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain. .,Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Av. Joan XXIII 27/31, 08028, Barcelona, Spain.
| |
Collapse
|
9
|
Temizer R, Chen YW, Aoki C. Individual differences in the positive outcome from adolescent ketamine treatment in a female mouse model of anorexia nervosa involve drebrin A at excitatory synapses of the medial prefrontal cortex. Synapse 2023; 77:e22253. [PMID: 36121749 PMCID: PMC9691557 DOI: 10.1002/syn.22253] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/16/2022] [Accepted: 09/04/2022] [Indexed: 01/29/2023]
Abstract
Anorexia nervosa (AN) is a mental illness with the highest rates of mortality and relapse, and no approved pharmacological treatment. Using an animal model of AN, called activity-based anorexia (ABA), we showed earlier that a single intraperitoneal injection of ketamine at a dose of 30 mg/kg (30mgKET), but not 3 mg/kg (3mgKET), has a long-lasting effect upon adolescent females of ameliorating anorexia-like symptoms through the following changes: enhanced food consumption and body weight; reduced running and anxiety-like behavior. However, there were also individual differences in the drug's efficacy. We hypothesized that individual differences in ketamine's ameliorative effects involve drebrin A, an F-actin-binding protein known to be required for the activity-dependent trafficking of NMDA receptors (NMDARs). We tested this hypothesis by electron microscopic quantifications of drebrin A immunoreactivity at excitatory synapses of pyramidal neurons (PN) and GABAergic interneurons (GABA-IN) in deep layer 1 of prefrontal cortex (PFC) of these mice. Results reveal that (1) the areal density of excitatory synapses on GABA-IN is greater for the 30mgKET group than the 3mgKET group; (2) the proportion of drebrin A+ excitatory synapses is greater for both PN and GABA-IN of 30mgKET than 3mgKET group. Correlation analyses with behavioral measurements revealed that (3) 30mgKET's protection is associated with reduced levels of drebrin A in the cytoplasm of GABA-IN and higher levels at extrasynaptic membranous sites of PN and GABA-IN; (5) altogether pointing to 30mgKET-induced homeostatic plasticity that engages drebrin A at excitatory synapses of both PN and GABA-IN.
Collapse
Affiliation(s)
- Rose Temizer
- Center for Neural Science, New York University, New York City, New York, USA
| | - Yi-Wen Chen
- Center for Neural Science, New York University, New York City, New York, USA
| | - Chiye Aoki
- Center for Neural Science, New York University, New York City, New York, USA
| |
Collapse
|
10
|
Chen C, Chu CH, Chu Y, Chang TY, Chen SW, Liang SY, Tsai YC, Chen BC, Tu HL, Cheng PL. Neuronal paxillin and drebrin mediate BDNF-induced force transduction and growth cone turning in a soft-tissue-like environment. Cell Rep 2022; 40:111188. [PMID: 35977504 DOI: 10.1016/j.celrep.2022.111188] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 03/23/2022] [Accepted: 07/20/2022] [Indexed: 11/26/2022] Open
Abstract
Soft tissue environments govern neuronal morphogenesis. However, the precise molecular mechanisms underlying chemotropism-directed axonal growth cone movement in extremely soft environments remain unclear. Here, we show that drebrin, a growth cone T-zone protein, modulates growth cone turning in response to brain-derived neurotrophic factor (BDNF) coated on a soft substrate. Structurally, axonal growth cones of rodent hippocampal neurons grown on 0.1 kPa hydrogels possess an expanded T zone in which drebrin is highly integrated with both F-actin and microtubules. Biochemically, we identify paxillin as interacting with drebrin in cells grown on 0.1 kPa hydrogels but not on glass coverslips. When grown on 0.1 kPa substrates, growth cones asymmetrically exposed to BDNF-bound stripes exhibit enhanced paxillin-drebrin interaction on the side facing the stripes, an activity that is PKA and AAK1 dependent but independent of Src kinase. Functionally, we show that BDNF-induced growth cone turning and force generation on soft substrates require drebrin phosphorylation and paxillin-drebrin association.
Collapse
Affiliation(s)
- Chen Chen
- Institute of Molecular Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Chien-Hsin Chu
- Institute of Molecular Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Ying Chu
- Institute of Molecular Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Ting-Ya Chang
- Institute of Molecular Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Sheng-Wen Chen
- Institute of Molecular Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Shu-Yang Liang
- Institute of Molecular Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Yun-Chi Tsai
- Research Center for Applied Sciences, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Bi-Chang Chen
- Research Center for Applied Sciences, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Hsiung-Lin Tu
- Institute of Chemistry, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Pei-Lin Cheng
- Institute of Molecular Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan.
| |
Collapse
|
11
|
Espinosa-Jiménez T, Busquets O, Cano A, Sánchez-López E, Verdaguer E, Parcerisas A, Olloquequi J, Auladell C, Folch J, Wahli W, Vázquez-Carrera M, Camins A, Ettcheto M. Peroxisomal Proliferator-Activated Receptor β/δ Deficiency Induces Cognitive Alterations. Front Pharmacol 2022; 13:902047. [PMID: 35899125 PMCID: PMC9310104 DOI: 10.3389/fphar.2022.902047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Peroxisome proliferator-activated receptor β/δ (PPARβ/δ), the most PPAR abundant isotype in the central nervous system, is involved in microglial homeostasis and metabolism, whose disturbances have been demonstrated to play a key role in memory impairment. Although PPARβ/δ function is well-established in metabolism, its contribution to neuronal and specifically memory process is underexplored. Therefore, the aim of the study is to determine the role of PPARβ/δ in the neuropathological pathways involved in memory impairment and as to whether a risk factor implicated in memory loss such as obesity modulates neuropathological markers. To carry out this study, 6-month-old total knock-out for the Ppard gene male mice with C57BL/6X129/SV background (PPARβ/δ-/-) and wild-type (WT) littermates with the same genetic background were used. Animals were fed, after the weaning (at 21 days old), and throughout their growth, either conventional chow (CT) or a palmitic acid-enriched diet (HFD). Thus, four groups were defined: WT CT, WT HFD, PPARβ/δ-/- CT, and PPARβ/δ-/- HFD. Before sacrifice, novel object recognition test (NORT) and glucose and insulin tolerance tests were performed. After that, animals were sacrificed by intracardiac perfusion or cervical dislocation. Different techniques, such as GolgiStain kit or immunofluorescence, were used to evaluate the role of PPARβ/δ in memory dysfunction. Our results showed a decrease in dendritic spine density and synaptic markers in PPARβ/δ-/- mice, which were corroborated in the NORT. Likewise, our study demonstrated that the lack of PPARβ/δ receptor enhances gliosis in the hippocampus, contributing to astrocyte and microglial activation and to the increase in neuroinflammatory biomarkers. Additionally, alterations in the hippocampal insulin receptor pathway were found. Interestingly, while some of the disturbances caused by the lack of PPARβ/δ were not affected by feeding the HFD, others were exacerbated or required the combination of both factors. Taken together, the loss of PPARβ/δ-/- affects neuronal and synaptic structure, contributing to memory dysfunction, and they also present this receptor as a possible new target for the treatment of memory impairment.
Collapse
Affiliation(s)
- Triana Espinosa-Jiménez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Oriol Busquets
- Dominick P. Purpura Department of Neurosciences, Albert Einstein College of Medicine, New York City, NY, United States
| | - Amanda Cano
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
- Research Center and Memory Clinic, Fundació ACE Institut Català de Neurociències Aplicades—International University of Catalunya (UIC), Barcelona, Spain
| | - Elena Sánchez-López
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
- Unit of Synthesis and Biomedical Applications of Peptides, IQAC-CSIC, Barcelona, Spain
| | - Ester Verdaguer
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Antoni Parcerisas
- Departament of Basic Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Jordi Olloquequi
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
| | - Carme Auladell
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Jaume Folch
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Life Science, University Rovira i Virgili, Reus, Spain
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- ToxAlim (Research Center in Food Toxicology), INRAE, Toulouse Cedex, France
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
- *Correspondence: Miren Ettcheto,
| |
Collapse
|
12
|
Kubota H, Ogawa H, Miyazaki M, Ishii S, Oyama K, Kawamura Y, Ishiwata S, Suzuki M. Microscopic Temperature Control Reveals Cooperative Regulation of Actin-Myosin Interaction by Drebrin E. NANO LETTERS 2021; 21:9526-9533. [PMID: 34751025 DOI: 10.1021/acs.nanolett.1c02955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Drebrin E is a regulatory protein of intracellular force produced by actomyosin complexes, that is, myosin molecular motors interacting with actin filaments. The expression level of drebrin E in nerve cells decreases as the animal grows, suggesting its pivotal but unclarified role in neuronal development. Here, by applying the microscopic heat pulse method to actomyosin motility assay, the regulatory mechanism is examined from the room temperature up to 37 °C without a thermal denaturing of proteins. We show that the inhibition of actomyosin motility by drebrin E is eliminated immediately and reversibly during heating and depends on drebrin E concentration. The direct observation of quantum dot-labeled drebrin E implies its stable binding to actin filaments during the heat-induced sliding. Our results suggest that drebrin E allosterically modifies the actin filament structure to regulate cooperatively the actomyosin activity at the maintained in vivo body temperature.
