1
|
Wu XS, Zhang Z, Jin Y, Mushtaheed A, Wu LG. Actin maintains synaptic transmission by restraining vesicle release probability. iScience 2025; 28:112000. [PMID: 40109375 PMCID: PMC11919605 DOI: 10.1016/j.isci.2025.112000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/22/2024] [Accepted: 02/07/2025] [Indexed: 03/22/2025] Open
Abstract
Despite decades of pharmacological studies, how the ubiquitous cytoskeletal actin regulates synaptic transmission remains poorly understood. We addressed this issue with a tissue-specific knockout of actin β-isoform or γ-isoform, combined with recordings of postsynaptic EPSCs, presynaptic capacitance jumps or fluorescent synaptophysin-pHluorin changes, and electron microscopy in large calyx-type and small conventional hippocampal synapses. We found that actin restrains basal synaptic transmission during single action potential firings by lowering the readily releasable vesicle's release probability. Such an inhibition of basal synaptic transmission is turned into facilitation during repetitive firings by slowing down depletion of the readily releasable vesicle pool and, thus, short-term synaptic depression, leading to more effective synaptic transmission for a longer time. These mechanisms, together with the previous finding that actin promotes vesicle replenishment to the readily releasable pool, may control synaptic transmission and short-term synaptic plasticity at many synapses, contributing to neurological disorders caused by actin cytoskeleton impairment.
Collapse
Affiliation(s)
- Xin-Sheng Wu
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bethesda, MD 20892, USA
| | - Zhen Zhang
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bethesda, MD 20892, USA
- Office of Genetic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Yinghui Jin
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bethesda, MD 20892, USA
| | - Afreen Mushtaheed
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bethesda, MD 20892, USA
| | - Ling-Gang Wu
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bethesda, MD 20892, USA
| |
Collapse
|
2
|
Uppalapati A, Wang T, Nguyen LH. Evaluation of suitable reference genes for gene expression studies in the developing mouse cortex using RT-qPCR. BMC Neurosci 2025; 26:12. [PMID: 39966711 PMCID: PMC11837712 DOI: 10.1186/s12868-025-00934-y] [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: 08/13/2024] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Real-time quantitative PCR (RT-qPCR) is a widely used method to investigate gene expression in neuroscience studies. Accurate relative quantification of RT-qPCR requires the selection of reference genes that are stably expressed across the experimental conditions and tissues of interest. While RT-qPCR is often performed to investigate gene expression changes during neurodevelopment, few studies have examined the expression stability of commonly used reference genes in the developing mouse cortex. RESULTS Here, we evaluated the stability of five housekeeping genes, Actb, Gapdh, B2m, Rpl13a, and Hprt, in cortical tissue from mice at embryonic day 15 to postnatal day 0 to identify optimal reference genes with stable expression during late corticogenesis. The expression stability was assessed using five computational algorithms: BestKeeper, geNorm, NormFinder, DeltaCt, and RefFinder. Our results showed that B2m, Gapdh, and Hprt, or a combination of B2m/Gapdh and B2m/Hprt, were the most stably expressed genes or gene pairs. In contrast, Actb and Rpl13a were the least stably expressed. CONCLUSION This study identifies B2m, Gapdh, and Hprt as suitable reference genes for relative quantification in RT-qPCR-based cortical development studies spanning the period of embryonic day 15 to postnatal day 0.
Collapse
Affiliation(s)
- Ananya Uppalapati
- Department of Neuroscience, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Timothy Wang
- Department of Neuroscience, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Lena H Nguyen
- Department of Neuroscience, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, 75080, USA.
| |
Collapse
|
3
|
Wang W, Rui M. Advances in understanding the roles of actin scaffolding and membrane trafficking in dendrite development. J Genet Genomics 2024; 51:1151-1161. [PMID: 38925347 DOI: 10.1016/j.jgg.2024.06.010] [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: 04/24/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
Dendritic morphology is typically highly branched, and the branching and synaptic abundance of dendrites can enhance the receptive range of neurons and the diversity of information received, thus providing the basis for information processing in the nervous system. Once dendritic development is aberrantly compromised or damaged, it may lead to abnormal connectivity of the neural network, affecting the function and stability of the nervous system and ultimately triggering a series of neurological disorders. Research on the regulation of dendritic developmental processes has flourished, and much progress is now being made in its regulatory mechanisms. Noteworthily, dendrites are characterized by an extremely complex dendritic arborization that cannot be attributed to individual protein functions alone, requiring a systematic analysis of the intrinsic and extrinsic signals and the coordinated roles among them. Actin cytoskeleton organization and membrane vesicle trafficking are required during dendrite development, with actin providing tracks for vesicles and vesicle trafficking in turn providing material for actin assembly. In this review, we focus on these two basic biological processes and discuss the molecular mechanisms and their synergistic effects underlying the morphogenesis of neuronal dendrites. We also offer insights and discuss strategies for the potential preventive and therapeutic treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Wanting Wang
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210031, China
| | - Menglong Rui
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210031, China.
| |
Collapse
|
4
|
Micovic K, Canuel A, Remtulla A, Chuyen A, Byrsan M, McGarry DJ, Olson MF. Mical1 deletion in tyrosinase expressing cells affects mouse running gaits. GENES, BRAIN, AND BEHAVIOR 2024; 23:e70004. [PMID: 39344934 PMCID: PMC11440367 DOI: 10.1111/gbb.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 10/01/2024]
Abstract
Neuronal development is a highly regulated process that is dependent on the correct coordination of cellular responses to extracellular cues. In response to semaphorin axon guidance proteins, the MICAL1 protein is stimulated to produce reactive oxygen species that oxidize actin on specific methionine residues, leading to filamentous actin depolymerization and consequent changes in neuronal growth cone dynamics. Crossing genetically modified mice homozygous for floxed Mical1 (Mical1fl/fl) alleles with transgenic mice expressing Cre recombinase under the control of a tyrosinase gene enhancer/promoter (Tyr::Cre) enabled conditional Mical1 deletion. Immunohistochemical analysis showed Mical1 expression in the cerebellum, which plays a prominent role in the coordination of motor movements, with reduced Mical1 expression in Mical1fl/fl mice co-expressing Tyr::Cre. Analysis of the gaits of mice running on a treadmill showed that both male and female Mical1fl/fl, Tyr::Cre mutant mice had significant alterations to their striding patterns relative to wild-type mice, although the specific aspects of their altered gaits differed between the sexes. Additional motor tests that involved movement on a rotating rod, descending a vertical pole, or crossing a balance beam did not show significant differences between the genotypes, suggesting that the effect of the Mical1fl/fl, Tyr::Cre genetic modifications was only manifested during specific highly coordinated movements that contribute to running. These findings indicate that there is a behavioral consequence in Mical1fl/fl, Tyr::Cre mutant mice that affects motor control as manifested by alterations in their gait.
