1
|
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: 0] [Impact Index Per Article: 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
|
2
|
Guan W, Gao H, Sun S, Zheng T, Wu L, Wang X, Huang R, Li G. Multi-scale, multi-level anisotropic silk fibroin/metformin scaffolds for repair of peripheral nerve injury. Int J Biol Macromol 2023; 246:125518. [PMID: 37353122 DOI: 10.1016/j.ijbiomac.2023.125518] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
Silk fibroin (SF) as a natural polymer has a long history of application in various regenerative medicine fields, but there are still many shortcomings in silk fibroin for using as nerve scaffolds, which limit its clinical application in peripheral nerve regeneration (PNR). In this work, a multi-scale and multi-level metformin (MF)-loaded silk fibroin scaffold with anisotropic micro-nano composite topology was prepared by micromolding electrospinning for accelerating PNR. The scaffolds were characterized for morphology, wettability, mechanical properties, degradability, and drug release, and Schwann cells (SCs) and dorsal root ganglia (DRG) were cultured on the scaffolds to assess their effects on neural cell behavior. Finally, the gene expression differences of neural cells cultured on scaffolds were analyzed by gene sequencing and RT-qPCR to explore the possible signaling pathways and mechanisms. The results showed that the scaffolds had excellent mechanical properties and hydrophilicity, slow degradation rate and drug release rate, which were enough to support the repair of peripheral nerve injury for a long time. In Vitro cell experiments showed that the scaffolds could significantly promote the orientation of SCs and axons extension of DRG. Gene sequencing and RT-qPCR revealed that the scaffolds could up-regulate the expression of genes related to SCs proliferation, adhesion, migration, and myelination. In summary, the scaffolds hold great potential for promoting PNR at the micro/nano multiscale and physical/chemical levels and show promising application for the treatment of peripheral nerve injury in the future.
Collapse
Affiliation(s)
- Wenchao Guan
- Key laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Hongxia Gao
- Key laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Shaolan Sun
- Key laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Tiantian Zheng
- Key laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Linliang Wu
- Key laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Xiaolu Wang
- Suzhou Simatech CO.,LTD., Suzhou 215123, China
| | - Ran Huang
- Zhejiang Silkseekers Biotechnology CO., LTD., Hangzhou 310004, China
| | - Guicai Li
- Key laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.
| |
Collapse
|
3
|
Ghrelin/GHS-R1A antagonism in memory test and its effects on central molecular signaling involved in addiction in rats. Pharmacol Biochem Behav 2023; 224:173528. [PMID: 36870422 DOI: 10.1016/j.pbb.2023.173528] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/23/2022] [Accepted: 02/12/2023] [Indexed: 03/06/2023]
Abstract
Central ghrelin signaling seems to play important role in addiction as well as memory processing. Antagonism of the growth hormone secretagogue receptor (GHS-R1A) has been recently proposed as a promising tool for the unsatisfactory drug addiction therapy. However, molecular aspects of GHS-R1A involvement in specific brain regions remain unclear. The present study demonstrated for the first time that acute as well as subchronic (4 days) administration of the experimental GHS-R1A antagonist JMV2959 in usual intraperitoneal doses including 3 mg/kg, had no influence on memory functions tested in the Morris Water Maze in rats as well as no significant effects on the molecular markers linked with memory processing in selected brain areas in rats, specifically on the β-actin, c-Fos, two forms of the calcium/calmodulin-dependent protein kinase II (CaMKII, p-CaMKII) and the cAMP-response element binding protein (CREB, p-CREB), within the medial prefrontal cortex (mPFC), nucleus accumbens (NAc), dorsal striatum, and hippocampus (HIPP). Furthermore, following the methamphetamine intravenous self-administration in rats, the 3 mg/kg JMV2959 pretreatment significantly reduced or prevented the methamphetamine-induced significant decrease of hippocampal β-actin and c-Fos as well as it prevented the significant decrease of CREB in the NAC and mPFC. These results imply, that the GHS-R1A antagonist/JMV2959 might reduce/prevent some of the memory-linked molecular changes elicited by methamphetamine addiction within brain structures associated with memory (HIPP), reward (NAc), and motivation (mPFC), which may contribute to the previously observed significant JMV2959-induced reduction of the methamphetamine self-administration and drug-seeking behavior in the same animals. Further research is necessary to corroborate these results.
Collapse
|
4
|
Actin dynamics in protein homeostasis. Biosci Rep 2022; 42:231720. [PMID: 36043949 PMCID: PMC9469105 DOI: 10.1042/bsr20210848] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/22/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
Cell homeostasis is maintained in all organisms by the constant adjustment of cell constituents and organisation to account for environmental context. Fine-tuning of the optimal balance of proteins for the conditions, or protein homeostasis, is critical to maintaining cell homeostasis. Actin, a major constituent of the cytoskeleton, forms many different structures which are acutely sensitive to the cell environment. Furthermore, actin structures interact with and are critically important for the function and regulation of multiple factors involved with mRNA and protein production and degradation, and protein regulation. Altogether, actin is a key, if often overlooked, regulator of protein homeostasis across eukaryotes. In this review, we highlight these roles and how they are altered following cell stress, from mRNA transcription to protein degradation.
Collapse
|
5
|
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: 1.5] [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
|
6
|
Xie X, Mahmood SR, Gjorgjieva T, Percipalle P. Emerging roles of cytoskeletal proteins in regulating gene expression and genome organization during differentiation. Nucleus 2020; 11:53-65. [PMID: 32212905 PMCID: PMC7289583 DOI: 10.1080/19491034.2020.1742066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the eukaryotic cell nucleus, cytoskeletal proteins are emerging as essential players in nuclear function. In particular, actin regulates chromatin as part of ATP-dependent chromatin remodeling complexes, it modulates transcription and it is incorporated into nascent ribonucleoprotein complexes, accompanying them from the site of transcription to polyribosomes. The nuclear actin pool is undistinguishable from the cytoplasmic one in terms of its ability to undergo polymerization and it has also been implicated in the dynamics of chromatin, regulating heterochromatin segregation at the nuclear lamina and maintaining heterochromatin levels in the nuclear interiors. One of the next frontiers is, therefore, to determine a possible involvement of nuclear actin in the functional architecture of the cell nucleus by regulating the hierarchical organization of chromatin and, thus, genome organization. Here, we discuss the repertoire of these potential actin functions and how they are likely to play a role in the context of cellular differentiation.