Collapse
Affiliation(s)
- Hiroaki Kubota
- Department of Physics, Faculty of Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, 3-24-1 Hyakunincho, Shinjuku-ku, Tokyo 169-0073, Japan
| | - Hiroyuki Ogawa
- Department of Physics, Faculty of Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Makito Miyazaki
- Hakubi Center for Advanced Research, Kyoto University, Yoshida-honmachi, Sakyo-ku, Kyoto 606-8501, Japan
- Department of Physics, Graduate School of Science, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan
- PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris F-75005, France
| | - Shuya Ishii
- Department of Physics, Faculty of Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
- Quantum Beam Science Research Directorate, National Institutes for Quantum and Radiological Science and Technology, 1233 Watanuki-machi, Takasaki, Gunma 370-1292, Japan
| | - Kotaro Oyama
- Department of Physics, Faculty of Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
- PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
- Quantum Beam Science Research Directorate, National Institutes for Quantum and Radiological Science and Technology, 1233 Watanuki-machi, Takasaki, Gunma 370-1292, Japan
| | - Yuki Kawamura
- Department of Physics, Faculty of Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Shin'ichi Ishiwata
- Department of Physics, Faculty of Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Madoka Suzuki
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
13
|
Karnam S, Maddala R, Stiber JA, Rao PV. Drebrin, an actin-binding protein, is required for lens morphogenesis and growth. Dev Dyn 2021; 250:1600-1617. [PMID: 33896079 PMCID: PMC8542647 DOI: 10.1002/dvdy.353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Lens morphogenesis, architecture, and clarity are known to be critically dependent on actin cytoskeleton organization and cell adhesive interactions. There is limited knowledge, however regarding the identity and role of key proteins regulating actin cytoskeletal organization in the lens. This study investigated the role of drebrin, a developmentally regulated actin-binding protein, in mouse lens development by generating and characterizing a conditional knockout (cKO) mouse model using the Cre-LoxP recombination approach. RESULTS Drebrin E, a splice variant of DBN1 is a predominant isoform expressed in the mouse lens and exhibits a maturation-dependent downregulation. Drebrin co-distributes with actin in both epithelium and fibers. Conditional deficiency (both haploinsufficiency and complete absence) of drebrin results in disrupted lens morphogenesis leading to cataract and microphthalmia. The drebrin cKO lens reveals a dramatic decrease in epithelial height and width, E-cadherin, and proliferation, and increased apoptotic cell death and expression of α-smooth muscle actin, together with severely impaired fiber cell organization, polarity, and cell-cell adhesion. CONCLUSIONS This study demonstrates the requirement of drebrin in lens development and growth, with drebrin deficiency leading to impaired lens morphogenesis and microphthalmia.
Collapse
Affiliation(s)
- Shruthi Karnam
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC. USA
| | - Rupalatha Maddala
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC. USA
| | - Jonathan A Stiber
- Department of Medicine, Duke University School of Medicine, Durham, NC. USA
| | - Ponugoti V Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC. USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC. USA
| |
Collapse
|
14
|
Cuestas Torres DM, Cardenas FP. Synaptic plasticity in Alzheimer's disease and healthy aging. Rev Neurosci 2021; 31:245-268. [PMID: 32250284 DOI: 10.1515/revneuro-2019-0058] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022]
Abstract
The strength and efficiency of synaptic connections are affected by the environment or the experience of the individual. This property, called synaptic plasticity, is directly related to memory and learning processes and has been modeled at the cellular level. These types of cellular memory and learning models include specific stimulation protocols that generate a long-term strengthening of the synapses, called long-term potentiation, or a weakening of the said long-term synapses, called long-term depression. Although, for decades, researchers have believed that the main cause of the cognitive deficit that characterizes Alzheimer's disease (AD) and aging was the loss of neurons, the hypothesis of an imbalance in the cellular and molecular mechanisms of synaptic plasticity underlying this deficit is currently widely accepted. An understanding of the molecular and cellular changes underlying the process of synaptic plasticity during the development of AD and aging will direct future studies to specific targets, resulting in the development of much more efficient and specific therapeutic strategies. In this review, we classify, discuss, and describe the main findings related to changes in the neurophysiological mechanisms of synaptic plasticity in excitatory synapses underlying AD and aging. In addition, we suggest possible mechanisms in which aging can become a high-risk factor for the development of AD and how its development could be prevented or slowed.
Collapse
Affiliation(s)
- Diana Marcela Cuestas Torres
- Departamento de Psicología and Departamento de Biología, Laboratorio de Neurociencia y Comportamiento, Universidad de los Andes, Cra 1 N° 18A-12, CP 111711, Bogotá, Colombia
| | - Fernando P Cardenas
- Departamento de Psicología, Laboratorio de Neurociencia y Comportamiento, Universidad de los Andes, Cra 1 N° 18A-12, CP 111711, Bogotá, Colombia
| |
Collapse
|
15
|
Alvarez-Suarez P, Nowak N, Protasiuk-Filipunas A, Yamazaki H, Prószyński TJ, Gawor M. Drebrin Regulates Acetylcholine Receptor Clustering and Organization of Microtubules at the Postsynaptic Machinery. Int J Mol Sci 2021; 22:9387. [PMID: 34502296 PMCID: PMC8430516 DOI: 10.3390/ijms22179387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 01/07/2023] Open
Abstract
Proper muscle function depends on the neuromuscular junctions (NMJs), which mature postnatally to complex "pretzel-like" structures, allowing for effective synaptic transmission. Postsynaptic acetylcholine receptors (AChRs) at NMJs are anchored in the actin cytoskeleton and clustered by the scaffold protein rapsyn, recruiting various actin-organizing proteins. Mechanisms driving the maturation of the postsynaptic machinery and regulating rapsyn interactions with the cytoskeleton are still poorly understood. Drebrin is an actin and microtubule cross-linker essential for the functioning of the synapses in the brain, but its role at NMJs remains elusive. We used immunohistochemistry, RNA interference, drebrin inhibitor 3,5-bis-trifluoromethyl pyrazole (BTP2) and co-immunopreciptation to explore the role of this protein at the postsynaptic machinery. We identify drebrin as a postsynaptic protein colocalizing with the AChRs both in vitro and in vivo. We also show that drebrin is enriched at synaptic podosomes. Downregulation of drebrin or blocking its interaction with actin in cultured myotubes impairs the organization of AChR clusters and the cluster-associated microtubule network. Finally, we demonstrate that drebrin interacts with rapsyn and a drebrin interactor, plus-end-tracking protein EB3. Our results reveal an interplay between drebrin and cluster-stabilizing machinery involving rapsyn, actin cytoskeleton, and microtubules.
Collapse
Affiliation(s)
- Paloma Alvarez-Suarez
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (P.A.-S.); (N.N.); (A.P.-F.); (T.J.P.)
| | - Natalia Nowak
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (P.A.-S.); (N.N.); (A.P.-F.); (T.J.P.)
| | - Anna Protasiuk-Filipunas
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (P.A.-S.); (N.N.); (A.P.-F.); (T.J.P.)
| | - Hiroyuki Yamazaki
- Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan;
| | - Tomasz J. Prószyński
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (P.A.-S.); (N.N.); (A.P.-F.); (T.J.P.)
| | - Marta Gawor
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (P.A.-S.); (N.N.); (A.P.-F.); (T.J.P.)
| |
Collapse
|
16
|
Jia Y, Zhao Q, Yin H, Guo S, Sun M, Yang Z, Zhao X. Reaction-Diffusion Model-Based Research on Formation Mechanism of Neuron Dendritic Spine Patterns. Front Neurorobot 2021; 15:563682. [PMID: 34194309 PMCID: PMC8236519 DOI: 10.3389/fnbot.2021.563682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 05/17/2021] [Indexed: 11/13/2022] Open
Abstract
The pattern abnormalities of dendritic spine, tiny protrusions on neuron dendrites, have been found related to multiple nervous system diseases, such as Parkinson's disease and schizophrenia. The determination of the factors affecting spine patterns is of vital importance to explore the pathogenesis of these diseases, and further, search the treatment method for them. Although the study of dendritic spines is a hot topic in neuroscience in recent years, there is still a lack of systematic study on the formation mechanism of its pattern. This paper provided a reinterpretation of reaction-diffusion model to simulate the formation process of dendritic spine, and further, study the factors affecting spine patterns. First, all four classic shapes of spines, mushroom-type, stubby-type, thin-type, and branched-type were reproduced using the model. We found that the consumption rate of substrates by the cytoskeleton is a key factor to regulate spine shape. Moreover, we found that the density of spines can be regulated by the amount of an exogenous activator and inhibitor, which is in accordance with the anatomical results found in hippocampal CA1 in SD rats with glioma. Further, we analyzed the inner mechanism of the above model parameters regulating the dendritic spine pattern through Turing instability analysis and drew a conclusion that an exogenous inhibitor and activator changes Turing wavelength through which to regulate spine densities. Finally, we discussed the deep regulation mechanisms of several reported regulators of dendritic spine shape and densities based on our simulation results. Our work might evoke attention to the mathematic model-based pathogenesis research for neuron diseases which are related to the dendritic spine pattern abnormalities and spark inspiration in the treatment research for these diseases.