Collapse
Affiliation(s)
- Katarina Micovic
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Alicia Canuel
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoOntarioCanada
| | - Aasiya Remtulla
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoOntarioCanada
| | - Alexandre Chuyen
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Margarita Byrsan
- Biomedical Engineering ProgramToronto Metropolitan UniversityTorontoOntarioCanada
| | - David J. McGarry
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Michael F. Olson
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoOntarioCanada
- Biomedical Engineering ProgramToronto Metropolitan UniversityTorontoOntarioCanada
| |
Collapse
|
5
|
Greve JN, Marquardt A, Heiringhoff R, Reindl T, Thiel C, Di Donato N, Taft MH, Manstein DJ. The non-muscle actinopathy-associated mutation E334Q in cytoskeletal γ-actin perturbs interaction of actin filaments with myosin and ADF/cofilin family proteins. eLife 2024; 12:RP93013. [PMID: 38446501 PMCID: PMC10942649 DOI: 10.7554/elife.93013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024] Open
Abstract
Various heterozygous cytoskeletal γ-actin mutations have been shown to cause Baraitser-Winter cerebrofrontofacial syndrome, non-syndromic hearing loss, or isolated eye coloboma. Here, we report the biochemical characterization of human cytoskeletal γ-actin carrying mutation E334Q, a mutation that leads to a hitherto unspecified non-muscle actinopathy. Following expression, purification, and removal of linker and thymosin β4 tag sequences, the p.E334Q monomers show normal integration into linear and branched actin filaments. The mutation does not affect thermal stability, actin filament nucleation, elongation, and turnover. Model building and normal mode analysis predict significant differences in the interaction of p.E334Q filaments with myosin motors and members of the ADF/cofilin family of actin-binding proteins. Assays probing the interactions of p.E334Q filaments with human class 2 and class 5 myosin motor constructs show significant reductions in sliding velocity and actin affinity. E334Q differentially affects cofilin-mediated actin dynamics by increasing the rate of cofilin-mediated de novo nucleation of actin filaments and decreasing the efficiency of cofilin-mediated filament severing. Thus, it is likely that p.E334Q-mediated changes in myosin motor activity, as well as filament turnover, contribute to the observed disease phenotype.
Collapse
Affiliation(s)
- Johannes N Greve
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz Hartmann Centre for MedicalHannoverGermany
| | - Anja Marquardt
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz Hartmann Centre for MedicalHannoverGermany
| | - Robin Heiringhoff
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz Hartmann Centre for MedicalHannoverGermany
| | - Theresia Reindl
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz Hartmann Centre for MedicalHannoverGermany
| | - Claudia Thiel
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz Hartmann Centre for MedicalHannoverGermany
| | | | - Manuel H Taft
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz Hartmann Centre for MedicalHannoverGermany
| | - Dietmar J Manstein
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz Hartmann Centre for MedicalHannoverGermany
- Division for Structural Biochemistry, Hannover Medical SchoolHannoverGermany
- RESiST, Cluster of Excellence 2155, Hannover Medical SchoolHannoverGermany
| |
Collapse
|
6
|
Artimagnella O, Mazzon E, Salamone S, Pollastro F, Gugliandolo A, Chiricosta L. Cannabinerol (CBNR) Influences Synaptic Genes Associated with Cytoskeleton and Ion Channels in NSC-34 Cell Line: A Transcriptomic Study. Biomedicines 2024; 12:189. [PMID: 38255294 PMCID: PMC10813620 DOI: 10.3390/biomedicines12010189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Cannabinoids are receiving great attention as a novel approach in the treatment of cognitive and motor disabilities, which characterize neurological disorders. To date, over 100 phytocannabinoids have been extracted from Cannabis sativa, and some of them have shown neuroprotective properties and the capacity to influence synaptic transmission. In this study, we investigated the effects of a less-known phytocannabinoid, cannabinerol (CBNR), on neuronal physiology. Using the NSC-34 motor-neuron-like cell line and next-generation sequencing analysis, we discovered that CBNR influences synaptic genes associated with synapse organization and specialization, including genes related to the cytoskeleton and ion channels. Specifically, the calcium, sodium, and potassium channel subunits (Cacna1b, Cacna1c, Cacnb1, Grin1, Scn8a, Kcnc1, Kcnj9) were upregulated, along with genes related to NMDAR (Agap3, Syngap1) and calcium (Cabp1, Camkv) signaling. Moreover, cytoskeletal and cytoskeleton-associated genes (Actn2, Ina, Trio, Marcks, Bsn, Rtn4, Dgkz, Htt) were also regulated by CBNR. These findings highlight the important role played by CBNR in the regulation of synaptogenesis and synaptic transmission, suggesting the need for further studies to evaluate the neuroprotective role of CBNR in the treatment of synaptic dysfunctions that characterize motor disabilities in many neurological disorders.
Collapse
Affiliation(s)
- Osvaldo Artimagnella
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy (E.M.)
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy (E.M.)
| | - Stefano Salamone
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy; (S.S.); (F.P.)
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy; (S.S.); (F.P.)
| | - Agnese Gugliandolo
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy (E.M.)
| | - Luigi Chiricosta
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy (E.M.)
| |
Collapse
|
7
|
Okyar Baş A, Yardımcı GK, Güner Oytun M, Ceylan S, Kahyaoğlu Z, Öztürk Y, Çöteli S, Halil MG, Cankurtaran M, Kılıç L, Doğu Yavuz BB. Cognitive Status is Better in Older Adults under Colchicine Treatment: A Case-Control Study. Exp Aging Res 2024; 50:17-27. [PMID: 36373385 DOI: 10.1080/0361073x.2022.2145164] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/04/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE We aimed to investigate the effects of colchicine, an important anti-inflammatory agent, on cognitive functions in a geriatric population diagnosed with gout or osteoarthritis by comparing it to non-colchicine users. METHODS 138 geriatric patients (67 colchicine users and 71 non-users) were enrolled. Within comprehensive geriatric assessment (CGA), cognitive status assessment via Mini-Mental State Examination test (MMSE), Quick Mild Cognitive Impairment Screening test (Qmci), clock drowning test (CDT), and digit span tests were performed. RESULTS Median age was 68 (65-72), and there were 82 female (59.4%) patients. The scores of CDT, Backward Digit Span Test, MMSE-Total, MMSE-Attention, MMSE-Motor Function, Qmci-Total Score, Qmci-Clock drawing, and Qmci-Logical Memory were significantly higher in the colchicine user group (p < .005), showing better cognitive function. Adjusted model analysis showed that colchicine usage is independently correlated with higher Qmci-Total Score and Qmci-Logical Memory Score (For Qmci total score β = 7.87 95%CI = 5.48-10.27, p = <0.0001, and for Qmci Logical memory score β = 3.52, 95%CI = 2.12-4.91, p = <0.0001). CONCLUSION To the best of our knowledge, this is the first study revealing that colchicine usage is associated with better cognitive performance in older adults. Further investigations with a prospective, larger-sampled and randomized design are needed to show the causal relationship between colchicine and cognition.
Collapse
Affiliation(s)
- Arzu Okyar Baş
- Department of Internal Medicine, Division of Geriatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Gözde Kübra Yardımcı
- Department of Internal Medicine, Division of Rheumatology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Merve Güner Oytun
- Department of Internal Medicine, Division of Geriatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Serdar Ceylan
- Department of Internal Medicine, Division of Geriatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Zeynep Kahyaoğlu
- Department of Internal Medicine, Division of Geriatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Yelda Öztürk
- Department of Internal Medicine, Division of Geriatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Süheyla Çöteli
- Department of Internal Medicine, Division of Geriatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Meltem Gülhan Halil
- Department of Internal Medicine, Division of Geriatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Mustafa Cankurtaran
- Department of Internal Medicine, Division of Geriatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Levent Kılıç
- Department of Internal Medicine, Division of Rheumatology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Burcu Balam Doğu Yavuz
- Department of Internal Medicine, Division of Geriatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
8
|
Zhang Q, Jiu Y. The regulation of host cytoskeleton during SARS-CoV-2 infection in the nervous system. BRAIN SCIENCE ADVANCES 2023. [DOI: 10.26599/bsa.2023.9050004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
The global economy and public health are currently under enormous pressure since the outbreak of COVID-19. Apart from respiratory discomfort, a subpopulation of COVID-19 patients exhibits neurological symptoms such as headache, myalgia, and loss of smell. Some have even shown encephalitis and necrotizing hemorrhagic encephalopathy. The cytoskeleton of nerve cells changes drastically in these pathologies, indicating that the cytoskeleton and its related proteins are closely related to the pathogenesis of nervous system diseases. In this review, we present the up-to-date association between host cytoskeleton and coronavirus infection in the context of the nervous system. We systematically summarize cytoskeleton-related pathogen-host interactions in both the peripheral and central nervous systems, hoping to contribute to the development of clinical treatment in COVID-19 patients.