Collapse
Affiliation(s)
- Xin Xie
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
| | - S Raza Mahmood
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates.,Department of Biology, New York University, New York, NY, USA
| | - Tamara Gjorgjieva
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
| | - Piergiorgio Percipalle
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
7
|
Vanslembrouck B, Ampe C, Hengel J. Time for rethinking the different β‐actin transgenic mouse models? Cytoskeleton (Hoboken) 2020; 77:527-543. [DOI: 10.1002/cm.21647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 11/19/2020] [Accepted: 11/24/2020] [Indexed: 01/23/2023]
Affiliation(s)
- Bieke Vanslembrouck
- Medical Cell Biology Research Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences Ghent University Ghent Belgium
| | - Christophe Ampe
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences Ghent University Ghent Belgium
| | - Jolanda Hengel
- Medical Cell Biology Research Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences Ghent University Ghent Belgium
| |
Collapse
|
8
|
Lechuga S, Naydenov NG, Feygin A, Cruise M, Ervasti JM, Ivanov AI. Loss of β-Cytoplasmic Actin in the Intestinal Epithelium Increases Gut Barrier Permeability in vivo and Exaggerates the Severity of Experimental Colitis. Front Cell Dev Biol 2020; 8:588836. [PMID: 33195251 PMCID: PMC7644907 DOI: 10.3389/fcell.2020.588836] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/06/2020] [Indexed: 12/21/2022] Open
Abstract
Intestinal epithelial barrier is critical for the maintenance of normal gut homeostasis and disruption of this barrier may trigger or exaggerate mucosal inflammation. The actin cytoskeleton is a key regulator of barrier structure and function, controlling the assembly and permeability of epithelial adherens and tight junctions. Epithelial cells express two actin isoforms: a β-cytoplasmic actin and γ-cytoplasmic actin. Our previous in vitro studies demonstrated that these actin isoforms play distinctive roles in establishing the intestinal epithelial barrier, by controlling the organization of different junctional complexes. It remains unknown, whether β-actin and γ-actin have unique or redundant functions in regulating the gut barrier in vivo. To address this question, we selectively knocked out β-actin expression in mouse intestinal epithelium. Mice with intestinal epithelial knockout of β-actin do not display gastrointestinal abnormalities or gross alterations of colonic mucosal architecture. This could be due to compensatory upregulation of γ-actin expression. Despite such compensation, β-actin knockout mice demonstrate increased intestinal permeability. Furthermore, these animals show more severe clinical symptoms during dextran sodium sulfate induced colitis, compared to control littermates. Such exaggerated colitis is associated with the higher expression of inflammatory cytokines, increased macrophage infiltration in the gut, and accelerated mucosal cell death. Consistently, intestinal organoids generated from β-actin knockout mice are more sensitive to tumor necrosis factor induced cell death, ex vivo. Overall, our data suggests that β-actin functions as an essential regulator of gut barrier integrity in vivo, and plays a tissue protective role during mucosal injury and inflammation.
Collapse
Affiliation(s)
- Susana Lechuga
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Nayden G Naydenov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Alex Feygin
- School of Nursing, Virginia Commonwealth University School of Nursing, Richmond, VA, United States
| | - Michael Cruise
- Department of Pathology, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - James M Ervasti
- Department of Biochemistry and Molecular Biology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Andrei I Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
9
|
Loss of serum response factor in mature neurons in the dentate gyrus alters the morphology of dendritic spines and hippocampus-dependent behavioral tasks. Brain Struct Funct 2019; 224:2691-2701. [PMID: 31375980 PMCID: PMC6778544 DOI: 10.1007/s00429-019-01925-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 07/19/2019] [Indexed: 12/12/2022]
Abstract
Serum response factor (SRF) is a major transcription factor that regulates the expression of several plasticity-associated genes in the brain. Although the developmental expression of SRF in excitatory neurons is crucial for establishing proper hippocampal circuitry, no substantial evidence of its role in unstimulated mature neurons has been provided. The present study used time-controlled, conditional SRF knockout mice and found that the lack of SRF in adult neurons led to decreased actin levels and inactivation of the actin-severing protein cofilin 1 through its increase in phosphorylation at Ser3. The augmentation of cofilin 1 phosphorylation correlated with an alteration of dendritic spine morphology in the dentate gyrus, which was reflected by an increase in the number of spines that clustered into the long-spine category. The changes in spine morphology coincided with a lower amplitude and frequency of miniature excitatory postsynaptic currents. Moreover, SRF knockout animals were hyperactive and exhibited impairments in hippocampus-dependent behaviors, such as digging, marble burying, and nesting. Altogether, our data indicate that the adult deletion of neuronal SRF leads to alterations of spine morphology and function and hippocampus-dependent behaviors. Thus, SRF deletion in adult neurons recapitulates some aspects of morphological, electrophysiological, and behavioral changes that are observed in such psychiatric disorders as schizophrenia and autism spectrum disorders.
Collapse
|
10
|
Multiple roles of the actin and microtubule-regulating formins in the developing brain. Neurosci Res 2019; 138:59-69. [DOI: 10.1016/j.neures.2018.09.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 08/22/2018] [Accepted: 08/23/2018] [Indexed: 01/08/2023]
|
11
|
β-actin regulates a heterochromatin landscape essential for optimal induction of neuronal programs during direct reprograming. PLoS Genet 2018; 14:e1007846. [PMID: 30557298 PMCID: PMC6312353 DOI: 10.1371/journal.pgen.1007846] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 12/31/2018] [Accepted: 11/21/2018] [Indexed: 02/02/2023] Open
Abstract
During neuronal development, β-actin serves an important role in growth cone mediated axon guidance. Consistent with this notion, in vivo ablation of the β-actin gene leads to abnormalities in the nervous system. However, whether β-actin is involved in the regulation of neuronal gene programs is not known. In this study, we directly reprogramed β-actin+/+ WT, β-actin+/- HET and β-actin-/- KO mouse embryonic fibroblast (MEFs) into chemically induced neurons (CiNeurons). Using RNA-seq analysis, we profiled the transcriptome changes among the CiNeurons. We discovered that induction of neuronal gene programs was impaired in KO CiNeurons in comparison to WT ones, whereas HET CiNeurons showed an intermediate levels of induction. ChIP-seq analysis of heterochromatin markers demonstrated that the impaired expression of neuronal gene programs correlated with the elevated H3K9 and H3K27 methylation levels at gene loci in β-actin deficient MEFs, which is linked to the loss of chromatin association of the BAF complex ATPase subunit Brg1. Together, our study shows that heterochromatin alteration in β-actin null MEFs impedes the induction of neuronal gene programs during direct reprograming. These findings are in line with the notion that H3K9Me3-based heterochromatin forms a major epigenetic barrier during cell fate change. Although β-actin plays an important role in growth cone mediated axon guidance in neurons, the potential role of β-actin in controlling neuron differentiation remains unknown. Here, we converted β-actin+/+ WT, β-actin+/- HET and β-actin-/- KO mouse embryonic fibroblast (MEFs) into chemically induced neurons (CiNeurons) by direct reprograming. We found that the up-regulation of neuronal programs was impaired in β-actin-/- CiNeurons in comparison to WT ones. β-actin+/- HET CiNeurons showed an intermediate level of neuronal program expression, suggesting that β-actin dosage plays an important role during direct neuronal reprograming. Importantly, the impaired up-regulation of neuron-related genes was associated with the elevated H3K9 and H3K27 methylation levels at gene loci in KO MEFs. These epigenetic changes were accompanied by the impaired chromatin association of Brg1-containing chromatin remodeling BAF complex in β-actin null cells. Together our study demonstrates that β-actin is required for the optimal induction of neuronal gene programs during direct reprograming by presetting a favorable chromatin status.