Collapse
Affiliation(s)
- Yiqing Jia
- Institute of Robotics and Automatic Information Systems, College of Artificial Intelligence, Nankai University, Tianjin, China
| | - Qili Zhao
- Institute of Robotics and Automatic Information Systems, College of Artificial Intelligence, Nankai University, Tianjin, China
| | - Hongqiang Yin
- State Key Laboratory of Medicinal Chemical Biology, School of Medicine, Nankai University, Tianjin, China
| | - Shan Guo
- Institute of Robotics and Automatic Information Systems, College of Artificial Intelligence, Nankai University, Tianjin, China
| | - Mingzhu Sun
- Institute of Robotics and Automatic Information Systems, College of Artificial Intelligence, Nankai University, Tianjin, China
| | - Zhuo Yang
- State Key Laboratory of Medicinal Chemical Biology, School of Medicine, Nankai University, Tianjin, China
| | - Xin Zhao
- Institute of Robotics and Automatic Information Systems, College of Artificial Intelligence, Nankai University, Tianjin, China
| |
Collapse
|
17
|
Regulation of actin dynamics in dendritic spines: Nanostructure, molecular mobility, and signaling mechanisms. Mol Cell Neurosci 2020; 109:103564. [DOI: 10.1016/j.mcn.2020.103564] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/04/2020] [Indexed: 12/16/2022] Open
|
18
|
Dombroski TCD, Peixoto-Santos JE, Maciel K, Baqui MMA, Velasco TR, Sakamoto AC, Assirati JA, Carlotti CG, Machado HR, Sousa GKD, Hanamura K, Leite JP, Costa da Costa J, Palmini AL, Paglioli E, Neder L, Spreafico R, Shirao T, Garbelli R, Martins AR. Drebrin expression patterns in patients with refractory temporal lobe epilepsy and hippocampal sclerosis. Epilepsia 2020; 61:1581-1594. [PMID: 32662890 DOI: 10.1111/epi.16595] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/08/2020] [Accepted: 06/08/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Drebrins are crucial for synaptic function and dendritic spine development, remodeling, and maintenance. In temporal lobe epilepsy (TLE) patients, a significant hippocampal synaptic reorganization occurs, and synaptic reorganization has been associated with hippocampal hyperexcitability. This study aimed to evaluate, in TLE patients, the hippocampal expression of drebrin using immunohistochemistry with DAS2 or M2F6 antibodies that recognize adult (drebrin A) or adult and embryonic (pan-drebrin) isoforms, respectively. METHODS Hippocampal sections from drug-resistant TLE patients with hippocampal sclerosis (HS; TLE, n = 33), of whom 31 presented with type 1 HS and two with type 2 HS, and autopsy control cases (n = 20) were assayed by immunohistochemistry and evaluated for neuron density, and drebrin A and pan-drebrin expression. Double-labeling immunofluorescences were performed to localize drebrin A-positive spines in dendrites (MAP2), and to evaluate whether drebrin colocalizes with inhibitory (GAD65) and excitatory (VGlut1) presynaptic markers. RESULTS Compared to controls, TLE patients had increased pan-drebrin in all hippocampal subfields and increased drebrin A-immunopositive area in all hippocampal subfields but CA1. Drebrin-positive spine density followed the same pattern as total drebrin quantification. Confocal microscopy indicated juxtaposition of drebrin-positive spines with VGlut1-positive puncta, but not with GAD65-positive puncta. Drebrin expression in the dentate gyrus of TLE cases was associated negatively with seizure frequency and positively with verbal memory. TLE patients with lower drebrin-immunopositive area in inner molecular layer (IML) than in outer molecular layer (OML) had a lower seizure frequency than those with higher or comparable drebrin-immunopositive area in IML compared with OML. SIGNIFICANCE Our results suggest that changes in drebrin-positive spines and drebrin expression in the dentate gyrus of TLE patients are associated with lower seizure frequency, more preserved verbal memory, and a better postsurgical outcome.
Collapse
Affiliation(s)
| | - Jose Eduardo Peixoto-Santos
- Discipline of Neuroscience, Department of Neurology and Neurosurgery, Paulista Medical School, UNIFESP, São Paulo, Brazil
| | - Karina Maciel
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Munira Muhammad Abdel Baqui
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Tonicarlo Rodrigues Velasco
- Ribeirao Preto Epilepsy Surgery Center, Clinics Hospital, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Americo Ceiki Sakamoto
- Ribeirao Preto Epilepsy Surgery Center, Clinics Hospital, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João Alberto Assirati
- Department of Surgery, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Carlos Gilberto Carlotti
- Department of Surgery, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Hélio Rubens Machado
- Department of Surgery, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Gleice Kelly de Sousa
- Graduate Program of Health Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Kenji Hanamura
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - João Pereira Leite
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jaderson Costa da Costa
- Department of Internal Medicine, School of Medicine, Epilepsy Surgery Program and Brain Institute, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - André Luiz Palmini
- Department of Internal Medicine, School of Medicine, Epilepsy Surgery Program and Brain Institute, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Eliseu Paglioli
- Department of Surgery, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Luciano Neder
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Roberto Spreafico
- Clinical Epileptology and Experimental Neurophysiology Unit, Scientific Institute for Research and Health Care Foundation Carlo Besta Neurological Institute, Milan, Italy
| | - Tomoaki Shirao
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Rita Garbelli
- Clinical Epileptology and Experimental Neurophysiology Unit, Scientific Institute for Research and Health Care Foundation Carlo Besta Neurological Institute, Milan, Italy
| | - Antonio Roberto Martins
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Institute for Neuroscience and Behavior, Ribeirão Preto, Brazil
| |
Collapse
|
19
|
Three-dimensional real time imaging of amyloid β aggregation on living cells. Sci Rep 2020; 10:9742. [PMID: 32546691 PMCID: PMC7297742 DOI: 10.1038/s41598-020-66129-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 05/13/2020] [Indexed: 01/17/2023] Open
Abstract
Alzheimer’s disease (AD) is a progressive disorder of the brain that gradually decreases thinking, memory, and language abilities. The aggregation process of amyloid β (Aβ) is a key step in the expression of its neurocytotoxicity and development of AD because Aβ aggregation and accumulation around neuronal cells induces cell death. However, the molecular mechanism underlying the neurocytotoxicity and cell death by Aβ aggregation has not been clearly elucidated. In this study, we successfully visualized real-time process of Aβ42 aggregation around living cells by applying our established QD imaging method. 3D observations using confocal laser microscopy revealed that Aβ42 preferentially started to aggregate at the region where membrane protrusions frequently formed. Furthermore, we found that inhibition of actin polymerization using cytochalasin D reduced aggregation of Aβ42 on the cell surface. These results indicate that actin polymerization-dependent cell motility is responsible for the promotion of Aβ42 aggregation at the cell periphery. 3D observation also revealed that the aggregates around the cell remained in that location even if cell death occurred, implying that amyloid plaques found in the AD brain grew from the debris of dead cells that accumulated Aβ42 aggregates.