Collapse
Affiliation(s)
- Qian Zhang
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yaming Jiu
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
9
|
Sîrbulescu RF, Ilieş I, Amelung L, Zupanc GKH. Proteomic characterization of spontaneously regrowing spinal cord following injury in the teleost fish Apteronotus leptorhynchus, a regeneration-competent vertebrate. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2022; 208:671-706. [PMID: 36445471 DOI: 10.1007/s00359-022-01591-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/30/2022] [Accepted: 11/01/2022] [Indexed: 11/30/2022]
Abstract
In adult mammals, spontaneous repair after spinal cord injury (SCI) is severely limited. By contrast, teleost fish successfully regenerate injured axons and produce new neurons from adult neural stem cells after SCI. The molecular mechanisms underlying this high regenerative capacity are largely unknown. The present study addresses this gap by examining the temporal dynamics of proteome changes in response to SCI in the brown ghost knifefish (Apteronotus leptorhynchus). Two-dimensional difference gel electrophoresis (2D DIGE) was combined with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) and tandem mass spectrometry (MS/MS) to collect data during early (1 day), mid (10 days), and late (30 days) phases of regeneration following caudal amputation SCI. Forty-two unique proteins with significant differences in abundance between injured and intact control samples were identified. Correlation analysis uncovered six clusters of spots with similar expression patterns over time and strong conditional dependences, typically within functional families or between isoforms. Significantly regulated proteins were associated with axon development and regeneration; proliferation and morphogenesis; neuronal differentiation and re-establishment of neural connections; promotion of neuroprotection, redox homeostasis, and membrane repair; and metabolism or energy supply. Notably, at all three time points examined, significant regulation of proteins involved in inflammatory responses was absent.
Collapse
Affiliation(s)
- Ruxandra F Sîrbulescu
- School of Engineering and Science, Jacobs University Bremen, 28725, Bremen, Germany
- Laboratory of Neurobiology, Department of Biology, Northeastern University, Boston, MA, 02115, USA
- Vaccine and Immunotherapy Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Iulian Ilieş
- School of Humanities and Social Sciences, Jacobs University Bremen, 28725, Bremen, Germany
- Laboratory of Neurobiology, Department of Biology, Northeastern University, Boston, MA, 02115, USA
| | - Lisa Amelung
- Laboratory of Neurobiology, Department of Biology, Northeastern University, Boston, MA, 02115, USA
| | - Günther K H Zupanc
- School of Engineering and Science, Jacobs University Bremen, 28725, Bremen, Germany.
- Laboratory of Neurobiology, Department of Biology, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
10
|
Romaus-Sanjurjo D, Saikia JM, Kim HJ, Tsai KM, Le GQ, Zheng B. Overexpressing eukaryotic elongation factor 1 alpha (eEF1A) proteins to promote corticospinal axon repair after injury. Cell Death Discov 2022; 8:390. [PMID: 36123349 PMCID: PMC9485247 DOI: 10.1038/s41420-022-01186-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 11/08/2022] Open
Abstract
Although protein synthesis is hypothesized to have a pivotal role in axonal repair after central nervous system (CNS) injury, the role of core components of the protein synthesis machinery has not been examined. Notably, some elongation factors possess non-canonical functions that may further impact axonal repair. Here, we examined whether overexpressing eukaryotic elongation factor 1 alpha (eEF1A) proteins enhances the collateral sprouting of corticospinal tract (CST) neurons after unilateral pyramidotomy, along with the underlying molecular mechanisms. We found that overexpressing eEF1A proteins in CST neurons increased the levels of pS6, an indicator for mTOR activity, but not pSTAT3 and pAKT levels, in neuronal somas. Strikingly, overexpressing eEF1A2 alone, but neither eEF1A1 alone nor both factors simultaneously, increased protein synthesis and actin rearrangement in CST neurons. While eEF1A1 overexpression only slightly enhanced CST sprouting after pyramidotomy, eEF1A2 overexpression substantially enhanced this sprouting. Surprisingly, co-overexpression of both eEF1A1 and eEF1A2 led to a sprouting phenotype similar to wild-type controls, suggesting an antagonistic effect of overexpressing both proteins. These data provide the first evidence that overexpressing a core component of the translation machinery, eEF1A2, enhances CST sprouting, likely by a combination of increased protein synthesis, mTOR signaling and actin cytoskeleton rearrangement.
Collapse
Affiliation(s)
- Daniel Romaus-Sanjurjo
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratories (LINCs), Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Junmi M Saikia
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Neurosciences Graduate Program, University of California San Diego, La Jolla, CA, 92093, USA
| | - Hugo J Kim
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Kristen M Tsai
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Geneva Q Le
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Binhai Zheng
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
- VA San Diego Research Service, San Diego, CA, 92161, USA.
| |
Collapse
|
11
|
Sun C, Dong Y, Wei J, Cai M, Liang D, Fu Y, Zhou Y, Sui Y, Wu F, Mikhaylov R, Wang H, Fan F, Xie Z, Stringer M, Yang Z, Wu Z, Tian L, Yang X. Acoustically Accelerated Neural Differentiation of Human Embryonic Stem Cells. Acta Biomater 2022; 151:333-345. [DOI: 10.1016/j.actbio.2022.07.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/14/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022]
|
12
|
Wu LG, Chan CY. Multiple Roles of Actin in Exo- and Endocytosis. Front Synaptic Neurosci 2022; 14:841704. [PMID: 35308832 PMCID: PMC8931529 DOI: 10.3389/fnsyn.2022.841704] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/11/2022] [Indexed: 11/20/2022] Open
Abstract
Cytoskeletal filamentous actin (F-actin) has long been considered a molecule that may regulate exo- and endocytosis. However, its exact roles remained elusive. Recent studies shed new light on many crucial roles of F-actin in regulating exo- and endocytosis. Here, this progress is reviewed from studies of secretory cells, particularly neurons and endocrine cells. These studies reveal that F-actin is involved in mediating all kinetically distinguishable forms of endocytosis, including ultrafast, fast, slow, bulk, and overshoot endocytosis, likely via membrane pit formation. F-actin promotes vesicle replenishment to the readily releasable pool most likely via active zone clearance, which may sustain synaptic transmission and overcome short-term depression of synaptic transmission during repetitive firing. By enhancing plasma membrane tension, F-actin promotes fusion pore expansion, vesicular content release, and a fusion mode called shrink fusion involving fusing vesicle shrinking. Not only F-actin, but also the F-actin assembly pathway, including ATP hydrolysis, N-WASH, and formin, are involved in mediating these roles of exo- and endocytosis. Neurological disorders, including spinocerebellar ataxia 13 caused by Kv3.3 channel mutation, may involve impairment of F-actin and its assembly pathway, leading in turn to impairment of exo- and endocytosis at synapses that may contribute to neurological disorders.
Collapse
Affiliation(s)
- Ling-Gang Wu
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
| | | |
Collapse
|
13
|
Subramanian S, Biswas A, Alves C, Sudhakar S, Shekdar K, Krishnan P, Shroff M, Taranath A, Arrigoni F, Aldinger K, Leventer R, Dobyns W, Mankad K. ACTA2-Related Dysgyria: An Under-Recognized Malformation of Cortical Development. AJNR Am J Neuroradiol 2022; 43:146-150. [PMID: 34857515 PMCID: PMC8757559 DOI: 10.3174/ajnr.a7364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/27/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND PURPOSE Pathogenic variants in the ACTA2 gene cause a distinctive arterial phenotype that has recently been described to be associated with brain malformation. Our objective was to further characterize gyral abnormalities in patients with ACTA2 pathogenic variants as per the 2020 consensus recommendations for the definition and classification of malformations of cortical development. MATERIALS AND METHODS We performed a retrospective, multicentric review of patients with proved ACTA2 pathogenic variants, searching for the presence of malformations of cortical development. A consensus read was performed for all patients, and the type and location of cortical malformation were noted in each. The presence of the typical ACTA2 arterial phenotype as well as demographic and relevant clinical data was obtained. RESULTS We included 13 patients with ACTA2 pathogenic variants (Arg179His mutation, n = 11, and Arg179Cys mutation, n = 2). Ninety-two percent (12/13) of patients had peri-Sylvian dysgyria, 77% (10/13) had frontal dysgyria, and 15% (2/13) had generalized dysgyria. The peri-Sylvian location was involved in all patients with dysgyria (12/12). All patients with dysgyria had a characteristic arterial phenotype described in ACTA2 pathogenic variants. One patient did not have dysgyria or the characteristic arterial phenotype. CONCLUSIONS Dysgyria is common in patients with ACTA2 pathogenic variants, with a peri-Sylvian and frontal predominance, and was seen in all our patients who also had the typical ACTA2 arterial phenotype.