Collapse
|
12
|
Abstract
The highly similar cytoplasmic β- and γ-actins differ by only four functionally similar amino acids, yet previous in vitro and in vivo data suggest that they support unique functions due to striking phenotypic differences between Actb and Actg1 null mouse and cell models. To determine whether the four amino acid variances were responsible for the functional differences between cytoplasmic actins, we gene edited the endogenous mouse Actb locus to translate γ-actin protein. The resulting mice and primary embryonic fibroblasts completely lacked β-actin protein, but were viable and did not present with the most overt and severe cell and organismal phenotypes observed with gene knockout. Nonetheless, the edited mice exhibited progressive high-frequency hearing loss and degeneration of actin-based stereocilia as previously reported for hair cell-specific Actb knockout mice. Thus, β-actin protein is not required for general cellular functions, but is necessary to maintain auditory stereocilia.
Collapse
|
13
|
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: 2.0] [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
|
14
|
Self-Complementary AAV9 Gene Delivery Partially Corrects Pathology Associated with Juvenile Neuronal Ceroid Lipofuscinosis (CLN3). J Neurosci 2017; 36:9669-82. [PMID: 27629717 DOI: 10.1523/jneurosci.1635-16.2016] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 07/29/2016] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Juvenile neuronal ceroid lipofuscinosis (JNCL) is a fatal lysosomal storage disease caused by autosomal-recessive mutations in CLN3 for which no treatment exists. Symptoms appear between 5 and 10 years of age, beginning with blindness and seizures, followed by progressive cognitive and motor decline and premature death (late teens to 20s). We explored a gene delivery approach for JNCL by generating two self-complementary adeno-associated virus 9 (scAAV9) constructs to address CLN3 dosage effects using the methyl-CpG-binding protein 2 (MeCP2) and β-actin promoters to drive low versus high transgene expression, respectively. This approach was based on the expectation that low CLN3 levels are required for cellular homeostasis due to minimal CLN3 expression postnatally, although this had not yet been demonstrated in vivo One-month-old Cln3(Δex7/8) mice received one systemic (intravenous) injection of scAAV9/MeCP2-hCLN3 or scAAV9/β-actin-hCLN3, with green fluorescent protein (GFP)-expressing viruses as controls. A promoter-dosage effect was observed in all brain regions examined, in which hCLN3 levels were elevated 3- to 8-fold in Cln3(Δex7/8) mice receiving scAAV9/β-actin-hCLN3 versus scAAV9/MeCP2-hCLN3. However, a disconnect occurred between CLN3 levels and disease improvement, because only the scAAV9 construct driving low CLN3 expression (scAAV9/MeCP2-hCLN3) corrected motor deficits and attenuated microglial and astrocyte activation and lysosomal pathology. This may have resulted from preferential promoter usage because transgene expression after intravenous scAAV9/MeCP2-GFP injection was primarily detected in NeuN(+) neurons, whereas scAAV9/β-actin-GFP drove transgene expression in GFAP(+) astrocytes. This is the first demonstration of a systemic delivery route to restore CLN3 in vivo using scAAV9 and highlights the importance of promoter selection for disease modification in juvenile animals. SIGNIFICANCE STATEMENT Juvenile neuronal ceroid lipofuscinosis (JNCL) is a fatal lysosomal storage disease caused by CLN3 mutations. We explored a gene delivery approach using two self-complementary adeno-associated virus 9 (scAAV9) constructs to address CLN3 dosage effects using the methyl-CpG-binding protein 2 (MeCP2) and β-actin promoters. hCLN3 levels were elevated 3- to 8-fold in Cln3(Δex7/8) mice receiving scAAV9/β-actin-hCLN3 versus scAAV9/MeCP2-hCLN3 after a single systemic injection. However, only scAAV9/MeCP2-hCLN3 corrected motor deficits and attenuated glial activation and lysosomal pathology. This may reflect preferential promoter usage because transgene expression with scAAV9/MeCP2-green fluorescent protein (GFP) was primarily in neurons, whereas scAAV9/β-actin-GFP drove transgene expression in astrocytes. This is the first demonstration of systemic delivery for CLN3 using scAAV9 and highlights the importance of promoter selection for disease modification in juvenile animals.
Collapse
|
15
|
Moradi M, Sivadasan R, Saal L, Lüningschrör P, Dombert B, Rathod RJ, Dieterich DC, Blum R, Sendtner M. Differential roles of α-, β-, and γ-actin in axon growth and collateral branch formation in motoneurons. J Cell Biol 2017; 216:793-814. [PMID: 28246119 PMCID: PMC5346967 DOI: 10.1083/jcb.201604117] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 11/11/2016] [Accepted: 01/17/2017] [Indexed: 12/17/2022] Open
Abstract
α-, β-, and γ-actin differentially regulate cytoskeletal dynamics and stability in axons of motoneurons. Locally translated α-actin contributes to stable actin filaments in axonal branches, whereas β- and γ-actin give rise to highly dynamic filaments that modulate growth cone dynamics. Axonal branching and terminal arborization are fundamental events during the establishment of synaptic connectivity. They are triggered by assembly of actin filaments along axon shafts giving rise to filopodia. The specific contribution of the three actin isoforms, Actα, Actβ, and Actγ, to filopodia stability and dynamics during this process is not well understood. Here, we report that Actα, Actβ, and Actγ isoforms are expressed in primary mouse motoneurons and their transcripts are translocated into axons. shRNA-mediated depletion of Actα reduces axonal filopodia dynamics and disturbs collateral branch formation. Knockdown of Actβ reduces dynamic movements of growth cone filopodia and impairs presynaptic differentiation. Ablation of Actβ or Actγ leads to compensatory up-regulation of the two other isoforms, which allows maintenance of total actin levels and preserves F-actin polymerization. Collectively, our data provide evidence for specific roles of different actin isoforms in spatial regulation of actin dynamics and stability in axons of developing motoneurons.