Collapse
|
20
|
Pinho J, Marcut C, Fonseca R. Actin remodeling, the synaptic tag and the maintenance of synaptic plasticity. IUBMB Life 2020; 72:577-589. [DOI: 10.1002/iub.2261] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 02/06/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Júlia Pinho
- Cellular and Systems Neurobiology, Chronic Disease Research CenterNOVA Medical School Lisbon Portugal
| | - Cristina Marcut
- Cellular and Systems Neurobiology, Chronic Disease Research CenterNOVA Medical School Lisbon Portugal
| | - Rosalina Fonseca
- Cellular and Systems Neurobiology, Chronic Disease Research CenterNOVA Medical School Lisbon Portugal
| |
Collapse
|
21
|
Dendritic Spines in Alzheimer's Disease: How the Actin Cytoskeleton Contributes to Synaptic Failure. Int J Mol Sci 2020; 21:ijms21030908. [PMID: 32019166 PMCID: PMC7036943 DOI: 10.3390/ijms21030908] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/24/2020] [Accepted: 01/26/2020] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by Aβ-driven synaptic dysfunction in the early phases of pathogenesis. In the synaptic context, the actin cytoskeleton is a crucial element to maintain the dendritic spine architecture and to orchestrate the spine’s morphology remodeling driven by synaptic activity. Indeed, spine shape and synaptic strength are strictly correlated and precisely governed during plasticity phenomena in order to convert short-term alterations of synaptic strength into long-lasting changes that are embedded in stable structural modification. These functional and structural modifications are considered the biological basis of learning and memory processes. In this review we discussed the existing evidence regarding the role of the spine actin cytoskeleton in AD synaptic failure. We revised the physiological function of the actin cytoskeleton in the spine shaping and the contribution of actin dynamics in the endocytosis mechanism. The internalization process is implicated in different aspects of AD since it controls both glutamate receptor membrane levels and amyloid generation. The detailed understanding of the mechanisms controlling the actin cytoskeleton in a unique biological context as the dendritic spine could pave the way to the development of innovative synapse-tailored therapeutic interventions and to the identification of novel biomarkers to monitor synaptic loss in AD.
Collapse
|
22
|
High-content imaging analysis for detecting the loss of drebrin clusters along dendrites in cultured hippocampal neurons. J Pharmacol Toxicol Methods 2019; 99:106607. [DOI: 10.1016/j.vascn.2019.106607] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 06/27/2019] [Accepted: 06/27/2019] [Indexed: 11/18/2022]
|
23
|
Gan YJ, Fang AW, Liu C, Liu BJ, Yang FM, Guan JT, Lan CL, Dai XD, Li T, Cao Y, Ran Y, Gong XH, Jin ZB, Cui RZ, Iwata T, Qu J, Lu F, Chi ZL. Elevated Plasma Levels of Drebrin in Glaucoma Patients With Neurodegeneration. Front Neurosci 2019; 13:326. [PMID: 31001081 PMCID: PMC6456690 DOI: 10.3389/fnins.2019.00326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/21/2019] [Indexed: 11/21/2022] Open
Abstract
Glaucoma is an optic neuropathy characterized by progressive degeneration of retinal ganglion cells (RGCs). Aberrations in several cytoskeletal proteins, such as tau have been implicated in the pathogenesis of neurodegenerative diseases, could be initiating factors in glaucoma progression and occurring prior to axon degeneration. Developmentally regulated brain protein (Drebrin or DBN1) is an evolutionarily conserved actin-binding protein playing a prominent role in neurons and is implicated in neurodegenerative diseases. However, the relationship between circulating DBN1 levels and RGC degeneration in glaucoma patients remains unclear. In our preliminary study, we detected drebrin protein in the plasma of glaucoma patients using proteomic analysis. Subsequently, we recruited a total of 232 patients including primary angle-closure glaucoma (PACG), primary open-angle glaucoma (POAG) and Posner-Schlossman syndrome (PS) and measured its DBN1 plasma levels. We observed elevated DBN1 plasma levels in patients with primary glaucoma but not in patients with PS compared to nonaxonopathic controls. Interestingly, in contrast to tau plasma levels increased in all groups of patients, elevated drebrin plasma levels correlated with retinal nerve fiber layer defect (RNFLD) in glaucoma patients. To further explore the expression of DBN1 in neurodegeneration, we conducted experiment of optic nerve crush (ONC) models, and observed increased expression of DBN1 in the serum as well as in the retina and then decreased after ONC. This result reinforces the potentiality of circulating DBN1 levels are increased in glaucoma patients with neurodegeneration. Taken together, our findings suggest that circulating DBN1 levels correlated with RNFLD and may reflect the severity of RGCs injury in glaucoma patients. Combining measurement of circulating drebrin and tau levels may be a useful indicator for monitoring progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi-Jing Gan
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ai-Wu Fang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chang Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bai-Jing Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Feng-Mei Yang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ji-Tian Guan
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chun-Lin Lan
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao-Dan Dai
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tong Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ying Cao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yun Ran
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xian-Hui Gong
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zi-Bing Jin
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China.,International Joint Research Center for Regenerative Medicine and Neurogenetics, Wenzhou Medical University, Wenzhou, China
| | - Ren-Zhe Cui
- Department of Ophthalmology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Takeshi Iwata
- Division of Molecular and Cellular Biology, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Jia Qu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fan Lu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China.,International Joint Research Center for Regenerative Medicine and Neurogenetics, Wenzhou Medical University, Wenzhou, China
| | - Zai-Long Chi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, China.,International Joint Research Center for Regenerative Medicine and Neurogenetics, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
24
|
ATM phosphorylation of the actin-binding protein drebrin controls oxidation stress-resistance in mammalian neurons and C. elegans. Nat Commun 2019; 10:486. [PMID: 30700723 PMCID: PMC6353951 DOI: 10.1038/s41467-019-08420-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 12/27/2018] [Indexed: 12/13/2022] Open
Abstract
Drebrin (DBN) regulates cytoskeletal functions during neuronal development, and is thought to contribute to structural and functional synaptic changes associated with aging and Alzheimer’s disease. Here we show that DBN coordinates stress signalling with cytoskeletal dynamics, via a mechanism involving kinase ataxia-telangiectasia mutated (ATM). An excess of reactive oxygen species (ROS) stimulates ATM-dependent phosphorylation of DBN at serine-647, which enhances protein stability and accounts for improved stress resilience in dendritic spines. We generated a humanized DBN Caenorhabditis elegans model and show that a phospho-DBN mutant disrupts the protective ATM effect on lifespan under sustained oxidative stress. Our data indicate a master regulatory function of ATM-DBN in integrating cytosolic stress-induced signalling with the dynamics of actin remodelling to provide protection from synapse dysfunction and ROS-triggered reduced lifespan. They further suggest that DBN protein abundance governs actin filament stability to contribute to the consequences of oxidative stress in physiological and pathological conditions. Drebrin is an actin-binding protein known to play a role in neuronal dendritic spines but its precise regulation is unclear. Here, the authors report that DBN is activated by oxidative stress in an ATM-kinase dependent manner and increases resistance to oxidative stress in mice and in C. elegans.
Collapse
|
25
|
Borovac J, Bosch M, Okamoto K. Regulation of actin dynamics during structural plasticity of dendritic spines: Signaling messengers and actin-binding proteins. Mol Cell Neurosci 2018; 91:122-130. [PMID: 30004015 DOI: 10.1016/j.mcn.2018.07.001] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 06/25/2018] [Accepted: 07/06/2018] [Indexed: 12/17/2022] Open
Abstract
Activity-dependent plasticity of synaptic structure and function plays an essential role in neuronal development and in cognitive functions including learning and memory. The formation, maintenance and modulation of dendritic spines are mainly controlled by the dynamics of actin filaments (F-actin) through interaction with various actin-binding proteins (ABPs) and postsynaptic signaling messengers. Induction of long-term potentiation (LTP) triggers a cascade of events involving Ca2+ signaling, intracellular pathways such as cAMP and cGMP, and regulation of ABPs such as CaMKII, Cofilin, Aip1, Arp2/3, α-actinin, Profilin and Drebrin. We review here how these ABPs modulate the rate of assembly, disassembly, stabilization and bundling of F-actin during LTP induction. We highlight the crucial role that CaMKII exerts in both functional and structural plasticity by directly coupling Ca2+ signaling with F-actin dynamics through the β subunit. Moreover, we show how cAMP and cGMP second messengers regulate postsynaptic structural potentiation. Brain disorders such as Alzheimer's disease, schizophrenia or autism, are associated with alterations in the regulation of F-actin dynamics by these ABPs and signaling messengers. Thus, a better understanding of the molecular mechanisms controlling actin cytoskeleton can provide cues for the treatment of these disorders.