Collapse
Affiliation(s)
- S. Subramanian
- From the Division of Pediatric Radiology (S.S.), Department of Radiology, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - A. Biswas
- Department of Diagnostic Imaging (A.B., P.K., M.S.), The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | | | - S.V. Sudhakar
- Department of Radiology (S.V.S., K.M.), Great Ormond Street Hospital, NHS Foundation Trust, London, UK
| | - K.V. Shekdar
- Department of Radiology, and Department of Radiology (K.V.S.), Perelman School of Medicine at the University of Pennsylvania, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - P. Krishnan
- Department of Diagnostic Imaging (A.B., P.K., M.S.), The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - M. Shroff
- Department of Diagnostic Imaging (A.B., P.K., M.S.), The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - A. Taranath
- Department of Medical Imaging (A.T.), Women’s and Children’s Hospital, Adelaide, South Australia, Australia
| | - F. Arrigoni
- Neuroimaging Lab (F.A.), Scientific Institute, Istituto di Ricovero e Cura a Carattere Scientifico Eugenio Medea, Bosisio Parini, Italy
| | - K.A. Aldinger
- Department of Pediatrics (K.A.A.), University of Washington School of Medicine, Seattle, Washington,Center for Integrative Brain Research (K.A.A., W.B.D.), Seattle Children’s Research Institute, Seattle, Washington
| | - R.J. Leventer
- Department of Neurology (R.J.L.), Royal Children’s Hospital and Murdoch Children’s Research Institute, Parkville, Victoria, Australia,Department of Pediatrics (R.J.L.), University of Melbourne, Melbourne, Victoria, Australia
| | - W.B. Dobyns
- Center for Integrative Brain Research (K.A.A., W.B.D.), Seattle Children’s Research Institute, Seattle, Washington,Division of Genetics and Metabolism (W.B.D.), Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - K. Mankad
- Department of Radiology (S.V.S., K.M.), Great Ormond Street Hospital, NHS Foundation Trust, London, UK
| |
Collapse
|
14
|
Tabusi M, Thorsdottir S, Lysandrou M, Narciso AR, Minoia M, Srambickal CV, Widengren J, Henriques-Normark B, Iovino F. Neuronal death in pneumococcal meningitis is triggered by pneumolysin and RrgA interactions with β-actin. PLoS Pathog 2021; 17:e1009432. [PMID: 33760879 PMCID: PMC7990213 DOI: 10.1371/journal.ppat.1009432] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/28/2021] [Indexed: 12/14/2022] Open
Abstract
Neuronal damage is a major consequence of bacterial meningitis, but little is known about mechanisms of bacterial interaction with neurons leading to neuronal cell death. Streptococcus pneumoniae (pneumococcus) is a leading cause of bacterial meningitis and many survivors develop neurological sequelae after the acute infection has resolved, possibly due to neuronal damage. Here, we studied mechanisms for pneumococcal interactions with neurons. Using human primary neurons, pull-down experiments and mass spectrometry, we show that pneumococci interact with the cytoskeleton protein β-actin through the pilus-1 adhesin RrgA and the cytotoxin pneumolysin (Ply), thereby promoting adhesion and invasion of neurons, and neuronal death. Using our bacteremia-derived meningitis mouse model, we observe that RrgA- and Ply-expressing pneumococci co-localize with neuronal β-actin. Using purified proteins, we show that Ply, through its cholesterol-binding domain 4, interacts with the neuronal plasma membrane, thereby increasing the exposure on the outer surface of β-actin filaments, leading to more β-actin binding sites available for RrgA binding, and thus enhanced pneumococcal interactions with neurons. Pneumococcal infection promotes neuronal death possibly due to increased intracellular Ca2+ levels depending on presence of Ply, as well as on actin cytoskeleton disassembly. STED super-resolution microscopy showed disruption of β-actin filaments in neurons infected with pneumococci expressing RrgA and Ply. Finally, neuronal death caused by pneumococcal infection could be inhibited using antibodies against β-actin. The generated data potentially helps explaining mechanisms for why pneumococci frequently cause neurological sequelae.
Collapse
Affiliation(s)
- Mahebali Tabusi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, BioClinicum J7:20, Stockholm, Sweden
- Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Sigrun Thorsdottir
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, BioClinicum J7:20, Stockholm, Sweden
- Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Lysandrou
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, BioClinicum J7:20, Stockholm, Sweden
- Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Ana Rita Narciso
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, BioClinicum J7:20, Stockholm, Sweden
- Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Melania Minoia
- Department of Molecular Biosciences, The Wenner-Gren Institutet, Stockholm University, Stockholm, Sweden
| | | | - Jerker Widengren
- Department of Applied Physics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Birgitta Henriques-Normark
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, BioClinicum J7:20, Stockholm, Sweden
- Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Federico Iovino
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, BioClinicum J7:20, Stockholm, Sweden
- Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
15
|
Rodriguez CM, Chun SY, Mills RE, Todd PK. Translation of upstream open reading frames in a model of neuronal differentiation. BMC Genomics 2019; 20:391. [PMID: 31109297 PMCID: PMC6528255 DOI: 10.1186/s12864-019-5775-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/07/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Upstream open reading frames (uORFs) initiate translation within mRNA 5' leaders, and have the potential to alter main coding sequence (CDS) translation on transcripts in which they reside. Ribosome profiling (RP) studies suggest that translating ribosomes are pervasive within 5' leaders across model systems. However, the significance of this observation remains unclear. To explore a role for uORF usage in a model of neuronal differentiation, we performed RP on undifferentiated and differentiated human neuroblastoma cells. RESULTS Using a spectral coherence algorithm (SPECtre), we identify 4954 consistently translated uORFs across 31% of all neuroblastoma transcripts. These uORFs predominantly utilize non-AUG initiation codons and exhibit translational efficiencies (TE) comparable to annotated coding regions. On a population basis, the global impact of both AUG and non-AUG initiated uORFs on basal CDS translation were small, even when analysis is limited to conserved and consistently translated uORFs. However, uORFs did alter the translation of a subset of genes, including the Diamond-Blackfan Anemia associated ribosomal gene RPS24. With retinoic acid induced differentiation, we observed an overall positive correlation in translational shifts between uORF/CDS pairs. However, CDSs downstream of uORFs show smaller shifts in TE with differentiation relative to CDSs without a predicted uORF, suggesting that uORF translation buffers cell state dependent fluctuations in CDS translation. CONCLUSION This work provides insights into the dynamic relationships and potential regulatory functions of uORF/CDS pairs in a model of neuronal differentiation.