Collapse
Affiliation(s)
- Mehri Moradi
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| | - Rajeeve Sivadasan
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| | - Lena Saal
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| | - Patrick Lüningschrör
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| | - Benjamin Dombert
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| | - Reena Jagdish Rathod
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| | - Daniela C Dieterich
- Institute for Pharmacology and Toxicology, Medical Faculty, University of Magdeburg, 39120 Magdeburg, Germany.,Center for Behavioral Brain Sciences, Medical Faculty, University of Magdeburg, 39120 Magdeburg, Germany
| | - Robert Blum
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| |
Collapse
|
16
|
Patrinostro X, O'Rourke AR, Chamberlain CM, Moriarity BS, Perrin BJ, Ervasti JM. Relative importance of β cyto- and γ cyto-actin in primary mouse embryonic fibroblasts. Mol Biol Cell 2017; 28:771-782. [PMID: 28077619 PMCID: PMC5349784 DOI: 10.1091/mbc.e16-07-0503] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 11/29/2016] [Accepted: 01/05/2017] [Indexed: 12/14/2022] Open
Abstract
The highly homologous β (βcyto) and γ (γcyto) cytoplasmic actins are hypothesized to carry out both redundant and unique essential functions, but studies using targeted gene knockout and siRNA-mediated transcript knockdown to examine βcyto- and γcyto-isoform--specific functions in various cell types have yielded conflicting data. Here we quantitatively characterized actin transcript and protein levels, as well as cellular phenotypes, in both gene- and transcript-targeted primary mouse embryonic fibroblasts. We found that the smooth muscle αsm-actin isoform was the dominantly expressed actin isoform in WT primary fibroblasts and was also the most dramatically up-regulated in primary βcyto- or β/γcyto-actin double-knockout fibroblasts. Gene targeting of βcyto-actin, but not γcyto-actin, led to greatly decreased cell proliferation, decreased levels of cellular ATP, and increased serum response factor signaling in primary fibroblasts, whereas immortalization induced by SV40 large T antigen supported fibroblast proliferation in the absence of βcyto-actin. Consistent with in vivo gene-targeting studies in mice, both gene- and transcript-targeting approaches demonstrate that the loss of βcyto-actin protein is more disruptive to primary fibroblast function than is the loss of γcyto-actin.
Collapse
Affiliation(s)
- Xiaobai Patrinostro
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455
| | - Allison R O'Rourke
- Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota, Minneapolis, MN 55455
| | - Christopher M Chamberlain
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455
| | | | - Benjamin J Perrin
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46022
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455 .,Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
17
|
Yates TM, Turner CL, Firth HV, Berg J, Pilz DT. Baraitser-Winter cerebrofrontofacial syndrome. Clin Genet 2016; 92:3-9. [PMID: 27625340 DOI: 10.1111/cge.12864] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 09/07/2016] [Accepted: 09/07/2016] [Indexed: 01/31/2023]
Abstract
Baraitser-Winter cerebrofrontofacial syndrome (BWCFF) (BRWS; MIM #243310, 614583) is a rare developmental disorder affecting multiple organ systems. It is characterised by intellectual disability (mild to severe) and distinctive facial appearance (metopic ridging/trigonocephaly, bilateral ptosis, hypertelorism). The additional presence of cortical malformations (pachygyria/lissencephaly) and ocular colobomata are also suggestive of this syndrome. Other features include moderate short stature, contractures, congenital cardiac disease and genitourinary malformations. BWCFF is caused by missense mutations in the cytoplasmic beta- and gamma-actin genes ACTB and ACTG1. We provide an overview of the clinical characteristics (including some novel findings in four recently diagnosed patients), diagnosis, management, mutation spectrum and genetic counselling.
Collapse
Affiliation(s)
- T M Yates
- Department of Medical Genetics, University of Glasgow, Glasgow, UK
| | - C L Turner
- Peninsula Clinical Genetics Service, Royal Devon and Exeter Hospital, Exeter, UK
| | - H V Firth
- Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, UK
| | - J Berg
- Department of Clinical Genetics, Ninewells Hospital, Dundee, UK
| | - D T Pilz
- West of Scotland Genetics Service, Queen Elizabeth University Hospital, Glasgow, UK
| |
Collapse
|
18
|
Uemura T, Mori T, Kurihara T, Kawase S, Koike R, Satoga M, Cao X, Li X, Yanagawa T, Sakurai T, Shindo T, Tabuchi K. Fluorescent protein tagging of endogenous protein in brain neurons using CRISPR/Cas9-mediated knock-in and in utero electroporation techniques. Sci Rep 2016; 6:35861. [PMID: 27782168 PMCID: PMC5080626 DOI: 10.1038/srep35861] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 10/05/2016] [Indexed: 12/23/2022] Open
Abstract
Genome editing is a powerful technique for studying gene functions. CRISPR/Cas9-mediated gene knock-in has recently been applied to various cells and organisms. Here, we successfully knocked in an EGFP coding sequence at the site immediately after the first ATG codon of the β-actin gene in neurons in the brain by the combined use of the CRISPR/Cas9 system and in utero electroporation technique, resulting in the expression of the EGFP-tagged β-actin protein in cortical layer 2/3 pyramidal neurons. We detected EGFP fluorescence signals in the soma and neurites of EGFP knock-in neurons. These signals were particularly abundant in the head of dendritic spines, corresponding to the localization of the endogenous β-actin protein. EGFP knock-in neurons showed no detectable changes in spine density and basic electrophysiological properties. In contrast, exogenously overexpressed EGFP-β-actin showed increased spine density and EPSC frequency, and changed resting membrane potential. Thus, our technique provides a potential tool to elucidate the localization of various endogenous proteins in neurons by epitope tagging without altering neuronal and synaptic functions. This technique can be also useful for introducing a specific mutation into genes to study the function of proteins and genomic elements in brain neurons.