Collapse
Affiliation(s)
- Jelena Borovac
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Miquel Bosch
- Institute for Bioengineering of Catalonia, Barcelona 08028, Spain.
| | - Kenichi Okamoto
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON M5G 1X5, Canada.
| |
Collapse
|
26
|
Yamazaki H, Sasagawa Y, Yamamoto H, Bito H, Shirao T. CaMKIIβ is localized in dendritic spines as both drebrin-dependent and drebrin-independent pools. J Neurochem 2018; 146:145-159. [PMID: 29675826 PMCID: PMC6099455 DOI: 10.1111/jnc.14449] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 03/14/2018] [Accepted: 04/04/2018] [Indexed: 12/21/2022]
Abstract
Drebrin is a major F-actin binding protein in dendritic spines that is critically involved in the regulation of dendritic spine morphogenesis, pathology, and plasticity. In this study, we aimed to identify a novel drebrin-binding protein involved in spine morphogenesis and synaptic plasticity. We confirmed the beta subunit of Ca2+ /calmodulin-dependent protein kinase II (CaMKIIβ) as a drebrin-binding protein using a yeast two-hybrid system, and investigated the drebrin-CaMKIIβ relationship in dendritic spines using rat hippocampal neurons. Drebrin knockdown resulted in diffuse localization of CaMKIIβ in dendrites during the resting state, suggesting that drebrin is involved in the accumulation of CaMKIIβ in dendritic spines. Fluorescence recovery after photobleaching analysis showed that drebrin knockdown increased the stable fraction of CaMKIIβ, indicating the presence of drebrin-independent, more stable CaMKIIβ. NMDA receptor activation also increased the stable fraction in parallel with drebrin exodus from dendritic spines. These findings suggest that CaMKIIβ can be classified into distinct pools: CaMKIIβ associated with drebrin, CaMKIIβ associated with post-synaptic density (PSD), and CaMKIIβ free from PSD and drebrin. CaMKIIβ appears to be anchored to a protein complex composed of drebrin-binding F-actin during the resting state. NMDA receptor activation releases CaMKIIβ from drebrin resulting in CaMKIIβ association with PSD.
Collapse
Affiliation(s)
- Hiroyuki Yamazaki
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yoshio Sasagawa
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Hideyuki Yamamoto
- Department of Biochemistry, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tomoaki Shirao
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
27
|
Drebrin in Neuronal Migration and Axonal Growth. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1006:141-155. [PMID: 28865019 DOI: 10.1007/978-4-431-56550-5_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
During development, production of neurons from neural stem cells, migration of neurons from their birthplace to their final location, and extension of neurites, axons, and dendrites are important for the formation of functional neuronal circuits. The actin cytoskeleton has major roles in the morphological development of neurons. In this chapter, we focused on the distribution and function of the actin-binding protein, drebrin, to elucidate the importance of drebrin-bound F-actin in neurons during early developmental stages of neurons in embryonic, postnatal, and adult brains. There are three major isoforms of drebrin in the chicken brain (E1, E2, and A) and two major isoforms in the mammalian brain (E and A). Among these drebrin isoforms, drebrin E1 and E2 in chicken and drebrin E in the mammalian brain are involved in these neuronal stages. In migrating neurons of the developing and adult brain, drebrin is localized at the base of filopodia of leading processes, to regulate neuronal migration. In axonal growth cones, drebrin is localized in the transitional zone to regulate axonal growth by inhibiting actomyosin interactions and mediating the interactions between F-actin and microtubules. For axonal collateral branching, drebrin is localized at axonal actin patches and the base of filopodia, to accelerate the transition from actin patches to filopodia and stabilize the filopodia.
Collapse
|
28
|
Del Mar Masdeu M, Armendáriz BG, Torre AL, Soriano E, Burgaya F, Ureña JM. Identification of novel Ack1-interacting proteins and Ack1 phosphorylated sites in mouse brain by mass spectrometry. Oncotarget 2017; 8:101146-101157. [PMID: 29254152 PMCID: PMC5731862 DOI: 10.18632/oncotarget.20929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/26/2017] [Indexed: 12/04/2022] Open
Abstract
Ack1 (activated Cdc42-associated tyrosine kinase) is a non-receptor tyrosine kinase that is highly expressed in brain. This kinase contains several protein-protein interaction domains and its action is partially regulated by phosphorylation. As a first step to address the neuronal functions of Ack1, here we screened mouse brain samples to identify proteins that interact with this kinase. Using mass spectrometry analysis, we identified new putative partners for Ack1 including cytoskeletal proteins such as Drebrin or MAP4; adhesion regulators such as NCAM1 and neurabin-2; and synapse mediators such as SynGAP, GRIN1 and GRIN3. In addition, we confirmed that Ack1 and CAMKII both co-immunoprecipitate and co-localize in neurons. We also identified that adult and P5 samples contained the phosphorylated residues Thr 104 and Ser 825, and only P5 samples contained phosphorylated Ser 722, a site linked to cancer and interleukin signaling when phosphorylated. All these findings support the notion that Ack1 could be involved in neuronal plasticity.
Collapse
Affiliation(s)
- Maria Del Mar Masdeu
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain.,Present address: Francis Crick Institute, Mill Hill Laboratory, Mill Hill, London NW7 1AA, United Kingdom
| | - Beatriz G Armendáriz
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain
| | - Anna La Torre
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain.,Present address: Department of Cell Biology and Human Anatomy, University of California Davis, 95616 Davis, California, USA
| | - Eduardo Soriano
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain.,Vall d´Hebron Institute of Research, Barcelona 08035, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain
| | - Ferran Burgaya
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain
| | - Jesús Mariano Ureña
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain
| |
Collapse
|
29
|
Kajita Y, Kojima N, Koganezawa N, Yamazaki H, Sakimura K, Shirao T. Drebrin E regulates neuroblast proliferation and chain migration in the adult brain. Eur J Neurosci 2017; 46:2214-2228. [DOI: 10.1111/ejn.13668] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 08/11/2017] [Accepted: 08/15/2017] [Indexed: 02/04/2023]
Affiliation(s)
- Yuki Kajita
- Department of Neurobiology and Behavior; Gunma University Graduate School of Medicine; 3-39-22 Showa-machi Maebashi 371-8511 Japan
| | - Nobuhiko Kojima
- Department of Neurobiology and Behavior; Gunma University Graduate School of Medicine; 3-39-22 Showa-machi Maebashi 371-8511 Japan
| | - Noriko Koganezawa
- Department of Neurobiology and Behavior; Gunma University Graduate School of Medicine; 3-39-22 Showa-machi Maebashi 371-8511 Japan
| | - Hiroyuki Yamazaki
- Department of Neurobiology and Behavior; Gunma University Graduate School of Medicine; 3-39-22 Showa-machi Maebashi 371-8511 Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology; Brain Research Institute; Niigata University; Niigata Japan
| | - Tomoaki Shirao
- Department of Neurobiology and Behavior; Gunma University Graduate School of Medicine; 3-39-22 Showa-machi Maebashi 371-8511 Japan
| |
Collapse
|
30
|
Nikolaienko O, Patil S, Eriksen MS, Bramham CR. Arc protein: a flexible hub for synaptic plasticity and cognition. Semin Cell Dev Biol 2017; 77:33-42. [PMID: 28890419 DOI: 10.1016/j.semcdb.2017.09.006] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/06/2017] [Accepted: 09/06/2017] [Indexed: 02/08/2023]
Abstract
Mammalian excitatory synapses express diverse types of synaptic plasticity. A major challenge in neuroscience is to understand how a neuron utilizes different types of plasticity to sculpt brain development, function, and behavior. Neuronal activity-induced expression of the immediate early protein, Arc, is critical for long-term potentiation and depression of synaptic transmission, homeostatic synaptic scaling, and adaptive functions such as long-term memory formation. However, the molecular basis of Arc protein function as a regulator of synaptic plasticity and cognition remains a puzzle. Recent work on the biophysical and structural properties of Arc, its protein-protein interactions and post-translational modifications have shed light on the issue. Here, we present Arc protein as a flexible, multifunctional and interactive hub. Arc interacts with specific effector proteins in neuronal compartments (dendritic spines, nuclear domains) to bidirectionally regulate synaptic strength by distinct molecular mechanisms. Arc stability, subcellular localization, and interactions are dictated by synaptic activity and post-translational modification of Arc. This functional versatility and context-dependent signaling supports a view of Arc as a highly specialized master organizer of long-term synaptic plasticity, critical for information storage and cognition.