Collapse
Affiliation(s)
- Caitlin M Rodriguez
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Sang Y Chun
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ryan E Mills
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Peter K Todd
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
- VA Ann Arbor Healthcare System, Ann Arbor, MI, USA.
| |
Collapse
|
16
|
D'Arco F, Alves CA, Raybaud C, Chong WKK, Ishak GE, Ramji S, Grima M, Barkovich AJ, Ganesan V. Expanding the Distinctive Neuroimaging Phenotype of ACTA2 Mutations. AJNR Am J Neuroradiol 2018; 39:2126-2131. [PMID: 30262641 DOI: 10.3174/ajnr.a5823] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/16/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND PURPOSE Arg179His mutations in ACTA2 are associated with a distinctive neurovascular phenotype characterized by a straight course of intracranial arteries, absent basal Moyamoya collaterals, dilation of the proximal internal carotid arteries, and occlusive disease of the terminal internal carotid arteries. We now add to the distinctive neuroimaging features in these patients by describing their unique constellation of brain malformative findings that could flag the diagnosis in cases in which targeted cerebrovascular imaging has not been performed. MATERIALS AND METHODS Neuroimaging studies from 13 patients with heterozygous Arg179His mutations in ACTA2 and 1 patient with pathognomonic clinicoradiologic findings for ACTA2 mutation were retrospectively reviewed. The presence and localization of brain malformations and other abnormal brain MR imaging findings are reported. RESULTS Characteristics bending and hypoplasia of the anterior corpus callosum, apparent absence of the anterior gyrus cinguli, and radial frontal gyration were present in 100% of the patients; flattening of the pons on the midline and multiple indentations in the lateral surface of the pons were demonstrated in 93% of the patients; and apparent "squeezing" of the cerebral peduncles in 85% of the patients. CONCLUSIONS Because α-actin is not expressed in the brain parenchyma, only in vascular tissue, we speculate that rather than a true malformative process, these findings represent a deformation of the brain during development related to the mechanical interaction with rigid arteries during the embryogenesis.
Collapse
Affiliation(s)
- F D'Arco
- From the Departments of Radiology (F.D'A., W.K.K.C.)
| | - C A Alves
- Radiology Department (C.A.A.), Hospital Das Clinicas, Sao Paulo, Brazil
| | - C Raybaud
- Department of Diagnostic Imaging (C.R.), Hospital for Sick Children, Toronto, Ontario, Canada
| | - W K K Chong
- From the Departments of Radiology (F.D'A., W.K.K.C.)
| | - G E Ishak
- Department of Radiology (G.E.I.), Seattle Children's Hospital, University of Washington School of Medicine, Seattle, Washington
| | - S Ramji
- Department of Radiology (S.R.), Imperial College Healthcare National Health Service Trust, London, UK
| | - M Grima
- Department of Radiology (M.G.), University Hospital of North Staffordshire National Health Service Trust, Stoke-on-Trent, UK
| | - A J Barkovich
- Department of Radiology and Diagnostic Imaging (A.J.B.), University of California, San Francisco, San Francisco, California
| | - V Ganesan
- Neurology (V.G.), Great Ormond Street Hospital for Children National Health Service Foundation Trust, London, UK
- Neuroscience Unit (V.G.), UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
17
|
Li HQ, Tan L, Yang HP, Pang W, Xu T, Jiang YG. Changes of hippocampus proteomic profiles after blueberry extracts supplementation in APP/PS1 transgenic mice. Nutr Neurosci 2018; 23:75-84. [PMID: 29781405 DOI: 10.1080/1028415x.2018.1471251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Objective: To examine protein changes in the hippocampus of APP/PS1 transgenic mice after blueberry extracts (BB) intervention.Methods: Eight APP/PS1 transgenic mice were randomly assigned to Alzheimer's disease (AD)+BB group (n=4) and AD+control group (n=4). After a 16-week treatment, 2-DE and MALDI-TOF-MS were used to compare the proteomic profiles of the hippocampus in the two groups and Western blot was used to confirm the important differentially expressed proteins.Results: Twelve proteins were differentially expressed between the two groups. Nine of them were identified. Cytochrome b-c1 complex subunit 6, beta-actin, dynamin 1, and heat shock cognate 71 were up-regulated in AD+BB group, while a-enolase, stress-induced-phosphoprotein 1, malate dehydrogenase (MDH), MDH 1, and T-complex protein 1 subunit beta were down-regulated, respectively. Importantly, some of the identified proteins (e.g. dynamin 1) are known to be involved in cognitive impairment. Western blot analysis of hippocampus dynamin 1 expression confirmed the proteomic findings.Conclusions: The consumption of BB modulates the expression of proteins that are linked to the improvements of cognitive dysfunction in hippocampus of APP/PS1 transgenic mice.
Collapse
Affiliation(s)
- Hai-Qiang Li
- Department of Nutrition, Tianjin Institute of Health and Environmental Medicine, Tianjin, People's Republic of China.,Yantai Economic and Technological Development Area Hospital, Yantai, People's Republic of China
| | - Long Tan
- Department of Nutrition and Food Security, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
| | - Hong-Peng Yang
- Tianjin Agricultural College, Tianjin, People's Republic of China
| | - Wei Pang
- Department of Nutrition, Tianjin Institute of Health and Environmental Medicine, Tianjin, People's Republic of China
| | - Tong Xu
- Department of Nutrition, Tianjin Institute of Health and Environmental Medicine, Tianjin, People's Republic of China
| | - Yu-Gang Jiang
- Department of Nutrition, Tianjin Institute of Health and Environmental Medicine, Tianjin, People's Republic of China
| |
Collapse
|
18
|
Snow WM, Cadonic C, Cortes-Perez C, Roy Chowdhury SK, Djordjevic J, Thomson E, Bernstein MJ, Suh M, Fernyhough P, Albensi BC. Chronic dietary creatine enhances hippocampal-dependent spatial memory, bioenergetics, and levels of plasticity-related proteins associated with NF-κB. ACTA ACUST UNITED AC 2018; 25:54-66. [PMID: 29339557 PMCID: PMC5772392 DOI: 10.1101/lm.046284.117] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 11/03/2017] [Indexed: 12/23/2022]
Abstract
The brain has a high demand for energy, of which creatine (Cr) is an important regulator. Studies document neurocognitive benefits of oral Cr in mammals, yet little is known regarding their physiological basis. This study investigated the effects of Cr supplementation (3%, w/w) on hippocampal function in male C57BL/6 mice, including spatial learning and memory in the Morris water maze and oxygen consumption rates from isolated mitochondria in real time. Levels of transcription factors and related proteins (CREB, Egr1, and IκB to indicate NF-κB activity), proteins implicated in cognition (CaMKII, PSD-95, and Egr2), and mitochondrial proteins (electron transport chain Complex I, mitochondrial fission protein Drp1) were probed with Western blotting. Dietary Cr decreased escape latency/time to locate the platform (P < 0.05) and increased the time spent in the target quadrant (P < 0.01) in the Morris water maze. This was accompanied by increased coupled respiration (P < 0.05) in isolated hippocampal mitochondria. Protein levels of CaMKII, PSD-95, and Complex 1 were increased in Cr-fed mice, whereas IκB was decreased. These data demonstrate that dietary supplementation with Cr can improve learning, memory, and mitochondrial function and have important implications for the treatment of diseases affecting memory and energy homeostasis.