Collapse
Affiliation(s)
- Takeshi Uemura
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano 390-8621, Japan
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano 390-8621, Japan
- CREST, JST, Saitama 332-0012, Japan
| | - Takuma Mori
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano 390-8621, Japan
| | - Taiga Kurihara
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano 390-8621, Japan
| | - Shiori Kawase
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano 390-8621, Japan
- CREST, JST, Saitama 332-0012, Japan
| | - Rie Koike
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano 390-8621, Japan
- CREST, JST, Saitama 332-0012, Japan
| | - Michiru Satoga
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano 390-8621, Japan
| | - Xueshan Cao
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano 390-8621, Japan
| | - Xue Li
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano 390-8621, Japan
| | - Toru Yanagawa
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Takayuki Sakurai
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano 390-8621, Japan
| | - Takayuki Shindo
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano 390-8621, Japan
| | - Katsuhiko Tabuchi
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano 390-8621, Japan
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano 390-8621, Japan
- PRESTO, JST, Saitama 332-0012, Japan
| |
Collapse
|
19
|
Zhen Y, Li W. Impairment of autophagosome-lysosome fusion in the buff mutant mice with the VPS33A(D251E) mutation. Autophagy 2016; 11:1608-22. [PMID: 26259518 DOI: 10.1080/15548627.2015.1072669] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The HOPS (homotypic fusion and protein sorting) complex functions in endocytic and autophagic pathways in both lower eukaryotes and mammalian cells through its involvement in fusion events between endosomes and lysosomes or autophagosomes and lysosomes. However, the differential molecular mechanisms underlying these fusion processes are largely unknown. Buff (bf) is a mouse mutant that carries an Asp251-to-Glu point mutation (D251E) in the VPS33A protein, a tethering protein and a core subunit of the HOPS complex. Bf mice showed impaired spontaneous locomotor activity, motor learning, and autophagic activity. Although the gross anatomy of the brain was apparently normal, the number of Purkinje cells was significantly reduced. Furthermore, we found that fusion between autophagosomes and lysosomes was defective in bf cells without compromising the endocytic pathway. The direct association of mutant VPS33A(D251E) with the autophagic SNARE complex, STX17 (syntaxin 17)-VAMP8-SNAP29, was enhanced. In addition, the VPS33A(D251E) mutation enhanced interactions with other HOPS subunits, namely VPS41, VPS39, VPS18, and VPS11, except for VPS16. Reduction of the interactions between VPS33A(Y440D) and several other HOPS subunits led to decreased association with STX17. These results suggest that the VPS33A(D251E) mutation plays dual roles by increasing the HOPS complex assembly and its association with the autophagic SNARE complex, which selectively affects the autophagosome-lysosome fusion that impairs basal autophagic activity and induces Purkinje cell loss.
Collapse
Affiliation(s)
- Yuanli Zhen
- a State Key Laboratory of Molecular Developmental Biology; Institute of Genetics & Developmental Biology; Chinese Academy of Sciences ; Beijing , China.,b University of Chinese Academy of Sciences ; Beijing , China
| | - Wei Li
- a State Key Laboratory of Molecular Developmental Biology; Institute of Genetics & Developmental Biology; Chinese Academy of Sciences ; Beijing , China.,c Center of Alzheimer Disease; Beijing Institute for Brain Disorders ; Beijing China.,d Beijing Children's Hospital; Capital Medical University ; Beijing , China
| |
Collapse
|
20
|
Abstract
Actin is the central building block of the actin cytoskeleton, a highly regulated filamentous network enabling dynamic processes of cells and simultaneously providing structure. Mammals have six actin isoforms that are very conserved and thus share common functions. Tissue-specific expression in part underlies their differential roles, but actin isoforms also coexist in various cell types and tissues, suggesting specific functions and preferential interaction partners. Gene deletion models, antibody-based staining patterns, gene silencing effects, and the occurrence of isoform-specific mutations in certain diseases have provided clues for specificity on the subcellular level and its consequences on the organism level. Yet, the differential actin isoform functions are still far from understood in detail. Biochemical studies on the different isoforms in pure form are just emerging, and investigations in cells have to deal with a complex and regulated system, including compensatory actin isoform expression.
Collapse
Affiliation(s)
- Christophe Ampe
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, A. Baertsoenkaai 3, 9000, Ghent, Belgium.
| | - Marleen Van Troys
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, A. Baertsoenkaai 3, 9000, Ghent, Belgium
| |
Collapse
|
21
|
Snow WM, Pahlavan PS, Djordjevic J, McAllister D, Platt EE, Alashmali S, Bernstein MJ, Suh M, Albensi BC. Morris Water Maze Training in Mice Elevates Hippocampal Levels of Transcription Factors Nuclear Factor (Erythroid-derived 2)-like 2 and Nuclear Factor Kappa B p65. Front Mol Neurosci 2015; 8:70. [PMID: 26635523 PMCID: PMC4649017 DOI: 10.3389/fnmol.2015.00070] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/03/2015] [Indexed: 11/30/2022] Open
Abstract
Research has identified several transcription factors that regulate activity-dependent plasticity and memory, with cAMP-response element binding protein (CREB) being the most well-studied. In neurons, CREB activation is influenced by the transcription factor nuclear factor kappa B (NF-κB), considered central to immunity but more recently implicated in memory. The transcription factor early growth response-2 (Egr-2), an NF-κB gene target, is also associated with learning and memory. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2), an antioxidant transcription factor linked to NF-κB in pathological conditions, has not been studied in normal memory. Given that numerous transcription factors implicated in activity-dependent plasticity demonstrate connections to NF-κB, this study simultaneously evaluated protein levels of NF-κB, CREB, Egr-2, Nrf2, and actin in hippocampi from young (1 month-old) weanling CD1 mice after training in the Morris water maze, a hippocampal-dependent spatial memory task. After a 6-day acquisition period, time to locate the hidden platform decreased in the Morris water maze. Mice spent more time in the target vs. non-target quadrants of the maze, suggestive of recall of the platform location. Western blot data revealed a decrease in NF-κB p50 protein after training relative to controls, whereas NF-κB p65, Nrf2 and actin increased. Nrf2 levels were correlated with platform crosses in nearly all tested animals. These data demonstrate that training in a spatial memory task results in alterations in and associations with particular transcription factors in the hippocampus, including upregulation of NF-κB p65 and Nrf2. Training-induced increases in actin protein levels caution against its use as a loading control in immunoblot studies examining activity-dependent plasticity, learning, and memory.