Collapse
Affiliation(s)
- Oleksii Nikolaienko
- Department of Biomedicine and KG Jebsen Center for Neuropsychiatric Disorders, University of Bergen, Jonas Lies vei 91, N-5009, Bergen, Norway
| | - Sudarshan Patil
- Department of Biomedicine and KG Jebsen Center for Neuropsychiatric Disorders, University of Bergen, Jonas Lies vei 91, N-5009, Bergen, Norway
| | - Maria Steene Eriksen
- Department of Biomedicine and KG Jebsen Center for Neuropsychiatric Disorders, University of Bergen, Jonas Lies vei 91, N-5009, Bergen, Norway
| | - Clive R Bramham
- Department of Biomedicine and KG Jebsen Center for Neuropsychiatric Disorders, University of Bergen, Jonas Lies vei 91, N-5009, Bergen, Norway.
| |
Collapse
|
31
|
Biochemistry of Drebrin and Its Binding to Actin Filaments. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1006:37-47. [DOI: 10.1007/978-4-431-56550-5_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
32
|
Tai LM, Balu D, Avila-Munoz E, Abdullah L, Thomas R, Collins N, Valencia-Olvera AC, LaDu MJ. EFAD transgenic mice as a human APOE relevant preclinical model of Alzheimer's disease. J Lipid Res 2017; 58:1733-1755. [PMID: 28389477 PMCID: PMC5580905 DOI: 10.1194/jlr.r076315] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/06/2017] [Indexed: 01/12/2023] Open
Abstract
Identified in 1993, APOE4 is the greatest genetic risk factor for sporadic Alzheimer's disease (AD), increasing risk up to 15-fold compared with APOE3, with APOE2 decreasing AD risk. However, the functional effects of APOE4 on AD pathology remain unclear and, in some cases, controversial. In vivo progress to understand how the human (h)-APOE genotypes affect AD pathology has been limited by the lack of a tractable familial AD-transgenic (FAD-Tg) mouse model expressing h-APOE rather than mouse (m)-APOE. The disparity between m- and h-apoE is relevant for virtually every AD-relevant pathway, including amyloid-β (Aβ) deposition and clearance, neuroinflammation, tau pathology, neural plasticity and cerebrovascular deficits. EFAD mice were designed as a temporally useful preclinical FAD-Tg-mouse model expressing the h-APOE genotypes for identifying mechanisms underlying APOE-modulated symptoms of AD pathology. From their first description in 2012, EFAD mice have enabled critical basic and therapeutic research. Here we review insights gleaned from the EFAD mice and summarize future directions.
Collapse
Affiliation(s)
- Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Evangelina Avila-Munoz
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Riya Thomas
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Nicole Collins
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612.
| |
Collapse
|
33
|
Nair RR, Patil S, Tiron A, Kanhema T, Panja D, Schiro L, Parobczak K, Wilczynski G, Bramham CR. Dynamic Arc SUMOylation and Selective Interaction with F-Actin-Binding Protein Drebrin A in LTP Consolidation In Vivo. Front Synaptic Neurosci 2017; 9:8. [PMID: 28553222 PMCID: PMC5426369 DOI: 10.3389/fnsyn.2017.00008] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 04/21/2017] [Indexed: 01/21/2023] Open
Abstract
Activity-regulatedcytoskeleton-associated protein (Arc) protein is implicated as a master regulator of long-term forms of synaptic plasticity and memory formation, but the mechanisms controlling Arc protein function are little known. Post-translation modification by small ubiquitin-like modifier (SUMO) proteins has emerged as a major mechanism for regulating protein-protein interactions and function. We first show in cell lines that ectopically expressed Arc undergoes mono-SUMOylation. The covalent addition of a single SUMO1 protein was confirmed by in vitro SUMOylation of immunoprecipitated Arc. To explore regulation of endogenous Arc during synaptic plasticity, we induced long-term potentiation (LTP) in the dentate gyrus of live anesthetized rats. Using coimmunoprecipitation of native proteins, we show that Arc synthesized during the maintenance phase of LTP undergoes dynamic mono-SUMO1-ylation. Levels of unmodified Arc increase in multiple subcellular fractions (cytosol, membrane, nuclear and cytoskeletal), whereas enhanced Arc SUMOylation was specific to the synaptoneurosomal and the cytoskeletal fractions. Dentate gyrus LTP consolidation requires a period of sustained Arc synthesis driven by brain-derived neurotrophic factor (BDNF) signaling. Local infusion of the BDNF scavenger, TrkB-Fc, during LTP maintenance resulted in rapid reversion of LTP, inhibition of Arc synthesis and loss of enhanced Arc SUMO1ylation. Furthermore, coimmunoprecipitation analysis showed that SUMO1-ylated Arc forms a complex with the F-actin-binding protein drebrin A, a major regulator of cytoskeletal dynamics in dendritic spines. Although Arc also interacted with dynamin 2, calcium/calmodulindependentprotein kinase II-beta (CaMKIIβ), and postsynaptic density protein-95 (PSD-95), these complexes lacked SUMOylated Arc. The results support a model in which newly synthesized Arc is SUMOylated and targeted for actin cytoskeletal regulation during in vivo LTP.
Collapse
Affiliation(s)
- Rajeevkumar R Nair
- Department of Biomedicine and KG Jebsen Centre for Neuropsychiatric Disorders, University of BergenBergen, Norway
| | - Sudarshan Patil
- Department of Biomedicine and KG Jebsen Centre for Neuropsychiatric Disorders, University of BergenBergen, Norway
| | - Adrian Tiron
- Department of Biomedicine and KG Jebsen Centre for Neuropsychiatric Disorders, University of BergenBergen, Norway
| | - Tambudzai Kanhema
- Department of Biomedicine and KG Jebsen Centre for Neuropsychiatric Disorders, University of BergenBergen, Norway
| | - Debabrata Panja
- Department of Biomedicine and KG Jebsen Centre for Neuropsychiatric Disorders, University of BergenBergen, Norway
| | - Lars Schiro
- Department of Biomedicine and KG Jebsen Centre for Neuropsychiatric Disorders, University of BergenBergen, Norway
| | - Kamil Parobczak
- Laboratory of Molecular and Systemic Neuromorphology, Department of Neurophysiology, Nencki Institute of Experimental BiologyWarsaw, Poland
| | - Grzegorz Wilczynski
- Laboratory of Molecular and Systemic Neuromorphology, Department of Neurophysiology, Nencki Institute of Experimental BiologyWarsaw, Poland
| | - Clive R Bramham
- Department of Biomedicine and KG Jebsen Centre for Neuropsychiatric Disorders, University of BergenBergen, Norway
| |
Collapse
|
34
|
Cho C, MacDonald R, Shang J, Cho MJ, Chalifour LE, Paudel HK. Early growth response-1-mediated down-regulation of drebrin correlates with loss of dendritic spines. J Neurochem 2017; 142:56-73. [DOI: 10.1111/jnc.14031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/28/2017] [Accepted: 03/22/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Chulmin Cho
- Department of Neurology and Neurosurgery; McGill University; Montreal Quebec Canada
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
| | - Ryen MacDonald
- Department of Neurology and Neurosurgery; McGill University; Montreal Quebec Canada
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
| | - Jijun Shang
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
| | - Moon Jeong Cho
- Department of Neurology and Neurosurgery; McGill University; Montreal Quebec Canada
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
| | - Lorraine E. Chalifour
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
- Department of Medicine; McGill University; Montreal Quebec Canada
| | - Hemant K. Paudel
- Department of Neurology and Neurosurgery; McGill University; Montreal Quebec Canada
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
- Department of Medicine; McGill University; Montreal Quebec Canada
| |
Collapse
|
35
|
Shirao T, Hanamura K, Koganezawa N, Ishizuka Y, Yamazaki H, Sekino Y. The role of drebrin in neurons. J Neurochem 2017; 141:819-834. [PMID: 28199019 DOI: 10.1111/jnc.13988] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 12/27/2016] [Accepted: 01/11/2017] [Indexed: 01/13/2023]
Abstract
Drebrin is an actin-binding protein that changes the helical pitch of actin filaments (F-actin), and drebrin-decorated F-actin shows slow treadmilling and decreased rate of depolymerization. Moreover, the characteristic morphology of drebrin-decorated F-actin enables it to respond differently to the same signals from other actin cytoskeletons. Drebrin consists of two major isoforms, drebrin E and drebrin A. In the developing brain, drebrin E appears in migrating neurons and accumulates in the growth cones of axons and dendrites. Drebrin E-decorated F-actin links lamellipodium F-actin to microtubules in the growth cones. Then drebrin A appears at nascent synapses and drebrin A-decorated F-actin facilitates postsynaptic molecular assembly. In the adult brain, drebrin A-decorated F-actin is concentrated in the central region of dendritic spines. During long-term potentiation initiation, NMDA receptor-mediated Ca2+ influx induces the transient exodus of drebrin A-decorated F-actin via myosin II ATPase activation. Because of the unique physical characteristics of drebrin A-decorated F-actin, this exodus likely contributes to the facilitation of F-actin polymerization and spine enlargement. Additionally, drebrin reaccumulation in dendritic spines is observed after the exodus. In our drebrin exodus model of structure-based synaptic plasticity, reestablishment of drebrin A-decorated F-actin is necessary to keep the enlarged spine size during long-term potentiation maintenance. In this review, we introduce the genetic and biochemical properties of drebrin and the roles of drebrin in early stage of brain development, synaptic formation and synaptic plasticity. Further, we discuss the pathological relevance of drebrin loss in Alzheimer's disease. This article is part of the mini review series "60th Anniversary of the Japanese Society for Neurochemistry".