Collapse
Affiliation(s)
- Wanda M Snow
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba R2H 2A6, Canada
| | - Chris Cadonic
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba R2H 2A6, Canada
| | - Claudia Cortes-Perez
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba R2H 2A6, Canada
| | - Subir K Roy Chowdhury
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba R2H 2A6, Canada
| | - Jelena Djordjevic
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba R2H 2A6, Canada
| | - Ella Thomson
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba R2H 2A6, Canada
| | - Michael J Bernstein
- Department of Psychological and Social Sciences, Pennsylvania State University Abington, Abington, Pennsylvania 19001, USA
| | - Miyoung Suh
- Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Paul Fernyhough
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba R2H 2A6, Canada.,Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, Manitoba R3E 0T6, Canada
| | - Benedict C Albensi
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba R2H 2A6, Canada.,Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, Manitoba R3E 0T6, Canada
| |
Collapse
|
19
|
Simhadri PK, Malwade R, Vanka R, Nakka VP, Kuppusamy G, Babu PP. Dysregulation of LIMK-1/cofilin-1 pathway: A possible basis for alteration of neuronal morphology in experimental cerebral malaria. Ann Neurol 2017; 82:429-443. [PMID: 28843047 DOI: 10.1002/ana.25028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 08/02/2017] [Accepted: 08/18/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Loss of cognition even after survival is the salient feature of cerebral malaria (CM). Currently, the fate of neuronal morphology is not studied at the ultrastructural level in CM. Recent studies suggest that maintenance of neuronal morphology and dendritic spine density (actin dynamics in particular) are essential for proper cognitive function. LIMK-1/cofilin-1 signaling pathway is known to be involved in the maintenance of actin dynamics through regulation of cofilin-1, and in executing learning and memory functions. METHODS Using an experimental mouse model, we analyzed the behavioral parameters of asymptomatic mice with CM by performing a rapid murine coma and behavior scale experiment. We performed Golgi-Cox staining to assess neuronal morphology, dendritic spine density, and arborization in brain cortex subjected to Plasmodium berghei ANKA infection compared to asymptomatic, anemic, and control groups. We studied the neural gene expression pattern of LIMK-1, cofilin-1, and β-actin in all the experimental groups by semiquantitative and quantitative polymerase chain reaction followed by immunoblotting and immunofluorescence. RESULTS We observed significant loss of dendritic spine density, abnormal spine morphology, reduced dendritic arborization, and extensive dendritic varicosities in the cortical neurons of CM-infected brain. Furthermore, these observations correlated with diminished protein levels of LIMK-1, cofilin-1, phospho-cofilin-1, and β-actin in the whole brain lysates as well as formation of actin-cofilin rods in the brain sections of symptomatic mice with CM. INTERPRETATION Overall, our findings suggest that the altered neuronal morphology and dysregulation of LIMK-1/cofilin-1 pathway could affect the cognitive outcome after experimental CM. Therefore, this study could help to establish newer therapeutic strategies addressing long-term cognitive impairment after CM. Ann Neurol 2017;82:429-443.
Collapse
Affiliation(s)
- Praveen Kumar Simhadri
- Department of Biotechnology and Bioinformatics, School of life Sciences, University of Hyderabad, Hyderabad, Telangana
| | - Ruchi Malwade
- Department of Biotechnology and Bioinformatics, School of life Sciences, University of Hyderabad, Hyderabad, Telangana
| | - Ravisankar Vanka
- Department of Pharmaceutics, JSS College of Pharmacy, Udhagamandalam, Tamil Nadu, India
| | - Venkata Prasuja Nakka
- Department of Biotechnology and Bioinformatics, School of life Sciences, University of Hyderabad, Hyderabad, Telangana
| | | | - Phanithi Prakash Babu
- Department of Biotechnology and Bioinformatics, School of life Sciences, University of Hyderabad, Hyderabad, Telangana
| |
Collapse
|
20
|
Lei W, Myers KR, Rui Y, Hladyshau S, Tsygankov D, Zheng JQ. Phosphoinositide-dependent enrichment of actin monomers in dendritic spines regulates synapse development and plasticity. J Cell Biol 2017; 216:2551-2564. [PMID: 28659327 PMCID: PMC5551708 DOI: 10.1083/jcb.201612042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/03/2017] [Accepted: 05/18/2017] [Indexed: 12/17/2022] Open
Abstract
Dendritic spines are small postsynaptic compartments of excitatory synapses in the vertebrate brain that are modified during learning, aging, and neurological disorders. The formation and modification of dendritic spines depend on rapid assembly and dynamic remodeling of the actin cytoskeleton in this highly compartmentalized space, but the precise mechanisms remain to be fully elucidated. In this study, we report that spatiotemporal enrichment of actin monomers (G-actin) in dendritic spines regulates spine development and plasticity. We first show that dendritic spines contain a locally enriched pool of G-actin that can be regulated by synaptic activity. We further find that this G-actin pool functions in spine development and its modification during synaptic plasticity. Mechanistically, the relatively immobile G-actin pool in spines depends on the phosphoinositide PI(3,4,5)P3 and involves the actin monomer-binding protein profilin. Together, our results have revealed a novel mechanism by which dynamic enrichment of G-actin in spines regulates the actin remodeling underlying synapse development and plasticity.
Collapse
Affiliation(s)
- Wenliang Lei
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
- Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA
| | - Kenneth R Myers
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
- Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA
| | - Yanfang Rui
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
- Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA
| | - Siarhei Hladyshau
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
| | - Denis Tsygankov
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
| | - James Q Zheng
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA
- Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
21
|
Axonal transport deficits in multiple sclerosis: spiraling into the abyss. Acta Neuropathol 2017; 134:1-14. [PMID: 28315956 PMCID: PMC5486629 DOI: 10.1007/s00401-017-1697-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 12/16/2022]
Abstract
The transport of mitochondria and other cellular components along the axonal microtubule cytoskeleton plays an essential role in neuronal survival. Defects in this system have been linked to a large number of neurological disorders. In multiple sclerosis (MS) and associated models such as experimental autoimmune encephalomyelitis (EAE), alterations in axonal transport have been shown to exist before neurodegeneration occurs. Genome-wide association (GWA) studies have linked several motor proteins to MS susceptibility, while neuropathological studies have shown accumulations of proteins and organelles suggestive for transport deficits. A reduced effectiveness of axonal transport can lead to neurodegeneration through inhibition of mitochondrial motility, disruption of axoglial interaction or prevention of remyelination. In MS, demyelination leads to dysregulation of axonal transport, aggravated by the effects of TNF-alpha, nitric oxide and glutamate on the cytoskeleton. The combined effect of all these pathways is a vicious cycle in which a defective axonal transport system leads to an increase in ATP consumption through loss of membrane organization and a reduction in available ATP through inhibition of mitochondrial transport, resulting in even further inhibition of transport. The persistent activity of this positive feedback loop contributes to neurodegeneration in MS.
Collapse
|
22
|
Keskindemirci G, Eskikurt G, Ayaz NA, Çakan M, Ermutlu N, İşoğlu Alkaç Ü. Does familial Mediterranean fever affect cognitive function in children? Electrophysiological preliminary study. Int J Neurosci 2017; 128:10-14. [PMID: 28581880 DOI: 10.1080/00207454.2017.1338697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVES Familial Mediterranean fever (FMF) is a periodic autoinflammatory disease with subclinical inflammation occurring between attacks. The aim of the study was to prospectively evaluate the cognitive function of children diagnosed with FMF that were under colchicine therapy and compare them with healthy controls through electrophysiologically event-related potentials (ERPs) study. METHODS Twelve children with FMF and 12 healthy controls were included in the study. During the electroencephalography recordings, all participants were instructed to discriminate rare stimuli (target stimuli) from frequent stimuli (standard stimuli) by pressing a botton on a mouse immediately following the target stimulus. P300, the cognitive component of ERP, was obtained in response to target stimuli and its amplitude and latency were measured. RESULTS The amplitude of the P300 of the FMF patients was higher and the latencies of the P300 of the FMF patients were shorter than the amplitudes and latencies of control patients, respectively. The difference between the groups was statistically significant for amplitude but not for latency. CONCLUSIONS Cognitive processing reflecting allocation of attention and visual processing speed seems not to be negatively affected in FMF patients with homozygous M694V mutations undergoing colchicine treatment. As this study is unique in its evaluation of the cognitive function of children with FMF, these findings may be helpful for counseling families and patients affected by the condition.