Collapse
Affiliation(s)
- Wanda M Snow
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research Winnipeg, MB, Canada ; Faculty of Health Sciences, Department of Pharmacology and Therapeutics, College of Medicine, University of Manitoba Winnipeg, MB, Canada
| | - Payam S Pahlavan
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research Winnipeg, MB, Canada ; Faculty of Health Sciences, Department of Pharmacology and Therapeutics, College of Medicine, University of Manitoba Winnipeg, MB, Canada
| | - Jelena Djordjevic
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research Winnipeg, MB, Canada ; Faculty of Health Sciences, Department of Pharmacology and Therapeutics, College of Medicine, University of Manitoba Winnipeg, MB, Canada
| | - Danielle McAllister
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research Winnipeg, MB, Canada
| | - Eric E Platt
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research Winnipeg, MB, Canada
| | - Shoug Alashmali
- Department of Human Nutritional Sciences, University of Manitoba Winnipeg, MB, Canada
| | - Michael J Bernstein
- Department of Psychological and Social Sciences, Pennsylvania State University Abington Abington, PA, USA
| | - Miyoung Suh
- Department of Human Nutritional Sciences, University of Manitoba Winnipeg, MB, Canada
| | - Benedict C Albensi
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research Winnipeg, MB, Canada ; Faculty of Health Sciences, Department of Pharmacology and Therapeutics, College of Medicine, University of Manitoba Winnipeg, MB, Canada ; Faculty of Engineering, Department of Electrical and Computer Engineering, University of Manitoba Winnipeg, MB, Canada
| |
Collapse
|
22
|
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: 2.9] [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
|
23
|
Whish S, Dziegielewska KM, Møllgård K, Noor NM, Liddelow SA, Habgood MD, Richardson SJ, Saunders NR. The inner CSF-brain barrier: developmentally controlled access to the brain via intercellular junctions. Front Neurosci 2015; 9:16. [PMID: 25729345 PMCID: PMC4325900 DOI: 10.3389/fnins.2015.00016] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 01/12/2015] [Indexed: 12/04/2022] Open
Abstract
In the adult the interface between the cerebrospinal fluid and the brain is lined by the ependymal cells, which are joined by gap junctions. These intercellular connections do not provide a diffusional restrain between the two compartments. However, during development this interface, initially consisting of neuroepithelial cells and later radial glial cells, is characterized by “strap” junctions, which limit the exchange of different sized molecules between cerebrospinal fluid and the brain parenchyma. Here we provide a systematic study of permeability properties of this inner cerebrospinal fluid-brain barrier during mouse development from embryonic day, E17 until adult. Results show that at fetal stages exchange across this barrier is restricted to the smallest molecules (286Da) and the diffusional restraint is progressively removed as the brain develops. By postnatal day P20, molecules the size of plasma proteins (70 kDa) diffuse freely. Transcriptomic analysis of junctional proteins present in the cerebrospinal fluid-brain interface showed expression of adherens junctional proteins, actins, cadherins and catenins changing in a development manner consistent with the observed changes in the permeability studies. Gap junction proteins were only identified in the adult as was claudin-11. Immunohistochemistry was used to localize at the cellular level some of the adherens junctional proteins of genes identified from transcriptomic analysis. N-cadherin, β - and α-catenin immunoreactivity was detected outlining the inner CSF-brain interface from E16; most of these markers were not present in the adult ependyma. Claudin-5 was present in the apical-most part of radial glial cells and in endothelial cells in embryos, but only in endothelial cells including plexus endothelial cells in adults. Claudin-11 was only immunopositive in the adult, consistent with results obtained from transcriptomic analysis. These results provide information about physiological, molecular and morphological-related permeability changes occurring at the inner cerebrospinal fluid-brain barrier during brain development.
Collapse
Affiliation(s)
- Sophie Whish
- Department of Pharmacology and Therapeutics, University of Melbourne Parkville, VIC, Australia
| | | | - Kjeld Møllgård
- Department of Cellular and Molecular Medicine, Institute of Cellular and Molecular Medicine, University of Copenhagen Copenhagen, Denmark
| | - Natassya M Noor
- Department of Pharmacology and Therapeutics, University of Melbourne Parkville, VIC, Australia
| | - Shane A Liddelow
- Department of Pharmacology and Therapeutics, University of Melbourne Parkville, VIC, Australia ; Department of Neurobiology, Stanford University Palo Alto, CA, USA
| | - Mark D Habgood
- Department of Pharmacology and Therapeutics, University of Melbourne Parkville, VIC, Australia
| | | | - Norman R Saunders
- Department of Pharmacology and Therapeutics, University of Melbourne Parkville, VIC, Australia
| |
Collapse
|
24
|
Abstract
Neurons begin their life as simple spheres, but can ultimately assume an elaborate morphology with numerous, highly arborized dendrites, and long axons. This is achieved via an astounding developmental progression which is dependent upon regulated assembly and dynamics of the cellular cytoskeleton. As neurites emerge out of the soma, neurons break their spherical symmetry and begin to acquire the morphological features that define their structure and function. Neurons regulate their cytoskeleton to achieve changes in cell shape, velocity, and direction as they migrate, extend neurites, and polarize. Of particular importance, the organization and dynamics of actin and microtubules directs the migration and morphogenesis of neurons. This review focuses on the regulation of intrinsic properties of the actin and microtubule cytoskeletons and how specific cytoskeletal structures and dynamics are associated with the earliest phase of neuronal morphogenesis—neuritogenesis.
Collapse
|
25
|
Genomic cloning and promoter analysis of the β-actin gene from Korean rose bitterling (Rhodeus uyekii). Genes Genomics 2014. [DOI: 10.1007/s13258-014-0221-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
26
|
Ghorbani M, Mohamadpour AH, Mehri S, Abnous K, Hassanzadeh-Khayyat M. Proteomics screening of molecular targets of granulocyte colony stimulating factor in the mouse brain and PC12 cell line. Life Sci 2014; 108:44-7. [PMID: 24831556 DOI: 10.1016/j.lfs.2014.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 04/10/2014] [Accepted: 05/02/2014] [Indexed: 10/25/2022]
Abstract
AIMS Granulocyte colony stimulating factor (G-CSF), a new neuroprotective agent, binds to its specific receptors in the brain. In this study we hypothesized that at least a part of G-CSF's neuroprotective effect may be mediated through its interaction with other proteins in the brain. MAIN METHODS Using an immunoprecipitation (IP) kit, at first the antibody of G-CSF was covalently crosslinked to protein A/G agarose. Then the mouse brain or PC12 cell lysate mixed with G-CSF was added to the agarose beads plus antibody. After immunoaffinity isolation of target proteins, gel electrophoresis was performed and protein bands were identified using MALDI-TOF/TOF and MASCOT software. KEY FINDINGS Our data show that G-CSF physically binds to cellular proteins like sodium/potassium-transporting ATPase, beta actin, aldehyde dehydrogenase, regucalcin and glutathione-s-transferase. These proteins are involved in membrane transportation, cell structure, signal transduction, enzymes involve in calcium related cell signaling and redox homeostasis. SIGNIFICANCE Interaction of G-CSF with these proteins can explain some of its pharmacological effects in the CNS.