Collapse
Affiliation(s)
- Tomoaki Shirao
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Kenji Hanamura
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Noriko Koganezawa
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yuta Ishizuka
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Hiroyuki Yamazaki
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yuko Sekino
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan.,Division of Pharmacology, National Institute of Health Sciences, Tokyo, Japan
| |
Collapse
|
36
|
Koganezawa N, Hanamura K, Shirao T. Progress in applications of iPSC-derived neurons for evaluation of drugs. Nihon Yakurigaku Zasshi 2017; 149:104-109. [PMID: 28260738 DOI: 10.1254/fpj.149.104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
37
|
Koganezawa N, Hanamura K, Sekino Y, Shirao T. The role of drebrin in dendritic spines. Mol Cell Neurosci 2017; 84:85-92. [PMID: 28161364 DOI: 10.1016/j.mcn.2017.01.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/06/2016] [Accepted: 01/18/2017] [Indexed: 11/17/2022] Open
Abstract
Dendritic spines form typical excitatory synapses in the brain and their shapes vary depending on synaptic inputs. It has been suggested that the morphological changes of dendritic spines play an important role in synaptic plasticity. Dendritic spines contain a high concentration of actin, which has a central role in supporting cell motility, and polymerization of actin filaments (F-actin) is most likely involved in spine shape changes. Drebrin is an actin-binding protein that forms stable F-actin and is highly accumulated within dendritic spines. Drebrin has two isoforms, embryonic-type drebrin E and adult-type drebrin A, that change during development from E to A. Inhibition of drebrin A expression results in a delay of synapse formation and inhibition of postsynaptic protein accumulation, suggesting that drebrin A has an important role in spine maturation. In mature synapses, glutamate stimulation induces rapid spine-head enlargement during long-term potentiation (LTP) formation. LTP stimulation induces Ca2+ entry through N-methyl-d-aspartate (NMDA) receptors, which causes drebrin exodus from dendritic spines. Once drebrin exits from dendritic spine heads, the dynamic actin pool increases in spine heads to facilitate F-actin polymerization. To maintain enlarged spine heads, drebrin-decorated F-actin is thought to reform within the spine heads. Thus, drebrin plays a pivotal role in spine plasticity through regulation of F-actin.
Collapse
Affiliation(s)
- Noriko Koganezawa
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Kenji Hanamura
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Yuko Sekino
- Division of Pharmacology, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Tomoaki Shirao
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan.
| |
Collapse
|
38
|
Homer, Spikar, and Other Drebrin-Binding Proteins in the Brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1006:249-268. [PMID: 28865024 DOI: 10.1007/978-4-431-56550-5_14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Drebrin is a major F-actin-binding protein in the brain. In the past two decades, many drebrin-binding proteins in addition to F-actin have been identified in several research fields including neuroscience, oncology, and immunology. Among the drebrin-binding proteins, there are various kinds of proteins including scaffold proteins, nuclear proteins, phosphatases, microtubule-binding proteins, G-actin-binding proteins, gap junction proteins, chemokine receptors, and cell-adhesion-related proteins. The interaction between drebrin and its binding partners seems to play important roles in higher brain functions, because drebrin is involved in the pathogenesis of some neurological diseases with cognitive defects. In this chapter, we will first review the interaction of Homer and spikar with drebrin, particularly focusing on spine morphogenesis and synaptic function. Homer contributes to spine morphogenesis by cooperating with shank and activated Cdc42 small GTPase, suggesting a novel signaling pathway comprising Homer, drebrin, shank, and Cdc42 for spine morphogenesis. Drebrin sequesters spikar in the cytoplasm and stabilizes it in dendritic spines, leading to spine formation. Finally, we will introduce some other drebrin-binding proteins including end-binding protein 3 (EB3), profilin, progranulin, and phosphatase and tensin homologue (PTEN). These proteins are involved in Alzheimer's disease and cancer. Therefore, further studies on drebrin and its binding proteins will be of great importance to elucidate the pathologies of various diseases and may contribute to their medical treatment and diagnostics development.
Collapse
|
39
|
Aoki C, Sherpa AD. Making of a Synapse: Recurrent Roles of Drebrin A at Excitatory Synapses Throughout Life. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1006:119-139. [DOI: 10.1007/978-4-431-56550-5_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
40
|
Sekino Y, Koganezawa N, Mizui T, Shirao T. Role of Drebrin in Synaptic Plasticity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1006:183-201. [DOI: 10.1007/978-4-431-56550-5_11] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
41
|
|
42
|
|
43
|
Gallo G. Coordination of the axonal cytoskeleton during the emergence of axon collateral branches. Neural Regen Res 2016; 11:709-11. [PMID: 27335541 PMCID: PMC4904448 DOI: 10.4103/1673-5374.182684] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Gianluca Gallo
- Temple University, Lewis Kats School of Medicine, Department of Anatomy and cell Biology, Shriners Hospitals Pediatric Research Center, Philadelphia, PA, USA
| |
Collapse
|
44
|
Brettle M, Patel S, Fath T. Tropomyosins in the healthy and diseased nervous system. Brain Res Bull 2016; 126:311-323. [PMID: 27298153 DOI: 10.1016/j.brainresbull.2016.06.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/09/2016] [Accepted: 06/10/2016] [Indexed: 12/25/2022]
Abstract
Regulation of the actin cytoskeleton is dependent on a plethora of actin-associated proteins in all eukaryotic cells. The family of tropomyosins plays a key role in controlling the function of several of these actin-associated proteins and their access to actin filaments. In order to understand the regulation of the actin cytoskeleton in highly dynamic subcellular compartments of neurons such as growth cones of developing neurons and the synaptic compartment of mature neurons, it is pivotal to decipher the functional role of tropomyosins in the nervous system. In this review, we will discuss the current understanding and recent findings on the regulation of the actin cytoskeleton by tropomyosins and potential implication that this has for the dysregulation of the actin cytoskeleton in neurological diseases.
Collapse
Affiliation(s)
- Merryn Brettle
- Neurodegeneration and Repair Unit, School of Medical Sciences, University of New South Wales, 2052 Sydney, New South Wales, Australia
| | - Shrujna Patel
- Neurodegeneration and Repair Unit, School of Medical Sciences, University of New South Wales, 2052 Sydney, New South Wales, Australia
| | - Thomas Fath
- Neurodegeneration and Repair Unit, School of Medical Sciences, University of New South Wales, 2052 Sydney, New South Wales, Australia.
| |
Collapse
|
45
|
Connexin43 Forms Supramolecular Complexes through Non-Overlapping Binding Sites for Drebrin, Tubulin, and ZO-1. PLoS One 2016; 11:e0157073. [PMID: 27280719 PMCID: PMC4900556 DOI: 10.1371/journal.pone.0157073] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 05/24/2016] [Indexed: 02/06/2023] Open
Abstract
Gap junctions are membrane specialization domains identified in most tissue types where cells abut each other. The connexin channels found in these membrane domains are conduits for direct cell-to-cell transfer of ions and molecules. Connexin43 (Cx43) is the most ubiquitous connexin, with critical roles in heart, skin, and brain. Several studies described the interaction between Cx43 and the cytoskeleton involving the actin binding proteins Zonula occludens (ZO-1) and drebrin, as well as with tubulin. However, a direct interaction has not been identified between drebrin and Cx43. In this study, co-IP and NMR experiments were used to demonstrate that the Cx43-CT directly interacts with the highly conserved N-terminus region of drebrin. Three Cx43-CT areas were found to be involved in drebrin binding, with residues 264–275 being critical for the interaction. Mimicking Src phosphorylation within this region (Y265) significantly disrupted the interaction between the Cx43-CT and drebrin. Immunofluorescence showed colocalization of Cx43, drebrin, and F-actin in astrocytes and Vero cells membrane, indicating that Cx43 forms a submembrane protein complex with cytoskeletal and scaffolding proteins. The co-IP data suggest that Cx43 indirectly interacts with F-actin through drebrin. Along with the known interaction of the Cx43-CT with ZO-1 and tubulin, the data presented here for drebrin indicate non-overlapping and separated binding sites for all three proteins for which simultaneous binding could be important in regulating cytoskeleton rearrangements, especially for neuronal migration during brain development.