Collapse
Affiliation(s)
- Gonca Keskindemirci
- a Pediatric Rheumatology Clinic , Kanuni Sultan Süleyman Training and Research Hospital , İstanbul , Turkey
| | - Gökçer Eskikurt
- b Department of Neuroscience , Institude of Experimental Medicine, İstanbul University , İstanbul , Turkey
| | - Nuray Aktay Ayaz
- a Pediatric Rheumatology Clinic , Kanuni Sultan Süleyman Training and Research Hospital , İstanbul , Turkey
| | - Mustafa Çakan
- a Pediatric Rheumatology Clinic , Kanuni Sultan Süleyman Training and Research Hospital , İstanbul , Turkey
| | - Numan Ermutlu
- c Department of Physiology, Faculty of Medicine , İstanbul Bilim University , İstabul , Turkey
| | - Ümmühan İşoğlu Alkaç
- d Department of Physiology, İstanbul Medical Faculty , İstanbul University , İstanbul , Turkey
| |
Collapse
|
23
|
Actin Is Crucial for All Kinetically Distinguishable Forms of Endocytosis at Synapses. Neuron 2016; 92:1020-1035. [PMID: 27840001 DOI: 10.1016/j.neuron.2016.10.014] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 06/16/2016] [Accepted: 10/04/2016] [Indexed: 01/18/2023]
Abstract
Mechanical force is needed to mediate endocytosis. Whether actin, the most abundant force-generating molecule, is essential for endocytosis is highly controversial in mammalian cells, particularly synapses, likely due to the use of actin blockers, the efficiency and specificity of which are often unclear in the studied cell. Here we addressed this issue using a knockout approach combined with measurements of membrane capacitance and fission pore conductance, imaging of vesicular protein endocytosis, and electron microscopy. We found that two actin isoforms, β- and γ-actin, are crucial for slow, rapid, bulk, and overshoot endocytosis at large calyx-type synapses, and for slow endocytosis and bulk endocytosis at small hippocampal synapses. Polymerized actin provides mechanical force to form endocytic pits. Actin also facilitates replenishment of the readily releasable vesicle pool, likely via endocytic clearance of active zones. We conclude that polymerized actin provides mechanical force essential for all kinetically distinguishable forms of endocytosis at synapses.
Collapse
|
24
|
Wen PJ, Grenklo S, Arpino G, Tan X, Liao HS, Heureaux J, Peng SY, Chiang HC, Hamid E, Zhao WD, Shin W, Näreoja T, Evergren E, Jin Y, Karlsson R, Ebert SN, Jin A, Liu AP, Shupliakov O, Wu LG. Actin dynamics provides membrane tension to merge fusing vesicles into the plasma membrane. Nat Commun 2016; 7:12604. [PMID: 27576662 PMCID: PMC5013665 DOI: 10.1038/ncomms12604] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 07/13/2016] [Indexed: 01/22/2023] Open
Abstract
Vesicle fusion is executed via formation of an Ω-shaped structure (Ω-profile), followed by closure (kiss-and-run) or merging of the Ω-profile into the plasma membrane (full fusion). Although Ω-profile closure limits release but recycles vesicles economically, Ω-profile merging facilitates release but couples to classical endocytosis for recycling. Despite its crucial role in determining exocytosis/endocytosis modes, how Ω-profile merging is mediated is poorly understood in endocrine cells and neurons containing small ∼30–300 nm vesicles. Here, using confocal and super-resolution STED imaging, force measurements, pharmacology and gene knockout, we show that dynamic assembly of filamentous actin, involving ATP hydrolysis, N-WASP and formin, mediates Ω-profile merging by providing sufficient plasma membrane tension to shrink the Ω-profile in neuroendocrine chromaffin cells containing ∼300 nm vesicles. Actin-directed compounds also induce Ω-profile accumulation at lamprey synaptic active zones, suggesting that actin may mediate Ω-profile merging at synapses. These results uncover molecular and biophysical mechanisms underlying Ω-profile merging. As vesicles fuse to the plasma membrane, they form intermediate Ω-shaped structures followed by either closure of the pore or full merging with the plasma membrane. Here Wen et al. show that dynamic actin assembly provides membrane tension to promote Ω merging in neuroendocrine cells and synapses.
Collapse
Affiliation(s)
- Peter J Wen
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| | - Staffan Grenklo
- Center of Excellence in Developmental Biology, Department of Neuroscience, Karolinska Institutet, S-171 77 Stockholm, Sweden.,Department of Cell Biology, WGI, Stockholm University, 106 91 Stockholm, Sweden
| | - Gianvito Arpino
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA.,Center of Excellence in Developmental Biology, Department of Neuroscience, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Xinyu Tan
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Hsien-Shun Liao
- National Institute of Biomedical Imaging and Bioengineering (NIBIB), Bethesda, Maryland 20892, USA
| | - Johanna Heureaux
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Shi-Yong Peng
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| | - Hsueh-Cheng Chiang
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| | - Edaeni Hamid
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| | - Wei-Dong Zhao
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| | - Wonchul Shin
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| | - Tuomas Näreoja
- Center of Excellence in Developmental Biology, Department of Neuroscience, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Emma Evergren
- Center of Excellence in Developmental Biology, Department of Neuroscience, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Yinghui Jin
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| | - Roger Karlsson
- Department of Cell Biology, WGI, Stockholm University, 106 91 Stockholm, Sweden
| | - Steven N Ebert
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Boulevard, Orlando, Florida 32827, USA
| | - Albert Jin
- National Institute of Biomedical Imaging and Bioengineering (NIBIB), Bethesda, Maryland 20892, USA
| | - Allen P Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Oleg Shupliakov
- Center of Excellence in Developmental Biology, Department of Neuroscience, Karolinska Institutet, S-171 77 Stockholm, Sweden.,Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia
| | - Ling-Gang Wu
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| |
Collapse
|
25
|
Activation of the cAMP Pathway Induces RACK1-Dependent Binding of β-Actin to BDNF Promoter. PLoS One 2016; 11:e0160948. [PMID: 27505161 PMCID: PMC4978483 DOI: 10.1371/journal.pone.0160948] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 07/27/2016] [Indexed: 11/28/2022] Open
Abstract
RACK1 is a scaffolding protein that contributes to the specificity and propagation of several signaling cascades including the cAMP pathway. As such, RACK1 participates in numerous cellular functions ranging from cell migration and morphology to gene transcription. To obtain further insights on the mechanisms whereby RACK1 regulates cAMP-dependent processes, we set out to identify new binding partners of RACK1 during activation of the cAMP signaling using a proteomics strategy. We identified β-actin as a direct RACK1 binding partner and found that the association between β-actin and RACK1 is increased in response to the activation of the cAMP pathway. Furthermore, we show that cAMP-dependent increase in BDNF expression requires filamentous actin. We further report that β-actin associates with the BDNF promoter IV upon the activation of the cAMP pathway and present data to suggest that the association of β-actin with BDNF promoter IV is RACK1-dependent. Taken together, our data suggest that β-actin is a new RACK1 binding partner and that the RACK1 and β-actin association participate in the cAMP-dependent regulation of BDNF transcription.
Collapse
|
26
|
Yeh CW, Hsu LS. Zebrafish diras1 Promoted Neurite Outgrowth in Neuro-2a Cells and Maintained Trigeminal Ganglion Neurons In Vivo via Rac1-Dependent Pathway. Mol Neurobiol 2015; 53:6594-6607. [PMID: 26635085 DOI: 10.1007/s12035-015-9550-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 11/18/2015] [Indexed: 01/25/2023]
Abstract
The small GTPase Ras superfamily regulates several neuronal functions including neurite outgrowth and neuron proliferation. In this study, zebrafish diras1a and diras1b were identified and were found to be mainly expressed in the central nervous system and dorsal neuron ganglion. Overexpression of green fluorescent protein (GFP)-diras1a or GFP-diras1b triggered neurite outgrowth of Neuro-2a cells. The wild types, but not the C terminus truncated forms, of diras1a and diras1b elevated the protein level of Ras-related C3 botulinum toxin substrate 1 (Rac1) and downregulated Ras homologous member A (RhoA) expression. Glutathione S-transferase (GST) pull-down assay also revealed that diras1a and diras1b enhanced Rac1 activity. Interfering with Rac1, Pak1, or cyclin-dependent kinase 5 (CDK5) activity or with the Arp2/3 inhibitor prevented diras1a and diras1b from mediating the neurite outgrowth effects. In the zebrafish model, knockdown of diras1a and/or diras1b by morpholino antisense oligonucleotides not only reduced axon guidance but also caused the loss of trigeminal ganglion without affecting the precursor markers, such as ngn1 and neuroD. Co-injection with messenger RNA (mRNA) derived from mouse diras1 or constitutively active human Rac1 restored the population of trigeminal ganglion. In conclusion, we provided preliminary evidence that diras1 is involved in neurite outgrowth and maintains the number of trigeminal ganglions through the Rac1-dependent pathway.