Collapse
Affiliation(s)
- Maryam Ghorbani
- Department of Pharmacology and Toxicology, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amir Hooshang Mohamadpour
- Pharmaceutical Research Center, Department of Clinical Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soghra Mehri
- Pharmaceutical Research Center, Department of Pharmacodynamy and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Department of Medicinal Chemistry, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Hassanzadeh-Khayyat
- Pharmaceutical Research Center, Department of Medicinal Chemistry, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
27
|
Tondeleir D, Noelanders R, Bakkali K, Ampe C. Beta-actin is required for proper mouse neural crest ontogeny. PLoS One 2014; 9:e85608. [PMID: 24409333 PMCID: PMC3883714 DOI: 10.1371/journal.pone.0085608] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 12/05/2013] [Indexed: 02/06/2023] Open
Abstract
The mouse genome consists of six functional actin genes of which the expression patterns are temporally and spatially regulated during development and in the adult organism. Deletion of beta-actin in mouse is lethal during embryonic development, although there is compensatory expression of other actin isoforms. This suggests different isoform specific functions and, more in particular, an important function for beta-actin during early mammalian development. We here report a role for beta-actin during neural crest ontogeny. Although beta-actin null neural crest cells show expression of neural crest markers, less cells delaminate and their migration arrests shortly after. These phenotypes were associated with elevated apoptosis levels in neural crest cells, whereas proliferation levels were unchanged. Specifically the pre-migratory neural crest cells displayed higher levels of apoptosis, suggesting increased apoptosis in the neural tube accounts for the decreased amount of migrating neural crest cells seen in the beta-actin null embryos. These cells additionally displayed a lack of membrane bound N-cadherin and dramatic decrease in cadherin-11 expression which was more pronounced in the pre-migratory neural crest population, potentially indicating linkage between the cadherin-11 expression and apoptosis. By inhibiting ROCK ex vivo, the knockout neural crest cells regained migratory capacity and cadherin-11 expression was upregulated. We conclude that the presence of beta-actin is vital for survival, specifically of pre-migratory neural crest cells, their proper emigration from the neural tube and their subsequent migration. Furthermore, the absence of beta-actin affects cadherin-11 and N-cadherin function, which could partly be alleviated by ROCK inhibition, situating the Rho-ROCK signaling in a feedback loop with cadherin-11.
Collapse
Affiliation(s)
- Davina Tondeleir
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Rivka Noelanders
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Karima Bakkali
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Christophe Ampe
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- * E-mail:
| |
Collapse
|
28
|
Kalous A, Stake JI, Yisraeli JK, Holt CE. RNA-binding protein Vg1RBP regulates terminal arbor formation but not long-range axon navigation in the developing visual system. Dev Neurobiol 2013; 74:303-18. [PMID: 23853158 DOI: 10.1002/dneu.22110] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 06/28/2013] [Accepted: 07/05/2013] [Indexed: 12/21/2022]
Abstract
Local synthesis of β-actin is required for attractive turning responses to guidance cues of growth cones in vitro but its functional role in axon guidance in vivo is poorly understood. The transport and translation of β-actin mRNA is regulated by the RNA-binding protein, Vg1RBP (zipcode-binding protein-1). To examine whether Vg1RBP plays a role in axon navigation in vivo, we disrupted Vg1RBP function in embryonic Xenopus laevis retinal ganglion cells by expressing a dominant-negative Vg1RBP and by antisense morpholino knockdown. We found that attractive turning to a netrin-1 gradient in vitro was abolished in Vg1RBP-deficient axons but, surprisingly, the long-range navigation from the retina to the optic tectum was unaffected. Within the tectum, however, the branching and complexity of axon terminals were significantly reduced. High-resolution time-lapse imaging of axon terminals in vivo revealed that Vg1RBP-GFP-positive granules accumulate locally in the axon shaft immediately preceding the emergence a filopodial-like protrusion. Comparative analysis of branch dynamics showed that Vg1RBP-deficient axons extend far fewer filopodial-like protrusions than control axons and indicate that Vg1RBP promotes filopodial formation, an essential step in branch initiation. Our findings show that Vg1RBP is required for terminal arborization but not long-range axon navigation and suggest that Vg1RBP-regulated mRNA translation promotes synaptic complexity.
Collapse
Affiliation(s)
- Adrianna Kalous
- Department of Physiology, Development, and Neuroscience, University of Cambridge, CB2 3DY, United Kingdom
| | | | | | | |
Collapse
|
29
|
Jablonka S, Dombert B, Asan E, Sendtner M. Mechanisms for axon maintenance and plasticity in motoneurons: alterations in motoneuron disease. J Anat 2013; 224:3-14. [PMID: 24007389 DOI: 10.1111/joa.12097] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2013] [Indexed: 12/12/2022] Open
Abstract
In motoneuron disease and other neurodegenerative disorders, the loss of synapses and axon branches occurs early but is compensated by sprouting of neighboring axon terminals. Defective local axonal signaling for maintenance and dynamics of the axonal microtubule and actin cytoskeleton plays a central role in this context. The molecular mechanisms that lead to defective cytoskeleton architecture in two mouse models of motoneuron disease are summarized and discussed in this manuscript. In the progressive motor neuropathy (pmn) mouse model of motoneuron disease that is caused by a mutation in the tubulin-specific chaperone E gene, death of motoneuron cell bodies appears as a consequence of axonal degeneration. Treatment with bcl-2 overexpression or with glial-derived neurotrophic factor prevents loss of motoneuron cell bodies but does not influence the course of disease. In contrast, treatment with ciliary neurotrophic factor (CNTF) significantly delays disease onset and prolongs survival of pmn mice. This difference is due to the activation of Stat-3 via the CNTF receptor complex in axons of pmn mutant motoneurons. Most of the activated Stat-3 protein is not transported to the nucleus to activate transcription, but interacts locally in axons with stathmin, a protein that destabilizes microtubules. This interaction plays a major role in CNTF signaling for microtubule dynamics in axons. In Smn-deficient mice, a model of spinal muscular atrophy, defects in axonal translocation of β-actin mRNA and possibly other mRNA species have been observed. Moreover, the regulation of local protein synthesis in response to signals from neurotrophic factors and extracellular matrix proteins is altered in motoneurons from this model of motoneuron disease. These findings indicate that local signals are important for maintenance and plasticity of axonal branches and neuromuscular endplates, and that disturbances in these signaling mechanisms could contribute to the pathophysiology of motoneuron diseases.