Collapse
|
46
|
Masdeu MDM, Armendáriz BG, Soriano E, Ureña JM, Burgaya F. New partners and phosphorylation sites of focal adhesion kinase identified by mass spectrometry. Biochim Biophys Acta Gen Subj 2016; 1860:1388-94. [PMID: 27033120 DOI: 10.1016/j.bbagen.2016.02.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/22/2015] [Accepted: 02/23/2016] [Indexed: 01/29/2023]
Abstract
The regulation of focal adhesion kinase (FAK) involves phosphorylation and multiple interactions with other signaling proteins. Some of these pathways are relevant for nervous system functions such as branching, axonal guidance, and plasticity. In this study, we screened mouse brain to identify FAK-interactive proteins and phosphorylatable residues as a first step to address the neuronal functions of this kinase. Using mass spectrometry analysis, we identified new phosphorylated sites (Thr 952, Thr 1048, and Ser 1049), which lie in the FAT domain; and putative new partners for FAK, which include cytoskeletal proteins such as drebrin and MAP 6, adhesion regulators such as neurabin-2 and plakophilin 1, and synapse-associated proteins such as SynGAP and a NMDA receptor subunit. Our findings support the participation of brain-localized FAK in neuronal plasticity.
Collapse
Affiliation(s)
- Maria del Mar Masdeu
- Developmental Neurobiology and Neural Regeneration Group, Department of Cell Biology, Faculty of Biology, University of Barcelona, Diagonal 643, 08038 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain
| | - Beatriz G Armendáriz
- Developmental Neurobiology and Neural Regeneration Group, Department of Cell Biology, Faculty of Biology, University of Barcelona, Diagonal 643, 08038 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain
| | - Eduardo Soriano
- Developmental Neurobiology and Neural Regeneration Group, Department of Cell Biology, Faculty of Biology, University of Barcelona, Diagonal 643, 08038 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain; Vall d´Hebron Institute of Research, 08035 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Jesús Mariano Ureña
- Developmental Neurobiology and Neural Regeneration Group, Department of Cell Biology, Faculty of Biology, University of Barcelona, Diagonal 643, 08038 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain
| | - Ferran Burgaya
- Developmental Neurobiology and Neural Regeneration Group, Department of Cell Biology, Faculty of Biology, University of Barcelona, Diagonal 643, 08038 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain.
| |
Collapse
|
47
|
Kojima N, Yasuda H, Hanamura K, Ishizuka Y, Sekino Y, Shirao T. Drebrin A regulates hippocampal LTP and hippocampus-dependent fear learning in adult mice. Neuroscience 2016; 324:218-26. [PMID: 26970584 DOI: 10.1016/j.neuroscience.2016.03.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 02/24/2016] [Accepted: 03/05/2016] [Indexed: 10/25/2022]
Abstract
Structural plasticity of dendritic spines, which underlies higher brain functions including learning and memory, is dynamically regulated by the actin cytoskeleton and its associated proteins. Drebrin A is an F-actin-binding protein preferentially expressed in the brain and localized in the dendritic spines of mature neurons. Isoform conversion from drebrin E to drebrin A and accumulation of the latter in dendritic spines occurs during synapse maturation. We have previously demonstrated that drebrin A plays a pivotal role in spine morphogenesis and plasticity. However, it is unclear whether drebrin A plays a specific role in processes required for structural plasticity, and whether drebrin E can substitute in this role. To answer these questions, we analyzed mutant mice (named DAKO mice), in which isoform conversion from drebrin E to drebrin A is disrupted. In DAKO mouse brain, drebrin E continues to be expressed throughout life instead of drebrin A. Electrophysiological studies using hippocampal slices revealed that long-term potentiation of CA1 synapses was impaired in adult DAKO mice, but not in adolescents. In parallel with this age-dependent impairment, DAKO mice exhibited impaired hippocampus-dependent fear learning in an age-dependent manner; the impairment was evident in adult mice, but not in adolescents. In addition, histological investigation revealed that the spine length of the apical dendrite of CA1 pyramidal cells was significantly longer in adult DAKO mice than in wild-type mice. Our data indicate that the roles of drebrin E and drebrin A in brain function are different from each other, that the isoform conversion of drebrin is critical, and that drebrin A is indispensable for normal synaptic plasticity and hippocampus-dependent fear memory in the adult brain.
Collapse
Affiliation(s)
- N Kojima
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan; Faculty of Life Sciences, Toyo University, Itakura, Gunma 374-0193, Japan; Institute of Life Innovation Studies, Toyo University, Itakura, Gunma 374-0193, Japan
| | - H Yasuda
- Education and Research Support Center, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - K Hanamura
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Y Ishizuka
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Y Sekino
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan; Division of Pharmacology, National Institute of Health Sciences, Tokyo, Tokyo 158-8501, Japan
| | - T Shirao
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan.
| |
Collapse
|
48
|
Ketschek A, Spillane M, Dun XP, Hardy H, Chilton J, Gallo G. Drebrin coordinates the actin and microtubule cytoskeleton during the initiation of axon collateral branches. Dev Neurobiol 2016; 76:1092-110. [PMID: 26731339 DOI: 10.1002/dneu.22377] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 12/07/2015] [Accepted: 01/01/2016] [Indexed: 11/10/2022]
Abstract
Drebrin is a cytoskeleton-associated protein which can interact with both actin filaments and the tips of microtubules. Its roles have been studied mostly in dendrites, and the functions of drebrin in axons are less well understood. In this study, we analyzed the role of drebrin, through shRNA-mediated depletion and overexpression, in the collateral branching of chicken embryonic sensory axons. We report that drebrin promotes the formation of axonal filopodia and collateral branches in vivo and in vitro. Live imaging of cytoskeletal dynamics revealed that drebrin promotes the formation of filopodia from precursor structures termed axonal actin patches. Endogenous drebrin localizes to actin patches and depletion studies indicate that drebrin contributes to the development of patches. In filopodia, endogenous drebrin localizes to the proximal portion of the filopodium. Drebrin was found to promote the stability of axonal filopodia and the entry of microtubule plus tips into axonal filopodia. The effects of drebrin on the stabilization of filopodia are independent of its effects on promoting microtubule targeting to filopodia. Inhibition of myosin II induces a redistribution of endogenous drebrin distally into filopodia, and further increases branching in drebrin overexpressing neurons. Finally, a 30 min treatment with the branch-inducing signal nerve growth factor increases the levels of axonal drebrin. This study determines the specific roles of drebrin in the regulation of the axonal cytoskeleton, and provides evidence that drebrin contributes to the coordination of the actin and microtubule cytoskeleton during the initial stages of axon branching. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1092-1110, 2016.
Collapse
Affiliation(s)
- Andrea Ketschek
- Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, 3500 N. Broad St, Philadelphia, Pennsylvania, 19140
| | - Mirela Spillane
- Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, 3500 N. Broad St, Philadelphia, Pennsylvania, 19140
| | - Xin-Peng Dun
- Peninsula Schools of Medicine and Dentistry, University of Plymouth, Plymouth Science Park, Research Way, Plymouth, PL6 8BU, United Kingdom
| | - Holly Hardy
- RILD Building, University of Exeter Medical School, Wellcome Wolfson Medical Research Centre, Barrack Road, Exeter, EX2 5DW, United Kingdom
| | - John Chilton
- RILD Building, University of Exeter Medical School, Wellcome Wolfson Medical Research Centre, Barrack Road, Exeter, EX2 5DW, United Kingdom
| | - Gianluca Gallo
- Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, 3500 N. Broad St, Philadelphia, Pennsylvania, 19140
| |
Collapse
|
49
|
Menon S, Gupton SL. Building Blocks of Functioning Brain: Cytoskeletal Dynamics in Neuronal Development. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 322:183-245. [PMID: 26940519 PMCID: PMC4809367 DOI: 10.1016/bs.ircmb.2015.10.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neural connectivity requires proper polarization of neurons, guidance to appropriate target locations, and establishment of synaptic connections. From when neurons are born to when they finally reach their synaptic partners, neurons undergo constant rearrangment of the cytoskeleton to achieve appropriate shape and polarity. Of particular importance to neuronal guidance to target locations is the growth cone at the tip of the axon. Growth-cone steering is also dictated by the underlying cytoskeleton. All these changes require spatiotemporal control of the cytoskeletal machinery. This review summarizes the proteins that are involved in modulating the actin and microtubule cytoskeleton during the various stages of neuronal development.
Collapse
Affiliation(s)
- Shalini Menon
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Stephanie L Gupton
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, United States of America; Neuroscience Center and Curriculum in Neurobiology, University of North Carolina, Chapel Hill, NC, United States of America; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States of America.
| |
Collapse
|
50
|
Mazur AJ, Radaszkiewicz T, Makowiecka A, Malicka-Błaszkiewicz M, Mannherz HG, Nowak D. Gelsolin interacts with LamR, hnRNP U, nestin, Arp3 and β-tubulin in human melanoma cells as revealed by immunoprecipitation and mass spectrometry. Eur J Cell Biol 2016; 95:26-41. [DOI: 10.1016/j.ejcb.2015.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 10/08/2015] [Accepted: 11/04/2015] [Indexed: 01/25/2023] Open
|