Collapse
Affiliation(s)
- Chi-Wei Yeh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, No. 110, Sec.1, Jianguo N. Rd., Taichung, 40201, Taiwan
| | - Li-Sung Hsu
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, No. 110, Sec.1, Jianguo N. Rd., Taichung, 40201, Taiwan.
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan.
| |
Collapse
|
27
|
Schreiber J, Végh MJ, Dawitz J, Kroon T, Loos M, Labonté D, Li KW, Van Nierop P, Van Diepen MT, De Zeeuw CI, Kneussel M, Meredith RM, Smit AB, Van Kesteren RE. Ubiquitin ligase TRIM3 controls hippocampal plasticity and learning by regulating synaptic γ-actin levels. J Cell Biol 2015; 211:569-86. [PMID: 26527743 PMCID: PMC4639863 DOI: 10.1083/jcb.201506048] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 10/06/2015] [Indexed: 12/24/2022] Open
Abstract
TRIM3 regulates synaptic γ-actin levels. TRIM3-deficient mice consequently have higher hippocampal spine densities, increased long-term potentiation, and enhanced contextual fear memory consolidation, indicating that temporal control of ACTG1 levels by TRIM3 is required to constrain hippocampal plasticity within physiological boundaries. Synaptic plasticity requires remodeling of the actin cytoskeleton. Although two actin isoforms, β- and γ-actin, are expressed in dendritic spines, the specific contribution of γ-actin in the expression of synaptic plasticity is unknown. We show that synaptic γ-actin levels are regulated by the E3 ubiquitin ligase TRIM3. TRIM3 protein and Actg1 transcript are colocalized in messenger ribonucleoprotein granules responsible for the dendritic targeting of messenger RNAs. TRIM3 polyubiquitylates γ-actin, most likely cotranslationally at synaptic sites. Trim3−/− mice consequently have increased levels of γ-actin at hippocampal synapses, resulting in higher spine densities, increased long-term potentiation, and enhanced short-term contextual fear memory consolidation. Interestingly, hippocampal deletion of Actg1 caused an increase in long-term fear memory. Collectively, our findings suggest that temporal control of γ-actin levels by TRIM3 is required to regulate the timing of hippocampal plasticity. We propose a model in which TRIM3 regulates synaptic γ-actin turnover and actin filament stability and thus forms a transient inhibitory constraint on the expression of hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- Joerg Schreiber
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Marlene J Végh
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Julia Dawitz
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Tim Kroon
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Maarten Loos
- Sylics (Synaptologics BV), 1008 BA Amsterdam, Netherlands
| | - Dorthe Labonté
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Pim Van Nierop
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Michiel T Van Diepen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus Medical Center, 3000 DR Rotterdam, Netherlands Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Science, 1105 BA Amsterdam, Netherlands
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Rhiannon M Meredith
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Ronald E Van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| |
Collapse
|
28
|
Kline CF, Scott J, Curran J, Hund TJ, Mohler PJ. Ankyrin-B regulates Cav2.1 and Cav2.2 channel expression and targeting. J Biol Chem 2014; 289:5285-95. [PMID: 24394417 DOI: 10.1074/jbc.m113.523639] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
N-type and P/Q-type calcium channels are documented players in the regulation of synaptic function; however, the mechanisms underlying their expression and cellular targeting are poorly understood. Ankyrin polypeptides are essential for normal integral membrane protein expression in a number of cell types, including neurons, cardiomyocytes, epithelia, secretory cells, and erythrocytes. Ankyrin dysfunction has been linked to defects in integral protein expression, abnormal cellular function, and disease. Here, we demonstrate that ankyrin-B associates with Cav2.1 and Cav2.2 in cortex, cerebellum, and brain stem. Additionally, using in vitro and in vivo techniques, we demonstrate that ankyrin-B, via its membrane-binding domain, associates with a highly conserved motif in the DII/III loop domain of Cav2.1 and Cav2.2. Further, we demonstrate that this domain is necessary for proper targeting of Cav2.1 and Cav2.2 in a heterologous system. Finally, we demonstrate that mutation of a single conserved tyrosine residue in the ankyrin-binding motif of both Cav2.1 (Y797E) and Cav2.2 (Y788E) results in loss of association with ankyrin-B in vitro and in vivo. Collectively, our findings identify an interaction between ankyrin-B and both Cav2.1 and Cav2.2 at the amino acid level that is necessary for proper Cav2.1 and Cav2.2 targeting in vivo.
Collapse
|
29
|
Erythrocyte shape abnormalities, membrane oxidative damage, and β-actin alterations: an unrecognized triad in classical autism. Mediators Inflamm 2013; 2013:432616. [PMID: 24453417 PMCID: PMC3880759 DOI: 10.1155/2013/432616] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 10/22/2013] [Indexed: 12/27/2022] Open
Abstract
Autism spectrum disorders (ASDs) are a complex group of neurodevelopment disorders steadily rising in frequency and treatment refractory, where the search for biological markers is of paramount importance. Although red blood cells (RBCs) membrane lipidomics and rheological variables have been reported to be altered, with some suggestions indicating an increased lipid peroxidation in the erythrocyte membrane, to date no information exists on how the oxidative membrane damage may affect cytoskeletal membrane proteins and, ultimately, RBCs shape in autism. Here, we investigated RBC morphology by scanning electron microscopy in patients with classical autism, that is, the predominant ASDs phenotype (age range: 6–26 years), nonautistic neurodevelopmental disorders (i.e., “positive controls”), and healthy controls (i.e., “negative controls”). A high percentage of altered RBCs shapes, predominantly elliptocytes, was observed in autistic patients, but not in both control groups. The RBCs altered morphology in autistic subjects was related to increased erythrocyte membrane F2-isoprostanes and 4-hydroxynonenal protein adducts. In addition, an oxidative damage of the erythrocyte membrane β-actin protein was evidenced. Therefore, the combination of erythrocyte shape abnormalities, erythrocyte membrane oxidative damage, and β-actin alterations constitutes a previously unrecognized triad in classical autism and provides new biological markers in the diagnostic workup of ASDs.
Collapse
|
30
|
Ma M. Role of calpains in the injury-induced dysfunction and degeneration of the mammalian axon. Neurobiol Dis 2013; 60:61-79. [PMID: 23969238 PMCID: PMC3882011 DOI: 10.1016/j.nbd.2013.08.010] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 07/17/2013] [Accepted: 08/08/2013] [Indexed: 12/21/2022] Open
Abstract
Axonal injury and degeneration, whether primary or secondary, contribute to the morbidity and mortality seen in many acquired and inherited central nervous system (CNS) and peripheral nervous system (PNS) disorders, such as traumatic brain injury, spinal cord injury, cerebral ischemia, neurodegenerative diseases, and peripheral neuropathies. The calpain family of proteases has been mechanistically linked to the dysfunction and degeneration of axons. While the direct mechanisms by which transection, mechanical strain, ischemia, or complement activation trigger intra-axonal calpain activity are likely different, the downstream effects of unregulated calpain activity may be similar in seemingly disparate diseases. In this review, a brief examination of axonal structure is followed by a focused overview of the calpain family. Finally, the mechanisms by which calpains may disrupt the axonal cytoskeleton, transport, and specialized domains (axon initial segment, nodes, and terminals) are discussed.
Collapse
Affiliation(s)
- Marek Ma
- Department of Emergency Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Resuscitation Science, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|