Collapse
Affiliation(s)
- Sibylle Jablonka
- Institute for Clinical Neurobiology, University Hospital, University of Wuerzburg, Wuerzburg, Germany
| | | | | | | |
Collapse
|
30
|
Abstract
Increasing evidence points to the importance of local protein synthesis for axonal growth and responses to axotomy, yet there is little insight into the functions of individual locally synthesized proteins. We recently showed that expression of a reporter mRNA with the axonally localizing β-actin mRNA 3'UTR competes with endogenous β-actin and GAP-43 mRNAs for binding to ZBP1 and axonal localization in adult sensory neurons (Donnelly et al., 2011). Here, we show that the 3'UTR of GAP-43 mRNA can deplete axons of endogenous β-actin mRNA. We took advantage of this 3'UTR competition to address the functions of axonally synthesized β-actin and GAP-43 proteins. In cultured rat neurons, increasing axonal synthesis of β-actin protein while decreasing axonal synthesis of GAP-43 protein resulted in short highly branched axons. Decreasing axonal synthesis of β-actin protein while increasing axonal synthesis of GAP-43 protein resulted in long axons with few branches. siRNA-mediated depletion of overall GAP-43 mRNA from dorsal root ganglia (DRGs) decreased the length of axons, while overall depletion of β-actin mRNA from DRGs decreased the number of axon branches. These deficits in axon growth could be rescued by transfecting with siRNA-resistant constructs encoding β-actin or GAP-43 proteins, but only if the mRNAs were targeted for axonal transport. Finally, in ovo electroporation of axonally targeted GAP-43 mRNA increased length and axonally targeted β-actin mRNA increased branching of sensory axons growing into the chick spinal cord. These studies indicate that axonal translation of β-actin mRNA supports axon branching and axonal translation of GAP-43 mRNA supports elongating growth.
Collapse
|
31
|
Actin isoforms in neuronal development and function. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 301:157-213. [PMID: 23317819 DOI: 10.1016/b978-0-12-407704-1.00004-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The actin cytoskeleton contributes directly or indirectly to nearly every aspect of neuronal development and function. This diversity of functions is often attributed to actin regulatory proteins, although how the composition of the actin cytoskeleton itself may influence its function is often overlooked. In neurons, the actin cytoskeleton is composed of two distinct isoforms, β- and γ-actin. Functions for β-actin have been investigated in axon guidance, synaptogenesis, and disease. Insight from loss-of-function in vivo studies has also revealed novel roles for β-actin in select brain structures and behaviors. Conversely, very little is known regarding functions of γ-actin in neurons. The dysregulation or mutation of both β- and γ-actin has been implicated in multiple human neurological disorders, however, demonstrating the critical importance of these still poorly understood proteins. This chapter highlights what is currently known regarding potential distinct functions for β- and γ-actin in neurons as well as the significant areas that remain unexplored.
Collapse
|
32
|
Lorson MA, Lorson CL. SMN-inducing compounds for the treatment of spinal muscular atrophy. Future Med Chem 2012; 4:2067-84. [PMID: 23157239 PMCID: PMC3589915 DOI: 10.4155/fmc.12.131] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a leading genetic cause of infant mortality. A neurodegenerative disease, it is caused by loss of SMN1, although low, but essential, levels of SMN protein are produced by the nearly identical gene SMN2. While no effective treatment or therapy currently exists, a new wave of therapeutics has rapidly progressed from cell-based and preclinical animal models to the point where clinical trials have initiated for SMA-specific compounds. There are several reasons why SMA has moved relatively rapidly towards novel therapeutics, including: SMA is monogenic; the molecular understanding of SMN gene regulation has been building for nearly 20 years; and all SMA patients retain one or more copies of SMN2 that produces low levels of full-length, fully functional SMN protein. This review primarily focuses upon the biology behind the disease and examines SMN1- and SMN2-targeted therapeutics.
Collapse
Affiliation(s)
- Monique A Lorson
- Department of Veterinary Pathobiology, Bond Life Sciences Center, Room 440C, University of Missouri, MO 65211 USA
| | - Christian L Lorson
- Department of Veterinary Pathobiology, Bond Life Sciences Center, Room 471G, University of Missouri, Columbia, MO 65211, USA
- Department of Molecular Microbiology & Immunology, University of Missouri, MO, USA
| |
Collapse
|
33
|
Rathod R, Havlicek S, Frank N, Blum R, Sendtner M. Laminin induced local axonal translation of β-actin mRNA is impaired in SMN-deficient motoneurons. Histochem Cell Biol 2012; 138:737-48. [PMID: 22810847 DOI: 10.1007/s00418-012-0989-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2012] [Indexed: 01/10/2023]
Abstract
Reduced levels of the SMN (survival of motoneuron) protein cause spinal muscular atrophy, the main form of motoneuron disease in children and young adults. In cultured motoneurons, reduced SMN levels lead to disturbed axon growth that correlates with reduced actin mRNA and protein in growth cones, indicating that anterograde transport and local translation of β-actin mRNA are altered in this disease. However, it is not fully understood how local translation of the β-actin mRNA is regulated in SMN-deficient motoneurons. Here, we established a lentiviral GFP-based reporter construct to monitor local translation of β-actin mRNA. Time-lapse imaging of fluorescence recovery after photobleaching (FRAP) in living motoneurons revealed that β-actin is locally translated in the growth cones of embryonic motoneurons. Interestingly, local translation of the β-actin reporter construct was differentially regulated by various Laminin isoforms, indicating that Laminins provide extracellular cues for the regulation of local translation in growth cones. Notably, local translation of β-actin mRNA was deregulated in motoneurons from a mouse model for the most severe form of SMA (Smn(-/-);SMN2). Taken together our findings suggest that local translation of β-actin in growth cones of motoneurons is regulated by Laminin signalling and that this signalling is disturbed in SMA.
Collapse
Affiliation(s)
- Reena Rathod
- Institute for Clinical Neurobiology, University of Wuerzburg, Building E4, Versbacherstr. 5, 97078, Wuerzburg, Germany
| | | | | | | | | |
Collapse
|